2020-Autoimmunity and Organ Damage in SLE
2020-Autoimmunity and Organ Damage in SLE
2020-Autoimmunity and Organ Damage in SLE
Author manuscript
Nat Immunol. Author manuscript; available in PMC 2021 May 20.
Author Manuscript
Abstract
Author Manuscript
Impressive progress has been made over the last several years toward understanding how almost
every aspect of the immune system contributes to the expression of systemic autoimmunity. In
parallel, studies have shed light on the mechanisms that contribute to organ inflammation and
damage. New approaches that address the complicated interaction between genetic variants,
epigenetic processes, sex and the environment promise to enlighten the multitude of pathways that
lead to what is clinically defined as systemic lupus erythematosus. It is expected that each patient
owns a unique ‘interactome’, which will dictate specific treatment.
It took almost 100 years to realize that lupus erythematosus, which was initially thought to
be a skin entity, is a systemic disease that spares no organ and that an aberrant autoimmune
response is involved in its pathogenesis. The involvement of vital organs and tissues such as
Author Manuscript
the brain, blood and the kidney in most patients, the vast majority of whom are women of
childbearing age, impels efforts to develop diagnostic tools and effective therapeutics. The
prevalence ranges from 20 to 150 cases per 100,000 people and appears to be increasing as
the disease is recognized more readily and survival rates improve. In the United States,
people of African, Hispanic or Asian ancestry, as compared to those of other racial or ethnic
groups, tend to have an increased prevalence of systemic lupus erythematosus (SLE) and
greater involvement of vital organs. The 10-year survival rate has increased significantly
over the last 50 years to more than 70%, mostly because of greater awareness of the disease,
the extensive and wiser use of immunosuppressive drugs and a more efficient treatment of
infections, the major cause of death1–3.
Although low levels of autoreactivity and autoimmunity are necessary for lymphocyte
Author Manuscript
selection and, in general, for the regulation of the immune system, in certain individuals,
autoimmunity advances through multiple pathways (reviewed in ref. 4) and leads to organ
inflammation and damage. The diverse mechanisms do not contribute equally to the
expression of disease in all patients with SLE, as will be discussed below. It appears that the
✉
Correspondence should be addressed to G.C.T. [email protected].
Competing interests
The author declares no competing interests.
Reprints and permissions information is available at www.nature.com/reprints.
Editor recognition statement L. A. Dempsey was the primary editor on this article and managed its editorial process and peer review
in collaboration with the rest of the editorial team.
Tsokos Page 2
clinical heterogeneity of the disease is matched by the multiple pathogenic processes, which
Author Manuscript
justifies the call for the development of personalized medicine (Fig. 1).
specific clinical manifestations, pointing strongly to the heterogeneity of the disease. Several
genes linked to the immune response are regulated through long-distance chromatin
interactions10,11. Studies addressing long-distance interactions between gene variants in SLE
are still missing, but, with the advent of new technologies, such studies will emerge.
CD8 locus is methylated in T cells from patients with SLE, resulting in the generation of
CD3+CD4−CD8− T cells, whereas the Il2 locus is hypermethylated, resulting in low
production of IL-2 (reviewed in ref. 12). Delivery of a demethylating agent specifically to
either CD4+ or CD8+ cells in lupus-prone mice suppresses disease expression by enhancing
the expression of FoxP3 and sustaining the expression of CD813.
In SLE T cells, the cyclic AMP response element modifier (CREMα) binds to various
regulatory elements within the CD8 cluster and recruits histone modifiers, including
DNMT3a and the histone methyltransferase G9a, causing stable silencing of CD8A and
CD8B14. A similar process takes place in the Il2 locus, resulting in decreased IL-2
production15. In contrast, STAT3 recruitment to Il10 regulatory regions mediates the
recruitment of histone acetyltransferase p300, resulting in enhanced gene expression16.
Author Manuscript
A large number of microRNAs are suspected to control at least one-third of human mRNA
stability and translation, and, reasonably, they have been studied in SLE. A limited list is
presented in Table 2. MicroRNA serum concentrations may serve as disease biomarkers17,
and the development of antagomirs, used to silence endogenous micoRNAs, may help to
control the disease.
the genetic susceptibility. Better understanding of the biochemical processes involved should
offer unique opportunities to develop new therapeutics, and in a personalized manner.
Environment
Approximately a third of monozygotic twins are clinically concordant for SLE, which
signifies the importance of environmental factors in the expression of the disease. The
environment, including ultraviolet (UV) light, and cross-reactivity between self-antigens and
molecules defined by viruses and other pathogens is important in the pathogenesis of SLE.
Microbes, including innocuous commensal organisms that colonize the gut, skin, nasal
cavities and the vagina, may trigger and sustain autoimmune inflammation in genetically
susceptible hosts. Microbiota have been amply shown to shape the immune response,
including the development of T helper type 1 (TH1), TH2 and regulatory T (Treg) cells and
Author Manuscript
Cross-reactivity between bacterial species and autoantigens has been long claimed to
contribute to the expression of disease in susceptible individuals. The bacterium
Propionibacterium propionicum, which encodes an ortholog of the RNA-binding protein
Ro60, was found in cutaneous lesions of patients with subcutaneous lupus erythematosus
and was shown to stimulate memory T cells from patients with SLE19, suggesting a direct
involvement of pathogens in T cell proliferation and the production of autoantibodies.
Segmented filamentous bacteria induce intestinal IL-17-producing TH17 cells20, and gut
microbiota drive autoimmune arthritis by promoting the generation of follicular helper T
(TFH) cells21. Microbiota apparently use Toll-like receptor (TLR) signaling because
TNFAIP3 (A20)-deficient mice, which develop autoimmunity, fail to do so when MyD88 (a
Author Manuscript
central TLR signaling molecule) is genetically deleted, or the mice are treated with
antibiotics22. Microbiota translocate from the gut to the mesenteric lymph nodes, spleen23
and the liver and induce TH17 and TFH cells as well as innate immune pathways, including
the plasmacytoid dendritic cell (pDC)–type I interferon (IFNα/β) axis. Interestingly, certain
microbiota can be found in the liver of patients with SLE or autoimmune hepatitis24.
Microbiota contribute to disease expression through a number of mechanisms, including
molecular mimicry, engagement of the innate immune response and the propagation of
proinflammatory TH17 cells. Accordingly, better understanding of the role of microbiota in
the expression of SLE should reveal simple approaches for controlling autoimmunity though
dietary changes or changing the distribution of microbiota in the gut and elsewhere.
