Journal List > J Bacteriol Virol > v.54(3) > 1516088419

Park, Kim, and Jo: Host Immune Pathways to Mycobacterium tuberculosis Infection

Abstract

In the post-COVID-19 era, tuberculosis caused by Mycobacterium tuberculosis (Mtb) remains of global concern given the high mortality rates of infectious diseases caused by single pathogens. This has created an urgent need to develop new therapeutic paradigms, i.e., host-directed therapeutics, which can be achieved using several strategies involving host-protective immune system components and pathways. These include the formation of protective granulomas by macrophages, which are the main guardian cells of the innate immune system; balanced regulation of the pro- and anti-inflammatory cytokine networks; and autophagy, which is a lysosome-dependent catabolic process that degrades intracytoplasmic cargos, including intracellular Mtb. In this review, we briefly discuss recent insights into the host innate immune mechanisms that control intracellular Mtb infections, including regulation of granuloma formation, and the roles played by crucial cytokines and autophagy in host–Mtb interactions. A deeper understanding of molecular strategies that might improve anti-Mtb responses would facilitate the development of novel and improved therapeutics based on knowledge of the tug-of-war between the host and the pathogen.

INTRODUCTION

Mycobacterium tuberculosis (Mtb), the causal pathogen of human tuberculosis (TB), remains a major killer among infectious diseases caused by single bacterial pathogens (1). Mtb can be transmitted via an aerosol and infects alveolar macrophages (AMs). Mtb is then disseminated to other myeloid cells, among which macrophages and neutrophils are known to eradicate Mtb (2). However, they also serve as niches in which Mtb may persist (2). Mtb employs several strategies to evade the host protective immune responses, thereby progressing to human TB. Mtb virulence factors include several cell surface components, enzymes, and transcriptional factors (3, 4). Among them, the ESAT-6 secretion system-1 (ESX-1), also known as the type VII secretion system, which is induced by region of difference (RD) 1, plays a critical role in TB pathogenesis (5, 6). Host cell–Mtb interactions are mediated by dynamic dialogs between Mtb and host cell components (7). The protective host response against TB is complex.
To date, several host immune and non-immune components involved in protection against intracellular Mtb have been reported. Among these host protective factors/pathways, we discuss the major immune strategies that together constitute the host defense against Mtb infection. These include TB granulomas; innate and adaptive immune responses; macrophages, neutrophils, and dendritic cells (DCs); and autophagy. Although these are all essential for protection against TB, changes in the regulation thereof may be detrimental to the host. Additionally, we discuss several cytokine biomarkers in the context of Mtb infection. It is crucial to understand the intricate controls that govern the expression of protective components that enhance host defense and prevent disease progression.

Overview of Mtb infection

Mtb, a facultative intracellular pathogen, is a rod-shaped bacillus that belongs to the slow-growing mycobacteria of the Mtb complex, which includes mycobacteria other than Mtb, and M. leprae (the leprosy pathogen) (8, 9). Although systemic Mtb dissemination is common, primary infection in adults involves lung tissue; Mtb interacts with the resident AMs (2, 10, 11). The Mtb has characteristic cell wall composition which hampers the success to treat TB with the actions of several antibiotics (12, 13). The mycobacterial cell wall architecture has been extensively studied since the 1960s (14, 15). The wall has a thick peptidoglycan-arabinogalactan complex, mycolic acid, capsular components including polysaccharides (e.g., α-glucan), lipids (e.g., PDIMs and PIMs), and proteins (e.g., GroEL2) (12, 16). Given the unusually lipid-rich cell wall, mycobacteria exhibit distinct staining characteristics and are known as acid-fast bacteria (17). These cell wall components of the outermost Mtb compartments interact with host cells to suppress or activate immune responses (16).
Stepwise Mtb adaptation and survival within host lungs, which mount major immune system-based attacks against infection, employ specific bacterial strategies using signal transduction systems, redox sensors, and secretion complexes (18, 19, 20). One such strategy, the two-component system (TCS), which is mostly exhibited by bacteria, is a phosphorelay signal transduction pathway that senses signals from histidine kinase and mounts an appropriate transcriptional response via a cognate response regulator (21, 22). Mtb harbors 12 pairs of TCSs that combat host immune system function after the establishment of an infection (22, 23). Each system produces proteins to construct a favorable granulomatous environment from the harsh conditions such as hypoxia, depleting energy sources, lowering the pH, and producing reactive oxygen species (ROS) or reactive nitrogen intermediates. Mtb thus persists for some time without growth and is not recognized by host immune sensors (8, 21, 24). For example, the DosS-DosR system, a well-known TCS of Mtb, plays a crucial role in Mtb survival under hypoxic conditions (25, 26). After O2 depletion, DosS senses the low O2 level and then activates DosR to upregulate the transcription of approximately 50 downstream genes, including hspX (Rv2031c), which encodes the chaperone alpha-crystallin and contributes to maintenance of the dormant state by stiffening its cell wall (26, 27). Recent studies have shown that the HspX protein may serve as a vaccine against TB (28, 29, 30).
Most recent attention has focused on drug-resistant TB, which imposes a major global burden through direct infection with drug-resistant Mtb or resistance acquired via Mtb genetic mutation triggered by adverse side-effects, or a failure to comply with the drug regimen during prolonged treatment (31, 32). In 2022, TB was recognized as the most common cause of airway drug-resistant epidemics in the past two centuries (1, 31, 33). Thus, there is an urgent need for the development of new and simple curative strategies, i.e., effective anti-mycobacterial drugs. Many reviews have highlighted the importance of new host-directed therapeutics for TB and the need to study the association between Mtb and the host immune system (7, 34, 35).
The host–Mtb interaction is characterized by granulomatous inflammation, which contains and localizes Mtb at the site of infection (36). A granuloma, first described by Rich in 1951, is the hallmark lesion of TB, walling off Mtb from the host environment (37, 38). A granulomatous lesion is an organized structure containing various immune cells and other types of cells, i.e., macrophages, lymphocytes, monocytes, neutrophils, and fibroblasts (39). Granuloma formation is initiated by the innate immune system, although CD4+ T lymphocytes play a major role in integration of the granulomatous structure (36, 39, 40). Although debate continues, the host granulomatous response to Mtb infection involves both protective and destructive granulomas (36). Protective granuloma formation is mediated by the cellular and humoral arms of the immune system to eradicate Mtb. Hypoxia within a granuloma stresses Mtb; cellular genes regulated by the hypoxia inducible factor 1 subunit alpha (HIF-1α) pathway are activated to clear intracellular Mtb, and HIF-1α serves as a marker of hypoxia within a granuloma. Previous studies reported correlations between TB progression and HIF-1α production in both TB patients and healthy controls (41). HIF-1α activates lactate dehydrogenase (LDH), which converts intracellular pyruvate from glycolysis into lactate (42). Earlier studies found increased LDH levels in the bronchoalveolar lavage fluid or sputum of patients with active TB (43, 44). Moreover, HIF-1α-depletion, which lacks HIF-1α-activated LDH-A, increased the level of intracellular pyruvate (compared to wild-type) in bone marrow-derived macrophages, thus facilitating Mtb growth using host-derived pyruvate as an energy source (45). However, if the host immune response is dysregulated, irreversible tissue damage can trigger necrosis, cavitation, and the spread of infected Mtb into lung parenchymal tissues (36, 46). Indeed, excessive pro-inflammatory responses may cause extensive tissue damage and the persistence of non-replicating bacilli. In this context, destruction of lung tissue extracellular matrix, principally by matrix metalloproteinases, triggers necrosis of TB lesions and unfavorable outcomes (46).

Innate immune responses

The front-line lung defenses include various cells in the immune system and barrier structures. Mtb primarily infects lung innate immune cells, exploiting them to create an environment that aids Mtb replication. Alveolar epithelial cells (AECs) are the first physical barrier that counters Mtb lung infection (47). Type 1 AECs are involved in gas exchange. Type 2 AECs exhibit immunomodulatory functions, secreting lactoferrin, defensins, cathelicidins, ROS, metalloproteinase, and surfactant proteins that enhance macrophagic phagocytosis and inhibit Mtb growth (48, 49, 50, 51). AECs recognize Mtb via bacterial pattern recognition receptors (PRRs) and then produce chemotactic cytokines, such as interferon (IFN)-γ, tumor necrosis factor (TNF)-α, and granulocyte–macrophage colony-stimulating factor (GM-CSF), which recruit immune cells. Mtb can invade AECs, creating a replication-conducive environment and thus crossing the mucosal barrier that initially inhibited dissemination.
Bacterial Toll-like receptor (TLR) signaling activates the nuclear factor kappa B (NF-κB) and MAPK pathways (50, 52). Mtb replication within AECs triggers cell necrosis; bacteria are released and infect surrounding cells (53). The endothelial cells (ECs) that line the vasculature facilitate immune cell transport during inflammation. ECs regulate metabolic homeostasis, vascular permeability, and hemodynamics (54). ECs recognize pathogens through PRRs on pathogen surfaces and then produce ROS, reactive nitrogen intermediates, and antimicrobial peptides (54). However, Mtb can enter ECs through Rab5 and Rab7, aiding bacterial dissemination through the bloodstream and lymphatic system (55, 56). The pathogen-associated molecular patterns of Mtb, such as phthiocerol dimycocerosates and mannosylated lipoarabinomannan (Man-LAM), promote recognition via EC mannose receptors and subsequent EC infection (57, 58). Vascular endothelial growth factor (VEGF), secreted by macrophages infected with Mtb expressing the RD1 virulence factor, promotes angiogenesis and facilitates bacterial spread (59). The serum VEGF levels of TB patients are elevated (60, 61, 62), and a VEGF-inhibitory therapeutic strategy has been used to normalize granuloma status and reduce bacterial dissemination (63, 64). Therefore, an understanding of the interaction between Mtb and the host is crucial to develop effective therapeutic strategies. This section focuses on the interactions between Mtb and innate immune cells, including macrophages, DCs, and neutrophils.
Macrophages
Macrophages play key roles in the immune response against Mtb via bacterial phagocytosis, initiation of inflammatory responses, and coordination with other immune cells. AMs and interstitial macrophages (IMs) play different roles during fetal lung development (65, 66). AMs maintain barrier integrity and regulate surfactant production, whereas IMs control the actions of T cells and DCs during infection (67). Mtb-infected macrophages undergo metabolic changes; AMs exhibit increased fatty acid oxidation, and IMs exhibit more glycolysis, affecting the ability of cells to control bacterial growth (68). Macrophages are classically activated by Th1 cytokines (M1 macrophages), which are pro-inflammatory and bactericidal; alternatively, they are activated by Th2 cytokines (M2 macrophages), which are anti-inflammatory and involved in tissue repair (69). Mtb effectors such as ESAT-6 drive M1-type polarization at an early stage but subsequently induce an M2-like anti-inflammatory phenotype in AMs to facilitate Mtb replication (70, 71). However, IMs exhibit a pro-inflammatory M1 phenotype, effectively restricting bacterial growth (71). Other Mtb proteins, such as RP105, Rv1987, PPE36, and EspC, influence macrophage activation and polarization via various intracellular pathways (72, 73, 74, 75).
Macrophages, major components of granulomas, play a critical role in Mtb death and contribute to immunopathology in a context-dependent manner. Granulomas contain Mtb infection but may also serve as niches within which bacteria can survive and replicate. Macrophages are involved in the initiation of granulomatous lesions and can develop into both epithelioid cells and multinucleated giant cells (76). Macrophages are recruited to sites of infection by the ESX-1/RD1 region of Mtb; they phagocytose the bacteria and then undergo apoptosis, which is associated with new macrophage recruitment and propagation of the infection (38). A computational model suggests that the macrophage polarization ratio predicts granuloma outcomes. M1 polarization early in infection and certain dynamics of the NF-κB response may provide a favorable outcome (77).
Macrophages produce a range of cytokines, including TNF-α, interleukin (IL)-1β, and IL-6, which recruit other immune cells and maintain the inflammatory environment required to control Mtb infection. Macrophages also interact with other immune cells to enhance antigen presentation and T cell activation, recruit neutrophils to sites of infection, and support B cell activation, thus bridging the gap between innate and adaptive immunity. An understanding of how macrophage polarization is regulated in infective microenvironments and the factors that control macrophage functions in granulomas is essential to determine the fate of granulomas and influence disease outcomes.
Macrophages employ several mechanisms to defend against Mtb, including autophagy, apoptosis, and pyroptosis (78). Autophagy aids elimination of intracellular pathogens, while apoptosis and pyroptosis are forms of cell death that also enhance Mtb removal by recruiting and activating new phagocytes (79, 80, 81). However, Mtb has evolved mechanisms that manipulate these processes to its own advantage; Mtb inhibits autophagy via upregulation of IL-10 and histone modification (82, 83). Mtb also employs various evasion strategies, including inhibition of phagosome maturation by the iron-sulfur cluster protein WhiB3 and regulation of non-coding RNA expression (78, 84). An understanding of these mechanisms will inform the development of new therapeutic strategies.
Neutrophils
Neutrophils, one of the most common immune cells in the airways of individuals with active TB, are also key players in the innate immune response against Mtb (85). Their primary function is phagocytosis; the cells engulf and attempt to kill Mtb using various agents such as ROS and antimicrobial peptides. Neutrophils secrete pro-inflammatory cytokines and chemokines, which recruit and activate other immune cells at a site of infection. Neutrophils can undergo apoptosis and be phagocytosed by macrophages, thus transferring their antimicrobial contents to the macrophages and aiding bacterial killing within the latter cells (86). Further, neutrophils interact with DCs to enhance antigen presentation and T cell activation, bridging innate and adaptive immunity. However, Mtb has evolved mechanisms that neutralize ROS, including the enhanced intracellular survival (eis) gene and a type I NADH dehydrogenase (NuoG) that inhibits ROS production and thus protects the bacteria (82, 87, 88). Additionally, Mtb induces neutrophil necrosis, such that Mtb survives and the infection spreads (89, 90). Mtb also resists neutrophil extracellular traps (NETs); these web-like structures are composed of DNA and antimicrobial proteins that immobilize and kill pathogens (91). Also, excessive neutrophil activity and NET production can trigger tissue damage and necrosis, impairing the ability to control Mtb infection (92, 93). Therefore, the development of therapies that target the evasive Mtb mechanisms might enhance the effectiveness of neutrophil-mediated Mtb killing.
DCs
DCs are essential antigen-presenting cells (APCs) that bridge the gap between the innate and adaptive immune systems. DCs process and present antigens to T cells via major histocompatibility complex (MHC) molecules, thus both initiating and modulating the adaptive immune response. DCs initially exist as immature cells below the lung epithelial layer, but when they encounter antigens they mature and migrate to lymph nodes to prime T cells. The Mtb NuoG protein delays DC migration and antigen uptake by inhibiting the apoptosis of infected neutrophils (87). Although DCs produce pro-inflammatory cytokines such as IL-12, which is essential for differentiation of T helper 1 (Th1) cells, the Man-LAM, ESX-1, and glycolipid Di-O-acyl trehalose of Mtb manipulate the protective function of DCs (5, 94, 95, 96). In addition, Mtb promotes Th17 cell expansion rather than expansion of IFN-γ-producing CD4+ T cells (97). DCs also contribute to the formation and maintenance of the granulomas that isolate Mtb and suppress Mtb growth. However, granulomas also “hide” Mtb, protecting the bacteria from host aggression (98, 99). Therefore, further studies on DCs and their interactions with other immune cells during Mtb infection might provide valuable insights into the pathogenesis of TB and suggest potential therapeutic strategies.

