Next Article in Journal
Differential Regulation of DC Function, Adaptive Immunity, and MyD88 Dependence by MF59 and AS03-like Adjuvants
Previous Article in Journal
Determinants of Parental Intention to Vaccinate Young Adolescent Girls against the Human Papillomavirus in Taiwan: An Online Survey Study
Previous Article in Special Issue
Development and Evaluation of an Immunoinformatics-Based Multi-Peptide Vaccine against Acinetobacter baumannii Infection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Live Attenuated Vaccines against Tuberculosis: Targeting the Disruption of Genes Encoding the Secretory Proteins of Mycobacteria

by
Raja Veerapandian
1,
Shrikanth S. Gadad
2,
Chinnaswamy Jagannath
3,* and
Subramanian Dhandayuthapani
1,*
1
Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
2
Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
3
Department of Pathology and Genomic Medicine, Houston Methodist Research Institute & Weill Cornell Medical College, Houston, TX 77030, USA
*
Authors to whom correspondence should be addressed.
Vaccines 2024, 12(5), 530; https://doi.org/10.3390/vaccines12050530
Submission received: 8 April 2024 / Revised: 7 May 2024 / Accepted: 8 May 2024 / Published: 12 May 2024
(This article belongs to the Special Issue Novel Vaccines for Infectious Pathogens)

Abstract

:
Tuberculosis (TB), a chronic infectious disease affecting humans, causes over 1.3 million deaths per year throughout the world. The current preventive vaccine BCG provides protection against childhood TB, but it fails to protect against pulmonary TB. Multiple candidates have been evaluated to either replace or boost the efficacy of the BCG vaccine, including subunit protein, DNA, virus vector-based vaccines, etc., most of which provide only short-term immunity. Several live attenuated vaccines derived from Mycobacterium tuberculosis (Mtb) and BCG have also been developed to induce long-term immunity. Since Mtb mediates its virulence through multiple secreted proteins, these proteins have been targeted to produce attenuated but immunogenic vaccines. In this review, we discuss the characteristics and prospects of live attenuated vaccines generated by targeting the disruption of the genes encoding secretory mycobacterial proteins.

1. Introduction

Tuberculosis (TB) is caused by a Gram-positive bacterial pathogen, Mycobacterium tuberculosis (Mtb), which is known for its thick cell wall made up of long-chain fatty acids called mycolic acids [1]. Despite effective control measures, neither the death rate nor the incidence rate of TB has shown any sign of decline in recent years, and they remain at 1.3 million and 10.6 million in 2022 [2]. In addition, a quarter of the world population is estimated to have latent TB infection (LTBI) [3]. This situation is further aggravated by the emergence of multidrug-resistant tuberculosis (MDR-TB) and extensively drug-resistant tuberculosis (XDR-TB) strains, which require second-line drugs and prolonged care. In addition, HIV-TB coinfection contributes to about 12.8% of the total deaths due to TB, because HIV-1 depletes the CD4+ T cells associated with Th1 immune response [2]. The goal of the World Health Organization (WHO) is to reduce the number of new TB cases by 90% in the year 2035, and achieving this goal requires the development of novel drugs and therapeutic agents for the treatment of the disease and, importantly, efficacious preventive and therapeutic vaccines.
Currently, Bacille Calmette–Guerin (BCG) is the only approved vaccine for TB. It was derived from the virulent Mycobacterium bovis, the pathogen that causes TB in cattle, when it was passaged in culture media over two hundred times [4,5]. An estimated four billion doses of BCG have been administered to infants with very few adverse incidences, indicating that it is the safest vaccine available [6]. However, a major weakness of BCG is its variable efficacy (0–80%) in different populations of different ethnicities [5,7]. Several reasons account for this discrepancy, including (a) the geographical location of the populations tested [8], (b) environmental mycobacteria that share antigens with BCG [7], and (c) variation in the antigenic profile of BCG sub-strains due to genetic differences [9]. In addition, it has been proposed that humans exposed to atypical mycobacteria [10] and helminthic infections will have increased Th2 response, which can also reduce the efficacy of BCG vaccination [11,12]. Regardless of these factors, the consensus is that BCG prevents TB meningitis in children but has no effect on the most common pulmonary TB in children and adults [5,7]. A recent meta-review also reiterates that infant vaccination using BCG can prevent tuberculosis in young children but is ineffective in adolescents and adults [13]. This underscores the need for a better primary vaccine and a booster that can induce BCG-induced primary immunity.
The past two decades have witnessed the development of many vaccines against TB, and some of these vaccines have advanced to clinical trials [14]. These include platforms based on protein subunits, viral vectors, recombinant mycobacterial live attenuated vaccines (LAVs), killed whole cell vaccines, and an mRNA-based vaccine [6,15,16,17,18,19,20,21]. Compared to other vaccines that transiently express antigens, LAVs are considered superior to others because they tend to persist for extended periods, encoding antigens and enabling the longer-lasting stimulation of immune cells to induce protective immune responses [22,23]. Further, the antigens produced by LAVs are closer to native antigens, which are properly folded proteins, carbohydrates, and lipids [24,25]. Moreover, these diverse antigens will likely stimulate multiple immune cell populations like subsets of T and B cells and phenotypes such as NK and innate T cells. Another advantage of using LAVs is that their cell wall components can stimulate innate immunity [26,27]. Thus, cell wall lipids such as trehalose dimycolate (TDM) can activate Mincle receptor-inducing trained immunity [28,29], whereas other glycolipids and several pathogen-associated molecular patterns (PAMPs) can trigger pattern recognition receptors (PRR) like TLR-2 and TLR-4 [30]. However, great caution should be exercised when considering lipids as inducers of the immune response because some essential Mtb lipids, like sulfoglycolipids, inhibit the innate immune response [31]. More importantly, the loss of phthiocerol dimycocerosates (PDIMs) and phenolic glycolipids (PGLs) in the BCG Pasteur strain reduces the efficacy of the BCG vaccine, highlighting the importance of PDIMs/PGLs [32]. Another attractive strategy for modifying live attenuated vaccines (LAVs) is the addition of major Mtb proteins to BCG to create recombinant BCG vaccines for better efficacy. However, our review is focused explicitly on Mtb or BCG knockouts as LAVs.
A major approach to derive LAVs against TB depends on a ‘rational deletion of genes’ in the chromosomes of Mtb and BCG [33]. Often, the target genes play a critical role in immune evasion by Mtb. Importantly, there is a strong link between immune evasion mechanisms and the ‘secretory proteins’ of mycobacteria, because many of these seem to be released into the host environment to modulate phagolysosomal (PL) fusion, autophagy, apoptosis, the modulation of cytokines, the intracellular survival of pathogens, and other related antimicrobial pathways [34,35,36,37] (Figure 1, Figure 2 and Figure 3). Coincidentally, many LAVs for TB are based on deleting genes encoding ‘secretory proteins’ or their regulators and transporters (Table 1 and Table 2). In this review, we describe the immunological parameters of LAVs deficient in secreted protein(s) and their efficacy compared with the BCG vaccine, which also secretes several antigenic proteins [38]. We specifically address LAVs deficient in secretory antigens, since other gene knockout mutants have been described by others [15,18,39,40].

2. Secretory Systems of Mycobacteria

To determine the significance of host immune modulation by Mtb, it is imperative to understand the secretory systems of mycobacteria. Bacteria generally have intriguing mechanisms to transport some of their proteins across the cytoplasmic membrane, which are called secretion systems or secretory pathways [41]. They transport proteins that need to be localized in the periplasm, outer membrane, or surface or released in the extracellular environment and into the host cells. The two major secretory pathways in bacteria are the general secretory pathway (or Sec pathway) and the twin-arginine translocation pathway or TAT pathway [41]. Both are highly conserved systems and are present in most bacteria [42,43]. In addition to Sec and TAT pathways, Gram-negative pathogenic bacteria have special pathways to transport virulence factors. There are six such special pathways, which are named type I through type VI secretory systems. Some of these systems, like the type III secretion system in Salmonella species, form microneedles to inject the effector proteins directly into the host cells [41]. The Gram-positive mycobacteria were initially thought to have only the Sec and TAT pathways to secrete proteins. However, a new special secretory system was later identified in mycobacteria [44] and was named the type VII secretion system [45], in line with the previously labeled secretion systems.
In contrast to other species, mycobacteria have two Sec pathways, SecA1 and SecA2 [44]. While transporting proteins through SecA1 requires a signal sequence, no signal sequence is required for transporting proteins through SecA2. The SecA2 pathway in Mtb appears to mainly transport proteins related to pathogenesis, such as SodA, SapM, and PknG [46]. On the other hand, the TAT pathway requires a TAT signal sequence to transport proteins [41]. In Mtb, the TAT pathway transports fewer proteins associated with pathogenesis, including phospholipase A and C [46].
Further, the Type VII secretion system in Mtb and related mycobacteria has five export systems, and they are named ESX-1 through ESX-5 [47,48,49]. Each system has a cluster of genes to encode the proteins, facilitated by the structural proteins required for transportation. The ESX systems recognize the substrates or the proteins to be transported by the YxxxD/E signal (amino acid) motif in their sequence [50]. Among the five ESX systems, ESX-4 seems to be the oldest system, and others appear to have evolved through duplication events [51,52]. Notably, the gene cluster for this system lacks genes encoding PE/PPE proteins, although the other four systems do have genes to encode these proteins.
Nonetheless, the ESX-1 system seems to be the most well-studied and is responsible for the secretion of EsxA (ESAT-6) and EsxB (CFP-10) proteins in Mtb and related pathogens [53,54,55,56]. These proteins are both virulence factors and immunodominant antigens, and coincidentally, their absence leads to an avirulent phenotype in Mtb [55]. ESAT-6 enables the bacteria to lyse the phagosomal membrane with the aid of the chaperone CFP-10, a strategy that is missing in BCG vaccine strains [57]. The ESX-1 system also constitutes the Region of Difference 1 (RD1), deleted in the BCG vaccine strains [54]. Indeed, the attenuation of BCG appears to be due to the absence of the RD1 region in its chromosome, because the complementation of BCG with the RD1 region restores its virulence [54]. Both ESX-1 and ESX-2 systems are intact in Mtb and other related pathogens, but little information is available about its secreted products and role in pathogenesis [58].
Further, ESX-3 and ESX-5 systems participate in the secretion of their proteins, and the latter plays a significant role in immune modulation or inflammasome activation [59,60,61,62,63]. ESX-4 is different in functionality, and unlike other systems, it plays a vital role in conjugation between bacteria [64]. Overall, mycobacteria use Sec, TAT, and ESX systems to transport proteins that regulate virulence and immunogenicity. However, some secreted proteins do not seem to have specific export signatures.
Figure 1. Subversion of phagocytosis and related processes by mycobacterial secreted proteins. Phagosome maturation: pattern recognition receptors (PRRs) of macrophages recognize mycobacteria through mycobacterial pathogen-associated molecular patterns (PAMPs), resulting in the engulfment of bacilli by a phagosome, an organelle derived from the plasma membrane. Phagosomes undergo a series of steps called phagosome maturation to digest the engulfed bacilli and present the antigens to the immune cells. Additionally, phagosomes fuse with early endosomes, late endosomes, and, subsequently, with lysosomes to acquire the materials/properties required for the killing/digestion of the pathogen. However, intracellular mycobacteria like M. tuberculosis (Mtb), M. bovis, M. marinum, and BCG have multiple strategies to protect them against the phagocytic processes. M. marinum secretes PPE38 [65] to block the phagocytosis of the bacilli. Mtb secretes several secretory proteins to inhibit the phagosome maturation process. SapM [34] and PtpB [66] dephosphorylate phosphatidylinositol-3-phosphate (PI3P) to inhibit phagosome maturation, whereas PtpA protein inhibits phagosome acidification by blocking Vacuolar-type ATPase (V-ATPase) [67]. TlyA inhibits Early Endosomal Antigen-1 (EEA1), Ras-related protein 5 (RAB5), and RAB7 recruitment [68]. NdkA inhibits RAB5 and RAB7 [69], while PknG inhibits RAB7L1 [70]. Additionally, Mtb secretes UreC to alkalize the phagosomes [71]. In addition, Mtb perforates the phagosome and escapes to the cytosol using the concerted action of phthiocerol dimycocerosates (PDIM) and ESX-1 system [72]. Host efforts to repair phagosomal rupture were blocked by the EsxH protein [59]. ROS/iNOS: Host NADPH oxidases (NOX) from the cytoplasm and mitochondrial electron transport chain are the primary sources of reactive oxygen species (ROS) production. ROS is blocked by Mtb proteins like Eis, ESAT-6/CFP-10, NuoG, NdkA, PPE2, SodA [73,74,75,76], and inducible nitric oxide synthase (iNOS), while the mediated production of NO is blocked by PtpB, PPE2, PE_PGRS62, PE5, PE15, and PE4 [77,78,79,80]. Epigenetic regulators: Mtb secretes proteins like Rv1988 [81] and Rv2966c [82] to methylate host DNA and proteins like Eis [83] and Rv3423.1 [84] to acetylate host DNA to manipulate the host immune response. Proinflammatory cytokines: Mtb secretes proteins like PtpA, PtpB, ESAT-6/CFP-10, Eis, EchA1, and PknG [77,85,86,87,88,89] to inhibit proinflammatory cytokines. Cell death pathways: Cytosolic escape of the pathogen leads to activation of various cell death pathways like necrosis mediated by Zmp1, CpnT, and PE25:PPE41 [90,91,92] or ferroptosis via PtpA which benefits the pathogen [93]. The cytosolic presence of bacilli DNA or RNA triggers various pathways like apoptosis, autophagy/xenophagy, and pyroptosis, which are detrimental to the pathogen. Mtb secretes NuoG, PtpA, PtpB, NdkA, Rv3654c, Rv3655c, and Rv3033 to block apoptosis [36,75,77,94,95,96]. Proteins like Zmp1, PknF, PtpB, and Rv3364c block inflammasome activation and/or pyroptosis [35,97,98,99]. Autophagy/xenophagy pathways are blocked by proteins like NuoG, Eis, SapM, PE_PGRS20, PE_PGRS47, PPE51, LprE, and PknG [86,100,101,102,103,104,105]; however, these proteins block autophagy indirectly by blocking early/late phagosome proteins. Note: Some live attenuated Mtb or BCG vaccines described in this review lack one or more secretory proteins mentioned above, and they are designated in this figure with rose-colored oval shapes.
Figure 1. Subversion of phagocytosis and related processes by mycobacterial secreted proteins. Phagosome maturation: pattern recognition receptors (PRRs) of macrophages recognize mycobacteria through mycobacterial pathogen-associated molecular patterns (PAMPs), resulting in the engulfment of bacilli by a phagosome, an organelle derived from the plasma membrane. Phagosomes undergo a series of steps called phagosome maturation to digest the engulfed bacilli and present the antigens to the immune cells. Additionally, phagosomes fuse with early endosomes, late endosomes, and, subsequently, with lysosomes to acquire the materials/properties required for the killing/digestion of the pathogen. However, intracellular mycobacteria like M. tuberculosis (Mtb), M. bovis, M. marinum, and BCG have multiple strategies to protect them against the phagocytic processes. M. marinum secretes PPE38 [65] to block the phagocytosis of the bacilli. Mtb secretes several secretory proteins to inhibit the phagosome maturation process. SapM [34] and PtpB [66] dephosphorylate phosphatidylinositol-3-phosphate (PI3P) to inhibit phagosome maturation, whereas PtpA protein inhibits phagosome acidification by blocking Vacuolar-type ATPase (V-ATPase) [67]. TlyA inhibits Early Endosomal Antigen-1 (EEA1), Ras-related protein 5 (RAB5), and RAB7 recruitment [68]. NdkA inhibits RAB5 and RAB7 [69], while PknG inhibits RAB7L1 [70]. Additionally, Mtb secretes UreC to alkalize the phagosomes [71]. In addition, Mtb perforates the phagosome and escapes to the cytosol using the concerted action of phthiocerol dimycocerosates (PDIM) and ESX-1 system [72]. Host efforts to repair phagosomal rupture were blocked by the EsxH protein [59]. ROS/iNOS: Host NADPH oxidases (NOX) from the cytoplasm and mitochondrial electron transport chain are the primary sources of reactive oxygen species (ROS) production. ROS is blocked by Mtb proteins like Eis, ESAT-6/CFP-10, NuoG, NdkA, PPE2, SodA [73,74,75,76], and inducible nitric oxide synthase (iNOS), while the mediated production of NO is blocked by PtpB, PPE2, PE_PGRS62, PE5, PE15, and PE4 [77,78,79,80]. Epigenetic regulators: Mtb secretes proteins like Rv1988 [81] and Rv2966c [82] to methylate host DNA and proteins like Eis [83] and Rv3423.1 [84] to acetylate host DNA to manipulate the host immune response. Proinflammatory cytokines: Mtb secretes proteins like PtpA, PtpB, ESAT-6/CFP-10, Eis, EchA1, and PknG [77,85,86,87,88,89] to inhibit proinflammatory cytokines. Cell death pathways: Cytosolic escape of the pathogen leads to activation of various cell death pathways like necrosis mediated by Zmp1, CpnT, and PE25:PPE41 [90,91,92] or ferroptosis via PtpA which benefits the pathogen [93]. The cytosolic presence of bacilli DNA or RNA triggers various pathways like apoptosis, autophagy/xenophagy, and pyroptosis, which are detrimental to the pathogen. Mtb secretes NuoG, PtpA, PtpB, NdkA, Rv3654c, Rv3655c, and Rv3033 to block apoptosis [36,75,77,94,95,96]. Proteins like Zmp1, PknF, PtpB, and Rv3364c block inflammasome activation and/or pyroptosis [35,97,98,99]. Autophagy/xenophagy pathways are blocked by proteins like NuoG, Eis, SapM, PE_PGRS20, PE_PGRS47, PPE51, LprE, and PknG [86,100,101,102,103,104,105]; however, these proteins block autophagy indirectly by blocking early/late phagosome proteins. Note: Some live attenuated Mtb or BCG vaccines described in this review lack one or more secretory proteins mentioned above, and they are designated in this figure with rose-colored oval shapes.
Vaccines 12 00530 g001

3. Mycobacterial Vaccines Deficient in Secreted Protein(s)

As noted above, the preference for LAVs over others is due to their superior number of antigens and ability to stimulate immune response for a more prolonged period in vivo [22]. BCG, an excellent example of an LAV, is poorly effective in adults, requiring a replacement or a booster. Initial studies to improve BCG through the recombinant overexpression of antigenic genes did improve its efficacy against TB in the mouse model [106,107,108,109,110]. In particular rBCG30 and BCG85B vaccines showed remarkable efficacy against TB in mice but did not advance to clinical trials. One possible reason is that they lacked the immunogenic RD1 region that encodes ESAT6 and CFP10, which are paradoxically related to virulence in Mtb. An extensively explored approach is deleting genes in BCG and Mtb to derive LAVs. In the past two decades, over fifty mycobacterial mutant strains with deletion or disrupted gene(s) in the chromosome have been tested for their vaccine efficacy in animal models, regardless of their attenuation status [18,110]. Interestingly, a large proportion of the LAVs tested so far are those that lack secretory proteins (Table 1 and Table 2). The primary candidate LAVs lacking secretory proteins are discussed below.

3.1. Ag85 Complex

Mtb expresses three secreted fibronectin proteins (Fbp), namely FbpA (Rv3804), FbpB (Rv1886c), and FbpC (Rv0129c) [111,112]. All three of them are major antigens (Ag), and hence, they are also known as Ag85A (31 kDa), Ag85B (30 kDa), and Ag85C (31.5 kDa) and collectively as the Ag85 complex of proteins [111,112]. The amino acid sequences of these proteins are highly conserved among mycobacterial species and, in particular, the Mtb complex. Besides fibronectin-binding activity, these proteins show mycolyl transferase activity essential in assembling the mycobacterial cell walls [113]. Armitige et al. [114] disrupted the genes fbpA and fbpB in Mtb and assessed the mutant strains (ΔfbpA and ΔfbpB) for their growth and survival in culture, macrophages, and mice [114,115]. The ΔfbpA strain was later shown to protect C57BL/6 mice against challenges with an efficacy similar to BCG [115]. To our knowledge, this is the first study demonstrating that an Mtb mutant lacking a secretory protein antigen is effective as a vaccine against TB. The ΔfbpA candidate vaccine induced high levels of phagosome maturation, proinflammatory cytokines, and Th1 immune response and an increased expansion of CD4+ CXCR3+ IFN-γ+ cells in mice after vaccination [116]. Although recombinant mycobacterial strains over-expressing Ag85B (FbpB) and Ag85C (FbpC) also protect mice against TB [106,117], thus far, Mtb mutants ‘lacking’ these proteins have not been tested for vaccine efficacy. Notably, a BCG strain lacking the Ag85B protein showed efficacy against TB in mice, like BCG [118]. This is not surprising, because BCG naturally has a mutation in Ag85B, which affects the mycolyl transferase activity and its stability [119]. It is intriguing to note that Ag85 complex proteins are immunogenic in mice, guinea pigs, and human immune cells, and individual deletion yields mutants defective in cell wall lipids due to their mycology transferase activity. The ΔfbpA has reduced levels of trehalose dimycolate (TDM) in its cell wall [120] and TDM has been thought to be a virulence factor [121]. Therefore, we are pursuing the hypothesis that the selective deletion of Ag85 complex genes in combination with other functionally characterized genes will lead to markedly immunogenic and concurrently attenuated vaccines [122] (under submission). It should be noted that the MTBVAC vaccine, which is currently undergoing clinical trials, shows a higher secretion of Ag85 proteins, indicating its role in inducing an immune response [123].
Figure 2. Subversion of LC3-associated phagocytosis (LAP) by secreted proteins of mycobacteria. LC3-associated phagocytosis is initiated by macrophage via engaging specific receptors like TLR1/2, TLR2/6, TLR4, Fc receptors, CLEC7A/Dectin-1, and TIM4, and it also recognizes apoptotic, necrotic, and entotic cells [124]. Once the bacilli are engulfed within a single membraned phagosome, phosphatidylinositol-3-phosphate (PI3P) is recruited over the phagosome, which is generated by the PtdIns3K (Class lll phosphatidylinositol 3-kinase) complex. LAP and canonical autophagy share common features and unique features in their pathway. Both require PI(3)P production and common machinery like BECLIN, VPS 34, ATG5, ATG7, ATG16L, TSG101, and RAB7 to recruit LC3 over the phagosome [125]. Recruitment of LC3 over a single-layered membrane is called LAPosome. LAPosome subsequently fuses with lysosome to eliminate M. tuberculosis (Mtb). Some unique proteins involved in the LAP pathway are Rac, p47, p40, p67, p22, gp91phox, and Rubicon [125]. However, Mtb secretes multiple proteins to block the LAP pathway. Mtb proteins like CpsA, NdkA, and PPE2 inhibit the phagosomal recruitment of NADPH oxidase (NOX-2 complex) to the phagosome, whereas NuoG and KatG proteins neutralize reactive oxygen species (ROS) [126]. Note: some live attenuated Mtb or BCG vaccines described in this review lack one or more secretory proteins mentioned above, and they are designated in this figure with rose-colored oval shapes.
Figure 2. Subversion of LC3-associated phagocytosis (LAP) by secreted proteins of mycobacteria. LC3-associated phagocytosis is initiated by macrophage via engaging specific receptors like TLR1/2, TLR2/6, TLR4, Fc receptors, CLEC7A/Dectin-1, and TIM4, and it also recognizes apoptotic, necrotic, and entotic cells [124]. Once the bacilli are engulfed within a single membraned phagosome, phosphatidylinositol-3-phosphate (PI3P) is recruited over the phagosome, which is generated by the PtdIns3K (Class lll phosphatidylinositol 3-kinase) complex. LAP and canonical autophagy share common features and unique features in their pathway. Both require PI(3)P production and common machinery like BECLIN, VPS 34, ATG5, ATG7, ATG16L, TSG101, and RAB7 to recruit LC3 over the phagosome [125]. Recruitment of LC3 over a single-layered membrane is called LAPosome. LAPosome subsequently fuses with lysosome to eliminate M. tuberculosis (Mtb). Some unique proteins involved in the LAP pathway are Rac, p47, p40, p67, p22, gp91phox, and Rubicon [125]. However, Mtb secretes multiple proteins to block the LAP pathway. Mtb proteins like CpsA, NdkA, and PPE2 inhibit the phagosomal recruitment of NADPH oxidase (NOX-2 complex) to the phagosome, whereas NuoG and KatG proteins neutralize reactive oxygen species (ROS) [126]. Note: some live attenuated Mtb or BCG vaccines described in this review lack one or more secretory proteins mentioned above, and they are designated in this figure with rose-colored oval shapes.
Vaccines 12 00530 g002

3.2. LpqH

Proteomics identified the LpqH (Rv3763) protein in the cell wall and the culture filtrates of Mtb. LpqH is a 19 kDa lipoprotein that plays multiple roles in the virulence of Mtb, such as TLR-2 interaction, the induction of humoral and T cell-mediated responses, and apoptosis [127,128]. Macrophages infected with a mutant strain of Mtb lacking this protein (Δ19) show a reduced surface expression of MHC-II molecules and the secretion of cytokines IL-1β, IL12p40, and TNF-α [129]. The Δ19 strain was highly attenuated for growth in C57BL/6 mice compared to wild-type Mtb H37Rv. Despite poor in vivo growth, in mice, LpqH elicited a protective efficacy equal to BCG (1 log10 reduction in Mtb counts in lungs) [130]. Reduced Mtb growth correlated with increased IFN-γ-secreting CD4+ and CD8+ T cells comparable to mice receiving the BCG vaccine, although the lung granulomas of Δ19 strain-vaccinated mice had more lymphocytes than BCG-vaccinated mice, which had more vacuolated foamy macrophages.

3.3. LprG

This is another mycobacterial lipoprotein agonist of TLR-2 receptors in macrophages [131], which, in combination with immunomodulatory lipids, seems to prevent PL fusion in macrophages and limit antigen presentation through the MHC-II pathway [132]. The Rv1411c gene encoding LprG is transcriptionally linked to Rv1410c, which encodes a transmembrane efflux pump, and the deletion of the 1411c-1410c locus in Mtb (ΔlprG) attenuates its growth in immunodeficient mice [133]. The growth attenuation appears to be due to the accumulation of intracellular triacylglycerides (TAG) and altered bacterial metabolism [133]. The ΔlprG mutant was evaluated for its immunogenicity and vaccine efficacy in three mouse strains [134]. While ΔlprG-induced protection was comparable to BCG in C57BL/6 and BALB/c mice, it showed a 0.9 log10 better decrease in Mtb load in the lungs of C3HeB/FeJ mice; the latter developed necrotizing TB granulomas, similar to humans [134]. Variable protection was also reflected in pathology as ΔlprG-vaccinated mice showed fewer granulomas than mice given BCG. Further, compared to BCG, increased Ag-specific CD4+ positive T cell responses, lower percentages of PD-1 positive T cells, and increased antigen-specific IL-17A-secreting T cells were found in the lungs of ΔlprG-immunized mice [134]. These data were confirmed by a recent study where increased protection was observed in ΔlprG-immunized C3HeB/FeJ mice given a low dose aerosol infection. Protection correlated with elevated serum levels of IL-17A, IL-6, CXCL2, CCL2, IFN-γ, and CXCL1 [135]. The ΔlprG mutant illustrates that LAVs may show variable protection in mouse strains of different genetic backgrounds.

3.4. BfrB

Iron limitation is a major factor affecting host–pathogen interactions [136]. Though Mtb has multiple mechanisms to sequester iron from the host, it uses bacterioferritin (BfrB, Rv3841), a secretory protein [137,138], to store the iron when it is abundant and to release it when required. A ΔbfrB mutant of Mtb could not establish a chronic infection in mice [139]. When it was used as a vaccine against Mtb H37Rv in mice, the mutant generated protection comparable to BCG, although they differed in organ pathology and lung transcriptomic signatures [140]. Further, at eight weeks post-vaccination using ΔbfrB, mice had reduced inflammation, smaller lung granulomas, and extensive fibrosis [140].

3.5. CpsA

Encoded by the gene Rv3484, CpsA belongs to the LytR-CpsA-Psr family of conserved proteins related to cell wall assembly in Gram-positive bacteria [141]. Koster et al. reported that Mtb lacking the CpsA protein (ΔcpsA) enhances LC3-associated phagocytosis (LAP) and the recruitment of NADPH oxidase to their phagosomes, suggesting that secreted CpsA protein inhibits these innate immune responses to survive inside the host [125]. In addition, the ΔcpsA strain showed defective growth in mice, which was restored by complementation using a functional cpsA gene [125]. Since LAP can increase antigen presentation through the MHC-II pathway, the authors proposed that the ΔcpsA could be a potential vaccine candidate and created a new mutant mc26206ΔcpsA by deleting cpsA in mc26206, which is an auxotrophic mutant strain (ΔleuCD and ΔpanCD) [142]. Despite increased LC3 trafficking in BMDMs, mc26206ΔcpsA showed protection similar to BCG in C57BL/6 mice challenged with Mtb H37Rv [142]. Given its ability to increase LC3 trafficking, it is unclear why there was no better protection than BCG, which uses a sapM-dependent mechanism to evade autophagy. A deletion of cpsA in Mtb may generate a more protective phenotype.

3.6. BioA

The enzyme 7,8-diaminopelargonic acid synthase, also known as BioA (Rv1568), is one of the four enzymes associated with synthesizing biotin molecules in Mtb [143]. It appears to be critical for the acute and chronic infection of mice infected with Mtb [144]. Mtb ΔbioA was severely attenuated in guinea pigs regardless of aerosol or intradermal route of infection, and intriguingly, the lungs of ΔbioA-infected guinea pigs did not show live bacteria after six weeks post-infection [145]. Although vaccinating guinea pigs using this mutant significantly reduced the bacillary burden in the lungs and spleens, repeated vaccination before the Mtb challenge reduced its efficacy [145]. This study illustrates that the hyperattenuation of Mtb may not always correlate with vaccine efficacy.

3.7. Gln Proteins

Glutamine synthetase activity in the culture filtrate of Mtb has been reported [146]. The Mtb genome has multiple genes encoding glutamine synthetase, namely GlnA1 (Rv2220), GlnA2 (Rv2222c), GlnA3 (Rv1878), GlnA4 (Rv2860c), and a regulator protein GlnE (Rv2221c). However, only GlnA1, GlnA3, and GlnA4 were identified in the culture filtrate of Mtb using proteomics [138,147]. Lee et al. [148] characterized the GlnA1, GlnA2, GlnA3, and GlnA4 mutants and a triple mutant for GlnA1EA2. Of these, only glnA1 and glnA1EA2 were essential for the growth of Mtb, and they were auxotrophic to glutamine. Both ΔglnA1 and ΔglnA1EA2 showed attenuated growth in immunodeficient SCID mice and immunocompetent C57BL/6 mice [148]. Additional studies using C57BL/6 mice revealed that they protected mice like BCG [148]. The inability of these mutants to multiply in vivo appears to have discouraged their further validation. GlnA1 is abundant in mycobacterial extracellular vesicles (MEV), suggesting that it may serve as a diagnostic marker [149].

3.8. SapM

Among the three known secreted phosphatases (SapM, PtpA, and PtpB) of Mtb, SapM (Rv3310) is the first to be identified as an acid phosphatase [150]. After Mtb infects human macrophages, its survival within macrophages depends upon the inhibition of phagosome maturation. SapM is one of the key enzymes implicated in phagosomal maturation arrest during Mtb–macrophage interactions [46,151]. SapM dephosphorylates phagosomal membrane-bound phosphatidylinositol 3-phosphate (PI3P), a lipid molecule associated with the recruitment of downstream effector Rab proteins essential for phagosomal maturation, also called phagosome–lysosome (PL) fusion [34]. Essentially, the SapM-mediated removal of a phosphate molecule from PI3P affects its structure and reduces the recruitment of effector proteins, inhibiting PL fusion. Further, SapM also inhibits autophagy by blocking Rab7, a small GTPase required for lysosomal fusion [100]. Two independent studies used MtbΔsapM to define the role of SapM during PL fusion [152,153]. Upon the infection of macrophages, these mutants were seen more frequently in the matured phagolysosomal compartments enriched with lysosomal markers like LAMP1 when compared to macrophages infected with wild-type Mtb [152,153]. The MtbΔsapM mutant also showed attenuated growth in macrophages and in the lungs and spleen of guinea pigs [152,153]. Consistent with PL fusion competence and attenuation, the mutant showed increased in vitro and in vivo immunogenicity; its immunogenicity was further increased when fbpA was deleted, yielding an MtbΔfbpAsapM double knockout (DKO) mutant [152]. Notably, mice vaccinated with the DKO strain and challenged with Mtb showed better protection (>1 log10) than mice receiving the BCG vaccine [122]. Others reported similar results when guinea pigs were vaccinated using a triple knockout strain of Mtb (MtbΔmms) that lacked three phosphatases (PtpA, PtpB, and SapM) [154]. MtbΔmms was more effective in decreasing Mtb CFUs in the lungs of guinea pigs (3.60 log10) compared to the lungs of guinea pigs receiving BCG vaccination (4.43 log10) [154]. These two studies suggest that the deletion of the same gene increases the vaccine efficacy in the KO mutants, even if the other deleted genes are functionally different, mainly because the deletion of the bioA gene in MtbΔmms (MtbΔmmsb) had no additional benefit [155]. It is relevant to recall here that a BCG sapM mutant (BCGΔsapM) transposon mutant also shows enhanced protection as a vaccine against Mtb in BALB/c mice [156]. BCGΔsapM enriched CD11c+MHC-II intCD40int dendritic cells (DCs) in the draining lymph nodes and was found to be safer than the BCG in SCID mice [157]. Since sapM deletion increased the vaccine efficacy of both Mtb- and BCG-derived mutants, it may be an essential target for more effective vaccines.

3.9. Ptp

PtpA (Rv2234) and PtpB (Rv0153c) are the only two secreted phosphotyrosine protein phosphatases (Ptp) identified in Mtb [158]. By interacting with host signaling partners, they can modulate the cellular pathways of the host cells [159]. The genes encoding PtpA and PtpB have been disrupted in the chromosome of Mtb, and their roles in pathogenicity are reported [160,161]. PtbA primarily affects phagosomal maturation by interacting with H subunit V-ATPase, an enzyme required for phagosomal lumen acidification, and PL fusion by dephosphorylating the vacuolar protein sorting-associated protein 33B (VPS33B), a late endosomal molecule [67,160]. In addition, PtpA plays a critical role in suppressing innate immune responses by interacting with ubiquitin, dephosphorylating JNK, and regulating host genes such as GADD45A [85,158]. Similarly, PtpB has been reported to promote the survival of Mtb H37Rv by suppressing iNOS, IL-1β, and IL-6 [77], thus suppressing innate immune responses through ERK1/2 and Akt pathways [162] and interacting with ubiquitin and inhibiting host cell pyroptosis [97]. Coincidentally, the ΔptpB strain is attenuated for growth in macrophages and guinea pigs compared to wild-type Mtb [161]. Further, an Mtb strain with triple deletions (ptpA, ptpB, and sapM) protected against TB in a guinea pig challenge model better than BCG. [154]. Although the individual roles of ΔptpA or ΔptpB in contributing to vaccine efficacy remain unclear, the fact that they affect multiple host processes justifies their deletion as a strategy to derive vaccines.

3.10. Zmp1

Zinc-containing metalloprotease 1, or Zmp1, is encoded by the gene Rv0198c. Masters et al. [35] first generated a zmp1 deletion mutant in Mtb and BCG, showing that it is crucial in preventing inflammasome activation in macrophages and phagosomal maturation. Macrophages infected with an MtbΔzmp1 mutant not only secreted more IL-1β but also enhanced PL fusion, indicating that it is a key virulence factor [35]. In addition, Zmp1 causes necrotic cell death and the dissemination of Mtb [90]. Interestingly, the deletion of zmp1 has similar effects in both Mtb and BCG fields [35]. This led Sanders et al. to evaluate zmp1 deletion in the BCG vaccine [163,164]. Mouse bone marrow-derived dendritic cells (DCs) infected with BCG and BCGΔzmp1 were compared for their antigen presentation to T cells using Mtb Ag85A-specific MHC-II restricted hybridoma T cells [163]. As expected, DCs infected with BCGΔzmp1 displayed enhanced antigen presentation, suggesting that the BCGΔzmp1 strain is immunogenic [163]. BCGΔzmp1-vaccinated mice showed a stronger delayed-type hypersensitivity (DTH) reaction, and splenocytes showed heightened IFN-γ levels in response to PPD stimulation when compared to splenocytes from BCG-immunized mice [163].
Further, BCG Pasteur lacking Zmp1 and BCG Denmark strain were compared with wild-type BCG Denmark (Danish) for efficacy against TB in guinea pigs [164]. Both mutants showed impressive protection against the Mtb challenge and reduced the lung Mtb burden by approximately 0.5 log10 CFU compared to BCG Denmark. This observation is remarkable because BCG Denmark, on its own, showed about 1.8 log10 CFU reduction compared to unvaccinated controls [164]. BCGΔzmp1 has a high safety profile in SCID mice, particularly the Danish BCGΔzmp1, which is hyper-attenuated [164]. Although the mechanisms underlying protection are unclear, BCGΔzmp1 strain was about to enter into a phase I clinical trial in 2017 [21].

3.11. Eis

The enhanced intracellular survival (EIS) protein is a secretory protein of Mtb encoded by the gene Rv2416c [165]. The name EIS is because it enhances the survival of M. smegmatis within macrophages [166]. Paradoxically, an eis deletion mutant of Mtb (MtbΔeis) showed no defect in intracellular survival within macrophages but induced higher levels of proinflammatory cytokines [167]. Such macrophages also showed increased reactive oxygen species (ROS) generation, autophagy, and cell death [86]. The Eis protein is an enzyme with aminoglycoside N-acetyltransferase activity. Thus, it seems capable of modulating or inhibiting proinflammatory responses, JNK-dependent autophagy, ROS generation, and, to some extent, phagosome maturation by acetylating the host phosphatase protein DUSP16/MKP-7 [73]. The inhibition of autophagy by Eis also seems to be due to the acetylation of histone H3 (Ac-H3), which can upregulate the expression of IL-10 and, as a consequence, activate the Akt/mTOR/p70S6K pathway [83]. Eis is the first secreted protein of Mtb to epigenetically modify macrophages. Recently, its homolog from BCG (BCG_2432c) was knocked out in BCG (China sub-strain), and the mutant ΔBCG_2432c was tested as a vaccine against TB in a C57BL/6 mice [168]. Remarkably, ΔBCG_2432c-immunized mice showed approximately a 2.0 log10 reduction in CFU in the lungs compared to mice immunized with wild-type BCG (China sub-strain). This enhanced protection was likely due to elevated levels of IFN-γ+ CD4+ TEM (effector memory T cells) and IL2+CD4+TCM (Central memory T cells) in the lungs and spleens of ΔBCG_2432c-immunized mice. This is the first study demonstrating a significant reduction in Mtb CFU in mice by a BCG vaccine with a single gene deletion in the chromosome. Because of its effect in mice, the deletion of the eis gene in Mtb appears to be a promising approach for TB vaccines.

3.12. Esx5

The products of the Esx5 system activate the inflammasome pathway in the host cells, facilitating the death of the cells and escape of the Mtb [49,61]. This system comprises 17 genes, including five encoding PPE25, PE18, PPE26, PPE27, and PE19 proteins, all containing strong T cell epitopes showing cross-reactivity with other non-Esx PE/PPE proteins [63,169]. Deleting the five ppe-pe genes (from ppe25 to pe19) of the esx5 renders the Mtb attenuated for growth in immunocompetent mice [169]. Further, C57BL/6 mice immunized with an Mtb Δppe25-pe19 and challenged with H37Rv had a reduced bacterial load in the lungs and spleen compared to BCG-vaccinated mice [169], indicating moderately better protection. In contrast, mice and guinea pigs immunized with the Δesx5 strain, which has a deletion of 17 genes in the esx5 locus (Rv1782-Rv1798), showed similar levels of protection to BCG-vaccinated mice exposed to a virulent HN878 strain of Mtb [170]. Better protection by the Δesx5 vaccine was noted only when administered using a prime-boost strategy with BCG as the prime vaccination [170]. Enhanced protection correlated with increased numbers of activated monocytes, central memory T cells (TCM), and follicular T cells (TFH) [170], although the mechanisms behind the increased protection by prime-boost vaccination remain unclear. In the same study, an MtbΔesx-3 mutant, which has a deletion of 11 genes, was also tested, but its protection seems to be lower than that of the Δesx5 strain. The authors of this study opined that the large-scale deletion of genes in the Δesx5 vaccine could safeguard its potential reversion to virulence. However, this may lead to the loss of protective T cell epitopes in proteins encoded by the deleted genes. Since Δesx5 is highly attenuated in immunocompromised SCID mice [170], it has an interesting potential to be developed as a vaccine for HIV-infected children who are susceptible to TB.
Figure 3. Subversion of xenophagy/autophagy by secreted proteins of mycobacteria. M. tuberculosis (Mtb) is well-known for inhibiting the maturation of phagosomes and their subsequent fusion with lysosomes. Through the ESX-1 secretion system and the cell envelope lipid PDIM, Mtb perforates the phagosome membrane and escapes to the cytosol. The first defensive step of a host is initiating the autophagy pathway by successfully binding ubiquitin to bacteria, followed by the recruitment of autophagy adaptors such as p62, OPTN, TAX18P1, NBR1, and TOLLIP. Subsequently, these autophagy adaptors engross with microtubule-associated-protein-1 light chain 3 (LC3) to deliver Mtb to autophagosomes. However, Mtb has multiple evasion strategies to escape from the host autophagic pathway via the secretion of various protein effectors. Mtb SapM inhibits Rab7, a late endosome marker required for autophagosomes to fuse with the lysosomes [100]. Mtb protein Eis inhibits autophagy via suppression of c-Jun N-terminal kinase (JNK)-mediated reactive oxygen species (ROS) signaling [86] or through the acetylation of host histone, which upregulates IL-10 and activates the Akt/mTOR/p70S6K pathway [83]. Mtb secretes LprE to suppress autophagy by inhibiting the expression of cathelicidin antimicrobial peptide (CAMP) via the p38 MAPK pathway [104]. Mtb protein PE_PGRS47 suppresses autophagy by inhibiting LC3 colocalization [171]. PknG blocks Rab14 to inhibit the autophagosome maturation [103]. PE_PGRS20 and PE_PGRS47 inhibit autophagy by interacting with RAB1A, which recruits the ULK1 (unc-51-like autophagy activating kinase 1) complex to the pre-autophagosome [102]. PPE51 inhibits autophagy by blocking the activation of the extracellular signal-regulated kinase 1/2 (ERK1/2) pathway [105]. ESAT-6 released by Mtb perforates the lysosomes and autophagosomes, leading to perturbation of membranes. Damage to membrane releases cathepsin B in the cytosol and causes the subsequent activation of NLRP3-inflammasome and release of matured IL-1β [172]. Secreted ESAT-6 also affects autophagy flux in dendritic cells [173]. Note: Some live attenuated Mtb or BCG vaccines described in this review lack one or more secretory proteins mentioned above, and they are designated in this figure with rose-colored oval shapes.
Figure 3. Subversion of xenophagy/autophagy by secreted proteins of mycobacteria. M. tuberculosis (Mtb) is well-known for inhibiting the maturation of phagosomes and their subsequent fusion with lysosomes. Through the ESX-1 secretion system and the cell envelope lipid PDIM, Mtb perforates the phagosome membrane and escapes to the cytosol. The first defensive step of a host is initiating the autophagy pathway by successfully binding ubiquitin to bacteria, followed by the recruitment of autophagy adaptors such as p62, OPTN, TAX18P1, NBR1, and TOLLIP. Subsequently, these autophagy adaptors engross with microtubule-associated-protein-1 light chain 3 (LC3) to deliver Mtb to autophagosomes. However, Mtb has multiple evasion strategies to escape from the host autophagic pathway via the secretion of various protein effectors. Mtb SapM inhibits Rab7, a late endosome marker required for autophagosomes to fuse with the lysosomes [100]. Mtb protein Eis inhibits autophagy via suppression of c-Jun N-terminal kinase (JNK)-mediated reactive oxygen species (ROS) signaling [86] or through the acetylation of host histone, which upregulates IL-10 and activates the Akt/mTOR/p70S6K pathway [83]. Mtb secretes LprE to suppress autophagy by inhibiting the expression of cathelicidin antimicrobial peptide (CAMP) via the p38 MAPK pathway [104]. Mtb protein PE_PGRS47 suppresses autophagy by inhibiting LC3 colocalization [171]. PknG blocks Rab14 to inhibit the autophagosome maturation [103]. PE_PGRS20 and PE_PGRS47 inhibit autophagy by interacting with RAB1A, which recruits the ULK1 (unc-51-like autophagy activating kinase 1) complex to the pre-autophagosome [102]. PPE51 inhibits autophagy by blocking the activation of the extracellular signal-regulated kinase 1/2 (ERK1/2) pathway [105]. ESAT-6 released by Mtb perforates the lysosomes and autophagosomes, leading to perturbation of membranes. Damage to membrane releases cathepsin B in the cytosol and causes the subsequent activation of NLRP3-inflammasome and release of matured IL-1β [172]. Secreted ESAT-6 also affects autophagy flux in dendritic cells [173]. Note: Some live attenuated Mtb or BCG vaccines described in this review lack one or more secretory proteins mentioned above, and they are designated in this figure with rose-colored oval shapes.
Vaccines 12 00530 g003

3.13. UreC

Secreted urease C (Rv1850) of mycobacteria neutralizes the acidic environment of the phagosome of the macrophages, contributing to phagosomal maturation arrest [174,175]. Although BCG lacks the RD1 locus and is attenuated, it retains several genes related to phagosomal maturation arrest, including ureC. Consequently, BCG is sequestered within the neutral pH phagosome [176]. To nullify this effect, Kaufmann and his colleagues used a novel strategy of integrating the gene hly, which encodes the Listeria monocytogenes derived listeriolysin (LLO) toxin, into the chromosome of BCG, concurrently deleting ureC in its chromosome [177]. They proposed that BCG-secreted LLO could perforate the phagosomal membrane and allow the bacteria into the cytosol for bacterial antigen processing through the MHC-I pathway. The deletion of ureC, on the other hand, could favor the vATPase-mediated acidic pH required for LLO activity. They constructed two isogenic strains, BCG::hly and BCG::hlyΔureC, and assessed their vaccine efficacy in BALB/c mice [177]. Both showed better protection over the parental strain, although protection by BCG::hlyΔureC was superior [177]. This was due to the induction of apoptosis, increased antigen processing through MHC-I, the elicitation of T cells like TCM, TFH, and Th17, and high IgG antibody levels produced by BCG::hlyΔureC [177,178]. Further, the vaccine had an excellent safety profile in mice, guinea pigs, newborn rabbits, and non-human primates [179]. Currently named VPM1002, it is one of the few live mycobacterial vaccines undergoing clinical trials in sub-Saharan Africa and India [14].

3.14. NuoG

This protein is one of the subunits of type-I NADH dehydrogenase of Mtb and BCG and was identified by screening for anti-apoptotic genes in Mtb [36]. Mtb uses NuoG (Rv3151) to inhibit host apoptosis by neutralizing the ROS derived from NOX2 [74]. Since the vaccine-induced apoptosis of macrophages increases vaccine efficacy [177], nuoG gene deletion in BCG and BCG::hlyΔureC was created to improve vaccine efficacy [101]. Both BCGΔnuoG and BCG::hlyΔureCΔnuoG seem to enhance apoptosis in murine lymph nodes and improve autophagy activity in macrophages [101]. Consequently, both strains showed enhanced immunogenicity and efficacy against TB in mice challenged with Mtb H37Rv [101]. Specifically, the lung CFUs of BCGΔnuoG- and BCG::hlureCΔnuoG-immunized mice demonstrated significant protection against TB. Protection against the more virulent Mtb Bejing W strain was also observed in mice immunized with BCG::hlyΔureCΔnuoG after 90 and 180 days post-challenge. The microarray analysis of draining lymph nodes from vaccinated mice showed an increased expression of genes related to GTPase activity, inflammatory responses, cell activation, and cell proliferation [101].
Additionally, the vaccine increased CD4+ TEM cells, TFH cells, germinal center B cells, and CD4+ TCM cells. Paradoxically, a nuoG mutant made in BCG China sub-strain had no significant protection in the Mtb-challenged mice [168]. These data suggest the possibility that the deletion of a gene in BCG sub-strains may give different effects, and this seems to be important because at least five major sub-strains of BCG (Copenhagen/Danish, Russian, Shanghai/China, Japan, Moreau) are used around the world for the primary immunization of infants.
Table 1. Efficacy of live Mycobacterium tuberculosis vaccines with gene(s) deleted for secreted protein(s).
Table 1. Efficacy of live Mycobacterium tuberculosis vaccines with gene(s) deleted for secreted protein(s).
Vaccine Name Vaccine Components Secreted Protein(s)
Absent
Immunization Route/Dose Challenge Mtb Strain Challenge Route/Dose Animal Model (Strain) Efficacy in Relation to BCG log10 CFU/LUNGS Reduction Than BCG Ref.
ΔfbpAH37Rv strain with single gene (fbpA) knockout.FbpA or Ag85ASC/105 CFU/mouse ErdmanAerosol/2.5 log10 CFU per mouseMouse (C57BL/6)Better than BCG ~1.5[115]
ΔglnA1H37Rv strain with single gene (glnA1) knockout.Glutamine synthetase A1SC/106 CFU/mouseErdmanAerosol/200 CFU per mouseMouse (C57BL/6)Equal to BCG-[148]
ΔglnA1EA2H37Rv strain with 3 genes (glnA1, glnE, and glnA2) knockout.Glutamine synthetase A, E and A2SC/106 CFU/mouseErdmanAerosol/200 CFU per mouseMouse (C57BL/6)Equal to BCG-[148]
Δ19H37Rv strain with single gene (lpqH) knockout.Lipoprotein LpqHSC/106 CFU/mouseH37RvAerosol/100 CFU per mouseMouse (C57BL/6)Equal to BCG-[130]
ΔmmsH37Rv strain with 3 genes (ptpA, ptpB, and sapM) knockout.Phosphatases PtpA, PtpB, and SapMID/5 × 105 CFU/guinea pigH37RvAerosol/10–30 CFU per guinea pigGuinea pigsBetter than BCG 0.83–4 weeks post-challenge;
1.41–12 weeks post-challenge
[154]
ΔbfrBH37Rv strain with single gene (bfrB) knockoutBacterio-ferritin BSC/106 CFU/mouseH37RvAerosol/100 CFU per mouseMouse (C57BL/6)Equal to BCG-[140]
ΔbioAH37Rv strain with single gene (bioA) knockoutBioA or 7,8-diaminopelargonic acid synthaseID/106 CFU/guinea pig (single or double dose with 6 week interval)ErdmanAerosol/50 CFU per guinea pigGuinea pigsEqual to BCG-[145]
ΔmmsbH37Rv strain with 4 genes (ptpA, ptpB, sapM, and bioA) knockout.Phosphatases PtpA, PtpB, SapM and BioAID/5 × 105 CFU/guinea pigH37RvAerosol/10–30 CFU per guinea pigGuinea pigsLess than BCG-[155]
mc26206ΔcpsAH37Rv strain with 3 genes (leuD, panCD, and cpsA) knockout.CpsASC/106 CFU/mouseH37RvAerosol/400 CFU per mouseMouse (C57BL/6)Equal to BCG-[142]
ΔlprGH37Rv strain with two genes (lprG and Rv1410c) knockout.Lipoprotein LprGSC/~106 CFU/mouseH37Rv and ErdmanAerosol/75 CFU per mouse
or Aerosol/1 Median Infectious Dose (1MID50).
Mouse (C57BL/6, BALB/c and C3HeB/FeJ)Equal or better than BCG0.67–0.9 (in C3HeB/
FeJ mice);
[134,135]
SO2MT103 strain with single gene (phoP) knockout.All secreted proteins that are affected by PhoPSC/107 CFU/mouseH37RvIV/2.5 × 105 CFU per mouseMouse (BALB/c)Equal to BCG-[180,181]
SC/5 × 104 CFU/ guinea pigH37RvAerosol/10–50 CFU or 500 CFU per guinea pigGuinea pigs (Dunkin Hartley)Better than BCG in high-dose challenge>1
ID/5 × 105 CFU/macaquesErdmanIT/1000 CFU per macaquesRhesus macaques (Macaca mulatta)Better than BCG0.77
Δppe25-pe19H37Rv strain with 5 genes (ppe25, pe18, ppe26, ppe27, and pe19) knock out.PPE25, PE18, PPE26, PPE27 and PE19SC/106 CFU/mouseH37RvAerosol/100 CFU per mouseMouse (C57BL/6)Better than BCG~0.5[169]
ΔsecA2mc23112 strain with single gene (secA2) knock out. SC/106 CFU/mouseBeijing/W (HN878) or ErdmanAerosol/50–100 CFU per mouseMouse (C57BL/6)Better than BCG0.72[182]
ID/103 CFU/guinea pigH37RvAerosol/10–30 CFU per guinea pigGuinea pigs (Dunkin Hartley)Better than BCG in lymph node but not in lungs-
ΔsecA2ΔlysAmc23112 strain with double gene (secA2 and lysA) knockout.Proteins secreted by SecA2 secretion system SC/106 CFU/mouseErdmanAerosol/50–100 CFU per mouseMouse (C57BL/6)Better than BCG0.66[183]
MTBVACMT103 strain with double gene (phoP and fadD26) knockout.All secreted proteins affected by PhoPSC/5 × 105 CFU/mouseH37RvIN/100 CFU per mouseMouse (C57BL/6)Better than BCG~0.5[184,185]
SC/5 × 103 − 5 × 105 − CFU/guinea pigH37RvAerosol/10–50 CFU per guinea pigGuinea pigs (Dunkin Hartley)Equal to BCG-
ID/8.2 × 105 CFU/macaquesErdmanAerosol/14–30 CFU per macaquesRhesus macaques (Macaca mulatta)Better than BCG but not in CFU-
MTBVAC erp-MT103 strain with triple gene (phoP, fadD26, and erp) knock out.All secreted proteins which are affected by PhoP and ErpID/105 CFU/mouseH37RvIT/103 CFU per mouseMouse (C57BL/6)Equal to BCG-[186]
Δesx-5H37Rv strain with 17 genes (eccB5, eccc5, cyp143, Rv1786, ppe25, pe18, ppe26, ppe27, pe19, esxM, esxN, ncRv11793, Rv1794, eccD5, mycP5, eccE5, and eccA5) knock out.ECCB5, ECCC5, CYP143, RV1786, PPE25, PE18, PPE26, PPE27, PE19, ESXM, ESXN, NCRV11793, RV1794, ECCD5, MYCP5, ECCE5, and ECCA5IM/106 CFU/mouse (2 dose with 6 week interval)HN878, and H37RvAerosol/40–100 CFU per mouseMouse (C57BL/6)Equal to BCG-[170]
IM/104 CFU/guinea pigBeijing 212Aerosol/10–20 CFU per guinea pigGuinea pigs (Dunkin Hartley)Equal to BCG-
Δesx-3H37Rv strain with 11 genes (eccA3, eccB3, eccC3, pe5, ppe4, esxG, esxH, espG3, eccD3, mycP3, and eccE3) knock outECCA3, ECCB3, ECCC3, PE5, PPE4, ESXG, ESXH, ESPG3, ECCD3, MYCP3, and ECCE3IM
/104 CFU/guinea pig
Beijing 212Aerosol/10–20 CFU per guinea pigGuinea pigs (Dunkin Hartley)Not mentioned-[170]
Note: subcutaneous (SC); intradermal (ID); intramuscular (IM); intravenous (IV); intranasal (IN); intratracheal (IT). Efficacy was determined based on lung CFU load in comparison with the wild-type BCG strain at least at one-time point. Log10 CFU was not mentioned for some of the studies for which we stated the approximate values based on bar graphs.

3.15. SecA2

As discussed above, Mtb possesses two secretory transport systems: the primary SecA1 (Rv3241c) and the accessory SecA2 (Rv1821). The latter is predicted to be a unique system in Mtb to transport a small subset of Mtb proteins related to its pathogenicity. Consistent with this prediction, an secA2 deletion mutant in Mtb H37Rv revealed its role in pathogenicity [187]. An secA2 mutant showed diminished SodA expression and relatively attenuated growth in immunocompetent and SCID mice and in mouse macrophages derived from Phox(−/−) and Nos2(−/−) mice [188]. Further, macrophages infected with this strain induced relatively higher levels of TNF-α, IL-2, IFN-γ, and reactive nitrogen intermediates (RNI) than macrophages infected with Mtb H37Rv, suggesting an immunosuppressive role for SecA2 [188].
Interestingly, the secA2 mutant enhanced apoptosis in macrophages and the priming of antigen-specific CD8+ T cells in mice [182], which led to its evaluation as a vaccine in mice and guinea pigs. C57BL/6 mice and guinea pigs immunized with a ΔsecA2 strain and challenged with HN878 and H37Rv showed a better reduction in CFUs against TB compared to BCG [182]. The lung CFUs of ΔsecA2-immunized mice were 0.72 log10 lower than those lung CFUs of mice vaccinated with BCG. This reduction in CFUs was also accompanied by a decrease in histopathological scores for the lungs in ΔsecA2-immunized mice [182]. Interestingly, it was proposed that ΔsecA2 mutant effects were due to SodA, the only enzyme affected by secA2 deletion at that time. However, current literature reveals that SecA2 is responsible for transporting several host effector proteins released by Mtb, including SapM, Ndk, PknG, LdpC, and others [46]. Thus, secA2 deletion is likely to decrease the expression of multiple proteins.
Nonetheless, an Mtb mutant lacking both SecA2 and LysA proteins showed increased protection against TB in mice compared to BCG [183]. Despite the efficacy of the ΔsecA2lysA mutant, it has not advanced as a vaccine candidate, which is slightl surprising. In addition, secA2 deletion in BCG had no impact on its immunogenicity [183], suggesting that secA2 may differently affect the genes of Mtb and BCG.

