Next Article in Journal
Antibiotic Use for Sepsis in Hospitalized Neonates in Botswana: Factors Associated with Guideline-Divergent Prescribing
Next Article in Special Issue
Differential Regulation of TFEB-Induced Autophagy during Mtb Infection and Starvation
Previous Article in Journal
Impacts of Surface Characteristics and Dew Point on the Blue-Light (BL405) Inactivation of Viruses
Previous Article in Special Issue
Evaluation of the Toxic Activity of the Graphene Oxide in the Ex Vivo Model of Human PBMC Infection with Mycobacterium tuberculosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Possible Mechanisms of Lymphopenia in Severe Tuberculosis

Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
*
Author to whom correspondence should be addressed.
Microorganisms 2023, 11(11), 2640; https://doi.org/10.3390/microorganisms11112640
Submission received: 14 September 2023 / Revised: 20 October 2023 / Accepted: 24 October 2023 / Published: 27 October 2023
(This article belongs to the Special Issue Immunometabolism in Mycobacterium tuberculosis (M.tb) Infection)

Abstract

:
Tuberculosis (TB) is a chronic infectious disease caused by Mycobacterium tuberculosis (M. tuberculosis). In lymphopenia, T cells are typically characterized by progressive loss and a decrease in their count results. Lymphopenia can hinder immune responses and lead to systemic immunosuppression, which is strongly associated with mortality. Lymphopenia is a significant immunological abnormality in the majority of patients with severe and advanced TB, and its severity is linked to disease outcomes. However, the underlying mechanism remains unclear. Currently, the research on the pathogenesis of lymphopenia during M. tuberculosis infection mainly focuses on how it affects lymphocyte production, survival, or tissue redistribution. This includes impairing hematopoiesis, inhibiting T-cell proliferation, and inducing lymphocyte apoptosis. In this study, we have compiled the latest research on the possible mechanisms that may cause lymphopenia during M. tuberculosis infection. Lymphopenia may have serious consequences in severe TB patients. Additionally, we discuss in detail potential intervention strategies to prevent lymphopenia, which could help understand TB immunopathogenesis and achieve the goal of preventing and treating severe TB.

1. Introduction

Tuberculosis (TB) is a major infectious disease caused by Mycobacterium tuberculosis (M. tuberculosis) [1]. TB is an ancient disease that originated in 17th–18th century Europe and is commonly referred to as the “white plague”, with an almost 100% infection rate and a mortality rate of 25% [2]. However, with the development of anti-TB drugs, improved sanitation, and better living conditions, the incidence and mortality of TB have significantly decreased. However, since the 1990s, due to the emergence of drug-resistant TB, the use of immunosuppressants, poverty, and population mobility, TB has resurged. According to the World Health Organization (WHO) Global TB Report 2022, about a quarter of the world’s population is infected with M. tuberculosis, there are about 10.6 million new TB cases worldwide each year, and 1.6 million people die from TB [3]. China is a high-TB-burden country, with the number of TB cases second only to India and Indonesia [3]. Therefore, TB remains a global public health problem.
Antigen-specific T/B lymphocytes play a critical role in the host response to M. tuberculosis infection. M. tuberculosis infection activates the body’s protective Th1-type cell-mediated immune responses by secreting cytokines such as IFN-γ and IL-2, which further activate the macrophage system to effectively control and eradicate M. tuberculosis [4,5]. Although Th1 cells play a major role in controlling intracellular pathogen infection, Th2 cells and CD8+ T cells are also important in combating M. tuberculosis infection. Th2 cells mainly release IL-4 and IL-10, participate in the activation and proliferation of B cells, and assist in the regulation of humoral immunity [6]. Upon infection, CD8+ T cells are also activated and differentiate into cytotoxic T lymphocytes (CTLs), which dissolve macrophages that have lost immune activity by releasing perforin and granulysin, eventually eliminating M. tuberculosis [7].
Severe forms of TB include military TB, multidrug-resistant tuberculosis (MDR-TB), and TB meningitis [8]. Some severe TB patients even need to be admitted to the intensive care unit (ICU) [9]. Severe TB can cause various complications, including severe weight loss, anemia, and lymphopenia, ultimately leading to death [10,11,12]. The mortality rates for these severe forms range from 15.5% to 65.9% [13,14]. Lymphopenia is a common sign in clinical patients with advanced and severe TB, but its underlying causes are still unclear. Therefore, it is necessary to investigate new mechanisms of lymphopenia to explore potential therapeutic targets.

2. Lymphopenia Is Common in TB Patients

The cellular immune response, which is mediated by T lymphocytes, plays a crucial role in anti-TB immunity. To understand the immune response following M. tuberculosis infection, it is important to examine the phenotypes and function of T lymphocytes in human peripheral blood [15]. T cell counts are known to change in response to M. tuberculosis infection. Severe TB patients typically experience a decrease in the number of CD4+ T cells [16] along with an increase in the expression of immune checkpoint molecules and a decrease in function [17]. Additionally, both CD4+ and CD8+ T cell counts are decreased in active TB patients [15,18]. Lymphopenia is defined as lymphocyte percentage (the percentage of lymphocyte count to leukocyte count) < 20% or absolute lymphocyte count < 109 cells/L [10,19]. Specifically, lymphopenia was found in 46% of untreated pulmonary TB patients [20] and 76.92% of patients with miliary TB [10], revealing that lymphopenia may be a prominent feature of severe TB infection [21]. More recently, CD4+ T cell lymphopenia has emerged as a significant risk factor in TB-infected patients [22]. Therefore, lymphopenia is a common occurrence in TB patients.
Furthermore, the degree of lymphopenia is associated with disease severity [23,24]. The more severe the disease, the lower the number of CD4+ T cell counts. The counts of leukocytes and the ratios of lymphocytes and monocytes during TB therapy are used clinically to evaluate the efficacy of treatment [25]. However, the pathogenesis of CD4+ T cell lymphopenia still lacks in-depth research, and further studies are needed to investigate the mechanisms of lymphopenia during severe M. tuberculosis infection and the possible treatment implications for these patients.

3. The Possible Mechanisms That Cause Lymphopenia

The various mechanisms mentioned here may work together and overlap in some instances, leading to lymphopenia in TB patients. The potential mechanisms of M. tuberculosis-induced lymphopenia are discussed below (Figure 1). However, further in vitro and in vivo studies are required to determine the importance of each component in the occurrence of lymphopenia.

3.1. Macrophages Inhibit Lymphocyte Proliferation

M. tuberculosis is an intracellular pathogen that resides predominantly within macrophages, which serve as the first line of host defense against bacterial infections. After M. tuberculosis infects the body, macrophages recognize the corresponding pathogen-associated molecular patterns (PAMPs) through pattern recognition receptors (PRRs) such as Toll-like receptors (TLRs) on the surface, thereby activating macrophages. After a series of signal transductions, activated macrophages generate a large number of cytokines, which further activate T cells to trigger an adaptive immune response and eradicate pathogens. In addition, IFN-γ has a central role in host defense against M. tuberculosis by activating macrophages [26]. However, activated macrophages can also inhibit T cell proliferation through Stat6-dependent PD-L2 expression [27].
The immunomodulatory properties of alveolar macrophages are relatively selective. Alveolar macrophages selectively inhibit the proliferation of T cells but permit T cell activation and effector functions such as cytokine secretion within the lungs [28,29]. Upham et al. demonstrated that human alveolar macrophages can inhibit T-cell proliferation in vitro [30]. Since IL-2 interacting with its specific receptor can trigger T cell proliferation, the mechanism may be that alveolar macrophages block the protein tyrosine’s phosphorylation related to IL-2 receptors, interfering with the normal function of IL-2 receptors, and ultimately inhibiting T cell proliferation [28].
The cytokines secreted by macrophages can also inhibit the proliferation of T cells and affect the host’s immunity against M. tuberculosis. M. tuberculosis can induce macrophages, T cells, and dendritic cells to secrete cytokines, such as tumor necrosis factor-α (TNF-α), which plays a pivotal role in T-cell-mediated adaptive immunity [31]. In addition, TNF-α is also a negative regulator that can inhibit T cell proliferation and reduce the T cell activation, thereby regulating the immune response [28,32].