Adaptive immune response

The adaptive immune response, particularly the roles played by T cells and B cells, is crucial to control Mtb infection and prevent TB progression. T cells directly kill infected cells and maintain granulomas; B cells produce antibodies and assist in T cell activation. Appropriate interplay between innate and adaptive immunity is vital to mount a comprehensive defense against Mtb. Upon exposure to Mtb, humans and mice require approximately 4–6 and 2–3 weeks, respectively, to develop antigen-specific T cell responses in lymph nodes. These delays highlight the critical role played by the innate immune response in the early stages of infection.
CD4+ T cells, also known as helper T cells, are essential for orchestration of the immune response against Mtb. These cells are activated by APCs that present Mtb antigens via MHC-II molecules. Once activated, CD4+ T cells produce cytokines, such as IFN-γ and TNF-α, which activate macrophages and enhance their bactericidal activities (100, 101). Cytokines are crucial for the formation and maintenance of granulomas, which contain the infection. Granulomas are composed of infected macrophages surrounded by T cells; granuloma integrity is vital to prevent Mtb spread (102, 103). CD8+ T cells, also known as cytotoxic T lymphocytes (CTLs), recognize and kill Mtb-infected cells after the T cells are activated by Mtb antigens presented via MHC-I molecules. CD8+ T cells release perforin and granzymes, which induce the apoptosis of infected cells and thereby limit the bacterial load. Like CD4+ T cells, CD8+ T cells contribute to granuloma formation and maintenance by secreting cytokines and exerting cytotoxic effects on infected macrophages (104). Both CD4+ and CD8+ T cells are involved in granuloma formation (105). CD8+ T cells from the blood of TB patients were more active than those from healthy controls (106). Regulatory T cells (Tregs) play a dual role in TB; these cells modulate the immune response to prevent excessive inflammation, which can lead to tissue damage. However, Mtb can cause Tregs to suppress the immune response (107, 108, 109). An understanding of the balance between effector T cell and Treg actions is crucial to develop therapies that enhance protective immunity without causing excessive tissue damage.
B cells produce antibodies that neutralize and thus aid the clearance of Mtb. Antibodies also opsonize bacteria, enhancing Mtb recognition and phagocytosis by macrophages. B cells contribute to long-term immunity and serve as APCs that present antigens to T cells. Recent studies have reported an Mtb-specific IgM reaction and Mtb-specific B cell responses in macaques and ex vivo human assays (110, 111). A better understanding of these mechanisms will further improve the vaccines and other therapeutic strategies that aim to control TB.

Autophagy restricts intracellular Mtb infection

In response to various stresses, autophagy plays a critical role in maintaining homeostasis. This lysosome-dependent catabolic process sequesters and degrades cytosolic macromolecular components (112). In early studies, macroautophagy (hereinafter autophagy) was viewed as a form of nonspecific degradation of damaged organelles and large macromolecules. Autophagy involves several consecutive steps, including phagophore formation, elongation into autophagosomes, and fusion thereof with lysosomes (113). The autophagy pathway (including macro- and micro-autophagy) is mediated by several autophagy-related genes (ATGs). Each ATG plays a critical role; sometimes, several ATGs combine to activate various autophagic processes (114). Of the various ATGs, the mammalian Atg8s play essential roles in both autophagy induction and lysosomal biogenesis by activating the EB transcription factor (115). Although many studies have explored the different functions of the ATGs that activate autophagy, more work is needed to fully understand the autophagy-dependent and -independent roles of various ATGs during infection.
In this section, we briefly discuss attempts to activate autophagy for elimination of intracellular Mtb, the use of xenophagy to control Mtb infection, and noncanonical autophagy during mycobacterial infection. Finally, we discuss several ways in which Mtb evades host autophagy, although we do not deal with all Mtb components that modulate host autophagy.
Activation of autophagy to control intracellular Mtb
Many exogenous stimuli, small molecules, and other agents have been reported to activate autophagy (Fig. 1), presumably nonselectively (116). An early study by Deretic’s group showed that IFN-γ promoted autophagic clearance of intracellular Mtb (117). Several PRRs, including TLR4 and TLR7, activate autophagy and thus promote the innate host defense (118, 119, 120, 121). In human monocytes/macrophages, vitamin D3 strongly induces autophagy and thus enhances intracellular Mtb control by LL-37 (an antimicrobial peptide) (122). A previous study found that FDA-approved drugs, such as carbamazepine and valproic acid, induced autophagic clearance of intracellular Mtb from primary human macrophages (123). Importantly, carbamazepine upregulated antimicrobial autophagic activities even against multidrug-resistant Mtb (123). Several natural products induced autophagy that suppressed intracellular Mtb survival in host cells. Natural marine clionamines enhanced autophagy and suppressed intracellular Mtb survival by targeting Pik1 (124). Many drugs and other agents improved antimicrobial activities by inducing autophagy (116), suggesting potential utility of autophagy-based adjunctive host-directed therapeutics for TB.
Fig. 1

Autophagy protects against Mtb infection. Autophagy is a key means by which a host eliminates intracellular bacteria, including Mtb. After Mtb phagocytosis occurs via various routes, autophagy is activated. Activated autophagosomes fuse with lysosomes to degrade and clear Mtb. Macroautophagy can be triggered by several extrinsic signals including PRR activation; IFN-γ generation; and treatment with vitamin D3, several drugs, or natural products. Xenophagy and LAP (distinct autophagic pathways) contribute to intracellular Mtb elimination. IFN, interferon; LAP, LC3-associated phagocytosis; LAPosome, LC3-associated phagosome; Mtb, Mycobacterium tuberculosis; NOX2, NADPH oxygenase 2; ROS, reactive oxygen species; TLR, Toll-like receptor; VitD3, vitamin D3.

JBV_2024_v54n3_167_f001.tif
Targeting of xenophagy to combat Mtb infection
Recent studies have revealed selective forms of autophagy targeting different cargos and organelles; the forms include mitophagy, xenophagy, and reticulophagy (125). Xenophagy is a specialized form of autophagy that focuses on foreign organisms (126). Several autophagic receptors, including p62/SQSTM1, NDP52, and OPTN, are involved in target recognition, linkage to the autophagic machinery, and eradication of intracellular bacteria (127, 128, 129). In addition, galectin-8 senses phagosomal damage and then recruits TAX1BP1, a xenophagy receptor, to target Mtb and limit its intracellular replication (130).
Interestingly, the host protein ubiquilin-1 interacts with Mtb via ESAT-6/EsxA, thereby facilitating ubiquitin, p62, and microtubule-associated protein 1 light chain 3 (LC3) accumulation around Mtb and thus bacterial control (131). Additionally, the ubiquitin ligase TRIM32 enhances autophagic clearance of Mtb in macrophages by recruiting the autophagic proteins NDP52 and BECN1 (132). Chemical mimics of N-degron (Nt-Arg) activate xenophagy to enhance the host defense against multidrug-resistant Mtb and Salmonella both in vivo and in vitro (133). Although recent studies have highlighted the crucial role played by xenophagy activation in Mtb infection control, more work is required to determine whether these newly emerging drugs/agents could control drug-resistant TB.
LC3-associated phagocytosis during Mtb infection
As mentioned above, canonical autophagy is triggered by a variety of stress signals (such as nutrient deprivation) and is regulated by the induction of adenosine monophosphate (AMP)-activated kinase (AMPK) production (134, 135). LC3-associated phagocytosis (LAP) is a noncanonical autophagic pathway that differs in several ways from canonical autophagy. One unique characteristic of LAP is lipidation of LC3/Atg8 within single-membrane vesicles (136) via a process that requires RUBCN, a RUN-domain-containing protein that plays an inhibitory role during canonical autophagy (137). Another distinct feature of LAP is the production of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-2 (NOX2)-derived ROS during phagosome maturation (137). In addition, LAP operates independently of AMPK–mammalian target of rapamycin (mTOR) complex 1 (mTORC1) pathways (137).
Accumulating evidence suggests that, during various bacterial infections, LAP is activated upon recognition of PRRs and then contributes to the host defense against a variety of intracellular bacterial infections (135). In the Mtb context, CpsA, a LytR-CpsA-Psr domain-containing protein, aids Mtb evasion of LAP and thus reduces Mtb killing by NOX2 (138). However, our knowledge of noncanonical (compared to canonical) autophagy during Mtb infection is limited. As LAP is important in terms of immune regulation and dead cell removal in several pathogenic conditions (136), future studies are warranted to elucidate the Mtb LAP effectors and the molecular mechanisms by which LAP’s actions are orchestrated during a hyperinflammatory TB infection. Such efforts would aid the development of new therapeutic strategies that specifically modulate LAP during the different clinical stages of infection.
Mtb effectors of autophagic evasion
Mtb is a successful human pathogen that evades host autophagy/xenophagy using many different tactics (Table 1). For example, the eukaryotic-like serine/threonine protein kinase G (PknG; Rv0410c) of Mtb inhibits autophagosome maturation by interacting with RAB14, a small GTPase of the host, thereby promoting Mtb survival within host cells (139). Additionally, Mtb produces a terpenyl nucleoside, 1-tuberculosinyladenosine (1-TbAd; Rv3377c-Rv3378c), which blocks both autophagy and lysosomal maturation in association with lipid storage in foamy macrophages (140). The Mtb acyl carrier protein (Rv2244) suppresses the nuclear translocation of transcription factor of EB (TFEB) and phagolysosomal fusion within macrophages (141). The Mtb effector PPE51 (Rv3136) suppresses the TLR2 signaling response, particularly that of extracellular signal-regulated kinase 1/2 (ERK1/2), and inhibits autophagic activation (142). A previous study showed that PE_PGRS20 (Rv1068c) and PE_PGRS47 (Rv2741) blocked the initiation of canonical autophagy by interacting with the Ras-related protein Rab1A (143). Although we have not cited all research in this field, further studies are required to identify more Mtb effectors and the molecular mechanisms by which host canonical and noncanonical autophagy are finely regulated. These efforts will uncover the mechanisms by which Mtb survives in host cells during infection, and may suggest new therapeutic strategies.
Table 1.

