1
|
Guo F, Song Y, Dong S, Wei J, Li B, Xu T, Wang H. Characterization and anti-tuberculosis effects of γδ T cells expanded and activated by Mycobacterium tuberculosis heat-resistant antigen. Virulence 2025; 16:2462092. [PMID: 39921673 PMCID: PMC11810100 DOI: 10.1080/21505594.2025.2462092] [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: 08/09/2024] [Revised: 12/01/2024] [Accepted: 01/20/2025] [Indexed: 02/10/2025] Open
Abstract
Tuberculosis (TB) is a chronic infectious disease caused by Mycobacterium tuberculosis (Mtb) that poses a severe threat to human health. A variety of highly immunogenic tuberculosis proteins have been used as targets in vaccine development to mitigate the spread of TB. Although Th1-type immunity has long been considered a crucial part of resistance to Mtb, γδ T cells, the predominant source of IL-17, are not negligible in controlling the early stages of TB infection. In addition to classical phosphoantigens, Mycobacterium tuberculosis heat-resistant antigens (HAg), a complex containing 564 proteins obtained from live tuberculosis bacteria after heat treatment at 121 °C for 20 min, have been confirmed to be highly effective γδ T cell stimulators as well. Several studies have demonstrated that HAg-activated γδ T cells can participate in TB immunity by secreting multiple cytokines against Mtb or by interacting with other innate immune cells. In this review, we present a possible mechanism of HAg stimulation of γδ T cells and the role of HAg-activated γδ T cells in anti-TB immunity. We also highlight the limitations of studies on HAg activation of γδ T cells and suggest further research directions on the relationship between HAg and γδ T cells.
Collapse
Affiliation(s)
- Fangzheng Guo
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
| | - Yamin Song
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
| | - Sihang Dong
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
| | - Jing Wei
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
| | - Baiqing Li
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Clinical Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
| | - Tao Xu
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Clinical Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
| | - Hongtao Wang
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Clinical Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
| |
Collapse
|
2
|
Yang J, Dan H, Chen Y, Zou L, Liu S, Wang F, Musa MB. miRNA Differential Expression Profile Analysis and Identification of Potential Key Genes in Active Tuberculosis. J Cell Mol Med 2025; 29:e70567. [PMID: 40387517 PMCID: PMC12087295 DOI: 10.1111/jcmm.70567] [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] [Received: 11/04/2024] [Revised: 03/19/2025] [Accepted: 04/17/2025] [Indexed: 05/20/2025] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium TB (MTB), remains a significant global health issue, particularly in developing nations. MicroRNAs (miRNAs) are non-coding RNAs (ncRNAs) that modulate immune responses and play a pivotal role in the pathogenesis of MTB by altering host immune defences. Insights into the regulatory functions of these miRNAs have revealed mechanisms through which MTB evades immune surveillance and establishes persistent infections, highlighting the critical role of miRNA networks in TB pathogenesis. The purpose of this study was to analyse miRNA expression in plasma from TB patients, to predict target genes, and to construct regulatory networks to elucidate the roles of miRNAs in TB pathogenesis. Plasma samples from three patients with active TB and three healthy controls were analysed using high-throughput small RNA sequencing. DEMs were identified using DESeq2, and target genes were predicted via TargetScan and miRWalk. Protein-protein interaction (PPI) networks were constructed using STRING and Cytoscape. Functional enrichment analyses were performed using Gene Ontology (GO) and KEGG databases. A total of 23 DEMs were identified, including 17 upregulated and 6 downregulated miRNAs. hsa-miR-15a-5p emerged as the most significantly upregulated miRNA. PPI network analysis highlighted CCND1, CDK6 and CCND2 as central genes, potentially regulated by miR-15a-5p. GO and KEGG analyses revealed enrichment in pathways related to cell cycle regulation, kinase activity and protein complex formation, suggesting their involvement in TB pathogenesis. This study identifies hsa-miR-15a-5p and its target genes as key components in the regulatory landscape of TB. These findings offer new insights into the molecular mechanisms of TB and propose potential biomarkers and therapeutic targets for future research.
Collapse
Affiliation(s)
- Jun Yang
- Faculty of Applied SciencesUniversiti Teknologi MARAShah AlamSelangorMalaysia
- Department of Laboratory MedicineGuilin Medical University Affiliated HospitalGuilinGuangxiChina
| | - Hanliang Dan
- Faculty of Applied SciencesUniversiti Teknologi MARAShah AlamSelangorMalaysia
- Nanxishan Hospital of Guangxi Zhuang Autonomous Region (The Second People's Hospital of Guangxi Zhuang Autonomous Region)GuilinGuangxiChina
| | - Yeng Chen
- Faculty of DentistryUniversity of MalaiyaKuala LumpurMalaysia
| | - Linrong Zou
- Department of Laboratory MedicineGuilin Medical University Affiliated HospitalGuilinGuangxiChina
| | - Shanshan Liu
- Department of Laboratory MedicineGuilin Medical University Affiliated HospitalGuilinGuangxiChina
| | - Feng Wang
- Department of Laboratory MedicineGuilin Medical University Affiliated HospitalGuilinGuangxiChina
| | - Maslinda Binti Musa
- Faculty of Applied SciencesUniversiti Teknologi MARAShah AlamSelangorMalaysia
| |
Collapse
|
3
|
Penã Avila J, Simmons J, Figueiredo MC, Turner M, Cordeiro-Santos M, Rolla VC, Kristki AL, Gangula R, Nochowicz C, Ram R, Bailin S, Mallal S, Gaudieri S, Alves E, Barreto-Duarte BB, Queiroz ATL, Nakaya HI, Andrade BB, Sterling TR, Kalams SA. Single-cell immune profiling at time of M. tuberculosis exposure reveals antigen-reactive programs that predict progression to active disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.29.25326433. [PMID: 40343021 PMCID: PMC12060959 DOI: 10.1101/2025.04.29.25326433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Early delineation of host immune responses at the moment of Mycobacterium tuberculosis (Mtb) exposure and infection is critical to identify individuals at risk of progressing to active tuberculosis (TB). We performed single-cell transcriptional profiling of over 500,000 peripheral blood mononuclear cells from 57 HIV-negative close contacts of TB cases in Brazil, including 25 individuals who developed active disease within two years (progressors) and 32 matched controls who remained disease-free (non-progressors). Cells were stimulated separately with the MTB300 peptide pool or irradiated Mtb (gRV), enabling resolution of antigen-reactive states across adaptive (CD4⁺ T-cells expressing abundant cytokines including IFNG, TNF, and IL17F) and trained-innate lineages, such as NK cells (producing GM-CSF, IFNG, CCL3, CCL4) and monocytes (GM-CSF, IL12B, IL36G). Progressors exhibited early hyper-metabolic CD4⁺ T-cell programs and proliferative NK cell signatures, whereas non-progressors preferentially upregulated complement activation and CCL3/4-driven chemokine signaling in monocytes. Notably, among progressors, gene expression profiles within antigen-reactive CD4⁺ T-cells and monocytes predicted the timing of progression to active TB. Together, these findings reveal high frequencies and functional diversity of antigen-reactive cells in Mtb-exposed individuals and nominate tractable immune correlates for the rational design of next-generation TB vaccines.
Collapse
|
4
|
Shao M, Chen Q, Zhang X, Dong S, Wei R, Shi H, Yi F. Dynamic Alterations in DNA Methylation of CD4 + T Cells and Macrophages in a Murine Model of Tuberculous Pleural Infection Induced by BCG Vaccination. MedComm (Beijing) 2025; 6:e70166. [PMID: 40170749 PMCID: PMC11959155 DOI: 10.1002/mco2.70166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 04/03/2025] Open
Abstract
Tuberculous pleural effusion (TPE) is a prevalent form of extrapulmonary tuberculosis, and immune abnormalities play a crucial role in promoting its development. However, the dynamic changes and regulatory characteristics of immune cells during TPE progression remain incompletely understood. This study analyzed DNA methylation and transcriptome data from macrophages and CD4+ T cells from pleural lavage fluid of BCG-induced tuberculous pleurisy mouse models at specific time points (Days 0, 1, 7, and 14). The results revealed substantial alterations in DNA methylation patterns associated with inflammatory factors and interferon genes. Notably, macrophages exhibited the most pronounced differences in DNA methylation profiles on Day 1, while CD4+ T cells demonstrated gradual changes over time. The investigation further indicated that DNA methylation primarily regulated the differentiation of Th1, Th17, and Th22 cells but not Th9 cells. Additionally, single-cell RNA sequencing analysis revealed an increasing expression of C1q during infection, which was regulated by DNA methylation. Importantly, C1q+ and C1q- macrophages demonstrated distinct roles in modulating immune responses during infection. This research provides valuable insights into the DNA methylation profile of immune cells during Mycobacterium bovis infection-induced pleurisy in a mouse model, enhancing our understanding of the upstream regulatory mechanisms underlying immune response development in TPE.
Collapse
Affiliation(s)
- Ming‐Ming Shao
- Department of Respiratory and Critical Care MedicineBeijing Institute of Respiratory Medicine and Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
- Medical Research CenterBeijing Institute of Respiratory Medicine and Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Qing‐Yu Chen
- Department of Respiratory and Critical Care MedicineBeijing Institute of Respiratory Medicine and Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Xin Zhang
- Department of Respiratory and Critical Care MedicineBeijing Institute of Respiratory Medicine and Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Shu‐Feng Dong
- Department of Respiratory and Critical Care MedicineBeijing Institute of Respiratory Medicine and Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Rui‐Qi Wei
- Department of Respiratory and Critical Care MedicineBeijing Institute of Respiratory Medicine and Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Huan‐Zhong Shi
- Department of Respiratory and Critical Care MedicineBeijing Institute of Respiratory Medicine and Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Feng‐Shuang Yi
- Department of Respiratory and Critical Care MedicineBeijing Institute of Respiratory Medicine and Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
- Medical Research CenterBeijing Institute of Respiratory Medicine and Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
5
|
Chancharoenthana W, Kamolratanakul S, Rotcheewaphan S, Leelahavanichkul A, Schultz MJ. Recent advances in immunopathogenesis and clinical practice: mastering the challenge-managing of non-tuberculous mycobacteria. Front Immunol 2025; 16:1554544. [PMID: 40176807 PMCID: PMC11961655 DOI: 10.3389/fimmu.2025.1554544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/26/2025] [Indexed: 04/04/2025] Open
Abstract
Non-tuberculous mycobacteria (NTM) are widespread environmental pathogens that can lead to significant disease burden, particularly in immunocompromised individuals, but also in those with a normal immune system. The global incidence of NTM is increasing rapidly, with Mycobacterium avium complex (MAC) being one of the most common types. The immunopathogenesis of the MAC involves a complex interaction between the bacteria and the host immune system. MAC survives and replicates within macrophages by preventing the fusion of phagosomes and lysosomes. The mycobacteria can neutralize reactive oxygen and nitrogen species produced by the macrophages through their own enzymes. Additionally, MAC modulates cytokine production, allowing it to suppress or regulate the immune response. Diagnosing MAC infections can be challenging, and the effectiveness of available treatments may be limited due to MAC's unpredictable resistance to various antimycobacterial drugs in different regions. Treating MAC infection requires a collaborative approach involving different healthcare professionals and ensuring patient compliance. This review aims to shed light on the complexities of MAC infection treatment, discussing the challenges of MAC infection diagnosis, pharmacological considerations, such as drug regimens, drug monitoring, drug interactions, and the crucial role of a multidisciplinary healthcare team in achieving the best possible treatment outcomes for patients.
Collapse
Affiliation(s)
- Wiwat Chancharoenthana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Tropical Immunology and Translational Research Unit (TITRU), Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Supitcha Kamolratanakul
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Tropical Immunology and Translational Research Unit (TITRU), Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence on Translational Research in Inflammatory and Immunology (CETRII), Department of Microbiology, Chulalongkorn University, Bangkok, Thailand
| | - Marcus J. Schultz
- Department of Intensive Care & Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A), Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom
- Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
6
|
Gong Z, Xu H, Zhang Q, Wang G, Fan L, Wang Z, Fan L, Liu C, Yu Y, Liu Z, Zhou Q, Xiao H, Hou R, Zhao Y, Chen Y, Xie J. Unveiling the immunological landscape of disseminated tuberculosis: a single-cell transcriptome perspective. Front Immunol 2025; 16:1527592. [PMID: 40092995 PMCID: PMC11906432 DOI: 10.3389/fimmu.2025.1527592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/05/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Hematogenous disseminated tuberculosis (DTB) has an unclear etiology that likely involves multiple factors. Understanding the underlying immunological characteristics of DTB is crucial for elucidating its pathogenesis. Methods We conducted single-cell RNA transcriptome and T cell receptor (TCR) sequencing on samples from seven DTB patients. Additionally, we integrated and analyzed data from two published profiles of latent TB infection, three active TB cases, and two healthy controls. Results Our analysis revealed a significantly higher proportion of inflammatory immune cells (e.g., monocytes and macrophages) in DTB patients, along with a notably lower abundance of various lymphocytes (including T cells, B cells, and plasma cells), suggesting that lymphopenia is a prominent feature of the disease. T cell pseudotime analysis indicated a decrease in the expression of most hypervariable genes over time, pointing to T cell functional exhaustion. Furthermore, a marked absence of mucosal-associated invariant T (MAIT) cells was observed in the peripheral blood of DTB patients. In the TCR repertoire, specific polymorphisms (TRAV9-2, TRAV13-1, TRBV20-1, and TRBV5-1) and dominant clones (TRAJ49, TRBJ2-7, and TRBJ2-1) were identified. Analysis of the complementarity determining region 3 (CDR3) showed that the most frequent combination was TRAV1-2/TRAJ33, with the motif "CAAMD" being significantly reduced in DTB patients. Discussion These findings suggest that lymphopenia and T cell exhaustion, along with unique TCR signatures, may play critical roles in DTB pathogenesis. The reduced "CAAMD" motif and altered TCR clonotypes provide novel insights into the complex cellular dynamics associated with the disease, potentially offering new avenues for targeted immunological interventions.
