Vous êtes sur la page 1sur 10

Cell Host & Microbe

Review

Tuberculosis in Africa: Learning from


Pathogenesis for Biomarker Identification
Stefan H.E. Kaufmann1,* and Shreemanta K. Parida1
1Max Planck Institute for Infection Biology, Department of Immunology, Charitéplatz 1, D-10117 Berlin, Germany

*Correspondence: kaufmann@mpiib-berlin.mpg.de
DOI 10.1016/j.chom.2008.08.002

In Africa, more than 4 million people suffer from active tuberculosis (TB) resulting in an estimated 650,000
deaths every year. The etiologic agent of TB, Mycobacterium tuberculosis, survives in resting macrophages,
which control the pathogen after activation by specific T lymphocytes. Here, we describe the basic mecha-
nisms underlying the host response to TB with an emphasis on immunity and discuss diagnostics, drugs, and
vaccines for TB. Moreover, we outline our attempts to develop biomarkers, which could help the monitoring
of TB clinical trials, provide the basis for new diagnostics, and allow prognosis of outcome of infection and of
drug treatment.

Introduction research leading to the discovery of Mtb as the etiologic agent


Although tuberculosis (TB) is a health threat of global dimension, more than 125 years ago, the development of the tuberculin
it is most on the rampage in Africa with a focus in the Sub- skin test almost 120 years ago, both by Robert Koch, and the de-
Saharan part of the continent (WHO, 2008). Between 1990 and velopment of a vaccine against TB named after its discoverers,
2006, TB prevalence in Africa rose from 1.7 million to 4.2 million bacille Calmette-Guérin (BCG) ca. 80 years ago (Kaufmann
active cases with no signs of relief, as dramatically illustrated by and Winau, 2005; Kaufmann and Parida, 2007). All these mea-
the more than three-fold increase in new cases from 800,000 in sures are still in use today. It is no wonder that they cannot satisfy
1990 to 2.8 million in 2006. Much of this problem is due to the our state-of-the-art demands. The acid-fast staining of Mtb is the
human immunodeficiency virus/acquired immune deficiency basis of sputum diagnosis, which in Africa detects less than half
syndrome (HIV/AIDS) plague as illustrated by the enormous of all patients with active TB (Box S1 available online). The tuber-
HIV prevalence among TB patients of 22% (Table 1). In today’s culin skin test forms the basis for population-wide assessment of
Africa, Mycobacterium tuberculosis (Mtb) is the number one killer infection with Mtb but cannot distinguish between latent infec-
of HIV-infected individuals with one third of the 640,000 deaths tion and active disease (Lalvani, 2007) (Box S1). The vaccine
due to TB occurring in HIV-coinfected sufferers. Better health BCG protects newborns and young children from developing
care ranging from prevention to therapy for TB patients is ur- severe disseminated TB (miliary TB) but cannot protect against
gently required. Because of the high coincidence of TB and adult pulmonary TB, which today is the most prevalent form of
AIDS, Mtb and HIV can no longer be viewed exclusively by them- the disease (Kaufmann, 2006; Fine, 1995). Accordingly, this vac-
selves but must also be seen as two diseases in a single patient cine has virtually no impact on transmission of TB (Corbett et al.,
(Kaufmann and McMichael, 2005). The sufferers of HIV–Mtb 2003). Most drugs used to treat TB were developed between
coinfection need particular care since, historically, they have 1950 and 1970 (Box S2). The need for 3 to 4 drugs over a period
often been treated at different health facilities by different health of 6 to 9 months—combined with poor compliance—has led to
care providers. To make the situation worse, we are faced with the emergence of single drug-resistant, multidrug-resistant
an increasing occurrence of immune reconstitution inflammatory (MDR), and extensively drug-resistant (XDR) strains of Mtb. An
syndrome (IRIS) in HIV–Mtb-coinfected individuals under antire- effective response to this increasing threat requires new drugs
troviral therapy (ART), which exacerbates TB reactivation. targeting novel structures and pathways in Mtb that are not con-
It has occasionally been proposed that efforts should be con- quered by current drugs. Without doubt, better housing condi-
centrated on prevention and therapy of HIV/AIDS and that TB tions for those living in townships and densely crowded parts
would disappear once HIV control has been successfully imple- of cities in Sub-Saharan Africa and improved food and nutrient
mented. Unfortunately, the different modes of transmission of supply are needed to raise socioeconomic standards (Schaible
HIV and Mtb make the success of such a proposal unlikely. and Kaufmann, 2007). Yet, in a continent where a large propor-
HIV is primarily transmitted by sexual intercourse and thus can tion of the inhabitants live on less than $2 US a day, it is unlikely
be prevented to a large degree by changes in behavior such as that these goals will be achieved in the near future. They need to
abstinence or use of condoms accompanied by appropriate be complemented by better medical intervention measures.
education. In contrast, behavioral changes are largely inefficient In the short-term, broad coverage of BCG vaccination of
for TB because the pathogen is transmitted by aerosols, and it is newborns and complete coverage of TB drug supply, along
simply impossible to stop breathing. with better diagnostic tools in the field, are needed to face the
In some regions of Sub-Saharan Africa, TB has reached a level threat. In the long-term, satisfactory control will depend on an
reminiscent of that of European cities at the turn of the last cen- efficacious vaccine to prevent pulmonary TB in adults and trans-
tury. The European TB situation improved not only due to better mission of Mtb (Kaufmann, 2006; Kaufmann and McMichael,
living conditions. These were also the times of highly active TB 2005). A similar long-term strategy is hoped for HIV, namely

Cell Host & Microbe 4, September 11, 2008 ª2008 Elsevier Inc. 219
Cell Host & Microbe

Review

Table 1. TB and HIV/AIDS-TB in Africa and in African States, Where GC6-74 Studies Are Being Performed
Incidence of All Incidence of All Prevalence of All HIV Prevalence
Forms of TB 2006 Forms of TB 2006 Forms of TB in Incident TB Population
Nation (Numbers) (Rate per 100,000) (per 100,000) Cases (%) (Millions)
Ethiopia 306,330 379 643 6.3 81
Gambia 4,278 257 423 7.5 2
Malawi 51,172 377 322 70 14
South Africa 453,929 940 998 44 50
Uganda 106,037 355 561 16 30
Total Africa 2,807,688(921,746)a 363 547 22 775
Total Global 9,157,021 139 219 7.7 6,590
Source: (WHO, 2008).
a
Total of five countries depicted in Table 1.

