Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Jun 12.
Published in final edited form as: Cell. 2022 Dec 8;185(25):4682–4702. doi: 10.1016/j.cell.2022.10.025

Immune cell interactions in tuberculosis

JoAnne L Flynn 1,*, John Chan 2,*
PMCID: PMC12162144  NIHMSID: NIHMS1858516  PMID: 36493751

SUMMARY

Despite having been identified as the organism that causes tuberculosis in 1882, Mycobacterium tuberculosis has managed to still evade our understanding of the protective immune response against it, defying the development of an effective vaccine. Technology and novel experimental models have revealed much new knowledge, particularly with respect to the heterogeneity of the bacillus and the host response. This review focuses on certain immunological elements that have recently yielded exciting data and highlights the importance of taking a holistic approach to understanding the interaction of M. tuberculosis with the many host cells that contribute to the development of protective immunity.

INTRODUCTION

Tuberculosis has plagued humans for centuries. The bacillus Mycobacterium tuberculosis (Mtb) is transmitted primarily by aerosols expelled by a person with active tuberculosis. Tuberculosis is primarily considered a pulmonary disease, but the bacillus can infect nearly all organs in the body. Although there are still 10 million new cases of active tuberculosis every year worldwide and an estimated 1.3 million deaths (WHO, 2022), the majority of humans who are infected with Mtb control the infection without intervention. This state of clinically quiescent infection is known as latent tuberculosis (LTBI) and a small percentage of those with LTBI will progress to active tuberculosis years to decades after primary infection. It is now recognized that Mtb infection outcomes encompass a full spectrum from subclinical infection to mild, moderate, or severe active disease. Although controversial, it is possible and even likely that some of those classified with LTBI have cleared the infection (Behr et al., 2019), while others are at extremely low risk of reactivation. It is difficult to classify clinically the state of Mtb infection in humans. This is in part due to the diagnostics used to define Mtb infection or disease, which are based on reported symptoms, the host T cell response to mycobacterial antigens, chest X-rays or CT scans, and/or microscopy or culture of sputum or other patient samples. Thus, the various outcomes of Mtb infection are complex and likely due to a combination of several factors, including host immune responses, bacterial strain, and extent of exposure. Heterogeneity in outcome is prominent not only among infected subjects but also within a single infected person, where disease can regress in one area of the lung and progress simultaneously in another (Lin et al., 2014) (Figure 1).

Figure 1. The heterogeneous M. tuberculosis-host interaction in granulomas determine infection and disease outcomes.

Figure 1.

Cellular immunological and scRNA-seq analyses of granulomas, together with imaging and the use of barcoded bacilli, have revealed a high degree of heterogeneity of the granulomatous response among infected non-human primates and humans. In Mtb infection of naive hosts (left panel), the spectrum of heterogeneity spans resolution in restrictive granulomas, i.e., those that kill or limit replication of Mtb, to progression in permissive granulomas with increased bacterial growth and limited killing and dissemination to form new granulomas, representing the extremes on a continuum of disease states. Granulomas harbor a wide variety of immune cells and immunological factors (left and right panels), whose interactions likely govern whether the lesions progress or regress. The significance of the ability of specific cellular and immunological parameters of a granuloma in determining disease outcomes is seen in subjects of immunization-challenge studies (right panel), which can reflect vaccine efficacies. The granulomas developed upon challenge with virulent Mtb after immunization with highly protective vaccines such as i.v. BCG, which affords near sterilizing protection, are absent or smaller relative to unimmunized subjects and contain fewer or no bacilli (right panel). In addition, the lung parenchyma of non-human primates subjected to the i.v. BCG immunization-challenge model harbors CD4 T cells and CD8 T cells, and the airway is populated with both T cells and antibodies, suggesting an important role for these immunological elements in mediating vaccine-engendered protection against Mtb (right panel).

There have been decades of research into the immune responses that dictate protection against tuberculosis, yet we still do not have a firm grasp on the constellation of immune responses necessary to prevent infection or progression to disease. The immune response to Mtb infection is complex, involving many cell types and functions. The field has looked for the “magic bullet” to target for an effective vaccine, without much success. Often tuberculosis research focuses on the role that one cell type, one cytokine, or one pathway plays in control or exacerbation of infection. Instead, we posit that the various aspects of the immune response interact with and influence each other within several different anatomical compartments to lead to the ultimate outcome of the infection—clearance, containment, or progressive disease. That said, there are likely several “combinations” of factors that can result in a successful outcome, just as there are likely other combinations that result in progression to disease (Cadena et al., 2017). A holistic approach to the immunology of Mtb infection and disease is needed to identify mechanisms of protection. This is challenging, as it requires integration of multiple data types across different model systems and in humans. Because the primary battlefield between the host and the bacillus occurs in the tissues (airways, lung, granulomas, and lymph nodes), it can be difficult to extrapolate from samples easily obtained from humans, such as blood and sometimes bronchoalveolar lavage (BAL) fluid. Nonetheless, this is the challenge for modern research in the tuberculosis field—devising methods to interrogate what control of infection represents without a focus on one cell type or function, but instead to identify the collection of responses that can lead to effective elimination of the bacillus either at the time of infection or in persons who are already infected.

In this review, we discuss the various immunologic players in Mtb infection and disease but highlight recent studies that support the notion that the sum of immune responses is more important than any one particular cell type or function. It is also relevant that immune responses in the tissues can evolve over time, highlighting the plasticity of the immune system.

THE GRANULOMA

The granuloma is the pathologic hallmark of Mtb infection. Mtb is transmitted via aerosols from a person with active tuberculosis. Infection most often occurs upon inhalation of individual bacilli into the airways, where the bacillus encounters alveolar macrophages. These macrophages can engulf the bacilli and, in some cases, prevent productive infection. However, when the bacilli (likely in alveolar macrophages or maybe other phagocytes) transit to the lung parenchyma, the host mounts an immune response. Monocytes, macrophages, neutrophils, and dendritic cells are attracted to the site of infection, likely due to chemokine and cytokine signals from the infected cells, initiating granuloma formation. The bacilli are carried to the lung draining lymph nodes where priming of the adaptive immune response is initiated. Murine studies indicate this occurs within 7–14 days of infection (Reiley et al., 2008), although in humans and non-human primates adaptive T cell responses in the periphery are usually not observed until 4–6 weeks post-infection (Capuano et al., 2003; Poulsen, 1950). Primed T cells (and likely B cells) then migrate to the site of infection, resulting in the formation of an organized granuloma, with lymphocytes surrounding a macrophage layer, often with neutrophils surrounding the necrotic center (Figure 1). In non-human primates, the fully formed granuloma can be observed by 3–4 weeks post-infection (Lin et al., 2006). Within granulomas, Mtb can be intracellular, primarily within macrophages or extracellular, primarily within the caseous necrotic center. The immune responses necessary for controlling infection may differ depending on the location of the bacteria within granulomas.

Using DNA barcoded strains of Mtb in macaques, we demonstrated that each granuloma is initiated by a single bacillus; over the next 4–6 weeks that bacillus multiplies to reach ~105 CFU/granuloma, which appears to be the “carrying capacity” of any single granuloma (Lin et al., 2014). At that point, replication can be restrained by the granuloma or the Mtb can escape (disseminate) to form new granulomas in the lung. In most cases, bacterial killing is not robust until ~10 weeks of infection, likely due to a delay in a functioning adaptive immune response in granulomas (Grant et al., 2022; Lin et al., 2014).

Granulomas can form in other tissues as well, most commonly in thoracic lymph nodes. Dissemination of initial infection to the thoracic lymph nodes is common in Mtb infection. Because thoracic lymph nodes are important lymphoid structures, the migration of Mtb to the thoracic lymph nodes is likely critical for the initiation of adaptive immunity. However, lymph nodes can be a prominent site of bacterial persistence and potential source of reactivation (Ganchua et al., 2018, 2020). Humans with Mtb infection often have involved thoracic lymph nodes as seen by imaging and, particularly in children, an infected lymph node can be quite enlarged and cause lung lobe collapse (Cong et al., 2022; Navani et al., 2011). We showed previously that macaque thoracic lymph nodes are relatively poor at killing Mtb, and the data suggest that lymph nodes may be a source of bacterial dissemination (Ganchua et al., 2018).

Granuloma structure and function play a critical role in containment of the infection, yet many of the mechanisms by which this occurs and what leads to failure are still unclear. Holistic studies of granulomas and other pathologies in tuberculosis are required to dissect the complexity of the immune factor interplay in granulomas.

MACROPHAGES

Although the macrophage was discovered a century and a half ago, the biology of this highly complex phagocyte, which plays an important role in defense against microbes and tissue homeostasis, remains incompletely defined (Ginhoux and Guilliams, 2016; Guilliams et al., 2020). Nevertheless, with the help of novel technologies, we now know that there are distinct subsets of tissue-resident macrophages that vary in their ontogeny (bone marrow versus embryonic in origin), homeostasis maintenance, and functions, thus revising the concept of the mononuclear phagocyte system and the M/M2 paradigm of functional polarization (Martinez and Gordon, 2014; Murray, 2017). Importantly, cellular source of macrophage subsets and their tissue-specific niche bear functional consequences (reviewed in Ginhoux and Guilliams, 2016; Guilliams et al., 2020). It is apparent that organ-specific niche can modulate the development and maintenance of tissue-resident macrophages, imprinting upon them distinct attributes and functions, including metabolic activities. Elucidation of the mechanisms by which these processes are regulated will likely provide information that can lead to the design of macrophage-targeting intervention in treating diseases.

As the primary host cell for Mtb, macrophages have been widely used to study the Mtb-host interaction. Based on our current understanding of tissue-resident macrophage biology (Ginhoux and Guilliams, 2016; Guilliams et al., 2020), the variability of results generated by in vitro studies, which use cell lines and primary macrophages derived from different tissue compartments, is not unexpected. Relevant to Mtb, a pulmonary pathogen, lung-resident macrophages are made up of distinct subsets—broadly designated as alveolar macrophages (AM) and interstitium macrophages (IM) (reviewed in Ginhoux and Guilliams, 2016; Guilliams et al., 2020), which are located in the alveolus and the interstitium of the lungs, respectively (Figure 2). In mice, AM are of embryonic origin and thus bone marrow independent, and they are self-replenishing (Figure 2). Under certain experimental conditions, however, including irradiation and infections that lead to depletion of AM, monocytes can participate in replenishing this population (Ginhoux and Guilliams, 2016; Guilliams et al., 2020). Emerging evidence supports the notion that subsets of AM exist (Pisu et al., 2021). Similarly, IM are heterogeneous and of both embryonic and monocytic origin (Gibbings et al., 2017; Sabatel et al., 2017; Tan and Krasnow, 2016; Ural et al., 2020) with subsets that are functionally different from one another and from AM (Gibbings et al., 2017; Schyns et al., 2019; Ural et al., 2020). How do these spatially and functionally distinct macrophage subsets modulate infection and disease outcome in tuberculosis?

Figure 2. The interaction between lung macrophages and M. tuberculosis shapes the host response to the tubercle bacillus and influences the development of drug tolerance.

Figure 2.

In a mammalian host, alveolar macrophages (AM) of embryonic origin populate the air space prior to birth. (1) The AM population is self-renewing and represents the host cell that Mtb apparently targets primarily upon entering into the lung alveolar space. (2) Mtb-infected AM, through IL-1 receptor (IL-1R) signaling-dependent mechanisms (2), translocate from the alveolar space into the lung interstitium (3), serving as the vehicle that carries bacilli into the parenchyma. The translocation of infected AM is dependent on IL-1R, MyD88, and ASC in a macrophage-extrinsic fashion through non-hematopoietic cells. Translocated Mtb-infected AM multiplies in the interstitium, forming cellular aggregates (4), followed by recruitment from the vasculature (5) of neutrophils and monocyte-derived macrophages (6), which are then infected. The parenchyma of the lungs contains interstitial macrophages (IM), which are of bone marrow origin (7) and can be infected with Mtb (8). IM are a highly heterogeneous population, consisting of multiple subsets that interact with Mtb differentially, raising the possibility that distinct macrophage sub-populations may contribute to granuloma heterogeneity (Figure 1). One functional characteristic that reflects subset heterogeneity of AM and IM macrophage populations is the levels of stress these phagocytes imposed on Mtb, as assessed by fitness reporter mycobacterial strains (9). Mouse studies have shown that the AM and IM lineages display distinct metabolism (possibly pre-programed as a result of their different origins [1 and 7] and niche [2 and 8)] that differentially regulates replication of intracellular bacilli.) These cellular parameters translate to AM being more permissive for Mtb growth compared with IM. The levels of stress encountered by Mtb intracellularly can promote drug tolerance (10), an intermediate state conducive to the development of drug resistance. Permissive macrophages are depicted by higher number of paler bacilli in the phagosomes, while restrictive macrophages are those with lower number and darker bacilli in the vacuoles. The darker color of the bacilli denotes higher levels of macrophage-imposed intracellular stress.

Using Mtb fluorescent reporter strains that enable the assessment of mycobacterial fitness via determination of the replication status of intracellular bacilli and of the levels of stress they are subjected to, in conjunction with in vivo modeling and transcriptome analysis, Huang et al. (2018) reported that interaction of Mtb with AM and IM in mice leads to remarkably divergent infection outcomes. Compared with IM, AM exert less stress on intracellular Mtb and are relatively permissive for bacterial replication. In line with this observation, depletion of AM leads to reduced lung bacterial burden, while the opposite is observed upon ablation of blood monocytes and IM (Huang et al., 2018). Transcriptional data showed that metabolically, the permissive AM, which harbor relatively higher number of Mtb, exhibit a preference for fatty acid oxidation, while the relatively restrictive IM that carry a lower bacterial burden displays a propensity for the glycolytic pathway. This metabolism/Mtb control observation is congruent with the known preferred lipid diet of intracellular bacilli (Laval et al., 2021) and is in line with the results of studies involving inhibition of specific metabolic pathways in macrophages and in mice. The significance of these observations is buttressed by the results of a dual RNA sequencing (RNA-seq) study that enabled simultaneous characterization of the intracellular bacterial and host cell transcriptomes using sorted AM and IM from infected mice (Pisu et al., 2020). This latter study extends the previously observed differential metabolisms displayed by AM and IM (Huang et al., 2018) to include a range of distinctive transcriptome signatures, including varied levels of stress signals imposed by reactive nitrogen intermediates and restriction of iron availability (Pisu et al., 2020). The dual RNA-seq platform constitutes a versatile approach that should support detailed analysis of the host and bacillary in vivo responses during Mtb infection. Indeed, using the dual RNA-seq platform, together with the fitness reporter Mtb strains and detection of specific cell surface marker expression to study in vivo host-cell-bacterial interaction at the single-cell level, a recent study revealed that infected AM and IM in the lungs of mice3 weeks after an intranasal infection with a relatively high inoculum are composed of heterogeneous subpopulations(four each for AM and IM), with AM and IM containing both mycobacterial growth-restrictive and -permissive subsets; this functional diversity is further supported by their transcriptome profiles (Pisu et al., 2021).While the precise characteristics of these pulmonary macrophage subsets remain to be determined, the fact that individual subsets harbor bacilli with varied fitness phenotypes suggests that interaction of Mtb with distinct subpopulations may differentially modulate infection and disease outcome. The alveolar macrophage subsets identified in this mouse study can be detected in BAL fluid obtained from healthy uninfected humans, suggesting their relevance in human tuberculosis (Pisu et al., 2021). It is noteworthy that transposase-accessible chromatin sequencing (ATAC-seq) data suggest that preexisting epigenetic imprinting contributes to how each lung macrophage subset responds to Mtb infection (Pisu et al., 2021). Importantly, a recent study involving single-cell RNA-seq (scRNA-seq) analysis has revealed diverse macrophages subsets in the lungs of Mtb-infected macaques (Gideon et al., 2022). The results of these highly informative studies underscore the complexity of the lung macrophage landscape, characterized by distinct subsets that display differential interaction with Mtb (Huang et al., 2018; Pisu et al., 2021), raising the possibility that (1) the resulting differential interactions between subsets of these phagocytes with Mtb contribute, at least in part, to the heterogeneity in disease outcome in tuberculosis and the variability of granulomas observed in the lungs of a single infected individual (Cadena et al., 2017) and (2) targeting specific macrophage subsets and their local niche may enable manipulation of infection and disease outcomes and should pave the way for future experiments designed to unravel the relative contributions of the various lung-resident macrophage subpopulations in regulating the host response to the pathogen (Huang et al., 2018; Pisu et al., 2021).