Mtb effectors aiding autophagic evasion

Effectors Evasion strategy Outcome Ref.
PknG Interaction with RAB14 Inhibition of autophagosome maturation (139)
1-TbAd Inhibition of autophagy and lysosomal maturation Lipid storage in foamy macrophages (140)
AcpM Inhibition of TFEBa nuclear translocation Suppression of phagolysosomal fusion (141)
PPE51 Inhibition of the TLRb signaling response Inhibition of autophagic activation (142)
PE_PGRS20
PE_PGRS47
Interaction with Rab1A Blockage of canonical autophagy initiation (143)

aTFEB, transcription factor EB; bTLR, Toll-like receptor.

Cytokine networks: Host protection, pathologies, and biomarkers

During Mtb infection, both macrophages and neutrophils, two sentinels of innate immunity, play critical roles in mounting a host defense via activation of cytokine and chemokine production and the oxidative burst (144). Although early activation of pro-inflammatory mediators is critical to combat intracellular Mtb in host cells, this also induces tissue damage (144). One review suggested that the key immune markers of different stages of TB infection included various cytokines, such as IL-1β, IL-6, IL-8, IL-10, and IL-12, as well as matrix metalloproteinases and inhibitors thereof (145). A recent study on the plasma biomarkers of TB patients revealed that distinct inflammatory cytokine profiles, including elevated BAFF, LIGHT, sTNF-R1 and sTNF-R2, and IP-10 levels, were associated with the clinical severity of TB (146). A systematic review found that the actions the cytokines IFN-γ, IL-2, and TNF-α had been most frequently investigated, although further well-designed studies are warranted (147). Therefore, many cytokines/chemokines may play critical functions in infective outcomes and might also serve as biomarkers of different infective TB stages, as well as the positive outcomes of adequate pharmacotherapy.
In this section, we discuss four major innate immune cytokines that have been widely studied in terms of their actions in human TB (Fig. 2).
Fig. 2

Schematic showing the roles played by inflammatory cytokines during Mtb infection. Cytokines are crucial in host cell protection against Mtb infection. TNF-α upregulation recruits immune cells and stimulates granuloma formation. Mtb activates the NLRP3 inflammasome via ESAT6, followed by maturation of caspase-1, IL-1β, and IL-18. Colchicine-induced IL-1β production triggers Cox2/PGE2 signaling, suppressing Mtb growth. The Mtb protein UreC inhibits host DNA repair and thus activates cGAS-STING signaling. The level of IFN-β then increases, upregulating LD formation and facilitating Mtb replication. TREM2 blocks TNF-α, IL-1β, and ROS generation and promotes IFN-β and IL-10 synthesis. cGAS-STING, cyclic GMP-AMP synthase/stimulator of interferon genes; Cox2, cyclooxygenase 2; IFN, interferon; LD, lipid droplet; Mtb, Mycobacterium tuberculosis; PGE2, prostaglandin 2; PRR, pattern recognition receptor; TREM2, triggering receptor expressed on myeloid cells 2.

JBV_2024_v54n3_167_f002.tif
TNF-α
TNF-α is one of the most important cytokines in terms of TB progression. TNF-α is involved in M1 macrophage activation, apoptosis induction and regulation, and immune cell recruitment (148, 149). Also, TNF-α plays a critical role in host protection via granuloma formation and immune cell recruitment (148, 150). Anti-TNF therapy for patients with autoimmune diseases often predisposes them to an increased risk of TB re-activation (151). Such findings strongly suggest that TNF-α aids the host defense against TB. However, excessive TNF-α production leads to pathological responses, including cachexia, which is a major adverse event in humans with TB (148). Therefore, appropriate TNF-α production is required to maintain the host immune defense, without any toxic effects, during Mtb infection.
In patients with both pulmonary and intracerebral Mtb infections, TNF-α is critical in terms of resistance (150, 152). Infants are more susceptible to TB infection than older cases because several important cytokines, including TNF-α and IL-1, are produced only at low levels, and precursor cell differentiation into IFN-γ-producing T cells (153) is thus minimal. More studies are needed to understand the multifaceted roles played by TNF-α at different stages of various mycobacterial infections.
IL-1
Secretion of IL-1 (including IL-1β and IL-18) is modulated by inflammasome activation of innate immune cells. The inflammasome complex is a protein assembly with a sensor, an adaptor, and a caspase that is triggered by inflammatory signals. This multimeric protein platform activating caspase-1 is also responsible for the maturation of IL-1β and IL-18 (154). Of the many inflammasome complexes, the NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome has been the most widely studied.
Previous studies examined Mtb-manipulated, IL-1-derived pathogenic responses. The Mtb protein ESAT-6 triggered NLRP3 inflammasome activation in different cell types, leading to caspase-1 maturation and IL-1β release (155, 156). An Mtb ESAT-6 mutation was associated with no increase in the IL-1β or IL-18 level (157). Prolonged elevation of IL-1 levels during Mtb infection can damage tissue (158, 159). By modulating the IL-1β concentration, Mtb ensures its own survival and increases disease development associated with necrotic cell death (160, 161). The vaccine strain M. bovis Bacillus Calmette–Guérin (BCG) does not activate NLRP3 inflammasome assembly. Gain-of-function genetic changes in the NLRP3 gene of human macrophages increase Mtb growth restriction in such macrophages (162). Based on the pathogenic role played by IL-1 in TB, novel preventative strategies have been developed. TB anti-IL-1 therapy (IL-1 neutralizing antibodies) controlled inflammation by mitigating IL-1 signaling (158, 163, 164). However, such immunosuppressive treatment enhances the susceptibility of TB patients; a therapy that promotes IL-1 pathway activity without inducing a cytokine storm may be needed to avoid TB progression (165, 166). A recent study revealed that IL-1 blockade alleviated side effects associated with anti-TB linezolid treatment (159).
An early study showed that IL-1β production was required to protect against Mtb infection, regardless of the caspase-1 level (167). Mtb infection of mice lacking the Il1a, Il1b, or even genes of IL-1 receptor type 1 (IL-1R1) was associated with higher Mtb loads and severe lung pathology (168, 169, 170). Blockade of the adaptor myeloid differentiation factor 88 was also associated with high mortality after Mtb infection, reflecting dysfunctional IL-1R signaling (171). Furthermore, IL-1 plays a critical role in host resistance against Mtb infection by inducing eicosanoids via suppression of type I IFN synthesis (172). Another recent study showed that colchicine, a widely used inhibitor of NLRP3 inflammasome activation, rather induced Nlrp3 and Il1b gene expression. The secreted IL-1β increased IL-1R1 expression, and that of cyclooxygenase-2 (Cox2) and the lipid immune mediator prostaglandin E, effects associated with anti-Mtb actions (173). Additionally, Mtb promoted NLRP3-independent IL-1β release in lung tissues (174). Together, these data suggest that fine control of IL-1 signaling and inflammasome activation may aid the development of host-specific therapeutics.
IL-6
IL-6 is a pleiotropic cytokine that plays diverse roles in T and B cell regulation, and pro- and anti-inflammatory responses, in a context-dependent manner (175, 176). Thus, IL-6 is critical for the induction of chronic inflammation and the host defense against a variety of infections (176, 177). Recent studies have suggested that IL-6 may serve as a biomarker of the various infectious stages of human TB. Importantly, patient studies indicated that the pre-treatment IL-6 level may be a biomarker of unfavorable outcomes and disease progression during TB treatment (178, 179). An increased plasma IL-6 level was associated with a higher risk of lung cavitation in patients with drug-resistant TB (180).
Moreover, IL-6 production caused by the Mtb protein Rv0183 (a lysophospholipase) was higher in the whole blood of patients with active pulmonary TB than controls, suggesting that the protein might serve as an immunodiagnostic biomarker (181). A recent study showed that both IL-6 and IL-27 were potentially useful biomarkers of human pleural TB infections (182). In patients with tuberculous meningitis, the IL-6 level appears to be upregulated, and it may thus be a valuable biomarker (183).
However, IL-6 is a double-edged sword. Mtb inhibits host regulation of IL-6 production to suppress the activation of type I IFN pathways, thus exacerbating the disease (184). IL-6 suppressed the autophagic pathway activity promoted by IFN-γ by counteracting the protective effect of IFN-γ in Mtb-infected macrophages, thus increasing Mtb survival (185). Moreover, Mtb uses the ESAT-6 protein to upregulate IL-6 production, in turn activating the signal transducere and activator of transcription 3 (STAT3) protein; the action of this transcription factor is enhanced by multiple cytokines, including IL-6, in both mouse and human macrophage Mtb infections (186, 187).
IL-6 plays important protective roles against Mtb infection. Early studies showed that Mtb infection of mice with a genetic IL-6 deficiency increased the bacterial loads (compared to those of wild-type mice) by changing the responses of type I T helper cells (188, 189, 190). Additionally, IL-6 induced and maintained a well-controlled Th17 immune response, which has been suggested to be crucial to protect against TB (191). Moreover, IL-6 was required to prime IFN-γ-secreting T cells during TB vaccination of Il6-knockout mice or mice in which IL-6 was specifically neutralized; such mice were otherwise not well-protected by vaccination (192). IL-6 is also associated with differentiation of CXCR5+ T follicular helper cells; IL-6 mediates appropriate polarization of T follicular helper cells to macrophages containing Mtb, which optimizes the anti-mycobacterial response (193). Future studies should determine whether IL-6 bioactivity might serve as a clinically predictive biomarker in various TB infection settings.
Type I IFNs: IFN-α and IFN-β
Excessive production of type I IFN during TB infection potentiates disease pathogenesis and exacerbation (172). An important question concerns how type I IFN is produced during infection. A recent study showed that Mtb urease C (UreC, Rv1850) suppressed the host DNA repair pathway and thus activated the cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS)/stimulator of IFN genes (STING) pathway. The resultant activation of IFN-β production facilitated lipid droplet (LD) formation, thus contributing to Mtb replication (194). Additionally, transcriptome analysis of M. marinum-infected animal models showed that type I IFN-associated pathways were enriched in an inoculum-dose-dependent manner and became activated at high multiplicities of infection (195). Mtb carrying a rifampicin drug-resistance-conferring single-nucleotide polymorphism (SNP) in the RNA polymerase gene H445Y SNP inhibited lung myeloid and lymphoid immune responses via type I IFN-dependent signaling pathways to a greater extent than did wild-type Mtb (196). These data suggest that the bacterium itself, and its heterogeneity, are strongly associated with type I IFN production.
Several host factors activate type I IFN production during Mtb infection. Triggering receptor expressed on myeloid cells 2 (TREM2), a transmembrane surface receptor, suppressed TNF-α, IL-1β, and ROS production but increased the production of IFN-β and IL-10. Mtb binds to TREM2, rendering the intracellular conditions more favorable for bacterial survival via continuation of type I IFN signaling, thereby suppressing macrophage antimicrobial responses (197). Another recent study showed that IRGM1 deficiency increased IRGM3-dependent type I IFN signaling, suppressing Mtb-specific T cell expansion. Irgm1-deficient mice cannot control Mtb infections of neutrophils and AMs (198). Another recent study revealed that type I IFN, produced by plasmacytoid DCs derived via the formation of NETs, is released when control of Mtb replication fails and thus contributes to TB pathogenesis (199). These studies strongly suggest that type I IFNs aid bacterial survival and replication in lung tissues by inhibiting appropriate (protective) T cell helper 1 responses during infection.
Certain protective roles played by type I IFN during Mtb infection were apparent under specific conditions (200). Case reports showed that administration of both IFN-α and an antimycobacterial drug combination exhibited protective effects in patients with mycobacterial infections who responded poorly to the conventional drug regimen or suffered disease recurrence (201). Type I IFN exerted beneficial effects in the absence of IFN-γ signaling both in vivo and in vitro. Young patients struggling to control mycobacterial infections because of a lack of IFN-γ pathway activity improved after combined administration of IFN-α and antimycobacterial chemotherapy (202). In Mtb-infected mouse models, type I IFN is required for blockade of the early pulmonary pathology and mortality in the absence of IFN-γ receptors. Moreover, type I IFN enhanced BCG vaccine efficacy against Mtb infection by increasing the production of protective cytokines including TNF-α, IL-12, and IFN-γ (203, 204). Another study found that a basal level of type I IFN signaling was required for optimal generation of TNF-α and IL-12 by macrophages and DCs that counter Mtb infection (205, 206).
Further studies are required to identify the key cytokines during TB infection and where they function within the innate immune regulatory network. Do they protect the host, and could they serve as diagnostic and/or predictive markers?

