ArticlePDF AvailableLiterature Review

Th1 and Th17 Cells in Tuberculosis: Protection, Pathology, and Biomarkers

Wiley
Mediators of Inflammation
Authors:
  • Koltzov Institute of Developmental Biology of Russian Academy of Science

Abstract

The outcome of Mycobacterium tuberculosis (Mtb) infection ranges from a complete pathogen clearance through asymptomatic latent infection (LTBI) to active tuberculosis (TB) disease. It is now understood that LTBI and active TB represent a continuous spectrum of states with different degrees of pathogen “activity,” host pathology, and immune reactivity. Therefore, it is important to differentiate LTBI and active TB and identify active TB stages. CD4+ T cells play critical role during Mtb infection by mediating protection, contributing to inflammation, and regulating immune response. Th1 and Th17 cells are the main effector CD4+ T cells during TB. Th1 cells have been shown to contribute to TB protection by secreting IFN-γ and activating antimycobacterial action in macrophages. Th17 induce neutrophilic inflammation, mediate tissue damage, and thus have been implicated in TB pathology. In recent years new findings have accumulated that alter our view on the role of Th1 and Th17 cells during Mtb infection. This review discusses these new results and how they can be implemented for TB diagnosis and monitoring.
Review Article
Th1 and Th17 Cells in Tuberculosis:
Protection, Pathology, and Biomarkers
I. V. Lyadova and A. V. Panteleev
Immunology Department, Central Tuberculosis Research Institute, Yauza Alley 2, Moscow 107564, Russia
Correspondence should be addressed to I. V. Lyadova; ivlyadova@mail.ru
Received 7 August 2015; Accepted 11 October 2015
Academic Editor: Shen-An Hwang
Copyright © 2015 I. V. Lyadova and A. V. Panteleev. is is an open access article distributed under the Creative Commons
Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is
properly cited.
e outcome of Mycobacterium tuberculosis (Mtb) infection ranges from a complete pathogen clearance through asymptomatic
latent infection (LTBI) to active tuberculosis (TB) disease. It is now understood that LTBI and active TB represent a continuous
spectrum of states with dierent degrees of pathogen “activity,” host pathology, and immune reactivity. erefore, it is important
to dierentiate LTBI and active TB and identify active TB stages. CD4+T cells play critical role during Mtb infection by mediating
protection, contributing to inammation, and regulating immune response. 1 and 17 cells are the main eector CD4+Tcells
during TB. 1 cells have been shown to contribute to TB protection by secreting IFN-𝛾and activating antimycobacterial action in
macrophages. 17 induce neutrophilic inammation, mediate tissue damage, and thus have been implicated in TB pathology. In
recent years new ndings have accumulated that alter our view on the role of 1 and 17 cells during Mtb infection. is review
discusses these new results and how they can be implemented for TB diagnosis and monitoring.
1. Introduction
Tuberculosis (TB) is one of the most common infections in
the world. Infection with Mycobacterium tuberculosis (Mtb)
most oen aects the lungs. e outcome of the pulmonary
infection is divergent and can range from the complete
pathogen clearance through asymptomatic latent infection
(LTBI) to active TB disease.
LTBI has long been considered as a uniform state char-
acterized by the immunological sensitization in the absence
of clinical signs and symptoms of active TB disease. Recently
it has become clear that LTBI includes a broad spectrum of
states that dier by the degree of host immune resistance,
inammatory markers, and pathogen replication (i.e., by
LTBI “activity”) [1].
e characteristics of the TB disease are in turn very
diverse. ey dier by the type of pathology developed
in the lungs (e.g., tuberculoma, cavitary TB, and caseous
pneumonia), the size of the aected lung tissue, the rate of
disease progression, the characteristics of immune activation
and inammation, and Mtb load and replication (i.e., the
presence, quantity, and replication activity of Mtb in the
sputum). Overall, the outcome of Mtb infection is not
a simple two-state distribution which includes LTBI and
active TB but represents a continuous spectrum of states
that dier by pathogen and host “activity,” have a dierent
contagiousness, and require dierent treatment strategies.
erefore, it is important to accurately diagnose the stage and
the status of Mtb infection.
It is generally assumed that the outcome of Mtb infection
depends on natural variations in host immune response
to mycobacteria, in particular on the type and extent of
immune activation and inammation. Immune activation
and inammatory reactions are essential for host protection
against mycobacteria [2–6]. On the other hand, unrestricted
immune responses are deleterious and may lead to the TB
exacerbation [7, 8]. Overall, the interplay between immune
activation, inammation, and TB pathogenesis is complex
and not completely understood. In spite of the complexity,
associations between Mtb infection activity and some param-
eters of immune/inammatory reactions have recently been
described; several dierent immunological parameters have
been suggested as markers of TB activity. Many of them are
related to T helper cells (discussed below).
Hindawi Publishing Corporation
Mediators of Inflammation
Volume 2015, Article ID 854507, 13 pages
http://dx.doi.org/10.1155/2015/854507
2Mediators of Inammation
ere are several populations (dierentiation lineages)
of T helper cells. e rst two populations, 1 and 2,
were described over 20 years ago [9]. Later, additional T
helper subsets were identied, including 17, 22, 9,
and TFH. Among dierent populations of T helper cells, 1
and 17 are the main eector populations which mediate
protection and pathology during TB. In this review we discuss
therolefor1and17cellsinprotectiveandinammatory
responses during Mtb infection and the prospective use of
their markers for the evaluation of pulmonary TB activity.
2. Th1 Cells in TB Protection and Pathology
Distinct populations of T helper cells dier by the expressed
cytokines and transcription factors and by their response to
dierent classes of pathogens. 1 cells produce IFN-𝛾and
depend on the transcription factor T-bet. ey are induced
upon infection with intracellular pathogens and mediate
protection mainly by activating macrophages which destroy
intracellular pathogens. 2 cells produce IL-4, IL-5, and
IL-13, depend on the transcription factor GATA3, and ght
extracellular parasites [9]. M. tuberculosis is an intracellular
pathogen and elicits 1 response.
2.1. 1 Induction and Dierentiation. Eector 1 cells
dierentiate from na¨
ıve lymphocytes. In general, the eector
cell dierentiation is driven by the signals received from
TCR, costimulatory receptors, and cytokines. In the most
simplied model, signals mediated by TCR and costimu-
latory receptors induce T cell activation and proliferation.
Cytokine-derived signals are the main signals determining
the dierentiation lineage of antigen-activated T cells. Lig-
ation of cytokines with their receptors activates the signal
transducer and activator of transcription (STAT) factors. e
STATs translocate to the nucleus and bind genes encoding
lineage-specifying transcription factors (“master regulators”)
and eector cytokines. ese events determine the lineage of
dierentiating T helper cells [10].
e main 1 inducing cytokines are IL-12 and IFN-𝛾.
IL-12 is produced by antigen-presenting cells. Interaction of
IL-12 with the IL-12 receptor, expressed on the surface of T
cells, induces STAT4, which in turn induces T-bet, a master
regulator of 1 cells. T-bet binds directly to many 1-
specic genes (Ifng,cxcr3,Il18r1,Il12rb2, etc.) and positively
regulates their expression [11]. T-bet also negatively regulates
the expression of 2- and 17-specic genes and inhibits
the dierentiation of 2 and 17 cells. In the absence
of T-bet, T helper cells dierentiate towards 2-like cells.
STAT4 also directly binds to Ifng locus and stimulates IFN-𝛾
production.STAT4andT-betcooperatetoinducemaximal
IFN-𝛾production. In the absence of STAT4, T-bet does not
induce optimal IFN-𝛾levels, and the combination of T-bet
and STAT4 deciency abolishes IFN-𝛾production [11–13].
IFN-𝛾acts by inducing STAT1. STAT1 synergizes with
STAT4 in activating T-bet in T helper cells and also can
directly activate 1-associated genes [14, 15]. IL-4 and IL-
10 inhibit 1 cell dierentiation and induce 2 cells. us,
generally, 1 and 2 are alternate and mutually exclusive
lineages of T helper cells. In some conditions, however, 1
and 2 cells retain their plasticity, can transdierentiate,
and even coexpress 1- and 2-specic cytokines and
transcription factors [16].
Cytokine milieu is the main factor that determines the
lineage of T helper cell dierentiation. However the dieren-
tiationisalsoaectedbythedoseandthestrengthofTCR
agonistic ligand. It has been shown that stimulation with a
high dose of TCR agonistic peptide or a strongly agonistic
ligand favors generation of 1 cells whereas a low dose or
a weakly agonistic ligand favors 2 cells [17]. erefore,
we may speculate that pathogen load, pathogen metabolic
activity, and the stage of the disease aect the composition
of the responding T helper population.
2.2. 1 Cells and IFN-𝛾during TB. It has long been assumed
that the central role of 1 cells in the defense against TB
is due to the ability of IFN-𝛾to activate macrophages and
stimulate phagocytosis, phagosome maturation, production
of reactive nitrogen intermediates, and antigen presentation
[18–20]. is has been supported by many observations.
In particular, mice decient in CD4+subset and 1 type
cytokines (i.e., IL-12p40, IFN-𝛾)succumbearlytoMtb
infection with high bacterial loads [21–23]. Similar eects are
observed in mice with the defects in enzymes involved in
the generation of host-bactericidal molecules, dependent on
IFN-𝛾axis [24–27]. Humans with the mutations in molecules
involved in 1 immunity (i.e., the IL-12p40 subunit, the IL-12
receptor 𝛽1 chain, the IFN-𝛾-receptor ligand binding chain,
and STAT1) exhibit extremely high susceptibility to infec-
tions induced by Mtb,Bacillus Calmette-Guerin (BCG), or
environmental mycobacteria [28–30]. HIV-infected patients
decient in CD4+cells have increased reactivation of latent
Mtb infection and altered histopathological characteristics
of TB disease (i.e., diuse necrotic lesions instead of struc-
tured granulomas) [31]. ese and other studies supported
the concept that 1 are prerequisite for TB defense and
act by stimulating the antimycobacterial immunity through
the 1 IFN-𝛾→macrophage activation Mtb
killing/restriction pathway. However some new unexpected
results, that contradict this paradigm, are now emerging.
Several studies in mice reported lack of correlation
between the degree of protection and the frequencies of Mtb-
specic IFN-𝛾producing cells or IFN-𝛾levels [32–35]. In
some studies, antigen-specic IFN-𝛾production by CD4+
T cells correlated with the decrease in Mtb load but did
not reect the strength of protection [34]. In other studies,
1 mediated protection against Mtb replication was IFN-𝛾
independent [36, 37]. Next, some authors reported that IFN-
𝛾mediated protection by inhibiting a deleterious response
of 17 cells rather than by inhibiting Mtb replication [38].
Finally, in some experimental models CD4+Tcellswere
deleterious [39, 40].
For example, Scanga and coauthors reported that in the
murine model of latent tuberculosis, depletion of CD4+cells
resulted in uncontrolled Mtb growthbutitdidnotalter
IFN-𝛾response [32]. Gallegos and colleagues [37] studied
protective properties of ESAT-6 specic 1, 2, and 17
cellsderivedfromthewildtypemiceormicedecientfor
factors associated with 1 activity (e.g., IFN-𝛾,TNF-𝛼). In
Mediators of Inammation 3
an adoptive transfer model, 1 cells were more protective
than 17 cells; 2 cells were not exhibiting any protective
activity against Mtb replication. Yet the ability of 1 cells to
mediate protection did not depend on IFN-𝛾or TNF-𝛼.
Nandi and Behar adoptively transferred IFN-𝛾−/− mem-
ory CD4+TcellsintoMtb-infected recipients and assessed
bacterial load and mice survival [38]. e latter is an
integrated parameter of TB severity that largely depends on
the extent of pathological inammatory response. IFN-𝛾−/−
memory CD4+T cells retained their antimicrobial activity
but failed to protect recipient mice against severe inamma-
tion and death. It was suggested that IFN-𝛾acts by inhibiting
IL-17 response and pathogenic neutrophil accumulation in
theinfectedlungs,thatis,thatIFN-𝛾mediates protection by
limiting host inammatory response rather than by inducing
macrophage antibacterial activity.
Finally, a deleterious role for the CD4+Tcellsduring
TB has been demonstrated. Several studies documented
that mice decient in PD-1 and infected with Mtb exhibit
unaltered or even increased CD4 and IFN-𝛾responses but
diebecauseofthesevereinfectioncharacterizedbythe
uncontrolled bacterial proliferation, increased lung tissue
pathology, neutrophilic inltration, and high lung expression
of proinammatory cytokines. Of note, CD4+T cell depletion
reduced production of IFN-𝛾and other proinammatory
cytokines and rescued PD-1−/− mice from early mortality [39,
40]. Interestingly, resistance to viral infections is increased
following PD-1 blockade [41], indicating that in TB an
imbalancedTcellresponsemaybemoredeleteriousthan
during other infections.
us, one important aspect of TB immunity is that 1
subset may mediate protection by mechanisms other than
IFN-𝛾production or activation of host antibacterial activity
and even may contribute to pathology.
Another puzzle comes from the studies in humans that
investigated 1/IFN-𝛾responses during LTBI and active TB.
LTBI is usually considered as a state of Mtb infection
that develops due to an eective anti-TB immune defense.
It is believed that the comparison of immune responses
during LTBI and active TB may help with uncovering
immunological correlates of protection. However, multiple
studies of 1/IFN-𝛾responsesinhumanshaveresultedin
contradictory conclusions. Some studies reported that the
antigen-driven secretion of IFN-𝛾by blood mononuclear
cells is reduced in TB patients and normalized upon TB
treatment [42–44]. On the other hand, frequencies of circu-
lating IFN-𝛾producing cells are generally increased in TB
patientsanddecreasefollowingthetreatment.Inlinewith
this, some studies showed increased plasma levels of IFN-𝛾
in TB patients; these levels correlated with disease activity
and normalized during the treatment [45]. Furthermore,
patients with newly diagnosed TB were reported to have
higher levels of IFN-𝛾compared to patients with chronic TB.