3.2. M. tuberculosis-Induced Apoptosis of T Lymphocyte

Lymphopenia is also driven by lymphocyte apoptosis [33]. Lymphocyte apoptosis, an important immune regulatory mechanism, is linked to cell-mediated effector dysfunction in infectious diseases [34]. The increased apoptosis of CD4+ T lymphocytes in peripheral blood is related to lymphopenia. The apoptosis of Th1 lymphocytes leads to disease progression, while the apoptosis of macrophages favors disease protection [35]. T-cell apoptosis can significantly reduce the number of cells, thereby weakening the body’s anti-TB immune response, leading to the occurrence and development of TB [36]. The apoptosis rate of CD4+ T cells in peripheral blood from patients with sputum-positive pulmonary TB is significantly higher than that of patients with sputum-negative pulmonary TB and healthy controls. Moreover, immunodeficiency in patients with acutely progressive TB is associated with the apoptosis of immunocompetent cells, and the main clinical marker of apoptosis is the significant reduction in lymphocytes (4–10%) [37]. Virulent Mycobacterium avium (M. avium) infection also causes progressive severe lymphopenia due to increased apoptosis rates [38]. The apoptosis of CD4+ T cells and CD8+ T cells during M. avium infection is dependent on Fas/Fas ligand (FasL) and Bcl-2-sensitive pathways, respectively [39]. The intervention of certain factors that cause apoptosis is expected to bring new hope and breakthroughs in TB therapy.
M. tuberculosis antigens can directly cause T lymphocyte apoptosis. Studies have demonstrated that M. tuberculosis heat shock protein (HSP)-70, 65, and 16 could induce the apoptosis of peripheral blood monocytes and CD4+ and CD8+ lymphocytes in TB patients. The apoptosis in TB may be associated with the levels of TNF-α and NO, which are controlled by SLC11A1 (formerly NRAMP) [40]. The purified protein derivative of M. tuberculosis (Mtb-PPD) can induce T cell apoptosis by exploiting the Fas/FasL system [41]. Furthermore, M. tuberculosis antigen MPT64 could induce the apoptosis of EL4 lymphoma cells in a dose-dependent manner, which may be related to the elevated levels of IL-4, transforming growth factor-β (TGF-β), and Fas [42]. When CD4+ and γδ T cells from TB patients are cultured with M. tuberculosis, T cell apoptosis increases [36]. It has also been shown that the H37Rv strain causes a progressive increase in apoptotic Th1 lymphocytes mediated by Fas/FasL interaction [35]. Besides that, macrophages infected with M. tuberculosis may induce specific and non-specific T-cell apoptosis by nitric oxide (NO) and TNF-α [43].
The alteration of cytokines in TB patients is linked to lymphocyte apoptosis. A clinical study has shown a positive correlation between the apoptotic rate of CD4+ T cells in TB patients and the secretion levels of IFN-γ, IL-4, TGF-β, and TNF-α [44]. IFN-γ is thought to be the cause of T cell death in mycobacterial infections [45]. IFN-γ can stimulate macrophages to produce NO, which induces apoptosis in T cells, thus IFN-γ causes apoptosis of activated CD4+ T cells during mycobacterial infection by activating macrophages and NO [45]. IFN-γ-dependent apoptosis is observed in T cells. Some studies have also demonstrated that Fas and cleaved caspase 8 are involved in IFN-γ-mediated apoptosis [46]. Meanwhile, TNF-α signaling is also an important part of T cell-induced apoptosis by recruiting FADD and caspase-8 to the receptor complex II [47]. The response of peripheral blood lymphocytes from healthy donors to sonicated M. tuberculosis antigens shows an increase in IL-4 gene expression and apoptosis, while inhibiting IL-4 expression could reduce TNF-α-mediated lymphocyte apoptosis [34]. The mechanism may be that M. tuberculosis-activated lymphocytes induce IL-4 expression, thereby promoting the CD30+ population, which reduces TNF receptor-associated factor 2 (TRAF2) expression and enhances TNF-α-mediated apoptosis in lymphocytes [34]. These data suggest that the reduction of peripheral blood lymphocytes in TB patients may be mediated by cytokines, but further studies are needed to understand the mechanism.
In addition, upregulated perforin/granzyme expression may also be associated with the increased apoptosis of T cells in TB patients by synergizing with other genes, including caspase-3 and caspase-10 [48]. Effector CD8+ T cells from patients with severe TB also overexpress a variety of cytotoxic genes, like GZMA, GZMB, etc. Likewise, significantly increased expression of caspase-3 is detected in severe TB patients [21]. Moreover, it is reported that the pro-apoptotic gene X-linked inhibitor of apoptosis-associated factor 1 (XAF-1) [49] cooperates with caspase-3, TP53, and BCL2L11 to induce cell apoptosis. TNF can activate XAF1, which acts as an alternative pathway for TNF-associated apoptosis [50]. XAF1 also promotes TP53-related apoptosis by post-translational modification [21]. Furthermore, the immune checkpoint molecule Tim-3/Galectin-9 axis plays a critical role in inhibiting Th1 cell immunity and can lead to the apoptosis of Th1 cells by activating caspase-1 [51,52]. Using anti-Tim-3 antibodies to blockade Tim-3 could decrease the severity of the inflammatory responses and reduce CD8+ T cell apoptosis [53].

3.3. M. tuberculosis-Mediated Bone Marrow Hematopoietic Dysfunction

Chronic infections significantly affect the expansion and differentiation of bone marrow hematopoietic cells [54,55], which further influence the production of lymphocytes. Hematopoietic stem cells (HSCs), a population of stem cells, can give rise to all lineages of blood cells [56,57]. Infection-specific changes in the hematopoietic system may limit or promote the production of specific lineages, thereby centrally affecting the immune response. For instance, M. tuberculosis reprograms HSCs to limit myelopoiesis and impairs the development of protective trained immunity to M. tuberculosis infection in a type I interferon (IFN-I)-dependent manner [58]. Repeated infections of mice with M. avium depleted the pool of HSCs and made mice develop pancytopenia [59]. Moreover, Mycobacterium bovis Bacillus Calmette–Guérin (BCG) can train HSCs to enhance myelopoiesis at the expense of lymphopoiesis via the activation of IFN-II (IFN-γ) signaling and generate protective innate immunity against M. tuberculosis infection [60,61]. However, severe TB causes hematopoietic dysfunction and immune exhaustion due to continuous M. tuberculosis stimulation [10,62].
Some inflammatory cytokines are critical for regulating hematopoiesis and determining the differentiation fate of bone marrow hematopoietic cells. Among them, IFN-γ impairs the proliferation and reconstitution of HSCs and affects the differentiation from long-term HSCs (LT-HSCs) to short-term HSCs (ST-HSCs) [63,64]. Furthermore, chronic infection with M. avium and persistent M. tuberculosis antigen stimulation deplete lymphoid commitment due to persistent IFN-γ stimulation and induce lymphopenia [10]. IFN-γ promotes myelopoiesis by upregulating the expression of Batf2 [59]. Similar to IFN-γ, TNF-α also promotes myeloid differentiation [65]. Unlike IFN-γ, IL-7 is a crucial cytokine in lymphopoiesis and T cell homeostasis [66,67], which is essential for normal thymopoiesis [68], inhibiting apoptosis and promoting T cell survival [69]. IL-7 can rescue B-lymphopoiesis by driving the proliferation and expansion of B-cell progenitors [70,71]. Interestingly, a molecule called chromodomain helicase DNA-binding protein 4 (CHD4) is required for IL-7 receptor signaling and is also essential for transcriptional repression and lineage commitment during B lymphopoiesis [72]. Similarly, IL-2 can also enhance lymphoid differentiation, thereby reversing impaired hemopoiesis and lymphopenia [10,12]. As hematopoietic cells are the source of immune cells, the occurrence of lymphopenia is largely attributed to hematopoietic dysfunction.
Chemokines also play a critical role in the process of hematopoiesis, thereby affecting the production of T cells [73]. During chronic infection, the expression of chemokine CXCL10 can be induced by innate immune cell cytokines IFN-α/β as well as adaptive immune stimuli IFN-γ. CXCL10 is upregulated in TB patients and could inhibit the development of hematopoietic precursors [74], which may contribute to TB-associated lymphopenia. In the hematopoietic system, CXCR4 binds to its chemokine ligand, CXCL12, to mediate leukocyte transport, distribution, survival, activation, and proliferation. CXCR4 has been reported to help thymic progenitors homing from bone marrow to the thymus and emerges as a promising proposed TB target [75].
Post-translational modifications of proteins, particularly ubiquitination, play a significant role in the hematopoietic process and are closely associated with lymphopenia. Most E3 ubiquitin ligases have a negative impact on the development and function of HSCs. The E3 ubiquitin ligase c-Cbl is a novel negative regulator of HSC development and function. When c-Cbl is absent, it increases the number of HSCs and enhances the capacity to rebuild the hematopoietic system [76]. C-Cbl deficiency can lead to a decline in lymphocyte development and function in aging individuals [77]. Similarly, another E3 ubiquitin ligase Itch can also negatively regulate the development and function of HSCs, and its deficiency can also result in hematopoietic abnormalities [78]. The E3 ubiquitin ligase Fbw7 plays a crucial role in regulating HSC quiescence and self-renewal. Fbw7 is expressed in quiescent HSCs. Once it loses its self-renewal ability, its expression is downregulated. Deletion of Fbw7 in bone marrow stem cells and progenitors results in decreased hematopoietic progenitors and impaired self-renewal ability [79]. Recently, studies have demonstrated that the absence of ubiquitin editing enzyme A20 can lead to the overexpression of IFN-γ, leading to abnormal differentiation of lymphoid cells and promoting the differentiation into the myeloid lineage, ultimately causing lymphopenia [80].

