NEPHSAP - Renal Pathology Cases
NEPHSAP - Renal Pathology Cases
NEPHSAP - Renal Pathology Cases
NephSAP
®
Renal Pathology
Guest Co-Editors:
Glen S. Markowitz, MD,
M. Barry Stokes, MD,
Neeraja Kambham, MD,
Leal C. Herlitz, MD, and
Vivette D. D’Agati, MD
■ Editor-in-Chief: Stanley Goldfarb, MD
■ Deputy Editor: Raymond R. Townsend, MD
NephSAP
®
EDITOR-IN-CHIEF
Stanley Goldfarb, MD
University of Pennsylvania Medical School
Preface
Philadelphia, PA NephSAP® is one of the three major publications of the American Society of Nephrology
(ASN). Its primary goals are self-assessment, education, and the provision of Continuing
DEPUTY EDITOR Medical Education (CME) credits and Maintenance of Certification (MOC) credits for
Raymond R. Townsend, MD individuals certified by the American Board of Internal Medicine. Members of the ASN
University of Pennsylvania Medical School
Philadelphia, PA
automatically receive NephSAP with their monthly issue of The Journal of the American
Society of Nephrology (JASN).
MANAGING EDITOR
EDUCATION: Medical and Nephrologic information continually accrues at a rapid pace.
Gisela Deuter, BSN, MSA
Washington, DC
Bombarded from all sides with demands on their time, busy practitioners, academicians, and
trainees at all levels are increasingly challenged to review and understand all this new material.
ASSOCIATE EDITORS Each bimonthly issue of NephSAP is dedicated to a specific theme, i.e., to a specific area
Rajiv Agarwal, MD of clinical nephrology, hypertension, dialysis, and transplantation, and consists of an Editorial,
Indiana University School of Medicine a Syllabus, a Commentary on the Syllabus, and self-assessment questions. Over the course of
Indianapolis, IN
24 months, all clinically relevant and key elements of nephrology will be reviewed and updated.
David J. Cohen, MD
The authors of each issue digest, assimilate, and interpret key publications from the previous
Columbia University
New York, NY issues of other years and integrate this new material with the body of existing information.
Michael J. Choi, MD SELF-ASSESSMENT: Twenty-five single-best-answer questions will follow the 50 to 75 pages
Johns Hopkins University School of Medicine
Baltimore, MD
of Syllabus text. The examination is available online with immediate feedback. Those answer-
Michael Emmett, MD ing ⬎75% correctly will receive CME credit, and receive the answers to all the questions along
Baylor University with brief discussions and an updated bibliography. To help answer the questions, readers may
Dallas, TX go to the ASN web site, where relevant material from UpToDate in nephrology will be posted.
Linda F. Fried, MD, MPH Thus, members will find a new area reviewed every 2 months, and they will be able to test their
University of Pittsburgh understanding with our quiz. This format will help readers stay abreast of developing areas of
Pittsburgh, PA
clinical nephrology, hypertension, dialysis, and transplantation, and the review and update will
Richard J. Glassock, MD
Professor Emeritus, The David Geffen School
support those taking certification and recertification examinations.
of Medicine at the University of California CONTINUING MEDICAL EDUCATION: Most state and local medical agencies as well as
Los Angeles, CA
hospitals are demanding documentation of requisite CME credits for licensure and for staff
Kathleen D. Liu, MD
University of California San Francisco appointments. A maximum of 48 credits annually can be obtained by successfully completing
San Francisco, CA the NephSAP examination. In addition, individuals certified by the American Board of Internal
Kevin J. Martin, MBBCh Medicine may obtain credits towards Maintenance of Certification (MOC) by successfully
St. Louis University School of Medicine completing the self-assessment portion of NephSAP.
St. Louis, MO
Rajnish Mehrotra, MD BOARD CERTIFICATION AND INSERVICE EXAMINATION PREPARATION: Each issue
Harbor UCLA Research and Education Institute will also contain 5 questions and answers examining core topics in the particular discipline
Torrance, CA reviewed in the Syllabus. These questions are designed to provide trainees with challenging
Patrick T. Murray, MD questions to test their knowledge of key areas of nephrology.
University College Dublin
Dublin, Ireland ⬁ This paper meets the requirements of ANSI/NISO Z39.48-1921 (Permanence of Paper),
Patrick H. Nachman, MD effective with July 2002, Vol. 1, No. 1.
University of North Carolina
Chapel Hill, NC GUEST CO-EDITORS
Aldo J. Peixoto, MD
Yale University Vivette D. D’Agati, MD Glen S. Markowitz, MD
West Haven, CT Columbia University Medical Center Columbia University Medical Center
New York, NY New York, NY
Richard H. Sterns, MD
University of Rochester School of Medicine Leal C. Herlitz, MD M. Barry Stokes, MD
and Dentistry Columbia University Medical Center Columbia University Medical Center
Rochester, NY New York, NY New York, NY
John P. Vella, MD
Neeraja Kambham, MD
Maine Medical Center
Portland, ME Stanford University School of Medicine
Stanford, CA
FOUNDING EDITORS
Richard J. Glassock, MD, MACP
Editor-in-Chief Emeritus NephSAP®
Robert G. Narins, MD, MACP ©2011 by The American Society of Nephrology
NephSAP
®
Volume 10, Number 4, July 2011
Renal Pathology
Editorial 271
Problem-based learning- A new approach for NephSAP—
Ronald J. Falk, MD
Syllabus 272
Learning Objectives—
Glen S. Markowitz, MD, M. Barry Stokes, MD,
Neeraja Kambham, MD, Leal C. Herlitz, MD, and
Vivette D. D’Agati, MD
Section 1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .273
Section 2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .279
Section 3 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .285
Section 4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .291
Section 5 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .297
Section 6 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .305
Section 7 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .311
Section 8 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .317
Section 9 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .323
Section 10 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .329
Section 11 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .335
Section 12 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .341
Section 13 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .347
Section 14 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .353
Section 15 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .359
Section 16 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .367
NephSAP
®
Volume 10, Number 4, July 2011
Section 17 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .373
Section 18 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .379
Section 19 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .385
Section 20 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .393
Section 21 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .401
Section 22 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .407
Upcoming Issues
Chronic Kidney Disease and Progression—
Linda F. Fried, MD, and Michael J. Choi, MD . . . . . . .September 2011
Transplantation—
John P. Vella, MD, and David J. Cohen, MD . . . . . . . .November 2011
Pediatric Nephrology—
Howard Trachtman, MD, and Debbie Gipson, MD . . . . . .January 2012
Hypertension—
Raymond R. Townsend, MD, and Aldo J. Piexoto, MD . . . .March 2012
Diabetic Nephropathy—
Kumar Sharma, MD . . . . . . . . . . . . . . . . . . . . . . . . . . . . .July 2012
The Editorial Board of NephSAP extends its sincere appreciation to the following reviewers. Their efforts and insights have helped to
improve the quality of this postgraduate education offering.
NephSAP Review Panel
Nihal Y. Abosaif, MBBCh Rajiv Dhamija, MD Pranay Kathuria, MD, FASN
St. James University Hospital Walk in Medical Care University of Oklahoma College of Medicine
Leeds, United Kingdom Artesia, CA Tulsa, OK
Georgi Abraham, MBBS Susan R. DiGiovanni, MD Quresh T. Khairullah, MD, FASN
Pondicherry Institute of Medical Sciences Virginia Commonwealth University St. Clair Specialty Physicians
Madras Medical Mission Richmond, VA Detroit, MI
Chennai, India
Francis Dumler, MD Apurv Khanna, MD
Pablo H. Abrego, MD, FASN William Beaumont Hospital SUNY Upstate Medical University
Marshfield Clinic Royal Oak, MI Syracuse, NY
Wausau, WI
Mahmoud T. El-Khatib, MD, PhD, FASN Ramesh Khanna, MD
Anil K. Agarwal, MD, FASN University of Cincinnati Medical Center University of Missouri at Columbia
Ohio State University Medical Center Cincinnati, OH School of Medicine
Columbus, OH Columbia, MO
Lynda A. Frassetto, MD, FASN
Mustafa Ahmad, MD University of California at San Francisco Edgar V. Lerma, MD, FASN
King Fahad Medical City San Francisco, CA University of Illinois at Chicago
Riyadh, Saudi Arabia College of Medicine
Duvuru Geetha, MD
Chicago, IL
Jafar Al-Said, MD, FASN Johns Hopkins University
Bahrain Specialist Hospital Baltimore, MD Meyer D. Lifschitz, MD
Manama, Bahrain Shaare Zedek Medical Center
Carl S. Goldstein, MD
Jerusalem, Israel
Dante Amato-Martinez, MD, PhD Robert Wood Johnson Medical School
Universidad Nacional Autónoma de México New Brunswick, NJ Philippe S. Madhoun, MD
Tlalnepantla, Mexico Chu Charleroi
Nabil G. Guirguis, MD
Charleroi, Belgium
Anis U. Ansari, MD Kidney Dialysis and Transplant Group
Medical Associates Bridgeport, WV Jolanta Malyszko, MD, PhD, FASN
Clinton, IA Medical University
Pawan K. Gupta, MD
Bialystok, Poland
Akhtar Ashfaq, MD, FASN Altoona Regional Health System
North Shore University Hospital Altoona, PA Naveed N. Masani, MD
Great Neck, NY Winthrop University Hospital
Carsten Hafer, MD
Mineola, NY
Azra Bihorac, MD, FASN University of Hannover
University of Florida Hannover, Germany Hanna W. Mawad, MD, FASN
Gainesville, FL University of Kentucky Medical Center
Richard N. Hellman, MD
Lexington, KY
Mona B. Brake, MD Indiana University School of Medicine
Robert J. Dole VA Medical Center Indianapolis, IN Pascal Meier, MD, FASN
Wichita, KS Centre Hospitalier Universitaire Vaudois
Ekambaram M. Ilamathi, MD, FASN
Lausanne, Switzerland
Mauro Braun, MD Suffolk Nephrology Consultants
Cleveland Clinic Florida Stony Brook, NY Beckie Michael, DO, FASN
Weston, FL Marlton Nephrology and Hypertension
Viswanathan S. Iyer, MD, FASN
Marlton, NJ
Chokchai Chareandee, MD, FASN AKD-HTN LLC
Regions Hospital Harrisburg, PA Shahriar Moossavi, MD, PhD
Saint Paul, MN Wake Forest University Baptist
Bernard G. Jaar, MD
Medical Center
W. James Chon, MD, FASN Johns Hopkins Medical Institutions and
Winston-Salem, NC
University of Chicago Medical Center Nephrology Center of Maryland
Chicago, IL Baltimore, MD Scott R. Mullaney, MD
University of California at San Diego
Devasmita Choudhury, MD Avanelle V. Jack, MD
San Diego, CA
University of Texas Southwestern Louisiana State University Health
Medical School Sciences Center Quaid J. Nadri, MD, FASN
Dallas, TX New Orleans, LA King Faisal Specialist Hospital and
Research Center
Bulent Cuhaci, MD, FASN Sharon L. Karp, MD
Riiyadh, Saudi Arabia
Drexel University College of Medicine Indiana University School of Medicine
Philadelphia, PA Indianapolis, IN
Suzanne M. Norby, MD, FASN Karthik M. Ranganna, MD Robert J. Shay, MD, FASN
Mayo Clinic Drexel University College of Medicine East Georgia Kidney and
Rochester, MN Philadelphia, PA Hypertension Group
Augusta, GA
Michal Nowicki, MD Pawan K. Rao, MD, FASN
Medical University of Łódź St. Joseph’s Hospital Health Center Bhupinder Singh, MD, FASN
Łódź, Poland Syracuse, NY Southwest Kidney Institute
Tempe, AZ
Macaulay A. Onuigbo, MD, FASN Joel C. Reynolds, MD, FASN
Mayo Clinic Brooke Army Medical Center Rolf A.K. Stahl, MD
Eau Claire, WI San Antonio, TX University of Hamburg
Hamburg, Germany
Than N. Oo, MD Robert M.A. Richardson, MD
Nephrology Center University of Toronto Harold M. Szerlip, MD, FASN
Kalamazoo, MI Toronto, ON, Canada Medical College of Georgia
Augusta, GA
Kevin P. O’Reilly, MD Bijan Roshan, MD
Columbus Nephrology, Inc. Joslin Diabetes Center Bekir Tanriover, MD
Columbus, OH Harvard Medical School Dialysis Nephrology Associates
Boston, MA Dallas, TX
Malvinder S. Parmar, MB, MS, FASN
Northern Ontario School of Medicine Abinash C. Roy, MD Tushar J. Vachharajani, MD, FASN
Timmins, ON, Canada University of Utah School of Medicine Wake Forest University
Saint George, UT School of Medicine
Pairach Pintavorn, MD, FASN
Winston-Salem, NC
East Georgia Kidney and Hypertension Mario F Rubin, MD
Augusta, GA Massachusetts General Hospital Allen W. Vander, MD, FASN
Boston, MA Kidney Center of South Louisiana
Paul H. Pronovost, MD, FASN
Thibodaux, LA
Yale University School of Medicine Ehab R. Saad, MD, FASN
Waterbury, CA Medical College of Wisconsin Luigi Vernaglione, MD
Milwaukee, WI M. Giannuzzi Hospital
Mohammad A. Quasem, MD, FASN
Manduria, Italy
State University of New York Mohammad G. Saklayen, MD
Binghamton, NY Wright State University Medical School Shefali Vyas, MD
Dayton, OH Saint Barnabas Medical Center
Wajeh Y. Qunibi, MD
Livingston, NJ
University of Texas Health Sciences Center Ramesh Saxena, MD, PhD
San Antonio, TX University of Texas Southwestern Alexander Woywodt, MD, FASN
Medical Center Lancashire Teaching Hospitals NHS
Venkat Ramanathan, MD, FASN
Dallas, TX Foundation Trust
Baylor College of Medicine
Preston, United Kingdom
Houston, TX Gaurang M. Shah, MD
Long Beach VA Healthcare System
Long Beach, CA
NephSAP
®
Volume 10, Number 4, July 2011
Instructions to access the ASN website, NephSAP, and the UpToDate link
Compatible Browser: The ASN website (asn-online.org) has been formatted for cross-browser functionality, and should
display correctly in all modern web browsers. We recommend that you use Internet Explorer.
Monitor Settings: The ASN website was designed to be viewed in a 1024 ⫻ 768 or higher resolution.
Technical Support: If you are having difficulty viewing any of the pages, please refer to the ASN technical support page
for possible solutions. If you continue having problems, contact Hal Nesbitt at [email protected].
UpToDate provides an additional source of information that should help you answer up to 5 selected NephSAP
questions from each issue. The link is free and will remain active for the first 60 days after publication for each
issue.
● On the ASN home page, double click on the NephSAP link (NephSAP cover) on the bottom side of the page.
● On the NephSAP page, click on the UpToDate button on the left hand side to access the current links.
NephSAP
®
Volume 10, Number 4, July 2011
Disclosure Information
The ASN is responsible for identifying and resolving all conflicts of interest prior to presenting any educational activity to learners to ensure that
ASN CME activities promote quality and safety, are effective in improving medical practice, are based on valid contents, and are independent of the
control from commercial interests and free of commercial bias. All faculty are instructed to provide balanced, scientifically rigorous and evidence-
based presentations. In accordance with the disclosure policies of the Accreditation Council for Continuing Medical Education (ACCME) as well as
guidelines of the Food and Drug Administration (FDA), individuals who are in a position to control the content of an educational activity are re-
quired to disclose relationships with a commercial interest if (a) the relation is financial and occurred within the past 12 months; and (b) the individ-
ual had the opportunity to affect the content of continuing medical education with regard to that commercial interest. For this purpose, ASN consider
the relationships of the person involved in the CME activity to include financial relationships of a spouse or partner. Peer reviewers are asked to
abstain from reviewing topics if they have a conflict of interest. Disclosure information is made available to learners prior to the start of any ASN
educational activity.
Agarwal, Rajiv—Research funding: Abbott; Consultant/scientific advisor: Rockwell Medical, Watson Pharma; Honoraria: Abbott, Astra-Zeneca, Merck
Cohen, David J.—Research funding: Life Cycle Pharma, Novartis, Roche, Wyeth; Honoraria: Bristol-Myers-Squibb, Novartis, Roche
Emmett, Michael—Honoraria: Braintree Laboratories Fresenuis; Editorial board membership: American Journal of Cardiology, Clinical
Nephrology
Fried, Linda F.—Research funding: Merck, Reata; Honoraria: Pfizer
Fuchs, Elissa (Medical Editor)—none
Glassock, Richard J.—Consultant: Bio-Marin (inactive), Eli Lilly (active), FibroGen (inactive), Genentech (active), Lighthouse Learning (active),
Novartis (active), QuestCor (active), Wyeth (inactive); Ownership interests: LaJolla Pharmaceutical, Reata Inc.; Honoraria: American Society of
Nephrology, various medical schools for lectures and/or visiting professor; Membership board of directors/scientific advisor: American Renal
Associates, Los Angeles Biomedical Institute, University Kidney Research Associates (UKRO), Wyeth; Editorial board: UpToDate, American
Journal of Nephrology; Royalties: Oxford University Press; Paid expert testimony: Various legal firms regarding product liability
Goldfarb, Stanley—Consultant: Marval Scientific; Honoraria: GE Healthcare, Fresenius; Editorial board: Clinical Nephrology.
Liu, Kathleen D.—Ownership interest: Amgen; Honoraria: University of Virginia; Editorial board: Advances in Chronic Kidney Disease;
Other relationship: two biomarker studies, Abbott, CMIC.
Martin, Kevin J.—Consultant: Abbott, Cytochroma, Kai, Shire; Honoraria: Abbott, Genzyme, Kai, Shire; Scientific advisor: Abbott,
Cytochroma, Kai
Mehrotra, Rajnish—Research funding: Amgen, Baxter, Shire; Consultant: Novartis; Honoraria: AMAG, Baxter, Healthcare Shire
Murray, Patrick T.—Employment: spouse, Merck, Sharpe, & Dohme (Europe); Consultant: FAST Diagnostics (USA); Research funding:
Abbott, Alere, Argutus Medical; Honoraria: Sanofi Aventis; Editorial board: Clinical Journal of the American Society of Nephrology.
Nachman, Patrick H.—Honoraria: QuestCor; Multicenter clinical trial participation: Otsuka
Peixoto, Aldo J.—Consultant: Abbott, Sanofi-Aventis; Research funding: Pulsemetric; Honoraria: Boehringer-Ingelheim, Merck, Novartis,
Takeda; Scientific advisor/membership: Associate Editor–Blood Pressure Monitoring; Editorial Board: American Journal of Nephrology,
Brazilian Journal of Nephrology
Sterns, Richard H.—Honoraria: Astellas, Otsuka, Merck
Townsend, Raymond R.—Consultant: Daiichi-Sankyo, GlaxoSmithKline, Merck, Nicox, Novartis, Roche; Research funding: Novartis;
Honoraria: American Society of Hypertension, National Kidney Foundation
Vella, John P.—none
Editorial author:
Falk, Ronald J.—none
Guest Co-editors:
D’Agati, Vivette D.—none
Herlitz, Leal C.—none
Kambham, Neeraja—none
Markowitz, Glen S.—Consultant: Salix Pharmaceutical; Honoraria: Genzyme
Stokes, M. Barry—none
Commercial Support
There is no commercial support for this issue.
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Editorial
Problem-based learning- A new approach for NephSAP
Ronald J. Falk, MD
Department of Medicine, Division of Nephrology, University of North Carolina, Chapel Hill,
Chapel Hill, North Carolina
Optimally, problem-based learning simulates au- the accumulated evidence. The questions mimic Ameri-
thentic cases found in the practice of clinical medicine. can Board of Internal Medicine questions found on initial
This approach to learning promotes analytical and eval- or recertification exams.
uative thinking while emphasizing new information es- In reality, kidney biopsy conferences that use prob-
sential for understanding complex concepts. In nephrol- lem-based learning as their base are, in fact, examples of
ogy, it has been nephropathologists who have, by the “team-based learning.” Ideally, cases are not just de-
very nature of their craft, promoted and refined problem- scribed by the clinician and displayed by the pathologist
based learning. Many of us who became passionate about but also ignite lively discourse between experienced
glomerular diseases were introduced to the subject during physicians from multiple disciplines, each pushing the
engaging kidney biopsy conferences. There, the clinician other to hone in on specific clinical or pathologic detail,
presented a complex clinical scenario and the nephro- pathogenic considerations, and analysis of how best to
pathologist would reveal the captivating, dynamic im- manage the patient. In medical schools, problem-based
ages of glomerular, vascular, or tubulointerstitial com- learning has evolved into team-based learning to better
partments ravaged by a disease process. Typically, the emulate collaborative patient management. Groups of
nephropathologist artfully unfolded the diagnosis using students optimize learning when each member explores
images of light, immunofluorescence, and electron mi- different aspects of the presenting problem. The group
croscopy only after inviting a trainee to describe the must reach consensus on specific patient management
pathology, provide a differential diagnosis, and suggest a decisions and then articulate the justification of their
diagnosis. The case provided a springboard for the as- decision to the other students. Team-based learning is a
sembled cognoscenti to wax eloquent on the nature of the practical and highly effective learning strategy that max-
disease process, its natural history, and potential therapy. imizes active learning in the context of authentic clinical
The treating clinician was energized by these events and challenges. It also accommodates the best of didactic
gained a better understanding of a particular patient’s instruction where critical information is gained; expert
disease and optimal therapy. Kidney biopsy conferences insights or “pearls” are shared, collectively providing
remain wonderful examples of problem-based learning opportunities for learners to exercise critical thinking and
where clinical and pathologic correlations are illumi- evidence-based, decision-making skills to solve intrigu-
nated, new understandings of disease considered, and ing and complex clinical scenarios.
patient care improved. All of these NephSAP cases will be available on the
In this issue of NephSAP, Dr. Markowitz and col- American Society of Nephrology website and download-
leagues have recreated a virtual kidney biopsy confer- able for group discussion or team-based learning. One
ence. Using a problem-based learning approach, 22 cases can readily imagine these cases as the basis of a virtual
are presented spanning the spectrum of glomerular dis- kidney biopsy conference and an excellent resource
ease with varying degrees of diagnostic difficulty. A for nephrology trainees and for those in practice who
clinical vignette is followed by artfully displayed pa- wish to “revisit” their understanding of glomerular
thology. The critical question is asked: “What is the disease or plan to take recertifying exams. This edition
diagnosis?” The rationale for the consensus answer is of NephSAP provides an alternative approach to learn-
considered and then a succinct, well-referenced, state-of- ing that differs from that which NephSAP readers have
the-art discussion ensues. The learner can readily appre- come to expect. Periodic change is refreshing, and one
ciate the clinicopathological correlates and frame the can imagine this format will be used in other spheres
question as to how best to manage the patient based on of nephrology.
271
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Syllabus
Learning Objectives
Glen S. Markowitz, MD*, M. Barry Stokes, MD*, Neeraja Kambham, MD†,
Leal C. Herlitz, MD*, and Vivette D. D’Agati, MD*
*Department of Pathology & Cell Biology, Columbia University Medical Center and the
New York Presbyterian Hospital, New York, New York; and †Department of Pathology,
Stanford University School of Medicine, Stanford, California
1. To recognize the light microscopic, immunofluo- pathologic findings and pathogenesis, briefly touched
rescence, and ultrastructural findings in the most on clinical presentation, and avoided the topic of
common forms of glomerular disease. treatment. At the end of the last section on page 422,
2. To discuss recent advances in the pathogenesis of we provide a reverse table of contents that will
glomerular, tubulointerstitial, and vascular dis- allow the reader to look up specific diseases of
eases. interest.
3. To examine the most common secondary etiolo- The first four authors of this issue of NephSAP
gies of the major patterns of glomerular disease. share the great fortune of having been trained and/or
4. To formulate a differential diagnosis based on
mentored by our senior author, Vivette D’Agati. So
integration of clinical and pathologic findings in
much emphasis is placed on the founders of the field
medical renal disease.
of renal pathology, and yet not enough is placed on the
This issue of NephSAP is the first to cover the
individuals who have brought us into the modern era.
topic of renal pathology. In putting this together, we
There is no better example than Vivette, who has been
were given the freedom to create a unique format.
Each of the 22 sections in this issue begins with a instrumental in refining the pathologic classification of
clinical history and a set of renal biopsy images; the numerous medical renal diseases, in defining new
reader is then asked to determine the diagnosis. The disease entities, and in continuously providing insights
following page provides the reasoning behind the di- into disease pathogenesis. We wish to dedicate this
agnosis, followed by a discussion of the disease entity. issue to Dr. D’Agati, an unusual decision considering
We chose this format because it has been our experi- that she is the senior author. As in the many other
ence that renal pathology is learned best by clinical- projects we have undertaken jointly, Vivette selflessly
pathologic correlation. In an effort not to overlap with poured her energy into this project and brought it to a
the outstanding NephSAP on “Glomerular, Vascular, significantly higher level. She has been and continues
and Tubulointerstitial Diseases” written by Patrick to be a role model, knowing no limits when it comes
Nachman and Richard Glassock, we focused on renal to the pursuit of excellence.
272
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Section 1: CASE PRESENTATION 100 mmHg, 2⫹ pitting edema of the lower extremities, and a
weight of 273 lb. The patient had a creatinine level of 0.82
A 54-year-old African-American woman presents with full mg/dl and bland urine sediment. All serologies were negative
nephrotic syndrome including 24-hour urine protein of 5.8 g, or normal including anti-nuclear antibody, anti-DNA antibody,
serum albumin of 2.2 g/dl, edema, and a recent 25-lb weight hepatitis B surface antigen (HbsAg), hepatitis C antibody,
gain. Past medical history was significant for hypertension for anti-neutrophil cytoplasmic antibody (ANCA), rheumatoid
20 years, arthritis of the knees for which she had been treated factor, C3, and C4. There was no evidence of a monoclonal
with nonsteroidal anti-inflammatory drugs (NSAIDs), and serum or urine spike. Initial treatment consisted of discontin-
morbid obesity. Her medications included meloxicam, hydro- uation of meloxicam and the addition of an angiotensin recep-
chlorothiazide, lisinopril, lovastatin, and propoxyphene with tor blocker. Two months later, the patient’s nephrotic param-
acetaminophen. Physical examination revealed a BP of 140/ eters remained unchanged. Renal biopsy was performed.
Figure 1. Figure 2.
Figure 4.
Figure 3. IgG
ties. Slight vacuolization of the GBM may be demon- or stage 3, and multiple stages can coexist in a single
strable with silver stain, and minute trichrome-red biopsy or even a single glomerulus.
deposits may be visible on high power. The GBM is
usually normal in thickness, although it may appear Clinical-Pathologic Correlation
rigid with associated podocyte swelling. Stage 2 MN Multiple pathologic findings have been reported
is the most frequently encountered in biopsy practice to predict reduced renal survival in MN, including the
and is characterized by global subepithelial deposits degree of tubular atrophy and interstitial fibrosis, the
separated by intervening projections of the GBM, development of segmental glomerulosclerosis (7,8),
referred to as “GBM spikes.” In stage 3, the subepi- the intensity of complement deposition by immuno-
thelial deposits and GBM spikes become covered and fluorescence (9,10), and the presence of deposits in
enclosed by overlying neomembrane formation, pro- multiple stages (“heterogeneous deposits”) (11). Al-
ducing chain-like thickenings (Figure 5). By stage 4, though these pathologic findings may be associated
the deposits have been incorporated into an irregularly with a worse prognosis, they do not appear to predict
thickened GBM. In stage 4 and, to a lesser extent, clinical outcomes independent of baseline clinical
stage 3, deposits become more electron lucent as variables (10).
they undergo resorption. In some cases of stage 4
MN, deposits are no longer visible, making it diffi- Secondary Forms of MN
Secondary forms of MN, which represent ap-
cult to establish a definitive diagnosis of MN.
proximately 25% of cases (12), fall into four main
Importantly, the Ehrenreich & Churg staging of
categories. First and foremost, MN may occur second-
MN provides a useful framework to understand the
ary to autoimmune/collagen vascular diseases, most
process of subepithelial deposit formation and incor-
notably systemic lupus erythematosus (i.e., “membra-
poration into the GBM, but its clinical relevance is
nous lupus nephritis”) but also rheumatoid arthritis,
limited because there is no good correlation with the
Sjögren syndrome, mixed connective tissue disease,
degree of proteinuria, renal function, or prognosis. sarcoidosis, Grave disease, and Hashimoto thyroiditis.
Furthermore, “progression” from one stage to another Second, MN may be associated with malignancy, most
(e.g., stage 2 to stage 3) can be associated with commonly carcinomas of the lung, prostate, breast,
improvement or worsening of proteinuria. In clinical and gastrointestinal tract (3,13,14). A recent study
practice, more than 80% of cases of MN are in stage 2 from France found that 10% of adult patients with MN
had evidence of malignancy, which was often occult at
the time of renal biopsy. Malignancy was more com-
mon in patients with MN who were older than 60 years
and had a history of significant tobacco use, suggest-
ing an increased need for screening in this population
(3,12). Multiple therapeutic agents have been impli-
cated in the development of MN, including gold,
penicillamine, multiple NSAIDs, COX-2 inhibitors,
and captopril. Infectious causes of MN include hepa-
titis B, hepatitis C, syphilis, and multiple parasites.
Primary MN typically reveals isolated subepithe-
lial immune deposits that stain predominantly for IgG.
Any deviations from this pathologic picture should
suggest a possible secondary form of disease. For
Figure 5. A case of stage 3 MN exhibits complex thickening instance, mesangial or endocapillary proliferation, de-
of the glomerular capillary walls by subepithelial and in- posits at other sites (e.g., mesangial, subendothelial,
tramembranous deposits ranging from electron dense to tubular basement membranes, vessel walls), the im-
electron lucent. Most of the deposits are separated by spikes
and incorporated into the GBM by an overlying new layer
munofluorescence finding of a “full house” of staining
of basement membrane material. The foot processes are for immunoglobulins (IgG, IgM, and IgA) and com-
diffusely effaced. Magnification, ⫻2500. plement (C3 and C1q), and the ultrastructural finding
276 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
of endothelial tubuloreticular inclusions favor a sec- Gp330/megalin antibodies generated against tubular
ondary form of MN, in particular membranous lupus brush border antigen cross-react with Gp330 ex-
nephritis (12) (Figure 6). An exception to this rule is pressed on the soles of podocyte foot processes. In this
neoplasia-associated MN, which is usually indistin- model, immune complexes formed in situ are capped
guishable morphologically from primary MN. and shed off the podocyte’s clathrin-coated pits into
Multiple reports have attempted to use staining the subepithelial region, producing MN. Subsequent
for the subclasses of IgG as a means of differentiating studies on the passive HN model have demonstrated
primary from secondary MN (15–17). In primary MN, that proteinuria is complement dependent (20,21) and
IgG4 predominates, but IgG1 is also commonly pres- involves formation of the C5b-C9 membrane attack
ent and is thought to play a role in complement complex which inserts into the podocyte cell mem-
activation. In membranous lupus nephritis, all sub- brane, inciting podocytes to synthesize proteases, ox-
classes of IgG1 (i.e., IgG1 through IgG4) are com- idants, TGF-, and extracellular matrix (22,23). Al-
monly seen with staining for IgG4 typically least though Gp330 is not expressed by human podocytes,
intense. In MN related to malignancy, positivity for the HN model has been critically important to eluci-
IgG1, IgG2, IgG4, and rarely IgG3 is found. Given the date the role of in situ immune complex formation in
significant overlap in findings, we have not found the pathogenesis of MN.
staining for IgG subclasses to be particularly helpful as An advance in the search for a human Heymann
a diagnostic tool, although it is extensively used in the antigen occurred in 2002, when Debiec et al. (24)
research context. identified a newborn male with severe nephrotic syn-
drome caused by MN associated with antibodies
Etiology and Pathogenesis against neutral endopeptidase (NEP), a protein ex-
Heymann nephritis (HN) is the classic animal
pressed on both the proximal tubular brush border and
model of MN, which is produced by immunizing rats
podocytes. The patient’s mother was NEP deficient
with fractionated renal cortex enriched in proximal
and had developed antibodies against NEP during a
tubular brush borders (Fx1A). Passive transfer of im-
previous pregnancy that ended in miscarriage. Trans-
mune serum to naive rats produces a passive model of
placental transfer of anti-NEP antibodies led to MN in
HN. The critical immunogenic component of Fx1A is
the immediate postnatal period. On the basis of this
Gp330/megalin (18,19), known as the “Heymann an-
family, NEP became the first human podocyte antigen
tigen.” Gp330/megalin is expressed on rat proximal
shown to act as a “Heymann antigen” (24). Additional
tubular brush border and podocyte foot processes,
families with antenatal MN resulting from transpla-
where it functions as an LDL receptor. The anti-
cental transfer of anti-NEP antibodies have been iden-
tified (25).
A tremendous breakthrough in our understanding
of the pathogenesis of primary MN occurred in 2009,
when Beck and colleagues (26,27) identified antibod-
ies against a conformation-dependent epitope of the
M-type phospholipase A2 receptor (PLA2R) in 75% of
patients with primary (idiopathic) MN. PLA2R is a
mannose-containing transmembrane receptor glyco-
protein expressed on podocytes and at lower levels on
type II pneumocytes and leukocytes (28). Patients with
idiopathic MN have circulating, predominantly IgG4
antibodies against PLA2R but no evidence of circulat-
Figure 6. In this case of MN, the global subepithelial ing immune complexes, supporting the concept of in
deposits are accompanied by prominent mesangial and sub- situ immune complex formation (26). Antibodies
endothelial deposits, as well as focal endothelial tubulore- eluted from the deposits of primary MN have speci-
ticular inclusions (not shown). The patient was found to
have evidence of systemic lupus erythematosus, and the
ficity for PLA2R (26). The presence of anti-PLA2R
final diagnosis was membranous lupus nephritis. Magnifi- antibodies in the serum of the majority of patients with
cation, ⫻6000. primary MN and their absence in secondary forms
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 277
identify PLA2R as the major target antigen in primary cance of focal segmental glomerulosclerosis. Am J Kidney Dis 41:
38 – 48, 2003
MN. Anti-PLA2R antibody serum levels seem to be 9. Wehrmann M, Bohle A, Bogenschutz O, Eissele R, Freislederer A,
specific for primary MN and to correlate with disease Ohlschlegel C, Schumm G, Batz C, Gartner HV: Long-term progno-
activity (26,27). Treatment-induced remissions are as- sis of chronic idiopathic membranous glomerulonephritis. Clin Neph-
rol 31: 67–76, 1989
sociated with reduction in the levels of circulating 10. Troyanov S, Roasio L, Pandes M, Herzenberg AM, Cattran DC:
anti-PLA2R autoantibody (26). Recurrence of primary Renal pathology in idiopathic membranous nephropathy: A new
MN in the allograft has been associated with reappear- perspective. Kidney Int 69: 1641–1648, 2006
11. Yoshimoto K, Yokoyama H, Wada T, Furuichi K, Sakai N, Iwata Y,
ance of anti-PLA2R antibodies (29). More recently, Goshima S, Kida H: Pathologic findings of initial biopsies reflect the
Stanescu et al. have demonstrated that MN is linked to outcomes of membranous nephropathy. Kidney Int 65: 148 –153,
the HLA-DQA1 allele on chromosome 6p21, a finding 2004
12. Glassock RJ: Secondary membranous glomerulonephritis. Nephrol
which supports a central role for autoimmunity in the Dial Transplant [Suppl 1]: 64 –71, 1992
pathogenesis of MN (30). 13. Burstein DM, Korbet SM, Schwartz MM: Membranous glomerulo-
The pathogenesis of most forms of secondary nephritis and malignancy. Am J Kidney Dis 22: 5–10, 1993
14. Eagen JW, Lewis EJ: Glomerulopathies of neoplasia. Kidney Int 11:
MN is likely to involve immune responses to nonglo- 297–303, 1977
merular antigens (e.g., infectious pathogens, tumors, 15. Doi T, Mayumi M, Kanatsu K, Suehiro F, Hamashima Y: Distribu-
nuclear autoantigens). The target antigen could be- tion of IgG subclass in membranous nephropathy. Clin Exp Immunol
58: 57– 62, 1984
come “planted” on the subepithelial aspect of the 16. Kuroki A, Shibata T, Honda H, Totsuka D, Kobayashi K, Sugisaki T:
GBM favored by its small size and cationic charge, Glomerular and serum IgG subclasses in diffuse proliferative lupus
followed by in situ binding of a circulating antibody. It nephritis, membranous lupus nephritis, and idiopathic membranous
nephropathy. Intern Med 41: 936 –942, 2002
is also possible that some patients have circulating 17. Ohtani H, Wakui H, Komatsuda A, Okuyama S, Masai R, Maki N,
preformed immune complexes that have the potential Kigawa A, Sawada K, Imai H: Distribution of IgG subclass deposits
to dissociate and reform in the subepithelial space in malignancy-associated membranous nephropathy. Nephrol Dial
Transplant 19: 574 –579, 2004
because of their low avidity and cationic charge. The 18. Heymann W, Hackel DB, Harwood S, Wilson SG, Hunter JL:
responsible antigens are probably diverse depending Production of nephrotic syndrome in rats by Freund’s adjuvants
on the condition and include nucleosomal antigens in and rat kidney suspensions. Proc Soc Exp Biol Med 100: 660 –
664, 1959
membranous lupus nephritis or HbsAg in hepatitis 19. Kerjaschki D, Farquhar MG: The pathogenic antigen of Heymann
B–associated MN (31). nephritis is a membrane glycoprotein of the renal proximal tubule
brush border. Proc Natl Acad Sci 79: 5557–5561, 1982
20. Baker PJ, Ochi RF, Schulze M, Johnson RJ, Campbell C, Couser
References WG: Depletion of C6 prevents development of proteinuria in exper-
1. Cattran DC, Pei Y, Greenwood CM, Ponticelli C, Passerini P, imental membranous nephropathy in rats. Am J Pathol 135: 185–194,
Honkanen E: Validation of a predictive model of idiopathic mem- 1989
branous nephropathy: Its clinical and research implications. Kidney 21. Cybulsky AV, Rennke HG, Feintzeig ID, Salant DJ: Complement-
Int 51: 901–907, 1997 induced glomerular epithelial cell injury: Role of the membrane
2. Jennette JC, Iskandar SS, Dalldorf FG: Pathologic differentiation attack complex in rat membranous nephropathy. J Clin Invest 77:
between lupus and nonlupus membranous glomerulopathy. Kidney 1096 –1107, 1986
Int 24: 377–385, 1983 22. Couser WG: Mediation of immune glomerular injury. J Am Soc
3. Lefaucheur C, Stengel B, Nochy D, Martel P, Hill GS, Jacquot C, Nephrol 1: 13–29, 1990
Rossert J: Membranous nephropathy and cancer: Epidemiologic 23. Nangaku M, Shankland SJ, Couser WG: Cellular response to injury
evidence and determinants of high-risk cancer association. Kidney Int in membranous nephropathy. J Am Soc Nephrol 16: 1195–1204, 2005
70: 1510 –1517, 2006 24. Debiec H, Guigonis V, Mougenot B, Decobert F, Haymann JP,
4. Klassen J, Elwood C, Grossberg AL, Milgrom F, Montes M, Bensman A, Deschenes G, Ronco PM: Antenatal membranous glo-
Sepulveda M, Andres G: Evolution of membranous nephropathy into merulonephritis due to anti-neutral endopeptidase antibodies. N Engl
anti-glomerular-basement-membrane glomerulonephritis. N Engl J Med 346: 2053–2060, 2002
J Med 290: 1340 –1344, 1974 25. Debiec H, Nauta J, Coulet F, van der Burg M, Guigonis V, Schur-
5. Nasr SH, Said M, Valeri AM, Stokes MB, Masani NN, D’Agati VD, mans T, de Heer E, Soubrier F, Janssen F, Ronco P: Role of
Markowitz GS: Membranous glomerulonephritis with ANCA-asso- truncating mutations in MME gene in fetomaternal alloimmunisation
ciated necrotizing and crescentic glomerulonephritis. Clin J Am Soc and antenatal glomerulopathies. Lancet 364: 1252–1259, 2004
Nephrol 4: 299 –308, 2009 26. Beck LH, Bonegio RG, Lambeau G, Beck DM, Powell DW, Cum-
6. Ehrenreich T, Churg J: Pathology of membranous nephropathy. In: mins TD, Klein JB, Salant DJ: M-Type phospholipase A2 receptor as
Pathology Annual, edited by Sommers SC, New York, Appleton- target antigen in idiopathic membranous nephropathy. N Engl J Med
Century-Crofts, 1968 361: 11–21, 2009
7. Wakai S, Magil AB: Focal glomerulosclerosis in idiopathic membra- 27. Beck LH, Salant DJ: Membranous nephropathy: Recent travels and
nous glomerulonephritis. Kidney Int 41: 428 – 434, 1992 new roads ahead. Kidney Int 77: 765–770, 2010
8. Dumoulin A, Hill GS, Montseny JJ, Meyrier A: Clinical and mor- 28. Hanasaki K: Mammalian phospholipase A2:phospholipase A2 recep-
phological prognostic factors in membranous nephropathy: Signifi- tor. Biol Pharm Bull 27: 1165–1167, 2004
278 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
29. Stahl R, Hoxha E, Fechner K: PLA2R autoantibodies and recurrent Powis SH, Brenchley P, Feehally J, Rees AJ, Debiec H, Wetzels JF,
membranous nephropathy after transplantation. N Engl J Med 363: Ronco P, Mathieson PW, Kleta R: Risk HLA-DQA1 and PLA2R1
496 – 498, 2010 alleles in idiopathic membranous nephropathy. N Engl J Med 364:
30. Stanescu HC, Arcos-Burgos M, Medlar A, Bockenhauer D, Kottgen 616 – 626, 2011
A, Dragomirescu L, Voinescu C, Patel N, Pearce K, Hubank M, 31. Lai FM, Lai KN, Tam JS, To KF, Li PK: Primary glomerulonephritis
Stephens HA, Laundy V, Padmanabhan S, Zawadzka A, Hofstra JM, with detectable hepatitis B virus antigens. Am J Surg Pathol 18:
Coenen MJ, den Heijer M, Kiemeney LA, Bacq-Daian D, Stengel B, 175–186, 1994
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Section 2: CASE PRESENTATION albumin was 1.7 g/dl, and urinalysis showed 4⫹ protein
and 5 to 10 erythrocytes per high-power field with no
A 58-year-old African American man presents with an cellular casts. A 24-hour urine collection contained
85-lb weight gain over the past month as a result of 12.4 g of protein. Serologic evaluation revealed negative
progressive anasarca. Past medical history is significant or normal anti-nuclear antibody, anti-DNA antibody, C3,
for 12 years of hypertension and diet-controlled diabetes. C4, hepatitis B surface antigen, hepatitis C antibody, and
Baseline serum creatinine 1 year prior was 1.1 mg/dl. At HIV serology. No monoclonal serum or urine spikes were
the time of biopsy, serum creatinine was 6.3 mg/dl, serum detected by serum and urine protein electrophoresis.
Figure 1. Figure 2.
Figure 3. Figure 4.
(6,10). At Columbia University Medical Center, no cases C1. The protein resorption droplets within podocyte
of collapsing FSGS were found before 1979, but the and tubular epithelial cytoplasm often stain with anti-
incidence of primary (idiopathic) collapsing FSGS pro- sera to IgG, IgA, and albumin. True immune complex
gressively increased through the 1980s and by the mid- deposition within the glomerular tuft is absent, how-
1990s represented 24% of cases of primary FSGS (10). ever.
Serologic evaluation of patients with collapsing Electron Microscopy. By electron microscopy, the
FSGS, particularly in those of African and Hispanic collapsed segments display wrinkling and retraction of
descent, should include HIV testing because HIV- the GBMs, causing luminal narrowing or obliteration.
associated nephropathy is a common secondary cause Overlying the collapsed tuft, the podocytes exhibit
of collapsing FSGS. Drug nephrotoxicity, most nota- complete foot process effacement with foci of detach-
bly with pamidronate (11) and IFN therapy (12), can ment from the GBM and intervening accumulation of
cause collapsing FSGS, as discussed in the section on loose extracellular matrix. The pseudocrescents con-
Secondary Forms of Collapsing FSGS. tain plump glomerular epithelial cells that often appear
confluent with the parietal epithelial layer, obliterating
Pathology the urinary space. These large epithelial cells typi-
Light Microscopy. Light microscopic features of cally contain abundant intracytoplasmic electron-
collapsing FSGS are distinctive. Although glomerular dense rounded, membrane-bound inclusions repre-
collapse may be segmental in distribution, more typi- senting protein resorption droplets. Glomeruli
cally the entire glomerular globe is affected, producing without collapse usually have extensive podocyte
a picture of global collapsing sclerosis. Collapsing foot process effacement involving ⬎50% of the
lesions are characterized by occlusion of capillary glomerular capillary surface area. Endothelial cells
lumina as a result of implosive wrinkling and retrac- should be examined for the presence of tubuloreticular
tion of the GBMs, best seen with the Jones methena- inclusions, which are commonly seen in the setting of
mine silver or periodic acid–Schiff stains. Accompa- HIV infection and IFN therapy but not in primary
nying the tuft collapse are dramatic proliferation and collapsing FSGS. Endothelial tubuloreticular inclu-
hypertrophy of overlying visceral epithelial cells, sions, which are considered “interferon footprints,”
which typically contain large intracytoplasmic protein consist of 24 nm interanastamosing tubular structures
resorption droplets. Whereas other subtypes of FSGS located within the dilated endoplasmic reticulum of
often show expansion of extracellular matrix, intracap- the endothelial cell cytoplasm (Figure 5).
illary foam cells, hyalinosis, and segmental adhesions
to Bowman’s capsule, these features are typically
absent, at least in the early stages of collapsing FSGS.
In the late stages, the collapsed tuft retracts into a solid
sphere crowned by a monolayer of hypertrophied vis-
ceral epithelial cells. Tubulointerstitial abnormalities
are also prominent and may appear out of proportion
to the degree of glomerulosclerosis. Tubular degener-
ative and regenerative changes include loss of brush
border, shedding of cytoplasmic fragments into tubu-
lar lumina, tubular epithelial hypertrophy, and en-
larged vesicular nuclei with prominent nucleoli. The
formation of tubular microcysts containing loose pro-
teinaceous casts is a distinctive feature of collapsing
FSGS. Figure 5. In the setting of collapsing FSGS, the finding of
endothelial tubuloreticular inclusions (TRI), which are in-
Immunofluorescence. Within collapsing segments,
teranastamosing tubular structures within the dilated endo-
immunofluorescence typically shows staining of the plasmic reticulum of endothelial cells, raises the possibility
glomerular tuft with IgM, C3, and, less commonly, of HIV infection. Magnification, ⫻20,000.