Finally, it is now well documented that interferon-gamma-
release assays (IGRAs) do not discriminate LTBI and active
TB [46–48]. is indicates that the two groups do not dier
consistently by the levels of Mtb-specic IFN-𝛾secretion
(detected in Quantiferon-TB Gold-in tube assay, QFT) or
by the frequencies of Mtb-specic IFN-𝛾producing cells
(dened in T-SPOT-TB assay). us, no steady dierences
in IFN-𝛾responses have been identied so far between LTBI
andactiveTB.
Considering TB patients, they are characterized by a great
variability in the immune responses. It has been shown that
IGRAs have suboptimal sensitivity for active TB, supposedly
due to the immune suppression developed during severe
disease. To verify this hypothesis, we have recently analysed
whether the levels of IFN-𝛾secretion determined in QFT
assay and the frequency of the antigen-specic IFN-𝛾produc-
ing cells determined by ow cytometry are associated with
TB severity. For that, we checked the correlation of IFN-𝛾
responses with the following characteristics of TB disease: the
presence and the quantities of Mtb in the sputum, the forms
of pulmonary pathology (i.e., tuberculoma, inltrative TB,
cavitary TB, caseous pneumonia), the degree of pulmonary
destruction, TB extent, clinical disease severity, and hemato-
logic abnormalities. We have found no signicant association
between these parameters and IFN-𝛾responses, which argues
against the direct association between TB disease severity
and the extent of IFN-𝛾response ([49], and our unpublished
results).
2.3. Section Summary. Altogether, 1 mediated IFN-𝛾
response activates Mtb killing in vitro and contributes to
the restriction of Mtb growth in vivo but also plays anti-
inammatory role during TB and can participate in TB
exacerbation. Assays based on the evaluation of Mtb-specic
IFN-𝛾responses are currently used to identify Mtb infection,
but these assays do not allow discriminating LTBI and TB
disease. Numerous comparisons of IFN-𝛾responses during
active TB and LTBI have revealed no consistent patterns.
ere are several possible explanations for that: dierent
anatomical distributions of eector 1 cells during active TB
and LTBI, functional exhaustion of eector 1 cells during
chronic TB disease, diversity of Mtb antigens expressed
duringLTBIandactiveTB,andagreatvariabilityofTB
patients with regard to TB stage, host immune reactivity,
and genetic and other factors. Irrespective of the underlying
mechanisms, IFN-𝛾response seems to be unreliable for the
evaluation of TB activity. However other markers of 1
reactivity may be valuable and will be discussed below.
3. Th17 Cells in TB Defense and Pathology
17 were rst described as a distinct population of the T
helper cells that are controlled by the transcription factor
ROR𝛾t and develop independent of T-bet, STAT4, GATA-3,
and STAT6 transcription factors that are critical for the 1
and 2 development [50, 51]. e main eector cytokine of
17 is IL-17; other cytokines are IL-22, IL-26, and GM-CSF
[52–56].
e family of IL-17 cytokines consists of the six similar
members, designated from IL-17A (oen referred to as IL-
17) to IL-17F. 17 cells produce IL-17A and IL-17F. Both
have similar biological activities and are the most thoroughly
studied members of the IL-17 family. e family of IL-17
receptors includes ve members (IL-17RA–IL17RE) that are
4Mediators of Inammation
expressed on dendritic cells (DC), macrophages, lympho-
cytes, epithelial cells, keratinocytes, and broblasts and allow
dierent organs to respond to IL-17. IL-17A and IL-17F bind
to IL-17RA [53, 57].
17 mediate pleiotropic activities that involve induction
of proinammatory genes (cytokines, chemokines, and met-
alloproteinases) and antimicrobial peptides, modulation of
extracellular matrix, stimulation of granulopoiesis, recruit-
ment, and activation of neutrophil granulocytes. erefore,
the main functions of 17 cells are protection against
extracellular pathogens and mediation of the inamma-
tory response, particularly during autoimmune and chronic
inammatory diseases.
3.1. 17 Induction and Dierentiation. e dierentiation,
expansion, and maintenance of 17 cells depend on TGF-
𝛽,IL-1𝛽, IL-6, IL-21, and IL-23. e role of these cytokines
in the induction and maintenance/stabilization of 17
cells is dierent in some species, in particular in mice
and humans [53, 58–63]. In general, the dierentiation of
mouse 17 cells depends on IL-6 and TGF-𝛽.IL-23,IL-
1𝛽,andTNF-𝛼maintain and amplify 17 cells. Data on
the dierentiation requirements for human 17 cells are
contradictory. In most studies, human 17 cells depended
on dierent combinations of IL-1𝛽, IL-6, and IL-23. TGF-
𝛽was dispensable or even inhibitory for their develop-
ment(reviewedindetailin[64]).Inbothhumansand
mice dierent combinations of cytokines may result in
the generation of dierent 17 subsets (see below). IFN-
𝛾, IL-12, IL-27, IL-4, and IL-2 inhibit 17 dierentiation
[54, 58, 65–67].
At the molecular level, the generation of 17 requires
induction of ROR𝛾t,akeyregulatorof17dierentiation,
which depends on the activation of the “pioneering factor”
STAT3[10].IL-6,IL-21,andIL-23activateSTAT3,thus
inducing the expression of ROR-𝛾t. Of note, IL-23 is able
to induce the phosphorylation of both STAT3 and STAT4,
but STAT3 phosphorylation is much stronger and biases T
cell dierentiation towards the formation of 17 cells (the
opposite is true for IL-12: it induces strong phosphorylation
of STAT4 but relatively weak phosphorylation of STAT3 [53]).
e role of TGF-𝛽in 17 dierentiation is largely mediated
through its capacity to inhibit the dierentiation of 1 cells
(which suppress 17) [68, 69]. It is assumed that, in the pres-
ence of TGF-𝛽, IL-6 contributes to the generation of 17 by
inhibiting the dierentiation of Treg. Indeed, TGF-𝛽is crit-
ical for the generation of Treg. IL-6 inhibits Treg generation
and thus in inammatory conditions favors the generation of
17; that is, it regulates 17/Treg balance [70]. IL-21 was
showntoupregulateitsownexpressionandthatofIL-21R
and IL-23R. Besides ROR𝛾t, optimal generation of the 17
cells also depends on other cofactors, such as IRF4, BATF, and
RUNX1, which cooperate with ROR-𝛾tin17dierentiation
[10].
3.2. 17/IL-17 Biological Activity. Pleiotropic activities of
17 cells involve activation and recruitment of neutrophils,
stimulation of granulopoietic lineage of dierentiation, and
supportofinammation.
Neutrophil recruitment is mediated through the produc-
tion of CXCL8 and GM-CSF [54, 71] and the induction of
CXCL1, CXCL2, CXCL5, CXCL6, CXCL8, and G-CSF in tis-
sue resident cells, in particular in human bronchial epithelial
and venous endothelial cells [72]. CXCL1, CXCL2, CXCL5,
CXCL6, and CXCL8 are neutrophil-attracting chemokines;
GM-CSF and G-CSF stimulate granulopoiesis and granulo-
cyte activation [73].
e ability of IL-17 to recruit neutrophils to mucosal
sites has been demonstrated in a number of studies [74–
77]. e role for IL-17 in modulating granulopoiesis is
evident from the expansion of the neutrophil progenitors
in the bone marrow and mature neutrophils in peripheral
blood of the mice overexpressing IL-17 [78]. In the model
of lung infection induced by Klebsiella pneumoniae, lack
of IL-17 hampered stress-induced granulopoiesis suggesting
that during infections intact IL-17 response may be required
for the eective granulocyte generation [79]. In primary
human bronchial epithelial cells 17 induce the expres-
sion of mucins MUC5AC and MUC5B [80] and antimi-
crobial peptides (e.g., human beta-defensins) [81, 82]. e
activity is mediated by IL-17 and IL-22 [83] and together
with neutrophil recruitment serves to clear extracellular
pathogens. Recent studies suggest that 17 are also involved
in host protection against some intracellular pathogens
[84].
17/IL-17 mediated induction of chemokines and
cytokines in tissue resident cells, as well as the recruitment
of neutrophils, promote local inammatory responses and
mayleadtotheserioustissuedamage.Accordingly,17
cellshavebeenimplicatedinthepathogenesisofseveral
autoimmune and chronic inammatory diseases. It has been
suggested that in autoimmunity and chronic inammation,
IL-17 synergizes with other proinammatory cytokines
abundantly produced in these pathological conditions, such
as TNF-𝛼and IL-1𝛽[53]. 17 cells may even coexpress
IL-17 and TNF-𝛼. It has been suggested that IL-17 rather
sustains preexisting inammation than induces it. is
means that 17 may be protective during acute infection
and deleterious during chronic one [85]. is concept may
be relevant to TB, as TB is accompanied by the abundant
inammatory responses.
3.3. 17 Phenotype and Subpopulations. Na¨
ıve and antigen-
experienced cells dier by the expression of their dierentia-
tion markers. Distinct lineages (populations) of T helper cells
dier by the expression of chemokine receptors. Finally, some
T cell lineages express lineage-specic markers. e majority
of IL-17 producing cells express CD45RAphenotype of
antigen-experienced cells [86]. Characteristic feature of IL-
17 producing cells and their precursors is the expression of
CD161, the lectin receptor, and the human ortholog of murine
NK1.1 [87, 88].
With regard to chemokine receptor expression, the 17
population is heterogeneous. It contains cells expressing lym-
phoid tissue homing receptor CCR7 (in a higher proportion
than IFN-𝛾producing cells), B follicle homing receptor
CXCR5, and nonlymphoid tissue homing receptors CCR4,
CCR5, and CXCR6. Heterogeneous expression of these
Mediators of Inammation 5
receptors by 17 cells underlies their capacity to migrate to
dierent sites [89].
Chemokine receptors CXCR3, CCR4, and CCR6 discrim-
inate dierent lineages of T helper cells. 1 cells are char-
acterized by the expression of CXCR3, CXCR6, and CCR5;
2 cells express CCR3, CCR4, and CCR8. e chemokine
receptor associated with 17 is CCR6, the receptor for
CCL20 and defensins, mediating homing to skin and mucosal
sites. Correlation between the expression of CCR6, the
production of IL-17, and the expression of RORC (the human
ortholog of mouse ROR𝛾t) was directly demonstrated, and
17 cells were shown to express the CCR6+CCR4hiCXCR3lo
phenotype [90].
Further analysis, however, identied a population of
CCR6+CCR4loCXCR3hi cells that coexpress 17 (CCR6)
and 1 (CXCR3) associated receptors [91]. is phenotype
is associated with the coexpression of two master regulators,
ROR𝛾tandT-bet[10].Functionalanalysisshowedthat
CCR6+CCR4loCXCR3hi population contains cells producing
only IFN-𝛾,onlyIL-17,andpoly-functionalcellsableto
produce both IFN-𝛾and IL-17. Due to the simultaneous
production of IFN-𝛾and IL-17, the latter population was
named 1/17 [64, 90, 92–95].
Further heterogeneity of 17 cells comes from their
inammatory properties. In mice, “pathogenic” and “non-
pathogenic” populations of IL-17 producing cells have been
described. Generation of pathogenic cells depended on IL-
23.ecellsproducedIL-17,andtheiradoptivetransfer
caused experimental autoimmune encephalomyelitis. Non-
pathogenic cells coexpressed IL-17A and IL-10, were able
to suppress T cell proliferation and were called immune-
suppressive or regulatory 17 cells (r17) [96–99].
In humans, a population of 17.1 cells similar to the
“pathogenic” mouse 17 cells has recently been described.
e cells coexpressed CCR6 and CXCR3 and exhibited
transient expression of c-kit and stable expression of the mul-
tidrug transporter MDR1. Transcriptionally and functionally
17.1 cells resembled the pathogenic mouse 17 cells; that is,
they were highly sensitive to IL-23 stimulation and produced
both 17 and 1 cytokines [91]. Similarly, in some studies,
“pathogenic” 17 cells in mice were also characterized by
the coexpression of IL-17A, TNF-𝛼,andIL-2.Altogether,the
data raise questions on whether pathogenic potential of 17
cellsisassociatedwiththeCCR6
+CXCR3+population and
whether it depends on IL-17 or on a combination of factors
that these cells produce.
To summarize, the main phenotypic characteristic of IL-
17 producing cells is the expression of CD161 and CCR6.
17 are phenotypically and functionally heterogeneous and
contain at least two subpopulations, 17 and 1/17, that
dier by the expression of chemokine receptors (i.e., CCR4
and CXCR3) and eector cytokines (i.e., IL-17, IL-17 and IFN-
𝛾, IL-17 and IL-10) and may play dierent roles in immune
pathology and protection. eir exact role in these processes
is yet to be determined.
3.4.17andIL-17inTBProtectionandPathology. e fact
that 17 cells mediate both antibacterial and proinamma-
tory responses suggests that their role during infection is
complex. is is particularly true for TB, as the pathogenesis
of TB critically depends on the extent of inammation. Data
on 17 responses during TB are numerous but not uniform
and quite likely depend on the model and the degree of
inammation.
Following vaccination, 17 seem to contribute to mem-
ory response and protection. In mice immunized with BCG,
IL-17 supported 1 reactivity by downregulating IL-10 and
upregulating IL-12 production in dendritic cells [100, 101].