3.16. PhoP

PhoP is a sensor component of the PhoP–PhoR two-component regulatory system and an important virulence factor [189], regulating approximately 2% of the genes in the Mtb genome [190]. The loss of virulence in Mtb H37Ra (avirulent strain) is partly related to a point mutation in the phoP gene (Rv0757) [191]. Although PhoP is not a secretory protein, evidence indicates that it indirectly controls the translocation of the secreted proteins ESAT-6 and CFP-10 of the ESX1 secretion system [192]. It appears that the translocation of ESAT-6 and CFP-10 to the bacterium’s surface requires the products of espACD genes located within the ESX1 system, whose expression is controlled by phoP [193]. Thus, the deletion of the phoP gene could affect the expression of the espACD genes and, consequently, the translocation of ESAT-6 and CFP-10 [194,195]. The absence of phoP results in the secretion of CFP-10 independently of ESAT-6, eliciting immune responses to CFP-10 in both mice and non-human primates upon MTBVAC exposure [184,196]. A ΔphoP vaccine SO2 developed from a clinical strain of Mtb MT103 belongs to this category [180]. The SO2 vaccine was severely attenuated in SCID mice and conferred superior protection against TB compared to BCG in mice, guinea pigs, and non-human primates [180]. Later, to meet the stipulations of the Geneva Consensus [197], the SO2 vaccine was genetically modified as a marker-less double mutant with deletions in phoP and fadD26, and this new construct was named the MTBVAC vaccine [185]. Compared to BCG, MTBVAC showed enhanced safety, increased immunogenicity, and efficacy in animal models such as mice, guinea pigs, and non-human primates [184,185,196,198]. The MTBVAC vaccine protects macaques better than BCG by reducing disease pathology measured by in vivo imaging using CT scans, macroscopic pathological lesions examined at necropsy, and studying the frequency and severity of pulmonary granulomas [184]. The immune signatures after MTBVAC vaccination also included higher levels of Th1 cytokines response, especially poly- (IFN-γ+TNF-α+IL2+) and multi-(IFN-γ+TNF-α+) functional CD4+ T cells. After rigorous preclinical studies, the MTBVAC vaccine entered a clinical trial in Africa [199]. MTBVAC is an example of a vaccine developed through the rational deletion of genes in Mtb for eventual human application.

3.17. Mpt

Two homologous secreted proteins, namely Mpt70 (Rv2875) and Mpt 83 (Rv2873), were found to be highly immunogenic in humans and mice [200]. The genes encoding these proteins are conserved in both Mtb and BCG. A recent study evaluated a mutant in which these two genes and esxS, espC, and espA of the ESX system in BCG create a five-gene knockout ΔBCG-TK [201]. Interestingly, ΔBCG-TK showed vaccine efficacy similar to that of wild-type BCG in mice, and one can propose a contradictory observation that knocking out genes encoding secretory proteins may not affect vaccine efficacy. However, as noted above, with ΔlprG and ΔcpsA mutants, the genetic background of BCG or Mtb used to create a mutant may decide the vaccine’s immunogenicity.

3.18. Erp

Mtb encodes a 28 kDa secretory protein named exported repeated protein (Erp; Rv3810) that contains 11 proline–glycine–leucine–threonine–serine (PGLTS) repeats [202]. Mycobacterial erp mutants are attenuated in macrophages, zebrafish embryos, mice, and leopard frogs [203,204]. Erp is found to interact with another gene called Rv2212, an adenylyl cyclase. Through this interaction, Erp enhances Rv2212-mediated cyclic AMP (cAMP) production, which seems to lower the intracellular survival of Mtb Δerp. We note here that the deletion of the erp gene in the MTBVAC strain (MTBVAC erp (-) further attenuated its growth in SCID mice compared to MTBVAC and BCG. Although MTBVACΔerp generates protection similar to BCG, because of its higher safety profile, MTBVAC erp (-) has been recommended for the vaccination of immune-suppressed populations, such as people with HIV, where BCG causes disseminated disease, also known as BCGosis [186].

3.19. BCG_1419c

BCG_1419c is a cyclic dimeric GMP (c-di-GMP) phosphodiesterase (PDE) protein with phosphodiesterase activity [205]. This protein is encoded by gene BCG_1419c in the BCG Pasteur strain and by the gene Rv1357c in Mtb H37Rv and it was reported to degrade bis-(30–50)-c-di-GMP, which is linked to biofilm formation and virulence [205]. BCG_1419c is not a secretory protein. However, similar to the phoP mutant, which affects the secretion of ESAT-6 and CFP10, the BCG∆BCG1419c mutant vaccine increases the expression of secreted proteins like Tuf, GroEL1, DnaK, and GroES, while showing reduced levels of GroEL2 and AhcY/SahH [206]. Remarkably, the BCGΔBCG1419c vaccine strain demonstrates a better control of both active and chronic TB in murine and guinea pig models compared to saline control [207,208,209,210,211,212,213]. However, it does not provide better protection for animals in terms of reducing lung CFU compared to BCG. Interestingly, in chronic type 2 diabetes (T2D), murine model BCGΔBCG1419c effectively reduces pneumonia in comparison to BCG-vaccinated mice [213].
Table 2. Efficacy of live BCG vaccines with gene(s) deleted for secreted protein(s).
Table 2. Efficacy of live BCG vaccines with gene(s) deleted for secreted protein(s).
Vaccine Name Vaccine Components Secreted Protein(s)
Absent
Immunization
Route/Dose
Challenge Mtb Strain Challenge Route/Dose Animal Model (Strain) Efficacy in
Relation to BCG
log10 CFU/LUNGS
Reduction Than BCG
Ref.
VPM1002
(ΔureC::hly)
BCG Pasteur strain with single gene (ureC) knockout, which expresses listeriolysin (hly).Urease CIV/106 CFU/mouseH37Rv or Beijing/WAerosol/30 or 200 CFU per mouseMouse (BALB/c)Better than BCG~0.5–2[177]
sapM::TBCG 1721 strain with single gene (sapM) knockout.SapM phosphataseSC/105 CFU/mouse H37RvIV/5 × 104 CFU per mouse
(or)
IT/ 2 × 105 CFU per mouse
Mouse (BALB/c)Better than BCG~0.5 (Luminescence)[156]
Δzmp1BCG Pasteur or Denmark strain with single gene (zmp1) knockout.Zmp1 or Zinc containing metalloprotease 1SC/5 × 104 CFU/ guinea pigH37RvAerosol/10–50 CFU per guinea pigGuinea pigs (Dunkin Hartley)Better than BCG~0.91[164]
BCG:Δ85BBCG Pasteur strain with single gene (fbpB) knockout.FbpB/Ag85BSC/5 × 105 CFU/mouseH37RvAerosol/100 CFU per mouseMouse (C57BL/6)Equal to BCG-[118]
ΔnuoGBCG Pasteur strain with single gene (nuoG) knockout.NuoG
type-I NADH dehydrogenase subunit G
SC/106 CFU/mouseH37RvAerosol/100–200 CFU per mouseMouse (C57BL/6)Better than BCG ~0.5[101]
ΔureC::hly ΔnuoGBCG Pasteur strain with double gene (ureC, nuoG) knockout, which expresses listeriolysin.UreC and NuoGSC/106 CFU/mouseH37Rv or Beijing/WAerosol/100–200 CFU per mouseMouse (C57BL/6)Better than BCG~0.8–2[101]
ΔBCG TK (triple knock-out)BCG Danish strain with five gene (esxS, mpt70, mpt83, espC and espA) knockoutEsxS, Mpt70, Mpt83, EspC, and EspASC/5 × 104 CFU/guinea pigM. bovis AF2122/97Aerosol/10–20 CFU per guinea pigGuinea pigs (Dunkin Hartley)Equal to BCG-[201]
ΔBCG2432cBCG China strain with eis gene (BCG2432c) knockoutEIS or Enhanced Intracellular Survival proteinSC/106 CFU/mouseH37RvIN/100 CFU per mouseMouse (C57BL/6)Better than BCG~1–2[168]
ΔBCG3174BCG China strain with nuoG gene knockout (BCG3174)NuoGSC/106 CFU/mouseH37RvIntranasal/100 CFU per mouseMouse (C57BL/6)Equal to BCG-[168]
ΔBCG 1419cBCG Pasteur strain with single gene (c-di-GMP phosphodiesterase) knockout.All secreted proteins affected by (c-di-GMP phosphodiesterase).SC/8 × 103 or 2.5 × 102 or 5 × 104 or 106 or ~105 or 107 CFU/mouseH37Rv or M2 or HN878IT/2.5 × 105 or 103 or ~170 CFU per mouse or Aerosol/100–200 CFU per mouse.Mouse (BALB/c, B6D2F1, C57BL/6, I/StSnEgYCit)Equal to BCG or Better than BCG in chronic infection model~0.8 (chronic infection model)[207,208,209,210,211,212,213]
ID/103 CFU per guinea pigH37RvAerosol/10–20 CFU per guinea pigGuinea pigsEqual to BCG-
Note: subcutaneous (SC); intradermal (ID); intramuscular (IM); intravenous (IV); intranasal (IN); intratracheal (IT). Efficacy was determined based on lung CFU load in comparison with the wild-type BCG strain at least at one time point. Log10 CFU was not mentioned for some of the studies for which we stated the approximate values based on bar graphs.

4. Status of Mycobacterial Vaccines Deficient in Secreted Protein(s)

It is apparent from these studies that BCG is a ‘natural’ vaccine arising out of the deletion of genes encoding secretory proteins, as it lacks the Region of Difference 1 (RD1) that encodes several secretory proteins, including the immunodominant antigens ESAT-6 and CFP-10 [54]. Studies discussed above specifically targeted genes that played a role during pathogenesis for developing vaccines. Surprisingly, only a few Mtb-derived vaccines showed higher efficacy than BCG, and most were comparable to BCG. In contrast, many BCG mutants with deletions of secretory genes showed increased efficacy compared to the wild type. We recall here that Mtb- or BCG-derived vaccines that showed higher efficacy than BCG had deletions in fpbA, sapM, zmp1, ureC, nuoG, secA2, and eis genes. Intriguingly, except for the fbpA-encoded product, other genes modulated PL fusion, autophagy, apoptosis, and inflammasomes in the antigen-presenting cells (APCs) [34,35,36,61,83,214]. Although the disruption of fbpA in Mtb also led to increased PL fusion in APCs, the underlying mechanism remains unclear [215].
Because PL fusion, autophagy, apoptosis, and inflammasome activation ensure the efficient processing and presentation of vaccine antigens to the T cells by the APCs [216,217,218], it is apparent that the deletion of these genes enhanced the efficacy of the mutants against tuberculosis, justifying the strategy of secretory protein gene knockout. However, we need to recognize the caveat that these deletions of proteins that contain potent T and B cell epitopes like ESAT6 and CFP10 may lead to reduced immunogenicity. In addition, an in vitro phenotype may not always lead to increased immunogenicity. An example is Mtb lacking cpsA; although MtbΔcpsA enhanced autophagy in APCs, an increase in vaccine efficacy was not observed [142]. In this regard, deletion strategies should focus on those that interfere with PL fusion and autophagy (SapM, Zmp, PtpA, and PtpB).
It also appears that gene knockouts in BCG and Mtb will likely have different consequences because of the genetic background of the attenuated vaccine vs. virulent pathogen. Because BCG has an excellent safety record, the deletion of sapM, zmp1, and eis is likely to improve immunogenicity, since BCG contains multiple immunogenic genes, an Antigen85 complex, and others listed above. Intriguingly, eis deletion in BCG markedly reduced the Mtb load in the lungs of nasally challenged mice [168]. Further, the double deletion of ureC and nuoG synergistically enhanced the efficacy of BCG [101], suggesting that highly effective BCG mutants can be produced through multiple deletions of genes selected based on their functions. Herein, we again emphasize the need to exercise caution in selecting the parent platform, since the ΔnuoG BCG Pasteur strain was effective against TB [101] but not the ΔnuoG BCG China sub-strain [168].
Similar to BCG vaccine manipulations, using the Mtb platform to derive vaccines deleting secretory products appears encouraging. The ΔsecAlysA and ΔfbpA-sapM and mutants were more effective than BCG [116,152,183] and are potential booster vaccines for BCG-vaccinated infants. Among these, the ΔsecA-lysA strain has an excellent safety profile in mice [183] compared to ΔfbpA-sapM, qualifying it for clinical trials. Our unpublished observations indicate that the safety profile of the ΔfbpA-sapM mutant in mice is lower than that of BCG and may need additional gene deletions. Although ΔlprG derived from Mtb also seems to be a strong candidate vaccine, its superior efficacy over BCG is apparent only in TB-susceptible C3HeB/FeJ mice but not in the TB-resistant C57/BL6 mice [134].

5. Future Directions and Conclusions

A significant impediment to developing vaccines against TB is the lack of reliable immune correlates of protection [219,220,221,222]. Studies with mice, guinea pigs, and rabbits revealed that CD4+ T cells secreting IFN-γ, TNF-γ, and IL-1β and CD8+ T cells secreting granulysin and perforin play a critical role in defending Mtb infection [223,224]. Recent studies show that TH17 T cells secreting IL-17 are also a protective parameter [225].
All new TB vaccines aim to enhance T cell-dependent immune responses in the host. Interestingly, the role of innate immunity in designing more efficacious vaccines has received less attention [226]. An intriguing example is the induction of trained immunity by the BCG vaccine, which activates dectin signaling, generating protection against TB through epigenetic modifications of macrophages, neutrophils, and DCs [227]. Although BCG-induced trained immunity seems more effective against nontuberculous infections, it remains unclear to what extent trained immunity affects adaptive immunity during tuberculosis vaccination. It has been reported that BCG-induced protection wanes by year 5 in children, and TB continues to occur despite vaccination [228].
In this context, we noted that deleting multiple secretory proteins led to enhanced autophagy and inflammasome activation that delivered vaccines to lysosomes for better immune responses (our unpublished work). Autophagy and inflammasome pathways are major innate immunity pathways triggered by multiple mechanisms of mycobacteria, including TLR, NLR, and C-type lectin (also known as dectins) signaling. Intriguingly, dectin-dependent trained immunity induced by BCG cell wall components is regulated by autophagy [229]. There is, therefore, a pressing need to investigate whether new-generation tuberculosis vaccines can be genetically manipulated to activate both innate (autophagy and inflammasome) and adaptive T cell-dependent arms of immunity. For example, Mtb was reported to secrete a lysine acetyltransferase that epigenetically modified the ability of macrophages to secrete anti-inflammatory cytokines like IL-10.
Moreover, the acetylation of histones associated with the genes regulating autophagy regulates the induction of autophagy [230]. A lysine acetyltransferase mutant of Mtb may induce robust autophagy and show better protection. Further, Mtb-derived methyl transferases hyermethylate the DNA of tuberculosis patients, reducing their immune responses [231]. We propose that deletion mutants of Mtb that lack acetylase, methylase, or both may serve as promising vaccine candidates.
An attractive alternative approach to enhance innate immunity is to integrate ‘adjuvant’ active molecules such as TLR agonists into candidate vaccines to overcome immune-suppressing proteins. We recently fused a TLR2-activating CFP-10-derived peptide C5 with Ag85B and expressed it in BCG through a plasmid. The recombinant BCG85BC5 vaccine enhanced protection against TB and induced significant levels of T-effector and T-central memory cell response in mice [108]. We propose that integrating adjuvant constructs like this into ΔsapM or Δzmp1 mutants will markedly improve protection associated with long-term memory.
Finally, recent reports indicate that the humoral immune response against Mtb may also play a significant role in protection against TB [101,184]. In this direction, we propose that deletion mutants can be made to express antibody-inducing peptide epitopes.

Author Contributions

S.D. and C.J. conceived the project; R.V., S.S.G., C.J. and S.D. wrote and edited the manuscript; R.V. created the figures. All authors have read and agreed to the published version of the manuscript.

Funding

This project was funded by NIH grants R01AI175837 (S.D., C.J., and S.S.G.) and R15AI156647 (S.D.). Additionally, C.J. was supported by NIH R01AI161015 and S.S.G. was supported by NIH R16GM149497, Cancer Prevention Institute of Texas (CPRIT) RR170020, and American Cancer Society RSG-22-170-01-RMC grants. The funders had no role in the study design, data collection, analysis, interpretation, or conclusions presented in this manuscript.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

All figures in this manuscript were created using www.BioRender.com.

Conflicts of Interest

The authors declare no competing interests.