3.4. T Cell Exhaustion

In many chronic infections and cancer, persistent antigen stimulation can lead to T cell exhaustion, which is a state of T cell dysfunction characterized by progressive loss of effector function and memory T cell potential as well as high and sustained expression of inhibitory immune checkpoint receptors [81,82]. In severe TB, the levels of IFN-γ and TNF from activated CD4+ and CD8+ T cells are lower, while the expression of multiple inhibitory molecules, including PD-1, Tim-3, etc., are high. These data suggest that T cells become dysfunctional in severe TB and fail to control M. tuberculosis infection [21]. In addition, T cell exhaustion is accompanied by alteration in transcription factor expression [83,84], and exhausted T cells have distinct transcriptional profiles, which include a subset of transcription factors, like positive regulatory domain 1 gene (PRDM1)/B lymphocyte-induced maturation protein-1 (Blimp-1) [85], basic leucine zipper ATF-like transcription factor (BATF) [86], and nuclear factor of activated T cells (NFAT) [87]. As such, significantly elevated expression of PRDM1 is found in severe TB patients compared with healthy donors [21]. Furthermore, the cells that lack CD28 expression are considered severely exhausted, and CD28-exhausted CD4+ T cells are linked to lymphocyte counts and poor prognosis [88].

3.5. Tissue Redistribution of Lymphocytes

M. tuberculosis infection could induce lymphocyte trafficking to secondary lymphoid tissues [89]. The decrease in blood lymphocytes may be due to T cells redistributing to the lungs and gastrointestinal tract. Chemokines and chemotactic cytokines can stimulate the migration of T lymphocytes to the lung [90]. TNF coordinates the induction of chemokines to recruit leukocytes to form granulomas in TB [91]. It has been suggested that the rapid increase in lymphocyte count during recovery is not a result of newly produced lymphocytes in the thymus but rather the result of lymphocyte recirculation between organs and peripheral blood.
Cell migration pathways contribute to the reduction of lymphocytes. Migration-associated genes like CXCL10, CXCL8, CCL3, etc., are upregulated in CD8+ T cells from severe TB patients, indicating that T cells display a significant activation of migration. Consistent with the results above, NK cells from severe TB patients also show high migration and death, which may be associated with the reduction in the NK population [21].

3.6. Aging-Related Immunosuppression

The aging population faces a significantly increased risk of infection and cancer. Immunosenescence can affect innate immunity and adaptive immunity. Aging is associated with a decrease in the number of hematopoietic precursors and the degeneration of the thymus, both of which lead to the reduced ability of newborn T cells. Therefore, the functional capacity of T cells is affected by aging, with a limited available T cell pool and a reduced number of naïve T cells [92,93]. Thymus activity persists into adulthood, and as the thymus shrinks, its function gradually decreases with age. The underlying mechanisms causing thymic atrophy still need to be investigated. Additionally, several age-related comorbidities like obesity, diabetes, malnutrition, chronic respiratory diseases, and cancer can increase the risk of developing active TB [94].
Various agents for bone marrow transplantation and aging research represent promising strategies for combating T-cell deficiencies in the aging population. Preclinical and clinical studies on the effects of keratinocyte growth factor, sex steroid ablation, and the cytokine IL-7 on T cell reconstitution suggest that these strategies can be utilized to alleviate the effects of T cell deficiencies and enhance T cell reconstitution in the elderly [95].

3.7. Activation-Induced Cell Death in Lymphocytes

Activation-induced cell death (AICD) in T lymphocytes has been recognized as one of the mechanisms of lymphopenia. T cells undergo AICD during contraction, which is triggered by the death receptor and results in apoptosis [96,97]. AICD is mediated by the Fas/FasL pathway [98]. Fas and FasL are a pair of receptors/ligands that are critically involved in lymphocyte homeostasis. The interaction between Fas and FasL triggers apoptosis in Fas-expressing cells [99]. The levels of Fas in PBMC are associated with the severity of TB [100]. The FasL surface and mRNA expression in peripheral blood mononuclear cells (PBMCs) from TB patients are higher compared to controls. Additionally, neutralizing FasL can eliminate T-cell apoptosis [44]. Furthermore, the loss of γδT cells is a consequence of M. tuberculosis antigen-mediated AICD through the interaction between Fas and FasL [101].
Except for FasL, AICD is also mediated by other receptor/ligand pathways such as TNF and TNF-receptor-related apoptosis-inducing ligand (TRAIL) [102]. In a mouse model of M. avium infection, both CD4+ and CD8+ T cells suffered a higher rate of apoptosis. Among them, low expression of Bcl-2 and excessive expression of TNF-α, TGF-β, and FasL are involved in the apoptosis of CD4+ T cells [44]. This suggests that there may be differences in the apoptotic pathways of CD4+ and CD8+ T cells, which are closely related to the expression levels of TNF receptors and Bcl-2 [39].

3.8. Myeloid-Deprived Suppressor Cells

Myeloid-deprived suppressor cells (MDSCs) are an immature population of myeloid cells that enter a pathological activation state and can suppress T-cell responses [103]. The levels of monocytic MDSCs are increased significantly in the peripheral blood of patients with severe TB [21,104]. In mycobacterial infections, such as M. tuberculosis, Mycobacterium smegmatis (M. smeg), and BCG, MDSC is induced [105]. Due to MDSC immunosuppressive activity, T cell functions like cytokine production, T cell activation, and modulation of T cell trafficking are inhibited [106], consequently decreasing T cell counts. Human MDSC upregulates PD-L1 during in vitro mycobacterial infection and utilizes this checkpoint molecule to limit T cell proliferation [107]. MDSC may promote TB reactivation by exacerbating the immunosuppressive effects of therapies such as anti-TNF drugs, and the deficiency of TNF-α is associated with T cell immunosuppression [108].

4. Potential Immunotherapies

As discussed above, substantial evidence supports a significant correlation between M. tuberculosis-induced lymphopenia and lymphocyte production, function, and survival. Therefore, targeting the specific TB immune profiles, such as reducing apoptosis or enhancing lymphopoiesis, is a promising treatment strategy to rescue lymphopenia and also compensate for the lymphocyte counts in severe TB. Therapy that enhances lymphopoiesis includes IL-7/IL-2 therapy. To inhibit apoptosis, strategies such as TNF-α or TGF-β antibodies can be employed (Table 1).

4.1. Reducing Apoptosis

Targeting the inhibition of apoptosis may prevent lymphopenia and compensate for the low number of lymphocytes in severe TB patients. FasL-mediated apoptosis plays a critical role in lymphocyte homeostasis and results in lymphopenia. Therefore, inhibiting the interaction between Fas and FasL could prevent the depletion of lymphocytes [109]. Fas–FasL interaction also induces T-cell apoptosis in AICD. Furthermore, the apoptosis of CD4+ T cells is closely related to the low expression of Bcl-2 and excessive expression of TNF-α, TGF-β, and FasL [44]. Thus, defective Fas or Bcl-2 overexpression protects T cells from TNF receptor-related apoptosis. Moreover, the use of IFN-γ, TNF-α, or TGF-β antibodies can reduce spontaneous or M. tuberculosis-induced T cell apoptosis [34,44,46]. Biologically, taurine is a semi-essential amino acid synthesized from methionine and cysteine. Taurine attenuates CD3/IL-2-induced T cell apoptosis in AICD and augments the immunotherapeutic potential of IL-2 [110]. Several strategies involved in inhibiting lymphocyte egress from secondary lymphoid tissues may also reverse lymphopenia.
However, the drawbacks of these treatment strategies cannot be ignored. For instance, a subset of cancers originating from the lymphatic system displays defective Fas/FasL-mediated apoptosis, rendering them resistant to chemotherapy [111]. Bcl-2 is commonly overexpressed in various types of cancers, especially in lymphoma. Therefore, treatment with Bcl-2 overexpression may be linked to therapeutic resistance and poor prognosis [112]. Considering that TNF-α is a cytokine involved in immune responses, blocking TNF-α may pose a unique risk of latent tuberculosis infection (LTBI) reactivation and new infections due to impaired effector immune responses [113]. Additionally, TGF-β plays a crucial role in the expansion of Th17 cells against mycobacterial infection [114]. Consequently, the extent to which the TGF-β blockade interferes with immune response in reducing apoptosis therapy is still unclear and requires further investigation.
Furthermore, there exists another view about the impact of T cell apoptosis, which suggests that T cell apoptosis might serve as a host regulatory mechanism to restrict excessive inflammation and prevent tissue damage, rather than an M. tuberculosis-induced immune deviation [115]. In line with this, certain studies have shown that mesenchymal stem cells (MSCs) can induce activated T cell apoptosis by producing NO, thereby weakening the delayed-type hypersensitivity (DTH) response [116]. Hence, additional research is required to ascertain the relevance of apoptosis in the development of TB and to uncover the underlying mechanisms.