Collapse
Affiliation(s)
- Zhen Gong
- Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, China
- Department of Clinical Laboratory, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hongxiang Xu
- Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, China
| | - Qiao Zhang
- Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, China
| | - Guirong Wang
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| | - Lin Fan
- Shanghai Clinical Research Center for Tuberculosis, Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zilu Wang
- Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, China
| | - Lichao Fan
- Shenyang Tenth People’s Hospital, Shenyang Chest Hospital, Shenyang, Liaoning, China
| | - Chang Liu
- Shenyang Tenth People’s Hospital, Shenyang Chest Hospital, Shenyang, Liaoning, China
| | - Yanhong Yu
- Shenyang Tenth People’s Hospital, Shenyang Chest Hospital, Shenyang, Liaoning, China
| | - Zhou Liu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Qiang Zhou
- Department of Clinical Laboratory, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | | | - Rui Hou
- Shanghai Biotechnology Corporation, Shanghai, China
| | - Ying Zhao
- Shanghai Biotechnology Corporation, Shanghai, China
| | - Yu Chen
- Shenyang Tenth People’s Hospital, Shenyang Chest Hospital, Shenyang, Liaoning, China
| | - Jianping Xie
- Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, China
| |
Collapse
|
7
|
Fu Y, Yang X, Ling Q, Huang Y, You X, Nie D, Sheng J, Chen Y, Wen Q, Zhou X, Zhou C, Hu S, Ma L. USP25 Promotes the Antimycobacterial Response of Macrophages Through Stabilizing B-Raf and C-Raf. J Infect Dis 2025; 231:366-377. [PMID: 39110031 DOI: 10.1093/infdis/jiae352] [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: 03/28/2024] [Accepted: 07/18/2024] [Indexed: 02/21/2025] Open
Abstract
Ubiquitin-specific peptidase 25 (USP25) is one of the best-characterized deubiquitinating enzymes and plays a vital regulatory role in various biological processes, especially in cancer development and immune regulation. However, the exact role of USP25 and its underlying mechanisms in macrophage activation and immunogenicity during Mycobacterium tuberculosis infection remain unclear. In this study, we found that M tuberculosis infection induced USP25 expression in human and mouse macrophages. In particular, USP25 expression is elevated in multiple cell types, especially monocytes, in patients with tuberculosis. Additionally, USP25 deficiency in macrophages and mice resulted in compromised immunity against M tuberculosis infection, accompanied by reduced expressions of various proinflammatory cytokines and chemokines. Mechanistically, USP25 in macrophages promoted the activation of the ERK signaling pathway through deubiquitination and stabilization of B-Raf and C-Raf. These findings collectively suggest the critical roles of USP25 in M tuberculosis infection and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Yuling Fu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University
| | - Xiaodan Yang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University
| | - Qiao Ling
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University
| | - Yulan Huang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University
| | - Xiaolong You
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University
| | - Dingnai Nie
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University
| | - Junli Sheng
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University
| | - Yitian Chen
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University
| | - Qian Wen
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University
| | - Xinying Zhou
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University
| | - Chaoying Zhou
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University
| | - Shengfeng Hu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University
- The Second Affiliated Hospital, The Second School of Clinical Medicine, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Guangzhou Medical University, China
| | - Li Ma
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University
| |
Collapse
|
8
|
Yang X, Chen Y, Pu B, Yuan X, Wang J, Chen C. YY1 Contributes to the Inflammatory Responses of Mycobacterium tuberculosis-Infected Macrophages Through Transcription Activation-Mediated Upregulation TLR4. Mol Biotechnol 2025; 67:778-789. [PMID: 38492118 DOI: 10.1007/s12033-024-01093-x] [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: 12/27/2023] [Accepted: 01/26/2024] [Indexed: 03/18/2024]
Abstract
Tuberculosis (TB) is a chronic respiratory infectious disease and is induced by Mycobacterium tuberculosis (M.tb) infection. Macrophages serve as the cellular home in immunoreaction against M.tb infection, which is tightly regulated through Toll-like receptor 4 (TLR4) expression. Therefore, this study is designed to explore the role and mechanism of TLR4 in mycobacterial injury in human macrophages (THP-1 cells) after M.tb infection. Cell proliferation and apoptosis were assessed using MTT, EdU, and flow cytometry assays. ELISA kits were utilized to assess the levels of Interleukin-6 (IL-6), IL-1β, and tumor necrosis factor α (TNF-α). The binding between Yin-Yang-1 (YY1) and TLR4 promoter was predicted by JASPAR and verified using Chromatin immunoprecipitation (ChIP) and dual-luciferase reporter assays. M.tb infection might repress THP-1 cell proliferation, and induce cell apoptosis and inflammatory response in a multiplicity of infection (MOI)-dependent manner. Moreover, M.tb infection increased the expression of TLR4 in HTP-1 cells in an MOI-dependent way, and its downregulation might overturn M.tb infection-mediated HTP-1 cell damage and inflammatory response. At the molecular level, YY1 was a transcription factor of TLR4 and promoted TLR4 transcription via binding to its promoter region. Besides, YY1 might activate the NF-kB signaling pathway via regulating TLR4. Meanwhile, TLR4 inhibitor BAY11-7082 might overturn the repression effect of TLR4 on M.tb-infected HTP-1 cell damage. YY1-activated TLR4 might aggravate mycobacterial injury in human macrophages after M.tb infection by the NF-kB pathway, providing a promising therapeutic target for TB treatment.
Collapse
Affiliation(s)
- Xing Yang
- Department of Preventive Health Care, Ren Huai People's Hospital, 2802, Building 3, Shengjie Community Harmony Square, Luban Street, Renhuai, Zunyi, Guizhou, China.
| | - Yu Chen
- Department of Health Management Division, Ren Huai People's Hospital, Zunyi, 564500, Guizhou, China
| | - Bingshuang Pu
- Department of Infectious Diseases, Ren Huai People's Hospital, Zunyi, 564500, Guizhou, China
| | - Xuan Yuan
- Department of Preventive Health Care, Ren Huai People's Hospital, 2802, Building 3, Shengjie Community Harmony Square, Luban Street, Renhuai, Zunyi, Guizhou, China
| | - Jiaojiao Wang
- Department of Preventive Health Care, Ren Huai People's Hospital, 2802, Building 3, Shengjie Community Harmony Square, Luban Street, Renhuai, Zunyi, Guizhou, China
| | - Chun Chen
- Department of Preventive Health Care, Ren Huai People's Hospital, 2802, Building 3, Shengjie Community Harmony Square, Luban Street, Renhuai, Zunyi, Guizhou, China
| |
Collapse
|
9
|
Mohammadnabi N, Shamseddin J, Emadi M, Bodaghi AB, Varseh M, Shariati A, Rezaei M, Dastranj M, Farahani A. Mycobacterium tuberculosis: The Mechanism of Pathogenicity, Immune Responses, and Diagnostic Challenges. J Clin Lab Anal 2024; 38:e25122. [PMID: 39593272 PMCID: PMC11632860 DOI: 10.1002/jcla.25122] [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: 03/20/2024] [Revised: 09/27/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND The infection caused by Mycobacterium tuberculosis arises from a complex interplay between the host immune system and the bacteria. Early and effective treatment of this disease is of great importance in order to prevent the emergence of drug-resistant strains. This necessitates the availability of fast and reliable diagnostic methods for managing affected cases. One reason why this study is significant is the lack of a comprehensive review in this field that thoroughly examines the importance, pathogenesis, and diagnosis of M. tuberculosis. Therefore, the aim of this review is to provide updated information on M. tuberculosis. METHODS We investigate the virulence factors, pathogenicity, and diagnostic methods of this bacterium, alongside the clinical symptoms and interpretation of different types of tuberculosis, including cerebral, miliary, nerve, and tubercular tuberculosis. RESULTS Mycobacterium tuberculosis acts as the causative agent of human tuberculosis and is regarded as one of the most adaptable human pathogens. M. tuberculosis possesses several virulence factors that help the bacterium evade mucous barriers. The rise of multidrug-resistant tuberculosis (MDR-TB) in both developing and industrialized countries emphasizes the need for rapid diagnostic methods. CONCLUSIONS Non-protein virulence factors play a crucial role in the pathogenicity of Mycobacterium tuberculosis (M. tuberculosis). The bacterial cell membrane contains proteins that modulate the host immune response. For instance, ESAT-6, either alone or in combination with CFP-10, reduces immune activity. While molecular techniques-such as DNA microarray, luciferase reporter assay, polymerase chain reaction (PCR), DNA and RNA probes, next-generation sequencing, and whole-genome sequencing-offer rapid, sensitive, and specific detection of M. tuberculosis, these methods are expensive and require technical expertise.
Collapse
Affiliation(s)
- Noura Mohammadnabi
- Student Research CommitteeKhomein University of Medical SciencesKhomeinIran
| | - Jebreil Shamseddin
- Infectious and Tropical Diseases Research Center, Hormozgan Health InstituteHormozgan University of Medical SciencesBandar AbbasIran
| | - Mobina Emadi
- Student Research CommitteeKhomein University of Medical SciencesKhomeinIran
| | - Ali Bayat Bodaghi
- Student Research CommitteeKhomein University of Medical SciencesKhomeinIran
| | - Mahdieh Varseh
- Student Research CommitteeKhomein University of Medical SciencesKhomeinIran
| | - Aref Shariati
- Infectious Diseases Research Center (IDRC)Arak University of Medical SciencesArakIran
| | - Mina Rezaei
- School of Environment, College of EngineeringUniversity of TehranTehranIran
| | - Mahsa Dastranj
- Microbiology Department, Kurdistan Science and Research BranchIslamic Azad UniversitySanandajIran
| | - Abbas Farahani
- Molecular Medicine Research CenterKhomein University of Medical SciencesKhomeinIran
- Department of Basic SciencesKhomein University of Medical SciencesKhomeinIran
| |
Collapse
|
10
|
Grace PS, Peters JM, Sixsmith J, Lu R, Luedeman C, Fenderson BA, Vickers A, Slein MD, Irvine EB, McKitrick T, Wei MH, Cummings RD, Wallace A, Cavacini LA, Choudhary A, Proulx MK, Sundling C, Källenius G, Reljic R, Ernst JD, Casadevall A, Locht C, Pinter A, Sasseti CM, Bryson BD, Fortune SM, Alter G. Antibody-Fab and -Fc features promote Mycobacterium tuberculosis restriction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617070. [PMID: 39416184 PMCID: PMC11482752 DOI: 10.1101/2024.10.07.617070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), is a leading cause of death by an infectious disease globally, with no efficacious vaccine. Antibodies are implicated in Mtb control, but the mechanisms of antibody action remain poorly understood. We assembled a library of TB monoclonal antibodies (mAb) and screened for the ability to restrict Mtb in mice, identifying protective antibodies targeting known and novel antigens. To dissect the mechanism of mAb-mediated Mtb restriction, we optimized a protective lipoarabinomannan-specific mAb through Fc-swapping. In vivo analysis of these Fc-variants revealed a critical role for Fc-effector function in Mtb restriction. Restrictive Fc-variants altered distribution of Mtb across innate immune cells. Single-cell transcriptomics highlighted distinctly activated molecular circuitry within innate immune cell subpopulations, highlighting early activation of neutrophils as a key signature of mAb-mediated Mtb restriction. Therefore, improved antibody-mediated restriction of Mtb is associated with reorganization of the tissue-level immune response to infection and depends on the collaboration of antibody Fab and Fc.
Collapse
|
11
|
Du W, Zhao Y, Zhang C, Zhang L, Zhou L, Sun Z, Huang X, Zhang N, Liu Z, Li K, Che N. Association of bacteriomes with drug susceptibility in lesions of pulmonary tuberculosis patients. Heliyon 2024; 10:e37583. [PMID: 39309911 PMCID: PMC11414563 DOI: 10.1016/j.heliyon.2024.e37583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/25/2024] Open
Abstract
Understanding how the bacteriomes in tuberculous lesions can be influenced by the susceptibility of Mycobacterium tuberculosis (MTB) can provide valuable information for preventing and treating drug resistant tuberculosis (DR-TB). High-throughput 16S rRNA sequencing was employed to analyze the bacteriome in pulmonary TB lesions from 14 patients with DR-TB and 47 patients with drug sensitive tuberculosis (DS-TB), along with 18 normal lung tissues (NT) from 18 lung cancer patients serving as the bacterial baseline. The phylogenetic investigation of communities by reconstruction of unobserved states2 (PICRUSt2) algorithm was utilized to predict bacterial metabolic functions. The major phyla of pulmonary bacteriomes included Proteobacteria, Actinobacteria, Bacteroidetes, Firmicutes and Fusobacteria. Alpha diversity indices, including ACE, Chao1, Shannon and OTU observed, all demonstrated different bacterial communities of DS-TB samples from that of NT samples; while only Shannon indicated difference between DR-TB and NT samples. The analysis of similarity (ANOSIM) showed significantly different bacterial communities within TB lesions compared to NT samples (R = 0.418, p = 0.001). However, difference was not observed between DR-TB and DS-TB samples (ANOSIM, R = 0.069, p = 0.173). The bacterial profiles within each DR-TB individual appeared unique, with no obvious clusters corresponding to drug-resistant phenotypes. Nevertheless, indicator genera identified in DR-TB and DS-TB lesions demonstrated distinctive micro-ecological environments. Most COG functions were enriched in TB lesions, and the most significant one was [J] translation, ribosomal structure and biogenesis. The distinct enrichment patterns of bacterial enzymes in DR-TB and DS-TB lesions suggest that pulmonary bacterial activities can be modulated by the susceptibility of MTB bacilli. This study provides fresh perspectives and strategies for the precise diagnosis and assessment of drug resistance tuberculosis.
