Immunology of Mycobacterium

Download as pdf or txt
Download as pdf or txt
You are on page 1of 37

Immunology of Mycobacterium

tuberculosis Infections
JONATHAN KEVIN SIA1 and JYOTHI RENGARAJAN2
1
Emory Vaccine Center, Emory University School of Medicine, Emory University, Atlanta, GA 30329;
2
Department of Medicine, Division of Infectious Diseases and Emory Vaccine Center,
Emory University School of Medicine, Emory University, Atlanta, GA 30329

ABSTRACT Tuberculosis (TB) is a serious global public health bacterial replication, is a well-known antimycobacte-
challenge that results in significant morbidity and mortality rial contribution by adaptive immune cells such as CD4
worldwide. TB is caused by infection with the bacilli
and CD8 T cells. Despite pressures from host immunity,
Mycobacterium tuberculosis (M. tuberculosis), which has evolved
a wide variety of strategies in order to thrive within its host.
M. tuberculosis is able to persist in the host. M. tuber-
Understanding the complex interactions between M. tuberculosis culosis infection results in hallmark lesions called gran-
and host immunity can inform the rational design of better ulomas, which are initially aggregates of infected and
TB vaccines and therapeutics. This chapter covers innate and uninfected myeloid cells circumscribed by a lymphocytic
adaptive immunity against M. tuberculosis infection, including cuff. The granuloma is thought to prevent bacterial dis-
insights on bacterial immune evasion and subversion garnered semination to extrapulmonary sites but can also become
from animal models of infection and human studies. In addition, a niche for long-term bacterial persistence. M. tubercu-
this chapter discusses the immunology of the TB granuloma,
losis has evolved myriad strategies to evade and subvert
TB diagnostics, and TB comorbidities. Finally, this chapter
provides a broad overview of the current TB vaccine pipeline. immune responses to persist within a host, and it is be-
coming increasingly clear that the immune response
to M. tuberculosis infection involves contributions from
INTRODUCTION a wide variety of innate and adaptive immune cells. A
Mycobacterium tuberculosis, the etiologic agent of tuber- clearer understanding of the complex cross talk between
culosis (TB), remains a significant global public health M. tuberculosis and host immunity is essential for the
burden (1). In 2016, there were 10.4 million new TB development of efficacious TB vaccines. Despite being
cases reported globally and nearly 1.7 million TB-related
deaths (1). Understanding the host response to M. tu- Received: 30 April 2018, Accepted: 10 May 2018,
Published: 12 July 2019
berculosis infection is a key aspect of efforts to eradi- Editors: Vincent A. Fischetti, The Rockefeller University, New York,
cate TB through the development of effective vaccines NY; Richard P. Novick, Skirball Institute for Molecular Medicine, NYU
and immune therapeutics. M. tuberculosis is an intra- Medical Center, New York, NY; Joseph J. Ferretti, Department of
Microbiology & Immunology, University of Oklahoma Health
cellular pathogen transmitted via inhalation of aerosol- Science Center, Oklahoma City, OK; Daniel A. Portnoy, Department
ized, bacteria-containing droplets. Innate immune cells of Molecular and Cellular Microbiology, University of California,
in the lungs, primarily macrophages, dendritic cells, Berkeley, Berkeley, CA; Miriam Braunstein, Department of
Microbiology and Immunology, University of North Carolina-Chapel
monocytes, and neutrophils, readily phagocytose M. Hill, Chapel Hill, NC, and Julian I. Rood, Infection and Immunity
tuberculosis and are the earliest defenders against the Program, Monash Biomedicine Discovery Institute, Monash
University, Melbourne, Australia
pathogen. The transformation of bacteria-containing
Citation: Sia JK, Rengarajan J.2019.Immunology of Mycobacterium
phagosomes into acidified, antimicrobial compartments tuberculosis infections. Microbiol Spectrum 7(4):GPP3-0022-2018.
is a central tenet of defense against M. tuberculosis. doi:10.1128/microbiolspec.GPP3-0022-2018.
In this regard, the production of interferon-γ (IFN-γ), Correspondence: Jyothi Rengarajan, [email protected]
© 2019 American Society for Microbiology. All rights reserved.
which can activate infected myeloid cells and inhibit

ASMscience.org/MicrobiolSpectrum 1
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Sia and Rengarajan

developed nearly a century ago, Mycobacterium bovis able to control infection in the form of LTBI, an esti-
bacillus Calmette-Guérin (BCG), an attenuated strain of mated 5 to 10% of people exposed to M. tuberculosis
M. bovis, remains the only licensed vaccine against TB. develop ATB, which is characterized by a persistent
Vaccination with BCG provides protection against se- cough accompanied by sputum production, weight loss,
vere forms of disseminated TB in children but has vari- weakness, and night sweats (9). Clinical diagnosis and
able efficacy in preventing pulmonary disease in children treatment of M. tuberculosis infection is complicated by
and adults (2–4). However, the immunological basis for a variety of coinfections and comorbidities.
the poor efficacy of BCG remains unclear. Moreover, Comorbidities that modulate immune function can
long-held concepts regarding the nature of desired im- exacerbate TB disease or contribute to progression of
mune responses in an ideal TB vaccine, namely, the in- individuals with LTBI to ATB. HIV coinfection in la-
duction of antigen-specific CD4 T cells producing IFN-γ, tently infected individuals increases the risk of develop-
are being updated to reflect the expanding knowledge ing TB from a 5 to 10% lifetime risk to a 10% annual
of host immunity to M. tuberculosis infection gathered risk, and HIV infection is the single greatest risk factor
from animal models and human cohort studies. Ad- for the development of TB (10–14). The relevance of
vances in imaging and single-cell technologies combined HIV coinfection to global TB mortality is highlighted by
with high-throughput approaches and systems-based the fact that more than a fifth of all TB-related deaths in
analyses are providing more information on the immune 2016 were in HIV-positive individuals (1). Progressive
response to M. tuberculosis infection at increasingly depletion and dysfunction of CD4 T cells following HIV
higher resolutions. As our understanding of the host re- infection leads to immune suppression and negatively
sponse to M. tuberculosis infection grows, opportunities impacts immunity to M. tuberculosis. Specific depletion
to leverage knowledge of the immunology of M. tuber- of M. tuberculosis-specific CD4 T cells has been reported
culosis infection toward improving therapeutics and in the peripheral blood (15, 16) and broncheoalveolar
vaccines for TB are increasing. lavage (BAL) samples (17, 18) of HIV-infected individ-
This article will cover integral features of the innate uals with LTBI. Several studies indicate that specific
and adaptive immune response to M. tuberculosis in- depletion may be a consequence of enhanced HIV co-
fection. Additionally, it will highlight recent findings on receptor expression in CD4 T cells, particularly CCR5,
the hallmark granuloma and novel cellular players con- in TB patients (15, 19–24). Alternative hypotheses to ex-
tributing to the host response to M. tuberculosis infec- plain specific depletion of M. tuberculosis-specific CD4
tion. Finally, it will provide an overview of the state of T cells include differential functionality of specific T
TB vaccine research, including a summary of BCG-based cells. In HIV coinfected LTBI, M. tuberculosis-specific
vaccines and the TB vaccine pipeline. CD4 T cells are reported to secrete interleukin 2 (IL-2) in
contrast to MIP-1β (macrophage inflammatory protein 1
beta) secreted by cytomegalovirus-specific CD4 T cells
IMMUNOPATHOGENESIS OF TB IN (16). Analysis of viral loads in HIV coinfected LTBI
HUMANS AND ANIMAL MODELS showed an inverse correlation between viral load and
Overview of Human TB the frequency of M. tuberculosis-specific CD4 T cells
Disease and Comorbidities secreting IL-2 (25), suggesting that IL-2 producing M.
Transmission of M. tuberculosis occurs after inhalation tuberculosis-specific CD4 T cells may be specifically
of aerosolized droplets containing live bacteria into the depleted in the context of HIV coinfection. Relatedly,
lungs. Successful transmission is influenced by a variety HIV-coinfected individuals have lower frequencies of
of conditions, including proximity and duration of cytokine-producing M. tuberculosis-specific CD4 T cells
contact with an individual with active TB (ATB) disease with impaired proliferative capacity compared to HIV-
and the immune-competency of the individual infected uninfected individuals with LTBI (26–28), suggesting
with M. tuberculosis (5–7). We now appreciate that in a M. tuberculosis-specific CD4 T-cell dysfunction during
clinical setting, M. tuberculosis infection presents as a HIV-infection. The relative contributions of depletion
continuum of diseased/infected states ranging from versus dysfunction of M. tuberculosis-specific CD4 T
asymptomatic latent TB infection (LTBI) to ATB disease. cells to enhanced TB risk following HIV infection remains
This complexity, combined with remarkable heteroge- unclear. Further, HIV infection may perturb protective
neity in lesions within a single patient, has presented immunity to M. tuberculosis in other immune compart-
unique challenges to the eradication of TB (8). While the ments, such as CD8 T cells. For instance, M. tuberculosis-
majority of individuals exposed to M. tuberculosis are specific CD8 T cells from individuals with LTBI are

2 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Immunology of M. tuberculosis Infections

reported to have impaired proliferation and degranulation Other animal models of M. tuberculosis infection in-
in HIV-infected compared to HIV-uninfected individuals clude guinea pigs, rabbits, fish, and non-human primates
(29). Studies have also described associations between TB (NHP). Each has distinct advantages and disadvantages
and many other conditions or activities, including smok- that make their use particularly suitable for different
ing, malnutrition, diabetes, helminth infections, chronic types of research questions. Following infection, guinea
lung diseases, and cancer (30, 31). Further investigations pigs exhibit pathological features, such as the organi-
will be required to fully understand the basis of identified zation and development of caseous necrotic granulomas,
associations with other infections and morbidities. that more accurately recapitulate the human granulo-
matous response compared to mice (41). Further, guinea
Animal Models of Infection pigs are very susceptible to M. tuberculosis infection and,
Knowledge of the host response to M. tuberculosis in- thus, are a good choice for testing candidate drugs and
fection has benefited greatly from the development vaccines and studying dissemination dynamics. Simi-
of animal models of infection. The variable outcomes larly, rabbits develop a well-organized granuloma that
of M. tuberculosis infection in humans are challenging can become necrotic following mycobacterial infection.
to model in a single animal model. Many experimen- However, rabbits are resistant to M. tuberculosis, and
tal animals are susceptible to M. tuberculosis infection high numbers of bacteria during inoculation or use of
and can inform us about aspects of human disease. The more virulent strains are needed (42–45). Nevertheless,
mouse model for TB benefits from many advantages: the rabbit model has been leveraged to study relatively
ease of manipulation and housing, availability of well- rarer forms of TB, such as cutaneous and meningeal
characterized inbred strains, sophisticated techniques TB (46, 47). The usefulness of both the guinea pig and
for the generation of mutant strains, availability of im- rabbit models is hampered by the scarcity of immuno-
munological and other reagents, and relatively low cost. logic reagents relative to mice. The zebrafish model has
Mice have been utilized to model host responses to provided novel insights into the establishment of the
M. tuberculosis infection, to evaluate drug and vaccine mycobacterial granuloma. Infection of transparent zeb-
candidates, and to study the immune response to mutant rafish larvae with the natural fish pathogen Mycobac-
strains of mycobacteria. Experimental infection can be terium marinum leads to the establishment of well-
delivered through multiple routes: intravenously, intra- organized granulomas that become necrotic and can
peritoneally, intratracheally, or via aerosolized particles. be visually monitored (48). The primary advantage of
The latter method, especially low-dose aerosol infection, the zebrafish model is the transparency of the zebra-
is the most physiologically relevant and has become the fish larvae, which, alongside facile manipulation of
preferred method. Different mouse strains have well- host and bacterial genetics, has been leveraged for in-
characterized lung pathologies and levels of suscepti- sight into early innate immune events leading to the
bility (32–36). Typically, following bacterial deposition formation of the granuloma as well as insights into
into the lungs, it takes approximately 2 weeks to begin human disease. Adaptive immunity is present in adult
priming adaptive immune responses in the lung-draining zebrafish, and different populations of CD4 T cells
lymph nodes and a further 1 to 2 weeks for robust par- have recently been described (49, 50), but these ani-
ticipation in the lungs by adaptive immune cells, but mals are no longer transparent, and relevance of the
bacterial burdens continue to be maintained at a high adult zebrafish immune response to human TB have yet
level in the lungs of M. tuberculosis-infected mice. There to be established.
are limitations to what can be gleaned from mouse The NHP model of M. tuberculosis infection reflects
models of M. tuberculosis infection due to the differences much of the heterogeneity observed in human TB. In-
in lung pathology between mice and humans. Further, fection of NHPs is typically performed by aerosol or
true latent infection and significant immune control of direct bronchoscopic deposition into the lungs of rhesus
infection are difficult to establish in the mouse model, or cynomolgus macaques and, depending on the dose of
though chemotherapeutically induced models of pauci- the inoculation and the strain of bacteria utilized, leads
bacillary disease in mice exist (37, 38). The development to symptomatic ATB disease or asymptomatic infection
of humanized mice that can recapitulate the heteroge- in which bacteria persist at low levels akin to LTBI. The
neity of human lung pathology may extend the advan- NHP model accurately recapitulates many of the hall-
tages of the mouse model, but humanized mice are also mark granulomas seen in humans, including the het-
reported to display aberrant T-cell responses and be erogeneity of granulomas that can be present in the same
unable to control bacterial burden (39, 40). animal (51), and presents clinical symptoms similar to

ASMscience.org/MicrobiolSpectrum 3
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Sia and Rengarajan

those seen in humans (52–57). The NHP model is (e.g., TLR-2, TLR-4, TLR-9) (72–74). Mannosylated
regarded as an important preclinical model for TB re- lipoarabinomannan, phosphatidyl-inositol mannosides,
search and is an excellent model for studying immunity phthiocerol dimycocerosates, phenolic glycolipids (PGLs),
to M. tuberculosis and assessing candidate drug and trehalose dimycolate, peptidoglycan, and other mycobac-
vaccine efficacies (58–63). Further, the NHP model can terial components are recognized by an array of cell sur-
be used to study reactivation in the setting of simian face and intracellular receptors that mediate phagocytosis
immunodeficiency virus coinfection or other types of and/or antimicrobial defenses. M. tuberculosis DNA (75,
immune modulation, such as anti-tumor necrosis-α 76) or bacterial second messengers (77) can be recognized
(anti-TNF-α) treatment, CD4 depletion, or inhibition of by cytosolic pattern recognition receptors (PRRs), such
indoleamine 2,3-dioxygenase (IDO) (64–71). as cGAS and STING (78, 79), to induce downstream
cytokine production and autophagy. Further, nucleotide
oligomerization domain-like receptors (NLRs) are cyto-
INNATE IMMUNITY TO solic PRRs that recognize M. tuberculosis pathogen-
M. TUBERCULOSIS INFECTION associated molecular patterns, such as muramyl dipep-
The earliest encounter between host and pathogen in TB tide, to activate a multiprotein complex termed the
occurs at the interface between innate immune cells and inflammasome. Functional redundancies for many of the
M. tuberculosis. While innate immunity is critical for receptors are likely to exist due to promiscuous ligand
early antimycobacterial responses, it is also important binding by different receptors and the wide array of
for the progression of infection and long-term control of available ligands on M. tuberculosis. Indeed, single or
M. tuberculosis by continually priming and educating double knockouts for canonical scavenger receptors and
adaptive immune responses and by regulating inflam- C-type lectin receptors did not modulate susceptibility or
mation. However, innate immune cells are often niches attenuate immune responses following M. tuberculosis
for bacterial replication, and M. tuberculosis utilizes infection (80). However, increased susceptibility to M.
a variety of strategies that subvert innate immune re- tuberculosis infection in a variety of knockout mice
sponses to establish a chronic infection. Here, we will demonstrate that a number of PRRs and their associated
detail key features of the innate immune response to signaling pathways also play important, nonredundant
M. tuberculosis infection, starting from recognition of roles in host defense against M. tuberculosis infection.
the bacterium and phagosomal defenses within infected M. tuberculosis expresses a variety of known or pu-
macrophages to priming of adaptive immune responses tative TLR ligands, and TLR-2, TLR-4, and TLR-9 have
by professional antigen-presenting cells. In between, we been implicated in host recognition of M. tuberculosis
will highlight how neutrophils and monocytes are mo- (reviewed in 73, 74). Polymorphisms in specific TLRs
bilized after M. tuberculosis infection, the role that or TLR signaling proteins have also been strongly as-
natural killer (NK) cells play during infection, how the sociated with pulmonary TB in humans and have been
balance of inflammation is regulated by the innate im- shown to influence immunity against M. tuberculosis
mune system, and how cell death affects the immune (81–84). The contribution of individual TLRs to im-
response. In each section, we will also highlight some munity against M. tuberculosis infection is variable, but
of the myriad strategies that M. tuberculosis utilizes to the importance of the TLR signaling pathway to anti-
subvert or evade the host innate immune response. mycobacterial immunity is evident in studies showing
that mice lacking the common TLR adaptor protein,
Recognition of M. tuberculosis by myeloid differentiation factor 88 (MyD88), quickly
Pattern Recognition Receptors succumb to M. tuberculosis infection (85, 86). Suscep-
Pathogen-associated molecular patterns on M. tubercu- tibility of MyD88–/– mice to M. tuberculosis infection
losis are recognized via a variety of receptors to medi- has been attributed to deficient expression of NOS2
ate opsonic and nonopsonic bacterial uptake: C-type (86), impaired ability to activate the IL-1β or IL-1 re-
lectins (e.g., mannose receptors, DC-SIGN, Dectin-1, ceptor (IL1R) pathway (87, 88), impaired receptivity
Dectin-2, Mincle), complement receptors (e.g., comple- of macrophages to IFN-γ signaling (89), and impaired
ment receptor 3), collectins (e.g., surfactant proteins A IL-12 and TNF-α responses in macrophages and den-
and D, mannose-binding lectin), scavenger receptors dritic cells (DCs) (85). Gene-deletion studies in single
(e.g., MARCO, SR-A1, CD36, SR-B1), Fc receptors (e.g., TLRs have revealed that innate immune responses to
FcgR), glycophosphatidylinositol-anchored membrane M. tuberculosis are likely the result of the complex ac-
receptors (e.g., CD14), and Toll-like receptors (TLRs) tivation of multiple signaling pathways. For instance,

4 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Immunology of M. tuberculosis Infections

mice lacking both TLR-2 and TLR-9 are more suscep- controversial. Modulation of innate immune responses
tible to M. tuberculosis infection than mice lacking the by M. tuberculosis is also accomplished through the
ability to signal through either TLR by itself (90). The presence of immune-inhibitory lipid components that
susceptibility of MyD88–/– mice to M. tuberculosis in- compete with immune-activating mycobacterial compo-
fection is an example of the importance of common nents for the same receptors. For example, expression
adaptor molecules that integrate signals from multiple of tetraacylated sulfoglycolipids by the W-Beijing strain
PRRs and other innate immune pathways for the in- GC1237 can competitively bind TLR-2 to attenuate
duction of antimycobacterial immunity. Further evi- responses to canonical TLR-2 agonists, including my-
dence for this concept is demonstrated by the increased cobacterial lipomannans (104). Lastly, M. tuberculosis
susceptibility of M. tuberculosis-infected mice lacking can also impair innate immune responses to cell-
CARD9, an adapter molecule integrating signals from envelope components through enzymatic means. For
C-type lectin receptors, or PYCARD/ASC, an adapter instance, an M. tuberculosis serine-hydrolase, Hip1, was
molecule integrating signals from nucleotide oligomeri- found to cleave multimeric, cell wall-associated GroEL2
zation domain-like receptors for the induction of the to a secreted monomeric form to mediate attenuated
inflammasome (91, 92). macrophage and DC responses (105–109). Addition-
MyD88 signaling in innate immunity integrates sig- ally, M. tuberculosis mutants lacking hip1 or a putative
naling from TLR and IL-1 receptor families by bridging mycobacterial metalloprotease, zmp1, display enhanced
ligand-receptor binding to IL-1-receptor-associated kinases inflammasome activation (106, 110), suggesting that
and the activation of multiple downstream pathways, in- M. tuberculosis has multiple strategies for dampening
cluding NF-κB, mitogen-activated protein kinases, and activation of the inflammasome.
activator protein 1. The IL-1 signaling pathway is clearly Thus, in addition to the array of host receptors that
required for resistance to M. tuberculosis infection in mediate recognition of M. tuberculosis, innate immune
mouse models and is supported by human immunogenetics responses to infection likely depend on the strain of
studies (93–96). In mice, the absence of IL-1 signaling led M. tuberculosis, the presence of cell wall components
to severe susceptibility to M. tuberculosis infection. Both that can competitively inhibit the activation of PRRs,
IL-1α and IL-1β, as well as their common receptor, IL-1R1, and the presence of M. tuberculosis enzymes that modify
have been implicated in immunity to M. tuberculosis (87, the immunogenicity of cell envelope components.
88, 97–101). Secretion of the mature form of IL-1β requires
cleavage by the terminal inflammasome effector, caspase-1, Phagosomal Defense in Macrophages
but M. tuberculosis-infected mice lacking MyD88, ASC, or Macrophages are the first immune cells to encounter
caspase-1 signaling do not display impaired IL-1β levels M. tuberculosis during infection and also represent the
(87). Further, mice deficient in IL-1β are considerably more primary replicative niche for M. tuberculosis. Recognition
susceptible to M. tuberculosis infection than mice lacking of M. tuberculosis by macrophages leads to phagocytosis
ASC or caspase-1 (87). These findings suggest that IL-1β is and sequestration of the bacterium in phagosomes, which
a key mediator of resistance to M. tuberculosis infection typically eradicate pathogens via fusion with lysosomes
but also indicate that the basis for resistance conferred by and consequent acidification of the pathogen-containing
MyD88, CARD9, and PYCARD/ASC likely depend on phagolysosome. However, M. tuberculosis is able to sur-
additional factors beyond IL-1β. vive and replicate in the phagosome by inhibiting phago-
While host recognition of M. tuberculosis leads to the somal maturation and phagolysosomal generation through
activation of innate immunity, M. tuberculosis has also a variety of mechanisms (reviewed in 72, 111). Further,
evolved strategies that evade innate immune responses transcriptional profiling of intraphagosomal bacteria indi-
mediated by PRRs. Strain-specific expression of cell en- cated that M. tuberculosis readily counters the nitrosative,
velope components may be associated with differential oxidative, hypoxic, and nutrient-poor phagosomal envi-
immune responses. For example, the W-Beijing lineage ronment through the expression of stress-adaptive genes
strain, HN878, has been found to express polyketide (112), though a genome-wide transposon site hybridiza-
synthase-derived phenolic glycolipids that are missing tion screen for M. tuberculosis survival in macrophages
in lab-adapted H37Rv or other clinical isolates (i.e., suggested that M. tuberculosis constitutively expresses
CDC1551) (102). Expression of PGL by HN878 has genes required for its survival (113). Nevertheless, it is clear
been found to diminish production of multiple innate that M. tuberculosis has adapted for a lifestyle inside the
immune cytokines and chemokines (102, 103), though macrophage and employs many strategies to survive within
its role in the increased virulence of HN878 remains these cells.

ASMscience.org/MicrobiolSpectrum 5
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Sia and Rengarajan

M. tuberculosis glycolipids can prevent accumulation complement receptor 3 depended on host cholesterol,
of phosphatidylinositol 3-phosphate on phagosomal which mediated phagosomal association with coronin-1
membranes and prevent phagolysosome biosynthesis and consequent inhibition of phagolysosome forma-
(114). M. tuberculosis also secretes phosphatases (SapM tion through activation of host calcineurin (138, 139).
and PtpA) and serine/threonine kinases (PknG) that Alternatively, TLR-2 recognition of mycobacterial
are proposed to interfere with phagosomal maturation mannosylated lipoarabinomannan activates NF-κB and
(115–121). There is also evidence that M. tuberculosis NOS2 gene transcription that leads to antimycobacterial
lipids, in particular, phthiocerol dimycocerosates, can nitric oxide (NO) production (140). NO production is
mediate escape from the phagosome and host cell death strongly associated with resistance to M. tuberculosis,
(122). An M. tuberculosis secretion system, ESX-1, is though evidence for the antimycobacterial effects of
also known for mediating disruptions in phagosomal NO is stronger in the mouse model. In mice, reactive
integrity and preventing phagosome maturation. Pro- nitrogen intermediates are toxic to mycobacteria in vitro
motion of aberrant phagosomal integrity and bacterial (141–143), and infection can be exacerbated by the
replication by M. tuberculosis ESX-1 is countered by inhibition of NOS in vitro (144, 145) or in vivo (146–
IFN-γ-induced, Rab20-mediated phagosomal matura- 148). NO production following IFN-γ signaling has also
tion (123). ESX-1-mediated phagosomal escape of bac- been reported to limit overt inflammation by inhibiting
teria is hypothesized to work through disruption of the processing of IL-1β by the inflammasome (149). Re-
phagosome by the 6-kDa early secretory antigenic target latedly, mice with disrupted NOS2 alleles display exa-
(ESAT-6) (124–127), though recent evidence proposes cerbated disease following M. tuberculosis infection
a contact-dependent, ESAT-6-independent mechanism (146, 150). Although in vitro studies using human al-
for ESX-1-mediated phagosomal permeabilization (128). veolar macrophages and primary monocytes did not find
Nevertheless, ESX-1-mediated permeabilization of the an antimycobacterial role for NO (151–153), specific
phagosome exposes M. tuberculosis pathogen-associated staining for NOS2 in the BAL of TB patients reveals
molecular patterns, such as N-glycolyl-muramyl dipep- upregulation in infected individuals compared to healthy
tide, to cytoplasmic nucleotide oligomerization domain controls (154). Nevertheless, M. tuberculosis has several
2 receptors to induce type I IFNs (129, 130). ESX-1- strategies to cope with otherwise damaging reactive ni-
mediated permeabilization of the phagosome also exposes trogen and oxygen intermediates: M. tuberculosis KatG,
extracellular bacterial DNA to the cytosolic DNA-sensing a catalase-peroxidase, can inactivate phagosomal reac-
pathway, which leads to targeting of M. tuberculosis to tive oxygen (155), and the M. tuberculosis proteasome
autophagosomes for subsequent killing (75). M. tuber- can mediate resistance to nitrosative stresses (156). Pro-
culosis ESX-3 has also been implicated in modulating miscuous recognition of mycobacterial antigens by the
intracellular trafficking of bacteria to avoid phagosomal same receptor may also have convergent outcomes as in
maturation through inhibition of the host endosomal the case for TLR-2-mediated recognition of M. tuber-
sorting complex required for transport (131–133). Thus, culosis cell wall fractions leading to TNF-α production
studies of the M. tuberculosis ESX secretion system have in murine macrophages (157). TLR-mediated recogni-
provided evidence for its role in both bacterial evasion tion of M. tuberculosis is also reported to synergize
of phagosomal pressures and host sensing of bacterial with the vitamin D pathway to induce the antimicrobial
components. In addition to the ESX system, M. tubercu- peptide (AMP), cathelicidin, in human macrophages
losis also expresses two SecA ATPase protein homologues (158, 159). The biologically active vitamin D metabolite,
(SecA1 and SecA2) involved in protein export (134). calcitriol, induces hCAP-18, a gene encoding the pro-
SecA2, in particular, has been implicated in virulence and form of cathelicidin, following TLR ligation of macro-
intracellular growth (135, 136). Interestingly, both M. tu- phages (158–160). In addition to direct antimicrobial
berculosis and BCG ΔsecA2 mutants are enriched in acidi- activity, cathelicidin has been shown to exert antimi-
fied phagosomes, indicating that mycobacterial SecA2 is crobial functions by activating transcription of host
required for arrest of phagosome maturation (137). autophagy genes Beclin-1 and Atg5 (161). The vitamin
M. tuberculosis entry into macrophages through dif- D pathway also synergizes with IFN-γ secreted by T cells
ferent receptors can lead to distinct activation of path- to induce IL-15 autocrine signaling to promote auto-
ways that can inhibit or promote bacterial replication. phagy and phagosomal maturation in M. tuberculosis-
The overall effect of multiple receptors engaging distinct infected human macrophages (162).
or overlapping M. tuberculosis ligands is a complex and Autophagy is the process whereby cytoplasmic con-
dynamic issue. For example, M. tuberculosis uptake by stituents are degraded or recycled. A role for autophagy

6 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Immunology of M. tuberculosis Infections

in antimycobacterial immunity in macrophages has of Atg5, but not other autophagy genes, compromised
been extensively characterized. Initial studies utilizing control of M. tuberculosis (172, 173). Deletion of the
M. bovis suggested that autophagy plays a role in pro- autophagy-related genes Ulk1, Ulk2, Atg4B, or p62
moting phagosomal maturation to enhance bacterial compromised the ability to induce autophagy, but they
killing (163). Moreover, LRG-47, an IFN-γ inducible were dispensable for the control of M. tuberculosis
p47 GTPase reported to be critical for phagosomal (173). Analysis of lung sections from M. tuberculosis-
maturation and control of M. tuberculosis (164), is also infected Atg5 knockout mice indicated that Atg5 may be
involved in the induction of autophagy in M. bovis- involved in regulation of neutrophil responses during
infected macrophages (165). Autophagy-related genes infection, suggesting autophagy-independent roles for
were revealed to be involved in regulating the intracel- Atg5. Further, a recently described role for Atg5 in LC3-
lular bacterial load of lab-adapted and clinical isolates associated phagocytosis during M. tuberculosis infection
of M. tuberculosis in a genome-wide small interfering supports the notion that specific components of auto-
RNA screen in infected human macrophage-like THP-1 phagy can also overlap with other phagosomal path-
cells (166). Accumulating evidence indicates that auto- ways in immunity against mycobacteria (174).
phagy is integrated into the host response to M. tuber- Taken together, it is clear that macrophage recogni-
culosis infection by synergizing with pathogen sensing, tion and phagocytosis of M. tuberculosis lead to a dy-
phagosomal maturation, and IFN-γ inducible path- namic tug of war between antimycobacterial defenses
ways to mediate antimycobacterial immunity: STING- and M. tuberculosis immune evasion. Macrophage
dependent cytosolic sensing of M. tuberculosis DNA defenses include AMPs, nitrosative stresses, phagolyso-
is required to deliver bacteria to autophagosomes and somal fusion, and autophagy and may operate inde-
restrict bacterial replication (75); knockdown of cGAS pendently of or subsequent to IFN-γ signaling. On the
in infected macrophages attenuated the induction of other hand, M. tuberculosis can subvert macrophage
autophagy and survival during chronic M. tuberculosis defenses at the level of the bacterial cell wall components
infection (78); detection of cyclic-di-AMP secreted by that limit phagosomal maturation and the bacterial
M. tuberculosis in macrophages induced type I IFN pro- genes that combat or allow adaptation to intracellular
duction and autophagy to limit bacterial virulence (77); immune pressure.
PARKIN, a conserved ubiquitin ligase, was shown to
ubiquitinate M. tuberculosis-containing phagosomes to Recruitment and Function of Neutrophils and
facilitate ubiquitin-mediated autophagy and restrict bac- Monocytes Following M. tuberculosis Infection
terial replication (167); IFN-γ-induced host ubiquilin-1 Secretion of cytokines and chemokines early during
colocalizes with M. tuberculosis and mediates trafficking infection recruits additional phagocytes to the site of
of bacteria to autophagosomes (168); IFN-γ receptor infection. Early secretion of chemoattractants may be
signaling mediated by the MyD88 adaptor-like (Mal) attributed to infected alveolar macrophages as well as
molecule induced autophagy and killing of intracellular lung epithelial cells (175–177). Moreover, a recent study
M. tuberculosis in macrophages (169). Several studies suggests that cross talk between primary bronchial epi-
have also delineated strategies employed by M. tuber- thelial cells and infected macrophages may also promote
culosis to evade autophagy. M. tuberculosis is reported secretion of chemokines (178). Trafficking of additional
to induce the expression of microRNA-33 to inhibit monocytes and granulocytes to the lung exerts immune
autophagy and regulate intracellular lipid metabolism pressure on M. tuberculosis and is crucial for the initi-
to benefit bacterial replication (170). Further, a screen of ation of adaptive immune responses, but it may also
M. tuberculosis cosmid clones in search of genes that promote M. tuberculosis cell-to-cell transmission and
inhibited bone marrow-derived DC antigen presentation dissemination.
revealed M. tuberculosis PE_PGRS47 (Rv2741) as an Recruitment of neutrophils serves as an early line of
inhibitor of autophagy-mediated antigen presentation defense against M. tuberculosis infection via secretion
(171), suggesting that M. tuberculosis-mediated im- of antimicrobial molecules and inflammatory media-
pairment of innate immunity can also negatively impact tors, but neutrophils also serve as niches for bacterial
the generation of adaptive immunity. It is also becoming replication and can impede immunity against M. tuber-
clear that autophagy-related proteins are likely to per- culosis. In humans with active pulmonary TB, neutro-
form multiple functions, and care must be taken when phils have been found to be a significant population
interpreting specific knockouts or knockdowns of indi- of M. tuberculosis-infected phagocytes in the BAL and
vidual genes. For instance, myeloid cell-specific ablation sputum (179). Whole blood transcriptional profiling