Following Mtb challenge of BCG vaccinated mice, 17
induced chemokines, recruited CD4+Tcellstothesite
of infection, favored granuloma formation, and accelerated
pathogen clearance [102]. In humans, generation of 17
and 1/17 cells in response to a novel TB vaccine, the
modied vaccinia virus Ankara expressing antigen 85A, was
documented [103]. Interestingly, BCG vaccination induces
dierent levels of IL-17 in genetically dierent mice. Garcia-
Pelayo and coauthors [35] have demonstrated that BALB/c
mice produce more IFN-𝛾and IL-17 and less IL-10 in
response to BCG as compared to C57BL/6 mice. is suggests
that (i) the extent of 17 reactivity is aected by the host
genetic factors; (ii) the pattern of 1/2/17 responses
may dier during TB infection and following BCG vaccina-
tion.
Data on the role for 17 during primary Mtb infection
are conicting.
In mice challenged via aerosol route with a low dose
of laboratory Mtb strain, IL-17 was dispensable for primary
immunity [104]. On the other hand, 17/IL-17 responses
were involved in the protection against highly virulent Mtb
isolate, HN878. Gopal and coauthors [105] reported that
mice challenged with HN878 exhibited elevated IL-17/17
responses as compared to mice challenged with laboratory
adapted Mtb strain. IL-17−/− mice infected with HN878
had elevated lung bacterial burden, diminished chemokine
response (CXL13, in particular), defective formation of
ectopic lymphoid follicles, and hampered colocalization of
T-lympho c yte s and m acrophages [105]. e involvement of
17 in TB protection is also supported by partial inhibition
of the Mtb growth following the adoptive transfer of 17 cells
[37].
e role for 17/IL-17 responses in TB protection in
humans was mainly studied by comparing these responses
in TB patients and healthy individuals. e results are
contradictory. Some authors reported similar levels of IL-17
in the blood and bronchoalveolar uids (BAL) of TB patients
and healthy controls [106]. In other studies, the frequencies
of blood IL-17 producing cells were reduced in TB patients
suggesting that 17 contribute to the protection [107]. is
idea is supported by the observation showing that the low
level of IL-17 in serum is associated with high mortality of
TB patients [108].
Other studies, however, suggest the involvement of 17
in TB pathology. Jurado and coauthors reported an aug-
mented 17 response in TB patients. e major source
of IL-17 was represented by IFN-𝛾+IL-17+CD4+Tcells,
and their proportion directly correlated with the clinical
parameters associated with the disease severity [109]. In
6Mediators of Inammation
line with this, Basile and others have associated augmented
17 response with persistent and high antigen load and
pathogen drug resistance [110]. It should be noted that IL-
17hasbeenimplicatedinneutrophilrecruitmentandstress-
induced granulopoiesis. Neutrophils have been suggested
to play a pathogenic role and exacerbate TB disease [7, 8,
111–113]. Recent studies performed in a mouse model have
suggested that immature myeloid cells are strong correlates
of severe TB [114–116]. us, the involvement of 17/IL-17
responses in the pathogenesis of severe TB is not surprising.
An important question is the interaction/s between 17
and1cellsandtheirrelativerolesduringactiveTBand
LTBI. As discussed above, 1 cells and IFN-𝛾suppress
the generation of 17. On the other hand, 17 do not
inhibit the generation of 1 in vitro and may even favor 1
response in vivo. e relationships between 1 and 17 cells
during TB infection are complex and not well understood.
Comparative analysis of 1 and 17 responses was per-
formed in several studies and the results are contradicting.
Mar´
ın and coauthors [117] studied the frequencies of IL-17
and IFN-𝛾producing cells in patients with active TB, LTBI,
and noninfected control donors. e frequency of IFN-𝛾
producingcellswaselevatedinLTBI,andIL-17producing
cells were more frequent in TB patients. e authors con-
cluded that active TB biases the protective 1 prole toward
the pathological 17 response. Another study compared
cytokine levels in tuberculin skin test (TST) positive and
TST negative individuals in TB endemic area. 1 and 2
cytokines were not dierent between the two groups; IL-17,
IL-23, and ROR𝛾t expressions were downregulated in TST
positive individuals. e authors suggested that lack of 17
cells predisposes to latent infection [118]. However this study
did not investigate active TB. Li and coauthors evaluated
studies of 1 and 17 responses in TB patients [119]. Of
226 studies, nine met their criteria and were selected for the
analysis. eir systematic review showed that in TB patients
the levels of IL-17 and IFN-𝛾were low; during LTBI IL-17 and
IFN-𝛾levels were generally high compared to active TB. In
contrast to TB disease, BCG vaccination in children induced
high level of IL-17 and IFN-𝛾.eauthorsconcludedthat
aer BCG vaccination and during Mtb infection IL-17 acts
as an eector molecule similar to IFN-𝛾and together with
IFN-𝛾contributes to TB protection.
An interesting question is which cells represent the
major source of IL-17 during TB. As discussed above, a
population of 1/17 cells coexpressing IFN-𝛾and IL-
17 exists. 17 may acquire expression of T-bet and IFN-
𝛾and even Foxp3 during their development; that is, they
exhibit substantial plasticity [10, 16, 120]. Whether “multi-
functional” 17 cells dier from “classical” 17 cells in
their pathogenicity is not completely clear. As noted above,
in some pathological conditions (e.g., in the gut of Crohn’s
disease patients), cells coexpressing 17 and 1 cytokines
are pathogenic. Similarly, in TB patients the accumulation
of IFN-𝛾+IL-17+cells correlated with the disease severity
[109]. Arlehamn and coauthors [121] have demonstrated
that TB-specic memory T cells are predominantly present
within the CCR6+CXCR3+CCR4low population and that
this population is signicantly increased in LTBI donors
compared to healthy controls. e CCR6+CXCR3+CCR4low
populationisknowntocontain1/17cells.However,
the authors did not detect IL-17 production in TB-specic
CCR6+CXCR3+CCR4cells upon their ex vivo antigenic
stimulation. Note that in response to other antigens (e.g.,
Candida albicans)CCR6
+CXCR3+CCR4low cells readily pro-
duce IL-17 [90]. us, the accumulation of Mtb-specic
CCR6+CXCR3+CCR4IL-17cells may represent a charac-
teristic feature of Mtb infection. It would be interesting to
study if these cells also accumulate during active TB or if they
specically mark LTBI.
Another question concerns 17 responses that develop
locallyinthelungs.echaracteristicsoflocal17responses
couldprobablyshedalightontherolewhichthesecells
play in TB protection/pathology, but this aspect has not
been studied in detail yet. In one study, IL-17 was not
abundant in pleural or pericardial uid of TB patients; IL-17
expression by mycobacteria-specic disease site T cells was
not detected in healthy Mtb-infectedpersons,orpatientswith
TB pericarditis, allowing the authors to conclude that IL-17
does not play a major role at established TB disease sites in
humans [122].
3.5. Section Summary. To summarize, a number of studies
suggest that IL-17 producing T cells are eciently generated
following vaccination and involved in the memory response
to subsequent Mtb challenge. e role for these cells during
primary Mtb infection is less clear. In many studies, the extent
of 17 response during TB infection was low, which may be
interpreted as a defect in the protective response, but also
as a dispensable role for 17/IL-17 in TB protection and
pathology. However, other studies have associated 17/IL-
17 with TB pathology and progression. It is possible that
17 cells may play dierent roles at subsequent stages of
TB infection providing protection at early disease stage but
inducing pathology at advanced TB. Next, IL-17 producing T
helper cells contain at least two dierent populations, 17
and 1/17. eir role in TB protection and pathology
may dier and has not been evaluated separately as were
the peculiarities of local 17 responses. It should be noted
that experimental data on the role for 17 cells during Mtb
infection should be interpreted with caution. Indeed, unlike
1 and 2 cells, 17 cells dier between mice and humans.
In particular, mouse and human 17 cells depend on
dierent sets of cytokines, and the ecacy of their generation
during Mtb infection in mice and humans may dier. Next,
mouse models of TB infection do not fully reproduce pul-
monary TB in humans by the type of pulmonary pathology
and granuloma formation. us, in mice and humans cells
involved in granuloma formation may have a dierent impact
in local immune responses. Finally, clinical Mtb isolates and
laboratory Mtb strains induce a dierent extent of 17
response.
It should also be noted that discriminating between
protective and pathological responses during infectious dis-
eases is always extremely dicult, as the same cells may
be simultaneously involved in both processes. However
Mediators of Inammation 7
some parameters of cellular responses (regardless of their
protective or pathological impact) may be strongly associated
with the disease severity/activity and thus may be used as
biomarkers for disease evaluation and monitoring. Whether
biomarkers of 1 and 17 responses may be used to
assess activity of Mtb infection is discussed in the next
section.
4. T Cell Associated Biomarkers of TB Activity
As discussed in the introduction, evaluation of TB activity is
an important diagnostic goal, both to discriminate between
LTBIandactiveTBandtomonitorinfectionactivityatthe
dierent stages of TB disease. Whether and which parameters
oftheimmuneresponsemaybeusedasmarkersofTB
activity are an open question.
4.1. IFN-𝛾.Speaking about IFN-𝛾response, which has been
studied most extensively, the relations between its intensity
and Mtb infection activity are extremely complex. In the most
simplied model, the ecacy of IFN-𝛾response depends
on genetic and/or other Mtb infection-independent factors
(e.g., nutrition, stress, the use of immunosuppressive drugs,
and chronic infections) and this aects the outcome of Mtb
infection. In this model, the lower the IFN-𝛾response is,
thehighertheTBactivitywouldbe.Ontheotherhand,
all immune responses, including IFN-𝛾,arepathogen-driven
andthusthemoreactivetheinfectionis,thehigherthe
immune response should be. However, chronic infection
and persistent antigenic stimulation induce regulatory loops
and T cell exhaustion, aecting the relationships between
infection activity and IFN-𝛾production. e real situation is
even more complex because IFN-𝛾is produced by dierent
immune cells, including innate and adaptive cells, which may
react to the infection dierently. Finally, the IFN-𝛾producing
cells accumulate preferentially at the sites of infection, so their
reduced (or increased) response in peripheral blood may be
associated with increased (or diminished) local responses.
is complexity explains why a simple measure of IFN-𝛾
response does not allow to evaluate TB activity. Indeed, the
levels of antigen-specic IFN-𝛾production by mononuclear
cells or the frequencies of Mtb-specic IFN-𝛾producing
cells, evaluated in IGRAs or by ow cytometry, do not
discriminate between LTBI and active TB. Also, the levels of
IFN-𝛾response are not associated with the disease severity
in TB patients ([47–49, 123]; our unpublished observations).
However, some other parameters of 1 response may be used
as TB biomarkers.
4.2. Markers of 1 Dierentiation and Activation. Several
studies have demonstrated that phenotypic markers of T cell
activation and dierentiation are promising biomarkers of TB
activity. Following antigen-driven dierentiation, T lympho-
cytes pass through several stages (early, late, and terminally
dierentiated eector cells). Each stage is characterized by the
set of markers expressed on the surface of T lymphocytes. As
the dierentiation process depends on antigenic stimulation,
markers of T cell dierentiation may serve as indicators of TB
activity. Among such markers CD27 seems to be best studied.
CD27isamemberofTNFreceptorsuperfamily.Itis
constitutively expressed by the naive T cells and early eector
lymphocytes but downregulated at the late stages of eec-
tor cell dierentiation. erefore, late eector lymphocytes
exhibit low to no CD27 expression [124–130].
In mice, the CD27low phenotype has been linked to
ecient IFN-𝛾production and lung-homing properties of
the eector CD4+T cells [130, 131]. Moreover, it was demon-
strated that CD27low eector CD4+T cells can dierentiate
from CD27hi eector precursors directly in the lungs infected
with Mtb [131].edatasuggestthatCD27low population may
serve as a measure of pulmonary TB activity. is, indeed,
was demonstrated in several studies performed by several
scientic teams.
In the pilot study, Streitz and others [132] examined
the percentages of CD27cells within the population of
blood Mtb-reactive CD4+T cells that were identied as
CD4+cells producing IFN-𝛾in response to the tuber-
culin stimulation. High (>49%) percentage of CD27(IFN-
𝛾+CD4+) cells discriminated patients with smear and/or cul-
ture positive pulmonary TB from patients with smear/culture
negative TB and LTBI with 100% sensitivity and 85.7%
specicity.
e potential of “CD27/IFN-𝛾” approach to discriminate
active TB and LTBI was also reported by other authors [49,
133–135]. Some studies found an association between the
accumulation of blood CD27Mtb-reactive CD4+Tcells
and bacillary load in TB patients [133]. In our study, the
frequencies of CD27IFN-𝛾+CD4+cells strongly correlated
with a degree of pulmonary destruction. Evaluation of blood
CD27IFN-𝛾+CD4+cells allowed not only to separate active
TB and LTBI, but also to assess the degree of pulmonary
destruction, that is, the activity of pathological process ongo-
ing in the lung tissue during TB and following the treatment
[49].Schuetzandcoauthorsmodiedthisapproachand
extended it to HIV+patients. e authors reported that
HIV+TBpatients have higher proportion of CD27IFN-𝛾+
CD4+cells than HIVTBpatients and suggested that in
HIV-infected individuals the accumulation of Mtb-reactive
CD27CD4+cells mirrors a degree of Mtb replication
and may help to identify subclinical Mtb infection [134].
e same group has recently extended their study made in
adults to children. e assay the authors called “TAM-TB”
(for “T cell activation marker-tuberculosis”) detected culture
conrmed TB cases in children with 83.3% sensitivity and
96.8% specicity [135]. Petruccioli and coauthors compared
diagnostic accuracy of dierent variations of “CD27/IFN-𝛾
approach and concluded that CD27 expression is a robust
biomarker for discriminating between TB stages [136].