4.2. Targeting Lymphocyte Generation

Some inflammatory cytokines can regulate the homeostasis and differentiation of hematopoietic cells [117]. Cytokines IL-7, IL-2, and stem cell factor (SCF) are proposed therapies for enhancing lymphopoiesis [118]. Infection with M. tuberculosis could impact the differentiation of hematopoietic cells, leading to a decrease in lymphocyte production [10,60]. In addition, the differentiation of hematopoietic cells is tightly regulated by lineage-determining transcription factors [119]. Among the transcription factors, interferon regulatory factor-8 (IRF8) and Batf2 tend to promote myeloid differentiation [59], while GATA2 and NOTCH1 are critical for T cell commitment [12,120]. Moreover, interferon regulatory factor-4 (IRF4) or paired box-5 (Pax5) have been associated with lymphopoiesis [121,122]. Therefore, upregulating the expressions of transcription factors, such as NOTCH1 and GATA2, involved in T-cell lineage commitment and down-regulating IFR8 and Batf2 participation in myeloid differentiation are seen as possibly useful strategies for enhancing lymphopoiesis and reversing lymphopenia.
Maintaining the homeostasis of hemopoiesis is crucial for establishing an effective immune response against TB [12]. When homeostasis is disrupted, it can impact the differentiation of HSCs. The objective of enhancing lymphopoiesis is to restore a steady state of hematopoiesis. Insufficient production of lymphocytes can result in a poor therapeutic effect, while excessive production can lead to side effects such as lymphocytosis. Therefore, immunotherapy needs to investigate the timing and dosage to achieve the optimal therapeutic outcome.
Table 1. Promising therapeutic strategies for severe TB.
Table 1. Promising therapeutic strategies for severe TB.
StrategyMajor ChangesIntervention MethodsIntervention Mechanism
Reducing apoptosisFasL ↑Fas antibody [109]Attenuates CD3/IL-2-induced T cell apoptosis
Bcl-2 ↓Bcl-2 overexpression [44]Protects T cells from TNF receptor-related apoptosis
IFN-γ, IL-4, TNF-α, or TGF-β ↑IFN-γ antibody [46], IL-4 antibody [34], TNF-α antibody [34], TGF-β antibody [44]Reduces the cytokines-mediated lymphocyte apoptosis
Targeting lymphocyte generationIL-7, IL-2, SCF, NOTCH1, GATA2, IRF4, Pax5 ↓IL-7, IL-2, SCF [118], NOTCH1 enhancer, GATA2 enhancer [12,120], IRF4 enhancer, and Pax5 enhancer [121,122]Promote lymphopoiesis
IFR8, Batf2 ↑IFR8 and Batf2 inhibitors [59]Reduces myeloid differentiation to promote lymphopoiesis
Notes: The arrow ↑ indicates up-regulated gene expression, while the arrow ↓ indicates down-regulated gene expression.

5. Conclusions and Perspectives

Tuberculosis is a chronic wasting disease, and its immune response level changes with the development of the disease. Peripheral blood lymphocytes are important cells involved in the body’s anti-TB immunity. Their quantity and function can be used to evaluate the body’s immune function and determine whether the host is infected by M. tuberculosis, which is closely related to the clinical severity of the disease. Lymphopenia often occurs in TB patients. Multiple mechanisms may be involved in the occurrence of lymphopenia by affecting lymphocyte production, function, and survival. The main mechanisms include the inhibition of lymphocyte proliferation by macrophages, inducing T lymphocyte apoptosis, and bone marrow hematopoietic dysfunction.
Lymphopenia may have serious consequences in severe TB patients. Due to the crucial role of M. tuberculosis-specific T cell-mediated immune responses in bacterial elimination, lymphopenia can cause bacterial persistence, replication, and tissue damage. Additionally, destroying a large number of lymphocytes can lead to general immunosuppression in TB patients. In accordance with COVID-19, lymphopenia also plays a key role in TB pathogenesis. In lymphopenia, the number of Treg cells (as the main regulator of immune responses, exhibiting immunosuppressive function) also decreases, thereby strengthening excessive inflammatory responses. Collectively, lymphopenia may lead to delayed elimination of M. tuberculosis, unrestricted Th1 response, uncontrolled cytokine production, and even cytokine storms, ultimately leading to multiple organ failure and death. Therefore, eliminating the factors that cause lymphopenia may be an effective therapeutic strategy for severe TB. However, further investigation on the pathogenesis of lymphopenia is still necessary to understand the interaction mechanism between M. tuberculosis and hosts. Conducting relevant studies will offer new insights for the prevention and treatment of severe TB.

Author Contributions

Conceptualization, Q.Z. and F.L.; writing—original draft preparation, F.L. and D.C.; writing—review and editing, F.L.; visualization, Y.D.; supervision, F.L.; project administration, F.L.; funding acquisition, F.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Gansu Science and Technology Project of China, grant number 23JRRA1100; the Youth Science Fund Project of the National Natural Science Foundation of China, grant number 82001675; and Fundamental Research Funds for the Central Universities of the Lanzhou University, grant number lzujbky-2023-17.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Acknowledgments

The authors would like to thank the editor and anonymous reviewers for positive and constructive comments and suggestions.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

TB: Tuberculosis; M. tuberculosis: Mycobacterium tuberculosis; MDR-TB: multidrug-resistant tuberculosis; HIV: human immunodeficiency virus; PAMP: pathogen-associated molecular pattern; PRR: pattern recognition receptor; TLR: Toll-like receptor; M. avium: Mycobacterium avium; TGF-β: transforming growth factor-β; NO: nitric oxide; XAF-1: X-linked inhibitor of apoptosis-associated factor 1; HSCs: hematopoietic stem cells; IFN-I: I interferon; BCG: Mycobacterium bovis Bacillus Calmette–Guérin; CHD4: Chromodomain helicase DNA-binding protein 4; PRDM1: positive regulatory domain 1; Blimp-1: B lymphocyte-induced maturation protein-1; BATF: basic leucine zipper ATF-like transcription factor; NFAT: nuclear factor of activated T cells; AICD: activation-induced cell death; PBMC: peripheral blood mononuclear cell; TRAIL: TNF-receptor-related apoptosis-inducing ligand; MDSC: myeloid-deprived suppressor cell; M. smeg: Mycobacterium smegmatis; MSCs: mesenchymal stem cells; DTH: delayed-type hypersensitivity; SCF: stem cell factor; IRF8: interferon regulatory factor-8; IRF4: interferon regulatory factor-4; Pax5: paired box-5.

References

  1. Furin, J.; Cox, H.; Pai, M. Tuberculosis. Lancet 2019, 393, 1642–1656. [Google Scholar] [CrossRef] [PubMed]
  2. Zumla, A. The white plague returns to London—With a vengeance. Lancet 2011, 377, 10–11. [Google Scholar] [CrossRef] [PubMed]
  3. Bagcchi, S. WHO’s Global Tuberculosis Report 2022. Lancet Microbe 2023, 4, e20. [Google Scholar] [CrossRef]
  4. Flynn, J.L.; Chan, J. Immune cell interactions in tuberculosis. Cell 2022, 185, 4682–4702. [Google Scholar] [CrossRef] [PubMed]
  5. Zhuang, L.; Ye, Z.; Li, L.; Yang, L.; Gong, W. Next-generation TB vaccines: Progress, challenges, and prospects. Vaccines 2023, 11, 1304. [Google Scholar] [CrossRef]
  6. Prezzemolo, T.; Guggino, G.; La Manna, M.P.; Di Liberto, D.; Dieli, F.; Caccamo, N. Functional signatures of human CD4 and CD8 T cell responses to Mycobacterium tuberculosis. Front. Immunol. 2014, 5, 180. [Google Scholar] [CrossRef]
  7. Wang, Y.; Sun, M.; He, M.; Cui, H.; Zhang, J.; Shi, L.; Wang, W.; Xu, W.; Gao, B.; Ding, J. Weak binder for MHC molecule is a potent Mycobacterium tuberculosis-specific CTL epitope in the context of HLA-A24 allele. Microb. Pathog. 2012, 53, 162–167. [Google Scholar] [CrossRef]
  8. Ruiz-Bedoya, C.A.; Mota, F.; Tucker, E.W.; Mahmud, F.J.; Reyes-Mantilla, M.I.; Erice, C.; Bahr, M.; Flavahan, K.; de Jesus, P.; Kim, J.; et al. High-dose rifampin improves bactericidal activity without increased intracerebral inflammation in animal models of tuberculous meningitis. J. Clin. Investig. 2022, 132, e155851. [Google Scholar] [CrossRef]
  9. Erbes, R.; Oettel, K.; Raffenberg, M.; Mauch, H.; Schmidt-Ioanas, M.; Lode, H. Characteristics and outcome of patients with active pulmonary tuberculosis requiring intensive care. Eur. Respir. J. 2006, 27, 1223–1228. [Google Scholar] [CrossRef]
  10. Li, F.; Ma, Y.; Li, X.; Zhang, D.; Han, J.; Tan, D.; Mi, Y.; Yang, X.; Wang, J.; Zhu, B. Severe persistent mycobacteria antigen stimulation causes lymphopenia through impairing hematopoiesis. Front. Cell. Infect. Microbiol. 2023, 13, 1079774. [Google Scholar] [CrossRef]
  11. Achi, H.V.; Ahui, B.J.; Anon, J.C.; Kouassi, B.A.; Dje-Bi, H.; Kininlman, H. Pancytopenia: A severe complication of miliary tuberculosis. Rev. Des Mal. Respir. 2013, 30, 33–37. [Google Scholar] [CrossRef]
  12. Li, F.; Liu, X.; Niu, H.; Lv, W.; Han, X.; Zhang, Y.; Zhu, B. Persistent stimulation with Mycobacterium tuberculosis antigen impairs the proliferation and transcriptional program of hematopoietic cells in bone marrow. Mol. Immunol. 2019, 112, 115–122. [Google Scholar] [CrossRef]
  13. Duro, R.P.; Figueiredo Dias, P.; Ferreira, A.A.; Xerinda, S.M.; Lima Alves, C.; Sarmento, A.C.; Dos Santos, L.C. Severe tuberculosis requiring intensive care: A descriptive analysis. Crit. Care Res. Pract. 2017, 2017, 9535463. [Google Scholar] [CrossRef] [PubMed]
  14. Davis, A.G.; Rohlwink, U.K.; Proust, A.; Figaji, A.A.; Wilkinson, R.J. The pathogenesis of tuberculous meningitis. J. Leukoc. Biol. 2019, 105, 267–280. [Google Scholar] [CrossRef] [PubMed]
  15. Rodrigues, D.S.; Medeiros, E.A.; Weckx, L.Y.; Bonnez, W.; Salomão, R.; Kallas, E.G. Immunophenotypic characterization of peripheral T lymphocytes in Mycobacterium tuberculosis infection and disease. Clin. Exp. Immunol. 2002, 128, 149–154. [Google Scholar] [CrossRef] [PubMed]
  16. Imperiale, B.R.; García, A.; Minotti, A.; González Montaner, P.; Moracho, L.; Morcillo, N.S.; Palmero, D.J.; Sasiain, M.D.C.; de la Barrera, S. Th22 response induced by Mycobacterium tuberculosis strains is closely related to severity of pulmonary lesions and bacillary load in patients with multi-drug-resistant tuberculosis. Clin. Exp. Immunol. 2021, 203, 267–280. [Google Scholar] [CrossRef]
  17. Liu, X.; Li, F.; Niu, H.; Ma, L.; Chen, J.; Zhang, Y.; Peng, L.; Gan, C.; Ma, X.; Zhu, B. IL-2 restores T-cell dysfunction induced by persistent Mycobacterium tuberculosis antigen stimulation. Front. Immunol. 2019, 10, 2350. [Google Scholar] [CrossRef]
  18. Li, K.; Ran, R.; Jiang, Z.; Fan, C.; Li, T.; Yin, Z. Changes in T-lymphocyte subsets and risk factors in human immunodeficiency virus-negative patients with active tuberculosis. Infection 2020, 48, 585–595. [Google Scholar] [CrossRef]
  19. Sheu, T.T.; Chiang, B.L. Lymphopenia, lymphopenia-induced proliferation, and autoimmunity. Int. J. Mol. Sci. 2021, 22, 4152. [Google Scholar] [CrossRef]
  20. Deveci, F.; Akbulut, H.H.; Celik, I.; Muz, M.H.; Ilhan, F. Lymphocyte subpopulations in pulmonary tuberculosis patients. Mediat. Inflamm. 2006, 2006, 89070. [Google Scholar] [CrossRef]
  21. Wang, Y.; Sun, Q.; Zhang, Y.; Li, X.; Liang, Q.; Guo, R.; Zhang, L.; Han, X.; Wang, J.; Shao, L.; et al. Systemic immune dysregulation in severe tuberculosis patients revealed by a single-cell transcriptome atlas. J. Infect. 2023, 86, 421–438. [Google Scholar] [CrossRef] [PubMed]
  22. Lao, M.; Chen, D.; Wu, X.; Chen, H.; Qiu, Q.; Yang, X.; Zhan, Z. Active tuberculosis in patients with systemic lupus erythematosus from Southern China: A retrospective study. Clin. Rheumatol. 2019, 38, 535–543. [Google Scholar] [CrossRef] [PubMed]
  23. Panteleev, A.V.; Nikitina, I.Y.; Burmistrova, I.A.; Kosmiadi, G.A.; Radaeva, T.V.; Amansahedov, R.B.; Sadikov, P.V.; Serdyuk, Y.V.; Larionova, E.E.; Bagdasarian, T.R.; et al. Severe tuberculosis in humans correlates best with neutrophil abundance and lymphocyte deficiency and does not correlate with antigen-specific CD4 T-cell response. Front. Immunol. 2017, 8, 963. [Google Scholar] [CrossRef] [PubMed]
  24. Jones, B.E.; Oo, M.M.; Taikwel, E.K.; Qian, D.; Kumar, A.; Maslow, E.R.; Barnes, P.F. CD4 cell counts in human immunodeficiency virus-negative patients with tuberculosis. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 1997, 24, 988–991. [Google Scholar] [CrossRef]
  25. Chedid, C.; Kokhreidze, E.; Tukvadze, N.; Banu, S.; Uddin, M.K.M.; Biswas, S.; Russomando, G.; Acosta, C.C.D.; Arenas, R.; Ranaivomanana, P.P.; et al. Association of baseline white blood cell counts with tuberculosis treatment outcome: A prospective multicentered cohort study. Int. J. Infect. Dis. IJID Off. Publ. Int. Soc. Infect. Dis. 2020, 100, 199–206. [Google Scholar] [CrossRef]
  26. Shanmuganathan, G.; Orujyan, D.; Narinyan, W.; Poladian, N.; Dhama, S.; Parthasarathy, A.; Ha, A.; Tran, D.; Velpuri, P.; Nguyen, K.H.; et al. Role of interferons in Mycobacterium tuberculosis infection. Clin. Pract. 2022, 12, 788–796. [Google Scholar] [CrossRef]
  27. Huber, S.; Hoffmann, R.; Muskens, F.; Voehringer, D. Alternatively activated macrophages inhibit T-cell proliferation by Stat6-dependent expression of PD-L2. Blood 2010, 116, 3311–3320. [Google Scholar] [CrossRef]
  28. Upham, J.W.; Strickland, D.H.; Robinson, B.W.; Holt, P.G. Selective inhibition of T cell proliferation but not expression of effector function by human alveolar macrophages. Thorax 1997, 52, 786–795. [Google Scholar] [CrossRef]
  29. Upham, J.W.; Strickland, D.H.; Bilyk, N.; Robinson, B.W.; Holt, P.G. Alveolar macrophages from humans and rodents selectively inhibit T-cell proliferation but permit T-cell activation and cytokine secretion. Immunology 1995, 84, 142–147. [Google Scholar]
  30. Grange, J.M. Inhibition of T cell proliferation by human alveolar macrophages. Thorax 1998, 53, 437. [Google Scholar] [CrossRef]
  31. Holbrook, J.; Lara-Reyna, S.; Jarosz-Griffiths, H.; McDermott, M. Tumour necrosis factor signalling in health and disease. F1000Research 2019, 8, 111. [Google Scholar] [CrossRef]
  32. Zganiacz, A.; Santosuosso, M.; Wang, J.; Yang, T.; Chen, L.; Anzulovic, M.; Alexander, S.; Gicquel, B.; Wan, Y.; Bramson, J.; et al. TNF-α is a critical negative regulator of type 1 immune activation during intracellular bacterial infection. J. Clin. Investig. 2004, 113, 401–413. [Google Scholar] [CrossRef] [PubMed]
  33. Silva, E.E.; Skon-Hegg, C.; Badovinac, V.P.; Griffith, T.S. The calm after the storm: Implications of sepsis immunoparalysis on host immunity. J. Immunol. 2023, 211, 711–719. [Google Scholar] [CrossRef] [PubMed]
  34. Seah, G.T.; Rook, G.A. Il-4 influences apoptosis of mycobacterium-reactive lymphocytes in the presence of TNF-alpha. J. Immunol. 2001, 167, 1230–1237. [Google Scholar] [CrossRef]
  35. Ríos-Barrera, V.A.; Campos-Peña, V.; Aguilar-León, D.; Lascurain, L.R.; Meraz-Ríos, M.A.; Moreno, J.; Figueroa-Granados, V.; Hernández-Pando, R. Macrophage and T lymphocyte apoptosis during experimental pulmonary tuberculosis: Their relationship to mycobacterial virulence. Eur. J. Immunol. 2006, 36, 345–353. [Google Scholar] [CrossRef]
  36. Hirsch, C.S.; Toossi, Z.; Vanham, G.; Johnson, J.L.; Peters, P.; Okwera, A.; Mugerwa, R.; Mugyenyi, P.; Ellner, J.J. Apoptosis and T cell hyporesponsiveness in pulmonary tuberculosis. J. Infect. Dis. 1999, 179, 945–953. [Google Scholar] [CrossRef]
  37. Khomenko, A.G.; Koval’chuk, L.V.; Mishin, V.I.; Pavliuk, A.S.; Veselova, A.V. Increased apoptosis of immunocompetent cells as a possible mechanism in the development of immunodeficiency in patients with acutely progressive tuberculosis. Probl. Tuberk. 1996, 6, 6–10. [Google Scholar]
  38. Flórido, M.; Pearl, J.E.; Solache, A.; Borges, M.; Haynes, L.; Cooper, A.M.; Appelberg, R. Gamma interferon-induced T-cell loss in virulent Mycobacterium avium infection. Infect. Immun. 2005, 73, 3577–3586. [Google Scholar] [CrossRef]
  39. Zhong, J.; Gilbertson, B.; Cheers, C. Apoptosis of CD4+ and CD8+ T cells during experimental infection with Mycobacterium avium is controlled by Fas/FasL and Bcl-2-sensitive pathways, respectively. Immunol. Cell Biol. 2003, 81, 480–486. [Google Scholar] [CrossRef]
  40. Dubaniewicz, A. Mycobacterial heat shock proteins in sarcoidosis and tuberculosis. Int. J. Mol. Sci. 2023, 24, 5084. [Google Scholar] [CrossRef]
  41. Soruri, A.; Schweyer, S.; Radzun, H.J.; Fayyazi, A. Mycobacterial antigens induce apoptosis in human purified protein derivative-specific alphabeta T lymphocytes in a concentration-dependent manner. Immunology 2002, 105, 222–230. [Google Scholar] [CrossRef] [PubMed]
  42. Wang, Q.; Tang, Y.; Liu, S. Effect of MPT64 from Mycobacterium tuberculosis on EL4 lymphocytes. Mil. Med. J. S. Chin. 2016, 30, 77–80. [Google Scholar] [CrossRef]
  43. Sharma, S.; Sharma, M.; Bose, M. Mycobacterium tuberculosis infection of human monocyte-derived macrophages leads to apoptosis of T cells. Immunol. Cell Biol. 2009, 87, 226–234. [Google Scholar] [CrossRef] [PubMed]
  44. Hirsch, C.S.; Johnson, J.L.; Okwera, A.; Kanost, R.A.; Wu, M.; Peters, P.; Muhumuza, M.; Mayanja-Kizza, H.; Mugerwa, R.D.; Mugyenyi, P.; et al. Mechanisms of apoptosis of T-cells in human tuberculosis. J. Clin. Immunol. 2005, 25, 353–364. [Google Scholar] [CrossRef] [PubMed]
  45. Dalton, D.K.; Haynes, L.; Chu, C.Q.; Swain, S.L.; Wittmer, S. Interferon-γ eliminates responding CD4 T cells during mycobacterial infection by inducing apoptosis of activated CD4 T cells. J. Exp. Med. 2000, 192, 117–122. [Google Scholar] [CrossRef]
  46. Stenger, E.O.; Rosborough, B.R.; Mathews, L.R.; Ma, H.; Mapara, M.Y.; Thomson, A.W.; Turnquist, H.R. IL-12hi rapamycin-conditioned dendritic cells mediate IFN-γ-dependent apoptosis of alloreactive CD4+ T cells in vitro and reduce lethal graft-versus-host disease. Biol. Blood Marrow Transplant. J. Am. Soc. Blood Marrow Transplant. 2014, 20, 192–201. [Google Scholar] [CrossRef]
  47. Micheau, O.; Tschopp, J. Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell 2003, 114, 181–190. [Google Scholar] [CrossRef]
  48. Elmore, S. Apoptosis: A review of programmed cell death. Toxicol. Pathol. 2007, 35, 495–516. [Google Scholar] [CrossRef]
  49. Liston, P.; Fong, W.G.; Kelly, N.L.; Toji, S.; Miyazaki, T.; Conte, D.; Tamai, K.; Craig, C.G.; McBurney, M.W.; Korneluk, R.G. Identification of XAF1 as an antagonist of XIAP anti-Caspase activity. Nat. Cell Biol. 2001, 3, 128–133. [Google Scholar] [CrossRef]
  50. Jeong, S.I.; Kim, J.W.; Ko, K.P.; Ryu, B.K.; Lee, M.G.; Kim, H.J.; Chi, S.G. XAF1 forms a positive feedback loop with IRF-1 to drive apoptotic stress response and suppress tumorigenesis. Cell Death Dis. 2018, 9, 806. [Google Scholar] [CrossRef]
  51. Kang, J.; Wei, Z.F.; Li, M.X.; Wang, J.H. Modulatory effect of Tim-3/Galectin-9 axis on T-cell-mediated immunity in pulmonary tuberculosis. J. Biosci. 2020, 45, 60. [Google Scholar] [CrossRef]
  52. Schlichtner, S.; Meyer, N.H.; Yasinska, I.M.; Aliu, N.; Berger, S.M.; Gibbs, B.F.; Fasler-Kan, E.; Sumbayev, V.V. Functional role of galectin-9 in directing human innate immune reactions to Gram-negative bacteria and T cell apoptosis. Int. Immunopharmacol. 2021, 100, 108155. [Google Scholar] [CrossRef] [PubMed]
  53. Liu, S.; Wang, C.; Jiang, Z.; Deng, X.; Bo, L. Tim-3 blockade decreases the apoptosis of CD8+ T cells and reduces the severity of sepsis in mice. J. Surg. Res. 2022, 279, 8–16. [Google Scholar] [CrossRef] [PubMed]
  54. Takizawa, H.; Boettcher, S.; Manz, M.G. Demand-adapted regulation of early hematopoiesis in infection and inflammation. Blood 2012, 119, 2991–3002. [Google Scholar] [CrossRef]
  55. Pal, R.; Mukhopadhyay, S. PPE2 protein of Mycobacterium tuberculosis affects myeloid hematopoiesis in mice. Immunobiology 2021, 226, 152051. [Google Scholar] [CrossRef] [PubMed]
  56. Ribatti, D.; d’Amati, A. Hematopoiesis and mast cell development. Int. J. Mol. Sci. 2023, 24, 10679. [Google Scholar] [CrossRef]
  57. Xia, J.; Liu, M.; Zhu, C.; Liu, S.; Ai, L.; Ma, D.; Zhu, P.; Wang, L.; Liu, F. Activation of lineage competence in hemogenic endothelium precedes the formation of hematopoietic stem cell heterogeneity. Cell Res. 2023, 33, 448–463. [Google Scholar] [CrossRef]
  58. Khan, N.; Downey, J.; Sanz, J.; Kaufmann, E.; Blankenhaus, B.; Pacis, A.; Pernet, E.; Ahmed, E.; Cardoso, S.; Nijnik, A.; et al. M. tuberculosis reprograms hematopoietic stem cells to limit myelopoiesis and impair trained immunity. Cell 2020, 183, 752–770.e22. [Google Scholar] [CrossRef]
  59. Matatall, K.A.; Jeong, M.; Chen, S.; Sun, D.; Chen, F.; Mo, Q.; Kimmel, M.; King, K.Y. Chronic infection depletes hematopoietic stem cells through stress-induced terminal differentiation. Cell Rep. 2016, 17, 2584–2595. [Google Scholar] [CrossRef]
  60. Kaufmann, E.; Sanz, J.; Dunn, J.L.; Khan, N.; Mendonça, L.E.; Pacis, A.; Tzelepis, F.; Pernet, E.; Dumaine, A.; Grenier, J.C.; et al. BCG educates hematopoietic stem cells to generate protective innate immunity against tuberculosis. Cell 2018, 172, 176–190.e19. [Google Scholar] [CrossRef]
  61. Cirovic, B.; de Bree, L.C.J.; Groh, L.; Blok, B.A.; Chan, J.; van der Velden, W.; Bremmers, M.E.J.; van Crevel, R.; Händler, K.; Picelli, S.; et al. BCG vaccination in humans elicits trained immunity via the hematopoietic progenitor compartment. Cell Host Microbe 2020, 28, 322–334.e5. [Google Scholar] [CrossRef] [PubMed]
  62. Khan, N.; Vidyarthi, A.; Amir, M.; Mushtaq, K.; Agrewala, J.N. T-cell exhaustion in tuberculosis: Pitfalls and prospects. Crit. Rev. Microbiol. 2017, 43, 133–141. [Google Scholar] [CrossRef] [PubMed]
  63. Qin, Y.; Fang, K.; Lu, N.; Hu, Y.; Tian, Z.; Zhang, C. Interferon gamma inhibits the differentiation of mouse adult liver and bone marrow hematopoietic stem cells by inhibiting the activation of notch signaling. Stem Cell Res. Ther. 2019, 10, 210. [Google Scholar] [CrossRef] [PubMed]
  64. de Bruin, A.M.; Voermans, C.; Nolte, M.A. Impact of interferon-γ on hematopoiesis. Blood 2014, 124, 2479–2486. [Google Scholar] [CrossRef]
  65. Yamashita, M.; Passegué, E. TNF-α coordinates hematopoietic stem cell survival and myeloid regeneration. Cell Stem Cell 2019, 25, 357–372.e7. [Google Scholar] [CrossRef]