development of a vaccine that provides protection (Kaufmann S3). The countries involved are all severely affected by Mtb/TB
and McMichael, 2005). However, despite major efforts to de- and HIV/AIDS. TB incidences range from 940 per 100,000 in
velop an efficacious HIV vaccine, this goal seems to be more dis- South Africa as the highest to 257 per 100,000 in The Gambia
tant than ever (Lagakos and Gable, 2008; Walker and Burton, as the lowest, and from 70% HIV in TB patients as the highest
2008; Boasso et al., 2008). Although the situation for developing in Malawi to 6.3% in Ethiopia (See Table 1). Newly diagnosed
a successful TB vaccine is improving, much still needs to be acute pulmonary TB patients are the entry point of our studies,
done (Kaufmann, 2006). and their healthy household contacts infected with Mtb as evi-
denced by positive tuberculin skin testing are the focus of our
Why Biomarkers for TB? studies. Several cohorts have been included in longitudinal pro-
We have argued that the vast majority (approx. 80% in Sub- spective case control studies spanning a follow-up period of
Saharan Africa) of all who become infected with Mtb will control 2 years from the time of recruitment. An integrated interdisciplin-
the pathogen lifelong without developing clinical signs of dis- ary immunologic and multiomics approach has been adopted
ease. Thus, the host immune system has mechanisms at hand for blood leukocytes from both HIV ve and HIV+ve subjects.
to control the pathogen effectively although it only rarely seems HIV+ve cohorts (with and without active TB disease) are con-
to achieve sterile eradication of Mtb (McKinney et al., 1998). Will founded by several factors such as the use of chemoprophylaxis
it be possible to learn from this naturally induced host response and ART (particularly in subjects without active TB) that would in-
and better understand those mechanisms that control infection fluence the outcome of TB (re)activation. Nevertheless, they pro-
and exploit their value as potential indicators of protection? If vide a trend in comparison to the HIV ve cohorts in the same
so, then a biomarker of protection against TB could be defined population.
and serve as a guideline for monitoring vaccine-induced immu- A major goal of our strategy is the definition of a surrogate
nity. Such a biomarker of protection against disease could end-point for a phase II/III clinical trial with novel TB vaccines
provide valuable information about vaccine efficacy prior to the and perhaps also with new drugs against TB. Several TB vac-
clinical end-point of TB disease outbreak. In the area of clinical cines are due to enter, or have already completed, clinical phase
trials, a biomarker is defined as a characteristic feature that is I trials and a few candidates have reached phase II trials (Box
objectively measured and evaluated as an indicator of a normal S4). Some are expected to enter phase III clinical trials in the
or a pathological process or of the response to an intervention near future. The greatest obstacle will thus come at the end of
(Biomarkers Definitions Working Group, 2001). Thus, our goals, the research-and-development pipeline with a phase III clinical
to identify biomarkers of protection and susceptibility to natural trial aimed at definitively determining protective vaccine efficacy
infection with Mtb leading to the adoption of a marker of in adults. Currently, the success of a vaccine in a phase III clinical
vaccine-induced immunity against TB, fall into this definition trial is measured by the prevention of active TB outbreak in study
(Table 2). group participants. In other words, clinical diagnosis of TB dis-
With support from the Bill & Melinda Gates Foundation under ease serves as the clinical end-point of such a trial. Even with
the Grand Challenges in Global Health program (http://www. a TB incidence rate of 1 in 100 as seen in the worst-affected parts
gcgh.org/NewVaccines/Challenges/LearnaboutImmunological of South Africa and Swaziland, large groups of individuals would
Responses/Pages/default.aspx), we attempt to define bio- need to be enrolled to reach adequate power in the study to ob-
markers for TB in the context of HIV/AIDS in five African nations, tain conclusive evidence. Added to this is the long incubation
namely, Ethiopia, The Gambia, Malawi, South Africa, and time of latent Mtb infection ranging from months to lifelong.
Uganda (http://www.biomarkers-for-tb.net/). This collaborative About 10%–20% of those who will become infected with Mtb
project falls under the broader thematic goal ‘‘To Create New during a phase III trial in Africa will develop active TB at a later
Vaccines’’ among seven major goal-oriented themes and specif- time point with 5%–10% probably occurring within the first
ically under Grand Challenge 6 (GC6) out of the 14 defined Grand 2 years. Thus, it has been estimated that clinical trials for TB vac-
Challenges topics: ‘‘Learn which immunological responses pro- cines may last several years (possibly a decade if not longer)
vide protective immunity’’; referred as GC6-74 hereafter (Box comprising thousands of study subjects. In view of this grave

220 Cell Host & Microbe 4, September 11, 2008 ª2008 Elsevier Inc.
Cell Host & Microbe

Review

Table 2. Biomarker Definitions in the lung parenchyma. Monocytes and granulocytes are at-
tracted to the site of bacterial deposition in the lung parenchyma
Term Definition
where accumulating cells form loosely packed lesions. In the
Biomarker Characteristic that is objectively measured
draining lymph nodes, T lymphocytes with specificity for myco-
and evaluated as an indicator of normal
biological processes, pathological processes,
bacterial antigens are stimulated. Antigen-specific T cells, by
or physiological/pharmacological responses way of their recirculation and tissue surveillance, recognize
to an intervention. Mtb-infected macrophages and DCs in lung lesions and induce
Correlate of Measurable sign in a host in response to formation of productive granulomas where Mtb is contained
protection an infectious agent indicating whether the and thus prevented from causing active disease (Ulrichs and
individual is being protected against Kaufmann, 2006; Russell, 2007). Different combinations of che-
becoming infected and/or developing mokines produced by cells in the inflamed and infected area
disease. attract different leukocyte populations to the site of action (Charo
Surrogate of Validated marker of correlate of protection. and Ransohoff, 2006; Li and Flavell, 2008; Wolf et al., 2008). Re-
protection ciprocally, cells that respond to the chemokine gradient need to
Clinical endpoint Characteristic or variable that reflects the express the homologous set of chemokine receptors (Charo and
final outcome of disease in terms of function, Ransohoff, 2006).
effect, progress, recovery, survival, or death.
Surrogate endpoint Biomarker that is intended to substitute for a T Cell Subsets and Their Cytokines
clinical endpoint, predicting clinical outcome Activated T lymphocytes comprise CD4 T cells and CD8 T cells
in terms of benefit, or harm, or lack of benefit as well as unconventional T cells, such as the gd T lymphocytes
or harm. and the CD1-restricted T lymphocytes (Flynn and Chan, 2001).
Source: (Biomarkers Definitions Working Group, 2001; Colburn, 2000). The CD4 T cells that are restricted by gene products of the major
histocompatibility complex (MHC) class II represent major
players in protection against TB (Figure 2). Of greatest relevance
situation, biomarkers could help to accelerate the development are the T helper (Th) cells that produce interferon gamma (IFN-g),
process at different stages of clinical trials. which have been termed Th1 cells. IFN-g activates macro-
Even before a phase III clinical trial, biomarkers that provide phages (Nathan et al., 1983). As a consequence, the resting
robust information for educated decisions about entry into, or macrophage, which primarily serves as a fertile habitat for Mtb,
termination of, phase II clinical vaccine trials as well as informa- is transformed into an activated macrophage that can control
tion for decisions on combinations of different vaccine candi- replication of Mtb. Still, even activated macrophages fail to erad-
dates in heterologous prime-boost schemes would be extraordi- icate their predators (Vandal et al., 2008). Thus, the macrophage
narily helpful. As an added value, information on biomarkers for serves as both host and effector cell. Recent evidence suggests
TB can provide the basis for (1) novel diagnostic tools to more that interleukin (IL) 17-producing Th cells (so-called Th17 cells)
reliably diagnose active TB (Box S1), (2) prognostic markers of participate in protection against TB, although their role is less
infection outcome, i.e., TB disease reactivation or latent infec- clear (Khader et al., 2007). Probably they provide a first line of
tion, and (3) biomarkers that can predict drug treatment out- defense against Mtb and participate in the early steps of granu-
come, i.e., treatment success, drug failure, or relapse (Figure 1). loma formation (Ouyang et al., 2008).
The CD8 T cells recognize mycobacterial antigens in the con-
From the Site of Infection and Disease text of MHC class I and contribute to protection first by produc-
to the Peripheral Blood ing cytokines, notably IFN-g, and second by direct cytolytic and
In the following we will describe our rationale for gathering infor- mycobacteriolytic mechanisms. Accordingly, they have been
mation about the host response in naturally infected individuals termed cytolytic T lymphocytes (CTLs). Molecules involved in
for definition of biomarkers in TB. these CTL mechanisms include granzymes and perforins. The
In Africa, almost 60% of all TB cases are sputum smear neg- gd T cells recognize phosphorylated ligands in apparent ab-
ative (WHO, 2008). This not only represents a severe impediment sence of restriction elements and the CD1-restricted T cells
to active case finding and hence for efficacious drug treatment, are specific for mycobacterial glycolipids, such as lipoarabino-
but it also suggests that there is a high proportion of extrapulmo- mannan (LAM) and mycolic acids (Figure 2). The role of these
nary TB (WHO, 2008). Extrapulmonary TB can progress to a gen- unconventional T cells in protective immunity is unclear. It is
eralized form such as miliary TB in AIDS patients or be confined most likely that they serve as back-up cells producing similar
to distinct organs with the most prevalent and deadly form being cytokines as the CD4 Th1 and Th17 cells as well as the
meningeal TB (Maher, 2008). Yet, in the majority of cases, includ- CD8 CTLs.
ing all sputum smear-positive and a significant proportion of The Th1 cells produce additional cytokines, notably IL-2,
smear-negative cases, Mtb resides in the lung, which therefore which promotes stimulation of both Th1 cells and CTLs, lympho-
serves as the origin of transmission via aerosols. Enclosed in toxin (LT), tumor necrosis factor alpha (TNF-a), which synergizes
minute droplets, bacteria are expelled by coughing of an active with IFN-g in macrophage activation, and granulocyte-macro-
pulmonary TB patient and in this way enter lung alveoli of other phage colony-stimulating factor (GM-CSF), which stimulates
individuals. There, alveolar macrophages, as well as interstitial granulocyte and macrophage maturation (Figure 2). The Th17
dendritic cells (DCs), engulf Mtb (Figure 1). Some bacteria are cells also produce IL-6 as proinflammatory mediator as well as
transported to draining lymph nodes while others are trapped IL-22, which stimulates defensin production in epithelial cells