CONCLUSION

Balanced regulation of the host innate immune and inflammatory responses is critical to protect the host against TB. A granuloma is the hallmark of TB. Protective granuloma formation is critical to ensure immunometabolic limitation of bacterial proliferation at infection sites. Dysregulation of granuloma formation leads to Mtb spread. Future studies should explore how protective granulomas are maintained and the factors involved in TB persistence and progression.
It is clear that the proinflammatory cytokine TNF-α plays critical roles in granuloma formation and the development of protective immunity against TB infection. However, excessive TNF-α production triggers pathological inflammation and cell death. IL-1 is another protective cytokine produced via inflammasome activation. IL-6 is a pleiotropic cytokine that has recently served as a diagnostic and predictive marker in various stages of TB infection. Finally, the actions of pathological type I IFNs during TB infections have been widely studied. It may be that no single cytokine or chemokine can serve as a biomarker of disease progression or protection. Future studies should explore the cytokine networks that finely tune the protective and pathological immune responses.
Both canonical and noncanonical autophagy are now being recognized as crucial immune system contributors to the progression/control of mycobacterial infections. However, further preclinical and clinical studies are required to develop different autophagy-based adjunctive therapeutics for patients with drug-resistant infections and those in different stages of TB infection, and more mechanistic studies are needed to elucidate the Mtb effector–host interactions that regulate autophagy and other defensive pathways. More extensive investigations of host–pathogen interactions are required to develop more efficacious adjunctive treatment modalities. We must move to a host-directed therapeutic approach.

ABBREVIATIONS

AM, alveolar macrophage; AEC, alveolar epithelial cell; AMP, adenosine monophosphate; AMPK, adenosine monophosphate kinase; APC, antigen presenting cell; ATG, Autophagy-related gene; BCG, bacillus Calmette- Guérin; cGAS/STING, cyclic GMP-AMP synthase/stimulator of interferon genes; Cox2, cyclooxygenase-2; CTL, cytotoxic T lymphocyte; DC, dendritic cell; EC, epithelial cell; ERK1/2, extracellular signal-regulated kinase 1/2; ESX-1, ESAT-6 secretion system; GM-CSF, granulocyte-marophage colony stimulating factor; GMP, guanosine monophosphate; HIF, hypoxia-inducible factor; IFN, interferon; IL, interleukin; IL-1R1, interleukin 1 receptor, type I; IM, interstitial macrophage; LAP, LC3-associated phagocytosis; LC3, microtubule-associated protein 1 light chain 3; LD, lipid droplet; LDH, lactate dehydrogenase; Man-LAM, mannosylated lipoarabinomannan; MHC, major histocompatibility complex; Mtb, Mycobacterium tuberculosis; mTOR, mammalian target of rapamycin; mTORC1, mammalian target of rapamycin complex 1; NADPH, nicotinamide adenine dinucleotide phosphate; NET, neutrophil extracellular trap; NF-κB, nuclear factor kappa B; NLRP3, NOD-, LRR-, and pyrin domain-containing protein 3; NOX2, NADPH oxidase-2; PRR, pattern recognition receptor; RD, region of difference; ROS, reactive oxygen species; SNP, small nucleotide polymorphism; STAT3, signal transducer and activator of transcription 3; TB, tuberculosis; 1-TbAd. 1-tuberculosinyladonosine; TCS, two-component system; TFEB, transcription factor EB; Th1, T helper 11; TLR, Toll-like receptor; TNF, tumor necrosis factor; Treg, regulatory T cell; TREM2, triggering receptor expressed on myeloid cells 2; VEGF, vascular endothelial growing factor.

ACKNOWLEDGMENTS

This work was supported by research fund of Chungnam National University.

References

1. World Health Organization. Global tuberculosis report 2023. Geneva. 2023.
2. Chandra P, Grigsby SJ, Philips JA. Immune evasion and provocation by Mycobacterium tuberculosis. Nat Rev Microbiol. 2022;20(12):750-766.DOI: 10.1038/s41579-022-00763-4.
3. Passos BBS, Araujo-Pereira M, Vinhaes CL, Amaral EP, Andrade BB. The role of ESAT-6 in tuberculosis immunopathology. Front Immunol. 2024;15:1383098.DOI: 10.3389/fimmu.2024.1383098.
4. Ramon-Luing LA, Palacios Y, Ruiz A, Tellez-Navarrete NA, Chavez-Galan L. Virulence factors of Mycobacterium tuberculosis as modulators of cell death mechanisms. Pathogens. 2023;12(6):839.DOI: 10.3390/pathogens12060839.
5. Romagnoli A, Etna MP, Giacomini E, Pardini M, Remoli ME, Corazzari M, et al. ESX-1 dependent impairment of autophagic flux by Mycobacterium tuberculosis in human dendritic cells. Autophagy. 2012;8(9):1357-1370.DOI: 10.4161/auto.20881.
6. Tiwari S, Casey R, Goulding CW, Hingley-Wilson S, Jacobs WR Jr. Infect and inject: How Mycobacterium tuberculosis exploits its major virulence-associated Type VII secretion system, ESX-1. Microbiol Spectr. 2019;7(3).DOI: 10.1128/microbiolspec.BAI-0024-2019.
7. Saini S, Gangwar A, Sharma R. Harnessing host-pathogen interactions for innovative drug discovery and host-directed therapeutics to tackle tuberculosis. Microbiol Res. 2023;275:127466.DOI: 10.1016/j.micres.2023.127466.
8. Orgeur M, Sous C, Madacki J, Brosch R. Evolution and emergence of Mycobacterium tuberculosis. FEMS Microbiol Rev. 2024;48(2):fuae006.DOI: 10.1093/femsre/fuae006.
9. Franco-Paredes C, Marcos LA, Henao-Martinez AF, Rodriguez-Morales AJ, Villamil-Gomez WE, Gotuzzo E, et al. Cutaneous mycobacterial infections. Clin Microbiol Rev. 2018;32(1):e00069-18.DOI: 10.1128/CMR.00069-18.
10. Moule MG, Cirillo JD. Mycobacterium tuberculosis dissemination plays a critical role in pathogenesis. Front Cell Infect Microbiol. 2020;10:65.DOI: 10.3389/fcimb.2020.00065.
11. Zumla A, Chakaya J, Centis R, D'Ambrosio L, Mwaba P, Bates M, et al. Tuberculosis treatment and management--an update on treatment regimens, trials, new drugs, and adjunct therapies. Lancet Respir Med. 2015;3(3):220-234.DOI: 10.1016/S2213-2600(15)00063-6.
12. Dulberger CL, Rubin EJ, Boutte CC. The mycobacterial cell envelope - a moving target. Nat Rev Microbiol. 2020;18(1):47-59.DOI: 10.1038/s41579-019-0273-7.
13. Singh R, Dwivedi SP, Gaharwar US, Meena R, Rajamani P, Prasad T. Recent updates on drug resistance in Mycobacterium tuberculosis. J Appl Microbiol. 2020;128:1547-1567.DOI: 10.1111/jam.14478.
14. Imaeda T, Kanetsuna F, Galindo B. Ultrastructure of cell walls of genus Mycobacterium. J Ultrastruct Res. 1968;25(1):46-63.DOI: 10.1016/S0022-5320(68)80059-0.
15. Winder FG, Collins PB. Inhibition by isoniazid of synthesis of mycolic acids in Mycobacterium tuberculosis. J Gen Microbiol. 1970;63(1):41-48.DOI: 10.1099/00221287-63-1-41.
16. Abrahams KA, Besra GS. Synthesis and recycling of the mycobacterial cell envelope. Curr Opin Microbiol. 2021;60:58-65.DOI: 10.1016/j.mib.2021.01.012.
17. Francis RJ, Robb G, McCann L, Khatri B, Keeble J, Dagg B, et al. Three-dimensional in situ morphometrics of Mycobacterium tuberculosis infection within lesions by optical mesoscopy and novel acid-fast staining. Sci Rep. 2020;10(1):21774.DOI: 10.1038/s41598-020-78640-4.
18. Frando A, Boradia V, Grundner C. Regulatory intersection of two-component system and Ser/Thr protein kinase signaling in Mycobacterium tuberculosis. J Mol Biol. 2024;436(2):168379.DOI: 10.1016/j.jmb.2023.168379.
19. Oh Y, Lee HN, Ko EM, Jeong JA, Park SW, Oh JI. Mycobacterial regulatory systems involved in the regulation of gene expression under respiration-inhibitory conditions. J Microbiol. 2023;61(3):297-315.DOI: 10.1007/s12275-023-00026-8.
20. Roy S, Ghatak D, Das P, BoseDasgupta S. ESX secretion system: The gatekeepers of mycobacterial survivability and pathogenesis. Eur J Microbiol Immunol (Bp). 2020;10(4):202-209.DOI: 10.1556/1886.2020.00028.
21. Stupar M, Furness J, De Voss CJ, Tan L, West NP. Two-component sensor histidine kinases of Mycobacterium tuberculosis: Beacons for niche navigation. Mol Microbiol. 2022;117(5):973-985.DOI: 10.1111/mmi.14899.
22. Frando A, Grundner C. More than two components: complexities in bacterial phosphosignaling. mSystems. 2024;9(5):e0028924.DOI: 10.1128/msystems.00289-24.
23. Luo G, Ming T, Yang L, He L, Tao T, Wang Y. Modulators targeting protein-protein interactions in Mycobacterium tuberculosis. Microbiol Res. 2024;284:127675.DOI: 10.1016/j.micres.2024.127675.
24. Verma A, Ghoshal A, Dwivedi VP, Bhaskar A. Tuberculosis: The success tale of less explored dormant Mycobacterium tuberculosis. Front Cell Infect Microbiol. 2022;12:1079569.DOI: 10.3389/fcimb.2022.1079569.
25. Kundu M, Basu J. Applications of transcriptomics and proteomics for understanding dormancy and resuscitation in Mycobacterium tuberculosis. Front Microbiol. 2021;12:642487.DOI: 10.3389/fmicb.2021.642487.
26. Kim MJ, Park KJ, Ko IJ, Kim YM, Oh JI. Different roles of DosS and DosT in the hypoxic adaptation of Mycobacteria. J Bacteriol. 2010;192(19):4868-4875.DOI: 10.1128/JB.00550-10.
27. Yuan Y, Crane DD, Simpson RM, Zhu YQ, Hickey MJ, Sherman DR, et al. The 16-kDa alpha-crystallin (Acr) protein of Mycobacterium tuberculosis is required for growth in macrophages. Proc Natl Acad Sci U S A. 1998;95(16):9578-9583.DOI: 10.1073/pnas.95.16.9578.
28. Karbalaei M, Mosavat A, Soleimanpour S, Farsiani H, Ghazvini K, Amini AA, et al. Production and evaluation of Ag85B:HspX:hFcgamma1 immunogenicity as an Fc fusion recombinant multi-stage vaccine candidate against Mycobacterium tuberculosis. Curr Microbiol. 2024;81(5):127.DOI: 10.1007/s00284-024-03655-3.
29. Mansury D, Ghazvini K, Amel Jamehdar S, Badiee A, Tafaghodi M, Nikpoor AR, et al. Increasing cellular immune response in liposomal formulations of DOTAP encapsulated by fusion protein Hspx, PPE44, and EsxV, as a potential tuberculosis vaccine candidate. Rep Biochem Mol Biol. 2019;7(2):156-166.
30. Liu W, Li J, Niu H, Lin X, Li R, Wang Y, et al. Immunogenicity and protective efficacy of multistage vaccine candidates (Mtb8.4-HspX and HspX-Mtb8.4) against Mycobacterium tuberculosis infection in mice. Int Immunopharmacol. 2017;53:83-89.DOI: 10.1016/j.intimp.2017.10.015.
31. Dheda K, Mirzayev F, Cirillo DM, Udwadia Z, Dooley KE, Chang KC, et al. Multidrug-resistant tuberculosis. Nat Rev Dis Primers. 2024;10(1):22.DOI: 10.1038/s41572-024-00504-2.
32. Grobbel HP, Merker M, Kohler N, Andres S, Hoffmann H, Heyckendorf J, et al. Design of multidrug-resistant tuberculosis treatment regimens based on DNA sequencing. Clin Infect Dis. 2021;73(7):1194-1202.DOI: 10.1093/cid/ciab359.
33. The Economist Inteligence Unit. It's time to end drug-resistant tuberculosis. The Economist. 2019. Available at https://www.eiu.com/graphics/marketing/pdf/its-time-to-end-drug-resistant-tuberculosis-full-report.pdf. [accessed on 22 July 2024].
34. Ahmed S, Raqib R, Guethmundsson GH, Bergman P, Agerberth B, Rekha RS. Host-directed therapy as a novel treatment strategy to overcome tuberculosis: targeting immune modulation. Antibiotics (Basel). 2020;9(1):21.DOI: 10.3390/antibiotics9010021.
35. Ayodele S, Kumar P, van Eyk A, Choonara YE. Advances in immunomodulatory strategies for host-directed therapies in combating tuberculosis. Biomed Pharmacother. 2023;162:114588.DOI: 10.1016/j.biopha.2023.114588.
36. Kiran D, Podell BK, Chambers M, Basaraba RJ. Host-directed therapy targeting the Mycobacterium tuberculosis granuloma: a review. Semin Immunopathol. 2016;38(2):167-183.DOI: 10.1007/s00281-015-0537-x.
37. Ramakrishnan L. Revisiting the role of the granuloma in tuberculosis. Nat Rev Immunol. 2012;12(5):352-366.DOI: 10.1038/nri3211.
38. Davis JM, Ramakrishnan L. The role of the granuloma in expansion and dissemination of early tuberculous infection. Cell. 2009;136(1):37-49.DOI: 10.1016/j.cell.2008.11.014.
39. Weeratunga P, Moller DR, Ho LP. Immune mechanisms of granuloma formation in sarcoidosis and tuberculosis. J Clin Invest. 2024;134(1):e175264.DOI: 10.1172/JCI175264.
40. Davis JM, Clay H, Lewis JL, Ghori N, Herbomel P, Ramakrishnan L. Real-time visualization of mycobacterium-macrophage interactions leading to initiation of granuloma formation in zebrafish embryos. Immunity. 2002;17(6):693-702.DOI: 10.1016/S1074-7613(02)00475-2.
41. Belton M, Brilha S, Manavaki R, Mauri F, Nijran K, Hong YT, et al. Hypoxia and tissue destruction in pulmonary TB. Thorax. 2016;71(12):1145-1153.DOI: 10.1136/thoraxjnl-2015-207402.
42. Rabinowitz JD, Enerback S. Lactate: the ugly duckling of energy metabolism. Nat Metab. 2020;2(7):566-571.DOI: 10.1038/s42255-020-0243-4.
43. Emad A, Rezaian GR. Lactate dehydrogenase in bronchoalveolar lavage fluid of patients with active pulmonary tuberculosis. Respiration. 1999;66(1):41-45.DOI: 10.1159/000029335.
44. Sharma PR, Jain S, Bamezai RN, Tiwari PK. Utility of serum LDH isoforms in the assessment of Mycobacterium tuberculosis induced pathology in TB patients of Sahariya tribe. Indian J Clin Biochem. 2010;25(1):57-63.DOI: 10.1007/s12291-010-0012-3.
45. Osada-Oka M, Goda N, Saiga H, Yamamoto M, Takeda K, Ozeki Y, et al. Metabolic adaptation to glycolysis is a basic defense mechanism of macrophages for Mycobacterium tuberculosis infection. Int Immunol. 2019;31(12):781-793.DOI: 10.1093/intimm/dxz048.
46. Elkington PT, D'Armiento JM, Friedland JS. Tuberculosis immunopathology: the neglected role of extracellular matrix destruction. Sci Transl Med. 2011;3(71):71ps6.DOI: 10.1126/scitranslmed.3001847.
47. Li Y, Wang Y, Liu X. The role of airway epithelial cells in response to mycobacteria infection. Clin Dev Immunol. 2012;2012:791392.DOI: 10.1155/2012/791392.
48. Ghio AJ, Carter JD, Dailey LA, Devlin RB, Samet JM. Respiratory epithelial cells demonstrate lactoferrin receptors that increase after metal exposure. Am J Physiol. 1999;276(6):L933-40.DOI: 10.1152/ajplung.1999.276.6.L933.
49. Doss M, White MR, Tecle T, Hartshorn KL. Human defensins and LL-37 in mucosal immunity. J Leukoc Biol. 2010;87(1):79-92.DOI: 10.1189/jlb.0609382.
50. Elkington PT, Emerson JE, Lopez-Pascua LD, O'Kane CM, Horncastle DE, Boyle JJ, et al. Mycobacterium tuberculosis up-regulates matrix metalloproteinase-1 secretion from human airway epithelial cells via a p38 MAPK switch. J Immunol. 2005;175(8):5333-5340.DOI: 10.4049/jimmunol.175.8.5333.
51. Hiemstra PS. Epithelial antimicrobial peptides and proteins: their role in host defence and inflammation. Paediatr Respir Rev. 2001;2(4):306-310.DOI: 10.1053/prrv.2001.0165.
52. Lutay N, Hakansson G, Alaridah N, Hallgren O, Westergren-Thorsson G, Godaly G. Mycobacteria bypass mucosal NF-kB signalling to induce an epithelial anti-inflammatory IL-22 and IL-10 response. PLoS One. 2014;9(1):e86466.DOI: 10.1371/journal.pone.0086466.
53. Kinhikar AG, Verma I, Chandra D, Singh KK, Weldingh K, Andersen P, et al. Potential role for ESAT6 in dissemination of M. tuberculosis via human lung epithelial cells. Mol Microbiol. 2010;75(1):92-106.DOI: 10.1111/j.1365-2958.2009.06959.x.
54. Mai J, Virtue A, Shen J, Wang H, Yang XF. An evolving new paradigm: endothelial cells--conditional innate immune cells. J Hematol Oncol. 2013;6:61.DOI: 10.1186/1756-8722-6-61.
55. Baltierra-Uribe SL, Garcia-Vasquez Mde J, Castrejon-Jimenez NS, Estrella-Pinon MP, Luna-Herrera J, Garcia-Perez BE. Mycobacteria entry and trafficking into endothelial cells. Can J Microbiol. 2014;60(9):569-577.DOI: 10.1139/cjm-2014-0087.