  66. Fry, T.J.; Connick, E.; Falloon, J.; Lederman, M.M.; Liewehr, D.J.; Spritzler, J.; Steinberg, S.M.; Wood, L.V.; Yarchoan, R.; Zuckerman, J.; et al. A potential role for interleukin-7 in T-cell homeostasis. Blood 2001, 97, 2983–2990. [Google Scholar] [CrossRef]
  67. Fry, T.J.; Mackall, C.L. The many faces of IL-7: From lymphopoiesis to peripheral T cell maintenance. J. Immunol. 2005, 174, 6571–6576. [Google Scholar] [CrossRef]
  68. Murray, R.; Suda, T.; Wrighton, N.; Lee, F.; Zlotnik, A. IL-7 is a growth and maintenance factor for mature and immature thymocyte subsets. Int. Immunol. 1989, 1, 526–531. [Google Scholar] [CrossRef]
  69. Chen, D.; Tang, T.X.; Deng, H.; Yang, X.P.; Tang, Z.H. Interleukin-7 biology and its effects on immune cells: Mediator of generation, differentiation, survival, and homeostasis. Front. Immunol. 2021, 12, 747324. [Google Scholar] [CrossRef]
  70. Blin-Wakkach, C.; Wakkach, A.; Quincey, D.; Carle, G.F. Interleukin-7 partially rescues B-lymphopoiesis in osteopetrotic oc/oc mice through the engagement of B220+ CD11b+ progenitors. Exp. Hematol. 2006, 34, 851–859. [Google Scholar] [CrossRef]
  71. Kaiser, F.M.P.; Janowska, I.; Menafra, R.; de Gier, M.; Korzhenevich, J.; Pico-Knijnenburg, I.; Khatri, I.; Schulz, A.S.; Kuijpers, T.W.; Lankester, A.C.; et al. IL-7 receptor signaling drives human B-cell progenitor differentiation and expansion. Blood 2023, 142, 113. [Google Scholar] [CrossRef]
  72. Arends, T.; Dege, C.; Bortnick, A.; Danhorn, T.; Knapp, J.R.; Jia, H.; Harmacek, L.; Fleenor, C.J.; Straign, D.; Walton, K.; et al. CHD4 is essential for transcriptional repression and lineage progression in B lymphopoiesis. Proc. Natl. Acad. Sci. USA 2019, 116, 10927–10936. [Google Scholar] [CrossRef]
  73. Franczak, S.; Ulrich, H.; Ratajczak, M.Z. Hematopoietic stem cells on the crossroad between purinergic signaling and innate immunity. Purinergic Signal. 2023. [Google Scholar] [CrossRef]
  74. Sarris, A.H.; Broxmeyer, H.E.; Wirthmueller, U.; Karasavvas, N.; Cooper, S.; Lu, L.; Krueger, J.; Ravetch, J.V. Human interferon-inducible protein 10: Expression and purification of recombinant protein demonstrate inhibition of early human hematopoietic progenitors. J. Exp. Med. 1993, 178, 1127–1132. [Google Scholar] [CrossRef]
  75. Irham, L.M.; Adikusuma, W.; Perwitasari, D.A. Genomic variants-driven drug repurposing for tuberculosis by utilizing the established bioinformatic-based approach. Biochem. Biophys. Rep. 2022, 32, 101334. [Google Scholar] [CrossRef]
  76. Rathinam, C.; Thien, C.B.; Langdon, W.Y.; Gu, H.; Flavell, R.A. The E3 ubiquitin ligase c-Cbl restricts development and functions of hematopoietic stem cells. Genes Dev. 2008, 22, 992–997. [Google Scholar] [CrossRef]
  77. Rathinam, C.; Flavell, R.A. c-Cbl deficiency leads to diminished lymphocyte development and functions in an age-dependent manner. Proc. Natl. Acad. Sci. USA 2010, 107, 8316–8321. [Google Scholar] [CrossRef]
  78. Rathinam, C.; Matesic, L.E.; Flavell, R.A. The E3 ligase Itch is a negative regulator of the homeostasis and function of hematopoietic stem cells. Nat. Immunol. 2011, 12, 399–407. [Google Scholar] [CrossRef]
  79. Thompson, B.J.; Jankovic, V.; Gao, J.; Buonamici, S.; Vest, A.; Lee, J.M.; Zavadil, J.; Nimer, S.D.; Aifantis, I. Control of hematopoietic stem cell quiescence by the E3 ubiquitin ligase Fbw7. J. Exp. Med. 2008, 205, 1395–1408. [Google Scholar] [CrossRef]
  80. Nakagawa, M.M.; Rathinam, C.V. A20 deficiency in hematopoietic stem cells causes lymphopenia and myeloproliferation due to elevated Interferon-γ signals. Sci. Rep. 2019, 9, 12658. [Google Scholar] [CrossRef]
  81. Wherry, E.J.; Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 2015, 15, 486–499. [Google Scholar] [CrossRef]
  82. Belk, J.A.; Daniel, B.; Satpathy, A.T. Epigenetic regulation of T cell exhaustion. Nat. Immunol. 2022, 23, 848–860. [Google Scholar] [CrossRef]
  83. McLane, L.M.; Abdel-Hakeem, M.S.; Wherry, E.J. CD8 T cell exhaustion during chronic viral infection and cancer. Annu. Rev. Immunol. 2019, 37, 457–495. [Google Scholar] [CrossRef]
  84. Tian, W.; Qin, G.; Jia, M.; Li, W.; Cai, W.; Wang, H.; Zhao, Y.; Bao, X.; Wei, W.; Zhang, Y.; et al. Hierarchical transcriptional network governing heterogeneous T cell exhaustion and its implications for immune checkpoint blockade. Front. Immunol. 2023, 14, 1198551. [Google Scholar] [CrossRef]
  85. Scharping, N.E.; Rivadeneira, D.B.; Menk, A.V.; Vignali, P.D.A.; Ford, B.R.; Rittenhouse, N.L.; Peralta, R.; Wang, Y.; Wang, Y.; DePeaux, K.; et al. Mitochondrial stress induced by continuous stimulation under hypoxia rapidly drives T cell exhaustion. Nat. Immunol. 2021, 22, 205–215. [Google Scholar] [CrossRef]
  86. Utzschneider, D.T.; Gabriel, S.S.; Chisanga, D.; Gloury, R.; Gubser, P.M.; Vasanthakumar, A.; Shi, W.; Kallies, A. Early precursor T cells establish and propagate T cell exhaustion in chronic infection. Nat. Immunol. 2020, 21, 1256–1266. [Google Scholar] [CrossRef]
  87. Seo, W.; Jerin, C.; Nishikawa, H. Transcriptional regulatory network for the establishment of CD8+ T cell exhaustion. Exp. Mol. Med. 2021, 53, 202–209. [Google Scholar] [CrossRef]
  88. Huang, Y.; Zheng, H.; Zhu, Y.; Hong, Y.; Zha, J.; Lin, Z.; Li, Z.; Wang, C.; Fang, Z.; Yu, X.; et al. Loss of CD28 expression associates with severe T-cell exhaustion in acute myeloid leukemia. Front. Immunol. 2023, 14, 1139517. [Google Scholar] [CrossRef]
  89. Goetzl, E.J.; Gräler, M.H. Sphingosine 1-phosphate and its type 1 G protein-coupled receptor: Trophic support and functional regulation of T lymphocytes. J. Leukoc. Biol. 2004, 76, 30–35. [Google Scholar] [CrossRef]
  90. Algood, H.M.; Lin, P.L.; Yankura, D.; Jones, A.; Chan, J.; Flynn, J.L. TNF influences chemokine expression of macrophages in vitro and that of CD11b+ cells in vivo during Mycobacterium tuberculosis infection. J. Immunol. 2004, 172, 6846–6857. [Google Scholar] [CrossRef]
  91. Roach, D.R.; Bean, A.G.; Demangel, C.; France, M.P.; Briscoe, H.; Britton, W.J. TNF regulates chemokine induction essential for cell recruitment, granuloma formation, and clearance of mycobacterial infection. J. Immunol. 2002, 168, 4620–4627. [Google Scholar] [CrossRef] [PubMed]
  92. Akbar, A.N.; Fletcher, J.M. Memory T cell homeostasis and senescence during aging. Curr. Opin. Immunol. 2005, 17, 480–485. [Google Scholar] [CrossRef] [PubMed]
  93. Olmo-Fontánez, A.M.; Turner, J. Tuberculosis in an aging world. Pathogens 2022, 11, 1101. [Google Scholar] [CrossRef] [PubMed]
  94. Byng-Maddick, R.; Noursadeghi, M. Does tuberculosis threaten our ageing populations? BMC Infect. Dis. 2016, 16, 119. [Google Scholar] [CrossRef]
  95. Holland, A.M.; van den Brink, M.R. Rejuvenation of the aging T cell compartment. Curr. Opin. Immunol. 2009, 21, 454–459. [Google Scholar] [CrossRef]
  96. Green, D.R.; Droin, N.; Pinkoski, M. Activation-induced cell death in T cells. Immunol. Rev. 2003, 193, 70–81. [Google Scholar] [CrossRef]
  97. Shin, S.Y.; Kim, M.W.; Cho, K.H.; Nguyen, L.K. Coupled feedback regulation of nuclear factor of activated T-cells (NFAT) modulates activation-induced cell death of T cells. Sci. Rep. 2019, 9, 10637. [Google Scholar] [CrossRef]
  98. Masuda, A.; Isobe, Y.; Sugimoto, K.; Yoshimori, M.; Arai, A.; Komatsu, N. Efficient recruitment of c-FLIP(L) to the death-inducing signaling complex leads to Fas resistance in natural killer-cell lymphoma. Cancer Sci. 2020, 111, 807–816. [Google Scholar] [CrossRef]
  99. Ju, S.T.; Matsui, K.; Ozdemirli, M. Molecular and cellular mechanisms regulating T and B cell apoptosis through Fas/FasL interaction. Int. Rev. Immunol. 1999, 18, 485–513. [Google Scholar] [CrossRef]