Cell Host & Microbe 4, September 11, 2008 ª2008 Elsevier Inc. 221
Cell Host & Microbe

Review

Figure 1. From the Site of Action in the Lung to Biomarkers in the Peripheral Blood
The figure depicts the major pathologic and protective mechanisms in the lung during latent Mtb infection and active TB disease. In addition, the picture indicates
determination of immunologic, transcriptomic, and metabolomic biomarkers.

(McGeachy and Cua, 2008; Ouyang et al., 2008). Another Th cell Antigen Processing/Presentation
population, the Th2 cells, produce B cell-stimulating factors, no- Mtb resides in the early phagosome and prevents it from further
tably IL-4 and IL-5, and hence promote antibody production; yet, maturation (Armstrong and Hart, 1971). Thus, mycobacterial an-
the role of antibodies produced by plasma cells as progeny of tigens can be loaded on MHC-II molecules, which then present
activated B cells in protection against TB is minor. CD4 T cells the antigenic peptides to CD4 T cells. The MHC-I molecules
can also produce cytokines that impair immunity to TB such as are loaded with antigens in the cytosolic compartment (Figure 2).
IL-4, produced by Th2 cells as well as IL-10 and tumor growth Because Mtb persists within the phagosome, efficient loading of
factor beta (TGF-b) produced by regulatory T (Treg) cells (Belkaid MHC-I molecules remained a mystery until recently. Recent find-
and Rouse, 2005). Some cytokines such as IL-21 that are pro- ings indicate egress of Mtb into the cytosol (van der Wel et al.,
duced by different CD4 T cell sets express pleiotropic activities 2007); however this issue is still controversial. Evidence from
on various T cell populations. Phenotype studies using knockout our laboratory suggests that macrophages infected with Mtb
mutant mice or blocking agent(s) for cytokines represent a reduc- undergo apoptosis. This leads to the formation of vesicles
tionist approach in a well-defined model system. In contrast, containing mycobacterial antigens. DCs in the vicinity of such
in vivo activities of multiple intertwined processes, which pro- apoptotic macrophages engulf these vesicles and present the
ceed concurrently, have to be addressed using a global systems antigens in the context of MHC-II, MHC-I, and CD1 in a highly
biology approach. Obviously, characterization of these different efficacious way (Schaible et al., 2003; Winau et al., 2006). This
Th cell populations and their cytokine patterns provide promising process, termed crosspriming, probably plays a major role in
candidates for biomarker research. Recent findings suggest that T cell stimulation during Mtb infection.
polyfunctional T cells, which produce multiple cytokines such
as IFN-g, IL-2, TNF-a, LT, and/or GM-CSF, are best suited as T Cell Polarization by Cytokines
biomarker candidates for protection in TB (Seder et al., 2008; Polarization into different T cell sets is mostly dictated by distinct
Mueller et al., 2008). These polyfunctional T cells probably repre- cytokines (Figure 2) (Kaufmann, 2007; Keir et al., 2008). Thus,
sent T memory (Tm) cells, which sustain protective immunity TGF-b, together with IL-6, promotes development of Th17 cells
(Sallusto et al., 2004). Treg cells are interesting candidates for in mice while in humans IL-21 replaces IL-6 (Yang et al., 2008).
biomarker research as well because their increase may correlate IL-1b, IL-6, and IL-23 can further support growth of Th17 cells.
with disease reactivation as a consequence of dampened IL-12, IL-18, and IFN-g promote Th1 cell development. IL-4 as
immune responses (Belkaid and Rouse, 2005). well as IL-25 and IL-33 induce Th2 cell development. TGF-b, in

222 Cell Host & Microbe 4, September 11, 2008 ª2008 Elsevier Inc.
Cell Host & Microbe

Review

Figure 2. Three Steps to T Cell Activation


The figure shows three critical steps toward T cell activation and polarization.
(1) T cells recognize antigen not in its natural form; rather the antigen is presented by reference structures encoded by the major histocompatibility complex
(MHC). Antigens that reside in phagosomes are presented by MHC-II molecules to CD4 T cells. Because Mtb resides in phagosomes, this is the main antigen
presentation/recognition pathway in TB. Antigens in the cytosol are loaded onto MHC-I molecules that present their antigens to CD8 T cells. Unconventional
T cells comprising gd T cells and CD1-restricted T cells are activated during Mtb infection, as well. Human gd T cells recognize small molecules containing
pyrophosphate residues, also termed phospholigands. The CD1-restricted T cells recognize mycobacterial glycolipids in the context of CD1 molecules.
(2) CD4 T cells can be further subdivided into different T helper (Th) cells according to their cytokine expression pattern. The Th1 cells are critical for protection
against TB. They typically produce interferon gamma (IFN-g), tumor necrosis factor alpha (TNF-a), lymphotoxin (LT), interleukin (IL)-2, and granulocyte-macro-
phage colony-stimulating factor (GM-CSF). The Th2 cells stimulate humoral immune responses via secretion of IL-4 and IL-5. The Th17 cells produce IL-6, IL-17,
IL-21, and IL-22, which probably contribute to early protection. The regulatory T cells (Treg) produce tumor growth factor beta (TGF-b) and IL-10, which suppress
immune responses. Cytokines are also important for polarization of CD4 Th cells. Thus, IL-12, IL-18, and IFN-g promote Th1 cell development; IL-33 together
with IL-4 and IL-25 induce Th2 cell development; TGF-b, together with IL-6, promotes Th17 cell polarization in mice and TGF-b and IL-21 do so in humans, and
TGF-b together with vitamin A derivates (retinoic acid, RA) favors Treg cell maturation.
(3) For complete activation, T cells need to be costimulated by surface molecules. Many of these molecules belong to the B7 family. Thus, interactions of CD28
on T cells with B7.1 and/or B7.2 on antigen-presenting cells (APCs) promote T cell stimulation as do interactions of the inducible T cell costimulator (ICOS) on
activated T cells with ICOSL (B7.h) on APCs. Other mechanisms are inhibitory such as interactions of B7.1 and B7.2 with CTLA-4 and interactions with PD-1 on
activated T cells with PD-L on APCs.