Collapse
Affiliation(s)
- Weili Du
- Department of Pathology, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beiguandajie 9#, Tongzhou Dist, Beijing, 101149, China
| | - Yingli Zhao
- Department of Pathology, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beiguandajie 9#, Tongzhou Dist, Beijing, 101149, China
| | - Chen Zhang
- Department of Pathology, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beiguandajie 9#, Tongzhou Dist, Beijing, 101149, China
| | - Li Zhang
- Department of Pathology, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beiguandajie 9#, Tongzhou Dist, Beijing, 101149, China
| | - Lijuan Zhou
- Department of Pathology, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beiguandajie 9#, Tongzhou Dist, Beijing, 101149, China
| | - Zuyu Sun
- Department of Pathology, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beiguandajie 9#, Tongzhou Dist, Beijing, 101149, China
| | - Xiaojie Huang
- Department of Pathology, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beiguandajie 9#, Tongzhou Dist, Beijing, 101149, China
| | - Nana Zhang
- Department of Pathology, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beiguandajie 9#, Tongzhou Dist, Beijing, 101149, China
| | - Zichen Liu
- Department of Pathology, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beiguandajie 9#, Tongzhou Dist, Beijing, 101149, China
| | - Kun Li
- Department of Pathology, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beiguandajie 9#, Tongzhou Dist, Beijing, 101149, China
| | - Nanying Che
- Department of Pathology, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beiguandajie 9#, Tongzhou Dist, Beijing, 101149, China
| |
Collapse
|
12
|
Shen J, Han L, Yao J, Qiu X, Xu S, Liu X, Li F, Li Z. Infection route influence the consequences of Nocardia farcinica infection in BALB/c mice. BMC Infect Dis 2024; 24:1016. [PMID: 39304798 DOI: 10.1186/s12879-024-09877-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 09/04/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Nocardia, a rare but potentially fatal pathogen, can induce systemic infections with diverse manifestations. This study aimed to investigate the tissue and organ damage caused by Nocardia farcinica (N. farcinica) in mice via different infection routes, evaluate the resulting host immune responses, and assess its invasiveness in brain tissue. METHODS BALB/c mice were infected with N. farcinica through intranasal, intraperitoneal, and intravenous routes (doses: 1 × 10^8, 1 × 10^7, 1 × 10^7 CFU in 50 µl PBS). Over a 7-day period, body temperature, weight, and mortality were monitored, and samples were collected for histopathological analysis and bacterial load assessment. Serum was isolated for cytokine detection via ELISA. For RNA-seq analysis, mice were infected with 1 × 107 CFU through three infection routes, after which brain tissue was harvested. RESULTS Intraperitoneal and intravenous N. farcinica infections caused significant clinical symptoms, mortality, and neural disruption in mice, resulting in severe systemic infection. Conversely, intranasal infection primarily affected the lungs without causing significant damage to other organs. Intraperitoneal and intravenous infections significantly increased serum cytokines, particularly TNF-α and IFN-γ. RNA-seq analysis of brains from intravenously infected mice revealed significant differential gene expression, whereas the intranasal and intraperitoneal routes showed limited differences (only three genes). The enriched Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways in the intravenous group were primarily related to immune processes. CONCLUSION The study demonstrated that intravenous N. farcinica infection induces significant clinical symptoms, triggers an inflammatory response, damages multiple organs, and leads to systemic infections.
Collapse
Affiliation(s)
- Jirao Shen
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Lichao Han
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jiang Yao
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaotong Qiu
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Shuai Xu
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xueping Liu
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Fang Li
- Department of Medicine, Tibet University, Lhasa, Tibet, 850000, PR China
| | - Zhenjun Li
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
| |
Collapse
|
13
|
Peng X, Pu F, Zhou F, Dai X, Xu F, Wang J, Feng J, Xia P. Exploring expression levels of the cGAS-STING pathway genes in peripheral blood mononuclear cells of spinal tuberculosis patients. BMC Infect Dis 2024; 24:915. [PMID: 39232642 PMCID: PMC11373091 DOI: 10.1186/s12879-024-09815-x] [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: 04/04/2024] [Accepted: 08/27/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND This study aimed to investigate the differential expression levels of the cGAS-STING pathway in peripheral blood mononuclear cells (PBMCs) of spinal tuberculosis (TB) patients with different progression and its feasibility as a diagnostic marker. METHODS Peripheral blood and medical records of 25 patients with spinal TB and 10 healthy individuals, were prospectively collected and analyzed. PBMCs and serum were extracted from peripheral blood and the expression levels of the cGAS-STING pathway in PBMCs were measured by real-time PCR (RT-PCR) and serum interferon β (IFN-β) expression levels were measured by enzyme-linked immunosorbent assay (ELISA). The expression of Interferon regulatory Factor 3 (IRF3) in PBMCs was measured using western blot. Statistical analysis was performed using the SPSS 26.0 statistical package. RESULTS The results showed that the expression level of the TANK-binding kinase 1 (TBK1) and IRF3 was significantly higher in PBMCs (P < 0.05), in patients with active lesions than in patients with stable lesions. The serum concentration of IFN-β was significantly higher in patients with active lesions (P = 0.028). Compared with healthy individuals, the expression level of the cGAS-STING pathway was elevated in PBMCs of TB patients (P < 0.05), and the difference in the expression level of IFN-β was not statistically significant (P > 0.05), and the serum IFN-β concentration was elevated (P < 0.05). The calculated AUC values for TBK1 and IRF3 in PBMCs, IFN-β in serum and erythrocyte sedimentation rate (ESR) to distinguish between patients with active and stable lesions were 0.732, 0.714, 0.839, and 0.714 respectively. CONCLUSIONS The expression level of TBK1 and IRF3 in PBMCs, and IFN-β in the serum of patients with spinal TB is positively correlated with disease activity. TBK1 has higher specificity and IFN-β in serum has higher sensitivity when used to differentiate between patients with active and stable lesions.
Collapse
Affiliation(s)
- Xianglin Peng
- Department of Orthopedics, Tongji Medical College, Traditional Chinese and Western Medicine Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan, 430022, China
| | - Feifei Pu
- Department of Orthopedics, Tongji Medical College, Traditional Chinese and Western Medicine Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan, 430022, China
| | - Fangzheng Zhou
- Department of Orthopedics, Tongji Medical College, Traditional Chinese and Western Medicine Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan, 430022, China
| | - Xiyong Dai
- Wuhan Pulmonary Hospital, Wuhan Institute for Tuberculosis Control, Wuhan, 430022, China
| | - Feng Xu
- Wuhan Pulmonary Hospital, Wuhan Institute for Tuberculosis Control, Wuhan, 430022, China
| | - Junwen Wang
- Department of Orthopedics, Wuhan Fourth Hospital, Puai Hospital, Wuhan, 430030, China
| | - Jing Feng
- Department of Orthopedics, Tongji Medical College, Traditional Chinese and Western Medicine Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan, 430022, China.
| | - Ping Xia
- Department of Orthopedics, Wuhan Fourth Hospital, Puai Hospital, Wuhan, 430030, China.
| |
Collapse
|
14
|
Nduba V, Njagi LN, Murithi W, Mwongera Z, Byers J, Logioia G, Peterson G, Segnitz RM, Fennelly K, Hawn TR, Horne DJ. Mycobacterium tuberculosis cough aerosol culture status associates with host characteristics and inflammatory profiles. Nat Commun 2024; 15:7604. [PMID: 39217183 PMCID: PMC11365933 DOI: 10.1038/s41467-024-52122-x] [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] [Received: 12/15/2023] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
Interrupting transmission events is critical to tuberculosis control. Cough-generated aerosol cultures predict tuberculosis transmission better than microbiological or clinical markers. We hypothesize that highly infectious individuals with pulmonary tuberculosis (positive for cough aerosol cultures) have elevated inflammatory markers and unique transcriptional profiles compared to less infectious individuals. We performed a prospective, longitudinal study using cough aerosol sampling system. We enrolled 142 participants with treatment-naïve pulmonary tuberculosis in Kenya and assessed the association of clinical, microbiologic, and immunologic characteristics with Mycobacterium tuberculosis aerosolization and transmission in 129 household members. Contacts of the forty-three aerosol culture-positive participants (30%) are more likely to have a positive interferon-gamma release assay (85% vs 53%, P = 0.006) and higher median IFNγ level (P < 0.001, 4.28 IU/ml (1.77-5.91) vs. 0.71 (0.01-3.56)) compared to aerosol culture-negative individuals. We find that higher bacillary burden, younger age, larger mean upper arm circumference, and host inflammatory profiles, including elevated serum C-reactive protein and lower plasma TNF levels, associate with positive cough aerosol cultures. Notably, we find pre-treatment whole blood transcriptional profiles associate with aerosol culture status, independent of bacillary load. These findings suggest that tuberculosis infectiousness is associated with epidemiologic characteristics and inflammatory signatures and that these features may identify highly infectious persons.
Collapse
Affiliation(s)
- Videlis Nduba
- Centre for Respiratory Diseases Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Lilian N Njagi
- Centre for Respiratory Diseases Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Wilfred Murithi
- Centre for Respiratory Diseases Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Zipporah Mwongera
- Centre for Respiratory Diseases Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Jodi Byers
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Gisella Logioia
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Glenna Peterson
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - R Max Segnitz
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Kevin Fennelly
- Division of Intramural Research, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, MD, USA
| | - Thomas R Hawn
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - David J Horne
- Department of Global Health, University of Washington, Seattle, WA, USA.
- Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
15
|
Niu L, Wang H, Luo G, Zhou J, Hu Z, Yan B. Advances in understanding immune homeostasis in latent tuberculosis infection. WIREs Mech Dis 2024; 16:e1643. [PMID: 38351551 DOI: 10.1002/wsbm.1643] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/19/2024] [Accepted: 01/19/2024] [Indexed: 07/13/2024]
Abstract
Nearly one-fourth of the global population is infected by Mycobacterium tuberculosis (Mtb), and approximately 90%-95% remain asymptomatic as latent tuberculosis infection (LTBI), an estimated 5%-10% of those with latent infections will eventually progress to active tuberculosis (ATB). Although it is widely accepted that LTBI transitioning to ATB results from a disruption of host immune balance and a weakening of protective immune responses, the exact underlying immunological mechanisms that promote this conversion are not well characterized. Thus, it is difficult to accurately predict tuberculosis (TB) progression in advance, leaving the LTBI population as a significant threat to TB prevention and control. This article systematically explores three aspects related to the immunoregulatory mechanisms and translational research about LTBI: (1) the distinct immunocytological characteristics of LTBI and ATB, (2) LTBI diagnostic markers discovery related to host anti-TB immunity and metabolic pathways, and (3) vaccine development focus on LTBI. This article is categorized under: Infectious Diseases > Molecular and Cellular Physiology Infectious Diseases > Genetics/Genomics/Epigenetics Immune System Diseases > Genetics/Genomics/Epigenetics.
Collapse
Affiliation(s)
- Liangfei Niu
- Center for Tuberculosis Research, Shanghai Public Health Clinical Center, Fudan University, Shanghai, People's Republic of China
| | - Hao Wang
- Center for Tuberculosis Research, Shanghai Public Health Clinical Center, Fudan University, Shanghai, People's Republic of China
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China
| | - Geyang Luo
- Center for Tuberculosis Research, Shanghai Public Health Clinical Center, Fudan University, Shanghai, People's Republic of China
| | - Jing Zhou
- Department of Pathology, Center for Tuberculosis Research, Shanghai Public Health Clinical Center, Fudan University, Shanghai, People's Republic of China
| | - Zhidong Hu
- Center for Tuberculosis Research, Shanghai Public Health Clinical Center, Fudan University, Shanghai, People's Republic of China
| | - Bo Yan
- Center for Tuberculosis Research, Shanghai Public Health Clinical Center, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
16
|
Dhanyalayam D, Thangavel H, Sidrat T, Oswal N, Lizardo K, Mauro M, Zhao X, Xue HH, Desai JV, Nagajyothi JF. The Influence of Body Fat Dynamics on Pulmonary Immune Responses in Murine Tuberculosis: Unraveling Sex-Specific Insights. Int J Mol Sci 2024; 25:6823. [PMID: 38999932 PMCID: PMC11241512 DOI: 10.3390/ijms25136823] [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: 04/08/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
The World Health Organization (WHO) highlights a greater susceptibility of males to tuberculosis (TB), a vulnerability attributed to sex-specific variations in body fat and dietary factors. Our study delves into the unexplored terrain of how alterations in body fat influence Mycobacterium tuberculosis (Mtb) burden, lung pathology, immune responses, and gene expression, with a focus on sex-specific dynamics. Utilizing a low-dose Mtb-HN878 clinical strain infection model, we employ transgenic FAT-ATTAC mice with modulable body fat to explore the impact of fat loss (via fat ablation) and fat gain (via a medium-fat diet, MFD). Firstly, our investigation unveils that Mtb infection triggers severe pulmonary pathology in males, marked by shifts in metabolic signaling involving heightened lipid hydrolysis and proinflammatory signaling driven by IL-6 and localized pro-inflammatory CD8+ cells. This stands in stark contrast to females on a control regular diet (RD). Secondly, our findings indicate that both fat loss and fat gain in males lead to significantly elevated (1.6-fold (p ≤ 0.01) and 1.7-fold (p ≤ 0.001), respectively) Mtb burden in the lungs compared to females during Mtb infection (where fat loss and gain did not alter Mtb load in the lungs). This upsurge is associated with impaired lung lipid metabolism and intensified mitochondrial oxidative phosphorylation-regulated activity in lung CD8+ cells during Mtb infection. Additionally, our research brings to light that females exhibit a more robust systemic IFNγ (p ≤ 0.001) response than males during Mtb infection. This heightened response may either prevent active disease or contribute to latency in females during Mtb infection. In summary, our comprehensive analysis of the interplay between body fat changes and sex bias in Mtb infection reveals that alterations in body fat critically impact pulmonary pathology in males. Specifically, these changes significantly reduce the levels of pulmonary CD8+ T-cells and increase the Mtb burden in the lungs compared to females. The reduction in CD8+ cells in males is linked to an increase in mitochondrial oxidative phosphorylation and a decrease in TNFα, which are essential for CD8+ cell activation.
Collapse
Affiliation(s)
- Dhanya Dhanyalayam
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Hariprasad Thangavel
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Tabinda Sidrat
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Neelam Oswal
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Kezia Lizardo
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Michael Mauro
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Xin Zhao
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Hai-Hui Xue
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Jigar V Desai
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Jyothi F Nagajyothi
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| |
Collapse
|
17
|
Yu Y, Hua J, Chen L. Autophagy-related molecular clusters identified as indicators for distinguishing active and latent TB infection in pediatric patients. BMC Pediatr 2024; 24:398. [PMID: 38890657 PMCID: PMC11186109 DOI: 10.1186/s12887-024-04881-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 06/11/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Autophagy is crucial for controlling the manifestation of tuberculosis. This study intends to discover autophagy-related molecular clusters as biomarkers for discriminating between latent tuberculosis (LTBI) and active tuberculosis (ATB) in children through gene expression profile analysis. METHODS The expression of autophagy modulators was examined in pediatric patients with LTBI and ATB utilizing public datasets from the Gene Expression Omnibus (GEO) collection (GSE39939 and GSE39940). RESULTS In a training dataset (GSE39939), patients with LTBI and ATB exhibited the expression of autophagy-related genes connected with their active immune responses. Two molecular clusters associated with autophagy were identified. Compared to Cluster 1, Cluster 2 was distinguished through decreased adaptive cellular immune response and enhanced inflammatory activation, according to single-sample gene set enrichment analysis (ssGSEA). Per the study of gene set variation, Cluster 2's differentially expressed genes (DEGs) played a role in synthesizing transfer RNA, DNA repair and recombination, and primary immunodeficiency. The peak variation efficiency, root mean square error, and area under the curve (AUC) (AUC = 0.950) were all lowered in random forest models. Finally, a seven-gene-dependent random forest profile was created utilizing the CD247, MAN1C1, FAM84B, HSZFP36, SLC16A10, DTX3, and SIRT4 genes, which performed well against the validation dataset GSE139940 (AUC = 0.888). The nomogram calibration and decision curves performed well in identifying ATB from LTBI. CONCLUSIONS In summary, according to the present investigation, autophagy and the immunopathology of TB might be correlated. Furthermore, this investigation established a compelling prediction expression profile for measuring autophagy subtype development risks, which might be employed as possible biomarkers in children to differentiate ATB from LTBI.