Sex
Author Manuscript
Despite the fact that more than 90% of the people affected with SLE are women, we still do
not have a clear understanding of the causative mechanisms. It is well known that people
with XXX (Klinefelter syndrome) are prone to SLE and that epigenetic changes in certain
pathogenic genes (for example, TNFSF5, encoding CD40L) contribute to disease
expression. Also, six SLE susceptibility loci map to the X chromosome, four of which
Estrogen alters the thresholds for B cell apoptosis and activation, and estrogen receptor α
contributes to T cell–mediated autoimmune inflammation by promoting T cell activation,
and it also promotes lupus in NZB × NZW F1 mice. At the molecular level, estrogen
upregulates CREMα expression, which is known to control the expression of Il2 and Il17
(reviewed in ref. 26). Gene expression analysis revealed a female-biased autoimmunity-
related network driven by the transcription factor VGLL3 that is linked with autoimmune
diseases, including SLE, Sjogren’s syndrome and scleroderma27. While we still do not
understand why women represent the vast majority of people with SLE, new evidence points
to distinct molecular processes and the probable activation of X-chromosome-defined genes
that are genetically linked to SLE.
Author Manuscript
DCs link innate and adaptive immune responses and have been identified in the expression
of SLE, as their uncontrolled activation may drive autoimmunity31. Although the numbers
Author Manuscript
are decreased in the periphery, they are found activated in the inflamed tissues, producing
inflammatory cytokines and helping T and B cells. Immune complexes containing RNA
induce OX40-ligand expression by conventional SLE DCs. Subsequently, they drive the
differentiation of naive and memory CD4+ T cells into TFH cells, which are able to help B
cells32 and impair Treg function33. Conventional DCs in SLE instruct the differentiation of
IgG and IgA plasmablasts and contribute to the formation of ectopic lymphoid structures34.
pDCs are distinguished from conventional DCs by morphology and cell surface markers and
Author Manuscript
are similarly low in the periphery, probably because they lodge inflamed areas. Triggered by
TLR7/9 agonists, they produce IFN type I35 to contribute to disease expression, and
duplication of TLR7 promotes disease36. Specific depletion of pDCs in mice reduces disease
manifestations such as autoantibody production, glomerulonephritis and expression of IFN-
inducible genes37. At the clinical level, targeting of pDCs with a BDCA2 antibody
ameliorates skin disease in patients with SLE38, while chronic triggering of pDCs through
TLR7 and TLR9 renders pDCs resistant to inhibition of the NF-κB pathway and leads to
steroid resistance39.
Marginal zone macrophages surrounding the splenic follicles are crucial for the efficient
clearance of apoptotic cells and for the induction of tolerance to autoantigens. Phagocytosis
of apoptotic cells by splenic marginal zone macrophages requires the megakaryoblastic
leukemia 1 transcriptional coactivator–mediated mechanosensing pathway40. The production
Author Manuscript
Type I IFN, which affects multiple components of the immune system, has been
demonstrated to contribute to the pathogenesis of adult and pediatric SLE43 and to reflect
disease activity (reviewed in detail in ref. 44). Yet type I IFN alone may not be sufficient to
cause disease expression45 and, in some murine strains, may even be beneficial46.
Author Manuscript
Proper processing of apoptotic material involves the activation of the transcription factor aryl
hydrocarbon receptor (AhR) following engagement of TLR9, which leads to a series of
events that suppress inflammation, including the production of IL-10. Deletion of AhR in
myeloid cells causes autoimmunity, and its transcription signature correlates with disease
activity in mice and humans47. This observation may explain why TLR9 has a protective
effect against autoimmunity.
Platelets are activated in patients and mice with SLE through a number of mechanisms,
including the action of immune complexes and contact with injured endothelial cells, and
they display a type I IFN signature48. Once activated, platelets express and release CD40L
and modulate adaptive immunity by activating antigen-presenting cells, including DCs.
Platelets interact with pDCs in patients to increase the secretion of type I IFN by triggering
TLR9 and TLR749. Understanding of the contribution of platelets may reveal adjuvant tools
Author Manuscript
Autoreactive IgE causes basophils to home to lymph nodes, promote TH2 cell differentiation
and enhance the production of self-reactive antibodies that cause lupus-like nephritis in mice
lacking the protein tyrosine kinase Lyn. Patients with SLE with elevated concentrations of
self-reactive IgEs and activated basophils have increased disease activity and active lupus
nephritis50.
Studies in mice have shown, in a definitive manner, the roles of neutrophils, basophils,
Author Manuscript
pDCs, TLR activation and IFN type I production in the expression of SLE. Several of these
contribute directly to organ damage, whereas others instruct, directly or indirectly, the
aberrations of the adaptive immune response. The diversity of the involved pathways
underlines the wide clinical spectrum of the disease, and it is quite possible that each cellular
element contributes to the expression of disease, to varying degrees.
Age-associated B cells (which include IgD−CD27− and CD21lo B cells) have been detected
in human autoimmune disorders, including SLE. Their expansion is controlled by the
transcription factor IRF5, variants of which are linked to SLE, through IL-21 expression and
unique landscape remodeling56. In mice, age-associated B cells are expanded in the absence
of GTPase regulatory proteins (DEF6 and SWAP70), and DEF6 variants have been
identified as conferring increased susceptibility to SLE57.
The SLE molecular signature in B cells appears to become established during the resting
naive phase and is dominated by the enrichment of accessible chromatin motifs for the
transcription factors AP-1 and EGR58, which is facilitated by, probably among other factors,
IFN-γ59. B cells that lack IgD and CD27 are known to be expanded in patients with SLE
Author Manuscript
and to produce autoantibodies. These cells are hyperresponsive to TLR7 agonists and IL-21,
lack the TLR regulator TRAF560 and have features of freshly activated naive B cells61.
Although allergic reactions are not more frequent in people with SLE as compared to normal
subjects, IgE antibodies specific for dsDNA are present in the sera of patients with SLE.
These IgE antibodies bind to the high-affinity FcγRI receptor for IgE, can activate pDCs and
transfer DNA to TLR9 in phagosomes. This activation results in the secretion of substantial
amounts of IFN-α62.