combination with molecules other than cytokines, such as vita- of the B7 family, notably B7.1 (CD80) and B7.2 (CD86)
min A derivates, favors development of Treg cells that can be fur- expressed on the surface of antigen-presenting cells (APCs),
ther subdivided into regulatory subsets. Additional control is are costimulatory molecules that interact with CD28 on T cells
achieved by counteractive effects of cytokines: IFN-g promotes and in this way initiate T cell activation. In contrast, CTLA-4,
Th1 cells and blocks Th2 cells whereas IL-4 promotes Th2 cells which is also recognized by B7.1 and B7.2, inhibits T cell acti-
but blocks Th1 cells. IL-27 favors Treg cells and impairs Th17 vation. The crosstalk between CD40 on APCs and CD40 ligand
cells (Bettelli et al., 2008). (CD40L, CD154) on T cells can contribute to T cell activation,
and the inducible T cell costimulator (ICOS) expressed on acti-
T Cell Activation by Costimulatory Molecules vated T cells sustains the activation status when it interacts
T cell activation is regulated by costimulatory molecules with ICOS-L (B7.h) on APCs. In contrast, the programmed
(Hutloff et al., 1999; Keir et al., 2008) (Figure 2). Thus, members death 1 molecule (PD-1) causes exhaustion of activated

Cell Host & Microbe 4, September 11, 2008 ª2008 Elsevier Inc. 223
Cell Host & Microbe

Review

T cells (primarily CD8 T cells) when it reacts with its ligand PD-L gene-expression profiles revealed overlaps as well as unique
on APCs. expression profiles at the different sites. Our findings strongly
support the notion that in patients suffering from active TB, dif-
Instruction of Acquired Immunity by Innate Immunity ferent disease entities exist, ranging from dormant Mtb in pro-
Surface expression of the different accessory molecules and ductive granulomas to active Mtb in cavitary lesions. Currently
production of the various cytokines that determine activation available drugs primarily affect metabolically active Mtb, and it
and polarization of T cell populations are strongly influenced has been speculated that after eradication of these active popu-
by the type of the invading pathogen (Pulendran and Ahmed, lations the dormant population is resuscitated and continues
2006). Thus, the innate immune system is critical for instructing the vicious cycle: a new population of Mtb organisms grows to
the acquired immune response so that it is best tailored to coun- enormous numbers and forms new lesions. This differential
teract the unique survival stratagem of the invading pathogen. In drug susceptibility of Mtb during different stages of its life cycle,
the case of Mtb, members of the toll-like receptor family (TLR) which coexist in a patient with active TB disease, is one reason
recognize CpG motifs of bacterial DNA as general molecular for the prolonged treatment time of at least 6 months required
patterns for bacteria, and lipoproteins and glycolipids as more to cure TB (Box S2).
specific characteristic patterns for mycobacteria. The respective It will be important to understand whether during latent infec-
TLR members, TLR-9, TLR-2, and TLR-4, are central to the tion, protective T cells respond to antigens representing the met-
buildup of an effective immune response against Mtb (Flynn abolically active stage of Mtb, the dormant stage, or both stages.
and Chan, 2001). It will be equally important to determine the immune mechanisms
that precede reactivation. Do Treg cells arise prior to reactivation
The Life Cycle of Mtb in Infected Individuals and if so, which antigens do they recognize? Do effector and/or
During latent infection, Mtb is contained in productive granulo- Tm cells become exhausted, e.g., through PD-1–PD-L interac-
mas where it transforms into a dormant state (Figure 1). It is be- tions? There is experimental evidence for both types of immuno-
lieved that the bacilli are in a state of nonreplicating persistence suppression in TB (Kursar et al., 2007; Jurado et al., 2008; Hegasy
although recent observations suggest that they may indeed et al., unpublished data). Moreover, it will be interesting to know
slowly replicate (Manabe and Bishai, 2000; Munoz-Elias et al., whether latent Mtb infection is sustained by effector T cells that
2005). In vitro studies have provided compelling evidence that recognize the antigens expressed during latency, i.e., dormancy
in this state, Mtb expresses a distinct gene expression program and starvation gene products, or whether protection is sustained
characterized by upregulation of DosR genes and starvation by Tm cells that, for example, recognize antigens expressed early
genes (Voskuil et al., 2003; Rosenkrands et al., 2002; Betts after host invasion by Mtb such as the secreted antigens Antigen
et al., 2002). 85 and ESAT-6 (Demissie et al., 2006). Ottenhoff and coworkers
TB becomes clinically apparent either when dormant Mtb have determined human T cell responses to DosR-controlled
organisms are resuscitated to cause disease reactivation or sub- antigens (Leyten et al., 2006; Lin et al., 2007). Recent work from
sequent to reinfection with Mtb. Disease reactivation is paral- our group using sensitive T cell stimulation assays revealed re-
leled by transformation of the Mtb gene-expression profile sponses to subdominant latency antigens of Mtb allowing dis-
from a dormant to a metabolically active type. It is likely that crimination of latently infected individuals from active TB patients
this latter expression profile resembles that of Mtb when it enters (Schuck et al., unpublished data). Within GC6-74, a pilot study
the host. With the onset of full-blown TB disease, granulomas liq- has analyzed T cell responses at different sites in Africa to dor-
uefy and Mtb organisms flourish in the caseous cellular detritus mancy and starvation antigens and selected the most potent
to numbers as high as 1012 or more bacteria per lesion (Canetti, ones for further longitudinal studies (GC6 Consortium, unpub-
1965). Yet, it is likely that even during active disease some lished data). Such analyses will determine the value of antigens
lesions remain productive and succeed in containing Mtb in expressed during dormancy for monitoring of immune responses
a dormant state. Hence, the lung of a TB patient may comprise during progression of latent infection to active TB disease.
different independent entities of infection ranging from minute Determination of antigen-specific responses is further compli-
productive granulomas containing few dormant mycobacteria cated by prevalence of distinct lineages of Mtb in different parts
to large cavities housing an enormous amount of highly active of the world (Gagneux and Small, 2007). These lineages seem
Mtb microorganisms. Probably, even a single lesion can contain to express different degrees of virulence. Accordingly, different
Mtb in different metabolic states. lineages and even sublineages or strains may express different
The global gene-expression profile of Mtb residing in different antigen expression patterns. Preliminary evidence obtained
sites of lungs from patients suffering from severe TB has been within GC6-74 indeed indicates that the different Mtb isolates
determined (Fenhalls et al., 2002; Rachman et al., 2006). Overall, prevalent at field sites of our collaborative project show marked
in our study more than 20% of the genome of Mtb was differen- differences at the transcription level under different in vitro con-
tially regulated in vivo as compared to in vitro-cultured samples ditions mimicking the natural history of the disease (Schoolnik
of the same Mtb microbes (Rachman et al., 2006). These genes et al., unpublished data). Further studies are in progress to
were primarily involved in (1) cell wall synthesis, (2) energy pro- determine the implications of these observations in terms of
duction through fatty acid degradation under microaerophilic Mtb-specific immune responses.
conditions, and (3) active synthesis of proteins involved in eva-
sion and fortification of Mtb. Subsequent analyses compared Immunologic Biomarkers in TB
Mtb (1) in the center of caseous lesions, (2) in the pericavity, All of these questions are important for biomarker research
and (3) at distant sites of the lung (Rachman et al., 2006). These focusing on immunologic mechanisms and phenotypes.