ASMscience.org/MicrobiolSpectrum 7
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Sia and Rengarajan

also identified a neutrophil signature in ATB patients therapy (191). Further, as discussed later in the chapter,
that is associated with type I and type II IFN-inducible dysregulation of neutrophil recruitment by unrestrained
genes (180) and expression of the inhibitory molecule IL-17 responses during M. tuberculosis infection can in-
PD-L1 (181), suggesting that neutrophils may play an cur pathological consequences by driving lung-damaging
immunomodulatory role in human TB. In mice, the ki- inflammation. Thus, the overall effect of neutrophil re-
netics and magnitude of neutrophil recruitment follow- cruitment to the site of M. tuberculosis infection may be
ing M. tuberculosis infection depends on the strain of determined by host genetics, the context of infection
mouse infected. Evidence for a pathogenic role for neu- (pulmonary versus extrapulmonary), or timing and du-
trophils is shown in studies comparing neutrophil re- ration of neutrophil activity.
cruitment in resistant versus susceptible mouse strains In addition to neutrophils, monocytes are recruited to
(182, 183). When comparing resistant C57BL/6 mice to the site of M. tuberculosis infection. Similar to neutro-
susceptible DBA/2 mice after M. tuberculosis infection, phils, monocyte recruitment is important for innate im-
a study found that neutrophils were rapidly recruited munity during M. tuberculosis infection but may also
into the broncheoalveolar space at higher magnitudes inadvertently promote M. tuberculosis dissemination.
in susceptible mice. Depletion of neutrophils at the onset C-C chemokine receptor type 2 (CCR2) is a chemokine
of M. tuberculosis infection specifically extended the life receptor expressed on monocytes and is responsible for
spans of DBA/2 mice, suggesting that early neutrophil CCL2-mediated recruitment of monocytes to sites of
involvement was pathogenic in genetically susceptible bacterial infection (192). CCR2 was found to mediate
mice (183). Similarly, neutrophil depletion in susceptible immunity against M. tuberculosis depending on the dose
I/St mice shortly after M. tuberculosis infection reduced of infection. CCR2 knockout mice were more suscepti-
lung pathology and bacterial growth and improved ble to high-dose intravenous M. tuberculosis infection
survival compared to C57BL/6 mice (184). In a separate (193), but not after low-dose infection (194). Monocytes
study, depletion of neutrophils 5 weeks after aerosol have been shown to differentiate into macrophages and
M. tuberculosis infection of resistant BALB/c mice en- DCs following M. tuberculosis infection, and monocytes
hanced the levels of lung IL-6 and IL-17 without im- transferred into M. tuberculosis-infected mice were shown
pacting IFN-γ and modestly enhanced control of bacterial to be the predominant population of innate immune cells
burden (185). Neutrophil depletion in the first 4 days producing iNOS (195). Additionally, monocyte delivery
following intravenous M. tuberculosis infection of BALB/ of M. tuberculosis to pulmonary lymph nodes can coor-
c mice, however, led to enhanced bacterial growth at dinate with DCs to prime CD4 T cells after infection (196).
extrapulmonary sites, suggesting that antimycobacterial Monocytes may therefore represent a recruited popula-
immunity conferred by neutrophils may be dependent on tion of innate cells that combat M. tuberculosis infection
the route of infection and the kinetics of neutrophil in- through the production of reactive nitrogen intermediates
volvement (186). Utilizing fluorescently labeled bacteria, and priming of adaptive immunity. However, monocyte
a recent study demonstrated that bacterial distribution in recruitment following M. tuberculosis infection may also
myeloid cells shifts from CD11b+Ly6G– monocytes and be detrimental to the host by providing an environment
macrophages to CD11b+Ly6G+ neutrophils in Nos2–/– full of permissive cells. Treatment of M. tuberculosis-
animals infected with M. tuberculosis, suggesting that infected mice with polyinosinic-polycytidylic acid (polyIC)
neutrophil influx can create a growth-permissive envi- led to CCR2-dependent recruitment of a population of
ronment for M. tuberculosis under NO-deficient condi- M. tuberculosis-permissive monocytes, severe susceptibil-
tions (187). Evidence for beneficial roles that neutrophils ity, and early mortality (197). Interestingly, susceptibility
play in antimycobacterial defense focus on neutrophil of polyIC-treated mice to M. tuberculosis infection was
secretion of AMPs such as cathelicidin and lipocalin-2 to dependent on type I IFN signaling and was not due to any
restrict bacterial replication (188) or via uptake of AMP- particular alteration to the T-cell response. The recruit-
containing apoptotic neutrophils by M. tuberculosis- ment of neutrophils and monocytes to the site of M. tu-
infected macrophages (189). Neutrophils can also release berculosis infection represents a host strategy to contain
chromatin scaffolds that trap extracellular bacteria in an bacterial replication that is co-opted by the bacterium to
AMP-containing mesh. M. tuberculosis has been shown facilitate its growth and dissemination.
to induce the formation of neutrophil extracellular traps
in vitro (190), and levels of neutrophil extracellular traps NK Cells in M. tuberculosis Infection
detected in the plasma of ATB patients were associ- NK cells are innate lymphocytes with the capacity to
ated with disease severity and decreased with antibiotic secrete IFN-γ and perform cytolytic functions to mediate

8 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Immunology of M. tuberculosis Infections

control of a variety of pathogens, including M. tuber- In addition to induction of type I IFNs, neutrophils have
culosis. Various components of the M. tuberculosis cell also been reported to drive lung destruction through the
wall can bind directly to NKp44 found on NK cells (198), secretion of matrix metalloproteinase 8 (216). The ma-
and NK cells can also recognize stress molecules upregu- trix metalloproteinase family of enzymes has been im-
lated on the surface of M. tuberculosis-infected cells (199). plicated in lung tissue destruction during M. tuberculosis
NK cells can mediate direct killing of M. tuberculosis- infection (217–220) but has also been shown to promote
infected macrophages (199) but can also restrict intra- macrophage recruitment and bacterial dissemination
cellular bacterial replication via secretion of IL-22 (200) during infection of zebrafish (221).
and IFN-γ (201) to increase phagolysosomal fusion of Eicosanoids are lipid mediators of inflammation de-
M. tuberculosis-containing phagosomes. Additionally, rived from the oxidation of arachidonic acid. The bal-
NK cells can enhance immunity against M. tuberculosis ance between proinflammatory prostaglandin E2 (PGE2)
indirectly by enhancing CD8 T-cell production of IFN-γ and anti-inflammatory lipoxin A4 (LXA4), two members
(202) by promoting the expansion of γδ T cells (203) and of the eicosanoid family of signaling molecules, can de-
by lysing M. tuberculosis-expanded regulatory T cells termine the outcome of M. tuberculosis infection (222–
(204). The cytolytic capacity of NK cells is diminished 224). During M. tuberculosis infection, mice incapable
in ATB patients relative to healthy controls and can be of synthesizing PGE2 display increased susceptibility
reconstituted following antibiotic therapy (205). Further, (223), and absence of the enzyme 5-lipoxygenase, which
NK cell function in TB patients can be attenuated by metabolizes arachidonic acid to LXA4, confers resistance
monocyte-derived IL-10 (201). Interestingly, a popula- (222). Importantly, therapeutic correction of low PGE2
tion of IL-21-dependent NK cells that appears following levels can confer enhanced survival in highly susceptible
BCG vaccination has been shown to expand following mice infected with M. tuberculosis (100). Leukotriene
M. tuberculosis challenge (206), suggesting that NK cells A4 hydrolase is an enzyme that catalyzes the production
may also display some hallmark characteristics of mem- of proinflammatory leukotriene B4 from leukotriene A4,
ory cells. which can also be converted to anti-inflammatory LXA4
as a counterbalance. In zebrafish, LTA4H mutants were
Inflammation and Cell Death found to be hypersusceptible to M. marinum infection
During M. tuberculosis Infection due to dysregulation of the balance between leukotrienes
The regulation of inflammation is a critical factor that and lipoxins; increased levels of LXA4 in LTA4H mu-
determines the outcome of M. tuberculosis infection. tants impaired TNF-α responses and promoted sus-
Overexuberant inflammation impairs cellular immunity, ceptibility (225). The relevance of this finding to humans
damages lung tissue, and can lead to lung cavitation and is highlighted in a TB meningitis cohort in Vietnam
enhanced transmission. Inversely, too little inflamma- where heterozygosity for six LTA4H polymorphisms
tion can impair control of bacterial burden by delaying conferred a survival advantage over homozygosity (225).
the induction of innate and adaptive immunity. While Indeed, anti-inflammatory glucocorticoid treatment effi-
neutrophil recruitment and activity during M. tubercu- cacy in TB meningitis patients can be differentiated by
losis infection can help contain bacterial replication, a single nucleotide polymorphism in the LTA4H pro-
sustained neutrophilic inflammation can mediate dam- moter controlling transcriptional activity, which suggests
aging inflammation and promote disease. Importantly, that the balance of inflammation is critical to disease
whole blood transcriptomics identified a neutrophil- progression and treatment outcomes in TB meningitis
driven type I IFN-inducible signature in human TB that (226).
decreased upon treatment (180). Excessive type I IFN TNF-α is a critical proinflammatory cytokine in im-
signaling has been shown to promote disease in mouse munity against M. tuberculosis infection and can be se-
models and human samples. Mice lacking type I IFN creted by a number of innate and adaptive immune cells.
signaling are more resistant to M. tuberculosis infection The importance of TNF-α in antimycobacterial immu-
(207–210), though signaling through type I IFNs may nity is clearly demonstrated by heightened susceptibil-
play a protective role in the absence of IFN-γ (211, 212). ity of TNF-α antibody-depleted animals or in animals
Mechanisms underlying the pathogenic role of type I lacking TNF receptor signaling following M. tubercu-
IFNs during M. tuberculosis infection include inhibition losis infection (227). TNF-α is also a critical mediator of
of IL-1β production (213, 214), induction of IL-10 to immunity against TB in humans. This is demonstrated
impair innate cytokine production (215), and loss of by increased rates of progression to ATB in LTBI pa-
IFN-γ responsiveness in infected macrophages (215). tients receiving anti-TNF treatment for inflammatory

ASMscience.org/MicrobiolSpectrum 9
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Sia and Rengarajan

disorders (228), which can be recapitulated in the NHP lowing low-dose aerosol infection with virulent M. tuber-
model of infection (70). The effects of anti-TNF treat- culosis (223). Host cell necrosis following M. tuberculosis
ment in humans and NHPs, as well as in mice (229– infection can be induced through activation of the cyto-
231), suggests that TNF-α is critical for maintaining solic receptor interacting protein kinase 3 pathway, which
sequestration of M. tuberculosis in the granuloma. inhibits apoptosis of infected macrophages through Bcl-
Histopathological evidence from gene-disrupted or xL and promotes necrosis through upregulation of ROS
antibody-depleted mice infected with M. tuberculosis (242). Additionally, macrophages infected with virulent
also suggests that TNF-α signaling may be playing a H37Rv, but not avirulent H37Ra, undergo proteolysis
role in modulating apoptotic or necrotic cell death at the N-terminal of annexin-1, which prevents the com-
following infection (229, 231). pletion of the apoptotic envelope and drives macrophage
Cell death can be a means of restricting bacterial re- necrosis (235). Taken together, apoptosis represents a
plication by the host or a way to disseminate to second- strategy by the host to limit infection through the com-
ary loci of infection for M. tuberculosis. Apoptosis of bination of bacterial sequestration in apoptotic vesicles
M. tuberculosis-infected cells leads to fewer viable bac- and the induction of adaptive immune responses, but
teria and effective cross-presentation of bacterial antigens M. tuberculosis may delay apoptosis or promote necrosis
(224, 232, 233), whereas necrosis of M. tuberculosis- to facilitate replication and dissemination.
infected cells allows viable bacteria to exit and disseminate
(223, 234, 235). Proapoptotic M. tuberculosis mutants Initiation of Adaptive Immunity
lacking secA2 (236) and nuoG (237) were attenuated to M. tuberculosis by DCs
in vivo, and mice infected with these strains displayed An important function of innate immunity during M.
enhanced priming of adaptive immunity compared to in- tuberculosis infection is the priming of adaptive immune
fection with wild-type M. tuberculosis, suggesting that responses. DCs are professional antigen-presenting cells
prevention of host cell apoptosis is an M. tuberculosis vir- that initiate adaptive immunity by presenting M. tuber-
ulence strategy. Relatedly, M. tuberculosis-infected mu- culosis antigens in the context of major histocompati-
rine neutrophils can aid in DC trafficking to the draining bility complex (MHC), costimulatory molecules, and
lymph nodes to initiate antigen-specific CD4 T-cell re- cytokines. Depletion of cells expressing the pan-DC
sponses (238), but M. tuberculosis delays CD4 T-cell marker, CD11c, following M. tuberculosis infection
priming by inhibiting neutrophil apoptosis (239). Infec- impaired control of bacterial burden and delayed the
tion with the proapoptotic nuoG mutant M. tuberculosis initiation of adaptive immunity, illustrating the impor-
resulted in earlier DC trafficking to lung-draining lymph tance of DCs in mobilizing adaptive immune responses
nodes and earlier priming of antigen-specific CD4 T cells, that can control bacterial replication (243). There is
but enhanced priming was abrogated upon neutrophil abundant evidence that M. tuberculosis is able to infect
depletion (239). Additionally, uninfected macrophages murine (244–246) and human DCs (247–249). In mice
performing a constitutive housekeeping function called infected with green fluorescent protein GFP-expressing
efferocytosis can uptake M. tuberculosis-containing apo- M. tuberculosis, DCs were found to be the major pop-
ptotic bodies, which leads to delivery and killing of bac- ulation of phagocytes infected by bacteria after 4 weeks
teria in lysosomes (240). This suggests that apoptosis may (246). Upon M. tuberculosis infection, DCs mature and
be a host strategy to limit bacterial replication by se- migrate to the lung-draining lymph nodes to initiate
questering bacteria in vesicles that can be safely degraded antigen-specific T-cell responses, which depended on
by nearby innate immune cells. Inhibition of apoptosis the chemokine receptor CCR7 and its corresponding
by M. tuberculosis is driven by host intrinsic factors fol- chemokines CCL19 and CCL21 (250–252). Further, IL-
lowing infection with virulent strains. The proinflamma- 12, a cytokine secreted by myeloid cells and important for
tory eicosanoid PGE2 has been demonstrated to regulate the induction of IFN-γ responses, is required for DC mi-
synaptotagmin-7, a calcium sensor that maintains plasma gration during M. tuberculosis infection (253). Priming of
membrane integrity (241). Human macrophages infected adaptive immune responses requires the transport of live
with virulent M. tuberculosis H37Rv, but not avirulent bacteria to the lung-draining lymph nodes (246, 250), but
H37Ra, promote LXA4 production and inhibition of antigen-specific T cells can be primed by both the infected
PGE2 biosynthesis, which impairs resealing of plasma migratory DC and uninfected lymph node resident DC. A
membrane disruptions to preferentially induce host cell study demonstrated that infected DCs migrate to the
necrosis instead of apoptosis (241). Mice lacking PGE2 lung-draining lymph nodes, where they secrete soluble,
also suffered from increased lung bacterial burden fol- unprocessed M. tuberculosis antigens that are summarily

10 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Immunology of M. tuberculosis Infections

phagocytosed by uninfected lymph node resident DCs M. tuberculosis subversion of DC functions can interfere
(254). The exportation of M. tuberculosis antigens was with antigen presentation and delay or impair the initi-
initially proposed to benefit the host by circumventing ation of the adaptive immune response. Improving DC
inefficient antigen presentation by infected DCs. How- functions during M. tuberculosis infection may improve
ever, secretion of M. tuberculosis antigens by infected innate and adaptive immunity and enhance immune
DCs may also benefit the pathogen by diverting antigen control of bacterial burden. A study that exoge-
away from MHC class II antigen presentation (255). nously engaged the CD40 costimulation pathway in
Effective interaction between DCs and T cells is de- M. tuberculosis-infected DCs improved DC functions
pendent on appropriate function of antigen presentation and promoted antigen-specific CD4 T-cell responses that
machinery, including expression of MHC, costimula- augmented control of lung bacterial burden (268). Fur-
tory molecules, and cytokines following M. tuberculosis ther, mucosal transfer of Ag85B-loaded DCs following
infection. However, there is abundant evidence that challenge with M. tuberculosis augmented the efficacy of
M. tuberculosis infection impairs antigen presentation BCG vaccination (269), suggesting that early antigen
to evade antigen-specific T-cell responses. It is well rec- presentation by DCs is an important component that
ognized that M. tuberculosis infection leads to impaired determines the efficacy of vaccine-induced immunity.
MHC class II antigen presentation by macrophages DCs are critical players that initiate adaptive immune
(reviewed in 256). M. tuberculosis-mediated inhibition responses to M. tuberculosis and determine the outcome
of phagosomal maturation has been implicated in at- of infection. Interventions or therapies that improve DC
tenuating processing of M. tuberculosis antigen 85 functions may provide benefits by augmenting cross talk
(Ag85) and the MHC class II-associated invariant chain between DCs and antigen-specific T cells.
(257). Multiple studies have also reported that M. tu-
berculosis infection impairs MHC class II expression
in macrophages through inhibition of class II transacti- ADAPTIVE IMMUNITY AGAINST
vator, a master transcriptional regulator controlling M. TUBERCULOSIS
expression of MHC class II molecules (258–261), al- Protective immunity to M. tuberculosis and control
though there is little evidence of similar inhibition of bacterial replication requires adaptive immune re-
of MHC class II in DCs. Nevertheless, M. tuberculosis sponses. This is best exemplified by the extreme sus-
infection of DCs leads to functional impairment of an- ceptibility to mycobacterial infections of lymphopenic
tigen presentation. M. tuberculosis infection has been HIV patients and gene-deleted mice lacking MHC
shown to impair DC maturation of human (reviewed class II or T cells in general. Cytokine secretion and di-
in 262) and murine DC functions (reviewed in 263, rect antimicrobial actions of antigen-specific T cells are
264). Studies examining proliferation of T-cell recep- key features of the adaptive immune response against
tor transgenic CD4 T cells specific for M. tuberculosis M. tuberculosis infection. Further, the long-lived nature
Ag85 as a proxy for functional antigen presentation of antigen-specific memory T cells provides the basis for
have demonstrated that M. tuberculosis EsxH can im- developing vaccines that induce antimycobacterial im-
pair antigen processing through inhibition of the host munity. There are also expanding roles for B cells, γδ
endosomal sorting complex required for transport T cells, and CD1-restricted T cells that provide specific
(ESCRT) (265). Additionally, M. tuberculosis promotes responses to a diverse set of M. tuberculosis antigens
suboptimal antigen presentation in vitro and in vivo that complement antigens classically presented through
without detectable differences in the expression levels MHC class I and II. However, adaptive immune re-
of costimulatory molecules when compared to BCG- sponses can also become malignant by promoting ex-
infected DCs (266). Interestingly, studies using a mutant cessive inflammation or be rendered ineffective from
M. tuberculosis strain lacking hip1 (discussed above) chronic antigen exposure. Here, we cover the impor-
indicate that M. tuberculosis readily impairs DC costi- tance of timing, location, and quality of CD4 T-cell
mulation and cytokine production to evade antigen- responses during M. tuberculosis infection, how CD8
specific CD4 T-cell responses (107, 109), and a recent T cells contribute to immunity against M. tuberculosis,
study demonstrated that BCG hip1 retains similar im- the roles that inhibitory receptors play during infection,
mune evasion functions (267). Taken together, the ini- the phenotypes and functions of memory T cells, and the
tiation of the adaptive immune response requires the roles that B cells, γδ T cells, CD1-restricted lymphocytes,
participation of DCs, which themselves are readily and mucosal associated invariant T (MAIT) cells play in
infected and subverted by M. tuberculosis infection. immunity against M. tuberculosis.

ASMscience.org/MicrobiolSpectrum 11
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Sia and Rengarajan

Kinetics and Homing of CD4 T Cells to vasculature-restricted CD4 T cells that expressed
after M. tuberculosis Infection CX3CR1 (280). Interestingly, cells retained in the lung
In the mouse model of infection, CD4 T-cell responses vasculature secreted the highest amount of IFN-γ during
are absolutely required to control bacterial replication, infection (280). Adoptive transfer studies demonstrated
and animals lacking such responses succumb rapidly that IFN-γ accounted for greater control of bacterial
(270, 271). MHC class II knockout mice or CD4 de- burden in the spleen over the lung and drove immuno-
pletion led to abrupt mortality following M. tuberculosis pathology when overexpressed (281), suggesting that
infection (270, 271). CD8 T cells play a key role in im- the function of IFN-γ may be to mediate control of bac-
munity against M. tuberculosis but cannot compensate terial dissemination to extrapulmonary sites and that
for CD4 deficiency (270). Similarly, antibody depletion IFN-γ may be detrimental when unrestrained. The dis-
of CD4 in cynomolgus macaques severely compromised tinction between vasculature-restricted and parenchyma-
control of M. tuberculosis and led to reactivation in la- localizing CD4 T cells seems less important in rhesus
tently infected animals (69). Thus, the initiation of the macaques (282), where the majority of antigen-specific
CD4 T-cell response is a key feature defining the out- CD4 T cells can be found in the lung parenchyma but are
come of M. tuberculosis infection. There is a widely re- restricted to the outer lymphocytic cuff of granulomas.
cognized delay in the initiation of antigen-specific CD4 Notably, studies have demonstrated that expression
T-cell responses following low-dose aerosol infection of of IDO by cells in the granulomas of M. tuberculosis-
mice (250, 272–275) and NHPs (276). M. tuberculosis- infected rhesus macaques can mediate inhibition of T cell
infected cynomolgus macaques had detectable antigen- entrance into granuloma, and biochemical inhibition
specific responses 4 weeks postinfection (276). In mouse of IDO led to reorganization of the granuloma to include
models of infection, antigen-specific CD4 T-cell re- T cells localizing into the macrophage core (66, 283).
sponses are first detected in the lung-draining lymph Taken together, there is strong evidence that localization
nodes 2 weeks after infection. Significant antigen-specific of antigen-specific CD4 T cells into the lung tissues where
lung CD4 T-cell responses are subsequently detected in M. tuberculosis-infected myeloid cells reside is an impor-
the lungs 3 weeks after infection. This is in stark contrast tant feature of protective immunity to M. tuberculosis.
to antigen-specific responses to other bacterial (277) or
viral (278) pathogens, which are detected swiftly after Quality and Specificity of the CD4
infection. Adoptive transfer of ESAT-6-specific CD4 T-Cell Response to M. tuberculosis
T cells prior to aerosol M. tuberculosis infection have The quality of the T-cell response is an important feature
demonstrated an apparent kinetic bottleneck whereby determining the outcome of M. tuberculosis infection.
lung antigen-specific activation occurs only 7 days after Canonically, the production of IFN-γ by Th1 cells, CD8
infection despite the presence of antigen-specific T cells T cells, and other lymphocytes is considered essential
(279), suggesting that antigen-specific responses are for protection against mycobacterial infections. In hu-
delayed by mechanisms other than trafficking of CD4 man immunogenetics studies, Mendelian susceptibility
T cells from the mediastinal lymph nodes to the lungs. to mycobacterial disease (MSMD) is a spectrum of
Delay in the initiation of adaptive immune responses to genetic mutations in five autosomal genes (IFNGR1,
M. tuberculosis infection may be due to a variety of fac- IFNGR2, STAT1, IL12B, IL12RB) and an X-linked
tors, including slow growth of the bacterium, inhibited gene that confer susceptibility to avirulent environmen-
apoptosis of infected macrophages and neutrophils, and tal mycobacteria and BCG (284). Deficiencies related
delayed activation and migration of DCs, which cumu- to IFN-γ signaling in young patients with mutations
latively allow M. tuberculosis to establish a persistent in IFNGR1 and IFNGR2 confer fatal susceptibility to
infection in the lung. mycobacterial infections (285–288). STAT1 is an in-
CD4 T cells interact with infected macrophages to tracellular molecule important for IFN-γ signaling, and
restrict intracellular M. tuberculosis replication. Thus, individuals with heterozygous germline STAT1 muta-
the effectiveness of the CD4 T-cell response depends on tions lose gamma-interferon activating factor (GAF) ex-
proper homing of antigen-specific CD4 T cells from pression (289). GAF is an important transcription factor
lymphoid tissues to M. tuberculosis-infected cells in the that facilitates IFN-γ-induced gene expression. Individ-
lung. In M. tuberculosis-infected mice, antigen-specific uals with heterozygous STAT1 mutations have impaired
CD4 T cells expressing CXCR3 localized to the lung nuclear accumulation of GAF and suffer from recur-
parenchyma and were more efficient at controlling bac- rent mycobacterial infections (289). Additionally, mu-
teria following M. tuberculosis infection when compared tations affecting IL-12 expression levels and signaling

12 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Immunology of M. tuberculosis Infections

also confer susceptibility to mycobacterial infections. (314), it is possible that adaptive immunity at stages of
Two mutations in the leucine zipper domain of NEMO, infection when bacterial burden is high may be com-
an intracellular protein involved in NF-κB activation, promised by T cell-derived IL-10. T-bet is a member
impairs CD40-mediated IL-12 production in mono- of the T-box family of transcription factors that is en-
cytes and DCs (290) and leads to recurrent mycobacte- coded by Tbx21 and is the master transcriptional regu-
rial infections. Similarly, defects that impair IL-12p40 lator for lineage commitment to the Th1 subset (315).
lead to decreased IFN-γ levels and confer susceptibility Interestingly, adoptive transfer of T-bet knockout ESAT-
to mycobacterial infections (291–294). Mutations in 6-specific T-cell receptor-transgenic CD4 T cells skewed
IL12RB are the most frequent genetic factors associated toward Th1 in vitro retains the capacity for early pro-
with Mendelian susceptibility to mycobacterial disease, tection against M. tuberculosis infection (316), suggesting
but recurrent mycobacterial susceptibility in individ- that protection conferred by Th1 cells may be indepen-
uals with IL12RB mutations can be mitigated with dent of T-bet or IFN-γ production. Taken together, these
BCG vaccination or primary BCG disease (291, 292, studies demonstrate a clear requirement for the IL-12/
295–298), suggesting that IL-12/IL-23 signaling may not IFN-γ axis in immunity against M. tuberculosis infection
be completely required for secondary immunity. IFN-γ in humans and animal models. Further studies delineat-
is readily detected in human BAL in patients with TB ing the mechanisms underlying IFN-γ- and Th1-mediated
disease and decreases following therapy (299), which is immunity against M. tuberculosis are warranted.
likely a consequence of decreasing bacterial loads. In Although Th1 responses are important for immunity
contrast, studies of human peripheral blood mononu- against TB, studies have also demonstrated that CD4
clear cells show a decrease in IFN-γ responses in ATB T-cell subsets secreting IL-17 (Th17) and FoxP3+ regu-
patients compared to controls (300–305). Lower fre- latory CD4 T cells contribute to the response against
quencies of M. tuberculosis-specific IFN-γ responses in M. tuberculosis infection. There are context-dependent
ATB patients may reflect trafficking of these cells to the beneficial or detrimental roles for Th17s during infec-
lungs, resulting in specific depletion from the periphery. tion with M. tuberculosis. Infection with a W-Beijing
IFN-γ secretion is also an important tool leveraged for lineage strain of M. tuberculosis, HN878, induce Th17
the detection of M. tuberculosis-specific CD4 T-cell responses, and mice deficient in IL-17 display increased
responses in humans and in animal models. Genome- bacterial burden following infection (317). IL-17 re-
wide analysis of M. tuberculosis-specific CD4 T-cell ceptor A subunit knockout mice (318) and IL-17A
epitopes in LTBI individuals revealed three broadly knockout mice (319) also displayed impaired long-term
immunodominant antigenic islands related to bacterial control of high-dose infection with H37Rv. Transfer of
secretion systems recognized by IFN-γ secreting CD4 BCG-specific, IFN-γ knockout Th17 cells into M. tuber-
T cells (306). Animal models of TB also demonstrate a culosis infected, T cell-deficient mice conferred enhanced
key role for IFN-γ in immunity against M. tuberculosis protection and prolonged survival compared to transfer
infection. Mice deficient in IFN-γ succumb to low-dose of naive IFN-γ knockout CD4 T cells (320), suggesting
M. tuberculosis infection (307, 308). Correspondingly, that Th17 cells can mediate protection independently
mice lacking IL-12 are also unable to control M. tuber- of IFN-γ. In humans, significant frequencies of IL-17-
culosis infection (253, 309, 310). The antimycobacterial producing CD4 T cells were found in the peripheral
effects of IFN-γ in mouse models are broadly related to blood mononuclear cells and BAL of BCG-vaccinated
the induction of AMPs, iNOS, and cytokines that acti- healthy individuals and declined in patients with active
vate infected macrophages to restrict intracellular bac- disease (321). Further, individuals with bi-allelic RORC
terial replication, though other mechanisms underlying loss-of-function mutations displayed impaired IL-17
IFN-γ-mediated immunity to M. tuberculosis infection and IFN-γ responses and were susceptible to mycobac-
are still being elucidated. IL-10-deficient mice are less terial disease and candidiasis (322). The generation of
susceptible to M. tuberculosis infection due to an en- Th17 responses to M. tuberculosis in vitro requires
hanced Th1 response (311), suggesting that IL-10 limits costimulation through the CD40-CD40L pathway since
Th1 immunity during M. tuberculosis infection. How- the absence of CD40 on DCs or CD40L on CD4 T cells
ever, Th1 cells secreting IL-10 can also impair host con- attenuates antigen-specific IL-17 responses (268). Acti-
trol of M. tuberculosis infection (312), and CD4 T cells vation of M. tuberculosis-infected DCs through CD40
producing both IFN-γ and IL-10 are detected in the BAL promoted enhanced antigen-specific Th1 and Th17
of ATB patients (313). Given that IL-10 secretion by Th1 responses that contributed to better control of bacterial
cells has been shown to be a result of high antigen dose burden in vivo (268), suggesting that a balanced Th1

ASMscience.org/MicrobiolSpectrum 13
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Sia and Rengarajan

and Th17 response is desirable for immunity against (339), highlighting a role for TLR-2-mediated recruit-
M. tuberculosis. The precise role of Th17 cells in pro- ment of T-regs in limiting tissue pathology at chronic
tective immunity to M. tuberculosis remains unclear but stages of disease. Taken together, these results suggest
may be related to their role in the development of less that the functional contribution of T-regs to immunity
hypoxic granulomas (323), in the recruitment of Th1 against M. tuberculosis infection and outcome of disease
cells (324), or in the induction of CXC-chemokines and may be dependent on multiple factors, including strain of
B-cell follicles (325). However, unrestrained IL-17 res- bacteria and stage of infection.
ponses have also been shown to promote detrimental In humans and animal models, M. tuberculosis
immunopathology, typically through pathological neu- establishes a persistent infection despite the induction
trophilia. IFN-γR1 knockout animals (326) or IFN-γR1 of adaptive immune responses. Persistent inflammation
bone marrow chimeric mice selectively lacking the re- and chronic antigen exposure precedes functional ex-
ceptor in nonhematopoietic cells (327) display amplified haustion due to chronic antigenic stimulation. In con-
Th17 responses following M. tuberculosis infection that trast to the expression of Ag85B, which decreases early
lead to a pathogenic accumulation of neutrophils detri- following infection, ESAT-6 is expressed by M. tuber-
mental to the host, suggesting that IFN-γ signaling serves culosis throughout infection (340, 341). Multiple studies
a regulatory role by limiting excessive IL-17-mediated examining CD4 T-cell responses to ESAT-6 and Ag85B
neutrophilia. have suggested that antigen-specific responses are dic-
FoxP3+ CD4 T cells, or T-regulatory cells (T-regs), tated by bacterial expression of those antigens through-
can impair antimycobacterial T-cell responses and con- out infection. CD4 T cells specific for ESAT-6 display a
tribute to disease but can also limit overt inflammation. terminally differentiated phenotype with evidence for
FoxP3+ T-regs can be found in the peripheral blood and functional exhaustion, which runs in contrast to Ag85B-
airways of M. tuberculosis-infected macaques (328) and specific CD4 T cells that appear functional but are quickly
humans (329–334). In mice, T-regs accumulate in the diminished (272, 342–347). Indeed, a vaccine that con-
lung-draining lymph nodes and the lungs following low- tains ESAT-6, Ag85B, and Rv2660c, which is expressed
dose aerosol M. tuberculosis infection (335). Importantly, at late stages of infection, demonstrated enhanced efficacy
FoxP3+ T-regs localized to pulmonary areas adjacent to compared to BCG or to a vaccine containing ESAT-6 and
effector CD4 T cells and depletion of T-regs before and Ag85B (348), suggesting that rational incorporation of
early after infection-enhanced control of bacterial burden antigens present at different stages of infection may im-
(335). Further, M. tuberculosis-specific T-regs delay the prove vaccine efficacy. A clearer understanding of pro-
expansion of antimycobacterial CD4 and CD8 T cells tective CD4 T-cell immunity will require further studies
and, consequently, transfer of M. tuberculosis-specific of the spectrum of antigens recognized by CD4 T cells
T-regs confers increased susceptibility to infection (336). following infection with M. tuberculosis in animal models
Regulation of T-regs during M. tuberculosis infection and in humans.
may be mediated by Th1 responses since M. tuberculosis-
specific T-regs are selectively eliminated following IL-12 Role of CD8 T Cells in M. tuberculosis Infection
driven T-bet expression (337). The functional properties Mice with gene deletion of β2 microglobulin, which
of T-regs responsible for limiting antimycobacterial CD4 abrogates MHC class I antigen presentation, or mice
and CD8 responses remains unclear. IL-10 was not found depleted of CD8 T cells live longer than corresponding
to be secreted by T-regs in mice infected with H37Rv disruptions to the MHC class II pathway or CD4 T-cell
(335). In contrast, T-regs from mice infected with the responses following M. tuberculosis infection (270).
W-Beijing strain, HN878, were found to secrete IL-10, Regardless, CD8 T cells contribute significantly to im-
express inhibitory receptors, and expand to greater de- munity against M. tuberculosis infection. Mice lacking
grees compared to infection with H37Rv (209), sug- TAP-1 (transporter associated with antigen processing
gesting that IL-10 secretion by T-regs may be dependent 1) antigen presentation molecules have deficient CD8
on bacterial strain. Notably, the expansion of T-regs in T-cell responses and succumb more rapidly following
the lungs of mice and outbred guinea pigs infected with M. tuberculosis infection compared to wild-type con-
W-Beijing strains occurred concurrently with a loss of trols (349, 350). Depletion of CD8 T cells in rhesus
Th1 responses and is associated with severe pulmo- macaques compromises protective immunity from BCG
nary pathology (209, 338). However, progressive loss vaccination or chemotherapeutic interventions (57), sug-
of T-regs in chronically infected TLR-2 knockout mice gesting that CD8 T cells are important components of
was associated with increased pulmonary inflammation recall responses to M. tuberculosis infection. Similarly,