282 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Secondary Forms of Collapsing FSGS (11). Proteinuria and renal insufficiency typically im-
Collapsing FSGS is associated with a variety of prove after drug withdrawal but do not reach baseline
systemic conditions. The most common and best un- levels. Patients treated with IFN-␣ (for hepatitis C
derstood association is HIV infection, producing HIV- virus infection), IFN- (for multiple sclerosis), or
associated nephropathy (HIVAN). HIV-1 directly in- IFN-␥ (for idiopathic pulmonary fibrosis or chronic
fects renal epithelial cells (including podocytes, granulomatous disease) may develop collapsing FSGS
parietal epithelial cells, and tubular epithelial cells), with severe nephrotic syndrome that improves after
where it is capable of active replication and quasispecies drug discontinuation (12).
evolution (13,14). Once the virus infects renal epithe- Acute vaso-occlusive injury in the setting of
lium, it causes dysregulation of host genes to produce cholesterol embolization (23) and severe renovascular
nephropathy. Genetic engineering in the mouse has iden- disease (24) have also been reported to cause severe
tified several specific HIV-1 genes as particularly impor- proteinuria and collapsing glomerulopathy. In renal
tant in HIVAN pathogenesis. Podocyte-specific ex- allografts, collapsing FSGS has been described in a
pression of vpr and nef acts synergistically to produce variety of settings (25), most notably prominent mi-
nephropathy in double transgenic mice (15). Vpr plays crovascular disease, suggesting a role for ischemic
a role in intranuclear transport of the HIV-1 preinte- injury (26). Finally, rare familial forms of collapsing
gration complex and causes G2 cell-cycle arrest. Tu- FSGS have been described (27), including a new
bular epithelial cells transfected with vpr develop association with action myoclonus renal failure syn-
cell-cycle arrest and inability to undergo cytokinesis, drome caused by autosomal recessive mutations in
resulting in increased chromosomal copy number and lysosomal membrane protein SCARB2/LIMP-2 (28).
enlarged nuclei, accounting for the typical tubular cell
enlargement and “reactivity” seen histologically in Etiology and Pathogenesis
human and murine HIVAN (16). Nef, a major HIV-1 Podocyte injury is a central pathogenetic mech-
virulence factor, contains a proline-rich motif that anism in all types of FSGS. The unique morphology of
interacts with the SH3 domain of Src family kinases, collapsing FSGS with implosion of the glomerular tuft
activating signal transduction events that cause disrup- and proliferation of overlying epithelial cells supports
tion of the actin cytoskeleton, podocyte dedifferentia- a severe and rapid podocyte injury. Podocytes are
tion, and proliferation (17,18). The strong African highly differentiated cells with a unique cytoarchitec-
American racial predominance of HIV-associated ne- ture required for structural support of the glomerular
phropathy supports the importance of genetic suscep- capillaries, synthesis of the GBM, and regulation of
tibility (see Etiology and Pathogenesis section). The glomerular permselectivity. Differentiated podocytes
use of antiretroviral therapy in HIV-1–infected pa- are postmitotic cells, and podocyte development is
tients has reduced the incidence and attenuated the characterized by the expression of maturity markers
course of HIVAN (15). such as Wilms’ tumor 1, podocalyxin, and synaptopo-
Other viral infections that have been associated din. In collapsing FSGS, both idiopathic and HIV-
with collapsing FSGS include cytomegalovirus (19), associated podocytes become dedifferentiated and
parvovirus B19 (20), and simian virus 40 (21). Rare dysregulated, losing expression of these maturity
cases have also been described in association with markers and acquiring markers of proliferation, such
tuberculosis, Epstein-Barr virus infection, and eryth- as Ki-67 (29). In contrast, mature podocyte markers
rophagocytosis syndrome (22). In some of these are retained in membranous nephropathy and minimal
forms, there is a potential for podocyte infection by the change disease, despite severe proteinuria and foot
inciting virus (20,21), whereas in others, podocyte process effacement.
injury probably results from indirect cytokine effects The podocyte dedifferentiation in collapsing
or innate immunity (20). FSGS has made it difficult to identify the origin of the
Drug nephrotoxicity is another secondary cause proliferating glomerular epithelial cells within the
of collapsing FSGS. Treatment with high-dosage pseudocrescents. Using lineage-specific markers for
pamidronate has been associated with the development parietal epithelial cells, such as cytokeratins, there is
of collapsing FSGS in patients being treated for hy- increasing evidence that parietal epithelial cells con-
percalcemia of malignancy and osteolytic metastases tribute to the cellular proliferation seen in collapsing
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 283
FSGS of various causes (30 –32). The recent identifi- drome, progressive irreversible renal failure, and glomerular “col-
lapse”: A new clinicopathologic entity? Am J Kidney Dis 7: 20 –28,
cation of a stem cell reservoir in Bowman’s capsule 1986
that can repopulate podocytes supports the concept 5. Schwartz MM, Lewis EJ: Focal segmental glomerular sclerosis: The
that both immature podocytes and cells of parietal cellular lesion. Kidney Int 28: 968 –974, 1985
6. Haas M, Spargo BH, Coventry S: Increasing incidence of focal-
origin contribute to the extracapillary proliferation in segmental glomerulosclerosis among adult nephropathies: A 20-year
collapsing glomerulopathies (33,34). Microarray stud- renal biopsy study. Am J Kidney Dis 26: 740 –750, 1995
ies performed on glomeruli microdissected from hu- 7. Detwiler RK, Falk RJ, Hogan SL, Jennette JC: Collapsing glomeru-
lopathy: A clinically and pathologically distinct variant of focal
man renal biopsies with primary collapsing FSGS segmental glomerulosclerosis. Kidney Int 45: 1416 –1424, 1994
have confirmed reduced expression of podocyte ma- 8. Valeri A, Barisoni L, Appel GB, Seigle R, D’Agati V: Idiopathic
turity markers and slit diaphragm proteins, supporting collapsing focal segmental glomerulosclerosis: A clinicopathologic
study. Kidney Int 50: 1734 –1746, 1996
the concept of the dysregulated podocyte phenotype. 9. Laurinavicius A, Hurwitz S, Rennke HG: Collapsing glomerulopathy
They have also identified overrepresentation of pari- in HIV and non-HIV patients: A clinicopathological and follow-up
etal markers and genes involved in biologic processes study. Kidney Int 56: 2203–2213, 1999
10. Barisoni L, Valeri A, Radhakrishnan J, Nash M, Appel G, D’Agati V:
of development and differentiation, consistent with Focal segmental glomerulosclerosis: A 20-year epidemiologic study
expansion of the parietal cell compartment and recruit- [Abstract]. J Am Soc Nephrol 5: 347, 1994
ment of progenitor cells (35). 11. Markowitz GS, Appel GB, Fine PL, Fenves AZ, Loon NR, Jagannath
S, Kuhn JA, Dratch AD, D’Agati VD: Collapsing focal segmental
The genetic basis for the strong African Ameri-
glomerulosclerosis following treatment with high-dose pamidronate.
can racial predisposition to FSGS has been linked to J Am Soc Nephrol 12: 1164 –1172, 2001
allelic variants in a region of chromosome 22 includ- 12. Markowitz GS, Nasr SH, Stokes MB, D’Agati VD: Treatment with
IFN-alpha, -beta, or -gamma is associated with collapsing focal
ing MYH9 encoding myosin heavy chain 9 and
segmental glomerulosclerosis. Clin J Am Soc Nephrol 5: 607– 615,
APOL1 encoding apolipoprotein L-1 (36,37), which 2010
are in close linkage disequilibrium. Two specific se- 13. Bruggeman LA, Ross MD, Tanji N, Cara A, Dikman S, Gordon RE,
Burns GC, D’Agati VD, Winston JA, Klotman ME, Klotman PE:
quence variants of APOL1 show a strong correlation
Renal epithelium is a previously unrecognized site of HIV-1 infec-
with the development of FSGS. APOL1, a constituent tion. J Am Soc Nephrol 11: 2079 –2087, 2000
of HDL particles in the blood, confers resistance to 14. Marras D, Bruggeman LA, Gao F, Tanji N, Mansukhani MM, Cara
A, Ross MD, Gusella GL, Benson G, D’Agati VD, Hahn BH,
trypanosomal infection through its ability to lyse
Klotman ME, Klotman PE: Replication and compartmentalization of
Trypanosoma brucei brucei, the parasite that causes HIV-1 in kidney epithelium of patients with HIV-associated nephrop-
sleeping sickness. Subspecies of Trypanosoma athy. Nat Med 8: 522–526, 2002
evolved in Africa that were resistant to lysis, in turn 15. Zuo Y, Matsusaka T, Zhong J, Ma J, Ma LJ, Hanna Z, Jolicoeur P,
Fogo AB, Ichikawa I: HIV-1 genes vpr and nef synergistically
favoring evolutionary mutations in human APOL1 that damage podocytes, leading to glomerulosclerosis. J Am Soc Nephrol
could counteract the resistance. These allelic variants 17: 2832–2843, 2006
of APOL1, which confer protection against infection, 16. Rosenstiel PE, Gruosso T, Letourneau AM, Chan JJ, LeBlanc A,
Husain M, Najfeld V, Planelles V, D’Agati VD, Klotman ME,
come at the cost of increased risk for FSGS, although Klotman PE: HIV-1 Vpr inhibits cytokinesis in human proximal
the mechanistic basis for the renal effects remains tubule cells. Kidney Int 74: 1049 –1058, 2008
unknown (37) Thus, analogous to the emergence of 17. Lu TC, He JC, Wang ZH, Feng X, Fukumi-Tominaga T, Chen N, Xu
J, Iyengar R, Klotman PE: HIV-1 Nef disrupts the podocyte actin
sickle cell trait to resist malaria, the genetic resistance cytoskeleton by interacting with diaphanous interacting protein.
to the infectious pathogen is a dominant trait (present J Biol Chem 283: 8173– 8182, 2008
in heterozygotes), whereas the host disease is reces- 18. He JC, Husain M, Sunamoto M, D’Agati VD, Klotman ME, Iyengar
R, Klotman PE: Nef stimulates proliferation of glomerular podocytes
sive (present in homozygotes). through activation of Src-dependent Stat3 and MAPK1,2 pathways.
J Clin Invest 114: 643– 651, 2004
References 19. Tomlinson L, Boriskin Y, McPhee I, Holwill S, Rice P: Acute
1. Stokes MB, Valeri AM, Markowitz GS, D’Agati VD: Cellular focal cytomegalovirus infection complicated by collapsing glomerulopa-
segmental glomerulosclerosis: Clinical and pathologic features. Kid- thy. Nephrol Dial Transplant 18: 187–189, 2003
ney Int 70: 1783–1792, 2006 20. Moudgil A, Nast CC, Bagga A, Wei L, Nurmamet A, Cohen AH,
2. Thomas DB, Franceschini N, Hogan SL, Ten Holder S, Jennette CE, Jordan SC, Toyoda M: Association of parvovirus B19 infection with
Falk RJ, Jennette JC: Clinical and pathologic characteristics of focal idiopathic collapsing glomerulopathy. Kidney Int 59: 2126 –2133,
segmental glomerulosclerosis pathologic variants. Kidney Int 69: 2001
920 –926, 2006 21. Li RM, Branton MH, Tanawattanacharoen S, Falk RA, Jennette JC,
3. D’Agati VD, Fogo AB, Bruijn JA, Jennette JC: Pathologic classifi- Kopp JB: Molecular identification of SV40 infection in human
cation of focal segmental glomerulosclerosis: A working proposal. subjects and possible association with kidney disease. J Am Soc
Am J Kidney Dis 43: 368 –382, 2004 Nephrol 13: 2320 –2330, 2002
4. Weiss MA, Daquioag E, Margolin EG, Pollak VE: Nephrotic syn- 22. Thaunat O, Delahousse M, Fakhouri F, Martinez F, Stephan JL, Noel
284 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
LH, Karras A: Nephrotic syndrome associated with hemophagocytic Assmann KK, Steenbergen EJ, Wetzels JF: Proliferating cells in HIV
syndrome. Kidney Int 69: 1892–1898, 2006 and pamidronate-associated collapsing focal segmental glomerulo-
23. Greenberg A, Bastacky SI, Iqbal A, Borochovitz D, Johnson JP: sclerosis are parietal epithelial cells. Kidney Int 70: 338 –344, 2006
Focal segmental glomerulosclerosis associated with nephrotic syn- 32. Appel D, Kershaw DB, Smeets B, Yuan G, Fuss A, Frye B, Elger M, Kriz
drome in cholesterol atheroembolism: Clinicopathological correla- W, Floege J, Moeller MJ: Recruitment of podocytes from glomerular
tions. Am J Kidney Dis 29: 334 –344, 1997 parietal epithelial cells. J Am Soc Nephrol 20: 333–343, 2009
24. Thadhani R, Pascual M, Nickeleit V, Tolkoff-Rubin N, Colvin R: 33. Ronconi E, Sagrinati C, Angelotti ML, Lazzeri E, Mazzinghi B,
Preliminary description of focal segmental glomerulosclerosis in Ballerini L, Parente E, Becherucci F, Gacci M, Carini M, Maggi E,
patients with renovascular disease. Lancet 347: 231–233, 1996 Serio M, Vannelli GB, Lasagni L, Romagnani S, Romagnani P:
25. Stokes MB, Davis CL, Alpers CE: Collapsing glomerulopathy in Regeneration of glomerular podocytes by human renal progenitors.
renal allografts: A morphological pattern with diverse clinicopatho- J Am Soc Nephrol 20: 322–332, 2009
logic associations. Am J Kidney Dis 33: 658 – 666, 1999 34. Smeets B, Angelotti ML, Rizzo P, Dijkman H, Lazzeri E, Mooren F,
26. Nadasdy T, Allen C, Zand MS: Zonal distribution of glomerular Ballerini L, Parente E, Sagrinati C, Mazzinghi B, Ronconi E,
collapse in renal allografts: Possible role of vascular changes. Hum Becherucci F, Benigni A, Steenbergen E, Lasagni L, Remuzzi G,
Pathol 33: 437– 441, 2002 Wetzels J, Romagnani P: Renal progenitor cells contribute to hyper-
27. Avila-Casado MC, Vargas-Alarcon G, Soto ME, Hernandez G, Reyes plastic lesions of podocytopathies and crescentic glomerulonephritis.
PA, Herrera-Acosta J: Familial collapsing glomerulopathy: Clinical, J Am Soc Nephrol 20: 2593–2603, 2009
pathological and immunogenetic features. Kidney Int 63: 233–239, 2003 35. Hodgin JB, Borczuk AC, Nasr SH, Markowitz GS, Nair V, Martini S,
28. Berkovic SF, Dibbens LM, Oshlack A, Silver JD, Katerelos M, Vears Eichinger F, Vining C, Berthier CC, Kretzler M, D’Agati VD: A
DF, Lullmann-Rauch R, Blanz J, Zhang KW, Stankovich J, Kalnins molecular profile of focal segmental glomerulosclerosis from forma-
RM, Dowling JP, Andermann E, Andermann F, Faldini E, D’Hooge lin-fixed, paraffin-embedded tissue. Am J Pathol 177: 1674 –1686,
R, Vadlamudi L, Macdonell RA, Hodgson BL, Bayly MA, Savige J, 2010
Mulley JC, Smyth GK, Power DA, Saftig P, Bahlo M: Array-based 36. Kopp JB, Smith MW, Nelson GW, Johnson RC, Freedman BI,
gene discovery with three unrelated subjects shows SCARB2/ Bowden DW, Oleksyk T, McKenzie LM, Kajiyama H, Ahuja TS,
LIMP-2 deficiency causes myoclonus epilepsy and glomerulosclero- Berns JS, Briggs W, Cho ME, Dart RA, Kimmel PL, Korbet SM,
sis. Am J Hum Genet 82: 673– 684, 2008 Michel DM, Mokrzycki MH, Schelling JR, Simon E, Trachtman H,
29. Barisoni L, Kriz W, Mundel P, D’Agati V: The dysregulated podo- Vlahov D, Winkler CA: MYH9 is a major-effect risk gene for focal
cyte phenotype: A novel concept in the pathogenesis of collapsing segmental glomerulosclerosis. Nat Genet 40: 1175–1184, 2008
idiopathic focal segmental glomerulosclerosis and HIV-associated 37. Genovese G, Friedman DJ, Ross MD, Lecordier L, Uzureau P,
nephropathy. J Am Soc Nephrol 10: 51– 61, 1999 Freedman BI, Bowden DW, Langefeld CD, Oleksyk TK, Uscinski
30. Dijkman H, Smeets B, van der Laak J, Steenbergen E, Wetzels J: The Knob AL, Bernhardy AJ, Hicks PJ, Nelson GW, Vanhollebeke B,
parietal epithelial cell is crucially involved in human idiopathic focal Winkler CA, Kopp JB, Pays E, Pollak MR: Association of trypano-
segmental glomerulosclerosis. Kidney Int 68: 1562–1572, 2005 lytic ApoL1 variants with kidney disease in African Americans.
31. Dijkman HB, Weening JJ, Smeets B, Verrijp KC, van Kuppevelt TH, Science 329: 841– 845, 2010
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Section 3: CASE PRESENTATION count of 7200, platelets of 340,000, and normal serum electrolytes
(Na, K, Cl, CO2, and Ca). Urinalysis revealed 3⫹ protein and
A 74-year-old Caucasian woman presents with nephrotic syn- microhematuria without red blood cell casts. Serologies included
drome and microhematuria. Past medical history includes long- negative anti-nuclear antibody, anti-neutrophil cytoplasmic anti-
standing hypothyroidism, depression, and a 3-year history of body, hepatitis B surface antigen, hepatitis C virus antibody, HIV,
hypertension. The patient’s medications include levothyroxine, rheumatoid factor, and cryoglobulins. Serum complements (C3
metoprolol, irbesartan/hydrochlorothiazide, sertraline, montelu- and C4) were within the normal range. Serum protein electropho-
kast, and fexofenadine. Physical examination reveals a BP of resis revealed reduced albumin 3.0 (normal 3.6 to 4.7 g/dl) and
138/80 mmHg and 2⫹ lower extremity edema. The patient denies reduced gamma globulins 0.4 (normal 0.6 to 1.6 g/dl). Both serum
history of diabetes, fever, rash, arthralgias, or gross hematuria. protein electrophoresis and urine protein electrophoresis with
Laboratory studies include serum creatinine level of 1.3 mg/dl, immunofixation were negative for monoclonal protein. Kidney
blood urea nitrogen of 43 mg/dl, 24-hour urine protein of 5.12 g, size by ultrasound was 10.4 and 10.3 cm. A renal biopsy was
serum albumin of 3.3 g/dl, hematocrit of 31.4%, white blood cell performed.
Figure 1. Figure 2.
Figure 4.
Figure 3. IgG.
Figure 5. Figure 6.
285
286 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
depression of C3. In 30% of patients, there was a mono- and one IgG3) exhibiting the same heavy- and light-
clonal serum protein with the same heavy- and light- chain isotypes as had been detected in their native
chain isotype as was detected in the glomerular deposits, kidney biopsies. No patient had evidence of a serum
although only one patient had preexisting myeloma. One M-spike before or after transplantation. Recurrence
patient had primary renal amyloidosis (AL type) diag- was treated with a combination of high-dosage pred-
nosed at the same time as PGNMID on renal biopsy, nisone and rituximab in three patients and prednisone
despite a negative bone marrow for myeloma. Although and cyclophosphamide in one. After a mean posttrans-
several patients had a history of carcinoma, one had plantation follow-up of 43 months, all four patients
underlying HIV infection and one had autoimmune he- had a reduction in proteinuria and histologic activity of
molytic anemia, no consistent clinical profile that iden- the glomerulonephritis. Three patients also had im-
tifies patients at risk for developing PGNMID has proved serum creatinine, with the exception of the
emerged. single patient with intercurrent rejection.
Eighteen (56%) patients received immunomodula-
tory therapy, with or without concurrent renin-angioten- Pathology
sin system blockade (6). These 18 patients included nine Diagnostic criteria for PGNMID (6) include renal
of the 11 patients who had a positive M-spike. Treatment biopsy findings of glomerulonephritis with the following:
was not standardized and consisted of prednisone alone (1) glomerular immune deposits staining positive for ␥
in six patients, prednisone and an alkylating agent (cy- heavy chain (IgG), with negativity for ␣ (IgA) and
clophosphamide or chlorambucil) in three patients, tha- (IgM) heavy chains, indicating restriction to a single (␥)
lidomide in one, bortezomib in one, mycophenolate Ig class; (2) positive staining for a single ␥ (IgG) subclass
mofetil in three, and rituximab in four. On follow-up of (IgG1, IgG2, IgG3, or IgG4); (3) positive staining for a
a mean of 30.3 months (range 1 to 114), available for 32 single light-chain isotype ( or ), indicating monoclo-
patients, 37.5% had complete or partial recovery, 37.5% nality; (4) predominantly granular electron-dense depos-
had persistent renal dysfunction, and 21.9% progressed its in mesangial, subendothelial, and/or subepithelial lo-
to ESRD (6). On univariate analysis, correlates of ESRD cations by electron microscopy, resembling immune
were higher creatinine at biopsy, percentage glomerulo- complex glomerulonephritis; and (5) no clinical or labo-
sclerosis and degree of interstitial fibrosis, but not treat- ratory evidence of cryoglobulinemia.
ment or the presence of M-spike. On multivariate anal-
Light Microscopy. The most common histologic
ysis, a higher percentage of glomerulosclerosis emerged
pattern is membranoproliferative glomerulonephritis, fol-
as the only independent predictor of ESRD (6).
This retrospective, uncontrolled study was un- lowed by endocapillary proliferative glomerulonephritis
able to show a statistical benefit of treatment, perhaps (6,12). Irregular segmental membranous features are fre-
because of the small sample size and the tendency to quent in this setting. There are also rare examples of
treat patients with more severe pathologic features. Of predominantly mesangial proliferative (6,15) or membra-
interest, only one patient lacking M-spike at presenta- nous glomerulonephritis (6,9,16). The membranoprolif-
tion subsequently developed M-spike. In addition, no erative variant is characterized by mesangial expansion
patient with M-spike at presentation subsequently de- by increased cells and matrix as well as mesangial cell
veloped hematologic malignancy such as myeloma or interposition and duplication of GBMs. Infiltrating mac-
B cell lymphoproliferative disorder (6). Longer fol- rophages and neutrophils may be seen. Crescents may
low-ups on a larger number of patients are needed to occur in up to one third of cases but usually involve a
determine the risk for developing a hematologic ma- minority of glomeruli. Most cases are associated with
lignancy and the optimal therapeutic regimen. some degree of interstitial inflammation, fibrosis, and
PGNMID may recur in the allograft (13,17). A tubular atrophy ranging from mild to severe.
recent report described four patients with recurrent Immunofluorescence. The IgG deposits are located
PGNMID documented by allograft biopsy at a mean of exclusively in the glomeruli and usually involve both
3.8 months after transplantation. Recurrence was her- the mesangium and the peripheral glomerular capillary
alded by the development of proteinuria, hematuria, walls. The deposits are commonly granular but occa-
and allograft dysfunction. In all cases, there was glo- sionally exhibit a semilinear or smudgy texture. All
merular deposition of monoclonal IgG3 (three IgG3 cases show light-chain restriction, with sole staining
288 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
for in approximately three quarters of cases and sole membranoproliferative or diffuse proliferative glomeru-
staining for in the remaining one quarter. There may lonephritis pattern, typically with prominent infiltrating
be some nonspecific focal staining for IgM in areas of monocytes and large subendothelial and intracapillary
glomerulosclerosis, but no specific staining for IgM or immune deposits forming “immune thrombi” (2). Ul-
IgA is observed. The majority of cases exhibit depo- trastructurally, the deposits commonly show an annu-
sition of C3 (97%) and C1q (64%) in the distribution lar-tubular or fibrillar substructure. Serum cryoglobu-
of the IgG deposits. PGNMID should be confirmed by lin testing demonstrates a type 1 cryoglobulin
staining for the IgG subtypes (1 through 4) when the composed of monoclonal IgG, usually of isotype.
routine immunofluorescence panel shows light-chain The glomerular deposits in immunotactoid glomerulo-
restriction. By definition, the IgG deposits should stain nephritis are composed of microtubular structures with
for a single IgG isotype, indicating that they are a diameter of 30 to 50 nm often arranged in parallel
monotypic. In the largest series, the 32 cases studied stacks, whereas in fibrillary glomerulonephritis they
included nine IgG1, two IgG2, and 21 IgG3 (6). There are composed of Congo red–negative, randomly ori-
were no examples of monoclonal IgG4. Interestingly, ented fibrils measuring 16 to 24 nm in diameter (3).
IgG1 and IgG2 deposits were more commonly ob- Light- and heavy-chain amyloidosis is extremely rare,
served in patients with a demonstrable M-spike, and, like light-chain amyloidosis, exhibits deposits of
whereas only two of the 21 patients with IgG3 had a Congo red–positive haphazardly oriented fibrils 8 to
positive M-spike. Thus, IgG3 was the most common 14 nm in diameter (4).
subtype in patients lacking an M-spike, suggesting that
it may have particular pathogenetic significance in this Pathogenesis
subgroup. The cause of PGNMID seems to involve the
Electron Microscopy. Non-organized granular exclusively glomerular deposition of a monoclonal
electron-dense deposits are confined to the glomerular IgG molecule. Most cases have deposition of mono-
compartment, without involvement of Bowman’s cap- clonal IgG3. Fewer have been reported with mono-
sule, tubular basement membranes, interstitium, or clonal IgG3, IgG1, IgG1, or IgG2. By immuno-
vessel walls. Most cases have a combination of mes- staining, the deposited IgG molecule contains all three
angial and subendothelial electron-dense deposits in a constant domains (CH1, CH2, and CH3), suggesting
diffuse and global distribution. Subepithelial deposits the deposition of a complete (nondeleted) Ig molecule
occur in approximately one half of cases and are more (5). Only sequencing of the pathogenic IgG would
irregularly distributed, in a predominantly segmental determine whether there are unique mutations or sub-
pattern. The few cases with global subepithelial de- stitutions that predispose to glomerular deposition by
posits are those with predominantly membranous fea- promoting self-aggregability, enhancing charge inter-
tures by light microscopy. In some cases, the deposits actions with glomerular constituents, or increasing
have a variegated texture, with areas of greater or hydrophobicity. Because the CH2 domain is present, it
lesser electron density. However, organized fibrils and is not surprising that activation of complement is
microtubules are not typically observed. commonly observed as glomerular co-deposits of C1q
and C3, which may lead to hypocomplementemia.
Differential Diagnosis The four IgG subtypes differ in their quantity and
Other glomerular diseases with monoclonal IgG chemical properties. IgG3 comprises only 8% of total
deposits can be distinguished from PGNMID on the circulating IgG, but it has properties that could make it
basis of their combined pathologic and clinical fea- intrinsically “nephritogenic” (18 –20). For example, it is
tures. Randall-type LHCDD is characterized by nod- the most positively charged subclass (pI 8.2 to 9.0),
ular glomerulosclerosis by light microscopy; diffuse which could promote binding to negative charge sites in
linear staining of glomerular and tubular basement the glomerular capillary wall. It has the largest molecular
membranes for a single heavy chain and a single light weight (170,000 Da), which makes it likely to become
chain by immunofluorescence; and nonfibrillar, punc- concentrated in the glomerular capillaries during filtra-
tate electron-dense deposits in the GBMs and tubular tion. IgG3 also has the capacity to self-aggregate via
basement membranes by electron microscopy (1). Fc–Fc interactions, which might favor formation of in-
Type 1 cryoglobulinemic glomerulonephritis exhibits a traglomerular electron-dense aggregates. Interestingly,
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 289
IgG3 also is the most common isotype in murine and Appel GB, Aucouturier P, D’Agati VD: Proliferative glomerulone-
phritis with monoclonal IgG deposits: A distinct entity mimicking
human lupus nephritis, cryoglobulinemia, and IgG my- immune-complex glomerulonephritis. Kidney Int 65: 85–96, 2004
eloma hyperviscosity syndrome (19,20). 6. Nasr SH, Satoskar A, Markowitz GS, Valeri AM, Appel GB, Stokes
The ability of some monoclonal IgG to self-aggre- MB, Nadasdy T, D’Agati VD: Proliferative glomerulonephritis with
monoclonal IgG deposits. J Am Soc Nephrol 20: 2055–2064, 2009
gate in vitro suggests that the glomerular IgG deposits 7. Evans DJ, Macanovic M, Dunn MJ, Pusey CD: Membranous glo-
may not be complexed to antigen (16). However, because merulonephritis associated with follicular B-cell lymphoma and sub-
only 30% of patients have a corresponding M-spike and epithelial deposition of IgG1-kappa paraprotein. Nephron Clin Pract
93: c112— c118, 2003
evidence of myeloma is rare, it remains unclear whether 8. Lee JG, Moon KC, Lee JE, Kim P, Lee JG, Kim JH, Lee KY: A case
the monoclonal IgG in some patients could deposit in the of proliferative glomerulonephritis with monoclonal IgG deposits.
course of an antigen-driven immune response, perhaps as Korean J Nephrol 23: 987–991, 2004
9. Komatsuda A, Masai R, Ohtani H, Togashi M, Maki N, Sawada K,
part of an antigen-antibody complex formed by specific Wakui H: Monoclonal immunoglobulin deposition disease associated
B cell clones. Challenge by foreign antigen typically with membranous features. Nephrol Dial Transplant 23: 3888 –3894,
elicits a polyclonal immune response. Hypothetically, in 2008
10. Bridoux F, Zanetta G, Mougenot B, Goujon JM, Vanhille P, Bauwens
the course of an immunologic response to exogenous or M, Chevet D, Ronco P, Preud’homme JL, Touchard G: Glomeru-
endogenous antigens, one or more clones of B cells could lopathy with non-organized and non-Randall type monoclonal immu-
proliferate and produce monoclonal IgG molecules (par- noglobulin deposits: A rare entity [Abstract]. J Am Soc Nephrol 12:
94A, 2001
ticularly IgG3) with the ability to self-aggregate and
11. Geldenhuys L, Jones B: Glomerulonephritis with monoclonal immu-
rapidly deposit in glomeruli through entrapment and/or noglobulin deposits [Abstract]. J Am Soc Nephrol 19: 671A, 2008
interaction with negative charge sites in the glomerular 12. Masai R, Wakui H, Komatsuda A, Togashi M, Maki N, Ohtani H,
Oyama Y, Sawada K: Characteristics of proliferative glomerulo-
capillary walls. The small quantity of this monoclonal
nephritis with monoclonal IgG deposits associated with mem-
IgG may elude detection by serum protein electrophore- branoproliferative features. Clin Nephrol 72: 46 –54, 2009
sis with immunofixation because of its avidity for the 13. Albawardi A, Sataskar A, Brodsky S, Nadasdy GM, Nadasdy T:
Proliferative glomerulonephritis with monoclonal IgG deposits recurs
glomeruli and rapid aggregability favored by the mole-
or may develop de novo in renal allografts. [Abstract]. Mod Pathol
cule’s intrinsic physical properties and the high intraglo- 23: 337A, 2010
merular concentrations reached during glomerular siev- 14. Alpers CE, Tu WH, Hopper J Jr, Biava CG: Single light chain
subclass (kappa chain) immunoglobulin deposition in glomerulone-
ing. The possibility of an oligoclonal IgG response with
phritis. Hum Pathol 16: 294 –304, 1985
the same light- and heavy-chain isotypes also cannot be 15. Komatsuda A, Wakui H, Ohtani H, Nimura T, Sawada K: Steroid-
excluded on the basis of the renal biopsy findings be- responsive nephrotic syndrome in a patient with proliferative glomer-
ulonephritis with monoclonal IgG deposits with pure mesangial
cause monoclonality can be proved only by immunoblot-
proliferative features. NDT Plus May 2, 2010 [epub ahead of print]
ting/immunofixation techniques. 16. de Seigneux S, Bindi P, Debiec H, Alyanakian MA, Aymard B,
Callard P, Ronco P, Aucouturier P: Immunoglobulin deposition
References disease with a membranous pattern and a circulating monoclonal
1. Lin J, Markowitz GS, Valeri AM, Kambham N, Sherman WH, Appel immunoglobulin G with charge-dependent aggregation properties.
GB, D’Agati VD: Renal monoclonal immunoglobulin deposition Am J Kidney Dis 56: 117–121, 2010
disease: The disease spectrum. J Am Soc Nephrol 12: 1482–1492, 17. Nasr SH, Sethi S, Cornell LD, Fidler ME, Boelkins M, Fervenza FC,
2001 Cosio FG, D’Agati VD: Proliferative glomerulonephritis with mono-
2. Nasr SH, Markowitz GS, Reddy BS, Maesaka J, Swidler MA, clonal IgG deposits recurs in the renal transplant. Clin J Am Soc
D’Agati VD: Dysproteinemia, proteinuria, and glomerulonephritis. Nephrol 6: 122–132, 2011
Kidney Int 69: 772–775, 2006 18. Grey HM, Hirst JW, Cohn M: A new mouse immunoglobulin: IgG3.
3. Rosenstock JL, Markowitz GS, Valeri AM, Sacchi G, Appel GB, J Exp Med 133: 289 –304, 1971
D’Agati VD: Fibrillary and immunotactoid glomerulonephritis: Dis- 19. Capra JD, Kunkel HG: Aggregation of gamma-G3 proteins: Rele-
tinct entities with different clinical and pathologic features. Kidney vance to the hyperviscosity syndrome. J Clin Invest 49: 610 – 621,
Int 63: 1450 –1461, 2003 1970
4. Nasr SH, Colvin R, Markowitz GS: IgG1 lambda light and heavy 20. Abdelmoula M, Spertini F, Shibata T, Gyotoku Y, Luzuy S, Lambert
chain renal amyloidosis. Kidney Int 70: 7, 2006 PH, Izui S: IgG3 is the major source of cryoglobulins in mice.
5. Nasr SH, Markowitz GS, Stokes MB, Seshan SV, Valderrama E, J Immunol 143: 526 –532, 1989
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Section 4: CASE PRESENTATION RBC casts. Laboratory evaluation shows serum creatinine
level of 1.4 mg/dl, 24-hour urine protein of 7.2 g/d, serum
A 27-year-old African-American woman presents with albumin of 2.4 g/dl, decreased serum complements (C3 62
nephrotic syndrome during the first trimester of her first [normal 80 to 180]; C4 ⬍6 [normal 10 to 45]), positive
pregnancy. She has a past history of hepatitis C virus (HCV) rheumatoid factor (RF), and positive type 2 mixed cryo-
infection (presumed to be from tattoos) and poorly con- globulin (IgM-/IgG). All other serologic tests are negative
trolled hypertension of unknown duration. Physical exami- or normal, including anti-nuclear antibody, anti-DNA anti-
nation reveals BP of 160/100 and 2⫹ lower extremity body, antistreptolysin-O, anti-neutrophil cytoplasmic anti-
edema. Urinalysis shows 4⫹ protein and 15 red blood cells body, anti– glomerular basement membrane (anti-GBM) an-
(RBCs) per high-power field, with dysmorphic RBCs but no tibody, and hepatitis B surface antigen.
Figure 1. Figure 2.
tal glomerular sclerosis among patients infected with hepatitis C 15. Markowitz GS, Nasr SH, Stokes MB, D’Agati VD: Treatment with
virus. Nephron 81: 37– 40, 1999 IFN-alpha, -beta, or -gamma is associated with collapsing focal
4. Meyers CM, Seeff LB, Stehman-Breen CO, Hoofnagle JH: Hepatitis segmental glomerulosclerosis. Clin J Am Soc Nephrol 5: 607– 615,
C and renal disease: An update. Am J Kidney Dis 42: 631– 657, 2003 2010
5. Fabrizi F, Pozzi C, Farina M, Dattolo P, Lunghi G, Badalamenti S, 16. Feiner H, Gallo G: Ultrastructure in glomerulonephritis associated
Pagano A, Locatelli F: Hepatitis C virus infection and acute or with cryoglobulinemia: A report of six cases and review of the
chronic glomerulonephritis: An epidemiological and clinical ap- literature. Am J Pathol 88: 145–162, 1977
praisal. Nephrol Dial Transplant 13: 1991–1997, 1998 17. Brunkhorst R, Kliem V, Koch KM: Recurrence of membranoprolif-
6. Johnson RJ, Gretch DR, Yamabe H, Hart J, Bacchi CE, Hartwell P, erative glomerulonephritis after renal transplantation in a patient with
Couser WG, Corey L, Wener MH, Alpers CE, Willson R: Mem- chronic hepatitis C. Nephron 72: 465– 467, 1996
branoproliferative glomerulonephritis associated with hepatitis C 18. KDIGO clinical practice guidelines for the prevention, diagnosis,
virus infection. N Engl J Med 328: 465– 470, 1993 evaluation, and treatment of hepatitis C in chronic kidney disease.
7. Beddhu S, Bastacky S, Johnson JP: The clinical and morphologic Kidney Int Suppl S1–S99, 2008
spectrum of renal cryoglobulinemia. Medicine (Baltimore) 81: 398 – 19. Nachman PH, Glassock RJ: Glomerular, vascular, and tubulointer-
409, 2002 stitial diseases. NephSAP 9: 161, 2010
8. Bryce AH, Kyle RA, Dispenzieri A, Gertz MA: Natural history and 20. Fabrizi F, Lunghi G, Messa P, Martin P: Therapy of hepatitis C
therapy of 66 patients with mixed cryoglobulinemia. Am J Hematol virus-associated glomerulonephritis: current approaches. J Nephrol
81: 511–518, 2006
21: 813– 825, 2008
9. McGuire BM, Julian BA, Bynon JS Jr, Cook WJ, King SJ, Curtis JJ,
21. Stehman-Breen C, Alpers CE, Couser WG, Willson R, Johnson RJ:
Accortt NA, Eckhoff DE: Brief communication: Glomerulonephritis
Hepatitis C virus associated membranous glomerulonephritis. Clin
in patients with hepatitis C cirrhosis undergoing liver transplantation.
Nephrol 44: 141–147, 1995
Ann Intern Med 144: 735–741, 2006
22. Ray S, Rouse K, Appis A, Novak R, Haller NA: Fibrillary glomer-
10. Stokes MB, Chawla H, Brody RI, Kumar A, Gertner R, Goldfarb DS,
ulonephritis with hepatitis C viral infection and hypocomple-
Gallo G: Immune complex glomerulonephritis in patients coinfected
mentemia. Ren Fail 30: 759 –762, 2008
with human immunodeficiency virus and hepatitis C virus. Am J
23. Alpers CE, Smith KD: Cryoglobulinemia and renal disease. Curr
Kidney Dis 29: 514 –525, 1997
11. Cheng JT, Anderson HL Jr, Markowitz GS, Appel GB, Pogue VA, Opin Nephrol Hypertens 17: 243–249, 2008
D’Agati VD: Hepatitis C virus-associated glomerular disease in 24. Ferri S, Dal Pero F, Bortoletto G, Bianchi FB, Lenzi M, Alberti A,
patients with human immunodeficiency virus coinfection. J Am Soc Gerotto M: Detailed analysis of the E2-IgM complex in hepatitis
Nephrol 10: 1566 –1574, 1999 C-related type II mixed cryoglobulinaemia. J Viral Hepat 13: 166 –
12. Ahmed MS, Wong CF, Shawki H, Kapoor N, Pandya BK: Rapidly 176, 2006
deteriorating renal function with membranoproliferative glomerulo- 25. Fornasieri A, Armelloni S, Bernasconi P, Li M, de Septis CP, Sinico
nephritis type 1 associated with hepatitis C treated successfully with RA, D’Amico G: High binding of immunoglobulin M kappa rheu-
steroids and antiviral therapy: A case report and review of literature. matoid factor from type II cryoglobulins to cellular fibronectin: a
Clin Nephrol 69: 298 –301, 2008 mechanism for induction of in situ immune complex glomerulone-
13. Guerra G, Narayan G, Rennke HG, Jaber BL: Crescentic fibrillary phritis? Am J Kidney Dis 27: 476 – 483, 1996
glomerulonephritis associated with hepatitis C viral infection. Clin 26. Cao Y, Zhang Y, Wang S, Zou W: Detection of the hepatitis C virus
Nephrol 60: 364 –368, 2003 antigen in kidney tissue from infected patients with various glomer-
14. Baid S, Pascual M, Williams WW Jr, Tolkoff-Rubin N, Johnson SM, ulonephritis. Nephrol Dial Transplant 24: 2745–2751, 2009
Collins B, Chung RT, Delmonico FL, Cosimi AB, Colvin RB: Renal 27. Kowalewska J, Muhlfeld AS, Hudkins KL, Yeh MM, Farr AG,
thrombotic microangiopathy associated with anticardiolipin antibod- Ravetch JV, Alpers CE: Thymic stromal lymphopoietin transgenic
ies in hepatitis C-positive renal allograft recipients. J Am Soc Nephrol mice develop cryoglobulinemia and hepatitis with similarities to
10: 146 –153, 1999 human hepatitis C liver disease. Am J Pathol 170: 981–989, 2007
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Section 5: CASE PRESENTATION serum albumin level of 3.6 g/dl. Urinalysis contains 300
mg/dl protein, and microscopic examination of the uri-
A 57-year-old Caucasian man is discovered to have nary sediment reveals 8 red blood cells (RBCs) per
microscopic hematuria, subnephrotic proteinuria, and re- high-power field and no casts. Serum IgA level is ele-
nal insufficiency. Past medical history is significant for vated, but all other serologies are negative or normal,
hypertension for 3 years. There is no history of diabetes including serum complement levels (C3 and C4), anti-
and no family history of renal disease. Physical exami- nuclear antibody, hepatitis B surface antigen, hepatitis C
nation reveals no edema and BP of 120/92 mmHg on antibody, anti-neutrophil cytoplasmic antibody (ANCA),
antihypertensive medications. The patient denies any and anti– glomerular basement membrane (anti-GBM)
history of recent infection, rash, arthralgias, or abdominal antibody. No monoclonal spike is detected by serum
pain. Laboratory evaluation includes serum creatinine protein electrophoresis. The kidneys measure 11.7 and
level of 1.7 mg/dl, 24-hour urine protein of 1.0 g, and 12.2 cm in length by ultrasound.
Figure 3.
IgAN. The degree of tubular atrophy and interstitial biopsies. There is typically staining for complement
fibrosis ranges from mild to severe (Figures 8 and 9) C3, without C1q, consistent with complement acti-
and is usually commensurate with the degree of glomeru- vation via the alternative and/or lectin pathways.
losclerosis. Proximal tubules may contain RBC casts and Staining for light chain is usually stronger than ,
show degenerative and regenerative features. Arteriosclero- reflecting the normal predominance of the isotype
sis and arteriolosclerosis are more common in older patients in circulating IgA molecules. Peripheral capillary
and those with a history of hypertension. Features of acute wall staining for IgA is seen in cases with endocap-
thrombotic microangiopathy (i.e., intravascular thrombi) illary proliferative or membranoproliferative fea-
may be seen in cases of advanced chronic IgAN with severe tures. Rare cases of coexistent IgAN and membra-
hypertension (12). Arteritis is not a feature of IgAN and, if nous nephropathy have been described and can be
present, suggests the differential diagnoses of HSP nephritis identified by their diffuse granular capillary wall
or coexistent ANCA vasculitis. staining for IgG and mesangial staining for IgA
Immunofluorescence. Dominant or co-dominant (13).
mesangial staining for IgA is the hallmark of IgAN. Electron Microscopy. Electron-dense deposits are
Weaker staining for IgG and IgM is seen in some seen at sites corresponding to the immunofluorescence
staining. Mesangial deposits are the most typical find-
ing and tend to aggregate beneath the GBM reflection
over the mesangium, a location referred to as “parame-
sangial” (Figure 3). Peripheral capillary wall deposits
are less common and usually subendothelial, associ-
ated with endocapillary proliferation. Occasional seg-
mental intramembranous or subepithelial deposits may
also occur. Some cases have localized textural changes
in the GBM such as segmental thinning and basket-
weave lamellation. These findings may reflect remod-
eling of basement membranes following resorption of
capillary wall deposits and are a likely source of
glomerular hematuria. Diffuse thinning of GBMs
should suggest coexistent thin basement membrane
Figure 8. This case of IgAN has well-preserved tubules with nephropathy. Podocyte foot process effacement is gen-
no significant interstitial fibrosis. Magnification, ⫻200 erally mild and focal in mesangial IgAN but more
(trichrome). diffuse in endocapillary proliferative forms with wide-
spread peripheral capillary wall deposits. Rare cases of
IgAN show diffuse foot process effacement and ex-
clusively mesangial deposits; if this is accompanied by
a clinical history of abrupt onset of nephrotic syn-
drome, a diagnosis of minimal change disease with
IgA deposits is warranted.
The differential diagnosis of IgAN includes other
conditions in which dominant IgA glomerular deposits
may occur. HSP nephritis is considered to be a sys-
temic form of IgAN manifesting purpuric rash (typi-
cally of the lower extremities), arthralgias, abdominal
pain/hematochezia, and glomerulonephritis. Not all
patients with HSP exhibit the full clinical syndrome.
Figure 9. In this example of IgAN, interstitial fibrosis and
HSP can occur at any age but is most prevalent in
tubular atrophy occupy approximately 60% of the cortex. young children and is usually transient. In some pa-
Magnification, ⫻200 (trichrome). tients with HSP, the multisystem disease resolves but
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 301
active IgAN persists for years. Deposits of IgA are related with renal outcomes, neither has been uni-
detectable in the glomeruli and in vasculitic lesions in versally adopted (7). Moreover, pathologic class did
the skin and gut. Other diagnostic considerations in- not predict renal outcomes independent of clinical
clude lupus nephritis, IgA-dominant acute postinfec- presenting features (e.g., hypertension, proteinuria)
tious glomerulonephritis (14,15), and HIV-related im- (21) or show superiority to a semiquantitative as-
mune complex glomerulonephritis (16). In addition, sessment of the degree of tubular atrophy and inter-
IgA cryoglobulinemia and IgA monoclonal gammopa- stitial fibrosis (22). The Oxford Classification of
thy may give rise to glomerular disease with IgA- IgAN was designed to identify pathologic features
dominant deposits. Clinical-pathologic correlation is that show high interobserver reproducibility and
necessary to distinguish these conditions from IgAN. predict adverse clinical outcome (rate of decline of
estimated GFR) independent of clinical variables at
Clinical-Pathologic Correlation time of biopsy or during follow-up (6,7). Therefore,
Most patients who have IgAN and present with it is best considered a prognostic grading system
hematuria, mild proteinuria, and preserved kidney rather than a comprehensive classification. Impor-
function have biopsy findings of mesangial hyper- tantly, because patients with HSP, proteinuria ⬍0.5
cellularity, focal proliferation, and/or focal glomer- g/d, estimated GFR ⬍30 ml/min, or ESRD in ⬍12
ulosclerosis. Heavier proteinuria and renal insuffi- months from onset were excluded, the Oxford co-
ciency are associated with more diffuse hort does not represent the complete spectrum of
endocapillary proliferation and subendothelial cap- IgAN. Among the study group of 265 IgA biopsies,
illary wall IgA deposits, as well as more sclerosing one third from children who were younger than 18
glomerular lesions. Diffuse crescentic disease years, four pathologic variables that fulfilled the
(⬎50% crescents) is associated with rapidly pro- aforementioned criteria were identified: Mesangial
gressive renal failure and acute nephritic syndrome. hypercellularity, Endothelial hypercellularity, Seg-
Some cases of IgAN with gross hematuria and acute mental glomerulosclerosis, and Tubular atrophy-
renal failure show relatively mild glomerular dis- interstitial fibrosis. These pathologic lesions consti-
ease but severe acute tubular injury as a result of tute the Oxford-MEST score (Table 1) (6,7).
numerous RBC casts. In this context, the acute The M (mesangial) score is calculated as fol-
kidney injury usually resolves as the hematuria lows: Each glomerulus is graded as 0 (⬍4 mesangial
subsides. IgAN presenting with chronic renal failure cells/mesangial area), 1 (4 to 5 mesangial cells/
generally shows diffuse glomerulosclerosis and ad- mesangial area), 2 (6 to 7 mesangial cells/mesangial
vanced tubular atrophy and interstitial fibrosis. area), or 3 (ⱖ8 mesangial cells/mesangial area). An
Cases with severe/accelerated hypertension may de- average score ⱕ0.5 is assigned M0, and a score of
velop arteriolar changes of thrombotic microangi- ⬎0.5 is assigned M1. In practice, if ⬎50% glomer-
opathy. uli have ⬎3 cells in a mesangial area, then the score
Numerous studies have identified associations is M1 (6). E1 is defined by increased number of
between clinical features and outcomes in IgAN. Of cells within glomerular capillary lumina causing
these, the most significant are elevated serum creati- narrowing of the lumina. S1 is defined by subtotal
nine (or low GFR), degree of proteinuria, persistence capillary obliteration by sclerosis or a synechial
of proteinuria despite therapy, and elevated mean attachment to Bowman’s capsule. Tubular atrophy
arterial BP. Most— but not all (17)—studies indicate and interstitial fibrosis are graded on the basis of the
worse outcomes in male patients. area of cortex involved: T0, 0 to 25%; T1, 26 to
Overall, childhood IgAN seems to have a more 50%; or T2, ⬎50%. Each of these pathologic fea-
benign course than the adult disease, and proliferative tures (M ⬎0.5, E1, S1, T1, and T2) are indepen-
lesions seem to have better outcomes in children dently predictive of clinical outcome in both chil-
compared with adults (18). dren (23) and adults with IgAN. Compared with
Oxford Classification/MEST Score. Several adults, children had more severe mesangial prolif-
pathologic classifications of IgAN have been pro- eration, more endocapillary proliferation, less
posed, including those of Lee et al. (19) and Haas chronic tubulointerstitial disease, and less arterio-
(20). Although these two classification systems cor- sclerosis (23). Adoption of this schema should stan-
302 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
molecules or via molecular mimicry between the un- 11. Bollee G, Noel LH, Suarez F, Royal V, Gilardin L, de Serre NP,
El-Ghoul B, Lesavre P, Alyanakian MA, Fakhouri F: Pauci-immune
dergalactosylated IgA1 and microbial antigens. The crescentic glomerulonephritis associated with ANCA of IgA class.
defect in galactosylation also may promote self-aggre- Am J Kidney Dis 53: 1063–1067, 2009
gation of IgA1 molecules. The abnormal IgA1 mole- 12. Chang A, Kowalewska J, Smith KD, Nicosia RF, Alpers CE: A
clinicopathologic study of thrombotic microangiopathy in the setting
cules seem to be inherently more “sticky,” promoting of IgA nephropathy. Clin Nephrol 66: 397– 404, 2006
increased glomerular deposition and reduced mesan- 13. Stokes MB, Alpers CE: Combined membranous nephropathy and
gial clearance. In addition, mesangial deposition may IgA nephropathy. Am J Kidney Dis 32: 649 – 656, 1998
14. Nasr SH, Markowitz GS, Whelan JD, Albanese JJ, Rosen RM, Fein
be favored by interactions with transferrin receptor, DA, Kim SS, D’Agati VD: IgA-dominant acute poststaphylococcal
fibronectin, or Fc␣ receptor on mesangial cells. Sub- glomerulonephritis complicating diabetic nephropathy. Hum Pathol
sequent events include complement activation via the 34: 1235–1241, 2003
15. Satoskar AA, Nadasdy G, Plaza JA, Sedmak D, Shidham G, Hebert
alternative and mannose-binding lectin pathways and L, Nadasdy T: Staphylococcus infection-associated glomerulonephri-
release of cytokines that lead to glomerular hypercel- tis mimicking IgA nephropathy. Clin J Am Soc Nephrol 1: 1179 –
lularity, matrix production, podocyte injury, and scar- 1186, 2006
16. Kimmel PL, Phillips TM, Ferreira-Centeno A, Farkas-Szallasi T,
ring. Recent emphasis has been placed on podocyte Abraham AA, Garrett CT: Brief report: Idiotypic IgA nephropathy in
depletion as a pathway of progressive glomerular scar- patients with human immunodeficiency virus infection. N Engl J Med
327: 702–706, 1992
ring through the development of segmental lesions of
17. Cattran DC, Reich HN, Beanlands HJ, Miller JA, Scholey JW,
sclerosis (36). Troyanov S: The impact of sex in primary glomerulonephritis.
Nephrol Dial Transplant 23: 2247–2253, 2008
18. Haas M, Rahman MH, Cohn RA, Fathallah-Shaykh S, Ansari A,
References Bartosh SM: IgA nephropathy in children and adults: Comparison of
1. Tumlin JA, Madaio MP, Hennigar R: Idiopathic IgA nephropathy: histologic features and clinical outcomes. Nephrol Dial Transplant
Pathogenesis, histopathology, and therapeutic options. Clin J Am Soc 23: 2537–2545, 2008
Nephrol 2: 1054 –1061, 2007 19. Lee SM, Rao VM, Franklin WA, Schiffer MS, Aronson AJ, Spargo
2. Pouria S, Barratt J: Secondary IgA nephropathy. Semin Nephrol 28: BH, Katz AI: IgA nephropathy: Morphologic predictors of progres-
27–37, 2008 sive renal disease. Hum Pathol 13: 314 –322, 1982
3. D’Amico G: Natural history of idiopathic IgA nephropathy: Role of 20. Haas M: Histologic subclassification of IgA nephropathy: A clinico-
clinical and histological prognostic factors. Am J Kidney Dis 36: pathologic study of 244 cases. Am J Kidney Dis 29: 829 – 842, 1997
227–237, 2000 21. Bartosik LP, Lajoie G, Sugar L, Cattran DC: Predicting progression
4. Reich HN, Troyanov S, Scholey JW, Cattran DC: Remission of in IgA nephropathy. Am J Kidney Dis 38: 728 –735, 2001
proteinuria improves prognosis in IgA nephropathy. J Am Soc Neph- 22. Daniel L, Saingra Y, Giorgi R, Bouvier C, Pellissier JF, Berland Y:
rol 18: 3177–3183, 2007 Tubular lesions determine prognosis of IgA nephropathy. Am J
5. D’Amico G: Natural history of idiopathic IgA nephropathy and Kidney Dis 35: 13–20, 2000
factors predictive of disease outcome. Semin Nephrol 24: 179 –196, 23. Coppo R, Troyanov S, Camilla R, Hogg RJ, Cattran DC, Cook HT,
2004 Feehally J, Roberts IS, Amore A, Alpers CE, Barratt J, Berthoux F,
6. Cattran DC, Coppo R, Cook HT, Feehally J, Roberts IS, Troyanov S, Bonsib S, Bruijn JA, D’Agati V, D’Amico G, Emancipator SN,
Alpers CE, Amore A, Barratt J, Berthoux F, Bonsib S, Bruijn JA, Emma F, Ferrario F, Fervenza FC, Florquin S, Fogo AB, Geddes CC,
D’Agati V, D’Amico G, Emancipator S, Emma F, Ferrario F, Fer- Groene HJ, Haas M, Herzenberg AM, Hill PA, Hsu SI, Jennette JC,
venza FC, Florquin S, Fogo A, Geddes CC, Groene HJ, Haas M, Joh K, Julian BA, Kawamura T, Lai FM, Li LS, Li PK, Liu ZH,
Herzenberg AM, Hill PA, Hogg RJ, Hsu SI, Jennette JC, Joh K, Mezzano S, Schena FP, Tomino Y, Walker PD, Wang H, Weening JJ,
Julian BA, Kawamura T, Lai FM, Leung CB, Li LS, Li PK, Liu ZH, Yoshikawa N, Zhang H: The Oxford IgA nephropathy clinicopatho-
Mackinnon B, Mezzano S, Schena FP, Tomino Y, Walker PD, Wang logical classification is valid for children as well as adults. Kidney Int
H, Weening JJ, Yoshikawa N, Zhang H: The Oxford classification of 77: 921–927, 2010
IgA nephropathy: Rationale, clinicopathological correlations, and 24. Eitner F, Floege J: Glomerular disease: The Oxford classification—
classification. Kidney Int 76: 534 –545, 2009 Predicting progression of IgAN. Nat Rev Nephrol 5: 557–559, 2009
7. Roberts IS, Cook HT, Troyanov S, Alpers CE, Amore A, Barratt J, 25. Gharavi AG, Yan Y, Scolari F, Schena FP, Frasca GM, Ghiggeri GM,
Berthoux F, Bonsib S, Bruijn JA, Cattran DC, Coppo R, D’Agati V, Cooper K, Amoroso A, Viola BF, Battini G, Caridi G, Canova C, Farhi A,
D’Amico G, Emancipator S, Emma F, Feehally J, Ferrario F, Fer- Subramanian V, Nelson-Williams C, Woodford S, Julian BA, Wyatt RJ,
venza FC, Florquin S, Fogo A, Geddes CC, Groene HJ, Haas M, Lifton RP: IgA nephropathy, the most common cause of glomerulonephritis,
Herzenberg AM, Hill PA, Hogg RJ, Hsu SI, Jennette JC, Joh K, is linked to 6q22-23. Nat Genet 26: 354–357, 2000
Julian BA, Kawamura T, Lai FM, Li LS, Li PK, Liu ZH, Mackinnon 26. Liu XQ, Paterson AD, He N, St George-Hyslop P, Rauta V, Gron-
B, Mezzano S, Schena FP, Tomino Y, Walker PD, Wang H, Weening hagen-Riska C, Laakso M, Thibaudin L, Berthoux F, Cattran D, Pei
JJ, Yoshikawa N, Zhang H: The Oxford classification of IgA ne- Y: IL5RA and TNFRSF6B gene variants are associated with sporadic
phropathy: Pathology definitions, correlations, and reproducibility. IgA nephropathy. J Am Soc Nephrol 19: 1025–1033, 2008
Kidney Int 76: 546 –556, 2009 27. Feehally J, Farrall M, Boland A, Gale DP, Gut I, Heath S, Kumar A,
8. Galla JH: IgA nephropathy. Kidney Int 47: 377–387, 1995 Peden JF, Maxwell PH, Morris DL, Padmanabhan S, Vyse TJ,
9. Jennette JC, Wall SD, Wilkman AS: Low incidence of IgA nephrop- Zawadzka A, Rees AJ, Lathrop M, Ratcliffe PJ: HLA has strongest
athy in blacks. Kidney Int 28: 944 –950, 1985 association with IgA nephropathy in genome-wide analysis. J Am Soc
10. Ibels LS, Gyory AZ: IgA nephropathy: Analysis of the natural Nephrol 21: 1791–1797, 2010
history, important factors in the progression of renal disease, and a 28. Yamamoto R, Nagasawa Y, Shoji T, Inoue K, Uehata T, Kaneko T,
review of the literature. Medicine (Baltimore) 73: 79 –102, 1994 Okada T, Yamauchi A, Tsubakihara Y, Imai E, Isaka Y, Rakugi H:
304 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
A candidate gene approach to genetic prognostic factors of IgA 33. Chintalacharuvu SR, Yamashita M, Bagheri N, Blanchard TG, Ne-
nephropathy: A result of Polymorphism REsearch to DIstinguish drud JG, Lamm ME, Tomino Y, Emancipator SN: T cell cytokine
genetic factors Contributing To progression of IgA Nephropathy polarity as a determinant of immunoglobulin A (IgA) glycosylation
(PREDICT-IgAN). Nephrol Dial Transplant 24: 3686 –3694, 2009 and the severity of experimental IgA nephropathy. Clin Exp Immunol
29. Smerud HK, Fellstrom B, Hallgren R, Osagie S, Venge P, Kristjans- 153: 456 – 462, 2008
son G: Gluten sensitivity in patients with IgA nephropathy. Nephrol 34. Glassock RJ: Analyzing antibody activity in IgA nephropathy. J Clin
Dial Transplant 24: 2476 –2481, 2009 Invest 119: 1450 –1452, 2009
30. Qin W, Zhong X, Fan JM, Zhang YJ, Liu XR, Ma XY: External
35. Suzuki H, Fan R, Zhang Z, Brown R, Hall S, Julian BA, Chatham
suppression causes the low expression of the Cosmc gene in IgA
WW, Suzuki Y, Wyatt RJ, Moldoveanu Z, Lee JY, Robinson J,
nephropathy. Nephrol Dial Transplant 23: 1608 –1614, 2008
Tomana M, Tomino Y, Mestecky J, Novak J: Aberrantly glycosy-
31. Suzuki H, Moldoveanu Z, Hall S, Brown R, Vu HL, Novak L, Julian
lated IgA1 in IgA nephropathy patients is recognized by IgG anti-
BA, Tomana M, Wyatt RJ, Edberg JC, Alarcon GS, Kimberly RP,
Tomino Y, Mestecky J, Novak J: IgA1-secreting cell lines from bodies with restricted heterogeneity. J Clin Invest 119: 1668 –1677,
patients with IgA nephropathy produce aberrantly glycosylated IgA1. 2009
J Clin Invest 118: 629 – 639, 2008 36. El Karoui K, Hill GS, Karras A, Moulonguet L, Caudwell V,
32. Nishie T, Miyaishi O, Azuma H, Kameyama A, Naruse C, Hashimoto Loupy A, Bruneval P, Jacquot C, Nochy D: Focal segmental
N, Yokoyama H, Narimatsu H, Wada T, Asano M: Development of glomerulosclerosis plays a major role in the progression of IgA
immunoglobulin A nephropathy-like disease in beta-1,4-galactosyl- nephropathy. II. Light microscopic and clinical studies. Kidney Int
transferase-I-deficient mice. Am J Pathol 170: 447– 456, 2007 79: 643– 654, 2011
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Figure 1. Figure 2.
Figure 3. Figure 4.
References
1. Pirani CL, Silva F, Nasr SH, D’Agati VD, Chander P, Striker LM:
Renal lesions in plasma cell dyscrasias: Ultrastructural observation.
Am J Kidney Dis 10: 208 –221, 1987
2. Lin J, Markowitz GS, Valeri AM, Kambham N, Sherman WH, Appel
Figure 8. The ultrastructural finding of abundant, markedly GB, D’Agati VD: Renal monoclonal immunoglobulin deposition
disease: The disease spectrum. J Am Soc Nephrol 12: 1482–1492,
electron-dense, rounded and angulated intracellular crystals
2001
within the cytoplasm of proximal tubular epithelium is 3. Lasser EC, Lang JH, Zawadzki ZA: Myeloma protein precipitates in
highly characteristic of LCFS. Magnification, ⫻4000. urography. JAMA 198: 273–275, 1966
4. Cohen DJ, Sherman WH, Osserman EF, Appel GB: Acute renal
failure in patients with multiple myeloma. Am J Med 76: 247–256,
1984
5. Irish AB, Winearls CG, Littlewood T: Presentation and survival of
patients with severe renal failure and myeloma. QJM 90: 773–780,
1997
6. Kyle RA: Multiple myeloma: Review of 869 cases. Mayo Clin Proc
50: 29 – 40, 1975
7. Herrera GA, Joseph L, Gu X, Hough A, Barlogie B: Renal pathologic
spectrum in an autopsy series of patients with plasma cell dyscrasia.
Arch Pathol Lab Med 128: 875–987, 2004
8. Ivanyi B: Frequency of light chain deposition nephropathy relative to
renal amyloidosis and Bence Jones cast nephropathy in a necropsy
study of patients with myeloma. Arch Pathol Lab Med 114: 986 –987,
1990
9. Kapadia SB: Multiple myeloma: A clinicopathologic study of 62
Figure 9. Higher magnification of the intracellular crystals consecutively autopsied cases. Medicine (Baltimore) 59: 380 –392,
that characterize LCFS reveals a regular periodicity. Many 1980
of the crystals are larger than the adjacent mitochondria. 10. Paueksakon P, Revelo MP, Horn RG, Shappell S, Fogo AS: Mono-
Magnification, ⫻80,000. clonal gammopathy: Significance and possible causality in renal
disease. Am J Kidney Dis 42: 87–95, 2003
11. Sethi S, Hanna MH, Fervenza FC: Unusual casts in a case of multiple
catabolizes light chains into amino acids before their myeloma. Am J Kidney Dis 54: 970 –974, 2009
release into the circulation (25,26). As a result of the 12. Koss MN, Pirani CL, Osserman EF: Experimental Bence Jones cast
nephropathy. Lab Invest 34: 579 –591, 1976
resistance to cathepsin B, a truncated NH2-terminal 13. Smolens P, Barnes JL, Stein JH: Effect of chronic administration of
fragment of the light chain accumulates and pro- different Bence Jones proteins on rat kidney. Kidney Int 30: 874 –
882, 1986
motes intracellular crystal formation. Of note, intra-
14. Solomon A, Weiss DT, Kattine AA: Nephrotoxic potential of Bence
peritoneal injection into rats of monoclonal light Jones proteins. New Engl J Med 324: 1845–1851, 1991
chains from the urine of a patient with LCFS has been 15. Sanders PW, Booker BB, Bishop JB, Cheung HC: Mechanisms of
intranephronal proteinaceous cast formation by low molecular weight
shown to reproduce the proximal tubular crystals of proteins. J Clin Invest 85: 570 –576, 1990
LCFS (27). More recently, a transgenic model of 16. Huang ZQ, Sanders PW: Biochemical interaction between Tamm-
LCFS has been created in which the mouse JK region Horsfall glycoprotein and Ig light chains in the pathogenesis of cast
nephropathy. Lab Invest 73: 810 – 817, 1995
was replaced by the VK-JK gene segment from a 17. Huang ZQ, Sanders PW: Localization of a single binding site for
patient with LCFS. Identical proximal tubular crystals immunoglobulin light chains on human Tamm-Horsfall glycoprotein.
as had been seen in the patient’s biopsy formed in the J Clin Invest 99: 732–736, 1997
18. Ying WZ, Sanders PW: Mapping the binding domain of immuno-
transgenic mouse proximal tubular cells. A conditional globulin light chains for Tamm-Horsfall protein. Am J Pathol 158:
deletion of the VK-JK segment abrogated crystal for- 1859 –1866, 2001
mation in this model (28). 19. Pote A, Zwizinski C, Simon EE, Meleg-Smith S, Batuman V:
Cytotoxicity of myeloma light chains in cultured human kidney
The majority of patients with LCFS have evi- proximal tubule cells. Am J Kidney Dis 36: 735–744, 2000
dence of dysproteinemia, and most fulfill criteria for 20. Khan AM, Li M, Balamuthusamy S, Maderdrut JL, Simon EE,
310 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Batuman V: Myeloma light chain-induced renal injury in mice. 25. Aucouturier P, Bauwens M, Khamlichi AA, Denoroy L, Spinelli S,
Nephron Exp Nephrol 116: e32– e41, 2010 Touchard G, Preud’homme JL, Cogne M: Monoclonal Ig L chain and
21. Li M, Hering-Smith KS, Simon EE, Batuman V: Myeloma light L chain V domain fragment crystallization in myeloma-associated
chains induce epithelial-mesenchymal transition in human renal Fanconi’s syndrome. J Immunol 150: 3561–3568, 1993
proximal tubule epithelial cells. Nephrol Dial Transplant 23: 860 – 26. Leboulleux M, Lelongt B, Mougenot B, Touchard G, Makdassi R,
870, 2008 Rocca A, Noel LH, Ronco PM, Aucouturier P: Protease resistance
22. Maldonado JE, Velosa JA, Kyle RA, Wagoner RD, Holley KE, and binding of Ig light chains in myeloma-associated tubulopathies.
Salassa RM: Fanconi syndrome in adults: A manifestation of a latent Kidney Int 48: 72–79, 1995
form of myeloma. Am J Med 58: 354 –364, 1975 27. Clyne DH, Brendstrup L, First MR, Pesce AJ, Finkel PN, Pollak VE,
23. Messiaen T, Deret S, Mougenot B, Bridoux F, Dequiedt P, Dion JJ,
Pirani CL: Renal effects of intraperitoneal kappa chain injection:
Makdassi R, Meeus F, Pourrat J, Touchard G, Vanhille P, Zaoui P,
Induction of crystals in renal tubular cells. Lab Invest 31: 131–142,
Aucouturier P, Ronco PM: Adult Fanconi syndrome secondary to
1974
light chain gammopathy: Clinicopathologic heterogeneity and un-
28. Sirac C, Bridoux F, Carrion C, Devuyst O, Fernandez B, Goujon JM,
usual features in 11 patients. Medicine (Baltimore) 79: 135–154,
2000 El Hamel C, Aldigier JC, Touchard G, Cogne M: Role of the
24. Nasr SH, Galgano SJ, Markowitz GS, Stokes MB, D’Agati VD: monoclonal kappa chain V domain and reversibility of renal damage
Immunofluorescence on pronase-digested paraffin sections: A valu- in a transgenic model of acquired Fanconi syndrome. Blood 108:
able salvage technique for renal biopsies. Kidney Int 70: 2148 –2151, 536 –543, 2006
2006
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Figure 1. Figure 2.
Figure 4.
Figure 3.
immune reactants in the lamellated basement mem- tions to form hexamers: ␣1.␣1.␣2(IV)–␣1.␣1.␣2(IV),
branes (5). Immunofluorescent antibodies against the ␣3.␣4.␣5(IV)–␣ 3. ␣ 4. ␣ 5(IV), and ␣ 1. ␣ 1. ␣ 2(IV)–
␣1, ␣3, and ␣5 chains of collagen IV are commercially ␣5.␣5.␣6(IV). The ␣1.␣1.␣2(IV)–␣1.␣1.␣2(IV) net-
available (Wieslab, Lund, Sweden) and may help to work is expressed in all renal basement membranes
confirm the diagnosis of HN and provide prognostic including the GBM, tubular basement membranes
information (see Etiology and Pathogenesis section). (TBMs), and Bowman’s capsule. No known mutations
Electron Microscopy. Electron microscopy is re- in the ␣2 chain have been described, whereas rare
quired to establish a pathologic diagnosis of HN. patients with COL4A1 mutations and HANAC syn-
Classically, GBMs show prominent thickening and drome (Hereditary Angiopathy, Nephropathy, Aneu-
lamellation resulting in a “basket-weave” appearance, rysms, and muscle Cramps) have been reported (7).
and the podocytes overlying the abnormal basement The ␣3.␣4.␣5(IV)–␣3.␣4.␣5(IV) network is the main
membranes display variable foot process effacement. form of type IV collagen in the mature GBM and is
The lamellations form in the plane of the lamina densa also present in distal TBMs. The ␣1.␣1.␣2(IV)–
and typically enclose electron lucent zones. Minute ␣5.␣5.␣6(IV) hexamer is restricted to Bowman’s cap-
electron-dense granulations may collect between some sule and collecting duct TBMs.
of the lamellations. Early in the disease course in All known mutations that cause HN are found in
young males or at any point in adult females, the major COL4A3, COL4A4, or COL4A5. The majority of
ultrastructural finding may be GBM thinning (typi- cases of HN are X-linked and have mutations in
cally ⬍225 nm). Careful search under the electron COL4A5. In contrast, autosomal inheritance of HN is
microscope may reveal only rare foci of subtle textural associated with mutations in the ␣3 and/or ␣4 chain.
changes, such as early lamination, reticulation, and The ␣3.␣4.␣5(IV)–␣3.␣4.␣5(IV) network is the
scalloping, making differentiation from thin basement major collagen IV network in the mature GBM. In
membrane nephropathy difficult (if not impossible) on glomerular development, ␣1.␣1.␣2(IV)–␣1.␣1.␣2(IV)
morphologic grounds. In the setting of a strong family predominates in the immature GBM and must undergo
history of HN and ultrastructural findings of GBM a developmental switch with replacement by
thinning without well-developed lamellations, we of- ␣3.␣4.␣5(IV)–␣3.␣4.␣5(IV) as the GBM matures. In
ten use the term “hereditary nephritis with thin base- patients with HN, there is a failure of this normal
ment membrane phenotype” to indicate that it is not maturation process, leading to persistence of the im-
possible to differentiate with certainty between thin mature ␣1.␣1.␣2(IV)–␣1.␣1.␣2(IV) network in the
basement membrane nephropathy and early HN. GBM. As a result, the GBM of patients with HN
contains predominantly ␣1.␣1.␣2(IV)–␣1.␣1.␣2(IV)
Etiology and Pathogenesis rather than ␣3.␣4.␣5(IV)–␣3.␣4.␣5(IV). This is
Recent advances in our understanding of the clearly an oversimplification because intact or dis-
structure of type IV collagen coupled with the avail- rupted staining for the ␣3 and ␣5 chains of type IV
ability of genetic testing have led to dramatic progress collagen can be demonstrated by immunofluorescence
in unraveling the pathogenesis of HN, as reviewed by in a significant percentage of adult patients with HN.
Hudson et al. (6). The type IV collagen gene family It is likely that the size, type, and location of the
consists of six distinct ␣ chains (␣1 through ␣6), mutations within the COL4A3, COL4A4, and
each encoded by a separate gene. Genes for COL4A5 genes determine the relative contribution of
COL4A1 and COL4A2 are on chromosome 13, the ␣3.␣4.␣5(IV)–␣3.␣4.␣5(IV) and ␣1.␣1.␣2(IV)–
COL4A3 and COL4A4 are on chromosome 2, and ␣1.␣1.␣2(IV) to the GBM in individual patients
COL4A5 and COL4A6 are on the X chromosome. with HN.
These six ␣ chains assemble into triple helical protom- Our improved understanding of collagen IV dis-
ers that in turn form hexamers that are found in tribution in the kidney is useful in the renal biopsy
basement membranes. Despite the large number of diagnosis of HN (8). Antibodies directed against the
hypothetical combinations, only three triple helical ␣1, ␣3, and ␣5 chains of collagen IV have been
protomers are known to occur: ␣ 1. ␣ 1. ␣ 2(IV), commercially available for more than a decade (Wi-
␣3.␣4.␣5(IV), and ␣5.␣5.␣6(IV). Furthermore, these eslab). A normal staining pattern shows ␣1 in all
protomers are known to pair up in only three combina- basement membranes of the kidney; ␣3 in the GBM
314 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
and distal TBMs; and ␣5 in the GBM, distal and (Figure 5). In contrast, there was complete absence of
collecting tubular TBMs, and Bowman’s capsule. In ␣3 and ␣5 from glomerular and distal TBMs and
males with X-linked HN, ␣5 staining may be reduced complete absence of ␣5 from Bowman’s capsule
or lost in the GBM, distal TBMs, and Bowman’s (Figure 6). Female carriers of X-linked mutations
capsule. Staining for ␣3 is similarly altered because in typically show discontinuous staining (“skip areas”)
the absence of intact ␣5, the ␣3.␣4.␣5 protomer does for ␣3 and ␣5 in GBMs rather than complete loss. This
not assemble normally and fails to become incorpo- can be explained by random inactivation of the mu-
rated into basement membranes (9). In homozygous tated X chromosome in individual podocytes during
mutations of autosomally encoded COL4A3 or development, causing females to be mosaics.
COL4A4, decreased staining for ␣3 and ␣5 in the Immunofluorescence studies on skin biopsy speci-
GBM and TBMs is seen, but ␣5 staining within mens may also provide useful information in X-linked HN
Bowman’s capsule is preserved because the ␣5.␣5.␣6 because the ␣1.␣1.␣2(IV)–␣5.␣5.␣6(IV) network is nor-
protomer is not affected (Table 1) (10). mally expressed in the epidermal basement membrane.
Although immunostaining for the ␣1, ␣3, and ␣5 Complete loss of ␣5 may be seen in males with
chains of collagen IV is useful, this approach has COL4A5 mutations. Because females are a mosaic for
limitations. A significant minority of patients with HN the mutation, the loss of immunoreactivity for
exhibit normal staining for the subtypes of collagen COL4A5 is typically segmental or discontinuous (Ta-
IV; although the precise percentage is unclear, it has ble 1). Again, whereas the loss of staining in the
been suggested to be in the range of 20% (11) and in epidermal basement membrane seems to be specific
our experience is closer to 50%. The presence or for the diagnosis of X-linked HN, a normal staining
absence of staining for ␣3 and ␣5 likely depends on pattern is seen in approximately 30% of patients as
the severity of the mutation. Patients with large dele- well as in all patients with autosomal forms of HN
tions or frame shift or truncating mutations are likely
to have loss of staining. By contrast, patients with
smaller mutations (e.g., missense mutations leading to
single amino acid substitutions) may exhibit an essen-
tially normal staining pattern, making it impossible to
exclude the diagnosis of HN on the basis of immuno-
fluorescence alone. We previously stained for ␣3 and
␣5 type IV collagen in 16 cases of biopsy-proven HN
and found that the mean age at the time of biopsy was
16.3 years in patients with loss of staining (and there-
fore likely larger mutations), as compared with 36.6
years in patients with intact staining (and likely
smaller mutations; P ⫽ 0.03) (12).
Staining for the subtypes of type IV collagen was
performed in the case of the 20-year-old man pre- Figure 5. Immunofluorescence staining for ␣1 chain of type
sented herein. It revealed positivity for the ␣1 subunit IV collagen is positive in all renal basement membranes.
in all renal basement membranes, including the GBM Magnification, ⫻400.
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 315
References
1. Alport AC: Hereditary familial congenital haemorrhagic nephritis.
Figure 6. In contrast, staining for the ␣3 (A) and ␣5 (B) Br Med J 1: 504 –506, 1927
chains of type IV collagen is lost in glomerular and distal 2. Levy M, Feingold J: Estimating prevalence in single-gene kidney
TBMs. Magnification, ⫻400. diseases progressing to renal failure. Kidney Int 58: 925–943, 2000
3. Kashtan CE: Familial hematurias: What we know and what we
don’t. Pediatr Nephrol 20: 1027–1035, 2005
(13). Patients with autosomal forms retain staining for 4. Bekheirnia MR, Reed B, Gregory MC, McFann K, Shamshirsaz
COL4A5 because neither COL4A3 nor COL4A4 is AA, Masoumi A, Schrier RW: Genotype-phenotype correlation in
expressed in normal epidermal basement membrane. X-linked Alport syndrome. J Am Soc Nephrol 21: 876 – 883, 2010
5. Nasr SH, Markowitz GS, Goldstein CS, Fildes RD, D’Agati VD:
Clinical-Pathologic Correlations. Genetic testing Hereditary nephritis mimicking immune complex-mediated glomer-
ulonephritis. Hum Pathol 37: 547–554, 2006
has only recently become commercially available for
6. Hudson BG, Tryggvason K, Sundaramoorthy M, Neilson EG: Al-
the diagnosis of HN and is being used increasingly in port’s syndrome, Goodpasture’s syndrome and type IV collagen.
routine practice. Establishing genotype–phenotype N Engl J Med 348: 2543–2556, 2003
7. Plaisier E, Gribouval O, Alamowitch S, Mougenot B, Prost C,
correlations would significantly increase the prognos- Verpont MC, Marro B, Desmettre T, Cohen SY, Roullet E, Dracon
tic value of such testing. Bekheirnia et al. (4) exam- M, Fardeau M, Van Agtmael T, Kerjaschki D, Antignac C, Ronco
ined males with X-linked HN and showed a relation- P: COL4A1 mutations and hereditary angiopathy, nephropathy,
aneurysms, and muscle cramps. N Engl J Med 357: 2687–2695,
ship among mutation type, location within the gene, 2007
and severity of disease. Average age of onset of ESRD 8. Haas M: Alport syndrome and thin glomerular basement membrane
was 37 years in patients with missense mutations, 28 nephropathy: A practical approach to diagnosis. Arch Pathol Lab
Med 133: 224 –232, 2009
years for patients with splice-site mutations, and 25 9. Naito I, Kawai S, Nomura S, Sado Y, Osawa G: Relationship
years for those with truncating mutations (P ⬍ between COL4A5 gene mutation and distribution of type IV colla-
0.0001). The position of the mutation in the gene was gen in male X-linked Alport syndrome. Kidney Int 50: 304 –311,
1996
also prognostic with mutations at the 5⬘ end of 10. Gubler MC, Knebelmann B, Beziau A, Broyer M, Pirson Y, Had-
COL4A5 being associated with earlier onset of ESRD doum F, Kleppel MM, Antignac C: Autosomal recessive Alport
316 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
syndrome: Immunohistochemical study of type IV collagen chain roder C, Sanak M, Krejcova S, Carvalho MF, Saus J, Antignac C,
distribution. Kidney Int 47: 1142–1147, 1995 Smeets H, Gubler MC: X-linked Alport syndrome: Natural history in
11. Kashtan CE, Segal Y: Genetic disorders of glomerular basement 195 families and genotype-phenotype correlations in males. J Am Soc
membranes. Nephron Clin Pract 118: c9 – c18, 2011 Nephrol 11: 649 – 657, 2000
12. Markowitz GS, Gelber C, D’Agati VD: An 18-year-old male with 15. Jais JP, Knebelmann B, Giatras I, de Marchi M, Rizzoni G,
hematuria, renal insufficiency, and defective synthesis of type IV Renieri A, Weber M, Gross O, Netzer KO, Flinter F, Pirson Y,
collagen. Kidney Int 69: 2278 –2282, 2006 Dahan K, Wieslander J, Persson U, Tryggvason K, Martin P, Hertz
13. Patey-Mariaud de Serre N, Garfa M, Messieres B, Noel LH, Knebel- JM, Schroder C, Sanak M, Fernanda M, Carvalho MF, Saus J,
mann B: Collagen ␣5 and ␣2(IV) chain coexpression: Analysis of Antignac C, Smeets H, Gubler MC: X-linked Alport syndrome:
skin biopsies of Alport patients. Kidney Int 72: 512–516, 2007 Natural history and genotype–phenotype correlations in girls and
14. Jais JP, Knebelmann B, Giatras I, de Marchi M, Rizzoni G, Renieri women belonging to 195 families: A “European Community
A, Weber M, Gross O, Netzer KO, Flinter F, Pirson Y, Verellen C, Alport Syndrome Concerted Action” study. J Am Soc Nephrol 14:
Wieslander J, Persson U, Tryggvason K, Martin P, Hertz JM, Sch- 2603–2610, 2003
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Section 8: CASE PRESENTATION the abdomen, was normal. Recent urinalysis revealed no
proteinuria and 3⫹ heme; urinary microscopy showed 10 to
A 36-year-old Caucasian man without previous medical 20 red blood cells (RBCs) per high-power field, without
history was found to have microscopic hematuria with RBC casts. The patient has a stable serum creatinine level of
normal renal function during hospitalization for a motor 0.8 mg/dl. There is no history of hypertension or diabetes or
vehicle accident 4 years earlier. Since that time, he has had family history of renal disease. Serologic studies including
persistent microhematuria documented on routine urinaly- anti-nuclear antibody, anti-neutrophil cytoplasmic antibody,
ses, without episodes of gross hematuria or flank pain. A and antistreptolysin-O are negative, and serum complement
recent lower urinary tract workup for hematuria, including levels C3 and C4 are normal. A renal biopsy is performed for
urine culture, cystoscopy and computed tomography scan of investigation of persistent asymptomatic microhematuria.
Figure 1. Figure 2.
Figure 3. Figure 4.
generally diffuse and global GBM thinning in the A diagnosis of TBMN requires accurate tech-
absence of other ultrastructural abnormalities. The niques for GBM measurement. The GBM is a trilam-
normal range of GBM thickness varies among indi- inar structure composed of a central lamina densa, an
viduals and can be influenced by the patient’s age and inner lamina rara interna, and an outer lamina rara
gender and by the particular fixation methods and externa. To include the laminae rarae, the GBM thick-
embedding compounds used for electron microscopy ness is measured from the outer edge of the endothelial
(7,8). Thus, it is important for each renal pathology cell plasma membrane to the outer edge of the cell
laboratory to establish its own thresholds for “thin” membrane of the podocyte foot processes. The GBMs
GBM on the basis of a value for GBM thickness that are measured only in the peripheral capillaries, avoid-
is ⬎2 standard deviations below the normal mean ing areas of GBM folds and the reflection over the
(6,7). At Columbia University, the mean normal GBM mesangium. One technique of GBM thickness mea-
thickness is 350 ⫾ 50 nm for male adults and 300 ⫾ surement includes the orthogonal intercept/mean har-
40 nm for female adults (6,8). GBM thickness pro- monic thickness method (11). The electron micros-
gressively increases from birth to 9 to 11 years and copy prints are overlaid with a grid of perpendicularly
then plateaus. Detailed analyses of pediatric biopsies oriented lines of known dimension, and the GBM
have established the estimated normal ranges of GBM width is recorded in “classes.” The intersections be-
thickness in children (9,10). tween the grid lines and the endothelial cell plasma
According to World Health Organization guide- membranes are analyzed, and a mean harmonic thick-
lines for diagnosis of TBMN, the GBM thinning ness is calculated using a set of equations, all of which
should be diffuse with mean GBM thickness ⬍250 nm can be time-consuming. Alternately, a simple mea-
in adults and ⬍180 nm in children aged 2 to 11 years surement of GBM thickness can be made using a
magnification graticule or metric ruler applied to an
(6). However, several investigators prefer a thresh-
electron microscopic print after correction for the
old of 225 nm in adults, especially women. It should
magnification of the photographic negative and the
also be emphasized that thinning should affect
enlargement factor from negative to print (6). In this
⬎50% of the total glomerular capillary area. Alter-
era of digital cameras, GBM measurements are most
nating zones of thinning and thickening of the GBM
easily and rapidly performed using the appropriate
with lamellation of the lamina densa should prompt
software. It is extremely important that multiple rep-
a diagnosis of hereditary nephritis/Alport syndrome.
resentative GBM measurements be taken over multi-
ple capillaries of several glomeruli to calculate an
accurate mean value.
Because a diagnosis of TBMN requires electron
microscopic examination, inadequate biopsies that
lack glomeruli in the tissue allocated for electron
microscopy pose a diagnostic challenge. Salvage elec-
tron microscopy on deparaffinized, formalin-fixed tis-
sue, which is usually performed in this situation, is
helpful to diagnose many glomerular diseases but is
distinctly unreliable as a means of diagnosing TBMN.
This is because the reprocessing technique itself
causes tissue dehydration and artifactual thinning of
Figure 6. In a case of minimal change disease superimposed the GBM. Although it may be possible to exclude
on TBMN, there is diffuse effacement of foot processes TBMN, it is not possible to diagnose TBMN accu-
with condensation of the actin cytoskeleton. The underlying
GBM is uniformly thinned (180 to 200 nm). The patient, an rately in deparaffinized, reprocessed tissue (8).
82-year old woman with longstanding history of microhe- Immunofluorescence for Collagen IV Subtypes. Re-
maturia, presented with abrupt onset of nephrotic syndrome
(lower extremity edema, proteinuria of 8.0 g/day, and serum
nal biopsy staining with commercially available anti-
albumin of 2.1 g/dl), which responded to steroid therapy. sera to the ␣1, ␣3, and ␣5 subunits of collagen IV is
Magnification, ⫻10,000, electron micrograph. helpful to support a diagnosis of TBMN and rule out
320 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Alport syndrome. TBMN retains a normal pattern of may also cause TBMN in some kindreds, none have
immunoreactivity for all subunits (␣1 through ␣6) of yet been identified (17).
type IV collagen. The normal staining pattern for ␣3 is Recent genotypic studies suggest the existence of
diffuse linear positivity along the GBMs and distal a rare severe form of TBMN that does not represent
TBMs. The normal ␣5 staining pattern is similar to ␣3, misdiagnosed Alport syndrome. These patients have
with the exception that Bowman’s capsule, and col- diffuse thinning of the GBM with lesions of FSGS and
lecting duct TBMs are also positive. Because the clinically manifest with greater proteinuria, renal im-
GBMs are thin, the GBM staining for ␣3 and ␣5 pairment, and progression to end-stage kidney disease.
subunits in TBMN may appear weaker and more Molecular analyses have identified founder mutations
delicate than that of normal controls. Importantly, in in COL4A3 and COL4A4 genes in these families
TBMN no segmental or complete loss of staining for (4,18). It has been hypothesized that particular poly-
the ␣3 and ␣5 subunits typical of Alport syndrome is morphisms or unidentified modifier genes may under-
observed (6,7). It is important to recognize that stain- lie this more severe phenotype (19). For example,
ing for the ␣ subunits of collagen IV is not an absolute some investigators have identified mutations in
means to distinguish between TBMN and Alport syn- NPHS2 (encoding podocin) in unrelated patients with
drome because at least 20% (and in our experience TBMN and greater proteinuria (17,20), whereas others
closer to 50%) of patients with Alport syndrome retain have been unable to confirm NPHS2 mutations in
normal staining patterns (12,13) For more difficult pedigrees with TBMN and FSGS (21).
cases, genetic testing for mutations in COL4A3, Association with other Glomerular Diseases. GBM
COL4A4, and COL4A5 is commercially available. thinning may occur as a localized process in many
glomerular diseases, particularly when there is glo-
Pathogenesis merular capillary wall distension, rupture, and/or re-
Although TBMN is an inherited disorder with modeling. Unlike TBMN, these conditions exhibit
autosomal dominant transmission, a positive family short segments of thinning on a background of other,
history of hematuria can be elicited from only two obvious glomerular pathology. In addition, TBMN
thirds of patients. Seemingly sporadic cases may be with diffuse GBM thinning may coexist with another
due to undetected (subclinical) disease in other family glomerular disease as a dual glomerulopathy (5,6).
members, nonpenetrance of hematuria in affected fam- Approximately 30% of patients with IgAN have thin
ily members, or de novo mutations. Considerable ge- basement membranes, a frequency higher than in the
netic overlap exists between TBMN and Alport syn- general population, suggesting a possible pathogenetic
drome, and identification of type IV collagen gene link or selection bias for biopsy. In support of this
mutations in Alport syndrome has helped elucidate the hypothesis, patients with IgAN and TBMN often have
familial hematuria and do not manifest the usual ab-
pathogenesis of TBMN (12,14,15). It is now estab-
normalities of IgA glycosylation (5). Other reported
lished that TBMN is caused by heterozygous muta-
associations with TBMN are likely to occur by chance,
tions in the COL4A3 or COL4A4 gene located on
such as minimal change disease (Figure 6) and mem-
chromosome 2 (12,16). Mutations include missense or
branous nephropathy (6). In these cases, TBMN is
splice-site deletions or frame shifts. TBMN likely
usually an incidental finding and does not influence
represents a carrier state for autosomal recessive Al-
the overall prognosis of the superimposed glomerular
port syndrome, a disorder arising from similar ho-
disease.
mozygous or compound heterozygous COL4A3/
COL4A4 mutations (17). Too few mutations have
References
been described for autosomal dominant Alport syn- 1. Wang YY, Savige J: The epidemiology of thin basement membrane
drome to make genotype distinctions from TBMN. nephropathy. Semin Nephrol 25: 136 –139, 2005
2. Gregory MC: The clinical features of thin basement membrane
The mutation detection rate varies on the basis of the nephropathy. Semin Nephrol 25: 140 –145, 2005
technique used but is complicated by the large size of 3. Packham DK, Perkovic V, Savige J, Broome MR: Hematuria in thin
COL4A3/COL4A4 genetic loci and the existence of basement membrane nephropathy. Semin Nephrol 25: 146 –148, 2005
4. Voskarides K, Damianou L, Neocleous V, Zouvani I, Christodouli-
polymorphisms. Although it has been speculated that dou S, Hadjiconstantinou V, Ioannou K, Athanasiou Y, Patsias C,
mutations in other genes encoding GBM components Alexopoulos E, Pierides A, Kyriacou K, Deltas C: COL4A3/
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 321
COL4A4 mutations producing focal segmental glomerulosclerosis 14. Kashtan CE: Alport syndrome and thin glomerular basement mem-
and renal failure in thin basement membrane nephropathy. J Am Soc brane disease. J Am Soc Nephrol 9: 1736 –1750, 1998
Nephrol 18: 3004 –3016, 2007 15. Kashtan CE: Familial hematuria due to type IV collagen mutations:
5. Norby SM, Cosio FG: Thin basement membrane nephropathy asso- Alport syndrome and thin basement membrane nephropathy. Curr
ciated with other glomerular diseases. Semin Nephrol 25: 176 –179, Opin Pediatr 16: 177–181, 2004
2005 16. Lemmink HH, Nillesen WN, Mochizuki T, Schroder CH, Brunner
6. Foster K, Markowitz GS, D’Agati VD: Pathology of thin basement HG, van Oost BA, Monnens LA, Smeets HJ: Benign familial hema-
membrane nephropathy. Semin Nephrol 25: 149 –158, 2005 turia due to mutation of the type IV collagen alpha4 gene. J Clin
7. Haas M: Alport syndrome and thin glomerular basement membrane Invest 98: 1114 –1118, 1996
nephropathy: A practical approach to diagnosis. Arch Pathol Lab 17. Rana K, Wang YY, Buzza M, Tonna S, Zhang KW, Lin T, Sin L,
Med 133: 224 –232, 2009 Padavarat S, Savige J: The genetics of thin basement membrane
nephropathy. Semin Nephrol 25: 163–170, 2005
8. Nasr SH, Markowitz GS, Valeri AM, Yu Z, Chen L, D’Agati VD:
18. Pierides A, Voskarides K, Athanasiou Y, Ioannou K, Damianou L,
Thin basement membrane nephropathy cannot be diagnosed reliably
Arsali M, Zavros M, Pierides M, Vargemezis V, Patsias C, Zouvani
in deparaffinized, formalin-fixed tissue. Nephrol Dial Transplant 22:
I, Elia A, Kyriacou K, Deltas C: Clinico-pathological correlations in
1228 –1232, 2007
127 patients in 11 large pedigrees, segregating one of three heterozy-
9. Morita M, White RH, Raafat F, Barnes JM, Standring DM: Glomer-
gous mutations in the COL4A3/ COL4A4 genes associated with
ular basement membrane thickness in children: A morphometric
familial haematuria and significant late progression to proteinuria and
study. Pediatr Nephrol 2: 190 –195, 1988 chronic kidney disease from focal segmental glomerulosclerosis.