224 Cell Host & Microbe 4, September 11, 2008 ª2008 Elsevier Inc.
Cell Host & Microbe

Review

Obviously, concomitant determination of multiple cytokines (no- bial host molecules, (2) molecules involved in inflammation, (3)
tably IL-2, IFN-g, TNF-a, GM-CSF) and killer molecules (perforin, chemokines, and (4) molecules responsible for vesicle trafficking
granulysin), the phenotype of the T cells (CD4, CD8), expression (Jacobsen et al., 2007). Examples of differentially expressed
of memory markers, and T cell homing (various chemokine genes are formyl peptide receptor, bacteriocidal permeability
receptors notably CCR7) and adhesion molecules (notably increasing protein, lactoferrin, a-defensin 1, a-defensin 4,
CD62L) should be considered for multiplex profiling of immuno- CD64 (Fcg receptor 1 with high affinity), and Rab33A (Jacobsen
logic biomarkers (Figure 1). This functional assessment should et al., 2007, 2005). After appropriate deconfounding, linear dis-
be combined with concomitant determination of specificity for criminant analysis identified three genes among the ca. 30,000
a large panel of mycobacterial antigens characteristic of dor- genes of the human genome that allow discrimination between
mant and metabolically active Mtb. In an ideal world, these healthy latently infected individuals and active TB patients
assays would be performed by multicolor fluorescence-acti- (Jacobsen et al., 2006). These were: Rab33A, lactoferrin, and
vated cell sorting (FACS) analysis using antibodies for critical CD64. Hence, design of a custom-made transcriptome chip or
cell-surface molecules and cytokines combined with tetramer multiplex ligation-dependent probe amplification (MLPA) for TB
constructs presenting defined mycobacterial epitopes. A more measuring a restricted number of highly differentially expressed
realistic approach would be the combination of whole blood genes appears feasible (Box S6). We considered this observa-
cell assays using a panel of mycobacterial antigens for T cell tion sufficiently convincing to pursue a large transcriptome
restimulation (Black et al., unpublished data) with multiplex cyto- analysis within GC6-74.
kine determinations by commercially available assay systems, Two of the three genes identified, lactoferrin and CD64, may
such as the Luminex system, which allows determination of up be of little surprise for those involved in TB research. Lactoferrin
to 100 different cytokines (Box S5) (Vignali, 2000). is a transporter molecule with high affinity for iron that allows the
host to efficiently compete with Mtb for iron (Schaible and Kauf-
Regional or Central Immune Regulation in TB? mann, 2005). In a mouse model, lactoferrin has been shown to
Recent studies have revealed the emergence of lymphoid struc- reverse a high susceptibility to TB by correcting the iron overload
tures in the vicinity of granulomas in the lungs of patients suffer- (Schaible et al., 2002). CD64, the Fcg receptor with high affinity,
ing from TB (Ulrichs and Kaufmann, 2006) (Figure 1). In these is upregulated by IFN-g stimulation and may contribute to host
experiments, tissue from patients who had undergone surgical defense against TB by stimulating the respiratory burst during
resection of affected lung areas due to excessive multidrug- uptake of Mtb (Perussia et al., 1983; van de Winkel and Capel,
resistant (MDR)-TB were analyzed by immunohistology (Ulrichs 1993).
et al., 2004). These analyses revealed evidence for tertiary lym- More surprising was the identification of Rab33A as a discrim-
phoid structures in the vicinity of granulomatous lesions com- inating factor. This molecule is a member of the Rab family of
prising DCs, B cells, T cells, and macrophages. Consistent small GTPases. Although we were originally puzzled by the func-
with these observations, experimental studies with influenza tional role of this molecule in TB, more recent experiments (Tsu-
virus-infected mice have revealed that the infected lung can as- boi and Fukuda, 2006) identified Rab33A, together with other
sume functions of secondary lymphoid organs (Moyron-Quiroz Rab molecules, in dense core vesicles. In our studies, differential
et al., 2006; Randall et al., 2008). Most recent studies from our expression of Rab33A was restricted to CD8 T cells (Jacobsen
laboratory provide strong evidence that granulomas from Mtb- et al., 2007). Our current working hypothesis, therefore, is that
infected mice serve as tertiary lymphoid structures capable of Rab33A is involved in the release of lytic molecules by CTLs.
inducing and sustaining T cell responses to mycobacterial and As described above, such molecules comprise perforin and
bystander antigens (Walker et al., unpublished data). It will be granulysin, which have been implicated in lysis of host cells
important to determine whether such local immune responses and killing of Mtb (Stenger et al., 1998).
operate in the affected organ independently from the central im- The exclusive identification of Rab33A in CD8 T cells also
mune system or not. The more independent the affected tissue emphasizes the importance of deconfounding for transcriptome
is, the smaller the likelihood will be that relevant T lymphocytes analysis of data obtained with heterogenous surrogate cell pop-
recirculate through the bloodstream. This question therefore ulations such as peripheral blood leukocytes (Jacobsen et al.,
has a direct impact on the value of peripheral blood leukocytes 2006; Repsilber et al., 2005). The peripheral blood cell composi-
for immunologic biomarker characterization. tion of latently infected individuals and patients with active TB
differs profoundly. While monocytes and natural killer (NK) cells
Transcriptomics in TB are present at higher proportions in TB patients, T cells and B
Measurement of immune biomarkers as described above is cells are present in higher proportions in latently infected individ-
biased by the panels of antigens, cytokines, and cell-surface uals (Jacobsen et al., 2007). Appropriate integration of bioinfor-
receptors preselected for analysis. This approach should be matics is needed to correct for these differences if determination
complemented by an unbiased approach, namely, global gene- of the individual leukocyte populations in all samples is not
expression profiling of peripheral blood leukocytes (Jacobsen feasible as is the case within the GC6-74 project.
et al., 2005, 2008) (Figure 1). Before embarking on transcriptomic Glycolipids are well-known stimulators of immune-suppres-
biomarker studies within GC6-74, we have performed proof-of- sive mechanisms with mannose-capped LAMs being of particu-
principle experiments among healthy latently infected individ- lar importance. This molecule is known to stimulate suppressive
uals, as evidenced by positive tuberculin skin test responses, mechanisms by signaling through DC-SIGN (van Kooyk and Ra-
and patients suffering from active TB disease. Differentially reg- binovich, 2008; Geijtenbeek et al., 2003). Gene-expression pro-
ulated genes belong to the following four groups: (1) antimicro- filing of DCs stimulated by LAM cognates revealed upregulation

Cell Host & Microbe 4, September 11, 2008 ª2008 Elsevier Inc. 225
Cell Host & Microbe

Review

of PD-L1 and of molecules involved in tryptophan catabolism on prognostic diagnosis in TB. Yet, we consider this a worthwhile
the level of transcription (Hegasy et al., unpublished data). In the undertaking for the following reasons:
human system, PD-L1 is expressed at medium cell-surface den-
sity on DCs and activated T cells and at high density on mono- (1) Two billion inhabitants of this globe are infected with Mtb
cytes (Figure 2). Interestingly, the lung has also been found to ex- and more than 10% of them are at risk of developing
press high levels of PD-L1 (Keir et al., 2008). PD-1 is expressed active TB disease after weakening of the immune system.
on activated and exhausted T cells, DCs, and monocytes. It is The risk of developing active TB disease could be pre-
thus possible that during active TB, PD-1–PD-L interactions vented by prophylactic chemotherapy after diagnosis
lead to T cell exhaustion (Jurado et al., 2008; Hegasy et al., un- using prognostic biomarkers.
published data). Moreover, our metabolomic studies revealed (2) Several vaccine candidates have entered the pipeline and
a low abundance of tryptophan in serum of patients with active hence, biomarkers could accelerate clinical trials by pro-
TB as compared to serum of latently infected individuals (unpub- viding valuable information on vaccine efficacy prior to the
lished data). Tryptophan degradation is a well-described cata- clinical end-point of active TB outbreak (Table 2).
bolic pathway of immune suppression with indoleamine 2, 3-di- (3) None of the vaccine candidates in the pipeline has pro-
oxygenase (IDO) as a crucial enzyme (Munn and Mellor, 2007). vided evidence for inducing sterile eradication of Mtb,
This enzyme is critically involved in activation of Treg cells, as making heterologous prime-boost stratagems composed
well as in CD4 and CD8 T cell suppression. of novel prime vaccines (improved BCG) and new subunit
vaccines (protein/adjuvant formulations) a valid option
(Kaufmann, 2006). Biomarkers could help to provide
Metabolomic Biomarkers in TB relevant information for educated selection of the most
Metabolomics measures the global spectrum of metabolic reac- promising combined vaccination against TB.
tions of an organism during physiological or pathological condi- (4) Biomarkers could be equally helpful for monitoring of clin-
tions (Box S7) (Mitchell and Carmichael, 2005). It can be as- ical trials assessing anti-Mtb activities of existing drugs
sumed that Mtb strongly influences metabolism of affected already approved for infectious diseases other than TB
tissues. It is likely that the productive lung containing dormant as well as for novel drug candidates that are refilling
bacteria during latent infection and the lung granuloma associ- the dried-out pipeline of TB drug development, notably,
ated with excessive tissue destruction during full-blown TB because combinations with existing drug treatment
disease produce different types of metabolites. Similarly, Mtb schemes need to be assessed (Box S2).
at different stages of its life cycle may well produce different me-
tabolites. Some of the host- and pathogen-derived metabolites For basic TB researchers, biomarkers provide an added value,
enter the circulation where they can be measured. This is the ba- namely a holistic view of in vivo functions during latent Mtb infec-
sis for metabolomic biomarker studies in TB (Idle and Gonzalez, tion and active TB disease, which will open up new research
2007) (Figure 1). Although not part of GC6-74 we have decided to avenues (Young et al., 2008). Global biomarker studies have
perform proof-of-principle experiments in the area of metabolo- sometimes been criticized as being non-hypothesis driven. We
mics of TB in collaboration with Metabolon Inc, USA (http:// consider the identification of novel molecules and pathways in
www.metabolon.com), and we were gratified to observe that TB highly fertile soil for propagating research of the hypothe-
metabolomics allowed distinction between healthy latently in- sis-driven kind. For example, differential expression of Rab33A
fected individuals and patients with active TB (unpublished in CD8 T lymphocytes in the peripheral blood from patients
data). Currently, most of the 350 identified metabolites are of with active TB versus latently infected healthy individuals has
host origin. Of these almost two-thirds are unknown and one- prompted us not only to reassess the functional role of CD8
third has been identified unequivocally. Yet, both types of com- T cells in TB but also to study more precisely the role of the sus-
pounds can be used for metabolomic discrimination between pected CTL mechanisms expressed by CD8 T cells in TB. This is
healthy and diseased individuals. We are currently assembling only one of many examples that have fueled new research ave-
components of Mtb to broaden the analytical spectrum. We nues. We consider it rewarding that findings obtained in the field
assume that identification of unique Mtb molecules will allow are being transferred back to the academic laboratory thus re-
us to define one component of a biosignature useful for definitive translating field observations into hypothesis-driven elucidation
distinction between TB and other diseases unrelated to Mtb of basic mechanisms. We are optimistic that collaborations like
infection such as Sarcoidosis or Crohn’s disease. It is also pos- ours in GC6-74 between scientists in the northern and southern
sible that volatile compounds leave lung lesions and reach the al- hemispheres will benefit both partners equally by producing the
veolar space from where they are released into the environment basis for novel control measures as well as for innovative
via breathing, coughing, etc. (Figure 1). At this stage, although research on basic disease mechanisms.
measurement of volatile substances is not part of our research
strategy, we consider this an interesting avenue to pursue. SUPPLEMENTAL DATA

Indeed, recent evidence suggests the feasibility of such an Supplemental Data include seven boxes and can be found with this article online
approach (Syhre and Chambers, 2008). at http://www.cellhostandmicrobe.com/cgi/content/full/4/3/219/DC1/.

ACKNOWLEDGMENTS
Concluding Remarks
We are aware that a long and winding road lies ahead of us The authors are grateful to M.L. Grossman for editorial help, J. Maertzdorf for
before biomarkers can be used for clinical trial monitoring and preparation of the transcriptomics box, and D. Schad and L. Fehlig for graphic

226 Cell Host & Microbe 4, September 11, 2008 ª2008 Elsevier Inc.
Cell Host & Microbe

Review
support. We also thank members of our department for sharing unpublished Jacobsen, M., Mattow, J., Repsilber, D., and Kaufmann, S.H. (2008). Novel
work. Studies on TB in our department receive financial support from the fol- strategies to identify biomarkers in tuberculosis. Biol. Chem. 389, 487–495.
lowing: Bill & Melinda Gates Foundation Grand Challenges in Global Health
Grant No. 37772, the European Union Framework Program 6 Projects, Design Jacobsen, M., Repsilber, D., Gutschmidt, A., Neher, A., Feldmann, K., Mollen-
and Testing of Vaccine Candidates against Tuberculosis (TBVAC) and kopf, H.J., Ziegler, A., and Kaufmann, S.H.E. (2005). Ras-associated small
MUVAPRED (Mucosal Vaccines for Poverty Related Diseases); Sonderfor- GTPase 33A, a novel T cell factor, is down-regulated in patients with tubercu-
losis. J. Infect. Dis. 192, 1211–1218.
schungsbereich Protektive und pathologische Folgen der Antigen-Verarbei-
tung (SFB 421); and Bundesministerium für Bildung und Forschung Networks Jacobsen, M., Repsilber, D., Gutschmidt, A., Neher, A., Feldmann, K., Mollen-
PathoGenoMikPlus and Nationales Genomforschungsnetz (NGFN). The kopf, H.J., Kaufmann, S.H., and Ziegler, A. (2006). Deconfounding microarray
authors are particularly grateful for the collaborative efforts of GC6-74 consor- analysis - independent measurements of cell type proportions used in a
tium members. regression model to resolve tissue heterogeneity bias. Methods Inf. Med. 45,
557–563.
REFERENCES
Jacobsen, M., Repsilber, D., Gutschmidt, A., Neher, A., Feldmann, K., Mollen-
kopf, H.J., Ziegler, A., and Kaufmann, S.H. (2007). Candidate biomarkers for
Armstrong, J.A., and Hart, P.D. (1971). Response of cultured macrophages to discrimination between infection and disease caused by Mycobacterium
Mycobacterium tuberculosis, with observations on fusion of lysosomes with tuberculosis. J. Mol. Med. 85, 613–621.
phagosomes. J. Exp. Med. 134, 713–740.
Jurado, J.O., Alvarez, I.B., Pasquinelli, V., Martinez, G.J., Quiroga, M.F., Ab-
Belkaid, Y., and Rouse, B.T. (2005). Natural regulatory T cells in infectious dis- bate, E., Musella, R.M., Chuluyan, H.E., and Garcia, V.E. (2008). Programmed
ease. Nat. Immunol. 6, 353–360. death (PD)-1:PD-ligand 1/PD-ligand 2 pathway inhibits T cell effector functions
during human tuberculosis. J. Immunol. 181, 116–125.
Bettelli, E., Korn, T., Oukka, M., and Kuchroo, V.K. (2008). Induction and
effector functions of T(H)17 cells. Nature 453, 1051–1057. Kaufmann, S.H. (2007). The contribution of immunology to the rational design
of novel antibacterial vaccines. Nat. Rev. Microbiol. 5, 491–504.
Betts, J.C., Lukey, P.T., Robb, L.C., McAdam, R.A., and Duncan, K. (2002).
Evaluation of a nutrient starvation model of Mycobacterium tuberculosis Kaufmann, S.H., and Parida, S.K. (2007). Changing funding patterns in tuber-
persistence by gene and protein expression profiling. Mol. Microbiol. 43, culosis. Nat. Med. 13, 299–303.
717–731.
Kaufmann, S.H.E. (2006). Envisioning future strategies for vaccination against
Biomarkers Definitions Working Group (2001). Biomarkers and surrogate end- tuberculosis. Nat. Rev. Immunol. 6, 699–704.
points: preferred definitions and conceptual framework. Clin. Pharmacol. Ther.
69, 89–95. Kaufmann, S.H.E., and McMichael, A.J. (2005). Annulling a dangerous liaison:
vaccination strategies against AIDS and tuberculosis. Nat. Med. 11, S33–S44.
Boasso, A., Shearer, G.M., and Clerici, M. (2008). The hunt for an HIV vaccine:
time to rethink recent failures. Lancet 371, 1897–1898. Kaufmann, S.H.E., and Winau, F. (2005). From bacteriology to immunology:
the dualism of specificity. Nat. Immunol. 6, 1063–1066.
Canetti, G. (1965). Present aspects of bacterial resistance in tuberculosis. Am.
Rev. Respir. Dis. 92, 687–703. Keir, M.E., Butte, M.J., Freeman, G.J., and Sharpe, A.H. (2008). PD-1 and its
ligands in tolerance and immunity. Annu. Rev. Immunol. 26, 677–704.
Charo, I.F., and Ransohoff, R.M. (2006). The many roles of chemokines and
chemokine receptors in inflammation. N. Engl. J. Med. 354, 610–621. Khader, S.A., Bell, G.K., Pearl, J.E., Fountain, J.J., Rangel-Moreno, J., Cilley,
G.E., Shen, F., Eaton, S.M., Gaffen, S.L., Swain, S.L., et al. (2007). IL-23 and
Colburn, W.A. (2000). Optimizing the use of biomarkers, surrogate endpoints, IL-17 in the establishment of protective pulmonary CD4(+) T cell responses
and clinical endpoints for more efficient drug development. J. Clin. Pharmacol. after vaccination and during Mycobacterium tuberculosis challenge. Nat.
40, 1419–1427. Immunol. 8, 369–377.