Collapse
Affiliation(s)
- Yang Yu
- Department of Pediatric, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| | - Jie Hua
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liang Chen
- Department of Infectious Diseases, Taikang Xianlin Drum Tower Hospital, Affiliated Hospital of Medical College of Nanjing University, Qixia District, NO 188, Lingshan North Road, Qixia District, Nanjing, 210046, China.
| |
Collapse
|
18
|
Tong F, Lai J, Lu Z, Bao Z, Cao J. Clinical features, immunologic parameter and treatment outcome of Chinese tuberculosis patients with or without DM. Front Med (Lausanne) 2024; 11:1386124. [PMID: 38933114 PMCID: PMC11199526 DOI: 10.3389/fmed.2024.1386124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Background The coexistence of diabetes mellitus (DM) and pulmonary tuberculosis (PTB) poses a significant health concern globally, with their convergence presenting a considerable challenge to healthcare systems. Previous research has highlighted that comorbidities can mutually influence and exacerbate immune disorders. However, there is a paucity of data on the impact of DM on immunological features and treatment responses in the TB population in China. Methods From January 2020 to June 2022, 264 cases of pulmonary tuberculosis patients (82 DM patients and 182 non-DM patients) hospitalized in our center were selected. 80 patients with TB with DM (TB-DM) and 80 patients with TB without DM (TB-NDM) were enrolled into the final analysis by propensity score matching for age, gender and involved lung field at a ratio of 1:1. The clinical characteristics, immunological features and treatment response were compared between the two groups. Results After propensity score matching, no differences in the general features such as age gender, involved lung field, the incidence of retreatment and WBC count were found between the two groups. Compared to TB-NDM group, the TB-DM group exhibited a higher positive rate of sputum smear and incidence of cavitary lesions. Immunological features analysis revealed that the TB-DM patients had higher levels of TNF-α [pg/ml; 8.56 (7.08-13.35) vs. 7.64 (6.38-10.14) p = 0.033] and IL-8 [pg/ml; 25.85 (11.63-58.40) vs. 17.56 (6.44-39.08) p = 0.003] but lower CD8+ T lymphocyte count [cells/mm3; 334.02 (249.35-420.71) VS 380.95 (291.73-471.25) p = 0.038]. However, there was no significant difference in serum IL-6 concentration and CD4+ T lymphocyte count between the two groups. After 2 months of anti-tuberculosis treatment, 39 (24.4%) cases had suboptimal treatment response, including 23 (28.7%) TB-DM patients and 16 (20%) TB-NDM patients. There was no difference in suboptimal response rate (SRR) was found between the two groups (p = 0.269). The multivariate logistic regression analysis indicated that retreatment for TB [AOR: 5.68 (95%CI: 2.01-16.08), p = 0.001], sputum smear positivity [AOR: 8.01 (95%CI: 2.62-24.50), p = 0.001] were associated with SRR in all participants, and in TB-DM group, only sputum smear positivity [AOR: 16.47 (1.75-155.12), p = 0.014] was positive with SRR. Conclusion DM is a risk factor for pulmonary cavity formation and sputum smear positivity in TB population. Additionally, TB-DM patients is characterized by enhanced cytokine responses and decreased CD8+ T lymphocytes. The retreatment for TB and sputum smear positivity were associated with the occurrence of suboptimal treatment response.
Collapse
Affiliation(s)
- Fengjun Tong
- Department of Infectious Diseases, Qingchun Hospital, Hangzhou, China
| | - Jie Lai
- Department of Infectious Diseases, Qingchun Hospital, Hangzhou, China
| | - Zhenhui Lu
- Department of Infectious Diseases, Qingchun Hospital, Hangzhou, China
| | - Zhijian Bao
- Zhejiang Tuberculosis Diagnosis and Treatment Center, Hangzhou Red Cross Hospital, Hangzhou, China
| | - Junyan Cao
- Department of Infectious Diseases, Qingchun Hospital, Hangzhou, China
| |
Collapse
|
19
|
Zhang J, Chen J, Zhang Y, Chen L, Mo W, Yang Q, Zhang M, Liu H. Exploring TSPAN4 promoter methylation as a diagnostic biomarker for tuberculosis. Front Genet 2024; 15:1380828. [PMID: 38680421 PMCID: PMC11048481 DOI: 10.3389/fgene.2024.1380828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/01/2024] [Indexed: 05/01/2024] Open
Abstract
Background Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is a persistent infectious disease threatening human health. The existing diagnostic methods still have significant shortcomings, including a low positivity rate in pathogen-based diagnoses and the inability of immunological diagnostics to detect active TB. Hence, it is urgent to develop new techniques to detect TB more accurate and earlier. This research aims to scrutinize and authenticate DNA methylation markers suitable for tuberculosis diagnosis. Concurrently, Providing a new approach for tuberculosis diagnosis. Methods Blood samples from patients with newly diagnosed tuberculosis and healthy controls (HC) were utilized in this study. Examining methylation microarray data from 40 whole blood samples (22TB + 18HC), we employed two procedures: signature gene methylated position analysis and signature region methylated position analysis to pinpoint distinctive methylated positions. Based on the screening results, diagnostic classifiers are constructed through machine learning, and validation was conducted through pyrosequencing in a separate queue (22TB + 18HC). Culminating in the development of a new tuberculosis diagnostic method via quantitative real-time methylation specific PCR (qMSP). Results The combination of the two procedures revealed a total of 10 methylated positions, all of which were located in the promoter region. These 10 signature methylated positions facilitated the construction of a diagnostic classifier, exhibiting robust diagnostic accuracy in both cross-validation and external test sets. The LDA model demonstrated the best classification performance, achieving an AUC of 0.83, specificity of 0.8, and sensitivity of 0.86 on the external test set. Furthermore, the validation of signature methylated positions through pyrosequencing demonstrated high agreement with screening outcomes. Additionally, qMSP detection of 2 potential hypomethylated positions (cg04552852 and cg12464638) exhibited promising results, yielding an AUC of 0.794, specificity of 0.720, and sensitivity of 0.816. Conclusion Our study demonstrates that the validated signature methylated positions through pyrosequencing emerge as plausible biomarkers for tuberculosis diagnosis. The specific methylation markers in the TSPAN4 gene, identified in whole blood samples, hold promise for improving tuberculosis diagnosis. This approach could significantly enhance diagnostic accuracy and speed, offering a new avenue for early detection and treatment.
Collapse
Affiliation(s)
- Jiahao Zhang
- National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Institute of Pathogen Biology and Center for Tuberculosis Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jilong Chen
- National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Institute of Pathogen Biology and Center for Tuberculosis Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Zhang
- National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Institute of Pathogen Biology and Center for Tuberculosis Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liuchi Chen
- National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Institute of Pathogen Biology and Center for Tuberculosis Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weiwei Mo
- National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Institute of Pathogen Biology and Center for Tuberculosis Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qianting Yang
- Shenzhen Clinical Research Center for Tuberculosis, Shenzhen, China
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Institute for Hepatology, Southern University of Science and Technology, Shenzhen, China
| | - Mingxia Zhang
- Shenzhen Clinical Research Center for Tuberculosis, Shenzhen, China
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Institute for Hepatology, Southern University of Science and Technology, Shenzhen, China
| | - Haiying Liu
- National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Institute of Pathogen Biology and Center for Tuberculosis Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
20
|
Matos ADO, Dantas PHDS, Queiroz HAGDB, Silva-Sales M, Sales-Campos H. TREM-2: friend or foe in infectious diseases? Crit Rev Microbiol 2024; 50:1-19. [PMID: 36403150 DOI: 10.1080/1040841x.2022.2146481] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 11/07/2022] [Indexed: 11/21/2022]
Abstract
The triggering receptor expressed on myeloid cells-2 (TREM-2) is an immune receptor expressed on immune and non-immune cells, more frequently investigated in neurodegenerative disorders and considered a marker for microglia activation. In infectious diseases, the receptor was initially believed to be an anti-inflammatory molecule, opposing the inflammation triggered by TREM-1. Currently, TREM-2 is associated with different aspects in response to infectious stimuli, including the induction of bacterial phagocytosis and clearance, containment of exacerbated pro-inflammatory responses, induction of M2 differentiation and activation of Th1 lymphocytes, besides of neurological damage after viral infection. Here, we present and discuss results published in the last two decades regarding the expression, activation and functions of TREM-2 during the course of bacterial, viral, fungal and parasitic infections. A surprisingly plasticity was observed regarding the roles of the receptor in the aforementioned contexts, which largely varied according to the cell/organ and pathogen type, besides influencing disease outcome. Therefore, our review aimed to critically overview the role of TREM-2 in infectious diseases, highlighting its potential to be used as a clinical biomarker or therapeutic target.
Collapse
Affiliation(s)
| | | | | | - Marcelle Silva-Sales
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
| | | |
Collapse
|
21
|
Panda S, Morgan J, Cheng C, Saito M, Gilman RH, Ciobanu N, Crudu V, Catanzaro DG, Catanzaro A, Rodwell T, Perera JSB, Chathuranga T, Gunasena B, DeSilva AD, Peters B, Sette A, Lindestam Arlehamn CS. Identification of differentially recognized T cell epitopes in the spectrum of tuberculosis infection. Nat Commun 2024; 15:765. [PMID: 38278794 PMCID: PMC10817963 DOI: 10.1038/s41467-024-45058-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 01/12/2024] [Indexed: 01/28/2024] Open
Abstract
There is still incomplete knowledge of which Mycobacterium tuberculosis (Mtb) antigens can trigger distinct T cell responses at different stages of infection. Here, a proteome-wide screen of 20,610 Mtb-derived peptides in 21 patients mid-treatment for active tuberculosis (ATB) reveals IFNγ-specific T cell responses against 137 unique epitopes. Of these, 16% are recognized by two or more participants and predominantly derived from cell wall and cell processes antigens. There is differential recognition of antigens, including TB vaccine candidate antigens, between ATB participants and interferon-gamma release assay (IGRA + /-) individuals. We developed an ATB-specific peptide pool (ATB116) consisting of epitopes exclusively recognized by ATB participants. This pool can distinguish patients with pulmonary ATB from IGRA + /- individuals from various geographical locations, with a sensitivity of over 60% and a specificity exceeding 80%. This proteome-wide screen of T cell reactivity identified infection stage-specific epitopes and antigens for potential use in diagnostics and measuring Mtb-specific immune responses.
Collapse
Affiliation(s)
- Sudhasini Panda
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Jeffrey Morgan
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Catherine Cheng
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Mayuko Saito
- Department of Virology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Robert H Gilman
- Johns Hopkins School of Public Health, Baltimore, MD, USA
- Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Nelly Ciobanu
- Phthisiopneumology Institute, Chisinau, Republic of Moldova
| | - Valeriu Crudu
- Phthisiopneumology Institute, Chisinau, Republic of Moldova
| | - Donald G Catanzaro
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR, USA
| | - Antonino Catanzaro
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Timothy Rodwell
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Judy S B Perera
- Faculty of Medicine, General Sir John Kotelawala Defense University, Ratmalana, Sri Lanka
| | - Teshan Chathuranga
- Faculty of Medicine, General Sir John Kotelawala Defense University, Ratmalana, Sri Lanka
| | - Bandu Gunasena
- National Hospital for Respiratory Diseases, Welisara, Sri Lanka
| | - Aruna D DeSilva
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Faculty of Medicine, General Sir John Kotelawala Defense University, Ratmalana, Sri Lanka
| | - Bjoern Peters
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | | |
Collapse
|
22
|
Dai X, Zhou L, He X, Hua J, Chen L, Lu Y. Identification of apoptosis-related gene signatures as potential biomarkers for differentiating active from latent tuberculosis via bioinformatics analysis. Front Cell Infect Microbiol 2024; 14:1285493. [PMID: 38312744 PMCID: PMC10834671 DOI: 10.3389/fcimb.2024.1285493] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/02/2024] [Indexed: 02/06/2024] Open
Abstract
Background Apoptosis is associated with the pathogenesis of Mycobacterium tuberculosis infection. This study aims to identify apoptosis-related genes as biomarkers for differentiating active tuberculosis (ATB) from latent tuberculosis infection (LTBI). Methods The tuberculosis (TB) datasets (GSE19491, GSE62525, and GSE28623) were downloaded from the Gene Expression Omnibus (GEO) database. The diagnostic biomarkers differentiating ATB from LTBI were identified by combining the data of protein-protein interaction network, differentially expressed gene, Weighted Gene Co-Expression Network Analysis (WGCNA), and receiver operating characteristic (ROC) analyses. Machine learning algorithms were employed to validate the diagnostic ability of the biomarkers. Enrichment analysis for biomarkers was established, and potential drugs were predicted. The association between biomarkers and N6-methyladenosine (m6A) or 5-methylated cytosine (m5C) regulators was evaluated. Results Six biomarkers including CASP1, TNFSF10, CASP4, CASP5, IFI16, and ATF3 were obtained for differentiating ATB from LTBI. They showed strong diagnostic performances, with area under ROC (AUC) values > 0.7. Enrichment analysis demonstrated that the biomarkers were involved in immune and inflammation responses. Furthermore, 24 drugs, including progesterone and emricasan, were predicted. The correlation analysis revealed that biomarkers were positively correlated with most m6A or m5C regulators. Conclusion The six ARGs can serve as effective biomarkers differentiating ATB from LTBI and provide insight into the pathogenesis of Mycobacterium tuberculosis infection.