T cells are key players in promoting the autoimmune response by providing help to B cells
and by activating antigen-presenting cells through cytokine release and direct cellular
contact. Additionally, they infiltrate tissues and promote local inflammation. Autoreactive
Author Manuscript
CD4+ T cells are presumed to respond to nucleosomal antigens and, in particular, to peptides
derived from histones63. An interesting T cell subset of unknown pathogenic importance was
recorded in patients with SLE and multiple sclerosis (CXCR3+CD38+CD39+PD-1+HLA-DR
+CD161+KLRG1−CD28+OX40+), which differs from T
FH cells and was first recognized in
the gut of patients with celiac disease by virtue of binding gluten64.
TFH cells promote B cell function and evolve from CD4+ T cells in the presence of IL-6,
IL-21 and inducible T cell costimulatory (ICOS)65. ICOS deficiency protects MRL-lpr mice
from disease66. A CD4+ cell subset that resembles TFH cells is expanded in the peripheral
blood of patients with active SLE65. The ATP-gated ionotropic P2X7 receptor restricts the
Author Manuscript
expansion of aberrant TFH cells, but TFH cells from patients with SLE are resistant to P2X7-
mediated inhibition of cytokine-driven expansion, pointing to a signaling defect67.
−
CXCR5 CXCR3+PD-1+ helper T cells, different from TFH cells, are present in the periphery
and in kidney tissues of people with SLE, and provide help to B cells by producing IL-10
and succinate68.
CD8+ T cell cytotoxic responses are decreased in SLE and contribute to increased rates of
infection7. A CD8+CD38+ T cell population is expanded in the peripheral blood of patients
with SLE. CD8+CD38+ T cells display decreased production of granzymes and perforin and
reduced cytotoxic capacity, and patients with SLE, for whom this population is expanded,
experience infections more frequently. CD38, a marker of T cell exhaustion, is an
ectonucleotidase that degrades NAD and, through the histone methyltransferase EZH2,
Author Manuscript
Treg cells are characterized by constitutive expression of the transcription factor FoxP3 and
high expression of the high-avidity IL-2 receptor α chain (CD25); in humans, some
activated T effector (Teff) cells also transiently express this molecule. The number of Treg
cells is reduced during the early phases of the disease, whereas the CD45RA−FoxP3lo non-
Treg cell population is increased in active SLE72. The realization that Treg cells have higher
Author Manuscript
affinity receptors for IL-2 and, therefore, stronger IL-2 receptor (IL-2R)–mediated signaling
than Teff cells, suggested that administration of IL-2 at a lower dose than that used for Teff
cells should promote Treg cell expansion and function. Administration of low-dose IL-2 to
lupus-prone mice expanded the population of Treg cells and shrank the pool of
CD3+CD4−CD8− IL-17-producing T cells73, which are known to contribute to the
development of lupus nephritis74. Low-dose IL-2 administered to people with SLE has been
reported75 to produce clinical benefit. A caveat to the apparent success of low-dose IL-2 is
evidence that the IL-2–IL-2R–p-STAT5 signaling pathway in SLE T cells is compromised76.
IL-2 has the potential to reverse several pathogenic processes involved in the development of
SLE, including poor Treg cell function, increased IL-17 production, increased TFH cell
activity and the expansion of the population of CD4−CD8− T cells77. The demonstration that
Treg cells contribute to tissue repair78 and the possibility that Treg cells are limited in the
Author Manuscript
kidneys of patients with lupus nephritis79 encourage the consideration of approaches that
enrich Treg cells in the kidneys or other tissues.
The phenotype and function of T cells isolated from patients with SLE has been extensively
studied in the search for clues to explain the pathogenesis of the disease and in an attempt to
identify molecules that can serve as biomarkers and/or therapeutic targets80. These studies
have revealed that, in the context of SLE, T cell function is severely compromised as a result
of a large number of signaling aberrations that distort gene expression profiles and skew the
cellular immune response toward a proinflammatory type (reviewed in refs. 80,81). In brief,
Author Manuscript
CD3-mediated T cell signaling is abnormal in people with SLE, and this is followed by
aberrant expression of kinases, phosphatases, transcription factors, chemokine receptors and
adhesion molecules and the production of chemokines and proinflammatory cytokines (Fig.
2).
Metabolic abnormalities have been recognized in people with SLE and in lupus-prone mice
and have been linked to abnormal T cell function. SLE T cells display increased oxidative
stress, as indicated by the depletion of glutathione (via NADPH loss), metabolic checkpoint
kinase complex mTORC1, glycolysis and glutaminolysis. Inhibition of increased mTORC1,
glycolysis or glutaminolysis mitigates disease in lupus-prone mice (reviewed in ref. 82).
Lupus nephritis
Author Manuscript
The kidney is involved in more than half of patients with SLE and contributes significantly
to morbidity. The contribution of autoantibodies with a number of reactivities and of
immune complexes in the expression of kidney inflammation has been reviewed extensively
over the years. Their role in instigating injury is considered to be mediated through the
activation of the complement system, which accounts for the inflammatory response83.
Podocytes express increased amounts of the serine/threonine kinase CaMK4, which, through
a distinct series of biochemical events, causes injury, and cell-targeted inhibition of CaMK4
in podocytes averts the deposition of immune complexes and nephritis84. These findings
signify the importance of resident cells in the initiation and propagation of kidney
inflammation.
infiltrating cells were thought to be exhausted87, clonally expanded CD4+ and CD8+ T cells
with memory effector cell markers are present in the kidneys of lupus nephritis. CD8+ T
cells are present in all biopsy samples and were found to adhere to the Bowman’s capsule
and to infiltrate the tubular epithelium88 and contribute to kidney damage. IL-17-producing
T cells have been found within kidney cell infiltrates of patients with lupus nephritis74, and
IL-17 is important for the development of lupus nephritis89. TFH cells are present in the
kidneys in close association with B cells in people with lupus nephritis, suggesting that they
may provide help to them90. Intrarenal B cells form germinal center–like structures produce
antibodies to vimentin, which is a dominant target in human lupus tubulointerstitial
nephritis91. Application of deep convolutional neural network methodology in specimens
from patients with lupus nephritis enabled cell-distance mapping, which confirmed that DCs
present antigen to CD4+ T cells92. Understanding the cellular architectures of in situ
Author Manuscript
immunity in lupus nephritis should expand our understanding of the involved pathogenic
processes.
the expanded circulating Gr1lo monocyte population94. Brief ischemia accelerates the
infiltration of Ly6Chi inflammatory macrophages into the kidneys of MRL-lpr mice95. DCs
Author Manuscript
also infiltrate the kidneys in people with lupus nephritis, probably propagating local adaptive
immune responses. Myeloid DC infiltration is associated with the accumulation of lymphoid
aggregates in the kidneys94. The identification of anti-inflammatory monocytes in the
kidneys of patients with lupus nephritis96 is especially important in considering prohealing
rather than immunosuppressive therapeutic approaches. Lastly, CD43hiCD11c
+F4/80loMHC-II− patrolling monocytes, which are known to orchestrate experimental
kidney inflammation97, are present in the kidneys of patients with lupus nephritis and lupus-
prone mice. Their function depends on TNFAIP3-interacting protein 1 (also referred to as
ABIN1) and its absence promotes lupus nephritis in a TLR-dependent manner98.
Single-cell RNA sequencing of kidney and skin biopsy material from patients with lupus
nephritis revealed type I IFN−response signatures in tubular cells and keratinocytes. Also,
Author Manuscript
high IFN response and fibrotic signatures in tubular cells were associated with failure to
respond to treatment99. Recent single-cell RNA sequencing of kidney samples from people
with lupus nephritis revealed 21 subsets of disease-active leukocytes, including multiple
populations of myeloid cells, T cells, natural killer cells and B cells, that demonstrated both
pro-inflammatory responses and inflammation-resolving responses. Also, evidence of
activated B cells and of progressive stages of monocyte differentiation were detected in the
kidney. A clear IFN type I response was observed in most cells. Two chemokine receptors,
CXCR4 and CX3CR1, were broadly expressed, implying they may have central roles in cell
trafficking100. Similar studies that investigate resident kidney cells in parallel may reveal
how the invasion of inflammatory cells alters the gene expression landscape and the function
of kidney cells.
Author Manuscript
There are a multitude of mechanisms that are involved in the expression of lupus nephritis,
including immune complexes, complement, infiltrating proinflammatory T cells and
antibody-producing B cells, and monocytes and the inherent processes of resident cells
account for the majority of the clinical heterogeneity of lupus nephritis and for the variable
response to drugs and biologics. Understanding the dominant operative mechanism in each
patient is the only way to develop personalized medicine.
The central nervous system is involved frequently in people and mice with SLE. The clinical
manifestations are quite diverse, apparently reflecting numerous immune and local
pathogenic processes102. So far, antibody-mediated neuronal injury, microglial cell
activation and infiltrating T cells are involved in the expression of brain injury. Antibodies
that recognize double-stranded DNA can cross-react with the NR2A and NR2B subunits of
the N-methyl-D-aspartate receptor and cause neuronal death, primarily via increased
neuronal calcium influx, which mimics glutamate excitotoxicity103. Transfer of these
antibodies to normal mice104 or immunization with the NMDAR-derived DWEYS
pentapeptide105 causes neuropsychiatric disease.
Within the brain, resident microglia are the predominant immune cells of the CNS and are
potent cytokine producers. It appears that neuronal injury is followed by microglial
Author Manuscript
Lymphocytes and other immune cells are probably important in the expression of CNS
disease. Tertiary lymphoid structures are present in the choroid plexus of lupus-prone mice
and people with SLE106, and lymphocytes apparently enter the brain parenchyma, but their
nature and function have not been studied. The recent reports on the presence of T cells in
the brains of people with autism107 and Alzheimer’s disease108 call for more attention on the
role of T cells in brain tissue injury.
Cutaneous lupus
Author Manuscript
TH17 cells, which are present in skin biopsy material, may contribute to the inflammatory
process112. pDCs have the unique capacity to rapidly produce huge amounts of IFN-α upon
recognition of viral RNA and DNA through TLR7 and TLR9 or through other pathogen
recognition receptors that are present in skin lesions113. The development of skin lesions
depends on FasL expressed on infiltrating TH1 cells recognizing cognate antigen and on
TLR7 in the absence of TLR9, revealing a complex regulation of skin inflammation in
lupus. pDCs and IFN-α have been shown to be abundant in skin lesions114.
The importance of TNF in the expression of skin lesions has been shown in experiments in
Author Manuscript
which lupus IgG was injected into the skin of various genetically modified mice. In brief,
monocytes but not lymphocytes or Ig were required for the induction of skin inflammation
(reviewed in ref. 115). Although TNF was required, only TNF receptor type I and not II
trimerization was needed for the induction of inflammation. While blockade of IL-17 may
have value in the treatment of patients with skin lupus, it is unclear whether TNF receptor
trimerization inhibitors may have any value, particularly given that TNF blockade may lead
to autoimmune manifestations.
production of endothelial nitric oxide synthase and to cause endothelial damage117. Low-
density granulocytes damage the vasculature because of their increased propensity for
NETosis and promote vascular leakage and endothelial-to-mesenchymal transition through
the degradation of vascular endothelial cadherin118. CCR5+T-bet+FoxP3+ CD4+ effector T
cells are present in atherosclerotic plaques119. Invariant natural killer T cells, however, were
recently claimed to interact with monocytes and promote an atheroprotective effect120.
sufficient to cause autoimmunity, reflecting a ‘house of cards’ effect, rather than what
actually happens in people with SLE. While the complete understanding of each pathway, be
it cellular or molecular, is of enormous significance, more important is the need to master
technology to identify which pathway is the driver in each individual person with SLE.
The list of failing clinical trials keeps expanding and has been regularly updated, but there
has been no effort to change the approach to clinical trials in a radical manner1. It is
unfortunate, although unavoidable, that we define the disease using diverse classification
Author Manuscript
criteria. There is no doubt that each biologic helps some people with SLE, mostly because
that pathway is central to the expression of the disease in the responding group of patients,
but this is not sufficient. There are two solutions: administer to each patient with SLE
multiple drugs/biologics in the hope that a greater number will experience clinical benefit, or
identify the driving pathway in each patient and treat her accordingly. A corollary to the first
approach is to administer biologics that correct multiple pathways.
The expanding use of whole-exome sequencing along with genome-wide association studies
has increased the number of patients with monogenic lupus, which will most probably
continue to increase. For these patients, besides treatment with drugs to control
manifestations, there is the possibility, with advancing technologies, of talking about a cure.
Although still early, it is important to identify patients in whom two or a few gene variants
contribute to the expression of the disease (oligogenic patients). New technologies should be
Author Manuscript
The expanding use of single-cell sequencing will help to identify previously unknown
functional immune cell subsets that may be upstream in immune system dysregulation and
could offer new targets for treatment. Single-cell sequencing of involved tissues may reveal
more features of the invading immune cells and, more importantly, that local resident cells
have both previously unrecognized functions100 and the ability to produce molecules that
cause organ destruction.
It is unfathomable that, although practically all patients are women (9 or more in 10), we
Author Manuscript
know very little of what lies behind this. The X chromosome packs numerous genes
involved in the immune response, and it appears that improper inactivation may perturb the
immune balance in favor of autoimmunity125. Are there master regulators encoded by genes
of the X chromosome that commandeer all that has been reported for SLE? Do these genes
need to be expressed in a homozygous fashion through unleashing the inactive allele? In the
same line of thinking, the study of the epigenome is still nascent in SLE.
Studies of the involved organs, including skin, the kidney and the brain, have revealed local
processes that are responsible for tissue damage. Without ignoring the role of the
autoimmune response in instigating organ damage, it is possible that the local inflammatory
processes proceed independently with little or no outside input. Better understanding of
these processes should open new approaches to disease treatment.
Author Manuscript
Acknowledgements
I thank my colleagues current and past who have helped me acquire a better understanding of this formidable
disease known as lupus. I want to thank M. Tsokos for her support and feedback during the preparation of this
report and N. Plummer for helping with the assembly of references. The work in my laboratory has been supported
by the NIH.
References
Author Manuscript
1. Durcan L, O’Dwyer T & Petri M Management strategies and future directions for systemic lupus
erythematosus in adults. Lancet 393, 2332–2343 (2019). [PubMed: 31180030]
2. Dörner T & Furie R Novel paradigms in systemic lupus erythematosus. Lancet 393, 2344–2358
(2019). [PubMed: 31180031]
3. Tsokos GC Systemic lupus erythematosus. N. Engl. J. Med. 365, 2110–2121 (2011). [PubMed:
22129255]
4. Theofilopoulos AN, Kono DH & Baccala R The multiple pathways to autoimmunity. Nat. Immunol.
18, 716–724 (2017). [PubMed: 28632714]
5. Deng Y & Tsao BP Updates in lupus genetics. Curr. Rheumatol. Rep. 19, 68 (2017). [PubMed:
28983873]
6. Langefeld CD et al. Transancestral mapping and genetic load in systemic lupus erythematosus. Nat.
Commun. 8, 16021 (2017). [PubMed: 28714469]
7. Tsokos GC, Lo MS, Costa Reis P & Sullivan KE New insights into the immunopathogenesis of
systemic lupus erythematosus. Nat. Rev. Rheumatol. 12, 716–730 (2016). [PubMed: 27872476]
Author Manuscript
health and disease. J. Biol. Chem. 288, 31880–31887 (2013). [PubMed: 24047902]
15. Hedrich CM, Rauen T & Tsokos GC cAMP-responsive element modulator (CREM)α protein
signaling mediates epigenetic remodeling of the human interleukin-2 gene: implications in
systemic lupus erythematosus. J. Biol. Chem. 286, 43429–43436 (2011). [PubMed: 21976679]
16. Hedrich CM et al. Stat3 promotes IL-10 expression in lupus T cells through trans-activation and
chromatin remodeling. Proc. Natl Acad. Sci. USA 111, 13457–13462 (2014). [PubMed:
25187566]
17. Jin F et al. Serum microRNA profiles serve as novel biomarkers for autoimmune diseases. Front.
Immunol. 9, 2381 (2018). [PubMed: 30459760]
18. Coit P & Sawalha AH The human microbiome in rheumatic autoimmune diseases: a
comprehensive review. Clin. Immunol. 170, 70–79 (2016). [PubMed: 27493014]
19. Greiling TM et al. Commensal orthologs of the human autoantigen Ro60 as triggers of
autoimmunity in lupus. Sci. Transl. Med. 10, eaan2306 (2018). [PubMed: 29593104]
20. Ivanov II et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 139,
Author Manuscript
25. Odhams CA et al. Interferon inducible X-linked gene CXorf21 may contribute to sexual
dimorphism in Systemic Lupus Erythematosus. Nat. Commun. 10, 2164 (2019). [PubMed:
Author Manuscript
31092820]
26. Moulton VR & Tsokos GC Why do women get lupus? Clin. Immunol. 144, 53–56 (2012).
[PubMed: 22659035]
27. Liang Y et al. A gene network regulated by the transcription factor VGLL3 as a promoter of sex-
biased autoimmune diseases. Nat. Immunol. 18, 152–160 (2017). [PubMed: 27992404]
28. Garcia-Romo GS et al. Netting neutrophils are major inducers of type I IFN production in pediatric
systemic lupus erythematosus. Sci. Transl. Med. 3, 73ra20 (2011).
29. Frangou E et al. REDD1/autophagy pathway promotes thromboinflammation and fibrosis in
human systemic lupus erythematosus (SLE) through NETs decorated with tissue factor (TF) and
interleukin-17A (IL-17A). Ann. Rheum. Dis. 78, 238–248 (2019). [PubMed: 30563869]
30. Puga I et al. B cell–helper neutrophils stimulate the diversification and production of
immunoglobulin in the marginal zone of the spleen. Nat. Immunol. 13, 170–180 (2012).
31. Blanco P, Palucka AK, Gill M, Pascual V & Banchereau J Induction of dendritic cell differentiation
by IFN-α in systemic lupus erythematosus. Science 294, 1540–1543 (2001). [PubMed: 11711679]
Author Manuscript
32. Jacquemin C et al. OX40 ligand contributes to human lupus pathogenesis by promoting T follicular
helper response. Immunity 42, 1159–1170 (2015). [PubMed: 26070486]
33. Jacquemin C et al. OX40L/OX40 axis impairs follicular and natural Treg function in human SLE.
JCI Insight 3, e122167 (2018).
34. Joo H et al. Serum from patients with SLE instructs monocytes to promote IgG and IgA
plasmablast differentiation. J. Exp. Med. 209, 1335–1348 (2012). [PubMed: 22689824]
35. Barrat FJ et al. Nucleic acids of mammalian origin can act as endogenous ligands for Toll-like
receptors and may promote systemic lupus erythematosus. J. Exp. Med. 202, 1131–1139 (2005).
[PubMed: 16230478]
36. Pisitkun P et al. Autoreactive B cell responses to RNA-related antigens due to TLR7 gene
duplication. Science 312, 1669–1672 (2006). [PubMed: 16709748]
37. Rowland SL et al. Early, transient depletion of plasmacytoid dendritic cells ameliorates
autoimmunity in a lupus model. J. Exp. Med. 211, 1977–1991 (2014). [PubMed: 25180065]
38. Furie R et al. Monoclonal antibody targeting BDCA2 ameliorates skin lesions in systemic lupus
Author Manuscript
45. Zhuang H, Szeto C, Han S, Yang L & Reeves WH Animal models of interferon signature positive
lupus. Front. Immunol. 6, 291 (2015). [PubMed: 26097482]
46. Schwarting A et al. Interferon-β: a therapeutic for autoimmune lupus in MRL-Faslpr mice. J. Am.
Soc. Nephrol. 16, 3264–3272 (2005). [PubMed: 16221871]
47. Shinde R et al. Apoptotic cell-induced AhR activity is required for immunological tolerance and
suppression of systemic lupus erythematosus in mice and humans. Nat. Immunol. 19, 571–582
(2018). [PubMed: 29760532]
48. Lood C et al. Platelet transcriptional profile and protein expression in patients with systemic lupus
erythematosus: up-regulation of the type I interferon system is strongly associated with vascular
Author Manuscript
61. Tipton CM et al. Diversity, cellular origin and autoreactivity of antibody-secreting cell population
expansions in acute systemic lupus erythematosus. Nat. Immunol. 16, 755–765 (2015). [PubMed:
26006014]
62. Henault J et al. Self-reactive IgE exacerbates interferon responses associated with autoimmunity.
Nat. Immunol. 17, 196–203 (2016). [PubMed: 26692173]
63. Mohan C, Adams S, Stanik V & Datta SK Nucleosome: a major immunogen for pathogenic
autoantibody-inducing T cells of lupus. J. Exp. Med. 177, 1367–1381 (1993). [PubMed: 8478612]
64. Christophersen A et al. Distinct phenotype of CD4+ T cells driving celiac disease identified in
multiple autoimmune conditions. Nat. Med. 25, 734–737 (2019). [PubMed: 30911136]
65. Kim SJ, Lee K & Diamond B Follicular helper T cells in systemic lupus erythematosus. Front.
Immunol. 9, 1793 (2018). [PubMed: 30123218]
66. Odegard JM et al. ICOS-dependent extrafollicular helper T cells elicit IgG production via IL-21 in
systemic autoimmunity. J. Exp. Med. 205, 2873–2886 (2008). [PubMed: 18981236]
67. Faliti CE et al. P2X7 receptor restrains pathogenic Tfh cell generation in systemic lupus
Author Manuscript
71. McKinney EF, Lee JC, Jayne DR, Lyons PA & Smith KG T-cell exhaustion, co-stimulation and
clinical outcome in autoimmunity and infection. Nature 523, 612–616 (2015). [PubMed:
Author Manuscript
26123020]
72. Sharabi A et al. Regulatory T cells in the treatment of disease. Nat. Rev. Drug Discov. 17, 823–844
(2018). [PubMed: 30310234]
73. Mizui M et al. IL-2 protects lupus-prone mice from multiple end-organ damage by limiting
CD4−CD8− IL-17–producing T cells. J. Immunol. 193, 2168–2177 (2014). [PubMed: 25063876]
74. Crispin JC et al. Expanded double negative T cells in patients with systemic lupus erythematosus
produce IL-17 and infiltrate the kidneys. J. Immunol. 181, 8761–8766 (2008). [PubMed:
19050297]
75. He J et al. Low-dose interleukin-2 treatment selectively modulates CD4+ T cell subsets in patients
with systemic lupus erythematosus. Nat. Med. 22, 991–993 (2016). [PubMed: 27500725]
76. Comte D et al. Brief report: CD4+ T cells from patients with systemic lupus erythematosus respond
poorly to exogenous interleukin-2. Arthritis Rheumatol. 69, 808–813 (2017). [PubMed: 27992687]
77. Moulton VR et al. Pathogenesis of human systemic lupus erythematosus: a cellular perspective.
Trends Mol. Med. 23, 615–635 (2017). [PubMed: 28623084]
Author Manuscript
78. Burzyn D et al. A special population of regulatory T cells potentiates muscle repair. Cell 155,
1282–1295 (2013). [PubMed: 24315098]
79. Afeltra A et al. The involvement of T regulatory lymphocytes in a cohort of lupus nephritis
patients: a pilot study. Intern. Emerg. Med. 10, 677–683 (2015). [PubMed: 25720575]
80. Crispin JC, Hedrich CM, Suárez-Fueyo A, Comte D & Tsokos GC SLE-associated defects
promote altered T cell function. Crit. Rev. Immunol. 37, 39–58 (2017). [PubMed: 29431078]
81. Moulton VR & Tsokos GC T cell signaling abnormalities contribute to aberrant immune cell
function and autoimmunity. J. Clin. Invest. 125, 2220–2227 (2015). [PubMed: 25961450]
82. Sharabi A & Tsokos GC T cell metabolism: new insights in systemic lupus erythematosus
pathogenesis and therapy. Nat. Rev. Rheumatol. 16, 100–112 (2020). [PubMed: 31949287]
83. Flores-Mendoza G, Sansón SP, Rodriguez-Castro S, Crispin JC & Rosetti F Mechanisms of tissue
injury in lupus nephritis. Trends Mol. Med. 24, 364–378 (2018). [PubMed: 29526595]
84. Maeda K et al. CaMK4 compromises podocyte function in autoimmune and nonautoimmune
kidney disease. J. Clin. Invest. 128, 3445–3459 (2018). [PubMed: 29985166]
Author Manuscript
85. Steinmetz OM et al. CXCR3 mediates renal Th1 and Th17 immune response in murine lupus
nephritis. J. Immunol. 183, 4693–4704 (2009). [PubMed: 19734217]
86. Peng SL et al. Murine lupus in the absence of alpha beta T cells. J. Immunol. 156, 4041–4049
(1996). [PubMed: 8621947]
87. Tilstra JS et al. Kidney-infiltrating T cells in murine lupus nephritis are metabolically and
functionally exhausted. J. Clin. Invest. 128, 4884–4897 (2018). [PubMed: 30130253]
88. Winchester R et al. Immunologic characteristics of intrarenal T cells: trafficking of expanded
CD8+ T cell β-chain clonotypes in progressive lupus nephritis. Arthritis Rheum. 64, 1589–1600
(2012). [PubMed: 22130908]
89. Amarilyo G, Lourenço EV, Shi FD & La Cava A IL-17 promotes murine lupus. J. Immunol. 193,
540–543 (2014). [PubMed: 24920843]
90. Liarski VM et al. Cell distance mapping identifies functional T follicular helper cells in inflamed
human renal tissue. Sci. Transl. Med. 6, 230ra246 (2014).