Corbett, E.L., Watt, C.J., Walker, N., Maher, D., Williams, B.G., Raviglione, Kursar, M., Koch, M., Mittrucker, H.W., Nouailles, G., Bonhagen, K., Kamradt,
M.C., and Dye, C. (2003). The growing burden of tuberculosis: global trends T., and Kaufmann, S.H. (2007). Cutting edge: regulatory T cells prevent effi-
and interactions with the HIV epidemic. Arch. Intern. Med. 163, 1009–1021. cient clearance of Mycobacterium tuberculosis. J. Immunol. 178, 2661–2665.

Demissie, A., Leyten, E.M., Abebe, M., Wassie, L., Aseffa, A., Abate, G., Lagakos, S.W., and Gable, A.R. (2008). Challenges to HIV prevention–seeking
Fletcher, H., Owiafe, P., Hill, P.C., Brookes, R., et al. (2006). Recognition of effective measures in the absence of a vaccine. N. Engl. J. Med. 358, 1543–
stage-specific mycobacterial antigens differentiates between acute and 1545.
latent infections with Mycobacterium tuberculosis. Clin. Vaccine Immunol.
13, 179–186. Lalvani, A. (2007). Diagnosing tuberculosis infection in the 21st century: new
tools to tackle an old enemy. Chest 131, 1898–1906.
Fenhalls, G., Stevens, L., Moses, L., Bezuidenhout, J., Betts, J.C., Helden,
P.P., Lukey, P.T., and Duncan, K. (2002). In situ detection of Mycobacterium Leyten, E.M., Lin, M.Y., Franken, K.L., Friggen, A.H., Prins, C., van Meijgaar-
tuberculosis transcripts in human lung granulomas reveals differential gene den, K.E., Voskuil, M.I., Weldingh, K., Andersen, P., Schoolnik, G.K., et al.
expression in necrotic lesions. Infect. Immun. 70, 6330–6338. (2006). Human T-cell responses to 25 novel antigens encoded by genes of
the dormancy regulon of Mycobacterium tuberculosis. Microbes Infect. 8,
Fine, P.E. (1995). Variation in protection by BCG: implications of and for 2052–2060.
heterologous immunity. Lancet 346, 1339–1345.
Li, M.O., and Flavell, R.A. (2008). Contextual regulation of inflammation: a duet
Flynn, J.L., and Chan, J. (2001). Immunology of tuberculosis. Annu. Rev. by transforming growth factor-beta and interleukin-10. Immunity 28, 468–476.
Immunol. 19, 93–129.
Lin, M.Y., Geluk, A., Smith, S.G., Stewart, A.L., Friggen, A.H., Franken, K.L.,
Gagneux, S., and Small, P.M. (2007). Global phylogeography of Mycobacte- Verduyn, M.J., van Meijgaarden, K.E., Voskuil, M.I., Dockrell, H.M., et al.
rium tuberculosis and implications for tuberculosis product development. (2007). Lack of immune responses to Mycobacterium tuberculosis DosR reg-
Lancet Infect. Dis. 7, 328–337. ulon proteins following Mycobacterium bovis BCG vaccination. Infect. Immun.
75, 3523–3530.
Geijtenbeek, T.B., van Vliet, S.J., Koppel, E.A., Sanchez-Hernandez, M.,
Vandenbroucke-Grauls, C.M., Appelmelk, B., and van Kooyk, Y. (2003). Myco- Maher, D. (2008). Tuberculosis Control: Good Clinical Care and Good Public
bacteria target DC-SIGN to suppress dendritic cell function. J. Exp. Med. 197, Health, S.H.E. Kaufmann and P.D. van Helden, eds. (Weinheim: Wiley-VCH),
7–17. pp. 115–130.

Hutloff, A., Dittrich, A.M., Beier, K.C., Eljaschewitsch, B., Kraft, R., Anagnosto- Manabe, Y.C., and Bishai, W.R. (2000). Latent Mycobacterium tuberculosis-
poulos, I., and Kroczek, R.A. (1999). ICOS is an inducible T-cell co-stimulator persistence, patience, and winning by waiting. Nat. Med. 6, 1327–1329.
structurally and functionally related to CD28. Nature 397, 263–266.
McGeachy, M.J., and Cua, D.J. (2008). Th17 cell differentiation: the long and
Idle, J.R., and Gonzalez, F.J. (2007). Metabolomics. Cell Metab. 6, 348–351. winding road. Immunity 28, 445–453.

Cell Host & Microbe 4, September 11, 2008 ª2008 Elsevier Inc. 227
Cell Host & Microbe

Review
McKinney, J.D., Jacobs, W.R., and Bloom, B.R. (1998). Persisting problems in tates antigen presentation to T lymphocytes through MHC-I and CD1 in tuber-
tuberculosis. In Emerging Infections, R. Krause, J.I. Gallin, and A.S. Fauci, eds. culosis. Nat. Med. 9, 1039–1046.
(New York: Academic Press), pp. 51–146.
Seder, R.A., Darrah, P.A., and Roederer, M. (2008). T-cell quality in memory
Mitchell, S.C., and Carmichael, P.L. (2005). Metabonomics and the endocrine and protection: implications for vaccine design. Nat. Rev. Immunol. 8,
system. Mol. Cell. Endocrinol. 244, 10–14. 247–258.

Moyron-Quiroz, J.E., Rangel-Moreno, J., Hartson, L., Kusser, K., Tighe, M.P., Stenger, S., Hanson, D.A., Teitelbaum, R., Dewan, P., Niazi, K.R., Froelich,
Klonowski, K.D., Lefrancois, L., Cauley, L.S., Harmsen, A.G., Lund, F.E., and C.J., Ganz, T., Thoma-Uszynski, S., Melian, A., Bogdan, C., et al. (1998). An
Randall, T.D. (2006). Persistence and responsiveness of immunologic memory antimicrobial activity of cytolytic T cells mediated by granulysin. Science
in the absence of secondary lymphoid organs. Immunity 25, 643–654. 282, 121–125.