10. Vogler C, McAdams AJ, Homan SM: Glomerular basement mem- Nephrol Dial Transplant 24: 2721–2729, 2009
brane and lamina densa in infants and children: An ultrastructural 19. Kashtan CE: The wages of thin. J Am Soc Nephrol 18: 2800 –2802,
evaluation. Pediatr Pathol 7: 527–534, 1987 2007
11. Dische FE: Measurement of glomerular basement membrane thick- 20. Tonna S, Wang YY, Wilson D, Rigby L, Tabone T, Cotton R, Savige
ness and its application to the diagnosis of thin-membrane nephrop- J: The R229Q mutation in NPHS2 may predispose to proteinuria in
athy. Arch Pathol Lab Med 116: 43– 49, 1992 thin-basement-membrane nephropathy. Pediatr Nephrol 23: 2201–
12. Tryggvason K, Patrakka J: Thin basement membrane nephropathy. 2207, 2008
J Am Soc Nephrol 17: 813– 822, 2006 21. Deltas C: Thin basement membrane nephropathy: Is there genetic
13. Kashtan CE, Segal Y: Genetic disorders of glomerular basement predisposition to more severe disease? Pediatr Nephrol 24: 877– 879,
membranes. Nephron Clin Pract 118: c9 – c18, 2011 2009
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Section 9: CASE PRESENTATION and 4⫹ protein, and urine sediment is active with too
numerous to count erythrocytes and occasional leukocytes.
A 10-year-old Caucasian girl presents with edema. Other Urine protein excretion is 6.5 g/day, and serum albumin is
than occasional upper respiratory tract infections, the pa- 1.7 g/dl. Serologic evaluation shows negative anti-nuclear
tient has no significant past medical history. There is no antibody, anti-DNA, hepatitis B surface antigen, hepatitis C
family history of renal disease. On examination, she is antibody, anti-neutrophil cytoplasmic antibody, and anti–
found to have new-onset hypertension (BP 137/76 mmHg; glomerular basement membrane (anti-GBM) antibodies.
⬎99th percentile for systolic BP), height of 146 cm, weight She has an elevated antistreptolysin-O titer, and both C3 and
of 45.7 kg, and 2⫹ lower extremity edema. She denies any C4 are depressed. The kidneys measure 11.6 and 12.1 cm by
episodes of gross hematuria. Urinalysis reveals 3⫹ blood ultrasound.
Figure 1. Figure 2.
Figure 5. Figure 6.
323
324 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
What is the BEST diagnosis? in children, whereas adults frequently have an identi-
A. Acute postinfectious glomerulonephritis (APIGN) fiable underlying condition. MPGN also is classically
B. Membranoproliferative glomerulonephritis (MPGN) divided into three types on the basis of the combined
type 1 findings of light microscopy, immunofluorescence,
C. Dense deposit disease and electron microscopy. MPGN type 1 is the focus of
D. MPGN type 3, Strife and Anders subtype this section. MPGN type 2 is preferentially called
E. MPGN type 3, Burkholder subtype dense deposit disease because of its distinctive patho-
genesis and frequent lack of membranoproliferative
Answer features. MPGN type 3 is divided into the Burkholder
The renal biopsy findings show a membranopro- subtype and the Strife and Anders subtype and is
liferative glomerulonephritis (MPGN) type 1 pattern discussed briefly in the Differential Diagnosis section.
of injury (answer B). Given the young age of the Many of these morphologic variants of MPGN were
patient, the absence of systemic disease, the low serum described in the 1970s, before there was an under-
C3 and C4, and the otherwise negative serologic standing of pathogenesis, associations with hepatitis C
workup, this most likely represents idiopathic MPGN infection, and inherited or acquired disorders of alter-
type 1. Figure 1 (hematoxylin and eosin, ⫻400) shows native pathway complement activation. Concepts
an enlarged hypercellular glomerulus with accentuated about the validity of these historical subtypes are
lobularity. Figure 2 (Jones methenamine silver, ⫻600) evolving as new pathogenetic insights emerge.
highlights the presence of mesangial and subendothe-
lial immune deposits, which have a glassy pink ap- Clinical Characteristics
pearance. Glomerular capillaries are narrowed by mes- MPGN type 1 is reported to account for approx-
angial expansion and proliferation and contain imately 6% of nephrotic syndrome in children accord-
scattered intraluminal leukocytes. GBMs show seg- ing to the International Study of Kidney Disease in
mental duplications with mesangial interposition. Fig- Children (ISKDC) (1). Among adults, the relative
ures 3 (immunofluorescence for C3) and 4 (immuno- frequency of MPGN varies significantly depending on
fluorescence for IgG) show the immunofluorescence geographic location. In the United States and many
results including diffuse granular mesangial and seg- other industrialized nations, the relative frequency of
mental to global semilinear peripheral capillary wall MPGN type 1 is declining, accounting for ⬍5% of
staining for C3 (3⫹ intensity) and IgG (1⫹). Electron cases of nephrotic syndrome in adults, whereas in
microscopy (Figures 5 and 6) reveals numerous elec- parts of Africa and South America, MPGN is the most
tron-dense deposits in the mesangium, accompanied commonly encountered primary glomerular disease
by mesangial cell proliferation that severely narrows (2). Clinical symptoms may be predominantly ne-
the glomerular capillary lumina. Prominent subendo- phrotic, nephritic, or both. Approximately half of pa-
thelial deposits are present. At higher magnification, a tients report a respiratory tract infection before onset
large subendothelial deposit is seen, and a thin layer of of symptoms. Proteinuria is almost universally present
neomembrane has formed between the endothelium with more than half of patients presenting with ne-
and the deposit to produce a GBM double contour phrotic syndrome. Persistent microscopic hematuria is
(Figure 6). Occasional small subepithelial deposits are found in most patients. Approximately 10 to 20% of
also present. patients with MPGN type 1 present with an acute
nephritic syndrome, and some of these develop recur-
Membranoproliferative Glomerulonephritis rent episodes of gross hematuria. Although not all
MPGN is a morphologic pattern of glomerular patients have depressed serum C3 (and in some cases
injury characterized by cellular proliferation and thick- depressed C4) levels at presentation, the majority will
ening of glomerular capillary walls as a result of develop hypocomplementemia at some point in their
immune complex deposition, peripheral mesangial cell course. Renal insufficiency is seen in approximately
interposition, and duplication of GBMs. MPGN is 25% of patients at the time of diagnosis but is more
divided into primary (idiopathic) and secondary forms common in adults than in children. Hypertension is
of disease, which most often relate to infection or seen in approximately one-third of patients at onset
systemic conditions. Primary MPGN is more common and is usually mild (3).
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 325
schistosomiasis, toxoplasmosis, malaria), and other with segmental GBM duplication. Immunofluores-
unusual organisms (e.g., mycoplasma, borreliosis, cence staining is typically dominant for C3 and often
leishmaniasis) (6). Cryoglobulinemic glomerulone- accompanied by IgG in both MPGN and APIGN. By
phritis of any cause classically displays membranopro- electron microscopy, type 1 MPGN should show more
liferative histology. Particularly in the adult popula- extensive subendothelial deposits and extensive redu-
tion, careful serologic workup and clinical correlation plication of GBMs with mesangial interposition,
are needed to exclude secondary causes of MPGN type whereas APIGN should display evidence of subepi-
1 before a diagnosis of idiopathic (primary) MPGN is thelial “hump” formation. Notably, occasional subep-
made. ithelial deposits may also be seen in type 1 MPGN.
Once MPGN is diagnosed, proper classification
Differential Diagnosis of the MPGN pattern is needed. MPGN type 1 is
When a membranoproliferative pattern of glo- typically dominated by subendothelial and mesangial
merular injury is identified by light microscopy, there deposits with GBM duplication and mesangial inter-
are many diagnostic considerations other than MPGN. position. MPGN type 3 has two subtypes, one de-
A subacute or chronic thrombotic microangiopathy scribed by Strife et al. (8) and Anders et al. (9) and
often displays a membranoproliferative appearance. It another defined by Burkholder et al. (10). The Burk-
can be distinguished by immunofluorescence positiv- holder subtype resembles MPGN type 1 but shows
ity for fibrin (often with nonspecific positivity for more pronounced subepithelial deposits, akin to a
IgM) and by the ultrastructural findings of electron- membranous pattern superimposed on a type 1 MPGN
lucent, flocculent material between the duplicated lay- (Figure 7). Like MPGN type 1, the Burkholder sub-
ers of GBM. Multiple immune complex–mediated type typically has deposits of Ig and complement, is
diseases may have a membranoproliferative appear- often associated with chronic infection, and likely has
ance by light microscopy including IgA nephropathy, a similar pathogenesis. In some cases, there is a
lupus nephritis, and fibrillary glomerulonephritis, al- continuum of findings between type 1 and type 3 in
though these entities can be distinguished from MPGN different glomeruli, supporting their similar pathogen-
type 1 on the basis of clinical history, serologies, the esis. The Strife and Anders subtype features more
composition of the deposits by immunofluorescence, complex intramembranous deposits that seem to ex-
and the ultrastructural appearance of the deposits. tend from the subendothelial to subepithelial aspects
Importantly, many forms of dysproteinemia-associ- of the GBM, causing irregular GBM thickening with
ated glomerular disease exhibit a membranoprolifera- disruption and fraying of the lamina densa (Figure 8).
tive appearance, including type 1 cryoglobulinemic It often has dominant or exclusive staining for C3 and
glomerulonephritis, immunotactoid glomerulopathy, may exhibit areas by electron microscopy that overlap
monoclonal Ig deposition disease, and “proliferative with features of dense deposit disease. Testing for
glomerulonephritis with monoclonal Ig deposits” (7). complement regulatory factors (factor H, factor I, and
In each of these conditions, the monoclonal nature of
the paraprotein deposits is often established by immu-
nofluorescence, which typically shows restricted pos-
itivity for a single light chain isotype and/or a single
heavy chain class. Furthermore, the deposits in type 1
cryoglobulinemic glomerulonephritis often have a dis-
tinctive substructure, whereas immunotactoid glo-
merulopathy is characterized by hollow microtubular
deposits with a diameter of 30 to 50 nm.
It may be particularly difficult to distinguish
MPGN from APIGN because both can present with a
mixed nephritic/nephrotic syndrome, history of pre-
Figure 7. MPGN type 3 of Burkholder. Note the presence of
ceding infection, and hypocomplementemia. On bi- prominent subendothelial and subepithelial deposits along
opsy, histologic features may overlap significantly, with segmental duplication of the glomerular basement
including mesangial and endocapillary proliferation membrane. Magnification, ⫻6000.
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 327
6. Rennke HG: Secondary membranoproliferative glomerulonephritis. and proliferative glomerulonephritis: A correlative light, immu-
Kidney Int 47: 643– 656, 1995 nofluorescence and electron microscopic study. Lab Invest 23:
7. Nasr SH, Satoskar A, Markowitz GS, Valeri AM, Appel GB, 459 – 479, 1970
Stokes MB, Nadasdy T, D’Agati VD: Proliferative glomerulone- 11. Gallo GR, Caulin-Glaser T, Lamm ME: Charge of circulating im-
phritis with monoclonal IgG deposits. J Am Soc Nephrol 20: mune complexes as a factor in glomerular basement membrane
1986 –1996, 2009 localization in mice. J Clin Invest 67: 305–313, 1981
8. Strife CF, McEnery PT, McAdams AJ, West CD: Membranoprolif- 12. Servais A, Fremeaux-Bacchi V, Lequintrec M, Salomon R, Blouin J,
erative glomerulonephritis with disruption of the glomerular base- Knebelmann B, Grunfeld JP, Lasavre P, Noel LH, Fakhouri F:
ment membrane. Clin Nephrol 7: 65–72, 1977 Primary glomerulonephritis with isolated C3 deposits: A new entity
9. Anders D, Agricola B, Sippel M, Thoenes W: Basement mem- which shares common genetic risk factors with heaemolyticuraemic
brane changes in membranoproliferative glomerulonephritis: syndrome. J Med Genet 44: 193–199, 2007
Characterization of a third type by silver impregnation of ultra 13. Boyer O, Noel LH, Balzamo E, Guest G, Biebuyck N, Charbit M,
thin sections. Virchows Arch A Patholol Anat Histol 376: 1–19, Salomon R, Fremeaux-Bacchi V, Niaudet P: Complement factor H
1977 deficiency and posttransplantation glomerulonephritis with isolated
10. Burkholder PM, Marchand A, Krueger RP: Mixed membranous C3 deposits. Am J Kidney Dis 51: 671– 677, 2008
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Section 10: CASE PRESENTATION edema. Laboratory workup shows a serum creatinine level of
0.5 mg/dl, 24-hour urine protein of 6.2 g, albumin of 1.5 g/dl,
A 12-year-old Caucasian boy presents with a 1-month his- and urinalysis with 4⫹ albumin, too numerous to count red
tory of periorbital edema and several episodes of gross hema- blood cells, and scattered red blood cell casts. Serologic
turia over the last 3 weeks. The patient’s past medical history workup shows negative anti-nuclear antibody and anti-DNA
is remarkable for an episode of streptococcal pharyngitis 2 antibody, negative hepatitis B surface antigen and hepatitis C
months earlier. There is no family history of renal disease. antibody, elevated antistreptolysin-O titer, a moderately de-
Physical examination reveals a BP of 115/70 mmHg and creased C3 complement level, and a normal C4.
Figure 1. Figure 2.
Figure 3. Figure 4. C3
Figure 5. Figure 6.
329
330 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
glomerulonephritis with isolated C3 deposits is en- opment of DDD, yet other patients with DDD have
countered (5). reduced function of these factors as a result of allelic
Monoclonal Ig deposition disease (MIDD) must variants or the formation of autoantibodies (9). Mice
also be considered in the differential diagnosis of adult deficient in factor H have uncontrolled C3 activation and
DDD. Both MIDD and DDD have markedly electron- develop a membranoproliferative glomerulonephritis re-
dense band-like deposits involving the lamina densa, sembling DDD (10).
mesangium, and other basement membranes of the kid- Another mechanism by which the alternative
ney. MIDD is readily distinguishable from DDD by the pathway can become dysregulated is through the for-
demonstration of monoclonal light and/or heavy chains mation of a C3 nephritic factor (C3NeF). C3NeF is an
on immunofluorescence. Interestingly, monoclonal gam- autoantibody that binds to and stabilizes C3 conver-
mopathy of undetermined significance (MGUS) is not tase, protecting it from factor H– or factor I– mediated
an infrequent comorbidity in adult patients with DDD decay. Whereas a C3NeF is detectable in most chil-
and was reported in four of 18 adults in one series (1). dren with DDD using the hemolytic test or the C3NeF
Another report found MGUS in 71% of patients who IgG solid phase assay, up to half of patients with
had a diagnosis of DDD and were older than 49 years MPGN types I or III also have detectable C3NeF
(6). The link between plasma cell dyscrasias and the activity, making this a nonspecific marker (11).
development of DDD may be more than coincidental. How excessive alternative complement pathway
One report suggests that some abnormal monoclonal activation causes the formation of dense deposits in
immunoglobulins have the ability to bind complement tissues is unclear. A recent study used laser capture
regulatory proteins, such as factor H, resulting in microdissection and mass spectrometry to analyze the
dysregulation of the alternative complement pathway composition of the glomerular deposits in DDD and
and the development of DDD (7). Another reported found various components of the alternative and ter-
case demonstrated the ability of a monoclonal light minal complement pathways, including C3, C5, C6,
chain isolated from a patient with MGUS and C3 C7, C8, and C9 (12). Notably, the deposits also con-
glomerulonephritis to initiate activation of the fluid tain certain terminal complement complex regulators,
phase of the alternative pathway (8). Despite the such as clusterin and vitronectin, that function primar-
presence of MGUS, these older adults have a renal ily in the fluid phase. These findings suggest that the
biopsy picture of DDD with glomerular deposits of C3 terminal complement components are activated in the
only (without co-deposits of monoclonal light or fluid phase and form deposits in glomeruli, possibly
heavy chain) (6). favored by glomerular ultrafiltration.
togenic lambda light chain dimer: A unique human miniautoantibody 10. Pickering MC, Cook HT, Warren J, Bygrave AE, Moss J, Walport
against complement factor H. J Immunol 163: 5490 –5496, 1999 MJ, Botto M: Uncontrolled C3 activation causes membranoprolifera-
8. Meri S, Koistinen V, Miettinen A, Tornroth T, Seppala IJ: Activation tive glomerulonephritis in mice deficient in complement factor H.
of the alternative pathway of complement by monoclonal lambda Nat Genet 31: 424 – 428, 2002
light chains in membranoproliferative glomerulonephritis. J Exp Med 11. Schwertz R, Rother U, Anders D, Gretz N, Scharer K, Kirschfink M:
175: 939 –950, 1992 Complement analysis in children with idiopathic membranoprolifera-
9. Abrera-Abeleda MA, Nishimura C, Smith JL, Sethi S, McRae JL, tive glomerulonephritis: A long-term follow-up. Pediatr Allergy
Murphy BF, Silvestri G, Skerka C, Jozsi M, Zipfel PF, Hageman GS, Immunol 12: 166 –172, 2001
Smith RJ: Variations in the complement regulatory genes factor H 12. Sethi S, Gamez JD, Vrana JA, Theis JD, Bergen HR, Zipfel PF,
(CFH) and factor H related 5 (CFHR5) are associated with mem- Dogan A, Smith RJH: Glomeruli of dense deposit disease contain
branoproliferative glomerulonephritis type II (dense deposit disease). components of the alternative and terminal complement pathway.
J Med Genet 43: 582–589, 2006 Kidney Int 75: 952–960, 2009
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Section 11: CASE PRESENTATION kocyturia, but no cellular casts. Serologies include positive anti-
nuclear antibody of 1:40 (speckled pattern), negative anti-DNA
A 49-year-old Caucasian man presents with right upper antibody and anti-Smith antibodies, and reduced serum C3 (⬍10)
quadrant pain and is found to have acute renal failure and and C4 (⬍5) complement levels. Other serologies are negative or
hematuria. He has a history of deep vein thrombosis 12 normal, including hepatitis B surface antigen, hepatitis C anti-
years earlier, aortic valve thrombosis several years earlier, body, HIV, and anti-neutrophil cytoplasmic antibody. Complete
and positive anticardiolipin antibody. He has had hyperten- blood count reveals a hematocrit of 28%, white blood cell count of
sion for ⬍1 year and no diabetes. Physical examination 9000, and platelet count of 56,000. Schistocytes were identified on
reveals livedo reticularis on the lower extremities, no peripheral blood smear. The kidneys measure 12.0 cm and 12.8
edema, and a BP of 158/78 mmHg. Laboratory tests show cm in length by ultrasound. Three days after admission, he
serum creatinine level of 6.0 mg/dl (increased from a baseline develops signs of encephalopathy, and brain computed tomogra-
value of 1.0 mg/dl 3 months earlier) and a serum albumin of 2.3 phy scan reveals multiple microinfarcts. A renal biopsy is
g/dl. Urinalysis reveals 1⫹ protein, microhematuria and leu- performed.
Figure 1. Figure 2.
Figure 3. Fibrin
Figure 4.
What is the BEST diagnosis?
A. Diffuse crescentic glomerulonephritis, pauci-immune type
B. Waldenstrom macroglobulinemia
C. Thrombotic thrombocytopenic purpura
D. Catastrophic antiphospholipid antibody (APA) syndrome
E. Hemolytic-uremic syndrome (HUS)
335
336 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Pathology
Light Microscopy. The renal pathologic findings in
APA-associated nephropathy encompass the spectrum
of renal changes seen in other causes of TMA (14).
Acute TMA is characterized by the presence of fibrin
thrombi (with entrapped red blood cells and platelets)
within glomeruli (Figure 1), arterioles (Figure 2), and
interlobular arteries. Arterial vessels may show mu-
coid intimal edema and hyperplasia (Figure 5). In
severe disease (e.g., in catastrophic APA syndrome),
there may be fibrinoid necrosis of arterial myocytes, Figure 6. A glomerulus shows duplication of GBMs, mim-
interstitial hemorrhage, and frank cortical necrosis. icking membranoproliferative glomerulonephritis, in a case
of chronic APA nephropathy. There is also ischemic wrin-
Although leukocytes may be marginated in the lumen kling of GBMs and dissolution of the mesangial matrix,
of thrombosed vessels, there is typically no evidence consistent with mesangiolysis. Magnification, ⫻600 (Jones
of vasculitis (transmural inflammation of the vessel methenamine silver).
338 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
liferative glomerulonephritis, mesangial C3 glomeru- of coagulation and loose matrix material. Mesangioly-
lonephritis, minimal change disease, and FSGS. sis has a similar electron-lucent appearance owing to
Whether these findings represent coincidental unre- dissolution of the mesangial matrix. The subendothe-
lated renal disease or incipient lupus nephritis or are lial fluffy material may be incorporated into the glo-
directly related to APA is often unclear at the time of merular capillary wall by an underlying thin layer of
biopsy. Some of these patients will subsequently de- newly formed basement membrane, leading to a dou-
velop SLE, and it has been suggested that membra- ble-contoured glomerular capillary wall. Partial or
nous glomerulonephritis, in particular, may be a man- circumferential mesangial interposition often occurs in
ifestation of primary APA syndrome (2). Lupus these areas, producing a membranoproliferative pat-
nephritis is commonly found in patients with lupus- tern (Figure 7). Fibrin tactoids may be seen in areas
related APA nephropathy (6). Any class of lupus with fresh thrombosis (Figure 4). The glomerular en-
nephritis may be seen, but proliferative forms (classes dothelium is often swollen with loss of fenestrations
III and IV) tend to predominate, with the exception of and lifting off the GBM. Cases with coexistent lupus
class V (membranous) lupus nephritis in patients who nephritis have endothelial tubuloreticular inclusions
present with nephrotic syndrome. and electron-dense deposits at the sites of immune
Immunofluorescence. Glomerular and arteriolar complex deposition detected by immunofluorescence.
thrombi stain for fibrin/fibrinogen (Figure 3). Weaker,
nonspecific staining for IgM and C3 (and in some Differential Diagnosis
cases C1q) may be present in areas of thrombosis and The differential diagnosis of renal TMA is broad
in sclerosing glomeruli. Immune deposits that stain for and requires careful clinical correlation to reach a
IgG (with possible co-deposits of IgA, IgM, C3, and more specific diagnosis. A major cause of renal TMA
C1q) are a feature of coexistent lupus nephritis but are is HUS caused by infections by shigatoxin-producing
not seen in primary APA nephropathy. Escherichia coli (mostly of the O157:H7 serotype),
neuraminidase-producing Streptococcus pneumoniae,
Electron Microscopy. A characteristic ultrastruc-
or HIV. There is increasing recognition that some
tural finding in all forms of TMA, including that
patients with HUS have inherited deficiencies in reg-
related to APA syndrome, is the presence of ill-
ulators of the alternative complement pathway such as
defined, flocculent electron-lucent material (some-
factor H, membrane co-factor protein, factor I, or
times termed “fluff”) in the subendothelial space be-
complement C3 (16). Other individuals with HUS may
tween endothelial cells and the GBM (Figure 7). This
develop inhibitory autoantibodies to complement reg-
fluffy material is thought to contain degraded products
ulatory proteins, particularly to factor H (17). Throm-
botic thrombocytopenic purpura typically manifests
with skin and neurologic involvement and may be due to
genetic deficiency of von Willebrand factor cleaving pro-
tease (ADAMTS13) or inhibitory autoantibodies. Other
major causes of renal TMA include acute scleroderma
renal crisis, malignant hypertension, eclampsia of
pregnancy, drug toxicity, radiation toxicity, and neo-
plasia. Drugs that can cause TMA include a host of
chemotherapeutic agents (e.g., gemcitabine, mitomy-
cin C, tamoxifen, bleomycin, cisplatin, cytosine arabi-
noside, daunorubicin, antivascular endothelial growth
factor agents) (18,19), as well as antiplatelet agents
(ticlopidine and clopidogrel), quinine, calcineurin inhib-
itors, and oral contraceptives. Some patients with rheu-
matoid arthritis receiving anti–TNF-␣ agents have been
Figure 7. Electron micrograph showing subendothelial elec-
tron-lucent “fluff” (arrow) and duplication of basement
reported to develop APA and renal TMA (20,21). Al-
membranes (arrowheads), producing a membranoprolifera- though pathogenesis is diverse, these various forms of
tive pattern. Magnification, ⫻6000. TMA share injury to endothelial cells as a common
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 339
16. Noris M, Remuzzi G: Hemolytic uremic syndrome. J Am Soc Neph- factor alpha antagonists in patients with rheumatoid arthritis induces
rol 16: 1035–1050, 2005 anticardiolipin antibodies. Ann Rheum Dis 63: 1075–1078, 2004
17. Dragon-Durey MA, Loirat C, Cloarec S, Macher MA, Blouin J, Nivet 22. Uthman I, Khamashta M: Ethnic and geographical variation in
H, Weiss L, Fridman WH, Fremeaux-Bacchi V: Anti-factor H auto- antiphospholipid (Hughes) syndrome. Ann Rheum Dis 64: 1671–
antibodies associated with atypical hemolytic uremic syndrome. J Am 1676, 2005
Soc Nephrol 16: 555–563, 2005 23. Vega-Ostertag ME, Pierangeli SS: Mechanisms of aPL-mediated
18. Eremina V, Jefferson JA, Kowalewska J, Hochster H, Haas M, thrombosis: Effects of aPL on endothelium and platelets. Curr
Weisstuch J, Richardson C, Kopp JB, Kabir MG, Backx PH, Gerber Rheumatol Rep 9: 190 –197, 2007
HP, Ferrara N, Barisoni L, Alpers CE, Quaggin SE: VEGF inhibition 24. Pierangeli SS, Vega-Ostertag ME, Raschi E, Liu X, Romay-
and renal thrombotic microangiopathy. N Engl J Med 358: 1129 – Penabad Z, De Micheli V, Galli M, Moia M, Tincani A, Borghi
1136, 2008 MO, Nguyen-Oghalai T, Meroni PL: Toll-like receptor and an-
19. Stokes MB, Erazo MC, D’Agati VD: Glomerular disease related to tiphospholipid mediated thrombosis: In vivo studies. Ann Rheum
anti-VEGF therapy. Kidney Int 74: 1487–1491, 2008 Dis 66: 1327–1333, 2007
20. Stokes MB, Foster K, Markowitz GS, Ebrahimi F, Hines W, Kauf- 25. Espinosa G, Cervera R: Antiphospholipid syndrome. Arthritis Res
man D, Moore B, Wolde D, D’Agati VD: Development of glomer- Ther 10: 230, 2008
ulonephritis during anti-TNF-alpha therapy for rheumatoid arthritis. 26. Blank M, Krause I, Fridkin M, Keller N, Kopolovic J, Goldberg I,
Nephrol Dial Transplant 20: 1400 –1406, 2005 Tobar A, Shoenfeld Y: Bacterial induction of autoantibodies to
21. Jonsdottir T, Forslid J, van Vollenhoven A, Harju A, Brannemark S, beta2-glycoprotein-I accounts for the infectious etiology of antiphos-
Klareskog L, van Vollenhoven RF: Treatment with tumour necrosis pholipid syndrome. J Clin Invest 109: 797– 804, 2002
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Section 12: CASE PRESENTATION examination reveals a BP of 130/64 mmHg, weight of 118 lb,
and no edema or cutaneous manifestations. Urinalysis reveals
An 86-year-old Caucasian woman presents with acute 2⫹ protein, four to 10 red blood cells per high-power field, and
kidney injury (AKI) and a creatinine level of 2.22 mg/dl. numerous white blood cells. She has a normal serum free light
Past medical history is significant for chronic kidney chain ratio, no evidence of a monoclonal serum spike, no
disease with a baseline creatinine of 1.3 mg/dl 10 months eosinophilia, a negative urine culture, serum albumin of 3.9
earlier, osteoporosis, and recent 10-lb weight loss. The g/dl, serum calcium of 10.2 mg/dl, hematocrit of 32.9%, and a
patient recently underwent urogynecologic evaluation for urine protein-creatinine ratio of 1.8. Negative serologies in-
frequent nocturia and urge incontinence. She was found clude anti-nuclear antibody, anti– glomerular basement mem-
to have sterile pyuria, a second-degree cystocele, and an brane antibody, anti-neutrophil cytoplasmic antibody, hepa-
elevated post-void residual volume. She was treated with titis B surface antigen, and hepatitis C virus antibody.
a pessary, leading to mild improvement in her symptoms. Serum C3 and C4 are normal. Her kidneys measure 9.0
Her medications are limited to multivitamins and vitamin and 9.1 cm in length by ultrasound, without evidence of
D, although she has previously been treated for osteopo- obstruction. Three weeks later, the patient’s creatinine
rosis with monthly risedronate. There is no history of level remains elevated at 2.2 mg/dl. Renal biopsy is
nonsteroidal anti-inflammatory drug (NSAID) use. Physical performed.
Figure 1. Figure 2.
Figure 3. Figure 4.
Multiple clinical and laboratory findings may Detailed clinical history, physical examination,
provide clues to the pathogenesis of AIN in individual and serologic evaluation may provide important clues
patients. The initial report of drug-induced AIN by as to the etiology of non– drug-induced forms of AIN.
Baldwin et al. (12) described seven patients who For instance, uveitis and AIN are seen in patients with
developed AIN after treatment with penicillin or meth- Sjögren syndrome, sarcoidosis, or TINU syndrome;
icillin and noted the presence of systemic signs of a pulmonary disease suggests the possibility of sarcoid-
hypersensitivity reaction including fever, rash, and osis; signs of systemic vasculitis may implicate We-
eosinophilia. Subsequent experience has shown that gener granulomatosis; and pancreatitis raises the pos-
this “classic triad” of symptoms of drug-induced AIN sibility of IgG4 immune complex tubulointerstitial
is seen much more commonly after treatment with nephritis (9). Infectious causes of AIN should be
-lactam antibiotics than with other classes of drugs carefully excluded in immunosuppressed and trans-
and that, overall, the full triad is seen in ⬍10% of plant patients.
patients (3,13). Eosinophiluria, when present, is also Many patients with AIN present only with unex-
regarded as a relatively specific sign of drug-induced plained renal insufficiency, and the biopsy findings of
AIN. AIN come as a surprise to the nephrologist. The
Given the high prevalence of drug-induced AIN, possibility of AIN should be considered for all patients
the most common inciting therapeutic agents deserve with unexplained renal insufficiency, in particular in
special consideration. The form of AIN induced by the setting of leukocyturia and subnephrotic protein-
NSAIDs has distinctive features including the typical uria.
absence of signs of a hypersensitivity reaction, a long
duration of treatment (typically ⬎6 months) before the Pathology
development of AIN, and the usual absence of inter- Light microscopy. The diagnosis of AIN can be
stitial eosinophils (14). The 5-aminosalicylates, an- established only by light microscopy, where the pre-
other class of anti-inflammatory drugs that are mainly dominant findings are interstitial inflammation with
used to treat inflammatory bowel disease, are also extension of the infiltrate across TBMs, producing
commonly associated with AIN including 42 pub- “tubulitis” (Figures 1 through 3). All areas of the
lished cases (15) and an incidence that has been cortex may be involved, sometimes with particularly
estimated to be greater than 1 in 500 patients (15–17). intense inflammation in the outer medulla. In most
Similar to NSAIDs, the AIN that follows use of forms of interstitial nephritis, lymphocytes are the
5-aminosalicylates is associated with a longer duration predominant component of the interstitial infiltrate,
of therapy and the usual absence of eosinophils. The although monocytes, plasma cells, eosinophils, and
most rapidly rising cause of drug-induced AIN and, in neutrophils also may be seen. The relative composi-
some countries, the most common etiology overall is tion of the interstitial infiltrate may provide clues to
proton pump inhibitors (PPIs) used to treated gastritis the cause of AIN. For instance, the presence of prom-
and gastroesophageal reflux disease. PPI-induced AIN inent interstitial eosinophils strongly favors a drug-
appears to represent a class effect because it can induced cause, although the absence of eosinophils
follow the use of most, if not all, available PPIs. The does not argue against this possibility. Plasma cells are
AIN that follows the use of PPIs presents at a mean of particularly prominent with renal parenchyma infec-
3 months following exposure, is commonly associated tion by BK polyoma virus (BK nephropathy) and IgG4
with eosinophilic inflammation, and may exhibit signs immune complex tubulointerstitial nephritis with au-
of a systemic hypersensitivity reaction (18 –20). There toimmune pancreatitis. Prominent neutrophils, in par-
are at least 64 reports of biopsy-proven AIN related to ticular when accompanied by neutrophilic tubulitis,
PPIs (18). AIN has been attributed to multiple classes neutrophil casts, or microabscesses, suggest renal pa-
of antibiotics other than the -lactams, including ri- renchymal infection, typically of bacterial origin (i.e.,
fampin, sulfonamides, and the fluoroquinolones, acute pyelonephritis) (Figure 5).
among many others. Importantly, drug-induced AIN The interstitial inflammation and tubulitis of
represents an idiosyncratic, non-dose-dependent reac- AIN are commonly accompanied by findings of acute
tion that can potentially follow the use of any thera- tubular injury, including luminal ectasia, cytoplasmic
peutic agent. simplification and vacuolization, irregular luminal
344 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
any GIN, fungal and mycobacterial infections should report from the UK MDC Glomerulonephritis Register and a review
of the literature. Nephrol Dial Transplant 13[Suppl 7]: 12–16, 1998
be excluded by performing special stains for micro- 3. Clarkson MR, Giblin L, O’Connell FP, O’Kelly P, Walshe JJ, Conlon
organisms (AFB and Gomori methenamine silver) and P, O’Meara Y, Dormon A, Campbell E, Donohue J: Acute interstitial
by urine cultures. nephritis: Clinical features and response to corticosteroid therapy.
Nephrol Dial Transplant 19: 2778 –2783, 2004
4. Schwarz A, Krause PH, Kunzerdorf U, Keller F, Distler A: The
Pathogenesis outcome of acute interstitial nephritis: Risk factors for the transition
The pathogenesis of AIN varies depending on from acute to chronic interstitial nephritis. Clin Nephrol 54: 179 –
the specific etiology. Inflammation is a basic response 190, 2000
5. Buysen JG, Houtlhoff HJ, Krediet RT, Arisz L: Acute interstitial
to infection in infectious forms of AIN. The interstitial nephritis: A clinical and morphological study in 27 patients. Nephrol
infiltrates in systemic, presumed autoimmune forms of Dial Transplant 5: 94 –99, 1990
AIN are part of a larger, multisystem inflammatory 6. Farrington K, Levison DA, Greenwood RN, Cattell WR, Baker LR:
Renal biopsy in patients with unexplained renal impairment and
response that, in the majority of cases, only second- normal kidney size. Q J Med 70: 221–233, 1989
arily involves the renal parenchyma. TINU, an idio- 7. Baker RJ, Pusey CD: The changing profile of acute tubulointerstitial
pathic condition that by definition involves the kidney, nephritis. Nephrol Dial Transplant 19: 8 –11, 2004
8. Kambham N, Markowitz GS, Tanji N, Mansukhani MM, Orazi A,
is thought to be mediated by dysregulated cell-medi- D’Agati VD: Idiopathic hypocomplementemic interstitial nephritis
ated immunity in patients with genetic susceptibility, as with extensive tubulointerstitial deposits. Am J Kidney Dis 37:
388 –399, 2001
evidenced by a strong association with the HLA-DQ and
9. Cornell LD, Chicano SL, Deshpande V, Collins AB, Selig MK,
HLA-DR alleles (27). Lauwers GY, Barisoni L, Colvin RB: Pseudotumors due to IgG4
Drug-induced AIN likely represents a type IV immune-complex tubulointerstitial nephritis associated with autoim-
mune pancreatocentric disease. Am J Surg Pathol 31: 1586 –1597,
(delayed-type) hypersensitivity response. This concept 2007
is supported by multiple clinical observations includ- 10. Saeki T, Nishi S, Imai N, Ito T, Yamazaki H, Kawano M, Yamamoto
ing the fact that a minimum of 7 to 10 days of M, Takahashi H, Matsui S, Nakada S, Origuchi T, Hirabayashi A,
Homma N, Tsubata Y, Takata T, Wada Y, Saito A, Fukase S, Ishioka
exposure typically precedes the development of AKI; K, Miyazaki Y, Umehara H, Sugai S, Narita I: Cliniopathologic
a shorter exposure time induces AIN after repeat characteristics of patients with IgG4-related tubulointerstitial nephri-
exposure; the common presence of systemic signs of a tis. Kidney Int 78: 1016 –1023, 2010
11. Cheuk W, Chan JK: IgG4-related sclerosing disease: A critical
hypersensitivity reaction; and the idiosyncratic, non– appraisal of an evolving clinicopathologic entity. Adv Anat Pathol 17:
dosage-dependent nature of this lesion (13). Patho- 303–332, 2010
logic findings that support a type IV hypersensitivity 12. Baldwin DS, Levine BB, McCluskey RT, Gallo GR: Renal failure
and interstitial nephritis due to penicillin and methicillin. N Engl
response include the predominance of T cells, frequent J Med 279: 1245–1252, 1968
presence of eosinophils, and the absence of Ig deposits 13. Rossert J: Drug-induced acute interstitial nephritis. Kidney Int 60:
by immunofluorescence. Of note, the interstitial infil- 804 – 817, 2001
14. Pirani CL, Valeri A, D’Agati V, Appel GB: Renal toxicity of nonsteroi-
trate in drug-induced AIN related to -lactam antibi- dal anti-inflammatory drugs. Contrib Nephrol 55: 159 –175, 1987
otics and NSAIDs has been shown to be composed of 15. Gisbert JP, Gonzalez-Lama Y, Mate J: 5-Aminosalicylates and renal
72% T cells with a relative equal admixture of CD4⫹ function in inflammatory bowel disease: A systemic review. Inflamm
Bowel Dis 13: 629 – 638, 2007
and CD8⫹ cells, 15% monocytes, and 7% B cells (28). 16. World MJ, Stevens PE, Ashton MA, Rainford DJ: Mesalamine-associ-
The kidney is vulnerable to drug-induced AIN ated interstitial nephritis. Nephrol Dial Transplant 11: 614 – 621, 1996
17. Arend LJ, Springate JE: Interstitial nephritis from mesalamine: Case
because of its important role in drug clearance and
report and literature review. Pediatr Nephrol 19: 550 –553, 2004
metabolism. The precise mechanism by which indi- 18. Brewster UC, Perazella MA: Proton pump inhibitors and the kidney:
vidual therapeutic agents elicit this response remains Critical review. Clin Nephrol 68: 65–72, 2007
19. Geevasinga N, Coleman PL, Webster AC, Roger SD: Proton pump
poorly understood. Hypotheses include tubular epithe- inhibitors and acute interstitial nephritis. Clin Gastroenterol Hepatol
lial uptake of the drug or its metabolites with binding 4: 597– 604, 2006
to cellular constituents or TBM, leading to drug- 20. Sierra F, Suarez M, Rey M, Vela MF: Systemic review: Proton pump
inhibitor-associated acute interstitial nephritis. Aliment Pharmacol
hapten formation. A cell-mediated immune response Ther 26: 545–553, 2007
ensues. Roles for molecular mimicry and individual 21. Border WA, Lehman DH, Egan JD, Sass HJ, Glode JE, Wilson CB:
host response genes have been proposed (13). Antitubular basement-membrane antibodies in methicillin-associated
interstitial nephritis. N Engl J Med 291: 381–384, 1974
22. Bracamonte E, Leca N, Smith KD, Nicosia RF, Nickeleit V, Kend-
References rick E, Furmanczyk PS, Davis CL, Alpers CE, Kowaleska J: Tubular
1. Councilman WT: Acute interstitial nephritis. J Exp Med 3: 393– 420, basement membrane immune deposits in association with BK polyo-
1898 mavirus nephropathy. Am J Transplant 7: 1552–1560, 2007
2. Davison AM, Jones CH: Acute interstitial nephritis in the elderly: A 23. Bijol V, Mendez GP, Nose V, Rennke HG: Granulomatous interstitial
346 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
nephritis: A clinicopathologic study of 46 cases from a single insti- 27. Levinson RD, Park MS, Rikkers SM, Reed EF, Smith JR, Martin TM,
tution. Int J Surg Pathol 14: 57– 63, 2006 Rosenbaum JT, Foster CS, Sherman MD, Holland GN: Strong
24. Joss N, Morris S, Young B, Geddes C: Granulomatous interstitial associations between specific HLA-DQ and HLA-DR alleles and the
nephritis. Clin J Am Soc Nephrol 2: 222–230, 2007 tubulointerstitial nephritis and uveitis syndrome. Invest Ophthalmol
25. Ram R, Swarnalatha G, Prasad N, Prayaga A, Dakshina Murthy KV: Vis Sci 44: 653– 657, 2003
Granulomatous interstitial nephritis following prolonged use of phe- 28. D’Agati VD, Theise ND, Pirani CL, Knowles DM, Appel GB:
nytoin. Saudi J Kidney Dis Transpl 20: 131–133, 2009 Interstitial nephritis related to nonsteroidal anti-inflammatory agents
26. Mignon F, Mery JP, Mougenot B, Ronco P, Roland J, Morel-Maroger and -lactam antibiotics: A comparative study of the interstitial
L: Granulomatous interstitial nephritis. Adv Nephrol Necker Hosp 13: infiltrates using monoclonal antibodies. Mod Pathol 2: 390 –396,
219 –245, 1984 1989
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Section 13: CASE PRESENTATION earlier. Physical examination reveals a BP of 187/92 mmHg,
3⫹ pitting lower extremity edema, and no hepatosplenomeg-
An 87-year-old Caucasian woman presents with abrupt aly or cutaneous manifestations. Laboratory evaluation reveals
onset of lower extremity edema and is found to have nephrotic a 24-hour urine protein of 18.0 g, serum albumin 1.7 g/dl,
syndrome and acute kidney injury (AKI) with a creatinine level hematocrit 34%, normal C3 and C4 complement levels, and no
of 2.84 mg/dl. Past medical history is significant for hyperten- evidence of a monoclonal serum or urine spike. Urinalysis
sion, depression, anemia, osteoarthritis, peptic ulcer disease, reveals 4⫹ protein with 8 to 10 red blood cells per high-power
and bladder incontinence. Her medications include furo- field, 30 to 50 white blood cells per high-power field, and no
semide, metoprolol, duloxetine, mirtazapine, alprazolam, hy- cellular casts. Urine cultures are positive for Escherichia coli.
droxycodone, pantoprazole, risedronate, aspirin, and polysac- The following serologies are negative: Anti-nuclear antibody,
charide-iron complex. There is no history of diabetes or recent hepatitis C antibody, hepatitis B surface antigen, anti-neutro-
nonsteroidal anti-inflammatory drug (NSAID) use, and the phil cytoplasmic antibody, and anti– glomerular basement
patient’s baseline serum creatinine was 0.6 mg/dl 3 months membrane (anti-GBM) antibody.
Figure 1. Figure 2.
Figure 4.
Figure 3.
sampled FSGS in a child. This is a particularly its can be seen in IgM nephropathy, C1q nephropathy,
important consideration in a kidney biopsy with sub- or coexistent IgA nephropathy.
optimal sampling of deep juxtamedullary glomeruli, Diffuse Mesangial Hypercellularity. DMH is a
where the first lesions of FSGS typically develop. variant of MCD that is predominantly seen in young
Prominent protein and lipid resorption droplets are children and is associated with higher rates of initial
often observed in the tubules of MCD. Usually no steroid resistance and a higher incidence of microhe-
significant interstitial inflammation is seen. The pres- maturia than the usual form of MCD (Figure 6).
ence of significant interstitial inflammation and tubu- However, the remission rates of both groups are sim-
litis, sometimes associated with eosinophils, should ilar after 52 weeks of follow-up, and long-term out-
raise the suspicion of NSAID-induced MCD and acute comes are comparable (6). The renal biopsy findings
interstitial nephritis. Varying severity of arterio- and are akin to MCD except for the presence of diffuse
arteriolosclerosis may be observed in older patients. mesangial hypercellularity defined as more than four
Acute tubular injury characterized by simplified mesangial cells per area of mesangium and affecting
tubular epithelium, sloughed epithelial cells, and loss ⬎80% of the glomeruli sampled (6), although the
of proximal tubular brush borders typically occurs in exact criteria vary in different studies (7). A recent
adults with MCD and the syndrome of “MCD with pediatric study suggested that DMH (defined as more
acute renal failure.” This combination of findings is than three cells per mesangial area) in the presence of
thought to be mediated by hemodynamic factors with NS predicts multiple relapses but preservation of nor-
intravascular volume depletion in the setting of pro- mal renal function (8). Immunofluorescence is usually
found hypoalbuminemia and edema, in turn causing negative but may occasionally demonstrate mesangial
renal hypoperfusion and ischemic tubular injury. IgM and C3 only. Ultrastructural features include diffuse
Older adults with history of hypertension and vascular podocyte injury similar to MCD. Small paramesangial
narrowing as a result of arteriosclerosis are most deposits may be present in some cases, corresponding to
susceptible to this complication (4,5). the IgM and C3 staining.
Immunofluorescence. Immunofluorescence staining Relationship with FSGS. One of the challenges in
for immunoglobulins and complements is usually neg- pediatric NS is to determine whether biopsy findings
ative. Tubular protein resorption droplets may be high- of apparent MCD in fact represent undersampled
lighted by staining for albumin. High-intensity mes- FSGS. This sampling error may explain a diagnosis of
angial IgM or C1q staining should suggest IgM FSGS rendered on a repeat biopsy in a patient with a
nephropathy and C1q nephropathy, respectively, both previous biopsy diagnosis of MCD. It has also been
of which may be considered variants of MCD in this argued that MCD and FSGS represent a continuum of
setting. The presence of significant mesangial IgA
deposits (at least 1⫹ intensity) in an otherwise classic
case of MCD suggests a dual diagnosis of MCD with
coexistent mesangial IgA nephropathy.