Collapse
Affiliation(s)
- Xiaoting Dai
- Department of Infectious Diseases, Nanjing Lishui People’s Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| | - Litian Zhou
- Department of Neurosugery, Nanjing Lishui People’s Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| | - Xiaopu He
- Department of Geriatric Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Hua
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liang Chen
- Department of Infectious Diseases, Nanjing Lishui People’s Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| | - Yingying Lu
- Department of Clinical Laboratory, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Clinical Laboratory, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| |
Collapse
|
23
|
Rohilla A, Singh AK, Koleske B, Srikrishna G, Bishai WR. Structure-based virtual screening and in vitro validation of inhibitors of cyclic dinucleotide phosphodiesterases ENPP1 and CdnP. Microbiol Spectr 2024; 12:e0201223. [PMID: 38095464 PMCID: PMC10783014 DOI: 10.1128/spectrum.02012-23] [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: 05/12/2023] [Accepted: 11/21/2023] [Indexed: 01/13/2024] Open
Abstract
IMPORTANCE In this paper, we describe novel inhibitors of cyclic dinucleotide phosphodiesterase enzymes from Mycobacterium tuberculosis (M.tb) (CdnP) and mammals (ENPP1). The phosphodiesterase enzymes hydrolyze cyclic dinucleotides, such as 2',3'-cyclic GMP-AMP and c-di-AMP, which are stimulator of interferon gene (STING) agonists. By blocking the hydrolysis of STING agonists, the cyclic GMP-AMP synthase (cGAS)-STING-IRF3 pathway is potentiated. There is strong evidence in tuberculosis and in cancer biology that potentiation of the cGAS-STING-IRF3 pathway leads to improved M.tb clearance and also improved antitumor responses in cancer. In addition to the identification of novel inhibitors and their biochemical characterization, we provide proof-of-concept evidence that our E-3 inhibitor potentiates the cGAS-STING-IRF3 pathway in both macrophage cell lines and also in primary human monocyte-derived macrophages.
Collapse
Affiliation(s)
- Akshay Rohilla
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alok Kumar Singh
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Benjamin Koleske
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Geetha Srikrishna
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - William R. Bishai
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
24
|
Ishida E, Corrigan DT, Chen T, Liu Y, Kim RS, Song L, Rutledge TM, Magee DM, LaBaer J, Lowary TL, Lin PL, Achkar JM. Mucosal and systemic antigen-specific antibody responses correlate with protection against active tuberculosis in nonhuman primates. EBioMedicine 2024; 99:104897. [PMID: 38096687 PMCID: PMC10758715 DOI: 10.1016/j.ebiom.2023.104897] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 11/15/2023] [Accepted: 11/15/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Increasing evidence supports that antibodies can protect against active tuberculosis (TB) but knowledge of potentially protective antigens, especially in the airways, is limited. The main objective of this study was to identify antigen-specific airway and systemic immunoglobulin isotype responses associated with the outcome of controlled latent Mycobacterium tuberculosis (Mtb) infection (LTBI) versus uncontrolled infection (TB) in nonhuman primates. METHODS In a case-control design, using non-parametric group comparisons with false discovery rate adjustments, we assessed antibodies in 57 cynomolgus macaques which, following low-dose airway Mtb infection, developed either LTBI or TB. We investigated airway and systemic IgG, IgA, and IgM responses in paired bronchoalveolar lavage and plasma samples prior to, two-, and 5-6-months post Mtb infection using an antigen-unbiased approach with Mtb glycan and proteome-wide microarrays. FINDINGS Macaques that developed LTBI (n = 36) had significantly increased airway and plasma IgA reactivities to specific arabinomannan (AM) motifs prior to Mtb infection compared to those that developed TB (n = 21; p < 0.01, q < 0.05). Furthermore, LTBI macaques had higher plasma IgG reactivity to protein MTB32A (Rv0125) early post Mtb infection (p < 0.05) and increasing airway IgG responses to some proteins over time. INTERPRETATION Our results support a protective role of pre-existing mucosal (lung) and systemic IgA to specific Mtb glycan motifs, suggesting that prior exposure to nontuberculous mycobacteria could be protective against TB. They further suggest that IgG to Mtb proteins early post infection could provide an additional protective mechanism. These findings could inform TB vaccine development strategies. FUNDING NIH/NIAID AI117927, AI146329, and AI127173 to JMA.
Collapse
Affiliation(s)
- Elise Ishida
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Devin T Corrigan
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Tingting Chen
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yanyan Liu
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ryung S Kim
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Lusheng Song
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Tara M Rutledge
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - D Mitchell Magee
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Joshua LaBaer
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Todd L Lowary
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada; Institute of Biological Chemistry, Academia Sinica, Nangang Taipei, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Philana Ling Lin
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jacqueline M Achkar
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
25
|
Peng YF. Pulmonary tuberculosis and diabetes mellitus: Epidemiology, pathogenesis and therapeutic management (Review). MEDICINE INTERNATIONAL 2024; 4:4. [PMID: 38204892 PMCID: PMC10777470 DOI: 10.3892/mi.2023.128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 11/22/2023] [Indexed: 01/12/2024]
Abstract
The dual burden of pulmonary tuberculosis (PTB) and diabetes mellitus (DM) is a major global public health concern. There is increasing evidence to indicate an association between PTB and DM. DM is associated with immune dysfunction and altered immune components. Hyperglycemia weakens the innate immune response by affecting the function of macrophages, dendritic cells, neutrophils, and natural killer cells, and also disrupts the adaptive immune response, thus promoting the susceptibility of PTB in patients with DM. Antituberculosis drugs often cause the impairment of liver and kidney function in patients with PTB, and the infection with Mycobacterium tuberculosis weaken pancreatic endocrine function by causing islet cell amyloidosis, which disrupts glucose metabolism and thus increases the risk of developing DM in patients with PTB. The present review discusses the association between PTB and DM from the perspective of epidemiology, pathogenesis, and treatment management. The present review aims to provide information for the rational formulation of treatment strategies for patients with PTB-DM.
Collapse
Affiliation(s)
- You-Fan Peng
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| |
Collapse
|
26
|
Suman SK, Chandrasekaran N, Priya Doss CG. Micro-nanoemulsion and nanoparticle-assisted drug delivery against drug-resistant tuberculosis: recent developments. Clin Microbiol Rev 2023; 36:e0008823. [PMID: 38032192 PMCID: PMC10732062 DOI: 10.1128/cmr.00088-23] [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] [Indexed: 12/01/2023] Open
Abstract
Tuberculosis (TB) is a major global health problem and the second most prevalent infectious killer after COVID-19. It is caused by Mycobacterium tuberculosis (Mtb) and has become increasingly challenging to treat due to drug resistance. The World Health Organization declared TB a global health emergency in 1993. Drug resistance in TB is driven by mutations in the bacterial genome that can be influenced by prolonged drug exposure and poor patient adherence. The development of drug-resistant forms of TB, such as multidrug resistant, extensively drug resistant, and totally drug resistant, poses significant therapeutic challenges. Researchers are exploring new drugs and novel drug delivery systems, such as nanotechnology-based therapies, to combat drug resistance. Nanodrug delivery offers targeted and precise drug delivery, improves treatment efficacy, and reduces adverse effects. Along with nanoscale drug delivery, a new generation of antibiotics with potent therapeutic efficacy, drug repurposing, and new treatment regimens (combinations) that can tackle the problem of drug resistance in a shorter duration could be promising therapies in clinical settings. However, the clinical translation of nanomedicines faces challenges such as safety, large-scale production, regulatory frameworks, and intellectual property issues. In this review, we present the current status, most recent findings, challenges, and limiting barriers to the use of emulsions and nanoparticles against drug-resistant TB.
Collapse
Affiliation(s)
- Simpal Kumar Suman
- School of Bio Sciences & Technology (SBST), Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Natarajan Chandrasekaran
- Centre for Nano Biotechnology (CNBT), Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - C. George Priya Doss
- Laboratory for Integrative Genomics, Department of Integrative Biology, School of Bio Sciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
27
|
Zmyslia M, Fröhlich K, Dao T, Schmidt A, Jessen-Trefzer C. Deep Proteomic Investigation of Metabolic Adaptation in Mycobacteria under Different Growth Conditions. Proteomes 2023; 11:39. [PMID: 38133153 PMCID: PMC10747050 DOI: 10.3390/proteomes11040039] [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: 09/26/2023] [Revised: 11/22/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
Understanding the complex mechanisms of mycobacterial pathophysiology and adaptive responses presents challenges that can hinder drug development. However, employing physiologically relevant conditions, such as those found in human macrophages or simulating physiological growth conditions, holds promise for more effective drug screening. A valuable tool in this pursuit is proteomics, which allows for a comprehensive analysis of adaptive responses. In our study, we focused on Mycobacterium smegmatis, a model organism closely related to the pathogenic Mycobacterium tuberculosis, to investigate the impact of various carbon sources on mycobacterial growth. To facilitate this research, we developed a cost-effective, straightforward, and high-quality pipeline for proteome analysis and compared six different carbon source conditions. Additionally, we have created an online tool to present and analyze our data, making it easily accessible to the community. This user-friendly platform allows researchers and interested parties to explore and interpret the results effectively. Our findings shed light on mycobacterial adaptive physiology and present potential targets for drug development, contributing to the fight against tuberculosis.
Collapse
Affiliation(s)
- Mariia Zmyslia
- Faculty of Chemistry and Pharmacy, University of Freiburg, Albertstrasse 21, 79104 Freiburg, Germany; (M.Z.); (T.D.)
| | - Klemens Fröhlich
- Proteomics Core Facility, Biozentrum Basel, University of Basel, Spitalstrasse 41, 4056 Basel, Switzerland; (K.F.); (A.S.)
| | - Trinh Dao
- Faculty of Chemistry and Pharmacy, University of Freiburg, Albertstrasse 21, 79104 Freiburg, Germany; (M.Z.); (T.D.)
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Albertstrasse 19A, 79104 Freiburg, Germany
- The Center for Integrative Biological Signaling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Alexander Schmidt
- Proteomics Core Facility, Biozentrum Basel, University of Basel, Spitalstrasse 41, 4056 Basel, Switzerland; (K.F.); (A.S.)
| | - Claudia Jessen-Trefzer
- Faculty of Chemistry and Pharmacy, University of Freiburg, Albertstrasse 21, 79104 Freiburg, Germany; (M.Z.); (T.D.)
- The Center for Integrative Biological Signaling Studies, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
28
|
Fu J, Luo X, Lin M, Xiao Z, Huang L, Wang J, Zhu Y, Liu Y, Tao H. Marine-Fungi-Derived Gliotoxin Promotes Autophagy to Suppress Mycobacteria tuberculosis Infection in Macrophage. Mar Drugs 2023; 21:616. [PMID: 38132937 PMCID: PMC10745037 DOI: 10.3390/md21120616] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
The Mycobacterium tuberculosis (MTB) infection causes tuberculosis (TB) and has been a long-standing public-health threat. It is urgent that we discover novel antitubercular agents to manage the increased incidence of multidrug-resistant (MDR) or extensively drug-resistant (XDR) strains of MTB and tackle the adverse effects of the first- and second-line antitubercular drugs. We previously found that gliotoxin (1), 12, 13-dihydroxy-fumitremorgin C (2), and helvolic acid (3) from the cultures of a deep-sea-derived fungus, Aspergillus sp. SCSIO Ind09F01, showed direct anti-TB effects. As macrophages represent the first line of the host defense system against a mycobacteria infection, here we showed that the gliotoxin exerted potent anti-tuberculosis effects in human THP-1-derived macrophages and mouse-macrophage-leukemia cell line RAW 264.7, using CFU assay and laser confocal scanning microscope analysis. Mechanistically, gliotoxin apparently increased the ratio of LC3-II/LC3-I and Atg5 expression, but did not influence macrophage polarization, IL-1β, TNF-a, IL-10 production upon MTB infection, or ROS generation. Further study revealed that 3-MA could suppress gliotoxin-promoted autophagy and restore gliotoxin-inhibited MTB infection, indicating that gliotoxin-inhibited MTB infection can be treated through autophagy in macrophages. Therefore, we propose that marine fungi-derived gliotoxin holds the promise for the development of novel drugs for TB therapy.
Collapse
Affiliation(s)
- Jun Fu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; (J.F.)
| | - Xiaowei Luo
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Miaoping Lin
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Zimin Xiao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; (J.F.)
| | - Lishan Huang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; (J.F.)
| | - Jiaxi Wang
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Yongyan Zhu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; (J.F.)
| | - Yonghong Liu
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Huaming Tao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China; (J.F.)
| |
Collapse
|
29
|
Mwebaza I, Shaw R, Li Q, Fletcher S, Achkar JM, Harding CV, Carpenter SM, Boom WH. Impact of Mycobacterium tuberculosis Glycolipids on the CD4+ T Cell-Macrophage Immunological Synapse. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1385-1396. [PMID: 37695687 PMCID: PMC10579150 DOI: 10.4049/jimmunol.2300107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 08/24/2023] [Indexed: 09/13/2023]
Abstract
Mycobacterium tuberculosis cell-wall glycolipids such as mannosylated lipoarabinomannan (ManLAM) can inhibit murine CD4+ T cells by blocking TCR signaling. This results in suppression of IL-2 production, reduced T cell proliferation, and induction of CD4+ T cell anergy. This study extended these findings to the interaction between primary human CD4+ T cells and macrophages infected by mycobacteria. Exposure of human CD4+ T cells to ManLAM before activation resulted in loss of polyfunctionality, as measured by IL-2, IFN-γ, and TNF-α expression, and reduced CD25 expression. This was not associated with upregulation of inhibitory receptors CTLA-4, PD-1, TIM-3, and Lag-3. By confocal microscopy and imaging flow cytometry, ManLAM exposure reduced conjugate formation between macrophages and CD4+ T cells. ManLAM colocalized to the immunological synapse (IS) and reduced translocation of lymphocyte-specific protein tyrosine kinase (LCK) to the IS. When CD4+ T cells and Mycobacterium bovis BCG-infected monocytes were cocultured, ManLAM colocalized to CD4+ T cells, which formed fewer conjugates with infected monocytes. These results demonstrate that mycobacterial cell-wall glycolipids such as ManLAM can traffic from infected macrophages to disrupt productive IS formation and inhibit CD4+ T cell activation, contributing to immune evasion by M. tuberculosis.
Collapse
Affiliation(s)
- Ivan Mwebaza
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Rachel Shaw
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Qing Li
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Shane Fletcher
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH
| | | | - Clifford V. Harding
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH
- Department of Pathology, Case Western Reserve University, Cleveland, OH
| | - Stephen M. Carpenter
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH
- Department of Pathology, Case Western Reserve University, Cleveland, OH
| | - W. Henry Boom
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH
- Department of Pathology, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
30
|
Pan J, Chang Z, Zhang X, Dong Q, Zhao H, Shi J, Wang G. Research progress of single-cell sequencing in tuberculosis. Front Immunol 2023; 14:1276194. [PMID: 37901241 PMCID: PMC10611525 DOI: 10.3389/fimmu.2023.1276194] [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: 08/14/2023] [Accepted: 09/29/2023] [Indexed: 10/31/2023] Open
Abstract
Tuberculosis is a major infectious disease caused by Mycobacterium tuberculosis infection. The pathogenesis and immune mechanism of tuberculosis are not clear, and it is urgent to find new drugs, diagnosis, and treatment targets. A useful tool in the quest to reveal the enigmas related to Mycobacterium tuberculosis infection and disease is the single-cell sequencing technique. By clarifying cell heterogeneity, identifying pathogenic cell groups, and finding key gene targets, the map at the single cell level enables people to better understand the cell diversity of complex organisms and the immune state of hosts during infection. Here, we briefly reviewed the development of single-cell sequencing, and emphasized the different applications and limitations of various technologies. Single-cell sequencing has been widely used in the study of the pathogenesis and immune response of tuberculosis. We review these works summarizing the most influential findings. Combined with the multi-molecular level and multi-dimensional analysis, we aim to deeply understand the blank and potential future development of the research on Mycobacterium tuberculosis infection using single-cell sequencing technology.
Collapse
Affiliation(s)
| | | | | | | | | | - Jingwei Shi
- Key Laboratory of Pathobiology Ministry of Education, College of Basic Medical Sciences/China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| | - Guoqing Wang
- Key Laboratory of Pathobiology Ministry of Education, College of Basic Medical Sciences/China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
31
|
Yang J, Zhang L, Qiao W, Luo Y. Mycobacterium tuberculosis: Pathogenesis and therapeutic targets. MedComm (Beijing) 2023; 4:e353. [PMID: 37674971 PMCID: PMC10477518 DOI: 10.1002/mco2.353] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 09/08/2023] Open
Abstract
Tuberculosis (TB) remains a significant public health concern in the 21st century, especially due to drug resistance, coinfection with diseases like immunodeficiency syndrome (AIDS) and coronavirus disease 2019, and the lengthy and costly treatment protocols. In this review, we summarize the pathogenesis of TB infection, therapeutic targets, and corresponding modulators, including first-line medications, current clinical trial drugs and molecules in preclinical assessment. Understanding the mechanisms of Mycobacterium tuberculosis (Mtb) infection and important biological targets can lead to innovative treatments. While most antitubercular agents target pathogen-related processes, host-directed therapy (HDT) modalities addressing immune defense, survival mechanisms, and immunopathology also hold promise. Mtb's adaptation to the human host involves manipulating host cellular mechanisms, and HDT aims to disrupt this manipulation to enhance treatment effectiveness. Our review provides valuable insights for future anti-TB drug development efforts.
Collapse
Affiliation(s)
- Jiaxing Yang
- Center of Infectious Diseases and State Key Laboratory of Biotherapy, West China HospitalSichuan UniversityChengduChina
| | - Laiying Zhang
- Center of Infectious Diseases and State Key Laboratory of Biotherapy, West China HospitalSichuan UniversityChengduChina
| | - Wenliang Qiao
- Department of Thoracic Surgery, West China HospitalSichuan UniversityChengduSichuanChina
- Lung Cancer Center, West China HospitalSichuan UniversityChengduSichuanChina
| | - Youfu Luo
- Center of Infectious Diseases and State Key Laboratory of Biotherapy, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
32
|
Saini S, Gangwar A, Sharma R. Harnessing host-pathogen interactions for innovative drug discovery and host-directed therapeutics to tackle tuberculosis. Microbiol Res 2023; 275:127466. [PMID: 37531813 DOI: 10.1016/j.micres.2023.127466] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/04/2023]
Abstract
Tuberculosis (TB) is a highly contagious bacterial infection caused by Mycobacterium tuberculosis (Mtb), which has been ranked as the second leading cause of death worldwide from a single infectious agent. As an intracellular pathogen, Mtb has well adapted to the phagocytic host microenvironment, influencing diverse host processes such as gene expression, trafficking, metabolism, and signaling pathways of the host to its advantage. These responses are the result of dynamic interactions of the bacteria with the host cell signaling pathways, whereby the bacteria attenuate the host cellular processes for their survival. Specific host genes and the mechanisms involved in the entry and subsequent stabilization of M. tuberculosis intracellularly have been identified in various genetic and chemical screens recently. The present understanding of the co-evolution of Mtb and macrophage system presented us the new possibilities for exploring host-directed therapeutics (HDT). Here, we discuss the host-pathogen interaction for Mtb, including the pathways adapted by Mtb to escape immunity. The review sheds light on different host-directed therapies (HDTs) such as repurposed drugs and vitamins, along with their targets such as granuloma, autophagy, extracellular matrix, lipids, and cytokines, among others. The article also examines the available clinical data on these drug molecules. In conclusion, the review presents a perspective on the current knowledge in the field of HDTs and the need for additional research to overcome the challenges associated HDTs.
Collapse
Affiliation(s)
- Sapna Saini
- Infectious Diseases Division, CSIR, Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Anjali Gangwar
- Infectious Diseases Division, CSIR, Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rashmi Sharma
- Infectious Diseases Division, CSIR, Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
33
|
Diatlova A, Linkova N, Lavrova A, Zinchenko Y, Medvedev D, Krasichkov A, Polyakova V, Yablonskiy P. Molecular Markers of Early Immune Response in Tuberculosis: Prospects of Application in Predictive Medicine. Int J Mol Sci 2023; 24:13261. [PMID: 37686061 PMCID: PMC10487556 DOI: 10.3390/ijms241713261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Tuberculosis (TB) remains an important public health problem and one of the leading causes of death. Individuals with latent tuberculosis infection (LTBI) have an increased risk of developing active TB. The problem of the diagnosis of the various stages of TB and the identification of infected patients in the early stages has not yet been solved. The existing tests (the tuberculin skin test and the interferon-gamma release assay) are useful to distinguish between active and latent infections. But these tests cannot be used to predict the development of active TB in individuals with LTBI. The purpose of this review was to analyze the extant data of the interaction of M. tuberculosis with immune cells and identify molecular predictive markers and markers of the early stages of TB. An analysis of more than 90 sources from the literature allowed us to determine various subpopulations of immune cells involved in the pathogenesis of TB, namely, macrophages, dendritic cells, B lymphocytes, T helper cells, cytotoxic T lymphocytes, and NK cells. The key molecular markers of the immune response to M. tuberculosis are cytokines (IL-1β, IL-6, IL-8, IL-10, IL-12, IL-17, IL-22b, IFNɣ, TNFa, and TGFß), matrix metalloproteinases (MMP-1, MMP-3, and MMP-9), and their inhibitors (TIMP-1, TIMP-2, TIMP-3, and TIMP-4). It is supposed that these molecules could be used as biomarkers to characterize different stages of TB infection, to evaluate the effectiveness of its treatment, and as targets of pharmacotherapy.
Collapse
Affiliation(s)
- Anastasiia Diatlova
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2–4, 191036 St. Petersburg, Russia
| | - Natalia Linkova
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2–4, 191036 St. Petersburg, Russia
- Biogerontology Department, St. Petersburg Institute of Bioregulation and Gerontology, Dynamo pr., 3, 197110 St. Petersburg, Russia
| | - Anastasia Lavrova
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2–4, 191036 St. Petersburg, Russia
- Department of Hospital Surgery, Faculty of Medicine, St. Petersburg State University, University Embankment, 7–9, 199034 St. Petersburg, Russia
| | - Yulia Zinchenko
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2–4, 191036 St. Petersburg, Russia
| | - Dmitrii Medvedev
- Biogerontology Department, St. Petersburg Institute of Bioregulation and Gerontology, Dynamo pr., 3, 197110 St. Petersburg, Russia
| | - Alexandr Krasichkov
- Department of Radio Engineering Systems, Electrotechnical University “LETI”, Prof. Popova Street 5F, 197022 St. Petersburg, Russia
| | - Victoria Polyakova
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2–4, 191036 St. Petersburg, Russia
| | - Piotr Yablonskiy
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2–4, 191036 St. Petersburg, Russia
- Department of Hospital Surgery, Faculty of Medicine, St. Petersburg State University, University Embankment, 7–9, 199034 St. Petersburg, Russia
| |
Collapse
|
34
|
Chen L, Hua J, He X. Identification of cuproptosis-related molecular subtypes as a biomarker for differentiating active from latent tuberculosis in children. BMC Genomics 2023; 24:368. [PMID: 37393262 DOI: 10.1186/s12864-023-09491-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 06/28/2023] [Indexed: 07/03/2023] Open
Abstract
BACKGROUND Cell death plays a crucial role in the progression of active tuberculosis (ATB) from latent infection (LTBI). Cuproptosis, a novel programmed cell death, has been reported to be associated with the pathology of various diseases. We aimed to identify cuproptosis-related molecular subtypes as biomarkers for distinguishing ATB from LTBI in pediatric patients. METHOD The expression profiles of cuproptosis regulators and immune characteristics in pediatric patients with ATB and LTBI were analyzed based on GSE39939 downloaded from the Gene Expression Omnibus. From the 52 ATB samples, we investigated the molecular subtypes based on differentially expressed cuproptosis-related genes (DE-CRGs) via consensus clustering and related immune cell infiltration. Subtype-specific differentially expressed genes (DEGs) were found using the weighted gene co-expression network analysis. The optimum machine model was then determined by comparing the performance of the eXtreme Gradient Boost (XGB), the random forest model (RF), the general linear model (GLM), and the support vector machine model (SVM). Nomogram and test datasets (GSE39940) were used to verify the prediction accuracy. RESULTS Nine DE-CRGs (NFE2L2, NLRP3, FDX1, LIPT1, PDHB, MTF1, GLS, DBT, and DLST) associated with active immune responses were ascertained between ATB and LTBI patients. Two cuproptosis-related molecular subtypes were defined in ATB pediatrics. Single sample gene set enrichment analysis suggested that compared with Subtype 2, Subtype 1 was characterized by decreased lymphocytes and increased inflammatory activation. Gene set variation analysis showed that cluster-specific DEGs in Subtype 1 were closely associated with immune and inflammation responses and energy and amino acids metabolism. The SVM model exhibited the best discriminative performance with a higher area under the curve (AUC = 0.983) and relatively lower root mean square and residual error. A final 5-gene-based (MAN1C1, DKFZP434N035, SIRT4, BPGM, and APBA2) SVM model was created, demonstrating satisfactory performance in the test datasets (AUC = 0.905). The decision curve analysis and nomogram calibration curve also revealed the accuracy of differentiating ATB from LTBI in children. CONCLUSION Our study suggested that cuproptosis might be associated with the immunopathology of Mycobacterium tuberculosis infection in children. Additionally, we built a satisfactory prediction model to assess the cuproptosis subtype risk in ATB, which can be used as a reliable biomarker for the distinguishment between pediatric ATB and LTBI.
Collapse
Affiliation(s)
- Liang Chen
- Department of Infectious Diseases, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, No.86, Chongwen Street, Lishui District, Nanjing City, 211002, China.
| | - Jie Hua
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaopu He
- Department of Geriatric Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
35
|
Adefisayo OO, Curtis ER, Smith CM. Mycobacterial Genetic Technologies for Probing the Host-Pathogen Microenvironment. Infect Immun 2023; 91:e0043022. [PMID: 37249448 PMCID: PMC10269127 DOI: 10.1128/iai.00430-22] [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] [Indexed: 05/31/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, is one of the oldest and most successful pathogens in the world. Diverse selective pressures encountered within host cells have directed the evolution of unique phenotypic traits, resulting in the remarkable evolutionary success of this largely obligate pathogen. Despite centuries of study, the genetic repertoire utilized by Mtb to drive virulence and host immune evasion remains to be fully understood. Various genetic approaches have been and continue to be developed to tackle the challenges of functional gene annotation and validation in an intractable organism such as Mtb. In vitro and ex vivo systems remain the primary approaches to generate and confirm hypotheses that drive a general understanding of mycobacteria biology. However, it remains of great importance to characterize genetic requirements for successful infection within a host system as in vitro and ex vivo studies fail to fully replicate the complex microenvironment experienced by Mtb. In this review, we evaluate the employment of the mycobacterial genetic toolkit to probe the host-pathogen interface by surveying the current state of mycobacterial genetic studies within host systems, with a major focus on the murine model. Specifically, we discuss the different ways that these tools have been utilized to examine various aspects of infection, including bacterial survival/virulence, bacterial evasion of host immunity, and development of novel antibacterial/vaccine strategies.
Collapse
Affiliation(s)
| | - Erin R. Curtis
- Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Clare M. Smith
- Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
36
|
Du X, Sheng J, Chen Y, He S, Yang Y, Huang Y, Fu Y, Lie L, Han Z, Zhu B, Liu H, Wen Q, Zhou X, Zhou C, Hu S, Ma L. The E3 ligase HERC5 promotes antimycobacterial responses in macrophages by ISGylating the phosphatase PTEN. Sci Signal 2023; 16:eabm1756. [PMID: 37279284 DOI: 10.1126/scisignal.abm1756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 05/16/2023] [Indexed: 06/08/2023]
Abstract
Innate immune signaling in macrophages during viral infection is regulated by ISGylation, the covalent attachment of the ubiquitin-like protein interferon-stimulated gene 15 (ISG15) to protein targets. Here, we explored the role of ISGylation in the macrophage response to infection with Mycobacterium tuberculosis. In human and mouse macrophages, the E3 ubiquitin ligases HERC5 and mHERC6, respectively, mediated the ISGylation of the phosphatase PTEN, which promoted its degradation. The decreased abundance of PTEN led to an increase in the activity of the PI3K-AKT signaling pathway, which stimulated the synthesis of proinflammatory cytokines. Bacterial growth was increased in culture and in vivo when human or mouse macrophages were deficient in the major E3 ISG15 ligase. The findings expand the role of ISGylation in macrophages to antibacterial immunity and suggest that HERC5 signaling may be a candidate target for adjunct host-directed therapy in patients with tuberculosis.
Collapse
Affiliation(s)
- Xialin Du
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Junli Sheng
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Yitian Chen
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Shitong He
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Yalong Yang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Yulan Huang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Yuling Fu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Linmiao Lie
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Zhenyu Han
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Bo Zhu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Honglin Liu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Qian Wen
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Xinying Zhou
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Chaoying Zhou
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Shengfeng Hu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Li Ma
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
37
|
Yang L, Chaves L, Kutscher HL, Karki S, Tamblin M, Kenney P, Reynolds JL. An immunoregulator nanomedicine approach for the treatment of tuberculosis. Front Bioeng Biotechnol 2023; 11:1095926. [PMID: 37304141 PMCID: PMC10249870 DOI: 10.3389/fbioe.2023.1095926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 05/12/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction: A nanoparticle composed of a poly (lactic-co-glycolic acid) (PLGA) core and a chitosan (CS) shell with surface-adsorbed 1,3 β-glucan (β-glucan) was synthesized. The exposure response of CS-PLGA nanoparticles (0.1 mg/mL) with surface-bound β-glucan at 0, 5, 10, 15, 20, or 25 ng or free β-glucan at 5, 10, 15, 20, or 25 ng/mL in macrophage in vitro and in vivo was investigated. Results: In vitro studies demonstrate that gene expression for IL-1β, IL-6, and TNFα increased at 10 and 15 ng surface-bound β-glucan on CS-PLGA nanoparticles (0.1 mg/mL) and at 20 and 25 ng/mL of free β-glucan both at 24 h and 48 h. Secretion of TNFα protein and ROS production increased at 5, 10, 15, and 20 ng surface-bound β-glucan on CS-PLGA nanoparticles and at 20 and 25 ng/mL of free β-glucan at 24 h. Laminarin, a Dectin-1 antagonist, prevented the increase in cytokine gene expression induced by CS-PLGA nanoparticles with surface-bound β-glucan at 10 and 15 ng, indicating a Dectin-1 receptor mechanism. Efficacy studies showed a significant reduction in intracellular accumulation of mycobacterium tuberculosis (Mtb) in monocyte-derived macrophages (MDM) incubated with on CS-PLGA (0.1 mg/ml) nanoparticles with 5, 10, and 15 ng surface-bound β-glucan or with 10 and 15 ng/mL of free β-glucan. β-glucan-CS-PLGA nanoparticles inhibited intracellular Mtb growth more than free β-glucan alone supporting the role of β-glucan-CS-PLGA nanoparticles as stronger adjuvants than free β-glucan. In vivo studies demonstrate that oropharyngeal aspiration (OPA) of CS-PLGA nanoparticles with nanogram concentrations of surface-bound β-glucan or free β-glucan increased TNFα gene expression in alveolar macrophages and TNFα protein secretion in bronchoalveolar lavage supernatants. Discussion: Data also demonstrate no damage to the alveolar epithelium or changes in the murine sepsis score following exposure to β-glucan-CS-PLGA nanoparticles only, indicating safety and feasibility of this nanoparticle adjuvant platform to mice by OPA.
Collapse
Affiliation(s)
- Luona Yang
- Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Lee Chaves
- Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Hilliard L. Kutscher
- Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Shanta Karki
- Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Maria Tamblin
- Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Patrick Kenney
- Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Jessica L. Reynolds
- Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
38
|
Bhaskar A, Pahuja I, Negi K, Verma A, Ghoshal A, Mathew B, Tripathi G, Maras JS, Chaturvedi S, Dwivedi VP. SIRT2 inhibition by AGK2 enhances mycobacteria-specific stem cell memory responses by modulating beta-catenin and glycolysis. iScience 2023; 26:106644. [PMID: 37192966 PMCID: PMC10182326 DOI: 10.1016/j.isci.2023.106644] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/27/2023] [Accepted: 04/06/2023] [Indexed: 05/18/2023] Open
Abstract
Bacille Calmette-Guerin (BCG) generates limited long-lasting adaptive memory responses leading to short-lived protection against adult pulmonary tuberculosis (TB). Here, we show that host sirtuin 2 (SIRT2) inhibition by AGK2 significantly enhances the BCG vaccine efficacy during primary infection and TB recurrence through enhanced stem cell memory (TSCM) responses. SIRT2 inhibition modulated the proteome landscape of CD4+ T cells affecting pathways involved in cellular metabolism and T-cell differentiation. Precisely, AGK2 treatment enriched the IFNγ-producing TSCM cells by activating β-catenin and glycolysis. Furthermore, SIRT2 specifically targeted histone H3 and NF-κB p65 to induce proinflammatory responses. Finally, inhibition of the Wnt/β-catenin pathway abolished the protective effects of AGK2 treatment during BCG vaccination. Taken together, this study provides a direct link between BCG vaccination, epigenetics, and memory immune responses. We identify SIRT2 as a key regulator of memory T cells during BCG vaccination and project SIRT2 inhibitors as potential immunoprophylaxis against TB.
Collapse
Affiliation(s)
- Ashima Bhaskar
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
- Corresponding author
| | - Isha Pahuja
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
- Department of Molecular Medicine, Jamia Hamdard University, New Delhi, India
| | - Kriti Negi
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Akanksha Verma
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Antara Ghoshal
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Babu Mathew
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Gaurav Tripathi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Jaswinder Singh Maras
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Shivam Chaturvedi
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Ved Prakash Dwivedi
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| |
Collapse
|
39
|
Chen L, Hua J, Dai X, He X. Assessment of ferroptosis-associated gene signatures as potential biomarkers for differentiating latent from active tuberculosis in children. Microb Genom 2023; 9. [PMID: 37163321 DOI: 10.1099/mgen.0.000997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
Ferroptotic cell death is a regulated process that is governed by iron-dependent membrane lipid peroxide accumulation that plays a pathogenic role in several disease-related settings. The use of ferroptosis-related genes (FRGs) to distinguish active tuberculosis (ATB) from latent tuberculosis infection (LTBI) among children, however, remains to be analysed. Tuberculosis-related gene expression data and FRG lists were obtained, respectively, from Gene Expression Omnibus (GEO) and FerrDb. Differentially expressed FRGs (DE-FRGs) detected when comparing samples from paediatric ATB and LTBI patients were explored using appropriate bioinformatics techniques, after which enrichment analyses were performed for these genes and hub genes were identified, with these genes then being used to explore potential drug interactions and construct competing endogenous RNA (ceRNA) networks. The GSE39939 dataset yielded 124 DE-FRGs that were primarily related to responses to oxidative, chemical and extracellular stimulus-associated stress. In total, the LASSO and SVM-RFE algorithms enabled the identification of nine hub genes (MAPK14, EGLN2, IDO1, USP11, SCD, CBS, PARP8, PARP16, CDC25A) that exhibited good diagnostic utility. Functional enrichment analyses of these genes suggested that they may govern ATB transition from LTBI through the control of many pathways, including the immune response, DNA repair, transcription, RNA degradation, and glycan and energy metabolism pathways. The CIBERSORT algorithm suggested that these genes were positively correlated with inflammatory and myeloid cell activity while being negatively correlated with the activity of lymphocytes. A total of 50 candidate drugs targeting 6 hub DE-FRGs were also identified, and a ceRNA network was used to explore the complex interplay among these hub genes. The nine hub FRGs defined in this study may serve as valuable biomarkers differentiating between ATB and LTBI in young patients.
Collapse
Affiliation(s)
- Liang Chen
- Department of Infectious Diseases, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, PR China
| | - Jie Hua
- Department of Gastroenterology, Liyang People's Hospital, Liyang Branch Hospital of Jiangsu Province Hospital, Nanjing, PR China
| | - Xiaoting Dai
- Department of Infectious Diseases, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, PR China
| | - Xiaopu He
- Department of Geriatric Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| |
Collapse
|
40
|
Jiang F, Peng C, Cheng P, Wang J, Lian J, Gong W. PP19128R, a Multiepitope Vaccine Designed to Prevent Latent Tuberculosis Infection, Induced Immune Responses In Silico and In Vitro Assays. Vaccines (Basel) 2023; 11:vaccines11040856. [PMID: 37112768 PMCID: PMC10145841 DOI: 10.3390/vaccines11040856] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/13/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Background: Latent tuberculosis infection (LTBI) is the primary source of active tuberculosis (ATB), but a preventive vaccine against LTBI is lacking. Methods: In this study, dominant helper T lymphocyte (HTL), cytotoxic T lymphocyte (CTL), and B-cell epitopes were identified from nine antigens related to LTBI and regions of difference (RDs). These epitopes were used to construct a novel multiepitope vaccine (MEV) based on their antigenicity, immunogenicity, sensitization, and toxicity. The immunological characteristics of the MEV were analyzed with immunoinformatics technology and verified by enzyme-linked immunospot assay and Th1/Th2/Th17 cytokine assay in vitro. Results: A novel MEV, designated PP19128R, containing 19 HTL epitopes, 12 CTL epitopes, 8 B-cell epitopes, toll-like receptor (TLR) agonists, and helper peptides, was successfully constructed. Bioinformatics analysis showed that the antigenicity, immunogenicity, and solubility of PP19128R were 0.8067, 9.29811, and 0.900675, respectively. The global population coverage of PP19128R in HLA class I and II alleles reached 82.24% and 93.71%, respectively. The binding energies of the PP19128R-TLR2 and PP19128R-TLR4 complexes were -1324.77 kcal/mol and -1278 kcal/mol, respectively. In vitro experiments showed that the PP19128R vaccine significantly increased the number of interferon gamma-positive (IFN-γ+) T lymphocytes and the levels of cytokines, such as IFN-γ, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and IL-10. Furthermore, positive correlations were observed between PP19128R-specific cytokines in ATB patients and individuals with LTBI. Conclusions: The PP19128R vaccine is a promising MEV with excellent antigenicity and immunogenicity and no toxicity or sensitization that can induce robust immune responses in silico and in vitro. This study provides a vaccine candidate for the prevention of LTBI in the future.
Collapse
Affiliation(s)
- Fan Jiang
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing 100091, China
- The Second Brigade of Cadet, Basic Medical Science Academy of Air Force Medical University, Xi'an 710032, China
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an 710032, China
| | - Cong Peng
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing 100091, China
| | - Peng Cheng
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing 100091, China
| | - Jie Wang
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing 100091, China
| | - Jianqi Lian
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an 710032, China
| | - Wenping Gong
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing 100091, China
| |
Collapse
|
41
|
Panda S, Morgan J, Cheng C, Saito M, Gilman RH, Ciobanu N, Crudu V, Catanzaro DG, Catanzaro A, Rodwell T, Perera JS, Chathuranga T, Gunasena B, DeSilva AD, Peters B, Sette A, Lindestam Arlehamn CS. Identification of differentially recognized T cell epitopes in the spectrum of Mtb infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.12.536550. [PMID: 37090558 PMCID: PMC10120689 DOI: 10.1101/2023.04.12.536550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Tuberculosis caused by Mycobacterium tuberculosis is one of the leading causes of death from a single infectious agent. Identifying dominant epitopes and comparing their reactivity in different tuberculosis (TB) infection states can help design diagnostics and vaccines. We performed a proteome-wide screen of 20,610 Mtb derived peptides in 21 Active TB (ATB) patients 3-4 months post-diagnosis of pulmonary TB (mid-treatment) using an IFNγ and IL-17 Fluorospot assay. Responses were mediated exclusively by IFNγ and identified a total of 137 unique epitopes, with each patient recognizing, on average, 8 individual epitopes and 22 epitopes (16%) recognized by 2 or more participants. Responses were predominantly directed against antigens part of the cell wall and cell processes category. Testing 517 peptides spanning TB vaccine candidates and ESAT-6 and CFP10 antigens also revealed differential recognition between ATB participants mid-treatment and healthy IGRA+ participants of several vaccine antigens. An ATB-specific peptide pool consisting of epitopes exclusively recognized by participants mid-treatment, allowed distinguishing participants with active pulmonary TB from healthy interferon-gamma release assay (IGRA)+/- participants from diverse geographical locations. Analysis of longitudinal samples indicated decreased reactivity during treatment for pulmonary TB. Together, these results show that a proteome-wide screen of T cell reactivity identifies epitopes and antigens that are differentially recognized depending on the Mtb infection stage. These have potential use in developing diagnostics and vaccine candidates and measuring correlates of protection.
Collapse
Affiliation(s)
- Sudhasini Panda
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Jeffrey Morgan
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Catherine Cheng
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Mayuko Saito
- Department of Virology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Robert H. Gilman
- Johns Hopkins School of Public Health, Baltimore, MD, USA
- Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Nelly Ciobanu
- Phthisiopneumology Institute, Chisinau, Republic of Moldova
| | - Valeriu Crudu
- Phthisiopneumology Institute, Chisinau, Republic of Moldova
| | - Donald G Catanzaro
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR, USA
| | - Antonino Catanzaro
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Timothy Rodwell
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Judy S.B. Perera
- Faculty of Medicine, General Sir John Kotelawala Defense University, Ratmalana, Sri Lanka
| | - Teshan Chathuranga
- Faculty of Medicine, General Sir John Kotelawala Defense University, Ratmalana, Sri Lanka
| | - Bandu Gunasena
- National Hospital for Respiratory Diseases, Welisara, Sri Lanka
| | - Aruna D. DeSilva
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Faculty of Medicine, General Sir John Kotelawala Defense University, Ratmalana, Sri Lanka
| | - Bjoern Peters
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | | |
Collapse
|
42
|
Murdaca G, Paladin F, Mangini G, Tiso D, Gangemi S. TBC and COVID: an interplay between two infections. Expert Opin Drug Saf 2023; 22:303-311. [PMID: 37079022 DOI: 10.1080/14740338.2023.2205638] [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: 02/28/2023] [Accepted: 04/18/2023] [Indexed: 04/21/2023]
Abstract
INTRODUCTION In a historical era dominated by the SARS-CoV-2 pandemic, a fact of growing interest emerges regarding co-infection with Mycobacterium tuberculosis (M. tuberculosis). This represents today an important clinical and diagnostic challenge, as the two pathogens are capable, through specific immunopathological mechanisms, of interacting with each other, determining a severe respiratory condition with a severe prognosis. AREAS COVERED With this review, we wanted to collect and analyze the latest scientific evidence concerning the main immunopathogenetic mechanisms shared by these two respiratory pathogens, with particular interest in the possible iatrogenic factors favoring coinfection and the need to define multidisciplinary and standardized screening tools aimed to identify coinfection early, ensuring the best clinical and therapeutic management. EXPERT OPINION The existence of a direct immunopathogenetic link between COVID-19 and TB indirectly contributes to mutual morbidity and mortality. The identification and application of early and standardized screening tools aimed at the identification of this condition is essential, in addition to vaccine prevention.
Collapse
Affiliation(s)
- Giuseppe Murdaca
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- Department of Internal Medicine (DIMI), Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Francesca Paladin
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- Department of Internal Medicine (DIMI), Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Gloria Mangini
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- Department of Internal Medicine (DIMI), Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Debora Tiso
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- Department of Internal Medicine (DIMI), Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| |
Collapse
|
43
|
Liao KM, Lee CS, Wu YC, Shu CC, Ho CH. Prior treated tuberculosis and mortality risk in lung cancer. Front Med (Lausanne) 2023; 10:1121257. [PMID: 37064038 PMCID: PMC10090669 DOI: 10.3389/fmed.2023.1121257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 03/03/2023] [Indexed: 03/31/2023] Open
Abstract
BackgroundLung cancer is one of the leading causes of cancer death worldwide, and tuberculosis (TB) is a common pre-existing disease. However, there is scarce literature studying the mortality risk in patients with prior TB and subsequent lung cancer.MethodsWe recruited lung cancer patients from the Taiwan Cancer Registry from 2011 to 2015 and classified them into two groups according to presence or absence of prior TB. We then matched them in a ratio of 1:4 using the exact matching approach. The mortality risk within 3 years after diagnosis of lung cancer was analyzed and compared between these two groups.ResultsDuring the study period, 43,472 patients with lung cancer were recruited, and of these, 1,211 (2.79%) patients had prior TB. After matching, this cohort included 5,935 patients with lung cancer in two groups: patients with prior TB before lung cancer (n = 1,187) and those without (n = 4,748). After controlling for demographic factors and comorbidities, the patients with prior TB had increased adjusted hazard ratios of 1.13 (95% CI: 1.04–1.23) and 1.11 (1.02–1.21) for all-cause and cancer-specific 3-year mortality, respectively, compared to the lung cancer patients without prior TB. Duration between TB and lung cancer (<1 year vs. 1–3 years vs. >3 years) had no differences for mortality risk.ConclusionIn the present study, 2.79% patients with lung cancer had prior TB, which was associated with higher 3-year mortality after they developed lung cancer. The mortality risk with prior TB did not decrease even if >3 years passed before diagnosis of lung cancer.
Collapse
Affiliation(s)
- Kuang-Ming Liao
- Department of Internal Medicine, Chi Mei Medical Center, Chiali, Taiwan
| | - Chung-Shu Lee
- Department of Pulmonary and Critical Care Medicine, New Taipei Municipal Tu Cheng Hospital, New Taipei City, Taiwan
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, School of Medicine, Chang Gung University, Taipei, Taiwan
| | - Yu-Cih Wu
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Chin-Chung Shu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- *Correspondence: Chin-Chung Shu,
| | - Chung-Han Ho
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
- Department of Information Management, Southern Taiwan University of Science and Technology, Tainan, Taiwan
- Cancer Center, Taipei Municipal Wanfang Hospital, Taipei Medical University, Taipei, Taiwan
- Chung-Han Ho,
| |
Collapse
|
44
|
Kathamuthu GR, Moideen K, Sridhar R, Baskaran D, Babu S. Systemic Levels of Pro-Inflammatory Cytokines and Post-Treatment Modulation in Tuberculous Lymphadenitis. Trop Med Infect Dis 2023; 8:tropicalmed8030150. [PMID: 36977151 PMCID: PMC10053505 DOI: 10.3390/tropicalmed8030150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/13/2023] [Accepted: 02/21/2023] [Indexed: 03/08/2023] Open
Abstract
Pro-inflammatory cytokines are potent stimulators of inflammation and immunity and markers of infection severity and bacteriological burden in pulmonary tuberculosis (PTB). Interferons could have both host-protective and detrimental effects on tuberculosis disease. However, their role has not been studied in tuberculous lymphadenitis (TBL). Thus, we evaluated the systemic pro-inflammatory (interleukin (IL)-12, IL-23, interferon (IFN)α, and IFNβ) cytokine levels in TBL, latent tuberculosis (LTBI), and healthy control (HC) individuals. In addition, we also measured the baseline (BL) and post-treatment (PT) systemic levels in TBL individuals. We demonstrate that TBL individuals are characterized by increased pro-inflammatory (IL-12, IL-23, IFNα, IFNβ) cytokines when compared to LTBI and HC individuals. We also show that after anti-tuberculosis treatment (ATT) completion, the systemic levels of pro-inflammatory cytokines were significantly modulated in TBL individuals. A receiver operating characteristic (ROC) analysis revealed IL-23, IFNα, and IFNβ significantly discriminated TBL disease from LTBI and/or HC individuals. Hence, our study demonstrates the altered systemic levels of pro-inflammatory cytokines and their reversal after ATT, suggesting that they are markers of disease pathogenesis/severity and altered immune regulation in TBL disease.
Collapse
Affiliation(s)
- Gokul Raj Kathamuthu
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai 600 031, India
- National Institute for Research in Tuberculosis (NIRT), Chennai 600 031, India
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77 Solna, Sweden
- Correspondence:
| | - Kadar Moideen
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai 600 031, India
| | | | - Dhanaraj Baskaran
- National Institute for Research in Tuberculosis (NIRT), Chennai 600 031, India
| | - Subash Babu
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai 600 031, India
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-0425, USA
| |
Collapse
|
45
|
Harnessing Innate Immunity to Treat Mycobacterium tuberculosis Infections: Heat-Killed Caulobacter crescentus as a Novel Biotherapeutic. Cells 2023; 12:cells12040560. [PMID: 36831226 PMCID: PMC9954702 DOI: 10.3390/cells12040560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/29/2023] [Accepted: 02/02/2023] [Indexed: 02/12/2023] Open
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis (Mtb), is a serious and devastating infectious disease worldwide. Approximately a quarter of the world population harbors latent Mtb infection without pathological consequences. Exposure of immunocompetent healthy individuals with Mtb does not result in active disease in more than 90% individuals, suggesting a defining role of host immunity to prevent and/or clear early infection. However, innate immune stimulation strategies have been relatively underexplored for the treatment of tuberculosis. In this study, we used cell culture and mouse models to examine the role of a heat-killed form of a non-pathogenic microbe, Caulobacter crescentus (HKCC), in inducing innate immunity and limiting Mtb infection. We also examined the added benefits of a distinct chemo-immunotherapeutic strategy that incorporates concurrent treatments with low doses of a first-line drug isoniazid and HKCC. This therapeutic approach resulted in highly significant reductions in disseminated Mtb in the lungs, liver, and spleen of mice compared to either agent alone. Our studies demonstrate the potential of a novel innate immunotherapeutic strategy with or without antimycobacterial drugs in controlling Mtb infection in mice and open new avenues for the treatment of tuberculosis in humans.
Collapse
|
46
|
Kumar NP, Babu S. Impact of diabetes mellitus on immunity to latent tuberculosis infection. FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2023; 4:1095467. [PMID: 36993821 PMCID: PMC10012073 DOI: 10.3389/fcdhc.2023.1095467] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/16/2023] [Indexed: 01/27/2023]
Abstract
Tuberculosis (TB) is an infectious disease that poses a major health threat and is one of the leading causes of death worldwide. Following exposure to Mycobacterium tuberculosis (M.tb) bacilli, hosts who fail to clear M.tb end up in a state of latent tuberculosis infection (LTBI), in which the bacteria are contained but not eliminated. Type 2 diabetes mellitus (DM) is a noncommunicable disease that can weaken host immunity and lead to increased susceptibility to various infectious diseases. Despite numerous studies on the relationship between DM and active TB, data on the association between DM and LTBI remains limited. Immunological data suggest that LTBI in the presence of DM leads to an impaired production of protective cytokines and poly-functional T cell responses, accounting for a potential immunological mechanism that could leads to an increased risk of active TB. This review highlights the salient features of the immunological underpinnings influencing the interaction between TB and DM in humans.
Collapse
Affiliation(s)
- Nathella Pavan Kumar
- ICMR-National Institute for Research in Tuberculosis, Chennai, India
- *Correspondence: Nathella Pavan Kumar, ,
| | - Subash Babu
- International Centre for Excellence in Research, National Institutes of Health, Chennai, India
- Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
47
|
Pathogenesis, Diagnostic Challenges, and Risk Factors of Pott's Disease. Clin Pract 2023; 13:155-165. [PMID: 36826156 PMCID: PMC9955044 DOI: 10.3390/clinpract13010014] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/14/2023] [Accepted: 01/17/2023] [Indexed: 01/27/2023] Open
Abstract
Tuberculosis (TB) prevalence is increasing in developed nations and continuing to cause significant mortality in low- and middle-income countries. As a result of the uptick in cases, there also exists an increased prevalence of extrapulmonary TB. TB is caused by Mycobacterium tuberculosis (M. tb). When M. tb disseminates to the vertebral column, it is called Pott's disease or spinal TB. The frequency, symptoms, and severity of the disease range by the location of the spine and the region of the affected vertebrae. While the current literature shows that timely diagnosis is crucial to reduce the morbidity and mortality from Pott's disease, there is a lack of specific clinical diagnostic criteria for Pott's disease, and the symptoms may be very non-specific. Studies have shown that novel molecular diagnostic methods are effective and timely choices. Research has implicated the risk factors for the susceptibility and severity of Pott's disease, such as HIV and immunosuppression, poverty, and malnutrition. Based on the current literature available, our group aims to summarize the pathogenesis, clinical features, diagnostic challenges, as well as the known risk factors for Pott's disease within this literature review.
Collapse
|
48
|
Poladian N, Orujyan D, Narinyan W, Oganyan AK, Navasardyan I, Velpuri P, Chorbajian A, Venketaraman V. Role of NF-κB during Mycobacterium tuberculosis Infection. Int J Mol Sci 2023; 24:1772. [PMID: 36675296 PMCID: PMC9865913 DOI: 10.3390/ijms24021772] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/10/2023] [Accepted: 01/14/2023] [Indexed: 01/18/2023] Open
Abstract
Mycobacterium tuberculosis (M. tb) causes tuberculosis infection in humans worldwide, especially among immunocompromised populations and areas of the world with insufficient funding for tuberculosis treatment. Specifically, M. tb is predominantly exhibited as a latent infection, which poses a greater risk of reactivation for infected individuals. It has been previously shown that M. tb infection requires pro-inflammatory and anti-inflammatory mediators to manage its associated granuloma formation via tumor necrosis factor-α (TNF-α), interleukin-12 (IL-12), interferon-γ (IFN-γ), and caseum formation via IL-10, respectively. Nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) has been found to play a unique mediator role in providing a pro-inflammatory response to chronic inflammatory disease processes by promoting the activation of macrophages and the release of various cytokines such as IL-1, IL-6, IL-12, and TNF-α. NF-κB's role is especially interesting in its mechanism of assisting the immune system's defense against M. tb, wherein NF-κB induces IL-2 receptors (IL-2R) to decrease the immune response, but has also been shown to crucially assist in keeping a granuloma and bacterial load contained. In order to understand NF-κB's role in reducing M. tb infection, within this literature review we will discuss the dynamic interaction between M. tb and NF-κB, with a focus on the intracellular signaling pathways and the possible side effects of NF-κB inactivation on M. tb infection. Through a thorough review of these interactions, this review aims to highlight the role of NF-κB in M. tb infection for the purpose of better understanding the complex immune response to M. tb infection and to uncover further potential therapeutic methods.
Collapse
Affiliation(s)
- Nicole Poladian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Davit Orujyan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - William Narinyan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Armani K. Oganyan
- College of Osteopathic Medicine, Des Moines University, 3200 Grand Ave, Des Moines, IA 50312, USA
| | - Inesa Navasardyan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Prathosh Velpuri
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Abraham Chorbajian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
49
|
Mouse Models for Mycobacterium tuberculosis Pathogenesis: Show and Do Not Tell. Pathogens 2022; 12:pathogens12010049. [PMID: 36678397 PMCID: PMC9865329 DOI: 10.3390/pathogens12010049] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 11/29/2022] [Accepted: 12/25/2022] [Indexed: 12/29/2022] Open
Abstract
Science has been taking profit from animal models since the first translational experiments back in ancient Greece. From there, and across all history, several remarkable findings have been obtained using animal models. One of the most popular models, especially for research in infectious diseases, is the mouse. Regarding research in tuberculosis, the mouse has provided useful information about host and bacterial traits related to susceptibility to the infection. The effect of aging, sexual dimorphisms, the route of infection, genetic differences between mice lineages and unbalanced immunity scenarios upon Mycobacterium tuberculosis infection and tuberculosis development has helped, helps and will help biomedical researchers in the design of new tools for diagnosis, treatment and prevention of tuberculosis, despite various discrepancies and the lack of deep study in some areas of these traits.
Collapse
|
50
|
Weng S, Zhang J, Ma H, Zhou J, Jia L, Wan Y, Cui P, Ruan Q, Shao L, Wu J, Wang H, Zhang W, Xu Y. B21 DNA vaccine expressing ag85b, rv2029c, and rv1738 confers a robust therapeutic effect against latent Mycobacterium tuberculosis infection. Front Immunol 2022; 13:1025931. [PMID: 36569899 PMCID: PMC9768437 DOI: 10.3389/fimmu.2022.1025931] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022] Open
Abstract
Latent tuberculosis infection (LTBI) treatment is known to accelerate the decline in TB incidence, especially in high-risk populations. Mycobacterium tuberculosis (M. tb) expression profiles differ at different growth periods, and vaccines protective and therapeutic effects may increase when they include antigenic compositions from different periods. To develop a post-exposure vaccine that targets LTBI, we constructed four therapeutic DNA vaccines (A39, B37, B31, and B21) using different combinations of antigens from the proliferation phase (Ag85A, Ag85B), PE/PPE family (Rv3425), and latent phase (Rv2029c, Rv1813c, Rv1738). We compared the immunogenicity of the four DNA vaccines in C57BL/6j mice. The B21 vaccine stimulated the strongest cellular immune responses, namely Th1/Th17 and CD8+ cytotoxic T lymphocyte responses. It also induced the generation of strengthened effector memory and central memory T cells. In latently infected mice, the B21 vaccine significantly reduced bacterial loads in the spleens and lungs and decreased lung pathology. In conclusion, the B21 DNA vaccine can enhance T cell responses and control the reactivation of LTBI.
Collapse
Affiliation(s)
- Shufeng Weng
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China
| | - Jinyi Zhang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China
| | - Huixia Ma
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China
| | - Jingyu Zhou
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Liqiu Jia
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanmin Wan
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China,Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Peng Cui
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China,Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiaoling Ruan
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Lingyun Shao
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Wu
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Honghai Wang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China
| | - Wenhong Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China,Key Laboratory of Medical Molecular Virology (MOE/MOH), Shanghai Medical College, Fudan University, Shanghai, China,*Correspondence: Ying Xu, ; Wenhong Zhang,
| | - Ying Xu
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China,Shanghai Huashen Institute of Microbes and Infections, Shanghai, China,*Correspondence: Ying Xu, ; Wenhong Zhang,
| |
Collapse
|