Mueller, C., Muller, B., and Perruchoud, A.P. (2008). Biomarkers: past, pres- Syhre, M., and Chambers, S.T. (2008). The scent of Mycobacterium tubercu-
ent, and future. Swiss Med. Wkly. 138, 225–229. losis. Tuberculosis (Edinb.) 88, 317–323.
Munn, D.H., and Mellor, A.L. (2007). Indoleamine 2,3-dioxygenase and tumor- Tsuboi, T., and Fukuda, M. (2006). Rab3A and Rab27A cooperatively regulate
induced tolerance. J. Clin. Invest. 117, 1147–1154. the docking step of dense-core vesicle exocytosis in PC12 cells. J. Cell Sci.
119, 2196–2203.
Munoz-Elias, E.J., Timm, J., Botha, T., Chan, W.T., Gomez, J.E., and
McKinney, J.D. (2005). Replication dynamics of Mycobacterium tuberculosis Ulrichs, T., and Kaufmann, S.H.E. (2006). New insights into the function of
in chronically infected mice. Infect. Immun. 73, 546–551. granulomas in human tuberculosis. J. Pathol. 208, 261–269.
Nathan, C.F., Murray, H.W., Wiebe, M.E., and Rubin, B.Y. (1983). Identification Ulrichs, T., Kosmiadi, G.A., Trusov, V., Jörg, S., Pradl, L., Titukhina, M., Mis-
of interferon-gamma as the lymphokine that activates human macrophage henko, V., Gushina, N., and Kaufmann, S.H.E. (2004). Human tuberculous
oxidative metabolism and antimicrobial activity. J. Exp. Med. 158, 670–689. granulomas induce peripheral lymphoid follicle-like structures to orchestrate
local host defence in the lung. J. Pathol. 204, 217–228.
Ouyang, W., Kolls, J.K., and Zheng, Y. (2008). The biological functions of
T helper 17 cell effector cytokines in inflammation. Immunity 28, 454–467. van de Winkel, J.G., and Capel, P.J. (1993). Human IgG Fc receptor hetero-
geneity: molecular aspects and clinical implications. Immunol. Today 14,
Perussia, B., Dayton, E.T., Fanning, V., Thiagarajan, P., Hoxie, J., and
215–221.
Trinchieri, G. (1983). Immune interferon and leukocyte-conditioned medium
induce normal and leukemic myeloid cells to differentiate along the monocytic van der Wel, N., Hava, D., Houben, D., Fluitsma, D., van Zon, M., Pierson, J.,
pathway. J. Exp. Med. 158, 2058–2080. Brenner, M., and Peters, P.J. (2007). M. tuberculosis and M. leprae trans-
locate from the phagolysosome to the cytosol in myeloid cells. Cell 129,
Pulendran, B., and Ahmed, R. (2006). Translating innate immunity into immu-
1287–1298.
nological memory: implications for vaccine development. Cell 124, 849–863.
van Kooyk, Y., and Rabinovich, G.A. (2008). Protein-glycan interactions in
Rachman, H., Strong, M., Ulrichs, T., Grode, L., Schuchhardt, J., Mollenkopf,
H., Kosmiadi, G.A., Eisenberg, D., and Kaufmann, S.H.E. (2006). Unique the control of innate and adaptive immune responses. Nat. Immunol. 9,
transcriptome signature of Mycobacterium tuberculosis in pulmonary tubercu- 593–601.
losis. Infect. Immun. 74, 1233–1242.
Vandal, O.H., Pierini, L.M., Schnappinger, D., Nathan, C.F., and Ehrt, S. (2008).
Randall, T.D., Carragher, D.M., and Rangel-Moreno, J. (2008). Development of A membrane protein preserves intrabacterial pH in intraphagosomal Myco-
secondary lymphoid organs. Annu. Rev. Immunol. 26, 627–650. bacterium tuberculosis. Nat. Med. 14, 849–854.

Repsilber, D., Fink, L., Jacobsen, M., Blasing, O., and Ziegler, A. (2005). Sam- Vignali, D.A. (2000). Multiplexed particle-based flow cytometric assays.
ple selection for microarray gene expression studies. Methods Inf. Med. 44, J. Immunol. Methods 243, 243–255.
461–467.
Voskuil, M.I., Schnappinger, D., Visconti, K.C., Harrell, M.I., Dolganov, G.M.,
Rosenkrands, I., Slayden, R.A., Crawford, J., Aagaard, C., Barry, C.E., III, and Sherman, D.R., and Schoolnik, G.K. (2003). Inhibition of respiration by nitric
Andersen, P. (2002). Hypoxic response of Mycobacterium tuberculosis studied oxide induces a Mycobacterium tuberculosis dormancy program. J. Exp.
by metabolic labeling and proteome analysis of cellular and extracellular Med. 198, 705–713.
proteins. J. Bacteriol. 184, 3485–3491.
Walker, B.D., and Burton, D.R. (2008). Toward an AIDS vaccine. Science 320,
Russell, D.G. (2007). Who puts the tubercle in tuberculosis? Nat. Rev. Micro- 760–764.
biol. 5, 39–47.
WHO (2008). Global tuberculosis control: surveillance, planning, financing.
Sallusto, F., Geginat, J., and Lanzavecchia, A. (2004). Central memory and http://www.who.int/tb/publications/global_report/en/index.html. Geneva, World
effector memory T cell subsets: function, generation, and maintenance. Health Organization.
Annu. Rev. Immunol. 22, 745–763.
Winau, F., Weber, S., Sad, S., de Diego, J., Hoops, S.L., Breiden, B., Sandhoff,
Schaible, U.E., and Kaufmann, S.H.E. (2005). A nutritive view on the host-path- K., Brinkmann, V., Kaufmann, S.H.E., and Schaible, U.E. (2006). Apoptotic
ogen interplay. Trends Microbiol. 13, 373–380. vesicles crossprime CD8 T cells and protect against tuberculosis. Immunity
24, 105–117.
Schaible, U.E., and Kaufmann, S.H. (2007). Malnutrition and infection: com-
plex mechanisms and global impacts. PLoS Med. 4, e115. 10.1371/journal. Wolf, M., Albrecht, S., and Marki, C. (2008). Proteolytic processing of chemo-
pmed.0040115. kines: implications in physiological and pathological conditions. Int. J. Bio-
chem. Cell Biol. 40, 1185–1198.
Schaible, U.E., Collins, H.L., Priem, F., and Kaufmann, S.H.E. (2002). Correc-
tion of the iron overload defect in beta-2-microglobulin knockout mice by Yang, L., Anderson, D.E., Baecher-Allan, C., Hastings, W.D., Bettelli, E.,
lactoferrin abolishes their increased susceptibility to tuberculosis. J. Exp. Oukka, M., Kuchroo, V.K., and Hafler, D.A. (2008). IL-21 and TGF-beta are
Med. 196, 1507–1513. required for differentiation of human T(H)17 cells. Nature 454, 350–352.

Schaible, U.E., Winau, F., Sieling, P.A., Fischer, K., Collins, H.L., Hagens, K., Young, D., Stark, J., and Kirschner, D. (2008). Systems biology of persistent
Modlin, R.L., Brinkmann, V., and Kaufmann, S.H.E. (2003). Apoptosis facili- infection: tuberculosis as a case study. Nat. Rev. Microbiol. 6, 520–528.

228 Cell Host & Microbe 4, September 11, 2008 ª2008 Elsevier Inc.

Vous aimerez peut-être aussi