Electron Microscopy. The ultrastructural abnor-
malities are limited to extensive podocyte foot process
effacement, usually involving ⬎75% of the total cap-
illary surface area examined. A sheet of podocyte
cytoplasm overlies normal GBMs, often with conden-
sations of actin cytoskeleton parallel to the GBM.
Villous projections of the podocyte cytoplasm into the
urinary space are often described as “microvillous
transformation” of the podocyte cell bodies. These
podocyte changes may be milder in partially treated
MCD but become extensive in the setting of a relapse. Figure 6. Diffuse mesangial hypercellularity is a variant of
No immune-type electron-dense deposits are identified MCD characterized by four or more mesangial cells per
in MCD. Significant mesangial electron-dense depos- mesangial area. Magnification, ⫻400 (H&E).
350 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
podocyte disease (podocytopathy) depending on the cytes can respond to external stimuli by activating
severity and reversibility of the podocyte injury and enzymatic pathways that dysregulate the actin cyto-
that some cases of MCD may evolve into FSGS over skeleton and filtration slit diaphragm.
time. In the majority of patients, however, the different Abnormalities in the proportion of T cell subsets
clinical course and pathologic findings at the outset have been documented in the serum of patients with
suggest that MCD and FSGS are two distinct entities. MCD. In general, patients with MCD have predomi-
Several studies have attempted to apply immu- nance of (T helper 2) Th2 cytokine response that has
nohistochemical methods to improve the accuracy of a role in antibody production and allergic reactions.
biopsy diagnosis of MCD and FSGS, but these tests Th2 cytokines are produced by lymphocytes and mast
are not used in clinical practice. A few investigators cells. MCD has also been linked to atopy and elevated
have reported reduced or redistributed GBM nephrin serum IgE levels (16). Levamisole, an immunostimu-
staining, but other studies could not confirm these lant and antihelminth drug used to reduce the relapse
findings (9). ␣-Dystroglycan, a podocyte protein im- rate of MCD is known to augment the Th1 response
portant in adhesion to GBM, may be reduced in MCD, and thus reset the Th1/Th2 balance (11). Although
but additional confirmatory studies are needed to es- most of the data focus on T cell abnormalities in
tablish the clinical utility of this immunostain (10). MCD, B cell responses may also play a role. Hypo-
gammaglobulinemia is seen in MCD with particularly
Pathogenesis suppressed levels of IgG1 and IgG2 and class-switch-
The pathogenesis of MCD is poorly understood. ing of B cells to increase production of IgG4 and IgE
It is thought to be mediated by targeted podocyte (17,18). It remains uncertain to what extent some of
injury, possibly through the presence of a circulating these immunologic abnormalities are cause or conse-
“permeability factor” with the ability to alter podocyte quence of the NS (19).
structure and glomerular permselectivity. There is sub- Hypotheses on pathogenesis include the presence
stantial evidence that patients with MCD have dys- of a circulating soluble factor and/or lack of an inhib-
regulated immunity, especially involving T lympho- itory molecule causing reorganization of the podo-
cytes. Treatment response to immunosuppressive cyte’s actin cytoskeleton, leading to reversible foot
agents such as corticosteroids and occurrence of MCD process effacement and proteinuria (20). Circumstan-
in association with viral infections (e.g., measles), tial evidence for such a theory includes the disappear-
Hodgkin’s lymphoma, thymoma, and T cell lympho- ance of NS when a kidney with MCD is transplanted
mas support such T cell theories. In the absence of into a patient without NS and reversal of recurrent
glomerular inflammation, the injury is likely cytokine MCD-associated NS in the allograft by plasmaphere-
mediated. Podocytes express receptors for interleukins sis. A circulating “permeability factor” was initially
(IL-4, -10, and -13) and chemokines (CCR and described in patients with FSGS (21) but has not been
CXCR), as well as Toll-like receptors (11), and in fully characterized. T cell hybridomas derived from T
vitro studies have demonstrated that particular cyto- cells of patients with MCD produce a glomerular
kines have direct effects on podocyte signaling, cyto- permeability factor, presumably a lymphokine, that
skeletal organization, motility and maintenance of the induces proteinuria in rats (22). Recent studies have
filtration barrier. Overexpression of IL-13 can induce shown that hemopexin, a heme-scavenging molecule,
MCD like nephropathy in rats (12). The induction of may be linked to MCD. Activated hemopexin func-
CD80 (B7–1) expression by podocytes after lipopoly- tions as a serine protease causing remodeling of podo-
saccharide exposure in mice causes reversible protein- cytes and alters the glomerular filtration barrier. It
uria and foot process effacement resembling human causes reorganization of the podocyte’s actin-based
MCD (13). CD80 also has been identified in the urine cytoskeleton and requires the slit diaphragm protein
of patients with MCD but not FSGS, suggesting it may nephrin for this function. Hemopexin is abundantly
be a useful biomarker (14). Podocyte expression of present in plasma and is an acute-phase reactant but is
CD80 has been shown to induce cathepsin L–mediated also synthesized by mesangial cells. It has been pro-
cleavage of large GTPase dynamin and synaptopodin, posed that lack of a putative inhibitory/protective fac-
leading to disruption of the actin cytoskeleton and foot tor in MCD may expose podocytes to the effects of
process effacement (15). These data suggest that podo- activated hemopexin (23).
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 351
Recent advances in the pathogenesis of FSGS are reported associations with Epstein-Barr virus and
have highlighted the significance of mutations in HIV infection. Allergic reactions associated with bee
podocyte and slit diaphragm genes. However, to date, stings, pollen exposure, and envenomations can pre-
no homozygous or compound heterozygous mutations cede the development of MCD. These secondary as-
of podocyte genes have been observed in MCD. Some sociations suggest that diverse precipitants may result
patients with MCD have heterozygous allelic varia- in common pathogenic pathways to podocyte injury
tions with amino acid substitutions in nephrin and and the development of MCD.
podocin genes, but their clinical significance is unclear
because there is no correlation between these genetic References
variations and steroid responsiveness or relapse rates 1. Nephrotic syndrome in children: Prediction of histopathology from
(24). It is possible that heterozygous variations of podo- clinical and laboratory characteristics at time of diagnosis. A report of
the International Study of Kidney Disease in Children. Kidney Int 13:
cyte proteins may predispose these individuals to develop 159 –165, 1978
NS when exposed to an extrinsic stimulus. A single 2. Nair R, Bell JM, Walker PD: Renal biopsy in patients aged 80 years
report of familial MCD in three families identified a gene and older. Am J Kidney Dis 44: 618 – 626, 2004
3. Hiraoka M, Takeda N, Tsukahara H, Kimura K, Takagi K, Hayashi
locus (SSNS1) for steroid-sensitive NS on chromosome S, Kato E, Ohta K, Sudo M: Favorable course of steroid-responsive
2 (25). However, overall, when compared with FSGS, nephrotic children with mild initial attack. Kidney Int 47: 1392–1393,
1995
MCD seems to be an acquired disease with genetic
4. Jennette JC, Falk RJ: Adult minimal change glomerulopathy with
susceptibility playing a minor role. acute renal failure. Am J Kidney Dis 16: 432– 437, 1990
Although the unifying hypothesis in the patho- 5. Moutzouris DA, Herlitz L, Appel GB, Markowitz GS, Freudenthal B,
Radhakrishnan J, D’Agati VD: Renal biopsy in the very elderly. Clin
genesis of MCD and FSGS involves podocyte injury, J Am Soc Nephrol 4: 1073–1082, 2009
the severity of injury may determine the disease phe- 6. Primary nephrotic syndrome in children: Clinical significance of
notype (26,27). Unlike FSGS, the podocyte injury in histopathologic variants of minimal change and of diffuse mesangial
hypercellularity. A Report of the International Study of Kidney
MCD is sublethal in that there is no podocyte loss or Disease in Children. Kidney Int 20: 765–771, 1981
podocyturia. The podocytes are thought to undergo 7. Childhood nephrotic syndrome associated with diffuse mesangial
foot process effacement via alterations in podocyte hypercellularity. A report of the Southwest Pediatric Nephrology
Study Group. Kidney Int 24: 87–94, 1983
signaling and motility, leading to altered permselec- 8. Silverstein DM, Craver RD: Mesangial hypercellularity in children:
tivity of the filtration barrier (27,28). These changes Presenting features and outcomes. Pediatr Nephrol 23: 921–928,
2008
are potentially reversible in MCD after immunosup-
9. Hingorani SR, Finn LS, Kowalewska J, McDonald RA, Eddy AA:
pressive therapy. Recent insights suggest that gluco- Expression of nephrin in acquired forms of nephrotic syndrome in
corticoids, cyclosporine, angiotensin-converting en- childhood. Pediatr Nephrol 19: 300 –305, 2004
10. Giannico G, Yang H, Neilson EG, Fogo AB: Dystroglycan in the
zyme inhibitors, and angiotensin II receptor blockers diagnosis of FSGS. Clin J Am Soc Nephrol 4: 1747–1753, 2009
exert direct protective effects on the podocyte in ad- 11. Mathieson PW: Minimal change nephropathy and focal segmental
dition to their systemic immunosuppressive or antihy- glomerulosclerosis. Semin Immunopathol 29: 415– 426, 2007
12. Lai KW, Wei CL, Tan LK, Tan PH, Chiang GS, Lee CG, Jordan
pertensive effects (11,29,30). SC, Yap HK: Overexpression of interleukin-13 induces minimal-
Secondary MCD. MCD can occur in association change-like nephropathy in rats. J Am Soc Nephrol 18: 1476 –
1485, 2007
with several underlying diseases such as drugs, neo- 13. Reiser J, von Gersdorff G, Loos M, Oh J, Asanuma K, Giardino L,
plasia, infections, and atopy (31). Certain drugs have Rastaldi MP, Calvaresi N, Watanabe H, Schwarz K, Faul C, Kretzler
M, Davidson A, Sugimoto H, Kalluri R, Sharpe AH, Kreidberg JA,
been reported to cause MCD either by direct podocyte
Mundel P: Induction of B7–1 in podocytes is associated with ne-
effects (e.g., lithium, IFN, pamidronate) or by hyper- phrotic syndrome. J Clin Invest 113: 1390 –1397, 2004
sensitivity reaction (e.g., NSAID). Neoplastic diseases 14. Garin EH, Mu W, Arthur JM, Rivard CJ, Araya CE, Shimada M,
Johnson RJ: Urinary CD80 is elevated in minimal change disease but
that cause MCD include Hodgkin’s and non-Hodg- not in focal segmental glomerulosclerosis. Kidney Int 78: 296 –302,
kin’s lymphoma, leukemia, carcinomas, and thymo- 2010
mas, among others (31–33). There may be a molecular 15. Mundel P, Reiser J: Proteinuria: An enzymatic disease of the podo-
cyte? Kidney Int 77: 571–580, 2010
link between classical Hodgkin’s disease and MCD, 16. Shao YN, Chen YC, Jenq CC, Hsu HH, Chang MY, Tian YC, Fang
with evidence of induction of a regulatory molecule, JT, Yang CW: Serum immunoglobulin E can predict minimal change
c-mip, in Hodgkin’s and Reed-Sterberg cells and disease before renal biopsy. Am J Med Sci 338: 264 –267, 2009
17. Warshaw BL, Check IJ: IgG subclasses in children with nephrotic
podocytes (34). The exact relationship between MCD syndrome. Am J Clin Pathol 92: 68 –72, 1989
and infections may be difficult to establish, and there 18. Mosmann TR, Coffman RL: TH1 and TH2 cells: Different patterns of
352 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
lymphokine secretion lead to different functional properties. Annu (SSNS1) for steroid-sensitive nephrotic syndrome on chromosome
Rev Immunol 7: 145–173, 1989 2p. J Am Soc Nephrol 14: 1897–1900, 2003
19. Eddy AA, Symons JM: Nephrotic syndrome in childhood. Lancet 26. Wiggins RC: The spectrum of podocytopathies: A unifying view of
362: 629 – 639, 2003 glomerular diseases. Kidney Int 71: 1205–1214, 2007
20. Garin EH: Circulating mediators of proteinuria in idiopathic minimal 27. D’Agati VD: The spectrum of focal segmental glomerulosclerosis:
lesion nephrotic syndrome. Pediatr Nephrol 14: 872– 878, 2000 New insights. Curr Opin Nephrol Hypertens 17: 271–281, 2008
21. Savin VJ, Sharma R, Sharma M, McCarthy ET, Swan SK, Ellis E, 28. Shankland SJ: The podocyte’s response to injury: Role in proteinuria
Lovell H, Warady B, Gunwar S, Chonko AM, Artero M, Vincenti F: and glomerulosclerosis. Kidney Int 69: 2131–2147, 2006
Circulating factor associated with increased glomerular permeability 29. Lane JC, Kaskel FJ: Pediatric nephrotic syndrome: From the simple
to albumin in recurrent focal segmental glomerulosclerosis. N Engl to the complex. Semin Nephrol 29: 389 –398, 2009
30. Reiser J, Gupta V, Kistler AD: Toward the development of podocyte-
J Med 334: 878 – 883, 1996
specific drugs. Kidney Int 77: 662– 668, 2010
22. Koyama A, Fujisaki M, Kobayashi M, Igarashi M, Narita M: A
31. Glassock RJ: Secondary minimal change disease. Nephrol Dial
glomerular permeability factor produced by human T cell hybrid-
Transplant 18[Suppl 6]: vi52–vi58, 2003
omas. Kidney Int 40: 453– 460, 1991
32. Karras A, de Montpreville V, Fakhouri F, Grunfeld JP, Lesavre P:
23. Lennon R, Singh A, Welsh GI, Coward RJ, Satchell S, Ni L, Renal and thymic pathology in thymoma-associated nephropathy:
Mathieson PW, Bakker WW, Saleem MA: Hemopexin induces neph- Report of 21 cases and review of the literature. Nephrol Dial
rin-dependent reorganization of the actin cytoskeleton in podocytes. Transplant 20: 1075–1082, 2005
J Am Soc Nephrol 19: 2140 –2149, 2008 33. Dabbs DJ, Striker LM, Mignon F, Striker G: Glomerular lesions in
24. Lahdenkari AT, Suvanto M, Kajantie E, Koskimies O, Kestila M, lymphomas and leukemias. Am J Med 80: 63–70, 1986
Jalanko H: Clinical features and outcome of childhood minimal 34. Audard V, Zhang SY, Copie-Bergman C, Rucker-Martin C, Ory V,
change nephrotic syndrome: Is genetics involved? Pediatr Nephrol Candelier M, Baia M, Lang P, Pawlak A, Sahali D: Occurrence of
20: 1073–1080, 2005 minimal change nephrotic syndrome in classical Hodgkin lymphoma
25. Ruf RG, Fuchshuber A, Karle SM, Lemainque A, Huck K, Wienker is closely related to the induction of c-mip in Hodgkin-Reed Stern-
T, Otto E, Hildebrandt F: Identification of the first gene locus berg cells and podocytes. Blood 115: 3756 –3762, 2010
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Section 14: CASE PRESENTATION 132/88 mmHg and 3⫹ lower extremity edema. Laboratory
values include serum creatinine level of 0.5 mg/dl, 24-hour
A 21-year-old Caucasian woman presents with new onset urine protein of 10.7 g, and serum albumin of 1.7 g/dl.
of lower extremity edema of approximately 3 to 4 weeks’ Urinalysis shows 4⫹ protein with no heme by dipstick and
duration. She had been diagnosed with mild hypertension an inactive urinary sediment. All serologies are negative or
and anemia (hematocrit 28%) 6 months earlier. She is taking normal, including anti-nuclear antibody, hepatitis B surface
no prescription medications and denies using over-the- antigen, hepatitis C antibody, and anti-neutrophil cytoplas-
counter medications. Physical examination reveals BP of mic antibody. A renal biopsy is performed.
Figure 1. Figure 2.
Figure 4.
Figure 3. IgM
Answer
Figure 1 (Jones methenamine silver, ⫻400)
shows a glomerulus with a segmental lesion (arrow) at
the tubular pole, adjacent to the origin of the proximal
tubule. The capillary lumina are filled with endocap-
illary cells that have a foamy cytoplasm, consistent
with a segmental “cellular lesion.” There is swelling of
the overlying podocytes, which are confluent with prox-
imal tubular epithelial cells (arrowhead). The adjacent
glomerular capillaries are unremarkable. Figure 2 (peri-
odic acid–Schiff, ⫻400) shows a different glomerulus
from the same biopsy with a small segmental cellular
lesion that contains endocapillary foam cells, involves Figure 5. A glomerulus shows segmental sclerosis of the
the peripheral tuft, and forms a small adhesion to usual type, with obliteration of glomerular capillary lumina
Bowman’s capsule (arrow). This lesion does not ex- by extracellular matrix and hyaline, loss of overlying podo-
tend to the tubular pole in this section or in adjacent cytes, and broad adhesion to Bowman’s capsule. The scle-
rotic segments demonstrate no relationship to either the
levels. The other glomeruli in the biopsy showed no tubular pole or the vascular pole in the plane of section.
abnormalities by light microscopy. There was no ev- These findings are consistent with FSGS, NOS. Magnifica-
idence of tubular atrophy, interstitial fibrosis, or arte- tion, ⫻400 (periodic acid–Schiff).
riosclerosis. Immunofluorescence microscopy (Figure 3,
⫻400) shows segmental tuft positivity for IgM, con-
sistent with nonspecific trapping, probably in the dis-
tribution of a segmental lesion. Electron microscopy
(Figure 4, ⫻8000) shows diffuse foot process efface-
ment and no immune-type electron-dense deposits.
The biopsy findings support a diagnosis of the tip
variant of focal segmental glomerulosclerosis (FSGS),
also referred to as “glomerular tip lesion” (GTL;
answer D).
tip variant, and collapsing variant. In this schema, the prevalence in a series of 111 biopsies with segmental
tip variant of FSGS is defined by the presence of at lesions in Birmingham, United Kingdom, performed
least one tip lesion (as described above), in the ab- between 1980 and 1989 and a 66% prevalence in a
sence of collapsing lesions or perihilar sclerosis (2). later series (1985 through 2003), in a study population
Thus, the tip variant may contain other glomeruli with whose origin was not stated (10). Interestingly, the tip
segmental lesions of sclerosis that are located in the variant seems to be less common in African Ameri-
peripheral tuft or whose relationship to the vascular cans (13 to 15% of cases) (3–5), despite the higher
and tubular poles is indeterminate. Although mesan- overall frequency of FSGS in this population. How-
gial hypercellularity may be present (2), no mesangial ever, in one study, the majority (55%) of patients with
hypercellularity was reported in the largest series of tip tip lesion were African American (6). The reported
variant (3). frequency of the tip variant in children is low (2%
[11], 6% [12], and 0% [4]), possibly because renal
Clinical Characteristics biopsy is rarely performed in children with steroid-
FSGS, tip variant, typically presents with recent responsive nephrotic syndrome (4,11,12). Indeed, both
onset or short duration of full nephrotic syndrome/ of the pediatric series with cases of the tip variant of
heavy proteinuria. Among North Americans, the tip FSGS were performed in children who had steroid-
variant predominantly affects middle-aged or older resistant nephrotic syndrome and/or an atypical pre-
Caucasians and is rare in children and non-Caucasians sentation (11,12) and who might be expected to have
(3–5). Compared with FSGS NOS, the tip variant a longer duration of symptoms before biopsy and an
shows higher initial proteinuria and shorter duration of innately more aggressive course compared with those
symptoms before biopsy, akin to MCD (3). Both tip who underwent biopsy before starting therapy. Of
variant and collapsing variant show more frequent and note, GTL was identified retrospectively in 10% of
more severe nephrotic syndrome compared with other children who initially received a diagnosis of MCD
FSGS variants; however, tip variant has the best initial (13) and in five of eight autopsies of children dying
and final renal function and the highest rate of com- with nephrosis before 1950 (14). Thus, the tip variant
plete remission and renal survival at 5 years (5). Tip of FSGS may be underrecognized in children with
variant has the least severe chronic tubulointerstitial nephrotic syndrome because of biopsy selection bias.
injury, the least severe arteriosclerosis, and the lowest
frequency of hypertension (5). Although renal func-
Pathology
tion is usually normal at presentation, some patients
develop acute renal failure that resolves with remis- Light Microscopy. GTL is a focal finding, with
sion of nephrotic syndrome, suggesting a role for lesions identified in 12% (3) to 24% (6) of glomeruli
hemodynamic factors associated with severe hy- in adult series. In a review of autopsies of children
poalbuminemia and edema, similar to adult MCD with dying with nephrosis, Haas and Yousefzadeh (14)
acute kidney injury (3). As seen in MCD, some pa- identified GTL in five of eight cases, with 0.3 to 4.4%
tients with FSGS tip variant who achieve full remis- of total glomeruli involved. Although the early de-
sions following steroid therapy subsequently develop scriptions suggested that GTL involved all glomeruli
one or more relapses of nephrotic syndrome. (1,9), in the latest series from Howie et al. (10), only
The incidence of FSGS tip variant is unknown, two of 15 cases had tip lesions in ⬎50% glomeruli,
but the biopsy prevalence among US patients with and none showed 100% glomerular involvement.
FSGS ranges from 13% (6) to 17% (5). In a study of These findings emphasize the need to examine care-
225 patients with primary FSGS that excluded FSGS fully multiple biopsy levels for evidence of tip lesions
perihilar variant, the prevalences of the other variants and underscore the possibility of sampling error in
were tip variant, 26.6%; cellular variant, 9.7%; col- small biopsy specimens. Tip lesions are typically “cel-
lapsing variant, 24.8%; and NOS variant, 38.6% (7). lular” in appearance, with endocapillary foam cell
Deegens et al. (8) reported a 37% prevalence of tip accumulation and swelling and hyperplasia of overly-
variant in a predominantly Caucasian Dutch popula- ing visceral epithelial cells. Less frequently (19% of
tion with FSGS. Howie et al. (9), using the original, FSGS tip variant in one study [3]), the tip lesions have
more restrictive definition of GTL, reported a 9% a “sclerosing” appearance, with accumulation of hya-
356 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
line and acellular matrix material. Both cellular and Clinical Pathologic Correlation
sclerosing tip lesions may coexist in the same biopsy, Some cases of FSGS, tip variant progress to
but all are localized to the peripheral (tubular) pole ESRD, and tip variant may recur in the renal allograft
and demonstrate a relationship to the origin of the (10). Factors that predict poor renal outcome include
proximal tubule. The latter includes synechial attach- absence of remission of proteinuria (6) and greater
ment of the glomerular tuft to Bowman’s capsule, number of non-tip segmental lesions in the initial
prolapse or herniation of the glomerular tuft into the biopsy (3,10). Some studies have shown higher remis-
proximal tubule, and/or confluence of hyperplastic sion rates and less ESRD in FSGS, tip variant, com-
podocytes with proximal tubular cells. The Columbia pared with FSGS NOS (3,5), but others did not show
Classification requires that tip lesions involve ⬍25% any relationship between FSGS variant and outcome
of the glomerular tuft to differentiate lesions that (6,11,12). Some of these differences between studies
may be due to the different histologic criteria used for
originate at the tubular pole from larger lesions that
subclassification of FSGS. In a comparative series of
may have begun elsewhere in the glomerular globe but
patients with FSGS variants at Columbia University
reach the tubular pole only as they evolve.
(7), the percentage of complete and partial remis-
In the Columbia Classification of FSGS, tip vari-
sions was greatest for tip variant (76%), lowest for
ant, may show other segmental lesions in the same
collapsing variant (13%), and intermediate for cel-
biopsy as long as they are not collapsing or perihilar. lular variant (44%), compared with 39% for FSGS
In the only study of tip variant FSGS in which a NOS. There was an inverse relationship between
detailed pathologic analysis of all glomeruli was pro- remission status and development of ESRD across
vided, only 26% of cases showed “pure” tip lesions, subtypes. Thus, the incidence of ESRD was lowest
whereas other (non-tip) segmental lesions were seen in for tip variant (6%), highest for collapsing variant
74% of cases (3). However, no perihilar lesions were (65%), and intermediate for cellular variant (28%)
identified. Most of the non-tip lesions identified were compared with 35% for FSGS NOS. The values for
cellular and were located in the peripheral tuft or had tip variant approach the values for adult MCD,
in indeterminate relationship to the tubular pole and indicating that it has the best outcome of all FSGS
glomerular hilum. Conceivably, some of these lesions subtypes (7). For individual patients with nephrotic
represent tip lesions where the defining relationship to syndrome, remission response (to steroids and/or
the proximal tubule was not seen. Focal global glo- other therapies) remains the best predictor of out-
merulosclerosis may occur, but this is generally mild come in tip and other variants of FSGS (6).
and commensurate with the patient’s age. FSGS tip
variant typically shows no more than mild tubular Etiology and Pathogenesis
atrophy, interstitial fibrosis, and arteriosclerosis. A FSGS, tip variant, can be considered within the
subset shows acute tubular injury, and some display spectrum of primary podocytopathies. The etiology
patchy interstitial edema and mononuclear interstitial and pathogenesis of tip lesion are unknown. Like
inflammatory cell infiltrates. This acute tubular injury MCD and primary FSGS, it is thought to be the result
and interstitial edema are most often identified in of a yet unidentified circulating factor(s) that targets
biopsies from patients presenting with severe ne- the podocyte, leading to foot process effacement and
phrotic syndrome and acute renal failure. loss of filtration barrier (see pathogenesis section for
Case 13). It is likely that podocytes whose attachment
Immunofluorescence and Electron Microscopy. Non- to the glomerular basement membrane has been weak-
specific segmental staining for IgM and C3 is common ened by foot process effacement are more susceptible
in the distribution of the segmental lesions (Figure 3). to mechanical strain at the tubular pole, where there is
Electron microscopy typically shows diffuse foot pro- a convergence of protein-rich ultrafiltrate emanating
cess effacement involving patent capillaries (mean from all portions of the glomerular globe. Because of
93.5%; range 50 to 100%; Figure 4) (3). The podo- its contiguity to the tubular orifice, the tip region is
cytes may exhibit microvillous transformation of their particularly vulnerable to shear stress and turbulence,
cytoplasm. No immune-type electron-dense deposits which could pull the podocyte cell bodies toward the
are identified. tubular lumen, leading to podocyte detachment (15).
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 357
In addition, localized trauma from tuft prolapse into response to therapy of the histologic variants. J Am Soc Nephrol 15:
2169 –2177, 2004
the tubular pole may promote podocyte detachment 7. Stokes MB, Valeri AM, Markowitz GS, D’Agati VD: Cellular focal
and formation of adhesions at this site. In support of segmental glomerulosclerosis: Clinical and pathologic features. Kid-
this hypothesis, tip lesions also have been reported to ney Int 70: 1783–1792, 2006
8. Deegens JK, Steenbergen EJ, Borm GF, Wetzels JF: Pathological
occur in other proteinuric human glomerulopathies variants of focal segmental glomerulosclerosis in an adult Dutch
(e.g., membranous glomerulopathy, IgA nephropathy, population: Epidemiology and outcome. Nephrol Dial Transplant 23:
diabetic nephropathy) (16 –18) and in various experi- 186 –192, 2008
9. Howie AJ, Lee SJ, Green NJ, Newbold KM, Kizaki T, Koram A,
mental models of glomerular proteinuria (19). Richards NT, Michael J, Adu D: Different clinicopathological types
In conclusion, the tip variant of FSGS is a dis- of segmental sclerosing glomerular lesions in adults. Nephrol Dial
Transplant 8: 590 –599, 1993
tinctive pathologic finding that falls within the spec-
10. Howie AJ, Pankhurst T, Sarioglu S, Turhan N, Adu D: Evolution of
trum of primary podocytopathies encompassing MCD nephrotic-associated focal segmental glomerulosclerosis and relation
and FSGS. Most patients have abrupt onset of ne- to the glomerular tip lesion. Kidney Int 67: 987–1001, 2005
11. El-Refaey AM, Bakr A, Hammad A, Elmougy A, El-Houseeny F,
phrotic syndrome and respond to immunosuppressive Abdelrahman A, Sarhan A: Primary focal segmental glomeruloscle-
therapy with preservation of renal function, closely rosis in Egyptian children: A 10-year single-centre experience. Pe-
approximating the presentation and outcome of MCD. diatr Nephrol 25: 1369 –1373, 2010
12. Paik KH, Lee BH, Cho HY, Kang HG, Ha IS, Cheong HI, Jin DK,
In other individuals, tip lesion may be an early man- Moon KC, Choi Y: Primary focal segmental glomerular sclerosis in
ifestation of progressive FSGS. Most investigators children: Clinical course and prognosis. Pediatr Nephrol 22: 389 –
agree that tip variant is the most favorable prognostic 395, 2007
13. Howie AJ, Agarwal A, Sebire NJ, Trompeter RS: Glomerular tip
subgroup of FSGS. changes in childhood minimal change nephropathy. Pediatr Nephrol
23: 1281–1286, 2008
14. Haas M, Yousefzadeh N: Glomerular tip lesion in minimal change
References nephropathy: A study of autopsies before 1950. Am J Kidney Dis 39:
1. Howie AJ, Brewer DB: The glomerular tip lesion: A previously 1168 –1175, 2002
undescribed type of segmental glomerular abnormality. J Pathol 142: 15. Friedrich C, Endlich N, Kriz W, Endlich K: Podocytes are sensitive
205–220, 1984 to fluid shear stress in vitro. Am J Physiol Renal Physiol 291:
2. D’Agati VD, Fogo AB, Bruijn JA, Jennette JC: Pathologic classifi- F856 –F865, 2006
cation of focal segmental glomerulosclerosis: A working proposal. 16. Howie AJ: Changes at the glomerular tip: A feature of membranous
Am J Kidney Dis 43: 368 –382, 2004 nephropathy and other disorders associated with proteinuria. J Pathol
3. Stokes MB, Markowitz GS, Lin J, Valeri AM, D’Agati VD: Glomer- 150: 13–20, 1986
ular tip lesion: A distinct entity within the minimal change disease/ 17. El Karoui K, Hill GS, Karras A, Moulonguet L, Caudwell V,
focal segmental glomerulosclerosis spectrum. Kidney Int 65: 1690 – Loupy A, Bruneval P, Jacquot C, Nochy D: Focal segmental
1702, 2004 glomerulosclerosis plays a major role in the progression of IgA
4. Silverstein DM, Craver R: Presenting features and short-term outcome nephropathy: II. Light microscopic and clinical studies. Kidney Int
according to pathologic variant in childhood primary focal segmental 79: 643– 654, 2011
glomerulosclerosis. Clin J Am Soc Nephrol 2: 700 –707, 2007 18. Najafian B, Kim Y, Crosson JT, Mauer M: Atubular glomeruli and
5. Thomas DB, Franceschini N, Hogan SL, Ten Holder S, Jennette CE, glomerulotubular junction abnormalities in diabetic nephropathy.
Falk RJ, Jennette JC: Clinical and pathologic characteristics of focal J Am Soc Nephrol 14: 908 –917, 2003
segmental glomerulosclerosis pathologic variants. Kidney Int 69: 19. Howie AJ, Kizaki T, Beaman M, Morland CM, Birtwistle RJ, Adu D,
920 –926, 2006 Michael J, Williams AJ, Walls J, Matsuyama M, et al.: Different
6. Chun MJ, Korbet SM, Schwartz MM, Lewis EJ: Focal segmental types of segmental sclerosing glomerular lesions in six experimental
glomerulosclerosis in nephrotic adults: Presentation, prognosis, and models of proteinuria. J Pathol 157: 141–151, 1989
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Section 15: CASE PRESENTATION IU/ml, depressed C3 and C4 complement levels, a positive
IgG anti-cardiolipin antibody, and a negative lupus antico-
A 27-year-old Caucasian woman with a 10-year history agulant. Examination of the urine sediment discloses four to
of systemic lupus erythematosus (SLE) but no previous 10 red blood cells per high-power field, no significant white
history of renal disease presents with new onset of hema- blood cells, and no cellular casts. Previous manifestations of
turia and proteinuria. Laboratory evaluation reveals a serum SLE included pericarditis, alopecia, a malar rash, and ar-
creatinine level of 0.5 mg/dl, 24-hour urine protein of 2.9 g, thralgias. Physical examination reveals a BP of 122/76, an
serum albumin of 2.8 g/dl, white blood cell count of 2.4 ⫻ erythematous facial rash, and no edema. Medications in-
109/L, hematocrit of 36.8%, and platelet count of 127 ⫻ clude methylprednisolone 6 mg/day and plaquenil 200 mg/
109/L. Serologic evaluation reveals an anti-nuclear antibody day. The kidneys measure 12.1 and 11.5 cm in length by
(ANA) titer of ⬎1:1280, an anti-DNA antibody of ⬎500 ultrasound. Renal biopsy is performed.
Figure 1. Figure 2.
Figure 3. Figure 4.
What is the BEST diagnosis? Lupus nephritis (LN) refers to the spectrum of
A. Lupus nephritis (LN) class V immune complex (IC)-mediated renal disease seen in
B. LN class IV-S patients with SLE (1,2). The classification of LN has
gone through multiple revisions since the original
C. LN class IV-S and V
1974 World Health Organization Classification (3).
D. LN class IV-G
The most recent version, referred to as the Interna-
E. LN class IV-G and V tional Society of Nephrology/Renal Pathology Society
(ISN/RPS) 2003 Classification of Lupus Nephritis
Answer (4,5) (Table 1), has gained widespread acceptance and
In Figures 1 and 2, the glomeruli exhibit segmen-
is the approach used in this discussion.
tal endocapillary proliferation with infiltrating neutro-
Renal biopsy plays an important role in the diag-
phils and foci of karyorrhexis. The endocapillary pro-
nosis and treatment of patients with LN. Early in the
liferation has a distinctly segmental distribution,
disease process, biopsy may help to establish the diag-
involving ⬍50% of the glomerular tuft (hematoxylin,
nosis of SLE or the diagnosis of LN in a patient with
⫻400 for both). In Figure 3, the segmental endocap-
known SLE. An initial biopsy is useful to guide therapy.
illary proliferation is accompanied by a segmental
Repeat biopsies at 6-month intervals or longer are often
cellular crescent (Jones methenamine silver [JMS],
performed to assess the response to therapy and guide
⫻400). In contrast to the previous images, the glom-
subsequent treatment. In this section, the findings in LN
erulus in Figure 4 lacks endocapillary proliferation but
classes I through VI are reviewed, followed by a section
exhibits glomerular basement membrane (GBM)
on the clinical manifestations of the various classes. A
thickening with spike formation (JMS, ⫻600). Immu-
briefer discussion focuses on other lupus-related renal
nofluorescence reveals granular global mesangial and
disorders that fall outside the spectrum of LN.
granular to semilinear, segmental to global peripheral
capillary wall positivity for IgG (Figure 5, ⫻400). Pathology
There was similar staining for IgM, IgA, C3, and C1q Mesangial LN. Mesangial LN, which is the mildest
(not shown). Electron microscopy reveals global sub- form of LN, is defined by IC-mediated disease limited
epithelial and segmental subendothelial electron-dense to the mesangium. Mesangial LN is divided into LN
deposits (⫻6000). In the setting of SLE, the findings classes I and II. In class I, also referred to as “minimal
of segmental endocapillary proliferation involving the
majority of glomeruli are diagnostic of LN class IV-S. Table 1. Summary of ISN/RPS (2003) Classification of
The membranous changes, which are best seen on Lupus Nephritis
ultrastructural evaluation, merit an additional diagno-
sis of LN class V. Thus, the final diagnosis is LN class Class I Minimal mesangial LN
Class II Mesangial proliferative LN
IV-S and V (answer C).
Class III Focal LN (⬍50% of glomeruli)
III (A): active
Lupus Nephritis III (A/C): active and chronic
SLE is an autoimmune disease that involves III (C): chronic
multiple organ systems, most notably the kidney. Re- Class IV Diffuse LN (ⱖ50% of glomeruli)
nal disease can be seen at any time in the course of Diffuse segmental LN (IV-S)
SLE, is most frequently encountered during the first IV-S (A): active
year, and is associated with significant morbidity and IV-S (A/C): active and chronic
mortality. The percentage of patients who have SLE IV-S (C): chronic
Diffuse global LN (IV-G)
and develop renal disease is uncertain because many
IV-G (A): active
patients do not undergo renal biopsy and renal in- IV-G (A/C): active and chronic
volvement can be clinically silent. It has been esti- IV-G (C): chronic
mated that ⬎50% of patients with SLE will develop Class V Membranous LN
renal manifestations, which may include hematuria, Class VI Advanced sclerosing LN (ⱖ90% globally
proteinuria, full nephrotic syndrome, and/or renal in- sclerosed glomeruli without residual
sufficiency, at some point in their course. activity)
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 361
Figure 8. In contrast to LN IV-S, LN IV-G has diffuse and Figure 9. A characteristic feature of LN IV is large suben-
global endocapillary proliferation involving ⬎50% and usu- dothelial immune deposits forming “wire loops” and intra-
ally closer to 100% of the glomerular tuft. Magnification, capillary immune aggregates known as “hyaline thrombi.”
⫻400 (H&E). Magnification, ⫻400 (JMS).
362 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
difficult to determine whether a sclerotic glomerulus is these observations, all patients with LN and with
the result of LN or ischemic obsolescence. necrosis and crescent formation out of proportion to
Diffuse LN (LN IV) is further subdivided on the the degree of immune deposition should undergo
basis of whether the endocapillary lesions are predom- ANCA testing, preferably by ELISA because ANA
inantly segmental (S) or global (G) in distribution. The can mimic a perinuclear ANCA pattern by indirect
rationale for this distinction is based on the findings of immunofluorescence, and only ELISA can determine
the Lupus Nephritis Collaborative Study Group (6). In the ANCA specificity. Future studies are needed to
this prospective study, patients who met current crite- address the frequency and significance of ANCA pos-
ria for LN IV-S had a lower remission rate and poorer itivity in patients with LN, particular those with a
renal survival than patients with LN IV-G (6). Patients “pauci-immune” lesion.
with LN IV-S had a greater degree of fibrinoid necro- Renal biopsy reports from patients with LN
sis and less extensive immune deposition, leading the classes III and IV should include an assessment of
authors to propose the possibility of a distinct patho- disease activity and chronicity. The most widely ac-
genesis analogous to pauci-immune glomerulonephri- cepted and reproducible system is the activity and
tis (6). chronicity indices formulated by Austin et al. (17).
Subsequent studies using the ISN/RPS classifi- This system is particularly useful when comparing
cation of LN have failed to establish a difference in repeat biopsies. In general, the chronicity index is
prognosis between LN class IV-S and class IV-G considered to be more predictive of clinical outcome
(7–11). These studies did identify differences in pre- than the activity index (11,18).
senting clinical and pathologic features, however, with Membranous LN. Membranous LN, also referred to
LN class IV-G having a greater degree of proteinuria as LN class V, is characterized by segmental to global
and renal insufficiency and more abundant IC deposi- subepithelial electron-dense deposits. These membra-
tion, whereas LN class IV-S has more fibrinoid necro- nous changes are commonly accompanied by mesangial
sis (7,9). Among five studies that compared outcomes proliferation and deposits. Of note, endocapillary prolif-
in LN class IV-G versus LN class IV-S, one showed a eration should not be seen in LN class V; if present, then
better outcome in LN IV-G (10), one showed a better an additional diagnosis of LN class III or IV is warranted
outcome in LN IV-S (8), and three showed similar depending on the distribution of the endocapillary le-
outcomes between the two groups (7,9,11). Of note, sions. Because scattered subepithelial deposits can also
two of the studies documented transformations be- be found in patients with LN class III and class IV, an
tween LN class IV-S and class IV-G on repeat biopsies additional diagnosis of LN class V requires that subepi-
(7,9), supporting the concept of a similar pathogenesis. thelial deposits involve ⱖ50% of the glomerular tuft of
One possible explanation for why patients with ⱖ50% of glomeruli (4,5).
LN class IV-S exhibit more prominent necrosis and
less abundant immune deposition than patients with Advanced sclerosing LN. Advanced sclerosing
LN class IV-G is the role of anti-neutrophil cytoplas- LN, also referred to as LN class VI, is narrowly
mic antibodies (ANCA) (6,7,9). ANCA seropositivity defined as the presence of ⬎90% of glomeruli with
is seen in approximately 20% of patients with SLE by global sclerosis and no evidence of residual endocap-
indirect immunofluorescence (12) and 10% by ELISA illary proliferation in the remaining glomeruli (4,5). In
(13). Multiple reports have described renal biopsies our experience, ⬍1% of biopsies from patients with
from patients with SLE in which there was significant SLE fall into this category.
necrosis and crescent formation with only rare or Immunofluorescence. All cases of active LN and
absent subendothelial deposits (14 –16). This “pauci- even those with chronic lesions have identifiable im-
immune” picture led to testing for and discovery of mune deposits by immunofluorescence. The standard
ANCA seropositivity, most commonly anti-myeloper- immunofluorescence panel used by renal pathology
oxidase ANCA (14,16). Not surprising, the clinical laboratories includes antisera to three Ig classes—IgG,
presentation of patients with “necrotizing and crescen- IgM, and IgA—and two complement components—C3
tic LN with ANCA seropositivity” is typically one of and C1q. Positivity for all five immune reactants is
rapidly progressive glomerulonephritis, warranting ag- found in the majority of biopsies of LN, a finding
gressive immunosuppressive therapy (16). In light of referred to as “full house” staining. IgG is dominant or
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 363
higher incidence of podocytopathy in SLE than in the in LN, comprise 50% of the chronicity index score (17),
general population and the rapid remission of ne- and have a negative impact on clinical outcome (24).
phrotic syndrome after steroid therapy in all 11 pa-
tients with MCD and three of seven with FSGS sup- Pathogenesis
port the concept of a “lupus podocytopathy” (20,21). LN is a chronic IC-mediated disorder with com-
Lupus podocytopathy is seen most common early in plex pathogenesis. The predominance of SLE in fe-
the course of SLE or after a disease flare. A role for T males and the ability to attenuate the disease in murine
cell dysregulation, heightened cytokine milieu, or in models by hormonal manipulation underscore the im-
some cases nonsteroidal anti-inflammatory use has portance of hormonal factors. A role for genetic fac-
been proposed. Importantly, this entity should be rec- tors is supported by the increased incidence of SLE in
ognized as a highly steroid-responsive lesion (19 –21). first-degree relatives of patients with SLE, in monozy-
gotic than dizygotic twins, and in certain racial groups,
Vascular and tubulointerstitial lesions of SLE. A
such as African-Americans and Asians (25). In addi-
complex spectrum of vascular lesions is seen in pa-
tion, there are associations with certain HLA haplo-
tients with SLE. One of the most important is throm-
types (e.g., DR2, DR3, B8) (26) and inherited com-
botic microangiopathy, which is usually associated
plement deficiencies (e.g., C2 and C4).
with anti-phospholipid antibodies. This topic is cov-
LN is characterized by a broad range of autoan-
ered in Case 11 of this issue of NephSAP.
tibodies, the most pathogenic of which are anti-DNA
Vessel wall immune deposits are the most fre-
antibodies. The deposition of ICs in the glomeruli of
quent vascular lesion in LN and can occur in any class.
patients with LN is thought to involve three major
These deposits are detected by immunofluorescence
mechanisms, none of which are mutually exclusive:
and electron microscopy but do not elicit an inflam- (1) Autoantibodies against circulating antigens that
matory response and usually do not compromise the have become “planted” within the glomerulus, leading
vessel lumen. Thus, they are usually clinically silent. to in situ IC formation; (2) autoantibodies cross-react-
A minority of cases of LN, in particular active class ing with normal glomerular constituents, leading to in
IV, develop massive vascular immune deposits ad- situ IC formation; and (3) passive glomerular deposi-
mixed with fibrin that narrow the vessel lumen and are tion of preformed circulating IC. Many factors influ-
accompanied by endothelial and myocyte degenera- ence glomerular IC deposition, including the class and
tion. Because there is no inflammatory reaction, this subclass of Ig, the electric charge of the Ig, autoanti-
lesion has been termed “lupus vasculopathy” rather body specificity and cross-reactivity with glomerular
than vasculitis (22). It is usually associated with severe constituents, binding of autoantigens to glomerular
renal failure and hypertension. In contrast, the rare constituents through charge interactions, and clearance
entity of true inflammatory vasculitis in SLE, which of immune deposits through FcR interactions.
may be systemic, exhibits transmural, often necrotiz- Nucleosomes, which consist of DNA bound to
ing vessel wall inflammation without significant im- histones, are thought to play an important role in the
mune deposit formation, resembling microscopic development of LN. Patients with SLE have impaired
polyangiitis. Of note, thrombotic microangiopathy, lu- clearance of apoptotic cells, which frequently harbor
pus vasculopathy, and lupus vasculitis are associated nucleosomes on their surface (27). Because of their
with an increased risk for progression to ESRD (23). positive charge, nucleosomes have affinity for the nega-
TBM and interstitial deposits are common in LN, tively charged glomerular capillary wall, where they may
in particular LN class IV (24). The deposits are not become planted followed by in situ IC formation. There
usually visible by light microscopy, although there is is also evidence that anti-DNA antibodies exhibit a broad
often associated interstitial inflammation. Rarely, range of cross-reactivities to normal glomerular constit-
abundant tubulointerstitial immune deposits and se- uents such as mesangial cells, ␣-actinin-4, heparan sul-
vere interstitial inflammation occur in the absence of fate proteoglycans, laminin, and type IV collagen (28).
significant glomerular lesions. This unusual picture has Chromatin fragments generated may stimulate the innate
been referred to as “lupus interstitial nephritis.” In gen- immune system via Toll-like receptors, which are ex-
eral, tubular atrophy and interstitial fibrosis are prognos- pressed on infiltrating leukocytes and indigenous glomer-
tically important markers of chronic irreversible scarring ular cells (29).
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 365
The accumulation of immune deposits within Ueki K, Nojima Y: Revised classification of lupus nephritis is valuable in
predicting renal outcome with an indication of the proportion of glomeruli
glomeruli leads to complement activation and recruit- affected by chronic lesions. Rheumatology 47: 702–707, 2008
ment of monocytes/macrophages and neutrophils. The 12. Sen D, Isenberg DA: Antineutrophil cytoplasmic autoantibodies in
release of inflammatory mediators promotes glomeru- systemic lupus erythematosus. Lupus 12: 651– 658, 2003
13. Galeazzi M, Morozzi G, Sebastiani GD, Bellisai F, Marcolongo R, Cervera
lar cellular proliferation and matrix synthesis. Neutro-
R, De Ramon Garrido E, Fernandez-Nebro A, Houssiau F, Jedryka-Goral A,
phil release is likely to be important in the develop- Mathieu A, Papasteriades C, Piette JC, Scorza R, Smolen J: Anti-neutrophil
ment of endothelial injury, necrotizing lesions, rupture cytoplasmic antibodies in 566 European patients with systemic lupus ery-
of GBM, and crescent formation. The importance of thematosus: Prevalence, clinical associations and correlation with other
autoantibodies. Clin Exp Rheumatol 16: 541–546, 1998
infiltrating macrophages has been demonstrated by 14. Marshall S, Dressler R, D’Agati VD: Membranous lupus nephritis with
elegant studies in NZB/NZW Fc␥R⫺/⫺ mice. Kidneys antineutrophil cytoplasmic antibody-associated segmental necrotizing
from these Fc␥R-deficient lupus mice contain immune and crescentic glomerulonephritis. Am J Kidney Dis 29: 119 –124, 1997
15. Charney DA, Nassar G, Truong L, Nadasdy T: “Pauci-immune”
deposits and fix complement but are protected from proliferative and necrotizing glomerulonephritis with thrombotic mi-
development of proliferative LN (30). Microarray croangiopathy in patients with systemic lupus erythematosus and
analysis of laser-captured glomeruli from patients with lupus-like syndrome. Am J Kidney Dis 35: 1193–1206, 2000
16. Nasr SH, D’Agati VD, Park HR, Sterman PL, Goyzueta JD, Dressler
LN has uncovered overrepresentation of many inter-
RM, Hazlett SM, Pursell RN, Caputo C, Markowitz GS: Necrotizing
esting inflammatory and cellular pathways, including and crescentic lupus nephritis with antineutrophic cytoplasmic anti-
IFN response elements, matrix synthesis, and fibro- body seropositivity. Clin J Am Soc Nephrol 3: 682– 690, 2008
blast and glomerular epithelial cell proliferation (31). 17. Austin HA 3rd, Muenz LR, Joyce KM, Antonovych TT, Balow JE:
Diffuse proliferative lupus nephritis: Identification of specific patho-
logic features affecting renal outcome. Kidney Int 25: 689 – 695, 1984
References 18. Korbet SM, Lewis EJ, Schwartz MM, Reichlin M, Evans J, Rohde
1. Pollak VE, Pirani CL, Schwartz FD: The natural history of the renal RD: Factors predictive of outcome in severe lupus nephritis. Am J
manifestations of systemic lupus erythematosus. J Lab Clin Med 63: Kidney Dis 35: 904 –914, 2000
537–550, 1964 19. Dube GK, Markowitz GS, Radhakrishnan J, Appel GB, D’Agati VD:
2. Baldwin DS, Lowenstein J, Rothfield NJ, Gallo G, McCluskey RT: Minimal change disease in systemic lupus erythematosus. Clin Neph-
The clinical course of proliferative and membranous forms of lupus rol 57: 120 –126, 2002
nephritis. Ann Intern Med 73: 929 –940, 1970 20. Hertig A, Droz D, Lesavre P, Grunfeld JP, Rieu P: SLE and
3. McCluskey RT: Lupus nephritis. In: Kidney Pathology Decennial idiopathic nephrotic syndrome: Coincidence or not? Am J Kidney Dis
1966 –1975, edited by Sommers SC, East Norwalk, CT, Appleton- 40: 1179 –1184, 2002
Century-Crofts, 1975, pp 435– 450 21. Kraft SW, Schwartz MM, Korbet SM, Lewis EJ: Glomerular podo-
4. Weening JJ, D’Agati VD, Schwartz MM, Seshan SV, Alpers CE, cytopathy in patients with systemic lupus erythematosus. J Am Soc
Appel GB, Balow JE, Bruijn JA, Cook T, Ferrario F, Fogo AB, Nephrol 16: 175–179, 2004
Ginzler EM, Hebert L, Hill G, Hill P, Jennette JC, Kong NC, Lesavre 22. Appel GB, Pirani CL, D’Agati VD: Renal vascular complications of
P, Lockshin M, Looi LM, Makino H, Moura LA, Nagata M: The systemic lupus erythematosus. J Am Soc Nephrol 4: 1499 –1515, 1994
classification of glomerulonephritis in systemic lupus erythematosus 23. Banfi G, Bertani T, Boeri V, Faraggiana T, Mazzucco G, Monga G,
revisited. Kidney Int 65: 521–530, 2004 Sacchi G: Renal vascular lesions as a marker of poor prognosis in
5. Weening JJ, D’Agati VD, Schwartz MM, Seshan SV, Alpers CE, patients with lupus nephritis. Am J Kidney Dis 18: 240 –248, 1991
Appel GB, Balow JE, Bruijn JA, Cook T, Ferrario F, Fogo AB, 24. Yu F, Wu LH, Tan Y, Li LH, Wang CI, Wang WK, Qu Z, Chen MH,
Ginzler EM, Hebert L, Hill G, Hill P, Jennette JC, Kong NC, Lesavre Gao JJ, Li ZY, Zheng X, Ao J, Zhu SN, Wang SX, Zhao MH, Zou
P, Lockshin M, Looi LM, Makino H, Moura LA, Nagata M: The WZ, Liu G: Tubulointerstitial lesions of patients with lupus nephritis
classification of glomerulonephritis in systemic lupus erythematosus classified by the 2003 International Society of Nephrology and Renal
revisited. J Am Soc Nephrol 15: 241–250, 2004 Pathology Society system. Kidney Int 77: 820 – 829, 2010
6. Najafi CC, Korbet SM, Lewis EJ, Schwartz MM, Reichlin M, Evans 25. Lawrence JS, Martins CL, Drake GL: A family survey of lupus
J: Significance of histologic patterns of glomerular injury upon erythematosus. J Rheumatol 14: 913–921, 1987
long-term prognosis in severe lupus glomerulonephritis. Kidney Int 26. Reinertsen JL, Klippel JH, Johnson AH, Steinberg AD, Decker JL,
59: 2156 –2163, 2001 Mann DL: B-lymphocyte alloantigens associated with systemic lupus
7. Mittal B, Hurwitz S, Rennke H, Singh AK: New subcategories of erythematosus. N Engl J Med 299: 515–518, 1978
class IV lupus nephritis: Are there clinical, histologic, and outcome 27. Muller S, Dieker J, Tincani A, Meroni PL: Pathogenic anti-nucleo-
differences? Am J Kidney Dis 44: 1050 –1059, 2004 some antibodies. Lupus 17: 431– 436, 2008
8. Yokoyama H, Wada T, Hara A, Yamahana J, Nakaya I, Kobayashi M, 28. Yung S, Chan TM: Anti-DNA antibodies in the pathogenesis of lupus
Kitagawa K, Kokubo S, Iwata Y, Yoshimoto K, Shimizu K, Sakai N, nephritis: The emerging mechanisms. Autoimmun Rev 7: 317–321, 2008
Furuichi K: The outcome and a new ISN/RPS 2003 classification of 29. Mortensen ES, Rekvig OP: Nephritogenic potential of anti-DNA
lupus nephritis in Japanese. Kidney Int 66: 2382–2388, 2004 antibodies against necrotic nucleosomes. J Am Soc Nephrol 20:
9. Hill GS, Delahousse M, Nochy D, Bariety J. Class IV-S versus class 696 –704, 2009
IV-G lupus nephritis: Clinical and morphologic differences suggest- 30. Clynes R, Dumitru C, Ravetch JV: Uncoupling of immune complex
ing different pathogenesis. Kidney Int 68: 2288 –2297, 2005 formation and kidney damage in autoimmune glomerulonephritis.
10. Kim YG, Kim HW, Cho YM, Oh JS, Nah SS, Lee CK, Yoo B: The Science 279: 1052–1054, 1998
difference between lupus nephritis class IV-G and IV-S in Koreans: 31. Peterson KS, Huang JF, Zhu J, D’Agati V, Liu X, Miller N, Erlander
Focus on the response to cyclophosphamide induction treatment. MG, Jackson MR, Winchester RJ: Characterization of heterogeneity in
Rheumatology 47: 311–314, 2008 the molecular pathogenesis of lupus nephritis from transcriptional pro-
11. Hiramatsu N, Kuroiwa T, Ikeuchi H, Maeshima A, Kaneko Y, Hiromura K, files of laser captured glomeruli. J Clin Invest 113: 1722–1733, 2004
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Section 16: CASE PRESENTATION manifestations. The patient has a 24-hour urine protein of
2.6 g, albumin of 2.7 g/dl, hematocrit of 27%, and an
A 37-year-old Caucasian woman presents with hema- active urine sediment with many red blood cells (RBCs)
turia, proteinuria, new-onset hypertension, a 12-lb weight but no RBC casts. Serologic evaluation reveals depressed
gain over 1 week, and oliguric acute kidney injury with a C3 and CH50 complement levels, normal C4, negative
serum creatinine level of 2.0 mg/dl. Past medical history anti-nuclear antibody and anti-neutrophil cytoplasmic an-
is notable for anxiety, depression, joint pains, iron defi- tibody, and no evidence of cryoglobulinemia. The kid-
ciency anemia related to menorrhagia, and tobacco use. neys measure 13.6 and 14.0 cm in length by ultrasound,
Her only medication is escitalopram (Lexapro). Physical without evidence of obstruction. One week later her
examination reveals a BP of 154/94 mmHg, height of creatinine remains elevated at 1.5 mg/dl, and renal biopsy
5⬘6⬙, weight of 197 lb, mild edema, and no cutaneous is performed.
Figure 1. Figure 2.
Figure 4.
Figure 3. C3
100,000 in industrialized nations (6). In additional to ent large global capillary wall deposits with scant or
sporadic cases, there have been multiple reports of absent mesangial deposits. As the name suggests, the
epidemic outbreaks of APIGN in areas of lower so- “mesangial pattern” of APIGN is characterized by a
cioeconomic status with crowded living conditions predominance of mesangial deposits and likely repre-
and poor hygiene (6). sents a stage in the resolution of the starry sky pattern.
This interpretation is supported by the observation that
Pathology the starry sky pattern is typically seen in the initial 2
weeks, whereas the mesangial pattern occurs later (8).
Light Microscopy. At the light microscopic level,
With respect to the composition of the deposits, stain-
APIGN most commonly manifests as a diffuse endo-
ing is typically positive for C3 and IgG. Staining for
capillary proliferative glomerulonephritis (DPGN;
C3 is usually of equal or greater intensity than IgG;
Figure 1). This pattern of glomerulonephritis, which is
this dominant or co-dominant staining for C3 is a
seen in ⬎70% of cases (1,2,4,7), is characterized by
highly characteristic feature of APIGN that is seen in
global mesangial and endocapillary proliferation with
most cases, including all 82 cases recently evaluated at
infiltrating leukocytes. The DPGN often has an exu-
our center (2). As expected for a polyclonal immune
dative appearance, a term denoting the distinctive
response, cases with positive staining for IgG usually
presence of numerous intraglomerular neutrophils
have a similar intensity of staining for and light
(Figures 1 and 2). In severe cases, the proliferative
changes may be accompanied by cellular crescents. chains. Low-intensity staining for IgM, IgA, and/or
Trichrome stain may demonstrate large subepithelial C1q is seen in fewer than half of cases. Staining for C3
fuchsinophilic deposits, which correspond to the typically persists longer than that for IgG. This ex-
hump-shaped deposits seen ultrastructurally. Over plains why isolated C3 positivity is frequently encoun-
time, the proliferative changes in APIGN begin to tered in the mesangial pattern, which is thought to
regress toward a mesangial pattern of glomerulone- represent a resolving phase (8).
phritis. Thus, the light microscopic finding of purely Electron Microscopy. The most specific and dis-
mesangial proliferation is seen more often in biopsies tinctive pathologic finding in APIGN is subepithelial
performed at a longer interval after infection and hump-shaped electron-dense deposits (Figure 4).
frequently represents an intermediate stage in the res- These hump-shaped deposits are often large and typ-
olution of APIGN. APIGN may also manifest as a ically appear confluent with the garland pattern of
focal endocapillary proliferative glomerulonephritis, a immunofluorescence, somewhat less prominent with
term that is defined by endocapillary proliferation in the starry sky pattern, and infrequent to absent with the
⬍50% of glomeruli. Membranoproliferative features mesangial pattern. Importantly, the subepithelial de-
(i.e., glomerular basement membrane [GBM] duplica- posits are frequently accompanied by less conspicuous
tion with mesangial interposition), large intracapillary mesangial and subendothelial deposits and do not
deposits, and fibrinoid necrosis are not typically seen elicit the spike formation seen in membranous ne-
in classic post-streptococcal APIGN. Tubulointersti- phropathy.
tial changes frequently accompany APIGN, including
acute tubular injury, RBC casts, interstitial edema, and
inflammation. If the glomerulonephritis fails to re- Clinical and Pathologic Criteria to Diagnose
solve, then chronic changes of segmental and global APIGN
glomerular scarring, fibrous crescents, tubular atro- In contrast to most forms of DPGN, which are
phy, and interstitial fibrosis may develop. treated with immunomodulatory therapy, the most
common approach to APIGN is antibiotic therapy and
Immunofluorescence. Three patterns of immuno- supportive measures. While it is critical to distinguish
fluorescence positivity have been described in patients APIGN from other forms of glomerulonephritis for
with APIGN (8). The “starry sky pattern,” which is therapeutic purposes, a definitive diagnosis may be
most frequently encountered, is characterized by difficult in some cases lacking the full constellation of
finely granular deposits distributed in peripheral cap- clinical and pathologic findings. Our diagnostic ap-
illary walls and the mesangium (Figure 3). In contrast, proach is to require at least three of the following five
the “garland pattern” is characterized by more conflu- clinical and pathologic findings to establish a diag-
370 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
rheumatoid factor, and some have anti-neutrophil cy- are consistent with a more complex scenario in which
toplasmic antibody seropositivity. bacterial antigens, both circulating and planted at the
subepithelial and subendothelial aspects of the GBM,
Pathogenesis directly activate both the alternative complement path-
A brief overview of the complement system way (19) and the MBL pathway (20).
provides a framework for understanding the pathogen- The putative pathogenic antigen in most cases of
esis of APIGN. The complement system is a major APSGN is streptococcal pyrogenic exotoxin B (SPEB)
component of the innate, nonadaptive immune re- and its zymogen precursor (21–23). Circulating anti-
sponse and is mediated by a series of sequential bodies to SPEB can be identified in the overwhelming
enzymatic reactions that also greatly enhance, or majority of patients infected by nephritogenic group A
“complement,” the adaptive (or humoral) immune re- streptococci, including all 53 patients tested in one
sponse (18). Activation of the complement system study (21), and antibody levels correlate better with
leads to the formation of C3b from C3, which binds to infection than the more commonly available serum
the target of the immune response (i.e., bacteria) and markers such as anti–streptolysin-O or anti-DNAse B
promotes phagocytosis. The generation of chemotactin (24). SPEB has been localized to the glomerular de-
C5a recruits inflammatory cells to sites of inflamma- posits in APSGN and, more specifically, to the sub-
tion. The generation of C5b-9 can destroy target cells epithelial humps, where it co-localizes with C3 (21).
via formation of the membrane attack complex. The ability of SPEB to reach the subepithelial aspect
There are three distinct pathways of complement of the GBM is favored by its highly cationic charge,
activation (18). The classical pathway is activated by which would facilitate binding to anionic sites in the
the formation of antigen–antibody immune com- glomerular capillary wall. However, it is probably the
plexes, particularly those containing IgG of the IgG1 activation of complement on the subendothelial and
and IgG3 subtypes. In the classical pathway, the for- mesangial deposits, which are in contact with the
mation of C4b2b serves as C3 convertase. The man- circulation, that drives the influx of inflammatory cells
nose-binding lectin (MBL) pathway is activated inde- essential for the initiation of glomerulonephritis.
pendent of antibody by the binding of circulating An animal model of APSGN has been produced
lectins to polysaccharide residues on the surface of in mice by the passive transfer of anti-SPEB antibod-
bacteria, which in turn activates MBL-associated ser- ies, leading to acute glomerulonephritis, acute renal
ine proteases that activate complement components C2 failure, and proteinuria (25). A subsequent study by
and C4, leading to formation of C4b2b as C3 conver- the same group showed that anti-SPEB antibodies in
tase. The alternative complement pathway functions APSGN cross-react with human endothelial cells via
independent of antibodies, lectins, and the C4b2b C3 molecular mimicry and exhibit properties of autoanti-
convertase. It is continuously active at low levels as a bodies (26). Importantly, SPEB does not play a central
result of spontaneous ongoing C3 hydrolysis, binding role in all forms of APSGN, as evidenced by the fact
to factor B, and cleavage by factor D, leading to the that anti-SPEB antibodies were not detectable in a
formation of C3bBb as C3 convertase. C3bBb activa- recent Brazilian outbreak of APSGN associated with
tion is well controlled on human cells but is signifi- Streptococcus zooepidemicus (27). Thus, additional
cantly amplified on foreign cells, such that bacteria are nephritogenic antigens remain to be identified.
potent stimulators of the alternative pathway.
Multiple lines of evidence suggest that APIGN is References
not mediated exclusively by glomerular deposition of 1. Nasr SH, Fidler ME, Valeri AM, Cornell LD, Sethi S, Zoller A,
Stokes MB, Markowitz GS, D’Agati VD: Postinfectious glomerulo-
preformed circulating immune complexes, leading to nephritis in the elderly. J Am Soc Nephrol 22: 187–195, 2011
classical pathway complement activation. First, C3 2. Nasr SH, Markowitz GS, Stokes MB, Said SM, Valeri AM, D’Agati
staining by immunofluorescence is typically of greater VD: Acute postinfectious glomerulonephritis in the modern era.
Medicine (Baltimore) 87: 21–32, 2008
intensity than IgG, and, in some cases, staining for 3. Keller CK, Andrassy K, Waldherr R, Ritz E: Postinfectious glomer-
Ig is undetectable. Second, while hypocomple- ulonephritis: Is there a link to alcoholism? Q J Med 87: 97–102, 1994
mentemia is commonly encountered in APIGN, de- 4. Montseny JJ, Meyrier A, Kleinknecht D, Callard P: The current
spectrum of infectious glomerulonephritis: Experience with 76 pa-
pression of C3 is more common and typically more tients and review of the literature. Medicine (Baltimore) 74: 63–73,
profound than depression of C4. These observations 1995
372 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
5. Carapetis JR, Steer AC, Mulholland EK, Weber M: The global 17. Fukuda M, Motokawa M, Usami T, Oikawa T, Morozumi K, Yoshida
burden of group A streptococcal diseases. Lancet Infect Dis 5: A, Kimura G: PR3-ANCA-positive crescentic necrotizing glomeru-
685– 694, 2005 lonephritis accompanied by isolated pulmonic valve infective endo-
6. Rodriguez-Iturbe B, Musser JM: The current state of poststreptococ- carditis, with reference to previous reports of renal pathology. Clin
cal glomerulonephritis. J Am Soc Nephrol 19: 1855–1864, 2008 Nephrol 66: 202–209, 2006
7. Moroni G, Pozzi C, Quaglini S, Segagni S, Banfi G, Baroli A, Picardi 18. Ricklin D, Hajishengallis G, Yang K, Lambris JD: Complement: A
L, Colzani S, Simonini P, Mihatsch MJ, Ponticelli C: Long-term key stem for immune surveillance and homeostasis. Nat Immunol 11:
prognosis of diffuse proliferative glomerulonephritis associated with 785–797, 2010
infection in adults. Nephrol Dial Transplant 17: 1204 –1211, 2002 19. Westberg NG, Naff GB, Boyer JT, Michael AF: Glomerular deposi-
8. Sorger K, Gessler U, Hubner FK, Kohler H, Schulz W, Stuhlinger W, tion of properdin in acute and chronic glomerulonephritis with
Thoenes GH, Thoenes W: Subtypes of acute postinfectious glomer- hypocomplementemia. J Clin Invest 50: 642– 649, 1971
ulonephritis: Synopsis of clinical and pathological features. Clin 20. Ohsawa I, Ohi H, Endo M, Fujita T, Matsushita M, Fujita T:
Nephrol 17: 114 –128, 1982 Evidence of lectin complement pathway activation in poststreptococ-
9. Mazzucco G, Bertani T, Fortunato M, Bernardi M, Leutner M, cal glomerulonephritis. Kidney Int 56: 1158 –1160, 1999
Boldorini R, Monga G: Different patterns of renal damage in type 2 21. Batsford SR, Mezzano S, Mihatsch M, Schiltz E, Rodriguez-Iturbe B:
diabetes mellitus: A multicentric study on 393 biopsies. Am J Kidney Is the nephritogenic antigen in post-streptococcal glomerulonephritis
Dis 39: 713–720, 2002 pyrogenic exotoxin B (SPE B) or GAPDH? Kidney Int 68: 1120 –
10. Nasr SH, Markowitz GS, Whelan JD, Albanese JJ, Rosen RM, Fein 1129, 2005
DA, Kim SS, D’Agati VD: IgA-dominant acute poststaphylococcal 22. Rodriguez-Iturbe B, Batsford S: Pathogenesis of poststreptococcal
glomerulonephritis complicating diabetic nephropathy. Hum Pathol glomerulonephritis a century after Clemens von Pirquet. Kidney Int
34: 1235–1241, 2003 71: 1094 –1104, 2007
11. Satoskar AA, Nadasdy G, Plaza JA, Sedmak D, Shidham G, Hebert 23. Kanjanabuch T, Kittikowit W: Elam-Ong S: An update on acute
L, Nadasdy T: Staphylococcus infection-associated glomerulonephri- postinfectious glomerulonephritis worldwide. Nat Rev Nephrol 5:
tis mimicking IgA nephropathy. Clin J Am Soc Nephrol 1: 1179 – 259 –269, 2009
1186, 2006 24. Parra G, Rodriguez-Iturbe B, Batsford S, Vogt A, Mezzano S,
12. Haas M, Racusen LC, Bagnasco SM: IgA-dominant postinfectious Olavarria F, Exeni R, Laso M, Orta N: Antibody to streptococcal
glomerulonephritis: A report of 13 cases with common ultrastructural zymogen in the serum of patients with acute glomerulonephritis: A
features. Hum Pathol 39: 1309 –1316, 2008 multicentric study. Kidney Int 54: 509 –517, 1998
13. Arze RS, Rashid H, Morley R, Ward MK, Kerr DN: Shunt nephritis: 25. Luo YH, Kuo CF, Huang KJ, Wu JJ, Lei HY, Lin MT, Chuang WJ,
Report of two cases and review of the literature. Clin Nephrol 19: Liu CC, Lin CF, Lin YS: Streptococcal pyrogenic exotoxin B
48 –53, 1983 antibodies in a mouse model of glomerulonephritis. Kidney Int 72:
14. Haffner D, Schindera F, Aschoff A, Matthias S, Waldherr R, Scharer 716 –724, 2007
K: The clinical spectrum of shunt nephritis. Nephrol Dial Transplant 26. Luo YH, Chuang WJ, Wu JJ, Lin MT, Liu CC, Lin PY, Roan JN,
12: 1143–1148, 1997 Wong TW, Chen YL, Lin YS: Molecular mimicry between strepto-
15. Kiryluk K, Preddie D, D’Agati VD, Isom R: A young man with coccal pyrogenic exotoxin B and endothelial cells. Lab Invest 90:
Propionibacterium acnes-induced shunt nephritis. Kidney Int 73: 1492–1506: 2010
1434 –1440, 2008 27. Beres SB, Sesso R, Pinto SW, Hoe NP, Porcella SF, Deleo FR,
16. Majumdar A, Chowdhary S, Ferreira MA, Hammond LA, Howie AJ, Musser JM: Genome sequence of a Lancefield group C Streptococcus
Lipkin GW, Littler WA: Renal pathological findings in infective zooepidemicus strain causing epidemic nephritis: New information
endocarditis. Nephrol Dial Transplant 15: 1782–1787, 2000 about an old disease. PLoS One 3: e3026, 2008
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Section 17: CASE PRESENTATION significant only for chronic back pain. There is no history of
hypertension, diabetes, obesity, or tobacco use. The patient
A 62-year-old Caucasian man presents with a 2-week his- takes no prescription medications or nonsteroidal anti-inflam-
tory of low-grade fever, myalgias, cough, and decreased oral matory agents on an ongoing basis. Physical examination
intake, more recently accompanied by oliguria and a single reveals a BP of 146/79 mmHg and no edema or cutaneous
episode of gross hematuria. The patient was seen 5 days earlier manifestations. Chest x-ray is unremarkable. Urinalysis reveals
and given a course of ciprofloxacin, without symptomatic 2⫹ protein with too numerous to count red blood cells (RBCs)
improvement. Laboratory evaluation reveals acute kidney in- and rare white blood cells. Serologic results include a negative
jury with a creatinine level of 8.6 mg/dl, albumin 2.5 g/dl, anti-nuclear antibody and normal C3 and C4 complement
cholesterol 113 mg/dl, hematocrit 31%, white blood cell count levels. Additional serologies are pending at the time of renal
11,000, and platelet count 452,000. Past medical history is biopsy.
Figure 1. Figure 2.
Figure 4. Fibrin
Figure 3. IgG
centic glomerulonephritis (defined as ⬎50% of glom- scale of 0 to 4). Low-level linear staining for IgG, by
eruli containing crescents) is the most characteristic contrast, may be a nonspecific finding, especially in
and frequent histologic feature, affecting ⬎80% of older patients, allografts, and autopsy specimens. The
patients at the time of biopsy (7). Crescents tend to be clinical context must also be taken into account be-
of similar age and range from segmental to circumfer- cause patients with diabetes, obesity, or history of
ential. Isolated segmental fibrinoid necrosis of the cigarette smoking may have thickened GBMs with
glomerular tuft without crescents may be observed in low-intensity linear staining for IgG (usually not
biopsies performed early in the disease course (1). The ⬎1⫹) in the absence of crescentic glomerulonephri-
ruptured GBM is readily identified with periodic acid– tis. In equivocal cases, correlation with anti-GBM
Schiff or JMS stains, and the extravasated fibrin serology is needed. In the presence of pulmonary
within the urinary space and crescent stains bright disease, similar linear alveolar basement membrane
orange-red (fuchsinophilic) with trichrome stain. Mild staining for IgG can be demonstrated on lung biopsy,
glomerular neutrophil infiltration is often seen adja- but the immunofluorescence staining is more easily
cent to foci of glomerular necrosis. Glomerular tufts interpretable on a kidney biopsy. Distal tubular base-
unaffected by necrosis or crescents tend to be normal ment membranes occasionally have linear IgG staining
by light microscopy, with no evidence of mesangial or as well because they carry the same collagen IV
endocapillary proliferation. The cellular crescents of- isotypes as GBM, and this may account in part for the
ten have extensive destruction of Bowman’s capsule, associated severe tubulointerstitial inflammation. C3
merging with the adjacent interstitium. Periglomerular staining is typically more granular to semilinear and
inflammation and rare multinucleated giant cells are discontinuous. Rare patients have isolated linear GBM
likely a response to severe glomerular injury and may staining for IgA, rather than IgG, and the circulating
be identified in the absence of associated ANCA IgA anti-GBM antibodies have been shown to target
seropositivity. Severe tubulointerstitial inflammation, novel GBM antigenic determinants (8 –10).
interstitial edema, and acute tubular injury are fre- Electron Microscopy. In the acute phase, cellular
quently present, as are RBC casts. Vasculitis is not a crescents are associated with ruptures of the GBM,
feature of anti-GBM nephritis unless the patient has intracapillary fibrin, and extravasation of fibrin into
concurrent ANCA. The chronic phase of anti-GBM the urinary space. Fibrin is often intermingled with the
nephritis, as seen in late biopsies or more subacute cells of the crescent. There may be widening of the
presentation, often exhibits older fibrocellular/fibrous subendothelial zone by lucent material and glomerular
crescents and variable tubular atrophy and interstitial capillary infiltration by neutrophils or mononuclear
fibrosis. Even in the setting of extensive segmental leukocytes. Importantly, no granular immune-type
and global glomerulosclerosis with subcapsular fibro- electron-dense deposits are identified. This absence of
sis and no residual activity, identification of GBM and deposits is probably due to the uniform distribution of
Bowman’s capsule ruptures with periodic acid–Schiff the anti-GBM antibody, which does not aggregate to
or JMS stains should signal a previous necrotizing and form definable electron densities. Neutrophils and
crescentic glomerulonephritis as the initial process. RBCs gain entry into Bowman’s space through the
Immunofluorescence. In the context of a crescentic broad gaps in the GBM. Glomeruli with more chronic
glomerulonephritis, the finding of intense linear stain- features have progressive obliteration of the tuft by
ing of the GBM for IgG is diagnostic of anti-GBM extracellular matrix, leading to segmental and global
nephritis. The linear IgG staining may appear inter- glomerulosclerosis in association with fibrocellular or
rupted as a result of broad ruptures of the GBMs. In fibrous crescents. The ultrastructural findings are sim-
glomeruli with exuberant crescents and extensive de- ilar to those seen in ANCA-associated glomerulone-
struction of the tuft, it may be difficult to identify phritis, despite the different immunofluorescence find-
linear staining because no residual glomerular capil- ings.
laries are represented in the plane of section. Foci of Concurrent Disease. Up to one third of patients
necrosis and cellular crescents are usually highlighted with anti-GBM nephritis have concurrent circulating
with antisera to fibrin/fibrinogen. Most cases of anti- ANCA, usually with specificity for myeloperoxidase.
GBM nephritis have intense IgG staining (ⱖ2⫹ on a They can be detected either before or after the appear-
376 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
ance of anti-GBM antibodies. Some of these patients culating autoantibody is a mainstay of therapy, and
have arteritis or medullary capillaritis on renal biopsy, immunosuppressive therapy with corticosteroids and
and there may be other manifestations of systemic cyclophosphamide is aimed at reduction in inflamma-
vasculitis. Previous studies suggested that patients tion and antibody synthesis. T cells and macrophages
with concurrent anti-GBM nephritis and ANCA have may also play an important role in antigen recognition,
less severe disease and better renal and patient survival cell-mediated immunity, and exacerbation of inflam-
(even though patients tend to be older) compared with mation (1,18). Activated neutrophils and monocytes
patients with pure anti-GBM nephritis (11,12). Con- recruited by antibody binding and complement fixa-
versely, a more recent series demonstrated that such tion are thought to cause GBM perforations by release
“dual-positive” patients have a renal prognosis that is of reactive oxygen species and lytic proteases. The
similar to patients with isolated anti-GBM nephritis extravasation of fibrin and other growth factors into
(13). Relapse rates, however, are higher in the dual- Bowman’s space elicits the final pathway of crescent
positive group, similar to patients with isolated ANCA formation and destruction of the glomerulus.
disease. Given the therapeutic and long-term implica- The factors that trigger the autoimmune re-
tions of concurrent disease, it is recommended that all sponse include genetic predisposition and exposure
patients with anti-GBM nephritis be tested for ANCA. to environmental factors. There is a strong associa-
There are infrequent reports of concurrent anti- tion with major histocompatibility antigens DR (es-
GBM nephritis and membranous nephropathy (MGN). Ac- pecially DR2, carried by 85% of patients with anti-
curate diagnosis in such cases requires examination of GBM nephritis) and DQ (19). Environmental
immunofluorescence on high magnification to identify exposure to hydrocarbons before the onset of clin-
a layer of finely granular capillary wall staining for ical disease has been documented in several cases.
IgG above the linear GBM positivity for IgG. Ultra- Cigarette smoking and respiratory viral infections
structural identification of subepithelial membranous also seem to trigger autoimmunity and precipitate
deposits is needed for confirmation of coexistent dis- pulmonary manifestations.
ease. In these cases, MGN may precede or follow The target antigen for circulating anti-GBM an-
anti-GBM nephritis, or the two may develop simulta- tibodies is the NC1 domain of type IV collagen net-
neously. Although the concurrence of these two enti- works within the GBM. The same collagen networks
ties may be purely coincidental, experimental data are found within the basement membranes of pulmo-
based on animal models suggest a possible pathoge- nary alveolar capillaries. The elegant work of Ped-
netic link, possibly through epitope spreading (14). chenko et al. (20) elucidated the molecular architec-
The immunologic injury that is caused by anti-GBM ture of the “Goodpasture antigen.” The disease is now
disease or MGN may increase autoantigen synthesis understood to be an autoimmune “conformeropathy”
and exposure of potentially immunogenic GBM in which autoantibodies form to a conformational
epitopes that promote the development of a dual au- alteration in the ␣-3 and ␣-5 subunits of the NC1
toimmune glomerulopathy (15,16). It is unknown domain of collagen IV (20). Patients with Goodpasture
whether the specificity of the antibodies deposited in syndrome and anti-GBM nephritis form antibodies to
the subepithelial region is to phospholipase A2 recep- distinct epitopes encompassing the region EA and EB
tor, as seen in primary MGN, or to another antigenic of the ␣-3 NC1 monomer and region EA of the ␣-5
determinant. NC1 monomer. Interestingly, antibodies do not bind to
the native cross-linked ␣345NC1 hexamer, indicating
Pathogenesis that the target antigens must undergo a conformational
Anti-GBM nephritis (and Goodpasture syn- change to become autoantigens and to elicit an auto-
drome) is caused by development of autoimmunity to immune response (20,21).
a component of GBM. The pathogenic role of anti- Normally, the ␣-3, -4, and -5 chains of type IV
GBM antibodies was first demonstrated by Lerner et collagen assemble to form triple helical protomers,
al. in 1967 (17). There is a correlation between the which in turn dimerize by association through their
antibody titers and severity of disease, and persistently C-terminal NC1 domains to form interface hexamers
high levels are associated with immediate recurrence (22). The cross-linked hexamer is reinforced by sul-
in kidney transplants. Plasmapharesis to remove cir- filimine bonds and needs to be dissociated for autoan-
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 377
tibody binding to occur. A triggering event may alter 7. Johnson JP, Moore J Jr, Austin HA 3rd, Balow JE, Antonovych TT,
Wilson CB: Therapy of anti-glomerular basement membrane anti-
the quartenary structure of the ␣345NC1 hexamer, body disease: Analysis of prognostic significance of clinical, patho-
exposing neo-epitopes and eliciting autoantibody pro- logic and treatment factors. Medicine (Baltimore) 64: 219 –227, 1985
duction and binding. Putative triggers include enzy- 8. Ho J, Gibson IW, Zacharias J, Fervenza F, Colon S, Borza DB:
Antigenic heterogeneity of IgA anti-GBM disease: New renal targets
matic and nonenzymatic posttranslational modifica- of IgA autoantibodies. Am J Kidney Dis 52: 761–765, 2008
tions (e.g., oxidation, nitrosylation, glycation), rise in 9. Border WA, Baehler RW, Bhathena D, Glassock RJ: IgA antibase-
body temperature, and proteolytic cleavage (20). It has ment membrane nephritis with pulmonary hemorrhage. Ann Intern
Med 91: 21–25, 1979
been proposed that environmental exposure to infec-
10. Shaer AJ, Stewart LR, Cheek DE, Hurray D, Self SE: IgA antiglo-
tious agents, cigarette smoke, or organic solvents may merular basement membrane nephritis associated with Crohn’s dis-
inhibit the enzymes that catalyze the sulfilimine bond ease: A case report and review of glomerulonephritis in inflammatory
bowel disease. Am J Kidney Dis 41: 1097–1109, 2003
formation and promote hexamer dissociation (20,21).
11. Clyne S, Frederick C, Arndt F, Lewis J, Fogo AB: Concurrent and
Approximately 5 to 15% of patients with ESRD discrete clinicopathological presentations of Wegener granulomatosis
from Alport syndrome develop anti-GBM nephritis and anti-glomerular basement membrane disease. Am J Kidney Dis
after transplantation. Because of a heritable mutation 54: 1116 –1120, 2009
12. Bosch X, Mirapeix E, Font J, Borrellas X, Rodriguez R, Lopez-Soto
in ␣-5 collagen IV, these patients lack normal ␣345 A, Ingelmo M, Revert L: Prognostic implication of anti-neutrophil
hexamers in their native GBM and develop an immune cytoplasmic autoantibodies with myeloperoxidase specificity in anti-
response to normally expressed GBM antigens pre- glomerular basement membrane disease. Clin Nephrol 36: 107–113,
1991
sented in the renal allograft. Although the clinical and 13. Rutgers A, Slot M, van Paassen P, van Breda Vriesman P, Heeringa
pathologic features are similar to those of native anti- P, Tervaert JW: Coexistence of anti-glomerular basement membrane
GBM nephritis, the alloantibodies generated in post- antibodies and myeloperoxidase-ANCAs in crescentic glomerulone-
phritis. Am J Kidney Dis 46: 253–262, 2005
transplantation nephritis bind to the EA region of ␣-5 14. Fukatsu A, Brentjens JR, Killen PD, Kleinman HK, Martin GR,
NC1 of the intact hexamer, not to ␣-3 NC1. Unlike the Andres GA: Studies on the formation of glomerular immune deposits
situation in native anti-GBM nephritis, autoantibody in brown Norway rats injected with mercuric chloride. Clin Immunol
Immunopathol 45: 35– 47, 1987
binding is lessened upon dissociation of the hexamer, 15. Nasr SH, Ilamathi ME, Markowitz GS, D’Agati VD: A dual pattern
suggesting that the antibody binds to a surface epitope. of immunofluorescence positivity. Am J Kidney Dis 42: 419 – 426,
Thus, posttransplantation anti-GBM nephritis can oc- 2003
16. Troxell ML, Saxena AB, Kambham N: Concurrent anti-glomerular
cur in the absence of triggering events for hexamer basement membrane disease and membranous glomerulonephritis: A
dissociation (20). case report and literature review. Clin Nephrol 66: 120 –127, 2006
17. Lerner RA, Glassock RJ, Dixon FJ: The role of anti-glomerular
References basement membrane antibody in the pathogenesis of human glomer-
1. Jennette JC: Rapidly progressive crescentic glomerulonephritis. Kid- ulonephritis. J Exp Med 126: 989 –1004, 1967
ney Int 63: 1164 –1177, 2003 18. Bolton WK, Innes DJ Jr, Sturgill BC, Kaiser DL: T-cells and
2. Bolton WK: Goodpasture’s syndrome. Kidney Int 50: 1753–1766, macrophages in rapidly progressive glomerulonephritis: Clinicopath-
1996 ologic correlations. Kidney Int 32: 869 – 876, 1987
3. Cui Z, Zhao MH, Singh AK, Wang HY: Antiglomerular basement 19. Huey B, McCormick K, Capper J, Ratliff C, Colombe BW, Garovoy
membrane disease with normal renal function. Kidney Int 72: 1403– MR, Wilson CB: Associations of HLA-DR and HLA-DQ types with
1408, 2007 anti-GBM nephritis by sequence-specific oligonucleotide probe hy-
4. Saxena R, Bygren P, Cederholm B, Wieslander J: Circulating anti- bridization. Kidney Int 44: 307–312, 1993
entactin antibodies in patients with glomerulonephritis. Kidney Int 20. Pedchenko V, Bondar O, Fogo AB, Vanacore R, Voziyan P, Kitching
39: 996 –1004, 1991 AR, Wieslander J, Kashtan C, Borza DB, Neilson EG, Wilson CB,
5. Saxena R, Bygren P, Rasmussen N, Wieslander J: Circulating auto- Hudson BG: Molecular architecture of the Goodpasture autoantigen
antibodies in patients with extracapillary glomerulonephritis. Nephrol in anti-GBM nephritis. N Engl J Med 363: 343–354, 2010
Dial Transplant 6: 389 –397, 1991 21. Lionaki S, Jennette JC, Falk RJ: Anti-neutrophil cytoplasmic
6. Salama AD, Dougan T, Levy JB, Cook HT, Morgan SH, Naudeer S, (ANCA) and anti-glomerular basement membrane (GBM) autoanti-
Maidment G, George AJ, Evans D, Lightstone L, Pusey CD: Good- bodies in necrotizing and crescentic glomerulonephritis. Semin Im-
pasture’s disease in the absence of circulating anti-glomerular base- munopathol 29: 459 – 474, 2007
ment membrane antibodies as detected by standard techniques. Am J 22. Salant DJ: Goodpasture’s disease: New secrets revealed. N Engl
Kidney Dis 39: 1162–1167, 2002 J Med 363: 388 –391, 2010
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Section 18: CASE PRESENTATION cytoplasmic antibodies (P-ANCA; 1:320) with specificity
for anti-myeloperoxidase (MPO; 971 AU/ml; normal range
A 47-year-old man with history of psoriasis presents with 0 to 19). Other serologies are negative or normal, including
cough, headache, chills, and a petechial rash of the lower anti-nuclear antibody, anti– double stranded DNA antibody,
extremities. Laboratory evaluation reveals acute renal fail- hepatitis B surface antigen, hepatitis C antibody, and serum
ure with serum creatinine level of 4.8 mg/dl (increased from complement C3 and C4 levels. Magnetic resonance imaging
baseline value of 0.9 mg/dl 2 months earlier). Urinalysis reveals leptomeningeal enhancement, and chest x-ray shows
reveals microhematuria with red blood cell (RBC) casts and increased interstitial markings. Skin biopsy of the leg re-
3⫹ proteinuria; 24-hour urine protein excretion is 2.0 g. veals a leukocytoclastic vasculitis. A renal biopsy is
Serologic studies reveal positive perinuclear anti-neutrophil performed.
Figure 1. Figure 2.
Figure 3. Figure 4.
What is the BEST diagnosis? segmental glomerular fibrinoid necrosis and crescent
A. Henoch-Schönlein purpura nephritis formation, without significant immune deposits by
B. Goodpasture syndrome immunofluorescence or electron microscopy. More
C. Diffuse proliferative lupus nephritis than 85% of pauci-immune GN cases are associated
D. Cryoglobulinemic glomerulonephritis (GN) with serum ANCA, in which case the term ANCA-
E. Pauci-immune necrotizing and crescentic GN associated GN is used synonymously. This entity rep-
resents small-vessel vasculitis that may be either lim-
Answer ited to the kidney (renal-limited pauci-immune GN) or
The renal biopsy findings are most consistent associated with a systemic disease such as Wegener
with ANCA-associated pauci-immune necrotizing and granulomatosis (more recently referred to as “Granu-
crescentic GN with necrotizing arteritis (answer E). Of lomatosis with Polyangiitis (Wegner’s)”), microscopic
the 26 glomeruli sampled for light microscopy, five polyangiitis, or Churg-Strauss syndrome. All these
are globally sclerotic with evidence of old fibrous variants share certain clinical and pathologic features
crescents, and nine (35%) have foci of necrosis and/or and require prompt institution of immunosuppressive
cellular crescents. Figure 1 (Jones methenamine silver therapy.
[JMS], ⫻400) shows segmental fibrinoid necrosis of
the glomerular tuft with rupture of glomerular base- Clinical presentation
ment membranes (GBMs) and extravasation of fibrin Pauci-immune GN is the most common cause of
into the urinary space, associated with a nearly cir- crescentic GN, especially in individuals who are older
cumferential cellular crescent (Figure 1). Figure 2 than 65 years (1,2) and very elderly patients who are
(JMS, ⫻400) illustrates severe segmental destruction older than 80 years (3,4). The disease is more common
of the tuft by a voluminous cellular crescent with in Caucasians and males. In addition to the renal
broad rupture of Bowman’s capsule and periglomeru- manifestations of rapidly progressive GN, most pa-
lar inflammation (Figure 2). The earliest lesion is a tients report a prodrome of fever, arthralgias, myal-
small segmental focus of fibrinoid necrosis with rup- gias, and “flu-like” illness. These symptoms are prob-
ture of the GBM and mild segmental proliferation of ably due in part to antecedent infections and elevated
the overlying glomerular epithelial cells, forming an levels of proinflammatory cytokines. In the presence
incipient crescent (Figure 3, JMS, ⫻400). The pre- of systemic vasculitis, the symptoms vary on the basis
served glomeruli have no evidence of mesangial or of the organ involvement and include pulmonary hem-
endocapillary proliferation (Figure 4, periodic acid– orrhage and hemoptysis (alveolar capillaritis), mono-
Schiff, ⫻400). There was prominent interstitial inflam- neuritis multiplex (peripheral nerve arteritis), palpable
mation by lymphocytes, monocytes, and plasma cells skin purpura (leukocytoclastic vasculitis), abdominal
associated with focal acute tubular injury. Several RBC pain, and occult blood in the stool (intestinal arteritis
casts were identified. Two interlobular arteries show and ischemia) (5). Although all ANCA-associated
necrotizing vasculitis with fibrinoid necrosis of the in- GN can be considered “vasculitis” of the glomerular
tima and transmural inflammation (Figure 5, JMS, capillaries, associated systemic small-vessel vascu-
⫻400). Except for sparse focal segmental glomerular tuft litis often manifests as distinct syndromes. Patients
staining for IgM and C3, no significant immune deposits with Wegener granulomatosis typically have necro-
were identified by immunofluorescence microscopy. tizing granulomatous inflammation of the upper and
However, there is intense (3⫹) staining for fibrin/fibrin- lower respiratory tracts, resulting in sinusitis, otitis
ogen in the distribution of the necrotizing lesions and media, saddle nose deformity, subglottic stenosis,
crescents (Figure 6, immunofluorescence for fibrin, and/or cavitary nodular lung lesions. Microscopic
⫻400). On electron microscopy, no glomerular or vas- polyangiitis also presents as pulmonary-renal syn-
cular immune-type electron-dense deposits were seen. drome but lacks granulomatous inflammation. Pa-
tients with Churg-Strauss syndrome often have a
Pauci-immune Crescentic GN history of asthma and blood eosinophilia (⬎10%) that
Pauci-immune crescentic GN usually presents may precede the renal symptoms by many years.
with rapidly progressive renal failure, hematuria, and Renal involvement occurs commonly in We-
subnephrotic proteinuria. Renal biopsy reveals focal gener granulomatosis and microscopic polyangiitis
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 381
(ⱖ80% of cases) but less frequently in Churg-Strauss More than one fourth of patients with anti-GBM
syndrome (⬍50%) (2). Urinalysis reveals hematuria, disease have ANCA in addition to anti-GBM antibod-
mild subnephrotic proteinuria, and RBC casts. Ne- ies. When compared with patients with anti-GBM
phrotic-range proteinuria is seen in a minority of cases nephritis alone, these patients have better prognosis
but is more common in those with a coexistent defined and renal survival rates that are intermediate between
immune complex–mediated disease (e.g., IgA ne- anti-GBM disease and ANCA vasculitis, but they are
phropathy, lupus nephritis) or nonspecific associated prone to relapses of ANCA disease (11,12). When a
immune deposits (6). Higher serum creatinine level at patient with positive ANCA serology has a mixed
biopsy, African American race, higher percentage of picture of necrotizing and crescentic GN with immune
total glomerular involvement, and more arteriosclero- deposits, overlaps with membranous nephropathy, IgA
nephropathy, lupus nephritis, and acute postinfectious
sis are negative predictors of renal survival (3,7,8).