14 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Immunology of M. tuberculosis Infections

in a mouse model of latency induced by antibiotic treat- itory molecules, including PD-1 and CTLA-4, among
ment, CD8 T-cell responses were found to be important M. tuberculosis-specific CD4 T cells has been shown to
in preventing reactivation (351). The importance of decrease following treatment (366, 367). Importantly,
CD8 T cells during M. tuberculosis infection is related expression of PD-1 on antigen-specific CD4 T cells from
to their secretion of cytokines and cytolytic effector LTBI was not associated with decreased effector func-
molecules that can limit bacterial replication. In addi- tions, and these cells proved to be polyfunctional with
tion to IFN-γ and TNF-α, CD8 T cells secrete perforin to respect to cytokine production upon antigen restimula-
lyse M. tuberculosis-infected macrophages (352). CD8 tion (368), suggesting that PD-1 may be an indicator of
T cells can also release granulysin in cytotoxic granules bacterial burden and CD4 T-cell activation rather than
to directly kill intracellular M. tuberculosis (353, 354). functional exhaustion. However, there is some in vitro
The use of anti-TNF-α therapy in patients with rheu- evidence from human samples suggesting that blockade
matoid arthritis depletes a subset of effector memory of PD-1/PD-L1 interaction can prevent M. tuberculosis-
CD8 T cells that secrete granulysin and express cell specific CD4 T-cell apoptosis (369) and enhance CD8
surface TNF (355), which may partially explain the in- T-cell degranulation and antigen-specific IFN-γ responses
creased progression from LTBI to ATB in patients under- from the peripheral blood mononuclear cells of a subset
going anti-TNF-α therapy. Human CD8 T cells respond of high-responding ATB patients (370). There is evidence
to epitopes in CFP10 (356), ESAT-6 (357, 358), and the that T-cell responses during ATB disease are less poly-
Ag85 complex (359, 360). A variety of human CD8 T-cell functional and have limited proliferative capacity com-
clones tested against a panel of synthetic peptides derived pared to LTBI individuals (371, 372), but whether this
from immunodominant M. tuberculosis antigens revealed functional impairment is mediated by inhibitory recep-
that CD8 T-cell responses are concentrated toward a tors such as PD-1 remains unclear. PD-1-deficient mice
limited set of epitopes and are generally restricted by the infected with M. tuberculosis have increased bacterial
HLA-B allele (361, 362). M. tuberculosis escape from the burden, neutrophilic infiltration, overt inflammation,
phagosome and induction of apoptosis by M. tuberculosis- tissue necrosis, and diminished lifespan compared to
infected macrophages can promote cross-presentation of wild-type mice (373), suggesting that PD-1 is required to
M. tuberculosis antigens to CD8 T cells. However, as pre- prevent aberrant inflammation during M. tuberculosis in-
viously discussed, virulent M. tuberculosis has been shown fection. Further, adoptive transfer studies demonstrated
to inhibit host apoptosis and favor necrosis to circumvent that PD-1-expressing CD4 T cells are highly proliferative
efficient induction of CD8 T-cell responses. All considered, (342, 346), and CD4 T cells lacking PD-1 can drive pa-
CD8 T cells are a critical component of adaptive immunity thology and mortality following M. tuberculosis infec-
to M. tuberculosis infection and play an important role in tion (374), together suggesting that PD-1 may mark
different disease contexts by limiting reactivation during functional CD4 T cells with intrinsic capacity for im-
latency and by directly participating in antimicrobial munoregulation. T-cell immunoglobulin and mucin do-
functions during active infections. main-containing 3 (Tim-3) is another inhibitory receptor
shown to play a role in mediating antimicrobial re-
Inhibitory Receptors During sponses by binding to one of its ligands, galectin-9 (375),
M. tuberculosis Infection and inducing the production of IL-1β by human and
Chronic viral infections, such as HIV, induce the ex- murine macrophages infected with M. tuberculosis (375,
pression of coinhibitory receptors on the surface of 376). In contrast to PD-1, Tim-3-deficient mice were
T cells that can dampen T-cell functionality. Abrogation less susceptible to M. tuberculosis infection, and Tim-3
of coinhibitory receptor ligation has been shown to blockade was shown to improve antigen-specific CD4
be a viable strategy to revitalize functionally exhausted and CD8 T-cell cytokine expression (377), suggesting
virus-specific T-cell responses. The evidence for the im- that Tim-3 may play a role in limiting T-cell responses
portance of coinhibitory receptors during M. tubercu- by promoting functional exhaustion. However, Tim-3-
losis infection in animal models and in human samples expressing, M. tuberculosis-specific T cells from ATB
varies between human and small animal models and patients were functionally superior to T cells expressing
between the specific inhibitory receptor studied. PD-1, low levels of Tim-3. Further, small interfering RNA- or
CD160, and 2B4 are inhibitory receptors associated antibody-mediated disruption of Tim-3 signaling on the
with CD8 T-cell dysfunction in chronic viral infections T cells from ATB patients led to attenuated IFN-γ and
(363, 364) but are expressed at low levels on M. tuber- TNF-α production by Tim-3-expressing T cells, while
culosis-specific CD8 T cells (365). Expression of inhib- Tim-3 ligation augmented IFN-γ production (378). The

ASMscience.org/MicrobiolSpectrum 15
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Sia and Rengarajan

mechanisms underlying the roles of receptors such as revealed significant changes in B-cell-associated genes in
PD-1 and Tim-3 require further study and may deviate TB patients after initiation of TB treatment (394). No-
from their role in viral immunity. The evidence accu- tably, antibodies to M. tuberculosis proteins have been
mulated thus far suggests that these molecules mark reported in the sera of TB patients (395), and antibodies
functional T cells that play important roles in antimi- identified in a subset of health care workers exposed
crobial activity and prevention of uncontrolled inflam- to M. tuberculosis provide modest protection in vitro
mation following M. tuberculosis infection. and in a mouse model of infection (396). Utilization
of a high-throughput approach to identifying antibody
Memory T-Cell Responses targets in the M. tuberculosis proteome revealed a set
In humans, antigen-specific memory T-cell responses of extracellular antigens recognized by antibodies in
have been detected in individuals with LTBI and in TB the plasma of patients with ATB (397), suggesting that
patients following successful treatment and cure. Mem- B cells are active participants in immunity to M. tuber-
ory T-cell subsets can be identified according to their culosis infection. B-cell-deficient mice have elevated
cell surface phenotype and functional properties, and neutrophilic recruitment and exacerbated lung immu-
distinct populations of antigen-specific memory T cells nopathology following M. tuberculosis infection (398),
can be categorized based on their expression of a panel which is mediated through enhanced IL-17 responses
of cell surface activation markers and chemokine recep- in M. tuberculosis-infected B-cell-deficient or B-cell-
tors (379). Characterization of M. tuberculosis-specific depleted animals (399). These studies suggest that B cells
memory CD4 T cells in LTBI indicated that these cells can influence the outcome of M. tuberculosis infection
did not express activation markers and were largely of by moderating inflammatory responses. Antibody pro-
a CD45RA-CCR7 phenotype descriptive of T effector duction by B cells can promote divergent outcomes
memory cells (368, 380). In contrast, analysis of LTBI (400). Binding of antibody to the inhibitory Fc gamma
individuals using MHC class II tetramers revealed a pop- receptor II B attenuates macrophage IL-12 produc-
ulation of tetramer+CD45RA-CCR7+ central memory tion and negatively impacts Th1 responses (401), while
CD4 T cells that further expressed CXCR3+CCR6+ passive transfer of monoclonal antibodies specific for
(306), highlighting the heterogeneity of memory CD4 M. tuberculosis cell wall components can improve the
T-cell phenotypes that can vary based on antigen spec- outcome of infection in mice (390). B-cell secretion of
ificity, disease status, and manner in which specific re- cytokines can also influence M. tuberculosis-infected
sponses are identified. Human memory CD8 T cells are macrophages. Type I IFN expression by murine B cells
predominantly terminally differentiated effector memory and B cells from pleural effusion of TB patients altered
T cells in individuals with LTBI (365, 381). Memory macrophage polarization toward an anti-inflammatory
T-cell responses have also been studied in the context phenotype (402). Taken together, these studies highlight
of “memory-immune” mice, which are M. tuberculosis- a role for B cells, which constitute a significant popula-
infected mice that subsequently receive antibiotic treat- tion of lymphocytes around lung granulomas in the
ment. In this context, both memory CD4 (382, 383) and adaptive immune response to M. tuberculosis infection
CD8 (384, 385) T cells play a role in immunity against by modulating inflammation through the secretion of
M. tuberculosis infection. T cells from memory-immune antibodies and cytokines.
mice expanded rapidly, secreted IFN-γ, and conferred a
significant level of protection at early timepoints after γδ, CD1-Restricted T Cells, and MAIT Cells
infection (383, 386–388) but are ultimately unable to in Immunity against M. tuberculosis
confer long-term protection (389), suggesting that mem- γδ T cells are a population of T cells that express a re-
ory T cells generated after primary M. tuberculosis in- stricted repertoire of T-cell receptor genes, recognize
fection have limited capacity to protect from reinfection. nonpeptide antigens such as microbial metabolites and
phosphoantigens (403), and can be found at mucosal
B-Cell and Antibody Responses surfaces including the lung (404). γδ T cells proliferate
During M. tuberculosis Infection when exposed to M. tuberculosis-infected monocytes
There is a body of evidence suggesting that humoral (405). Multiple M. tuberculosis metabolites, includ-
immunity plays a role in defense against M. tuberculosis ing pyrophosphate, prenyl pyrophosphate derivatives
infection (reviewed in 390). B cells can be found along- (406, 407), and triphosphorylated thymidine-containing
side T cells in the lymphocytic cuff in human granulomas compounds (408), are recognized by human γδ T cells.
(391–393), and whole blood gene expression analysis Human γδ T cells can also respond to mycobacterial

16 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Immunology of M. tuberculosis Infections

heat shock proteins (409), though this response may family of M. tuberculosis lipopeptides called didehydroxy-
be dependent on BCG immunization (410). Vγ9Vδ2- mycobactins are presented by CD1a (435), and a variety
expressing γδ T cells represent a significant proportion of phospholipid antigens are presented by CD1c (425).
of M. tuberculosis-reactive T cells in peripheral blood The precise role of CD1-restricted T cells in immunity
(411–413) and can restrict intracellular M. tuberculosis during M. tuberculosis infection remains unclear, and
replication in macrophages (414). Interestingly, Vγ9Vδ2 further studies of their function in the periphery and es-
T cells can function as antigen presentation cells via pecially in BAL would inform their potential as targets for
provision of CD40 costimulation to promote the ex- TB vaccines.
pansion of αβ T cells with enhanced capacity to restrict MAIT cells are a subset of T cells with innate-like
intracellular BCG replication (415). Additionally, human qualities enriched in mucosal tissues, including the in-
Vγ2Vδ2 T cells recognize M. tuberculosis (416), and in testinal mucosa, lung, and liver (436–438). These cells
NHPs, Vγ2Vδ2 T cells are expanded by phosphoantigen recognize antigen through a nonpolymorphic MHC class
and IL-2 administration (417). Adoptively transferred I-related molecule 1 (439) presenting pterin-containing
Vγ2Vδ2 T cells into naive animals confer protection byproducts of riboflavin synthesis in bacteria and fungi
against M. tuberculosis infection (418). γδ T cells have (440). In humans, MAIT cells express a semi-invariant
been shown to mediate direct killing of M. tuberculosis Vα7.2 and CD161 and can either be double negative
via secretion of granulysin and perforin (419) or through for CD4 and CD8 or CD4-CD8+ (436, 441). MAIT cells
the induction of TNF-α by monocytes (420). There is also have been described in the peripheral blood of healthy
evidence that γδ T cells can influence DC cross talk with individuals and are depleted in ATB patients (442),
T cells by promoting DC maturation and expression of possibly reflecting migration into the lung. These cells
costimulatory molecules (421). In mice, γδ T cells accu- produce IFN-γ and TNF-α upon activation (442, 443),
mulate in the lung-draining lymph nodes, are responsive but their contribution to the immune response to M. tu-
to M. tuberculosis antigen independent of MHC class II berculosis infection requires further study.
(422), and are significant sources of early IL-17 produc-
tion following M. tuberculosis infection (423).
Due to the large repertoire of glycolipids present on INITIATION AND HETEROGENEITY
the mycobacterial cell wall, a significant T-cell response OF THE GRANULOMA
is directed at glycolipid antigens presented by the CD1 The granuloma is a hallmark histopathological structure
family of molecules. CD1 molecules are a family of in TB. It represents host sequestration of bacteria to limit
MHC class I-like antigen presentation molecules that dissemination as well as a niche for long-term persis-
present glycolipid antigens to T cells. There are five CD1 tence of M. tuberculosis. Further, the selectively drug-
family members in humans, split into two groups based permeable nature of the TB granuloma can diminish
on sequence homology. Group 1 molecules include CD1a, the efficacy of drugs meant to treat persistent bacteria
CD1b, CD1c, and CD1e. CD1d is the sole inclusion in (444). The granuloma is composed of an aggregate of
group 2 (424). Mycobacterial lipids are readily presented M. tuberculosis-infected and -uninfected macrophages
by CD1 molecules in human cells, but mechanistic studies in varying stages of maturation and differentiation
of this family of molecules is limited because mice only (445–447). Macrophages in the granuloma can undergo
express two orthologs of CD1d and do not express group an epithelioid transformation, become lipid-filled foamy
1 molecules. Nevertheless, studies in human cells revealed macrophages, or merge into multinucleated giant cells.
that mycobacterial lipids presented by group 1 CD1 mole- This central core of macrophages is accompanied by
cules promote T-cell proliferation and cytokine produc- neutrophils, DCs, and fibroblasts circumscribed by T
tion (425–432). Mycobacterial glycerol monomycolate, and B lymphocytes and progressively becomes a hypoxic
glucose monomycolate, sulphoglycolipids, and mycolic environment where many cells undergo necrotic death to
acid can be presented through CD1b (426–428, 433). form an acellular core termed the caseum (448). The
CD1b-restricted T cells expand and secrete IFN-γ and IL-2 granuloma is a hallmark structure in human TB that is
upon interaction with cognate antigen and contract fol- modeled variably among available animal models.
lowing anti-TB therapy (430). Interestingly, use of CD1b C57BL/6 and BALB/C mice do not naturally recapitulate
tetramers loaded with glucose monomycolate revealed the human granuloma in that lung lesions are rarely
that CD1b-restricted T cells are antigen-specific and also necrotic and caseating. The animal models that most
express CD4 (429, 434). M. tuberculosis lipids presented closely recapitulate the heterogeneity of human granu-
through CD1a and CD1c have also been identified. A lomas include certain susceptible inbred mouse strains

ASMscience.org/MicrobiolSpectrum 17
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Sia and Rengarajan

that present with necrotizing granulomas (C3HeB/FeJ, (454). T-cell functionality in the granuloma may there-
DBA/2, CBA/J, I/St), guinea pigs, rabbits, and the NHP fore be a function of disease status and proximity to the
model. Additionally, the zebrafish model has also bacteria-containing, hypoxic, and necrotic core of the
yielded fundamental insights into the initiation and dy- TB granuloma. Additionally, T cells near the granuloma
namics of the tuberculous granuloma. can be negatively impacted by the depletion of key
The transparency of zebrafish larvae has made direct amino acids required for proper function. As mentioned
visualization of the initiation of the granuloma possible previously, IDO, an enzyme that functions in the ca-
following infection with M. marinum (48). Studies based tabolism of tryptophan, is expressed by cells in the core
on this model have revealed that the innate immune re- of the granulomas of rhesus macaques infected with
sponse is sufficient to initiate the granuloma following M. tuberculosis (283), and inhibition of IDO promoted
infection. Recruitment of additional macrophages me- granuloma reorganization and attenuated disease (66).
diated, in part, by mycobacterial ESX-1 proteins initiates The functionality of T cells within granulomas may also
a cascade of events that leads to the establishment of be regulated by direct cross talk with infected myeloid
the mycobacterial granuloma (221, 449). Importantly, cells, including macrophages and DCs. Intravital imag-
recruited macrophages can traffic through the initial ing of mycobacteria-induced liver granulomas revealed
granuloma to phagocytose apoptotic infected macro- limited antigen-specific T-cell migration arrest in re-
phages and egress to form distal secondary granulomas sponse to infected myeloid cells (345), suggesting that
(450). Mycobacterial lipids play a key role in establish- T cells do not interact meaningfully with infected cells in
ing the granuloma by limiting macrophage effector granulomas. Taken together, these studies highlight the
functions and promoting the recruitment of additional vast complexity and heterogeneity of the TB granuloma.
macrophages to facilitate dissemination. In particular,
mycobacterial phthiocerol dimycocerosate can mask
TLR-signaling and prevent induction of nitrosative IMMUNOLOGY OF TB DIAGNOSTICS
stresses (451), and mycobacterial PGL can induce mac- TB diagnosis relies on evaluation of clinical symptoms
rophage production of CCL2 to recruit CCR2+ mono- and patient history combined with radiographic exam-
cytes that permit bacterial dissemination (452). These ination and detection of bacteria in sputum (9). The
studies collectively indicate that the initiation of the presence of acid-fast bacilli in sputum smears by mi-
mycobacterial granuloma is dependent on recruitment croscopy does not specifically indicate infection with
of bacteria-permissive macrophages and monocytes fol- M. tuberculosis; microbiological culture and nucleic
lowing initial infection and can be mediated by myco- acid amplification-based tests are required to confirm the
bacterial secreted factors and membrane lipids. presence of M. tuberculosis infection. Xpert MTB/RIF, a
TB granulomas can vary in their cellular composition, cartridge-based near-patient diagnostic assay utilizing
oxygenation levels, inflammatory milieu, and bacterial real-time nucleic acid amplification of M. tuberculosis
burden. This heterogeneity can exist between and within DNA, which also detects drug resistance to the first-line
infected hosts. Infection of cynomolgus macaques with drug rifampicin, is recommended by the World Health
a panel of M. tuberculosis isolates that differed by a Organization for TB diagnosis (455, 456). IFN-γ release
single nucleotide polymorphism revealed that individual assays (IGRAs), which leverage the specificity of the
granulomas can be founded by a single bacterium and immune response to M. tuberculosis, are the basis of
can vary in their bacterial burden compared to other the QuantiFERON-TB Gold In-Tube and T-SPOT.TB
granulomas within the same host (51). Analysis of T-cell diagnostic assays. IGRAs measure IFN-γ produced by
functionality between sterile and nonsterile granulomas antigen-specific T cells in blood that recognize M. tuber-
revealed a modest association between IL-10 and IL-17 culosis antigens (ESAT-6, CFP-10, TB7.7) (457). IGRAs
responses and clearance of M. tuberculosis in sterile provide increased specificity over traditional Mantoux
granulomas (453). However, in the context of TNF-α skin tests that depend on delayed-type hypersensitivity
neutralization in latently infected macaques, IL-10 and reactions to purified protein derivative, which is not
IL-17 responses were associated with animals at higher specific to M. tuberculosis infection, and positive results
risk of reactivation (71). Proteome analysis of laser- may be due to BCG vaccination or exposure to envi-
capture microdissected human and rabbit lung lesions ronmental mycobacteria. However, IGRAs do not dif-
suggests that inflammatory responses typical of the ferentiate between active and latent TB and cannot be
center of the TB granuloma are physically segregated used to diagnose TB disease. While sputum-based smear
from anti-inflammatory responses in adjacent lung tissue and culture techniques are established worldwide for

18 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Immunology of M. tuberculosis Infections

clinical indication of M. tuberculosis infection, collec- aim for in a TB vaccine. MVA85A is a modified vaccinia
tion of sputum, especially from children, can be chal- Ankara virus expressing Ag85A from M. tuberculosis
lenging and is not completely reliable. Therefore, there that was utilized as a booster vaccine in infants previ-
is interest in developing non-sputum-based diagnostic ously vaccinated with BCG (472). Notably, vaccination
approaches for TB. Detection of urinary lipoarabino- with MVA85A enhanced frequencies of antigen-specific,
mannan in suspected TB cases is being investigated in polyfunctional CD4 T cells co-expressing IFN-γ, IL-2,
HIV-infected (458) and -uninfected (459) individuals. and TNF-α (472). Although MVA85A vaccination en-
Blood-based biomarkers discriminating between LTBI and hanced antigen-specific CD4 T-cell responses, it did not
ATB are being investigated for potential application to TB provide added protection against TB disease in infants
diagnostics and treatment response (460–462). HLA-DR, (472). Other viral-vectored vaccines in various stages of
CD38, and Ki67 expression on M. tuberculosis-specific development include Ad5Ag85A (473), ChAdOx1.85A
CD4 T cells from peripheral blood is reported to be a + MVA85A (474), MVA85A-IMX313 (475), and TB/
highly specific and sensitive method to discriminate LTBI FLU-04L. Additionally, a recent study utilizing a recom-
and ATB and evaluate treatment response (460). A recent binant cytomegalovirus demonstrated protection in rhesus
study suggests that HLA-DR could function as a robust macaques (476). While viral vectors do not require the use
marker distinguishing LTBI and ATB in HIV-infected of adjuvants, previous exposure to the vector may attenu-
populations (461). Further understanding of the spectrum ate vaccine-induced responses and represents a potential
of antigen-specific responses to M. tuberculosis infection complication to the use of viral vectors. Whole-cell vaccines
can be leveraged to develop diagnostics that can monitor currently under development include killed Mycobacte-
infection and treatment response. rium vaccae, DAR-901 (477), VPM1002, MTBVAC, and
RUTI. VPM1002 is an approach to improve BCG immu-
nogenicity and vaccine potential by engineering BCG to
TB VACCINES express lysteriolysin from Listeria monocytogenes to es-
The only currently licensed vaccine against TB is ba- cape the phagosome and carry a urease deletion mutation
cillus Calmette-Guérin (BCG), an attenuated strain of that facilitates phagosomal acidification, thereby enhancing
M. bovis (463, 464). BCG confers protection against MHC class I antigen presentation to CD8 T cells (478).
severe forms of TB, including miliary TB and TB men- MTBVAC is a genetically attenuated M. tuberculosis strain
ingitis (465), but does not reliably protect against pul- lacking phoP and fadD26 that abrogates synthesis of var-
monary TB in children or adults (3, 4, 466). The lack ious surface lipids (479). Lastly, the therapeutic vaccine
of validated correlates of protection against TB is a se- candidate RUTI was developed by growing M. tuberculosis
vere limitation to TB vaccine development. Despite the under stress prior to fragmentation, detoxification, and
importance of IFN-γ responses in resistance against delivery in liposomes to individuals with LTBI to prevent
M. tuberculosis infection in humans and animal models, progression to ATB (480–482).
accumulating evidence suggests that induction of en- There have been substantial advances in our under-
hanced IFN-γ responses is not sufficient to obtain a more standing of immunity against M. tuberculosis from the
efficacious TB vaccine. Indeed, the frequency and func- days of Drs. Calmette and Guérin. Nevertheless, the ab-
tional profile of BCG-specific CD4, CD8, and γδ T cells sence of suitable alternatives to BCG highlights the
from whole blood, including IFN-γ-producing T cells, challenges before us. M. tuberculosis is adept at sub-
did not correlate with protection against TB in newborns verting the cross talk between innate and adaptive im-
(467). As of 2017, there are 14 TB vaccine candidates in munity, and it will be important to understand that cross
varying phases of clinical development representing talk for the rational development of better vaccines. Even
three broad strategies: subunit vaccines pairing M. tu- in the absence of protective correlates and in the face of
berculosis antigens with adjuvants, viral-vectored vac- disappointing preliminary results for MVA85A, the state
cines utilizing an attenuated virus for antigen delivery, of TB vaccine development is resurgent now more than
and whole-cell vaccines utilizing attenuated M. tuber- ever and provides cause for optimism for the development
culosis or related mycobacterial species. Protein subunit of more efficacious vaccines and therapeutics against TB.
vaccines currently under clinical development include
M72/AS01E (468), H4:IC31 (469), H56:IC31 (470), REFERENCES
1. WHO. 2017. Global Tuberculosis Report. World Health Organization,
and ID93/GLA-SE (471). Among viral-vectored vac- Geneva, Switzerland.
cines, results from the MVA85A phase IIb clinical trial 2. Abubakar I, Pimpin L, Ariti C, Beynon R, Mangtani P, Sterne JA,
have prompted reevaluation of immune correlates to Fine PE, Smith PG, Lipman M, Elliman D, Watson JM, Drumright LN,

ASMscience.org/MicrobiolSpectrum 19
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Sia and Rengarajan

Whiting PF, Vynnycky E, Rodrigues LC. 2013. Systematic review and 16. Geldmacher C, Ngwenyama N, Schuetz A, Petrovas C, Reither K,
meta-analysis of the current evidence on the duration of protection by Heeregrave EJ, Casazza JP, Ambrozak DR, Louder M, Ampofo W,
bacillus Calmette-Guérin vaccination against tuberculosis. Health Tech- Pollakis G, Hill B, Sanga E, Saathoff E, Maboko L, Roederer M, Paxton
nol Assess 17:1–372, v–vi http://dx.doi.org/10.3310/hta17370. WA, Hoelscher M, Koup RA. 2010. Preferential infection and depletion
3. Roy A, Eisenhut M, Harris RJ, Rodrigues LC, Sridhar S, Habermann S, of Mycobacterium tuberculosis-specific CD4 T cells after HIV-1 infection.
Snell L, Mangtani P, Adetifa I, Lalvani A, Abubakar I. 2014. Effect of J Exp Med 207:2869–2881 http://dx.doi.org/10.1084/jem.20100090.
BCG vaccination against Mycobacterium tuberculosis infection in chil- 17. Kalsdorf B, Scriba TJ, Wood K, Day CL, Dheda K, Dawson R,
dren: systematic review and meta-analysis. BMJ 349:g4643 http://dx.doi Hanekom WA, Lange C, Wilkinson RJ. 2009. HIV-1 infection impairs the
.org/10.1136/bmj.g4643. bronchoalveolar T-cell response to mycobacteria. Am J Respir Crit Care
4. Mangtani P, Abubakar I, Ariti C, Beynon R, Pimpin L, Fine PE, Rodrigues Med 180:1262–1270 http://dx.doi.org/10.1164/rccm.200907-1011OC.
LC, Smith PG, Lipman M, Whiting PF, Sterne JA. 2014. Protection by BCG 18. Bunjun R, Riou C, Soares AP, Thawer N, Müller TL, Kiravu A,
vaccine against tuberculosis: a systematic review of randomized controlled Ginbot Z, Oni T, Goliath R, Kalsdorf B, von Groote-Bidlingmaier F,
trials. Clin Infect Dis 58:470–480 http://dx.doi.org/10.1093/cid/cit790. Hanekom W, Walzl G, Wilkinson RJ, Burgers WA. 2017. Effect of HIV
5. Turner RD, Chiu C, Churchyard GJ, Esmail H, Lewinsohn DM, Gandhi on the frequency and number of Mycobacterium tuberculosis-specific
NR, Fennelly KP. 2017. Tuberculosis infectiousness and host susceptibil- CD4+ T cells in blood and airways during latent M. tuberculosis infection.
ity. J Infect Dis 216(Suppl 6):S636–S643 http://dx.doi.org/10.1093/infdis J Infect Dis 216:1550–1560 http://dx.doi.org/10.1093/infdis/jix529.
/jix361. 19. Rosas-Taraco AG, Arce-Mendoza AY, Caballero-Olín G, Salinas-
6. Churchyard G, Kim P, Shah NS, Rustomjee R, Gandhi N, Mathema B, Carmona MC. 2006. Mycobacterium tuberculosis upregulates coreceptors
Dowdy D, Kasmar A, Cardenas V. 2017. What we know about tuber- CCR5 and CXCR4 while HIV modulates CD14 favoring concurrent in-
culosis transmission: an overview. J Infect Dis 216(Suppl 6):S629–S635 fection. AIDS Res Hum Retroviruses 22:45–51 http://dx.doi.org/10.1089
http://dx.doi.org/10.1093/infdis/jix362. /aid.2006.22.45.
7. Mathema B, Andrews JR, Cohen T, Borgdorff MW, Behr M, Glynn JR, 20. Juffermans NP, Speelman P, Verbon A, Veenstra J, Jie C, van Deventer
Rustomjee R, Silk BJ, Wood R. 2017. Drivers of tuberculosis trans- SJ, van Der Poll T. 2001. Patients with active tuberculosis have increased
mission. J Infect Dis 216(Suppl 6):S644–S653 http://dx.doi.org/10.1093 expression of HIV coreceptors CXCR4 and CCR5 on CD4(+) T cells. Clin
/infdis/jix354. Infect Dis 32:650–652 http://dx.doi.org/10.1086/318701.
8. Barry CE III, Boshoff HI, Dartois V, Dick T, Ehrt S, Flynn J, 21. Mayanja-Kizza H, Wajja A, Wu M, Peters P, Nalugwa G, Mubiru
Schnappinger D, Wilkinson RJ, Young D. 2009. The spectrum of latent F, Aung H, Vanham G, Hirsch C, Whalen C, Ellner J, Toossi Z. 2001.
tuberculosis: rethinking the biology and intervention strategies. Nat Rev Activation of beta-chemokines and CCR5 in persons infected with human
Microbiol 7:845–855 http://dx.doi.org/10.1038/nrmicro2236. immunodeficiency virus type 1 and tuberculosis. J Infect Dis 183:1801–
9. Dheda K, Gumbo T, Maartens G, Dooley KE, McNerney R, Murray 1804 http://dx.doi.org/10.1086/320724.
M, Furin J, Nardell EA, London L, Lessem E, Theron G, van Helden P, 22. Morris L, Cilliers T, Bredell H, Phoswa M, Martin DJ. 2001. CCR5 is
Niemann S, Merker M, Dowdy D, Van Rie A, Siu GK, Pasipanodya JG, the major coreceptor used by HIV-1 subtype C isolates from patients with
Rodrigues C, Clark TG, Sirgel FA, Esmail A, Lin HH, Atre SR, Schaaf active tuberculosis. AIDS Res Hum Retroviruses 17:697–701 http://dx
HS, Chang KC, Lange C, Nahid P, Udwadia ZF, Horsburgh CR Jr, .doi.org/10.1089/088922201750236979.
Churchyard GJ, Menzies D, Hesseling AC, Nuermberger E, McIlleron H, 23. Santucci MB, Bocchino M, Garg SK, Marruchella A, Colizzi V, Saltini
Fennelly KP, Goemaere E, Jaramillo E, Low M, Jara CM, Padayatchi N, C, Fraziano M. 2004. Expansion of CCR5+ CD4+ T-lymphocytes in the
Warren RM. 2017. The epidemiology, pathogenesis, transmission, diag- course of active pulmonary tuberculosis. Eur Respir J 24:638–643 http://
nosis, and management of multidrug-resistant, extensively drug-resistant, dx.doi.org/10.1183/09031936.04.000105403.
and incurable tuberculosis. Lancet Respir Med 5:291–360.
24. Wolday D, Tegbaru B, Kassu A, Messele T, Coutinho R, van
10. Jones BE, Young SM, Antoniskis D, Davidson PT, Kramer F, Barnes Baarle D, Miedema F. 2005. Expression of chemokine receptors CCR5
PF. 1993. Relationship of the manifestations of tuberculosis to CD4 cell and CXCR4 on CD4+ T cells and plasma chemokine levels during treat-
counts in patients with human immunodeficiency virus infection. Am Rev ment of active tuberculosis in HIV-1-coinfected patients. J Acquir Immune
Respir Dis 148:1292–1297 http://dx.doi.org/10.1164/ajrccm/148.5.1292. Defic Syndr 39:265–271 http://dx.doi.org/10.1097/01.qai.0000163027
11. Leroy V, Salmi LR, Dupon M, Sentilhes A, Texier-Maugein J, Dequae .47147.2e.
L, Dabis F, Salamon R. 1997. Progression of human immunodeficiency 25. Day CL, Mkhwanazi N, Reddy S, Mncube Z, van der Stok M,
virus infection in patients with tuberculosis disease. A cohort study in Klenerman P, Walker BD. 2008. Detection of polyfunctional Mycobac-
Bordeaux, France, 1988-1994. The Groupe d’Epidémiologie Clinique du terium tuberculosis-specific T cells and association with viral load in HIV-
Sida en Aquitaine (GECSA). Am J Epidemiol 145:293–300 http://dx.doi 1-infected persons. J Infect Dis 197:990–999 http://dx.doi.org/10.1086
.org/10.1093/oxfordjournals.aje.a009105. /529048.
12. Havlir DV, Barnes PF. 1999. Tuberculosis in patients with human 26. Riou C, Bunjun R, Müller TL, Kiravu A, Ginbot Z, Oni T, Goliath R,
immunodeficiency virus infection. N Engl J Med 340:367–373 http://dx Wilkinson RJ, Burgers WA. 2016. Selective reduction of IFN-γ single
.doi.org/10.1056/NEJM199902043400507. positive mycobacteria-specific CD4+ T cells in HIV-1 infected individuals
13. Toossi Z. 2003. Virological and immunological impact of tuberculosis with latent tuberculosis infection. Tuberculosis (Edinb) 101:25–30 http://
on human immunodeficiency virus type 1 disease. J Infect Dis 188:1146– dx.doi.org/10.1016/j.tube.2016.07.018.
1155 http://dx.doi.org/10.1086/378676. 27. Strickland N, Müller TL, Berkowitz N, Goliath R, Carrington MN,
14. Sonnenberg P, Glynn JR, Fielding K, Murray J, Godfrey-Faussett P, Wilkinson RJ, Burgers WA, Riou C. 2017. Characterization of Myco-
Shearer S. 2005. How soon after infection with HIV does the risk of tu- bacterium tuberculosis-specific cells using MHC class II tetramers reveals
berculosis start to increase? A retrospective cohort study in South African phenotypic differences related to HIV infection and tuberculosis disease.
gold miners. J Infect Dis 191:150–158 http://dx.doi.org/10.1086/426827. J Immunol 199:2440–2450.
15. Geldmacher C, Schuetz A, Ngwenyama N, Casazza JP, Sanga E, 28. Day CL, Abrahams DA, Harris LD, van Rooyen M, Stone L, de Kock
Saathoff E, Boehme C, Geis S, Maboko L, Singh M, Minja F, Meyerhans M, Hanekom WA. 2017. HIV-1 infection is associated with depletion
A, Koup RA, Hoelscher M. 2008. Early depletion of Mycobacterium and functional impairment of Mycobacterium tuberculosis-specific CD4
tuberculosis-specific T helper 1 cell responses after HIV-1 infection. J T cells in individuals with latent tuberculosis infection. J Immunol 199:
Infect Dis 198:1590–1598 http://dx.doi.org/10.1086/593017. 2069–2080 http://dx.doi.org/10.4049/jimmunol.1700558.

20 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Immunology of M. tuberculosis Infections

29. Kalokhe AS, Adekambi T, Ibegbu CC, Ray SM, Day CL, Rengarajan 44. Manabe YC, Dannenberg AM Jr, Tyagi SK, Hatem CL, Yoder M,
J. 2015. Impaired degranulation and proliferative capacity of Mycobac- Woolwine SC, Zook BC, Pitt ML, Bishai WR. 2003. Different strains
terium tuberculosis-specific CD8+ T cells in HIV-infected individuals with of Mycobacterium tuberculosis cause various spectrums of disease in the
latent tuberculosis. J Infect Dis 211:635–640 http://dx.doi.org/10.1093 rabbit model of tuberculosis. Infect Immun 71:6004–6011 http://dx.doi
/infdis/jiu505. .org/10.1128/IAI.71.10.6004-6011.2003.
30. Fox GJ, Menzies D. 2013. Epidemiology of tuberculosis immunology. 45. Dorman SE, Hatem CL, Tyagi S, Aird K, Lopez-Molina J, Pitt ML,
Adv Exp Med Biol 783:1–32 http://dx.doi.org/10.1007/978-1-4614 Zook BC, Dannenberg AM Jr, Bishai WR, Manabe YC. 2004. Suscepti-
-6111-1_1. bility to tuberculosis: clues from studies with inbred and outbred New
31. Marais BJ, Lönnroth K, Lawn SD, Migliori GB, Mwaba P, Glaziou P, Zealand white rabbits. Infect Immun 72:1700–1705 http://dx.doi.org
Bates M, Colagiuri R, Zijenah L, Swaminathan S, Memish ZA, Pletschette /10.1128/IAI.72.3.1700-1705.2004.
M, Hoelscher M, Abubakar I, Hasan R, Zafar A, Pantaleo G, Craig G, 46. Tsenova L, Ellison E, Harbacheuski R, Moreira AL, Kurepina N,
Kim P, Maeurer M, Schito M, Zumla A. 2013. Tuberculosis comorbidity Reed MB, Mathema B, Barry CE III, Kaplan G. 2005. Virulence of se-
with communicable and non-communicable diseases: integrating health lected Mycobacterium tuberculosis clinical isolates in the rabbit model
services and control efforts. Lancet Infect Dis 13:436–448 http://dx.doi of meningitis is dependent on phenolic glycolipid produced by the bacilli.
.org/10.1016/S1473-3099(13)70015-X. J Infect Dis 192:98–106 http://dx.doi.org/10.1086/430614.
32. Cooper AM. 2014. Mouse model of tuberculosis. Cold Spring 47. Sun H, Ma X, Zhang G, Luo Y, Tang K, Lin X, Yu H, Zhang Y, Zhu
Harb Perspect Med 5:a018556 http://dx.doi.org/10.1101/cshperspect B. 2012. Effects of immunomodulators on liquefaction and ulceration in
.a018556. the rabbit skin model of tuberculosis. Tuberculosis (Edinb) 92:345–350
33. Fortin A, Abel L, Casanova JL, Gros P. 2007. Host genetics of my- http://dx.doi.org/10.1016/j.tube.2012.03.005.
cobacterial diseases in mice and men: forward genetic studies of BCG-osis 48. Davis JM, Clay H, Lewis JL, Ghori N, Herbomel P, Ramakrishnan L.
and tuberculosis. Annu Rev Genomics Hum Genet 8:163–192 http://dx 2002. Real-time visualization of mycobacterium-macrophage interactions
.doi.org/10.1146/annurev.genom.8.080706.092315. leading to initiation of granuloma formation in zebrafish embryos. Im-
34. Medina E, North RJ. 1998. Resistance ranking of some common munity 17:693–702 http://dx.doi.org/10.1016/S1074-7613(02)00475-2.
inbred mouse strains to Mycobacterium tuberculosis and relationship 49. Dee CT, Nagaraju RT, Athanasiadis EI, Gray C, Fernandez Del
to major histocompatibility complex haplotype and Nramp1 genotype. Ama L, Johnston SA, Secombes CJ, Cvejic A, Hurlstone AF. 2016. CD4-
Immunology 93:270–274 http://dx.doi.org/10.1046/j.1365-2567.1998 transgenic zebrafish reveal tissue-resident Th2- and regulatory T cell-like
.00419.x. populations and diverse mononuclear phagocytes. J Immunol 197:3520–
35. Sánchez F, Radaeva TV, Nikonenko BV, Persson AS, Sengul S, 3530 http://dx.doi.org/10.4049/jimmunol.1600959.
Schalling M, Schurr E, Apt AS, Lavebratt C. 2003. Multigenic control of 50. Kasheta M, Painter CA, Moore FE, Lobbardi R, Bryll A, Freiman E,
disease severity after virulent Mycobacterium tuberculosis infection in Stachura D, Rogers AB, Houvras Y, Langenau DM, Ceol CJ. 2017.
mice. Infect Immun 71:126–131 http://dx.doi.org/10.1128/IAI.71.1.126 Identification and characterization of T reg-like cells in zebrafish. J Exp
-131.2003. Med 214:3519–3530 http://dx.doi.org/10.1084/jem.20162084.
36. Marquis JF, Lacourse R, Ryan L, North RJ, Gros P. 2009. Genetic and 51. Lin PL, Ford CB, Coleman MT, Myers AJ, Gawande R, Ioerger T,
functional characterization of the mouse Trl3 locus in defense against Sacchettini J, Fortune SM, Flynn JL. 2014. Sterilization of granulomas is
tuberculosis. J Immunol 182:3757–3767 http://dx.doi.org/10.4049 common in active and latent tuberculosis despite within-host variability in
/jimmunol.0802094. bacterial killing. Nat Med 20:75–79 http://dx.doi.org/10.1038/nm.3412.
37. McCune RM Jr, McDermott W, Tompsett R. 1956. The fate 52. Flynn JL, Gideon HP, Mattila JT, Lin PL. 2015. Immunology studies
of Mycobacterium tuberculosis in mouse tissues as determined by the in non-human primate models of tuberculosis. Immunol Rev 264:60–73
microbial enumeration technique. II. The conversion of tuberculous in- http://dx.doi.org/10.1111/imr.12258.
fection to the latent state by the administration of pyrazinamide and a 53. Kaushal D, Mehra S. 2012. Faithful experimental models of human
companion drug. J Exp Med 104:763–802 http://dx.doi.org/10.1084 Mycobacterium Tuberculosis infection. Mycobact Dis 2:2 http://dx.doi
/jem.104.5.763. .org/10.4172/2161-1068.1000e108.
38. Scanga CA, Mohan VP, Joseph H, Yu K, Chan J, Flynn JL. 1999. 54. Hunter RL, Actor JK, Hwang SA, Khan A, Urbanowski ME, Kaushal
Reactivation of latent tuberculosis: variations on the Cornell murine D, Jagannath C. 2018. Pathogenesis and animal models of post-primary
model. Infect Immun 67:4531–4538. (bronchogenic) tuberculosis: a review. Pathogens 7:7 http://dx.doi.org
39. Calderon VE, Valbuena G, Goez Y, Judy BM, Huante MB, Sutjita P, /10.3390/pathogens7010019.
Johnston RK, Estes DM, Hunter RL, Actor JK, Cirillo JD, Endsley JJ. 55. Mothé BR, Lindestam Arlehamn CS, Dow C, Dillon MBC, Wiseman
2013. A humanized mouse model of tuberculosis. PLoS One 8:e63331 RW, Bohn P, Karl J, Golden NA, Gilpin T, Foreman TW, Rodgers MA,
http://dx.doi.org/10.1371/journal.pone.0063331. Mehra S, Scriba TJ, Flynn JL, Kaushal D, O’Connor DH, Sette A. 2015.
40. Heuts F, Gavier-Widén D, Carow B, Juarez J, Wigzell H, Rottenberg The TB-specific CD4(+) T cell immune repertoire in both cynomolgus
ME. 2013. CD4+ cell-dependent granuloma formation in humanized mice and rhesus macaques largely overlap with humans. Tuberculosis (Edinb)
infected with mycobacteria. Proc Natl Acad Sci USA 110:6482–6487 95:722–735 http://dx.doi.org/10.1016/j.tube.2015.07.005.
http://dx.doi.org/10.1073/pnas.1219985110. 56. Lai X, Shen Y, Zhou D, Sehgal P, Shen L, Simon M, Qiu L, Letvin NL,
41. McMurray DN. 2001. Disease model: pulmonary tuberculosis. Chen ZW. 2003. Immune biology of macaque lymphocyte populations
Trends Mol Med 7:135–137 http://dx.doi.org/10.1016/S1471-4914(00) during mycobacterial infection. Clin Exp Immunol 133:182–192 http://
01901-8. dx.doi.org/10.1046/j.1365-2249.2003.02209.x.
42. Allison MJ, Zappasodi P, Lurie MB. 1962. Host-parasite relationships 57. Chen CY, Huang D, Wang RC, Shen L, Zeng G, Yao S, Shen Y,
in natively resistant and susceptible rabbits on quantitative inhalation of Halliday L, Fortman J, McAllister M, Estep J, Hunt R, Vasconcelos D,
tubercle bacilli. Their significance for the nature of genetic resistance. Am Du G, Porcelli SA, Larsen MH, Jacobs WR Jr, Haynes BF, Letvin NL,
Rev Respir Dis 85:553–569. Chen ZW. 2009. A critical role for CD8 T cells in a nonhuman primate
43. Bishai WR, Dannenberg AM Jr, Parrish N, Ruiz R, Chen P, Zook BC, model of tuberculosis. PLoS Pathog 5:e1000392 http://dx.doi.org
Johnson W, Boles JW, Pitt ML. 1999. Virulence of Mycobacterium tu- /10.1371/journal.ppat.1000392.
berculosis CDC1551 and H37Rv in rabbits evaluated by Lurie’s pulmo- 58. Kaushal D, Foreman TW, Gautam US, Alvarez X, Adekambi T,
nary tubercle count method. Infect Immun 67:4931–4934. Rangel-Moreno J, Golden NA, Johnson AM, Phillips BL, Ahsan MH,

ASMscience.org/MicrobiolSpectrum 21
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Sia and Rengarajan

Russell-Lodrigue KE, Doyle LA, Roy CJ, Didier PJ, Blanchard JL, culosis while reactivation of latent infection is dependent on severity of
Rengarajan J, Lackner AA, Khader SA, Mehra S. 2015. Mucosal vacci- tissue depletion in cynomolgus macaques. AIDS Res Hum Retroviruses
nation with attenuated Mycobacterium tuberculosis induces strong cen- 28:1693–1702 http://dx.doi.org/10.1089/aid.2012.0028.
tral memory responses and protects against tuberculosis. Nat Commun 70. Lin PL, Myers A, Smith L, Bigbee C, Bigbee M, Fuhrman C, Grieser H,
6:8533 http://dx.doi.org/10.1038/ncomms9533. Chiosea I, Voitenek NN, Capuano SV, Klein E, Flynn JL. 2010. Tumor
59. Verreck FA, Vervenne RA, Kondova I, van Kralingen KW, Remarque necrosis factor neutralization results in disseminated disease in acute and
EJ, Braskamp G, van der Werff NM, Kersbergen A, Ottenhoff TH, Heidt latent Mycobacterium tuberculosis infection with normal granuloma
PJ, Gilbert SC, Gicquel B, Hill AV, Martin C, McShane H, Thomas AW. structure in a cynomolgus macaque model. Arthritis Rheum 62:340–350.
2009. MVA.85A boosting of BCG and an attenuated, phoP deficient M. 71. Lin PL, Maiello P, Gideon HP, Coleman MT, Cadena AM, Rodgers
tuberculosis vaccine both show protective efficacy against tuberculosis in MA, Gregg R, O’Malley M, Tomko J, Fillmore D, Frye LJ, Rutledge T,
rhesus macaques. PLoS One 4:e5264 http://dx.doi.org/10.1371/journal DiFazio RM, Janssen C, Klein E, Andersen PL, Fortune SM, Flynn JL.
.pone.0005264. 2016. PET CT identifies reactivation risk in cynomolgus macaques with
60. Rahman S, Magalhaes I, Rahman J, Ahmed RK, Sizemore DR, Scanga latent M. tuberculosis. PLoS Pathog 12:e1005739 http://dx.doi.org/10.1371
CA, Weichold F, Verreck F, Kondova I, Sadoff J, Thorstensson R, /journal.ppat.1005739.
Spångberg M, Svensson M, Andersson J, Maeurer M, Brighenti S. 2012. 72. Philips JA, Ernst JD. 2012. Tuberculosis pathogenesis and immunity.
Prime-boost vaccination with rBCG/rAd35 enhances CD8+ cytolytic T-cell Annu Rev Pathol 7:353–384 http://dx.doi.org/10.1146/annurev-pathol
responses in lesions from Mycobacterium tuberculosis-infected primates. -011811-132458.
Mol Med 18:647–658 http://dx.doi.org/10.2119/molmed.2011.00222. 73. Jo EK, Yang CS, Choi CH, Harding CV. 2007. Intracellular signalling
61. Lin PL, Coleman T, Carney JP, Lopresti BJ, Tomko J, Fillmore D, cascades regulating innate immune responses to Mycobacteria: branching
Dartois V, Scanga C, Frye LJ, Janssen C, Klein E, Barry CE III, Flynn JL. out from Toll-like receptors. Cell Microbiol 9:1087–1098 http://dx.doi
2013. Radiologic responses in cynomolgus macaques for assessing tu- .org/10.1111/j.1462-5822.2007.00914.x.
berculosis chemotherapy regimens. Antimicrob Agents Chemother 57: 74. Kleinnijenhuis J, Oosting M, Joosten LA, Netea MG, Van Crevel R.
4237–4244 http://dx.doi.org/10.1128/AAC.00277-13. 2011. Innate immune recognition of Mycobacterium tuberculosis. Clin
62. White AG, Maiello P, Coleman MT, Tomko JA, Frye LJ, Scanga CA, Dev Immunol 2011:405310 http://dx.doi.org/10.1155/2011/405310.
Lin PL, Flynn JL. 2017. Analysis of 18FDG PET/CT Imaging as a tool 75. Watson RO, Manzanillo PS, Cox JS. 2012. Extracellular M. tuber-
for studying Mycobacterium tuberculosis infection and treatment in non- culosis DNA targets bacteria for autophagy by activating the host DNA-
human primates. J Vis Exp (127):10.3791/56375. sensing pathway. Cell 150:803–815 http://dx.doi.org/10.1016/j.cell.2012
63. Lin PL, Dartois V, Johnston PJ, Janssen C, Via L, Goodwin MB, Klein .06.040.
E, Barry CE III, Flynn JL. 2012. Metronidazole prevents reactivation of 76. Manzanillo PS, Shiloh MU, Portnoy DA, Cox JS. 2012. Mycobacte-
latent Mycobacterium tuberculosis infection in macaques. Proc Natl Acad rium tuberculosis activates the DNA-dependent cytosolic surveillance
Sci U S A 109:14188–14193 http://dx.doi.org/10.1073/pnas.1121497109. pathway within macrophages. Cell Host Microbe 11:469–480 http://dx
64. Mehra S, Golden NA, Dutta NK, Midkiff CC, Alvarez X, Doyle LA, .doi.org/10.1016/j.chom.2012.03.007.
Asher M, Russell-Lodrigue K, Monjure C, Roy CJ, Blanchard JL, Didier 77. Dey B, Dey RJ, Cheung LS, Pokkali S, Guo H, Lee JH, Bishai WR.
PJ, Veazey RS, Lackner AA, Kaushal D. 2011. Reactivation of latent 2015. A bacterial cyclic dinucleotide activates the cytosolic surveillance
tuberculosis in rhesus macaques by coinfection with simian immuno- pathway and mediates innate resistance to tuberculosis. Nat Med 21:401–
deficiency virus. J Med Primatol 40:233–243 http://dx.doi.org/10.1111 406 http://dx.doi.org/10.1038/nm.3813.
/j.1600-0684.2011.00485.x. 78. Collins AC, Cai H, Li T, Franco LH, Li XD, Nair VR, Scharn CR,
65. Foreman TW, Mehra S, LoBato DN, Malek A, Alvarez X, Golden Stamm CE, Levine B, Chen ZJ, Shiloh MU. 2015. Cyclic GMP-AMP
NA, Bucşan AN, Didier PJ, Doyle-Meyers LA, Russell-Lodrigue KE, synthase is an innate immune DNA sensor for Mycobacterium tubercu-
Roy CJ, Blanchard J, Kuroda MJ, Lackner AA, Chan J, Khader SA, Jacobs losis. Cell Host Microbe 17:820–828 http://dx.doi.org/10.1016/j.chom
WR Jr, Kaushal D. 2016. CD4+ T-cell-independent mechanisms suppress .2015.05.005.
reactivation of latent tuberculosis in a macaque model of HIV coinfection. 79. Watson RO, Bell SL, MacDuff DA, Kimmey JM, Diner EJ, Olivas J,
Proc Natl Acad Sci U S A 113:E5636–E5644 http://dx.doi.org/10.1073 Vance RE, Stallings CL, Virgin HW, Cox JS. 2015. The cytosolic sensor
/pnas.1611987113. cGAS detects Mycobacterium tuberculosis DNA to induce type I inter-
66. Gautam US, Foreman TW, Bucsan AN, Veatch AV, Alvarez X, ferons and activate autophagy. Cell Host Microbe 17:811–819 http://dx
Adekambi T, Golden NA, Gentry KM, Doyle-Meyers LA, Russell- .doi.org/10.1016/j.chom.2015.05.004.
Lodrigue KE, Didier PJ, Blanchard JL, Kousoulas KG, Lackner AA, 80. Court N, Vasseur V, Vacher R, Frémond C, Shebzukhov Y, Yeremeev
Kalman D, Rengarajan J, Khader SA, Kaushal D, Mehra S. 2018. VV, Maillet I, Nedospasov SA, Gordon S, Fallon PG, Suzuki H, Ryffel B,
In vivo inhibition of tryptophan catabolism reorganizes the tuberculoma Quesniaux VF. 2010. Partial redundancy of the pattern recognition
and augments immune-mediated control of Mycobacterium tuberculosis. receptors, scavenger receptors, and C-type lectins for the long-term control
Proc Natl Acad Sci U S A 115:E62–E71 http://dx.doi.org/10.1073/pnas of Mycobacterium tuberculosis infection. J Immunol 184:7057–7070
.1711373114. http://dx.doi.org/10.4049/jimmunol.1000164.
67. Diedrich CR, Mattila JT, Klein E, Janssen C, Phuah J, Sturgeon TJ, 81. Velez DR, Wejse C, Stryjewski ME, Abbate E, Hulme WF, Myers
Montelaro RC, Lin PL, Flynn JL. 2010. Reactivation of latent tuber- JL, Estevan R, Patillo SG, Olesen R, Tacconelli A, Sirugo G, Gilbert JR,
culosis in cynomolgus macaques infected with SIV is associated with early Hamilton CD, Scott WK. 2010. Variants in toll-like receptors 2 and 9
peripheral T cell depletion and not virus load. PLoS One 5:e9611 http:// influence susceptibility to pulmonary tuberculosis in Caucasians, African-
dx.doi.org/10.1371/journal.pone.0009611. Americans, and West Africans. Hum Genet 127:65–73 http://dx.doi.org
68. Mattila JT, Diedrich CR, Lin PL, Phuah J, Flynn JL. 2011. Simian /10.1007/s00439-009-0741-7.
immunodeficiency virus-induced changes in T cell cytokine responses in 82. Ma X, Liu Y, Gowen BB, Graviss EA, Clark AG, Musser JM. 2007.
cynomolgus macaques with latent Mycobacterium tuberculosis infection Full-exon resequencing reveals toll-like receptor variants contribute to
are associated with timing of reactivation. J Immunol 186:3527–3537 human susceptibility to tuberculosis disease. PLoS One 2:e1318 http://dx
http://dx.doi.org/10.4049/jimmunol.1003773. .doi.org/10.1371/journal.pone.0001318.
69. Lin PL, Rutledge T, Green AM, Bigbee M, Fuhrman C, Klein E, Flynn 83. Davila S, Hibberd ML, Hari Dass R, Wong HE, Sahiratmadja E,
JL. 2012. CD4 T cell depletion exacerbates acute Mycobacterium tuber- Bonnard C, Alisjahbana B, Szeszko JS, Balabanova Y, Drobniewski F,

22 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Immunology of M. tuberculosis Infections

van Crevel R, van de Vosse E, Nejentsev S, Ottenhoff TH, Seielstad M. 96. Flores-Villanueva PO, Ruiz-Morales JA, Song CH, Flores LM, Jo EK,
2008. Genetic association and expression studies indicate a role of toll-like Montaño M, Barnes PF, Selman M, Granados J. 2005. A functional
receptor 8 in pulmonary tuberculosis. PLoS Genet 4:e1000218 http://dx promoter polymorphism in monocyte chemoattractant protein-1 is asso-
.doi.org/10.1371/journal.pgen.1000218. ciated with increased susceptibility to pulmonary tuberculosis. J Exp Med
84. Khor CC, Chapman SJ, Vannberg FO, Dunne A, Murphy C, Ling EY, 202:1649–1658 http://dx.doi.org/10.1084/jem.20050126.
Frodsham AJ, Walley AJ, Kyrieleis O, Khan A, Aucan C, Segal S, Moore 97. Juffermans NP, Florquin S, Camoglio L, Verbon A, Kolk AH, Speelman
CE, Knox K, Campbell SJ, Lienhardt C, Scott A, Aaby P, Sow OY, P, van Deventer SJ, van Der Poll T. 2000. Interleukin-1 signaling is essential
Grignani RT, Sillah J, Sirugo G, Peshu N, Williams TN, Maitland K, for host defense during murine pulmonary tuberculosis. J Infect Dis 182:
Davies RJ, Kwiatkowski DP, Day NP, Yala D, Crook DW, Marsh K, 902–908 http://dx.doi.org/10.1086/315771.
Berkley JA, O’Neill LA, Hill AV. 2007. A Mal functional variant is as- 98. Sugawara I, Yamada H, Hua S, Mizuno S. 2001. Role of interleukin
sociated with protection against invasive pneumococcal disease, bacter- (IL)-1 type 1 receptor in mycobacterial infection. Microbiol Immunol
emia, malaria and tuberculosis. Nat Genet 39:523–528 http://dx.doi.org 45:743–750 http://dx.doi.org/10.1111/j.1348-0421.2001.tb01310.x.
/10.1038/ng1976. 99. Yamada H, Mizumo S, Horai R, Iwakura Y, Sugawara I. 2000.
85. Fremond CM, Yeremeev V, Nicolle DM, Jacobs M, Quesniaux VF, Protective role of interleukin-1 in mycobacterial infection in IL-1 alpha/
Ryffel B. 2004. Fatal Mycobacterium tuberculosis infection despite beta double-knockout mice. Lab Invest 80:759–767 http://dx.doi.org
adaptive immune response in the absence of MyD88. J Clin Invest 114: /10.1038/labinvest.3780079.
1790–1799 http://dx.doi.org/10.1172/JCI200421027.
100. Mayer-Barber KD, Andrade BB, Oland SD, Amaral EP, Barber DL,
86. Scanga CA, Bafica A, Feng CG, Cheever AW, Hieny S, Sher A. 2004. Gonzales J, Derrick SC, Shi R, Kumar NP, Wei W, Yuan X, Zhang G,
MyD88-deficient mice display a profound loss in resistance to Myco- Cai Y, Babu S, Catalfamo M, Salazar AM, Via LE, Barry CE III, Sher A.
bacterium tuberculosis associated with partially impaired Th1 cytokine 2014. Host-directed therapy of tuberculosis based on interleukin-1 and
and nitric oxide synthase 2 expression. Infect Immun 72:2400–2404 type I interferon crosstalk. Nature 511:99–103 http://dx.doi.org/10.1038
http://dx.doi.org/10.1128/IAI.72.4.2400-2404.2004. /nature13489.
87. Mayer-Barber KD, Barber DL, Shenderov K, White SD, Wilson MS, 101. Di Paolo NC, Shafiani S, Day T, Papayannopoulou T, Russell DW,
Cheever A, Kugler D, Hieny S, Caspar P, Núñez G, Schlueter D, Flavell Iwakura Y, Sherman D, Urdahl K, Shayakhmetov DM. 2015. Inter-
RA, Sutterwala FS, Sher A. 2010. Caspase-1 independent IL-1beta pro- dependence between interleukin-1 and tumor necrosis factor regulates
duction is critical for host resistance to Mycobacterium tuberculosis TNF-dependent control of Mycobacterium tuberculosis infection. Immu-
and does not require TLR signaling in vivo. J Immunol 184:3326–3330 nity 43:1125–1136 http://dx.doi.org/10.1016/j.immuni.2015.11.016.
http://dx.doi.org/10.4049/jimmunol.0904189.
102. Reed MB, Domenech P, Manca C, Su H, Barczak AK, Kreiswirth
88. Fremond CM, Togbe D, Doz E, Rose S, Vasseur V, Maillet I, Jacobs BN, Kaplan G, Barry CE III. 2004. A glycolipid of hypervirulent tuber-
M, Ryffel B, Quesniaux VF. 2007. IL-1 receptor-mediated signal is an culosis strains that inhibits the innate immune response. Nature 431:84–
essential component of MyD88-dependent innate response to Mycobac- 87 http://dx.doi.org/10.1038/nature02837.
terium tuberculosis infection. J Immunol 179:1178–1189 http://dx.doi 103. Sinsimer D, Huet G, Manca C, Tsenova L, Koo MS, Kurepina N,
.org/10.4049/jimmunol.179.2.1178. Kana B, Mathema B, Marras SA, Kreiswirth BN, Guilhot C, Kaplan G.
89. Shi S, Nathan C, Schnappinger D, Drenkow J, Fuortes M, Block E, 2008. The phenolic glycolipid of Mycobacterium tuberculosis differ-
Ding A, Gingeras TR, Schoolnik G, Akira S, Takeda K, Ehrt S. 2003. entially modulates the early host cytokine response but does not in itself
MyD88 primes macrophages for full-scale activation by interferon- confer hypervirulence. Infect Immun 76:3027–3036 http://dx.doi.org
gamma yet mediates few responses to Mycobacterium tuberculosis. J Exp /10.1128/IAI.01663-07.
Med 198:987–997 http://dx.doi.org/10.1084/jem.20030603. 104. Blanc L, Gilleron M, Prandi J, Song OR, Jang MS, Gicquel B,
90. Bafica A, Scanga CA, Feng CG, Leifer C, Cheever A, Sher A. 2005. Drocourt D, Neyrolles O, Brodin P, Tiraby G, Vercellone A, Nigou J. 2017.
TLR9 regulates Th1 responses and cooperates with TLR2 in mediating Mycobacterium tuberculosis inhibits human innate immune responses via
optimal resistance to Mycobacterium tuberculosis. J Exp Med 202:1715– the production of TLR2 antagonist glycolipids. Proc Natl Acad Sci U S A
1724 http://dx.doi.org/10.1084/jem.20051782. 114:11205–11210 http://dx.doi.org/10.1073/pnas.1707840114.
91. Dorhoi A, Desel C, Yeremeev V, Pradl L, Brinkmann V, Mollenkopf 105. Rengarajan J, Murphy E, Park A, Krone CL, Hett EC, Bloom BR,
HJ, Hanke K, Gross O, Ruland J, Kaufmann SH. 2010. The adaptor Glimcher LH, Rubin EJ. 2008. Mycobacterium tuberculosis Rv2224c
molecule CARD9 is essential for tuberculosis control. J Exp Med 207: modulates innate immune responses. Proc Natl Acad Sci U S A 105:264–
777–792 http://dx.doi.org/10.1084/jem.20090067. 269 http://dx.doi.org/10.1073/pnas.0710601105.
92. McElvania Tekippe E, Allen IC, Hulseberg PD, Sullivan JT, McCann 106. Madan-Lala R, Peixoto KV, Re F, Rengarajan J. 2011. Mycobacte-
JR, Sandor M, Braunstein M, Ting JP. 2010. Granuloma formation and rium tuberculosis Hip1 dampens macrophage proinflammatory responses
host defense in chronic Mycobacterium tuberculosis infection requires by limiting Toll-like receptor 2 activation. Infect Immun 79:4828–4838
PYCARD/ASC but not NLRP3 or caspase-1. PLoS One 5:e12320 http:// http://dx.doi.org/10.1128/IAI.05574-11.
dx.doi.org/10.1371/journal.pone.0012320. 107. Madan-Lala R, Sia JK, King R, Adekambi T, Monin L, Khader SA,
93. Eklund D, Welin A, Andersson H, Verma D, Söderkvist P, Stendahl Pulendran B, Rengarajan J. 2014. Mycobacterium tuberculosis impairs
O, Särndahl E, Lerm M. 2014. Human gene variants linked to enhanced dendritic cell functions through the serine hydrolase Hip1. J Immunol
NLRP3 activity limit intramacrophage growth of Mycobacterium tuber- 192:4263–4272 http://dx.doi.org/10.4049/jimmunol.1303185.
culosis. J Infect Dis 209:749–753 http://dx.doi.org/10.1093/infdis/jit572. 108. Naffin-Olivos JL, Georgieva M, Goldfarb N, Madan-Lala R, Dong
94. Bellamy R, Ruwende C, Corrah T, McAdam KP, Whittle HC, Hill L, Bizzell E, Valinetz E, Brandt GS, Yu S, Shabashvili DE, Ringe D, Dunn
AV. 1998. Assessment of the interleukin 1 gene cluster and other candi- BM, Petsko GA, Rengarajan J. 2014. Mycobacterium tuberculosis Hip1
date gene polymorphisms in host susceptibility to tuberculosis. Tuber modulates macrophage responses through proteolysis of GroEL2. PLoS
Lung Dis 79:83–89 http://dx.doi.org/10.1054/tuld.1998.0009. Pathog 10:e1004132 http://dx.doi.org/10.1371/journal.ppat.1004132.
95. Wilkinson RJ, Patel P, Llewelyn M, Hirsch CS, Pasvol G, Snounou 109. Georgieva M, Sia JK, Bizzell E, Madan-Lala R, Rengarajan J. 2018.
G, Davidson RN, Toossi Z. 1999. Influence of polymorphism in the Mycobacterium tuberculosis GroEL2 modulates dendritic cell responses.
genes for the interleukin (IL)-1 receptor antagonist and IL-1beta on tu- Infect Immun 86:e00387-17.
berculosis. J Exp Med 189:1863–1874 http://dx.doi.org/10.1084/jem.189 110. Master SS, Rampini SK, Davis AS, Keller C, Ehlers S, Springer B,
.12.1863. Timmins GS, Sander P, Deretic V. 2008. Mycobacterium tuberculosis

ASMscience.org/MicrobiolSpectrum 23
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Sia and Rengarajan

prevents inflammasome activation. Cell Host Microbe 3:224–232 http:// 125. van der Wel N, Hava D, Houben D, Fluitsma D, van Zon M, Pierson
dx.doi.org/10.1016/j.chom.2008.03.003. J, Brenner M, Peters PJ. 2007. M. tuberculosis and M. leprae translocate
111. Ehrt S, Schnappinger D. 2009. Mycobacterial survival strategies in from the phagolysosome to the cytosol in myeloid cells. Cell 129:1287–
the phagosome: defence against host stresses. Cell Microbiol 11:1170– 1298 http://dx.doi.org/10.1016/j.cell.2007.05.059.
1178 http://dx.doi.org/10.1111/j.1462-5822.2009.01335.x. 126. Houben D, Demangel C, van Ingen J, Perez J, Baldeón L, Abdallah
112. Schnappinger D, Ehrt S, Voskuil MI, Liu Y, Mangan JA, Monahan AM, Caleechurn L, Bottai D, van Zon M, de Punder K, van der Laan T,
IM, Dolganov G, Efron B, Butcher PD, Nathan C, Schoolnik GK. 2003. Kant A, Bossers-de Vries R, Willemsen P, Bitter W, van Soolingen D,
Transcriptional adaptation of Mycobacterium tuberculosis within macro- Brosch R, van der Wel N, Peters PJ. 2012. ESX-1-mediated translocation
phages: insights into the phagosomal environment. J Exp Med 198:693– to the cytosol controls virulence of mycobacteria. Cell Microbiol 14:1287–
704 http://dx.doi.org/10.1084/jem.20030846. 1298 http://dx.doi.org/10.1111/j.1462-5822.2012.01799.x.
113. Rengarajan J, Bloom BR, Rubin EJ. 2005. Genome-wide require- 127. De Leon J, Jiang G, Ma Y, Rubin E, Fortune S, Sun J. 2012. My-
ments for Mycobacterium tuberculosis adaptation and survival in macro- cobacterium tuberculosis ESAT-6 exhibits a unique membrane-interacting
phages. Proc Natl Acad Sci U S A 102:8327–8332 http://dx.doi.org/10.1073 activity that is not found in its ortholog from non-pathogenic Myco-
/pnas.0503272102. bacterium smegmatis. J Biol Chem 287:44184–44191 http://dx.doi.org
/10.1074/jbc.M112.420869.
114. Fratti RA, Chua J, Vergne I, Deretic V. 2003. Mycobacterium tu-
berculosis glycosylated phosphatidylinositol causes phagosome matura- 128. Conrad WH, Osman MM, Shanahan JK, Chu F, Takaki KK,
tion arrest. Proc Natl Acad Sci USA 100:5437–5442 http://dx.doi.org Cameron J, Hopkinson-Woolley D, Brosch R, Ramakrishnan L. 2017.
/10.1073/pnas.0737613100. Mycobacterial ESX-1 secretion system mediates host cell lysis through
115. Saleh MT, Belisle JT. 2000. Secretion of an acid phosphatase (SapM) bacterium contact-dependent gross membrane disruptions. Proc Natl Acad
Sci USA 114:1371–1376 http://dx.doi.org/10.1073/pnas.1620133114.
by Mycobacterium tuberculosis that is similar to eukaryotic acid phos-
phatases. J Bacteriol 182:6850–6853 http://dx.doi.org/10.1128/JB.182.23 129. Pandey AK, Yang Y, Jiang Z, Fortune SM, Coulombe F, Behr MA,
.6850-6853.2000. Fitzgerald KA, Sassetti CM, Kelliher MA. 2009. NOD2, RIP2 and IRF5
play a critical role in the type I interferon response to Mycobacterium
116. Vergne I, Chua J, Lee HH, Lucas M, Belisle J, Deretic V. 2005.
Mechanism of phagolysosome biogenesis block by viable Mycobacterium tuberculosis. PLoS Pathog 5:e1000500 http://dx.doi.org/10.1371/journal
.ppat.1000500.
tuberculosis. Proc Natl Acad Sci USA 102:4033–4038 http://dx.doi.org
/10.1073/pnas.0409716102. 130. Coulombe F, Divangahi M, Veyrier F, de Léséleuc L, Gleason JL,
117. Bach H, Papavinasasundaram KG, Wong D, Hmama Z, Av-Gay Y. Yang Y, Kelliher MA, Pandey AK, Sassetti CM, Reed MB, Behr MA. 2009.
Increased NOD2-mediated recognition of N-glycolyl muramyl dipeptide.
2008. Mycobacterium tuberculosis virulence is mediated by PtpA dephos-
J Exp Med 206:1709–1716 http://dx.doi.org/10.1084/jem.20081779.
phorylation of human vacuolar protein sorting 33B. Cell Host Microbe
3:316–322 http://dx.doi.org/10.1016/j.chom.2008.03.008. 131. Mehra A, Zahra A, Thompson V, Sirisaengtaksin N, Wells A,
118. Nguyen L, Pieters J. 2005. The Trojan horse: survival tactics of Porto M, Köster S, Penberthy K, Kubota Y, Dricot A, Rogan D, Vidal M,
pathogenic mycobacteria in macrophages. Trends Cell Biol 15:269–276 Hill DE, Bean AJ, Philips JA. 2013. Mycobacterium tuberculosis type VII
http://dx.doi.org/10.1016/j.tcb.2005.03.009. secreted effector EsxH targets host ESCRT to impair trafficking. PLoS
Pathog 9:e1003734 http://dx.doi.org/10.1371/journal.ppat.1003734.
119. Majlessi L, Combaluzier B, Albrecht I, Garcia JE, Nouze C, Pieters J,
Leclerc C. 2007. Inhibition of phagosome maturation by mycobacteria 132. Tinaztepe E, Wei JR, Raynowska J, Portal-Celhay C, Thompson V,
does not interfere with presentation of mycobacterial antigens by MHC Philips JA. 2016. Role of metal-dependent regulation of ESX-3 secretion
molecules. J Immunol 179:1825–1833 http://dx.doi.org/10.4049/jimmunol in intracellular survival of Mycobacterium tuberculosis. Infect Immun
.179.3.1825. 84:2255–2263 http://dx.doi.org/10.1128/IAI.00197-16.
120. Walburger A, Koul A, Ferrari G, Nguyen L, Prescianotto-Baschong 133. Tufariello JM, Chapman JR, Kerantzas CA, Wong KW, Vilchèze
C, Huygen K, Klebl B, Thompson C, Bacher G, Pieters J. 2004. Pro- C, Jones CM, Cole LE, Tinaztepe E, Thompson V, Fenyö D, Niederweis
tein kinase G from pathogenic mycobacteria promotes survival within M, Ueberheide B, Philips JA, Jacobs WR Jr. 2016. Separable roles for
macrophages. Science 304:1800–1804 http://dx.doi.org/10.1126/science Mycobacterium tuberculosis ESX-3 effectors in iron acquisition and
.1099384. virulence. Proc Natl Acad Sci U S A 113:E348–E357 http://dx.doi.org
/10.1073/pnas.1523321113.
121. Cowley S, Ko M, Pick N, Chow R, Downing KJ, Gordhan BG,
Betts JC, Mizrahi V, Smith DA, Stokes RW, Av-Gay Y. 2004. The My- 134. Hou JM, D’Lima NG, Rigel NW, Gibbons HS, McCann JR,
cobacterium tuberculosis protein serine/threonine kinase PknG is linked Braunstein M, Teschke CM. 2008. ATPase activity of Mycobacterium
to cellular glutamate/glutamine levels and is important for growth in vivo. tuberculosis SecA1 and SecA2 proteins and its importance for SecA2
Mol Microbiol 52:1691–1702 http://dx.doi.org/10.1111/j.1365-2958.2004 function in macrophages. J Bacteriol 190:4880–4887 http://dx.doi.org
.04085.x. /10.1128/JB.00412-08.
122. Quigley J, Hughitt VK, Velikovsky CA, Mariuzza RA, El-Sayed 135. Braunstein M, Espinosa BJ, Chan J, Belisle JT, Jacobs WR Jr. 2003.
NM, Briken V. 2017. The cell wall lipid PDIM contributes to phagosomal SecA2 functions in the secretion of superoxide dismutase A and in the
escape and host cell exit of Mycobacterium tuberculosis. MBio 8:e00148- virulence of Mycobacterium tuberculosis. Mol Microbiol 48:453–464
17 http://dx.doi.org/10.1128/mBio.00148-17. http://dx.doi.org/10.1046/j.1365-2958.2003.03438.x.
123. Schnettger L, Rodgers A, Repnik U, Lai RP, Pei G, Verdoes M, 136. Kurtz S, McKinnon KP, Runge MS, Ting JP, Braunstein M. 2006.
Wilkinson RJ, Young DB, Gutierrez MG. 2017. A Rab20-dependent The SecA2 secretion factor of Mycobacterium tuberculosis promotes
membrane trafficking pathway controls M. tuberculosis replication by growth in macrophages and inhibits the host immune response. Infect
regulating phagosome spaciousness and integrity. Cell Host Microbe 21: Immun 74:6855–6864 http://dx.doi.org/10.1128/IAI.01022-06.
619–628.e5 http://dx.doi.org/10.1016/j.chom.2017.04.004. 137. Sullivan JT, Young EF, McCann JR, Braunstein M. 2012. The
124. de Jonge MI, Pehau-Arnaudet G, Fretz MM, Romain F, Bottai D, Mycobacterium tuberculosis SecA2 system subverts phagosome matura-
Brodin P, Honoré N, Marchal G, Jiskoot W, England P, Cole ST, Brosch tion to promote growth in macrophages. Infect Immun 80:996–1006
R. 2007. ESAT-6 from Mycobacterium tuberculosis dissociates from its http://dx.doi.org/10.1128/IAI.05987-11.
putative chaperone CFP-10 under acidic conditions and exhibits membrane- 138. Gatfield J, Pieters J. 2000. Essential role for cholesterol in entry of
lysing activity. J Bacteriol 189:6028–6034 http://dx.doi.org/10.1128/JB mycobacteria into macrophages. Science 288:1647–1650 http://dx.doi
.00469-07. .org/10.1126/science.288.5471.1647.

24 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Immunology of M. tuberculosis Infections

139. Jayachandran R, Sundaramurthy V, Combaluzier B, Mueller P, Korf 155. Li Z, Kelley C, Collins F, Rouse D, Morris S. 1998. Expression of
H, Huygen K, Miyazaki T, Albrecht I, Massner J, Pieters J. 2007. Survival katG in Mycobacterium tuberculosis is associated with its growth and
of mycobacteria in macrophages is mediated by coronin 1-dependent ac- persistence in mice and guinea pigs. J Infect Dis 177:1030–1035 http://dx
tivation of calcineurin. Cell 130:37–50 http://dx.doi.org/10.1016/j.cell .doi.org/10.1086/515254.
.2007.04.043. 156. Darwin KH, Ehrt S, Gutierrez-Ramos JC, Weich N, Nathan CF.
140. Brightbill HD, Libraty DH, Krutzik SR, Yang RB, Belisle JT, 2003. The proteasome of Mycobacterium tuberculosis is required for re-
Bleharski JR, Maitland M, Norgard MV, Plevy SE, Smale ST, Brennan PJ, sistance to nitric oxide. Science 302:1963–1966 http://dx.doi.org/10.1126
Bloom BR, Godowski PJ, Modlin RL. 1999. Host defense mechanisms /science.1091176.
triggered by microbial lipoproteins through toll-like receptors. Science 157. Underhill DM, Ozinsky A, Smith KD, Aderem A. 1999. Toll-like
285:732–736 http://dx.doi.org/10.1126/science.285.5428.732. receptor-2 mediates mycobacteria-induced proinflammatory signaling in
141. Long R, Light B, Talbot JA. 1999. Mycobacteriocidal action of macrophages. Proc Natl Acad Sci U S A 96:14459–14463 http://dx.doi
exogenous nitric oxide. Antimicrob Agents Chemother 43:403–405. .org/10.1073/pnas.96.25.14459.
142. Yu K, Mitchell C, Xing Y, Magliozzo RS, Bloom BR, Chan J. 1999. 158. Liu PT, Stenger S, Li H, Wenzel L, Tan BH, Krutzik SR, Ochoa MT,
Toxicity of nitrogen oxides and related oxidants on mycobacteria: Schauber J, Wu K, Meinken C, Kamen DL, Wagner M, Bals R, Steinmeyer
M. tuberculosis is resistant to peroxynitrite anion. Tuber Lung Dis 79: A, Zügel U, Gallo RL, Eisenberg D, Hewison M, Hollis BW, Adams JS,
191–198 http://dx.doi.org/10.1054/tuld.1998.0203. Bloom BR, Modlin RL. 2006. Toll-like receptor triggering of a vitamin
143. O’Brien L, Carmichael J, Lowrie DB, Andrew PW. 1994. Strains of D-mediated human antimicrobial response. Science 311:1770–1773
Mycobacterium tuberculosis differ in susceptibility to reactive nitrogen http://dx.doi.org/10.1126/science.1123933.
intermediates in vitro. Infect Immun 62:5187–5190. 159. Liu PT, Stenger S, Tang DH, Modlin RL. 2007. Cutting edge: vita-
144. Flesch IE, Kaufmann SH. 1991. Mechanisms involved in mycobacte- min D-mediated human antimicrobial activity against Mycobacterium
rial growth inhibition by gamma interferon-activated bone marrow macro- tuberculosis is dependent on the induction of cathelicidin. J Immunol 179:
phages: role of reactive nitrogen intermediates. Infect Immun 59:3213–3218. 2060–2063 http://dx.doi.org/10.4049/jimmunol.179.4.2060.
145. Chan J, Xing Y, Magliozzo RS, Bloom BR. 1992. Killing of virulent 160. Martineau AR, Wilkinson KA, Newton SM, Floto RA, Norman AW,
Mycobacterium tuberculosis by reactive nitrogen intermediates produced Skolimowska K, Davidson RN, Sørensen OE, Kampmann B, Griffiths
by activated murine macrophages. J Exp Med 175:1111–1122 http://dx CJ, Wilkinson RJ. 2007. IFN-gamma- and TNF-independent vitamin
.doi.org/10.1084/jem.175.4.1111. D-inducible human suppression of mycobacteria: the role of cathelicidin
146. MacMicking JD, North RJ, LaCourse R, Mudgett JS, Shah SK, LL-37. J Immunol 178:7190–7198 http://dx.doi.org/10.4049/jimmunol
.178.11.7190.
Nathan CF. 1997. Identification of nitric oxide synthase as a protective
locus against tuberculosis. Proc Natl Acad Sci U S A 94:5243–5248 http:// 161. Yuk JM, Shin DM, Lee HM, Yang CS, Jin HS, Kim KK, Lee ZW,
dx.doi.org/10.1073/pnas.94.10.5243. Lee SH, Kim JM, Jo EK. 2009. Vitamin D3 induces autophagy in human
monocytes/macrophages via cathelicidin. Cell Host Microbe 6:231–243
147. Chan J, Tanaka K, Carroll D, Flynn J, Bloom BR. 1995. Effects of
http://dx.doi.org/10.1016/j.chom.2009.08.004.
nitric oxide synthase inhibitors on murine infection with Mycobacterium
tuberculosis. Infect Immun 63:736–740. 162. Fabri M, Stenger S, Shin DM, Yuk JM, Liu PT, Realegeno S,
Lee HM, Krutzik SR, Schenk M, Sieling PA, Teles R, Montoya D, Iyer SS,
148. Flynn JL, Scanga CA, Tanaka KE, Chan J. 1998. Effects of amino-
Bruns H, Lewinsohn DM, Hollis BW, Hewison M, Adams JS, Steinmeyer
guanidine on latent murine tuberculosis. J Immunol 160:1796–1803.
A, Zügel U, Cheng G, Jo EK, Bloom BR, Modlin RL. 2011. Vitamin D is
149. Mishra BB, Rathinam VA, Martens GW, Martinot AJ, Kornfeld H, required for IFN-gamma-mediated antimicrobial activity of human mac-
Fitzgerald KA, Sassetti CM. 2013. Nitric oxide controls the immunopa- rophages. Sci Transl Med 3:104ra102 http://dx.doi.org/10.1126/scitranslmed
thology of tuberculosis by inhibiting NLRP3 inflammasome-dependent .3003045.
processing of IL-1β. Nat Immunol 14:52–60 http://dx.doi.org/10.1038/ni
163. Gutierrez MG, Master SS, Singh SB, Taylor GA, Colombo MI,
.2474.
Deretic V. 2004. Autophagy is a defense mechanism inhibiting BCG
150. Scanga CA, Mohan VP, Tanaka K, Alland D, Flynn JL, Chan J. and Mycobacterium tuberculosis survival in infected macrophages. Cell
2001. The inducible nitric oxide synthase locus confers protection against 119:753–766 http://dx.doi.org/10.1016/j.cell.2004.11.038.
aerogenic challenge of both clinical and laboratory strains of Mycobac-
terium tuberculosis in mice. Infect Immun 69:7711–7717 http://dx.doi 164. MacMicking JD, Taylor GA, McKinney JD. 2003. Immune control
of tuberculosis by IFN-gamma-inducible LRG-47. Science 302:654–659
.org/10.1128/IAI.69.12.7711-7717.2001.
http://dx.doi.org/10.1126/science.1088063.
151. Thoma-Uszynski S, Stenger S, Takeuchi O, Ochoa MT, Engele M,
Sieling PA, Barnes PF, Rollinghoff M, Bolcskei PL, Wagner M, Akira S, 165. Singh SB, Davis AS, Taylor GA, Deretic V. 2006. Human IRGM
induces autophagy to eliminate intracellular mycobacteria. Science 313:
Norgard MV, Belisle JT, Godowski PJ, Bloom BR, Modlin RL. 2001.
Induction of direct antimicrobial activity through mammalian Toll-like 1438–1441 http://dx.doi.org/10.1126/science.1129577.
receptors. Science 291:1544–1547 http://dx.doi.org/10.1126/science.291 166. Kumar D, Nath L, Kamal MA, Varshney A, Jain A, Singh S, Rao KV.
.5508.1544. 2010. Genome-wide analysis of the host intracellular network that
152. Jung JY, Madan-Lala R, Georgieva M, Rengarajan J, Sohaskey CD, regulates survival of Mycobacterium tuberculosis. Cell 140:731–743
Bange FC, Robinson CM. 2013. The intracellular environment of human http://dx.doi.org/10.1016/j.cell.2010.02.012.
macrophages that produce nitric oxide promotes growth of mycobacteria. 167. Manzanillo PS, Ayres JS, Watson RO, Collins AC, Souza G, Rae
Infect Immun 81:3198–3209 http://dx.doi.org/10.1128/IAI.00611-13. CS, Schneider DS, Nakamura K, Shiloh MU, Cox JS. 2013. The ubiquitin
153. Aston C, Rom WN, Talbot AT, Reibman J. 1998. Early inhibition ligase parkin mediates resistance to intracellular pathogens. Nature 501:
of mycobacterial growth by human alveolar macrophages is not due to 512–516 http://dx.doi.org/10.1038/nature12566.
nitric oxide. Am J Respir Crit Care Med 157:1943–1950 http://dx.doi.org 168. Sakowski ET, Koster S, Portal Celhay C, Park HS, Shrestha E,
/10.1164/ajrccm.157.6.9705028. Hetzenecker SE, Maurer K, Cadwell K, Philips JA. 2015. Ubiquilin 1 pro-
154. Nicholson S, Bonecini-Almeida MG, Lapa e Silva JR, Nathan C, Xie motes IFN-γ-induced xenophagy of Mycobacterium tuberculosis. PLoS
QW, Mumford R, Weidner JR, Calaycay J, Geng J, Boechat N, Linhares Pathog 11:e1005076 http://dx.doi.org/10.1371/journal.ppat.1005076.
C, Rom W, Ho JL. 1996. Inducible nitric oxide synthase in pulmonary 169. Ní Cheallaigh C, Sheedy FJ, Harris J, Muñoz-Wolf N, Lee J, West
alveolar macrophages from patients with tuberculosis. J Exp Med 183: K, McDermott EP, Smyth A, Gleeson LE, Coleman M, Martinez N,
2293–2302 http://dx.doi.org/10.1084/jem.183.5.2293. Hearnden CH, Tynan GA, Carroll EC, Jones SA, Corr SC, Bernard NJ,

ASMscience.org/MicrobiolSpectrum 25
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Sia and Rengarajan

Hughes MM, Corcoran SE, O’Sullivan M, Fallon CM, Kornfeld H, 182. Eruslanov EB, Lyadova IV, Kondratieva TK, Majorov KB,
Golenbock D, Gordon SV, O’Neill LA, Lavelle EC, Keane J. 2016. A Scheglov IV, Orlova MO, Apt AS. 2005. Neutrophil responses to Myco-
common variant in the adaptor Mal regulates interferon gamma signaling. bacterium tuberculosis infection in genetically susceptible and resistant
Immunity 44:368–379 http://dx.doi.org/10.1016/j.immuni.2016.01.019. mice. Infect Immun 73:1744–1753 http://dx.doi.org/10.1128/IAI.73.3
170. Ouimet M, Koster S, Sakowski E, Ramkhelawon B, van Solingen .1744-1753.2005.
C, Oldebeken S, Karunakaran D, Portal-Celhay C, Sheedy FJ, Ray TD, 183. Keller C, Hoffmann R, Lang R, Brandau S, Hermann C, Ehlers S.
Cecchini K, Zamore PD, Rayner KJ, Marcel YL, Philips JA, Moore KJ. 2006. Genetically determined susceptibility to tuberculosis in mice caus-
2016. Mycobacterium tuberculosis induces the miR-33 locus to repro- ally involves accelerated and enhanced recruitment of granulocytes. Infect
gram autophagy and host lipid metabolism. Nat Immunol 17:677–686 Immun 74:4295–4309 http://dx.doi.org/10.1128/IAI.00057-06.
http://dx.doi.org/10.1038/ni.3434. 184. Yeremeev V, Linge I, Kondratieva T, Apt A. 2015. Neutrophils ex-
171. Saini NK, Baena A, Ng TW, Venkataswamy MM, Kennedy SC, acerbate tuberculosis infection in genetically susceptible mice. Tubercu-
Kunnath-Velayudhan S, Carreño LJ, Xu J, Chan J, Larsen MH, Jacobs losis (Edinb) 95:447–451 http://dx.doi.org/10.1016/j.tube.2015.03.007.
WR Jr, Porcelli SA. 2016. Suppression of autophagy and antigen pre-
185. Zhang X, Majlessi L, Deriaud E, Leclerc C, Lo-Man R. 2009.
sentation by Mycobacterium tuberculosis PE_PGRS47. Nat Microbiol Coactivation of Syk kinase and MyD88 adaptor protein pathways by
1:16133 http://dx.doi.org/10.1038/nmicrobiol.2016.133.
bacteria promotes regulatory properties of neutrophils. Immunity 31:
172. Castillo EF, Dekonenko A, Arko-Mensah J, Mandell MA, Dupont N, 761–771 http://dx.doi.org/10.1016/j.immuni.2009.09.016.
Jiang S, Delgado-Vargas M, Timmins GS, Bhattacharya D, Yang H, Hutt
186. Pedrosa J, Saunders BM, Appelberg R, Orme IM, Silva MT, Cooper
J, Lyons CR, Dobos KM, Deretic V. 2012. Autophagy protects against
AM. 2000. Neutrophils play a protective nonphagocytic role in systemic
active tuberculosis by suppressing bacterial burden and inflammation.
Mycobacterium tuberculosis infection of mice. Infect Immun 68:577–583
Proc Natl Acad Sci U S A 109:E3168–E3176 http://dx.doi.org/10.1073
http://dx.doi.org/10.1128/IAI.68.2.577-583.2000.
/pnas.1210500109.
187. Mishra BB, Lovewell RR, Olive AJ, Zhang G, Wang W, Eugenin E,
173. Kimmey JM, Huynh JP, Weiss LA, Park S, Kambal A, Debnath J,
Smith CM, Phuah JY, Long JE, Dubuke ML, Palace SG, Goguen JD,
Virgin HW, Stallings CL. 2015. Unique role for ATG5 in neutrophil-
Baker RE, Nambi S, Mishra R, Booty MG, Baer CE, Shaffer SA, Dartois
mediated immunopathology during M. tuberculosis infection. Nature
V, McCormick BA, Chen X, Sassetti CM. 2017. Nitric oxide prevents
528:565–569 http://dx.doi.org/10.1038/nature16451.
a pathogen-permissive granulocytic inflammation during tuberculosis.
174. Köster S, Upadhyay S, Chandra P, Papavinasasundaram K, Yang G, Nat Microbiol 2:17072 http://dx.doi.org/10.1038/nmicrobiol.2017.72.
Hassan A, Grigsby SJ, Mittal E, Park HS, Jones V, Hsu FF, Jackson M,
Sassetti CM, Philips JA. 2017. Mycobacterium tuberculosis is protected 188. Martineau AR, Newton SM, Wilkinson KA, Kampmann B, Hall
BM, Nawroly N, Packe GE, Davidson RN, Griffiths CJ, Wilkinson RJ.
from NADPH oxidase and LC3-associated phagocytosis by the LCP
2007. Neutrophil-mediated innate immune resistance to mycobacteria.
protein CpsA. Proc Natl Acad Sci U S A 114:E8711–E8720.
J Clin Invest 117:1988–1994 http://dx.doi.org/10.1172/JCI31097.
175. Lin Y, Zhang M, Barnes PF. 1998. Chemokine production by a
human alveolar epithelial cell line in response to Mycobacterium tuber- 189. Tan BH, Meinken C, Bastian M, Bruns H, Legaspi A, Ochoa MT,
culosis. Infect Immun 66:1121–1126. Krutzik SR, Bloom BR, Ganz T, Modlin RL, Stenger S. 2006. Macro-
phages acquire neutrophil granules for antimicrobial activity against intra-
176. Wickremasinghe MI, Thomas LH, Friedland JS. 1999. Pulmonary
cellular pathogens. J Immunol 177:1864–1871 http://dx.doi.org/10.4049
epithelial cells are a source of IL-8 in the response to Mycobacterium
/jimmunol.177.3.1864.
tuberculosis: essential role of IL-1 from infected monocytes in a NF-kappa
B-dependent network. J Immunol 163:3936–3947. 190. Ramos-Kichik V, Mondragón-Flores R, Mondragón-Castelán M,
Gonzalez-Pozos S, Muñiz-Hernandez S, Rojas-Espinosa O, Chacón-
177. Nouailles G, Dorhoi A, Koch M, Zerrahn J, Weiner J III, Faé KC,
Salinas R, Estrada-Parra S, Estrada-García I. 2009. Neutrophil extracel-
Arrey F, Kuhlmann S, Bandermann S, Loewe D, Mollenkopf HJ, Vogelzang
lular traps are induced by Mycobacterium tuberculosis. Tuberculosis
A, Meyer-Schwesinger C, Mittrücker HW, McEwen G, Kaufmann SH.
(Edinb) 89:29–37 http://dx.doi.org/10.1016/j.tube.2008.09.009.
2014. CXCL5-secreting pulmonary epithelial cells drive destructive neu-
trophilic inflammation in tuberculosis. J Clin Invest 124:1268–1282 http:// 191. Schechter MC, Buac K, Adekambi T, Cagle S, Celli J, Ray SM,
dx.doi.org/10.1172/JCI72030. Mehta CC, Rada B, Rengarajan J. 2017. Neutrophil extracellular trap
(NET) levels in human plasma are associated with active TB. PLoS One
178. Reuschl AK, Edwards MR, Parker R, Connell DW, Hoang L, Halliday
12:e0182587 http://dx.doi.org/10.1371/journal.pone.0182587.
A, Jarvis H, Siddiqui N, Wright C, Bremang S, Newton SM, Beverley P,
Shattock RJ, Kon OM, Lalvani A. 2017. Innate activation of human pri- 192. Serbina NV, Pamer EG. 2006. Monocyte emigration from bone
mary epithelial cells broadens the host response to Mycobacterium tuber- marrow during bacterial infection requires signals mediated by chemokine
culosis in the airways. PLoS Pathog 13:e1006577 http://dx.doi.org/10.1371 receptor CCR2. Nat Immunol 7:311–317 http://dx.doi.org/10.1038/ni1309.
/journal.ppat.1006577. 193. Peters W, Scott HM, Chambers HF, Flynn JL, Charo IF, Ernst JD.
179. Eum SY, Kong JH, Hong MS, Lee YJ, Kim JH, Hwang SH, Cho SN, 2001. Chemokine receptor 2 serves an early and essential role in resistance
Via LE, Barry CE III. 2010. Neutrophils are the predominant infected to Mycobacterium tuberculosis. Proc Natl Acad Sci U S A 98:7958–7963
phagocytic cells in the airways of patients with active pulmonary TB. http://dx.doi.org/10.1073/pnas.131207398.
Chest 137:122–128 http://dx.doi.org/10.1378/chest.09-0903. 194. Scott HM, Flynn JL. 2002. Mycobacterium tuberculosis in chemo-
180. Berry MP, Graham CM, McNab FW, Xu Z, Bloch SA, Oni T, kine receptor 2-deficient mice: influence of dose on disease progression.
Wilkinson KA, Banchereau R, Skinner J, Wilkinson RJ, Quinn C, Blankenship Infect Immun 70:5946–5954 http://dx.doi.org/10.1128/IAI.70.11.5946
D, Dhawan R, Cush JJ, Mejias A, Ramilo O, Kon OM, Pascual V, Banchereau -5954.2002.
J, Chaussabel D, O’Garra A. 2010. An interferon-inducible neutrophil-driven 195. Sköld M, Behar SM. 2008. Tuberculosis triggers a tissue-dependent
blood transcriptional signature in human tuberculosis. Nature 466:973–977 program of differentiation and acquisition of effector functions by circu-
http://dx.doi.org/10.1038/nature09247. lating monocytes. J Immunol 181:6349–6360 http://dx.doi.org/10.4049
181. McNab FW, Berry MP, Graham CM, Bloch SA, Oni T, Wilkinson /jimmunol.181.9.6349.
KA, Wilkinson RJ, Kon OM, Banchereau J, Chaussabel D, O’Garra A. 196. Samstein M, Schreiber HA, Leiner IM, Susac B, Glickman MS,
2011. Programmed death ligand 1 is over-expressed by neutrophils in the Pamer EG. 2013. Essential yet limited role for CCR2+ inflammatory
blood of patients with active tuberculosis. Eur J Immunol 41:1941–1947 monocytes during Mycobacterium tuberculosis-specific T cell priming.
http://dx.doi.org/10.1002/eji.201141421. eLife 2:e01086 http://dx.doi.org/10.7554/eLife.01086.

26 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Immunology of M. tuberculosis Infections

197. Antonelli LR, Gigliotti Rothfuchs A, Gonçalves R, Roffê E, Cheever ESX-1-mediated secretion and contributes to pathogenesis. J Immunol
AW, Bafica A, Salazar AM, Feng CG, Sher A. 2010. Intranasal poly-IC 178:3143–3152 http://dx.doi.org/10.4049/jimmunol.178.5.3143.
treatment exacerbates tuberculosis in mice through the pulmonary re- 211. Desvignes L, Wolf AJ, Ernst JD. 2012. Dynamic roles of type I
cruitment of a pathogen-permissive monocyte/macrophage population. and type II IFNs in early infection with Mycobacterium tuberculosis. J
J Clin Invest 120:1674–1682 http://dx.doi.org/10.1172/JCI40817. Immunol 188:6205–6215 http://dx.doi.org/10.4049/jimmunol.1200255.
198. Esin S, Counoupas C, Aulicino A, Brancatisano FL, Maisetta G, 212. Moreira-Teixeira L, Sousa J, McNab FW, Torrado E, Cardoso F,
Bottai D, Di Luca M, Florio W, Campa M, Batoni G. 2013. Interaction of Machado H, Castro F, Cardoso V, Gaifem J, Wu X, Appelberg R, Castro
Mycobacterium tuberculosis cell wall components with the human natural AG, O’Garra A, Saraiva M. 2016. Type I IFN inhibits alternative mac-
killer cell receptors NKp44 and Toll-like receptor 2. Scand J Immunol rophage activation during Mycobacterium tuberculosis infection and leads
77:460–469 http://dx.doi.org/10.1111/sji.12052. to enhanced protection in the absence of IFN-γ signaling. J Immunol
199. Vankayalapati R, Garg A, Porgador A, Griffith DE, Klucar P, Safi H, 197:4714–4726 http://dx.doi.org/10.4049/jimmunol.1600584.
Girard WM, Cosman D, Spies T, Barnes PF. 2005. Role of NK cell- 213. Novikov A, Cardone M, Thompson R, Shenderov K, Kirschman KD,
activating receptors and their ligands in the lysis of mononuclear phago- Mayer-Barber KD, Myers TG, Rabin RL, Trinchieri G, Sher A, Feng CG.
cytes infected with an intracellular bacterium. J Immunol 175:4611–4617 2011. Mycobacterium tuberculosis triggers host type I IFN signaling to
http://dx.doi.org/10.4049/jimmunol.175.7.4611. regulate IL-1β production in human macrophages. J Immunol 187:2540–
200. Dhiman R, Indramohan M, Barnes PF, Nayak RC, Paidipally P, Rao 2547 http://dx.doi.org/10.4049/jimmunol.1100926.
LV, Vankayalapati R. 2009. IL-22 produced by human NK cells inhibits 214. Mayer-Barber KD, Andrade BB, Barber DL, Hieny S, Feng CG,
growth of Mycobacterium tuberculosis by enhancing phagolysosomal Caspar P, Oland S, Gordon S, Sher A. 2011. Innate and adaptive inter-
fusion. J Immunol 183:6639–6645 http://dx.doi.org/10.4049/jimmunol ferons suppress IL-1α and IL-1β production by distinct pulmonary mye-
.0902587. loid subsets during Mycobacterium tuberculosis infection. Immunity 35:
201. Schierloh P, Alemán M, Yokobori N, Alves L, Roldán N, Abbate E, 1023–1034 http://dx.doi.org/10.1016/j.immuni.2011.12.002.
del C Sasiain M, de la Barrera S. 2005. NK cell activity in tuberculosis is 215. McNab FW, Ewbank J, Howes A, Moreira-Teixeira L, Martirosyan
associated with impaired CD11a and ICAM-1 expression: a regulatory A, Ghilardi N, Saraiva M, O’Garra A. 2014. Type I IFN induces IL-10
role of monocytes in NK activation. Immunology 116:541–552. production in an IL-27-independent manner and blocks responsiveness to
202. Vankayalapati R, Klucar P, Wizel B, Weis SE, Samten B, Safi H, IFN-γ for production of IL-12 and bacterial killing in Mycobacterium
Shams H, Barnes PF. 2004. NK cells regulate CD8+ T cell effector func- tuberculosis-infected macrophages. J Immunol 193:3600–3612 http://dx
tion in response to an intracellular pathogen. J Immunol 172:130–137 .doi.org/10.4049/jimmunol.1401088.
http://dx.doi.org/10.4049/jimmunol.172.1.130. 216. Ong CW, Elkington PT, Brilha S, Ugarte-Gil C, Tome-Esteban MT,
203. Zhang R, Zheng X, Li B, Wei H, Tian Z. 2006. Human NK cells Tezera LB, Pabisiak PJ, Moores RC, Sathyamoorthy T, Patel V, Gilman
positively regulate gammadelta T cells in response to Mycobacterium tu- RH, Porter JC, Friedland JS. 2015. Neutrophil-derived MMP-8 drives
berculosis. J Immunol 176:2610–2616 http://dx.doi.org/10.4049/jimmunol AMPK-dependent matrix destruction in human pulmonary tuberculosis.
.176.4.2610. PLoS Pathog 11:e1004917 http://dx.doi.org/10.1371/journal.ppat.1004917.
204. Roy S, Barnes PF, Garg A, Wu S, Cosman D, Vankayalapati R. 2008. 217. Elkington PT, Emerson JE, Lopez-Pascua LD, O’Kane CM,
NK cells lyse T regulatory cells that expand in response to an intracellular Horncastle DE, Boyle JJ, Friedland JS. 2005. Mycobacterium tuberculosis
pathogen. J Immunol 180:1729–1736 http://dx.doi.org/10.4049/jimmunol up-regulates matrix metalloproteinase-1 secretion from human airway
.180.3.1729. epithelial cells via a p38 MAPK switch. J Immunol 175:5333–5340 http://
205. Nirmala R, Narayanan PR, Mathew R, Maran M, Deivanayagam dx.doi.org/10.4049/jimmunol.175.8.5333.
CN. 2001. Reduced NK activity in pulmonary tuberculosis patients with/ 218. Elkington PT, Nuttall RK, Boyle JJ, O’Kane CM, Horncastle DE,
without HIV infection: identifying the defective stage and studying the Edwards DR, Friedland JS. 2005. Mycobacterium tuberculosis, but not
effect of interleukins on NK activity. Tuberculosis (Edinb) 81:343–352 vaccine BCG, specifically upregulates matrix metalloproteinase-1. Am J
http://dx.doi.org/10.1054/tube.2001.0309. Respir Crit Care Med 172:1596–1604 http://dx.doi.org/10.1164/rccm
206. Venkatasubramanian S, Cheekatla S, Paidipally P, Tripathi D, Welch .200505-753OC.
E, Tvinnereim AR, Nurieva R, Vankayalapati R. 2017. IL-21-dependent 219. Price NM, Farrar J, Tran TT, Nguyen TH, Tran TH, Friedland JS.
expansion of memory-like NK cells enhances protective immune responses 2001. Identification of a matrix-degrading phenotype in human tubercu-
against Mycobacterium tuberculosis. Mucosal Immunol 10:1031–1042 losis in vitro and in vivo. J Immunol 166:4223–4230 http://dx.doi.org
http://dx.doi.org/10.1038/mi.2016.105. /10.4049/jimmunol.166.6.4223.
207. Manca C, Tsenova L, Bergtold A, Freeman S, Tovey M, Musser JM, 220. Elkington P, Shiomi T, Breen R, Nuttall RK, Ugarte-Gil CA, Walker
Barry CE III, Freedman VH, Kaplan G. 2001. Virulence of a Mycobac- NF, Saraiva L, Pedersen B, Mauri F, Lipman M, Edwards DR, Robertson
terium tuberculosis clinical isolate in mice is determined by failure to in- BD, D’Armiento J, Friedland JS. 2011. MMP-1 drives immunopathology
duce Th1 type immunity and is associated with induction of IFN-alpha/ in human tuberculosis and transgenic mice. J Clin Invest 121:1827–1833
beta. Proc Natl Acad Sci U S A 98:5752–5757 http://dx.doi.org/10.1073 http://dx.doi.org/10.1172/JCI45666.
/pnas.091096998. 221. Volkman HE, Pozos TC, Zheng J, Davis JM, Rawls JF,
208. Manca C, Tsenova L, Freeman S, Barczak AK, Tovey M, Murray PJ, Ramakrishnan L. 2010. Tuberculous granuloma induction via interaction
Barry C III, Kaplan G. 2005. Hypervirulent M. tuberculosis W/Beijing of a bacterial secreted protein with host epithelium. Science 327:466–469
strains upregulate type I IFNs and increase expression of negative http://dx.doi.org/10.1126/science.1179663.
regulators of the Jak-Stat pathway. J Interferon Cytokine Res 25:694–701 222. Bafica A, Scanga CA, Serhan C, Machado F, White S, Sher A, Aliberti
http://dx.doi.org/10.1089/jir.2005.25.694. J. 2005. Host control of Mycobacterium tuberculosis is regulated by 5-
209. Ordway D, Henao-Tamayo M, Harton M, Palanisamy G, Troudt J, lipoxygenase-dependent lipoxin production. J Clin Invest 115:1601–1606
Shanley C, Basaraba RJ, Orme IM. 2007. The hypervirulent Myco- http://dx.doi.org/10.1172/JCI23949.
bacterium tuberculosis strain HN878 induces a potent TH1 response 223. Chen M, Divangahi M, Gan H, Shin DS, Hong S, Lee DM, Serhan
followed by rapid down-regulation. J Immunol 179:522–531 http://dx CN, Behar SM, Remold HG. 2008. Lipid mediators in innate immunity
.doi.org/10.4049/jimmunol.179.1.522. against tuberculosis: opposing roles of PGE2 and LXA4 in the induction
210. Stanley SA, Johndrow JE, Manzanillo P, Cox JS. 2007. The type I of macrophage death. J Exp Med 205:2791–2801 http://dx.doi.org
IFN response to infection with Mycobacterium tuberculosis requires /10.1084/jem.20080767.

ASMscience.org/MicrobiolSpectrum 27
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Sia and Rengarajan

224. Divangahi M, Desjardins D, Nunes-Alves C, Remold HG, Behar 238. Blomgran R, Ernst JD. 2011. Lung neutrophils facilitate activa-
SM. 2010. Eicosanoid pathways regulate adaptive immunity to Mycobac- tion of naive antigen-specific CD4+ T cells during Mycobacterium tuber-
terium tuberculosis. Nat Immunol 11:751–758 http://dx.doi.org/10.1038 culosis infection. J Immunol 186:7110–7119 http://dx.doi.org/10.4049
/ni.1904. /jimmunol.1100001.
225. Tobin DM, Vary JC Jr, Ray JP, Walsh GS, Dunstan SJ, Bang ND, 239. Blomgran R, Desvignes L, Briken V, Ernst JD. 2012. Mycobacterium
Hagge DA, Khadge S, King MC, Hawn TR, Moens CB, Ramakrishnan L. tuberculosis inhibits neutrophil apoptosis, leading to delayed activation of
2010. The lta4h locus modulates susceptibility to mycobacterial infection naive CD4 T cells. Cell Host Microbe 11:81–90 http://dx.doi.org/10.1016
in zebrafish and humans. Cell 140:717–730 http://dx.doi.org/10.1016/j.cell /j.chom.2011.11.012.
.2010.02.013. 240. Martin CJ, Booty MG, Rosebrock TR, Nunes-Alves C, Desjardins
226. Tobin DM, Roca FJ, Oh SF, McFarland R, Vickery TW, Ray JP, DM, Keren I, Fortune SM, Remold HG, Behar SM. 2012. Efferocytosis
Ko DC, Zou Y, Bang ND, Chau TT, Vary JC, Hawn TR, Dunstan SJ, is an innate antibacterial mechanism. Cell Host Microbe 12:289–300
Farrar JJ, Thwaites GE, King MC, Serhan CN, Ramakrishnan L. 2012. http://dx.doi.org/10.1016/j.chom.2012.06.010.
Host genotype-specific therapies can optimize the inflammatory response 241. Divangahi M, Chen M, Gan H, Desjardins D, Hickman TT, Lee DM,
to mycobacterial infections. Cell 148:434–446 http://dx.doi.org/10.1016 Fortune S, Behar SM, Remold HG. 2009. Mycobacterium tuberculosis
/j.cell.2011.12.023. evades macrophage defenses by inhibiting plasma membrane repair. Nat
227. Flynn JL, Goldstein MM, Chan J, Triebold KJ, Pfeffer K, Lowenstein Immunol 10:899–906 http://dx.doi.org/10.1038/ni.1758.
CJ, Schreiber R, Mak TW, Bloom BR. 1995. Tumor necrosis factor-alpha 242. Zhao X, Khan N, Gan H, Tzelepis F, Nishimura T, Park SY,
is required in the protective immune response against Mycobacterium Divangahi M, Remold HG. 2017. Bcl-xL mediates RIPK3-dependent ne-
tuberculosis in mice. Immunity 2:561–572 http://dx.doi.org/10.1016 crosis in M. tuberculosis-infected macrophages. Mucosal Immunol 10:
/1074-7613(95)90001-2. 1553–1568 http://dx.doi.org/10.1038/mi.2017.12.
228. Keane J, Gershon S, Wise RP, Mirabile-Levens E, Kasznica J, 243. Tian T, Woodworth J, Sköld M, Behar SM. 2005. In vivo depletion
Schwieterman WD, Siegel JN, Braun MM. 2001. Tuberculosis associated of CD11c+ cells delays the CD4+ T cell response to Mycobacterium
with infliximab, a tumor necrosis factor alpha-neutralizing agent. N Engl J tuberculosis and exacerbates the outcome of infection. J Immunol 175:
Med 345:1098–1104 http://dx.doi.org/10.1056/NEJMoa011110. 3268–3272 http://dx.doi.org/10.4049/jimmunol.175.5.3268.
229. Botha T, Ryffel B. 2003. Reactivation of latent tuberculosis infec- 244. Bodnar KA, Serbina NV, Flynn JL. 2001. Fate of Mycobacterium
tion in TNF-deficient mice. J Immunol 171:3110–3118 http://dx.doi.org tuberculosis within murine dendritic cells. Infect Immun 69:800–809
/10.4049/jimmunol.171.6.3110. http://dx.doi.org/10.1128/IAI.69.2.800-809.2001.
230. Chakravarty SD, Zhu G, Tsai MC, Mohan VP, Marino S, Kirschner 245. Jiao X, Lo-Man R, Guermonprez P, Fiette L, Dériaud E, Burgaud S,
DE, Huang L, Flynn J, Chan J. 2008. Tumor necrosis factor blockade Gicquel B, Winter N, Leclerc C. 2002. Dendritic cells are host cells for
in chronic murine tuberculosis enhances granulomatous inflammation mycobacteria in vivo that trigger innate and acquired immunity. J Immunol
and disorganizes granulomas in the lungs. Infect Immun 76:916–926 168:1294–1301 http://dx.doi.org/10.4049/jimmunol.168.3.1294.
http://dx.doi.org/10.1128/IAI.01011-07. 246. Wolf AJ, Linas B, Trevejo-Nuñez GJ, Kincaid E, Tamura T, Takatsu
231. Mohan VP, Scanga CA, Yu K, Scott HM, Tanaka KE, Tsang E, K, Ernst JD. 2007. Mycobacterium tuberculosis infects dendritic cells with
Tsai MM, Flynn JL, Chan J. 2001. Effects of tumor necrosis factor alpha high frequency and impairs their function in vivo. J Immunol 179:2509–
on host immune response in chronic persistent tuberculosis: possible role 2519 http://dx.doi.org/10.4049/jimmunol.179.4.2509.
for limiting pathology. Infect Immun 69:1847–1855 http://dx.doi.org 247. Henderson RA, Watkins SC, Flynn JL. 1997. Activation of human
/10.1128/IAI.69.3.1847-1855.2001. dendritic cells following infection with Mycobacterium tuberculosis.
232. Oddo M, Renno T, Attinger A, Bakker T, MacDonald HR, Meylan J Immunol 159:635–643.
PR. 1998. Fas ligand-induced apoptosis of infected human macrophages 248. Tailleux L, Neyrolles O, Honoré-Bouakline S, Perret E, Sanchez F,
reduces the viability of intracellular Mycobacterium tuberculosis. J Im- Abastado JP, Lagrange PH, Gluckman JC, Rosenzwajg M, Herrmann JL.
munol 160:5448–5454. 2003. Constrained intracellular survival of Mycobacterium tuberculosis
233. Keane J, Remold HG, Kornfeld H. 2000. Virulent Mycobacterium in human dendritic cells. J Immunol 170:1939–1948 http://dx.doi.org
tuberculosis strains evade apoptosis of infected alveolar macrophages. /10.4049/jimmunol.170.4.1939.
J Immunol 164:2016–2020 http://dx.doi.org/10.4049/jimmunol.164.4 249. Tailleux L, Schwartz O, Herrmann JL, Pivert E, Jackson M, Amara
.2016. A, Legres L, Dreher D, Nicod LP, Gluckman JC, Lagrange PH, Gicquel B,
234. Chen M, Gan H, Remold HG. 2006. A mechanism of virulence: Neyrolles O. 2003. DC-SIGN is the major Mycobacterium tuberculosis
virulent Mycobacterium tuberculosis strain H37Rv, but not attenuated receptor on human dendritic cells. J Exp Med 197:121–127 http://dx.doi
H37Ra, causes significant mitochondrial inner membrane disruption .org/10.1084/jem.20021468.
in macrophages leading to necrosis. J Immunol 176:3707–3716 http://dx 250. Wolf AJ, Desvignes L, Linas B, Banaiee N, Tamura T, Takatsu K,
.doi.org/10.4049/jimmunol.176.6.3707. Ernst JD. 2008. Initiation of the adaptive immune response to Mycobac-
235. Gan H, Lee J, Ren F, Chen M, Kornfeld H, Remold HG. 2008. terium tuberculosis depends on antigen production in the local lymph
Mycobacterium tuberculosis blocks crosslinking of annexin-1 and apo- node, not the lungs. J Exp Med 205:105–115 http://dx.doi.org/10.1084
ptotic envelope formation on infected macrophages to maintain virulence. /jem.20071367.
Nat Immunol 9:1189–1197 http://dx.doi.org/10.1038/ni.1654. 251. Olmos S, Stukes S, Ernst JD. 2010. Ectopic activation of Myco-
236. Hinchey J, Lee S, Jeon BY, Basaraba RJ, Venkataswamy MM, bacterium tuberculosis-specific CD4+ T cells in lungs of CCR7-/- mice.
Chen B, Chan J, Braunstein M, Orme IM, Derrick SC, Morris SL, Jacobs J Immunol 184:895–901 http://dx.doi.org/10.4049/jimmunol.0901230.
WR Jr, Porcelli SA. 2007. Enhanced priming of adaptive immunity by a 252. Bhatt K, Hickman SP, Salgame P. 2004. Cutting edge: a new ap-
proapoptotic mutant of Mycobacterium tuberculosis. J Clin Invest 117: proach to modeling early lung immunity in murine tuberculosis. J Immunol
2279–2288 http://dx.doi.org/10.1172/JCI31947. 172:2748–2751 http://dx.doi.org/10.4049/jimmunol.172.5.2748.
237. Velmurugan K, Chen B, Miller JL, Azogue S, Gurses S, Hsu T, 253. Khader SA, Partida-Sanchez S, Bell G, Jelley-Gibbs DM, Swain S,
Glickman M, Jacobs WR Jr, Porcelli SA, Briken V. 2007. Mycobacterium Pearl JE, Ghilardi N, Desauvage FJ, Lund FE, Cooper AM. 2006. Inter-
tuberculosis nuoG is a virulence gene that inhibits apoptosis of infected leukin 12p40 is required for dendritic cell migration and T cell priming
host cells. PLoS Pathog 3:e110 http://dx.doi.org/10.1371/journal.ppat after Mycobacterium tuberculosis infection. J Exp Med 203:1805–1815
.0030110. http://dx.doi.org/10.1084/jem.20052545.

28 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Immunology of M. tuberculosis Infections

254. Srivastava S, Ernst JD. 2014. Cell-to-cell transfer of M. tuberculosis 270. Mogues T, Goodrich ME, Ryan L, LaCourse R, North RJ. 2001. The
antigens optimizes CD4 T cell priming. Cell Host Microbe 15:741–752 relative importance of T cell subsets in immunity and immunopathology
http://dx.doi.org/10.1016/j.chom.2014.05.007. of airborne Mycobacterium tuberculosis infection in mice. J Exp Med
255. Srivastava S, Grace PS, Ernst JD. 2016. Antigen export reduces 193:271–280 http://dx.doi.org/10.1084/jem.193.3.271.
antigen presentation and limits T cell control of M. tuberculosis. Cell Host 271. Caruso AM, Serbina N, Klein E, Triebold K, Bloom BR, Flynn JL.
Microbe 19:44–54 http://dx.doi.org/10.1016/j.chom.2015.12.003. 1999. Mice deficient in CD4 T cells have only transiently diminished levels
256. Harding CV, Boom WH. 2010. Regulation of antigen presentation of IFN-gamma, yet succumb to tuberculosis. J Immunol 162:5407–5416.
by Mycobacterium tuberculosis: a role for Toll-like receptors. Nat Rev 272. Reiley WW, Calayag MD, Wittmer ST, Huntington JL, Pearl JE,
Microbiol 8:296–307 http://dx.doi.org/10.1038/nrmicro2321. Fountain JJ, Martino CA, Roberts AD, Cooper AM, Winslow GM,
257. Ramachandra L, Noss E, Boom WH, Harding CV. 2001. Processing Woodland DL. 2008. ESAT-6-specific CD4 T cell responses to aerosol
of Mycobacterium tuberculosis antigen 85B involves intraphagosomal Mycobacterium tuberculosis infection are initiated in the mediastinal
formation of peptide-major histocompatibility complex II complexes and lymph nodes. Proc Natl Acad Sci U S A 105:10961–10966 http://dx.doi
is inhibited by live bacilli that decrease phagosome maturation. J Exp Med .org/10.1073/pnas.0801496105.
194:1421–1432 http://dx.doi.org/10.1084/jem.194.10.1421. 273. Urdahl KB, Shafiani S, Ernst JD. 2011. Initiation and regulation of
258. Kincaid EZ, Ernst JD. 2003. Mycobacterium tuberculosis exerts T-cell responses in tuberculosis. Mucosal Immunol 4:288–293 http://dx
gene-selective inhibition of transcriptional responses to IFN-gamma .doi.org/10.1038/mi.2011.10.
without inhibiting STAT1 function. J Immunol 171:2042–2049 http://dx 274. Chackerian AA, Alt JM, Perera TV, Dascher CC, Behar SM. 2002.
.doi.org/10.4049/jimmunol.171.4.2042. Dissemination of Mycobacterium tuberculosis is influenced by host factors
259. Pai RK, Convery M, Hamilton TA, Boom WH, Harding CV. 2003. and precedes the initiation of T-cell immunity. Infect Immun 70:4501–
Inhibition of IFN-gamma-induced class II transactivator expression by a 4509 http://dx.doi.org/10.1128/IAI.70.8.4501-4509.2002.
19-kDa lipoprotein from Mycobacterium tuberculosis: a potential mech- 275. Winslow GM, Cooper A, Reiley W, Chatterjee M, Woodland DL.
anism for immune evasion. J Immunol 171:175–184 http://dx.doi.org 2008. Early T-cell responses in tuberculosis immunity. Immunol Rev
/10.4049/jimmunol.171.1.175. 225:284–299 http://dx.doi.org/10.1111/j.1600-065X.2008.00693.x.
260. Pennini ME, Liu Y, Yang J, Croniger CM, Boom WH, 276. Lin PL, Pawar S, Myers A, Pegu A, Fuhrman C, Reinhart TA,
Harding CV. 2007. CCAAT/enhancer-binding protein beta and delta Capuano SV, Klein E, Flynn JL. 2006. Early events in Mycobacterium
binding to CIITA promoters is associated with the inhibition of CIITA tuberculosis infection in cynomolgus macaques. Infect Immun 74:3790–
expression in response to Mycobacterium tuberculosis 19-kDa lipopro- 3803 http://dx.doi.org/10.1128/IAI.00064-06.
tein. J Immunol 179:6910–6918 http://dx.doi.org/10.4049/jimmunol.179 277. Kursar M, Bonhagen K, Köhler A, Kamradt T, Kaufmann SH,
.10.6910. Mittrücker HW. 2002. Organ-specific CD4+ T cell response during Lis-
261. Pennini ME, Pai RK, Schultz DC, Boom WH, Harding CV. 2006. teria monocytogenes infection. J Immunol 168:6382–6387 http://dx.doi
Mycobacterium tuberculosis 19-kDa lipoprotein inhibits IFN-gamma- .org/10.4049/jimmunol.168.12.6382.
induced chromatin remodeling of MHC2TA by TLR2 and MAPK sig- 278. Manicassamy B, Manicassamy S, Belicha-Villanueva A, Pisanelli
naling. J Immunol 176:4323–4330 http://dx.doi.org/10.4049/jimmunol G, Pulendran B, García-Sastre A. 2010. Analysis of in vivo dynamics
.176.7.4323. of influenza virus infection in mice using a GFP reporter virus. Proc
262. Sia JK, Georgieva M, Rengarajan J. 2015. Innate immune defenses in Natl Acad Sci U S A 107:11531–11536 http://dx.doi.org/10.1073/pnas
human tuberculosis: an overview of the interactions between Mycobac- .0914994107.
terium tuberculosis and innate immune cells. J Immunol Res 2015:747543 279. Gallegos AM, Pamer EG, Glickman MS. 2008. Delayed protection
http://dx.doi.org/10.1155/2015/747543. by ESAT-6-specific effector CD4+ T cells after airborne M. tuberculosis
263. Baena A, Porcelli SA. 2009. Evasion and subversion of antigen infection. J Exp Med 205:2359–2368 http://dx.doi.org/10.1084/jem
presentation by Mycobacterium tuberculosis. Tissue Antigens 74:189– .20080353.
204 http://dx.doi.org/10.1111/j.1399-0039.2009.01301.x. 280. Sakai S, Kauffman KD, Schenkel JM, McBerry CC, Mayer-Barber
264. Srivastava S, Ernst JD, Desvignes L. 2014. Beyond macrophages: the KD, Masopust D, Barber DL. 2014. Cutting edge: control of Mycobac-
diversity of mononuclear cells in tuberculosis. Immunol Rev 262:179–192 terium tuberculosis infection by a subset of lung parenchyma-homing CD4
http://dx.doi.org/10.1111/imr.12217. T cells. J Immunol 192:2965–2969 http://dx.doi.org/10.4049/jimmunol
265. Portal-Celhay C, Tufariello JM, Srivastava S, Zahra A, Klevorn T, .1400019.
Grace PS, Mehra A, Park HS, Ernst JD, Jacobs WR Jr, Philips JA. 281. Sakai S, Kauffman KD, Sallin MA, Sharpe AH, Young HA, Ganusov
2016. Mycobacterium tuberculosis EsxH inhibits ESCRT-dependent VV, Barber DL. 2016. CD4 T cell-derived IFN-γ plays a minimal role
CD4+ T-cell activation. Nat Microbiol 2:16232 http://dx.doi.org/10.1038 in control of pulmonary Mycobacterium tuberculosis infection and must
/nmicrobiol.2016.232. be actively repressed by PD-1 to prevent lethal disease. PLoS Pathog 12:
266. Grace PS, Ernst JD. 2016. Suboptimal antigen presentation e1005667 http://dx.doi.org/10.1371/journal.ppat.1005667.
contributes to virulence of Mycobacterium tuberculosisin vivo. J Immunol 282. Kauffman KD, Sallin MA, Sakai S, Kamenyeva O, Kabat J, Weiner
196:357–364 http://dx.doi.org/10.4049/jimmunol.1501494. D, Sutphin M, Schimel D, Via L, Barry CE III, Wilder-Kofie T, Moore I,
267. Bizzell E, Sia JK, Quezada M, Enriquez A, Georgieva M, Rengarajan Moore R, Barber DL. 2017. Defective positioning in granulomas but
J. 2017. Deletion of BCG Hip1 protease enhances dendritic cell and CD4 not lung-homing limits CD4 T-cell interactions with Mycobacterium
T cell responses. J Leukoc Biol 103:739–748. tuberculosis-infected macrophages in rhesus macaques. Mucosal Immunol
268. Sia JK, Bizzell E, Madan-Lala R, Rengarajan J. 2017. Engaging the 11:462–473.
CD40-CD40L pathway augments T-helper cell responses and improves 283. Mehra S, Alvarez X, Didier PJ, Doyle LA, Blanchard JL, Lackner AA,
control of Mycobacterium tuberculosis infection. PLoS Pathog 13: Kaushal D. 2013. Granuloma correlates of protection against tuberculosis
e1006530 http://dx.doi.org/10.1371/journal.ppat.1006530. and mechanisms of immune modulation by Mycobacterium tuberculosis.
269. Griffiths KL, Ahmed M, Das S, Gopal R, Horne W, Connell TD, J Infect Dis 207:1115–1127 http://dx.doi.org/10.1093/infdis/jis778.
Moynihan KD, Kolls JK, Irvine DJ, Artyomov MN, Rangel-Moreno J, 284. Ottenhoff TH, Kumararatne D, Casanova JL. 1998. Novel human
Khader SA. 2016. Targeting dendritic cells to accelerate T-cell activation immunodeficiencies reveal the essential role of type-I cytokines in immu-
overcomes a bottleneck in tuberculosis vaccine efficacy. Nat Commun nity to intracellular bacteria. Immunol Today 19:491–494 http://dx.doi
7:13894 http://dx.doi.org/10.1038/ncomms13894. .org/10.1016/S0167-5699(98)01321-8.

ASMscience.org/MicrobiolSpectrum 29
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Sia and Rengarajan

285. Jouanguy E, Altare F, Lamhamedi S, Revy P, Emile JF, Newport M, 296. Altare F, Ensser A, Breiman A, Reichenbach J, Baghdadi JE, Fischer
Levin M, Blanche S, Seboun E, Fischer A, Casanova JL. 1996. Interferon- A, Emile JF, Gaillard JL, Meinl E, Casanova JL. 2001. Interleukin-12
gamma-receptor deficiency in an infant with fatal bacille Calmette-Guérin receptor beta1 deficiency in a patient with abdominal tuberculosis. J Infect
infection. N Engl J Med 335:1956–1961 http://dx.doi.org/10.1056/NEJM Dis 184:231–236 http://dx.doi.org/10.1086/321999.
199612263352604. 297. Caragol I, Raspall M, Fieschi C, Feinberg J, Larrosa MN, Hernández
286. Newport MJ, Huxley CM, Huston S, Hawrylowicz CM, Oostra M, Figueras C, Bertrán JM, Casanova JL, Español T. 2003. Clinical tu-
BA, Williamson R, Levin M. 1996. A mutation in the interferon-gamma- berculosis in 2 of 3 siblings with interleukin-12 receptor beta1 deficiency.
receptor gene and susceptibility to mycobacterial infection. N Engl J Clin Infect Dis 37:302–306 http://dx.doi.org/10.1086/375587.
Med 335:1941–1949 http://dx.doi.org/10.1056/NEJM19961226335 298. Fieschi C, Bosticardo M, de Beaucoudrey L, Boisson-Dupuis S,
2602. Feinberg J, Santos OF, Bustamante J, Levy J, Candotti F, Casanova JL.
287. Dorman SE, Holland SM. 1998. Mutation in the signal-transducing 2004. A novel form of complete IL-12/IL-23 receptor beta1 deficiency
chain of the interferon-gamma receptor and susceptibility to mycobacte- with cell surface-expressed nonfunctional receptors. Blood 104:2095–
rial infection. J Clin Invest 101:2364–2369 http://dx.doi.org/10.1172 2101 http://dx.doi.org/10.1182/blood-2004-02-0584.
/JCI2901. 299. Tsao TC, Chen CH, Hong JH, Hsieh MJ, Tsao KC, Lee CH. 2002.
288. Jouanguy E, Lamhamedi-Cherradi S, Lammas D, Dorman SE, Shifts of T4/T8 T lymphocytes from BAL fluid and peripheral blood by
Fondanèche MC, Dupuis S, Döffinger R, Altare F, Girdlestone J, Emile JF, clinical grade in patients with pulmonary tuberculosis. Chest 122:1285–
Ducoulombier H, Edgar D, Clarke J, Oxelius VA, Brai M, Novelli V, 1291 http://dx.doi.org/10.1378/chest.122.4.1285.
Heyne K, Fischer A, Holland SM, Kumararatne DS, Schreiber RD, 300. Bhattacharyya S, Singla R, Dey AB, Prasad HK. 1999. Dichotomy of
Casanova JL. 1999. A human IFNGR1 small deletion hotspot associated cytokine profiles in patients and high-risk healthy subjects exposed to
with dominant susceptibility to mycobacterial infection. Nat Genet 21: tuberculosis. Infect Immun 67:5597–5603.
370–378 http://dx.doi.org/10.1038/7701. 301. Hirsch CS, Toossi Z, Othieno C, Johnson JL, Schwander SK,
289. Dupuis S, Dargemont C, Fieschi C, Thomassin N, Rosenzweig S, Robertson S, Wallis RS, Edmonds K, Okwera A, Mugerwa R, Peters P,
Harris J, Holland SM, Schreiber RD, Casanova JL. 2001. Impairment of Ellner JJ. 1999. Depressed T-cell interferon-gamma responses in pulmo-
mycobacterial but not viral immunity by a germline human STAT1 mu- nary tuberculosis: analysis of underlying mechanisms and modulation
tation. Science 293:300–303 http://dx.doi.org/10.1126/science.1061154. with therapy. J Infect Dis 180:2069–2073 http://dx.doi.org/10.1086
290. Filipe-Santos O, Bustamante J, Haverkamp MH, Vinolo E, Ku CL, /315114.
Puel A, Frucht DM, Christel K, von Bernuth H, Jouanguy E, Feinberg J, 302. Torres M, Herrera T, Villareal H, Rich EA, Sada E. 1998. Cytokine
Durandy A, Senechal B, Chapgier A, Vogt G, de Beaucoudrey L, Fieschi C, profiles for peripheral blood lymphocytes from patients with active pul-
Picard C, Garfa M, Chemli J, Bejaoui M, Tsolia MN, Kutukculer N, monary tuberculosis and healthy household contacts in response to the
Plebani A, Notarangelo L, Bodemer C, Geissmann F, Israël A, Véron M, 30-kilodalton antigen of Mycobacterium tuberculosis. Infect Immun 66:
Knackstedt M, Barbouche R, Abel L, Magdorf K, Gendrel D, Agou F, 176–180.
Holland SM, Casanova JL. 2006. X-linked susceptibility to mycobacteria 303. Vekemans J, Lienhardt C, Sillah JS, Wheeler JG, Lahai GP, Doherty
is caused by mutations in NEMO impairing CD40-dependent IL-12 MT, Corrah T, Andersen P, McAdam KP, Marchant A. 2001. Tubercu-
production. J Exp Med 203:1745–1759 http://dx.doi.org/10.1084/jem losis contacts but not patients have higher gamma interferon responses
.20060085. to ESAT-6 than do community controls in The Gambia. Infect Immun
291. Altare F, Durandy A, Lammas D, Emile JF, Lamhamedi S, Le Deist F, 69:6554–6557 http://dx.doi.org/10.1128/IAI.69.10.6554-6557.2001.
Drysdale P, Jouanguy E, Döffinger R, Bernaudin F, Jeppsson O, Gollob 304. Pathan AA, Wilkinson KA, Klenerman P, McShane H, Davidson RN,
JA, Meinl E, Segal AW, Fischer A, Kumararatne D, Casanova JL. 1998. Pasvol G, Hill AV, Lalvani A. 2001. Direct ex vivo analysis of antigen-
Impairment of mycobacterial immunity in human interleukin-12 receptor specific IFN-gamma-secreting CD4 T cells in Mycobacterium tuberculosis-
deficiency. Science 280:1432–1435 http://dx.doi.org/10.1126/science.280 infected individuals: associations with clinical disease state and effect of
.5368.1432. treatment. J Immunol 167:5217–5225 http://dx.doi.org/10.4049/jimmunol
292. Altare F, Lammas D, Revy P, Jouanguy E, Döffinger R, Lamhamedi .167.9.5217.
S, Drysdale P, Scheel-Toellner D, Girdlestone J, Darbyshire P, Wadhwa 305. Sodhi A, Gong J, Silva C, Qian D, Barnes PF. 1997. Clinical
M, Dockrell H, Salmon M, Fischer A, Durandy A, Casanova JL, correlates of interferon gamma production in patients with tuberculosis.
Kumararatne DS. 1998. Inherited interleukin 12 deficiency in a child with Clin Infect Dis 25:617–620 http://dx.doi.org/10.1086/513769.
bacille Calmette-Guérin and Salmonella enteritidis disseminated infection. 306. Lindestam Arlehamn CS, Gerasimova A, Mele F, Henderson R,
J Clin Invest 102:2035–2040 http://dx.doi.org/10.1172/JCI4950. Swann J, Greenbaum JA, Kim Y, Sidney J, James EA, Taplitz R,
293. Elloumi-Zghal H, Barbouche MR, Chemli J, Béjaoui M, Harbi A, McKinney DM, Kwok WW, Grey H, Sallusto F, Peters B, Sette A. 2013.
Snoussi N, Abdelhak S, Dellagi K. 2002. Clinical and genetic hetero- Memory T cells in latent Mycobacterium tuberculosis infection are
geneity of inherited autosomal recessive susceptibility to disseminated directed against three antigenic islands and largely contained in a CXCR3
Mycobacterium bovis bacille Calmette-Guérin infection. J Infect Dis 185: +CCR6+ Th1 subset. PLoS Pathog 9:e1003130 http://dx.doi.org/10.1371
1468–1475 http://dx.doi.org/10.1086/340510. /journal.ppat.1003130.
294. Picard C, Fieschi C, Altare F, Al-Jumaah S, Al-Hajjar S, Feinberg J, 307. Flynn JL, Chan J, Triebold KJ, Dalton DK, Stewart TA, Bloom BR.
Dupuis S, Soudais C, Al-Mohsen IZ, Génin E, Lammas D, Kumararatne 1993. An essential role for interferon gamma in resistance to Mycobac-
DS, Leclerc T, Rafii A, Frayha H, Murugasu B, Wah LB, Sinniah R, terium tuberculosis infection. J Exp Med 178:2249–2254 http://dx.doi
Loubser M, Okamoto E, Al-Ghonaium A, Tufenkeji H, Abel L, Casanova .org/10.1084/jem.178.6.2249.
JL. 2002. Inherited interleukin-12 deficiency: IL12B genotype and clinical 308. Cooper AM, Dalton DK, Stewart TA, Griffin JP, Russell DG, Orme
phenotype of 13 patients from six kindreds. Am J Hum Genet 70:336–348 IM. 1993. Disseminated tuberculosis in interferon gamma gene-disrupted
http://dx.doi.org/10.1086/338625. mice. J Exp Med 178:2243–2247 http://dx.doi.org/10.1084/jem.178.6
295. de Jong R, Altare F, Haagen IA, Elferink DG, Boer T, van Breda .2243.
Vriesman PJ, Kabel PJ, Draaisma JM, van Dissel JT, Kroon FP, Casanova 309. Cooper AM, Magram J, Ferrante J, Orme IM. 1997. Interleukin 12
JL, Ottenhoff TH. 1998. Severe mycobacterial and Salmonella infections (IL-12) is crucial to the development of protective immunity in mice
in interleukin-12 receptor-deficient patients. Science 280:1435–1438 intravenously infected with mycobacterium tuberculosis. J Exp Med 186:
http://dx.doi.org/10.1126/science.280.5368.1435. 39–45 http://dx.doi.org/10.1084/jem.186.1.39.

30 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Immunology of M. tuberculosis Infections

310. Feng CG, Jankovic D, Kullberg M, Cheever A, Scanga CA, Hieny S, Sultana T, Lyons-Weiler J, Reinhart TA, Arcos J, de la Luz Garcia-
Caspar P, Yap GS, Sher A. 2005. Maintenance of pulmonary Th1 effector Hernandez M, Mastrangelo MA, Al-Hammadi N, Townsend R, Balada-
function in chronic tuberculosis requires persistent IL-12 production. Llasat JM, Torrelles JB, Kaplan G, Horne W, Kolls JK, Artyomov MN,
J Immunol 174:4185–4192 http://dx.doi.org/10.4049/jimmunol.174.7 Rangel-Moreno J, Zúñiga J, Khader SA. 2017. Interleukin-17 limits
.4185. hypoxia-inducible factor 1α and development of hypoxic granulomas
311. Redford PS, Boonstra A, Read S, Pitt J, Graham C, Stavropoulos E, during tuberculosis. JCI Insight 2:92973 http://dx.doi.org/10.1172/jci.
Bancroft GJ, O’Garra A. 2010. Enhanced protection to Mycobacterium insight.92973.
tuberculosis infection in IL-10-deficient mice is accompanied by early 324. Khader SA, Bell GK, Pearl JE, Fountain JJ, Rangel-Moreno J, Cilley
and enhanced Th1 responses in the lung. Eur J Immunol 40:2200–2210 GE, Shen F, Eaton SM, Gaffen SL, Swain SL, Locksley RM, Haynes L,
http://dx.doi.org/10.1002/eji.201040433. Randall TD, Cooper AM. 2007. IL-23 and IL-17 in the establishment of
312. Moreira-Teixeira L, Redford PS, Stavropoulos E, Ghilardi N, protective pulmonary CD4+ T cell responses after vaccination and during
Maynard CL, Weaver CT, Freitas do Rosário AP, Wu X, Langhorne J, Mycobacterium tuberculosis challenge. Nat Immunol 8:369–377 http://
O’Garra A. 2017. T cell-derived IL-10 impairs host resistance to Myco- dx.doi.org/10.1038/ni1449.
bacterium tuberculosis infection. J Immunol 199:613–623 http://dx.doi 325. Khader SA, Guglani L, Rangel-Moreno J, Gopal R, Junecko BA,
.org/10.4049/jimmunol.1601340. Fountain JJ, Martino C, Pearl JE, Tighe M, Lin YY, Slight S, Kolls JK,
313. Gerosa F, Nisii C, Righetti S, Micciolo R, Marchesini M, Cazzadori Reinhart TA, Randall TD, Cooper AM. 2011. IL-23 is required for long-
A, Trinchieri G. 1999. CD4(+) T cell clones producing both interferon- term control of Mycobacterium tuberculosis and B cell follicle formation
gamma and interleukin-10 predominate in bronchoalveolar lavages of in the infected lung. J Immunol 187:5402–5407 http://dx.doi.org/10.4049
active pulmonary tuberculosis patients. Clin Immunol 92:224–234 http:// /jimmunol.1101377.
dx.doi.org/10.1006/clim.1999.4752. 326. Nandi B, Behar SM. 2011. Regulation of neutrophils by interferon-γ
314. Saraiva M, Christensen JR, Veldhoen M, Murphy TL, Murphy KM, limits lung inflammation during tuberculosis infection. J Exp Med 208:
O’Garra A. 2009. Interleukin-10 production by Th1 cells requires inter- 2251–2262 http://dx.doi.org/10.1084/jem.20110919.
leukin-12-induced STAT4 transcription factor and ERK MAP kinase ac- 327. Desvignes L, Ernst JD. 2009. Interferon-gamma-responsive non-
tivation by high antigen dose. Immunity 31:209–219 http://dx.doi.org hematopoietic cells regulate the immune response to Mycobacterium
/10.1016/j.immuni.2009.05.012. tuberculosis. Immunity 31:974–985 http://dx.doi.org/10.1016/j.immuni
315. Szabo SJ, Kim ST, Costa GL, Zhang X, Fathman CG, Glimcher .2009.10.007.
LH. 2000. A novel transcription factor, T-bet, directs Th1 lineage com- 328. Green AM, Mattila JT, Bigbee CL, Bongers KS, Lin PL, Flynn JL.
mitment. Cell 100:655–669 http://dx.doi.org/10.1016/S0092-8674(00) 2010. CD4(+) regulatory T cells in a cynomolgus macaque model of
80702-3. Mycobacterium tuberculosis infection. J Infect Dis 202:533–541 http://dx
316. Gallegos AM, van Heijst JW, Samstein M, Su X, Pamer EG, .doi.org/10.1086/654896.
Glickman MS. 2011. A gamma interferon independent mechanism of 329. Geffner L, Basile JI, Yokobori N, Sabio Y García C, Musella R,
CD4 T cell mediated control of M. tuberculosis infection in vivo. PLoS Castagnino J, Sasiain MC, de la Barrera S. 2014. CD4(+) CD25(high)
Pathog 7:e1002052 http://dx.doi.org/10.1371/journal.ppat.1002052. forkhead box protein 3(+) regulatory T lymphocytes suppress interferon-γ
317. Gopal R, Monin L, Slight S, Uche U, Blanchard E, Fallert Junecko and CD107 expression in CD4(+) and CD8(+) T cells from tuberculous
BA, Ramos-Payan R, Stallings CL, Reinhart TA, Kolls JK, Kaushal D, pleural effusions. Clin Exp Immunol 175:235–245 http://dx.doi.org
Nagarajan U, Rangel-Moreno J, Khader SA. 2014. Unexpected role for /10.1111/cei.12227.
IL-17 in protective immunity against hypervirulent Mycobacterium tu- 330. Guyot-Revol V, Innes JA, Hackforth S, Hinks T, Lalvani A. 2006.
berculosis HN878 infection. PLoS Pathog 10:e1004099 http://dx.doi.org Regulatory T cells are expanded in blood and disease sites in patients with
/10.1371/journal.ppat.1004099. tuberculosis. Am J Respir Crit Care Med 173:803–810 http://dx.doi.org
318. Freches D, Korf H, Denis O, Havaux X, Huygen K, Romano M. /10.1164/rccm.200508-1294OC.
2013. Mice genetically inactivated in interleukin-17A receptor are defec- 331. Ribeiro-Rodrigues R, Resende Co T, Rojas R, Toossi Z, Dietze R,
tive in long-term control of Mycobacterium tuberculosis infection. Im- Boom WH, Maciel E, Hirsch CS. 2006. A role for CD4+CD25+ T cells in
munology 140:220–231 http://dx.doi.org/10.1111/imm.12130. regulation of the immune response during human tuberculosis. Clin Exp
319. Okamoto Yoshida Y, Umemura M, Yahagi A, O’Brien RL, Ikuta K, Immunol 144:25–34 http://dx.doi.org/10.1111/j.1365-2249.2006.03027.x.
Kishihara K, Hara H, Nakae S, Iwakura Y, Matsuzaki G. 2010. Essential 332. Chen X, Zhou B, Li M, Deng Q, Wu X, Le X, Wu C, Larmonier N,
role of IL-17A in the formation of a mycobacterial infection-induced gran- Zhang W, Zhang H, Wang H, Katsanis E. 2007. CD4(+)CD25(+)FoxP3
uloma in the lung. J Immunol 184:4414–4422 http://dx.doi.org/10.4049 (+) regulatory T cells suppress Mycobacterium tuberculosis immunity in
/jimmunol.0903332. patients with active disease. Clin Immunol 123:50–59 http://dx.doi.org
320. Wozniak TM, Saunders BM, Ryan AA, Britton WJ. 2010. Myco- /10.1016/j.clim.2006.11.009.
bacterium bovis BCG-specific Th17 cells confer partial protection against 333. Hougardy JM, Place S, Hildebrand M, Drowart A, Debrie AS,
Mycobacterium tuberculosis infection in the absence of gamma interferon. Locht C, Mascart F. 2007. Regulatory T cells depress immune responses
Infect Immun 78:4187–4194 http://dx.doi.org/10.1128/IAI.01392-09. to protective antigens in active tuberculosis. Am J Respir Crit Care Med
321. Scriba TJ, Kalsdorf B, Abrahams DA, Isaacs F, Hofmeister J, Black 176:409–416 http://dx.doi.org/10.1164/rccm.200701-084OC.
G, Hassan HY, Wilkinson RJ, Walzl G, Gelderbloem SJ, Mahomed H, 334. Garg A, Barnes PF, Roy S, Quiroga MF, Wu S, García VE, Krutzik SR,
Hussey GD, Hanekom WA. 2008. Distinct, specific IL-17- and IL-22- Weis SE, Vankayalapati R. 2008. Mannose-capped lipoarabinomannan-
producing CD4+ T cell subsets contribute to the human anti-mycobacterial and prostaglandin E2-dependent expansion of regulatory T cells in human
immune response. J Immunol 180:1962–1970 http://dx.doi.org/10.4049 Mycobacterium tuberculosis infection. Eur J Immunol 38:459–469 http://
/jimmunol.180.3.1962. dx.doi.org/10.1002/eji.200737268.
322. Okada S, et al. 2015. Immunodeficiencies. Impairment of immunity 335. Scott-Browne JP, Shafiani S, Tucker-Heard G, Ishida-Tsubota K,
to Candida and Mycobacterium in humans with bi-allelic RORC muta- Fontenot JD, Rudensky AY, Bevan MJ, Urdahl KB. 2007. Expansion and
tions. Science 349:606–613 http://dx.doi.org/10.1126/science.aaa4282. function of Foxp3-expressing T regulatory cells during tuberculosis. J Exp
323. Domingo-Gonzalez R, Das S, Griffiths KL, Ahmed M, Bambouskova Med 204:2159–2169 http://dx.doi.org/10.1084/jem.20062105.
M, Gopal R, Gondi S, Muñoz-Torrico M, Salazar-Lezama MA, Cruz- 336. Shafiani S, Tucker-Heard G, Kariyone A, Takatsu K, Urdahl KB.
Lagunas A, Jiménez-Álvarez L, Ramirez-Martinez G, Espinosa-Soto R, 2010. Pathogen-specific regulatory T cells delay the arrival of effector

ASMscience.org/MicrobiolSpectrum 31
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Sia and Rengarajan

T cells in the lung during early tuberculosis. J Exp Med 207:1409–1420 class I-dependent cell populations in protection to tuberculosis infection in
http://dx.doi.org/10.1084/jem.20091885. mice. Proc Natl Acad Sci U S A 97:4204–4208 http://dx.doi.org/10.1073
337. Shafiani S, Dinh C, Ertelt JM, Moguche AO, Siddiqui I, Smigiel KS, /pnas.97.8.4204.
Sharma P, Campbell DJ, Way SS, Urdahl KB. 2013. Pathogen-specific 351. van Pinxteren LA, Cassidy JP, Smedegaard BH, Agger EM,
Treg cells expand early during mycobacterium tuberculosis infection but Andersen P. 2000. Control of latent Mycobacterium tuberculosis infection
are later eliminated in response to interleukin-12. Immunity 38:1261– is dependent on CD8 T cells. Eur J Immunol 30:3689–3698 http://dx.doi
1270 http://dx.doi.org/10.1016/j.immuni.2013.06.003. .org/10.1002/1521-4141(200012)30:12<3689::AID-IMMU3689>3.0
338. Shang S, Harton M, Tamayo MH, Shanley C, Palanisamy GS, .CO;2-4.
Caraway M, Chan ED, Basaraba RJ, Orme IM, Ordway DJ. 2011. In- 352. Stenger S, Mazzaccaro RJ, Uyemura K, Cho S, Barnes PF, Rosat JP,
creased Foxp3 expression in guinea pigs infected with W-Beijing strains Sette A, Brenner MB, Porcelli SA, Bloom BR, Modlin RL. 1997. Differ-
of M. tuberculosis. Tuberculosis (Edinb) 91:378–385 http://dx.doi.org ential effects of cytolytic T cell subsets on intracellular infection. Science
/10.1016/j.tube.2011.06.001. 276:1684–1687 http://dx.doi.org/10.1126/science.276.5319.1684.
339. McBride A, Konowich J, Salgame P. 2013. Host defense and re- 353. Stenger S, Hanson DA, Teitelbaum R, Dewan P, Niazi KR, Froelich
cruitment of Foxp3+ T regulatory cells to the lungs in chronic Mycobac- CJ, Ganz T, Thoma-Uszynski S, Melián A, Bogdan C, Porcelli SA, Bloom
terium tuberculosis infection requires toll-like receptor 2. PLoS Pathog 9: BR, Krensky AM, Modlin RL. 1998. An antimicrobial activity of cytolytic
e1003397 http://dx.doi.org/10.1371/journal.ppat.1003397. T cells mediated by granulysin. Science 282:121–125 http://dx.doi.org
340. Rogerson BJ, Jung YJ, LaCourse R, Ryan L, Enright N, North RJ. /10.1126/science.282.5386.121.
2006. Expression levels of Mycobacterium tuberculosis antigen-encoding 354. Ernst WA, Thoma-Uszynski S, Teitelbaum R, Ko C, Hanson DA,
genes versus production levels of antigen-specific T cells during stationary Clayberger C, Krensky AM, Leippe M, Bloom BR, Ganz T, Modlin RL.
level lung infection in mice. Immunology 118:195–201 http://dx.doi.org 2000. Granulysin, a T cell product, kills bacteria by altering membrane
/10.1111/j.1365-2567.2006.02355.x. permeability. J Immunol 165:7102–7108 http://dx.doi.org/10.4049
341. Shi L, North R, Gennaro ML. 2004. Effect of growth state on /jimmunol.165.12.7102.
transcription levels of genes encoding major secreted antigens of Myco- 355. Bruns H, Meinken C, Schauenberg P, Härter G, Kern P, Modlin
bacterium tuberculosis in the mouse lung. Infect Immun 72:2420–2424 RL, Antoni C, Stenger S. 2009. Anti-TNF immunotherapy reduces CD8+
http://dx.doi.org/10.1128/IAI.72.4.2420-2424.2004. T cell-mediated antimicrobial activity against Mycobacterium tuber-
342. Moguche AO, Shafiani S, Clemons C, Larson RP, Dinh C, Higdon culosis in humans. J Clin Invest 119:1167–1177 http://dx.doi.org/10.1172
LE, Cambier CJ, Sissons JR, Gallegos AM, Fink PJ, Urdahl KB. 2015. /JCI38482.
ICOS and Bcl6-dependent pathways maintain a CD4 T cell population 356. Shams H, Klucar P, Weis SE, Lalvani A, Moonan PK, Safi H, Wizel B,
with memory-like properties during tuberculosis. J Exp Med 212:715–728 Ewer K, Nepom GT, Lewinsohn DM, Andersen P, Barnes PF. 2004.
http://dx.doi.org/10.1084/jem.20141518. Characterization of a Mycobacterium tuberculosis peptide that is recog-
343. Moguche AO, Musvosvi M, Penn-Nicholson A, Plumlee CR, Mearns H, nized by human CD4+ and CD8+ T cells in the context of multiple HLA
Geldenhuys H, Smit E, Abrahams D, Rozot V, Dintwe O, Hoff ST, Kromann alleles. J Immunol 173:1966–1977 http://dx.doi.org/10.4049/jimmunol
I, Ruhwald M, Bang P, Larson RP, Shafiani S, Ma S, Sherman DR, Sette A, .173.3.1966.
Lindestam Arlehamn CS, McKinney DM, Maecker H, Hanekom WA, 357. Lalvani A, Brookes R, Wilkinson RJ, Malin AS, Pathan AA,
Hatherill M, Andersen P, Scriba TJ, Urdahl KB. 2017. Antigen availability Andersen P, Dockrell H, Pasvol G, Hill AV. 1998. Human cytolytic and
shapes T cell differentiation and function during tuberculosis. Cell Host Mi- interferon gamma-secreting CD8+ T lymphocytes specific for Mycobac-
crobe 21:695–706.e5 http://dx.doi.org/10.1016/j.chom.2017.05.012. terium tuberculosis. Proc Natl Acad Sci U S A 95:270–275 http://dx.doi
344. Bold TD, Banaei N, Wolf AJ, Ernst JD. 2011. Suboptimal activation .org/10.1073/pnas.95.1.270.
of antigen-specific CD4+ effector cells enables persistence of M. tuber- 358. Pathan AA, Wilkinson KA, Wilkinson RJ, Latif M, McShane H,
culosisin vivo. PLoS Pathog 7:e1002063 http://dx.doi.org/10.1371/journal Pasvol G, Hill AV, Lalvani A. 2000. High frequencies of circulating IFN-
.ppat.1002063. gamma-secreting CD8 cytotoxic T cells specific for a novel MHC class I-
345. Egen JG, Rothfuchs AG, Feng CG, Horwitz MA, Sher A, Germain restricted Mycobacterium tuberculosis epitope in M. tuberculosis-infected
RN. 2011. Intravital imaging reveals limited antigen presentation and subjects without disease. Eur J Immunol 30:2713–2721 http://dx.doi.org
T cell effector function in mycobacterial granulomas. Immunity 34:807– /10.1002/1521-4141(200009)30:9<2713::AID-IMMU2713>3.0.CO;2-4.
819 http://dx.doi.org/10.1016/j.immuni.2011.03.022. 359. Klein MR, Smith SM, Hammond AS, Ogg GS, King AS, Vekemans J,
346. Reiley WW, Shafiani S, Wittmer ST, Tucker-Heard G, Moon JJ, Jaye A, Lukey PT, McAdam KP. 2001. HLA-B*35-restricted CD8 T cell
Jenkins MK, Urdahl KB, Winslow GM, Woodland DL. 2010. Distinct epitopes in the antigen 85 complex of Mycobacterium tuberculosis. J In-
functions of antigen-specific CD4 T cells during murine Mycobacterium fect Dis 183:928–934 http://dx.doi.org/10.1086/319267.
tuberculosis infection. Proc Natl Acad Sci U S A 107:19408–19413 http:// 360. Caccamo N, Meraviglia S, La Mendola C, Guggino G, Dieli F,
dx.doi.org/10.1073/pnas.1006298107. Salerno A. 2006. Phenotypical and functional analysis of memory and
347. Winslow GM, Roberts AD, Blackman MA, Woodland DL. 2003. effector human CD8 T cells specific for mycobacterial antigens. J Immunol
Persistence and turnover of antigen-specific CD4 T cells during chronic 177:1780–1785 http://dx.doi.org/10.4049/jimmunol.177.3.1780.
tuberculosis infection in the mouse. J Immunol 170:2046–2052 http://dx 361. Lewinsohn DA, Winata E, Swarbrick GM, Tanner KE, Cook MS,
.doi.org/10.4049/jimmunol.170.4.2046. Null MD, Cansler ME, Sette A, Sidney J, Lewinsohn DM. 2007. Immu-
348. Aagaard C, Hoang T, Dietrich J, Cardona PJ, Izzo A, Dolganov nodominant tuberculosis CD8 antigens preferentially restricted by HLA-
G, Schoolnik GK, Cassidy JP, Billeskov R, Andersen P. 2011. A multistage B. PLoS Pathog 3:1240–1249 http://dx.doi.org/10.1371/journal.ppat
tuberculosis vaccine that confers efficient protection before and after ex- .0030127.
posure. Nat Med 17:189–194 http://dx.doi.org/10.1038/nm.2285. 362. Lewinsohn DM, Swarbrick GM, Cansler ME, Null MD, Rajaraman
349. Behar SM, Dascher CC, Grusby MJ, Wang CR, Brenner MB. 1999. V, Frieder MM, Sherman DR, McWeeney S, Lewinsohn DA. 2013.
Susceptibility of mice deficient in CD1D or TAP1 to infection with My- Human Mycobacterium tuberculosis CD8 T Cell antigens/epitopes iden-
cobacterium tuberculosis. J Exp Med 189:1973–1980 http://dx.doi.org tified by a proteomic peptide library. PLoS One 8:e67016 http://dx.doi
/10.1084/jem.189.12.1973. .org/10.1371/journal.pone.0067016.
350. Sousa AO, Mazzaccaro RJ, Russell RG, Lee FK, Turner OC, Hong S, 363. Wherry EJ. 2011. T cell exhaustion. Nat Immunol 12:492–499
Van Kaer L, Bloom BR. 2000. Relative contributions of distinct MHC http://dx.doi.org/10.1038/ni.2035.

32 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Immunology of M. tuberculosis Infections

364. Day CL, Kaufmann DE, Kiepiela P, Brown JA, Moodley ES, Reddy S, human macrophages infected with Mycobacterium tuberculosis. J Immu-
Mackey EW, Miller JD, Leslie AJ, DePierres C, Mncube Z, Duraiswamy J, nol 189:5896–5902 http://dx.doi.org/10.4049/jimmunol.1200990.
Zhu B, Eichbaum Q, Altfeld M, Wherry EJ, Coovadia HM, Goulder PJ, 377. Jayaraman P, Jacques MK, Zhu C, Steblenko KM, Stowell BL, Madi
Klenerman P, Ahmed R, Freeman GJ, Walker BD. 2006. PD-1 expression A, Anderson AC, Kuchroo VK, Behar SM. 2016. TIM3 mediates T cell
on HIV-specific T cells is associated with T-cell exhaustion and disease exhaustion during Mycobacterium tuberculosis infection. PLoS Pathog
progression. Nature 443:350–354 http://dx.doi.org/10.1038/nature05115. 12:e1005490 http://dx.doi.org/10.1371/journal.ppat.1005490.
365. Rozot V, Vigano S, Mazza-Stalder J, Idrizi E, Day CL, Perreau M, 378. Qiu Y, Chen J, Liao H, Zhang Y, Wang H, Li S, Luo Y, Fang D, Li G,
Lazor-Blanchet C, Petruccioli E, Hanekom W, Goletti D, Bart PA, Nicod Zhou B, Shen L, Chen CY, Huang D, Cai J, Cao K, Jiang L, Zeng G, Chen
L, Pantaleo G, Harari A. 2013. Mycobacterium tuberculosis-specific ZW. 2012. Tim-3-expressing CD4+ and CD8+ T cells in human tuber-
CD8+ T cells are functionally and phenotypically different between latent culosis (TB) exhibit polarized effector memory phenotypes and stronger
infection and active disease. Eur J Immunol 43:1568–1577 http://dx.doi anti-TB effector functions. PLoS Pathog 8:e1002984 http://dx.doi.org
.org/10.1002/eji.201243262. /10.1371/journal.ppat.1002984.
366. Saharia KK, Petrovas C, Ferrando-Martinez S, Leal M, Luque R, 379. Mahnke YD, Brodie TM, Sallusto F, Roederer M, Lugli E. 2013.
Ive P, Luetkemeyer A, Havlir D, Koup RA. 2016. Tuberculosis therapy The who’s who of T-cell differentiation: human memory T-cell subsets.
modifies the cytokine profile, maturation state, and expression of inhibi- Eur J Immunol 43:2797–2809 http://dx.doi.org/10.1002/eji.201343751.
tory molecules on Mycobacterium tuberculosis-specific CD4+ T-cells. 380. Sallusto F, Lenig D, Förster R, Lipp M, Lanzavecchia A. 1999. Two
PLoS One 11:e0158262 http://dx.doi.org/10.1371/journal.pone.0158262.
subsets of memory T lymphocytes with distinct homing potentials and
367. Hassan SS, Akram M, King EC, Dockrell HM, Cliff JM. 2015. PD-1, effector functions. Nature 401:708–712 http://dx.doi.org/10.1038/44385.
PD-L1 and PD-L2 gene expression on T-cells and natural killer cells 381. Caccamo N, Guggino G, Meraviglia S, Gelsomino G, Di Carlo P,
declines in conjunction with a reduction in PD-1 protein during the in-
Titone L, Bocchino M, Galati D, Matarese A, Nouta J, Klein MR, Salerno
tensive phase of tuberculosis treatment. PLoS One 10:e0137646 http://dx A, Sanduzzi A, Dieli F, Ottenhoff TH. 2009. Analysis of Mycobacterium
.doi.org/10.1371/journal.pone.0137646.
tuberculosis-specific CD8 T-cells in patients with active tuberculosis and
368. Adekambi T, Ibegbu CC, Kalokhe AS, Yu T, Ray SM, Rengarajan J. in individuals with latent infection. PLoS One 4:e5528 http://dx.doi.org
2012. Distinct effector memory CD4+ T cell signatures in latent Myco- /10.1371/journal.pone.0005528.
bacterium tuberculosis infection, BCG vaccination and clinically resolved
382. Griffin JP, Orme IM. 1994. Evolution of CD4 T-cell subsets fol-
tuberculosis. PLoS One 7:e36046 http://dx.doi.org/10.1371/journal.pone
lowing infection of naive and memory immune mice with Mycobacterium
.0036046.
tuberculosis. Infect Immun 62:1683–1690.
369. Singh A, Mohan A, Dey AB, Mitra DK. 2013. Inhibiting the
383. Andersen P, Smedegaard B. 2000. CD4(+) T-cell subsets that mediate
programmed death 1 pathway rescues Mycobacterium tuberculosis-
immunological memory to Mycobacterium tuberculosis infection in mice.
specific interferon γ-producing T cells from apoptosis in patients with
Infect Immun 68:621–629 http://dx.doi.org/10.1128/IAI.68.2.621-629
pulmonary tuberculosis. J Infect Dis 208:603–615 http://dx.doi.org
.2000.
/10.1093/infdis/jit206.
384. Serbina NV, Flynn JL. 2001. CD8(+) T cells participate in the
370. Jurado JO, Alvarez IB, Pasquinelli V, Martínez GJ, Quiroga MF,
memory immune response to Mycobacterium tuberculosis. Infect Immun
Abbate E, Musella RM, Chuluyan HE, García VE. 2008. Programmed
69:4320–4328 http://dx.doi.org/10.1128/IAI.69.7.4320-4328.2001.
death (PD)-1:PD-ligand 1/PD-ligand 2 pathway inhibits T cell effector
functions during human tuberculosis. J Immunol 181:116–125 http://dx 385. Kamath A, Woodworth JS, Behar SM. 2006. Antigen-specific CD8+
.doi.org/10.4049/jimmunol.181.1.116. T cells and the development of central memory during Mycobacterium
tuberculosis infection. J Immunol 177:6361–6369 http://dx.doi.org
371. Govender L, Abel B, Hughes EJ, Scriba TJ, Kagina BM, de Kock M,
/10.4049/jimmunol.177.9.6361.
Walzl G, Black G, Rosenkrands I, Hussey GD, Mahomed H, Andersen P,
Hanekom WA. 2010. Higher human CD4 T cell response to novel My- 386. Hubbard RD, Flory CM, Collins FM. 1991. Memory T cell-mediated
cobacterium tuberculosis latency associated antigens Rv2660 and Rv2659 resistance to Mycobacterium tuberculosis infection in innately susceptible
in latent infection compared with tuberculosis disease. Vaccine 29:51–57 and resistant mice. Infect Immun 59:2012–2016.
http://dx.doi.org/10.1016/j.vaccine.2010.10.022. 387. Andersen P, Heron I. 1993. Specificity of a protective memory im-
372. Day CL, Abrahams DA, Lerumo L, Janse van Rensburg E, Stone L, mune response against Mycobacterium tuberculosis. Infect Immun 61:
O’rie T, Pienaar B, de Kock M, Kaplan G, Mahomed H, Dheda K, 844–851.
Hanekom WA. 2011. Functional capacity of Mycobacterium tuberculosis- 388. Orme IM. 1988. Characteristics and specificity of acquired immu-
specific T cell responses in humans is associated with mycobacterial load. nologic memory to Mycobacterium tuberculosis infection. J Immunol
J Immunol 187:2222–2232 http://dx.doi.org/10.4049/jimmunol.1101122. 140:3589–3593.
373. Lázár-Molnár E, Chen B, Sweeney KA, Wang EJ, Liu W, Lin J, 389. Kamath AB, Behar SM. 2005. Anamnestic responses of mice fol-
Porcelli SA, Almo SC, Nathenson SG, Jacobs WR Jr. 2010. Programmed lowing Mycobacterium tuberculosis infection. Infect Immun 73:6110–
death-1 (PD-1)-deficient mice are extraordinarily sensitive to tuberculosis. 6118 http://dx.doi.org/10.1128/IAI.73.9.6110-6118.2005.
Proc Natl Acad Sci U S A 107:13402–13407 http://dx.doi.org/10.1073 390. Achkar JM, Chan J, Casadevall A. 2015. B cells and antibodies in
/pnas.1007394107. the defense against Mycobacterium tuberculosis infection. Immunol Rev
374. Barber DL, Mayer-Barber KD, Feng CG, Sharpe AH, Sher A. 2011. 264:167–181 http://dx.doi.org/10.1111/imr.12276.
CD4 T cells promote rather than control tuberculosis in the absence of 391. Ulrichs T, Kosmiadi GA, Trusov V, Jörg S, Pradl L, Titukhina M,
PD-1-mediated inhibition. J Immunol 186:1598–1607 http://dx.doi.org Mishenko V, Gushina N, Kaufmann SH. 2004. Human tuberculous
/10.4049/jimmunol.1003304. granulomas induce peripheral lymphoid follicle-like structures to orches-
375. Jayaraman P, Sada-Ovalle I, Beladi S, Anderson AC, Dardalhon V, trate local host defence in the lung. J Pathol 204:217–228 http://dx.doi
Hotta C, Kuchroo VK, Behar SM. 2010. Tim3 binding to galectin-9 .org/10.1002/path.1628.
stimulates antimicrobial immunity. J Exp Med 207:2343–2354 http://dx 392. Tsai MC, Chakravarty S, Zhu G, Xu J, Tanaka K, Koch C,
.doi.org/10.1084/jem.20100687. Tufariello J, Flynn J, Chan J. 2006. Characterization of the tuberculous
376. Sada-Ovalle I, Chávez-Galán L, Torre-Bouscoulet L, Nava-Gamiño granuloma in murine and human lungs: cellular composition and rela-
L, Barrera L, Jayaraman P, Torres-Rojas M, Salazar-Lezama MA, Behar tive tissue oxygen tension. Cell Microbiol 8:218–232 http://dx.doi.org
SM. 2012. The Tim3-galectin 9 pathway induces antibacterial activity in /10.1111/j.1462-5822.2005.00612.x.

ASMscience.org/MicrobiolSpectrum 33
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Sia and Rengarajan

393. Kozakiewicz L, Phuah J, Flynn J, Chan J. 2013. The role of B cells nonpeptidic mycobacterial ligands. Science 264:267–270 http://dx.doi
and humoral immunity in Mycobacterium tuberculosis infection. Adv .org/10.1126/science.8146660.
Exp Med Biol 783:225–250 http://dx.doi.org/10.1007/978-1-4614-6111 409. Haregewoin A, Soman G, Hom RC, Finberg RW. 1989. Human
-1_12. gamma delta+ T cells respond to mycobacterial heat-shock protein. Na-
394. Cliff JM, Lee JS, Constantinou N, Cho JE, Clark TG, Ronacher K, ture 340:309–312 http://dx.doi.org/10.1038/340309a0.
King EC, Lukey PT, Duncan K, Van Helden PD, Walzl G, Dockrell HM. 410. Kabelitz D, Bender A, Schondelmaier S, Schoel B, Kaufmann SH.
2013. Distinct phases of blood gene expression pattern through tubercu- 1990. A large fraction of human peripheral blood gamma/delta + T cells is
losis treatment reflect modulation of the humoral immune response. activated by Mycobacterium tuberculosis but not by its 65-kD heat shock
J Infect Dis 207:18–29 http://dx.doi.org/10.1093/infdis/jis499. protein. J Exp Med 171:667–679 http://dx.doi.org/10.1084/jem.171.3.667.
395. Lyashchenko K, Colangeli R, Houde M, Al Jahdali H, Menzies D, 411. Kabelitz D, Bender A, Prospero T, Wesselborg S, Janssen O,
Gennaro ML. 1998. Heterogeneous antibody responses in tuberculosis. Pechhold K. 1991. The primary response of human gamma/delta + T cells
Infect Immun 66:3936–3940. to Mycobacterium tuberculosis is restricted to V gamma 9-bearing cells.
396. Li H, Wang XX, Wang B, Fu L, Liu G, Lu Y, Cao M, Huang H, J Exp Med 173:1331–1338 http://dx.doi.org/10.1084/jem.173.6.1331.
Javid B. 2017. Latently and uninfected healthcare workers exposed to TB 412. Boom WH, Chervenak KA, Mincek MA, Ellner JJ. 1992. Role of the
make protective antibodies against Mycobacterium tuberculosis. Proc mononuclear phagocyte as an antigen-presenting cell for human gamma
Natl Acad Sci U S A 114:5023–5028 http://dx.doi.org/10.1073/pnas delta T cells activated by live Mycobacterium tuberculosis. Infect Immun
.1611776114. 60:3480–3488.
397. Kunnath-Velayudhan S, Salamon H, Wang HY, Davidow AL, 413. De Libero G, Casorati G, Giachino C, Carbonara C, Migone N,
Molina DM, Huynh VT, Cirillo DM, Michel G, Talbot EA, Perkins MD, Matzinger P, Lanzavecchia A. 1991. Selection by two powerful antigens
Felgner PL, Liang X, Gennaro ML. 2010. Dynamic antibody responses to may account for the presence of the major population of human peripheral
the Mycobacterium tuberculosis proteome. Proc Natl Acad Sci U S A gamma/delta T cells. J Exp Med 173:1311–1322 http://dx.doi.org/10.1084
107:14703–14708 http://dx.doi.org/10.1073/pnas.1009080107. /jem.173.6.1311.
398. Maglione PJ, Xu J, Chan J. 2007. B cells moderate inflamma- 414. Dieli F, Troye-Blomberg M, Ivanyi J, Fournié JJ, Bonneville M,
tory progression and enhance bacterial containment upon pulmonary Peyrat MA, Sireci G, Salerno A. 2000. Vgamma9/Vdelta2 T lympho-
challenge with Mycobacterium tuberculosis. J Immunol 178:7222–7234 cytes reduce the viability of intracellular Mycobacterium tuberculosis. Eur
http://dx.doi.org/10.4049/jimmunol.178.11.7222. J Immunol 30:1512–1519 http://dx.doi.org/10.1002/(SICI)1521-4141
399. Kozakiewicz L, Chen Y, Xu J, Wang Y, Dunussi-Joannopoulos K, (200005)30:5<1512::AID-IMMU1512>3.0.CO;2-3.
Ou Q, Flynn JL, Porcelli SA, Jacobs WR Jr, Chan J. 2013. B cells regu- 415. Abate G, Spencer CT, Hamzabegovic F, Blazevic A, Xia M, Hoft
late neutrophilia during Mycobacterium tuberculosis infection and BCG DF. 2015. Mycobacterium-specific γ9δ2 T cells mediate both pathogen-
vaccination by modulating the interleukin-17 response. PLoS Pathog 9: inhibitory and CD40 ligand-dependent antigen presentation effects im-
e1003472 http://dx.doi.org/10.1371/journal.ppat.1003472. portant for tuberculosis immunity. Infect Immun 84:580–589 http://dx
400. Maglione PJ, Chan J. 2009. How B cells shape the immune response .doi.org/10.1128/IAI.01262-15.
against Mycobacterium tuberculosis. Eur J Immunol 39:676–686 http:// 416. Panchamoorthy G, McLean J, Modlin RL, Morita CT, Ishikawa S,
dx.doi.org/10.1002/eji.200839148. Brenner MB, Band H. 1991. A predominance of the T cell receptor V
401. Maglione PJ, Xu J, Casadevall A, Chan J. 2008. Fc gamma receptors gamma 2/V delta 2 subset in human mycobacteria-responsive T cells
regulate immune activation and susceptibility during Mycobacterium tu- suggests germline gene encoded recognition. J Immunol 147:3360–3369.
berculosis infection. J Immunol 180:3329–3338 http://dx.doi.org/10.4049 417. Chen CY, Yao S, Huang D, Wei H, Sicard H, Zeng G, Jomaa H,
/jimmunol.180.5.3329. Larsen MH, Jacobs WR Jr, Wang R, Letvin N, Shen Y, Qiu L, Shen L,
402. Benard A, Sakwa I, Schierloh P, Colom A, Mercier I, Tailleux L, Chen ZW. 2013. Phosphoantigen/IL2 expansion and differentiation of
Jouneau L, Boudinot P, Al-Saati T, Lang R, Rehwinkel J, Loxton AG, Vγ2Vδ2 T cells increase resistance to tuberculosis in nonhuman primates.
Kaufmann SH, Anton-Leberre V, O’Garra A, Del Carmen Sasiain M, PLoS Pathog 9:e1003501 http://dx.doi.org/10.1371/journal.ppat.1003501.
Gicquel B, Fillatreau S, Neyrolles O, Hudrisier D. 2017. B cells producing 418. Qaqish A, Huang D, Chen CY, Zhang Z, Wang R, Li S, Yang E,
type I interferon modulate macrophage polarization in tuberculosis. Am J Lu Y, Larsen MH, Jacobs WR Jr, Qian L, Frencher J, Shen L, Chen
Respir Crit Care Med 197:801–813. ZW. 2017. Adoptive transfer of phosphoantigen-specific γδ T cell subset
403. Carding SR, Egan PJ. 2002. Gammadelta T cells: functional plasticity attenuates Mycobacterium tuberculosis infection in nonhuman primates. J
and heterogeneity. Nat Rev Immunol 2:336–345 http://dx.doi.org Immunol 198:4753–4763 http://dx.doi.org/10.4049/jimmunol.1602019.
/10.1038/nri797. 419. Dieli F, Troye-Blomberg M, Ivanyi J, Fournié JJ, Krensky AM,
404. Bonneville M, O’Brien RL, Born WK. 2010. Gammadelta T cell Bonneville M, Peyrat MA, Caccamo N, Sireci G, Salerno A. 2001.
effector functions: a blend of innate programming and acquired plasticity. Granulysin-dependent killing of intracellular and extracellular Mycobac-
Nat Rev Immunol 10:467–478 http://dx.doi.org/10.1038/nri2781. terium tuberculosis by Vgamma9/Vdelta2 T lymphocytes. J Infect Dis
405. Havlir DV, Ellner JJ, Chervenak KA, Boom WH. 1991. Selective 184:1082–1085 http://dx.doi.org/10.1086/323600.
expansion of human gamma delta T cells by monocytes infected with live 420. Spencer CT, Abate G, Sakala IG, Xia M, Truscott SM, Eickhoff CS,
Mycobacterium tuberculosis. J Clin Invest 87:729–733 http://dx.doi.org Linn R, Blazevic A, Metkar SS, Peng G, Froelich CJ, Hoft DF. 2013.
/10.1172/JCI115053. Granzyme A produced by γ(9)δ(2) T cells induces human macrophages to
406. Tanaka Y, Sano S, Nieves E, De Libero G, Rosa D, Modlin inhibit growth of an intracellular pathogen. PLoS Pathog 9:e1003119
RL, Brenner MB, Bloom BR, Morita CT. 1994. Nonpeptide ligands for http://dx.doi.org/10.1371/journal.ppat.1003119.
human gamma delta T cells. Proc Natl Acad Sci U S A 91:8175–8179 421. Meraviglia S, Caccamo N, Salerno A, Sireci G, Dieli F. 2010. Partial
http://dx.doi.org/10.1073/pnas.91.17.8175. and ineffective activation of V gamma 9V delta 2 T cells by Mycobacte-
407. Tanaka Y, Morita CT, Tanaka Y, Nieves E, Brenner MB, Bloom rium tuberculosis-infected dendritic cells. J Immunol 185:1770–1776
BR. 1995. Natural and synthetic non-peptide antigens recognized by hu- http://dx.doi.org/10.4049/jimmunol.1000966.
man gamma delta T cells. Nature 375:155–158 http://dx.doi.org/10.1038 422. Janis EM, Kaufmann SH, Schwartz RH, Pardoll DM. 1989. Acti-
/375155a0. vation of gamma delta T cells in the primary immune response to Myco-
408. Constant P, Davodeau F, Peyrat MA, Poquet Y, Puzo G, Bonneville bacterium tuberculosis. Science 244:713–716 http://dx.doi.org/10.1126
M, Fournié JJ. 1994. Stimulation of human gamma delta T cells by /science.2524098.

34 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Immunology of M. tuberculosis Infections

423. Lockhart E, Green AM, Flynn JL. 2006. IL-17 production is domi- 437. Gold MC, Lewinsohn DM. 2013. Co-dependents: MR1-restricted
nated by gammadelta T cells rather than CD4 T cells during Mycobac- MAIT cells and their antimicrobial function. Nat Rev Microbiol 11:14–19
terium tuberculosis infection. J Immunol 177:4662–4669 http://dx.doi http://dx.doi.org/10.1038/nrmicro2918.
.org/10.4049/jimmunol.177.7.4662. 438. Le Bourhis L, Mburu YK, Lantz O. 2013. MAIT cells, surveyors of a
424. Strominger JL. 2010. An alternative path for antigen presenta- new class of antigen: development and functions. Curr Opin Immunol
tion: group 1 CD1 proteins. J Immunol 184:3303–3305 http://dx.doi.org 25:174–180 http://dx.doi.org/10.1016/j.coi.2013.01.005.
/10.4049/jimmunol.1090008. 439. Treiner E, Duban L, Bahram S, Radosavljevic M, Wanner V, Tilloy
425. Moody DB, Ulrichs T, Mühlecker W, Young DC, Gurcha SS, Grant F, Affaticati P, Gilfillan S, Lantz O. 2003. Selection of evolutionarily
E, Rosat JP, Brenner MB, Costello CE, Besra GS, Porcelli SA. 2000. conserved mucosal-associated invariant T cells by MR1. Nature 422:164–
CD1c-mediated T-cell recognition of isoprenoid glycolipids in Myco- 169 http://dx.doi.org/10.1038/nature01433.
bacterium tuberculosis infection. Nature 404:884–888 http://dx.doi.org 440. Kjer-Nielsen L, Patel O, Corbett AJ, Le Nours J, Meehan B, Liu L,
/10.1038/35009119. Bhati M, Chen Z, Kostenko L, Reantragoon R, Williamson NA, Purcell
426. Ulrichs T, Moody DB, Grant E, Kaufmann SH, Porcelli SA. 2003. AW, Dudek NL, McConville MJ, O’Hair RA, Khairallah GN, Godfrey
T-cell responses to CD1-presented lipid antigens in humans with Myco- DI, Fairlie DP, Rossjohn J, McCluskey J. 2012. MR1 presents microbial
bacterium tuberculosis infection. Infect Immun 71:3076–3087 http://dx vitamin B metabolites to MAIT cells. Nature 491:717–723 http://dx.doi
.doi.org/10.1128/IAI.71.6.3076-3087.2003. .org/10.1038/nature11605.
427. Gilleron M, Stenger S, Mazorra Z, Wittke F, Mariotti S, Böhmer G, 441. Dusseaux M, Martin E, Serriari N, Péguillet I, Premel V, Louis D,
Prandi J, Mori L, Puzo G, De Libero G. 2004. Diacylated sulfoglycolipids Milder M, Le Bourhis L, Soudais C, Treiner E, Lantz O. 2011. Human
are novel mycobacterial antigens stimulating CD1-restricted T cells during MAIT cells are xenobiotic-resistant, tissue-targeted, CD161hi IL-17-
infection with Mycobacterium tuberculosis. J Exp Med 199:649–659 secreting T cells. Blood 117:1250–1259 http://dx.doi.org/10.1182/blood
http://dx.doi.org/10.1084/jem.20031097. -2010-08-303339.
428. Layre E, Collmann A, Bastian M, Mariotti S, Czaplicki J, Prandi J, 442. Gold MC, Cerri S, Smyk-Pearson S, Cansler ME, Vogt TM, Delepine
Mori L, Stenger S, De Libero G, Puzo G, Gilleron M. 2009. Mycolic J, Winata E, Swarbrick GM, Chua WJ, Yu YY, Lantz O, Cook MS, Null
acids constitute a scaffold for mycobacterial lipid antigens stimulating MD, Jacoby DB, Harriff MJ, Lewinsohn DA, Hansen TH, Lewinsohn
CD1-restricted T cells. Chem Biol 16:82–92 http://dx.doi.org/10.1016/j DM. 2010. Human mucosal associated invariant T cells detect bacterially
.chembiol.2008.11.008. infected cells. PLoS Biol 8:e1000407 http://dx.doi.org/10.1371/journal
429. Kasmar AG, van Rhijn I, Cheng TY, Turner M, Seshadri C, Schiefner .pbio.1000407.
A, Kalathur RC, Annand JW, de Jong A, Shires J, Leon L, Brenner M, 443. Gold MC, Eid T, Smyk-Pearson S, Eberling Y, Swarbrick GM,
Wilson IA, Altman JD, Moody DB. 2011. CD1b tetramers bind αβ T cell Langley SM, Streeter PR, Lewinsohn DA, Lewinsohn DM. 2013. Human
receptors to identify a mycobacterial glycolipid-reactive T cell repertoire thymic MR1-restricted MAIT cells are innate pathogen-reactive effectors
in humans. J Exp Med 208:1741–1747 http://dx.doi.org/10.1084/jem that adapt following thymic egress. Mucosal Immunol 6:35–44 http://dx
.20110665. .doi.org/10.1038/mi.2012.45.
430. Montamat-Sicotte DJ, Millington KA, Willcox CR, Hingley-Wilson 444. Prideaux B, Via LE, Zimmerman MD, Eum S, Sarathy J, O’Brien P,
S, Hackforth S, Innes J, Kon OM, Lammas DA, Minnikin DE, Besra GS, Chen C, Kaya F, Weiner DM, Chen PY, Song T, Lee M, Shim TS, Cho JS,
Willcox BE, Lalvani A. 2011. A mycolic acid-specific CD1-restricted T cell Kim W, Cho SN, Olivier KN, Barry CE III, Dartois V. 2015. The asso-
population contributes to acute and memory immune responses in human ciation between sterilizing activity and drug distribution into tuberculosis
tuberculosis infection. J Clin Invest 121:2493–2503 http://dx.doi.org lesions. Nat Med 21:1223–1227 http://dx.doi.org/10.1038/nm.3937.
/10.1172/JCI46216. 445. Ramakrishnan L. 2012. Revisiting the role of the granuloma in tu-
431. Ly D, Kasmar AG, Cheng TY, de Jong A, Huang S, Roy S, Bhatt A, berculosis. Nat Rev Immunol 12:352–366 http://dx.doi.org/10.1038
van Summeren RP, Altman JD, Jacobs WR Jr, Adams EJ, Minnaard AJ, /nri3211.
Porcelli SA, Moody DB. 2013. CD1c tetramers detect ex vivo T cell 446. Cadena AM, Fortune SM, Flynn JL. 2017. Heterogeneity in tuber-
responses to processed phosphomycoketide antigens. J Exp Med 210:729– culosis. Nat Rev Immunol 17:691–702 http://dx.doi.org/10.1038/nri
741 http://dx.doi.org/10.1084/jem.20120624. .2017.69.
432. Seshadri C, Turner MT, Lewinsohn DM, Moody DB, Van Rhijn I. 447. Russell DG, Cardona PJ, Kim MJ, Allain S, Altare F. 2009. Foamy
2013. Lipoproteins are major targets of the polyclonal human T cell re- macrophages and the progression of the human tuberculosis granuloma.
sponse to Mycobacterium tuberculosis. J Immunol 190:278–284 http:// Nat Immunol 10:943–948 http://dx.doi.org/10.1038/ni.1781.
dx.doi.org/10.4049/jimmunol.1201667. 448. Via LE, Lin PL, Ray SM, Carrillo J, Allen SS, Eum SY, Taylor K,
433. Beckman EM, Porcelli SA, Morita CT, Behar SM, Furlong Klein E, Manjunatha U, Gonzales J, Lee EG, Park SK, Raleigh JA, Cho
ST, Brenner MB. 1994. Recognition of a lipid antigen by CD1-restricted SN, McMurray DN, Flynn JL, Barry CE III. 2008. Tuberculous granu-
alpha beta+ T cells. Nature 372:691–694 http://dx.doi.org/10.1038/3726 lomas are hypoxic in guinea pigs, rabbits, and nonhuman primates. Infect
91a0. Immun 76:2333–2340 http://dx.doi.org/10.1128/IAI.01515-07.
434. Sieling PA, Ochoa MT, Jullien D, Leslie DS, Sabet S, Rosat JP, 449. Volkman HE, Clay H, Beery D, Chang JCW, Sherman DR,
Burdick AE, Rea TH, Brenner MB, Porcelli SA, Modlin RL. 2000. Evi- Ramakrishnan L. 2004. Tuberculous granuloma formation is enhanced
dence for human CD4+ T cells in the CD1-restricted repertoire: derivation by a mycobacterium virulence determinant. PLoS Biol 2:e367 http://dx
of mycobacteria-reactive T cells from leprosy lesions. J Immunol 164: .doi.org/10.1371/journal.pbio.0020367.
4790–4796 http://dx.doi.org/10.4049/jimmunol.164.9.4790. 450. Davis JM, Ramakrishnan L. 2009. The role of the granuloma in
435. Moody DB, Young DC, Cheng TY, Rosat JP, Roura-Mir C, expansion and dissemination of early tuberculous infection. Cell 136:37–
O’Connor PB, Zajonc DM, Walz A, Miller MJ, Levery SB, Wilson IA, 49 http://dx.doi.org/10.1016/j.cell.2008.11.014.
Costello CE, Brenner MB. 2004. T cell activation by lipopeptide antigens. 451. Cambier CJ, Takaki KK, Larson RP, Hernandez RE, Tobin DM,
Science 303:527–531 http://dx.doi.org/10.1126/science.1089353. Urdahl KB, Cosma CL, Ramakrishnan L. 2014. Mycobacteria manipulate
436. Martin E, Treiner E, Duban L, Guerri L, Laude H, Toly C, Premel V, macrophage recruitment through coordinated use of membrane lipids.
Devys A, Moura IC, Tilloy F, Cherif S, Vera G, Latour S, Soudais C, Lantz Nature 505:218–222 http://dx.doi.org/10.1038/nature12799.
O. 2009. Stepwise development of MAIT cells in mouse and human. PLoS 452. Cambier CJ, O’Leary SM, O’Sullivan MP, Keane J, Ramakrishnan
Biol 7:e54 http://dx.doi.org/10.1371/journal.pbio.1000054. L. 2017. Phenolic glycolipid facilitates mycobacterial escape from

ASMscience.org/MicrobiolSpectrum 35
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Sia and Rengarajan

microbicidal tissue-resident macrophages. Immunity 47:552–565.e4 tuberculosis. Meta-analysis of the published literature. JAMA 271:698–
http://dx.doi.org/10.1016/j.immuni.2017.08.003. 702 http://dx.doi.org/10.1001/jama.1994.03510330076038.
453. Gideon HP, Phuah J, Myers AJ, Bryson BD, Rodgers MA, Coleman 467. Kagina BM, Abel B, Scriba TJ, Hughes EJ, Keyser A, Soares A,
MT, Maiello P, Rutledge T, Marino S, Fortune SM, Kirschner DE, Lin PL, Gamieldien H, Sidibana M, Hatherill M, Gelderbloem S, Mahomed H,
Flynn JL. 2015. Variability in tuberculosis granuloma T cell responses Hawkridge A, Hussey G, Kaplan G, Hanekom WA, other members of the
exists, but a balance of pro- and anti-inflammatory cytokines is associated South African Tuberculosis Vaccine Initiative. 2010. Specific T cell fre-
with sterilization. PLoS Pathog 11:e1004603 http://dx.doi.org/10.1371 quency and cytokine expression profile do not correlate with protection
/journal.ppat.1004603. against tuberculosis after bacillus Calmette-Guérin vaccination of new-
454. Marakalala MJ, Raju RM, Sharma K, Zhang YJ, Eugenin EA, borns. Am J Respir Crit Care Med 182:1073–1079 http://dx.doi.org
Prideaux B, Daudelin IB, Chen PY, Booty MG, Kim JH, Eum SY, Via LE, /10.1164/rccm.201003-0334OC.
Behar SM, Barry CE III, Mann M, Dartois V, Rubin EJ. 2016. Inflam- 468. Gillard P, Yang PC, Danilovits M, Su WJ, Cheng SL, Pehme L,
matory signaling in human tuberculosis granulomas is spatially organized. Bollaerts A, Jongert E, Moris P, Ofori-Anyinam O, Demoitié MA, Castro
Nat Med 22:531–538 http://dx.doi.org/10.1038/nm.4073. M. 2016. Safety and immunogenicity of the M72/AS01E candidate tu-
455. Helb D, Jones M, Story E, Boehme C, Wallace E, Ho K, Kop J, berculosis vaccine in adults with tuberculosis: A phase II randomised
Owens MR, Rodgers R, Banada P, Safi H, Blakemore R, Lan NT, Jones- study. Tuberculosis (Edinb) 100:118–127 http://dx.doi.org/10.1016/j.tube
López EC, Levi M, Burday M, Ayakaka I, Mugerwa RD, McMillan B, .2016.07.005.
Winn-Deen E, Christel L, Dailey P, Perkins MD, Persing DH, Alland D. 469. Geldenhuys H, Mearns H, Miles DJ, Tameris M, Hokey D, Shi Z,
2010. Rapid detection of Mycobacterium tuberculosis and rifampin re- Bennett S, Andersen P, Kromann I, Hoff ST, Hanekom WA, Mahomed H,
sistance by use of on-demand, near-patient technology. J Clin Microbiol Hatherill M, Scriba TJ, van Rooyen M, Bruce McClain J, Ryall R, de
48:229–237 http://dx.doi.org/10.1128/JCM.01463-09. Bruyn G, H4:IC31 Trial Study Group. 2015. The tuberculosis vaccine H4:
456. Stevens WS, Scott L, Noble L, Gous N, Dheda K. 2017. Impact of IC31 is safe and induces a persistent polyfunctional CD4 T cell response in
the GeneXpert MTB/RIF technology on tuberculosis control. Microbiol South African adults: a randomized controlled trial. Vaccine 33:3592–
Spectr 5:10.1128/microbiolspec.TBTB2-0040-2016 http://dx.doi.org 3599 http://dx.doi.org/10.1016/j.vaccine.2015.05.036.
/10.1128/microbiolspec.TBTB2-0040-2016. 470. Luabeya AK, Kagina BM, Tameris MD, Geldenhuys H, Hoff ST, Shi
457. Mazurek GH, Jereb J, Vernon A, LoBue P, Goldberg S, Castro K, Z, Kromann I, Hatherill M, Mahomed H, Hanekom WA, Andersen P,
Committee IE, IGRA Expert Committee, Centers for Disease Control and Scriba TJ, Schoeman E, Krohn C, Day CL, Africa H, Makhethe L, Smit E,
Prevention (CDC). 2010. Updated guidelines for using interferon gamma Brown Y, Suliman S, Hughes EJ, Bang P, Snowden MA, McClain B,
release assays to detect Mycobacterium tuberculosis infection: United Hussey GD, H56-032 Trial Study Group. 2015. First-in-human trial of
States, 2010. MMWR Recomm Rep 59(RR-5):1–25. the post-exposure tuberculosis vaccine H56:IC31 in Mycobacterium tu-
berculosis infected and non-infected healthy adults. Vaccine 33:4130–
458. Lawn SD, Kerkhoff AD, Vogt M, Wood R. 2012. Diagnostic accu-
racy of a low-cost, urine antigen, point-of-care screening assay for HIV- 4140 http://dx.doi.org/10.1016/j.vaccine.2015.06.051.
associated pulmonary tuberculosis before antiretroviral therapy: a de- 471. Baldwin SL, Reese VA, Huang PW, Beebe EA, Podell BK, Reed SG,
scriptive study. Lancet Infect Dis 12:201–209 http://dx.doi.org/10.1016 Coler RN. 2015. Protection and long-lived immunity induced by the ID93/
/S1473-3099(11)70251-1. GLA-SE vaccine candidate against a clinical Mycobacterium tuberculosis
isolate. Clin Vaccine Immunol 23:137–147 http://dx.doi.org/10.1128
459. Paris L, Magni R, Zaidi F, Araujo R, Saini N, Harpole M, Coronel J,
/CVI.00458-15.
Kirwan DE, Steinberg H, Gilman RH, Petricoin EF III, Nisini R, Luchini
A, Liotta L. 2017. Urine lipoarabinomannan glycan in HIV-negative 472. Tameris MD, Hatherill M, Landry BS, Scriba TJ, Snowden MA,
patients with pulmonary tuberculosis correlates with disease severity. Sci Lockhart S, Shea JE, McClain JB, Hussey GD, Hanekom WA, Mahomed
Transl Med 9:9 http://dx.doi.org/10.1126/scitranslmed.aal2807. H, McShane H, MVA85A 020 Trial Study Team. 2013. Safety and effi-
460. Adekambi T, Ibegbu CC, Cagle S, Kalokhe AS, Wang YF, Hu Y, cacy of MVA85A, a new tuberculosis vaccine, in infants previously vac-
cinated with BCG: a randomised, placebo-controlled phase 2b trial. Lancet
Day CL, Ray SM, Rengarajan J. 2015. Biomarkers on patient T cells
diagnose active tuberculosis and monitor treatment response. J Clin Invest 381:1021–1028 http://dx.doi.org/10.1016/S0140-6736(13)60177-4.
125:1827–1838 http://dx.doi.org/10.1172/JCI77990. 473. Smaill F, Jeyanathan M, Smieja M, Medina MF, Thanthrige-Don N,
Zganiacz A, Yin C, Heriazon A, Damjanovic D, Puri L, Hamid J, Xie F,
461. Riou C, Berkowitz N, Goliath R, Burgers WA, Wilkinson RJ. 2017.
Analysis of the phenotype of Mycobacterium tuberculosis-specific CD4+ Foley R, Bramson J, Gauldie J, Xing Z. 2013. A human type 5 adenovirus-
based tuberculosis vaccine induces robust T cell responses in humans
T cells to discriminate latent from active tuberculosis in HIV-uninfected
and HIV-infected individuals. Front Immunol 8:968 http://dx.doi.org despite preexisting anti-adenovirus immunity. Sci Transl Med 5:205ra134
http://dx.doi.org/10.1126/scitranslmed.3006843.
/10.3389/fimmu.2017.00968.
474. Stylianou E, Griffiths KL, Poyntz HC, Harrington-Kandt R, Dicks
462. Wilkinson KA, Oni T, Gideon HP, Goliath R, Wilkinson RJ, Riou C.
2016. Activation profile of Mycobacterium tuberculosis-specific CD4(+) MD, Stockdale L, Betts G, McShane H. 2015. Improvement of BCG
protective efficacy with a novel chimpanzee adenovirus and a modified
T cells reflects disease activity irrespective of HIV status. Am J Respir
vaccinia Ankara virus both expressing Ag85A. Vaccine 33:6800–6808
Crit Care Med 193:1307–1310 http://dx.doi.org/10.1164/rccm.201601
-0116LE. http://dx.doi.org/10.1016/j.vaccine.2015.10.017.
475. Minhinnick A, Satti I, Harris S, Wilkie M, Sheehan S, Stockdale L,
463. Calmette A. 1931. Preventive vaccination against tuberculosis with
Manjaly Thomas ZR, Lopez-Ramon R, Poulton I, Lawrie A, Vermaak S,
BCG. Proc R Soc Med 24:1481–1490.
Le Vert A, Del Campo J, Hill F, Moss P, McShane H. 2016. A first-
464. Behr MA, Wilson MA, Gill WP, Salamon H, Schoolnik GK, in-human phase 1 trial to evaluate the safety and immunogenicity of
Rane S, Small PM. 1999. Comparative genomics of BCG vaccines by the candidate tuberculosis vaccine MVA85A-IMX313, administered to
whole-genome DNA microarray. Science 284:1520–1523 http://dx.doi BCG-vaccinated adults. Vaccine 34:1412–1421 http://dx.doi.org/10.1016
.org/10.1126/science.284.5419.1520. /j.vaccine.2016.01.062.
465. Rodrigues LC, Diwan VK, Wheeler JG. 1993. Protective effect of BCG 476. Hansen SG, Zak DE, Xu G, Ford JC, Marshall EE, Malouli D,
against tuberculous meningitis and miliary tuberculosis: a meta-analysis. Gilbride RM, Hughes CM, Ventura AB, Ainslie E, Randall KT, Selseth
Int J Epidemiol 22:1154–1158 http://dx.doi.org/10.1093/ije/22.6.1154. AN, Rundstrom P, Herlache L, Lewis MS, Park H, Planer SL, Turner JM,
466. Colditz GA, Brewer TF, Berkey CS, Wilson ME, Burdick E, Fineberg Fischer M, Armstrong C, Zweig RC, Valvo J, Braun JM, Shankar S, Lu L,
HV, Mosteller F. 1994. Efficacy of BCG vaccine in the prevention of Sylwester AW, Legasse AW, Messerle M, Jarvis MA, Amon LM, Aderem

36 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28
Immunology of M. tuberculosis Infections

A, Alter G, Laddy DJ, Stone M, Bonavia A, Evans TG, Axthelm MK, Früh efficacy against Mycobacterium tuberculosis in newborn mice. Tubercu-
K, Edlefsen PT, Picker LJ. 2018. Prevention of tuberculosis in rhesus losis (Edinb) 96:71–74 http://dx.doi.org/10.1016/j.tube.2015.10.010.
macaques by a cytomegalovirus-based vaccine. Nat Med 24:130–143 480. Cardona PJ. 2006. RUTI: a new chance to shorten the treatment of
http://dx.doi.org/10.1038/nm.4473. latent tuberculosis infection. Tuberculosis (Edinb) 86:273–289 http://dx
477. Lahey T, Laddy D, Hill K, Schaeffer J, Hogg A, Keeble J, Dagg B, .doi.org/10.1016/j.tube.2006.01.024.
Ho MM, Arbeit RD, von Reyn CF. 2016. Immunogenicity and protective 481. Vilaplana C, Montané E, Pinto S, Barriocanal AM, Domenech G,
efficacy of the DAR-901 booster vaccine in a murine model of tuber- Torres F, Cardona PJ, Costa J. 2010. Double-blind, randomized, placebo-
culosis. PLoS One 11:e0168521 http://dx.doi.org/10.1371/journal.pone controlled Phase I clinical trial of the therapeutical antituberculous vaccine
.0168521. RUTI. Vaccine 28:1106–1116 http://dx.doi.org/10.1016/j.vaccine.2009
478. Grode L, Ganoza CA, Brohm C, Weiner J III, Eisele B, Kaufmann .09.134.
SH. 2013. Safety and immunogenicity of the recombinant BCG vaccine 482. Nell AS, D’lom E, Bouic P, Sabaté M, Bosser R, Picas J, Amat M,
VPM1002 in a phase 1 open-label randomized clinical trial. Vaccine Churchyard G, Cardona PJ. 2014. Safety, tolerability, and immunogenicity
31:1340–1348 http://dx.doi.org/10.1016/j.vaccine.2012.12.053. of the novel antituberculous vaccine RUTI: randomized, placebo-controlled
479. Aguilo N, Uranga S, Marinova D, Monzon M, Badiola J, Martin C. phase II clinical trial in patients with latent tuberculosis infection. PLoS
2016. MTBVAC vaccine is safe, immunogenic and confers protective One 9:e89612 http://dx.doi.org/10.1371/journal.pone.0089612.

ASMscience.org/MicrobiolSpectrum 37
Downloaded from www.asmscience.org by
IP: 128.122.230.148
On: Wed, 17 Jul 2019 04:48:28

You might also like