91. Kinloch AJ et al. Vimentin is a dominant target of in situ humoral immunity in human lupus
Author Manuscript
95. Iwata Y et al. Aberrant macrophages mediate defective kidney repair that triggers nephritis in
lupus-susceptible mice. J. Immunol. 188, 4568–4580 (2012). [PubMed: 22467656]
Author Manuscript
96. Berthier CC et al. Cross-species transcriptional network analysis defines shared inflammatory
responses in murine and human lupus nephritis. J. Immunol. 189, 988–1001 (2012). [PubMed:
22723521]
97. Finsterbusch M et al. Patrolling monocytes promote intravascular neutrophil activation and
glomerular injury in the acutely inflamed glomerulus. Proc. Natl Acad. Sci. USA 113, E5172–
E5181 (2016). [PubMed: 27528685]
98. Kuriakose J et al. Patrolling monocytes promote the pathogenesis of early lupus-like
glomerulonephritis. J. Clin. Invest. 129, 2251–2265 (2019). [PubMed: 31033479]
99. Der E et al. Tubular cell and keratinocyte single-cell transcriptomics applied to lupus nephritis
reveal type I IFN and fibrosis relevant pathways. Nat. Immunol. 20, 915–927 (2019); erratum 20,
1556 (2019). [PubMed: 31110316]
100. Arazi A et al. The immune cell landscape in kidneys of patients with lupus nephritis. Nat.
Immunol. 20, 902–914 (2019). [PubMed: 31209404]
101. Ge Y et al. Cgnz1 allele confers kidney resistance to damage preventing progression of immune
Author Manuscript
108. Heneka MT An immune-cell signature marks the brain in Alzheimer’s disease. Nature 577, 322–
323 (2020). [PubMed: 31937952]
109. Menke J et al. Sunlight triggers cutaneous lupus through a CSF-1-dependent mechanism in MRL-
Faslpr mice. J. Immunol. 181, 7367–7379 (2008). [PubMed: 18981160]
110. Martens HA et al. Analysis of C1q polymorphisms suggests association with systemic lupus
erythematosus, serum C1q and CH50 levels and disease severity. Ann. Rheum. Dis. 68, 715–720
(2009). [PubMed: 18504288]
111. Ramirez-Ortiz ZG et al. The scavenger receptor SCARF1 mediates the clearance of apoptotic
cells and prevents autoimmunity. Nat. Immunol. 14, 917–926 (2013). [PubMed: 23892722]
112. Oh SH et al. Expression of interleukin-17 is correlated with interferon-α expression in cutaneous
lesions of lupus erythematosus. Clin. Exp. Dermatol. 36, 512–520 (2011). [PubMed: 21631571]
113. Guiducci C et al. Autoimmune skin inflammation is dependent on plasmacytoid dendritic cell
activation by nucleic acids via TLR7 and TLR9. J. Exp. Med. 207, 2931–2942 (2010). [PubMed:
21115693]
Author Manuscript
114. Mande P et al. Fas ligand promotes an inducible TLR-dependent model of cutaneous lupus-like
inflammation. J. Clin. Invest. 128, 2966–2978 (2018). [PubMed: 29889098]
115. Deng GM & Tsokos GC Pathogenesis and targeted treatment of skin injury in SLE. Nat. Rev.
Rheumatol. 11, 663–669 (2015). [PubMed: 26241186]
116. Liu Y & Kaplan MJ Cardiovascular disease in systemic lupus erythematosus: an update. Curr.
Opin. Rheumatol. 30, 441–448 (2018). [PubMed: 29870498]
117. Buie JJ, Renaud LL, Muise-Helmericks R & Oates JC IFN-α negatively regulates the expression
of endothelial nitric oxide synthase and nitric oxide production: implications for systemic lupus
erythematosus. J. Immunol. 199, 1979–1988 (2017). [PubMed: 28779021]
118. Carlucci PM et al. Neutrophil subsets and their gene signature associate with vascular
inflammation and coronary atherosclerosis in lupus. JCI Insight 3, 99276 (2018). [PubMed:
Author Manuscript
29669944]
119. Li J et al. CCR5+T-bet+FoxP3+ effector CD4 T cells drive atherosclerosis. Circ. Res. 118, 1540–
1552 (2016). [PubMed: 27021296]
120. Smith E et al. Cross-talk between iNKT cells and monocytes triggers an atheroprotective immune
response in SLE patients with asymptomatic plaque. Sci. Immunol. 1, eaah4081 (2016).
[PubMed: 28783690]
121. Lim H et al. Proatherogenic conditions promote autoimmune T helper 17 cell responses in vivo.
Immunity 40, 153–165 (2014). [PubMed: 24412615]
122. Choi JY et al. Circulating follicular helper-like T cells in systemic lupus erythematosus:
association with disease activity. Arthritis Rheumatol. 67, 988–999 (2015). [PubMed: 25581113]
123. Ryu H et al. Atherogenic dyslipidemia promotes autoimmune follicular helper T cell responses
via IL-27. Nat. Immunol. 19, 583–593 (2018); erratum 19, 1036 (2018). [PubMed: 29713015]
124. Hedrich CM & Tsokos G SNPs talk to genes using landlines: long-range chromatin interactions
link genetic risk with epigenetic patterns in Takayasu arteritis. Ann. Rheum. Dis. 78, 1293–1295
Author Manuscript
[PubMed: 22523428]
137. Levine B & Kroemer G Biological functions of autophagy genes: a disease perspective. Cell 176,
11–42 (2019). [PubMed: 30633901]
138. Wang S, Wen F, Wiley GB, Kinter MT & Gaffney PM An enhancer element harboring variants
associated with systemic lupus erythematosus engages the TNFAIP3 promoter to influence A20
expression. PLoS Genet. 9, e1003750 (2013). [PubMed: 24039598]
139. Vereecke L, Beyaert R & van Loo G Genetic relationships between A20/TNFAIP3, chronic
inflammation and autoimmune disease. Biochem. Soc. Trans. 39, 1086–1091 (2011). [PubMed:
21787353]
140. Salmond RJ, Brownlie RJ, Morrison VL & Zamoyska R The tyrosine phosphatase PTPN22
discriminates weak self peptides from strong agonist TCR signals. Nat. Immunol. 15, 875–883
Author Manuscript
146. Zhao X et al. MicroRNA-125a contributes to elevated inflammatory chemokine RANTES levels
via targeting KLF13 in systemic lupus erythematosus. Arthritis Rheum. 62, 3425–3435 (2010).