TRAFFICKING MTB—THE ALVEOLAR SACINTERSTITIUM RELOCATION

The early sequence of events that occur after Mtb enters the alveolar space has been poorly understood until recently (Figure 2). Using a fluorescent Mtb strain to track lung host cell dynamics in acute Mtb infection in mice, a recent report revealed that AM constitute the primary cell type that Mtb parasitizes as early as 48 h post-inoculation (Cohen et al., 2018). Infected AM gain access to the lung interstitium during the innate phase of infection, where these phagocytes proliferate to form aggregates, followed by dissemination of bacilli to varied lung myeloid cell populations, mostly involving recruited neutrophils and monocyte-derived macrophages (Cohen et al., 2018). Pursuing the observation that infected AM upregulate the expression of pro-IL-1β, the effect of elements involved in IL-1 production and the IL-1R signaling pathway (Cohen et al., 2018) on AM translocation was examined, targeting IL-1R, MyD88, and ASC (apoptosis-associated speck-like protein containing a caspase recruitment domain), factors that have been reported to play significant roles in mediating innate immunity in tuberculosis. Using specific gene-disrupted mice and mixed bone marrow chimeras, the studies revealed that AM translocation is dependent on IL-1R, MyD88, and ASC in a macrophage-extrinsic fashion through non-hematopoietic cells (Cohen et al., 2018). Further, in agreement with the observation that IL-1 expression in tuberculosis is dependent on the mycobacterial ESX-1 secretion system, AM translocation was markedly attenuated in cells infected with an RD1 deletion Mtb mutant (Cohen et al., 2018 and references therein). Together, the results suggest that IL-1β mediates a crosstalk between infected AM and pulmonic non-hematopoietic cells (possibly lung epithelial cells) to enable the entry of infected macrophages into the interstitium from the airway. It will be of interest to determine the cellular source of IL-1 in this early phase of infection and to identify the non-hematopoietic cells that participate in the ESX-1-dependent relocation process. Importantly, the IL-1R signaling pathway is relevant clinically. A recent report noted that varied transmission capacity of clinical Mtb isolates is related to their discrepant proficiency to promote the migration of infected AM from the air space into the interstitium in an IL1-R signaling-dependent manner (Lovey et al., 2022). The high transmission (HT) Mtb strain, relative to the low transmission (LT) counterpart, triggers a more rapid relocation of infected AM into the interstitium that is associated with earlier dissemination of bacilli to the draining lymph nodes, higher levels of Th1 priming, and enhanced ability to bacterial control. Mice with chronic HT infection develop necrotic lung lesions that in humans, may develop into cavities to promote transmission (Hunter, 2011). The highly complex and heterogeneous granulomas modulate both mycobacterial containment and immune-mediated inflammatory damage in tuberculosis (Bhattacharya et al., 2021; Cadena et al., 2017; Krug et al., 2021; McCaffrey et al., 2022; Roca and Ramakrishnan, 2013; Tobin et al., 2012; Wells et al., 2021). Understanding the mechanisms underlying the regulation of the exacerbated granulomatous response in the HT strain could illuminate how human tissue-damaging immunopathology develops (Hunter, 2011). Worthy of note, in the above-described macrophage studies, the results discussed have been generated from models involving Mtb infection of naive mice. Given that a significant proportion of humans have been vaccinated with BCG and/or exposed to Mtb, it would be of interest and important to study the early events of Mtb-macrophage interaction using animals that are not mycobacteria naive.

THE T CELL RESPONSE

It must be recognized that currently, there are no true correlates of protection against tuberculosis, and although many immune cell types have been implicated as being important in prevention of tuberculosis disease in animal models, the mechanisms by which the immune response can control or eliminate infection remain poorly understood. T cells have long been recognized as necessary for control of Mtb infection and progression to disease. However, as discussed below, recent data support that there is a wide range of T cell types and functions that can contribute to protection or even exacerbate disease. Critical to effective control of Mtb infection and disease is the balance of T cell responses—neither too little nor too much—so that the infection is controlled but inflammation is restrained (Barber et al., 2019; Gideon et al., 2015; Kauffman et al., 2021). Inflammation is a major factor in tuberculosis disease, so a delicate balance of immune responses in granulomas, lung tissue, and lymph nodes is necessary to prevent excessive inflammation.

Much of the attention over the past several decades has been on CD4 T cells and their importance in control of Mtb infection and disease. There is no doubt that CD4 T cells are a key factor in the immune response to this pathogen. However, emerging evidence indicates that they are not the only important cell type and which functions of CD4 T cells are most relevant is controversial. Mice deficient in CD4 T cells, either genetic knockouts or through antibody depletion, have higher Mtb bacterial burdens compared with control animals (Caruso et al., 1999; Saunders et al., 2002; Scanga et al., 2000). Th1 CD4 T cells (expressing cytokines IFN-γ, TNF, and/or IL-2) are generally considered to be most important against Mtb, since Th1 cells can activate macrophages to induce anti-bacterial activity. In addition, murine knockouts of IFN-γ, TNF, or IL-12 genes (reviewed in Cooper, 2009), neutralization of TNF in NHPs or in humans (as a treatment for inflammatory diseases) (Keane et al., 2001; Lin et al., 2016, 2010), and genetic studies of mycobacterial susceptibility in humans have demonstrated the importance of these cytokines in resistance to tuberculosis. However, potential vaccine candidates that induce CD4 T cells producing IFN-γ or other Th1 responses have not necessarily resulted in enhanced protection in animal models or in human clinical trials (Tameris et al., 2014, 2013). In murine models, there is evidence that IFN-γ produced by CD4 T cells contributes to protection (Green et al., 2013) but also evidence that there are IFN-γ-independent mechanisms of CD4 T cells that are important (Sakai et al., 2016). Depletion of CD4 T cells in acute infection in macaques leads to exacerbated disease in most animals (Lin et al., 2012) HIV infection in humans is a major risk factor for development of active tuberculosis, and the risk is increased even before CD4 T cells are substantially depleted (Gupta et al., 2013; Lawn et al., 2009). In macaques with LTBI, SIV infection, as a model for HIV, caused reactivation in a majority of animals, while CD4 depletion caused reactivation tuberculosis in only a subset of animals (Bucsxan et al., 2019; Diedrich et al., 2010, 2020). This suggests that SIV (and likely HIV) has additional effects on the immune system beyond CD4 T cells, leading to increased susceptibility to tuberculosis.

In addition to Th1 cytokines, CD4 T cell production of IL-17 (Th17 cells) has been shown in murine models to enhance chemokines in lungs that induce recruitment of IFN-γ producing CD4 T cells to the lungs during Mtb infection (Khader et al., 2007) and in response to BCG vaccination ( Gopal et al., 2012). Neutralization of IL-17 during the early phase of infection in mice did not exacerbate tuberculosis (Segueni et al., 2016b). IL-17 was implicated in control of chronic Mtb infection in murine models although the production was primarily from γδ T cells whereas Th17 cells specific for Mtb were rare. During acute infection, the primary source of IL-17 was also shown to be from γδ T cells (Lockhart et al., 2006). There is also evidence that IL-17 can contribute to expansion of γδ T cells in non-human primates (Shen et al., 2015). In macaque models and in humans, CD4 T cells producing IL-17 are at relatively low levels compared with Th1 cells in blood and in tissues (Gideon et al., 2015; Nikitina et al., 2018). Nonetheless, in a model of mucosal BCG vaccination, Th17 cells were implicated as a correlate of protection (Dijkman et al., 2019). In intravenous (i.v.) BCG-vaccinated animals, which show robust protection against infection and disease, Th17 cells were only a small population in blood or tissues (Darrah et al., 2020). In a recent study where scRNA-seq was applied to granulomas, a subset of T cells (termed T1/T17) that included both CD4 and CD8 T cells expressed transcription factors that are associated with Th17 or Th1 cells but that did not express IL-17 was shown to be significantly associated with reduced bacterial burden (Gideon et al., 2022). An intriguing study using resected human lung tissue suggested that tissue-resident IL-17 expressing cells could participate in restricting Mtb growth (Ogongo et al., 2021). However, another study of the tuberculin skin test (TST) response in humans demonstrated that an IL-17 signaling pathway was upregulated in the TST of persons with active tuberculosis compared with those with LTBI, although the source of IL-17 was not reported (Pollara et al., 2021). In that study, IL-17 was hypothesized to contribute to tuberculosis pathology. In a study combining human, non-human primate and murine tuberculosis data, innate lymphocytes of the ILC3 class which produce IL-17 and IL-22 were implicated in the formation of bronchial associated lymphoid tissue (iBALT) in lungs, and ILC3-deficient mice had a modest increase in lung bacterial burden (Ardain et al., 2019). In human lungs, tissue-resident CD4 T cells expressing IL-17 were expanded compared with blood although at a lower frequency than CD4 T cells expressing canonical Th1 cytokines and IL-17 in lungs was inversely correlated with inflammation (Ogongo et al., 2021). In this same study, adding IL-17 or IL-2 to a 3D in vitro granuloma model reduced Mtb bacterial growth. In summary, Th17 cells and other producers of IL-17 may have different effects depending on the stage of infection or in the context of vaccination. They may well play a supporting role through interactions with other cells important for protection.

Immune responses do not exist in a vacuum and CD4 T cells can also exert other functions, including help for inducing B cell and CD8 T cell responses that may play a role in prevention of tuberculosis disease and induction of migration signals for other cells. TNF is a cytokine known to be important in protection against infection and reactivation tuberculosis in humans, nonhuman primates, and mice (Flynn et al., 1995; Keane et al., 2001; Lin et al., 2010). CD4 T cells are one source of TNF in Mtb infections, although macrophages are likely a larger contributor (Allie et al., 2013). The receptor for TNF on myeloid cells is critical to control of Mtb infections in mice (Segueni et al., 2016a), reinforcing that TNF (produced by T cells or myeloid cells) is an activator of macrophages. Data from mice suggest that CD4 T cells are important for inducing appropriate CD8 T cell responses, possibly through production of IL-2 or by activation of antigen presenting cells (Serbina et al., 2001).

Although CD4 T cells are clearly important in control of Mtb infection and disease, the role of adaptive CD8 T cells is less well studied (reviewed in Jasenosky et al., 2015; Lin and Flynn, 2015). In mice, there are conflicting data regarding the importance of these cells in control of tuberculosis. MHC-class-I-and TAP-deficient mice were both more susceptible to tuberculosis disease (Behar et al., 1999; Flynn et al., 1992; Schaible et al., 2002), but depletion of CD8 T cells was reported to only increase disease in the chronic phase of infection (van Pinxteren et al., 2000). The Behar lab has demonstrated enhanced protection with Mtb-specific CD8 T cells in mice, although not all Mtb-specific CD8 T cells recognized infected macrophages (Woodworth et al., 2008; Yang et al., 2018). In one study, human CD8 T cell lines were shown to primarily recognize heavily infected macrophages (Lewinsohn et al., 2003) and recognition of Mtb-infected macrophages by T cells is an area of active research. In addition, human CD8 T cells induced by vaccination with an adenovirus expressing Mtb antigens often did not recognize Mtb-infected macrophages (Nyendak et al., 2016). Antigen cross-presentation on MHC class I could underlie this phenomenon, and it will be important to understand the role of CD8 T cells that recognize infected cells as well as those that recognized uninfected cells presenting Mtb antigens in the context of granuloma level control of infection. Understanding the mechanisms by which CD8 T cells do or fail to recognize Mtb-infected macrophages will be critical in designing CD8-focused vaccine strategies.

Many studies on CD8 T cells in tuberculosis have measured cytokines such as IFN-γ and TNF rather than the more canonical function of CD8 T cells, which is cytotoxicity. There are multiple cytotoxic effectors described for CD8 T cells (and also innate cytotoxic cells), including granzymes (A, B, H, K, and M), granulysin, and perforin. Perforin forms a pore that is used to deliver cytotoxic molecules to a target cell. A potential role for CD8 T cells in control of Mtb is recognizing and destroying infected macrophages or directly killing Mtb inside the cells. There is also growing evidence that granzymes can be secreted and have extracellular functions including modulation of other cells, which could be important in the context of the granuloma. The exact functions of the different granzymes are still being investigated in other disease systems; however, Robert Modlin’s group reported that granulysin could directly kill Mtb in vitro (Stenger et al., 1998). Recently, a small fragment of granulysin has been shown to have antimicrobial activity against extracellular Mtb, internalized by human macrophages in vitro and restricted replication of intracellular Mtb (Noschka et al., 2021). Unlike humans and non-human primates, mice do not express granulysin. “Poly-cytotoxic” CD8 T cells (expressing granzyme B or CCL5, granulysin, and perforin) in human blood were associated with improved control of Mtb infection (Stegelmann et al., 2005). Zheng Chen’s group implicated CD8 T cells in the modest protection provided by intradermal BCG vaccination in macaques (Chen et al., 2009). Several groups have reported a series of Mtb antigens that are recognized by human CD8 T cells. There is wide variability among humans in terms of antigen recognition, and there does not appear to be a single or a few immunodominant antigens (Lin and Flynn, 2015; Lindestam Arlehamn et al., 2014). There is growing interest in the potential roles of cytotoxic CD8 T cells in control of tuberculosis, although much remains to be understood. Recent studies on macaque granulomas using scRNA-seq demonstrate the diversity of CD8 T cell responses at the site of infection, including several cytotoxic subsets that are associated with local control or killing of Mtb (Gideon et al., 2022). Identifying strategies to enhance CD8 T cells and harness their cytotoxic potential in the context of vaccines could be a game changer. Such strategies might include viral vectors, including the CMV-based tuberculosis vaccine discussed below, but also adjuvants such as IL-15 superagonists (N803) that enhance CD8 T cell responses in other systems (Berrien-Elliott et al., 2022; Miller et al., 2022).

Non-classical CD8 T cells recognize glycolipid antigens instead of protein antigens in the context of the MHC class Ib molecules CD1a–c. Humans, non-human primates, and guinea pigs express MHC CD1a–c, but mice do not. Several mycobacterial lipid antigens have been identified for CD1-restricted T cells, including glucose monomycolate, mycoketides, diacyltrehalose, and sulfoglycolipids (James et al., 2018; Reijneveld et al., 2021a, 2021b; Van Rhijn et al., 2013). The TCRs that recognize mycolyl lipid in the context of CD1b are germline encoded, often referred to as GEM TCRs. CD1-restricted T cells are expanded in Mtb-infected humans and in macaques vaccinated with BCG via the i.v. route (Layton et al., 2021). The role of these cells in protection is less clear. Immunization of guinea pigs with certain lipids showed modest improvements in tuberculosis-related pathology (Dascher et al., 2003).

In addition to adaptive CD8 T cell responses, there are also several innate-like lymphocytes that can express cytokine and/or cytotoxic effectors, including NK T cells, γδ T cells, and mucosal associated invariant T cells (MAITs) (collectively referred to as donor unrestricted T cells), and NK and other innate-like lymphocytes (ILCs). NK cells in blood have been associated with LTBI in humans (Chowdhury et al., 2018). Innate-like lymphocytes in airways or lung tissue may be the first line of defense against Mtb infection by modulating the AMs to restrict or kill Mtb bacilli. Several groups have postulated a protective role for MAIT cells, which have a restricted TCR usage and recognize non-peptide antigens presented by the class Ib molecule MR1 (reviewed in Gold et al., 2015). MAITS originally were demonstrated to recognize riboflavin intermediates produced by bacteria but more recently have been shown to respond to stimulation with mycobacterial antigens and even lysates from bacteria that do not produce riboflavin (reviewed in Lepore et al., 2021). MAITs are present in airways of mice, humans, and non-human primates and can produce effector molecules including cytokines and cytotoxic molecules. Although they are considered to be innate-like T cells, they can also have adaptive qualities and may act as a bridge between the innate and immune response to pathogens (Napier et al., 2015). MAITs may be among the first cells to interact with pathogens at mucosal surfaces, such as the airways. It was recently shown that these cells are clonally expanded in the airways of humans with pulmonary tuberculosis (Wong et al., 2019). In contrast, the frequency of MAITs in human and non-human primate granulomas is quite low (Gideon et al., 2022; Ogongo et al., 2020). Increasing the frequency of MAITs in mice with a small molecule led to improved control of Mtb (Sakai et al., 2021a). However, a similar strategy in macaques was complicated by an unexpected blunting of the MAIT response with no effect on bacterial burden (Sakai et al., 2021b). Thus, the roles or importance of MAITS in control of Mtb infection remains unclear.

γδ T cells have received a fair amount of attention over the years in the context of Mtb infection. These T cells recognize phosphoantigens such as isopentenyl pyrophosphate and (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMBPP) and vγ9/vδ2 T cells, which are the majority of γδ T cells in primates, are expanded during Mtb infection or BCG vaccination in humans and non-human primates (Hoft et al., 1998). HMBPP is produced by mycobacterial species and may account for the increase in γδ T cells following mycobacterial exposure although mycobacterial glycolipids were also shown to activate vγ9/vδ2 T cells (Xia et al., 2016). Although generally considered as innate-like T cells, data in non-human primates indicate that they have adaptive T cell qualities and memory responses (Qaqish et al., 2017; Shen et al., 2019). Chen et al. expanded vγ9/vδ2 T cells ex vivo and adoptively transferred them into macaques followed by Mtb infection, which reduced overall lung bacterial burden ~3–4-fold compared with naive macaques. In a subsequent study, this same group immunized macaques with a Listeria monocytogenes strain expressing HMBPP and found similar reductions in bacterial burden following Mtb challenge providing evidence that vγ9/vδ2 T cells may play a role in limiting tuberculosis disease (Shen et al., 2019). Recent data from Mtb-infected humans showed that the γδ T cell repertoire was clonally expanded in lung tissue and dominated by Vδ1 (Ogongo et al., 2020). γδ T cells are also present in macaque granulomas and express cytotoxic molecules (Gideon et al., 2022). γδ T cells can activate dendritic cells, leading to enhanced T cell responses. There are conflicting results in mice regarding the role of γδ T cells in protection against Mtb. In mice lacking γδ T cells due to a knockout in the delta gene, some studies showed modest increases in bacterial burden early following low-dose infection (Ladel et al., 1995) but not following high-dose infection or during chronic infection (Turner et al., 2001). Another study showed no effect on Mtb bacterial burden but suggested a role for these cells in limiting pathology in the lungs (D’Souza et al., 1997). At this time, it is not possible to selectively deplete γδ T cells in non-human primates, and there are some dissimilarities between the murine and primate γδ T cell responses to Mtb. Although more studies are needed, these cells, which are also found in granulomas (Gideon et al., 2022; Napier et al., 2015), may be playing a beneficial role in control of Mtb infection as well as influencing other immune cells.