56. Garcia-Perez BE, Villagomez-Palatto DA, Castaneda-Sanchez JI, Coral-Vazquez RM, Ramirez-Sanchez I, Ordonez-Razo RM, et al. Innate response of human endothelial cells infected with mycobacteria. Immunobiology. 2011;216(8):925-935.DOI: 10.1016/j.imbio.2011.01.004.
57. Lerner TR, Queval CJ, Fearns A, Repnik U, Griffiths G, Gutierrez MG. Phthiocerol dimycocerosates promote access to the cytosol and intracellular burden of Mycobacterium tuberculosis in lymphatic endothelial cells. BMC Biol. 2018;16(1):1.DOI: 10.1186/s12915-017-0471-6.
58. East L, Isacke CM. The mannose receptor family. Biochim Biophys Acta. 2002;1572(2-3):364-386.DOI: 10.1016/S0304-4165(02)00319-7.
59. Polena H, Boudou F, Tilleul S, Dubois-Colas N, Lecointe C, Rakotosamimanana N, et al. Mycobacterium tuberculosis exploits the formation of new blood vessels for its dissemination. Sci Rep. 2016;6:33162.DOI: 10.1038/srep33162.
60. Saghazadeh A, Rezaei N. Vascular endothelial growth factor levels in tuberculosis: A systematic review and meta-analysis. PLoS One. 2022;17(5):e0268543.DOI: 10.1371/journal.pone.0268543.
61. Matsuyama W, Hashiguchi T, Matsumuro K, Iwami F, Hirotsu Y, Kawabata M, et al. Increased serum level of vascular endothelial growth factor in pulmonary tuberculosis. Am J Respir Crit Care Med. 2000;162(3 Pt 1):1120-1122.DOI: 10.1164/ajrccm.162.3.9911010.
62. Alatas F, Alatas O, Metintas M, Ozarslan A, Erginel S, Yildirim H. Vascular endothelial growth factor levels in active pulmonary tuberculosis. Chest. 2004;125(6):2156-2159.DOI: 10.1378/chest.125.6.2156.
63. Datta M, Via LE, Kamoun WS, Liu C, Chen W, Seano G, et al. Anti-vascular endothelial growth factor treatment normalizes tuberculosis granuloma vasculature and improves small molecule delivery. Proc Natl Acad Sci U S A. 2015;112(6):1827-1832.DOI: 10.1073/pnas.1424563112.
64. Harding J, Ritter A, Rayasam A, Fabry Z, Sandor M. Lymphangiogenesis is induced by mycobacterial granulomas via vascular endothelial growth factor receptor-3 and supports systemic T-cell responses against mycobacterial antigen. Am J Pathol. 2015;185(2):432-445.DOI: 10.1016/j.ajpath.2014.09.020.
65. Gomez Perdiguero E, Klapproth K, Schulz C, Busch K, Azzoni E, Crozet L, et al. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature. 2015;518(7540):547-551.DOI: 10.1038/nature13989.
66. Hoeffel G, Chen J, Lavin Y, Low D, Almeida FF, See P, et al. C-Myb(+) erythro-myeloid progenitor-derived fetal monocytes give rise to adult tissue-resident macrophages. Immunity. 2015;42(4):665-678.DOI: 10.1016/j.immuni.2015.03.011.
67. Aegerter H, Lambrecht BN, Jakubzick CV. Biology of lung macrophages in health and disease. Immunity. 2022;55(9):1564-1580.DOI: 10.1016/j.immuni.2022.08.010.
68. Huang L, Nazarova EV, Tan S, Liu Y, Russell DG. Growth of Mycobacterium tuberculosis in vivo segregates with host macrophage metabolism and ontogeny. J Exp Med. 2018;215(4):1135-1152.DOI: 10.1084/jem.20172020.
69. Gordon S. Alternative activation of macrophages. Nat Rev Immunol. 2003;3(1):23-35.DOI: 10.1038/nri978.
70. Cohen SB, Gern BH, Delahaye JL, Adams KN, Plumlee CR, Winkler JK, et al. Alveolar macrophages provide an early Mycobacterium tuberculosis niche and initiate dissemination. Cell Host Microbe. 2018;24(3):439-446. e4.DOI: 10.1016/j.chom.2018.08.001.
71. Redente EF, Higgins DM, Dwyer-Nield LD, Orme IM, Gonzalez-Juarrero M, Malkinson AM. Differential polarization of alveolar macrophages and bone marrow-derived monocytes following chemically and pathogen-induced chronic lung inflammation. J Leukoc Biol. 2010;88(1):159-168.DOI: 10.1189/jlb.0609378.
72. Blumenthal A, Kobayashi T, Pierini LM, Banaei N, Ernst JD, Miyake K, et al. RP105 facilitates macrophage activation by Mycobacterium tuberculosis lipoproteins. Cell Host Microbe. 2009;5(1):35-46.DOI: 10.1016/j.chom.2008.12.002.
73. Sha S, Shi Y, Tang Y, Jia L, Han X, Liu Y, et al. Mycobacterium tuberculosis Rv1987 protein induces M2 polarization of macrophages through activating the PI3K/Akt1/mTOR signaling pathway. Immunol Cell Biol. 2021;99(6):570-585.DOI: 10.1111/imcb.12436.
74. Gong Z, Han S, Liang T, Zhang H, Sun Q, Pan H, et al. Mycobacterium tuberculosis effector PPE36 attenuates host cytokine storm damage via inhibiting macrophage M1 polarization. J Cell Physiol. 2021;236(11):7405-7420.DOI: 10.1002/jcp.30411.
75. Guo Q, Bi J, Li M, Ge W, Xu Y, Fan W, et al. ESX secretion-associated protein C from Mycobacterium tuberculosis induces macrophage activation through the toll-like receptor-4/mitogen-activated protein kinase signaling pathway. Front Cell Infect Microbiol. 2019;9:158.DOI: 10.3389/fcimb.2019.00158.
76. Ahmad F, Rani A, Alam A, Zarin S, Pandey S, Singh H, et al. Macrophage: a cell with many faces and functions in Tuberculosis. Front Immunol. 2022;13:747799.DOI: 10.3389/fimmu.2022.747799.
77. Marino S, Cilfone NA, Mattila JT, Linderman JJ, Flynn JL, Kirschner DE. Macrophage polarization drives granuloma outcome during Mycobacterium tuberculosis infection. Infect Immun. 2015;83(1):324-338.DOI: 10.1128/IAI.02494-14.
78. Bo H, Moure UAE, Yang Y, Pan J, Li L, Wang M, et al. Mycobacterium tuberculosis-macrophage interaction: molecular updates. Front Cell Infect Microbiol. 2023;13:1062963.DOI: 10.3389/fcimb.2023.1062963.
79. Songane M, Kleinnijenhuis J, Netea MG, van Crevel R. The role of autophagy in host defence against Mycobacterium tuberculosis infection. Tuberculosis (Edinb). 2012;92(5):388-396.DOI: 10.1016/j.tube.2012.05.004.
80. Lee J, Hartman M, Kornfeld H. Macrophage apoptosis in tuberculosis. Yonsei Med J. 2009;50(1):1-11.DOI: 10.3349/ymj.2009.50.1.1.
81. Feng Y, Li M, Yangzhong X, Zhang X, Zu A, Hou Y, et al. Pyroptosis in inflammation-related respiratory disease. J Physiol Biochem. 2022;78(4):721-737.DOI: 10.1007/s13105-022-00909-1.
82. Shin DM, Jeon BY, Lee HM, Jin HS, Yuk JM, Song CH, et al. Mycobacterium tuberculosis Eis regulates autophagy, inflammation, and cell death through redox-dependent signaling. PLoS Pathog. 2010;6(12):e1001230.DOI: 10.1371/journal.ppat.1001230.
83. Duan L, Yi M, Chen J, Li S, Chen W. Mycobacterium tuberculosis eis gene inhibits macrophage autophagy through up-regulation of IL-10 by increasing the acetylation of histone H3. Biochem Biophys Res Commun. 2016;473(4):1229-1234.DOI: 10.1016/j.bbrc.2016.04.045.
84. Singh A, Crossman DK, Mai D, Guidry L, Voskuil MI, Renfrow MB, et al. Mycobacterium tuberculosis WhiB3 maintains redox homeostasis by regulating virulence lipid anabolism to modulate macrophage response. PLoS Pathog. 2009;5(8):e1000545.DOI: 10.1371/journal.ppat.1000545.
85. Moideen K, Kumar NP, Nair D, Banurekha VV, Bethunaickan R, Babu S. Heightened systemic levels of neutrophil and eosinophil granular proteins in pulmonary tuberculosis and reversal following treatment. Infect Immun. 2018;86(6):e00008-18.DOI: 10.1128/IAI.00008-18.
86. Monteith AJ, Miller JM, Maxwell CN, Chazin WJ, Skaar EP. Neutrophil extracellular traps enhance macrophage killing of bacterial pathogens. Sci Adv. 2021;7(37):eabj2101.DOI: 10.1126/sciadv.abj2101.
87. Blomgran R, Desvignes L, Briken V, Ernst JD. Mycobacterium tuberculosis inhibits neutrophil apoptosis, leading to delayed activation of naive CD4 T cells. Cell Host Microbe. 2012;11(1):81-90.DOI: 10.1016/j.chom.2011.11.012.
88. Corleis B, Korbel D, Wilson R, Bylund J, Chee R, Schaible UE. Escape of Mycobacterium tuberculosis from oxidative killing by neutrophils. Cell Microbiol. 2012;14(7):1109-1121.DOI: 10.1111/j.1462-5822.2012.01783.x.
89. Repasy T, Martinez N, Lee J, West K, Li W, Kornfeld H. Bacillary replication and macrophage necrosis are determinants of neutrophil recruitment in tuberculosis. Microbes Infect. 2015;17(8):564-574.DOI: 10.1016/j.micinf.2015.03.013.
90. Dallenga T, Repnik U, Corleis B, Eich J, Reimer R, Griffiths GW, et al. M. tuberculosis-induced necrosis of infected neutrophils promotes bacterial growth following phagocytosis by macrophages. Cell Host Microbe. 2017;22(4):519-530. e3.DOI: 10.1016/j.chom.2017.09.003.
91. Filio-Rodriguez G, Estrada-Garcia I, Arce-Paredes P, Moreno-Altamirano MM, Islas-Trujillo S, Ponce-Regalado MD, et al. In vivo induction of neutrophil extracellular traps by Mycobacterium tuberculosis in a guinea pig model. Innate Immun. 2017;23(7):625-637.DOI: 10.1177/1753425917732406.
92. Nouailles G, Dorhoi A, Koch M, Zerrahn J, Weiner J 3rd, Fae KC, et al. CXCL5-secreting pulmonary epithelial cells drive destructive neutrophilic inflammation in tuberculosis. J Clin Invest. 2014;124(3):1268-1282.DOI: 10.1172/JCI72030.
93. Kim TS, Jin YB, Kim YS, Kim S, Kim JK, Lee HM, et al. SIRT3 promotes antimycobacterial defenses by coordinating mitochondrial and autophagic functions. Autophagy. 2019;15(8):1356-1375.DOI: 10.1080/15548627.2019.1582743.
94. Geijtenbeek TB, Van Vliet SJ, Koppel EA, Sanchez-Hernandez M, Vandenbroucke-Grauls CM, Appelmelk B, et al. Mycobacteria target DC-SIGN to suppress dendritic cell function. J Exp Med. 2003;197(1):7-17.DOI: 10.1084/jem.20021229.
95. Chieppa M, Bianchi G, Doni A, Del Prete A, Sironi M, Laskarin G, et al. Cross-linking of the mannose receptor on monocyte-derived dendritic cells activates an anti-inflammatory immunosuppressive program. J Immunol. 2003;171(9):4552-60.DOI: 10.4049/jimmunol.171.9.4552.
96. Magallanes-Puebla A, Espinosa-Cueto P, Lopez-Marin LM, Mancilla R. Mycobacterial glycolipid Di-O-acyl trehalose promotes a tolerogenic profile in dendritic cells. PLoS One. 2018;13(12):e0207202.DOI: 10.1371/journal.pone.0207202.
97. Sondergaard JN, Laursen JM, Rosholm LB, Brix S. Mycobacterium tuberculosis promotes Th17 expansion via regulation of human dendritic cells toward a high CD14 and low IL-12p70 phenotype that reprograms upon exogenous IFN-gamma. Int Immunol. 2014;26(12):705-716.DOI: 10.1093/intimm/dxu085.
98. Cadena AM, Fortune SM, Flynn JL. Heterogeneity in tuberculosis. Nat Rev Immunol. 2017;17(11):691-702.DOI: 10.1038/nri.2017.69.
99. Ulrichs T, Kosmiadi GA, Jorg S, Pradl L, Titukhina M, Mishenko V, et al. Differential organization of the local immune response in patients with active cavitary tuberculosis or with nonprogressive tuberculoma. J Infect Dis. 2005;192(1)):89-97.DOI: 10.1086/430621.
100. Caruso AM, Serbina N, Klein E, Triebold K, Bloom BR, Flynn JL. Mice deficient in CD4 T cells have only transiently diminished levels of IFN-gamma, yet succumb to tuberculosis. J Immunol. 1999;162(9):5407-5416.DOI: 10.4049/jimmunol.162.9.5407.
101. Flynn JL, Goldstein MM, Chan J, Triebold KJ, Pfeffer K, Lowenstein CJ, et al. Tumor necrosis factor-alpha is required in the protective immune response against Mycobacterium tuberculosis in mice. Immunity. 1995;2(6):561-572.DOI: 10.1016/1074-7613(95)90001-2.
102. Ansari AW, Kamarulzaman A, Schmidt RE. Multifaceted impact of host C-C chemokine CCL2 in the immuno-pathogenesis of HIV-1/M. tuberculosis co-infection. Front Immunol. 2013;4:312.DOI: 10.3389/fimmu.2013.00312.
103. Diedrich CR, O'Hern J, Wilkinson RJ. HIV-1 and the Mycobacterium tuberculosis granuloma: A systematic review and meta-analysis. Tuberculosis (Edinb). 2016;98:62-76.DOI: 10.1016/j.tube.2016.02.010.
104. Lu YJ, Barreira-Silva P, Boyce S, Powers J, Cavallo K, Behar SM. CD4 T cell help prevents CD8 T cell exhaustion and promotes control of Mycobacterium tuberculosis infection. Cell Rep. 2021;36(11):109696.DOI: 10.1016/j.celrep.2021.109696.
105. Serbina NV, Liu CC, Scanga CA, Flynn JL. CD8+ CTL from lungs of Mycobacterium tuberculosis-infected mice express perforin in vivo and lyse infected macrophages. J Immunol. 2000;165(1):353-363.DOI: 10.4049/jimmunol.165.1.353.