GN should be considered (13–15).
ANCA serology. The diagnosis of pauci-immune
GN was advanced by the introduction of ANCA sero- Pathology
logic testing in the late 1980s. The predictive value of
Light microscopy. The early lesions of pauci-im-
ANCA positivity depends to a large extent on the
mune GN include focal and segmental glomerular
pretest probability of ANCA-associated vasculitis. In
fibrinoid necrosis with GBM rupture, sparse neutro-
the appropriate clinical context, positive ANCA has a
phil infiltration, and nuclear fragmentation (pyknosis
very high positive predictive value, but a negative
and karyorrhexis), followed by the rapid formation of
ANCA does not rule out pauci-immune GN (approx-
cellular crescents. The extent and severity of glomer-
imately 15% of cases are ANCA negative). Without ular involvement vary, but approximately two thirds of
clinical support for pauci-immune GN, a negative patients have ⬎50% crescents, qualifying as crescen-
ANCA has a high negative predictive value, whereas a tic GN (16). A brisk inflammatory infiltrate is often
positive test is a poor predictor of ANCA-associated seen adjacent to foci of glomerular necrosis and in-
disease. cludes lymphocytes, monocytes, plasma cells, neutro-
ANCA are autoantibodies that target proteins phils, and variable eosinophils. Glomeruli spared by
within neutrophil granules and monocyte lysosomes. the crescentic process usually appear unremarkable,
The more specific ELISA uses two purified substrates without evidence of mesangial or endocapillary pro-
that correspond to myeloperoxidase (MPO) and pro- liferation. Conversely, if ANCA-associated GN is su-
teinase 3 (PR3). The less specific (but more sensitive) perimposed on an immune complex–mediated disease,
indirect immunofluorescence method detects ANCA then variable glomerular proliferation may occur. Cir-
in patients’ serum using alcohol-fixed human neutro- cumferential crescents that cause destruction of Bow-
phils as substrate. Two patterns of neutrophil staining man’s capsule may result in periglomerular inflammation
are identified: Cytoplasmic staining corresponding to centered on the glomerulus, mimicking granulomas. Ep-
PR3-ANCA and perinuclear staining corresponding to ithelioid histiocytes and multinucleated giant cells may
MPO-ANCA. ANCA do not definitively distinguish contribute to the periglomerular inflammatory infiltrate.
categories of pauci-immune GN because of extensive However, true interstitial necrotizing granulomas are rare
overlap, but, in general, Wegener granulomatosis is in pauci-immune GN and are uncommon even in We-
more likely to be PR3-ANCA positive (75%), whereas gener granulomatosis and Churg-Strauss syndrome.
Churg-Strauss syndrome, microscopic polyangiitis, In addition to glomeruli, ANCA-associated small-
and renal limited disease more often tend to be MPO- vessel vasculitis can involve arterioles, interlobular arter-
ANCA positive (60, 50, and 60%, respectively) (9). ies, venules, and capillaries (16). Affected arteries show
Patients in all categories can have negative ANCA fibrinoid necrosis of the intima and media with mural
serologies, but Churg-Strauss syndrome has the high- inflammation and disruption of elastic lamellae. In-
est frequency of negativity. Although ANCA titers volvement of medullary vasa recta can cause necro-
correlate with disease activity in a high proportion of tizing interstitial capillaritis, medullary hemorrhage,
patients, it is not always predictive of relapses. In fact, and even papillary necrosis.
some patients have undergone successful renal trans- The chronic phase has segmental and global
plantation despite persistently high ANCA titers (10). glomerulosclerosis with evidence of old fibrous or
382 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
epigenetic modifications such as loss of DNA meth- and that cotrimazole therapy reduces the number of
ylation that result in increased surface expression of relapses (34). Thus, multiple lines of evidence support
MPO/PR3 (20,23,24). a role for infection in inciting and perpetuating
ANCA-mediated autoimmunity may be initiated ANCA-associated GN in genetically susceptible indi-
by anti-idiotypic antibodies, especially in PR3-ANCA viduals.
disease (22,25–27). The complementary peptide of
PR3, generated by the antisense DNA strand of PR3, References
1. Jennette JC: Rapidly progressive crescentic glomerulonephritis. Kid-
is itself capable of triggering an antibody response. ney Int 63: 1164 –1177, 2003
These anti-complementary antibodies in turn promote 2. Jennette JC, Falk RJ: Small-vessel vasculitis. N Engl J Med 337:
development of anti-idiotypic antibodies that can 1512–1523, 1997
3. Bomback AS, Appel GB, Radhakrishnan J, Shirazian S, Herlitz LC,
cross-react with sense peptide, the PR3 autoantigen. Stokes B, D’Agati VD, Markowitz GS: ANCA-associated glomeru-
Sequences similar to complementary PR3 peptide lonephritis in the very elderly. Kidney Int 79: 757–764, 2011
have been identified in microorganisms such as Ross 4. Moutzouris DA, Herlitz L, Appel GB, Markowitz GS, Freudenthal B,
Radhakrishnan J, D’Agati VD: Renal biopsy in the very elderly. Clin
River virus, Staphylococcus aureus, and Entamoeba J Am Soc Nephrol 4: 1073–1082, 2009
histolytica, among others, suggesting a role for molec- 5. Seo P, Stone JH: The antineutrophil cytoplasmic antibody-associated
vasculitides. Am J Med 117: 39 –50, 2004
ular mimicry in triggering autoimmunity (26). There is
6. Neumann I, Regele H, Kain R, Birck R, Meisl FT: Glomerular
also sequence homology between complementary PR3 immune deposits are associated with increased proteinuria in patients
protein and plasminogen, and cross-reacting antibod- with ANCA-associated crescentic nephritis. Nephrol Dial Transplant
18: 524 –531, 2003
ies may be responsible for the increased deep vein 7. Hogan SL, Falk RJ, Chin H, Cai J, Jennette CE, Jennette JC,
thrombosis seen in PR3-ANCA disease (28). Nachman PH: Predictors of relapse and treatment resistance in
Lysosomal membrane protein-2 (LAMP-2), a antineutrophil cytoplasmic antibody-associated small-vessel vasculi-
tis. Ann Intern Med 143: 621– 631, 2005
membrane-associated protein expressed on the surface 8. Berden AE, Ferrario F, Hagen EC, Jayne DR, Jennette JC, Joh K,
of neutrophils and endothelial cells, has been impli- Neumann I, Noel LH, Pusey CD, Waldherr R, Bruijn JA, Bajema IM:
cated in ANCA-associated GN (29,30). Autoantibod- Histopathologic classification of ANCA-associated glomerulonephri-
tis. J Am Soc Nephrol 21: 1628 –1636, 2010
ies against LAMP-2 were detected in ⬎90% of a 9. Jennette JC, Thomas DB: Pauci-immune and antineutrophil cytoplas-
patient cohort with pauci-immune GN, a prevalence mic autoantibody-mediated crescentic glomerulonephritis and vas-
rate twice that of MPO-ANCA or PR3-ANCA (30). culitis. In: Pathology of the Kidney, 6th Ed., edited by Jennette JC,
Philadelphia, Lippincott Williams & Wilkins, 2007, pp 664 – 673
LAMP-2 antibodies have been shown to cause endo- 10. Nachman PH, Segelmark M, Westman K, Hogan SL, Satterly KK,
thelial injury in vitro and to induce pauci-immune GN Jennette JC, Falk R: Recurrent ANCA-associated small vessel vas-
in vivo in animal studies (30). There is 100% homol- culitis after transplantation: A pooled analysis. Kidney Int 56: 1544 –
1550, 1999
ogy between one of the LAMP-2 epitopes and bacte- 11. Clyne S, Frederick C, Arndt F, Lewis J, Fogo AB: Concurrent and
rial fimbrial adhesin, FimH, suggesting a role for discrete clinicopathological presentations of Wegener granulomatosis
molecular mimicry induced by Gram-negative bacte- and anti-glomerular basement membrane disease. Am J Kidney Dis
54: 1116 –1120, 2009
rial infections (30). 12. Lionaki S, Jennette JC, Falk RJ: Anti-neutrophil cytoplasmic
It is known that neutrophils are capable of killing (ANCA) and anti-glomerular basement membrane (GBM) autoanti-
bodies in necrotizing and crescentic glomerulonephritis. Semin Im-
pathogens extracellularly by releasing chromatin fi-
munopathol 29: 459 – 474, 2007
bers during cell death, thereby creating a glutinous 13. Nasr SH, D’Agati VD, Park HR, Sterman PL, Goyzueta JD, Dressler
DNA web. These fibers, known as neutrophil extra- RM, Hazlett SM, Pursell RN, Caputo C, Markowitz GS: Necrotizing
and crescentic lupus nephritis with antineutrophil cytoplasmic anti-
cellular traps (NET), are formed after neutrophil re- body seropositivity. Clin J Am Soc Nephrol 3: 682– 690, 2008
spiratory burst (31). NET release was recently dem- 14. Nasr SH, Said SM, Valeri AM, Stokes MB, Masani NN, D’Agati VD,
onstrated in ANCA vasculitis, and these traps Markowitz GS: Membranous glomerulonephritis with ANCA-asso-
ciated necrotizing and crescentic glomerulonephritis. Clin J Am Soc
contained autoantigens PR3 and MPO (32). It has been Nephrol 4: 299 –308, 2009
proposed that the extracellular location of these anti- 15. Haas M, Jafri J, Bartosh SM, Karp SL, Adler SG, Meehan SM:
gens in response to a bacterial infection (e.g., S. ANCA-associated crescentic glomerulonephritis with mesangial IgA
deposits. Am J Kidney Dis 36: 709 –718, 2000
aureus) may trigger or exacerbate ANCA disease 16. Jennette JC, Falk RJ: The pathology of vasculitis involving the
(29,32). This hypothesis is consistent with previous kidney. Am J Kidney Dis 24: 130 –141, 1994
observations that chronic nasal infection with S. au- 17. Davies DJ, Moran JE, Niall JF, Ryan GB: Segmental necrotising
glomerulonephritis with antineutrophil antibody: possible arbovirus
reus is common in Wegener granulomatosis, that aetiology? Br Med J (Clin Res Ed) 285: 606, 1982
chronic nasal carriage is a risk factor for relapse (33), 18. Morgan MD, Harper L, Williams J, Savage C: Anti-neutrophil
384 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
cytoplasm-associated glomerulonephritis. J Am Soc Nephrol 17: 27. Shoenfeld Y: The idiotypic network in autoimmunity: Antibodies
1224 –1234, 2006 that bind antibodies that bind antibodies. Nat Med 10: 17–18, 2004
19. Xiao H, Heeringa P, Hu P, Liu Z, Zhao M, Aratani Y, Maeda N, Falk 28. Bautz DJ, Preston GA, Lionaki S, Hewins P, Wolberg AS, Yang JJ,
RJ, Jennette JC: Antineutrophil cytoplasmic autoantibodies specific Hogan SL, Chin H, Moll S, Jennette JC, Falk RJ: Antibodies with
for myeloperoxidase cause glomerulonephritis and vasculitis in mice. dual reactivity to plasminogen and complementary PR3 in PR3-
J Clin Invest 110: 955–963, 2002 ANCA vasculitis. J Am Soc Nephrol 19: 2421–2429, 2008
20. Falk RJ, Jennette JC: ANCA disease: Where is this field heading? 29. Bosch X: LAMPs and NETs in the pathogenesis of ANCA vasculitis.
J Am Soc Nephrol 21: 745–752, 2010 J Am Soc Nephrol 20: 1654 –1656, 2009
21. Xiao H, Schreiber A, Heeringa P, Falk RJ, Jennette JC: Alternative 30. Kain R, Exner M, Brandes R, Ziebermayr R, Cunningham D, Alder-
complement pathway in the pathogenesis of disease mediated by son CA, Davidovits A, Raab I, Jahn R, Ashour O, Spitzauer S,
anti-neutrophil cytoplasmic autoantibodies. Am J Pathol 170: 52– 64, Sunder-Plassmann G, Fukuda M, Klemm P, Rees AJ, Kerjaschki D:
2007
Molecular mimicry in pauci-immune focal necrotizing glomerulone-
22. Kallenberg CG: Pathogenesis of PR3-ANCA associated vasculitis. J
phritis. Nat Med 14: 1088 –1096, 2008
Autoimmun 30: 29 –36, 2008
31. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y,
23. Borgmann S, Haubitz M: Genetic impact of pathogenesis and prog-
Weiss DS, Weinrauch Y, Zychlinsky A: Neutrophil extracellular
nosis of ANCA-associated vasculitides. Clin Exp Rheumatol 22:
traps kill bacteria. Science 303: 1532–1535, 2004
S79 –S86, 2004
24. Ciavatta DJ, Yang J, Preston GA, Badhwar AK, Xiao H, Hewins P, 32. Kessenbrock K, Krumbholz M, Schonermarck U, Back W, Gross
Nester CM, Pendergraft WF 3rd, Magnuson TR, Jennette JC, Falk WL, Werb Z, Grone HJ, Brinkmann V, Jenne DE: Netting neutro-
RJ: Epigenetic basis for aberrant upregulation of autoantigen genes in phils in autoimmune small-vessel vasculitis. Nat Med 15: 623– 625,
humans with ANCA vasculitis. J Clin Invest 120: 3209 –3219, 2010 2009
25. Pendergraft WF 3rd, Pressler BM, Jennette JC, Falk RJ, Preston GA: 33. Stegeman CA, Tervaert JW, Sluiter WJ, Manson WL, de Jong PE,
Autoantigen complementarity: A new theory implicating comple- Kallenberg CG: Association of chronic nasal carriage of Staphylo-
mentary proteins as initiators of autoimmune disease. J Mol Med 83: coccus aureus and higher relapse rates in Wegener granulomatosis.
12–25, 2005 Ann Intern Med 120: 12–17, 1994
26. Pendergraft WF 3rd, Preston GA, Shah RR, Tropsha A, Carter CW 34. Stegeman CA, Tervaert JW, de Jong PE, Kallenberg CG: Trim-
Jr, Jennette JC, Falk RJ: Autoimmunity is triggered by cPR-3(105– ethoprim-sulfamethoxazole (co-trimoxazole) for the prevention of
201), a protein complementary to human autoantigen proteinase-3. relapses of Wegener’s granulomatosis. Dutch Co-Trimoxazole We-
Nat Med 10: 72–79, 2004 gener Study Group. N Engl J Med 335: 16 –20, 1996
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Section 19: CASE PRESENTATION history of hypertension, and BP is 110/60 mmHg. Serum
creatinine level is 1.2 mg/dl. All serologies are negative
A 72-year-old Caucasian woman presents with new onset or normal, including anti-nuclear antibody, C3, C4, hep-
of lower extremity edema for 1 month and is found to have atitis B surface antigen, hepatitis C antibody, and anti-
nephrotic syndrome (24-hour urine protein is 11.0 g, serum neutrophil cytoplasmic antibody. Serum protein electro-
albumin is 2.0 g/dl, serum cholesterol is 302 mg/dl). Her phoresis shows no evidence of a monoclonal spike. The
past medical history is notable for a diagnosis of diabetes 2 patient weighs 140 lb, body mass index is 27.3, and
years earlier. She has no evidence of diabetic retinopathy, kidney size is 11.5 cm bilaterally. A kidney biopsy is
and most recent hemoglobin A1c is 7.0. She has no performed.
Figure 1. Figure 2.
Figure 4.
Figure 3.
Figure 5. Figure 6.
385
386 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
What is the BEST diagnosis? confined to one organ or affect multiple organs, and
A. Diabetic glomerulosclerosis renal involvement is common in systemic amyloido-
B. Renal amyloidosis, AL type sis.
C. Fibrillary glomerulonephritis More than 26 amyloid precursor proteins have
D. Immunotactoid glomerulonephritis been identified, 12 of which may involve the kidney
E. Light-chain deposition disease (LCDD), type (1,2). These include light-chain amyloid (AL), heavy-
chain amyloid (AH), light- and heavy-chain amyloid
Answer (ALH), amyloid A (AA), 2 microglobulin, transthy-
The glomeruli appear normocellular. Mesangial retin, fibrinogen Aa, apolipoprotein A-I, apolipopro-
areas are moderately expanded by deposits of homo- tein A-II, lysozyme, cystatin C, gelsolin, and leuko-
geneous pale eosinophilic material (Figure 1, ⫻400). cyte chemotactic factor 2 (ALect2) (1) ALect2 is a
This material is weakly periodic acid–Schiff (PAS) newly recognized form of renal amyloidosis caused by
positive and segmentally thickens some glomerular deposition of leukocyte chemotactic factor 2 (LECT2)
capillary walls, without associated endocapillary pro- peptide, which is encoded by a distinct genetic poly-
liferation (Figure 2, ⫻400). The mesangial deposits morphism more prevalent in Mexican Americans (3).
are Congo red positive and give the typical apple- The most common form of renal amyloidosis (53
green birefringence of amyloid when viewed under to 63%) is derived from monoclonal Ig light chains
polarized light (Figure 3, Congo red, polarized light, (AL amyloid) (4,5). The second most common form is
⫻600). Immunofluorescence shows light-chain re- AA amyloidosis derived from deposition of serum
striction with negativity for light chain and intense amyloid A, an acute-phase reactant produced by the
staining for light chain in the distribution of the liver in chronic inflammatory states. Together, these
expanded mesangium and thickened glomerular cap- two etiologies account for ⬎95% of renal amyloidosis
illary walls (Figure 4, immunofluorescence staining encountered on renal biopsy. All forms of renal amy-
for and light chains, ⫻400). There was no loidosis appear similar by light microscopy and elec-
glomerular staining for immunoglobulins or com- tron microscopy. Diagnosis of a specific etiology re-
plement. Similar staining for light chain was seen quires demonstration of the precursor protein by
in some vessel walls. Electron microscopy reveals immunostaining using specific antisera to the patho-
fibrillar deposits infiltrating the glomerular base-
genic protein. In difficult cases, mass spectrometry can
ment membrane (GBM), forming focal subepithelial
be performed on tissue deposits extracted by laser
projections (Figure 5, ⫻25,000). Similar fibrillar de-
capture microdissection (6).
posits infiltrate the mesangial matrix (not shown). On
high-power magnification, the fibrils appear randomly
oriented and measure 8 to 12 nm in thickness Clinical Characteristics
(Figure 6, ⫻60,000). The biopsy findings are diagnos- AL amyloidosis typically affects older individu-
tic of renal amyloidosis, AL type (answer B). als (⬎50 years), reflecting the incidence of plasma cell
dyscrasia, with a male preponderance (5,7). In the
Renal Amyloidosis Mayo Clinic series of 474 patients with AL, the mean
The amyloidoses are a diverse group of diseases age was 64 years, and 69% were male (7). Most (73%)
characterized by extracellular tissue deposition of patients had proteinuria, 25% had nephrotic syndrome,
fibrillar proteins with mean diameter of 10 nm. Al- and 50% presented with renal insufficiency. The uri-
though the precursor proteins vary, all amyloid pro- nary sediment is typically bland, and microhematuria
teins share an antiparallel -pleated sheet structure on is uncommon. In an Italian study of 237 patients with
x-ray diffraction. In addition, all forms of amyloid AL, nephrotic syndrome was present in 59% and renal
contain amyloid P component, a member of the pen- insufficiency in 54% (8). In a renal biopsy series at
traxin family of serum proteins that plays a role in Columbia University, amyloidosis was the second
fibrillogenesis. The gold standard for pathologic diag- most common cause of nephrotic syndrome in very
nosis is the demonstration of characteristic apple- elderly patients who were older than 80 years (9).
green birefringence with Congo red stain when exam- Extrarenal manifestations of AL amyloid include re-
ined under polarized light (1). Amyloid may be strictive cardiomyopathy, orthostatic hypotension, pe-
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 387
inant involvement of the vessels or tubulointerstitial uniform, the deposits of AL amyloid are usually more
compartment may occur. Some of these cases have chunky, localized, and irregular.
disproportionately severe involvement of the medul- Electron Microscopy. Amyloid deposits are com-
lary interstitium. When vessels are affected, there is a posed of randomly oriented, nonbranching fibrils of
tendency for the amyloid to replace medial myocytes, indeterminate length, which usually measure 8 to 12
causing the media to appear hypocellular (Figure 9). nm in average thickness (Figures 5 and 6). Fibrils
Immunofluorescence. In AL amyloid, immunohis- typically deposit first in the mesangium, followed by
tochemical stains show monotypic light chain ( light GBM deposits. In early cases with small focal depos-
chain in 75% of cases). Rarely, deposits stain for a its, demonstration of the fibrillar deposits may be
single Ig heavy chain (␥, ␣, or ; AH), or a single light difficult without careful search. Fibrils often infiltrate
chain and heavy chain. By immunofluorescence, am- the entire GBM thickness and can aggregate in parallel
yloid deposits have a “smudged” appearance, which is to form perpendicular spicular projections toward the
distinct from the more granular texture of immune urinary space, corresponding to the hair-like projec-
complex deposits. Unlike Randall-type LCDD, in tions seen by light microscopy. Podocyte foot process
which the deposits tend to be linear, diffuse, and effacement is variable and tends to be most severe
overlying glomerular capillary walls infiltrated by am-
yloid. At the ultrastructural level, amyloid fibrils look
similar regardless of the renal compartment involved
and the precursor protein.
Special Studies. Positive Congo red staining is
diagnostic of amyloidosis and excludes other dis-
eases that give rise to acellular glomerular deposits,
such as diabetic glomerulosclerosis, LCDD, fibril-
lary glomerulonephritis, immunotactoid glomeru-
lopathy, fibronectin glomerulopathy, and collageno-
fibrotic glomerulopathy. For best results, Congo red
stain should be performed on thick tissue sections (at
least 6 m). In most cases of AL, the Congo red stain
is unequivocal. Interpretation may be difficult in cases
Figure 8. The amyloid fibrils infiltrate some GBMs, form- in which only small deposits are present. In such cases,
ing spicular projections that mimic spikes (arrows). Magni- electron microscopy may be diagnostic. The major
fication, ⫻600 (Jones methenamine silver).
cause of a false-positive Congo red stain is confusion
with interstitial collagen, which gives a yellow bire-
fringence under polarized light. Thus, interpretation of
Congo red stain is best performed by an experienced
observer.
Diagnosis of AL and AA amyloid can usually be
made by immunofluorescence staining for and
light chain and immunohistochemical staining for se-
rum amyloid A. Of note, a minority of AL cases do not
stain with commercial antibodies for Ig or light chains
(up to 35% in one series) (25). This occurs when the
truncated monoclonal light chains contain only the
variable region, which cannot be detected with com-
mercial antisera raised against the constant domain of
Figure 9. Arterial walls are replaced by amyloid deposits,
the light-chain molecule (1). In addition, up to 25% of
which appear blue-gray with trichrome stain. Magnification, patients with hereditary forms of amyloidosis have an
⫻200 (trichrome). unrelated monoclonal gammopathy (26). This may
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 389
lead to nonspecific trapping of the circulating mono- Most cases of AL (with or without kidney
clonal light chain in the amyloid deposits. Similarly, involvement) are derived from light chain. A
because of the “stickiness” of the amyloid deposits, recent study suggested that AL has a higher
AA amyloidosis may exhibit nonspecific immunoflu- incidence of nephrotic syndrome compared
orescence staining for circulating plasma proteins, with AL , whereas AL shows more cardiac and
including immunoglobulins, light chains, and comple- liver involvement (11). Although overall survival is
ment components, making interpretation difficult. similar in AL and , renal involvement is more
Thus, careful clinical correlation, staining for other common with AL and has worse outcomes (22).
amyloid proteins (e.g., transthyretin, fibrinogen Aa, Cardiac involvement is the major determinant of sur-
apolipoprotein AI and AII, lysozyme) (1), mass spec- vival in AL (31), but renal involvement is an indepen-
trometry, and genetic testing all are helpful in cases in dent predictor of mortality (22).
which an unequivocal diagnosis of AL or AA is not
possible. Etiology and Pathogenesis
Although renal biopsy expectedly has the highest AL is caused by release of an amyloidogenic
yield for diagnosing renal AL amyloidosis, there are monoclonal light chain or light chain fragment from a
less invasive means of tissue diagnosis. Subcutaneous neoplastic plasma cell clone in the bone marrow or
fat biopsy (via either fine-needle aspiration or core less commonly a neoplastic B cell clone in the lym-
biopsy) is a highly sensitive and specific technique for phoid tissue. Amino acid substitutions in AL light
diagnosing systemic amyloidosis (27,28). In addition, chains are thought to promote abnormal protein fold-
fat biopsies are a safe and easy method of obtaining ing favoring fibrillogenesis, and clones derived from
tissue for amyloid typing and novel investigations, the 6a (VI) germline gene are more likely to cause
such as proteomic studies (29). Renal involvement by renal involvement (32).
AA amyloid occurs in three settings: Chronic
AL amyloidosis can be inferred if amyloid is detected
infection, autoimmune inflammatory conditions, and
by biopsy of a nonrenal tissue, there is ⱖ0.5 g of
familial Mediterranean fever. Infectious causes in-
albumin in a 24-hour urine collection, and there is no
clude chronic bronchiectasis, osteomyelitis, decubitus
other explanation for the albuminuria (e.g., diabetes,
ulcers, tuberculosis, and intravenous drug use when
hypertension) (30).
the user resorts to skin popping. Autoimmune condi-
tions associated with AA amyloid include rheumatoid
Clinical-Pathologic Correlation arthritis, juvenile rheumatoid arthritis, inflammatory
Most patients who have AL and undergo renal bowel disease, and ankylosing spondylitis. Familial
biopsy for indications of heavy proteinuria or ne- Mediterranean fever is an autosomal recessive cause
phrotic syndrome exhibit significant glomerular amy- of AA amyloid with highest prevalence in Sephardic
loid deposits, with or without tubulointerstitial and Jews, Turks, Armenians, and Arabs and manifesting
vessel wall deposits. Interestingly, the amount of recurrent episodes of fever and serositis. Regardless of
glomerular amyloid deposition does not correlate the cause, the precursor protein in AA amyloid is the
well with the severity of proteinuria; occasional amino terminal region of serum amyloid A (SAA)
cases show minimal amyloid deposits despite ne- protein, an acute-phase reactant that is produced by the
phrotic-range proteinuria. Conversely, the level of liver and circulates in association with HDL.
renal insufficiency tends to correlate with the overall The factors that determine the potential for amy-
degree of amyloid infiltration, particularly when it loidogenesis in AL and AA include macrophage-de-
leads to glomerular obliteration, vascular narrowing, pendent processing to generate amyloid precursors,
and tubulointerstitial replacement. Cases with predom- the -pleated sheet structure of the precursor proteins,
inant tubulointerstitial involvement are more likely to capacity for amyloidogenic protein folding, stabiliza-
present with renal insufficiency than proteinuria and tion by amyloid P protein, and interaction with spe-
may develop specific tubular defects, such as renal cific sulfated glycosaminoglycans and proteoglycans.
tubular acidosis and nephrogenic diabetes insipidus. The factors governing tissue and organ tropisms are
Cases of AL amyloid tend to have less interstitial unknown. The pathogenic light chains have been
inflammation than cases of AA amyloid. shown in vitro to have proapoptotic effects on mesan-
390 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
gial cells, inhibit mesangial cell synthesis of TGF-, nostic in patients with primary systemic amyloidosis undergoing
peripheral blood stem cell transplantation. Blood 107: 3378 –3383,
and activate metalloproteinases, leading to replace- 2006
ment of mesangial cells and matrix by amyloid fibrils 14. Morris KL, Tate JR, Gill D, Kennedy G, Wellwood J, Marlton P, Bird
(33,34). Physical disruption of the glomerular filtra- R, Mills AK, Mollee P: Diagnostic and prognostic utility of the serum
free light chain assay in patients with AL amyloidosis. Intern Med J
tion barrier results in nonselective proteinuria. 37: 456 – 463, 2007
15. Lachmann HJ, Gallimore R, Gillmore JD, Carr-Smith HD, Bradwell
AR, Pepys MB, Hawkins PN: Outcome in systemic AL amyloidosis
References in relation to changes in concentration of circulating free immuno-
1. Picken MM: Amyloidosis: Where are we now and where are we globulin light chains following chemotherapy. Br J Haematol 122:
heading? Arch Pathol Lab Med 134: 545–551, 2010 78 – 84, 2003
2. Benson MD, James S, Scott K, Liepnieks JJ, Kluve-Beckerman B: 16. Sanchorawala V, Seldin DC, Magnani B, Skinner M, Wright DG:
Leukocyte chemotactic factor 2: A novel renal amyloid protein. Serum free light-chain responses after high-dose intravenous mel-
Kidney Int 74: 218 –222, 2008 phalan and autologous stem cell transplantation for AL (primary)
3. Murphy CL, Wang S, Kestler D, Larsen C, Benson D, Weiss DT, amyloidosis. Bone Marrow Transplant 36: 597– 600, 2005
Solomon A: Leukocyte chemotactic factor 2 (LECT2)-associated 17. Sanchorawala V: Light-chain (AL) amyloidosis: Diagnosis and treat-
renal amyloidosis: A case series. Am J Kidney Dis 56: 1100 –1107, ment. Clin J Am Soc Nephrol 1: 1331–1341, 2006
2010 18. Kumar SK, Gertz MA, Lacy MQ, Dingli D, Hayman SR, Buadi FK,
4. Bergesio F, Ciciani AM, Manganaro M, Palladini G, Santostefano M, Short-Detweiler K, Zeldenrust SR, Leung N, Greipp PR, Lust JA,
Brugnano R, Di Palma AM, Gallo M, Rosati A, Tosi PL, Salvadori Russell SJ, Kyle RA, Rajkumar SV, Dispenzieri A: Recent improve-
M: Renal involvement in systemic amyloidosis: An Italian collabor- ments in survival in primary systemic amyloidosis and the impor-
ative study on survival and renal outcome. Nephrol Dial Transplant tance of an early mortality risk score. Mayo Clin Proc 86: 12–18,
23: 941–951, 2008 2011
5. von Hutten H, Mihatsch M, Lobeck H, Rudolph B, Eriksson M, 19. Comenzo RL: Current and emerging views and treatments of sys-
Rocken C: Prevalence and origin of amyloid in kidney biopsies. Am J temic immunoglobulin light-chain (Al) amyloidosis. Contrib Nephrol
Surg Pathol 33: 1198 –1205, 2009 153: 195–210, 2007
6. Sethi S, Theis JD, Leung N, Dispenzieri A, Nasr SH, Fidler ME, 20. Sanchorawala V, Wright DG, Rosenzweig M, Finn KT, Fennessey S,
Cornell LD, Gamez JD, Vrana JA, Dogan A: Mass spectrometry- Zeldis JB, Skinner M, Seldin DC: Lenalidomide and dexamethasone
based proteomic diagnosis of renal immunoglobulin heavy chain in the treatment of AL amyloidosis: Results of a phase 2 trial. Blood
amyloidosis. Clin J Am Soc Nephrol 5: 2180 –2187, 2010 109: 492– 496, 2007
7. Kyle RA, Gertz MA: Primary systemic amyloidosis: Clinical and 21. Wechalekar AD, Lachmann HJ, Offer M, Hawkins PN, Gillmore JD:
laboratory features in 474 cases. Semin Hematol 32: 45–59, 1995 Efficacy of bortezomib in systemic AL amyloidosis with relapsed/
refractory clonal disease. Haematologica 93: 295–298, 2008
8. Bergesio F, Ciciani AM, Santostefano M, Brugnano R, Manganaro
22. Gertz MA, Leung N, Lacy MQ, Dispenzieri A, Zeldenrust SR,
M, Palladini G, Di Palma AM, Gallo M, Tosi PL, Salvadori M: Renal
Hayman SR, Buadi FK, Dingli D, Greipp PR, Kumar SK, Lust JA,
involvement in systemic amyloidosis: An Italian retrospective study
Rajkumar SV, Russell SJ, Witzig TE: Clinical outcome of immuno-
on epidemiological and clinical data at diagnosis. Nephrol Dial
globulin light chain amyloidosis affecting the kidney. Nephrol Dial
Transplant 22: 1608 –1618, 2007
Transplant 24: 3132–3137, 2009
9. Moutzouris DA, Herlitz L, Appel GB, Markowitz GS, Freudenthal B,
23. Dember LM, Sanchorawala V, Seldin DC, Wright DG, LaValley M,
Radhakrishnan J, D’Agati VD: Renal biopsy in the very elderly. Clin
Berk JL, Falk RH, Skinner M: Effect of dose-intensive intravenous
J Am Soc Nephrol 4: 1073–1082, 2009
melphalan and autologous blood stem-cell transplantation on al
10. Bochtler T, Hegenbart U, Heiss C, Benner A, Cremer F, Volkmann
amyloidosis-associated renal disease. Ann Intern Med 134: 746 –753,
M, Ludwig J, Perz JB, Ho AD, Goldschmidt H, Schonland SO:
2001
Evaluation of the serum-free light chain test in untreated patients with 24. Leung N, Dispenzieri A, Fervenza FC, Lacy MQ, Villicana R,
AL amyloidosis. Haematologica 93: 459 – 462, 2008 Cavalcante JL, Gertz MA: Renal response after high-dose melphalan
11. Kumar S, Dispenzieri A, Katzmann JA, Larson DR, Colby CL, Lacy and stem cell transplantation is a favorable marker in patients with
MQ, Hayman SR, Buadi FK, Leung N, Zeldenrust SR, Ramirez- primary systemic amyloidosis. Am J Kidney Dis 46: 270 –277, 2005
Alvarado M, Clark RJ, Kyle RA, Rajkumar SV, Gertz MA: Serum 25. Novak L, Cook WJ, Herrera GA, Sanders PW: AL-amyloidosis is
immunoglobulin free light chain measurement in AL amyloidosis: underdiagnosed in renal biopsies. Nephrol Dial Transplant 19: 3050 –
Prognostic value and correlations with clinical features. Blood 116: 3053, 2004
5126 –5129, 2010 26. Lachmann HJ, Booth DR, Booth SE, Bybee A, Gilbertson JA,
12. Dispenzieri A, Kyle R, Merlini G, Miguel JS, Ludwig H, Hajek R, Gillmore JD, Pepys MB, Hawkins PN: Misdiagnosis of hereditary
Palumbo A, Jagannath S, Blade J, Lonial S, Dimopoulos M, amyloidosis as AL (primary) amyloidosis. N Engl J Med 346:
Comenzo R, Einsele H, Barlogie B, Anderson K, Gertz M, Harous- 1786 –1791, 2002
seau JL, Attal M, Tosi P, Sonneveld P, Boccadoro M, Morgan G, 27. Kaplan B, Vidal R, Kumar A, Ghiso J, Gallo G: Immunochemical
Richardson P, Sezer O, Mateos MV, Cavo M, Joshua D, Turesson I, microanalysis of amyloid proteins in fine-needle aspirates of abdom-
Chen W, Shimizu K, Powles R, Rajkumar SV, Durie BG: Interna- inal fat. Am J Clin Pathol 112: 403– 407, 1999
tional Myeloma Working Group guidelines for serum-free light chain 28. Ansari-Lari MA, Ali SZ: Fine-needle aspiration of abdominal fat pad
analysis in multiple myeloma and related disorders. Leukemia 23: for amyloid detection: A clinically useful test? Diagn Cytopathol 30:
215–224, 2009 178 –181, 2004
13. Dispenzieri A, Lacy MQ, Katzmann JA, Rajkumar SV, Abraham RS, 29. Lavatelli F, Perlman DH, Spencer B, Prokaeva T, McComb ME,
Hayman SR, Kumar SK, Clark R, Kyle RA, Litzow MR, Inwards DJ, Theberge R, Connors LH, Bellotti V, Seldin DC, Merlini G, Skinner
Ansell SM, Micallef IM, Porrata LF, Elliott MA, Johnston PB, M, Costello CE: Amyloidogenic and associated proteins in systemic
Greipp PR, Witzig TE, Zeldenrust SR, Russell SJ, Gastineau D, Gertz amyloidosis proteome of adipose tissue. Mol Cell Proteomics 7:
MA: Absolute values of immunoglobulin free light chains are prog- 1570 –1583, 2008
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 391
30. Gertz MA, Comenzo R, Falk RH, Fermand JP, Hazenberg BP, 32. Comenzo RL, Zhang Y, Martinez C, Osman K, Herrera GA: The
Hawkins PN, Merlini G, Moreau P, Ronco P, Sanchorawala V, Sezer tropism of organ involvement in primary systemic amyloidosis:
O, Solomon A, Grateau G: Definition of organ involvement and Contributions of Ig V(L) germ line gene use and clonal plasma cell
treatment response in immunoglobulin light chain amyloidosis (AL): burden. Blood 98: 714 –720, 2001
A consensus opinion from the 10th International Symposium on 33. Teng J, Turbat-Herrera EA, Herrera GA: Role of translational re-
Amyloid and Amyloidosis, Tours, France, 18 –22 April 2004. Am J search advancing the understanding of the pathogenesis of light
Hematol 79: 319 –328, 2005 chain-mediated glomerulopathies. Pathol Int 57: 398 – 412, 2007
31. Bollee G, Guery B, Joly D, Snanoudj R, Terrier B, Allouache M, 34. Keeling J, Teng J, Herrera GA: AL-amyloidosis and light-chain
Mercadal L, Peraldi MN, Viron B, Fumeron C, Elie C, Fakhouri F: deposition disease light chains induce divergent phenotypic trans-
Presentation and outcome of patients with systemic amyloidosis formations of human mesangial cells. Lab Invest 84: 1322–1338,
undergoing dialysis. Clin J Am Soc Nephrol 3: 375–381, 2008 2004
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Section 20: CASE PRESENTATION nuclear antibody, anti-DNA antibody, hepatitis B surface
antigen, hepatitis C antibody, HIV serology, and serum
A 60-year-old Caucasian man with a history of morbid complements C3 and C4. Serum protein electrophoresis
obesity, hypertension, hyperlipidemia, and sleep apnea with immunofixation electrophoresis shows hypogam-
presents with nephrotic syndrome and renal insufficiency. maglobulinemia with no evidence of a monoclonal spike,
On physical examination, he is normotensive (on antihyper- and urinary protein electrophoresis is negative for a mono-
tensive medications) with 2⫹ pitting lower extremity clonal protein. Serum free light chain testing shows elevated
edema. Urinalysis shows 4⫹ protein, 1⫹ heme, and bland free light chains and a serum / ratio of 75.0 (normal
urinary sediment. Laboratory workup shows serum creati- range 0.26 to 1.65). Bone marrow biopsy shows a normo-
nine level of 2.36 mg/dl, 24-hour urine protein of 3.5 g, cellular marrow without evidence of plasmacytosis, and no
serum albumin of 2.1 g/dl, and hemoglobin of 10.7 g/dl. All clonal B cell population is detected by polymerase chain
serologic studies are negative or normal, including anti- reaction for Ig heavy chain gene rearrangement.
Figure 1.
Figure 2.
Figure 3.
Figure 4. IgG
393
394 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
C4) may occur in cases of HCDD or LHCDD diomyopathy, myocardial infarction, conduction de-
because of the complement-fixing ability of the fects, and arrhythmias.
heavy chains (particularly ␥1 or ␥3), which resides
in the CH2 domain (3,5). Pathology
Because the urine dipstick detects albumin but is Light microscopy. Nodular sclerosing glomeru-
insensitive to light and heavy chains, there is often a lopathy is the most characteristic histologic finding in
discrepancy between the quantification of proteinuria MIDD (3,15). The mesangium is diffusely and glo-
by dipstick and by a 24-hour urine collection. Immu- bally expanded by increased matrix admixed with
nofixation electrophoresis is superior to routine serum deposits, associated with milder mesangial hypercel-
and urine protein electrophoresis for the identification lularity. The mesangial expansion and presence of
of monoclonal light or heavy chains. This is particu- variable membranoproliferative features (focal mesan-
larly important in MIDD, in which the concentration gial interposition and duplication of GBMs) narrow
of monoclonal Ig in urine or serum may be low as a the capillary lumina. The deposits typically stain
result of strong tissue avidity. Overall, a monoclonal brightly eosinophilic, strongly periodic acid–Schiff
serum or urine protein can be identified in approxi- (PAS) positive, trichrome red, and weakly argyro-
mately 80% of patients with LCDD and 60% of philic. Some of the nodules are surrounded by glomer-
patients with HCDD (3). In some cases of HCDD, the ular capillary microaneurysms, resembling those seen
truncated heavy chain can be detected only on serum in diabetic nephropathy. However, the nodules of
fractionation and Western blotting (2,9). Even in cases MIDD tend to be more symmetrically distributed and
have different staining characteristics than diabetic
in which pathogenic light chains were undetectable in
nodules, which typically stain exclusively for extra-
serum or urine, in vitro biosynthetic labeling experi-
cellular matrix (ECM) as trichrome blue and strongly
ments have confirmed their secretion by bone marrow
argyrophilic. In contrast to amyloidosis, the deposits
plasma cells (1,10,11). The recent introduction of
of MIDD are Congo red negative.
serum free and light-chain quantification is prov-
There is ribbon-like thickening of the GBMs,
ing to be an extremely sensitive assay (12). Because
TBMs, and vascular basement membranes by PAS-
the vast majority of paraproteinemias are due to light-
positive material. In the arteries, the deposits have the
chain overproduction, this test has the potential to
distinctive feature of ringing the individual myocytes,
capture virtually all cases of LCDD or LHCDD.
producing a thick pericellular cuff (Figure 6). A rare
A bone marrow biopsy performed for documen- pattern of LCDD has been described with massive
tation of hematologic malignancy shows variable find- thickening of glomerular capillary walls, mimicking
ings. The criteria for multiple myeloma are met in 40 membranoproliferative glomerulonephritis, without
to 50% of cases of LCDD and 25% of cases of HCDD evidence of nodular glomerulosclerosis (16). Cres-
(1,3,13,14), whereas others have a mild plasma cell cents are uncommon in LCDD but may occur in up to
dyscrasia, often requiring immunohistochemical con- one third of cases of HCDD. Rarely, the deposits form
firmation. When MIDD is associated with cast ne- interstitial PAS-positive aggregates that are confluent
phropathy, multiple myeloma is diagnosed in ⬎90% with the thickened TBMs (Figure 7). Giant cell reac-
of cases. Waldenstrom macroglobulinemia and tion may form to some of the larger interstitial depos-
chronic lymphocytic leukemia are more rare causes of its. In more advanced cases, there is severe tubular
MIDD. atrophy and interstitial fibrosis.
MIDD is a systemic disease with clinically evi- Crystalline or fractured casts dominate the histo-
dent extrarenal involvement most commonly affecting logic picture in cases of MIDD with coexistent my-
the liver and heart. Other possible sites of involvement eloma cast nephropathy. Most such cases are due to
include the lymph nodes, salivary glands, spleen, gas- LCDD with light-chain restriction (3). Usually, the
trointestinal tract, lung, skin, thyroid gland, adrenal renal biopsies show no obvious nodular glomerulo-
glands, and peripheral nerves (4). Hepatic involvement sclerosis by light microscopy, although linear deposits
may present as cholestatic jaundice, elevated liver of MIDD are evident by immunofluorescence. In ad-
function tests, portal hypertension, and rarely liver dition, these cases usually lack identifiable electron-
failure. Cardiac manifestations include restrictive car- dense deposits at the ultrastructural level.
396 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Figure 6. An arteriole contains strongly eosinophilic depos- Figure 8. A case of ␥-HCDD illustrates positivity for IgG
its that form rings around the individual myocytes in a case that rings the individual myocytes of a small artery, pro-
of LCDD. Magnification, ⫻600 (hematoxylin and eosin). ducing a net-like pattern. Magnification, ⫻400 (immuno-
fluorescence, IgG).
the renal transplant, patients with ESRD should not undergo promote myofibroblastic transformation of mesangial
transplantation unless their synthesis of monoclonal protein cells, with increased mesangial synthesis of growth
has been controlled through effective therapy. factors, PDGF-, and TGF- (28,30). Whereas the
initial activation of PDGF- stimulates mesangial cell
Pathogenesis proliferation, the subsequent activation of TGF- in-
MIDD is caused by tissue deposition of patho- hibits proliferation while enhancing ECM production
genic Ig light and/or heavy chains, which in turn (30). Exposure to pathogenic light chains induces
promotes accumulation of ECM to produce the char- mesangial cell synthesis of tenascin-C, a major ECM
acteristic pathologic lesions (20). Sequence analysis of component of the nodules (28,30). Other ECM pro-
the pathogenic Ig fragments has identified altered teins synthesized include laminin, collagen IV, and
structural properties that promote tissue deposition and fibronectin. It is also likely that the normal degrada-
fibrotic responses. tion of tenascin-C is compromised by ineffective mes-
The vast majority of LCDD are restricted, in angial release of matrix metalloproteinase 7 (30).
contrast to the predominance of restriction in AL Light chains deposited in the glomerular capillary wall
amyloidosis. In the course of Ig production, a B cell are thought to mediate proteinuria by interference with
clone chooses from among four possible variable do- endothelial and podocyte functions required for main-
mains of the light chain, which are denoted VI tenance of filtration barrier.
through IV. Patients with LCDD have an overrepre- Sequence analysis of pathogenic heavy chains in
sentation of the VIV variability subgroup (21), with HCDD has revealed the consistent deletion of the CH1
fewer examples of the VI subgroup (22). The vari- constant domain in all cases of ␥-HCDD, as well as a
able domains contain highly variable complementari- case of ␣-HCDD (5,31). Rarely, the CH2 and hinge
ty-determining regions (CDR1 through 3) that further regions are also deleted (2). The CH1 domain normally
broaden the repertoire of antigenic determinants rec- binds to a chaperone protein, heavy chain– binding
ognized. Importantly, the rare VIV subgroup nor- protein, for assembly of the complete Ig molecule
mally has a longer CDR1 loop containing more hy- within the endoplasmic reticulum of the plasma cell
drophobic residues than other V subgroups. In (32). Heavy chains lacking a CH1 domain are prema-
LCDD, analysis of pathogenic VIV light chains at turely secreted into the circulation as free heavy chains
the cDNA and protein levels has identified specific in the case of HCDD or as heavy chain complexed to
amino acid substitutions (e.g., leucine, isoleucine, or light chain in the case of LHCDD. Similar to LCDD,
tyrosine at positions 27 and/or 31) that alter the light the pathogenic heavy chains often have unique amino
chain’s physiochemical properties (8). The resultant acid substitutions in the variable domain that affect
conformational changes and presence of hydrophobic charge and hydrophobicity, favoring tissue deposition
residues at sites exposed to solvent promote reduced (2,33).
protein solubility and enhanced tissue precipitation
(23) as well as ECM production (24). In addition,
References
charge alterations can increase the binding of cationic 1. Buxbaum JN, Chuba JV, Hellman GC, Solomon A, Gallo GR:
light chains to anionic basement membranes (25,26). Monoclonal immunoglobulin deposition disease: Light chain and
Posttranslational N-glycosylation of the pathogenic light and heavy chain deposition diseases and their relation to light
chain amyloidosis—Clinical features, immunopathology, and molec-
light chains is thought to increase tissue avidity in ular analysis. Ann Intern Med 112: 455– 464, 1990
patients with LCDD with undetectable serum levels 2. Aucouturier P, Khamlichi AA, Touchard G, Justrabo E, Cogne M,
(13,21). Chauffert B, Martin F, Preud’homme JL: Brief report: Heavy-chain
deposition disease. N Engl J Med 329: 1389 –1393, 1993
Mesangial cells play a critical role in the patho- 3. Lin J, Markowitz GS, Valeri AM, Kambham N, Sherman WH, Appel
genesis of LCDD-related nodular glomerulosclerosis GB, D’Agati VD: Renal monoclonal immunoglobulin deposition
(27). Investigators have established an in vitro model disease: The disease spectrum. J Am Soc Nephrol 12: 1482–1492,
2001
of LCDD by incubating human mesangial cells with 4. Randall RE, Williamson WC Jr, Mullinax F, Tung MY, Still WJ:
pathogenic light chains isolated from the urine of Manifestations of systemic light chain deposition. Am J Med 60:
patients with LCDD (28 –30). The mesangial cells 293–299, 1976
5. Kambham N, Markowitz GS, Appel GB, Kleiner MJ, Aucouturier P,
undergo nodular growth patterns in vitro, mimicking D’Agati VD: Heavy chain deposition disease: The disease spectrum.
the lesions seen on biopsy (30). The light chains Am J Kidney Dis 33: 954 –962, 1999
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 399
6. Markowitz GS: Dysproteinemia and the kidney. Adv Anat Pathol 11: light (heavy)-chain deposition disease: From molecular medicine to
49 – 63, 2004 pathophysiology-driven therapy. Clin J Am Soc Nephrol 1: 1342–
7. Pozzi C, D’Amico M, Fogazzi GB, Curioni S, Ferrario F, Pasquali S, 1350, 2006
Quattrocchio G, Rollino C, Segagni S, Locatelli F: Light chain 21. Denoroy L, Deret S, Aucouturier P: Overrepresentation of the V
deposition disease with renal involvement: Clinical characteristics kappa IV subgroup in light chain deposition disease. Immunol Lett
and prognostic factors. Am J Kidney Dis 42: 1154 –1163, 2003 42: 63– 66, 1994
8. Ronco PM, Alyanakian MA, Mougenot B, Aucouturier P: Light 22. Vidal R, Goni F, Stevens F, Aucouturier P, Kumar A, Frangione B,
chain deposition disease: A model of glomerulosclerosis defined at Ghiso J, Gallo G: Somatic mutations of the L12a gene in V-kappa(1)
the molecular level. J Am Soc Nephrol 12: 1558 –1565, 2001 light chain deposition disease: Potential effects on aberrant protein
9. Moulin B, Deret S, Mariette X, Kourilsky O, Imai H, Dupouet L, conformation and deposition. Am J Pathol 155: 2009 –2017, 1999
Marcellin L, Kolb I, Aucouturier P, Brouet JC, Ronco PM, Mougenot 23. Khamlichi AA, Rocca A, Touchard G, Aucouturier P, Preud’homme
B: Nodular glomerulosclerosis with deposition of monoclonal immu- JL, Cogne M: Role of light chain variable region in myeloma with
noglobulin heavy chains lacking C(H)1. J Am Soc Nephrol 10: light chain deposition disease: Evidence from an experimental model.
519 –528, 1999 Blood 86: 3655–3659, 1995
10. Cogne M, Preud’homme JL, Bauwens M, Touchard G, Aucouturier 24. Deret S, Chomilier J, Huang DB, Preud’homme JL, Stevens FJ,
P: Structure of a monoclonal kappa chain of the V kappa IV subgroup Aucouturier P: Molecular modeling of immunoglobulin light chains
in the kidney and plasma cells in light chain deposition disease. J Clin implicates hydrophobic residues in non-amyloid light chain deposi-
Invest 87: 2186 –2190, 1991 tion disease. Protein Eng 10: 1191–1197, 1997
11. Preud’homme JL, Morel-Maroger L, Brouet JC, Cerf M, Mignon F, 25. Kaplan B, Livneh A, Gallo G: Charge differences between in vivo
Guglielmi P, Seligmann M: Synthesis of abnormal immunoglobulins
deposits in immunoglobulin light chain amyloidosis and non-amyloid
in lymphoplasmacytic disorders with visceral light chain deposition.
light chain deposition disease. Br J Haematol 136: 723–728, 2007
Am J Med 69: 703–710, 1980
26. Picken MM: Immunoglobulin light and heavy chain amyloidosis
12. Pratt G: The evolving use of serum free light chain assays in
AL/AH: Renal pathology and differential diagnosis. Contrib Nephrol
haematology. Br J Haematol 141: 413– 422, 2008
153: 135–155, 2007
13. Ganeval D, Noel LH, Preud’homme JL, Droz D, Grunfeld JP:
27. Zhu L, Herrera GA, Murphy-Ullrich JE, Huang ZQ, Sanders PW:
Light-chain deposition disease: Its relation with AL-type amyloido-
Pathogenesis of glomerulosclerosis in light chain deposition disease:
sis. Kidney Int 26: 1–9, 1984
Role for transforming growth factor-beta. Am J Pathol 147: 375–385,
14. Tubbs RR, Gephardt GN, McMahon JT, Hall PM, Valenzuela R, Vidt
1995
DG: Light chain nephropathy. Am J Med 71: 263–269, 1981
15. Sanders PW, Herrera GA, Kirk KA, Old CW, Galla JH: Spectrum of 28. Herrera GA, Russell WJ, Isaac J, Turbat-Herrera EA, Tagouri YM,
glomerular and tubulointerstitial renal lesions associated with mono- Sanders PW, Picken MM, Dempsey S: Glomerulopathic light chain-
typical immunoglobulin light chain deposition. Lab Invest 64: 527– mesangial cell interactions modulate in vitro extracellular matrix
537, 1991 remodeling and reproduce mesangiopathic findings documented in
16. Salant DJ, Sanchorawala V, D’Agati VD: A case of atypical light vivo. Ultrastruct Pathol 23: 107–126, 1999
chain deposition disease: Diagnosis and treatment. Clin J Am Soc 29. Keeling J, Herrera GA: Matrix metalloproteinases and mesangial
Nephrol 2: 858 – 867, 2007 remodeling in light chain-related glomerular damage. Kidney Int 68:
17. Herrera GA, Turbat-Herrera EA: Ultrastructural immunolabeling in 1590 –1603, 2005
the diagnosis of monoclonal light-and heavy-chain-related renal dis- 30. Keeling J, Herrera GA: An in vitro model of light chain deposition
eases. Ultrastruct Pathol 34: 161–173, 2010 disease. Kidney Int 75: 634 – 645, 2009
18. Hassoun H, Flombaum C, D’Agati VD, Rafferty BT, Cohen A, 31. Cheng IK, Ho SK, Chan DT, Ng WK, Chan KW: Crescentic nodular
Klimek VM, Boruchov A, Kewalramani T, Reich L, Nimer SD, glomerulosclerosis secondary to truncated immunoglobulin alpha
Comenzo RL: High-dose melphalan and auto-SCT in patients with heavy chain deposition. Am J Kidney Dis 28: 283–288, 1996
monoclonal Ig deposition disease. Bone Marrow Transplant 42: 32. Hendershot L, Bole D, Kohler G, Kearney JF: Assembly and secre-
405– 412, 2008 tion of heavy chains that do not associate posttranslationally with
19. Firkin F, Hill PA, Dwyer K, Gock H: Reversal of dialysis-dependent immunoglobulin heavy chain-binding protein. J Cell Biol 104: 761–
renal failure in light-chain deposition disease by autologous periph- 767, 1987
eral blood stem cell transplantation. Am J Kidney Dis 44: 551–555, 33. Khamlichi AA, Aucouturier P, Preud’homme JL, Cogne M: Structure
2004 of abnormal heavy chains in human heavy-chain-deposition disease.
20. Ronco P, Plaisier E, Mougenot B, Aucouturier P: Immunoglobulin Eur J Biochem 229: 54 – 60, 1995
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Section 21: CASE PRESENTATION 1⫹ lower extremity edema. Urinalysis showed 3⫹ protein and
10 to 20 red blood cells per high-power field. All serologies are
A 55-year-old African American woman presents with in- negative or normal, including anti-nuclear antibody, anti-DNA
creasing serum creatinine, hematuria, and proteinuria. Past antibody, anti-neutrophil cytoplasmic antibody, rheumatoid
medical history is significant for a 20-year history of type 2 factor, hepatitis B surface antigen, hepatitis C antibody, and C3
diabetes (without retinopathy) and longstanding hypertension. and C4. There is no evidence of a monoclonal spike on serum
Six months prior to biopsy, the patient had a serum creatinine protein electrophoresis or urine protein electrophoresis with
of 1.6 mg/dl. At the time of presentation, the patient has a immunofixation. The patient’s medications include meto-
creatinine of 2.6 mg/dl and a 24-hour urine protein of 2.4 g. prolol, furosemide, glipizide, and esomeprazole. A renal bi-
Physical examination is notable for a BP 155/79 mmHg and opsy is performed.
Figure 1. Figure 2.
incidence of renal insufficiency at the time of diagnosis, a mean of 44, 20, and 7 months, respectively. These
most cases of FGN exhibit at least mild tubular atrophy, outcomes were significantly worse than for the MES
interstitial fibrosis, and interstitial inflammation. (80 months) and MGN (87 months) groups. In this
Immunofluorescence. With rare exceptions, the study, 45% of patients reached ESRD at a mean
fibrillar immune deposits of FGN stain dominantly follow-up time of 23 months, including 18% of the
with antisera to IgG. The texture of the staining is patients with a presenting creatinine level of ⬍1.5
often “smudged,” resembling the staining quality in mg/dl and 72% of the patients with a presenting
amyloidosis. This smudgy appearance differs from the creatinine level of ⬎2.0 mg/dl. In multivariate analy-
typical granular or linear staining observed in other sis, only serum creatinine at biopsy and the degree of
immune complex–mediated processes. Early studies interstitial fibrosis significantly correlated with out-
on the IgG subtypes found that the deposits of FGN come. Similarly, Iskander et al. (1) and Fogo et al. (2)
were mainly or exclusively composed of IgG4 (1). A reported a 52 and 44% incidence of ESRD over a
more recent series noted oligotypic deposits staining mean follow-up of 24 months.
for both IgG1 and IgG4 in the majority of cases, with Fibrillary Glomerulonephritis and HCV Infection.
occasional cases staining for only IgG1 or IgG4 (3). HCV infection is present in a subset of patients with
These findings support the presence of subtype-re- FGN (5), including up to 17% of patients in the largest
stricted IgG deposits, which may predispose to fibril- available series (3). Cases of HCV-associated FGN
logenesis. The fibrillar deposits in FGN are usually exhibit no distinguishing pathologic features when
polyclonal, with staining for both and light chains; compared with other FGN, and serum complement
however, occasional cases show light-chain restriction levels are typically normal. Of note, there is a single
(typically ). Staining for complement component C3 case of FGN and concurrent HCV infection in which
is typically strong, whereas staining for C1q is less proteinuria and renal insufficiency improved after
frequent and weaker. Low-intensity positivity for IgM treatment of HCV with ␣-IFN, supporting a pathoge-
is found in approximately half of cases, whereas IgA is netic association (6).
rarely identified (1,3).
Differential Diagnosis. Because of its broad range of
Electron Microscopy. Electron microscopy is re- histologic appearances, FGN is easily confused with
quired to establish a definitive diagnosis of FGN. The other glomerulonephritides. Immunofluorescence helps
diagnostic ultrastructural feature is the presence of to narrow the differential diagnosis to entities associated
randomly oriented fibrils typically measuring between with IgG-dominant staining; however, definitive diagno-
16 and 24 nm in diameter. The distribution of the sis requires electron microscopy. At the ultrastructural
deposits mimics the findings by light microscopy, with level, FGN must be distinguished from other diseases
mesangial fibrils predominating in the MES pattern that have organized fibrillar deposits, such as amyloido-
and mesangial and capillary wall fibrils seen in the sis, ITG, and, rarely, cryoglobulinemic glomerulonephri-
MPGN, DPGN, and MGN patterns. In the MPGN and tis and lupus nephritis. The fibrillar deposits of FGN
DPGN patterns, the fibrils typically diffusely permeate typically measure 16 to 24 nm in diameter, which is
the GBM, extending from the subendothelial to sub- approximately twice the diameter of amyloid fibrils (8 to
epithelial region and obscuring the lamina densa. In 12 nm). Importantly, Congo red and thioflavin T staining
some cases, the fibrils are admixed with nonorganized are positive in amyloidosis and negative in FGN. In
granular electron-dense deposits. Extraglomerular de- addition, immunofluorescence shows light-chain restric-
posits are extremely unusual (4). tion and negative staining for IgG in AL amyloidosis,
Clinical-Pathologic Correlation. Rosenstock et al. whereas FGN deposits are composed of polyclonal IgG
(3) examined the relationship among histologic pat- and complement.
terns of FGN, presenting features, and clinical out- ITG is a rare glomerular disease that can be con-
comes. The MPGN, DPGN, and DS patterns are as- fused with FGN because of its similar clinical presenta-
sociated with a higher serum creatinine level and a tion and similar light microscopic and immunofluorescence
greater degree of proteinuria at the time of biopsy findings. Differentiation requires electron microscopy,
compared with the MES and MGN patterns. Similarly, which reveals larger 30- to 50-nm microtubular depos-
the MPGN, DPGN, and DS patterns reached ESRD at its with a hollow center in ITG. Unlike FGN, the
404 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Figure 6. Comparison of the organized deposits in amyloidosis (A), FGN (B), and ITG (C). Magnification, ⫻80,000.
microtubular deposits of ITG often align to form contain IgG, , , C3, and amyloid P component (8).
parallel stacks in the subepithelial and/or subendothe- Amyloid P component is a normal serum glycoprotein
lial regions without infiltrating the GBM itself. Figure that can be found deposited in all types of amyloidosis.
6 shows the characteristic fibrils of amyloidosis (A), As in amyloidosis, it is likely that amyloid P plays a
FGN (B), and ITG (C), all photographed at ⫻80,000 role in fibrillogenesis and resistance to proteolysis.
magnification. In many cases, the IgG deposits of ITG The ability of cryoglobulins to produce orga-
are monoclonal, staining for a single light chain and a nized glomerular deposits of immunoglobulins raises
single IgG subtype. The distinction between FGN and the question whether the fibrillar deposits in FGN
ITG is clinically relevant because of the greater asso- represent a type of “slow” cryoglobulin. Rostango et
ciation of ITG with lymphoproliferative disorders, al. (9) studied a patient who had biopsy-proven FGN
monoclonal gammopathy, and hypocomplementemia. and whose serum formed a fibrillar precipitate after
Cryoglobulinemic glomerulonephritis occasionally prolonged cold storage at 4°C. Unlike cryoglobulin
exhibits deposits with an organized fibrillar substructure. precipitates, the fibrils did not dissolve on rewarming.
These deposits are more commonly microtubular and Using immunoelectron microscopy, amino acid se-
thus more likely to be confused with ITG than FGN. quencing, and immunoblotting, they demonstrated that
Therefore, cryoglobulin studies should be performed to the fibrils were composed of immunoglobulins, amy-
exclude the possibility of cryoglobulinemic glomerulo- loid P, and fibronectin. Fibronectin is found in plasma
nephritis in cases of FGN and ITG. In lupus nephritis, and extracellular matrix, shows a wide range of li-
organized fibrillar or microtubular structures may be seen gand-binding interactions, and is known to be associ-
focally in otherwise granular deposits. This finding prob- ated with immune complexes and cryoprecipitates.
ably reflects the presence of type 3 (mixed) cryoglobulins Study of additional cases is needed to determine the
in some patients with lupus nephritis. Electron micros- specificity of these findings for FGN.
copy is required to distinguish the MGN pattern of FGN The immunoelectron microscopic studies support
from membranous nephropathy (7). that IgG is an integral component of the fibrillary depos-
A variety of disorders of extracellular matrix also its. On the basis of the restricted IgG subtype staining, it
manifest organized fibrillar glomerular deposits, includ- has been proposed that the deposits may represent an
ing collagen type 3 glomerulopathy, fibronectin glomeru- oligoclonal immune response to an antigenic challenge
lopathy, nail-patella syndrome, and fibrillosis of scarring (e.g., to viral infection in the case of HCV) and that their
(as seen in diabetic nodules and nonspecific glomerulo- homogeneous composition favors fibrillogenesis.
sclerosis). These entities can be differentiated from FGN
on the basis of their negative staining for IgG and the References
collagenous appearance of the deposits. 1. Iskandar SS, Falk RJ, Jennette JC: Clinical and pathologic features of
fibrillary glomerulonephritis. Kidney Int 42: 1401–1407, 1992
Etiology and Pathogenesis 2. Fogo A, Qureshi N, Horn RG: Morphologic and clinical features of
The pathogenesis of FGN is poorly understood, fibrillary glomerulonephritis versus immunotactoid glomerulopathy.
Am J Kidney Dis 22: 367–377, 1993
in part because of the absence of an animal model. 3. Rosenstock JL, Markowitz GS, Valeri AM, Sacchi G, Appel GB,
Immunoelectron microscopy has shown that the fibrils D’Agati VD: Fibrillary and immunotactoid glomerulonephritis: Dis-
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 405
tinct entities with different clinical and pathologic features. Kidney Int 7. Muller-Hocker J, Weiss M, Sitter M, Samtleben W: Fibrillary glomer-
63: 1450 –1461, 2003 ulonephritis mimicking membranous nephropathy: A diagnostic pit-
4. Adeyi OA, Sethi S, Rennke HG: Fibrillary glomerulonephritis: A fall. Pathol Res Pract 205: 265–271, 2009
report of 2 cases with extensive glomerular and tubular deposits. Hum 8. Yang GC, Nieto R, Stachura I, Gallo GR: Ultrastructural immunohis-
Pathol 32: 660 – 663, 2001 tochemical localization of polyclonal IgG, C3, and amyloid P compo-
5. Markowitz GS, Cheng JT, Colvin RB, Trebbin WM, D’Agati VD: nent on the Congo-red negative amyloid-like fibrils of fibrillary
Hepatitis C viral infection is associated with fibrillary glomerulone- glomerulopathy. Am J Pathol 141: 409 – 419, 1992
phritis and immunotactoid glomerulopathy. J Am Soc Nephrol 9: 9. Rostango A, Vidal R, Asok K, Chuba J, Niederman G, Gold L,
2244 –2252, 1998 Frangione B, Ghiso J, Gallo G: Fibrillary glomerulonephritis related to
6. Coroneos E, Truong L, Olivero J: Fibrillary glomerulonephritis associated serum fibrillar immunoglobulin-fibronectin complexes. Am J Kidney
with hepatitis C viral infection. Am J Kidney Dis 29: 132–135, 1997 Dis 28: 676 – 684, 1996
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Section 22: CASE PRESENTATION reveals 2⫹ peripheral edema and BP of 130/90 mmHg.
Urinalysis shows 4⫹ protein, more than five red blood cells
A 57-year-old man with a history of hypertension for 25 per high-power field, more than five white blood cells per
years, 40 pack-years of smoking, hepatitis C infection for 20 high-power field, but no cellular casts. Laboratory evalua-
years, and type 2 diabetes for 10 years presents with slowly tion reveals hemoglobin A1c of 7.4, serum albumin level of
progressive renal failure and worsening proteinuria. He was 2.9 g/dl, positive hepatitis C antibody, negative hepatitis B
treated briefly with interferon 5 years earlier but is currently surface antigen, negative anti-nuclear antibody, negative
on no antiviral medications. Serum creatinine level rose anti-neutrophil cytoplasmic antibody, negative cryoglobu-
from 2.5 mg/dl 10 years ago to 4.0 mg/dl at presentation. lins, normal C3 and C4, and no monoclonal spike on serum
Proteinuria rose from 6.0 g/day 3 months earlier to 10.0 protein electrophoresis or urinary protein electrophoresis.
g/day. Medications include valsartan, hydrochlorothiazide, Hepatitis C viral titer is low, and liver function tests are
doxazosin, insulin, and esomeprazole. Physical examination normal. Renal biopsy is performed.
Figure 1. Figure 2.
Figure 3. Figure 4.
What is the BEST diagnosis? accounting for ⬎40% of all new cases (1). Type 1
A. Hepatitis C–associated membranoproliferative glomer- diabetes, which is caused by autoimmune destruction
ulonephritis of the pancreatic islets, typically has its onset in
B. Smoking-related glomerulopathy childhood and manifests insulin deficiency. Type 2
C. Light-chain deposition disease diabetes is more common in older adults but is also
D. Nodular diabetic glomerulosclerosis (NDG) increasingly recognized in young adults and children
E. Hypertensive arterionephrosclerosis with metabolic syndrome (including insulin resistance,
obesity, hyperlipidemia, and hypertension). The risk
Answer for developing nephropathy in type 1 and type 2
A representative glomerulus appears enlarged diabetes is similar. It has been estimated that approx-
with global nodular mesangial sclerosis, mild mesan- imately 30 to 40% of patients with type 1 and type 2
gial hypercellularity, and uniform thickening of the diabetes will develop nephropathy. Renal disease be-
glomerular basement membranes (GBMs; Figure 1; gins with microalbuminuria (albumin excretion 30 to
periodic acid–Schiff [PAS], ⫻400). The adjacent pre- 300 mg/24 h). Approximately 15% of individuals with
glomerular arteriole (arrow) shows mural hyalinosis. microalbuminuria develop heavier proteinuria and
Another glomerulus contains an eccentric large mes- progressive renal insufficiency (2– 4). The cumulative
angial nodule composed of acellular lamellated PAS- incidence of ESRD as a result of diabetic nephropathy
positive extracellular matrix (Figure 2, PAS, ⫻400). is 50% in type 1 diabetes (5) and 3 to 11% in type 2
Bowman’s capsule appears thickened and lamellated. diabetes (6). However, most cases of ESRD are due to
Silver stain reveals global silver-positive mesangial type 2 diabetes, reflecting its much greater prevalence.
sclerosis with segmental capillary microaneurysm for- NDG, characterized by acellular nodular accu-
mation (arrowheads). Mesangial interposition sur- mulations of mesangial matrix (also termed Kimmel-
rounds some of the nodules. (Figure 3, Jones methe- stiel-Wilson nodules), is the histologic hallmark of
namine silver, ⫻400). Forty percent of glomeruli were diabetic renal disease and is present in up to 40% of
globally sclerotic with prominent hyalinosis. Tubular renal biopsies from patients with diabetes (7). The
atrophy and interstitial fibrosis occupied approxi- other major pattern of glomerular involvement in di-
mately 60% of the cortex. An arteriole shows severe abetic nephropathy is diffuse mesangial sclerosis
hyalinosis (Figure 4, Jones methenamine silver, (without nodularity), which is termed “diffuse diabetic
⫻400). Immunofluorescence (IF) microscopy reveals glomerulosclerosis.” Both diffuse and nodular forms
diffuse linear staining of GBMs and tubular basement of diabetic nephropathy also exhibit GBM thickening,
membranes (TBMs) with antisera to albumin (1 to 2⫹; tubulointerstitial scarring, arteriosclerosis, and arteri-
Figure 5, ⫻100) and IgG (trace to 1⫹; data not olar hyalinosis. Similar glomerular lesions are found
shown), without significant positivity for other im- in patients with type 1 and type 2 diabetes. In addition,
mune reactants. By electron microscopy (EM), the nondiabetic kidney disease (NDKD) is not uncom-
mesangium is expanded by nodular accumulations of mon, either alone or superimposed on diabetic ne-
matrix material that compress the mesangial cells and phropathy (see NDKD in Patients with Diabetes sec-
narrow the glomerular capillaries, associated with uni- tion). NDKD is more frequently detected in patients
form thickening of GBMs (Figure 6, electron micro- with type 2 diabetes, reflecting the greater prevalence
graph, ⫻4000). No immune-type electron-dense de- of comorbid conditions, such as systemic hyperten-
posits or endothelial tubuloreticular inclusions were sion, cardiovascular disease, and older age.
identified. These findings are consistent with nodular Compared with diffuse diabetic glomerulosclerosis,
diabetic glomerulosclerosis (NDG) (answer D). It is NDG is characterized by longer duration of diabetes,
possible that longstanding hypertension and smoking heavier proteinuria, greater renal insufficiency, more fre-
contributed to the vascular disease and glomeruloscle- quent diabetic retinopathy, more severe chronic patho-
rosis. logic changes, and worse prognosis (8,9), consistent with
a more advanced stage of diabetic nephropathy (9).
Nodular Diabetic Glomerulosclerosis Nonetheless, it remains uncertain whether NDG evolves
Diabetic kidney disease (diabetic nephropathy) is from diffuse mesangial sclerosis or arises ab initio via
the single leading cause of ESRD in the United States, distinct pathogenetic pathways.
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 409
tension closely resembles NDG but usually has more onset of full nephrotic syndrome and whose biopsies
neovascularization by small capillaries within the mes- show only mild diabetic changes with severe foot
angial nodules and extending outward from the glo- process effacement and podocyte microvillous trans-
merular hilus (16). formation (24). IgA-dominant post-staphylococcal in-
NDKD in Patients with Diabetes. NDKD, either fectious glomerulonephritis is not uncommon in older
alone or superimposed on diabetic nephropathy, is not patients with diabetes and cellulitis or other suppura-
uncommon in patients with diabetes. The likelihood of tive infections (22). Tubulointerstitial diseases include
finding NDKD depends on clinical variables that in- acute tubular necrosis, acute interstitial nephritis, py-
elonephritis, and myeloma cast nephropathy, among
fluence the decision to biopsy, such as short duration
others. Vascular diseases include cholesterol emboli-
of diabetes, absence of retinopathy, active urine sedi-
zation and thrombotic microangiopathy.
ment, hypocomplementemia or other serologic abnor-
malities, sudden onset of heavy proteinuria, or rapidly
deteriorating kidney function. In a biopsy study of 34
Pathogenesis
The pathogenesis of NDG is multifactorial and
patients with type 2 diabetes and microalbuminuria,
complex. Genetic predisposition seems to play a role
41% showed absent or only mild diabetic changes and
in both type 1 and type 2 diabetes. Elevation in
disproportionately severe tubulointerstitial or vascular
intracellular glucose levels leads to glucose oxidation
disease, which correlated with poor glycemic control
in the tricarboxylic acid cycle, generation of more
(18). In a series of 51 patients with type 2 diabetes and
electron donors to the electron transport chain, and
⬎300 mg/d albuminuria, 68% of biopsies showed
overproduction of mitochondrial superoxide (25).
diabetic glomerulosclerosis, 18% showed normal glo-
These perturbations in turn activate a number of del-
merular structure, and 14% had NDKD (19). Among eterious metabolic pathways, including protein kinase
233 unselected patients who had type 2 diabetes and C, the polyol pathway, and the hexosamine pathway,
underwent renal biopsy, 28% had pure diabetic ne- all of which have been linked to hyperfiltration, endo-
phropathy, 19% had concurrent diabetic nephropathy thelial injury, microvascular disease, and albuminuria.
and NDKD, and 53% had NDKD alone, including a Importantly, hyperglycemia causes nonenzy-
broad variety of glomerular diseases (20). In an un- matic glycation of extracellular matrix proteins and
published series of 168 consecutive native kidney lipids, which are converted through the Maillard reac-
biopsies from patients with type 2 diabetes at our tion into Schiff bases, Amadori products, and ulti-
institution (74% of whom had nephrotic-range protein- mately advanced glycation end products (AGE), pro-
uria), diabetic nephropathy alone was seen in 86 moting irreversible cross-linking of matrix proteins.
(51%) cases, diabetic nephropathy with superimposed Although the GBMs become thickened, the reduction
NDKD in 39 (23%) cases, and NDKD alone in 43 in heparan sulfate proteoglycan content is thought to
(26%) cases. Clinical correlates of NDKD alone in- be important in the induction of albuminuria (26).
cluded shorter duration of diabetes (5 versus 10 years) AGE have been identified by immunohistochemistry
and less frequent diabetic retinopathy (17 versus in the thickened renal basement membranes of human
38%). Cases with combined diabetic nephropathy and diabetic nephropathy (27). Interaction with RAGE, a
NDKD were more likely to have active urinary sedi- multiligand receptor for AGE, in turn activates proin-
ment and rapid progression than those with diabetic flammatory and profibrotic pathways. RAGE is ex-
nephropathy alone. pressed on podocytes and glomerular endothelial cells,
NDKD in patients with diabetes comprises a where interaction with AGE generates podocyte oxi-
broad spectrum of glomerular, tubulointerstitial, and dative stress, endothelial dysfunction, proinflamma-
vascular diseases (21). Glomerular diseases include tory and profibrotic cytokine production, altered glo-
minimal change disease, acute postinfectious glomer- merular filtration barrier, and eventual podocyte
ulonephritis (22), pauci-immune crescentic glomeru- apoptosis (28).
lonephritis (23), IgA nephropathy, membranous ne- The hyperglycemic milieu also activates pro-
phropathy, and fibrillary glomerulonephritis, to name renin to generate angiotensin II (29). In addition to its
a few. Minimal change disease should be considered hemodynamic effects, angiotensin II promotes local
in patients who have diabetes and present with abrupt profibrotic cytokine production and glomerular matrix
412 Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
expansion. Recent evidence has focused on the roles with retinopathy. The Collaborative Study Group. Nephrol Dial
Transplant 13: 2547–2552, 1998
of high fructose intake and elevated uric acid in the 9. Hong D, Zheng T, Jia-qing S, Jian W, Zhi-hong L, Lei-shi L: Nodular
stimulation of NF-B–induced endothelial injury, ma- glomerular lesion: A later stage of diabetic nephropathy? Diabetes
trix production, and systemic hypertension in patients Res Clin Pract 78: 189 –195, 2007
10. Fioretto P, Caramori ML, Mauer M: The kidney in diabetes: Dynamic
with type 2 diabetes (30). pathways of injury and repair. The Camillo Golgi Lecture 2007.
The pathogenesis of nodules is thought to in- Diabetologia 51: 1347–1355, 2008
volve repeated episodes of mesangiolysis and endo- 11. Gall MA, Rossing P, Skott P, Damsbo P, Vaag A, Bech K, Dejgaard
A, Lauritzen M, Lauritzen E, Hougaard P, Beck-Nielsen H, Parving
thelial injury and repair, as evidenced by their frequent HH: Prevalence of micro- and macroalbuminuria, arterial hyperten-
association with microaneurysm formation, entrapped sion, retinopathy and large vessel disease in European type 2 (non-
red blood cells and fibrin, and increased glomerular insulin-dependent) diabetic patients. Diabetologia 34: 655– 661,
1991
expression of plasminogen activator inhibitor 1 (31). 12. Mogensen CE, Schmitz O: The diabetic kidney: From hyperfiltration
In a mouse model of diabetic nephropathy, control of and microalbuminuria to end-stage renal failure. Med Clin North Am
systemic BP ameliorated the formation of NDG but 72: 1465–1492, 1988
13. Tervaert TW, Mooyaart AL, Amann K, Cohen AH, Cook HT,
not diffuse mesangial sclerosis (32). Of note, reversal Drachenberg CB, Ferrario F, Fogo AB, Haas M, de Heer E, Joh K,
of NDG has been demonstrated after pancreas trans- Noel LH, Radhakrishnan J, Seshan SV, Bajema IM, Bruijn JA:
plantation, indicating the potential for regression of Pathologic classification of diabetic nephropathy. J Am Soc Nephrol
21: 556 –563, 2010
established diabetic lesions upon sustained euglycemia
14. Gallo GR: Elution studies in kidneys with linear deposition of
(33). Progression of diabetic glomerulosclerosis in- immunoglobulin in glomeruli. Am J Pathol 61: 377–394, 1970
volves podocyte loss and adhesions to Bowman’s 15. Gonul II, Gough J, Jim K, Benediktsson H: Glomerular mesangial
fibrillary deposits in a patient with diabetes mellitus. Int Urol Neph-
capsule. Adhesions often develop first at the tubular
rol 38: 767–772, 2006
pole, producing atubular glomeruli that have lost com- 16. Nasr SH, D’Agati VD: Nodular glomerulosclerosis in the nondiabetic
munication with the proximal tubules (34). Lesions smoker. J Am Soc Nephrol 18: 2032–2036, 2007
17. Markowitz GS, Lin J, Valeri AM, Avila C, Nasr SH, D’Agati VD:
often progress through stages of segmental to global
Idiopathic nodular glomerulosclerosis is a distinct clinicopathologic
glomerulosclerosis. Similar to other chronic renal dis- entity linked to hypertension and smoking. Hum Pathol 33: 826 – 835,
eases, irreversible podocyte stress and depletion are 2002
18. Fioretto P, Mauer M, Brocco E, Velussi M, Frigato F, Muollo B,
now recognized as important mediators of progressive
Sambataro M, Abaterusso C, Baggio B, Crepaldi G, Nosadini R:
glomerulosclerosis in diabetic nephropathy (35,36). Patterns of renal injury in NIDDM patients with microalbuminuria.
Diabetologia 39: 1569 –1576, 1996
19. Christensen PK, Larsen S, Horn T, Olsen S, Parving HH: Causes of
References
1. US Renal Data System: 2010 Annual Data Report: Atlas of Chronic albuminuria in patients with type 2 diabetes without diabetic retinop-
Kidney Disease and End-Stage Renal Disease, Bethesda, National athy. Kidney Int 58: 1719 –1731, 2000
Institute of Diabetes and Digestive and Kidney Diseases, 2010 20. Pham TT, Sim JJ, Kujubu DA, Liu IL, Kumar VA: Prevalence of
2. Hovind P, Tarnow L, Rossing P, Jensen BR, Graae M, Torp I, Binder nondiabetic renal disease in diabetic patients. Am J Nephrol 27:
C, Parving HH: Predictors for the development of microalbuminuria 322–328, 2007
and macroalbuminuria in patients with type 1 diabetes: Inception 21. Gambara V, Mecca G, Remuzzi G, Bertani T: Heterogeneous nature
cohort study. BMJ 328: 1105, 2004 of renal lesions in type II diabetes. J Am Soc Nephrol 3: 1458 –1466,
3. Amin R, Widmer B, Prevost AT, Schwarze P, Cooper J, Edge J, 1993
Marcovecchio L, Neil A, Dalton RN, Dunger DB: Risk of microalbu- 22. Nasr SH, Share DS, Vargas MT, D’Agati VD, Markowitz GS: Acute
minuria and progression to macroalbuminuria in a cohort with child- poststaphylococcal glomerulonephritis superimposed on diabetic glo-
hood onset type 1 diabetes: Prospective observational study. BMJ merulosclerosis. Kidney Int 71: 1317–1321, 2007
336: 697–701, 2008 23. Nasr SH, D’Agati VD, Said SM, Stokes MB, Appel GB, Valeri AM,
4. Adler AI, Stevens RJ, Manley SE, Bilous RW, Cull CA, Holman RR: Markowitz GS: Pauci-immune crescentic glomerulonephritis super-
Development and progression of nephropathy in type 2 diabetes: The imposed on diabetic glomerulosclerosis. Clin J Am Soc Nephrol 3:
United Kingdom Prospective Diabetes Study (UKPDS 64). Kidney 1282–1288, 2008
Int 63: 225–232, 2003 24. Stokes MB, Kwakye J, D’Agati VD: Nephrotic syndrome and ARF
5. Krolewski AS, Warram JH, Christlieb AR, Busick EJ, Kahn CR: The in a diabetic patient. Am J Kidney Dis 41: 1327–1333, 2003
changing natural history of nephropathy in type I diabetes. Am J Med 25. Brownlee M: The pathobiology of diabetic complications: A unifying
78: 785–794, 1985 mechanism. Diabetes 54: 1615–1625, 2005
6. Mogensen CE, Schmitz A, Christensen CK: Comparative renal 26. Vernier RL, Steffes MW, Sisson-Ross S, Mauer SM: Heparan sulfate
pathophysiology relevant to IDDM and NIDDM patients. Diabetes proteoglycan in the glomerular basement membrane in type 1 diabe-
Metab Rev 4: 453– 483, 1988 tes mellitus. Kidney Int 41: 1070 –1080, 1992
7. Wirta O, Helin H, Mustonen J, Kuittinen E, Savela T, Pasternack A: 27. Tanji N, Markowitz GS, Fu C, Kislinger T, Taguchi A, Pischetsrieder
Renal findings and glomerular pathology in diabetic subjects. M, Stern D, Schmidt AM, D’Agati VD: Expression of advanced
Nephron 84: 236 –242, 2000 glycation end products and their cellular receptor RAGE in diabetic
8. Schwartz MM, Lewis EJ, Leonard-Martin T, Lewis JB, Batlle D: nephropathy and nondiabetic renal disease. J Am Soc Nephrol 11:
Renal pathology patterns in type II diabetes mellitus: Relationship 1656 –1666, 2000
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 413
28. D’Agati V, Schmidt AM: RAGE and the pathogenesis of chronic lointerstitial injury, in diabetic eNOS knockout mice. Am J Pathol
kidney disease. Nat Rev Nephrol 6: 352–360, 2010 174: 1221–1229, 2009
29. Nguyen G, Danser AH: Prorenin and (pro)renin receptor: A review of 33. Fioretto P, Steffes MW, Sutherland DE, Goetz FC, Mauer M:
available data from in vitro studies and experimental models in Reversal of lesions of diabetic nephropathy after pancreas transplan-
rodents: Exp Physiol 93:557–563, 2008 tation. N Engl J Med 339: 69 –75, 1998
30. Johnson RJ, Perez-Pozo SE, Sautin YY, Manitius J, Sanchez-Lozada 34. Najafian B, Kim Y, Crosson JT, Mauer M: Atubular glomeruli and
LG, Feig DI, Shafiu M, Segal M, Glassock RJ, Shimada M, Roncal glomerulotubular junction abnormalities in diabetic nephropathy.
C, Nakagawa T: Hypothesis: Could excessive fructose intake and uric J Am Soc Nephrol 14: 908 –917, 2003
acid cause type 2 diabetes? Endocr Rev 30: 96 –116, 2009 35. Toyoda M, Najafian B, Kim Y, Caramori ML, Mauer M: Podocyte
31. Paueksakon P, Revelo MP, Ma LJ, Marcantoni C, Fogo AB: Mi- detachment and reduced glomerular capillary endothelial fenes-
croangiopathic injury and augmented PAI-1 in human diabetic ne- tration in human type 1 diabetic nephropathy. Diabetes 56: 2155–
phropathy. Kidney Int 61: 2142–2148, 2002 2160, 2007
32. Kosugi T, Heinig M, Nakayama T, Connor T, Yuzawa Y, Li Q, 36. Pagtalunan ME, Miller PL, Jumping-Eagle S, Nelson RG, Myers BD,
Hauswirth WW, Grant MB, Croker BP, Campbell-Thompson M, Rennke HG, Coplon NS, Sun L, Meyer TW: Podocyte loss and
Zhang L, Atkinson MA, Segal MS, Nakagawa T: Lowering blood progressive glomerular injury in type II diabetes. J Clin Invest 99:
pressure blocks mesangiolysis and mesangial nodules, but not tubu- 342–348, 1997
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Examination Questions
Instructions to obtain 12.0 AMA PRA Category 1 CreditsTM
Date of Original Release: July 2011
Examination Available Online: on or before Monday, July 18, 2011
Audio Files Available: There are no audio files for this issue.
CME Credit Eligible Through: June 30, 2012
Answers: Correct answers with explanations will be posted on the ASN website in July 2012 when the issue is archived.
UpToDate Links Active: July and August 2011
Core Nephrology question links active: No core questions for this issue
Target Audience: Nephrology Board and recertification candidates, practicing nephrologists, and internists.
Method of Participation:
● Read the syllabus that is supplemented by original articles in the reference lists, and complete the online self-assessment
examination.
● Examinations are available online only after the first week of the publication month. There is no fee. Each participant is
allowed two attempts to pass the examination (⬎75% correct) for CME credit.
● Upon completion, review your score and incorrect answers.
● Your CME certificate can be printed immediately after completion.
● Answers and explanations are provided with a passing score and/or after the second attempt.
● CME Credit will be posted to your transcript within 48 hours after checking the attestation box.
Instructions to Access the Online Examination and Evaluation:
● Go to the ASN website: www.asn-online.org.
● Click the CME tab at the top of the homepage.
● Click the ASN CME Center button on the left side of the page.
● Click on to the ASN CME Center icon.
● Login to the ASN website.
● Select Claim Credits for the NephSAP topic-activity you would like to complete.
● Complete the NephSAP examination.
● Complete the evaluation.
● Enter the number of CME credits commensurate with your participation in the activity.
● Check the box attesting that you have completed this activity.
● You can print your CME certificate immediately.
● CME credit will be posted to your transcript within 48 hours.
● View or print your full transcript anytime at “My CME Center.”
Instructions to Obtain American Board of Internal Medicine (ABIM) Maintenance of Certification
(MOC) Points:
Each issue of NephSAP provides 10 MOC points. Respondents must meet the following criteria:
● Be certified by ABIM in internal medicine and/or nephrology and must be enrolled in the ABIM–MOC program
via the ABIM website (www.abim.org).
● Take the self-assessment examination within the timeframe specified in this issue of NephSAP.
● Designate the issue for MOC points by clicking on the MOC link on the CME certificate page after passing the examination.
You will be leaving the ASN site and transferring the information directly to the ABIM in real-time.
● Provide your ABIM Certificate ID number and your date of birth.
● You will receive a confirmation message from the ABIM indicating the receipt of your information.
MOC points will be applied to only those ABIM candidates who have enrolled in the program. It is your responsibility to complete
the ABIM MOC enrollment process.
414
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011
Volume 10, Number 4, July 2010 —Renal Pathology and Examination Questions
mesangial proliferative glomerulonephritis. diffuse tubular injury and atypical, distal tubular
Electron microscopy reveals global mesangial casts that stain brightly with the hematoxylin
deposits. and eosin stain, appear pale with the periodic
The mesangial deposits seen by immunoflu- acid–Schiff stain, and are associated with adher-
orescence are likely to stain dominantly for ent monocytes and rare giant cells.
which ONE of the following immune reac- Which ONE of the following pathologic
tants? findings is MOST likely to be seen upon
A. IgG further evaluation?
B. IgM A. By immunofluorescence, the tubular casts
C. IgA stain similarly for and light chains.
D. C3 B. By immunofluorescence, the tubular casts
E. light chain stain dominantly for a single light chain
( or ) and with minimal intensity for the
reciprocal light chain.
夝 5. An 18-year-old Asian woman is found to have
microhematuria, a 24-hour urine protein of C. Electron microscopy reveals abundant
1.5 g, a serum creatinine of 0.8 mg/dl, an ANA mesangial randomly oriented fibrils with a
titer of 1:80, and a negative anti-DNA antibody. mean diameter of 8 to 12 nm.
Renal biopsy reveals a diffuse mesangial and D. Electron microscopy reveals global subepi-
focal endocapillary proliferative glomerulone- thelial electron-dense deposits.
phritis. By immunofluorescence, the global E. Immunofluorescence reveals linear staining
mesangial and segmental subendothelial depos- for in glomerular basement membranes
its stain co-dominantly for IgG and IgA (each (GBMs) and tubular basement membranes,
3⫹). Thus, the differential diagnosis includes
Bowman’s capsule, and blood vessels.
mainly lupus nephritis and IgA nephropathy
(IgAN).
夝 7. A 62-year-old woman with a 6-month history of
Which ONE of the following findings would
smoldering multiple myeloma is found to have a
favor the diagnosis of IgAN in this patient?
24-hour urine protein of 5 g, serum albumin
A. The ultrastructural finding of multiple en- level of 3.4 g/dl, creatinine of 1.4 mg/dl, and mild
dothelial tubuloreticular inclusions edema. Renal biopsy is performed, and light mi-
B. The serologic finding of normal serum croscopy reveals a nodular pattern of glomerulo-
complement levels sclerosis. Immunofluorescence reveals linear
C. The immunofluorescence finding of tubu- staining for light chain in glomerular and tubular
lar basement membrane and vessel wall basement membranes, Bowman’s capsule, and
deposits blood vessels. Electron microscopy reveals finely
D. The immunofluorescence finding of 2⫹ granular, nonfibrillar, punctate deposits in glomer-
global mesangial positivity for C1q ular and tubular basement membranes, the mesan-
E. The immunofluorescence finding of 2⫹ gium, and vessel walls.
global mesangial positivity for IgM, C3, These findings are diagnostic of which ONE
and C1q (i.e., a “full house”) of the following disease entities?
A. Light chain deposition disease
6. An 82-year-old woman presents with back pain, B. Amyloidosis
hypercalcemia, anemia, and acute renal failure
C. Light chain Fanconi syndrome
with a creatinine level of 5.1 mg/dl. She has a
24-hour urine protein of 4 g. Urinalysis reveals D. Proliferative glomerulonephritis with
1⫹ protein and a bland urine sediment. Renal monoclonal IgG deposits
biopsy is performed. Light microscopy reveals E. Nodular diabetic glomerulosclerosis
Nephrology Self-Assessment Program - Vol 10, No 4, July 2011 417
24. A 39-year-old Caucasian man presents with abrupt image of a representative glomerulus from the
onset of nephrotic syndrome. The light micro- biopsy is provided. Immunofluorescence was
scopic findings are provided. Immunofluorescence negative for all immune reactants, and electron
was negative, and electron microscopy revealed microscopy confirmed the absence of deposits.
complete foot process effacement.
Which ONE of the following is the COR- On the basis of these findings, which ONE of
RECT diagnosis? the following statement is CORRECT regard-
A. Minimal change disease ing the serologic workup on this patient?
B. FSGS, collapsing variant A. The probability of a positive anti-GBM
antibody is ⬎50%.
C. FSGS, cellular variant
B. The probability of a positive ANCA
D. FSGS, tip lesion variant
is ⬎75%.
E. Amyloidosis
C. The probability of a positive ANCA is 98
to 100%.
25. A 79-year-old woman is found to have a creat-
inine level of 4.4 mg/dl, an active urine sediment D. The probability of a positive ANA
with RBC casts, and subnephrotic proteinuria. is ⬎50%.
Renal biopsy is performed while awaiting re- E. The probability of hypocomplementemia
ceipt of the results of serologic studies. An is ⬎75%.
NephSAP
®
Volume 10, Number 4, July 2011
422