One of the conundrums in Mtb infection is the relatively slow adaptive T cell response to the pathogen in animal models and in humans. The TST, which relies on a delayed-type hypersensitivity response and therefore measures adaptive T cell responses, takes up to 6 weeks after infection to become positive in humans and in macaque models (Capuano et al., 2003; Poulsen, 1950). Mice also have a relatively slow adaptive T cell response. In mice, various studies have suggested this is due to inhibition of antigen presentation of Mtb antigens, delayed migration of the bacilli to the lung draining lymph nodes, and direct interference with T cell responses (Chackerian et al., 2002; Myers et al., 2013; Reiley et al., 2008). This delay in induction of T cell responses is likely due, at least in part, to the relatively small numbers of infecting bacilli and the slow growth of the organism, so that there is insufficient antigen detected by the immune system early after infection. A recent study from our group showed that in macaques adaptive T cells and innate lymphocytes are present in granulomas, but at early time points (4 weeks) innate lymphocytes are expressing the T-bet transcription factor that regulates cytokine and cytotoxic genes while the adaptive T cells are T-bet negative (Grant et al., 2022). At this early time point, a subset of CD4 T cells express the more global transcription factor RORα. By 12 weeks, the CD8 T cells in granulomas have a significant induction of Tbet, but it takes even longer for a reliably measurable frequency of T-bet+ CD4 T cells to appear. While the T-bet+ CD4 T cell population is enriched for cytokine production, the T-bet+ CD8 T cells produce minimal cytokines but express granzyme B. Thus, while innate lymphocytes likely contribute to early containment of Mtb infection in granulomas, there is a slow evolution of the functional adaptive T cell response. Even in mice, Mtb in lungs grows to a high level with minimal bacterial killing until after 4 weeks of infection. Mtb killing in macaque granulomas is not readily observed until ~10 weeks post-infection (Lin et al., 2014). This delay in functional adaptive T cell responses likely allows the Mtb bacillus to establish a foothold in the lungs resulting in either a persistent or progressive infection. Notable was that a majority of T cells in granulomas express none of the five transcription factors analyzed (Tbet, RORα, RORγ, GATA3, or Foxp3) (Grant et al., 2022). It has also been shown that a majority of T cells in granulomas do not express cytokines (Gideon et al., 2015; Grant et al., 2022). These findings suggest that many of the T cells are not stimulated in granulomas perhaps due to granuloma structure or that many are not Mtb antigen specific and perhaps traffic to the granuloma in response to inflammation (Millar et al., 2021). We previously investigated whether T cells in granulomas were functionally exhausted but did not find evidence for this (Wong et al., 2018); this is supported by the lack of exhaustion markers on T cells in human granulomas based on spatial imaging via MIBI-TOF (McCaffrey et al., 2022). An effective vaccine likely needs to either prevent infection or induce Mtb-specific T resident memory cells that can quickly limit or extinguish infection when the bacilli enter the lungs.

THE B CELL AND ANTIBODY RESPONSE

Antibodies (Ab) exert their antimicrobial activities via the antigen-binding fragment (Fab) and the interaction of the crystallizable fragment (Fc) with Fc receptors (FcRs) on effector cells (Bournazos et al., 2015; Chan et al., 2014; Lu et al., 2018), regulating neutralization, opsonization, opsonophagocytosis, complement fixation, and a broad range of effector functions of innate immune cells, including antigen presentation and cytokine production (Figure 3). The effect of the Fc-FcγR interaction is particularly far reaching, as FcγRs are expressed on a wide variety of immune cells (Bournazos et al., 2015; Chan et al., 2014; Lu et al., 2018). Ample evidence indicates that Ab and B cells (via antigen presentation and cytokine and Ab production) can protect against microbes (including intracellular pathogens) by influencing T cells responses, such as the development of memory, as well as by modulating vaccine efficacy (Bournazos et al., 2015; Chan et al., 2014; Lu et al., 2018; Lund, 2008; Lund and Randall, 2010). B cell and Ab can thus play a role in inducing protection and improving vaccine efficacy against infection for which cell-mediated immune responses are pivotal in controlling the pathogens (Figure 3).

Figure 3. The B cell and humoral immune response to Mtb.

Figure 3.

The host response to Mtb begins with the presentation of Mtb antigens (Ag) by antigen-presenting cells to cognate CD4+ and CD8+ T cells, initiating the development of specific T cell lineages (1). In parallel, naive B cells interact with Mtb component (2), which results in the development of activated B cells. The initial encounters of T cells and B cells with Mtb Ag are an essential first step that leads to the formation in the germinal center of T follicular helper cells (Tfh) and germinal center B cells (3), which interaction is critical for the generation of memory B cells (4), and long-lived antibody (Ab)-producing plasma cells (5). Memory B cells can react rapidly in response to Mtb antigenic challenge, and the plasma cells (5), which produce high quality Ab (6), are critical for the generation and maintenance of serological memory. These latter two components are pivotal to the development of a robust and effective humoral immune response to microbes and Ab-based vaccines. In addition, Mtb Ag-activated B cells (7) can produce soluble factors such as pro-inflammatory and anti-inflammatory cytokines to modulate the development of anti-tuberculosis immunity (8). Additionally, the multifunctional B cells can contribute to the regulation of the immune response to Mtb via other effector functions such as the presentation of Ag to T cells (9). Together, the capacity of B cells to present Ag and to produce cytokines and Mtb-specific Ab can contribute significantly to the regulation of the granulomatous response during Mtb infection. These properties of B cells, in turn, play a role in mediating interaction between B cell and humoral immunity to the broad gamut of immune cells present in the tuberculous granuloma (Figure 1) to determine infection and disease outcomes, granuloma heterogeneity, and the levels of immunopathology (10). Of the major effector functions of B cell and humoral immunity (6, 8, and 9), the role of Ab in modulating anti-tuberculosis immune responses is perhaps the best studied. Specific humoral responses are associated with distinct infection and disease outcomes in human subjects. These are likely related to diverse Ab-mediated effector functions (11) as regulated by differential glycosylation of the Fc fragment of immunoglobulins (Ig) and the distinct functionality of the range of Ab isotypes. The interaction between the Fc of Ab and Fc receptors (FcR) can significantly regulate Ab-mediated effector functions of Ig to (1) modulate opsonophagocytosis, (2) block invasion of the pathogen, (3) enhance dendritic cell (DC) function via engagement of immune complex (IC) by DC to augment T cell responses, (4) mediate Ab-dependent cell-mediated cytotoxicity (ADCC) by targeting NK cells, and (5) Ab-dependent cell-mediated phagocytosis (ADCP) by targeting macrophages and neutrophils. These Ab-mediated effector functions, in turn, modulate infection and disease outcomes, granuloma heterogeneity, and the levels of immunopathology (12).

The notion that B cells and Ab can protect against Mtb is still a topic of considerable debate (Chan et al., 2014), not least because of the apparent low risk for developing tuberculosis in individuals receiving rituximab, a B-cell-depleting mAb (Cantini et al., 2014). Since rituximab is not efficacious in depleting plasma cells, anti-tuberculous serological memory imparted by plasma cells could still be operative in persons at risks for developing tuberculosis, thereby averting overt disease manifestation. Nevertheless, passive immunization studies using Mtb antigen-specific monoclonal Ab (mAb) support a protective role for Ab in tuberculosis (Chan et al., 2014). Existing evidence supports that B cells and Ab modulate immune responses to Mtb (Chan et al., 2014) and are required for the development of optimal anti-tuberculosis immunity, regulating cytokine production, neutrophilic response and granulomatous inflammation, and modulating vaccine-elicited immunity in mice. A role for B cells and Ab in regulating immune responses to tuberculosis has also been observed in non-human primate tuberculosis models (Phuah et al., 2016, 2012). These studies prompted efforts in characterizing the anti-tuberculosis humoral immunity in persons manifesting a spectrum of infection and disease outcomes (Li et al., 2017; Lu et al., 2016, 2019; Chowdhury et al., 2018; Zimmermann et al., 2016) (Figure 3). Thus, IgG in sera from persons with LTBI is superior to that from patients with active disease in mediating NK cell and macrophage anti-tuberculosis effector functions, with a predilection for FcγRIII binding. These Abs exhibit enhanced capacity to activate NK cells that is associated with augmented Ab-dependent cell-mediated cytotoxicity (ADCC), and with increased ability to promote Ab-dependent cell-mediated phagocytosis (ADCP) and restrict Mtb growth in macrophages. These functional differences are associated with distinct patterns of Ab glycosylation (Lu et al., 2016, 2019), which is known to modulate IgG functions (Alter et al., 2018; Wang and Ravetch, 2019). The NK cell/ADCC observation is supported by an independent study examining three separate cohorts of varied demographics (Chowdhury et al., 2018). NK cells kill pathogen-infected cells by ADCC (Bournazos et al., 2015), a process initiated by binding of NK FcγRIII with the Fc portion of Ab that reacts with pathogen antigens expressed on the surface of infected host cells. NK cells (Heron et al., 2012) and IgG (Reynolds et al., 1991) (the key mediator of ADCC) are present in the alveolus, raising the possibility that ADCC may play a role in ablating infected macrophages at the early alveolar phase of infection (Bournazos et al., 2015). To leverage ADCC for Mtb control, identification of the mycobacterial antigens expressed on the surface of infected cells, particularly macrophages, the primary host cells for Mtb, is critical.

Further supporting a role for IgG in the control of tuberculosis, it has been shown that polyclonal IgG from persons with LTBI and the resister phenotype (see below) (Behr et al., 2021; Kroon et al., 2020; Simmons et al., 2018), but not from subjects with active tuberculosis, protects against Mtb challenge in mice (Chen et al., 2020; Li et al., 2017). Intriguingly, IgA mAb cloned from persons with active tuberculosis or LTBI inhibit Mtb invasion of epithelial cells and macrophages, while that of IgG1 subtype promote entry (Zimmermann et al., 2016), underscoring the importance of isotype-dependent Ab-mediated effector functions in controlling Mtb. Indeed, mice deficient in the inhibitory FcγRIIB exhibit enhanced Mtb containment and attenuated lung inflammation that was associated with an augmented pulmonary Th1 response (Maglione et al., 2008). Conversely, Mtb-infected mice deficient in the γ-chain shared by activating FcγRs exhibit enhanced susceptibility and exacerbated immunopathology concomitant with increased production of the immunosuppressive cytokine IL-10. Thus, engagement of distinct FcγR can divergently affect cytokine production and susceptibility during Mtb infection. Elucidation of the Fc-FcγR reaction and the downstream events triggered by this interaction will likely shed light on mechanisms that regulate the development of anti-tuberculosis immunity. Cloning and molecular characterization of mAb from infected hosts (Ishida et al., 2021; Watson et al., 2021; Zimmermann et al., 2016), particularly in full length, will advance our understanding of how Abs regulate anti-tuberculosis immunity.

A role for the less-studied IgM in protection against Mtb is supported by a recent i.v. BCG immunization study in NHP showing that this vaccination strategy elicits a superior Ab response in blood and BAL fluid, relative to that engendered via conventional intradermal vaccination, with IgM being the most prominent isotype associated with bacterial burden control (Irvine et al., 2021). Both natural and immune IgM can protect against microbes, including intracellular pathogens (Baumgarth, 2011; Racine and Winslow, 2009). Innate-like B-1 B cells, which can play a role in infection (Smith and Baumgarth, 2019), are the primary producer of natural IgM (Baumgarth, 2011; Racine and Winslow, 2009). While evidence exists that B-1 B cells contribute to immune responses to Mycobacterium (Acosta et al., 2021), their role in defense against Mtb remains to be examined. Ab, including IgM, are present in airways (Reynolds et al., 1991; Torrelles and Schlesinger, 2017) and thus can potentially mediate antimicrobial effects in the early innate phase of Mtb infection. Vaccines protocols that can engender rapid appearance of Mtb-specific Ab in the airway upon Mtb entry may prove protective.

Recent advances in our understanding of tuberculosis epidemiology and of the highly complex granulomas in animals and humans (Cadena et al., 2017; Cohen et al., 2022; Gideon et al., 2022; McCaffrey et al., 2022; Wells et al., 2021) have led to reconsideration of the definitions of the various Mtb infection and disease states. Much debate revolves around the immunological characteristics of resisters (Behr et al., 2021; Simmons et al., 2018). The level of heterogeneity of subjects categorized into distinct states likely parallels that of lung granulomas (Cadena et al., 2017; Gideon et al., 2022) (Figure 1); this may account, at least in part, for the variability of results in human tuberculosis studies. Precise definitions for the various infection and disease states will improve study design and can impact tuberculosis public health policy (Behr et al., 2021; Simmons et al., 2018). One such state that has recently become a focus of investigation is that of the resisters, defined as individuals who remain TST- or IGRA (interferon-γ release assay)-negative despite extensive Mtb exposure) (Behr et al., 2021; Simmons et al., 2018). However, the immunological and microbiological definition of this entity is unclear (Behr et al., 2021; Simmons et al., 2018). A recent study supports the notion that resisters are or were in fact infected and developed adaptive immunity to Mtb, as the subjects exhibit a non-IFN-γ T cell response to the diagnostic Mtb-specific antigens ESAT6 and CFP10, and their Mtb-specific Ab display class switching and relatively high binding avidity (Lu et al., 2019). Thus, resisters most likely represent a heretofore uncharacterized state of Mtb infection. The Ab response of resisters exhibits features distinct from that of LTBI subjects, with a preponderance of IgG1 that is associated with unique glycosylation patterns. The non-IFN-γ T cell response is associated with augmented CD40L expression. CD40L is expressed by a wide range of cells, which engages with its receptor CD40 to regulate innate and adaptive immunity, including T-cell-dependent B cell activation (Elgueta et al., 2009; Parker, 1993). It is thus possible that the CD40L-high CD4 T cells of resisters are poised to drive B cell development. More studies are required to assess the significance of the resister CD40L phenotype and the non-IFN-γ T cell response. It would be of interest to determine whether resisters remain infected long-term and whether their anti-tuberculosis immunity is superior to that of LTBI subjects. A protective role for resister IgG is reinforced by the observation that passive transfer of IgG from an independent “resister” cohort protects against Mtb in mice (Li et al., 2017).

The germinal center (GC) reaction is critical for the development of effective B cell and humoral immunity (Victora and Nussenzweig, 2022). Within the GC, developing B cells receive help from T follicular helper cells (Tfhs) to attain a durable, high-affinity Ab response, with the development of memory B cells and long-lived plasma cells (Victora and Nussenzweig, 2022). GC-like structures are present in the lungs of Mtb-infected hosts, including humans (Chan et al., 2014; Tsai et al., 2006; Ulrichs et al., 2004). There is evidence that these ectopic GCs (Pitzalis et al., 2014) are associated with favorable Mtb infection outcome (Foreman et al., 2016; Khader et al., 2009; Slight et al., 2013), but their significance in regulating immune responses to Mtb, particularly locally in the lungs, has not been formally evaluated. Despite the critical role of Tfh in promoting the development of humoral immunity and the recent recognition of B cells and Ab as important factors in defense against Mtb in humans (Chan et al., 2014; Li et al., 2017; Lu et al., 2016, 2019; Chowdhury et al., 2018; Zimmermann et al., 2016), the precise role of Tfh in tuberculosis remains incompletely defined. It is possible that subjects manifesting variable infection and disease outcomes with varied Ab profiles may harbor distinct Tfh subsets that can differentially regulate humoral responses in tuberculosis, thereby influencing the susceptibility to infection or disease.