  100. Roodgar, M.; Ross, C.T.; Tarara, R.; Lowenstine, L.; Dandekar, S.; Smith, D.G. Gene expression and TB pathogenesis in rhesus macaques: TR4, CD40, CD40L, FAS (CD95), and TNF are host genetic markers in peripheral blood mononuclear cells that are associated with severity of TB lesions. Infect. Genet. Evol. J. Mol. Epidemiol. Evol. Genet. Infect. Dis. 2015, 36, 396–409. [Google Scholar] [CrossRef]
  101. Li, B.; Bassiri, H.; Rossman, M.D.; Kramer, P.; Eyuboglu, A.F.; Torres, M.; Sada, E.; Imir, T.; Carding, S.R. Involvement of the Fas/Fas ligand pathway in activation-induced cell death of mycobacteria-reactive human gamma delta T cells: A mechanism for the loss of gamma delta T cells in patients with pulmonary tuberculosis. J. Immunol. 1998, 161, 1558–1567. [Google Scholar] [CrossRef] [PubMed]
  102. Maher, S.; Toomey, D.; Condron, C.; Bouchier-Hayes, D. Activation-induced cell death: The controversial role of Fas and Fas ligand in immune privilege and tumour counterattack. Immunol. Cell Biol. 2002, 80, 131–137. [Google Scholar] [CrossRef] [PubMed]
  103. Gabrilovich, D.I.; Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009, 9, 162–174. [Google Scholar] [CrossRef] [PubMed]
  104. Magcwebeba, T.; Dorhoi, A.; du Plessis, N. The emerging role of myeloid-derived suppressor cells in tuberculosis. Front. Immunol. 2019, 10, 917. [Google Scholar] [CrossRef]
  105. Martino, A.; Badell, E.; Abadie, V.; Balloy, V.; Chignard, M.; Mistou, M.Y.; Combadière, B.; Combadière, C.; Winter, N. Mycobacterium bovis bacillus Calmette-Guérin vaccination mobilizes innate myeloid-derived suppressor cells restraining in vivo T cell priming via IL-1R-dependent nitric oxide production. J. Immunol. 2010, 184, 2038–2047. [Google Scholar] [CrossRef]
  106. du Plessis, N.; Loebenberg, L.; Kriel, M.; von Groote-Bidlingmaier, F.; Ribechini, E.; Loxton, A.G.; van Helden, P.D.; Lutz, M.B.; Walzl, G. Increased frequency of myeloid-derived suppressor cells during active tuberculosis and after recent Mycobacterium tuberculosis infection suppresses T-cell function. Am. J. Respir. Crit. Care Med. 2013, 188, 724–732. [Google Scholar] [CrossRef]
  107. Agrawal, N.; Streata, I.; Pei, G.; Weiner, J.; Kotze, L.; Bandermann, S.; Lozza, L.; Walzl, G.; du Plessis, N.; Ioana, M.; et al. Human monocytic suppressive cells promote replication of Mycobacterium tuberculosis and alter stability of in vitro generated granulomas. Front. Immunol. 2018, 9, 2417. [Google Scholar] [CrossRef]
  108. Clay, H.; Volkman, H.E.; Ramakrishnan, L. Tumor necrosis factor signaling mediates resistance to mycobacteria by inhibiting bacterial growth and macrophage death. Immunity 2008, 29, 283–294. [Google Scholar] [CrossRef]
  109. Hao, Z.; Hampel, B.; Yagita, H.; Rajewsky, K. T cell-specific ablation of Fas leads to Fas ligand-mediated lymphocyte depletion and inflammatory pulmonary fibrosis. J. Exp. Med. 2004, 199, 1355–1365. [Google Scholar] [CrossRef]
  110. Maher, S.G.; Condron, C.E.; Bouchier-Hayes, D.J.; Toomey, D.M. Taurine attenuates CD3/interleukin-2-induced T cell apoptosis in an in vitro model of activation-induced cell death (AICD). Clin. Exp. Immunol. 2005, 139, 279–286. [Google Scholar] [CrossRef]
  111. Mustafa, N.; Mitxelena, J.; Infante, A.; Zenarruzabeitia, O.; Eriz, A.; Iglesias-Ara, A.; Zubiaga, A.M. E2f2 attenuates apoptosis of activated T lymphocytes and protects from immune-mediated injury through repression of Fas and FasL. Int. J. Mol. Sci. 2021, 23, 311. [Google Scholar] [CrossRef] [PubMed]
  112. Kapoor, I.; Bodo, J.; Hill, B.T.; Hsi, E.D.; Almasan, A. Targeting BCL-2 in B-cell malignancies and overcoming therapeutic resistance. Cell Death Dis. 2020, 11, 941. [Google Scholar] [CrossRef] [PubMed]
  113. Godfrey, M.S.; Friedman, L.N. Tuberculosis and biologic therapies: Anti-tumor necrosis factor-α and beyond. Clin. Chest Med. 2019, 40, 721–739. [Google Scholar] [CrossRef] [PubMed]
  114. Basile, J.I.; Kviatcovsky, D.; Romero, M.M.; Balboa, L.; Monteserin, J.; Ritacco, V.; Lopez, B.; Sabio y García, C.; García, A.; Vescovo, M.; et al. Mycobacterium tuberculosis multi-drug-resistant strain M induces IL-17(+) IFNγ(-) CD4(+) T cell expansion through an IL-23 and TGF-β-dependent mechanism in patients with MDR-TB tuberculosis. Clin. Exp. Immunol. 2017, 187, 160–173. [Google Scholar] [CrossRef]
  115. Rajavelu, P.; Pokkali, S.; Umashankar, P.; Bhatt, K.; Narayanan, P.R.; Salgame, P.; Das, S.D. Comparative evaluation of cytokines, T-cell apoptosis, and costimulatory molecule expression in tuberculous and nontuberculous pleurisy. Clin. Transl. Sci. 2008, 1, 209–214. [Google Scholar] [CrossRef]
  116. Lim, J.H.; Kim, J.S.; Yoon, I.H.; Shin, J.S.; Nam, H.Y.; Yang, S.H.; Kim, S.J.; Park, C.G. Immunomodulation of delayed-type hypersensitivity responses by mesenchymal stem cells is associated with bystander T cell apoptosis in the draining lymph node. J. Immunol. 2010, 185, 4022–4029. [Google Scholar] [CrossRef]
  117. Hajishengallis, G.; Li, X.; Chavakis, T. Immunometabolic control of hematopoiesis. Mol. Asp. Med. 2021, 77, 100923. [Google Scholar] [CrossRef]
  118. Harada, T.; Tsuboi, I.; Hino, H.; Yuda, M.; Hirabayashi, Y.; Hirai, S.; Aizawa, S. Age-related exacerbation of hematopoietic organ damage induced by systemic hyper-inflammation in senescence-accelerated mice. Sci. Rep. 2021, 11, 23250. [Google Scholar] [CrossRef]
  119. Paul, F.; Arkin, Y.; Giladi, A.; Jaitin, D.A.; Kenigsberg, E.; Keren-Shaul, H.; Winter, D.; Lara-Astiaso, D.; Gury, M.; Weiner, A.; et al. Transcriptional heterogeneity and lineage commitment in myeloid progenitors. Cell 2015, 163, 1663–1677. [Google Scholar] [CrossRef]
  120. Doulatov, S.; Notta, F.; Laurenti, E.; Dick, J.E. Hematopoiesis: A human perspective. Cell Stem Cell 2012, 10, 120–136. [Google Scholar] [CrossRef]
  121. Ottens, K.; Satterthwaite, A.B. IRF4 has a unique role in early B cell development and acts prior to CD21 expression to control marginal zone B cell numbers. Front. Immunol. 2021, 12, 779085. [Google Scholar] [CrossRef] [PubMed]
  122. Wang, T.; Chen, Z.; Li, C.; Zhang, W.; Huang, W.; Xue, J.; Wang, J.; Li, S. PAX5 and circ1857 affected DLBCL progression and B-cell proliferation through regulating GINS1. Cancer Sci. 2023, 114, 3203–3215. [Google Scholar] [CrossRef]
Figure 1. The possible mechanisms that may lead to lymphopenia in severe TB patients. Lymphopenia is a common sign in severe TB patients and is affected by various factors, ranging from impaired production to function and survival. The detailed mechanisms consist of macrophages inhibiting lymphocyte proliferation, M. tuberculosis-induced T lymphocyte apoptosis, hematopoietic dysfunction, activation-induced cell death, T cell exhaustion, tissue redistribution of lymphocytes, and aging-related immunosuppression.
Figure 1. The possible mechanisms that may lead to lymphopenia in severe TB patients. Lymphopenia is a common sign in severe TB patients and is affected by various factors, ranging from impaired production to function and survival. The detailed mechanisms consist of macrophages inhibiting lymphocyte proliferation, M. tuberculosis-induced T lymphocyte apoptosis, hematopoietic dysfunction, activation-induced cell death, T cell exhaustion, tissue redistribution of lymphocytes, and aging-related immunosuppression.
Microorganisms 11 02640 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Li, F.; Chen, D.; Zeng, Q.; Du, Y. Possible Mechanisms of Lymphopenia in Severe Tuberculosis. Microorganisms 2023, 11, 2640. https://doi.org/10.3390/microorganisms11112640

AMA Style

Li F, Chen D, Zeng Q, Du Y. Possible Mechanisms of Lymphopenia in Severe Tuberculosis. Microorganisms. 2023; 11(11):2640. https://doi.org/10.3390/microorganisms11112640

Chicago/Turabian Style

Li, Fei, Dandan Chen, Qingqing Zeng, and Yunjie Du. 2023. "Possible Mechanisms of Lymphopenia in Severe Tuberculosis" Microorganisms 11, no. 11: 2640. https://doi.org/10.3390/microorganisms11112640

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop