1
|
Menon AR, Prest RJ, Tobin DM, Champion PA. Mycobacterium marinum as a model for understanding principles of mycobacterial pathogenesis. J Bacteriol 2025; 207:e0004725. [PMID: 40304497 PMCID: PMC12096832 DOI: 10.1128/jb.00047-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025] Open
Abstract
Mycobacterium marinum is a fish pathogen that has become a powerful and well-established model that has accelerated our understanding of the mechanisms of mycobacterial disease. M. marinum is a versatile surrogate for understanding the closely related human pathogen M. tuberculosis, which causes tuberculosis in humans. M. marinum has defined key mechanisms of pathogenesis, both shared with M. tuberculosis and unique to this species. In this review, we discuss the discovery of M. marinum as an occasional human pathogen, the shared aspects of pathogenesis with M. tuberculosis, and how M. marinum has been exploited as a model to define the molecular mechanisms of mycobacterial pathogenesis across several phases of infection.
Collapse
Affiliation(s)
- Aruna R. Menon
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Rebecca J. Prest
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - David M. Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Patricia A. Champion
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, USA
| |
Collapse
|
2
|
Santamaria C, Biegas KJ, Lim PN, Cabral J, Kim CY, Lee JR, Gaidhane IV, Papson C, Gomard-Henshaw K, Rothchild AC, Swarts BM, Siegrist MS. Trehalose dimycolate inhibits phagosome maturation and promotes intracellular M. tuberculosis growth via noncanonical SNARE interactions. Proc Natl Acad Sci U S A 2025; 122:e2423292122. [PMID: 40354525 PMCID: PMC12107153 DOI: 10.1073/pnas.2423292122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 03/26/2025] [Indexed: 05/14/2025] Open
Abstract
Mycobacterial cell envelopes are rich in unusual lipids and glycans that play key roles during infection and vaccination. The most abundant envelope glycolipid is trehalose dimycolate (TDM). TDM compromises the host response to mycobacterial species via multiple mechanisms, including inhibition of phagosome maturation. The molecular mechanism by which TDM inhibits phagosome maturation has been elusive. We find that a clickable, photoaffinity TDM probe recapitulates key phenotypes of native TDM in macrophage host cells and binds several host Soluble N-ethylmaleimide-Sensitive Factor Attachment Proteins Receptor (SNARE) proteins, including Vesicle Transport through Interaction with t-SNAREs 1B (VTI1B), Syntaxin 8 (STX8), and Vesicle-Associated Membrane Protein 2 (VAMP2). VTI1B and STX8 normally promote endosome fusion by forming a complex with VAMP8. However, in the presence of Mycobacterium tuberculosis, VTI1B and STX8 complex with VAMP2, which in turn decreases VAMP8 binding. VAMP2 acts together with mycolate structure to inhibit phagosome maturation and promotes intracellular M. tuberculosis replication. Thus one mechanism by which TDM constrains the innate immune response to M. tuberculosis is via noncanonical SNARE complexation.
Collapse
Affiliation(s)
- Carolina Santamaria
- Molecular and Cellular Biology Program, College of Natural Sciences, University of Massachusetts, Amherst, MA01003
- Department of Microbiology, University of Massachusetts, Amherst, MA01003
| | - Kyle J. Biegas
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI48859
- Biochemistry, Cell, and Molecular Biology Graduate Program, College of Science and Engineering, Central Michigan University, Mount Pleasant, MI48859
| | - Pamelia N. Lim
- Molecular and Cellular Biology Program, College of Natural Sciences, University of Massachusetts, Amherst, MA01003
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA01003
| | - Jessica Cabral
- Department of Microbiology, University of Massachusetts, Amherst, MA01003
| | - Christi Y. Kim
- Department of Microbiology, University of Massachusetts, Amherst, MA01003
| | - James R. Lee
- Department of Microbiology, University of Massachusetts, Amherst, MA01003
| | - Ishani V. Gaidhane
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI48859
| | - Casey Papson
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI48859
| | - Kyla Gomard-Henshaw
- Molecular and Cellular Biology Program, College of Natural Sciences, University of Massachusetts, Amherst, MA01003
- Department of Microbiology, University of Massachusetts, Amherst, MA01003
| | - Alissa C. Rothchild
- Molecular and Cellular Biology Program, College of Natural Sciences, University of Massachusetts, Amherst, MA01003
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA01003
| | - Benjamin M. Swarts
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI48859
- Biochemistry, Cell, and Molecular Biology Graduate Program, College of Science and Engineering, Central Michigan University, Mount Pleasant, MI48859
| | - M. Sloan Siegrist
- Molecular and Cellular Biology Program, College of Natural Sciences, University of Massachusetts, Amherst, MA01003
- Department of Microbiology, University of Massachusetts, Amherst, MA01003
| |
Collapse
|
3
|
Zhang L, Fang F, Liu D, Xia G, Feng T, Lv J, Qi J, Li T, Liu H, Xu T, Wu F, Song C, Li W, Wang X, Chang X, Wang H, Wang T, Qian Z. Early secretory antigen target of 6-kDa of Mycobacterium tuberculosis inhibits macrophage apoptosis and host defense via TLR2. Respir Res 2025; 26:131. [PMID: 40205554 PMCID: PMC11983766 DOI: 10.1186/s12931-025-03210-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Abstract
Mycobacterium tuberculosis (M. tb) is an intracellular pathogen adept at evading the human immune system through a variety of mechanisms. During infection, M. tb secretes numerous virulence factors, including the 6 kDa early secretory antigen target (ESAT-6), which is produced by the ESX-1 secretion system. ESAT-6 plays a crucial role in host-pathogen interactions, either independently or in association with culture filtrate protein 10 (CFP-10). While some research has investigated the role of ESAT-6 in M. tb pathogenicity and vaccine development, its precise contribution to immune evasion and the cellular mechanisms involved remain poorly understood. To address this, we used cultured THP-1(A) macrophages to characterize the effects of secreted ESAT-6 on cellular host defenses and apoptosis. We found that ESAT-6 (5 μg/ml) inhibited M. tb-induced apoptosis in THP-1(A) macrophages by suppressing Toll-like receptor 2 (TLR2) through the Caspase-9/Caspase-3 pathway. Additionally, ESAT-6 reduced phagocytosis of M. tb by THP-1(A) macrophages by downregulating the production of interleukin-10 (IL-10), tumor necrosis factor-alpha (TNF-α), and interleukin-12 (IL-12). Furthermore, ESAT-6 diminished the bactericidal activity of macrophages by inducing reactive oxygen species (ROS) production. In parallel, our in silico analysis of differentially expressed genes in dendritic cells (DCs) infected with Bacille Calmette-Guérin (BCG) strains, with or without the region of difference-1 (RD1) gene, strongly suggests that ESAT-6, located within the RD1 region, modulates host defense functions and apoptosis in DCs during BCG infection. Collectively, these findings indicate that ESAT-6 plays a pivotal role in modulating the innate immune response of macrophages against M. tb by regulating macrophage recognition, phagocytosis, bactericidal activity, and apoptosis. Our study provides valuable insights into potential molecular targets for the development of innovative vaccines and therapeutic strategies against M. tb.
Collapse
Affiliation(s)
- Lin Zhang
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
- Yiwu Traditional Chinese Medicine Hospital, Jinhua, Zhejiang, China
| | - Fang Fang
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Danrui Liu
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Geman Xia
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Tong Feng
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Jingzhu Lv
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Jinying Qi
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Tengteng Li
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Hui Liu
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Tao Xu
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Fengjiao Wu
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Chuanwang Song
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Wei Li
- Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Department of Respiration, First Affiliated Hospital, Bengbu Medical University, Bengbu, Anhui, China
| | - Xiaojing Wang
- Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Department of Respiration, First Affiliated Hospital, Bengbu Medical University, Bengbu, Anhui, China
| | - Xianyou Chang
- The Infectious Disease Hospital of Bengbu City, Bengbu, Anhui, China
| | - Hongtao Wang
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Ting Wang
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA.
- Center for Translational Science, Florida International University, Port Saint Lucie, FL, USA.
| | - Zhongqing Qian
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China.
| |
Collapse
|
4
|
Rahim MA, Seo H, Barman I, Hossain MS, Shuvo MSH, Song HY. Insights into Autophagy in Microbiome Therapeutic Approaches for Drug-Resistant Tuberculosis. Cells 2025; 14:540. [PMID: 40214493 PMCID: PMC11989032 DOI: 10.3390/cells14070540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/23/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
Tuberculosis, primarily caused by Mycobacterium tuberculosis, is an airborne lung disease and continues to pose a significant global health threat, resulting in millions of deaths annually. The current treatment for tuberculosis involves a prolonged regimen of antibiotics, which leads to complications such as recurrence, drug resistance, reinfection, and a range of side effects. This scenario underscores the urgent need for novel therapeutic strategies to combat this lethal pathogen. Over the last two decades, microbiome therapeutics have emerged as promising next-generation drug candidates, offering advantages over traditional medications. In 2022, the Food and Drug Administration approved the first microbiome therapeutic for recurrent Clostridium infections, and extensive research is underway on microbiome treatments for various challenging diseases, including metabolic disorders and cancer. Research on microbiomes concerning tuberculosis commenced roughly a decade ago, and the scope of this research has broadened considerably over the last five years, with microbiome therapeutics now viewed as viable options for managing drug-resistant tuberculosis. Nevertheless, the understanding of their mechanisms is still in its infancy. Although autophagy has been extensively studied in other diseases, research into its role in tuberculosis is just beginning, with preliminary developments in progress. Against this backdrop, this comprehensive review begins by succinctly outlining tuberculosis' characteristics and assessing existing treatments' strengths and weaknesses, followed by a detailed examination of microbiome-based therapeutic approaches for drug-resistant tuberculosis. Additionally, this review focuses on establishing a basic understanding of microbiome treatments for tuberculosis, mainly through the lens of autophagy as a mechanism of action. Ultimately, this review aims to contribute to the foundational comprehension of microbiome-based therapies for tuberculosis, thereby setting the stage for the further advancement of microbiome therapeutics for drug-resistant tuberculosis.
Collapse
Affiliation(s)
- Md Abdur Rahim
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Hoonhee Seo
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
- Probiotics Microbiome Commercialization Research Center (PMC), Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Indrajeet Barman
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Mohammed Solayman Hossain
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Md Sarower Hossen Shuvo
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Ho-Yeon Song
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
- Probiotics Microbiome Commercialization Research Center (PMC), Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| |
Collapse
|
5
|
Santamaria C, Biegas KJ, Lim PN, Cabral J, Kim CY, Lee JR, Gaidhane IV, Papson C, Gomard-Henshaw K, Rothchild AC, Swarts BM, Siegrist MS. Trehalose dimycolate inhibits phagosome maturation and promotes intracellular M. tuberculosis growth via noncanonical SNARE interaction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.16.627577. [PMID: 39763847 PMCID: PMC11702582 DOI: 10.1101/2024.12.16.627577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Mycobacterial cell envelopes are rich in unusual lipids and glycans that play key roles during infection and vaccination. The most abundant envelope glycolipid is trehalose dimycolate (TDM). TDM compromises the host response to mycobacterial species via multiple mechanisms, including inhibition of phagosome maturation. The molecular mechanism by which TDM inhibits phagosome maturation has been elusive. We find that a clickable, photoaffinity TDM probe recapitulates key phenotypes of native TDM in macrophage host cells and binds several host SNARE proteins, including VTI1B, STX8, and VAMP2. VTI1B and STX8 normally promote endosome fusion by forming a complex with VAMP8. However, in the presence of Mycobacterium tuberculosis , VTI1B and STX8 complex with VAMP2, which in turn decreases VAMP8 binding. VAMP2 acts together with mycolate structure to inhibit phagosome maturation and promotes intracellular M. tuberculosis replication. Thus one mechanism by which TDM constrains the innate immune response to M. tuberculosis is via non-canonical SNARE complexation. Significance Statement Glycolipids from the Mycobacterium tuberculosis cell envelope, particularly trehalose dimycolate (TDM), play major roles in subverting the immune response to this intracellular pathogen. How subversion occurs is often unclear because glycans and lipids are technically challenging to study in cells. We discovered that a TDM-mimicking chemical probe interacts with three host SNARE proteins, including two that regulate endosome fusion and one that does not. The presence of TDM or M. tuberculosis triggers abnormal binding of these SNAREs, which in turn inhibits the fusion of M. tuberculosis -containing phagosomes with lysosomes and promotes M. tuberculosis replication. Our work provides an unusual example of a bacterial pathogen restricting the immune response via glycolipid-SNARE interactions.
Collapse
|
6
|
Augenstreich J, Phan AT, Allen CNS, Poddar A, Chen H, Srinivasan L, Briken V. Dynamic interplay of autophagy and membrane repair during Mycobacterium tuberculosis Infection. PLoS Pathog 2025; 21:e1012830. [PMID: 39746091 PMCID: PMC11731705 DOI: 10.1371/journal.ppat.1012830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 01/14/2025] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
Autophagy plays a crucial role in the host response to Mycobacterium tuberculosis (Mtb) infection, yet the dynamics and regulation of autophagy induction on Mtb-containing vacuoles (MCVs) remain only partially understood. We employed time-lapse confocal microscopy to investigate the recruitment of LC3B (LC3), a key autophagy marker, to MCVs at the single cell level with our newly developed workflow for single cell and single MCV tracking and fluorescence quantification. We show that approximately 70% of MCVs exhibited LC3 recruitment but that was lost in about 40% of those MCVs. The LC3 recruitment to MCVs displayed a high variability in timing that was independent of the size of the MCV or the bacterial burden. Most notably, the LC3-positive MCVs did not acidify, indicating that LC3 recruitment does not necessarily lead to the formation of mature autophagolysosomes. Interferon-gamma pre-treatment did not affect LC3 recruitment frequency or autophagosome acidification but increased the susceptibility of the macrophage to Mtb-induced cell death. LC3 recruitment and lysotracker staining were mutually exclusive events, alternating on some MCVs multiple times thus demonstrating a reversible aspect of the autophagy response. The LC3 recruitment was associated with galectin-3 and oxysterol-binding protein 1 staining, indicating a correlation with membrane damage and repair mechanisms. ATG7 knock-down did not impact membrane repair, suggesting that autophagy is not directly involved in this process but is coregulated by the membrane damage of MCVs. In summary, our findings provide novel insights into the dynamic and variable nature of LC3 recruitment to the MCVs over time during Mtb infection. Our data does not support a role for autophagy in either cell-autonomous defense against Mtb or membrane repair of the MCV in human macrophages. In addition, the combined dynamics of LC3 recruitment and Lysoview staining emerged as promising markers for investigating the damage and repair processes of phagosomal membranes.
Collapse
Affiliation(s)
- Jacques Augenstreich
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland United States of America
| | - Anna T. Phan
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland United States of America
| | - Charles N. S. Allen
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland United States of America
| | - Anushka Poddar
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland United States of America
| | - Hanzhang Chen
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland United States of America
| | - Lalitha Srinivasan
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland United States of America
| | - Volker Briken
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland United States of America
| |
Collapse
|
7
|
Mani R, Balu KE, Suzuki Y. Deficiencies of Inducible Costimulator (ICOS) During Chronic Infection with Toxoplasma gondii Upregulate the CD28-Dependent Cytotoxicity of CD8 + T Cells and Their Effector Function Against Tissue Cysts of the Parasite. Cells 2024; 13:1998. [PMID: 39682747 PMCID: PMC11640114 DOI: 10.3390/cells13231998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/21/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
We recently identified that the cerebral mRNA expression of inducible costimulator (ICOS) and its ligand, ICOSL, both significantly increase during the elimination of Toxoplasma gondii cysts from the brains of infected mice by the perforin-mediated cytotoxic activity of CD8+ T cells. In the present study, we examined the role of ICOS in activating the effector activity of CD8+ T cells in response to the presence of cysts in infected mice. Following the adoptive transfer of splenic CD8+ T cells from chronically infected ICOS-deficient (ICOS-/-) and wild-type (WT) mice to infected SCID mice, fewer CD8+ T cells were detected in the brains of the recipients of ICOS-/- CD8+ T cells than the recipients of WT CD8+ T cells. Interestingly, even with the lower migration rate of the ICOS-/- CD8+ T cells, those T cells eliminated T. gondii cysts more efficiently than WT CD8+ T cells did in the brains of the recipient mice. Consistently, the ICOS-/- CD8+ T cells secreted greater amounts of granzyme B in response to T. gondii antigens in vitro than WT CD8+ T cells did. We identified that CD8+ T cells of infected ICOS-/- mice express significantly greater levels of CD28 on their surface than CD8+ T cells of infected WT mice, and the relative expression of CD28 mRNA to CD8β mRNA levels in the brains of the recipients of those CD8+ T cells were strongly correlated with their relative expression levels of mRNA for T-bet transcription factors and perforin. Furthermore, blocking CD28 signaling using a combination of anti-CD80 and anti-CD86 antibodies eliminated the increased cytotoxic activity of the ICOS-/- CD8+ T cells in vitro. The present study uncovered notable compensatory interactions between ICOS and CD28, which protected the cytotoxic effector activity of CD8+ T cells against microbial infection in a murine model of chronic infection with T. gondii.
Collapse
Affiliation(s)
| | | | - Yasuhiro Suzuki
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA; (R.M.); (K.E.B.)
| |
Collapse
|
8
|
Jones BS, Pareek V, Hu DD, Weaver SD, Syska C, Galfano G, Champion MM, Champion PA. N - acetyl-transferases required for iron uptake and aminoglycoside resistance promote virulence lipid production in M. marinum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.05.602253. [PMID: 39005365 PMCID: PMC11245092 DOI: 10.1101/2024.07.05.602253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Phagosomal lysis is a key aspect of mycobacterial infection of host macrophages. Acetylation is a protein modification mediated enzymatically by N-acetyltransferases (NATs) that impacts bacterial pathogenesis and physiology. To identify NATs required for lytic activity, we leveraged Mycobacterium marinum, a nontubercular pathogen and an established model for M. tuberculosis. M. marinum hemolysis is a proxy for phagolytic activity. We generated M. marinum strains with deletions in conserved NAT genes and screened for hemolytic activity. Several conserved lysine acetyltransferases (KATs) contributed to hemolysis. Hemolysis is mediated by the ESX-1 secretion system and by phthiocerol dimycocerosate (PDIM), a virulence lipid. For several strains, the hemolytic activity was restored by the addition of second copy of the ESX-1 locus. Using thin-layer chromatography (TLC), we found a single NAT required for PDIM and phenolic glycolipid (PGL) production. MbtK is a conserved KAT required for mycobactin siderophore synthesis and virulence. Mycobactin J exogenously complemented PDIM/PGL production in the Δ mbtK strain. The Δ mbtK M. marinum strain was attenuated in macrophage and Galleria mellonella infection models. Constitutive expression of either eis or papA5, which encode a KAT required for aminoglycoside resistance and a PDIM/PGL biosynthetic enzyme, rescued PDIM/PGL production and virulence of the Δ mbtK strain. Eis N-terminally acetylated PapA5 in vitro , supporting a mechanism for restored lipid production. Overall, our study establishes connections between the MbtK and Eis NATs, and between iron uptake and PDIM and PGL synthesis in M. marinum . Our findings underscore the multifunctional nature of mycobacterial NATs and their connection to key virulence pathways. Significance Statement Acetylation is a modification of protein N-termini, lysine residues, antibiotics and lipids. Many of the enzymes that promote acetylation belong to the GNAT family of proteins. M. marinum is a well-established as a model to understand how M. tuberculosis causes tuberculosis. In this study we sought to identify conserved GNAT proteins required for early stages of mycobacterial infection. Using M. marinum, we determined that several GNAT proteins are required for the lytic activity of M. marinum. We uncovered previously unknown connections between acetyl-transferases required for iron uptake and antimicrobial resistance, and the production of the unique mycobacterial lipids, PDIM and PGLOur data support that acetyl-transferases from the GNAT family are interconnected, and have activities beyond those previously reported.
Collapse
|
9
|
Maure A, Lawarée E, Fiorentino F, Pawlik A, Gona S, Giraud-Gatineau A, Eldridge MJG, Danckaert A, Hardy D, Frigui W, Keck C, Gutierrez C, Neyrolles O, Aulner N, Mai A, Hamon M, Barreiro LB, Brodin P, Brosch R, Rotili D, Tailleux L. A host-directed oxadiazole compound potentiates antituberculosis treatment via zinc poisoning in human macrophages and in a mouse model of infection. PLoS Biol 2024; 22:e3002259. [PMID: 38683873 PMCID: PMC11081512 DOI: 10.1371/journal.pbio.3002259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 05/09/2024] [Accepted: 03/13/2024] [Indexed: 05/02/2024] Open
Abstract
Antituberculosis drugs, mostly developed over 60 years ago, combined with a poorly effective vaccine, have failed to eradicate tuberculosis. More worryingly, multiresistant strains of Mycobacterium tuberculosis (MTB) are constantly emerging. Innovative strategies are thus urgently needed to improve tuberculosis treatment. Recently, host-directed therapy has emerged as a promising strategy to be used in adjunct with existing or future antibiotics, by improving innate immunity or limiting immunopathology. Here, using high-content imaging, we identified novel 1,2,4-oxadiazole-based compounds, which allow human macrophages to control MTB replication. Genome-wide gene expression analysis revealed that these molecules induced zinc remobilization inside cells, resulting in bacterial zinc intoxication. More importantly, we also demonstrated that, upon treatment with these novel compounds, MTB became even more sensitive to antituberculosis drugs, in vitro and in vivo, in a mouse model of tuberculosis. Manipulation of heavy metal homeostasis holds thus great promise to be exploited to develop host-directed therapeutic interventions.
Collapse
Affiliation(s)
- Alexandra Maure
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Emeline Lawarée
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Francesco Fiorentino
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Alexandre Pawlik
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Saideep Gona
- Department of Genetic Medicine, University of Chicago, Chicago, Illinois, United States of America
| | | | | | - Anne Danckaert
- Institut Pasteur, Université Paris Cité, UTechS BioImaging-C2RT, Paris, France
| | - David Hardy
- Institut Pasteur, Université Paris Cité, Histopathology Platform, Paris, France
| | - Wafa Frigui
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Camille Keck
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Claude Gutierrez
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Olivier Neyrolles
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Nathalie Aulner
- Institut Pasteur, Université Paris Cité, UTechS BioImaging-C2RT, Paris, France
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
- Pasteur Institute, Cenci-bolognetti Foundation, Sapienza University of Rome, Rome, Italy
| | - Mélanie Hamon
- Institut Pasteur, Université Paris Cité, Chromatine et Infection unit, Paris, France
| | - Luis B. Barreiro
- Department of Genetic Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Priscille Brodin
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Roland Brosch
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Ludovic Tailleux
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| |
Collapse
|
10
|
McIntyre S, Warner J, Rush C, Vanderven HA. Antibodies as clinical tools for tuberculosis. Front Immunol 2023; 14:1278947. [PMID: 38162666 PMCID: PMC10755875 DOI: 10.3389/fimmu.2023.1278947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Tuberculosis (TB) is a leading cause of morbidity and mortality worldwide. Global research efforts to improve TB control are hindered by insufficient understanding of the role that antibodies play in protective immunity and pathogenesis. This impacts knowledge of rational and optimal vaccine design, appropriate diagnostic biomarkers, and development of therapeutics. Traditional approaches for the prevention and diagnosis of TB may be less efficacious in high prevalence, remote, and resource-poor settings. An improved understanding of the immune response to the causative agent of TB, Mycobacterium tuberculosis (Mtb), will be crucial for developing better vaccines, therapeutics, and diagnostics. While memory CD4+ T cells and cells and cytokine interferon gamma (IFN-g) have been the main identified correlates of protection in TB, mounting evidence suggests that other types of immunity may also have important roles. TB serology has identified antibodies and functional characteristics that may help diagnose Mtb infection and distinguish between different TB disease states. To date, no serological tests meet the World Health Organization (WHO) requirements for TB diagnosis, but multiplex assays show promise for improving the sensitivity and specificity of TB serodiagnosis. Monoclonal antibody (mAb) therapies and serum passive infusion studies in murine models of TB have also demonstrated some protective outcomes. However, animal models that better reflect the human immune response to Mtb are necessary to fully assess the clinical utility of antibody-based TB prophylactics and therapeutics. Candidate TB vaccines are not designed to elicit an Mtb-specific antibody response, but evidence suggests BCG and novel TB vaccines may induce protective Mtb antibodies. The potential of the humoral immune response in TB monitoring and control is being investigated and these studies provide important insight into the functional role of antibody-mediated immunity against TB. In this review, we describe the current state of development of antibody-based clinical tools for TB, with a focus on diagnostic, therapeutic, and vaccine-based applications.
Collapse
Affiliation(s)
- Sophie McIntyre
- Biomedical Sciences and Molecular Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Douglas, QLD, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Douglas, QLD, Australia
| | - Jeffrey Warner
- Biomedical Sciences and Molecular Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Douglas, QLD, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Douglas, QLD, Australia
| | - Catherine Rush
- Biomedical Sciences and Molecular Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Douglas, QLD, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Douglas, QLD, Australia
| | - Hillary A. Vanderven
- Biomedical Sciences and Molecular Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Douglas, QLD, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Douglas, QLD, Australia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
11
|
Nakayama H, Hanafusa K, Yamaji T, Oshima E, Hotta T, Takamori K, Ogawa H, Iwabuchi K. Phylactic role of anti-lipoarabinomannan IgM directed against mannan core during mycobacterial infection in macrophages. Tuberculosis (Edinb) 2023; 143:102391. [PMID: 37574397 DOI: 10.1016/j.tube.2023.102391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/15/2023]
Abstract
Mycobacteria enter host phagocytes, such as macrophages by binding to several receptors on phagocytes. Several mycobacterial species, including Mycobacterium tuberculosis have evolved systems to evade host bactericidal pathways. Lipoarabinomannan (LAM) is an essential mycobacterial molecule for both binding to phagocytes and escaping from bactericidal pathways. Integrin CD11b plays critical roles as a phagocytic receptor and contributes to host defense by mediating both nonopsonic and opsonic phagocytosis. However, the mechanisms by which CD11b-mediated phagocytosis associates with LAM and drives the phagocytic process of mycobacteria remain to be fully elucidated. We recently identified TMDU3 as anti-LAM IgM antibody against the mannan core of LAM. The present study investigated the roles of CD11b and TMDU3 in macrophage phagocytosis of mycobacteria and subsequent bactericidal lysosomal fusion to phagosomes. CD11b knockout cells generated by a CRISPR/Cas9 system showed significant attenuation of the ability to phagocytose non-opsonized mycobacteria and LAM-conjugated beads. Moreover, recombinant human CD11b protein was found to bind to LAM. TMDU3 markedly inhibited macrophage phagocytosis of non-opsonized mycobacteria. This antibody slightly increased the phagocytosis of mycobacteria under opsonized conditions, whereas it significantly enhanced CD11b-mediated bactericidal functions. Taken together, these results show a novel phylactic role of anti-LAM IgM during mycobacterial infection in macrophages.
Collapse
Affiliation(s)
- Hitoshi Nakayama
- Laboratory of Biochemistry, Juntendo University Faculty of Health Care and Nursing, Urayasu, Chiba, Japan; Institute for Environmental and Gender-specific Medicine, Juntendo University, Graduate School of Medicine, Urayasu, Chiba, Japan; Infection Control Nursing, Juntendo University Graduate School of Health Care and Nursing, Urayasu, Chiba, Japan.
| | - Kei Hanafusa
- Institute for Environmental and Gender-specific Medicine, Juntendo University, Graduate School of Medicine, Urayasu, Chiba, Japan
| | - Toshiyuki Yamaji
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Toyama, Shinjuku-ku, Tokyo, Japan
| | - Eriko Oshima
- Institute for Environmental and Gender-specific Medicine, Juntendo University, Graduate School of Medicine, Urayasu, Chiba, Japan
| | - Tomomi Hotta
- Institute for Environmental and Gender-specific Medicine, Juntendo University, Graduate School of Medicine, Urayasu, Chiba, Japan
| | - Kenji Takamori
- Institute for Environmental and Gender-specific Medicine, Juntendo University, Graduate School of Medicine, Urayasu, Chiba, Japan
| | - Hideoki Ogawa
- Institute for Environmental and Gender-specific Medicine, Juntendo University, Graduate School of Medicine, Urayasu, Chiba, Japan
| | - Kazuhisa Iwabuchi
- Laboratory of Biochemistry, Juntendo University Faculty of Health Care and Nursing, Urayasu, Chiba, Japan; Institute for Environmental and Gender-specific Medicine, Juntendo University, Graduate School of Medicine, Urayasu, Chiba, Japan; Infection Control Nursing, Juntendo University Graduate School of Health Care and Nursing, Urayasu, Chiba, Japan
| |
Collapse
|
12
|
Kaufmann SHE. Vaccine development against tuberculosis before and after Covid-19. Front Immunol 2023; 14:1273938. [PMID: 38035095 PMCID: PMC10684952 DOI: 10.3389/fimmu.2023.1273938] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/16/2023] [Indexed: 12/02/2023] Open
Abstract
Coronavirus disease (Covid-19) has not only shaped awareness of the impact of infectious diseases on global health. It has also provided instructive lessons for better prevention strategies against new and current infectious diseases of major importance. Tuberculosis (TB) is a major current health threat caused by Mycobacterium tuberculosis (Mtb) which has claimed more lives than any other pathogen over the last few centuries. Hence, better intervention measures, notably novel vaccines, are urgently needed to accomplish the goal of the World Health Organization to end TB by 2030. This article describes how the research and development of TB vaccines can benefit from recent developments in the Covid-19 vaccine pipeline from research to clinical development and outlines how the field of TB research can pursue its own approaches. It begins with a brief discussion of major vaccine platforms in general terms followed by a short description of the most widely applied Covid-19 vaccines. Next, different vaccination regimes and particular hurdles for TB vaccine research and development are described. This specifically considers the complex immune mechanisms underlying protection and pathology in TB which involve innate as well as acquired immune mechanisms and strongly depend on fine tuning the response. A brief description of the TB vaccine candidates that have entered clinical trials follows. Finally, it discusses how experiences from Covid-19 vaccine research, development, and rollout can and have been applied to the TB vaccine pipeline, emphasizing similarities and dissimilarities.
Collapse
Affiliation(s)
- Stefan H. E. Kaufmann
- Max Planck Institute for Infection Biology, Berlin, Germany
- Systems Immunology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, TX, United States
| |
Collapse
|
13
|
Liu Y, Chen T, Zhu Y, Furey A, Lowary TL, Chan J, Bournazos S, Ravetch JV, Achkar JM. Features and protective efficacy of human mAbs targeting Mycobacterium tuberculosis arabinomannan. JCI Insight 2023; 8:e167960. [PMID: 37733444 PMCID: PMC10619501 DOI: 10.1172/jci.insight.167960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 09/06/2023] [Indexed: 09/23/2023] Open
Abstract
A better understanding of the epitopes most relevant for antibody-mediated protection against tuberculosis (TB) remains a major knowledge gap. We have shown that human polyclonal IgG against the Mycobacterium tuberculosis (M. tuberculosis) surface glycan arabinomannan (AM) and related lipoarabinomannan (LAM) is protective against TB. To investigate the impact of AM epitope recognition and Fcγ receptor (FcγR) binding on antibody functions against M. tuberculosis, we isolated a high-affinity human monoclonal antibody (mAb; P1AM25) against AM and showed its binding to oligosaccharide (OS) motifs we previously found to be associated with in vitro functions of human polyclonal anti-AM IgG. Human IgG1 P1AM25, but not 2 other high-affinity human IgG1 anti-AM mAbs reactive with different AM OS motifs, enhanced M. tuberculosis phagocytosis by macrophages and reduced intracellular growth in an FcγR-dependent manner. P1AM25 in murine IgG2a, but neither murine IgG1 nor a non-FcγR-binding IgG, given intraperitoneally prior to and after aerosolized M. tuberculosis infection, was protective in C57BL/6 mice. Moreover, we demonstrated the protective efficacy of human IgG1 P1AM25 in passive transfer with M. tuberculosis-infected FcγR-humanized mice. These data enhance our knowledge of the important interplay between both antibody epitope specificity and Fc effector functions in the defense against M. tuberculosis and could inform development of vaccines against TB.
Collapse
Affiliation(s)
- Yanyan Liu
- Department of Microbiology and Immunology and
| | - Tingting Chen
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Yongqi Zhu
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Aisha Furey
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Todd L. Lowary
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - John Chan
- Public Health Research Institute at the International Center for Public Health, New Jersey Medical School – Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | | | | | - Jacqueline M. Achkar
- Department of Microbiology and Immunology and
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
14
|
Anes E, Pires D, Mandal M, Azevedo-Pereira JM. ESAT-6 a Major Virulence Factor of Mycobacterium tuberculosis. Biomolecules 2023; 13:968. [PMID: 37371548 DOI: 10.3390/biom13060968] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/31/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of human tuberculosis (TB), is one of the most successfully adapted human pathogens. Human-to-human transmission occurs at high rates through aerosols containing bacteria, but the pathogen evolved prior to the establishment of crowded populations. Mtb has developed a particular strategy to ensure persistence in the host until an opportunity for transmission arises. It has refined its lifestyle to obviate the need for virulence factors such as capsules, flagella, pili, or toxins to circumvent mucosal barriers. Instead, the pathogen uses host macrophages, where it establishes intracellular niches for its migration into the lung parenchyma and other tissues and for the induction of long-lived latency in granulomas. Finally, at the end of the infection cycle, Mtb induces necrotic cell death in macrophages to escape to the extracellular milieu and instructs a strong inflammatory response that is required for the progression from latency to disease and transmission. Common to all these events is ESAT-6, one of the major virulence factors secreted by the pathogen. This narrative review highlights the recent advances in understanding the role of ESAT-6 in hijacking macrophage function to establish successful infection and transmission and its use as a target for the development of diagnostic tools and vaccines.
Collapse
Affiliation(s)
- Elsa Anes
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - David Pires
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Center for Interdisciplinary Research in Health, Católica Medical School, Universidade Católica Portuguesa, Estrada Octávio Pato, 2635-631 Rio de Mouro, Portugal
| | - Manoj Mandal
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - José Miguel Azevedo-Pereira
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| |
Collapse
|
15
|
Leddy O, White FM, Bryson BD. Immunopeptidomics reveals determinants of Mycobacterium tuberculosis antigen presentation on MHC class I. eLife 2023; 12:e84070. [PMID: 37073954 PMCID: PMC10159623 DOI: 10.7554/elife.84070] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 04/17/2023] [Indexed: 04/20/2023] Open
Abstract
CD8+ T cell recognition of Mycobacterium tuberculosis (Mtb)-specific peptides presented on major histocompatibility complex class I (MHC-I) contributes to immunity to tuberculosis (TB), but the principles that govern presentation of Mtb antigens on MHC-I are incompletely understood. In this study, mass spectrometry (MS) analysis of the MHC-I repertoire of Mtb-infected primary human macrophages reveals that substrates of Mtb's type VII secretion systems (T7SS) are overrepresented among Mtb-derived peptides presented on MHC-I. Quantitative, targeted MS shows that ESX-1 activity is required for presentation of Mtb peptides derived from both ESX-1 substrates and ESX-5 substrates on MHC-I, consistent with a model in which proteins secreted by multiple T7SSs access a cytosolic antigen processing pathway via ESX-1-mediated phagosome permeabilization. Chemical inhibition of proteasome activity, lysosomal acidification, or cysteine cathepsin activity did not block presentation of Mtb antigens on MHC-I, suggesting involvement of other proteolytic pathways or redundancy among multiple pathways. Our study identifies Mtb antigens presented on MHC-I that could serve as targets for TB vaccines, and reveals how the activity of multiple T7SSs interacts to contribute to presentation of Mtb antigens on MHC-I.
Collapse
Affiliation(s)
- Owen Leddy
- Department of Biological Engineering, Massachusetts Institute of TechnologyCambridgeUnited States
- Ragon Institute of Massachusetts General Hospital, Harvard, and MITCambridgeUnited States
- Koch Institute for Integrative Cancer ResearchCambridgeUnited States
| | - Forest M White
- Department of Biological Engineering, Massachusetts Institute of TechnologyCambridgeUnited States
- Koch Institute for Integrative Cancer ResearchCambridgeUnited States
- Center for Precision Cancer MedicineCambridgeUnited States
| | - Bryan D Bryson
- Department of Biological Engineering, Massachusetts Institute of TechnologyCambridgeUnited States
- Ragon Institute of Massachusetts General Hospital, Harvard, and MITCambridgeUnited States
| |
Collapse
|
16
|
Pires D, Mandal M, Matos AI, Peres C, Catalão MJ, Azevedo-Pereira JM, Satchi-Fainaro R, Florindo HF, Anes E. Development of Chitosan Particles Loaded with siRNA for Cystatin C to Control Intracellular Drug-Resistant Mycobacterium tuberculosis. Antibiotics (Basel) 2023; 12:729. [PMID: 37107091 PMCID: PMC10135320 DOI: 10.3390/antibiotics12040729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
The golden age of antibiotics for tuberculosis (TB) is marked by its success in the 1950s of the last century. However, TB is not under control, and the rise in antibiotic resistance worldwide is a major threat to global health care. Understanding the complex interactions between TB bacilli and their host can inform the rational design of better TB therapeutics, including vaccines, new antibiotics, and host-directed therapies. We recently demonstrated that the modulation of cystatin C in human macrophages via RNA silencing improved the anti-mycobacterial immune responses to Mycobacterium tuberculosis infection. Available in vitro transfection methods are not suitable for the clinical translation of host-cell RNA silencing. To overcome this limitation, we developed different RNA delivery systems (DSs) that target human macrophages. Human peripheral blood-derived macrophages and THP1 cells are difficult to transfect using available methods. In this work, a new potential nanomedicine based on chitosan (CS-DS) was efficiently developed to carry a siRNA-targeting cystatin C to the infected macrophage models. Consequently, an effective impact on the intracellular survival/replication of TB bacilli, including drug-resistant clinical strains, was observed. Altogether, these results suggest the potential use of CS-DS in adjunctive therapy for TB in combination or not with antibiotics.
Collapse
Affiliation(s)
- David Pires
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Center for Interdisciplinary Research in Health, Católica Medical School, Universidade Católica Portuguesa, Estrada Octávio Pato, 2635-631 Rio de Mouro, Portugal
| | - Manoj Mandal
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Ana I. Matos
- Drug Delivery and Immunoengineering Unit, Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Carina Peres
- Drug Delivery and Immunoengineering Unit, Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Maria João Catalão
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - José Miguel Azevedo-Pereira
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv P.O. Box 39040, Israel
| | - Helena F. Florindo
- Drug Delivery and Immunoengineering Unit, Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Elsa Anes
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| |
Collapse
|
17
|
Duong VT, Skwarczynski M, Toth I. Towards the development of subunit vaccines against tuberculosis: The key role of adjuvant. Tuberculosis (Edinb) 2023; 139:102307. [PMID: 36706503 DOI: 10.1016/j.tube.2023.102307] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/22/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
According to the World Health Organization (WHO), tuberculosis (TB) is the leading cause of death triggered by a single infectious agent, worldwide. Bacillus Calmette-Guerin (BCG) is the only currently licensed anti-TB vaccine. However, other strategies, including modification of recombinant BCG vaccine, attenuated Mycobacterium tuberculosis (Mtb) mutant constructs, DNA and protein subunit vaccines, are under extensive investigation. As whole pathogen vaccines can trigger serious adverse reactions, most current strategies are focused on the development of safe anti-TB subunit vaccines; this is especially important given the rising TB infection rate in immunocompromised HIV patients. The whole Mtb genome has been mapped and major antigens have been identified; however, optimal vaccine delivery mode is still to be established. Isolated protein antigens are typically poorly immunogenic so adjuvants are required to induce strong and long-lasting immune responses. This article aims to review the developmental status of anti-TB subunit vaccine adjuvants.
Collapse
Affiliation(s)
- Viet Tram Duong
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia.
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia.
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia; Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia; School of Pharmacy, The University of Queensland, Woolloongabba, QLD, 4102, Australia.
| |
Collapse
|
18
|
Pires D, Mandal M, Pinho J, Catalão MJ, Almeida AJ, Azevedo-Pereira JM, Gaspar MM, Anes E. Liposomal Delivery of Saquinavir to Macrophages Overcomes Cathepsin Blockade by Mycobacterium tuberculosis and Helps Control the Phagosomal Replicative Niches. Int J Mol Sci 2023; 24:ijms24021142. [PMID: 36674655 PMCID: PMC9863908 DOI: 10.3390/ijms24021142] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/21/2022] [Accepted: 01/04/2023] [Indexed: 01/10/2023] Open
Abstract
Mycobacterium tuberculosis is able to establish a chronic colonization of lung macrophages in a controlled replication manner, giving rise to a so-called latent infection. Conversely, when intracellular bacteria undergo actively uncontrolled replication rates, they provide the switch for the active infection called tuberculosis to occur. Our group found that the pathogen is able to manipulate the activity of endolysosomal enzymes, cathepsins, directly at the level of gene expression or indirectly by regulating their natural inhibitors, cystatins. To provide evidence for the crucial role of cathepsin manipulation for the success of tuberculosis bacilli in their intracellular survival, we used liposomal delivery of saquinavir. This protease inhibitor was previously found to be able to increase cathepsin proteolytic activity, overcoming the pathogen induced blockade. In this study, we demonstrate that incorporation in liposomes was able to increase the efficiency of saquinavir internalization in macrophages, reducing cytotoxicity at higher concentrations. Consequently, our results show a significant impact on the intracellular killing not only to reference and clinical strains susceptible to current antibiotic therapy but also to multidrug- and extensively drug-resistant (XDR) Mtb strains. Altogether, this indicates the manipulation of cathepsins as a fine-tuning strategy used by the pathogen to survive and replicate in host cells.
Collapse
Affiliation(s)
- David Pires
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Center for Interdisciplinary Research in Health, Católica Medical School, Universidade Católica Portuguesa, Estrada Octávio Pato, 2635-631 Rio de Mouro, Portugal
| | - Manoj Mandal
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Jacinta Pinho
- Advanced Technologies for Drug Delivery, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Maria João Catalão
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - António José Almeida
- Advanced Technologies for Drug Delivery, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - José Miguel Azevedo-Pereira
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Maria Manuela Gaspar
- Advanced Technologies for Drug Delivery, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Elsa Anes
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Correspondence:
| |
Collapse
|
19
|
Sanchez KG, Prest RJ, Nicholson KR, Korotkov KV, Champion PA. Functional Analysis of EspM, an ESX-1-Associated Transcription Factor in Mycobacterium marinum. J Bacteriol 2022; 204:e0023322. [PMID: 36448785 PMCID: PMC9765225 DOI: 10.1128/jb.00233-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 11/01/2022] [Indexed: 12/03/2022] Open
Abstract
Pathogenic mycobacteria use the ESX-1 secretion system to escape the macrophage phagosome and survive infection. We demonstrated that the ESX-1 system is regulated by feedback control in Mycobacterium marinum, a nontuberculous pathogen and model for the human pathogen Mycobacterium tuberculosis. In the presence of a functional ESX-1 system, the WhiB6 transcription factor upregulates expression of ESX-1 substrate genes. In the absence of an assembled ESX-1 system, the conserved transcription factor, EspM, represses whiB6 expression by specifically binding the whiB6 promoter. Together, WhiB6 and EspM fine-tune the levels of ESX-1 substrates in response to the secretion system. The mechanisms underlying control of the ESX-1 system by EspM are unknown. Here, we conduct a structure and function analysis to investigate how EspM is regulated. Using biochemical approaches, we measured the formation of higher-order oligomers of EspM in vitro. We demonstrate that multimerization in vitro can be mediated through multiple domains of the EspM protein. Using a bacterial monohybrid system, we showed that EspM self-associates through multiple domains in Escherichia coli. Using this system, we performed a genetic screen to identify EspM variants that failed to self-associate. The screen yielded four EspM variants of interest, which we tested for activity in M. marinum. Our study revealed that the two helix-turn-helix domains are functionally distinct. Moreover, the helix bundle domain is required for wild-type multimerization in vitro. Our data support models where EspM monomers or hexamers contribute to the regulation of whiB6 expression. IMPORTANCE Pathogenic mycobacteria are bacteria that pose a large burden to human health globally. The ESX-1 secretion system is required for pathogenic mycobacteria to survive within and interact with the host. Proper function of the ESX-1 secretion system is achieved by tightly controlling the expression of secreted virulence factors, in part through transcriptional regulation. Here, we characterize the conserved transcription factor EspM, which regulates the expression of ESX-1 virulence factors. We define domains required for EspM to form multimers and bind DNA. These findings provide an initial characterization an ESX-1 transcription factor and provide insights into its mechanism of action.
Collapse
Affiliation(s)
- Kevin G. Sanchez
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, USA
| | - Rebecca J. Prest
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Kathleen R. Nicholson
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, USA
| | - Konstantin V. Korotkov
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Patricia A. Champion
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, USA
| |
Collapse
|
20
|
Wang Y, Ramos M, Jefferson M, Zhang W, Beraza N, Carding S, Powell PP, Stewart JP, Mayer U, Wileman T. Control of infection by LC3-associated phagocytosis, CASM, and detection of raised vacuolar pH by the V-ATPase-ATG16L1 axis. SCIENCE ADVANCES 2022; 8:eabn3298. [PMID: 36288298 PMCID: PMC9604538 DOI: 10.1126/sciadv.abn3298] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 07/07/2022] [Indexed: 05/29/2023]
Abstract
The delivery of pathogens to lysosomes for degradation provides an important defense against infection. Degradation is enhanced when LC3 is conjugated to endosomes and phagosomes containing pathogens to facilitate fusion with lysosomes. In phagocytic cells, TLR signaling and Rubicon activate LC3-associated phagocytosis (LAP) where stabilization of the NADPH oxidase leads to sustained ROS production and raised vacuolar pH. Raised pH triggers the assembly of the vacuolar ATPase on the vacuole membrane where it binds ATG16L1 to recruit the core LC3 conjugation complex (ATG16L1:ATG5-12). This V-ATPase-ATG16L1 axis is also activated in nonphagocytic cells to conjugate LC3 to endosomes containing extracellular microbes. Pathogens provide additional signals for recruitment of LC3 when they raise vacuolar pH with pore-forming toxins and proteins, phospholipases, or specialized secretion systems. Many microbes secrete virulence factors to inhibit ROS production and/or the V-ATPase-ATG16L1 axis to slow LC3 recruitment and avoid degradation in lysosomes.
Collapse
Affiliation(s)
- Yingxue Wang
- Norwich Medical School, University of East Anglia, Norwich, UK
- Quadram Institute Bioscience, Norwich, UK
| | - Maria Ramos
- Norwich Medical School, University of East Anglia, Norwich, UK
- Quadram Institute Bioscience, Norwich, UK
| | | | - Weijiao Zhang
- Norwich Medical School, University of East Anglia, Norwich, UK
| | | | | | - Penny P. Powell
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - James P. Stewart
- Department of Infection Biology, University of Liverpool, Liverpool, UK
| | - Ulrike Mayer
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Thomas Wileman
- Norwich Medical School, University of East Anglia, Norwich, UK
- Quadram Institute Bioscience, Norwich, UK
| |
Collapse
|
21
|
Cronin RM, Ferrell MJ, Cahir CW, Champion MM, Champion PA. Proteo-genetic analysis reveals clear hierarchy of ESX-1 secretion in Mycobacterium marinum. Proc Natl Acad Sci U S A 2022; 119:e2123100119. [PMID: 35671426 PMCID: PMC9214503 DOI: 10.1073/pnas.2123100119] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 05/06/2022] [Indexed: 12/14/2022] Open
Abstract
The ESX-1 (ESAT-6-system-1) system and the protein substrates it transports are essential for mycobacterial pathogenesis. The precise ways that ESX-1 substrates contribute to virulence remains unknown. Several known ESX-1 substrates are also required for the secretion of other proteins. We used a proteo-genetic approach to construct high-resolution dependency relationships for the roles of individual ESX-1 substrates in secretion and virulence in Mycobacterium marinum, a pathogen of humans and animals. Characterizing a collection of M. marinum strains with in-frame deletions in each of the known ESX-1 substrate genes and the corresponding complementation strains, we demonstrate that ESX-1 substrates are differentially required for ESX-1 activity and for virulence. Using isobaric-tagged proteomics, we quantified the degree of requirement of each substrate on protein secretion. We conclusively defined distinct contributions of ESX-1 substrates in protein secretion. Our data reveal a hierarchy of ESX-1 substrate secretion, which supports a model for the composition of the extracytoplasmic ESX-1 secretory machinery. Overall, our proteo-genetic analysis demonstrates discrete roles for ESX-1 substrates in ESX-1 function and secretion in M. marinum.
Collapse
Affiliation(s)
- Rachel M. Cronin
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556
| | - Micah J. Ferrell
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556
| | - Clare W. Cahir
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556
| | - Matthew M. Champion
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556
| | - Patricia A. Champion
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556
| |
Collapse
|
22
|
Rodriguez GM, Sharma N, Biswas A, Sharma N. The Iron Response of Mycobacterium tuberculosis and Its Implications for Tuberculosis Pathogenesis and Novel Therapeutics. Front Cell Infect Microbiol 2022; 12:876667. [PMID: 35646739 PMCID: PMC9132128 DOI: 10.3389/fcimb.2022.876667] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/25/2022] [Indexed: 12/16/2022] Open
Abstract
Most pathogenic bacteria require iron for growth. However, this metal is not freely available in the mammalian host. Due to its poor solubility and propensity to catalyze the generation of reactive oxygen species, host iron is kept in solution bound to specialized iron binding proteins. Access to iron is an important factor in the outcome of bacterial infections; iron limitation frequently induces virulence and drives pathogenic interactions with host cells. Here, we review the response of Mycobacterium tuberculosis to changes in iron availability, the relevance of this response to TB pathogenesis, and its potential for the design of new therapeutic interventions.
Collapse
|
23
|
Shantal CJN, Juan CC, Lizbeth BUS, Carlos HGJ, Estela GPB. Candida glabrata is a successful pathogen: an artist manipulating the immune response. Microbiol Res 2022; 260:127038. [DOI: 10.1016/j.micres.2022.127038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 04/02/2022] [Accepted: 04/07/2022] [Indexed: 02/07/2023]
|
24
|
Carpenter SM, Lu LL. Leveraging Antibody, B Cell and Fc Receptor Interactions to Understand Heterogeneous Immune Responses in Tuberculosis. Front Immunol 2022; 13:830482. [PMID: 35371092 PMCID: PMC8968866 DOI: 10.3389/fimmu.2022.830482] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/07/2022] [Indexed: 12/25/2022] Open
Abstract
Despite over a century of research, Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), continues to kill 1.5 million people annually. Though less than 10% of infected individuals develop active disease, the specific host immune responses that lead to Mtb transmission and death, as well as those that are protective, are not yet fully defined. Recent immune correlative studies demonstrate that the spectrum of infection and disease is more heterogenous than has been classically defined. Moreover, emerging translational and animal model data attribute a diverse immune repertoire to TB outcomes. Thus, protective and detrimental immune responses to Mtb likely encompass a framework that is broader than T helper type 1 (Th1) immunity. Antibodies, Fc receptor interactions and B cells are underexplored host responses to Mtb. Poised at the interface of initial bacterial host interactions and in granulomatous lesions, antibodies and Fc receptors expressed on macrophages, neutrophils, dendritic cells, natural killer cells, T and B cells have the potential to influence local and systemic adaptive immune responses. Broadening the paradigm of protective immunity will offer new paths to improve diagnostics and vaccines to reduce the morbidity and mortality of TB.
Collapse
Affiliation(s)
- Stephen M. Carpenter
- Division of Infectious Disease and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Cleveland Medical Center, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Lenette L. Lu
- Division of Geographic Medicine and Infectious Diseases, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, United States
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX, United States
- Parkland Health and Hospital System, Dallas, TX, United States
| |
Collapse
|
25
|
Walter I, Adam S, Gentilini MV, Kany AM, Brengel C, Thomann A, Sparwasser T, Köhnke J, Hartmann RW. Structure-Activity Relationship and Mode-Of-Action Studies Highlight 1-(4-Biphenylylmethyl)-1H-imidazole-Derived Small Molecules as Potent CYP121 Inhibitors. ChemMedChem 2021; 16:2786-2801. [PMID: 34010508 PMCID: PMC8519103 DOI: 10.1002/cmdc.202100283] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Indexed: 11/29/2022]
Abstract
CYP121 of Mycobacterium tuberculosis (Mtb) is an essential target for the development of novel potent drugs against tuberculosis (TB). Besides known antifungal azoles, further compounds of the azole class were recently identified as CYP121 inhibitors with antimycobacterial activity. Herein, we report the screening of a similarity-oriented library based on the former hit compound, the evaluation of affinity toward CYP121, and activity against M. bovis BCG. The results enabled a comprehensive SAR study, which was extended through the synthesis of promising compounds and led to the identification of favorable features for affinity and/or activity and hit compounds with 2.7-fold improved potency. Mode of action studies show that the hit compounds inhibit substrate conversion and highlighted CYP121 as the main antimycobacterial target of our compounds. Exemplified complex crystal structures of CYP121 with three inhibitors reveal a common binding site. Engaging in both hydrophobic interactions as well as hydrogen bonding to the sixth iron ligand, our compounds block a solvent channel leading to the active site heme. Additionally, we report the first CYP inhibitors that are able to reduce the intracellular replication of M. bovis BCG in macrophages, emphasizing their potential as future drug candidates against TB.
Collapse
Affiliation(s)
- Isabell Walter
- Department for Drug Design and OptimizationHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.166123SaarbrückenGermany
| | - Sebastian Adam
- Workgroup Structural Biology of Biosynthetic EnzymesHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI)Saarland UniversitySaarbrückenGermany
| | - Maria Virginia Gentilini
- Institute of Infection Immunology, TWINCORECentre for Experimental and Clinical Infection ResearchA Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI)HannoverGermany
| | - Andreas M. Kany
- Department for Drug Design and OptimizationHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.166123SaarbrückenGermany
| | - Christian Brengel
- Department for Drug Design and OptimizationHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.166123SaarbrückenGermany
| | - Andreas Thomann
- Department for Drug Design and OptimizationHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.166123SaarbrückenGermany
| | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORECentre for Experimental and Clinical Infection ResearchA Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI)HannoverGermany
| | - Jesko Köhnke
- Workgroup Structural Biology of Biosynthetic EnzymesHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI)Saarland UniversitySaarbrückenGermany
| | - Rolf W. Hartmann
- Department for Drug Design and OptimizationHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.166123SaarbrückenGermany
- Department of PharmacyPharmaceutical and Medicinal ChemistrySaarland UniversityCampus C2.366123SaarbrückenGermany
| |
Collapse
|
26
|
Mvubu NE, Chiliza TE. Exploring the Use of Medicinal Plants and Their Bioactive Derivatives as Alveolar NLRP3 Inflammasome Regulators during Mycobacterium tuberculosis Infection. Int J Mol Sci 2021; 22:ijms22179497. [PMID: 34502407 PMCID: PMC8431520 DOI: 10.3390/ijms22179497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/01/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022] Open
Abstract
Mycobacterium tuberculosis, the causative agent of tuberculosis (TB), is a successful intracellular pathogen that is responsible for the highest mortality rate among diseases caused by bacterial infections. During early interaction with the host innate cells, M. tuberculosis cell surface antigens interact with Toll like receptor 4 (TLR4) to activate the nucleotide-binding domain, leucine-rich-repeat containing family, pyrin domain-containing 3 (NLRP3) canonical, and non-canonical inflammasome pathways. NLRP3 inflammasome activation in the alveoli has been reported to contribute to the early inflammatory response that is needed for an effective anti-TB response through production of pro-inflammatory cytokines, including those of the Interleukin 1 (IL1) family. However, overstimulation of the alveolar NLRP3 inflammasomes can induce excessive inflammation that is pathological to the host. Several studies have explored the use of medicinal plants and/or their active derivatives to inhibit excessive stimulation of the inflammasomes and its associated factors, thus reducing immunopathological response in the host. This review describes the molecular mechanism of the NLRP3 inflammasome activation in the alveoli during M. tuberculosis infection. Furthermore, the mechanisms of inflammasome inhibition using medicinal plant and their derivatives will also be explored, thus offering a novel perspective on the alternative control strategies of M. tuberculosis-induced immunopathology.
Collapse
|
27
|
de Lima JB, da Silva Fonseca LP, Xavier LP, de Matos Macchi B, Cassoli JS, da Silva EO, da Silva Valadares RB, do Nascimento JLM, Santos AV, de Sena CBC. Culture of Mycobacterium smegmatis in Different Carbon Sources to Induce In Vitro Cholesterol Consumption Leads to Alterations in the Host Cells after Infection: A Macrophage Proteomics Analysis. Pathogens 2021; 10:pathogens10060662. [PMID: 34071265 PMCID: PMC8230116 DOI: 10.3390/pathogens10060662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/18/2021] [Accepted: 05/26/2021] [Indexed: 11/24/2022] Open
Abstract
During tuberculosis, Mycobacterium uses host macrophage cholesterol as a carbon and energy source. To mimic these conditions, Mycobacterium smegmatis can be cultured in minimal medium (MM) to induce cholesterol consumption in vitro. During cultivation, M. smegmatis consumes MM cholesterol and changes the accumulation of cell wall compounds, such as PIMs, LM, and LAM, which plays an important role in its pathogenicity. These changes lead to cell surface hydrophobicity modifications and H2O2 susceptibility. Furthermore, when M. smegmatis infects J774A.1 macrophages, it induces granuloma-like structure formation. The present study aims to assess macrophage molecular disturbances caused by M. smegmatis after cholesterol consumption, using proteomics analyses. Proteins that showed changes in expression levels were analyzed in silico using OmicsBox and String analysis to investigate the canonical pathways and functional networks involved in infection. Our results demonstrate that, after cholesterol consumption, M. smegmatis can induce deregulation of protein expression in macrophages. Many of these proteins are related to cytoskeleton remodeling, immune response, the ubiquitination pathway, mRNA processing, and immunometabolism. The identification of these proteins sheds light on the biochemical pathways involved in the mechanisms of action of mycobacteria infection, and may suggest novel protein targets for the development of new and improved treatments.
Collapse
Affiliation(s)
- Jaqueline Batista de Lima
- Laboratory of Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (J.B.d.L.); (E.O.d.S.)
- Laboratory of Biotechnology of Enzymes and Biotransformations, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (L.P.X.); (A.V.S.)
| | | | - Luciana Pereira Xavier
- Laboratory of Biotechnology of Enzymes and Biotransformations, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (L.P.X.); (A.V.S.)
| | - Barbarella de Matos Macchi
- Laboratory of Molecular and Cellular Neurochemistry, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (B.d.M.M.); (J.L.M.d.N.)
- National Institute of Science and Technology in Neuroimmunomodulation (INCT-NIM), Rio de Janeiro 21040-900, RJ, Brazil
| | - Juliana Silva Cassoli
- Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil;
| | - Edilene Oliveira da Silva
- Laboratory of Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (J.B.d.L.); (E.O.d.S.)
- National Institute of Science and Technology in Structural Biology and Bioimaging, Rio de Janeiro 21941-901, RJ, Brazil
| | | | - José Luiz Martins do Nascimento
- Laboratory of Molecular and Cellular Neurochemistry, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (B.d.M.M.); (J.L.M.d.N.)
- National Institute of Science and Technology in Neuroimmunomodulation (INCT-NIM), Rio de Janeiro 21040-900, RJ, Brazil
| | - Agenor Valadares Santos
- Laboratory of Biotechnology of Enzymes and Biotransformations, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (L.P.X.); (A.V.S.)
| | - Chubert Bernardo Castro de Sena
- Laboratory of Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (J.B.d.L.); (E.O.d.S.)
- National Institute of Science and Technology in Neuroimmunomodulation (INCT-NIM), Rio de Janeiro 21040-900, RJ, Brazil
- Correspondence:
| |
Collapse
|
28
|
Gioseffi A, Edelmann MJ, Kima PE. Intravacuolar Pathogens Hijack Host Extracellular Vesicle Biogenesis to Secrete Virulence Factors. Front Immunol 2021; 12:662944. [PMID: 33959131 PMCID: PMC8093443 DOI: 10.3389/fimmu.2021.662944] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/30/2021] [Indexed: 12/18/2022] Open
Abstract
Extracellular vesicles (EVs) have garnered significant interest in recent years due to their contributions to cell-to-cell communication and disease processes. EVs are composed of a complex profile of bioactive molecules, which include lipids, nucleic acids, metabolites, and proteins. Although the biogenesis of EVs released by cells under various normal and abnormal conditions has been well-studied, there is incomplete knowledge about how infection influences EV biogenesis. EVs from infected cells contain specific molecules of both host and pathogen origin that may contribute to pathogenesis and the elicitation of the host immune response. Intracellular pathogens exhibit diverse lifestyles that undoubtedly dictate the mechanisms by which their molecules enter the cell’s exosome biogenesis schemes. We will discuss the current understanding of the mechanisms used during infection to traffic molecules from their vacuolar niche to host EVs by selected intravacuolar pathogens. We initially review general exosome biogenesis schemes and then discuss what is known about EV biogenesis in Mycobacterium, Plasmodium, Toxoplasma, and Leishmania infections, which are pathogens that reside within membrane delimited compartments in phagocytes at some time in their life cycle within mammalian hosts. The review includes discussion of the need for further studies into the biogenesis of EVs to better understand the contributions of these vesicles to host-pathogen interactions, and to uncover potential therapeutic targets to control these pathogens.
Collapse
Affiliation(s)
- Anna Gioseffi
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Mariola J Edelmann
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Peter E Kima
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| |
Collapse
|
29
|
Rijnink WF, Ottenhoff THM, Joosten SA. B-Cells and Antibodies as Contributors to Effector Immune Responses in Tuberculosis. Front Immunol 2021; 12:640168. [PMID: 33679802 PMCID: PMC7930078 DOI: 10.3389/fimmu.2021.640168] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 01/29/2021] [Indexed: 12/19/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is still a major threat to mankind, urgently requiring improved vaccination and therapeutic strategies to reduce TB-disease burden. Most present vaccination strategies mainly aim to induce cell-mediated immunity (CMI), yet a series of independent studies has shown that B-cells and antibodies (Abs) may contribute significantly to reduce the mycobacterial burden. Although early studies using B-cell knock out animals did not support a major role for B-cells, more recent studies have provided new evidence that B-cells and Abs can contribute significantly to host defense against Mtb. B-cells and Abs exist in many different functional subsets, each equipped with unique functional properties. In this review, we will summarize current evidence on the contribution of B-cells and Abs to immunity toward Mtb, their potential utility as biomarkers, and their functional contribution to Mtb control.
Collapse
Affiliation(s)
- Willemijn F Rijnink
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Simone A Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
30
|
Kinsella RL, Zhu DX, Harrison GA, Mayer Bridwell AE, Prusa J, Chavez SM, Stallings CL. Perspectives and Advances in the Understanding of Tuberculosis. ANNUAL REVIEW OF PATHOLOGY 2021; 16:377-408. [PMID: 33497258 DOI: 10.1146/annurev-pathol-042120-032916] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), remains a leading cause of death due to infection in humans. To more effectively combat this pandemic, many aspects of TB control must be developed, including better point of care diagnostics, shorter and safer drug regimens, and a protective vaccine. To address all these areas of need, better understanding of the pathogen, host responses, and clinical manifestations of the disease is required. Recently, the application of cutting-edge technologies to the study of Mtb pathogenesis has resulted in significant advances in basic biology, vaccine development, and antibiotic discovery. This leaves us in an exciting era of Mtb research in which our understanding of this deadly infection is improving at a faster rate than ever, and renews hope in our fight to end TB. In this review, we reflect on what is known regarding Mtb pathogenesis, highlighting recent breakthroughs that will provide leverage for the next leaps forward in the field.
Collapse
Affiliation(s)
- Rachel L Kinsella
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA;
| | - Dennis X Zhu
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA;
| | - Gregory A Harrison
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA;
| | - Anne E Mayer Bridwell
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA;
| | - Jerome Prusa
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA;
| | - Sthefany M Chavez
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA;
| | - Christina L Stallings
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA;
| |
Collapse
|
31
|
Beg MA, Hejazi II, Thakur SC, Athar F. Domain-wise differentiation of Mycobacterium tuberculosis H 37 Rv hypothetical proteins: A roadmap to discover bacterial survival potentials. Biotechnol Appl Biochem 2021; 69:296-312. [PMID: 33469971 DOI: 10.1002/bab.2109] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 01/06/2021] [Indexed: 01/08/2023]
Abstract
Proteomic information revealed approximately 3,923 proteins in Mycobacterium tuberculosis H37 Rv genome of which around ∼25% of proteins are hypothetical proteins (HPs). The present work comprises computational approaches to identify and characterize the HPs of M. tuberculosis that symbolize the putative target for rationale development of a drug or antituberculosis strategy. Proteins were primarily classified based on motif and domain information, which were further analyzed for the presence of virulence factors (VFs), determination of localization, and signal peptide/enzymatic cleavage sites. 863 HPs were found, and 599 HPs were finalized based on motifs, that is, GTP (525), Trx (47), SAM (14), PE-PGRS (5), and CBD (8). 80 HPs contain virulence factor (VF), 24 HPs localized in membrane region, and 4 HPs contain signal peptide/enzymatic cleavage sites. The overall parametric study finalizes four HPs Rv0679c, Rv0906, Rv3627c, and Rv3811 that also comprise GTPase domain. Structure prediction, structure-based function prediction, molecular docking and mutation analysis of selected proteins were done. Docking studies revealed that GTP and GTPase inhibitor (mac0182344) were docked with all four proteins with high affinities. In silico point mutation studies showed that substitution of aspartate with glycine within a GTPase motif showed the largest decrease in stability and pH differentiation also affects protein's stability. This analysis thus fixes a roadmap in the direction of finding potential target of this bacterium for drug development and enlightens the efficacy of GTP as a major regulator of Mycobacterial cellular pathways.
Collapse
Affiliation(s)
- Md Amjad Beg
- Centre for Interdisciplinary Research in Basic Science, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Iram Iqbal Hejazi
- Centre for Interdisciplinary Research in Basic Science, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Sonu Chand Thakur
- Centre for Interdisciplinary Research in Basic Science, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Fareeda Athar
- Centre for Interdisciplinary Research in Basic Science, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| |
Collapse
|
32
|
Guo Q, Bi J, Wang H, Zhang X. Mycobacterium tuberculosis ESX-1-secreted substrate protein EspC promotes mycobacterial survival through endoplasmic reticulum stress-mediated apoptosis. Emerg Microbes Infect 2020; 10:19-36. [PMID: 33290182 PMCID: PMC7832037 DOI: 10.1080/22221751.2020.1861913] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
EsxA, secreted by the ESAT-6 secretion system 1 (ESX-1) secretion system, is considered the major Mycobacterium tuberculosis (Mtb) virulence determinant. However, the roles of the individual ESX-1 substrates, such as EspC, remain unclear due to their interdependency for secretion with EsxA. Here, we validated that EspC triggered ER stress-mediated apoptosis in macrophages. The EspC-mediated ER stress was involved in pro-inflammatory cytokines generation, intracellular Ca2+ release, and reactive oxygen species accumulation. Mitochondrial transmembrane potential dissipation and mitochondrial outer membrane permeabilization occurred in EspC-treated macrophages, causing apoptosis. Furthermore, ER stress-mediated apoptosis was effectively induced in EspC-overexpressing Mycobacterium smegmatis-infected macrophages and mice. EspC overexpression caused a significant increase in bacterial survival in the macrophages, spleens, and lungs, and accelerated mouse death was observed. Moreover, the increased viability of bacteria in the macrophages was significantly reduced by pretreatment with the apoptosis inhibitor. Overall, our results revealed that EspC is an essential ESX-1 protein for Mtb–host interactions and EspC-induced ER stress-mediated apoptosis may be employed by Mtb to establish and spread infection. Given the critical roles of the ESX systems in Mtb pathogenesis and immunity, our findings offer new perspectives on the complex host-pathogen interactions and mechanisms underlying ESX-1-mediated pathogenesis.
Collapse
Affiliation(s)
- Qinglong Guo
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, People's Republic of China.,National Clinical Research Center for Infectious Disease (Tuberculosis), Shenzhen Third People's Hospital, South University of Science and Technology of China, Shenzhen, People's Republic of China
| | - Jing Bi
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, People's Republic of China.,Key Laboratory of Medical Molecular Virology, Ministry of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Honghai Wang
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, People's Republic of China
| | - Xuelian Zhang
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, People's Republic of China.,Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
33
|
Modeling Tubercular ESX-1 Secretion Using Mycobacterium marinum. Microbiol Mol Biol Rev 2020; 84:84/4/e00082-19. [DOI: 10.1128/mmbr.00082-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pathogenic mycobacteria cause chronic and acute diseases ranging from human tuberculosis (TB) to nontubercular infections.
Mycobacterium tuberculosis
causes both acute and chronic human tuberculosis. Environmentally acquired nontubercular mycobacteria (NTM) cause chronic disease in humans and animals. Not surprisingly, NTM and
M. tuberculosis
often use shared molecular mechanisms to survive within the host. The ESX-1 system is a specialized secretion system that is essential for virulence and is functionally conserved between
M. tuberculosis
and
Mycobacterium marinum
.
Collapse
|
34
|
Khawbung JL, Nath D, Chakraborty S. Drug resistant Tuberculosis: A review. Comp Immunol Microbiol Infect Dis 2020; 74:101574. [PMID: 33249329 DOI: 10.1016/j.cimid.2020.101574] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 11/05/2020] [Indexed: 12/14/2022]
Abstract
Tuberculosis (TB) was announced as a global emergency in 1993. There was an alarming counter attack of TB worldwide. However, when it was known that TB can be cured completely, the general public became ignorant towards the infection. The pathogenic organism Mycobacterium tuberculosis continuously evolved to resist the antagonist drugs. This has led to the outbreak of resistant strain that gave rise to "Multi Drug Resistant-Tuberculosis" and "Extensively Drug Resistant Tuberculosis" that can still be cured with a lower success rate. While the mechanism of resistance proceeds further, it ultimately causes unmanageable totally drug resistant TB (TDR-TB). Studying the molecular mechanisms underlying the resistance to drugs would help us grasp the genetics and pathophysiology of the disease. In this review, we present the molecular mechanisms behind Mycobacterium tolerance to drugs and their approach towards the development of multi-drug resistant, extremely drug resistant and totally drug resistant TB.
Collapse
Affiliation(s)
| | - Durbba Nath
- Department of Biotechnology, Assam University, Silchar, 788011, Assam, India
| | - Supriyo Chakraborty
- Department of Biotechnology, Assam University, Silchar, 788011, Assam, India.
| |
Collapse
|
35
|
Tran AC, Diogo GR, Paul MJ, Copland A, Hart P, Mehta N, Irvine EB, Mussá T, Drake PMW, Ivanyi J, Alter G, Reljic R. Mucosal Therapy of Multi-Drug Resistant Tuberculosis With IgA and Interferon-γ. Front Immunol 2020; 11:582833. [PMID: 33193394 PMCID: PMC7606302 DOI: 10.3389/fimmu.2020.582833] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/29/2020] [Indexed: 12/31/2022] Open
Abstract
New evidence has been emerging that antibodies can be protective in various experimental models of tuberculosis. Here, we report on protection against multidrug-resistant Mycobacterium tuberculosis (MDR-TB) infection using a combination of the human monoclonal IgA 2E9 antibody against the alpha-crystallin (Acr, HspX) antigen and mouse interferon-gamma in mice transgenic for the human IgA receptor, CD89. The effect of the combined mucosal IgA and IFN-γ; treatment was strongest (50-fold reduction) when therapy was applied at the time of infection, but a statistically significant reduction of lung bacterial load was observed even when the therapy was initiated once the infection had already been established. The protection involving enhanced phagocytosis and then neutrophil mediated killing of infected cells was IgA isotype mediated, because treatment with an IgG version of 2E9 antibody was not effective in human IgG receptor CD64 transgenic mice. The Acr antigen specificity of IgA antibodies for protection in humans has been indicated by their elevated serum levels in latent tuberculosis unlike the lack of IgA antibodies against the virulence-associated MPT64 antigen. Our results represent the first evidence for potential translation of mucosal immunotherapy for the management of MDR-TB.
Collapse
Affiliation(s)
- Andy C Tran
- Institute for Infection and Immunity, St. George's University, London, United Kingdom
| | - Gil R Diogo
- Institute for Infection and Immunity, St. George's University, London, United Kingdom
| | - Matthew J Paul
- Institute for Infection and Immunity, St. George's University, London, United Kingdom
| | - Alastair Copland
- Institute for Infection and Immunity, St. George's University, London, United Kingdom
| | - Peter Hart
- Institute for Infection and Immunity, St. George's University, London, United Kingdom
| | - Nickita Mehta
- Ragon Institute, Harvard, Cambridge, MA, United States
| | | | - Tufária Mussá
- Department of Microbiology, Faculty of Medicine, Eduardo Mondlane University, Maputo, Mozambique.,Departamento de Plataformas Tecnológicas em Saúde, Instituto Nacional de Saúde, Maputo, Mozambique
| | - Pascal M W Drake
- Institute for Infection and Immunity, St. George's University, London, United Kingdom
| | - Juraj Ivanyi
- Departamento de Plataformas Tecnológicas em Saúde, Instituto Nacional de Saúde, Maputo, Mozambique.,Guy's Campus of King's College London, London, United Kingdom
| | - Galit Alter
- Ragon Institute, Harvard, Cambridge, MA, United States
| | - Rajko Reljic
- Institute for Infection and Immunity, St. George's University, London, United Kingdom
| |
Collapse
|
36
|
Sprenkeler EGG, Webbers SDS, Kuijpers TW. When Actin is Not Actin' Like It Should: A New Category of Distinct Primary Immunodeficiency Disorders. J Innate Immun 2020; 13:3-25. [PMID: 32846417 DOI: 10.1159/000509717] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022] Open
Abstract
An increasing number of primary immunodeficiencies (PIDs) have been identified over the last decade, which are caused by deleterious mutations in genes encoding for proteins involved in actin cytoskeleton regulation. These mutations primarily affect hematopoietic cells and lead to defective function of immune cells, such as impaired motility, signaling, proliferative capacity, and defective antimicrobial host defense. Here, we review several of these immunological "actinopathies" and cover both clinical aspects, as well as cellular mechanisms of these PIDs. We focus in particular on the effect of these mutations on human neutrophil function.
Collapse
Affiliation(s)
- Evelien G G Sprenkeler
- Department of Blood Cell Research, Sanquin Research, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, The Netherlands, .,Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, AUMC, University of Amsterdam, Amsterdam, The Netherlands,
| | - Steven D S Webbers
- Department of Blood Cell Research, Sanquin Research, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, AUMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Taco W Kuijpers
- Department of Blood Cell Research, Sanquin Research, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, AUMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
37
|
Fighting Persistence: How Chronic Infections with Mycobacterium tuberculosis Evade T Cell-Mediated Clearance and New Strategies To Defeat Them. Infect Immun 2020; 88:IAI.00916-19. [PMID: 32094248 DOI: 10.1128/iai.00916-19] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Chronic bacterial infections are caused by pathogens that persist within their hosts and avoid clearance by the immune system. Treatment and/or detection of such pathogens is difficult, and the resulting pathologies are often deleterious or fatal. There is an urgent need to develop protective vaccines and host-directed therapies that synergize with antibiotics to prevent pathogen persistence and infection-associated pathologies. However, many persistent pathogens, such as Mycobacterium tuberculosis, actively target the very host pathways activated by vaccination. These immune evasion tactics blunt the effectiveness of immunization strategies and are impeding progress to control these infections throughout the world. Therefore, it is essential that M. tuberculosis immune evasion-related pathogen virulence strategies are considered to maximize the effectiveness of potential new treatments. In this review, we focus on how Mycobacterium tuberculosis infects antigen-presenting cells and evades effective immune clearance by the adaptive response through (i) manipulating antigen presentation, (ii) repressing T cell-activating costimulatory molecules, and (iii) inducing ligands that drive T cell exhaustion. In this context, we will examine the challenges that bacterial virulence strategies pose to developing new vaccines. We will then discuss new approaches that will help dissect M. tuberculosis immune evasion mechanisms and devise strategies to bypass them to promote long-term protection and prevent disease progression.
Collapse
|
38
|
Giraud-Gatineau A, Coya JM, Maure A, Biton A, Thomson M, Bernard EM, Marrec J, Gutierrez MG, Larrouy-Maumus G, Brosch R, Gicquel B, Tailleux L. The antibiotic bedaquiline activates host macrophage innate immune resistance to bacterial infection. eLife 2020; 9:e55692. [PMID: 32369020 PMCID: PMC7200153 DOI: 10.7554/elife.55692] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/04/2020] [Indexed: 12/12/2022] Open
Abstract
Antibiotics are widely used in the treatment of bacterial infections. Although known for their microbicidal activity, antibiotics may also interfere with the host's immune system. Here, we analyzed the effects of bedaquiline (BDQ), an inhibitor of the mycobacterial ATP synthase, on human macrophages. Genome-wide gene expression analysis revealed that BDQ reprogramed cells into potent bactericidal phagocytes. We found that 579 and 1,495 genes were respectively differentially expressed in naive- and M. tuberculosis-infected macrophages incubated with the drug, with an over-representation of lysosome-associated genes. BDQ treatment triggered a variety of antimicrobial defense mechanisms, including phagosome-lysosome fusion, and autophagy. These effects were associated with activation of transcription factor EB, involved in the transcription of lysosomal genes, resulting in enhanced intracellular killing of different bacterial species that were naturally insensitive to BDQ. Thus, BDQ could be used as a host-directed therapy against a wide range of bacterial infections.
Collapse
Affiliation(s)
- Alexandre Giraud-Gatineau
- Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Institut PasteurParisFrance
- Université Paris Diderot, Sorbonne Paris Cité, Cellule PasteurParisFrance
| | | | - Alexandra Maure
- Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Institut PasteurParisFrance
- Université Paris Diderot, Sorbonne Paris Cité, Cellule PasteurParisFrance
| | - Anne Biton
- Bioinformatics and Biostatistics, Department of Computational Biology, USR 3756 CNRS, Institut PasteurParisFrance
| | - Michael Thomson
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Faculty of Natural Sciences, Imperial College LondonLondonUnited Kingdom
| | - Elliott M Bernard
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Jade Marrec
- Mycobacterial Genetics Unit, Institut PasteurParisFrance
| | - Maximiliano G Gutierrez
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Gérald Larrouy-Maumus
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Faculty of Natural Sciences, Imperial College LondonLondonUnited Kingdom
| | - Roland Brosch
- Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Institut PasteurParisFrance
| | - Brigitte Gicquel
- Mycobacterial Genetics Unit, Institut PasteurParisFrance
- Department of Tuberculosis Control and Prevention, Shenzhen Nanshan Center for Chronic Disease ControlShenzhenChina
| | - Ludovic Tailleux
- Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Institut PasteurParisFrance
- Mycobacterial Genetics Unit, Institut PasteurParisFrance
| |
Collapse
|
39
|
Jahagirdar PS, Gupta PK, Kulkarni SP, Devarajan PV. Intramacrophage Delivery of Dual Drug Loaded Nanoparticles for Effective Clearance of Mycobacterium tuberculosis. J Pharm Sci 2020; 109:2262-2270. [PMID: 32240695 DOI: 10.1016/j.xphs.2020.03.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/28/2020] [Accepted: 03/18/2020] [Indexed: 12/26/2022]
Abstract
The escalating global burden of tuberculosis necessitates radical strategies to curb its spread. In this study, rifampicin (RIF), a first line anti-tubercular antibiotic and curcumin (CUR), a promising antimycobacterial compound were co-encapsulated in polymeric nanoparticles to achieve intramacrophage delivery and improved Mycobacterium tuberculosis clearance. The dual loaded nanoparticles revealed average size ∼400 nm, low polydispersity and zeta potential of -26.89 ± 2.9 mV. Near complete release of both drugs from nanoparticles in artificial lysosomal fluid proposed drug release after macrophage internalisation. Nanoparticles were nontoxic to RAW 264.7 macrophages and aided 1.5-fold higher drug internalisation compared to free drugs. Enriched intracellular internalisation and lysosomal presence of nanoparticles was ascertained by confocal microscopy. Comparable minimum inhibitory concentration (MIC) of free RIF and CUR and nanoparticle encapsulated RIF and CUR confirmed retention of drug properties. High efficacy against Mycobacterium tuberculosis infected macrophages with RIF-CUR nanoparticles at 25× MIC (98.03 ± 2.5%), with complete clearance above 50× MIC suggests the dual loaded nanoparticles as a promising new nanosystem for tackling tuberculosis.
Collapse
Affiliation(s)
- Priyanka S Jahagirdar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Matunga, Mumbai, India
| | - Pramod K Gupta
- Radiation Medicine Centre, Bhabha Atomic Research Centre, TMH Annexe, Parel, Mumbai, India
| | - Savita P Kulkarni
- Radiation Medicine Centre, Bhabha Atomic Research Centre, TMH Annexe, Parel, Mumbai, India
| | - Padma V Devarajan
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Matunga, Mumbai, India.
| |
Collapse
|
40
|
An M. tuberculosis Metabolic Enzyme Moonlights as an Anti-Inflammatory Effector Protein. Cell Host Microbe 2020; 27:310-312. [PMID: 32164839 DOI: 10.1016/j.chom.2020.02.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The mechanisms by which Mycobacterium tuberculosis evades host immunity remain enigmatic. In a recent study in the journal Nature, Wang et al. report that an enzyme acting in the bacterial cytoplasm to degrade lipids also acts as an effector protein in the host cell cytoplasm to suppress inflammation.
Collapse
|
41
|
Sanchez KG, Ferrell MJ, Chirakos AE, Nicholson KR, Abramovitch RB, Champion MM, Champion PA. EspM Is a Conserved Transcription Factor That Regulates Gene Expression in Response to the ESX-1 System. mBio 2020; 11:e02807-19. [PMID: 32019792 PMCID: PMC7002343 DOI: 10.1128/mbio.02807-19] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 12/18/2019] [Indexed: 01/01/2023] Open
Abstract
Pathogenic mycobacteria encounter multiple environments during macrophage infection. Temporally, the bacteria are engulfed into the phagosome, lyse the phagosomal membrane, and interact with the cytosol before spreading to another cell. Virulence factors secreted by the mycobacterial ESX-1 (ESAT-6-system-1) secretion system mediate the essential transition from the phagosome to the cytosol. It was recently discovered that the ESX-1 system also regulates mycobacterial gene expression in Mycobacterium marinum (R. E. Bosserman, T. T. Nguyen, K. G. Sanchez, A. E. Chirakos, et al., Proc Natl Acad Sci U S A 114:E10772-E10781, 2017, https://doi.org/10.1073/pnas.1710167114), a nontuberculous mycobacterial pathogen, and in the human-pathogenic species M. tuberculosis (A. M. Abdallah, E. M. Weerdenburg, Q. Guan, R. Ummels, et al., PLoS One 14:e0211003, 2019, https://doi.org/10.1371/journal.pone.0211003). It is not known how the ESX-1 system regulates gene expression. Here, we identify the first transcription factor required for the ESX-1-dependent transcriptional response in pathogenic mycobacteria. We demonstrate that the gene divergently transcribed from the whiB6 gene and adjacent to the ESX-1 locus in mycobacterial pathogens encodes a conserved transcription factor (MMAR_5438, Rv3863, now espM). We prove that EspM from both M. marinum and M. tuberculosis directly and specifically binds the whiB6-espM intergenic region. We show that EspM is required for ESX-1-dependent repression of whiB6 expression and for the regulation of ESX-1-associated gene expression. Finally, we demonstrate that EspM functions to fine-tune ESX-1 activity in M. marinum Taking the data together, this report extends the esx-1 locus, defines a conserved regulator of the ESX-1 virulence pathway, and begins to elucidate how the ESX-1 system regulates gene expression.IMPORTANCE Mycobacterial pathogens use the ESX-1 system to transport protein substrates that mediate essential interactions with the host during infection. We previously demonstrated that in addition to transporting proteins, the ESX-1 secretion system regulates gene expression. Here, we identify a conserved transcription factor that regulates gene expression in response to the ESX-1 system. We demonstrate that this transcription factor is functionally conserved in M. marinum, a pathogen of ectothermic animals; M. tuberculosis, the human-pathogenic species that causes tuberculosis; and M. smegmatis, a nonpathogenic mycobacterial species. These findings provide the first mechanistic insight into how the ESX-1 system elicits a transcriptional response, a function of this protein transport system that was previously unknown.
Collapse
Affiliation(s)
- Kevin G Sanchez
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Micah J Ferrell
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Alexandra E Chirakos
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Kathleen R Nicholson
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Robert B Abramovitch
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Matthew M Champion
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Patricia A Champion
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| |
Collapse
|
42
|
McLean MR, Lu LL, Kent SJ, Chung AW. An Inflammatory Story: Antibodies in Tuberculosis Comorbidities. Front Immunol 2019; 10:2846. [PMID: 31921122 PMCID: PMC6913197 DOI: 10.3389/fimmu.2019.02846] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 11/19/2019] [Indexed: 12/20/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) resides in a quarter of the world's population and is the causative agent for tuberculosis (TB), the most common infectious reason of death in humans today. Although cellular immunity has been firmly established in the control of Mtb, there is growing evidence that antibodies may also modulate the infection. More specifically, certain antibody features are associated with inflammation and are divergent in different states of human infection and disease. Importantly, TB impacts not just the healthy but also those with chronic conditions. While HIV represents the quintessential comorbid condition for TB, recent epidemiological evidence shows that additional chronic conditions such as diabetes and kidney disease are rising. In fact, the prevalence of diabetes as a comorbid TB condition is now higher than that of HIV. These chronic diseases are themselves independently associated with pro-inflammatory immune states that encompass antibody profiles. This review discusses isotypes, subclasses, post-translational modifications and Fc-mediated functions of antibodies in TB infection and in the comorbid chronic conditions of HIV, diabetes, and kidney diseases. We propose that inflammatory antibody profiles, which are a marker of active TB, may be an important biomarker for detection of TB disease progression within comorbid individuals. We highlight the need for future studies to determine which inflammatory antibody profiles are the consequences of comorbidities and which may potentially contribute to TB reactivation.
Collapse
Affiliation(s)
- Milla R McLean
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Lenette L Lu
- Division of Infectious Disease and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Infectious Diseases Department, Melbourne Sexual Health Centre, Alfred Health, Central Clinical School, Monash University, Brisbane, VIC, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, SA, Australia
| | - Amy W Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
43
|
El-Badrawy MK, Arram EO, Abdalla DA, Al-Sagheer D, Zahran A, AboElEla MA, El-Badrawy A, Amin W. Effect of adding inhalation of sodium bicarbonate 8.4% to the usual treatment on smear-positive pulmonary tuberculosis: a prospective controlled study. THE EGYPTIAN JOURNAL OF BRONCHOLOGY 2019. [DOI: 10.4103/ejb.ejb_18_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
44
|
Bandara HMHN, Samaranayake LP. Viral, bacterial, and fungal infections of the oral mucosa: Types, incidence, predisposing factors, diagnostic algorithms, and management. Periodontol 2000 2019; 80:148-176. [PMID: 31090135 DOI: 10.1111/prd.12273] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
For millions of years, microbiota residing within us, including those in the oral cavity, coexisted in a harmonious symbiotic fashion that provided a quintessential foundation for human health. It is now clear that disruption of such a healthy relationship leading to microbial dysbiosis causes a wide array of infections, ranging from localized, mild, superficial infections to deep, disseminated life-threatening diseases. With recent advances in research, diagnostics, and improved surveillance we are witnessing an array of emerging and re-emerging oral infections and orofacial manifestations of systemic infections. Orofacial infections may cause significant discomfort to the patients and unnecessary economic burden. Thus, the early recognition of such infections is paramount for holistic patient management, and oral clinicians have a critical role in recognizing, diagnosing, managing, and preventing either new or old orofacial infections. This paper aims to provide an update on current understanding of well-established and emerging viral, bacterial, and fungal infections manifesting in the human oral cavity.
Collapse
Affiliation(s)
| | - Lakshman P Samaranayake
- Department of Oral and Craniofacial Health Sciences, College of Dental Medicine, University of Sharjah, Sharjah, UAE
| |
Collapse
|
45
|
Gupta S, Kumar A, Singh K, Kumari R, Sharma A, Singh RK, Pandey SK, Anupurba S. Rv1273c, an ABC transporter of Mycobacterium tuberculosis promotes mycobacterial intracellular survival within macrophages via modulating the host cell immune response. Int J Biol Macromol 2019; 142:320-331. [PMID: 31593717 DOI: 10.1016/j.ijbiomac.2019.09.103] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 08/18/2019] [Accepted: 09/13/2019] [Indexed: 02/06/2023]
Abstract
Mycobacterium proteins, especially cell wall associated proteins, interact with host macrophage to regulate the functions and cytokine production. So, identification and characterization of such proteins is essential for understanding tuberculosis pathogenesis. The role of the ABC transporter proteins in the pathophysiology and virulence of Mycobacterium tuberculosis is not clearly understood. In the present study, Rv1273c, an ABC transporter, has been expressed in a non-pathogenic and fast growing Mycobacterium smegmatis strain to explore its role in host pathogen interactions. Over expression of Rv1273c resulted in enhanced intracellular survival in macrophage as well as modified cell wall architecture. We found altered colony morphology and cell surface properties that might be linked with remodelling of bacterial cell wall which may help in the intracellular survival of mycobacterium. However, the enhanced intracellular survival was not found to be the consequence of an increased resistance to intracellular stresses. The activation of macrophage by Rv1273c was associated with perturbed cytokine production. Pharmacological inhibition experiment and western immunoblotting suggested that this altered cytokine profile was mediated possibly by NF-kB and p38 pathway in macrophage. Overall, the present findings indicated that Rv1273c enhanced mycobacterium persistence and mediated the evasion of immune responses during infection.
Collapse
Affiliation(s)
- Smita Gupta
- Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Arun Kumar
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Kamal Singh
- Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Richa Kumari
- Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ajay Sharma
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Rakesh K Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Satyendra K Pandey
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Shampa Anupurba
- Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India.
| |
Collapse
|
46
|
Abstract
Are antibodies important for protection against tuberculosis? The jury has been out for more than 100 years. B cell depletion in experimental Mycobacterium tuberculosis infection failed to identify a major role for these cells in immunity to tuberculosis. However, recent identification of naturally occurring antibodies in humans that are protective during M. tuberculosis infection has reignited the debate. Here, we discuss the evidence for a protective role for antibodies in tuberculosis and consider the feasibility of designing novel tuberculosis vaccines targeting humoral immunity.
Collapse
Affiliation(s)
- Hao Li
- Centre for Global Health and Infectious Diseases, Collaborative Innovation Centre for the Diagnosis and Treatment of Infectious Diseases, Tsinghua University School of Medicine, Beijing, China
| | - Babak Javid
- Centre for Global Health and Infectious Diseases, Collaborative Innovation Centre for the Diagnosis and Treatment of Infectious Diseases, Tsinghua University School of Medicine, Beijing, China.
| |
Collapse
|
47
|
Rab GTPases: Switching to Human Diseases. Cells 2019; 8:cells8080909. [PMID: 31426400 PMCID: PMC6721686 DOI: 10.3390/cells8080909] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023] Open
Abstract
Rab proteins compose the largest family of small GTPases and control the different steps of intracellular membrane traffic. More recently, they have been shown to also regulate cell signaling, division, survival, and migration. The regulation of these processes generally occurs through recruitment of effectors and regulatory proteins, which control the association of Rab proteins to membranes and their activation state. Alterations in Rab proteins and their effectors are associated with multiple human diseases, including neurodegeneration, cancer, and infections. This review provides an overview of how the dysregulation of Rab-mediated functions and membrane trafficking contributes to these disorders. Understanding the altered dynamics of Rabs and intracellular transport defects might thus shed new light on potential therapeutic strategies.
Collapse
|
48
|
Khan A, Bakhru P, Saikolappan S, Das K, Soudani E, Singh CR, Estrella JL, Zhang D, Pasare C, Ma Y, Sun J, Wang J, Hunter RL, Tony Eissa N, Dhandayuthapani S, Jagannath C. An autophagy-inducing and TLR-2 activating BCG vaccine induces a robust protection against tuberculosis in mice. NPJ Vaccines 2019; 4:34. [PMID: 31396406 PMCID: PMC6683161 DOI: 10.1038/s41541-019-0122-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 05/15/2019] [Indexed: 12/28/2022] Open
Abstract
Mycobacterium bovis BCG is widely used as a vaccine against tuberculosis due to M. tuberculosis (Mtb), which kills millions of people each year. BCG variably protects children, but not adults against tuberculosis. BCG evades phagosome maturation, autophagy, and reduces MHC-II expression of antigen-presenting cells (APCs) affecting T-cell activation. To bypass these defects, an autophagy-inducing, TLR-2 activating C5 peptide from Mtb-derived CFP-10 protein was overexpressed in BCG in combination with Ag85B. Recombinant BCG85C5 induced a robust MHC-II-dependent antigen presentation to CD4 T cells in vitro, and elicited stronger TH1 cytokines (IL-12, IL-1β, and TNFα) from APCs of C57Bl/6 mice increasing phosphorylation of p38MAPK and ERK. BCG85C5 also enhanced MHC-II surface expression of MΦs by inhibiting MARCH1 ubiquitin ligase that degrades MHC-II. BCG85C5 infected APCs from MyD88 or TLR-2 knockout mice showed decreased antigen presentation. Furthermore, BCG85C5 induced LC3-dependent autophagy in macrophages increasing antigen presentation. Consistent with in vitro effects, BCG85C5 markedly expanded both effector and central memory T cells in C57Bl/6 mice protecting them against both primary aerosol infection with Mtb and reinfection, but was less effective among TLR-2 knockout mice. Thus, BCG85C5 induces stronger and longer lasting immunity, and is better than BCG against tuberculosis of mice.
Collapse
Affiliation(s)
- Arshad Khan
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX USA
| | - Pearl Bakhru
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX USA
| | - Sankaralingam Saikolappan
- Molecular and Translational Medicine, Paul L. Foster School of Medicine Texas Tech University Health Sciences Center, El Paso, TX USA
| | - Kishore Das
- Molecular and Translational Medicine, Paul L. Foster School of Medicine Texas Tech University Health Sciences Center, El Paso, TX USA
| | - Emily Soudani
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX USA
| | - Christopher R. Singh
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX USA
| | - Jaymie L. Estrella
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX USA
| | - Dekai Zhang
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX USA
| | - Chandrashekhar Pasare
- Division of Immunobiology, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229 USA
| | - Yue Ma
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, Houston, TX USA
| | - Jianjun Sun
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, Houston, TX USA
| | - Jin Wang
- Methodist Hospital Research Institute, Houston, TX USA
| | - Robert L. Hunter
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX USA
| | | | - Subramanian Dhandayuthapani
- Molecular and Translational Medicine, Paul L. Foster School of Medicine Texas Tech University Health Sciences Center, El Paso, TX USA
| | - Chinnaswamy Jagannath
- Department of Pathology and Laboratory Medicine, University of Texas Health Sciences Center, Houston, TX USA
- Methodist Hospital Research Institute, Houston, TX USA
| |
Collapse
|
49
|
Tran AC, Kim MY, Reljic R. Emerging Themes for the Role of Antibodies in Tuberculosis. Immune Netw 2019; 19:e24. [PMID: 31501712 PMCID: PMC6722270 DOI: 10.4110/in.2019.19.e24] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/12/2019] [Accepted: 06/13/2019] [Indexed: 02/06/2023] Open
Abstract
The best way to debunk a scientific dogma is to throw irrefutable evidence at it. This is especially true if the dogma in question has been nurtured over many decades, as is the case with the apparent redundancy of antibodies (Abs) against intracellular pathogens. Although not fully compelling yet, that ‘hard core’ evidence is nevertheless now slowly beginning to emerge. This is true for several clinically relevant infections but none more so than Mycobacterium tuberculosis, the archetype intracellular pathogen that poses a great health challenge to the mankind. Here, prompted by a spate of recent high-profile reports on the effects of Abs in various experimental models of tuberculosis, we step back and take a critical look at the progress that has been made in the last 5 years and highlight some of the strengths and shortcomings of the presented evidence. We conclude that the tide of the opinion has begun to turn in favour of Abs but we also caution against overinterpreting the currently available limited evidence. For, until definitive evidence that can withstand even the most rigorous of experimental tests is produced, the dogma may yet survive. Or indeed, we may find that the truth is hidden somewhere in between the dogma and the unfulfilled scientific prophecy.
Collapse
Affiliation(s)
- Andy C Tran
- St George's, University of London, London SW17 0RE, UK
| | - Mi-Young Kim
- St George's, University of London, London SW17 0RE, UK.,Department of Molecular Biology and The Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju 54896, Korea
| | - Rajko Reljic
- St George's, University of London, London SW17 0RE, UK
| |
Collapse
|
50
|
Bussi C, Gutierrez MG. Mycobacterium tuberculosis infection of host cells in space and time. FEMS Microbiol Rev 2019; 43:341-361. [PMID: 30916769 PMCID: PMC6606852 DOI: 10.1093/femsre/fuz006] [Citation(s) in RCA: 217] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 03/26/2019] [Indexed: 12/16/2022] Open
Abstract
Tuberculosis (TB) caused by the bacterial pathogen Mycobacterium tuberculosis (Mtb) remains one of the deadliest infectious diseases with over a billion deaths in the past 200 years (Paulson 2013). TB causes more deaths worldwide than any other single infectious agent, with 10.4 million new cases and close to 1.7 million deaths in 2017. The obstacles that make TB hard to treat and eradicate are intrinsically linked to the intracellular lifestyle of Mtb. Mtb needs to replicate within human cells to disseminate to other individuals and cause disease. However, we still do not completely understand how Mtb manages to survive within eukaryotic cells and why some cells are able to eradicate this lethal pathogen. Here, we summarise the current knowledge of the complex host cell-pathogen interactions in TB and review the cellular mechanisms operating at the interface between Mtb and the human host cell, highlighting the technical and methodological challenges to investigating the cell biology of human host cell-Mtb interactions.
Collapse
Affiliation(s)
- Claudio Bussi
- Host-pathogen interactions in tuberculosis laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom
| | - Maximiliano G Gutierrez
- Host-pathogen interactions in tuberculosis laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom
| |
Collapse
|