References

  1. Brennan, P.J. Structure, function, and biogenesis of the cell wall of Mycobacterium tuberculosis. Tuberculosis 2003, 83, 91–97. [Google Scholar] [CrossRef]
  2. WHO. Global Tuberculosis Report 2022; World Health Organization: Geneva, Switzerland, 2023. [Google Scholar]
  3. Houben, R.M.; Dodd, P.J. The Global Burden of Latent Tuberculosis Infection: A Re-estimation Using Mathematical Modelling. PLoS Med. 2016, 13, e1002152. [Google Scholar] [CrossRef]
  4. Ahmed, A.; Rakshit, S.; Adiga, V.; Dias, M.; Dwarkanath, P.; D’Souza, G.; Vyakarnam, A. A century of BCG: Impact on tuberculosis control and beyond. Immunol. Rev. 2021, 301, 98–121. [Google Scholar] [CrossRef]
  5. Andersen, P.; Doherty, T.M. The success and failure of BCG—Implications for a novel tuberculosis vaccine. Nat. Rev. Microbiol. 2005, 3, 656–662. [Google Scholar] [CrossRef]
  6. Kaufmann, S.H.E. Vaccination Against Tuberculosis: Revamping BCG by Molecular Genetics Guided by Immunology. Front. Immunol. 2020, 11, 316. [Google Scholar] [CrossRef]
  7. Fine, P.E. Variation in protection by BCG: Implications of and for heterologous immunity. Lancet 1995, 346, 1339–1345. [Google Scholar] [CrossRef]
  8. Prentice, S.; Dockrell, H.M. Antituberculosis BCG vaccination: More reasons for varying innate and adaptive immune responses. J. Clin. Investig. 2020, 130, 5121–5123. [Google Scholar] [CrossRef]
  9. Behr, M.A. Comparative genomics of BCG vaccines. Tuberculosis 2001, 81, 165–168. [Google Scholar] [CrossRef]
  10. Rook, G.A.; Dheda, K.; Zumla, A. Do successful tuberculosis vaccines need to be immunoregulatory rather than merely Th1-boosting? Vaccine 2005, 23, 2115–2120. [Google Scholar] [CrossRef]
  11. Elias, D.; Akuffo, H.; Pawlowski, A.; Haile, M.; Schon, T.; Britton, S. Schistosoma mansoni infection reduces the protective efficacy of BCG vaccination against virulent Mycobacterium tuberculosis. Vaccine 2005, 23, 1326–1334. [Google Scholar] [CrossRef]
  12. Elias, D.; Britton, S.; Aseffa, A.; Engers, H.; Akuffo, H. Poor immunogenicity of BCG in helminth infected population is associated with increased in vitro TGF-beta production. Vaccine 2008, 26, 3897–3902. [Google Scholar] [CrossRef]
  13. Martinez, L.; Cords, O.; Liu, Q.; Acuna-Villaorduna, C.; Bonnet, M.; Fox, G.J.; Carvalho, A.C.C.; Chan, P.C.; Croda, J.; Hill, P.C.; et al. Infant BCG vaccination and risk of pulmonary and extrapulmonary tuberculosis throughout the life course: A systematic review and individual participant data meta-analysis. Lancet Glob. Health 2022, 10, e1307–e1316. [Google Scholar] [CrossRef]
  14. 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]
  15. Sable, S.B.; Posey, J.E.; Scriba, T.J. Tuberculosis Vaccine Development: Progress in Clinical Evaluation. Clin. Microbiol. Rev. 2019, 33, 10–1128. [Google Scholar] [CrossRef]
  16. Andersen, P.; Scriba, T.J. Moving tuberculosis vaccines from theory to practice. Nat. Rev. Immunol. 2019, 19, 550–562. [Google Scholar] [CrossRef]
  17. Tang, J.; Yam, W.C.; Chen, Z. Mycobacterium tuberculosis infection and vaccine development. Tuberculosis 2016, 98, 30–41. [Google Scholar] [CrossRef]
  18. Ng, T.W.; Saavedra-Avila, N.A.; Kennedy, S.C.; Carreno, L.J.; Porcelli, S.A. Current efforts and future prospects in the development of live mycobacteria as vaccines. Expert Rev. Vaccines 2015, 14, 1493–1507. [Google Scholar] [CrossRef]
  19. Schrager, L.K.; Vekemens, J.; Drager, N.; Lewinsohn, D.M.; Olesen, O.F. The status of tuberculosis vaccine development. Lancet Infect. Dis. 2020, 20, e28–e37. [Google Scholar] [CrossRef]
  20. Larsen, S.E.; Erasmus, J.H.; Reese, V.A.; Pecor, T.; Archer, J.; Kandahar, A.; Hsu, F.C.; Nicholes, K.; Reed, S.G.; Baldwin, S.L.; et al. An RNA-Based Vaccine Platform for Use against Mycobacterium tuberculosis. Vaccines 2023, 11, 130. [Google Scholar] [CrossRef]
  21. Soleimanpour, S.; Yaghoubi, A.; Sadat Seddighinia, F.; Rezaee, S.A.R. A century of attempts to develop an effective tuberculosis vaccine: Why they failed? Int. Immunopharmacol. 2022, 109, 108791. [Google Scholar] [CrossRef]
  22. Scriba, T.J.; Kaufmann, S.H.; Henri Lambert, P.; Sanicas, M.; Martin, C.; Neyrolles, O. Vaccination Against Tuberculosis with Whole-Cell Mycobacterial Vaccines. J. Infect. Dis. 2016, 214, 659–664. [Google Scholar] [CrossRef]
  23. Seder, R.A.; Hill, A.V. Vaccines against intracellular infections requiring cellular immunity. Nature 2000, 406, 793–798. [Google Scholar] [CrossRef]
  24. Porcelli, S.; Morita, C.T.; Brenner, M.B. CD1b restricts the response of human CD4-8-T lymphocytes to a microbial antigen. Nature 1992, 360, 593–597. [Google Scholar] [CrossRef]
  25. Van Rhijn, I.; Moody, D.B. CD1 and mycobacterial lipids activate human T cells. Immunol. Rev. 2015, 264, 138–153. [Google Scholar] [CrossRef]
  26. 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.; Handler, K.; Picelli, S.; et al. BCG Vaccination in Humans Elicits Trained Immunity via the Hematopoietic Progenitor Compartment. Cell Host Microbe 2020, 28, 322–334.e325. [Google Scholar] [CrossRef]
  27. Mourits, V.P.; Koeken, V.; de Bree, L.C.J.; Moorlag, S.; Chu, W.C.; Xu, X.; Dijkstra, H.; Lemmers, H.; Joosten, L.A.B.; Wang, Y.; et al. BCG-Induced Trained Immunity in Healthy Individuals: The Effect of Plasma Muramyl Dipeptide Concentrations. J. Immunol. Res. 2020, 2020, 5812743. [Google Scholar] [CrossRef]
  28. Schoenen, H.; Huber, A.; Sonda, N.; Zimmermann, S.; Jantsch, J.; Lepenies, B.; Bronte, V.; Lang, R. Differential control of Mincle-dependent cord factor recognition and macrophage responses by the transcription factors C/EBPbeta and HIF1alpha. J. Immunol. 2014, 193, 3664–3675. [Google Scholar] [CrossRef]
  29. Zhao, X.Q.; Zhu, L.L.; Chang, Q.; Jiang, C.; You, Y.; Luo, T.; Jia, X.M.; Lin, X. C-type lectin receptor dectin-3 mediates trehalose 6,6′-dimycolate (TDM)-induced Mincle expression through CARD9/Bcl10/MALT1-dependent nuclear factor (NF)-kappaB activation. J. Biol. Chem. 2014, 289, 30052–30062. [Google Scholar] [CrossRef] [PubMed]
  30. Tsuji, S.; Matsumoto, M.; Takeuchi, O.; Akira, S.; Azuma, I.; Hayashi, A.; Toyoshima, K.; Seya, T. Maturation of human dendritic cells by cell wall skeleton of Mycobacterium bovis bacillus Calmette-Guerin: Involvement of toll-like receptors. Infect. Immun. 2000, 68, 6883–6890. [Google Scholar] [CrossRef]
  31. Blanc, L.; Gilleron, M.; Prandi, J.; Song, O.R.; Jang, M.S.; Gicquel, B.; Drocourt, D.; Neyrolles, O.; Brodin, P.; Tiraby, G.; et al. Mycobacterium tuberculosis inhibits human innate immune responses via the production of TLR2 antagonist glycolipids. Proc. Natl. Acad. Sci. USA 2017, 114, 11205–11210. [Google Scholar] [CrossRef]
  32. Tran, V.; Ahn, S.K.; Ng, M.; Li, M.; Liu, J. Loss of Lipid Virulence Factors Reduces the Efficacy of the BCG Vaccine. Sci. Rep. 2016, 6, 29076. [Google Scholar] [CrossRef]
  33. Walker, K.B.; Brennan, M.J.; Ho, M.M.; Eskola, J.; Thiry, G.; Sadoff, J.; Dobbelaer, R.; Grode, L.; Liu, M.A.; Fruth, U.; et al. The second Geneva Consensus: Recommendations for novel live TB vaccines. Vaccine 2010, 28, 2259–2270. [Google Scholar] [CrossRef]
  34. Vergne, I.; Chua, J.; Lee, H.H.; Lucas, M.; Belisle, J.; Deretic, V. Mechanism of phagolysosome biogenesis block by viable Mycobacterium tuberculosis. Proc. Natl. Acad. Sci. USA 2005, 102, 4033–4038. [Google Scholar] [CrossRef]
  35. Master, S.S.; Rampini, S.K.; Davis, A.S.; Keller, C.; Ehlers, S.; Springer, B.; Timmins, G.S.; Sander, P.; Deretic, V. Mycobacterium tuberculosis prevents inflammasome activation. Cell Host Microbe 2008, 3, 224–232. [Google Scholar] [CrossRef]
  36. Velmurugan, K.; Chen, B.; Miller, J.L.; Azogue, S.; Gurses, S.; Hsu, T.; Glickman, M.; Jacobs, W.R., Jr.; Porcelli, S.A.; Briken, V. Mycobacterium tuberculosis nuoG is a virulence gene that inhibits apoptosis of infected host cells. PLoS Pathog. 2007, 3, e110. [Google Scholar] [CrossRef]
  37. Chai, Q.; Wang, L.; Liu, C.H.; Ge, B. New insights into the evasion of host innate immunity by Mycobacterium tuberculosis. Cell. Mol. Immunol. 2020, 17, 901–913. [Google Scholar] [CrossRef]
  38. Majlessi, L.; Prados-Rosales, R.; Casadevall, A.; Brosch, R. Release of mycobacterial antigens. Immunol. Rev. 2015, 264, 25–45. [Google Scholar] [CrossRef]
  39. Sambandamurthy, V.K.; Jacobs, W.R., Jr. Live attenuated mutants of Mycobacterium tuberculosis as candidate vaccines against tuberculosis. Microbes Infect. 2005, 7, 955–961. [Google Scholar] [CrossRef]
  40. Watt, J.; Liu, J. Preclinical Progress of Subunit and Live Attenuated Mycobacterium tuberculosis Vaccines: A Review following the First in Human Efficacy Trial. Pharmaceutics 2020, 12, 848. [Google Scholar] [CrossRef]
  41. Green, E.R.; Mecsas, J. Bacterial Secretion Systems: An Overview. Microbiol. Spectr. 2016, 4, 213–239. [Google Scholar] [CrossRef]
  42. Tsirigotaki, A.; De Geyter, J.; Sostaric, N.; Economou, A.; Karamanou, S. Protein export through the bacterial Sec pathway. Nat. Rev. Microbiol. 2017, 15, 21–36. [Google Scholar] [CrossRef]
  43. Berks, B.C. The twin-arginine protein translocation pathway. Annu. Rev. Biochem. 2015, 84, 843–864. [Google Scholar] [CrossRef]
  44. Converse, S.E.; Cox, J.S. A protein secretion pathway critical for Mycobacterium tuberculosis virulence is conserved and functional in Mycobacterium smegmatis. J. Bacteriol. 2005, 187, 1238–1245. [Google Scholar] [CrossRef]
  45. Bitter, W.; Houben, E.N.; Luirink, J.; Appelmelk, B.J. Type VII secretion in mycobacteria: Classification in line with cell envelope structure. Trends Microbiol. 2009, 17, 337–338. [Google Scholar] [CrossRef]
  46. Augenstreich, J.; Briken, V. Host Cell Targets of Released Lipid and Secreted Protein Effectors of Mycobacterium tuberculosis. Front. Cell. Infect. Microbiol. 2020, 10, 595029. [Google Scholar] [CrossRef]
  47. Ates, L.S.; Houben, E.N.G.; Bitter, W. Type VII Secretion: A Highly Versatile Secretion System. Microbiol. Spectr. 2016, 4, 357–384. [Google Scholar] [CrossRef]
  48. Tran, H.R.; Grebenc, D.W.; Klein, T.A.; Whitney, J.C. Bacterial type VII secretion: An important player in host-microbe and microbe-microbe interactions. Mol. Microbiol. 2021, 115, 478–489. [Google Scholar] [CrossRef]
  49. Abdallah, A.M.; Gey van Pittius, N.C.; Champion, P.A.; Cox, J.; Luirink, J.; Vandenbroucke-Grauls, C.M.; Appelmelk, B.J.; Bitter, W. Type VII secretion—Mycobacteria show the way. Nat. Rev. Microbiol. 2007, 5, 883–891. [Google Scholar] [CrossRef]
  50. Daleke, M.H.; Ummels, R.; Bawono, P.; Heringa, J.; Vandenbroucke-Grauls, C.M.; Luirink, J.; Bitter, W. General secretion signal for the mycobacterial type VII secretion pathway. Proc. Natl. Acad. Sci. USA 2012, 109, 11342–11347. [Google Scholar] [CrossRef]
  51. Dumas, E.; Christina Boritsch, E.; Vandenbogaert, M.; Rodriguez de la Vega, R.C.; Thiberge, J.M.; Caro, V.; Gaillard, J.L.; Heym, B.; Girard-Misguich, F.; Brosch, R.; et al. Mycobacterial Pan-Genome Analysis Suggests Important Role of Plasmids in the Radiation of Type VII Secretion Systems. Genome Biol. Evol. 2016, 8, 387–402. [Google Scholar] [CrossRef]
  52. Newton-Foot, M.; Warren, R.M.; Sampson, S.L.; van Helden, P.D.; Gey van Pittius, N.C. The plasmid-mediated evolution of the mycobacterial ESX (Type VII) secretion systems. BMC Evol. Biol. 2016, 16, 62. [Google Scholar] [CrossRef]
  53. Brodin, P.; Majlessi, L.; Marsollier, L.; de Jonge, M.I.; Bottai, D.; Demangel, C.; Hinds, J.; Neyrolles, O.; Butcher, P.D.; Leclerc, C.; et al. Dissection of ESAT-6 system 1 of Mycobacterium tuberculosis and impact on immunogenicity and virulence. Infect. Immun. 2006, 74, 88–98. [Google Scholar] [CrossRef]
  54. Pym, A.S.; Brodin, P.; Brosch, R.; Huerre, M.; Cole, S.T. Loss of RD1 contributed to the attenuation of the live tuberculosis vaccines Mycobacterium bovis BCG and Mycobacterium microti. Mol. Microbiol. 2002, 46, 709–717. [Google Scholar] [CrossRef]
  55. Lewis, K.N.; Liao, R.; Guinn, K.M.; Hickey, M.J.; Smith, S.; Behr, M.A.; Sherman, D.R. Deletion of RD1 from Mycobacterium tuberculosis mimics bacille Calmette-Guerin attenuation. J. Infect. Dis. 2003, 187, 117–123. [Google Scholar] [CrossRef]
  56. Hsu, T.; Hingley-Wilson, S.M.; Chen, B.; Chen, M.; Dai, A.Z.; Morin, P.M.; Marks, C.B.; Padiyar, J.; Goulding, C.; Gingery, M.; et al. The primary mechanism of attenuation of bacillus Calmette-Guerin is a loss of secreted lytic function required for invasion of lung interstitial tissue. Proc. Natl. Acad. Sci. USA 2003, 100, 12420–12425. [Google Scholar] [CrossRef]
  57. van der Wel, N.; Hava, D.; Houben, D.; Fluitsma, D.; van Zon, M.; Pierson, J.; Brenner, M.; Peters, P.J. M. tuberculosis and M. leprae translocate from the phagolysosome to the cytosol in myeloid cells. Cell 2007, 129, 1287–1298. [Google Scholar] [CrossRef]
  58. Roy, S.; Ghatak, D.; Das, P.; BoseDasgupta, S. ESX secretion system: The gatekeepers of mycobacterial survivability and pathogenesis. Eur. J. Microbiol. Immunol. 2020, 10, 202–209. [Google Scholar] [CrossRef]
  59. Portal-Celhay, C.; Tufariello, J.M.; Srivastava, S.; Zahra, A.; Klevorn, T.; Grace, P.S.; Mehra, A.; Park, H.S.; Ernst, J.D.; Jacobs, W.R., Jr.; et al. Mycobacterium tuberculosis EsxH inhibits ESCRT-dependent CD4(+) T-cell activation. Nat. Microbiol. 2016, 2, 16232. [Google Scholar] [CrossRef]
  60. Tufariello, J.M.; Chapman, J.R.; Kerantzas, C.A.; Wong, K.W.; Vilcheze, C.; Jones, C.M.; Cole, L.E.; Tinaztepe, E.; Thompson, V.; Fenyo, D.; et al. Separable roles for Mycobacterium tuberculosis ESX-3 effectors in iron acquisition and virulence. Proc. Natl. Acad. Sci. USA 2016, 113, E348–E357. [Google Scholar] [CrossRef]
  61. Abdallah, A.M.; Bestebroer, J.; Savage, N.D.; de Punder, K.; van Zon, M.; Wilson, L.; Korbee, C.J.; van der Sar, A.M.; Ottenhoff, T.H.; van der Wel, N.N.; et al. Mycobacterial secretion systems ESX-1 and ESX-5 play distinct roles in host cell death and inflammasome activation. J. Immunol. 2011, 187, 4744–4753. [Google Scholar] [CrossRef]
  62. Abdallah, A.M.; Savage, N.D.; van Zon, M.; Wilson, L.; Vandenbroucke-Grauls, C.M.; van der Wel, N.N.; Ottenhoff, T.H.; Bitter, W. The ESX-5 secretion system of Mycobacterium marinum modulates the macrophage response. J. Immunol. 2008, 181, 7166–7175. [Google Scholar] [CrossRef]
  63. Shah, S.; Briken, V. Modular Organization of the ESX-5 Secretion System in Mycobacterium tuberculosis. Front. Cell. Infect. Microbiol. 2016, 6, 49. [Google Scholar] [CrossRef]
  64. Gray, T.A.; Clark, R.R.; Boucher, N.; Lapierre, P.; Smith, C.; Derbyshire, K.M. Intercellular communication and conjugation are mediated by ESX secretion systems in mycobacteria. Science 2016, 354, 347–350. [Google Scholar] [CrossRef]
  65. Dong, D.; Wang, D.; Li, M.; Wang, H.; Yu, J.; Wang, C.; Liu, J.; Gao, Q. PPE38 modulates the innate immune response and is required for Mycobacterium marinum virulence. Infect. Immun. 2012, 80, 43–54. [Google Scholar] [CrossRef]
  66. Beresford, N.; Patel, S.; Armstrong, J.; Szoor, B.; Fordham-Skelton, A.P.; Tabernero, L. MptpB, a virulence factor from Mycobacterium tuberculosis, exhibits triple-specificity phosphatase activity. Biochem. J. 2007, 406, 13–18. [Google Scholar] [CrossRef]
  67. Wong, D.; Bach, H.; Sun, J.; Hmama, Z.; Av-Gay, Y. Mycobacterium tuberculosis protein tyrosine phosphatase (PtpA) excludes host vacuolar-H+-ATPase to inhibit phagosome acidification. Proc. Natl. Acad. Sci. USA 2011, 108, 19371–19376. [Google Scholar] [CrossRef]
  68. Mittal, E.; Kumar, S.; Rahman, A.; Krishnasastry, M.V. Modulation of phagolysosome maturation by bacterial tlyA gene product. J. Biosci. 2014, 39, 821–834. [Google Scholar] [CrossRef]
  69. Sun, J.; Wang, X.; Lau, A.; Liao, T.Y.; Bucci, C.; Hmama, Z. Mycobacterial nucleoside diphosphate kinase blocks phagosome maturation in murine RAW 264.7 macrophages. PLoS ONE 2010, 5, e8769. [Google Scholar] [CrossRef]
  70. Pradhan, G.; Shrivastva, R.; Mukhopadhyay, S. Mycobacterial PknG Targets the Rab7l1 Signaling Pathway to Inhibit Phagosome-Lysosome Fusion. J. Immunol. 2018, 201, 1421–1433. [Google Scholar] [CrossRef]
  71. Clemens, D.L.; Lee, B.Y.; Horwitz, M.A. Purification, characterization, and genetic analysis of Mycobacterium tuberculosis urease, a potentially critical determinant of host-pathogen interaction. J. Bacteriol. 1995, 177, 5644–5652. [Google Scholar] [CrossRef]
  72. Augenstreich, J.; Arbues, A.; Simeone, R.; Haanappel, E.; Wegener, A.; Sayes, F.; Le Chevalier, F.; Chalut, C.; Malaga, W.; Guilhot, C.; et al. ESX-1 and phthiocerol dimycocerosates of Mycobacterium tuberculosis act in concert to cause phagosomal rupture and host cell apoptosis. Cell. Microbiol. 2017, 19, e12726. [Google Scholar] [CrossRef]
  73. Kim, K.H.; An, D.R.; Song, J.; Yoon, J.Y.; Kim, H.S.; Yoon, H.J.; Im, H.N.; Kim, J.; Kim, D.J.; Lee, S.J.; et al. Mycobacterium tuberculosis Eis protein initiates suppression of host immune responses by acetylation of DUSP16/MKP-7. Proc. Natl. Acad. Sci. USA 2012, 109, 7729–7734. [Google Scholar] [CrossRef]
  74. Miller, J.L.; Velmurugan, K.; Cowan, M.J.; Briken, V. The type I NADH dehydrogenase of Mycobacterium tuberculosis counters phagosomal NOX2 activity to inhibit TNF-alpha-mediated host cell apoptosis. PLoS Pathog. 2010, 6, e1000864. [Google Scholar] [CrossRef]
  75. Sun, J.; Singh, V.; Lau, A.; Stokes, R.W.; Obregon-Henao, A.; Orme, I.M.; Wong, D.; Av-Gay, Y.; Hmama, Z. Mycobacterium tuberculosis nucleoside diphosphate kinase inactivates small GTPases leading to evasion of innate immunity. PLoS Pathog. 2013, 9, e1003499. [Google Scholar] [CrossRef]
  76. Piddington, D.L.; Fang, F.C.; Laessig, T.; Cooper, A.M.; Orme, I.M.; Buchmeier, N.A. Cu, Zn superoxide dismutase of Mycobacterium tuberculosis contributes to survival in activated macrophages that are generating an oxidative burst. Infect. Immun. 2001, 69, 4980–4987. [Google Scholar] [CrossRef]
  77. Fan, L.; Wu, X.; Jin, C.; Li, F.; Xiong, S.; Dong, Y. MptpB Promotes Mycobacteria Survival by Inhibiting the Expression of Inflammatory Mediators and Cell Apoptosis in Macrophages. Front. Cell. Infect. Microbiol. 2018, 8, 171. [Google Scholar] [CrossRef] [PubMed]
  78. Bhat, K.H.; Srivastava, S.; Kotturu, S.K.; Ghosh, S.; Mukhopadhyay, S. The PPE2 protein of Mycobacterium tuberculosis translocates to host nucleus and inhibits nitric oxide production. Sci. Rep. 2017, 7, 39706. [Google Scholar] [CrossRef] [PubMed]
  79. Thi, E.P.; Hong, C.J.; Sanghera, G.; Reiner, N.E. Identification of the Mycobacterium tuberculosis protein PE-PGRS62 as a novel effector that functions to block phagosome maturation and inhibit iNOS expression. Cell. Microbiol. 2013, 15, 795–808. [Google Scholar] [CrossRef] [PubMed]
  80. Fishbein, S.; van Wyk, N.; Warren, R.M.; Sampson, S.L. Phylogeny to function: PE/PPE protein evolution and impact on Mycobacterium tuberculosis pathogenicity. Mol. Microbiol. 2015, 96, 901–916. [Google Scholar] [CrossRef]
  81. Yaseen, I.; Kaur, P.; Nandicoori, V.K.; Khosla, S. Mycobacteria modulate host epigenetic machinery by Rv1988 methylation of a non-tail arginine of histone H3. Nat. Commun. 2015, 6, 8922. [Google Scholar] [CrossRef]
  82. Sharma, G.; Upadhyay, S.; Srilalitha, M.; Nandicoori, V.K.; Khosla, S. The interaction of mycobacterial protein Rv2966c with host chromatin is mediated through non-CpG methylation and histone H3/H4 binding. Nucleic Acids Res. 2015, 43, 3922–3937. [Google Scholar] [CrossRef]
  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, 1229–1234. [Google Scholar] [CrossRef] [PubMed]
  84. Jose, L.; Ramachandran, R.; Bhagavat, R.; Gomez, R.L.; Chandran, A.; Raghunandanan, S.; Omkumar, R.V.; Chandra, N.; Mundayoor, S.; Kumar, R.A. Hypothetical protein Rv3423.1 of Mycobacterium tuberculosis is a histone acetyltransferase. FEBS J. 2016, 283, 265–281. [Google Scholar] [CrossRef] [PubMed]
  85. Wang, J.; Li, B.X.; Ge, P.P.; Li, J.; Wang, Q.; Gao, G.F.; Qiu, X.B.; Liu, C.H. Mycobacterium tuberculosis suppresses innate immunity by coopting the host ubiquitin system. Nat. Immunol. 2015, 16, 237–245. [Google Scholar] [CrossRef] [PubMed]
  86. Shin, D.M.; Jeon, B.Y.; Lee, H.M.; Jin, H.S.; Yuk, J.M.; Song, C.H.; Lee, S.H.; Lee, Z.W.; Cho, S.N.; Kim, J.M.; et al. Mycobacterium tuberculosis eis regulates autophagy, inflammation, and cell death through redox-dependent signaling. PLoS Pathog. 2010, 6, e1001230. [Google Scholar] [CrossRef] [PubMed]
  87. Pathak, S.K.; Basu, S.; Basu, K.K.; Banerjee, A.; Pathak, S.; Bhattacharyya, A.; Kaisho, T.; Kundu, M.; Basu, J. Direct extracellular interaction between the early secreted antigen ESAT-6 of Mycobacterium tuberculosis and TLR2 inhibits TLR signaling in macrophages. Nat. Immunol. 2007, 8, 610–618. [Google Scholar] [CrossRef]
  88. Wang, L.; Wu, J.; Li, J.; Yang, H.; Tang, T.; Liang, H.; Zuo, M.; Wang, J.; Liu, H.; Liu, F.; et al. Host-mediated ubiquitination of a mycobacterial protein suppresses immunity. Nature 2020, 577, 682–688. [Google Scholar] [CrossRef] [PubMed]
  89. Wu, F.L.; Liu, Y.; Zhang, H.N.; Jiang, H.W.; Cheng, L.; Guo, S.J.; Deng, J.Y.; Bi, L.J.; Zhang, X.E.; Gao, H.F.; et al. Global Profiling of PknG Interactions Using a Human Proteome Microarray Reveals Novel Connections with CypA. Proteomics 2018, 18, e1800265. [Google Scholar] [CrossRef]
  90. Vemula, M.H.; Medisetti, R.; Ganji, R.; Jakkala, K.; Sankati, S.; Chatti, K.; Banerjee, S. Mycobacterium tuberculosis Zinc Metalloprotease-1 Assists Mycobacterial Dissemination in Zebrafish. Front. Microbiol. 2016, 7, 1347. [Google Scholar] [CrossRef]
  91. Sun, J.; Siroy, A.; Lokareddy, R.K.; Speer, A.; Doornbos, K.S.; Cingolani, G.; Niederweis, M. The tuberculosis necrotizing toxin kills macrophages by hydrolyzing NAD. Nat. Struct. Mol. Biol. 2015, 22, 672–678. [Google Scholar] [CrossRef]
  92. Tundup, S.; Mohareer, K.; Hasnain, S.E. Mycobacterium tuberculosis PE25/PPE41 protein complex induces necrosis in macrophages: Role in virulence and disease reactivation? FEBS Open Bio. 2014, 4, 822–828. [Google Scholar] [CrossRef]
  93. Qiang, L.; Zhang, Y.; Lei, Z.; Lu, Z.; Tan, S.; Ge, P.; Chai, Q.; Zhao, M.; Zhang, X.; Li, B.; et al. A mycobacterial effector promotes ferroptosis-dependent pathogenicity and dissemination. Nat. Commun. 2023, 14, 1430. [Google Scholar] [CrossRef]
  94. Wang, J.; Teng, J.L.; Zhao, D.; Ge, P.; Li, B.; Woo, P.C.; Liu, C.H. The ubiquitin ligase TRIM27 functions as a host restriction factor antagonized by Mycobacterium tuberculosis PtpA during mycobacterial infection. Sci. Rep. 2016, 6, 34827. [Google Scholar] [CrossRef]
  95. Danelishvili, L.; Yamazaki, Y.; Selker, J.; Bermudez, L.E. Secreted Mycobacterium tuberculosis Rv3654c and Rv3655c proteins participate in the suppression of macrophage apoptosis. PLoS ONE 2010, 5, e10474. [Google Scholar] [CrossRef]
  96. Zhang, W.; Lu, Q.; Dong, Y.; Yue, Y.; Xiong, S. Rv3033, as an Emerging Anti-apoptosis Factor, Facilitates Mycobacteria Survival via Inhibiting Macrophage Intrinsic Apoptosis. Front. Immunol. 2018, 9, 2136. [Google Scholar] [CrossRef]
  97. Chai, Q.; Yu, S.; Zhong, Y.; Lu, Z.; Qiu, C.; Yu, Y.; Zhang, X.; Zhang, Y.; Lei, Z.; Qiang, L.; et al. A bacterial phospholipid phosphatase inhibits host pyroptosis by hijacking ubiquitin. Science 2022, 378, eabq0132. [Google Scholar] [CrossRef]
  98. Rastogi, S.; Ellinwood, S.; Augenstreich, J.; Mayer-Barber, K.D.; Briken, V. Mycobacterium tuberculosis inhibits the NLRP3 inflammasome activation via its phosphokinase PknF. PLoS Pathog. 2021, 17, e1009712. [Google Scholar] [CrossRef]
  99. Danelishvili, L.; Everman, J.L.; McNamara, M.J.; Bermudez, L.E. Inhibition of the Plasma-Membrane-Associated Serine Protease Cathepsin G by Mycobacterium tuberculosis Rv3364c Suppresses Caspase-1 and Pyroptosis in Macrophages. Front. Microbiol. 2011, 2, 281. [Google Scholar] [CrossRef]
  100. Hu, D.; Wu, J.; Wang, W.; Mu, M.; Zhao, R.; Xu, X.; Chen, Z.; Xiao, J.; Hu, F.; Yang, Y.; et al. Autophagy regulation revealed by SapM-induced block of autophagosome-lysosome fusion via binding RAB7. Biochem. Biophys. Res. Commun. 2015, 461, 401–407. [Google Scholar] [CrossRef]
  101. Gengenbacher, M.; Nieuwenhuizen, N.; Vogelzang, A.; Liu, H.; Kaiser, P.; Schuerer, S.; Lazar, D.; Wagner, I.; Mollenkopf, H.J.; Kaufmann, S.H. Deletion of nuoG from the Vaccine Candidate Mycobacterium bovis BCG DeltaureC::hly Improves Protection against Tuberculosis. mBio 2016, 7. [Google Scholar] [CrossRef]
  102. Strong, E.J.; Ng, T.W.; Porcelli, S.A.; Lee, S. Mycobacterium tuberculosis PE_PGRS20 and PE_PGRS47 Proteins Inhibit Autophagy by Interaction with Rab1A. mSphere 2021, 6, e0054921. [Google Scholar] [CrossRef]
  103. Ge, P.; Lei, Z.; Yu, Y.; Lu, Z.; Qiang, L.; Chai, Q.; Zhang, Y.; Zhao, D.; Li, B.; Pang, Y.; et al. M. tuberculosis PknG manipulates host autophagy flux to promote pathogen intracellular survival. Autophagy 2022, 18, 576–594. [Google Scholar] [CrossRef]
  104. Padhi, A.; Pattnaik, K.; Biswas, M.; Jagadeb, M.; Behera, A.; Sonawane, A. Mycobacterium tuberculosis LprE Suppresses TLR2-Dependent Cathelicidin and Autophagy Expression to Enhance Bacterial Survival in Macrophages. J. Immunol. 2019, 203, 2665–2678. [Google Scholar] [CrossRef]
  105. Strong, E.J.; Wang, J.; Ng, T.W.; Porcelli, S.A.; Lee, S. Mycobacterium tuberculosis PPE51 Inhibits Autophagy by Suppressing Toll-Like Receptor 2-Dependent Signaling. mBio 2022, 13, e0297421. [Google Scholar] [CrossRef]
  106. Horwitz, M.A.; Harth, G.; Dillon, B.J.; Maslesa-Galic, S. Recombinant bacillus calmette-guerin (BCG) vaccines expressing the Mycobacterium tuberculosis 30-kDa major secretory protein induce greater protective immunity against tuberculosis than conventional BCG vaccines in a highly susceptible animal model. Proc. Natl. Acad. Sci. USA 2000, 97, 13853–13858. [Google Scholar] [CrossRef]
  107. Jagannath, C.; Lindsey, D.R.; Dhandayuthapani, S.; Xu, Y.; Hunter, R.L., Jr.; Eissa, N.T. Autophagy enhances the efficacy of BCG vaccine by increasing peptide presentation in mouse dendritic cells. Nat. Med. 2009, 15, 267–276. [Google Scholar] [CrossRef]
  108. Khan, A.; Bakhru, P.; Saikolappan, S.; Das, K.; Soudani, E.; Singh, C.R.; Estrella, J.L.; Zhang, D.; Pasare, C.; Ma, Y.; et al. An autophagy-inducing and TLR-2 activating BCG vaccine induces a robust protection against tuberculosis in mice. NPJ Vaccines 2019, 4, 34. [Google Scholar] [CrossRef]
  109. Singh, A.K.; Srikrishna, G.; Bivalacqua, T.J.; Bishai, W.R. Recombinant BCGs for tuberculosis and bladder cancer. Vaccine 2021, 39, 7321–7331. [Google Scholar] [CrossRef]
  110. Nieuwenhuizen, N.E.; Kaufmann, S.H.E. Next-Generation Vaccines Based on Bacille Calmette-Guerin. Front. Immunol. 2018, 9, 121. [Google Scholar] [CrossRef]
  111. Wiker, H.G.; Harboe, M. The antigen 85 complex: A major secretion product of Mycobacterium tuberculosis. Microbiol. Rev. 1992, 56, 648–661. [Google Scholar] [CrossRef]
  112. Karbalaei Zadeh Babaki, M.; Soleimanpour, S.; Rezaee, S.A. Antigen 85 complex as a powerful Mycobacterium tuberculosis immunogene: Biology, immune-pathogenicity, applications in diagnosis, and vaccine design. Microb. Pathog. 2017, 112, 20–29. [Google Scholar] [CrossRef]
  113. Belisle, J.T.; Vissa, V.D.; Sievert, T.; Takayama, K.; Brennan, P.J.; Besra, G.S. Role of the major antigen of Mycobacterium tuberculosis in cell wall biogenesis. Science 1997, 276, 1420–1422. [Google Scholar] [CrossRef]
  114. Armitige, L.Y.; Jagannath, C.; Wanger, A.R.; Norris, S.J. Disruption of the genes encoding antigen 85A and antigen 85B of Mycobacterium tuberculosis H37Rv: Effect on growth in culture and in macrophages. Infect. Immun. 2000, 68, 767–778. [Google Scholar] [CrossRef]
  115. Copenhaver, R.H.; Sepulveda, E.; Armitige, L.Y.; Actor, J.K.; Wanger, A.; Norris, S.J.; Hunter, R.L.; Jagannath, C. A mutant of Mycobacterium tuberculosis H37Rv that lacks expression of antigen 85A is attenuated in mice but retains vaccinogenic potential. Infect. Immun. 2004, 72, 7084–7095. [Google Scholar] [CrossRef]
  116. Roche, C.M.; Smith, A.; Lindsey, D.R.; Meher, A.; Schluns, K.; Arora, A.; Armitige, L.Y.; Jagannath, C. The DeltafbpA attenuated candidate vaccine from Mycobacterium tuberculosis, H37Rv primes for a stronger T-bet dependent Th1 immunity in mice. Tuberculosis 2011, 91 (Suppl. S1), S96–S104. [Google Scholar] [CrossRef]
  117. Jain, R.; Dey, B.; Dhar, N.; Rao, V.; Singh, R.; Gupta, U.D.; Katoch, V.M.; Ramanathan, V.D.; Tyagi, A.K. Enhanced and enduring protection against tuberculosis by recombinant BCG-Ag85C and its association with modulation of cytokine profile in lung. PLoS ONE 2008, 3, e3869. [Google Scholar] [CrossRef]
  118. Prendergast, K.A.; Counoupas, C.; Leotta, L.; Eto, C.; Bitter, W.; Winter, N.; Triccas, J.A. The Ag85B protein of the BCG vaccine facilitates macrophage uptake but is dispensable for protection against aerosol Mycobacterium tuberculosis infection. Vaccine 2016, 34, 2608–2615. [Google Scholar] [CrossRef]
  119. Copin, R.; Coscolla, M.; Efstathiadis, E.; Gagneux, S.; Ernst, J.D. Impact of in vitro evolution on antigenic diversity of Mycobacterium bovis bacillus Calmette-Guerin (BCG). Vaccine 2014, 32, 5998–6004. [Google Scholar] [CrossRef]
  120. Armitige, L.Y. Role of FbpA and FbpB in the Pathogenesis and Mycolyltransferase Activity of Mycobacterium Tuberculosis. Ph.D. Thesis, University of Texas Health Sciences Center Houston, Houston, TX, USA, 2002. [Google Scholar]
  121. Patin, E.C.; Geffken, A.C.; Willcocks, S.; Leschczyk, C.; Haas, A.; Nimmerjahn, F.; Lang, R.; Ward, T.H.; Schaible, U.E. Trehalose dimycolate interferes with FcgammaR-mediated phagosome maturation through Mincle, SHP-1 and FcgammaRIIB signalling. PLoS ONE 2017, 12, e0174973. [Google Scholar] [CrossRef]
  122. Mishra, A.; Singh, V.; Saikolappan, S.; Das, K.; Veerapandian, R.; Granica, O.; Khan, A.; Dhandayuthapani, S.; Jagannath, C. The ΔfbpAΔsapM candidate vaccine derived from Mycobacterium tuberculosis H37Rv is markedly immunogenic in macrophages and induces robust immunity to tuberculosis in mice. Front. Immunol. 2023. submitted. [Google Scholar]
  123. Solans, L.; Gonzalo-Asensio, J.; Sala, C.; Benjak, A.; Uplekar, S.; Rougemont, J.; Guilhot, C.; Malaga, W.; Martin, C.; Cole, S.T. The PhoP-dependent ncRNA Mcr7 modulates the TAT secretion system in Mycobacterium tuberculosis. PLoS Pathog. 2014, 10, e1004183. [Google Scholar] [CrossRef]
  124. Romao, S.; Munz, C. LC3-associated phagocytosis. Autophagy 2014, 10, 526–528. [Google Scholar] [CrossRef]
  125. Koster, S.; Upadhyay, S.; Chandra, P.; Papavinasasundaram, K.; Yang, G.; Hassan, A.; Grigsby, S.J.; Mittal, E.; Park, H.S.; Jones, V.; et al. Mycobacterium tuberculosis is protected from NADPH oxidase and LC3-associated phagocytosis by the LCP protein CpsA. Proc. Natl. Acad. Sci. USA 2017, 114, E8711–E8720. [Google Scholar] [CrossRef]
  126. Chandra, P.; Grigsby, S.J.; Philips, J.A. Immune evasion and provocation by Mycobacterium tuberculosis. Nat. Rev. Microbiol. 2022, 20, 750–766. [Google Scholar] [CrossRef]
  127. Brightbill, H.D.; Libraty, D.H.; Krutzik, S.R.; Yang, R.B.; Belisle, J.T.; Bleharski, J.R.; Maitland, M.; Norgard, M.V.; Plevy, S.E.; Smale, S.T.; et al. Host defense mechanisms triggered by microbial lipoproteins through toll-like receptors. Science 1999, 285, 732–736. [Google Scholar] [CrossRef]
  128. Ciaramella, A.; Cavone, A.; Santucci, M.B.; Garg, S.K.; Sanarico, N.; Bocchino, M.; Galati, D.; Martino, A.; Auricchio, G.; D’Orazio, M.; et al. Induction of apoptosis and release of interleukin-1 beta by cell wall-associated 19-kDa lipoprotein during the course of mycobacterial infection. J. Infect. Dis. 2004, 190, 1167–1176. [Google Scholar] [CrossRef]
  129. Stewart, G.R.; Wilkinson, K.A.; Newton, S.M.; Sullivan, S.M.; Neyrolles, O.; Wain, J.R.; Patel, J.; Pool, K.L.; Young, D.B.; Wilkinson, R.J. Effect of deletion or overexpression of the 19-kilodalton lipoprotein Rv3763 on the innate response to Mycobacterium tuberculosis. Infect. Immun. 2005, 73, 6831–6837. [Google Scholar] [CrossRef]
  130. Henao-Tamayo, M.; Junqueira-Kipnis, A.P.; Ordway, D.; Gonzales-Juarrero, M.; Stewart, G.R.; Young, D.B.; Wilkinson, R.J.; Basaraba, R.J.; Orme, I.M. A mutant of Mycobacterium tuberculosis lacking the 19-kDa lipoprotein Rv3763 is highly attenuated in vivo but retains potent vaccinogenic properties. Vaccine 2007, 25, 7153–7159. [Google Scholar] [CrossRef]
  131. Gehring, A.J.; Dobos, K.M.; Belisle, J.T.; Harding, C.V.; Boom, W.H. Mycobacterium tuberculosis LprG (Rv1411c): A novel TLR-2 ligand that inhibits human macrophage class II MHC antigen processing. J. Immunol. 2004, 173, 2660–2668. [Google Scholar] [CrossRef]
  132. Drage, M.G.; Tsai, H.C.; Pecora, N.D.; Cheng, T.Y.; Arida, A.R.; Shukla, S.; Rojas, R.E.; Seshadri, C.; Moody, D.B.; Boom, W.H.; et al. Mycobacterium tuberculosis lipoprotein LprG (Rv1411c) binds triacylated glycolipid agonists of Toll-like receptor 2. Nat. Struct. Mol. Biol. 2010, 17, 1088–1095. [Google Scholar] [CrossRef]
  133. Martinot, A.J.; Farrow, M.; Bai, L.; Layre, E.; Cheng, T.Y.; Tsai, J.H.; Iqbal, J.; Annand, J.W.; Sullivan, Z.A.; Hussain, M.M.; et al. Mycobacterial Metabolic Syndrome: LprG and Rv1410 Regulate Triacylglyceride Levels, Growth Rate and Virulence in Mycobacterium tuberculosis. PLoS Pathog. 2016, 12, e1005351. [Google Scholar] [CrossRef]
  134. Martinot, A.J.; Blass, E.; Yu, J.; Aid, M.; Mahrokhian, S.H.; Cohen, S.B.; Plumlee, C.R.; Larocca, R.A.; Siddiqi, N.; Wakabayashi, S.; et al. Protective efficacy of an attenuated Mtb DeltaLprG vaccine in mice. PLoS Pathog. 2020, 16, e1009096. [Google Scholar] [CrossRef]
  135. Vidal, S.J.; Sellers, D.; Yu, J.; Wakabayashi, S.; Sixsmith, J.; Aid, M.; Barrett, J.; Stevens, S.F.; Liu, X.; Li, W.; et al. Attenuated Mycobacterium tuberculosis vaccine protection in a low-dose murine challenge model. iScience 2023, 26, 106963. [Google Scholar] [CrossRef]
  136. Nairz, M.; Schroll, A.; Sonnweber, T.; Weiss, G. The struggle for iron—A metal at the host-pathogen interface. Cell. Microbiol. 2010, 12, 1691–1702. [Google Scholar] [CrossRef]
  137. Malen, H.; Berven, F.S.; Fladmark, K.E.; Wiker, H.G. Comprehensive analysis of exported proteins from Mycobacterium tuberculosis H37Rv. Proteomics 2007, 7, 1702–1718. [Google Scholar] [CrossRef]
  138. de Souza, G.A.; Leversen, N.A.; Malen, H.; Wiker, H.G. Bacterial proteins with cleaved or uncleaved signal peptides of the general secretory pathway. J. Proteomics 2011, 75, 502–510. [Google Scholar] [CrossRef]
  139. Pandey, R.; Rodriguez, G.M. A ferritin mutant of Mycobacterium tuberculosis is highly susceptible to killing by antibiotics and is unable to establish a chronic infection in mice. Infect. Immun. 2012, 80, 3650–3659. [Google Scholar] [CrossRef]
  140. Subbian, S.; Pandey, R.; Soteropoulos, P.; Rodriguez, G.M. Vaccination with an Attenuated Ferritin Mutant Protects Mice against Virulent Mycobacterium tuberculosis. J. Immunol. Res. 2015, 2015, 385402. [Google Scholar] [CrossRef]
  141. Stefanovic, C.; Hager, F.F.; Schaffer, C. LytR-CpsA-Psr Glycopolymer Transferases: Essential Bricks in Gram-Positive Bacterial Cell Wall Assembly. Int. J. Mol. Sci. 2021, 22, 908. [Google Scholar] [CrossRef]
  142. Koster, S.; Klevorn, T.; Papavinasasundaram, K.; Sassetti, C.M.; Portal-Celhay, C.; Philips, J.A. Consequence of enhanced LC3-trafficking for a live, attenuated M. tuberculosis vaccine. Vaccine 2018, 36, 939–944. [Google Scholar] [CrossRef]
  143. Dey, S.; Lane, J.M.; Lee, R.E.; Rubin, E.J.; Sacchettini, J.C. Structural characterization of the Mycobacterium tuberculosis biotin biosynthesis enzymes 7,8-diaminopelargonic acid synthase and dethiobiotin synthetase. Biochemistry 2010, 49, 6746–6760. [Google Scholar] [CrossRef]
  144. Woong Park, S.; Klotzsche, M.; Wilson, D.J.; Boshoff, H.I.; Eoh, H.; Manjunatha, U.; Blumenthal, A.; Rhee, K.; Barry, C.E., 3rd; Aldrich, C.C.; et al. Evaluating the sensitivity of Mycobacterium tuberculosis to biotin deprivation using regulated gene expression. PLoS Pathog. 2011, 7, e1002264. [Google Scholar] [CrossRef]
  145. Kar, R.; Nangpal, P.; Mathur, S.; Singh, S.; Tyagi, A.K. bioA mutant of Mycobacterium tuberculosis shows severe growth defect and imparts protection against tuberculosis in guinea pigs. PLoS ONE 2017, 12, e0179513. [Google Scholar] [CrossRef]
  146. Harth, G.; Clemens, D.L.; Horwitz, M.A. Glutamine synthetase of Mycobacterium tuberculosis: Extracellular release and characterization of its enzymatic activity. Proc. Natl. Acad. Sci. USA 1994, 91, 9342–9346. [Google Scholar] [CrossRef]
  147. Cole, S.T.; Brosch, R.; Parkhill, J.; Garnier, T.; Churcher, C.; Harris, D.; Gordon, S.V.; Eiglmeier, K.; Gas, S.; Barry, C.E., 3rd; et al. Deciphering the biology of Mycobacterium tuberculosis from the complete genome sequence. Nature 1998, 393, 537–544. [Google Scholar] [CrossRef]
  148. Lee, S.; Jeon, B.Y.; Bardarov, S.; Chen, M.; Morris, S.L.; Jacobs, W.R., Jr. Protection elicited by two glutamine auxotrophs of Mycobacterium tuberculosis and in vivo growth phenotypes of the four unique glutamine synthetase mutants in a murine model. Infect. Immun. 2006, 74, 6491–6495. [Google Scholar] [CrossRef]
  149. Schirmer, S.; Rauh, L.; Alebouyeh, S.; Delgado-Velandia, M.; Salgueiro, V.C.; Lerma, L.; Serrano-Mestre, J.L.; Azkargorta, M.; Elortza, F.; Lavin, J.L.; et al. Immunogenicity of Mycobacterial Extracellular Vesicles Isolated from Host-Related Conditions Informs About Tuberculosis Disease Status. Front. Microbiol. 2022, 13, 907296. [Google Scholar] [CrossRef]
  150. Saleh, M.T.; Belisle, J.T. Secretion of an acid phosphatase (SapM) by Mycobacterium tuberculosis that is similar to eukaryotic acid phosphatases. J. Bacteriol. 2000, 182, 6850–6853. [Google Scholar] [CrossRef]
  151. Pal, R.; Bisht, M.K.; Mukhopadhyay, S. Secretory proteins of Mycobacterium tuberculosis and their roles in modulation of host immune responses: Focus on therapeutic targets. FEBS J. 2022, 289, 4146–4171. [Google Scholar] [CrossRef]
  152. Saikolappan, S.; Estrella, J.; Sasindran, S.J.; Khan, A.; Armitige, L.Y.; Jagannath, C.; Dhandayuthapani, S. The fbpA/sapM double knock out strain of Mycobacterium tuberculosis is highly attenuated and immunogenic in macrophages. PLoS ONE 2012, 7, e36198. [Google Scholar] [CrossRef]
  153. Puri, R.V.; Reddy, P.V.; Tyagi, A.K. Secreted acid phosphatase (SapM) of Mycobacterium tuberculosis is indispensable for arresting phagosomal maturation and growth of the pathogen in guinea pig tissues. PLoS ONE 2013, 8, e70514. [Google Scholar] [CrossRef]
  154. Chauhan, P.; Reddy, P.V.; Singh, R.; Jaisinghani, N.; Gandotra, S.; Tyagi, A.K. Secretory phosphatases deficient mutant of Mycobacterium tuberculosis imparts protection at the primary site of infection in guinea pigs. PLoS ONE 2013, 8, e77930. [Google Scholar] [CrossRef]
  155. Bahal, R.K.; Mathur, S.; Chauhan, P.; Tyagi, A.K. An attenuated quadruple gene mutant of Mycobacterium tuberculosis imparts protection against tuberculosis in guinea pigs. Biol. Open 2018, 7, bio029546. [Google Scholar] [CrossRef]
  156. Festjens, N.; Bogaert, P.; Batni, A.; Houthuys, E.; Plets, E.; Vanderschaeghe, D.; Laukens, B.; Asselbergh, B.; Parthoens, E.; De Rycke, R.; et al. Disruption of the SapM locus in Mycobacterium bovis BCG improves its protective efficacy as a vaccine against M. tuberculosis. EMBO Mol. Med. 2011, 3, 222–234. [Google Scholar] [CrossRef]
  157. Festjens, N.; Vandewalle, K.; Houthuys, E.; Plets, E.; Vanderschaeghe, D.; Borgers, K.; Van Hecke, A.; Tiels, P.; Callewaert, N. SapM mutation to improve the BCG vaccine: Genomic, transcriptomic and preclinical safety characterization. Vaccine 2019, 37, 3539–3551. [Google Scholar] [CrossRef]
  158. Wang, J.; Ge, P.; Qiang, L.; Tian, F.; Zhao, D.; Chai, Q.; Zhu, M.; Zhou, R.; Meng, G.; Iwakura, Y.; et al. The mycobacterial phosphatase PtpA regulates the expression of host genes and promotes cell proliferation. Nat. Commun. 2017, 8, 244. [Google Scholar] [CrossRef]
  159. Tonks, N.K. Protein tyrosine phosphatases: From genes, to function, to disease. Nat. Rev. Mol. Cell. Biol. 2006, 7, 833–846. [Google Scholar] [CrossRef]
  160. Bach, H.; Papavinasasundaram, K.G.; Wong, D.; Hmama, Z.; Av-Gay, Y. Mycobacterium tuberculosis virulence is mediated by PtpA dephosphorylation of human vacuolar protein sorting 33B. Cell Host Microbe 2008, 3, 316–322. [Google Scholar] [CrossRef]
  161. Singh, R.; Rao, V.; Shakila, H.; Gupta, R.; Khera, A.; Dhar, N.; Singh, A.; Koul, A.; Singh, Y.; Naseema, M.; et al. Disruption of mptpB impairs the ability of Mycobacterium tuberculosis to survive in guinea pigs. Mol. Microbiol. 2003, 50, 751–762. [Google Scholar] [CrossRef]
  162. Zhou, B.; He, Y.; Zhang, X.; Xu, J.; Luo, Y.; Wang, Y.; Franzblau, S.G.; Yang, Z.; Chan, R.J.; Liu, Y.; et al. Targeting mycobacterium protein tyrosine phosphatase B for antituberculosis agents. Proc. Natl. Acad. Sci. USA 2010, 107, 4573–4578. [Google Scholar] [CrossRef]
  163. Johansen, P.; Fettelschoss, A.; Amstutz, B.; Selchow, P.; Waeckerle-Men, Y.; Keller, P.; Deretic, V.; Held, L.; Kundig, T.M.; Bottger, E.C.; et al. Relief from Zmp1-mediated arrest of phagosome maturation is associated with facilitated presentation and enhanced immunogenicity of mycobacterial antigens. Clin. Vaccine Immunol. 2011, 18, 907–913. [Google Scholar] [CrossRef]
  164. Sander, P.; Clark, S.; Petrera, A.; Vilaplana, C.; Meuli, M.; Selchow, P.; Zelmer, A.; Mohanan, D.; Andreu, N.; Rayner, E.; et al. Deletion of zmp1 improves Mycobacterium bovis BCG-mediated protection in a guinea pig model of tuberculosis. Vaccine 2015, 33, 1353–1359. [Google Scholar] [CrossRef]
  165. Dahl, J.L.; Wei, J.; Moulder, J.W.; Laal, S.; Friedman, R.L. Subcellular localization of the Iitracellular survival-enhancing Eis protein of Mycobacterium tuberculosis. Infect. Immun. 2001, 69, 4295–4302. [Google Scholar] [CrossRef]
  166. Wei, J.; Dahl, J.L.; Moulder, J.W.; Roberts, E.A.; O’Gaora, P.; Young, D.B.; Friedman, R.L. Identification of a Mycobacterium tuberculosis gene that enhances mycobacterial survival in macrophages. J. Bacteriol. 2000, 182, 377–384. [Google Scholar] [CrossRef]
  167. Samuel, L.P.; Song, C.H.; Wei, J.; Roberts, E.A.; Dahl, J.L.; Barry, C.E.; Jo, E.K.; Friedman, R.L. Expression, production and release of the Eis protein by Mycobacterium tuberculosis during infection of macrophages and its effect on cytokine secretion. Microbiology 2007, 153, 529–540. [Google Scholar] [CrossRef]
  168. Wu, Y.; Tian, M.; Zhang, Y.; Peng, H.; Lei, Q.; Yuan, X.; Liu, S.; Xiong, Y.; Lin, X.; Jo-Lewis, B.N.; et al. Deletion of BCG_2432c from the Bacillus Calmette-Guerin vaccine enhances autophagy-mediated immunity against tuberculosis. Allergy 2022, 77, 619–632. [Google Scholar] [CrossRef]
  169. Sayes, F.; Sun, L.; Di Luca, M.; Simeone, R.; Degaiffier, N.; Fiette, L.; Esin, S.; Brosch, R.; Bottai, D.; Leclerc, C.; et al. Strong immunogenicity and cross-reactivity of Mycobacterium tuberculosis ESX-5 type VII secretion: Encoded PE-PPE proteins predicts vaccine potential. Cell Host Microbe 2012, 11, 352–363. [Google Scholar] [CrossRef]
  170. Tiwari, S.; Dutt, T.S.; Chen, B.; Chen, M.; Kim, J.; Dai, A.Z.; Lukose, R.; Shanley, C.; Fox, A.; Karger, B.R.; et al. BCG-Prime and boost with Esx-5 secretion system deletion mutant leads to better protection against clinical strains of Mycobacterium tuberculosis. Vaccine 2020, 38, 7156–7165. [Google Scholar] [CrossRef]
  171. Saini, N.K.; Baena, A.; Ng, T.W.; Venkataswamy, M.M.; Kennedy, S.C.; Kunnath-Velayudhan, S.; Carreno, L.J.; Xu, J.; Chan, J.; Larsen, M.H.; et al. Suppression of autophagy and antigen presentation by Mycobacterium tuberculosis PE_PGRS47. Nat. Microbiol. 2016, 1, 16133. [Google Scholar] [CrossRef]
  172. Amaral, E.P.; Riteau, N.; Moayeri, M.; Maier, N.; Mayer-Barber, K.D.; Pereira, R.M.; Lage, S.L.; Kubler, A.; Bishai, W.R.; D’Imperio-Lima, M.R.; et al. Lysosomal Cathepsin Release Is Required for NLRP3-Inflammasome Activation by Mycobacterium tuberculosis in Infected Macrophages. Front. Immunol. 2018, 9, 1427. [Google Scholar] [CrossRef]
  173. Romagnoli, A.; Etna, M.P.; Giacomini, E.; Pardini, M.; Remoli, M.E.; Corazzari, M.; Falasca, L.; Goletti, D.; Gafa, V.; Simeone, R.; et al. ESX-1 dependent impairment of autophagic flux by Mycobacterium tuberculosis in human dendritic cells. Autophagy 2012, 8, 1357–1370. [Google Scholar] [CrossRef]
  174. Schaible, U.E.; Sturgill-Koszycki, S.; Schlesinger, P.H.; Russell, D.G. Cytokine activation leads to acidification and increases maturation of Mycobacterium avium-containing phagosomes in murine macrophages. J. Immunol. 1998, 160, 1290–1296. [Google Scholar] [CrossRef]
  175. Honer zu Bentrup, K.; Russell, D.G. Mycobacterial persistence: Adaptation to a changing environment. Trends Microbiol. 2001, 9, 597–605. [Google Scholar] [CrossRef]
  176. Lee, B.Y.; Jethwaney, D.; Schilling, B.; Clemens, D.L.; Gibson, B.W.; Horwitz, M.A. The Mycobacterium bovis bacille Calmette-Guerin phagosome proteome. Mol. Cell. Proteomics 2010, 9, 32–53. [Google Scholar] [CrossRef]
  177. Grode, L.; Seiler, P.; Baumann, S.; Hess, J.; Brinkmann, V.; Nasser Eddine, A.; Mann, P.; Goosmann, C.; Bandermann, S.; Smith, D.; et al. Increased vaccine efficacy against tuberculosis of recombinant Mycobacterium bovis bacille Calmette-Guerin mutants that secrete listeriolysin. J. Clin. Investig. 2005, 115, 2472–2479. [Google Scholar] [CrossRef]
  178. Desel, C.; Dorhoi, A.; Bandermann, S.; Grode, L.; Eisele, B.; Kaufmann, S.H. Recombinant BCG DeltaureC hly+ induces superior protection over parental BCG by stimulating a balanced combination of type 1 and type 17 cytokine responses. J. Infect. Dis. 2011, 204, 1573–1584. [Google Scholar] [CrossRef]
  179. Kaufmann, S.H.; Cotton, M.F.; Eisele, B.; Gengenbacher, M.; Grode, L.; Hesseling, A.C.; Walzl, G. The BCG replacement vaccine VPM1002: From drawing board to clinical trial. Expert Rev. Vaccines 2014, 13, 619–630. [Google Scholar] [CrossRef]
  180. Martin, C.; Williams, A.; Hernandez-Pando, R.; Cardona, P.J.; Gormley, E.; Bordat, Y.; Soto, C.Y.; Clark, S.O.; Hatch, G.J.; Aguilar, D.; et al. The live Mycobacterium tuberculosis phoP mutant strain is more attenuated than BCG and confers protective immunity against tuberculosis in mice and guinea pigs. Vaccine 2006, 24, 3408–3419. [Google Scholar] [CrossRef]
  181. Verreck, F.A.; Vervenne, R.A.; Kondova, I.; van Kralingen, K.W.; Remarque, E.J.; Braskamp, G.; van der Werff, N.M.; Kersbergen, A.; Ottenhoff, T.H.; Heidt, P.J.; et al. MVA.85A boosting of BCG and an attenuated, phoP deficient M. tuberculosis vaccine both show protective efficacy against tuberculosis in rhesus macaques. PLoS ONE 2009, 4, e5264. [Google Scholar] [CrossRef]
  182. Hinchey, J.; Lee, S.; Jeon, B.Y.; Basaraba, R.J.; Venkataswamy, M.M.; Chen, B.; Chan, J.; Braunstein, M.; Orme, I.M.; Derrick, S.C.; et al. Enhanced priming of adaptive immunity by a proapoptotic mutant of Mycobacterium tuberculosis. J. Clin. Investig. 2007, 117, 2279–2288. [Google Scholar] [CrossRef]
  183. Hinchey, J.; Jeon, B.Y.; Alley, H.; Chen, B.; Goldberg, M.; Derrick, S.; Morris, S.; Jacobs, W.R., Jr.; Porcelli, S.A.; Lee, S. Lysine auxotrophy combined with deletion of the SecA2 gene results in a safe and highly immunogenic candidate live attenuated vaccine for tuberculosis. PLoS ONE 2011, 6, e15857. [Google Scholar] [CrossRef]
  184. White, A.D.; Sibley, L.; Sarfas, C.; Morrison, A.; Gullick, J.; Clark, S.; Gleeson, F.; McIntyre, A.; Arlehamn, C.L.; Sette, A.; et al. MTBVAC vaccination protects rhesus macaques against aerosol challenge with M. tuberculosis and induces immune signatures analogous to those observed in clinical studies. NPJ Vaccines 2021, 6, 4. [Google Scholar] [CrossRef] [PubMed]
  185. Arbues, A.; Aguilo, J.I.; Gonzalo-Asensio, J.; Marinova, D.; Uranga, S.; Puentes, E.; Fernandez, C.; Parra, A.; Cardona, P.J.; Vilaplana, C.; et al. Construction, characterization and preclinical evaluation of MTBVAC, the first live-attenuated M. tuberculosis-based vaccine to enter clinical trials. Vaccine 2013, 31, 4867–4873. [Google Scholar] [CrossRef]
  186. Solans, L.; Uranga, S.; Aguilo, N.; Arnal, C.; Gomez, A.B.; Monzon, M.; Badiola, J.J.; Gicquel, B.; Martin, C. Hyper-attenuated MTBVAC erp mutant protects against tuberculosis in mice. Vaccine 2014, 32, 5192–5197. [Google Scholar] [CrossRef]
  187. Braunstein, M.; Espinosa, B.J.; Chan, J.; Belisle, J.T.; Jacobs, W.R., Jr. SecA2 functions in the secretion of superoxide dismutase A and in the virulence of Mycobacterium tuberculosis. Mol. Microbiol. 2003, 48, 453–464. [Google Scholar] [CrossRef]
  188. Kurtz, S.; McKinnon, K.P.; Runge, M.S.; Ting, J.P.; Braunstein, M. The SecA2 secretion factor of Mycobacterium tuberculosis promotes growth in macrophages and inhibits the host immune response. Infect. Immun. 2006, 74, 6855–6864. [Google Scholar] [CrossRef]
  189. Perez, E.; Samper, S.; Bordas, Y.; Guilhot, C.; Gicquel, B.; Martin, C. An essential role for phoP in Mycobacterium tuberculosis virulence. Mol. Microbiol. 2001, 41, 179–187. [Google Scholar] [CrossRef]
  190. Gonzalo-Asensio, J.; Mostowy, S.; Harders-Westerveen, J.; Huygen, K.; Hernandez-Pando, R.; Thole, J.; Behr, M.; Gicquel, B.; Martin, C. PhoP: A missing piece in the intricate puzzle of Mycobacterium tuberculosis virulence. PLoS ONE 2008, 3, e3496. [Google Scholar] [CrossRef]
  191. Lee, J.S.; Krause, R.; Schreiber, J.; Mollenkopf, H.J.; Kowall, J.; Stein, R.; Jeon, B.Y.; Kwak, J.Y.; Song, M.K.; Patron, J.P.; et al. Mutation in the transcriptional regulator PhoP contributes to avirulence of Mycobacterium tuberculosis H37Ra strain. Cell Host Microbe 2008, 3, 97–103. [Google Scholar] [CrossRef]
  192. Frigui, W.; Bottai, D.; Majlessi, L.; Monot, M.; Josselin, E.; Brodin, P.; Garnier, T.; Gicquel, B.; Martin, C.; Leclerc, C.; et al. Control of M. tuberculosis ESAT-6 secretion and specific T cell recognition by PhoP. PLoS Pathog. 2008, 4, e33. [Google Scholar] [CrossRef]
  193. Anil Kumar, V.; Goyal, R.; Bansal, R.; Singh, N.; Sevalkar, R.R.; Kumar, A.; Sarkar, D. EspR-dependent ESAT-6 Protein Secretion of Mycobacterium tuberculosis Requires the Presence of Virulence Regulator PhoP. J. Biol. Chem. 2016, 291, 19018–19030. [Google Scholar] [CrossRef]
  194. Broset, E.; Martin, C.; Gonzalo-Asensio, J. Evolutionary landscape of the Mycobacterium tuberculosis complex from the viewpoint of PhoPR: Implications for virulence regulation and application to vaccine development. mBio 2015, 6, e01289-15. [Google Scholar] [CrossRef] [PubMed]
  195. Gonzalo-Asensio, J.; Malaga, W.; Pawlik, A.; Astarie-Dequeker, C.; Passemar, C.; Moreau, F.; Laval, F.; Daffe, M.; Martin, C.; Brosch, R.; et al. Evolutionary history of tuberculosis shaped by conserved mutations in the PhoPR virulence regulator. Proc. Natl. Acad. Sci. USA 2014, 111, 11491–11496. [Google Scholar] [CrossRef] [PubMed]
  196. Aguilo, N.; Gonzalo-Asensio, J.; Alvarez-Arguedas, S.; Marinova, D.; Gomez, A.B.; Uranga, S.; Spallek, R.; Singh, M.; Audran, R.; Spertini, F.; et al. Reactogenicity to major tuberculosis antigens absent in BCG is linked to improved protection against Mycobacterium tuberculosis. Nat. Commun. 2017, 8, 16085. [Google Scholar] [CrossRef] [PubMed]
  197. Kamath, A.T.; Fruth, U.; Brennan, M.J.; Dobbelaer, R.; Hubrechts, P.; Ho, M.M.; Mayner, R.E.; Thole, J.; Walker, K.B.; Liu, M.; et al. New live mycobacterial vaccines: The Geneva consensus on essential steps towards clinical development. Vaccine 2005, 23, 3753–3761. [Google Scholar] [CrossRef]
  198. Clark, S.; Lanni, F.; Marinova, D.; Rayner, E.; Martin, C.; Williams, A. Revaccination of Guinea Pigs With the Live Attenuated Mycobacterium tuberculosis Vaccine MTBVAC Improves BCG’s Protection Against Tuberculosis. J. Infect. Dis. 2017, 216, 525–533. [Google Scholar] [CrossRef] [PubMed]
  199. Tameris, M.; Mearns, H.; Penn-Nicholson, A.; Gregg, Y.; Bilek, N.; Mabwe, S.; Geldenhuys, H.; Shenje, J.; Luabeya, A.K.K.; Murillo, I.; et al. Live-attenuated Mycobacterium tuberculosis vaccine MTBVAC versus BCG in adults and neonates: A randomised controlled, double-blind dose-escalation trial. Lancet Respir. Med. 2019, 7, 757–770. [Google Scholar] [CrossRef]
  200. Clemmensen, H.S.; Dube, J.Y.; McIntosh, F.; Rosenkrands, I.; Jungersen, G.; Aagaard, C.; Andersen, P.; Behr, M.A.; Mortensen, R. In vivo antigen expression regulates CD4 T cell differentiation and vaccine efficacy against Mycobacterium tuberculosis infection. bioRxiv 2021, 12, 10–1128. [Google Scholar] [CrossRef]
  201. Chandran, A.; Williams, K.; Mendum, T.; Stewart, G.; Clark, S.; Zadi, S.; Lanni, F.; McLeod, N.; Williams, A.; Villarreal-Ramos, B.; et al. Development of a diagnostic compatible BCG vaccine against Bovine tuberculosis. Sci. Rep. 2019, 9, 17791. [Google Scholar] [CrossRef]
  202. Bigi, F.; Alito, A.; Fisanotti, J.C.; Romano, M.I.; Cataldi, A. Characterization of a novel Mycobacterium bovis secreted antigen containing PGLTS repeats. Infect. Immun. 1995, 63, 2581–2586. [Google Scholar] [CrossRef]
  203. Berthet, F.X.; Lagranderie, M.; Gounon, P.; Laurent-Winter, C.; Ensergueix, D.; Chavarot, P.; Thouron, F.; Maranghi, E.; Pelicic, V.; Portnoï, D.; et al. Attenuation of virulence by disruption of the Mycobacterium tuberculosis erp gene. Science 1998, 282, 759–762. [Google Scholar] [CrossRef] [PubMed]
  204. Cosma, C.L.; Klein, K.; Kim, R.; Beery, D.; Ramakrishnan, L. Mycobacterium marinum Erp is a virulence determinant required for cell wall integrity and intracellular survival. Infect. Immun. 2006, 74, 3125–3133. [Google Scholar] [CrossRef] [PubMed]
  205. Flores-Valdez, M.A.; Aceves-Sanchez Mde, J.; Pedroza-Roldan, C.; Vega-Dominguez, P.J.; Prado-Montes de Oca, E.; Bravo-Madrigal, J.; Laval, F.; Daffe, M.; Koestler, B.; Waters, C.M. The Cyclic Di-GMP Phosphodiesterase Gene Rv1357c/BCG1419c Affects BCG Pellicle Production and In Vivo Maintenance. IUBMB Life 2015, 67, 129–138. [Google Scholar] [CrossRef] [PubMed]
  206. Velazquez-Fernandez, J.B.; Ferreira-Souza, G.H.M.; Rodriguez-Campos, J.; Aceves-Sanchez, M.J.; Bravo-Madrigal, J.; Vallejo-Cardona, A.A.; Flores-Valdez, M.A. Proteomic characterization of a second-generation version of the BCGDeltaBCG1419c vaccine candidate by means of electrospray-ionization quadrupole time-of-flight mass spectrometry. Pathog. Dis. 2021, 79, ftaa070. [Google Scholar] [CrossRef] [PubMed]
  207. Aceves-Sanchez, M.J.; Flores-Valdez, M.A.; Pedroza-Roldan, C.; Creissen, E.; Izzo, L.; Silva-Angulo, F.; Dawson, C.; Izzo, A.; Bielefeldt-Ohmann, H.; Segura-Cerda, C.A.; et al. Vaccination with BCGDeltaBCG1419c protects against pulmonary and extrapulmonary TB and is safer than BCG. Sci. Rep. 2021, 11, 12417. [Google Scholar] [CrossRef]
  208. Korotetskaya, M.; Baikuzina, P.; Segura-Cerda, C.A.; Aceves-Sánchez, M.J.; Apt, A.; Flores-Valdez, M.A. BCG and BCGΔBCG1419c transiently protect hypersusceptible I/St mice and induce different influx of macrophages and neutrophils during pulmonary tuberculosis. J. Med. Microbiol. 2022, 71, 001485. [Google Scholar] [CrossRef]
  209. Pedroza-Roldán, C.; Guapillo, C.; Barrios-Payán, J.; Mata-Espinosa, D.; Aceves-Sánchez Mde, J.; Marquina-Castillo, B.; Hernández-Pando, R.; Flores-Valdez, M.A. The BCGΔBCG1419c strain, which produces more pellicle in vitro, improves control of chronic tuberculosis in vivo. Vaccine 2016, 34, 4763–4770. [Google Scholar] [CrossRef] [PubMed]
  210. Flores-Valdez, M.A.; Pedroza-Roldán, C.; Aceves-Sánchez, M.J.; Peterson, E.J.R.; Baliga, N.S.; Hernández-Pando, R.; Troudt, J.; Creissen, E.; Izzo, L.; Bielefeldt-Ohmann, H.; et al. The BCGΔBCG1419c Vaccine Candidate Reduces Lung Pathology, IL-6, TNF-α, and IL-10 During Chronic TB Infection. Front. Microbiol. 2018, 9, 1281. [Google Scholar] [CrossRef]
  211. Kwon, K.W.; Aceves-Sánchez, M.J.; Segura-Cerda, C.A.; Choi, E.; Bielefeldt-Ohmann, H.; Shin, S.J.; Flores-Valdez, M.A. BCGΔBCG1419c increased memory CD8(+) T cell-associated immunogenicity and mitigated pulmonary inflammation compared with BCG in a model of chronic tuberculosis. Sci. Rep. 2022, 12, 15824. [Google Scholar] [CrossRef]
  212. Aceves-Sanchez, M.J.; Barrios-Payan, J.A.; Segura-Cerda, C.A.; Flores-Valdez, M.A.; Mata-Espinosa, D.; Pedroza-Roldan, C.; Yadav, R.; Saini, D.K.; de la Cruz, M.A.; Ares, M.A.; et al. BCG∆BCG1419c and BCG differ in induction of autophagy, c-di-GMP content, proteome, and progression of lung pathology in Mycobacterium tuberculosis HN878-infected male BALB/c mice. Vaccine 2023, 41, 3824–3835. [Google Scholar] [CrossRef]
  213. Segura-Cerda, C.A.; Marquina-Castillo, B.; Lozano-Ordaz, V.; Mata-Espinosa, D.; Barrios-Payán, J.A.; López-Torres, M.O.; Aceves-Sánchez, M.J.; Bielefeldt-Ohmann, H.; Hernández-Pando, R.; Flores-Valdez, M.A. BCG and BCGΔBCG1419c protect type 2 diabetic mice against tuberculosis via different participation of T and B lymphocytes, dendritic cells and pro-inflammatory cytokines. NPJ Vaccines 2020, 5, 21. [Google Scholar] [CrossRef]
  214. Zulauf, K.E.; Sullivan, J.T.; Braunstein, M. The SecA2 pathway of Mycobacterium tuberculosis exports effectors that work in concert to arrest phagosome and autophagosome maturation. PLoS Pathog. 2018, 14, e1007011. [Google Scholar] [CrossRef] [PubMed]
  215. Katti, M.K.; Dai, G.; Armitige, L.Y.; Rivera Marrero, C.; Daniel, S.; Singh, C.R.; Lindsey, D.R.; Dhandayuthapani, S.; Hunter, R.L.; Jagannath, C. The Delta fbpA mutant derived from Mycobacterium tuberculosis H37Rv has an enhanced susceptibility to intracellular antimicrobial oxidative mechanisms, undergoes limited phagosome maturation and activates macrophages and dendritic cells. Cell Microbiol. 2008, 10, 1286–1303. [Google Scholar] [CrossRef]
  216. Roche, P.A.; Furuta, K. The ins and outs of MHC class II-mediated antigen processing and presentation. Nat Rev Immunol 2015, 15, 203–216. [Google Scholar] [CrossRef]
  217. Kotsias, F.; Cebrian, I.; Alloatti, A. Antigen processing and presentation. Int. Rev. Cell Mol. Biol. 2019, 348, 69–121. [Google Scholar] [CrossRef]
  218. Germic, N.; Frangez, Z.; Yousefi, S.; Simon, H.U. Regulation of the innate immune system by autophagy: Monocytes, macrophages, dendritic cells and antigen presentation. Cell Death Differ. 2019, 26, 715–727. [Google Scholar] [CrossRef]
  219. Kuan, R.; Muskat, K.; Peters, B.; Lindestam Arlehamn, C.S. Is mapping the BCG vaccine-induced immune responses the key to improving the efficacy against tuberculosis? J. Intern. Med. 2020, 288, 651–660. [Google Scholar] [CrossRef] [PubMed]
  220. Brazier, B.; McShane, H. Towards new TB vaccines. Semin. Immunopathol. 2020, 42, 315–331. [Google Scholar] [CrossRef]
  221. Lewinsohn, D.A.; Lewinsohn, D.M.; Scriba, T.J. Polyfunctional CD4(+) T Cells As Targets for Tuberculosis Vaccination. Front. Immunol. 2017, 8, 1262. [Google Scholar] [CrossRef]
  222. Satti, I.; McShane, H. Current approaches toward identifying a correlate of immune protection from tuberculosis. Expert Rev. Vaccines 2019, 18, 43–59. [Google Scholar] [CrossRef]
  223. Andersen, P.; Woodworth, J.S. Tuberculosis vaccines--rethinking the current paradigm. Trends Immunol. 2014, 35, 387–395. [Google Scholar] [CrossRef]
  224. Andersen, P.; Kaufmann, S.H. Novel vaccination strategies against tuberculosis. Cold Spring Harb. Perspect. Med. 2014, 4, a018523. [Google Scholar] [CrossRef]
  225. Lin, Y.; Slight, S.R.; Khader, S.A. Th17 cytokines and vaccine-induced immunity. Semin. Immunopathol. 2010, 32, 79–90. [Google Scholar] [CrossRef]
  226. Khader, S.A.; Divangahi, M.; Hanekom, W.; Hill, P.C.; Maeurer, M.; Makar, K.W.; Mayer-Barber, K.D.; Mhlanga, M.M.; Nemes, E.; Schlesinger, L.S.; et al. Targeting innate immunity for tuberculosis vaccination. J. Clin. Investig. 2019, 129, 3482–3491. [Google Scholar] [CrossRef]
  227. Netea, M.G.; Joosten, L.A.; Latz, E.; Mills, K.H.; Natoli, G.; Stunnenberg, H.G.; O’Neill, L.A.; Xavier, R.J. Trained immunity: A program of innate immune memory in health and disease. Science 2016, 352, aaf1098. [Google Scholar] [CrossRef]
  228. Bettencourt, P.J.G. The 100(th) anniversary of bacille Calmette-Guérin (BCG) and the latest vaccines against COVID-19. Int. J. Tuberc. Lung Dis. 2021, 25, 611–613. [Google Scholar] [CrossRef]
  229. Buffen, K.; Oosting, M.; Quintin, J.; Ng, A.; Kleinnijenhuis, J.; Kumar, V.; van de Vosse, E.; Wijmenga, C.; van Crevel, R.; Oosterwijk, E.; et al. Autophagy controls BCG-induced trained immunity and the response to intravesical BCG therapy for bladder cancer. PLoS Pathog. 2014, 10, e1004485. [Google Scholar] [CrossRef]
  230. Xu, Y.; Wan, W. Acetylation in the regulation of autophagy. Autophagy 2023, 19, 379–387. [Google Scholar] [CrossRef]
  231. DiNardo, A.R.; Rajapakshe, K.; Nishiguchi, T.; Grimm, S.L.; Mtetwa, G.; Dlamini, Q.; Kahari, J.; Mahapatra, S.; Kay, A.; Maphalala, G.; et al. DNA hypermethylation during tuberculosis dampens host immune responsiveness. J. Clin. Investig. 2020, 130, 3113–3123. [Google Scholar] [CrossRef]
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

Veerapandian, R.; Gadad, S.S.; Jagannath, C.; Dhandayuthapani, S. Live Attenuated Vaccines against Tuberculosis: Targeting the Disruption of Genes Encoding the Secretory Proteins of Mycobacteria. Vaccines 2024, 12, 530. https://doi.org/10.3390/vaccines12050530

AMA Style

Veerapandian R, Gadad SS, Jagannath C, Dhandayuthapani S. Live Attenuated Vaccines against Tuberculosis: Targeting the Disruption of Genes Encoding the Secretory Proteins of Mycobacteria. Vaccines. 2024; 12(5):530. https://doi.org/10.3390/vaccines12050530

Chicago/Turabian Style

Veerapandian, Raja, Shrikanth S. Gadad, Chinnaswamy Jagannath, and Subramanian Dhandayuthapani. 2024. "Live Attenuated Vaccines against Tuberculosis: Targeting the Disruption of Genes Encoding the Secretory Proteins of Mycobacteria" Vaccines 12, no. 5: 530. https://doi.org/10.3390/vaccines12050530

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