106. Chavez-Galan L, Illescas-Eugenio J, Alvarez-Sekely M, Baez-Saldana R, Chavez R, Lascurain R. Tuberculosis patients display a high proportion of CD8(+) T cells with a high cytotoxic potential. Microbiol Immunol. 2019;63(8):316-327.DOI: 10.1111/1348-0421.12724.
107. Shafiani S, Tucker-Heard G, Kariyone A, Takatsu K, Urdahl KB. Pathogen-specific regulatory T cells delay the arrival of effector T cells in the lung during early tuberculosis. J Exp Med. 2010;207(7):1409-1420.DOI: 10.1084/jem.20091885.
108. Kursar M, Koch M, Mittrucker HW, Nouailles G, Bonhagen K, Kamradt T, et al. Cutting edge: regulatory T cells prevent efficient clearance of Mycobacterium tuberculosis. J Immunol. 2007;178(5):2661-2665.DOI: 10.4049/jimmunol.178.5.2661.
109. Scott-Browne JP, Shafiani S, Tucker-Heard G, Ishida-Tsubota K, Fontenot JD, Rudensky AY, et al. Expansion and function of Foxp3-expressing T regulatory cells during tuberculosis. J Exp Med. 2007;204(9):2159-2169.DOI: 10.1084/jem.20062105.
110. Irvine EB, O'Neil A, Darrah PA, Shin S, Choudhary A, Li W, et al. Robust IgM responses following intravenous vaccination with Bacille Calmette-Guerin associate with prevention of Mycobacterium tuberculosis infection in macaques. Nat Immunol. 2021;22(12):1515-1523.DOI: 10.1038/s41590-021-01066-1.
111. Watson A, Li H, Ma B, Weiss R, Bendayan D, Abramovitz L, et al. Human antibodies targeting a Mycobacterium transporter protein mediate protection against tuberculosis. Nat Commun. 2021;12(1):602.DOI: 10.1038/s41467-021-20930-0.
112. Klionsky DJ. The molecular machinery of autophagy: unanswered questions. J Cell Sci. 2005;118(Pt 1):7-18.DOI: 10.1242/jcs.01620.
113. Feng Y, Yao Z, Klionsky DJ. How to control self-digestion: transcriptional, post-transcriptional, and post-translational regulation of autophagy. Trends Cell Biol. 2015;25(6):354-363.DOI: 10.1016/j.tcb.2015.02.002.
114. Yamamoto H, Zhang S, Mizushima N. Autophagy genes in biology and disease. Nat Rev Genet. 2023;24(6):382-400.DOI: 10.1038/s41576-022-00562-w.
115. Kumar S, Jain A, Choi SW, Peixoto Duarte da Silva G, Allers L, Mudd MH, et al. Mammalian Atg8-family proteins are upstream regulators of the lysosomalsystem by controlling MTOR and TFEB. Autophagy. 2020;16(12):2305-2306.DOI: 10.1080/15548627.2020.1837423.
116. Kim YS, Silwal P, Kim SY, Yoshimori T, Jo EK. Autophagy-activating strategies to promote innate defense against mycobacteria. Exp Mol Med. 2019;51(12):1-10.DOI: 10.1038/s12276-019-0290-7.
117. Gutierrez MG, Master SS, Singh SB, Taylor GA, Colombo MI, Deretic V. Autophagy is a defense mechanism inhibiting BCG and Mycobacterium tuberculosis survival in infected macrophages. Cell. 2004;119(6):753-766.DOI: 10.1016/j.cell.2004.11.038.
118. Delgado M, Singh S, De Haro S, Master S, Ponpuak M, Dinkins C, et al. Autophagy and pattern recognition receptors in innate immunity. Immunol Rev. 2009;227(1):189-202.DOI: 10.1111/j.1600-065X.2008.00725.x.
119. Xu Y, Jagannath C, Liu XD, Sharafkhaneh A, Kolodziejska KE, Eissa NT. Toll-like receptor 4 is a sensor for autophagy associated with innate immunity. Immunity. 2007;27(1):135-144.DOI: 10.1016/j.immuni.2007.05.022.
120. Lee HK, Lund JM, Ramanathan B, Mizushima N, Iwasaki A. Autophagy-dependent viral recognition by plasmacytoid dendritic cells. Science. 2007;315(5817):1398-1401.DOI: 10.1126/science.1136880.
121. Delgado MA, Elmaoued RA, Davis AS, Kyei G, Deretic V. Toll-like receptors control autophagy. EMBO J. 2008;27(7):1110-1121.DOI: 10.1038/emboj.2008.31.
122. Yuk JM, Shin DM, Lee HM, Yang CS, Jin HS, Kim KK, et al. Vitamin D3 induces autophagy in human monocytes/macrophages via cathelicidin. Cell Host Microbe. 2009;6(3):231-243.DOI: 10.1016/j.chom.2009.08.004.
123. Schiebler M, Brown K, Hegyi K, Newton SM, Renna M, Hepburn L, et al. Functional drug screening reveals anticonvulsants as enhancers of mTOR-independent autophagic killing of Mycobacterium tuberculosis through inositol depletion. EMBO Mol Med. 2015;7(2):127-139.DOI: 10.15252/emmm.201404137.
124. Persaud R, Li SC, Chao JD, Forestieri R, Donohue E, Balgi AD, et al. Clionamines stimulate autophagy, inhibit Mycobacterium tuberculosis survival in macrophages, and target Pik1. Cell Chem Biol. 2022;29(5):870-882. e11.DOI: 10.1016/j.chembiol.2021.07.017.
125. Khaminets A, Behl C, Dikic I. Ubiquitin-dependent and independent signals in selective autophagy. Trends Cell Biol. 2016;26(1):6-16.DOI: 10.1016/j.tcb.2015.08.010.
126. McEwan DG. Host-pathogen interactions and subversion of autophagy. Essays Biochem. 2017;61(6):687-697.DOI: 10.1042/EBC20170058.
127. Wild P, Farhan H, McEwan DG, Wagner S, Rogov VV, Brady NR, et al. Phosphorylation of the autophagy receptor optineurin restricts Salmonella growth. Science. 2011;333(6039):228-233.DOI: 10.1126/science.1205405.
128. Jo EK, Yuk JM, Shin DM, Sasakawa C. Roles of autophagy in elimination of intracellular bacterial pathogens. Front Immunol. 2013;4:97.DOI: 10.3389/fimmu.2013.00097.
129. Castrejon-Jimenez NS, Leyva-Paredes K, Hernandez-Gonzalez JC, Luna-Herrera J, Garcia-Perez BE. The role of autophagy in bacterial infections. Biosci Trends. 2015;9(3):149-159.DOI: 10.5582/bst.2015.01035.
130. Bell SL, Lopez KL, Cox JS, Patrick KL, Watson RO. Galectin-8 senses phagosomal damage and recruits selective autophagy adapter TAX1BP1 to control Mycobacterium tuberculosis infection in macrophages. mBio. 2021;12(4):e0187120.DOI: 10.1128/mBio.01871-20.
131. Sakowski ET, Koster S, Portal Celhay C, Park HS, Shrestha E, Hetzenecker SE, et al. Ubiquilin 1 promotes IFN-gamma-induced xenophagy of Mycobacterium tuberculosis. PLoS Pathog. 2015;11(7):e1005076.DOI: 10.1371/journal.ppat.1005076.
132. Romagnoli A, Di Rienzo M, Petruccioli E, Fusco C, Palucci I, Micale L, et al. The ubiquitin ligase TRIM32 promotes the autophagic response to Mycobacterium tuberculosis infection in macrophages. Cell Death Dis. 2023;14(8):505.DOI: 10.1038/s41419-023-06026-1.
133. Lee YJ, Kim JK, Jung CH, Kim YJ, Jung EJ, Lee SH, et al. Chemical modulation of SQSTM1/p62-mediated xenophagy that targets a broad range of pathogenic bacteria. Autophagy. 2022;18(12):2926-2945.DOI: 10.1080/15548627.2022.2054240.
134. Mizushima N. The pleiotropic role of autophagy: from protein metabolism to bactericide. Cell Death Differ. 2005;12 Suppl 2:1535-1541.DOI: 10.1038/sj.cdd.4401728.
135. Pena-Martinez C, Rickman AD, Heckmann BL. Beyond autophagy: LC3-associated phagocytosis and endocytosis. Sci Adv. 2022;8(43):eabn1702.DOI: 10.1126/sciadv.abn1702.
136. Durgan J, Lystad AH, Sloan K, Carlsson SR, Wilson MI, Marcassa E, et al. Non-canonical autophagy drives alternative ATG8 conjugation to phosphatidylserine. Mol Cell. 2021;81(9):2031-2040. e8.DOI: 10.1016/j.molcel.2021.03.020.
137. Heckmann BL, Green DR. LC3-associated phagocytosis at a glance. J Cell Sci. 2019;132(5):jcs222984.DOI: 10.1242/jcs.222984.
138. Koster S, Upadhyay S, Chandra P, Papavinasasundaram K, Yang G, Hassan A, et al. Mycobacterium tuberculosis is protected from NADPH oxidase and LC3-associated phagocytosis by the LCP protein CpsA. Proc Natl Acad Sci U S A. 2017;114(41):E8711-E8720.DOI: 10.1073/pnas.1707792114.
139. Ge P, Lei Z, Yu Y, Lu Z, Qiang L, Chai Q, et al. M. tuberculosis PknG manipulates host autophagy flux to promote pathogen intracellular survival. Autophagy. 2022;18(3):576-594.DOI: 10.1080/15548627.2021.1938912.
140. Bedard M, van der Niet S, Bernard EM, Babunovic G, Cheng TY, Aylan B, et al. A terpene nucleoside from M. tuberculosis induces lysosomal lipid storage in foamy macrophages. J Clin Invest. 2023;133(6):e161944.DOI: 10.1172/JCI161944.
141. Paik S, Kim KT, Kim IS, Kim YJ, Kim HJ, Choi S, et al. Mycobacterial acyl carrier protein suppresses TFEB activation and upregulates miR-155 to inhibit host defense. Front Immunol. 2022;13:946929.DOI: 10.3389/fimmu.2022.946929.
142. Strong EJ, Wang J, Ng TW, Porcelli SA, Lee S. Mycobacterium tuberculosis PPE51 inhibits autophagy by suppressing Toll-like receptor 2-dependent signaling. mBio. 2022;13(3):e0297421.DOI: 10.1128/mbio.02974-21.
143. Strong EJ, Ng TW, Porcelli SA, Lee S. Mycobacterium tuberculosis PE_PGRS20 and PE_PGRS47 proteins inhibit autophagy by interaction with Rab1A. mSphere. 2021;6(4):e0054921.DOI: 10.1128/mSphere.00549-21.
144. Garcia-Bengoa M, Meurer M, Goethe R, Singh M, Reljic R, von Kockritz-Blickwede M. Role of phagocyte extracellular traps during Mycobacterium tuberculosis infections and tuberculosis disease processes. Front Microbiol. 2023;14:983299.DOI: 10.3389/fmicb.2023.983299.
145. Diatlova A, Linkova N, Lavrova A, Zinchenko Y, Medvedev D, Krasichkov A, et al. Molecular markers of rarly immune response in tuberculosis: prospects of application in predictive medicine. Int J Mol Sci. 2023;24(17):13261.DOI: 10.3390/ijms241713261.
146. Ashenafi S, Loreti MG, Bekele A, Aseffa G, Amogne W, Kassa E, et al. Inflammatory immune profiles associated with disease severity in pulmonary tuberculosis patients with moderate to severe clinical TB or anemia. Front Immunol. 2023;14:1296501.DOI: 10.3389/fimmu.2023.1296501.
147. Sudbury EL, Clifford V, Messina NL, Song R, Curtis N. Mycobacterium tuberculosis-specific cytokine biomarkers to differentiate active TB and LTBI: A systematic review. J Infect. 2020;81(6):873-881.DOI: 10.1016/j.jinf.2020.09.032.
148. Dorhoi A, Kaufmann SH. Tumor necrosis factor alpha in mycobacterial infection. Semin Immunol. 2014;26(3):203-209.DOI: 10.1016/j.smim.2014.04.003.
149. Ray JC, Flynn JL, Kirschner DE. Synergy between individual TNF-dependent functions determines granuloma performance for controlling Mycobacterium tuberculosis infection. J Immunol. 2009;182(6):3706-3717.DOI: 10.4049/jimmunol.0802297.
150. Roach DR, Bean AG, Demangel C, France MP, Briscoe H, Britton WJ. TNF regulates chemokine induction essential for cell recruitment, granuloma formation, and clearance of mycobacterial infection. J Immunol. 2002;168(9):4620-4627.DOI: 10.4049/jimmunol.168.9.4620.
151. Gomez-Reino JJ, Carmona L, Valverde VR, Mola EM, Montero MD, Group B. Treatment of rheumatoid arthritis with tumor necrosis factor inhibitors may predispose to significant increase in tuberculosis risk: a multicenter active-surveillance report. Arthritis Rheum. 2003;48(8):2122-2127.DOI: 10.1002/art.11137.
152. Francisco NM, Hsu NJ, Keeton R, Randall P, Sebesho B, Allie N, et al. TNF-dependent regulation and activation of innate immune cells are essential for host protection against cerebral tuberculosis. J Neuroinflammation. 2015;12:125.DOI: 10.1186/s12974-015-0345-1.
153. Vanden Driessche K, Persson A, Marais BJ, Fink PJ, Urdahl KB. Immune vulnerability of infants to tuberculosis. Clin Dev Immunol. 2013;2013:781320.DOI: 10.1155/2013/781320.
154. Swanson KV, Deng M, Ting JP. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat Rev Immunol. 2019;19(8):477-489.DOI: 10.1038/s41577-019-0165-0.
155. Mishra BB, Moura-Alves P, Sonawane A, Hacohen N, Griffiths G, Moita LF, et al. Mycobacterium tuberculosis protein ESAT-6 is a potent activator of the NLRP3/ASC inflammasome. Cell Microbiol. 2010;12(8):1046-1063.DOI: 10.1111/j.1462-5822.2010.01450.x.
156. Amaral EP, Riteau N, Moayeri M, Maier N, Mayer-Barber KD, Pereira RM, et al. Lysosomal cathepsin release is required for NLRP3-inflammasome activation by Mycobacterium tuberculosis in infected macrophages. Front Immunol. 2018;9:1427.DOI: 10.3389/fimmu.2018.01427.
157. Koo IC, Wang C, Raghavan S, Morisaki JH, Cox JS, Brown EJ. ESX-1-dependent cytolysis in lysosome secretion and inflammasome activation during mycobacterial infection. Cell Microbiol. 2008;10(9):1866-1878.DOI: 10.1111/j.1462-5822.2008.01177.x.
158. Silverio D, Goncalves R, Appelberg R, Saraiva M. Advances on the role and applications of interleukin-1 in tuberculosis. mBio. 2021;12(6):e0313421.DOI: 10.1128/mBio.03134-21.
159. Winchell CG, Mishra BB, Phuah JY, Saqib M, Nelson SJ, Maiello P, et al. Evaluation of IL-1 blockade as an adjunct to linezolid therapy for tuberculosis in mice and macaques. Front Immunol. 2020;11:891.DOI: 10.3389/fimmu.2020.00891.
160. Shah S, Bohsali A, Ahlbrand SE, Srinivasan L, Rathinam VA, Vogel SN, et al. Cutting edge: Mycobacterium tuberculosis but not nonvirulent mycobacteria inhibits IFN-beta and AIM2 inflammasome-dependent IL-1beta production via its ESX-1 secretion system. J Immunol. 2013;191(7):3514-3518.DOI: 10.4049/jimmunol.1301331.
161. Wong KW, Jacobs WR Jr. Critical role for NLRP3 in necrotic death triggered by Mycobacterium tuberculosis. Cell Microbiol. 2011;13(9):1371-1384.DOI: 10.1111/j.1462-5822.2011.01625.x.
162. Eklund D, Welin A, Andersson H, Verma D, Soderkvist P, Stendahl O, et al. Human gene variants linked to enhanced NLRP3 activity limit intramacrophage growth of Mycobacterium tuberculosis. J Infect Dis. 2014;209(5):749-753.DOI: 10.1093/infdis/jit572.
163. Dinarello CA, Simon A, van der Meer JW. Treating inflammation by blocking interleukin-1 in a broad spectrum of diseases. Nat Rev Drug Discov. 2012;11(8):633-652.DOI: 10.1038/nrd3800.
164. Dinarello CA, van der Meer JW. Treating inflammation by blocking interleukin-1 in humans. Semin Immunol. 2013;25(6):469-484.DOI: 10.1016/j.smim.2013.10.008.
165. Panda V, Ashar H, Srinath S. Antioxidant and hepatoprotective effect of Garcinia indica fruit rind in ethanol-induced hepatic damage in rodents. Interdiscip Toxicol. 2012;5(4):207-213.DOI: 10.2478/v10102-012-0034-1.
166. Brassard P, Kezouh A, Suissa S. Antirheumatic drugs and the risk of tuberculosis. Clin Infect Dis. 2006;43(6):717-722.DOI: 10.1086/506935.
167. Mayer-Barber KD, Barber DL, Shenderov K, White SD, Wilson MS, Cheever A, et al. Caspase-1 independent IL-1beta production is critical for host resistance to mycobacterium tuberculosis and does not require TLR signaling in vivo. J Immunol. 2010;184(7):3326-3330.DOI: 10.4049/jimmunol.0904189.
168. Yamada H, Mizumo S, Horai R, Iwakura Y, Sugawara I. Protective role of interleukin-1 in mycobacterial infection in IL-1 alpha/beta double-knockout mice. Lab Invest. 2000;80(5):759-767.DOI: 10.1038/labinvest.3780079.
169. Sugawara I, Yamada H, Hua S, Mizuno S. Role of interleukin (IL)-1 type 1 receptor in mycobacterial infection. Microbiol Immunol. 2001;45(11):743-750.DOI: 10.1111/j.1348-0421.2001.tb01310.x.
170. Bohrer AC, Tocheny C, Assmann M, Ganusov VV, Mayer-Barber KD. Cutting Edge: IL-1R1 mediates host resistance to Mycobacterium tuberculosis by trans-protection of infected cells. J Immunol. 2018;201(6):1645-1650.DOI: 10.4049/jimmunol.1800438.
171. Fremond CM, Togbe D, Doz E, Rose S, Vasseur V, Maillet I, et al. IL-1 receptor-mediated signal is an essential component of MyD88-dependent innate response to Mycobacterium tuberculosis infection. J Immunol. 2007;179(2):1178-1189.DOI: 10.4049/jimmunol.179.2.1178.
172. Mayer-Barber KD, Andrade BB, Oland SD, Amaral EP, Barber DL, Gonzales J, et al. Host-directed therapy of tuberculosis based on interleukin-1 and type I interferon crosstalk. Nature. 2014;511(7507):99-103.DOI: 10.1038/nature13489.
173. Kwon KW, Kim LH, Kang SM, Lee JM, Choi E, Park J, et al. Host-directed anti-mycobacterial activity of colchicine, an anti-gout drug, via strengthened host innate resistance reinforced by the IL-1beta/PGE2 axis. Br J Pharmacol. 2022;179(15):3951-3969.DOI: 10.1111/bph.15838.
174. Dorhoi A, Nouailles G, Jorg S, Hagens K, Heinemann E, Pradl L, et al. Activation of the NLRP3 inflammasome by Mycobacterium tuberculosis is uncoupled from susceptibility to active tuberculosis. Eur J Immunol. 2012;42(2):374-384.DOI: 10.1002/eji.201141548.
175. Wolf J, Rose-John S, Garbers C. Interleukin-6 and its receptors: a highly regulated and dynamic system. Cytokine. 2014;70(1):11-20.DOI: 10.1016/j.cyto.2014.05.024.
176. Gabay C. Interleukin-6 and chronic inflammation. Arthritis Res Ther. 2006;8 Suppl 2 (Suppl 2):S3.DOI: 10.1186/ar1917.
177. Rose-John S, Winthrop K, Calabrese L. The role of IL-6 in host defence against infections: immunobiology and clinical implications. Nat Rev Rheumatol. 2017;13(7):399-409.DOI: 10.1038/nrrheum.2017.83.
178. Gupte AN, Kumar P, Araujo-Pereira M, Kulkarni V, Paradkar M, Pradhan N, et al. Baseline IL-6 is a biomarker for unfavourable tuberculosis treatment outcomes: a multisite discovery and validation study. Eur Respir J. 2022;59(4):2100905.DOI: 10.1183/13993003.00905-2021.
179. Chowdhury IH, Choudhuri S, Sen A, Bhattacharya B, Ahmed AM, Hazra A, et al. Serum interleukin 6 (IL-6) as a potential biomarker of disease progression in active pulmonary tuberculosis following anti-tuberculosis drug therapy. Mol Immunol. 2015;63(2):601-602.DOI: 10.1016/j.molimm.2014.09.006.
180. Maseko TG, Ngubane S, Letsoalo M, Rambaran S, Archary D, Samsunder N, et al. Higher plasma interleukin - 6 levels are associated with lung cavitation in drug-resistant tuberculosis. BMC Immunol. 2023;24(1):26.DOI: 10.1186/s12865-023-00563-2.
181. Liu Y, Li X, Liu W, Liu Y, Zhong Z, Wang L, et al. IL-6 release of Rv0183 antigen-stimulated whole blood is a potential biomarker for active tuberculosis patients. J Infect. 2018;76(4):376-382.DOI: 10.1016/j.jinf.2017.11.004.
182. Antas P, Borchert J, Ponte C, Lima J, Georg I, Bastos M, et al. Interleukin-6 and -27 as potential novel biomarkers for human pleural tuberculosis regardless of the immunological status. Microbes Infect. 2024;26(1-2):105238.DOI: 10.1016/j.micinf.2023.105238.
183. Kruthika P. Role of IL 6 as a biomarker in the diagnosis of tuberculous meningitis - A systematic review. Int J Mycobacteriol. 2022;11(3):229-235.DOI: 10.4103/ijmy.ijmy_101_22.
184. Boni FG, Hamdi I, Koundi LM, Shrestha K, Xie J. Cytokine storm in tuberculosis and IL-6 involvement. Infect Genet Evol. 2022;97:105166.DOI: 10.1016/j.meegid.2021.105166.
185. Dutta RK, Kathania M, Raje M, Majumdar S. IL-6 inhibits IFN-gamma induced autophagy in Mycobacterium tuberculosis H37Rv infected macrophages. Int J Biochem Cell Biol. 2012;44(6):942-954.DOI: 10.1016/j.biocel.2012.02.021.
186. Jung BG, Wang X, Yi N, Ma J, Turner J, Samten B. Early secreted antigenic target of 6-kDa of Mycobacterium tuberculosis stimulates IL-6 production by macrophages through activation of STAT3. Sci Rep. 2017;7:40984.DOI: 10.1038/srep40984.
187. Rottenberg ME, Carow B. SOCS3 and STAT3, major controllers of the outcome of infection with Mycobacterium tuberculosis. Semin Immunol. 2014;26(6):518-532.DOI: 10.1016/j.smim.2014.10.004.
188. Ladel CH, Blum C, Dreher A, Reifenberg K, Kopf M, Kaufmann SH. Lethal tuberculosis in interleukin-6-deficient mutant mice. Infect Immun. 1997;65(11):4843-4849.DOI: 10.1128/iai.65.11.4843-4849.1997.
189. Saunders BM, Frank AA, Orme IM, Cooper AM. Interleukin-6 induces early gamma interferon production in the infected lung but is not required for generation of specific immunity to Mycobacterium tuberculosis infection. Infect Immun. 2000;68(6):3322-3326.DOI: 10.1128/IAI.68.6.3322-3326.2000.
190. Dienz O, Rincon M. The effects of IL-6 on CD4 T cell responses. Clin Immunol. 2009;130(1):27-33.DOI: 10.1016/j.clim.2008.08.018.
191. Linge I, Tsareva A, Kondratieva E, Dyatlov A, Hidalgo J, Zvartsev R, et al. Pleiotropic effect of IL-6 produced by B-Lymphocytes during early phases of adaptive immune responses against TB infection. Front Immunol. 2022;13:750068.DOI: 10.3389/fimmu.2022.750068.
192. Lindblad EB, Elhay MJ, Silva R, Appelberg R, Andersen P. Adjuvant modulation of immune responses to tuberculosis subunit vaccines. Infect Immun. 1997;65(2):623-629.DOI: 10.1128/iai.65.2.623-629.1997.
193. Slight SR, Rangel-Moreno J, Gopal R, Lin Y, Fallert Junecko BA, Mehra S, et al. CXCR5+ T helper cells mediate protective immunity against tuberculosis. J Clin Invest. 2013;123(2):712-726.DOI: 10.1172/JCI65728.
194. Liu S, Guan L, Peng C, Cheng Y, Cheng H, Wang F, et al. Mycobacterium tuberculosis suppresses host DNA repair to boost its intracellular survival. Cell Host Microbe. 2023;31(11):1820-1836. e10.DOI: 10.1016/j.chom.2023.09.010.
195. Ding Y, Bei C, Xue Q, Niu L, Tong J, Chen Y, et al. Transcriptomic analysis of mycobacterial infected macrophages reveals a high MOI specific type I IFN signaling. Infect Immun. 2023;91(7):e0015523.DOI: 10.1128/iai.00155-23.
196. Bobba S, Howard NC, Das S, Ahmed M, Tang L, Thirunavukkarasu S, et al. Mycobacterium tuberculosis carrying the rifampicin drug-resistance-conferring rpoB mutation H445Y is associated with suppressed immunity through type I interferons. mBio. 2023;14(5):e0094623.DOI: 10.1128/mbio.00946-23.
197. Dabla A, Liang YC, Rajabalee N, Irwin C, Moonen CGJ, Willis JV, et al. TREM2 promotes immune rvasion by Mycobacterium tuberculosis in human macrophages. mBio. 2022;13(4):e0145622.DOI: 10.1128/mbio.01456-22.
198. Naik SK, McNehlan ME, Mreyoud Y, Kinsella RL, Smirnov A, Chowdhury CS, et al. Type I IFN signaling in the absence of IRGM1 promotes M. tuberculosis replication in immune cells by suppressing T cell responses. bioRxiv. 2023;2023.DOI: 10.1101/2023.10.03.560720.
199. Kotov DI, Lee OV, Fattinger SA, Langner CA, Guillen JV, Peters JM, et al. Early cellular mechanisms of type I interferon-driven susceptibility to tuberculosis. Cell. 2023;186(25):5536-5553. e22.DOI: 10.1016/j.cell.2023.11.002.
200. Moreira-Teixeira L, Mayer-Barber K, Sher A, O'Garra A. Type I interferons in tuberculosis: Foe and occasionally friend. J Exp Med. 2018;215(5):1273-1285.DOI: 10.1084/jem.20180325.
201. Zarogoulidis P, Kioumis I, Papanas N, Manika K, Kontakiotis T, Papagianis A, et al. The effect of combination IFN-alpha-2a with usual antituberculosis chemotherapy in non-responding tuberculosis and diabetes mellitus: a case report and review of the literature. J Chemother. 2012;24(3):173-177.DOI: 10.1179/1973947812Y.0000000005.
202. Bax HI, Freeman AF, Ding L, Hsu AP, Marciano B, Kristosturyan E, et al. Interferon alpha treatment of patients with impaired interferon gamma signaling. J Clin Immunol. 2013;33(5):991-1001.DOI: 10.1007/s10875-013-9882-5.
203. Rivas-Santiago CE, Guerrero GG. IFN-alpha boosting of Mycobacterium bovis Bacillus Calmette Guerin-vaccine promoted Th1 type cellular response and protection against M. tuberculosis infection. Biomed Res Int. 2017;2017:8796760.DOI: 10.1155/2017/8796760.
204. Groschel MI, Sayes F, Shin SJ, Frigui W, Pawlik A, Orgeur M, et al. Recombinant BCG expressing ESX-1 of Mycobacterium marinum combines low virulence with cytosolic immune signaling and improved TB protection. Cell Rep. 2017;18(11):2752-2765.DOI: 10.1016/j.celrep.2017.02.057.
205. McNab FW, Ewbank J, Howes A, Moreira-Teixeira L, Martirosyan A, Ghilardi N, et al. Type I IFN induces IL-10 production in an IL-27-independent manner and blocks responsiveness to IFN-gamma for production of IL-12 and bacterial killing in Mycobacterium tuberculosis-infected macrophages. J Immunol. 2014;193(7):3600-3612.DOI: 10.4049/jimmunol.1401088.
206. Howes A, Taubert C, Blankley S, Spink N, Wu X, Graham CM, et al. Differential production of type I IFN determines the reciprocal levels of IL-10 and proinflammatory cytokines produced by C57BL/6 and BALB/c macrophages. J Immunol. 2016;197(7):2838-2853.DOI: 10.4049/jimmunol.1501923.
TOOLS
Similar articles