To summarize, the “CD27/IFN-𝛾”approachhasbeen
validated in several studies made in few independent lab-
oratories. In addition, it relies on the mechanisms that
are generally well understood and involve promoted local
generation of CD27cells in Mtb-infectedlungsandtheir
facilitatedemigrationfromthelungstothecirculationsystem
when lung tissue is destructed [49, 131].
Anothermarkerwhichcanbeusedtodierentiateactive
TB and LTBI is CD57. CD57, the human natural killer-1
8Mediators of Inammation
(HNK-1) glycoprotein, marks terminally dierentiated, pro-
liferatively incompetent cells [137]. Lee and others demon-
strated that increased frequencies of CD57+cells among
ESAT-6/CFP10-responding CD4+Tcellsdierentiateactive
TB from LTBI [138].
In contrast to dierentiation, which is an irreversible
process, the activation of T cells is a temporal state that
comesasaresultofTcellencounterwithacognateantigen.
T cell activation is characterized by upregulation of several
surface molecules. Some of them, in particular, CD38 and
HLA-DR,havebeenassociatedwithactiveTB.CD38isa
transmembrane glycoprotein that has ectoenzymatic proper-
ties and catalyzes the synthesis and hydrolysis of NAD and
cyclin ADP-ribose. HLA-DR is a member of MHC class II
molecules involved in antigen presentation. Both markers
are upregulated by antigen-responding T cells. Several recent
studies suggested that increased frequencies of CD38+and
HLA-DR+Mtb-reactive (IFN-𝛾producing) blood CD4 T
cells allow accurately separating active TB from LTBI and
even predicting the time of sputum conversion [123, 139].
4.3. Multifunctional T Cells. Besides IFN-𝛾,1cellsproduce
other cytokines, such as TNF-𝛼and IL-2. 1/17 cells
coexpress IFN-𝛾and IL-17. us, eector T cells may exhibit
multifunctional activities. It has been suggested that there
is an association between T cell multifunctionality and TB
activity.
Harari and coauthors reported that TB patients had
reduced frequencies of multifunctional (IFN-𝛾+TNF-𝛼+IL-
2+) cells and increased proportion of single-positive TNF-𝛼
producing cells (TNF-𝛼+IFN-𝛾IL-2) as compared to LTBI
patients [140]. e authors also showed that TB patients are
characterized by a higher content of Mtb-specic CD8+T
cells [141]. Combined determination of the frequencies of
single TNF-𝛼+cells and Mtb-specic CD8 T cells predicted
active TB with 86.5% specicity and 81.1% sensitivity [142].
Several other groups also reported reduced frequencies of
multifunctional cells in TB patients and their recovery fol-
lowing TB treatment [143].
In contrast, some other studies associated active TB
with increased proportions of multifunctional cells. Caccamo
and coauthors reported that patients with active TB had
high frequencies of multifunctional (IFN-𝛾+TNF-𝛼+IL-2+)
lymphocytes and these frequencies decreased following the
treatment; during LTBI single IFN-𝛾anddoubleIFN-𝛾+IL-2+
Mtb-responding cells predominated [144]. In another study,
active TB was associated with bifunctional (IFN-𝛾+TNF𝛼+)
CD4+T cells [145].
Chesov and coauthors using a novel dual cytokine detect-
ing uorescence-linked immunospot (FluoroSpot) assay
found that the number of single-positive IL2IFN-𝛾+cells
was higher in patients with active TB compared with past
TB and LTBI. However, there was the overlap in cytokine
responses, which precluded distinction between the cohorts
and suggested that the combined analysis of IL-2 and IFN-𝛾
producing cells does not allow to separate dierent states of
Mtb infection in clinical practice [146].
In HIV-infected individuals, analyses of T cell cytokine
prole also gave contradictory results. Some studies reported
higher frequencies of single functional TNF-𝛼-only-secreting
TcellsinHIV
+TB+patients [147], while others found
comparable cytokine proles of Mtb-reactive CD4+Tcellsin
HIV-infected patients with and without active TB [148].
A separate population of multifunctional cells is repre-
sented by 1/17 lymphocytes expressing CCR6+CXCR3+
phenotype. Recently, Sette group described a remarkable
expansion of CCR6+CXCR3+cells in LTBI [95]. e cells
were multifunctional as they produced IFN-𝛾,TNF-𝛼,and
IL-2 upon stimulation with TB-derived epitopes, but the
cells did not produce IL-17. Besides that, the authors did not
analyze CCR6+CXCR3+cells during active TB. us, whether
1/17 and/or CCR6+CXCR3+cellscanserveasmarkersof
LTBI or TB disease is still an open question.
Overall, current data on whether and how multifunction-
ality of eector T cells is associated with TB activity are con-
tradictory. It also remains unclear whether multifunctional
cells are more protective or more pathological compared to
single TNF-𝛼or IFN-𝛾producers, whether dierent cytokine
proles mirror dierent degrees of T cell maturation, and
whether they may serve as reliable biomarkers of TB activity.
us, more studies are required for the further implementa-
tion of this approach.
5. Conclusions
During the past decade a number of attempts have been made
to discover reliable TB biomarkers. In the eld of TB, studies
are generally aimed to identify biomarkers for (i) rapid TB
diagnosis, including diagnosis of sputum-negative TB cases;
(ii) dierentiation of active TB and LTBI; (iii) monitoring TB
activity; (iv) prediction of LTBI conversion to active TB; (v)
assessment of treatment ecacy.
In this review we have mainly focused on T cell associated
markers that have been suggested to discriminate active
TB and LTBI. Analysis of these biomarkers shows that
most of them are at Phase I of discovery according to the
classication of the National Institutes of Health, need to be
validated in several independent laboratories, and undergo
other evaluations. It should also be noted that the most
promising T cell associated biomarkers described above are
detected by means of ow cytometry. eir implementation
in clinical laboratories may be challenging and will require
assay simplication. However if the value of the developed
approaches is conrmed, this might be possible.
It is understood that immunological approaches for TB
diagnosis and monitoring are inferior to the direct identi-
cation of Mtb.Howeverimmunologicalapproachesmay
be of great value for detecting paucibacillary and paediatric
tuberculosis cases, TB screening, and monitoring treatment
ecacy beyond sputum conversion. Uncovering immunolog-
ical TB biomarkers will also allow a better understanding of
TB pathogenesis.
Conflict of Interests
e authors declare that they have no conict of interests
associated with this work.
Mediators of Inammation 9
Acknowledgment
e study was supported by the Russian Science Foundation
(no. 15-15-00136).
References
[1] C.E.BarryJr.,H.I.Bosho,V.Dartoisetal.,“espectrum
of latent tuberculosis: rethinking the biology and intervention
strategies,Nature Reviews Microbiology,vol.7,no.12,pp.845
855, 2009.
[2] N. W. Schluger and W. N. Rom, “e host immune response to
tuberculosis,American Journal of Respiratory and Critical Care
Medicine,vol.157,no.3,part1,pp.679691,1998.
[3] J. L. Flynn and J. Chan, “Immunology of tuberculosis,Annual
Review of Immunology,vol.19,pp.93129,2001.
[4] W.H.Boom,D.H.Canaday,S.A.Fulton,A.J.Gehring,R.E.
Rojas, and M. Torres, “Human immunity to M. tuberculosis:T
cell subsets and antigen processing,Tu berc ulosi s ,vol.83,no.
1–3, pp. 98–106, 2003.
[5] R.J.NorthandY.-J.Jung,“Immunitytotuberculosis,Annual
Review of Immunology,vol.22,pp.599623,2004.
[6] S. H. E. Kaufmann, “Tuberculosis: back on the immunologists
agenda,Immunity,vol.24,no.4,pp.351357,2006.
[7] I. V. Lyadova, “Inammation and immunopathogenesis
of tuberculosis progression,” in Understanding Tuberculosis—
Analyzing the Origin of Mycobacterium Tuberculosis Pathogenic-
ity, P.-J. Cardona, Ed., pp. 19–42, Intech, 2012, http://www
.intechopen.com/books/understanding-tuberculosis-analyzing
-the-origin-of-mycobacterium-tuberculosis-pathogenicity.
[8]A.Dorhoi,S.T.Reece,andS.H.E.Kaufmann,“Forbetter
or for worse: the immune response against Mycobacterium
tuberculosis balances pathology and protection,Immunological
Reviews,vol.240,no.1,pp.235251,2011.
[9]T.R.Mosmann,H.Cherwinski,M.W.Bond,M.A.Giedlin,
and R. L. Coman, “Two types of murine helper T cell clone.
I. Denition according to proles of lymphokine activities and
secreted proteins,e Journal of Immunology,vol.136,no.7,pp.
2348–2357, 1986.
[10] C. M. Evans and R. G. Jenner, “Transcription factor interplay in
T helper cell dierentiation,Briengs in Functional Genomics,
vol. 12, no. 6, Article ID elt025, pp. 499–511, 2013.
[11] J. Zhu, D. Jankovic, A. J. Oler et al., “e transcription
factor T-bet is induced by multiple pathways and prevents an
endogenousThelper-2programduringThelper-1responses,
Immunity,vol.37,no.4,pp.660673,2012.
[12] J. Zhu, H. Yamane, and W. E. Paul, “Dierentiation of eector
CD4 T cell populations,” Annual Review of Immunology,vol.28,
pp.445489,2010.
[13] L. Wei, G. Vahedi, H.-W. Sun et al., “Discrete roles of STAT4 and
STAT6 transcription factors in tuning epigenetic modications
and transcription during T helper cell dierentiation,Immu-
nity,vol.32,no.6,pp.840851,2010.
[14] A. A. Lighvani, D. M. Frucht, D. Jankovic et al., “T-bet is
rapidly induced by interferon-𝛾in lymphoid and myeloid cells,
Proceedings of the National Academy of Sciences of the United
States of America, vol. 98, no. 26, pp. 15137–15142, 2001.
[15] Z. Mikhak, C. M. Fleming, B. D. Medo et al., “STAT1 in
peripheral tissue dierentially regulates homing of antigen-
specic 1 and 2 cells,Journal of Immunology,vol.176,no.
8, pp. 4959–4967, 2006.
[16] J.Geginat,M.Paroni,S.Maglieetal.,“PlasticityofhumanCD4
Tcellsubsets,Frontiers in Immunology,vol.5,article630,2014.
[17] N. van Panhuys, F. Klauschen, and R. Germain, “T-cell-
receptor-dependent signal intensity dominantly controls CD4+
T cell polarization in vivo,Immunity,vol.41,no.1,pp.6374,
2014.
[18] J. Ma, T. Chen, J. Mandelin et al., “Regulation of macrophage
activation,Cellular and Molecular Life Sciences,vol.60,no.11,
pp. 2334–2346, 2003.
[19] J. L. Flynn, J. Chan, and P. L. Lin, “Macrophages and con-
trol of granulomatous inammation in tuberculosis,Mucosal
Immunology,vol.4,no.3,pp.271278,2011.
[20] G. Weiss and U. E. Schaible, “Macrophage defense mechanisms
against intracellular bacteria,Immunological Reviews,vol.264,
no. 1, pp. 182–203, 2015.
[21] A. M. Cooper, D. K. Dalton, T. A. Stewart, J. P. Grin, D. G.
Russell, and I. M. Orme, “Disseminated tuberculosis in inter-
feron gamma gene-disrupted mice,Journal of Experimental
Medicine,vol.178,no.6,pp.22432247,1993.
[22] A. M. Cooper, J. Magram, J. Ferrante, and I. M. Orme, “Inter-
leukin 12 (IL-12) is crucial to the development of protective
immunity in mice intravenously infected with Mycobacter ium
tuberculosis,” JournalofExperimentalMedicine,vol.186,no.1,
pp.3945,1997.
[23] T. Mogues, M. E. Goodrich, L. Ryan, R. LaCourse, and R. J.
North, “e relative importance of T cell subsets in immunity
and immunopathology of airborne Mycobacterium tuberculosis
infection in mice,eJournalofExperimentalMedicine,vol.
193, no. 3, pp. 271–280, 2001.
[24] J. D. MacMicking, R. J. North, R. LaCourse, J. S. Mudgett, S. K.
Shah, and C. F. Nathan, “Identication of nitric oxide synthase
as a protective locus against tuberculosis,Proceedings of the
National Academy of Sciences of the United States of America,
vol.94,no.10,pp.52435248,1997.
[25] A.M.Cooper,B.H.Segal,A.A.Frank,S.M.Holland,andI.M.
Orme, “Transient loss of resistance to pulmonary tuberculosis
in p47(phox-/-) mice,Infection and Immunity,vol.68,no.3,pp.
1231–1234, 2000.
[26]C.A.Scanga,V.P.Mohan,K.Tanaka,D.Alland,J.L.Flynn,
and J. Chan, “e inducible nitric oxide synthase locus confers
protection against aerogenic challenge of both clinical and
laboratory strains of Mycobacterium tuberculosis in mice,
Infection and Immunity,vol.69,no.12,pp.77117717,2001.
[27]Y.-J.Jung,R.LaCourse,L.Ryan,andR.J.North,“Virulent
but not avirulent Mycobacterium tuberculosis can evade the
growth inhibitory action of a T helper 1-dependent, nitric
oxide synthase 2-independent defense in mice,e Journal of
Experimental Medicine,vol.196,no.7,pp.991998,2002.
[28]F.Altare,A.Durandy,D.Lammasetal.,“Impairmentof
mycobacterial immunity in human interleukin-12 receptor
deciency,Science,vol.280,no.5368,pp.14321435,1998.
[29] S. E. Dorman and S. M. Holland, “Interferon-𝛾and interleukin-
12 pathway defects and human disease,Cytokine and Grow th
Factor Reviews,vol.11,no.4,pp.321333,2000.
[30] J.-L. Casanova and L. Abel, “Genetic dissection of immunity to
mycobacteria: the human model,Annual Review of Immunol-
ogy,vol.20,pp.581620,2002.
[31] J. E. Gallant and A. H. Ko, “Cavitary pulmonary lesions in
patients infected with human immunodeciency virus,Clinical
Infectious Diseases,vol.22,no.4,pp.671682,1996.
10 Mediators of Inammation
[32]C.A.Scanga,V.P.Mohan,K.Yuetal.,“DepletionofCD4
+
T cells causes reactivation of murine persistent tuberculosis
despite continued expression of interferon 𝛾and nitric oxide
synthase 2,e Journal of Experimental Medicine,vol.192,no.
3, pp. 347–358, 2000.
[33] L. Majlessi, M. Simsova, Z. Jarvis et al., “An increase in
antimycobacterial 1-cell responses by prime-boost protocols
of immunization does not enhance protection against tubercu-
losis,Infection and Immunity,vol.74,no.4,pp.21282137,2006.
[34] H.-W. Mittr¨
ucker,U.Steinho,A.K
¨
ohler et al., “Poor cor-
relation between BCG vaccination-induced T cell responses
and protection against tuberculosis,Proceedings of the National
Academy of Sciences of the United States of America,vol.104,no.
30, pp. 12434–12439, 2007.
[35] M. C. Garcia-Pelayo, V. S. Bachy, D. A. Kaveh, and P. J. Hogarth,
“BALB/c mice display more enhanced BCG vaccine induced
1 and 17 response than C57BL/6 mice but have equivalent
protection,” Tuber culos i s, vol. 95, no. 1, pp. 48S–53S, 2014.
[36] S. C. Cowley and K. L. Elkins, “CD4+ T cells mediate IFN-
𝛾-independent control of Mycobacterium tuberculosis infection
both in vitro and in vivo,e Journal of Immunology,vol.171,
no. 9, pp. 4689–4699, 2003.
[37]A.M.Gallegos,J.W.J.vanHeijst,M.Samstein,X.Su,E.G.
Pamer, and M. S. Glickman, “A gamma interferon independent
mechanism of CD4 T cell mediated control of M. tuberculosis
infection in vivo,PLoS Pathogens,vol.7,no.5,ArticleID
e1002052, 2011.
[38] B. Nandi and S. M. Behar, “Regulation of neutrophils by
interferon-𝛾limits lung inammation during tuberculosis
infection,e Journal of Experimental Medicine,vol.208,no.
11, pp. 2251–2262, 2011.
[39] E. L´
az´
ar-Moln´
ar, B. Chen, K. A. Sweeney et al., “Programmed
death-1 (PD-1)-decient mice are extraordinarily sensitive to
tuberculosis,Proceedings of the National Academy of Sciences
of the United States of America,vol.107,no.30,pp.1340213407,
2010.
[40] D.L.Barber,K.D.Mayer-Barber,C.G.Feng,A.H.Sharpe,and
A. Sher, “CD4 T cells promote rather than control tuberculosis
in the absence of PD-1-mediated inhibition,e Journal of
Immunology,vol.186,no.3,pp.15981607,2011.
[41] V. Velu, K. Titanji, B. Zhu et al., “Enhancing SIV-specic
immunity in vivo by PD-1 blockade,Nature,vol.458,no.7235,
pp.206210,2009.
[42] C. S. Hirsch, Z. Toossi, C. Othieno et al., “Depressed T-cell
interferon-𝛾responses in pulmonary tuberculosis: analysis of
underlying mechanisms and modulation with therapy,e
Journal of Infectious Diseases,vol.180,no.6,pp.20692073,
1999.
[43] E. Sahiratmadja, B. Alisjahbana, T. de Boer et al., “Dynamic
changes in pro- and anti-inammatory cytokine proles and
gamma interferon receptor signaling integrity correlate with
tuberculosis disease activity and response to curative treat-
ment,Infection and Immunity,vol.75,no.2,pp.820829,2007.
[44] J.S.Sutherland,M.K.Lalor,G.F.Blacketal.,“Analysisofhost
responses to Mycobacterium tuberculosis antigens in a multi-site
study of subjects with dierent TB and HIV infection states in
sub-Saharan Africa,PLoS ONE,vol.8,no.9,ArticleIDe74080,
2013.
[45] E. Sahiratmadja, B. Alisjahbana, S. Buccheri et al., “Plasma
granulysin levels and cellular interferon-𝛾production correlate
with curative host responses in tuberculosis, while plasma
interferon-𝛾levels correlate with tuberculosis disease activity in
adults,Tube rculo sis,vol.87,no.4,pp.312321,2007.
[46] M.Sester,G.Sotgiu,C.Langeetal.,“Interferon-𝛾release assays
for the diagnosis of active tuberculosis: a systematic review and
meta-analysis,European Respiratory Journal,vol.37,no.1,pp.
100–111, 2011.
[47] J.Z.Metcalfe,C.K.Everett,K.R.Steingartetal.,“Interferon-
𝛾release assays for active pulmonary tuberculosis diagnosis in
adults in low- and middle-income countries: systematic review
and meta-analysis,e Journal of Infectious Diseases,vol.204,
supplement 4, pp. S1120–S1129, 2011.
[48] M. Pai, C. M. Denkinger, S. V. Kik et al., “Gamma interferon
release assays for detection of Mycobacterium tuberculosis infec-
tion,Clinical Microbiology Reviews,vol.27,no.1,pp.320,2014.
[49]I.Y.Nikitina,N.A.Kondratuk,G.A.Kosmiadietal.,“Mtb-
specic CD27low CD4 T cells as markers of lung tissue destruc-
tion during pul monary tuberculosi s in humans,PLoS ONE,vol.
7, no. 8 , A r t i c le ID e 4 3 73 3, 2 0 1 2.
[50] L. A. Steinman, “A brief history of TH17, the rst major revision
in the TH1/TH2hypothesis of T cell-mediated tissue damage,
Nature Medicine,vol.13,pp.139145,2007.
[51] I. I. Ivanov, B. S. McKenzie, L. Zhou et al., “e orphan nuclear
receptor RORgammat directs the dierentiation program of
proinammatory IL-17+ T helper cell,Cell,vol.126,no.6,pp.
1121–1133, 2006.
[52] H. Park, Z. Li, X. O. Yang et al., “A distinct lineage of CD4 T
cells regulates tissue inammation by producing interleukin 17,
Nature Immunology, vol. 6, no. 11, pp. 1133–1141, 2005.
[53] J. F. Zambrano-Zaragoza, E. J. Romo-Mart´
ınez,M.D.J.Dur
´
an-
Avelar et al., “17 cells in autoimmune and infectious diseases,
International Journal of Inammation,vol.2014,ArticleID
651503, 12 pages, 2014.
[54] L. Codarri, G. Gy¨
ulv´
eszii, V. Tosevski et al., “ROR𝛾3t drives
production of the cytokine GM-CSF in helper T cells, which is
essential for the eector phase of autoimmune neuroinamma-
tion,Nature Immunology,vol.12,no.6,pp.560567,2011.
[55] S. C. Liang, X.-Y. Tan, D. P. Luxenberg et al., “Interleukin (IL)-
22 and IL-17 are coexpressed by 17 cells and cooperatively
enhance expression of antimicrobial peptides,JournalofExper-
imental Medicine,vol.203,no.10,pp.22712279,2006.
[56] P. Miossec, T. Korn, and V. K. Kuchroo, “Interleukin-17 and type
17 helper T cells,e New England Journal of Medicine,vol.361,
no. 9, pp. 888–898, 2009.
[57] H. Zhang, F. Bernuzzi, A. Lleo, X. Ma, and P. Invernizzi,
“erapeutic potential of IL-17-mediated signaling pathway in
autoimmune liver diseases,Mediators of Inammation,vol.
2015, Article ID 436450, 12 pages, 2015.
[58] P. R. Mangan, L. E. Harrington, D. B. O’Quinn et al., “Trans-
forming growth factor-beta induces development of the T H17
lineage,Nature,vol.441,no.7090,pp.231234,2006.
[59] R.Nurieva,X.O.Yang,G.Martinezetal.,“Essentialautocrine
regulation by IL-21 in the generation of inammatory T cells,
Nature, vol. 448, no. 7152, pp. 480–483, 2007.
[60]J.M.Coquet,S.Chakravarti,M.J.Smyth,andD.I.Godfrey,
“Cutting edge: IL-21 is not essential for 17 dierentiation
or experimental autoimmune encephalomyelitis,Journal of
Immunology,vol.180,no.11,pp.70977101,2008.
[61] S. Romagnani, E. Maggi, F. Liotta, L. Cosmi, and F. Annun-
ziato, “Properties and origin of human 17 cells,Molecular
Immunology,vol.47,no.1,pp.37,2009.
Mediators of Inammation 11
[62] E. Torrado and A. M. Cooper, “IL-17 and 17 cells in tuber-
culosis,CytokineandGrowthFactorReviews,vol.21,no.6,pp.
455–462, 2010.
[63] S. L. Gaen, “Recent advances in the IL-17 cytokine family,
Current Opinion in Immunology,vol.23,no.5,pp.613619,2011.
[64] F. Annunziato, L. Cosmi, F. Liotta, E. Maggi, and S. Romagnani,
“Human 17 cells: are they dierent from murine 17 cells?”
European Journal of Immunology,vol.39,no.3,pp.637640,
2009.
[65] A. Laurence, C. M. Tato, T. S. Davidson et al., “Interleukin-
2 signaling via STAT5 constrains T helper 17 cell generation,
Immunity,vol.26,no.3,pp.371381,2007.
[66] A. Cruz, S. A. Khader, E. Torrado et al., “Cutting edge: IFN-
𝛾regulates the induction and expansion of IL-17-producing
CD4 T cells during mycobacterial infection,e Journal of
Immunology,vol.177,no.3,pp.14161420,2006.
[67] C.Pot,L.Apetoh,A.Awasthi,andV.K.Kuchroo,“Induction
of regulatory Tr1 cells and inhibition of TH17 cells by IL-27,
Seminars in Immunology,vol.23,no.6,pp.438445,2011.
[68] E. Bettelli, Y. Carrier, W. Gao et al., “Reciprocal developmental
pathways for the generation of pathogenic eector TH17 and
regulatory T cells,” Nature,vol.441,no.7090,pp.235238,2006.
[69] V. Santarlasci, L. Maggi, M. Capone et al., “TGF-beta indirectly
favors the development of human 17 cells by inhibiting 1
cells,EuropeanJournalofImmunology,vol.39,no.1,pp.207
215, 2009.
[70] M. Oukka, “17 cells in immunity and autoimmunity,Annals
of the Rheumatic Diseases, vol. 67, supplement 3, pp. iii26–iii29,
2008.
[71] M. Pelletier, L. Maggi, A. Micheletti et al., “Evidence for a cross-
talk between human neutrophils and 17 cells,Blood, vol. 115,
no. 2, pp. 335–343, 2010.
[72] W.Ouyang,J.K.Kolls,andY.Zheng,“ebiologicalfunctions
of T helper 17 cell eector cytokines in inammation,” Immu-
nity,vol.28,no.4,pp.454467,2008.
[73] P. Schwarzenberger, W. Huang, Y. Peng et al., “Require-
ment of endogenous stem cell factor and granulocyte-colony-
stimulating factor for IL-17-mediated granulopoiesis,e Jour-
nal of Immunology,vol.164,no.9,pp.47834789,2000.
[74] J. Witowski, K. Pawlaczyk, A. Breborowicz et al., “IL-17 stimu-
lates intraperitoneal neutrophil inltration through the release
of GRO𝛼chemokine from mesothelial cells,e Journal of
Immunology,vol.165,no.10,pp.58145821,2000.
[75] H. Hoshino, M. Laan, M. Sj¨
ostrand, J. L¨
otvall, B.-E. Skoogh,
and A. Lind´
en, “Increased elastase and myeloperoxidase activity
associated with neutrophil recruitment by IL-17 in airways in
vivo,Journal of Allergy and Clinical Immunology,vol.105,no.1,
pp. 143–149, 2000.
[76] M. Miyamoto, O. Prause, M. Sj¨
ostrand, M. Laan, J. L¨
otvall,
and A. Lind´
en, “Endogenous IL-17 as a mediator of neutrophil
recruitment caused by endotoxin exposure in mouse airways,
e Journal of Immunology,vol.170,no.9,pp.46654672,2003.
[77] J. K. Kolls and S. A. Khader, “e role of 17 cytokines
in primary mucosal immunity,Cytokine & Growth Factor
Reviews,vol.21,no.6,pp.443448,2010.
[78] P. Schwarzenberger, V. La Russa, A. Miller et al., “IL-17 stim-
ulates granulopoiesis in mice: use of an alternate, novel gene
therapy-derived method for in vivo evaluation of cytokines,
e Journal of Immunology,vol.161,no.11,pp.63836389,1998.
[79]P.Ye,F.H.Rodriguez,S.Kanalyetal.,“Requirementof
interleukin 17 receptor signaling for lung CXC chemokine and
granulocyte colony-stimulating factor expression, neutrophil
recruitment, and host defense,e Journal of Experimental
Medicine,vol.194,no.4,pp.519527,2001.
[80] Y. Chen, P. ai, Y.-H. Zhao, Y.-S. Ho, M. M. DeSouza, and
R. Wu, “Stimulation of airway mucin gene expression by
interleukin (IL)-17 through IL-6 paracrine/autocrine loop,e
JournalofBiologicalChemistry,vol.278,no.19,pp.1703617043,
2003.
[81] C.-Y.Kao, Y. Chen, P. ai et al., “IL-17 markedly up-regulates𝛽-
defensin-2 expression in human airway epithelium via JAK and
NF-𝜅Bsignalingpathways,e Journal of Immunology,vol.173,
no.5,pp.34823491,2004.
[82] H.-C. Tsai, S. Velichko, L.-Y. Hung, and R. Wu, “IL-17A and
17 cells in lung inammation: an update on the role of 17
cell dierentiation and IL-17R signaling in host defense against
infection,Clinical and Developmental Immunology,vol.2013,
Article ID 267971, 12 pages, 2013.
[83] A.Li,Y.Gan,R.Wangetal.,“IL-22up-regulates𝛽-defensin-
2 expression in human alveolar epithelium via STAT3 but not
NF-𝜅B signaling pathway,Inammation,vol.38,no.3,pp.1191
1200, 2015.
[84] M. J. McGeachy and S. J. McSorley, “Microbial-induced 17:
superhero or supervillain?” e Journal of Immunology,vol.189,
no.7,pp.32853291,2012.
[85] F. Maione, N. Paschalidis, N. Mascolo, N. Duon, M. Perretti,
and F. D’Acquisto, “Interleukin 17 sustains rather than induces
inammation,Biochemical Pharmacology,vol.77,no.5,pp.
878–887, 2009.
[86] H. W. Lim, J. Lee, P. Hillsamer, and C. H. Kim, “Human
17 cells share major tracking receptors with both polarized
eector T cells and FOXP3+regulatory T cells,e Journal of
Immunology,vol.180,no.1,pp.122129,2008.
[87] L. Maggi, V. Santarlasci, M. Capone et al., “CD161 is a marker
of all human IL-17-producing T-cell subsets and is induced by
RORC,European Journal of Immunology,vol.40,no.8,pp.
2174–2181, 2010.
[88] L. Cosmi, R. De Palma, V. Santarlasci et al., “Human interleukin
17-producing cells originate from a CD161+CD4+Tcellprecur-
sor,e Journal of Experimental Medicine,vol.205,no.8,pp.
1903–1916, 2008.
[89] C. H. Kim, “Migration and function of 17 cells,Inammation
and Allergy—Drug Targets,vol.8,no.3,pp.221228,2009.
[90] E. V. Acosta-Rodriguez, L. Rivino, J. Geginat et al., “Surface
phenotype and antigenic specicity of human interleukin 17-
producing T helper memory cells,Nature Immunology,vol.8,
no. 6, pp. 639–646, 2007.
[91] R.Ramesh,L.Kozhaya,K.McKevittetal.,“Pro-inammatory
human 17 cells selectively express P-glycoprotein and
are refractory to glucocorticoids,Journal of Experimental
Medicine,vol.211,no.1,pp.89104,2014.
[92] F. Annunziato, L. Cosmi, V. Santarlasci et al., “Phenotypic
and functional features of human 17 cells,e Journal of
Experimental Medicine,vol.204,no.8,pp.18491861,2007.
[93] C. J. Cohen, S. Q. Crome, K. G. MacDonald, E. L. Dai, D. L.
Mager, and M. K. Levings, “Human 1 and 17 cells exhibit
epigenetic stability at signature cytokine and transcription
factor loci,Journal of Immunology,vol.187,no.11,pp.5615
5626, 2011.
[94] F.Sallusto,C.E.Zielinski,andA.Lanzavecchia,“Human17
subsets,European Journal of Immunology,vol.42,no.9,pp.
2215–2220, 2012.
12 Mediators of Inammation
[95] C. S. Lindestam Arlehamn, A. Gerasimova, F. Mele et al., “Mem-
ory T cells in latent Mycobacterium tuberculosis infection are
directed against three antigenic islands and largely contained
in a CXCR3+CCR6+ 1 subset,PLoS Pathogens,vol.9,no.1,
Article ID e1003130, 2013.
[96] K. Ghoreschi, A. Laurence, X.-P. Yang et al., “Generation of
pathogenic TH17 cells in the absence of TGF-𝛽signalling,
Nature,vol.467,no.7318,pp.967971,2010.
[97] E. Esplugues, S. Huber, N. Gagliani et al., “Control of TH17 cells
occurs in the small intestine,Nature,vol.475,no.7357,pp.514
518, 2011.
[98] Y. Lee, A. Awasthi, N. Yosef et al., “Induction and molecular
signature of pathogenic TH17 cells,Nature Immunology,vol.13,
no. 10, pp. 991–999, 2012.
[99] M. J. McGeachy, K. S. B ak-Jensen, Y. Chen et al., “TGF-𝛽and IL-
6 drive the production of IL-17 and IL-10 by T cells and restrain
TH-17 cell-mediated pathology,Nature Immunology,vol.8,no.
12, pp. 1390–1397, 2007.
[100]M.Umemura,A.Yahagi,S.Hamadaetal.,“IL-17-mediated
regulation of innate andacquired immune response against pul-
monary Mycobacterium bovis bacille Calmette-Gu´
erin infec-
tion,Journal of Immunology,vol.178,no.6,pp.37863796,
2007.
[101] R.Gopal,Y.Lin,N.Obermajeretal.,“IL-23-dependentIL-17
drives 1-cell responses following Mycobacterium bovis BCG
vaccination,European Journal of Immunology,vol.42,no.2,pp.
364–373, 2012.
[102] S.A.Khader,G.K.Bell,J.E.Pearletal.,“IL-23andIL-17inthe
establishment of protective pulmonary CD4+T cell responses
aer vaccination and during Mycobacterium tuberculosis chal-
lenge,Nature Immunology,vol.8,no.4,pp.369377,2007.
[103] K.L.Griths,A.A.Pathan,A.M.Minassianetal.,“1/17
cell induction and corresponding reduction in ATP consump-
tion following vaccination with the novel Mycobacterium tuber-
culosis vaccine MVA85A,PLoS ONE,vol.6,no.8,ArticleID
e23463, 2011.
[104] S. A. Khader, J. E. Pearl, K. Sakamoto et al., “IL-23 compensates
for the absence of IL-12p70 and is essential for the IL-17
response during tuberculosis but is dispensable for protection
and antigen-specic IFN-𝛾responsesifIL-12p70isavailable,
Journal of Immunology, vol. 175, no. 2, pp. 788–795, 2005.
[105] R. Gopal, L. Monin, S. Slight et al., “Unexpected role for IL-17
in protective immunity against hypervirulent Mycobacter ium
tuberculosis HN878 infection,PLoS Pathogens,vol.10,no.5,
Article ID e1004099, 2014.
[106] K.Dheda,J.-S.Chang,S.Lala,J.F.Huggett,A.Zumla,andG.
A. W. Rook, “Gene expression of IL17 and IL23 in the lungs of
patients with active tuberculosis,orax,vol.63,no.6,pp.566
568, 2008.
[107] T. J. Scriba, B. Kalsdorf, D.-A. Abrahams et al., “Distinct, specic
IL-17- and IL-22-producing CD4+T cell subsets contribute to
the human anti-mycobacterial immune response,Journal of
Immunology,vol.180,no.3,pp.19621970,2008.
[108] Y.-C. Chen, C.-H. Chin, S.-F. Liu et al., “Prognostic values of
serum IP-10 and IL-17 in patients with pulmonary tuberculosis,
Disease Markers, vol. 31, no. 2, pp. 101–110, 2011.
[109]J.O.Jurado,V.Pasquinelli,I.B.Alvarezetal.,“IL-17and
IFN-𝛾expression in lymphocytes from patients with active
tuberculosis correlates with the severity of the disease,Journal
of Leukocyte Biology,vol.91,no.6,pp.9911002,2012.
[110] J. I. Basile, L. J. Gener, M. M. Romero et al., “Outbreaks of
Mycobacterium tuberculosis MDR strains induce high IL-17 T-
cell response in patients with MDR tuberculosis that is closely
associated with high antigen load,Journal of Infectious Diseases,
vol. 204, no. 7, pp. 1054–1064, 2011.
[111] E. B. Eruslanov, I. V. Lyadova, T. K. Kondratieva et al.,
“Neutrophil responses to Mycobacterium tuberculosis infection
in genetically susceptible and resistant mice,Infection and
Immunity,vol.73,no.3,pp.17441753,2005.
[112] C. Keller, R. Homann, R. Lang, S. Brandau, C. Hermann, and
S. Ehlers, “Genetically determined susceptibility to tuberculosis
in mice causally involves accelerated and enhanced recruitment
of granuloc ytes,Infection and Immunity,vol.74,no.7,pp.4295
4309, 2006.
[113] A. Dorhoi, C. Desel, V. Yeremeev et al., “e adaptor molecule
CARD9 is essential for tuberculosis control,e Journal of
Experimental Medicine, vol. 207, no. 4, pp. 777–792, 2010.
[114]E.N.Tsiganov,E.M.Verbina,T.V.Radaevaetal.,
“Gr-1dimCD11b+immature myeloid-derived suppressor
cells but not neutrophils are markers of lethal tuberculosis
infection in mice,e Journal of Immunology,vol.192,no.10,
pp. 4718–4727, 2014.
[115] J. K. Knaul, S. J¨
org, D. Oberbeck-Mueller et al., “Lung-
residing myeloid-derived suppressors display dual functionality
in murine pulmonary tuberculosis,American Journal of Respi-
ratory and Critical Care Medicine,vol.190,no.9,pp.10531066,
2014.
[116] A.DorhoiandS.H.Kaufmann,“Versatilemyeloidcellsubsets
contribute to tuberculosis-associated inammation,” European
Journal of Immunology, vol. 45, no. 8, pp. 2191–2202, 2015.
[117] N. D. Mar´
ın, S. C. Par´
ıs,M.Rojas,andL.F.Garc
´
ıa, “Reduced
frequency of memory T cells and increased 17 responses
in patients with active tuberculosis,Clinical and Vaccine
Immunology,vol.19,no.10,pp.16671676,2012.
[118] S. Babu, S. Q. Bhat, N. P. Kumar, V. Kumaraswami, and T.
B. Nutman, “Regulatory T cells modulate 17 responses in
patients with positive tuberculin skin test results,Journal of
Infectious Diseases,vol.201,no.1,pp.2031,2010.
[119]Q.Li,J.Li,J.Tianetal.,“IL-17andIFN-𝛾production in
peripheral blood following BCG vaccination and Mycobac-
terium tuberculosis infection in human,European Review for
Medical and Pharmacological Sciences,vol.16,no.14,pp.2029
2036, 2012.
[120] S.N.Harbour,C.L.Maynard,C.L.Zindl,T.R.Schoeb,andC.
T. Weaver, “17 cells give rise to 1 cells that are required for
the pathogenesis of colitis,Proceedings of the National Academy
of Sciences,vol.112,no.22,pp.70617066,2015.
[121] C. L. Arlehamn, G. Seumois, A. Gerasimova et al., “Transcrip-
tional prole of tuberculosis antigen-specic T cells reveals
novel multifunctional features,e Journal of Immunology,vol.
193, no. 6, pp. 2931–2940, 2014.
[122] K. Matthews, K. A. Wilkinson, B. Kalsdorf et al., “Predominance
of interleukin-22 over interleukin-17 at the site of disease in
human tuberculosis,Tube r culo s is,vol.91,no.6,pp.587593,
2011.
[123] C. Riou, C. M. Gray, M. Lugongolo et al., “A subset of circulating
blood mycobacteria-specic CD4 T cells can predict the time to
Mycobacterium tuberculosis sputum culture conversion,PLoS
ONE,vol.9,no.7,ArticleIDe102178,2014.
[124] T. H. Watts, “THF/TNFR family members in costimulation of
T cell responses,Annual Review of Immunology,vol.23,no.1,
pp.2368,2005.
Mediators of Inammation 13
[125] D.Hamann,S.Kostense,K.C.Wolthersetal.,“Evidencethat
human CD8+CD45RA+CD27cells are induced by antigen
and evolve through extensive rounds of division,International
Immunology, vol. 11, no. 7, pp. 1027–1033, 1999.
[126] V.Appay,J.J.Zaunders,L.Papagnoetal.,“Characterizationof
CD4+CTLs ex vivo,e Journal of Immunology,vol.168,no.11,
pp. 5954–5958, 2002.
[127] R.D.Fritsch,X.Shen,G.P.Sims,K.S.Hathcock,R.J.Hodes,
and P. E. Lipsky, “Stepwise dierentiation of CD4 memory T
cells dened by expression of CCR7 and CD27,e Journal of
Immunology,vol.175,no.10,pp.64896497,2005.
[128]M.A.Nolte,R.W.vanOlen,K.P.J.M.vanGisbergen,
and R. A. W. van Lier, “Timing and tuning of CD27-CD70
interactions: the impact of signal strength in setting the balance
between adaptive responses and immunopathology,Immuno-
logical Reviews,vol.229,no.1,pp.216231,2009.
[129]D.A.Kaveh,V.S.Bachy,R.G.Hewinson,andP.J.Hogarth,
“Systemic BCG immunization induces persistent lung mucosal
multifunctional CD4 TEM cells which expand following viru-
lent mycobacterial challenge,PLoS ONE,vol.6,no.6,Article
ID e21566, 2011.
[130] I. V. Lyadova, S. Oberdorf, M. A. Kapina, A. S. Apt, S. L. Swain,
and P. C. Sayles, “CD4 T cells producing IFN-𝛾in the lungs
of mice challenged with mycobacteria express a CD27-negative
phenotype,Clinical & Experimental Immunology,vol.138,no.
1,pp.2129,2004.
[131] M. A. Kapina, G. S. Shepelkova, P. V. Bogacheva et al., “CD27low
CD4 T lymphocytes that accumulate in the mouse lungs during
mycobacterial infection dierentiate from CD27high precursors
in situ, produce IFN-𝛾, and protect the host against tuberculosis
infection,e Journal of Immunology,vol.178,no.2,pp.976
985, 2007.
[132] M. Streitz, L. Tesfa, V. Yildirim et al., “Loss of receptor on
tuberculin-reactive T-cells marks active pulmonary tuberculo-
sis,PLoS ONE,vol.2,no.8,articlee735,2007.
[133] J. Jiang, X. Wang, X. Wang et al., “Reduced CD27 expression on
antigen-specic CD4+T cells correlates with persistent active
tuberculosis,Journal of Clinical Immunology,vol.30,no.4,pp.
566–573, 2010.
[134] A. Schuetz, A. Haule, K. Reither et al., “Monitoring CD27
expression to evaluate mycobacterium tuberculosis activity in
HIV-1 infected individuals in vivo,PLoS ONE,vol.6,no.11,
Article ID e27284, 2011.
[135] D.Portevin,F.Moukambi,P.Clowesetal.,“Assessmentofthe
novel T-cell activation marker-tuberculosis assay for diagnosis
of active tuberculosis in children: a prospective proof-of-
concept study,” e Lancet Infectious Diseases, vol. 14, no. 10, pp.
931–938, 2014.
[136] E. Petruccioli, L. Petrone, V. Vanini et al., “Assessment of CD27
expression as a tool for active and latent tuberculosis diagnosis,
Journal of Infection,2015.
[137] J. M. Brenchley, N. J. Karandikar, M. R. Betts et al., “Expression
of CD57 denes replicative senescence and antigen-induced
apoptotic death of CD8+Tcells,Blood,vol.101,no.7,pp.2711
2720, 2003.
[138] J. Y. Lee, I. Jeong, J. Joh et al., “Dierential expression of CD57
in antigen-reactive CD4+Tcellsbetweenactiveandlatent
tuberculosis infection,Clinical Immunology,vol.159,no.1,pp.
37–46, 2015.
[139]T.Adekambi,C.C.Ibegbu,S.Cagleetal.,“Biomarkerson
patient T cells diagnose active tuberculosis and monitor treat-
ment response,e Journal of Clinical Investigation,vol.125,no.
5,pp.18271838,2015.
[140] A. Harari, V. Rozot, F. B. Enders et al., “Dominant TNF-𝛼+
Mycobacterium tuberculosis-specic CD4+T cell responses dis-
criminate between latent infection and active disease,Nature
Medicine, vol. 17, pp. 372–376, 2011.
[141] V. Rozot, S. Vigano, J. Mazza-Stalder et al., “Mycobacterium
tuberculosis-specic CD8+T cells are functionally and pheno-
typically dierent between latent infection and active disease,
European Journal of Immunology,vol.43,no.6,pp.15681577,
2013.
[142] V. Rozot, A. Patrizia, S. Vigano et al., “Combined use
of Mycobacterium tuberculosis-specic CD4 and CD8 T-cell
responsesisapowerfuldiagnostictoolofactivetuberculosis,
Clinical Infectious Diseases,vol.60,no.3,pp.432437,2015.
[143] C. L. Day, D. A. Abrahams, L. Lerumo et al., “Functional
capacity of Mycobacterium tuberculosis-specic T cell responses
in humans is associated with mycobacterial load,e Journal of
Immunology, vol. 187, no. 5, pp. 2222–2232, 2011.
[144] N. Caccamo, G. Guggino, S. A. Joosten et al., “Multifunctional
CD4+T cells correlate with active Mycobacterium tuberculosis
infection,European Journal of Immunology,vol.40,no.8,pp.
2211–2220, 2010.
[145] E. Petruccioli, L. Petrone, V. Vanini et al., “IFN𝛾/TNF𝛼specic-
cells and eector memory phenotype associate with active
tuberculosis,Journal of Infection,vol.66,no.6,pp.475486,
2013.
[146] D. Chesov, C. Lange, F. Daduna et al., “Combined antigen-
specic interferon-𝛾and interleukin-2 release assay (Fluo-
roSpot) for the diagnosis of Mycobacterium tuberculosis infec-
tion,PLoS ONE,vol.10,no.3,ArticleIDe0120006,2015.
[147] K.M.Pollock,H.S.Whitworth,D.J.Montamat-Sicotteetal.,
“T-cell immunophenotyping distinguishes active from latent
tuberculosis,eJournalofInfectiousDiseases,vol.208,no.6,
pp. 952–968, 2013.
[148] D.H.Canaday,S.Sridaran,P.VanEppsetal.,“CD4+Tcellpoly-
functional prole in HIV-TB coinfection are similar between
individuals with latent and active TB infection,Tuberc u losi s ,
vol. 95, no. 4, pp. 470–475, 2015.
... Low IL-17 production and corresponding amounts of IFN-γ enhance TB prophylaxis, whereas high levels of IL-17 production, even in the presence of IFN-γ, induce pathological delayed-type hypersensitivity (DTH) reactions [64,65]. IL-17 can enhance protective immune responses by enhancing IL-12 secretion [64]. ...
... In TB studies, introducing recombinant IL-12 into the liver of neutropenic mice reduced hyperactivation of infection [64,66]. IL-17 is a potent cytokine in neutrophil recruitment and activation; therefore, it can be concluded that early recruitment of neutrophils to the site of infection stimulates IL-12 release to induce Th1 responses [64,65]. ...
... Специфическое воспаление при туберкулезе возникает в результате сложного многофазного ответа, направленного на защиту организма от Mycobacterium tuberculosis, опосредуемого взаимодействием между иммунными клетками и растворимыми медиаторами. Эффективность защитных иммунных реакций при туберкулезе существенно зависит от определенных субпопуляций Т-лимфоцитов [24]. Мониторинг активности воспалительного процесса, субпопуляций лимфоцитов может уже на ранних этапах лечения помочь оценить эффективность интенсивной фазы противотуберкулезной терапии [1,2,35]. ...
Article
Full-text available
Monitoring activity of inflammatory process and lymphocyte subsets can help assess the effectiveness of intensive phase therapy (IPT) already in the early stages of treatment. The goal is to evaluate changes in the concentration and activity of enzymes associated with purine metabolism and peripheral blood lymphocyte subset composition, and to determine their relationship with IPT effectiveness in patients with newly diagnosed infiltrative pulmonary tuberculosis (IPTb). Materials and methods. In 141 IPTb patients, the IPT data were presented as follows: “significant improvement” (SI) — disappearance of intoxication symptoms, abacillation, closure of decay cavities; “less pronounced improvement” (LMI) — eliminated symptoms of intoxication, abacillation, pronounced resorption of focal and infiltrative changes, reduction of decay cavities. We assessed the activity of adenosine deaminase in blood serum (eADA-1, 2), mononuclear cells and neutrophils, the concentration of blood serum ecto-5'-nucleotidase (eHT5E), CD26 (DPPV) in blood serum (s, soluble form) and mononuclear cells (m, membrane form), subpopulation composition. Results. Patients exhibit increased concentrations of eNT5E, mCD26 (DPPIV) and eADA-2 activity, and decreased intracellular ADA-1 activity. In the “LMI” group, after IPT, an increased sCD26 (DPPV) level was noted. The groups differed in lymphocyte counts and percentage of CD3+CD8+ cells. eADA-2 activity was higher in the LMI group and increased after IPT, in contrast to comparison group. mCD26 (DPPIV) concentrations are higher in PD patients before therapy and after IPT. Conclusion. Thus, the outcome of IPT in IPTb patients is associated with altered T-lymphocyte populations and severity of the inflammatory process. Studying the activity of membrane and soluble eADA-2, CD26 (DPPIV) and percentage of CD3+CD8+ T-lymphocytes in the early stages of therapy can provide the necessary information for correcting personalized pathogenetic therapy of patients with newly diagnosed IPTb.
... Various cytokines and chemokines linked to the Th1/Th17 and Th2/Th9 signaling pathways are involved at different stages of the TB spectrum, either to contain infection in the early phase, or to contribute to Mtb elimination in TB disease (29)(30)(31)(32). However, a thorough description of the host immune response at different stages of treatment is needed for the development of more sensitive and specific non-sputum-based tests for monitoring TB treatment. ...
Article
Full-text available
Introduction New diagnostic tools are needed to rapidly assess the efficacy of pulmonary tuberculosis (PTB) treatment. The aim of this study was to evaluate several immune biomarkers in an observational and cross-sectional cohort study conducted in Paraguay. Methods Thirty-two patients with clinically and microbiologically confirmed PTB were evaluated before starting treatment (T0), after 2 months of treatment (T1) and at the end of treatment (T2). At each timepoint plasma levels of IFN-y, 17 pro- and anti-inflammatory cytokines/chemokines and complement factors C1q, C3 and C4 were assessed in unstimulated and Mtb-specific stimulated whole blood samples using QuantiFERON-TB gold plus and recombinant Mycobacterium smegmatis heparin binding hemagglutinin (rmsHBHA) as stimulation antigen. Complete blood counts and liver enzyme assays were also evaluated and correlated with biomarker levels in plasma. Results In unstimulated plasma, C1q (P<0.001), C4 (P<0.001), hemoglobin (P<0.001), lymphocyte proportion (P<0.001) and absolute white blood cell count (P=0.01) were significantly higher in PTB patients at baseline than in cured patients. C1q and C4 levels were found to be related to Mycobacterium tuberculosis load in sputum. Finally, a combinatorial analysis identified a plasma host signature comprising the detection of C1q and IL-13 levels in response to rmsHBHA as a tool differentiating PTB patients from cured TB profiles, with an AUC of 0.92 (sensitivity 94% and specificity 79%). Conclusion This observational study provides new insights on host immune responses throughout anti-TB treatment and emphasizes the role of host C1q and HBHA-specific IL-13 response as surrogate plasma biomarkers for monitoring TB treatment efficacy.
... For T cell populations, T-helper 1 (Th1) cells control mycobacteria by secreting IFN-γ and triggering antimycobacterial responses of macrophages [32][33][34], while Th17 cells are responsible for neutrophilic inflammation, lung tissue damage [35], and improved survival [36]. While Th 17 cells demonstrate multifaceted functions during TB infection (microbial control versus lung injury), Th1 activation seems to be mainly beneficial through microbial control [37], and Th 2 induction might enhance the complications (fibrosis and cavitation) partly through IL-4 production [38]. However, the comparable abundances of Th1, Th2, and Th17 cells between TB cases (LTBI and PTB) and healthy control implied less influence of Th cells on mycobacterial control compared with B cells and NK T cells. ...
Article
Full-text available
It is unclear how the immune system controls the transition from latent tuberculosis (TB) infection (LTBI) to active pulmonary infection (PTB). Here, we applied mass spectrometry cytometry time-of-flight (CyTOF) analysis of peripheral blood mononuclear cells to compare the immunological landscapes in patients with high tuberculous bacillary load PTB infections and LTBI. A total of 32 subjects (PTB [n = 12], LTBI [n = 17], healthy volunteers [n = 3]) were included. Participants with active PTBs were phlebotomized before administering antituberculosis treatment, whereas participants with LTBI progressed to PTB at the time of household screening. In the present study, CyTOF analysis identified significantly higher percentages of mucosal-associated invariant natural killer T (MAIT NKT) cells in subjects with LTBI than in those with active PTB and healthy controls. Moreover, 6 of 17 (35%) subjects with LTBI progressed to active PTB (LTBI progression) and had higher proportions of MAIT NKT cells and early NKT cells than those without progression (LTBI non-progression). Subjects with LTBI progression also showed a tendency toward low B cell levels relative to other subject groups. In conclusion, MAIT NKT cells were substantially more prevalent in subjects with LTBI, particularly those with progression to active PTB.
... IL-1b and TGF-b are the major polarizing cytokines for Th17 differentiation. On the other hand, Th17 cells have extreme plasticity and have the ability to transdifferentiate into the pathogenic hybrid Th1-Th17 cells, under the action of IL-12 (69). Recently, these hybrid Th1-Th17 cells have been identified as major pathogenic players, in several autoimmune diseases (70). ...
Article
Full-text available
Tuberculosis patients with diabetes, have higher sputum bacillary load, delayed sputum conversion, higher rates of drug resistance, higher lung cavitary involvement and extra-pulmonary TB infection, which is called as “Diabetes-Tuberculosis Nexus”. However, recently we have shown a reciprocal relationship between latent tuberculosis infection and insulin resistance, which has not been reported before. In this review, we would first discuss about the immune-endocrine network, which operates during pre-diabetes and incipient diabetes and how it confers protection against LTBI. The ability of IR to augment anti-TB immunity and the immunomodulatory effect of LTBI to quench IR were discussed, under IR-LTB antagonism. The ability of diabetes to impair anti-TB immunity and ability of active TB to worsen glycemic control, were discussed under “Diabetes-Tuberculosis Synergy”. The concept of “Fighter Genes” and how they confer protection against TB but susceptibility to IR was elaborated. Finally, we conclude with an evolutionary perspective about how IR and LTBI co-evolved in endemic zones, and have explained the molecular basis of “IR-LTB” Antagonism” and “DM-TB Synergy”, from an evolutionary perspective.
... However, Th1 and Th17 cross-regulation is essential for an optimized response against M. tuberculosis. When Th17 cell responses become pathogenic rather than protective, Th1 cells are induced to stop these harmful effects through IFN-y [28,29]. ...
Article
Full-text available
Tuberculosis has a major global impact. Immunocompetent hosts usually control this disease, resulting in an asymptomatic latent tuberculosis infection (LTBI). Because TNF inhibitors increase the risk of tuberculosis reactivation, current guidelines recommend tuberculosis screening before starting any biologic drug, and chemoprophylaxis if LTBI is diagnosed. Available evidence from clinical trials and real-world studies suggests that IL-17 and IL-23 inhibitors do not increase the risk of tuberculosis reactivation. To evaluate psoriasis patients with treated or untreated newly diagnosed LTBI who received IL-17 and IL-23 inhibitors and the tolerability/safety of tuberculosis chemoprophylaxis. This is a retrospective, observational, multinational study from a series of 14 dermatology centres based in Portugal, Spain, Italy, Greece and Brazil, which included adult patients with moderate-to-severe chronic plaque psoriasis and newly diagnosed LTBI who were treated with IL-23 or IL-17 inhibitors between January 2015 and March 2022. LTBI was diagnosed in the case of tuberculin skin test and/or interferon gamma release assay positivity, according to local guideline, prior to initiating IL-23 or IL-17 inhibitor. Patients with prior diagnosis of LTBI (treated or untreated) or treated active infection were excluded. A total of 405 patients were included; complete/incomplete/no chemoprophylaxis was administered in 62.2, 10.1 and 27.7% of patients, respectively. The main reason for not receiving or interrupting chemoprophylaxis was perceived heightened risk of liver toxicity and hepatotoxicity, respectively. The mean duration of biological treatment was 32.87 ± 20.95 months, and only one case of active tuberculosis infection (ATBI) was observed, after 14 months of treatment with ixekizumab. The proportion of ATBI associated with ixekizumab was 1.64% [95% confidence interval (CI): 0–5.43%] and 0% for all other agents and 0.46% (95% CI 0–1.06%) and 0% for IL-17 and IL-23 inhibitors, respectively (not statistically significant). The risk of tuberculosis reactivation in patients with psoriasis and LTBI does not seem to increase with IL-17 or IL-23 inhibitors. IL-17 or IL-23 inhibitors should be preferred over TNF antagonists when concerns regarding tuberculosis reactivation exists. In patients with LTBI considered at high risk for developing complications related to chemoprophylaxis, this preventive strategy may be waived before initiating treatment with IL-17 inhibitors and especially IL-23 inhibitors.
... Together, they suggest a defective innate immune response against TB infection with poor anti-microbial functionality. Another important cytokine IL-12 is involved in T helper type 1 (Th1) differentiation and IFN-γ production, which is required to resist mycobacteria and other intracellular pathogens (Lyadova and Panteleev, 2015). However, in our study, we did not find any significant difference in the PTB + DM group as compared to other study groups. ...
Article
Full-text available
Vaccinations are vital as they protect us from various illness-causing agents. Despite all the advancements in vaccine-related research, developing improved and safer vaccines against devastating infectious diseases including Ebola, tuberculosis and acquired immune deficiency syndrome (AIDS) remains a significant challenge. In addition, some of the current human vaccines can cause adverse reactions in some individuals, which limits their use for massive vaccination program. Therefore, it is necessary to design optimal vaccine candidates that can elicit appropriate immune responses but do not induce side effects. Subunit vaccines are relatively safe for the vaccination of humans, but they are unable to trigger an optimal protective immune response without an adjuvant. Although different types of adjuvants have been used for the formulation of vaccines to fight pathogens that have high antigenic diversity, due to the toxicity and safety issues associated with human-specific adjuvants, there are only a few adjuvants that have been approved for the formulation of human vaccines. Recently, nanoparticles (NPs) have gain specific attention and are commonly used as adjuvants for vaccine development as well as for drug delivery due to their excellent immune modulation properties. This review will focus on the current state of adjuvants in vaccine development, the mechanisms of human-compatible adjuvants and future research directions. We hope this review will provide valuable information to discovery novel adjuvants and drug delivery systems for developing novel vaccines and treatments.
Article
Full-text available
Interleukin 17 (IL-17)-producing T helper cells (T(H)-17 cells) have been characterized in mice as a distinct subset of effector cells, but their identity and properties in humans remain elusive. We report here that expression of CCR6 and CCR4 together identified human memory CD4+ T cells selectively producing IL-17 and expressing mRNA encoding the human ortholog of mouse RORgammat, a transcription factor, whereas CCR6 and CXCR3 identified T(H)1 cells producing interferon-gamma and T helper cells producing both interferon-gamma and IL-17. Memory T cells specific for Candida albicans were present mainly in the CCR6+CCR4+ T(H)-17 subset, whereas memory T cells specific for Mycobacterium tuberculosis were present in CCR6+CXCR3+ T helper type 1 subset. The elicitation of IL-17 responses correlated with the capacity of C. albicans hyphae to stimulate antigen-presenting cells for the priming of T(H)-17 responses in vitro and for the production of IL-23 but not IL-12. Our results demonstrate that human T(H)-17 cells have distinct migratory capacity and antigenic specificities and establish a link between microbial products, T helper cell differentiation and homing in response to fungal antigens.
Article
Full-text available
Emerging evidence reveals that various cytokines and tissue microenvironments contribute to liver inflammation and autoimmunity, and IL-17 family is one of highlights acknowledged. Although the implication of IL-17 family in most common autoimmune diseases (such as psoriasis, inflammatory bowel disease, and rheumatoid arthritis) has been extensively characterized, the role of this critical family in pathophysiology of autoimmune liver diseases (AILD) still needs to be clarified. In the review, we look into the intriguing biology of IL-17 family and further dissect on the intricate role of IL-17-mediated pathway in AILD. Considering encouraging data from preclinical and clinical trials, IL-17 targeted therapy has shown promises in several certain autoimmune conditions. However, blocking IL-17-mediated pathway is just beginning, and more fully investigation and reflection are required. Taking together, targeting IL-17-mediated responses may open up new areas of potential clinical treatment for AILD.
Article
Full-text available
Significance The Th17 subset of CD4 ⁺ T cells are important in the pathogenesis of inflammatory bowel disease (IBD), but the mechanisms of their actions, particularly the role of the development of IFN-γ–producing progeny of Th17 cells (Th1-like cells), are incompletely understood. Here, we show in a mouse model of Th17-driven IBD that transition of Th17 precursors to Th1-like cells is absolutely required for disease, because Th17 cells deficient in IFN-γ fail to induce intestinal inflammation. This transition is dependent on the transcription factors T-bet and, to a lesser extent, Stat4. These findings are relevant for clinical strategies that target IBD and suggest that focusing on both the Th17 and Th1-like arms of disease may be beneficial in therapy design.
Article
Full-text available
The identification and treatment of individuals with tuberculosis (TB) is a global public health priority. Accurate diagnosis of pulmonary active TB (ATB) disease remains challenging and relies on extensive medical evaluation and detection of Mycobacterium tuberculosis (Mtb) in the patient's sputum. Further, the response to treatment is monitored by sputum culture conversion, which takes several weeks for results. Here, we sought to identify blood-based host biomarkers associated with ATB and hypothesized that immune activation markers on Mtb-specific CD4+ T cells would be associated with Mtb load in vivo and could thus provide a gauge of Mtb infection. Using polychromatic flow cytometry, we evaluated the expression of immune activation markers on Mtb-specific CD4+ T cells from individuals with asymptomatic latent Mtb infection (LTBI) and ATB as well as from ATB patients undergoing anti-TB treatment. Frequencies of Mtb-specific IFN-γ+CD4+ T cells that expressed immune activation markers CD38 and HLA-DR as well as intracellular proliferation marker Ki-67 were substantially higher in subjects with ATB compared with those with LTBI. These markers accurately classified ATB and LTBI status, with cutoff values of 18%, 60%, and 5% for CD38+IFN-γ+, HLA-DR+IFN-γ+, and Ki-67+IFN-γ+, respectively, with 100% specificity and greater than 96% sensitivity. These markers also distinguished individuals with untreated ATB from those who had successfully completed anti-TB treatment and correlated with decreasing mycobacterial loads during treatment. We have identified host blood-based biomarkers on Mtb-specific CD4+ T cells that discriminate between ATB and LTBI and provide a set of tools for monitoring treatment response and cure. Registration is not required for observational studies. This study was funded by Emory University, the NIH, and the Yerkes National Primate Center.
Article
There are still no reliable tests to distinguish active tuberculosis (TB) from latent TB infection (LTBI). Assessment of CD27 modulation on CD4(+) T-cells has been suggested as a tool to diagnose different TB stages. To use several cytometric approaches to evaluate CD27 expression on Mycobacterium tuberculosis (Mtb)-specific CD4(+) T-cells to differentiate TB stages. 55 HIV-uninfected subjects were enrolled: 13 active TB; 12 cured TB; 30 LTBI. Whole blood was stimulated with RD1-proteins or Cytomegalovirus-lysate (CMV). Interferon (IFN)-γ response was evaluated by cytometry. The proportion of CD27(-/+) within the IFN-γ(+) CD4(+) T-cells or RATIO of the CD27-median fluorescence intensity (MFI) of CD4(+) T-cells over the CD27 MFI of IFN-γ(+) CD4(+) T-cells was evaluated. The greatest diagnostic accuracy in discriminating active TB vs. LTBI or cured TB was reached by evaluating the CD27(+) CD45RA(-) cells within the IFN-γ(+) CD4(+) T-cell subset (76.92 sensitivity for both, and 90% and 91.67% specificity, respectively), although the use of the CD27 MFI RATIO allows for stricter data analysis, independent of the operator. the study of CD27 expression using different approaches, whether it involves evaluation of CD45RA expression or not, is a robust biomarker for discriminating TB stages. Copyright © 2015 The British Infection Association. Published by Elsevier Ltd. All rights reserved.
Article
Tuberculosis (TB), a chronic bacterial infectious disease caused by Mycobacterium tuberculosis (Mtb), typically affects the lung and causes profound morbidity and mortality rates worldwide. Recent advances in cellular immunology emphasize the complexity of myeloid cell subsets controlling TB inflammation. The specialization of myeloid cell subsets for particular immune processes has tailored their roles in protection and pathology. Among myeloid cells, dendritic cells (DCs) are essential for the induction of adaptive immunity, macrophages predominantly harbor Mtb within TB granulomas and polymorphonuclear neutrophils (PMNs) orchestrate lung damage. However, within each myeloid cell population, diverse phenotypes with unique functions are currently recognized, differentially influencing TB pneumonia and granuloma functionality. More recently, myeloid-derived suppressor cells (MDSCs) have been identified at the site of Mtb infection. Along with PMNs, MDSCs accumulate within the inflamed lung, interact with granuloma-residing cells and contribute to exuberant inflammation. In this review, we discuss the contribution of different myeloid cell subsets to inflammation in TB by highlighting their interactions with Mtb and their role in lung pathology. Uncovering the manifold nature of myeloid cells in TB pathogenesis will inform the development of future immune therapies aimed at tipping the inflammation balance to the benefit of the host. This article is protected by copyright. All rights reserved. This article is protected by copyright. All rights reserved.
Article
The expression of protective immunity to Mycobacterium tuberculosis in mice is mediated by T lymphocytes that secrete cytokines. These molecules then mediate a variety of roles, including the activation of parasitized host macrophages, and the recruitment of other mononuclear phagocytes to the site of the infection in order to initiate granuloma formation. Among these cytokines, interferon gamma (IFN-gamma) is believed to play a key role is these events. In confirmation of this hypothesis, we show in this study that mice in which the IFN-gamma gene has been disrupted were unable to contain or control a normally sublethal dose of M. tuberculosis, delivered either intravenously or aerogenically. In such mice, a progressive and widespread tissue destruction and necrosis, associated with very high numbers of acid-fast bacilli, was observed. In contrast, despite the lack of protective immunity, some DTH-like reactivity could still be elicited. These data, therefore, indicate that although IFN-gamma may not be needed for DTH expression, it plays a pivotal and essential role in protective cellular immunity to tuberculosis infection.
Article
The development of diagnostic tests that predict the progression of latent tuberculosis infection to active disease is pivotal for the eradication of tuberculosis. As an initial step to achieve this goal, our study's aim was to identify biomarkers that differentiate active from latent tuberculosis infection. We compared active and latent tuberculosis infection groups in terms of the precursor frequency, functional subset differentiation, and senescence/exhaustion surface marker expression of antigen-specific CD4(+) T cells, which were defined as dividing cells upon their encountering with Mycobacterium (M.) tuberculosis antigens. Among several parameters shown to have statistically significant differences between the two groups, the frequency of CD57-expressing cells could differentiate effectively between active disease and latent infection. Our results suggest that the expression of CD57 in M. tuberculosis-reactive CD4(+) T cells could be a promising candidate biomarker with which to identify individuals with latent tuberculosis infection prone to progression to active disease. Copyright © 2015. Published by Elsevier Inc.
Article
For over 35 years, immunologists have divided T-helper (TH) cells into functional subsets. T-helper type 1 (TH1) cells - long thought to mediate tissue damage - might be involved in the initiation of damage, but they do not sustain or play a decisive role in many commonly studied models of autoimmunity, allergy and microbial immunity. A major role for the cytokine interleukin-17 (IL-17) has now been described in various models of immune-mediated tissue injury, including organ-specific autoimmunity in the brain, heart, synovium and intestines, allergic disorders of the lung and skin, and microbial infections of the intestines and the nervous system. A pathway named TH17 is now credited for causing and sustaining tissue damage in these diverse situations. The TH1 pathway antagonizes the T H17 pathway in an intricate fashion. The evolution of our understanding of the TH17 pathway illuminates a shift in immunologists' perspectives regarding the basis of tissue damage, where for over 20 years the role of TH1 cells was considered paramount.