Emerging evidence also suggests that B cells have the capacity to modulate immune responses to Mtb (Chan et al., 2014). B cell subsets with distinct phenotypic markers are associated with varied infection and disease states of tuberculosis (Joosten et al., 2016; Loxton and van Rensburg, 2021). The roles of these subsets in Mtb infection remain to be defined. B cells can regulate immune responses through the production of a broad range of cytokines (Chan et al., 2014; Fillatreau, 2018; Lund, 2008). For example, B cells generate IFN-β, a type I IFN, upon exposure to Mtb or its components (Bénard et al., 2018). Importantly, B cells in human tuberculous pleural fluid express high levels of type I IFN mRNA. Type I IFN promotes the development of anti-inflammatory human and mouse macrophages (Bénard et al., 2018), and Mtb can induce type I IFN expression to decrease resistance to host defense (Manca et al., 2001; Wiens and Ernst, 2016) by attenuating IL-1α/β production (Mayer-Barber et al., 2011; Novikov et al., 2011). It is thus possible that B cell type I IFN plays a role in diminishing anti-tuberculosis immunity. It will be of interest to determine the relative contribution of B cell type I IFN compared with that produced by other cells at the site of infection. In a tumor model, B cells generate the neurotransmitter GABA (γ-aminobutyric acid) to drive the development of anti-inflammatory IL-10-producing macrophages, resulting in attenuation of CD8 T cell cytotoxicity (Zhang et al., 2021). In a low-dose virulent Erdman Mtb infection in mice, B cell deficiency results in exacerbated granulomatous inflammation characterized by IL-17-driven neutrophilia (Kozakiewicz et al., 2013; Maglione et al., 2007). Conversely, in chronic tuberculosis induced by the Mtb CD1551 strain, the granulomatous response in B-cell-deficient mice is attenuated relative to wild type (Bosio et al., 2000). While these divergent results can be due to the use of dissimilar Mb strains, it is possible that B cells in acute and chronic tuberculosis are functionally different as a result of distinct Ab repertoires against antigens differentially expressed in the two phases of infection (Lavollay et al., 2008), varied cytokine profiles (Chan et al., 2014; Fillatreau, 2018; Lund, 2008) and/or capacity to present antigens (Lanzavecchia, 1990; Vascotto et al., 2007). In chronic tuberculosis, excessive inflammation can cause undesirable lung damage in the form of cavitary lesions which promote Mtb transmission (Bhattacharya et al., 2021; Hunter, 2020), a hindrance to tuberculosis control (Hunter, 2020; Modlin and Bloom, 2013). It is thus possible that B cells can be targeted to curb transmission.

B cells and Abs are present in abundance amid a wide variety of immune cells in the lungs of an infected host (Phuah et al., 2012, 2016; Tsai et al., 2006) and play a role in regulating anti-tuberculosis immunity (Carpenter and Lu, 2022; Chan et al., 2014). The local environment of airway, granuloma, or lymph node is likely to promote the interaction of the multifunctional B cells and Ab with other immunological factors (Figure 3). It is these intricate interactions between the diverse granuloma immune cell types and factors and cross-modulation that will ultimately shape the development of innate and adaptive immune responses to Mtb to influence infection and disease outcomes of tuberculosis.

MYCOBACTERIAL HETEROGENEITY AND ANTIBIOTIC TOLERANCE: FROM THE PERSPECTIVE OF MACROPHAGE-MTB INTERACTION

Microbes, including Mtb, exhibit antibiotic persistence, an innate phenotypic heterogeneity characterized by the survival of a small subpopulation (<1%) of drug-sensitive bacteria during in vitro treatment with a bactericidal antibiotic without being genetically resistant (Balaban et al., 2019; Bigger, 1944; Dhar and McKinney, 2007; Lewis, 2010). Bigger designated the surviving bacteria “persisters” and posited that this phenomenon is clinically relevant (Bigger, 1944). Studying the persisters in Mtb-infected mice treated with anti-tuberculosis drugs (McCune et al., 1956, 1966a, 1966b; McCune and Tompsett, 1956), McDermott espoused the significant relevance of this phenomenon in the treatment of infections, including that caused by Mtb. Accumulating evidence support that stresses exerted upon Mtb by infected macrophages augment bacterial heterogeneity, a state that supports survival and the development of antibiotic tolerance (Adams et al., 2011; Chung et al., 2022; Hicks et al., 2018; Liu et al., 2016; Manina et al., 2015; Pisu et al., 2021). Drug tolerance represents an intermediate step toward attaining drug resistance (Hicks et al., 2018; Levin-Reisman et al., 2017), which poses a major obstacle to effective tuberculosis control (WHO, 2022). Manina et al. showed that the levels of Mtb heterogeneity in standard in vitro growth condition can be augmented by a number of stresses, including those imposed by host immunity stemming from infected macrophages and in mice (Manina et al., 2015). A non-growing yet metabolically active Mtb subpopulation was only present in bacteria procured from the lungs of wild type but not immunocompromised IFN-γ-deficient mice, supporting a role for host immune pressure in the development of Mtb heterogeneity. This study also suggested the existence of mechanisms of drug persistence that go beyond the acquisition of a slow growth or non-replicative states, cellular parameters generally thought to be associated with this phenomenon (Balaban et al., 2013). Indeed, it has been shown that macrophage-induced expression of an efflux pump by actively growing Mtb can mediate drug tolerance (Adams et al., 2011).

The significance of host immunity in driving Mtb heterogeneity has been underscored in a recent study, showing that enhanced mutation rates are detected in isolates collected from HIV-negative but not HIV-positive subjects with active tuberculosis prior to the initiation of anti-tuberculous therapy (Liu et al., 2020). Using fitness reporter Mtb strain to probe the relationship of host immune pressure and the development of drug tolerance in vivo (Liu et al., 2016), it was shown that levels of Mtb tolerance to INH and rifampin derived from lung myeloid cells of Mtb-infected mice treated with these two agents are proportional to the degree of activation of the host cells (Liu et al., 2016). In addition, in vitro experiments demonstrated that activated macrophages promote the capacity of Mtb to develop tolerance to anti-tuberculosis drugs (Liu et al., 2016). Thus, it appears that the host macrophage environment contributes significantly to the development of drug tolerance, the latter an obstacle to tuberculosis treatment.

THE UNDERSTUDIED NON-MACROPHAGE MYELOID CELLS

Ample evidence support the notion that host-Mtb interaction involves a range of myeloid cells in the lungs of a tuberculous host, including macrophages, dendritic cells, monocytes, neutrophils, and even mast cells (Gideon et al., 2022; McCaffrey et al., 2022; Srivastava et al., 2014; Wolf et al., 2007), all of which are present in granulomatous tissues in mice (Srivastava et al., 2014; Tsai et al., 2006; Wolf et al., 2007), NHP (Gideon et al., 2022), and humans (McCaffrey et al., 2022). Thus, infected myeloid cell subsets can contribute to shaping the immune responses to Mtb and modulating the overall biology of Mtb, thereby influencing the outcome of the infection (Srivastava et al., 2014). Cohen et al. (2018) showed that in the acute phase of murine tuberculosis, the infection foci established in the interstitium by aggregates of translocated Mtb-infected AM mediate dissemination of bacilli into recruited neutrophils and monocyte-derived macrophages. What remains to be defined is how this initial bacterial dissemination into recruited myeloid cells occurs and the precise downstream events following this spread. In a zebra fish model of Mycobacterium marinum infection, resident macrophages in the hindbrain of the infected Danio are the first immune cells that the invading pathogen enters and therefore, could be considered the AM equivalent of Mtb lung infection (Cambier et al., 2017, 2014). M. marinum phenolic glycolipids triggered the production of CXCL2 by infected hindbrain macrophages, leading to the recruitment of the more growth permissive monocytes, to which bacilli within macrophages are transferred to cause disease dissemination (Cambier et al., 2014, 2017). These observations suggest that the neutrophils and monocytes contribute to the immune response to Mtb and to disease pathogenesis, but relative to macrophages, these versatile myeloid cells (Borregaard, 2010; Galli et al., 2011; Swirski et al., 2014; Xie et al., 2020) are understudied (Pisu et al., 2021; Srivastava et al., 2014; Wolf et al., 2007). For example, the precise role in tuberculosis of neutrophils, perhaps the first immune cells that arrive at the site of Mtb infection (Seiler et al., 2003; Tsai et al., 2006), remains to be defined. Evidence exists that neutrophilia may reflect failed Th1 immunity in tuberculosis (Nandi and Behar, 2011) and that neutrophils can modulate vaccine efficacy (Kozakiewicz et al., 2013). The interaction of Mtb with neutrophils can augment dendritic cell migration to lymph nodes to promote the initiation of adaptive immunity (Blomgran and Ernst, 2011). A role for neutrophils to protect against Mtb remains controversial and is likely contextual (see references in Blomgran and Ernst, 2011; Kozakiewicz et al., 2013). Neutrophilia has been associated with exuberant lung pathology and poor Mtb control in susceptible mice (Eruslanov et al., 2005; Keller et al., 2006). Neutrophils, recruited to infected lung via an IL-1b- and 12/15-lipoxygenase-dependent pathway, have been reported to provide an environment with nutrients that promote Mtb growth, and this phenomenon has clinical implications (Mishra et al., 2017). More recently, it has been shown that loss of the autophagy factor ATG5 in neutrophils enhances Mtb susceptibility and ATG5 can attenuate immunopathology caused by neutrophils (Kimmey et al., 2015), and specific apoptotic pathways in neutrophils can mediate protection against Mtb (Stutz et al., 2021). It is well established that a neutrophil and type 1 IFN signature is observed in blood from humans with active tuberculosis (Berry et al., 2010) and is recapitulated in mice (Moreira-Teixeira et al., 2020). A role for myeloid cells in shaping the immune response to Mtb is further underscored by a recent study designed to map specific microenvironments in tissues procured from humans with active tuberculosis using multiplexed ion beam imaging by time of flight (MIBI-TOF) to visualize 37 proteins (McCaffrey et al., 2022). Of note, one of the microenvironments, made up of myeloid cells comprising macrophages, neutrophils, monocytes, and dendritic cells, which displays expression of the immunoregulatory molecules indolamine 2,3-dioxygenase 1 and PD-L1, is characterized by the absence of IFN-g expression and enhanced levels of TGF-b and regulatory T cells. The immunoregulatory features of this myeloid microenvironment is apparent in the blood of tuberculosis patients as assessed by transcriptomic analysis. Importantly, PD-L1 expression is associated with progression to active tuberculosis. It is possible that the immunoregulatory cells in the myeloid microenvironment reported by McCaffrey et al. (2022) may be related to myeloid-derived suppressor cells described in tuberculosis (Dorhoi et al., 2020). The role of this myeloid niche in tuberculous granulomatous tissues in modulating the immune response to Mtb warrants further investigation.

TYPE 2 IMMUNE RESPONSES

Although historically, tuberculosis has been considered to be dominated by type 1 immune responses, including Th1 T cells, recent studies support that type 2 immune cells and responses may play a role in Mtb infection outcome. Eosinophils are found in human and macaque granulomas, although usually at low levels, and at least in mice appear to participate in control of Mtb (Bohrer et al., 2021). Mtb does not appear to infect eosinophils in mice, macaques, or human granulomas but instead these cells influence the microenvironment of the granuloma and are early to arrive at the site of infection. Mast cells are another key player in type 2 immune responses. From scRNA-seq data on macaque granulomas at 10 weeks post-infection, mast cells and plasma cells are found together in granulomas that arise early in infection and have high bacterial burdens (Gideon et al., 2022). Although there was no evidence for IgE production by the plasma cells, the mast cells demonstrated expression of IL-13 (Gideon et al., 2022). A fascinating study by Cronan et al. demonstrated in a zebrafish model using M. marinum that signaling through the IL-4 receptor (by IL-4 or IL-13) and Stat6 was critical to early granuloma formation, specifically by differentiation of macrophages into epithelioid macrophages (Cronan et al., 2021). Without Stat6 signaling, granulomas in the fish did not form properly and had poor control of M. marinum. Interaction analyses in the scRNA-seq granuloma study suggested that mast and plasma cells drive an early self-reinforcing type 2 immune environment (Gideon et al., 2022). The presence of fibroblasts at higher levels in conjunction with mast cells and plasma cells in high burden granulomas could indicate that this constellation of cells reflects a wound-healing environment, possibly trying to initiate and wall off the granuloma when adaptive responses have not yet begun to kill the bacilli. A recent study showed co-localization of Mtb antigens with mast cells in human lung tissue as well as production of cytokines including IL-17 that may influence migration of cells to granulomas and proteases that may contribute to fibrosis (Garcia-Rodriguez et al., 2021). Thus, type 2 immune responses may participate in control of infection through early granuloma formation and function and balancing of type 1 and other inflammatory responses but also may exacerbate disease depending on timing and location of the cytokines and cells during infection.

VACCINES AND CLINICAL CONUNDRUMS

To truly stem the tide of tuberculosis, we need new and effective vaccines. For the first time in decades, there have been positive developments in the vaccine space. In clinical trials, a subunit vaccine, M72, composed of two Mtb proteins with AS01E adjuvant was shown to reduce the incidence of active tuberculosis in people with clinically latent infection by 49.7% (Tait et al., 2019). Presumably, this subunit vaccine boosted CD4 T cell responses in LTBI subjects that reduced the chance of progressing to active tuberculosis. The same vaccine was tested in rhesus macaques, but as a prevention of infection or disease in BCG-vaccinated macaques. Naive (i.e., without prior Mtb infection) macaques were vaccinated intradermally with BCG and then boosted with M72/AS01 or M72 proteins expressed in an Ad5 viral vector (Darrah et al., 2019). In this scenario, M72 did not provide protection against tuberculosis disease beyond the small amount of protection provided by BCG. It remains to be seen in clinical trials whether M72/AS01E will provide protection against Mtb infection or disease in humans without LTBI.

Another recent clinical trial involved revaccination with BCG (Nemes et al., 2018). Humans previously vaccinated at birth with BCG were revaccinated intradermally with the standard human BCG dose or with an adjuvanted subunit vaccine H4. Although there was no difference in the rate of Mtb infection in any group, as assessed by the conversion of the IGRA, which is a blood-based assay that measures T cell IFN-γ production to Mtb-specific proteins, BCG revaccination showed a significant decrease in sustained IGRA conversion compared with those who were not revaccinated (46.3% versus 24.5%). In other words, a higher percentage of BCG revaccinated people who had converted their IGRA to positive then reverted to negative. H4 vaccination also increased the IGRA reversion rate, although not significantly different than the control group. As noted, the control group (BCG at birth but no subsequent vaccination) had a relatively high rate of IGRA reversion (25%).

What IGRA reversion signifies clinically is not clear. It could mean clearance of initial Mtb infection or lower Mtb bacterial burden, or it could point out the inadequacies of IGRA as a diagnostic measure. In fact, in human cohorts with known Mtb exposures who remain IGRA negative, known as “resisters,” it was demonstrated that in fact resisters do have T cell and antibody responses to Mtb proteins, but their T cells do not produce IFN-γ in response to ESAT-6 and CFP10 (Lu et al., 2019). Resisters may have a lower risk of progression to active tuberculosis (Stein et al., 2019). Recent data indicate that IGRA reverters do have T cell responses to Mtb proteins as measured by flow cytometry indicating that they were productively infected, and their responses are consistent with well-controlled infection (Mpande et al., 2021). It is possible that “resisters” and “reverters” are similar phenotypes, depending on when the IGRA assays were done post-exposure. More studies are needed to determine what IGRA reversion and persistent IGRA-negative results in Mtb-exposed persons signify in terms of risk of active tuberculosis. Since diagnosis of possible Mtb infection relies on measuring a small fraction of the immune responses to the pathogen rather than the actual presence of the pathogen, the interpretation of a positive or negative IGRA in terms of risk of disease remains challenging. This highlights the failings of our common diagnostic tools for detecting Mtb infection and disease, especially in developing countries. Recent studies have demonstrated the potential utility of transcriptional signatures of risk of disease in humans with LTBI, some that can identify development of active tuberculosis 12–18 months prior to symptoms (Penn-Nicholson et al., 2020; Suliman et al., 2018; Zak et al., 2016).

In the last several years, there have been studies in non-human primates demonstrating that robust protection against tuberculosis, even sterilizing immunity against infection, is possible, providing hope for a path to effective vaccines. Rhesus macaques are quite susceptible to tuberculosis, with nearly all animals succumbing to active tuberculosis within a few months post-infection with a virulent Mtb strain. A rhesus CMV vectored vaccine, expressing 6–9 Mtb proteins, provided nearly 70% protection against Mtb disease in rhesus macaques, and induced strong T cell responses (Hansen et al., 2018). An attenuated Mtb strain resulted in reduced disease following challenge with a less virulent Mtb strain in rhesus macaques (Kaushal et al., 2015). The only approved tuberculosis vaccine for humans, BCG, is administered intradermally to newborns. However, changing the route to i.v. and increasing the dose of BCG in rhesus macaques resulted in remarkable protection (90%), including sterilizing immunity, against Mtb infection or disease (Darrah et al., 2020). In contrast, intradermal BCG at the standard dose or high-dose aerosolized BCG did not provide protection against disease in macaques; high-dose intradermal BCG was not significantly better than low-dose BCG ID, but there was a subset of animals that showed some protection against disease. It is possible that higher doses of intradermal BCG in humans, possibly even in a revaccination trial, could improve protection, although additional studies would be needed. In contrast, delivery of BCG via bronchoscope directly in the airways of rhesus macaques resulted in protection against disease and perhaps even against infection in a subset of animals (Dijkman et al., 2019).

The rhesus macaque model sets a very high bar for protection as these animals are much more susceptible than humans to infection and disease. Thus, these recent successes offer both hope and opportunities to define what is necessary for an effective vaccine in humans. BCG delivered either by the i.v. route or via bronchoscope directly into the lungs resulted in much higher levels of mycobacteria-specific T resident memory cells in lungs compared with BCG delivered via other routes (Darrah et al., 2020). Although this has not been reported for the CMV-tuberculosis vaccine, one can speculate that the persistence of the CMV vector and the attenuated Mtb strain may also induce T resident memory cells or other protective factors in tissues. Unlike studies in mice, concurrent infection with Mtb in macaques provides robust protection against reinfection, providing yet one more model of protection that can be useful for delineating mechanisms of protection (Cadena et al., 2018). Ongoing and future studies including depletion of T cell subsets or B cells, neutralization of cytokines, and in-depth analyses of local immune responses in these vaccination and protection models can point to the critical protective mechanisms. These studies will provide crucial information about how best to construct an effective vaccine for humans.

It is also important to identify correlates of protection, which are not necessarily mechanisms of protection. Correlates of protection are critical for clinical vaccine trials and should be measurable in an easily accessible sample, such as blood (Nemes et al., 2022). Correlates may be a combination of innate responses, T cells, antibodies, or even gene expression or metabolic signatures. Such studies are being conducted in the clinical trials that have provided some measure of success. Correlates can also be assessed in the animal models of vaccine-induced protection, which will be important as more vaccines move into clinical trials.

It must be acknowledged that much of our understanding of events in the tissues, including formation, establishment, progression, and regression of granulomas, comes from animal models—mice, non-human primates, zebrafish, and others. Such studies are next to impossible to perform in humans. However, most studies in animal models are in naive (with respect to mycobacteria) animals, while in most countries humans are vaccinated at birth with BCG. In addition, in some parts of the world, exposure to non-tuberculous or environmental mycobacteria may influence subsequent exposure to Mtb. These present a limitation to translation of animal studies to humans. This is particularly important for vaccine studies, since vaccines must perform in humans who have been previously vaccinated with BCG. There are ongoing studies in macaques to vaccinate infants with BCG for future novel vaccine studies, but these are limited and expensive. Nonetheless, it is important to consider these limitations in interpreting the results from animal studies.

Due to the nature of Mtb infection and the lack of validated correlates of protection, clinical trials of vaccines are long and expensive, requiring large numbers of participants. Several infectious disease fields have benefited from human challenge models for assessing vaccines, including influenza and malaria (Minhinnick et al., 2016). Human challenges provide a rapid assessment of vaccine efficacy in a controlled setting with relatively few volunteers. However, a human challenge model of Mtb infection poses questions of safety and feasibility, including the difficulty in assessing infection or bacterial burden in vivo, the potential for latent infection that could reactivate later, and the long treatment regimen using drugs with known side effects. Still, there are efforts in this arena, mostly focusing on BCG challenge, either in skin (Minhinnick et al., 2016) or in lungs. PPD pulmonary challenge in persons with LTBI has been performed for several years to identify the recall response in lungs (Silver et al., 2003; Walrath et al., 2005). BCG skin challenge in the setting of vaccines has been done in humans with some modest success (Harris et al., 2014). However, whether the skin immune responses reflect the critical lung immune responses in terms of protection is unclear. BCG lung challenges have also been performed in proof-of-concept studies (Davids et al., 2020), although not yet in the setting of vaccines. Although difficult to develop, safe human challenge models for tuberculosis would be game changer in the vaccine world.

CONCLUDING REMARKS

The immune response to M. tuberculosis is complex and multilayered, and there are still many unanswered questions about protective or exacerbating immune responses. It is impossible to be truly comprehensive in a single review on the immunology of tuberculosis, and thus, there are many important studies that were not included in this short review. Our opinion is that the various host factors do not work in an independent fashion, either in airways, lungs, granulomas, or lymph nodes, but instead interact with each other. The sum total of immune interactions ultimately determines the outcome of infection, either by allowing or preventing initial infection, priming of the immune responses in lymph nodes, and influencing the outcome of each individual granuloma. There are multiple examples of cells and secreted factors, including chemokines, cytokines, and cytotoxic effector molecules, influencing other cells, not only in Mtb infection but in many other biological systems. We hypothesize that there are several interaction pathways that can lead to either success or failure in Mtb infection outcomes, involving multiple cell types and effectors. In other words, not all cell types or effectors are required in all situations, but that the long evolution of the human host with Mtb has resulted in multiple players that can contain or resolve infection. Similarly, Mtb survives and multiplies in humans and therefore there are pathogenic features of Mtb that resist elimination by the host, resulting in a host-pathogen standoff. The explosion of new data in the realm of tuberculosis immunology highlights various pathways to successful control or elimination of the bacillus through interventions or vaccines. It will likely require engagement of various arms of the immune system, as well as multiple cell types and effectors, with further research and translation of the findings to humans, to stem the tide of the tuberculosis epidemic.

ACKNOWLEDGMENTS

We thank the past and current members of the Flynn and Chan laboratories, our colleagues and collaborators, in particular, Sarah Fortune, for insightful discussions, interesting ideas, and support. We regret that, due to space limitation, many important references originically cited had to be removed. Funding is provided by NIH R01 AI114674, NIH R01 AI134183, NIH R01AI123093, NIH R56AI139053, NIH R01 AI143788, NIH NIAID 75N93019C00071, NIH R01 AI155495, NIH R01 AI169707, Bill and Melinda Gates Foundation, Wellcome Trust Delta LEAP to J.L.F.; and NIH R01 AI137344, NIH R01 AI146340 to J.C.; and NIH R01 AI139297 to J.C./J.L.F.

Footnotes

DECLARATION OF INTERESTS

Pending patent: A.K. Shalek, T. Hughes, M.H. Wadsworth, R. Seder, M. Roederer, J.L. Flynn, and P. Darrah, “COMPOSITIONS AND METHODS FOR TREATING BACTERIAL INFECTIONS,” US non-provisional patent application 17/137,481 claiming priority to US 62/954,998, filed December 30, 2020.