[PubMed: 20589685]
147. Thai TH et al. Deletion of microRNA-155 reduces autoantibody responses and alleviates lupus-
like disease in the Faslpr mouse. Proc. Natl Acad. Sci. USA 110, 20194–20199 (2013). [PubMed:
24282294]
148. Gonzalez-Martin A et al. The microRNA miR-148a functions as a critical regulator of B cell
tolerance and autoimmunity. Nat. Immunol. 17, 433–440 (2016). [PubMed: 26901150]
149. Zhu S et al. The microRNA miR-23b suppresses IL-17-associated autoimmune inflammation by
targeting TAB2, TAB3 and IKK-α. Nat. Med. 18, 1077–1086 (2012). [PubMed: 22660635]
150. Baumjohann D et al. The microRNA cluster miR-17~92 promotes TFH cell differentiation and
represses subset-inappropriate gene expression. Nat. Immunol. 14, 840–848 (2013). [PubMed:
23812098]
151. Kim SJ, Gregersen PK & Diamond B Regulation of dendritic cell activation by microRNA let-7c
Author Manuscript
rates of apoptosis and defects in mechanisms responsible for their clearance. The T cell
response to antigen is aberrant in and late signaling events (Fig. 2) and results in
misbalanced levels of cytokines, including decreased IL-2 and increased IL-17 production. T
cells, through distinct pathways, also acquire a greater ability to invade tissues and
contribute to the inflammatory response. B cells, in response to cognate and non-cognate
(cytokines) interactions with T cells, produce antibodies. Antibodies enter tissues directly or
in the form of immune complexes (IC), which contribute to tissue inflammation. Cells of the
innate immune response, under the influence of the involved pathogenic factors, produce
cytokines (including IFN-α) or, through the direct interaction with lymphocytes, contribute
significantly to the inflammatory organ-damaging response. The clinical heterogeneity of
the disease is highly correlated with the multitude of the pathways that lead to organ injury.
Although several pathways operate in each individual, the relative contribution of each
Author Manuscript
pathway varies from person to person. Finally, local factors dictate which organ will be
afflicted by the autoinflammatory response.
Fig. 2 |. T cell early and late signaling aberrations in T cells from patients with SLE.
Engagement of the CD3 complex either by autoantigen or circulating CD3/TCR antibodies
results in an increased intracytoplasmic calcium response. The CD3 complex is rewired,
with the CD3ζ chain replaced with the FcRγ chain, which recruits the kinase Syk. The
calcium-requiring kinase CaMKIV enhances the binding of CREMα/ICER to the Il2 and
Il17 or Il10 promoters to suppress and enhance their expression, respectively. Calcineurin
also dephosphorylates the transcription factor NFAT, which binds to the promoter of CD40L
and increases its expression. Phosphatase PP2A is increased in T cells with diverse effects:
in Treg cells, it dephosphorylates mTORC1 and promotes Treg cell function and, in effector
Author Manuscript
T cells, it enhances the binding of IRF4 to the Il17 promoter and it dephosphorylates p-
CREB. PP2A also promotes ROCK activity, which phosphorylates ERM, which in turn
enhances the ability of CD44 to bind its ligand hyaluronic acid (HA, expressed in the kidney
and other tissues). IL-2 signaling is defective with decreased amounts of p-STAT5, whereas
IL-6 signaling is increased with increased binding of p-STAT3 to the Il17 promoter. An
increased proportion of CD8+ T cells express the ectonucleotidase CD38, which suppresses
the level of NAD and the activity of the deacetylase SIRT1, which in turn enhances the
activity of the histone methyltransferase EZH2. As indicated by the green circles, a number
of the molecules have been considered as therapeutic targets: Syk, ROCK, calcineurin,
EZH2, IL-17, IL-23, JAK and mTOR as well as IL-2, which can be replenished with low
doses.
Author Manuscript
Table 1 |
STAT4 Binds the transcription factor HMGA1; shared with rheumatoid arthritis 128
PRDM1 Linked to increased production of IL-6 and cathepsin S (needed for loading peptides into MHC-II) 16,17
IKZF1 Involved in the expression of C1QB and five type I IFN–response genes 130
Fc receptor genes Clearance of apoptotic cells and immune complexes; antibody-mediated cell cytotoxicity 131
NCF1 Decreased number of copies predispose to, whereas increased protects against, SLE 135
ITGAM Impaired phagocytosis of complement-opsonized targets in monocytes, neutrophils and macrophages 136
TNFAIP3 Encodes the deubiquitinating enzyme A20, which is involved in the termination of NF-κB signaling; A20-deficient mice develop severe 138,139
PTPN22 Expansion of effector and memory T cells and increased production of IFN-γ, TNF and GM-CSF 140
Table 2 |
MicroRNAs control aspects of the immune response and organ damage in SLE
Author Manuscript
miR-155 Controls the expression of SH2-domain-containing inositol 5′-phosphatase 1 (SHIP1), which is important in B cell 147
activation; miR155−/−Faslpr mice have decreased autoimmunity and nephritis
miR-148a Impairs B cell tolerance by suppressing the expression of the autoimmune suppressor Gadd45a, the tumor suppressor 148
PTEN and the proapoptotic protein Bim, which promotes the survival of immature B cells after engagement of the B
cell antigen receptor
miR-23b Suppresses IL-17-associated autoimmune inflammation by targeting TAB2, TAB3 and IKKα mRNA 149
miR-17[sim]92 Promotes TFH cell differentiation; promotes DC activation through the microRNA let-7c and BLIMP-1 150
151
miR-150 Promotes renal fibrosis in lupus nephritis by downregulating expression of SOCS1 152
Author Manuscript
Author Manuscript
Author Manuscript