REFERENCES

  1. Acosta F, Fernández PL, and Goodridge A. (2021). Do B-1 cells play a role in response to Mycobacterium tuberculosis Beijing lineages? Virulence 13, 1–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Adams KN, Takaki K, Connolly LE, Wiedenhoft H, Winglee K, Humbert O, Edelstein PH, Cosma CL, and Ramakrishnan L. (2011). Drug tolerance in replicating mycobacteria mediated by a macrophage-induced efflux mechanism. Cell 145, 39–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Allie N, Grivennikov SI, Keeton R, Hsu NJ, Bourigault ML, Court N, Fremond C, Yeremeev V, Shebzukhov Y, Ryffel B, et al. (2013). Prominent role for T cell-derived tumour necrosis factor for sustained control of Mycobacterium tuberculosis infection. Sci. Rep 3, 1809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Alter G, Ottenhoff THM, and Joosten SA (2018). Antibody glycosylation in inflammation, disease and vaccination. Semin. Immunol 39, 102–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Ardain A, Domingo-Gonzalez R, Das S, Kazer SW, Howard NC, Singh A, Ahmed M, Nhamoyebonde S, Rangel-Moreno J, Ogongo P, et al. (2019). Group 3 innate lymphoid cells mediate early protective immunity against tuberculosis. Nature 570, 528–532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Balaban NQ, Gerdes K, Lewis K, and McKinney JD (2013). A problem of persistence: still more questions than answers? Nat. Rev. Microbiol 11, 587–591. [DOI] [PubMed] [Google Scholar]
  7. Balaban NQ, Helaine S, Lewis K, Ackermann M, Aldridge B, Andersson DI, Brynildsen MP, Bumann D, Camilli A, Collins JJ, et al. (2019).Definitions and guidelines for research on antibiotic persistence. Nat. Rev. Microbiol 17, 441–448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Barber DL, Sakai S, Kudchadkar RR, Fling SP, Day TA, Vergara JA, Ashkin D, Cheng JH, Lundgren LM, Raabe VN, et al. (2019). Tuberculosis following PD-1 blockade for cancer immunotherapy. Sci. Transl. Med 11, 475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Baumgarth N. (2011). The double life of a B-1 cell: self-reactivity selects for protective effector functions. Nat. Rev. Immunol 11, 34–46. [DOI] [PubMed] [Google Scholar]
  10. Behar SM, Dascher CC, Grusby MJ, Wang CR, and Brenner MB (1999). Susceptibility of mice deficient in CD1D or TAP1 to infection with Mycobacterium tuberculosis. J. Exp. Med 189, 1973–1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Behr MA, Edelstein PH, and Ramakrishnan L. (2019). Is Mycobacterium tuberculosis infection life long? BMJ 367, l5770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Behr MA, Kaufmann E, Duffin J, Edelstein PH, and Ramakrishnan L. (2021). Latent tuberculosis: two centuries of confusion. Am. J. Respir. Crit. Care Med 204, 142–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Bénard A, Sakwa I, Schierloh P, Colom A, Mercier I, Tailleux L, Jouneau L, Boudinot P, Al-Saati T, Lang R, et al. (2018). B cells producing Type I IFN modulate macrophage polarization in tuberculosis. Am. J. Respir. Crit. Care Med 197, 801–813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Berrien-Elliott MM, Becker-Hapak M, Cashen AF, Jacobs M, Wong P, Foster M, McClain E, Desai S, Pence P, Cooley S, et al. (2022). Systemic IL-15 promotes allogeneic cell rejection in patients treated with natural killer cell adoptive therapy. Blood 139, 1177–1183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Berry MP, Graham CM, McNab FW, Xu Z, Bloch SA, Oni T, Wilkinson KA, Banchereau R, Skinner J, Wilkinson RJ, et al. (2010). An interferon-inducible neutrophil-driven blood transcriptional signature in human tuberculosis. Nature 466, 973–977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Bhattacharya B, Xiao S, Chatterjee S, Urbanowski M, Ordonez A, Ihms EA, Agrahari G, Lun S, Berland R, Pichugin A, et al. (2021). The integrated stress response mediates necrosis in murine Mycobacterium tuberculosis granulomas. J. Clin. Invest 131, e130319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Bigger J. (1944). Treatment of staphylococcal infections with penicillin by intermittent sterilisation. Lancet 244, 497–500. [Google Scholar]
  18. Blomgran R, and Ernst JD (2011). Lung neutrophils facilitate activation of naive antigen-specific CD4+ T cells during Mycobacterium tuberculosis infection. J. Immunol 186, 7110–7119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Bohrer AC, Castro E, Hu Z, Queiroz ATL, Tocheny CE, Assmann M, Sakai S, Nelson C, Baker PJ, Ma H, et al. (2021). Eosinophils are part of the granulocyte response in tuberculosis and promote host resistance in mice. J. Exp. Med 218. e2021046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Borregaard N. (2010). Neutrophils, from marrow to microbes. Immunity 33, 657–670. [DOI] [PubMed] [Google Scholar]
  21. Bosio CM, Gardner D, and Elkins KL (2000). Infection of B cell-deficient mice with CDC 1551, a clinical isolate of Mycobacterium tuberculosis: delay in dissemination and development of lung pathology. J. Immunol 164, 6417–6425. [DOI] [PubMed] [Google Scholar]
  22. Bournazos S, DiLillo DJ, and Ravetch JV (2015). The role of Fc-FcγR interactions in IgG-mediated microbial neutralization. J. Exp. Med 212, 1361–1369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Bucsxan AN, Chatterjee A, Singh DK, Foreman TW, Lee TH, Threeton B, Kirkpatrick MG, Ahmed M, Golden N, Alvarez X, et al. (2019). Mechanisms of reactivation of latent tuberculosis infection due to SIV coinfection. J. Clin. Invest 129, 5254–5260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Cadena AM, Fortune SM, and Flynn JL (2017). Heterogeneity in tuberculosis. Nat. Rev. Immunol 17, 691–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Cadena AM, Hopkins FF, Maiello P, Carey AF, Wong EA, Martin CJ, Gideon HP, DiFazio RM, Andersen P, Lin PL, et al. (2018). Concurrent infection with Mycobacterium tuberculosis confers robust protection against secondary infection in macaques. PLoS Pathog. 14. e1007305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Cambier CJ, O’Leary SM, O’Sullivan MP, Keane J, and Ramakrishnan L. (2017). Phenolic glycolipid facilitates mycobacterial escape from microbicidal tissue-resident macrophages. Immunity 47, 552–565.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Cambier CJ, Takaki KK, Larson RP, Hernandez RE, Tobin DM, Urdahl KB, Cosma CL, and Ramakrishnan L. (2014). Mycobacteria manipulate macrophage recruitment through coordinated use of membrane lipids. Nature 505, 218–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Cantini F, Niccoli L, and Goletti D. (2014). Tuberculosis risk in patients treated with non-anti-tumor necrosis factor-α (TNF-α) targeted biologics and recently licensed TNF-α inhibitors: data from clinical trials and national registries. J. Rheumatol. Suppl 91, 56–64. [DOI] [PubMed] [Google Scholar]
  29. Capuano SV 3rd, Croix DA, Pawar S, Zinovik A, Myers A, Lin PL, Bissel S, Fuhrman C, Klein E, and Flynn JL (2003). Experimental Mycobacterium tuberculosis infection of cynomolgus macaques closely resembles the various manifestations of human M. tuberculosis infection. Infect. Immun 71, 5831–5844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Carpenter SM, and Lu LL (2022). Leveraging antibody, B cell and Fc receptor interactions to understand heterogeneous immune responses in tuberculosis. Front. Immunol 13, 830482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Caruso AM, Serbina N, Klein E, Triebold K, Bloom BR, and Flynn JL (1999). Mice deficient in CD4 T cells have only transiently diminished levels of IFN-gamma, yet succumb to tuberculosis. J. Immunol 162, 5407–5416. [PubMed] [Google Scholar]
  32. Chackerian AA, Alt JM, Perera TV, Dascher CC, and Behar SM (2002). Dissemination of Mycobacterium tuberculosis is influenced by host factors and precedes the initiation of T-cell immunity. Infect. Immun 70, 4501–4509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Chan J, Mehta S, Bharrhan S, Chen Y, Achkar JM, Casadevall A, and Flynn J. (2014). The role of B cells and humoral immunity in Mycobacterium tuberculosis infection. Semin. Immunol 26, 588–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Chen CY, Huang D, Wang RC, Shen L, Zeng G, Yao S, Shen Y, Halliday L, Fortman J, McAllister M, et al. (2009). A critical role for CD8 T cells in a nonhuman primate model of tuberculosis. PLoS Pathog. 5. e1000392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Chen T, Blanc C, Liu Y, Ishida E, Singer S, Xu J, Joe M, Jenny-Avital ER, Chan J, Lowary TL, and Achkar JM (2020). Capsular glycan recognition provides antibody-mediated immunity against tuberculosis. J. Clin. Invest 130, 1808–1822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Chung ES, Johnson WC, and Aldridge BB (2022). Types and functions of heterogeneity in mycobacteria. Nat. Rev. Microbiol 20, 529–541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Cohen SB, Gern BH, Delahaye JL, Adams KN, Plumlee CR, Winkler JK, Sherman DR, Gerner MY, and Urdahl KB (2018). Alveolar macrophages provide an early Mycobacterium tuberculosis Niche and initiate dissemination. Cell Host Microbe 24, 439–446.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Cohen SB, Gern BH, and Urdahl KB (2022). The tuberculous granuloma and preexisting immunity. Annu. Rev. Immunol 40, 589–614. [DOI] [PubMed] [Google Scholar]
  39. Cong CV, Ly TT, and Duc NM (2022). Primary lymphatic tuberculosis in children - Literature overview and case report. Radiol. Case Rep 17, 1656–1664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Cooper AM (2009). Cell-mediated immune responses in tuberculosis. Annu. Rev. Immunol 27, 393–422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Cronan MR, Hughes EJ, Brewer WJ, Viswanathan G, Hunt EG, Singh B, Mehra S, Oehlers SH, Gregory SG, Kaushal D, and Tobin DM (2021). A non-canonical type 2 immune response coordinates tuberculous granuloma formation and epithelialization. Cell 184, 1757–1774.e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Darrah PA, DiFazio RM, Maiello P, Gideon HP, Myers AJ, Rodgers MA, Hackney JA, Lindenstrom T, Evans T, Scanga CA, et al. (2019). Boosting BCG with proteins or rAd5 does not enhance protection against tuberculosis in rhesus macaques. NPJ Vaccines 4, 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Darrah PA, Zeppa JJ, Maiello P, Hackney JA, Wadsworth MH 2nd, Hughes TK, Pokkali S, Swanson PA 2nd, Grant NL, Rodgers MA, et al. (2020). Prevention of tuberculosis in macaques after intravenous BCG immunization. Nature 577, 95–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Dascher CC, Hiromatsu K, Xiong X, Morehouse C, Watts G, Liu G, McMurray DN, LeClair KP, Porcelli SA, and Brenner MB (2003). Immunization with a mycobacterial lipid vaccine improves pulmonary pathology in the guinea pig model of tuberculosis. Int. Immunol 15, 915–925. [DOI] [PubMed] [Google Scholar]
  45. Davids M, Pooran A, Hermann C, Mottay L, Thompson F, Cardenas J, Gu J, Koeuth T, Meldau R, Limberis J, et al. (2020). A human lung challenge model to evaluate the safety and immunogenicity of PPD and live Bacillus Calmette-Gué rin. Am. J. Respir. Crit. Care Med 201, 1277–1291. [DOI] [PubMed] [Google Scholar]
  46. Dhar N, and McKinney JD (2007). Microbial phenotypic heterogeneity and antibiotic tolerance. Curr. Opin. Microbiol 10, 30–38. [DOI] [PubMed] [Google Scholar]
  47. Diedrich CR, Mattila JT, Klein E, Janssen C, Phuah J, Sturgeon TJ, Montelaro RC, Lin PL, and Flynn JL (2010). Reactivation of latent tuberculosis in cynomolgus macaques infected with SIV is associated with early peripheral T cell depletion and not virus load. PLoS One 5, e9611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Diedrich CR, Rutledge T, Maiello P, Baranowski TM, White AG, Borish HJ, Karell P, Hopkins F, Brown J, Fortune SM, et al. (2020). SIV and Mycobacterium tuberculosis synergy within the granuloma accelerates the reactivation pattern of latent tuberculosis. PLoS Pathog. 16. e1008413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Dijkman K, Sombroek CC, Vervenne RAW, Hofman SO, Boot C, Remarque EJ, Kocken CHM, Ottenhoff THM, Kondova I, Khayum MA, et al. (2019). Prevention of tuberculosis infection and disease by local BCG in repeatedly exposed rhesus macaques. Nat. Med 25, 255–262. [DOI] [PubMed] [Google Scholar]
  50. Dorhoi A, Kotzé LA, Berzofsky JA, Sui Y, Gabrilovich DI, Garg A, Hafner R, Khader SA, Schaible UE, Kaufmann SH, et al. (2020). Therapies for tuberculosis and AIDS: myeloid-derived suppressor cells in focus. J. Clin. Invest 130, 2789–2799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. D’Souza CD, Cooper AM, Frank AA, Mazzaccaro RJ, Bloom BR, and Orme IM (1997). An anti-inflammatory role for gamma delta T lymphocytes in acquired immunity to Mycobacterium tuberculosis. J. Immunol 158, 1217–1221. [PubMed] [Google Scholar]
  52. Elgueta R, Benson MJ, de Vries VC, Wasiuk A, Guo Y, and Noelle RJ (2009). Molecular mechanism and function of CD40/CD40L engagement in the immune system. Immunol. Rev 229, 152–172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Eruslanov EB, Lyadova IV, Kondratieva TK, Majorov KB, Scheglov IV, Orlova MO, and Apt AS (2005). Neutrophil responses to Mycobacterium tuberculosis infection in genetically susceptible and resistant mice. Infect. Immun 73, 1744–1753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Fillatreau S. (2018). B cells and their cytokine activities implications in human diseases. Clin. Immunol 186, 26–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Flynn JL, Goldstein MM, Chan J, Triebold KJ, Pfeffer K, Lowenstein CJ, Schreiber R, Mak TW, and Bloom BR (1995). Tumor necrosis factor-alpha is required in the protective immune response against Mycobacterium tuberculosis in mice. Immunity 2, 561–572. [DOI] [PubMed] [Google Scholar]
  56. Flynn JL, Goldstein MM, Triebold KJ, Koller B, and Bloom BR (1992). Major histocompatibility complex class I-restricted T cells are required for resistance to Mycobacterium tuberculosis infection. Proc. Natl. Acad. Sci. USA 89, 12013–12017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Foreman TW, Mehra S, LoBato DN, Malek A, Alvarez X, Golden NA, Bucsxan AN, Didier PJ, Doyle-Meyers LA, Russell-Lodrigue KE, et al. (2016). CD4+ T-cell-independent mechanisms suppress reactivation of latent tuberculosis in a macaque model of HIV coinfection. Proc. Natl. Acad. Sci. USA 113, E5636–E5644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Galli SJ, Borregaard N, and Wynn TA (2011). Phenotypic and functional plasticity of cells of innate immunity: macrophages, mast cells and neutrophils. Nat. Immunol 12, 1035–1044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Ganchua SKC, Cadena AM, Maiello P, Gideon HP, Myers AJ, Junecko BF, Klein EC, Lin PL, Mattila JT, and Flynn JL (2018). Lymph nodes are sites of prolonged bacterial persistence during Mycobacterium tuberculosis infection in macaques. PLoS Pathog. 14. e1007337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Ganchua SKC, White AG, Klein EC, and Flynn JL (2020). Lymph nodes-The neglected battlefield in tuberculosis. PLoS Pathog. 16. e1008632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Garcia-Rodriguez KM, Bini EI, Gamboa-Domínguez A, Espitia-Pinzón CI, Huerta-Yepez S, Bulfone-Paus S, and Herná ndez-Pando R. (2021). Differential mast cell numbers and characteristics in human tuberculosis pulmonary lesions. Sci. Rep 11, 10687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Gibbings SL, Thomas SM, Atif SM, McCubbrey AL, Desch AN, Danhorn T, Leach SM, Bratton DL, Henson PM, Janssen WJ, and Jakubzick CV (2017). Three unique interstitial macrophages in the murine lung at steady state. Am. J. Respir. Cell Mol. Biol 57, 66–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Gideon HP, Hughes TK, Tzouanas CN, Wadsworth MH 2nd, Tu AA, Gierahn TM, Peters JM, Hopkins FF, Wei JR, Kummerlowe C, et al. (2022). Multimodal profiling of lung granulomas in macaques reveals cellular correlates of tuberculosis control. Immunity 55, 827–846.e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Gideon HP, Phuah J, Myers AJ, Bryson BD, Rodgers MA, Coleman MT, Maiello P, Rutledge T, Marino S, Fortune SM, et al. (2015). Variability in tuberculosis granuloma T cell responses exists, but a balance of pro- and anti-inflammatory cytokines is associated with sterilization. PLoS Pathog. 11. e1004603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Ginhoux F, and Guilliams M. (2016). Tissue-resident macrophage ontogeny and homeostasis. Immunity 44, 439–449. [DOI] [PubMed] [Google Scholar]
  66. Gold MC, Napier RJ, and Lewinsohn DM (2015). MR1-restricted mucosal associated invariant T (MAIT) cells in the immune response to Mycobacterium tuberculosis. Immunol. Rev 264, 154–166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Gopal R, Lin Y, Obermajer N, Slight S, Nuthalapati N, Ahmed M, Kalinski P, and Khader SA (2012). IL-23-dependent IL-17 drives Th1-cell responses following Mycobacterium bovis BCG vaccination. Eur. J. Immunol 42, 364–373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Grant NL, Maiello P, Klein E, Lin PL, Borish HJ, Tomko J, Frye LJ, White AG, Kirschner DE, Mattila JT, and Flynn JL (2022). T cell transcription factor expression evolves over time in granulomas from Mycobacterium tuberculosis-infected cynomolgus macaques. Cell Rep. 39, 110826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Green AM, Difazio R, and Flynn JL (2013). IFN-gamma from CD4 T cells is essential for host survival and enhances CD8 T cell function during Mycobacterium tuberculosis infection. J. Immunol 190, 270–277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Guilliams M, Thierry GR, Bonnardel J, and Bajenoff M. (2020). Establishment and maintenance of the macrophage niche. Immunity 52, 434–451. [DOI] [PubMed] [Google Scholar]
  71. Gupta RK, Lawn SD, Bekker LG, Caldwell J, Kaplan R, and Wood R. (2013). Impact of human immunodeficiency virus and CD4 count on tuberculosis diagnosis: analysis of city-wide data from Cape Town, South Africa. Int. J. Tuberc. Lung Dis 17, 1014–1022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Hansen SG, Zak DE, Xu G, Ford JC, Marshall EE, Malouli D, Gilbride RM, Hughes CM, Ventura AB, Ainslie E, et al. (2018). Prevention of tuberculosis in rhesus macaques by a cytomegalovirus-based vaccine. Nat. Med 24, 130–143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Harris SA, Meyer J, Satti I, Marsay L, Poulton ID, Tanner R, Minassian AM, Fletcher HA, and McShane H. (2014). Evaluation of a human BCG challenge model to assess antimycobacterial immunity induced by BCG and a candidate tuberculosis vaccine, MVA85A, alone and in combination. J. Infect. Dis 209, 1259–1268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Heron M, Grutters JC, ten Dam-Molenkamp KM, Hijdra D, van Heugten-Roeling A, Claessen AM, Ruven HJ, van den Bosch JM, and van Velzen-Blad H. (2012). Bronchoalveolar lavage cell pattern from healthy human lung. Clin. Exp. Immunol 167, 523–531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Hicks ND, Yang J, Zhang X, Zhao B, Grad YH, Liu L, Ou X, Chang Z, Xia H, Zhou Y, et al. (2018). Clinically prevalent mutations in Mycobacterium tuberculosis alter propionate metabolism and mediate multidrug tolerance. Nat. Microbiol 3, 1032–1042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Hoft DF, Brown RM, and Roodman ST (1998). Bacille calmette-guerin vaccination enhances human gamma Delta T cell responsiveness to mycobacteria suggestive of a memory-like phenotype. J. Immunol 161, 1045–1054. [PubMed] [Google Scholar]
  77. Huang L, Nazarova EV, Tan S, Liu Y, and Russell DG (2018). Growth of Mycobacterium tuberculosis in vivo segregates with host macrophage metabolism and ontogeny. J. Exp. Med 215, 1135–1152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Hunter RL (2011). Pathology of post primary tuberculosis of the lung: an illustrated critical review. Tuberculosis (Edinb) 91, 497–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Hunter RL (2020). The pathogenesis of tuberculosis-the Koch phenomenon reinstated. Pathogens 9, 813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Irvine EB, O’Neil A, Darrah PA, Shin S, Choudhary A, Li W, Honnen W, Mehra S, Kaushal D, Gideon HP, et al. (2021). Robust IgM responses following intravenous vaccination with bacille calmette-guerin associate with prevention of Mycobacterium tuberculosis infection in macaques. Nat. Immunol 22, 1515–1523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Ishida E, Corrigan DT, Malonis RJ, Hofmann D, Chen T, Amin AG, Chatterjee D, Joe M, Lowary TL, Lai JR, and Achkar JM (2021). Monoclonal antibodies from humans with Mycobacterium tuberculosis exposure or latent infection recognize distinct arabinomannan epitopes. Commun. Biol 4, 1181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. James CA, Yu KKQ, Gilleron M, Prandi J, Yedulla VR, Moleda ZZ, Diamanti E, Khan M, Aggarwal VK, Reijneveld JF, et al. (2018). CD1b tetramers identify T cells that recognize natural and synthetic diacylated sulfoglycolipids from Mycobacterium tuberculosis. Cell Chem. Biol 25, 392–402.e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Jasenosky LD, Scriba TJ, Hanekom WA, and Goldfeld AE (2015). T cells and adaptive immunity to Mycobacterium tuberculosis in humans. Immunol. Rev 264, 74–87. [DOI] [PubMed] [Google Scholar]
  84. Joosten SA, van Meijgaarden KE, Del Nonno F, Baiocchini A, Petrone L, Vanini V, Smits HH, Palmieri F, Goletti D, and Ottenhoff TH (2016). Patients with tuberculosis have a dysfunctional circulating b-cell compartment, which normalizes following successful treatment. PLoS Pathog. 12, e1005687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Kauffman KD, Sakai S, Lora NE, Namasivayam S, Baker PJ, Kamenyeva O, Foreman TW, Nelson CE, Oliveira-de-Souza D, Vinhaes CL, et al. (2021). PD-1 blockade exacerbates Mycobacterium tuberculosis infection in rhesus macaques. Sci. Immunol 6. eabf3861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Kaushal D, Foreman TW, Gautam US, Alvarez X, Adekambi T, Rangel-Moreno J, Golden NA, Johnson AM, Phillips BL, Ahsan MH, et al. (2015). Mucosal vaccination with attenuated Mycobacterium tuberculosis induces strong central memory responses and protects against tuberculosis. Nat. Commun 6, 8533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Keane J, Gershon S, Wise RP, Mirabile-Levens E, Kasznica J, Schwieterman WD, Siegel JN, and Braun MM (2001). Tuberculosis associated with infliximab, a tumor necrosis factor alpha-neutralizing agent. N. Engl. J. Med 345, 1098–1104. [DOI] [PubMed] [Google Scholar]
  88. Keller C, Hoffmann R, Lang R, Brandau S, Hermann C, and Ehlers S. (2006). Genetically determined susceptibility to tuberculosis in mice causally involves accelerated and enhanced recruitment of granulocytes. Infect. Immun 74, 4295–4309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Khader SA, Bell GK, Pearl JE, Fountain JJ, Rangel-Moreno J, Cilley GE, Shen F, Eaton SM, Gaffen SL, Swain SL, et al. (2007). IL-23 and IL-17 in the establishment of protective pulmonary CD4+ T cell responses after vaccination and during Mycobacterium tuberculosis challenge. Nat. Immunol 8, 369–377. [DOI] [PubMed] [Google Scholar]
  90. Khader SA, Rangel-Moreno J, Fountain JJ, Martino CA, Reiley WW, Pearl JE, Winslow GM, Woodland DL, Randall TD, and Cooper AM (2009). In a murine tuberculosis model, the absence of homeostatic chemokines delays granuloma formation and protective immunity. J. Immunol 183, 8004–8014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Kimmey JM, Huynh JP, Weiss LA, Park S, Kambal A, Debnath J, Virgin HW, and Stallings CL (2015). Unique role for ATG5 in neutrophil-mediated immunopathology during M. tuberculosis infection. Nature 528, 565–569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Kozakiewicz L, Chen Y, Xu J, Wang Y, Dunussi-Joannopoulos K, Ou Q, Flynn JL, Porcelli SA, Jacobs WR Jr., and Chan J. (2013). B cells regulate neutrophilia during Mycobacterium tuberculosis infection and BCG vaccination by modulating the interleukin-17 response. PLoS Pathog. 9. e1003472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Kroon EE, Kinnear CJ, Orlova M, Fischinger S, Shin S, Boolay S, Walzl G, Jacobs A, Wilkinson RJ, Alter G, et al. (2020). An observational study identifying highly tuberculosis-exposed, HIV-1-positive but persistently TB, tuberculin and IGRA negative persons with M. tuberculosis specific antibodies in Cape Town, South Africa. EBioMedicine 61, 103053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Krug S, Parveen S, and Bishai WR (2021). Host-directed therapies: modulating inflammation to treat tuberculosis. Front. Immunol 12, 660916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Ladel CH, Blum C, Dreher A, Reifenberg K, and Kaufmann SH (1995). Protective role of gamma/delta T cells and alpha/beta T cells in tuberculosis. Eur. J. Immunol 25, 2877–2881. [DOI] [PubMed] [Google Scholar]
  96. Lanzavecchia A. (1990). Receptor-mediated antigen uptake and its effect on antigen presentation to class II-restricted T lymphocytes. Annu. Rev. Immunol 8, 773–793. [DOI] [PubMed] [Google Scholar]
  97. Laval T, Chaumont L, and Demangel C. (2021). Not too fat to fight: the emerging role of macrophage fatty acid metabolism in immunity to Mycobacterium tuberculosis. Immunol. Rev 301, 84–97. [DOI] [PubMed] [Google Scholar]
  98. Lavollay M, Arthur M, Fourgeaud M, Dubost L, Marie A, Veziris N, Blanot D, Gutmann L, and Mainardi JL (2008). The peptidoglycan of stationary-phase Mycobacterium tuberculosis predominantly contains cross-links generated by L,D-transpeptidation. J. Bacteriol 190, 4360–4366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Lawn SD, Myer L, Edwards D, Bekker LG, and Wood R. (2009). Short-term and long-term risk of tuberculosis associated with CD4 cell recovery during antiretroviral therapy in South Africa. AIDS 23, 1717–1725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Layton ED, Barman S, Wilburn DB, Yu KKQ, Smith MT, Altman JD, Scriba TJ, Tahiri N, Minnaard AJ, Roederer M, et al. (2021). T cells specific for a mycobacterial glycolipid expand after intravenous Bacillus Calmette-Guerin vaccination. J. Immunol 206, 1240–1250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Lepore M, Lewinsohn DA, and Lewinsohn DM (2021). T cell receptor diversity, specificity and promiscuity of functionally heterogeneous human MR1-restricted T cells. Mol. Immunol 130, 64–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Levin-Reisman I, Ronin I, Gefen O, Braniss I, Shoresh N, and Balaban NQ (2017). Antibiotic tolerance facilitates the evolution of resistance. Science 355, 826–830. [DOI] [PubMed] [Google Scholar]
  103. Lewinsohn DA, Heinzel AS, Gardner JM, Zhu L, Alderson MR, and Lewinsohn DM (2003). Mycobacterium tuberculosis-specific CD8+ T cells preferentially recognize heavily infected cells. Am. J. Respir. Crit. Care Med 168, 1346–1352. [DOI] [PubMed] [Google Scholar]
  104. Lewis K. (2010). Persister cells. Annu. Rev. Microbiol 64, 357–372. [DOI] [PubMed] [Google Scholar]
  105. Li H, Wang XX, Wang B, Fu L, Liu G, Lu Y, Cao M, Huang H, and Javid B. (2017). Latently and uninfected healthcare workers exposed to TB make protective antibodies against Mycobacterium tuberculosis. Proc. Natl. Acad. Sci. USA 114, 5023–5028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Lin PL, and Flynn JL (2015). CD8 T cells and Mycobacterium tuberculosis infection. Semin. Immunopathol 37, 239–249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Lin PL, Ford CB, Coleman MT, Myers AJ, Gawande R, Ioerger T, Sacchettini J, Fortune SM, and Flynn JL (2014). Sterilization of granulomas is common in active and latent tuberculosis despite within-host variability in bacterial killing. Nat. Med 20, 75–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Lin PL, Maiello P, Gideon HP, Coleman MT, Cadena AM, Rodgers MA, Gregg R, O’Malley M, Tomko J, Fillmore D, et al. (2016). PET CT identifies reactivation risk in cynomolgus macaques with latent M. tuberculosis. PLoS Pathog. 12. e1005739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Lin PL, Myers A, Smith L, Bigbee C, Bigbee M, Fuhrman C, Grieser H, Chiosea I, Voitenek NN, Capuano SV, et al. (2010). Tumor necrosis factor neutralization results in disseminated disease in acute and latent Mycobacterium tuberculosis infection with normal granuloma structure in a cynomolgus macaque model. Arthritis Rheum. 62, 340–350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Lin PL, Pawar S, Myers A, Pegu A, Fuhrman C, Reinhart TA, Capuano SV, Klein E, and Flynn JL (2006). Early events in Mycobacterium tuberculosis infection in cynomolgus macaques. Infect. Immun 74, 3790–3803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Lin PL, Rutledge T, Green AM, Bigbee M, Fuhrman C, Klein E, and Flynn JL (2012). CD4 T cell depletion exacerbates acute Mycobacterium tuberculosis while reactivation of latent infection is dependent on severity of tissue depletion in cynomolgus macaques. AIDS Res. Hum. Retroviruses 28, 1693–1702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Lindestam Arlehamn CS, Lewinsohn D, Sette A, and Lewinsohn D. (2014). Antigens for CD4 and CD8 T cells in tuberculosis. Cold Spring Harb. Perspect. Med 4, a018465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Liu Q, Wei J, Li Y, Wang M, Su J, Lu Y, López MG, Qian X, Zhu Z, Wang H, et al. (2020). Mycobacterium tuberculosis clinical isolates carry mutational signatures of host immune environments. Sci. Adv 6. eaba4901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Liu Y, Tan S, Huang L, Abramovitch RB, Rohde KH, Zimmerman MD, Chen C, Dartois V, VanderVen BC, and Russell DG (2016). Immune activation of the host cell induces drug tolerance in Mycobacterium tuberculosis both in vitro and in vivo. J. Exp. Med 213, 809–825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Lockhart E, Green AM, and Flynn JL (2006). IL-17 production is dominated by gammadelta T cells rather than CD4 T cells during Mycobacterium tuberculosis infection. J. Immunol 177, 4662–4669. [DOI] [PubMed] [Google Scholar]
  116. Lovey A, Verma S, Kaipilyawar V, Ribeiro-Rodrigues R, Husain S, Palaci M, Dietze R, Ma S, Morrison RD, Sherman DR, et al. (2022). Early alveolar macrophage response and IL-1R-dependent T cell priming determine transmissibility of Mycobacterium tuberculosis strains. Nat. Commun 13, 884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Loxton AG, and van Rensburg IC (2021). FASL regulatory B-cells during Mycobacterium tuberculosis infection and TB disease. J. Mol. Biol 433, 166984. [DOI] [PubMed] [Google Scholar]
  118. Lu LL, Chung AW, Rosebrock TR, Ghebremichael M, Yu WH, Grace PS, Schoen MK, Tafesse F, Martin C, Leung V, et al. (2016). A functional role for antibodies in tuberculosis. Cell 167, 433–443.e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Lu LL, Smith MT, Yu KKQ, Luedemann C, Suscovich TJ, Grace PS, Cain A, Yu WH, McKitrick TR, Lauffenburger D, et al. (2019). IFN-γ-independent immune markers of Mycobacterium tuberculosis exposure. Nat. Med 25, 977–987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Lu LL, Suscovich TJ, Fortune SM, and Alter G. (2018). Beyond binding: antibody effector functions in infectious diseases. Nat. Rev. Immunol 18, 46–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Lund FE (2008). Cytokine-producing B lymphocytes-key regulators of immunity. Curr. Opin. Immunol 20, 332–338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Lund FE, and Randall TD (2010). Effector and regulatory B cells: modulators of CD4+ T cell immunity. Nat. Rev. Immunol 10, 236–247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Maglione PJ, Xu J, Casadevall A, and Chan J. (2008). Fc gamma receptors regulate immune activation and susceptibility during Mycobacterium tuberculosis infection. J. Immunol 180, 3329–3338. [DOI] [PubMed] [Google Scholar]
  124. Maglione PJ, Xu J, and Chan J. (2007). B cells moderate inflammatory progression and enhance bacterial containment upon pulmonary challenge with Mycobacterium tuberculosis. J. Immunol 178, 7222–7234. [DOI] [PubMed] [Google Scholar]
  125. Manca C, Tsenova L, Bergtold A, Freeman S, Tovey M, Musser JM, Barry CE 3rd, Freedman VH, and Kaplan G. (2001). Virulence of a Mycobacterium tuberculosis clinical isolate in mice is determined by failure to induce Th1 type immunity and is associated with induction of IFN-alpha /beta. Proc. Natl. Acad. Sci. USA 98, 5752–5757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Manina G, Dhar N, and McKinney JD (2015). Stress and host immunity amplify Mycobacterium tuberculosis phenotypic heterogeneity and induce nongrowing metabolically active forms. Cell Host Microbe 17, 32–46. [DOI] [PubMed] [Google Scholar]
  127. Martinez FO, and Gordon S. (2014). The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep. 6, 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Mayer-Barber KD, Andrade BB, Barber DL, Hieny S, Feng CG, Caspar P, Oland S, Gordon S, and Sher A. (2011). Innate and adaptive interferons suppress IL-1alpha and IL-1beta production by distinct pulmonary myeloid subsets during Mycobacterium tuberculosis infection. Immunity 35, 1023–1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. McCaffrey EF, Donato M, Keren L, Chen Z, Delmastro A, Fitzpatrick MB, Gupta S, Greenwald NF, Baranski A, Graf W, et al. (2022). The immunoregulatory landscape of human tuberculosis granulomas. Nat. Immunol 2, 318–329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. McCune RM, Feldmann FM, Lambert HP, and McDermott W. (1966a). Microbial persistence. I. The capacity of tubercle bacilli to survive sterilization in mouse tissues. J. Exp. Med 123, 445–468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. McCune RM, Feldmann FM, and McDermott W. (1966b). Microbial persistence. II. Characteristics of the sterile state of tubercle bacilli. J. Exp. Med 123, 469–486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. McCune RM Jr., McDermott W, and Tompsett R. (1956). The fate of Mycobacterium tuberculosis in mouse tissues as determined by the microbial enumeration technique. II. The conversion of tuberculous infection to the latent state by the administration of pyrazinamide and a companion drug. J. Exp. Med 104, 763–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. McCune RM Jr., and Tompsett R. (1956). Fate of Mycobacterium tuberculosis in mouse tissues as determined by the microbial enumeration technique. I. The persistence of drug-susceptible tubercle bacilli in the tissues despite prolonged antimicrobial therapy. J. Exp. Med 104, 737–762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Millar JA, Butler JR, Evans S, Grant NL, Mattila JT, Linderman JJ, Flynn JL, and Kirschner DE (2021). Corrigendum: spatial organization and recruitment of non-specific T cells may limit T cell-macrophage interactions within Mycobacterium tuberculosis granulomas. Front. Immunol 12, 790557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Miller JS, Davis ZB, Helgeson E, Reilly C, Thorkelson A, Anderson J, Lima NS, Jorstad S, Hart GT, Lee JH, et al. (2022). Safety and virologic impact of the IL-15 superagonist N-803 in people living with HIV: a phase 1 trial. Nat. Med 28, 392–400. [DOI] [PubMed] [Google Scholar]
  136. Minhinnick A, Harris S, Wilkie M, Peter J, Stockdale L, Manjaly-Thomas ZR, Vermaak S, Satti I, Moss P, and McShane H. (2016). Optimization of a human bacille calmette-guerin challenge model: A tool to evaluate antimycobacterial immunity. J. Infect. Dis 213, 824–830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Mishra BB, Lovewell RR, Olive AJ, Zhang G, Wang W, Eugenin E, Smith CM, Phuah JY, Long JE, Dubuke ML, et al. (2017). Nitric oxide prevents a pathogen-permissive granulocytic inflammation during tuberculosis. Nat. Microbiol 2, 17072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Modlin RL, and Bloom BR (2013). TB or not TB: that is no longer the question. Sci. Transl. Med 5. 213sr6. [DOI] [PubMed] [Google Scholar]
  139. Moreira-Teixeira L, Tabone O, Graham CM, Singhania A, Stavropoulos E, Redford PS, Chakravarty P, Priestnall SL, Suarez-Bonnet A, Herbert E, et al. (2020). Mouse transcriptome reveals potential signatures of protection and pathogenesis in human tuberculosis. Nat. Immunol 21, 464–476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Mpande CAM, Steigler P, Lloyd T, Rozot V, Mosito B, Schreuder C, Reid TD, Bilek N, Ruhwald M, Andrews JR, et al. (2021). Mycobacterium tuberculosis-specific T cell functional, memory, and activation profiles in QuantiFERON-reverters are consistent with controlled infection. Front. Immunol 12, 712480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Murray PJ (2017). Macrophage polarization. Annu. Rev. Physiol 79, 541–566. [DOI] [PubMed] [Google Scholar]
  142. Myers AJ, Marino S, Kirschner DE, and Flynn JL (2013). Inoculation dose of Mycobacterium tuberculosis does not influence priming of T cell responses in lymph nodes. J. Immunol 190, 4707–4716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Nandi B, and Behar SM (2011). Regulation of neutrophils by interferon-γ limits lung inflammation during tuberculosis infection. J. Exp. Med 208, 2251–2262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Napier RJ, Adams EJ, Gold MC, and Lewinsohn DM (2015). The role of mucosal associated invariant T cells in antimicrobial immunity. Front. Immunol 6, 344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Navani N, Molyneaux PL, Breen RA, Connell DW, Jepson A, Nankivell M, Brown JM, Morris-Jones S, Ng B, Wickremasinghe M, et al. (2011). Utility of endobronchial ultrasound-guided transbronchial needle aspiration in patients with tuberculous intrathoracic lymphadenopathy: a multicentre study. Thorax 66, 889–893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Nemes E, Fiore-Gartland A, Boggiano C, Coccia M, D’Souza P, Gilbert P, Ginsberg A, Hyrien O, Laddy D, Makar K, et al. (2022). The quest for vaccine-induced immune correlates of protection against tuberculosis. Vaccin. Insights 1, 165–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Nemes E, Geldenhuys H, Rozot V, Rutkowski KT, Ratangee F, Bilek N, Mabwe S, Makhethe L, Erasmus M, Toefy A, et al. (2018). Prevention of M. tuberculosis infection with H4:IC31 vaccine or BCG revaccination. N. Engl. J. Med 379, 138–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Nikitina IY, Panteleev AV, Kosmiadi GA, Serdyuk YV, Nenasheva TA, Nikolaev AA, Gorelova LA, Radaeva TV, Kiseleva YY, Bozhenko VK, and Lyadova IV (2018). Th1, Th17, and Th1Th17 lymphocytes during tuberculosis: Th1 lymphocytes predominate and appear as low-differentiated CXCR3+CCR6+ Cells in the Blood and Highly Differentiated CXCR3+/− CCR6− Cells in the Lungs. J. Immunol 200, 2090–2103. [DOI] [PubMed] [Google Scholar]
  149. Noschka R, Wondany F, Kizilsavas G, Weil T, Weidinger G, Walther P, Michaelis J, and Stenger S. (2021). Gran1: A granulysin-derived peptide with potent activity against intracellular Mycobacterium tuberculosis. Int. J. Mol. Sci 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Novikov A, Cardone M, Thompson R, Shenderov K, Kirschman KD, Mayer-Barber KD, Myers TG, Rabin RL, Trinchieri G, Sher A, and Feng CG (2011). Mycobacterium tuberculosis triggers host type I IFN signaling to regulate IL-1beta production in human macrophages. J. Immunol 187, 2540–2547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Nyendak M, Swarbrick GM, Duncan A, Cansler M, Huff EW, Hokey D, Evans T, Barker L, Blatner G, Sadoff J, et al. (2016). Adenovirally-induced polyfunctional T cells do not necessarily recognize the infected target: lessons from a Phase I trial of the AERAS-402 vaccine. Sci. Rep 6, 36355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Ogongo P, Steyn AJ, Karim F, Dullabh KJ, Awala I, Madansein R, Leslie A, and Behar SM (2020). Differential skewing of donor-unrestricted and γdelta T cell repertoires in tuberculosis-infected human lungs. J. Clin. Invest 130, 214–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Ogongo P, Tezera LB, Ardain A, Nhamoyebonde S, Ramsuran D, Singh A, Ng’oepe A, Karim F, Naidoo T, Khan K, et al. (2021). Tissue-residentlike CD4+ T cells secreting IL-17 control Mycobacterium tuberculosis in the human lung. J. Clin. Invest 131, e142014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Parker DC (1993). T cell-dependent B cell activation. Annu. Rev. Immunol 11, 331–360. [DOI] [PubMed] [Google Scholar]
  155. Penn-Nicholson A, Mbandi SK, Thompson E, Mendelsohn SC, Suliman S, Chegou NN, Malherbe ST, Darboe F, Erasmus M, Hanekom WA, et al. (2020). RISK6, a 6-gene transcriptomic signature of TB disease risk, diagnosis and treatment response. Sci. Rep 10, 8629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Phuah J, Wong EA, Gideon HP, Maiello P, Coleman MT, Hendricks MR, Ruden R, Cirrincione LR, Chan J, Lin PL, and Flynn JL (2016). Effects of B cell depletion on early Mycobacterium tuberculosis infection in cynomolgus macaques. Infect. Immun 84, 1301–1311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Phuah JY, Mattila JT, Lin PL, and Flynn JL (2012). Activated B cells in the granulomas of nonhuman primates infected with Mycobacterium tuberculosis. Am. J. Pathol 181, 508–514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Pisu D, Huang L, Grenier JK, and Russell DG (2020). Dual RNA-seq of Mtb-infected macrophages in vivo reveals ontologically distinct host-pathogen interactions. Cell Rep. 30, 335–350.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Pisu D, Huang L, Narang V, Theriault M, Lê-Bury G, Lee B, Lakudzala AE, Mzinza DT, Mhango DV, Mitini-Nkhoma SC, et al. (2021). Single cell analysis of M. tuberculosis phenotype and macrophage lineages in the infected lung. J. Exp. Med 218. e2021061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Pitzalis C, Jones GW, Bombardieri M, and Jones SA (2014). Ectopic lymphoid-like structures in infection, cancer and autoimmunity. Nat. Rev. Immunol 14, 447–462. [DOI] [PubMed] [Google Scholar]
  161. Pollara G, Turner CT, Rosenheim J, Chandran A, Bell LCK, Khan A, Patel A, Peralta LF, Folino A, Akarca A, et al. (2021). Exaggerated IL-17A activity in human in vivo recall responses discriminates active tuberculosis from latent infection and cured disease. Sci. Transl. Med 13. eabg7673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Poulsen A. (1950). Some clinical features of tuberculosis. 1. Incubation period. Acta Tuberc. Scand 24, 311–346. [PubMed] [Google Scholar]
  163. Qaqish A, Huang D, Chen CY, Zhang Z, Wang R, Li S, Yang E, Lu Y, Larsen MH, Jacobs WR Jr., et al. (2017). Adoptive Transfer of Phosphoantigen-Specific γdelta T cell Subset Attenuates Mycobacterium tuberculosis Infection in Nonhuman Primates. J. Immunol 198, 4753–4763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Racine R, and Winslow GM (2009). IgM in microbial infections: taken for granted? Immunol. Lett 125, 79–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Reijneveld JF, Holzheimer M, Young DC, Lopez K, Suliman S, Jimenez J, Calderon R, Lecca L, Murray MB, Ishikawa E, et al. (2021). Synthetic mycobacterial diacyl trehaloses reveal differential recognition by human T cell receptors and the C-type lectin Mincle. Sci. Rep 11, 1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Reijneveld JF, Marino L, Cao TP, Cheng TY, Dam D, Shahine A, Witte MD, Filippov DV, Suliman S, van der Marel GA, et al. (2021b). Rational design of a hydrolysis-resistant mycobacterial phosphoglycolipid antigen presented by CD1c to T cells. J. Biol. Chem 297, 101197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Reiley WW, Calayag MD, Wittmer ST, Huntington JL, Pearl JE, Fountain JJ, Martino CA, Roberts AD, Cooper AM, Winslow GM, and Woodland DL (2008). ESAT-6-specific CD4 T cell responses to aerosol Mycobacterium tuberculosis infection are initiated in the mediastinal lymph nodes. Proc. Natl. Acad. Sci. USA 105, 10961–10966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Reynolds SP, Edwards JH, Jones KP, and Davies BH (1991). Immunoglobulin and antibody levels in bronchoalveolar lavage fluid from symptomatic and asymptomatic pigeon breeders. Clin. Exp. Immunol 86, 278–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Roca FJ, and Ramakrishnan L. (2013). TNF dually mediates resistance and susceptibility to mycobacteria via mitochondrial reactive oxygen species. Cell 153, 521–534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Roy Chowdhury R, Vallania F, Yang Q, Lopez Angel CJ, Darboe F, Penn-Nicholson A, Rozot V, Nemes E, Malherbe ST, Ronacher K, et al. (2018). A multi-cohort study of the immune factors associated with M. tuberculosis infection outcomes. Nature 560, 644–648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Sabatel C, Radermecker C, Fievez L, Paulissen G, Chakarov S, Fernandes C, Olivier S, Toussaint M, Pirottin D, Xiao X, et al. (2017). Exposure to bacterial CpG DNA protects from airway allergic inflammation by expanding regulatory lung interstitial macrophages. Immunity 46, 457–473. [DOI] [PubMed] [Google Scholar]
  172. Sakai S, Kauffman KD, Oh S, Nelson CE, Barry CE 3rd, and Barber DL (2021a). MAIT cell-directed therapy of Mycobacterium tuberculosis infection. Mucosal Immunol. 14, 199–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Sakai S, Kauffman KD, Sallin MA, Sharpe AH, Young HA, Ganusov VV, and Barber DL (2016). CD4 T cell-derived IFN-gamma plays a minimal role in control of pulmonary Mycobacterium tuberculosis infection and must be actively repressed by PD-1 to prevent lethal disease. PLoS Pathog. 12. e1005667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Sakai S, Lora NE, Kauffman KD, Dorosky DE, Oh S, Namasivayam S, Gomez F, Fleegle JD, Arlehamn CSL, et al. ; Tuberculosis Imaging Program (2021b). Functional inactivation of pulmonary MAIT cells following 5-OP-RU treatment of non-human primates. Mucosal Immunol. 14, 1055–1066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Saunders BM, Frank AA, Orme IM, and Cooper AM (2002). CD4 is required for the development of a protective granulomatous response to pulmonary tuberculosis. Cell. Immunol 216, 65–72. [DOI] [PubMed] [Google Scholar]
  176. Scanga CA, Mohan VP, Yu K, Joseph H, Tanaka K, Chan J, and Flynn JL (2000). Depletion of CD4(+) T cells causes reactivation of murine persistent tuberculosis despite continued expression of interferon gamma and nitric oxide synthase 2. J. Exp. Med 192, 347–358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Schaible UE, Collins HL, Priem F, and Kaufmann SH (2002). Correction of the iron overload defect in beta-2-microglobulin knockout mice by lactoferrin abolishes their increased susceptibility to tuberculosis. J. Exp. Med 196, 1507–1513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Schyns J, Bai Q, Ruscitti C, Radermecker C, De Schepper S, Chakarov S, Farnir F, Pirottin D, Ginhoux F, Boeckxstaens G, et al. (2019). Non-classical tissue monocytes and two functionally distinct populations of interstitial macrophages populate the mouse lung. Nat. Commun 10, 3964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Segueni N, Benmerzoug S, Rose S, Gauthier A, Bourigault ML, Reverchon F, Philippeau A, Erard F, Le Bert M, Bouscayrol H, et al. (2016a). Innate myeloid cell TNFR1 mediates first line defence against primary Mycobacterium tuberculosis infection. Sci. Rep 6, 22454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Segueni N, Tritto E, Bourigault ML, Rose S, Erard F, Le Bert M, Jacobs M, Di Padova F, Stiehl DP, Moulin P, et al. (2016b). Controlled Mycobacterium tuberculosis infection in mice under treatment with anti-IL-17A or IL-17F antibodies, in contrast to TNFalpha neutralization. Sci. Rep 6, 36923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Seiler P, Aichele P, Bandermann S, Hauser AE, Lu B, Gerard NP, Gerard C, Ehlers S, Mollenkopf HJ, and Kaufmann SH (2003). Early granuloma formation after aerosol Mycobacterium tuberculosis infection is regulated by neutrophils via CXCR3-signaling chemokines. Eur. J. Immunol 33, 2676–2686. [DOI] [PubMed] [Google Scholar]
  182. Serbina NV, Lazarevic V, and Flynn JL (2001). CD4(+) T cells are required for the development of cytotoxic CD8(+) T cells during Mycobacterium tuberculosis infection. J. Immunol 167, 6991–7000. [DOI] [PubMed] [Google Scholar]
  183. Shen H, Wang Y, Chen CY, Frencher J, Huang D, Yang E, Ryan-Payseur B, and Chen ZW (2015). Th17-related cytokines contribute to recalllike expansion/effector function of HMBPP-specific Vg2Vd22 T cells after Mycobacterium tuberculosis infection or vaccination. Eur. J. Immunol 45, 442–451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Shen L, Frencher J, Huang D, Wang W, Yang E, Chen CY, Zhang Z, Wang R, Qaqish A, Larsen MH, et al. (2019). Immunization of Vγ2Vδ2 T cells programs sustained effector memory responses that control tuberculosis in nonhuman primates. Proc. Natl. Acad. Sci. USA 116, 6371–6378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Silver RF, Zukowski L, Kotake S, Li Q, Pozuelo F, Krywiak A, and Larkin R. (2003). Recruitment of antigen-specific Th1-like responses to the human lung following bronchoscopic segmental challenge with purified protein derivative of Mycobacterium tuberculosis. Am. J. Respir. Cell Mol. Biol 29, 117–123. [DOI] [PubMed] [Google Scholar]
  186. Simmons JD, Stein CM, Seshadri C, Campo M, Alter G, Fortune S, Schurr E, Wallis RS, Churchyard G, Mayanja-Kizza H, et al. (2018). Immunological mechanisms of human resistance to persistent Mycobacterium tuberculosis infection. Nat. Rev. Immunol 18, 575–589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Slight SR, Rangel-Moreno J, Gopal R, Lin Y, Fallert Junecko BA, Mehra S, Selman M, Becerril-Villanueva E, Baquera-Heredia J, Pavon L, et al. (2013). CXCR5+ T helper cells mediate protective immunity against tuberculosis. J. Clin. Invest 123, 712–726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Smith FL, and Baumgarth N. (2019). B-1 cell responses to infections. Curr. Opin. Immunol 57, 23–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Srivastava S, Ernst JD, and Desvignes L. (2014). Beyond macrophages: the diversity of mononuclear cells in tuberculosis. Immunol. Rev 262, 179–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Stegelmann F, Bastian M, Swoboda K, Bhat R, Kiessler V, Krensky AM, Roellinghoff M, Modlin RL, and Stenger S. (2005). Coordinate expression of CC chemokine ligand 5, granulysin, and perforin in CD8+ T cells provides a host defense mechanism against Mycobacterium tuberculosis. J. Immunol 175, 7474–7483. [DOI] [PubMed] [Google Scholar]
  191. Stein CM, Nsereko M, Malone LL, Okware B, Kisingo H, Nalukwago S, Chervenak K, Mayanja-Kizza H, Hawn TR, and Boom WH (2019). Long-term stability of resistance to latent Mycobacterium tuberculosis infection in highly exposed tuberculosis household contacts in Kampala, Uganda. Clin. Infect. Dis 68, 1705–1712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Stenger S, Hanson DA, Teitelbaum R, Dewan P, Niazi KR, Froelich CJ, Ganz T, Thoma-Uszynski S, Melia A, Boγδan C, et al. (1998). An antimicrobial activity of cytolytic T cells mediated by granulysin. Science 282, 121–125. [DOI] [PubMed] [Google Scholar]
  193. Stutz MD, Allison CC, Ojaimi S, Preston SP, Doerflinger M, Arandjelovic P, Whitehead L, Bader SM, Batey D, Asselin-Labat ML, et al. (2021). Macrophage and neutrophil death programs differentially confer resistance to tuberculosis. Immunity 54, 1758–1771.e7. [DOI] [PubMed] [Google Scholar]
  194. Suliman S, Thompson EG, Sutherland J, Weiner J 3rd, Ota MOC, Shankar S, Penn-Nicholson A, Thiel B, Erasmus M, Maertzdorf J, et al. (2018). Four-gene Pan-African blood signature predicts progression to tuberculosis. Am. J. Respir. Crit. Care Med 197, 1198–1208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Swirski FK, Hilgendorf I, and Robbins CS (2014). From proliferation to proliferation: monocyte lineage comes full circle. Semin. Immunopathol 36, 137–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Tait DR, Hatherill M, Van Der Meeren O, Ginsberg AM, Van Brakel E, Salaun B, Scriba TJ, Akite EJ, Ayles HM, Bollaerts A, et al. (2019). Final analysis of a trial of M72/AS01E vaccine to prevent tuberculosis. N. Engl. J. Med 381, 2429–2439. [DOI] [PubMed] [Google Scholar]
  197. Tameris M, Geldenhuys H, Luabeya AK, Smit E, Hughes JE, Vermaak S, Hanekom WA, Hatherill M, Mahomed H, McShane H, and Scriba TJ (2014). The candidate TB vaccine, MVA85A, induces highly durable Th1 responses. PLoS ONE 9, e87340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Tameris MD, Hatherill M, Landry BS, Scriba TJ, Snowden MA, Lockhart S, Shea JE, McClain JB, Hussey GD, Hanekom WA, et al. (2013). Safety and efficacy of MVA85A, a new tuberculosis vaccine, in infants previously vaccinated with BCG: a randomised, placebo-controlled phase 2b trial. Lancet 381, 1021–1028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  199. Tan SY, and Krasnow MA (2016). Developmental origin of lung macrophage diversity. Development 143, 1318–1327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Tobin DM, Roca FJ, Oh SF, McFarland R, Vickery TW, Ray JP, Ko DC, Zou Y, Bang ND, Chau TT, et al. (2012). Host genotype-specific therapies can optimize the inflammatory response to mycobacterial infections. Cell 148, 434–446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. Torrelles JB, and Schlesinger LS (2017). Integrating lung physiology, immunology, and tuberculosis. Trends Microbiol. 25, 688–697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Tsai MC, Chakravarty S, Zhu G, Xu J, Tanaka K, Koch C, Tufariello J, Flynn J, and Chan J. (2006). Characterization of the tuberculous granuloma in murine and human lungs: cellular composition and relative tissue oxygen tension. Cell. Microbiol 8, 218–232. [DOI] [PubMed] [Google Scholar]
  203. Turner J, Frank AA, Brooks JV, Marietta PM, Vesosky B, and Orme IM (2001). Tuberculosis in aged gammadelta T cell gene disrupted mice. Exp. Gerontol 36, 245–254. [DOI] [PubMed] [Google Scholar]
  204. Ulrichs T, Kosmiadi GA, Trusov V, Jörg S, Pradl L, Titukhina M, Mishenko V, Gushina N, and Kaufmann SH (2004). Human tuberculous granulomas induce peripheral lymphoid follicle-like structures to orchestrate local host defence in the lung. J. Pathol 204, 217–228. [DOI] [PubMed] [Google Scholar]
  205. Ural BB, Yeung ST, Damani-Yokota P, Devlin JC, de Vries M, Vera-Licona P, Samji T, Sawai CM, Jang G, Perez OA, et al. (2020). Identification of a nerve-associated, lung-resident interstitial macrophage subset with distinct localization and immunoregulatory properties. Sci. Immunol 5. eaax8756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. van Pinxteren LA, Cassidy JP, Smedegaard BH, Agger EM, and Andersen P. (2000). Control of latent Mycobacterium tuberculosis infection is dependent on CD8 T cells. Eur. J. Immunol 30, 3689–3698. [DOI] [PubMed] [Google Scholar]
  207. Van Rhijn I, Kasmar A, de Jong A, Gras S, Bhati M, Doorenspleet ME, de Vries N, Godfrey DI, Altman JD, de Jager W, et al. (2013). A conserved human T cell population targets mycobacterial antigens presented by CD1b. Nat. Immunol 14, 706–713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Vascotto F, Le Roux D, Lankar D, Faure-Andre G, Vargas P, Guermonprez P, and Lennon-Dumé nil AM (2007). Antigen presentation by B lymphocytes: how receptor signaling directs membrane trafficking. Curr. Opin. Immunol 19, 93–98. [DOI] [PubMed] [Google Scholar]
  209. Victora GD, and Nussenzweig MC (2022). Germinal centers. Annu. Rev. Immunol 40, 413–442. [DOI] [PubMed] [Google Scholar]
  210. Walrath J, Zukowski L, Krywiak A, and Silver RF (2005). Resident Th1-like effector memory cells in pulmonary recall responses to Mycobacterium tuberculosis. Am. J. Respir. Cell Mol. Biol 33, 48–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Wang TT, and Ravetch JV (2019). Functional diversification of IgGs through Fc glycosylation. J. Clin. Invest 129, 3492–3498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Watson A, Li H, Ma B, Weiss R, Bendayan D, Abramovitz L, BenShalom N, Mor M, Pinko E, Bar Oz M, et al. (2021). Human antibodies targeting a Mycobacterium transporter protein mediate protection against tuberculosis. Nat. Commun 12, 602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Wells G, Glasgow JN, Nargan K, Lumamba K, Madansein R, Maharaj K, Hunter RL, Naidoo T, Coetzer L, le Roux S, et al. (2021). Microcomputed tomography analysis of the human tuberculous lung reveals remarkable heterogeneity in three-dimensional granuloma morphology. Am. J. Respir. Crit. Care Med 204, 583–595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. WHO (2022). Global Tuberculosis reports: 1997–2022. https://www.who.int/tb/publications/global_report/en/. [Google Scholar]
  215. Wiens KE, and Ernst JD (2016). The mechanism for Type I interferon induction by Mycobacterium tuberculosis is bacterial strain-dependent. PLoS Pathog. 12. e1005809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Wolf AJ, Linas B, Trevejo-Nuñez GJ, Kincaid E, Tamura T, Takatsu K, and Ernst JD (2007). Mycobacterium tuberculosis infects dendritic cells with high frequency and impairs their function in vivo. J. Immunol 179, 2509–2519. [DOI] [PubMed] [Google Scholar]
  217. Wong EA, Joslyn L, Grant NL, Klein E, Lin PL, Kirschner DE, and Flynn JL (2018). Low levels of T cell exhaustion in tuberculous lung granulomas. Infect. Immun 86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Wong EB, Gold MC, Meermeier EW, Xulu BZ, Khuzwayo S, Sullivan ZA, Mahyari E, Rogers Z, Kløverpris H, Sharma PK, et al. (2019). TRAV1–2+ CD8+ T-cells including oligoconal expansions of MAIT cells are enriched in the airways in human tuberculosis. Commun. Biol 2, 203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Woodworth JS, Wu Y, and Behar SM (2008). Mycobacterium tuberculosis-specific CD8+ T cells require perforin to kill target cells and provide protection in vivo. J. Immunol 181, 8595–8603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  220. Xia M, Hesser DC, De P, Sakala IG, Spencer CT, Kirkwood JS, Abate G, Chatterjee D, Dobos KM, and Hoft DF (2016). A subset of protective g9d2 T cells is activated by novel mycobacterial glycolipid components. Infect. Immun 84, 2449–2462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  221. Xie X, Shi Q, Wu P, Zhang X, Kambara H, Su J, Yu H, Park SY, Guo R, Ren Q, et al. (2020). Single-cell transcriptome profiling reveals neutrophil heterogeneity in homeostasis and infection. Nat. Immunol 21, 1119–1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  222. Yang JD, Mott D, Sutiwisesak R, Lu YJ, Raso F, Stowell B, Babunovic GH, Lee J, Carpenter SM, Way SS, et al. (2018). Mycobacterium tuberculosis-specific CD4+ and CD8+ T cells differ in their capacity to recognize infected macrophages. PLoS Pathog. 14. e1007060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  223. Zak DE, Penn-Nicholson A, Scriba TJ, Thompson E, Suliman S, Amon LM, Mahomed H, Erasmus M, Whatney W, Hussey GD, et al. (2016). A blood RNA signature for tuberculosis disease risk: a prospective cohort study. Lancet 387, 2312–2322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. Zhang B, Vogelzang A, Miyajima M, Sugiura Y, Wu Y, Chamoto K, Nakano R, Hatae R, Menzies RJ, Sonomura K, et al. (2021). B cell-derived GABA elicits IL-10+ macrophages to limit anti-tumour immunity. Nature 599, 471–476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  225. Zimmermann N, Thormann V, Hu B, Köhler AB, Imai-Matsushima A, Locht C, Arnett E, Schlesinger LS, Zoller T, Schürmann M, et al. (2016). Human isotype-dependent inhibitory antibody responses against Mycobacterium tuberculosis. EMBO Mol. Med 8, 1325–1339. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES