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
|
Konjengbam BD, Meitei HN, Pandey A, Haobam R. Goals and strategies in vaccine development against tuberculosis. Mol Immunol 2025; 183:56-71. [PMID: 40327952 DOI: 10.1016/j.molimm.2025.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 04/16/2025] [Accepted: 04/27/2025] [Indexed: 05/08/2025]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), continues to be a major health problem globally. The emergence of multi-drug-resistant TB and extensively drug-resistant TB has become a severe threat to TB control programs. Currently, the Bacille Calmette-Guerin (BCG) vaccine protects a child from disease dissemination efficiently, but its efficiency wanes in adults. Despite all the limitations of BCG and accelerated TB vaccine research, BCG remains the only approved vaccine available for TB. Anti-TB drug treatment has been successful in combating the disease, but it has various side effects and requires an extended drug treatment period. So, vaccination is the finest outlook that can surpass the above-mentioned limitations. Several vaccine candidates are in the pipeline, and the hope for a potential candidate to either boost the BCG vaccine or replace BCG is underway. This review discusses different approaches to TB vaccine development. It summarizes all the challenges and limitations in vaccine development, and its preclinical and clinical trials. Additionally, DNA vaccines and their vaccination techniques are also discussed. Furthermore, the immunoinformatics approach and nanomaterial-based vaccine delivery with practical and productive endpoints are also discussed. Lastly, the potential prospects are also suggested for further studies, which would help bring positive outcomes.
Collapse
Affiliation(s)
| | | | - Anupama Pandey
- Department of Biotechnology, Manipur University, Canchipur, Imphal, Manipur 795003, India
| | - Reena Haobam
- Department of Biotechnology, Manipur University, Canchipur, Imphal, Manipur 795003, India.
| |
Collapse
|
3
|
Banerjee R. Tiny but Mighty: Small RNAs-The Micromanagers of Bacterial Survival, Virulence, and Host-Pathogen Interactions. Noncoding RNA 2025; 11:36. [PMID: 40407594 PMCID: PMC12101431 DOI: 10.3390/ncrna11030036] [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: 03/03/2025] [Revised: 04/07/2025] [Accepted: 04/28/2025] [Indexed: 05/26/2025] Open
Abstract
Bacterial pathogens have evolved diverse strategies to infect hosts, evade immune responses, and establish successful infections. While the role of transcription factors in bacterial virulence is well documented, emerging evidence highlights the significant contribution of small regulatory RNAs (sRNAs) in bacterial pathogenesis. These sRNAs function as posttranscriptional regulators that fine-tune gene expression, enabling bacteria to adapt rapidly to challenging environments. This review explores the multifaceted roles of bacterial sRNAs in host-pathogen interactions. Firstly, it examines how sRNAs regulate pathogenicity by modulating the expression of key virulence factors, including fimbriae, toxins, and secretion systems, followed by discussing the role of sRNAs in bacterial stress response mechanisms that counteract host immune defenses, such as oxidative and envelope stress. Additionally, this review investigates the involvement of sRNAs in antibiotic resistance by regulating efflux pumps, biofilm formation, and membrane modifications, which contribute to multi-drug resistance phenotypes. Lastly, this review highlights how sRNAs contribute to intra- and interspecies communication through quorum sensing, thereby coordinating bacterial behavior in response to environmental cues. Understanding these regulatory networks governed by sRNAs is essential for the development of innovative antimicrobial strategies. This review highlights the growing significance of sRNAs in bacterial pathogenicity and explores their potential as therapeutic targets for the treatment of bacterial infections.
Collapse
Affiliation(s)
- Rajdeep Banerjee
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
4
|
Khojasteh‐Kaffash S, Habibzadeh A, Moghaddam S, Afra F, Samieefar N, Fateh A. Tuberculosis Trends in the Post-COVID-19 Era: Is It Going to be a Global Concern? Health Sci Rep 2025; 8:e70792. [PMID: 40406648 PMCID: PMC12094973 DOI: 10.1002/hsr2.70792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 05/26/2025] Open
Abstract
Background and Aims Tuberculosis (TB), a leading cause of death from infectious diseases, faced considerable challenges during the coronavirus disease 2019 (COVID-19) pandemic. This review examines the impact of pandemic-related disruptions, including the diversion of healthcare resources, reduced access to TB diagnostics and treatment, and declining BCG vaccination rates, on TB trends. The aim is to forecast the post-COVID-19 TB burden, identify risk factors that exacerbate transmission, and propose strategies to prevent a global resurgence. Methods This narrative review incorporates epidemiological data, modeling research, and reports from the World Health Organization and national health systems. It examines TB trends before and after COVID-19, the outcomes of coinfection, and the pandemic's impact on immunology, socioeconomic factors, and health systems. The review also compares trends in India and South Africa-two countries facing significant challenges-to those observed during the COVID-19 pandemic. Results COVID-19 disruptions in healthcare led to an 18% decrease in TB notifications in 2020, resulting in delayed diagnoses, increased household transmission, and higher mortality. Immune dysregulation, including T-cell depletion and cytokine storms, contributed to a 12.3% mortality rate in COVID-19-TB coinfections. Models predict a 5%-15% rise in TB incidence and an additional 1.4 million deaths by 2025. Individuals with HIV, diabetes, and malnutrition were particularly vulnerable. Factors such as overcrowding, air pollution, and reduced Bacillus Calmette-Guérin (BCG) coverage in endemic regions have further heightened susceptibility to TB. Conclusion COVID-19 has undone years of progress in TB control, highlighting the need for a unified health strategy. Early diagnosis, treatment of latent TB, and BCG catch-up initiatives are crucial. Strengthening health systems, addressing socioeconomic factors such as poverty and hunger, and utilizing pandemic advancements like telemedicine and vaccine research will be key to preventing a resurgence of TB. Continued financial support and international cooperation are essential to eliminating TB by 2030.
Collapse
Affiliation(s)
- Soroush Khojasteh‐Kaffash
- Network of Interdisciplinarity in Neonates and Infants (NINI)Universal Scientific Education and Research Network (USERN)TehranIran
- Student Research CommitteeSchool of Medicine, Birjand University of Medical SciencesBirjandIran
| | - Adrina Habibzadeh
- Network of Interdisciplinarity in Neonates and Infants (NINI)Universal Scientific Education and Research Network (USERN)TehranIran
- Student Research CommitteeFasa University of Medical SciencesFasaIran
| | - Sina Moghaddam
- Network of Interdisciplinarity in Neonates and Infants (NINI)Universal Scientific Education and Research Network (USERN)TehranIran
- Department of Internal Medicine, Faculty of Veterinary MedicineUniversity of TehranTehranIran
- Department of Mycobacteriology and Pulmonary ResearchPasteur Institute of IranTehranIran
| | - Fatemeh Afra
- Network of Interdisciplinarity in Neonates and Infants (NINI)Universal Scientific Education and Research Network (USERN)TehranIran
- Clinical Pharmacy DepartmentSina Hospital, Tehran University of Medical Sciences (TUMS)TehranIran
| | - Noosha Samieefar
- Network of Interdisciplinarity in Neonates and Infants (NINI)Universal Scientific Education and Research Network (USERN)TehranIran
- Pediatric Chronic Kidney Disease Research Center, Gene, Cell & Tissue Research Institute, Children's Medical CenterTehran University of Medical SciencesTehranIran
| | - Abolfazl Fateh
- Department of Mycobacteriology and Pulmonary ResearchPasteur Institute of IranTehranIran
- Microbiology Research Center (MRC)Pasteur Institute of IranTehranIran
| |
Collapse
|
5
|
Ning J, Chi S, Zhang Y, Qiao L. Clinical characteristics and prognostic factors of pulmonary tuberculosis with interstitial changes. BMC Infect Dis 2025; 25:624. [PMID: 40301766 PMCID: PMC12039248 DOI: 10.1186/s12879-025-10970-y] [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: 07/20/2024] [Accepted: 04/14/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Pulmonary tuberculosis (PTB) remains a significant global public health challenge, particularly in its manifestation as interstitial lung disease. This form complicates clinical presentation, increasing the difficulty of diagnosis and treatment. However, studies on PTB with interstitial changes are relatively scarce, and their clinical significance and prognostic value have not been fully explored. The objective of the present study was to identify the key factors affecting clinical characteristics and prognosis in these patients. METHODS This retrospective study analyzed data from patients diagnosed with PTB with Interstitial Changes at Zigong First People's Hospital in Sichuan Province between January 2014 and January 2024. Sixteen patients meeting strict inclusion and exclusion criteria were enrolled. Clinical characteristics and key prognostic factors were identified using descriptive statistics and random forest analysis, with partial dependence plots generated to illustrate the independent contributions of each variable to adverse outcomes. RESULTS Among the 16 patients studied, 75.0% were male and 25.0% were female. The average number of pathogen species detected was 1.56 ± 0.73, and 31.3% of patients presented with fever symptoms at admission. Hospital stay durations ranged from 8 to 67 days, with a mean of 22.00 ± 16.02 days. Regarding drug resistance, 25.0% of patients exhibited rifampicin resistance, and approximately 31.2% had underlying diseases. Ultimately, 11 patients (68.8%) recovered, while 5 (31.2%) died. The random forest model identified age, rifampicin resistance, and the number of pathogen species as the main determinants of prognosis. Advanced age and drug resistance were significantly associated with a higher risk of death, and patients infected with multiple pathogens experienced worse outcomes. CONCLUSIONS This study enhances our understanding of the clinical characteristics and prognosis of tuberculosis patients presenting with interstitial lung disease, particularly identifying advanced age, rifampicin resistance, and a higher number of pathogen species as key prognostic factors. These findings provide valuable insights for the development of personalized treatment strategies and precision medicine approaches for this patient group.
Collapse
Affiliation(s)
- Junjie Ning
- Pediatric Intensive Care Unit, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, 610041, China
| | - Shenglin Chi
- Department of tuberculosis, First People's Hospital, Zigong City, Zigong, Sichuan Province, China
| | - Yuanwei Zhang
- Department of tuberculosis, First People's Hospital, Zigong City, Zigong, Sichuan Province, China
| | - Lina Qiao
- Pediatric Intensive Care Unit, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
6
|
Li M, Chen J, Zhang L, Chen X, Zhou J, Liu F, Zhou X, Xiao J, Yang K, Qi L, Han X, Liu T, Zhao H, Zhou Z, Chen X, Sun L. Clinicopathological characteristics and diagnostic performance of metagenomic pathogen detection technology in mycobacterial infections among HIV patients. Front Cell Infect Microbiol 2025; 15:1584189. [PMID: 40365535 PMCID: PMC12069358 DOI: 10.3389/fcimb.2025.1584189] [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: 02/26/2025] [Accepted: 04/04/2025] [Indexed: 05/15/2025] Open
Abstract
Background Mycobacterial infections represent a major cause of morbidity and mortality in HIV-infected individuals. This study evaluated diagnostic techniques for mycobacterial identification and compared clinicopathological features between HIV-positive and HIV-negative patients. Methods We analyzed 88 tissue samples (with 41 matched blood and 28 sputum samples) using histopathology (HE and acid-fast staining), bacterial culture, MTB-PCR (sputum/biopsy), PCR-reverse dot blot hybridization (RDBH), and metagenomic pathogen detection technology (MetaPath™). Logistic regression analyses were performed to identify factors affecting detection rates. Results Mycobacterial infection was detected in 95.5% (84/88) of patients. Among HIV-positive patients (n=63), 46% (29/63) had Mycobacterium tuberculosis (MTB) infections, and 44% (28/63) had non-tuberculous mycobacteria (NTM) infections, significantly higher than the 20% (5/25) NTM rate in HIV-negative patients. Univariate analysis identified HIV-positive status (p=0.009), lymph node involvement (p=0.020), and positive MetaPath™ results (p=0.002) as significant predictors of detection, while multivariate analysis confirmed these as independent factors (p=0.036; p=0.042; p=0.006). Lymph nodes were the most common infection site in HIV-positive patients (42.9%, 27/63), while lung tissue predominated in HIV-negative patients (48%, 12/25). MetaPath™ demonstrated superior sensitivity and specificity for detecting both MTB and NTM. Biopsy samples provided higher diagnostic accuracy than sputum or blood for lung and lymph node infections, but not for brain. In HIV-positive patients, NTM infections showed significantly more granuloma formation (p=0.032) and foam cells (p=0.005), but less necrosis (p=0.0005) compared to MTB infections. No significant differences were observed in HIV-negative patients. Conclusions MetaPath™ is a highly effective diagnostic tool for mycobacterial infections, particularly in tissue biopsies. HIV-positive status, lymph node involvement, and MetaPath™ positivity independently predict mycobacterial detection. HIV-positive patients exhibit distinct clinicopathological features, emphasizing the need for tailored diagnostic and therapeutic approaches based on immune status.
Collapse
Affiliation(s)
- Man Li
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Jiamin Chen
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Liang Zhang
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xiangmei Chen
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Jianfeng Zhou
- Department of Diagnostics, Beijing KingMed Center, Beijing, China
| | - Feifei Liu
- Department of Clinical Laboratory, Guangzhou Kingmed Center, Guangzhou, China
| | - Xingang Zhou
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Jiang Xiao
- Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Kun Yang
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Liming Qi
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xiaoyi Han
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Ting Liu
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Hongxin Zhao
- Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Zhen Zhou
- Department of Interventional Catheterization, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xiaoyou Chen
- Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Lei Sun
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Hilligan KL, Darrah PA, Seder RA, Sher A. Deconvoluting the interplay of innate and adaptive immunity in BCG-induced nonspecific and TB-specific host resistance. J Exp Med 2025; 222:e20240496. [PMID: 40100096 PMCID: PMC11917170 DOI: 10.1084/jem.20240496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/23/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025] Open
Abstract
BCG is the oldest vaccine in continuous use. While current intradermal vaccination regimens confer limited protection outside the context of pediatric extrapulmonary tuberculosis (TB), promising new data indicate that when administered mucosally or intravenously at a higher dose, BCG can induce sterilizing immunity against pulmonary TB in nonhuman primates. BCG is also known to promote nonspecific host resistance against a variety of unrelated infections and is a standard immunotherapy for bladder cancer, suggesting that this innate immune function may contribute to its protective role against TB. Here, we propose that both the mycobacterial-specific and off-target effects of BCG depend on the interplay of adaptive and innate cells and the cytokines they produce, and that the elucidation of this interaction should be a major strategy in the development of more effective BCG-based vaccines and immunotherapies.
Collapse
Affiliation(s)
| | - Patricia A. Darrah
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Robert A. Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alan Sher
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
8
|
Rahim MA, Seo H, Barman I, Hossain MS, Shuvo MSH, Song HY. Insights into Autophagy in Microbiome Therapeutic Approaches for Drug-Resistant Tuberculosis. Cells 2025; 14:540. [PMID: 40214493 PMCID: PMC11989032 DOI: 10.3390/cells14070540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/23/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
Tuberculosis, primarily caused by Mycobacterium tuberculosis, is an airborne lung disease and continues to pose a significant global health threat, resulting in millions of deaths annually. The current treatment for tuberculosis involves a prolonged regimen of antibiotics, which leads to complications such as recurrence, drug resistance, reinfection, and a range of side effects. This scenario underscores the urgent need for novel therapeutic strategies to combat this lethal pathogen. Over the last two decades, microbiome therapeutics have emerged as promising next-generation drug candidates, offering advantages over traditional medications. In 2022, the Food and Drug Administration approved the first microbiome therapeutic for recurrent Clostridium infections, and extensive research is underway on microbiome treatments for various challenging diseases, including metabolic disorders and cancer. Research on microbiomes concerning tuberculosis commenced roughly a decade ago, and the scope of this research has broadened considerably over the last five years, with microbiome therapeutics now viewed as viable options for managing drug-resistant tuberculosis. Nevertheless, the understanding of their mechanisms is still in its infancy. Although autophagy has been extensively studied in other diseases, research into its role in tuberculosis is just beginning, with preliminary developments in progress. Against this backdrop, this comprehensive review begins by succinctly outlining tuberculosis' characteristics and assessing existing treatments' strengths and weaknesses, followed by a detailed examination of microbiome-based therapeutic approaches for drug-resistant tuberculosis. Additionally, this review focuses on establishing a basic understanding of microbiome treatments for tuberculosis, mainly through the lens of autophagy as a mechanism of action. Ultimately, this review aims to contribute to the foundational comprehension of microbiome-based therapies for tuberculosis, thereby setting the stage for the further advancement of microbiome therapeutics for drug-resistant tuberculosis.
Collapse
Affiliation(s)
- Md Abdur Rahim
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Hoonhee Seo
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
- Probiotics Microbiome Commercialization Research Center (PMC), Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Indrajeet Barman
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Mohammed Solayman Hossain
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Md Sarower Hossen Shuvo
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Ho-Yeon Song
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
- Probiotics Microbiome Commercialization Research Center (PMC), Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| |
Collapse
|
9
|
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
|
10
|
Ssekamatte P, Sitenda D, Nabatanzi R, Nakibuule M, Kibirige D, Kyazze AP, Kateete DP, Bagaya BS, Sande OJ, van Crevel R, Cose S, Biraro IA. Isoniazid preventive therapy modulates Mycobacterium tuberculosis-specific T-cell responses in individuals with latent tuberculosis and type 2 diabetes. Sci Rep 2025; 15:10423. [PMID: 40140681 PMCID: PMC11947150 DOI: 10.1038/s41598-025-95386-z] [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: 12/21/2024] [Accepted: 03/20/2025] [Indexed: 03/28/2025] Open
Abstract
Diabetes mellitus (DM) is a significant contributor to tuberculosis (TB) incidence and poor treatment outcomes. This study explored the impact of isoniazid preventive therapy (IPT) on Mycobacterium tuberculosis (Mtb)-specific T-cell memory phenotypes and function among participants with latent TB infection and DM (LTBI-DM) at baseline and after 6 months of IPT; and compared the responses to healthy controls (HC). Peripheral blood mononuclear cells were stimulated with ESAT-6 and CFP-10 peptide pools to analyse CD4+ and CD8+ T-cell responses using flow cytometry. In LTBI-DM participants, effector memory CD4+ and CD8+ T cells were decreased post-IPT, suggesting a shift towards a less-activated state or differentiation into other subsets. CXCR5 expression on both CD4+ and CD8+ T cells was upregulated, while PD-1 expression was downregulated post-IPT, indicating reduced T-cell exhaustion and improved homing capabilities. Lastly, IL-17 A and IL-13 production in CD4+ and CD8+ T cells was increased post-IPT, respectively, which play a role in enhanced Mtb infection control. The post-IPT T-cell alterations were similar to normal HC levels. These findings suggest that IPT modulates and normalises specific T-cell memory phenotypes and functional responses in LTBI-DM participants, potentially contributing to improved long-term immunity and protection against TB. This study highlights the importance of preventive therapy in high-risk populations, and larger studies with more extended follow-up are needed to assess long-lasting IPT effects.
Collapse
Affiliation(s)
- Phillip Ssekamatte
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda.
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda.
| | - Diana Sitenda
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Rose Nabatanzi
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Marjorie Nakibuule
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Davis Kibirige
- Department of Medicine, Uganda Martyrs Hospital Lubaga, Kampala, Uganda
| | - Andrew Peter Kyazze
- Department of Internal Medicine, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - David Patrick Kateete
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Bernard Ssentalo Bagaya
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Obondo James Sande
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Reinout van Crevel
- Department of Internal Medicine and Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Kampala, Uganda
| | - Stephen Cose
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Irene Andia Biraro
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Internal Medicine, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| |
Collapse
|
11
|
Wang T, Quijada D, Ahmedna T, Castillo JR, Naji NS, Peske JD, Karakousis PC, Paul S, Karantanos T, Karanika S. Targeting CCRL2 enhances therapeutic outcomes in a tuberculosis mouse model. Front Immunol 2025; 16:1501329. [PMID: 40181978 PMCID: PMC11965133 DOI: 10.3389/fimmu.2025.1501329] [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: 09/24/2024] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
Tuberculosis (TB) remains among the leading infectious causes of death. Due to the limited number of antimicrobials in the TB drug discovery pipeline, interest has developed in host-directed approaches to improve TB treatment outcomes. C-C motif chemokine-like receptor 2 (CCRL2) is a unique seven-transmembrane domain receptor that is upregulated by inflammatory signals and mediates leucocyte migration. However, little is known about its role in TB infection. Here, we show that Mycobacterium tuberculosis (Mtb) infection increases CCRL2 protein expression in macrophages in vitro and alveolar macrophages (AMs), dendritic cells (DCs) and neutrophils in mouse lungs. To target selectively CCRL2-expressing cells in vivo, we developed a novel mouse anti-CCRL2 antibody-drug conjugate (ADC) linked with the cytotoxic drug SG3249. We tested its adjunctive therapeutic efficacy against TB when combined with the first-line regimen for drug-susceptible TB (isoniazid, rifampin, pyrazinamide, ethambutol; RHZE). The anti-CCRL2 ADC treatment potentiated RHZE efficacy in Mtb-infected mice and decreased gross lung inflammation. CCRL2 expression in lung DCs and AMs was lower in mice receiving anti-CCRL2 ADC treatment+RHZE compared to those receiving RHZE alone or the control group, although the total innate cell populations did not differ across treatment groups. Interestingly, neutrophils were completely absent in the anti-CCRL2 ADC treatment + RHZE group, unlike in the other treatment groups. IFN-γ+-and IL17-α+-T-cell responses, which are associated with optimal TB control, were also elevated in the anti-CCRL2 ADC treatment + RHZE group. Our findings suggest that CCRL2-targeting approaches may improve TB treatment outcomes, possibly through selective killing of Mtb-infected innate immune cells.
Collapse
Affiliation(s)
- Tianyin Wang
- Division of Infectious Diseases, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Darla Quijada
- Division of Infectious Diseases, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Taha Ahmedna
- Ludwig Center and Lustgarten Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jennie Ruelas Castillo
- Division of Infectious Diseases, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nour Sabiha Naji
- Division of Hematologic Malignancies, Department of Medical Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - J David Peske
- Division of Hematopathology, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Petros C. Karakousis
- Division of Infectious Diseases, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Suman Paul
- Ludwig Center and Lustgarten Laboratory, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Division of Hematopathology, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Theodoros Karantanos
- Division of Hematologic Malignancies, Department of Medical Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Styliani Karanika
- Division of Infectious Diseases, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
12
|
Gunasekaran H, Ranganathan UD, Bethunaickan R. The importance of inflammatory biomarkers in detecting and managing latent tuberculosis infection. Front Immunol 2025; 16:1538127. [PMID: 39981231 PMCID: PMC11839662 DOI: 10.3389/fimmu.2025.1538127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/17/2025] [Indexed: 02/22/2025] Open
Abstract
Infection with Mycobacterium tuberculosis (Mtb) triggers an autoimmune-like response in the host leading to further complications. One of the major concerns in eliminating Tuberculosis (TB) is identifying individuals with Latent Tuberculosis Infection (LTBI) who serve as major reservoirs of Mtb making them the important target group for TB eradication. Since no gold standard tests are available for detecting LTBI, the global burden of LTBI cannot be precisely determined. Since LTBI poses several challenges to worldwide healthcare, managing LTBI must be the key priority to achieve a TB-free status. The inflammatory mediators play a major role in determining the outcome of the Mtb infection and also their levels seem to change according to the disease severity. Identification of inflammatory mediators and utilizing them as diagnostic biomarkers for detecting the various stages of TB disease might help identify the reservoirs of Mtb infection even before they become symptomatic so that preventative treatment can be started early. In summary, this review primarily focuses on exploring different inflammatory markers along the course of the Mtb infection. Identifying LTBI-specific biomarkers helps to identify individuals who are at higher risk of developing TB and preparing them to adhere to preventive therapy thus minimizing the global burden of TB.
Collapse
Affiliation(s)
- Harinisri Gunasekaran
- Department of Immunology, ICMR-National Institute for Research in Tuberculosis, Chennai, India
- University of Madras, Chennai, India
| | - Uma Devi Ranganathan
- Department of Immunology, ICMR-National Institute for Research in Tuberculosis, Chennai, India
- University of Madras, Chennai, India
- Faculty of Medical Research, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Ramalingam Bethunaickan
- Department of Immunology, ICMR-National Institute for Research in Tuberculosis, Chennai, India
- University of Madras, Chennai, India
- Faculty of Medical Research, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
13
|
Gupta VK, Vaishnavi VV, Arrieta-Ortiz ML, Abhirami P, Jyothsna K, Jeyasankar S, Raghunathan V, Baliga NS, Agarwal R. 3D Hydrogel Culture System Recapitulates Key Tuberculosis Phenotypes and Demonstrates Pyrazinamide Efficacy. Adv Healthc Mater 2025; 14:e2304299. [PMID: 38655817 PMCID: PMC7616495 DOI: 10.1002/adhm.202304299] [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: 12/04/2023] [Revised: 03/29/2024] [Indexed: 04/26/2024]
Abstract
The mortality caused by tuberculosis (TB) infections is a global concern, and there is a need to improve understanding of the disease. Current in vitro infection models to study the disease have limitations such as short investigation durations and divergent transcriptional signatures. This study aims to overcome these limitations by developing a 3D collagen culture system that mimics the biomechanical and extracellular matrix (ECM) of lung microenvironment (collagen fibers, stiffness comparable to in vivo conditions) as the infection primarily manifests in the lungs. The system incorporates Mycobacterium tuberculosis (Mtb) infected human THP-1 or primary monocytes/macrophages. Dual RNA sequencing reveals higher mammalian gene expression similarity with patient samples than 2D macrophage infections. Similarly, bacterial gene expression more accurately recapitulates in vivo gene expression patterns compared to bacteria in 2D infection models. Key phenotypes observed in humans, such as foamy macrophages and mycobacterial cords, are reproduced in the model. This biomaterial system overcomes challenges associated with traditional platforms by modulating immune cells and closely mimicking in vivo infection conditions, including showing efficacy with clinically relevant concentrations of anti-TB drug pyrazinamide, not seen in any other in vitro infection model, making it reliable and readily adoptable for tuberculosis studies and drug screening.
Collapse
Affiliation(s)
- Vishal K. Gupta
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India
| | | | | | - P.S. Abhirami
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India
| | - K.M. Jyothsna
- Department of Electrical Communication Engineering, Indian Institute of Science, Bengaluru, India
| | | | - Varun Raghunathan
- Department of Electrical Communication Engineering, Indian Institute of Science, Bengaluru, India
| | | | - Rachit Agarwal
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
14
|
Dasan B, Rajamanickam A, Pandiarajan AN, Shanmugam S, Nott S, Babu S. Immunological mechanisms of tuberculosis susceptibility in TB-infected individuals with type 2 diabetes mellitus: insights from mycobacterial growth inhibition assay and cytokine analysis. Microbiol Spectr 2025; 13:e0144524. [PMID: 39656000 PMCID: PMC11705871 DOI: 10.1128/spectrum.01445-24] [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: 06/12/2024] [Accepted: 11/08/2024] [Indexed: 01/11/2025] Open
Abstract
Several studies have highlighted the increased risk of active tuberculosis (TB) in individuals with diabetes mellitus (DM), especially in TB-endemic regions. This dual burden poses significant challenges for TB control efforts. However, there is a lack of reliable laboratory tools to identify individuals at higher risk, and the immunological mechanisms underlying this susceptibility are poorly understood. In this study, we utilized the mycobacterial growth inhibition assay (MGIA) to assess immune response capacity against Mycobacterium tuberculosis (M.tb) in TB infection (TBI) in individuals with type 2 DM (T2DM) (n = 11) compared to those without type 2 DM (NDM) (n = 23). Additionally, we measured various cytokines using multiplex ELISA to understand the immune profile. Our findings revealed that TBI-T2DM individuals exhibited a lower capacity to inhibit M.tb growth compared to TBI-NDM, as evidenced by MGIA results (P = 0.0029). Cytokine analysis further demonstrated diminished production of key cytokines involved in protection, including type 1 (IFNγ, TNFα, IL-2), type 17 (IL-17A), and proinflammatory (IL-1α, IL-1β, IL-6, IL-12p70) cytokines in the TBI-T2DM group compared to TBI-NDM, upon M.tb infection. These findings suggest that MGIA holds promise as an in vitro marker for assessing M.tb immunological control in TBI individuals, particularly those with T2DM. The observed cytokine profile in TBI-T2DM individuals indicates a compromised immune response against M.tb activation, potentially explaining the heightened risk of active TB in this population. IMPORTANCE This study is important because it sheds light on the impaired immune response in individuals with type 2 diabetes mellitus (T2DM) who are infected with Mycobacterium tuberculosis (M.tb), offering critical insights into why they are at higher risk of developing active tuberculosis (TB). By demonstrating that T2DM individuals exhibit a weakened ability to control M.tb growth and a compromised cytokine profile, the research underscores the need for better diagnostic tools, such as the mycobacterial growth inhibition assay (MGIA), to identify those at greater risk of progression to active TB. The findings also highlight the importance of integrated care strategies for managing both T2DM and TB, particularly in TB-endemic regions, and point to the need for further research to develop more effective interventions tailored to this vulnerable population.
Collapse
Affiliation(s)
- Bindu Dasan
- Department of ICER, National Institute of Health-National Institute of Allergy and Infectious Diseases-International Center for Excellence in Research, Chennai, India
| | - Anuradha Rajamanickam
- Department of ICER, National Institute of Health-National Institute of Allergy and Infectious Diseases-International Center for Excellence in Research, Chennai, India
| | - Arul Nancy Pandiarajan
- Department of ICER, National Institute of Health-National Institute of Allergy and Infectious Diseases-International Center for Excellence in Research, Chennai, India
| | - Sivakumar Shanmugam
- Department of Bacteriology, ICMR-National Institute for Research in Tuberculosis, Chennai, India
| | - Sujatha Nott
- Infectious Diseases, Dignity Health, Chandler, Arizona, USA
| | - Subash Babu
- Department of ICER, National Institute of Health-National Institute of Allergy and Infectious Diseases-International Center for Excellence in Research, Chennai, India
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
15
|
Morrison T, Gunn-Moore DA, Hope JC, O’Halloran C. Cytokine and Chemokine Responses of Peripheral Blood Mononuclear Cells from Dogs Infected with Mycobacterium bovis. Pathogens 2024; 14:17. [PMID: 39860978 PMCID: PMC11768291 DOI: 10.3390/pathogens14010017] [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: 11/28/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Mycobacterial infections are an important emerging zoonosis in companion animals for which diagnostic options remain imperfect, and the canine immunological response to these infections has been poorly investigated. We sought to further define the cellular response of peripheral blood mononuclear cells (PBMCs) from dogs infected with Mycobacterium bovis, as determined using a commercial interferon-gamma response assay (IGRA). To this end, PBMCs from healthy or infected dogs were collected. Serum samples were tested to further classify dogs as seropositive or seronegative for circulating antibodies against M. bovis using the DPP® VetTB Assay, Idexx M. bovis antibody ELISA, and a novel purified protein derivative ELISA. Isolated PBMCs were stimulated with mycobacterial proteins (PPDB or ESAT-6/CFP-10), and 13 cytokines/chemokines were measured in the supernatant. These concentrations were determined using the CYTOMAG-90K MILLIPLEX MAP Canine Cytokine/Chemokine system. PBMCs from infected dogs released IFN-γ in response to stimulation, but this response was reduced in those that had seroconverted. Similarly, cells stimulated with PPDB secreted increased amounts of TNF-α when dogs were seronegative, but cells taken from seropositive dogs did not. Finally, the IL-18 response of seropositive dogs was reduced compared to those that were seronegative in response to PPDB, potentially suggesting that these dogs have a reduced macrophage functionality. This work demonstrates that the inflammatory cytokine response may wane following seroconversion with deleterious consequences for the host response. Overall, combining IFN-γ and TNF-α assessment during diagnosis may increase IGRA sensitivity, whilst further work is needed to better understand the prognostic and diagnostic implications of seroconversion in dogs.
Collapse
Affiliation(s)
| | | | | | - Conor O’Halloran
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian EH25 9RG, UK; (T.M.); (D.A.G.-M.); (J.C.H.)
| |
Collapse
|
16
|
Baqi A, Sumalani KK, Akhter N, Ahmed M, Ahmed K, Chawla D, Rizvi N. Frequency and Predictors of Diabetes Mellitus in Smear-Positive Pulmonary Tuberculosis Patients: A Cross-Sectional Study. Cureus 2024; 16:e75809. [PMID: 39816303 PMCID: PMC11734866 DOI: 10.7759/cureus.75809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2024] [Indexed: 01/18/2025] Open
Abstract
Introduction Tuberculosis (TB) is an infectious disease caused by Mycobacterium tuberculosis. Various studies have established an association between diabetes mellitus (DM) and pulmonary TB. This study describes the prevalence of DM and its predictors in smear-positive TB patients. Methods This descriptive cross-sectional study was conducted at the Department of Chest Medicine at Jinnah Postgraduate Medical Centre (JPMC), Karachi, Pakistan, from December 2015 to June 2016. The inclusion criteria were as follows: drug-sensitive pulmonary TB patients of either gender, age ≥ 18 years, registered at the TB clinic JPMC, who consented to participate in the study. Pregnant/lactating patients, drug-resistant TB patients, and those who did not consent to participate in the study were excluded. Fasting blood sugar (FBS) and glycated hemoglobin (HbA1c) were done for all patients included in the study. Sociodemographic data, along with FBS, random blood sugar (RBS), and HbA1c, were entered in a pre-designed proforma. A sedentary lifestyle was recorded as a subjective measure of the proforma. Diabetes was diagnosed if FBS was ≥126 mg/dL or HbA1c was ≥6.5%. Results Out of 100 TB patients included in the study, 18 (18%) were diagnosed with diabetes. Patients who were smokers, had a sedentary lifestyle, and had a family history of diabetes had a significantly high prevalence of diabetes (p-value < 0.0001). Age, gender, and body mass index (BMI) did not influence the prevalence of diabetes in TB patients. Conclusion The study revealed an increased occurrence of diabetes among patients with smear-positive pulmonary TB. Diabetes is more common in TB patients who smoke, have a sedentary lifestyle, and those who have a family history of diabetes. A comprehensive management plan shall be in place to cater to TB and diabetes comorbidity, including effective management of TB and optimal glycemic control.
Collapse
Affiliation(s)
- Abdul Baqi
- Pulmonology, Shaikh Khalifa Bin Zayed Al-Nahyan Medical Complex, Quetta, PAK
| | | | - Nousheen Akhter
- Pulmonology, Bahria University Medical and Dental College, Karachi, PAK
| | - Maqbool Ahmed
- Pulmonology, Fatima Jinnah General and Chest Hospital, Quetta, PAK
| | - Khalid Ahmed
- Pulmonology, Fatima Jinnah General and Chest Hospital, Quetta, PAK
| | - Dimple Chawla
- Pulmonology, Jinnah Postgraduate Medical Centre, Karachi, PAK
| | - Nadeem Rizvi
- Pulmonology, Jinnah Postgraduate Medical Centre, Karachi, PAK
| |
Collapse
|
17
|
Girmay G, Kiflie A, Alem M, Lemma M, Bewket G. Human-immunodeficiency virus infection associated with the impaired Th1 and pro-inflammatory cytokine response in latent tuberculosis-infected individuals: A comparative cross-sectional study. PLoS One 2024; 19:e0313306. [PMID: 39514524 PMCID: PMC11548834 DOI: 10.1371/journal.pone.0313306] [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: 02/20/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Tuberculosis (TB) and HIV co-infections are extensively overlapping, especially in developing countries. HIV infection is known as a major risk factor for the reactivation of latent TB into active TB. Although not fully understood and needs further study, HIV infection might enhance the reactivation of latent TB by breaching immune control mechanisms. We investigated the influence of HIV infection on the cytokine response of LTB-infected individuals. Heparinized venous blood was collected from 40 ART-naïve HIV-infected and 30 HIV-negative healthy controls for LTB screening, plasma collection, and PBMC isolation and stimulation. The level of cytokines in plasma and their production by PBMCs stimulated with purified protein derivative (PPD), staphylococcus enterotoxin B (SEB), or unstimulated PBMCs were analyzed using a cytometric bead array (CBA) assay. PPD-induced IL-2 by PBMCs was higher in LTB-infected groups compared with HIV-negative LTB-negative groups (p = 0.0015). When LTB-infected groups were co-infected with HIV (HIV+LTB+), the IL-2 (p < 0.0001) and IFN-gamma (p = 0.0144) production by PPD-stimulated PBMCs was reduced. The level of IL-2 (p = 0.0070), IL-6 (p = 0.0054), and TNF-alpha (p = 0.0045) in plasma were lower in HIV+LTB+ individuals compared with HIV-negative LTB-positive (HIV-LTB+) groups. Our findings suggested that HIV co-infection in LTB-positive individuals is associated with the diminished production of PPD-induced Th1 (IFN-gamma and IL-2) cytokines by PBMCs and in the plasma level of IL-2 and proinflammatory cytokines (IL-6 and TNF-alpha).
Collapse
Affiliation(s)
- Getu Girmay
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Science, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Amare Kiflie
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Science, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Meseret Alem
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Science, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Mulualem Lemma
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Science, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Gezahegn Bewket
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Science, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
18
|
Zhu L, Wang B, Gu J, Zhou J, Wu Y, Xu W, Yang M, Cai X, Shen H, Lu L, Wang F. IFNγ-secreting T cells that highly express IL-2 potently inhibit the growth of intracellular M. tuberculosis in macrophages. Front Immunol 2024; 15:1469118. [PMID: 39575242 PMCID: PMC11578947 DOI: 10.3389/fimmu.2024.1469118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/18/2024] [Indexed: 11/24/2024] Open
Abstract
Cytokine of interferon-gamma (IFNγ) plays a vital role in the immune response against Mycobacteria tuberculosis (Mtb) infection, yet the specific function of T cells producing IFNγ in this process remains unclear. In this study, we first isolated IFNγ+CD3+ T cells induced by Mtb antigens using surface staining assays. which showed a strong ability to inhibit the growth of intracellular mycobacteria in macrophages. Peripheral blood mononuclear cells (PBMCs) from healthy individuals were then challenged with Bacillus Calmette-Guérin (BCG) or Mtb, respectively, to sort IFNγ-secreting T cells for mRNA sequencing to analyze the gene expression patterns. The results of the integrated data analysis revealed distinct patterns of gene expression between IFNγ+CD3+ T cells induced by the BCG vaccine and those induced by Mtb pathogens. Further, unlike Mtb-induced cells, BCG-induced IFNγ+CD3+ T cells expressed high levels of interleukin-2 (IL-2), which increased the frequencies of these cells and the production of effector cytokines IFNγ and IL-2. Our findings suggested that IFNγ+CD3+ T cells with high IL-2 expression presented potent effector functions to inhibit intracellular Mtb growth, while Mtb infection impaired IL-2 expression in IFNγ+CD3+ T cells.
Collapse
Affiliation(s)
- Liying Zhu
- Shanghai Institute of Infectious Disease and Biosecurity and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Biosafety Level 3 Laboratory, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bo Wang
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jin Gu
- Shanghai Clinical Research Center for Infectious Disease (tuberculosis), Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China
| | - Jiayu Zhou
- Shanghai Institute of Infectious Disease and Biosecurity and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Biosafety Level 3 Laboratory, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuan Wu
- Shanghai Institute of Infectious Disease and Biosecurity and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Biosafety Level 3 Laboratory, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Xu
- Shanghai Institute of Infectious Disease and Biosecurity and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Biosafety Level 3 Laboratory, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Min Yang
- Shanghai Institute of Infectious Disease and Biosecurity and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Biosafety Level 3 Laboratory, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xia Cai
- Shanghai Institute of Infectious Disease and Biosecurity and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Biosafety Level 3 Laboratory, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongbo Shen
- Shanghai Clinical Research Center for Infectious Disease (tuberculosis), Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Institute for Advanced Study, Tongji University School of Medicine, Shanghai, China
- Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, China
| | - Lu Lu
- Shanghai Institute of Infectious Disease and Biosecurity and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Biosafety Level 3 Laboratory, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Feifei Wang
- Shanghai Institute of Infectious Disease and Biosecurity and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Biosafety Level 3 Laboratory, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Guo S, Ouyang J, Hu Z, Cao T, Huang C, Mou J, Gu X, Liu J. Intranasal vaccination with engineered BCG expressing CCL2 induces a stronger immune barrier against Mycobacterium tuberculosis than BCG. Mol Ther 2024; 32:3990-4005. [PMID: 39295146 PMCID: PMC11573603 DOI: 10.1016/j.ymthe.2024.09.021] [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: 03/23/2024] [Revised: 07/14/2024] [Accepted: 09/13/2024] [Indexed: 09/21/2024] Open
Abstract
Intradermal Mycobacterium bovis Bacillus Calmette-Guérin (BCG) vaccination is currently the only licensed strategy for preventing tuberculosis (TB). It provides limited protection against pulmonary TB. To enhance the efficacy of BCG, we developed a recombinant BCG expressing exogenous monocyte chemoattractant CC chemokine ligand 2 (CCL2) called rBCG-CCL2. Co-culturing macrophages with rBCG-CCL2 enhances their abilities in migration, phagocytosis, and effector molecule expression. In the mouse model, intranasal vaccination with rBCG-CCL2 induced greater immune cell infiltration and a more extensive innate immune response in lung compared to vaccination with parental BCG, as determined by multiparameter flow cytometry, transcriptomic analysis, and pathological assessments. Moreover, rBCG-CCL2 induced a high frequency of activated macrophages and antigen-specific T helper 1 (Th1) and Th17 T cells in lungs. The enhanced immune microenvironment responded more effectively to intravenous challenge with Mycobacterium tuberculosis (Mtb) H37Ra, leading to significant reductions in H37Ra burden and pathological damage to the lungs and spleen. Intranasal rBCG-CCL2-vaccinated mice rapidly initiated pro-inflammatory Th1 cytokine release and reduced pathological damage to the lungs and spleen during the early stage of H37Ra challenge. The finding that co-expression of CCL2 synergistically enhances the immune barrier induced by BCG provides a model for defining immune correlates and mechanisms of vaccine-elicited protection against TB.
Collapse
Affiliation(s)
- Shaohua Guo
- Center for Infectious Disease and Vaccine, West China Hospital, Sichuan University, Chengdu 610041, P.R. China
| | - Jiangshan Ouyang
- Center for Infectious Disease and Vaccine, West China Hospital, Sichuan University, Chengdu 610041, P.R. China
| | - Zhiming Hu
- Center for Infectious Disease and Vaccine, West China Hospital, Sichuan University, Chengdu 610041, P.R. China
| | - Ting Cao
- Center for Infectious Disease and Vaccine, West China Hospital, Sichuan University, Chengdu 610041, P.R. China
| | - Chunxu Huang
- Center for Infectious Disease and Vaccine, West China Hospital, Sichuan University, Chengdu 610041, P.R. China
| | - Jun Mou
- Center for Infectious Disease and Vaccine, West China Hospital, Sichuan University, Chengdu 610041, P.R. China
| | - Xinxia Gu
- Center for Infectious Disease and Vaccine, West China Hospital, Sichuan University, Chengdu 610041, P.R. China
| | - Jie Liu
- Center for Infectious Disease and Vaccine, West China Hospital, Sichuan University, Chengdu 610041, P.R. China.
| |
Collapse
|
20
|
Wang T, Quijada D, Ahmenda T, Castillo JR, Naji NS, Peske JD, Karakousis PC, Paul S, Karantanos T, Karanika S. Targeting CCRL2 enhances therapeutic outcomes in a tuberculosis mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614576. [PMID: 39386470 PMCID: PMC11463537 DOI: 10.1101/2024.09.23.614576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Tuberculosis (TB) remains among the leading infectious causes of death. Due to the limited number of antimicrobials in the TB drug discovery pipeline, interest has developed in host-directed approaches to improve TB treatment outcomes. C-C motif chemokine-like receptor 2 (CCRL2) is a unique seven-transmembrane domain receptor that is upregulated by inflammatory signals and mediates leucocyte migration. However, little is known about its role in the setting of TB infection. Here, we show that Mycobacterium tuberculosis (Mtb) infection increases CCRL2 protein expression in macrophages and in mouse lungs. To target selectively CCRL2-expressing cells in vivo, we developed a novel mouse anti-CCRL2 antibody-drug conjugate (ADC) linked with the cytotoxic drug SG3249. We tested its adjunctive therapeutic efficacy against TB when combined with the first-line regimen for drug-susceptible TB (isoniazid, rifampin, pyrazinamide, ethambutol; RHZE). The anti-CCRL2 ADC treatment potentiated RHZE efficacy in Mtb-infected mice and decreased gross lung inflammation. CCRL2 expression in lung dendritic cells and alveolar macrophages was lower in mice receiving anti-CCRL2 ADC treatment + RHZE compared to those receiving RHZE alone or the control group, although the total innate cell populations did not differ across treatment groups. Interestingly, neutrophils were completely absent in the anti-CCRL2 ADC treatment + RHZE group, unlike in the other treatment groups. IFN-γ+ and IL17-Α+ T-cell responses, which are associated with optimal TB control, were also elevated in the anti-CCRL2 ADC treatment + RHZE group. Collectively, our findings suggest that CCRL2-targeting approaches may improve TB treatment outcomes, possibly through selective killing of Mtb-infected innate immune cells.
Collapse
|
21
|
Karanika S, Wang T, Yilma A, Castillo JR, Gordy JT, Bailey H, Quijada D, Fessler K, Tasneen R, Rouse Salcido EM, Harris HT, Bates RE, Ton H, Meza J, Li Y, Taylor AD, Zheng JJ, Zhang J, Peske JD, Karantanos T, Maxwell AR, Nuermberger E, Markham RB, Karakousis PC. Therapeutic DNA Vaccine Targeting Mycobacterium tuberculosis Persisters Shortens Curative Tuberculosis Treatment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.03.611055. [PMID: 39282461 PMCID: PMC11398349 DOI: 10.1101/2024.09.03.611055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/11/2024]
Abstract
Mycobacterium tuberculosis ( Mtb) is one of the leading infectious causes of death worldwide. There is no available licensed therapeutic vaccine that shortens active tuberculosis (TB) disease drug treatment and prevents relapse, despite the World Health Organization's calls. Here, we show that an intranasal DNA vaccine containing a fusion of the stringent response rel Mtb gene with the gene encoding the immature dendritic cell-targeting chemokine, MIP-3α/CCL20, shortens the duration of curative TB treatment in immunocompetent mice. Compared to the first-line regimen for drug-susceptible TB alone, our novel adjunctive vaccine induced greater Rel Mtb -specific T-cell responses associated with optimal TB control in spleen, blood, lungs, mediastinal lymph nodes, and bronchoalveolar lavage (BAL) fluid. These responses were sustained, if not augmented, over time. It also triggered more effective dendritic cell recruitment, activation, and colocalization with T cells, implying enhanced crosstalk between innate and adaptive immunity. Moreover, it potentiated a 6-month TB drug-resistant regimen, rendering it effective across treatment regimens, and also showed promising results in CD4+ knockout mice, perhaps due to enhanced Rel-specific CD8+ T-cell responses. Notably, our novel fusion vaccine was also immunogenic in nonhuman primates, the gold standard animal model for TB vaccine studies, eliciting antigen-specific T-cell responses in blood and BAL fluid analogous to those observed in protected mice. Our findings have critical implications for therapeutic TB vaccine clinical development in immunocompetent and immunocompromised populations and may serve as a model for defining immunological correlates of therapeutic vaccine-induced protection. One sentence summary A TB vaccine shortens curative drug treatment in mice by eliciting strong TB-protective immune responses and induces similar responses in macaques.
Collapse
|
22
|
Ongarj J, Intapiboon P, Surasombatpattana S, Satti I, Harris SA, Morrison H, Sophonmanee R, McShane H, Tanner R, Pinpathomrat N. Evaluation of immune profiles associated with control of mycobacterial growth in systemic lupus erythematosus (SLE) patients. Tuberculosis (Edinb) 2024; 148:102533. [PMID: 38878478 DOI: 10.1016/j.tube.2024.102533] [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: 04/07/2024] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 09/09/2024]
Abstract
Tuberculosis (TB) is an infectious disease with the burden concentrated in low- and middle-income countries. Systemic lupus erythematosus (SLE) is an autoimmune disease associated with widespread inflammation that is prevalent in some TB endemic areas including East Africa and parts of Southeast Asia. SLE patients are known to be at higher risk of becoming infected with M. tb, developing TB disease. However, the immune mechanisms underlying this susceptibility are not well understood, particularly in the absence of immunosuppressive drugs. We present a pilot study in which we have evaluated intracellular cytokine responses and ex vivo ability to control mycobacterial growth using peripheral blood mononuclear cells (PBMC) collected from SLE patients before and during SLE treatment. After six months of treatment, SLE patients had the highest frequencies of CD8+ T cells, NK cells and NKT cells producing IFN-γ and/or TNF-α. This group also showed superior control of mycobacterial growth, and proinflammatory cytokine-producing NK and NKT cells correlated with mycobacterial growth inhibition at the individual patient level. These findings contribute to a better understanding of autoimmune profiles associated with control of mycobacterial growth in SLE patients, which may inform intervention strategies to reduce risk of TB disease in this population.
Collapse
Affiliation(s)
- Jomkwan Ongarj
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Porntip Intapiboon
- Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | | | - Iman Satti
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Hazel Morrison
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Ratchanon Sophonmanee
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Helen McShane
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Rachel Tanner
- Department of Biology, University of Oxford, Oxford, United Kingdom
| | - Nawamin Pinpathomrat
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand.
| |
Collapse
|
23
|
Banoo S, Yadav Y, Tyagi R, Manna A, Sagar R. Recent efforts in the development of glycoconjugate vaccine and available treatment for tuberculosis. Bioorg Chem 2024; 150:107610. [PMID: 38991488 DOI: 10.1016/j.bioorg.2024.107610] [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: 04/02/2024] [Revised: 06/23/2024] [Accepted: 06/30/2024] [Indexed: 07/13/2024]
Abstract
Tuberculosis (TB) continues to pose a grave threat to global health, despite relentless eradication efforts. In 1882, Robert Koch discovered that Mycobacterium tuberculosis (Mtb) is the bacterium responsible for causing tuberculosis. It is a fact that tuberculosis has claimed the lives of more than one billion people in the last few decades. It is imperative that we must take immediate and effective action to increase resources for TB research and treatment. Effective TB treatments demand an extensive investment of both time and finances, often requiring 6-9 months of rigorous antibiotic therapy. The most efficient way to control tuberculosis is by receiving a childhood Bacillus Calmette-Guérin (BCG) vaccination. Despite years of research on vaccine development, we still do not have any new approved vaccine for tuberculosis, except BCG, which is partially effective in young children. This review discusses briefly the available treatment for tuberculosis and remarkable advancements in glycoconjugate-based TB vaccine developments in recent years (2013-2024) and offers valuable direction for future research priorities.
Collapse
Affiliation(s)
- Sajida Banoo
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Yogesh Yadav
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rajdeep Tyagi
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Arunava Manna
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India.
| | - Ram Sagar
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India; Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
24
|
Abbasnia S, Hashem Asnaashari AM, Sharebiani H, Soleimanpour S, Mosavat A, Rezaee SA. Mycobacterium tuberculosis and host interactions in the manifestation of tuberculosis. J Clin Tuberc Other Mycobact Dis 2024; 36:100458. [PMID: 38983441 PMCID: PMC11231606 DOI: 10.1016/j.jctube.2024.100458] [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] [Indexed: 07/11/2024] Open
Abstract
The final step of epigenetic processes is changing the gene expression in a new microenvironment in the body, such as neuroendocrine changes, active infections, oncogenes, or chemical agents. The case of tuberculosis (TB) is an outcome of Mycobacterium tuberculosis (M.tb) and host interaction in the manifestation of active and latent TB or clearance. This comprehensive review explains and interprets the epigenetics findings regarding gene expressions on the host-pathogen interactions in the development and progression of tuberculosis. This review introduces novel insights into the complicated host-pathogen interactions, discusses the challengeable results, and shows the gaps in the clear understanding of M.tb behavior. Focusing on the biological phenomena of host-pathogen interactions, the epigenetic changes, and their outcomes provides a promising future for developing effective TB immunotherapies when converting gene expression toward appropriate host immune responses gradually becomes attainable. Overall, this review may shed light on the dark sides of TB pathogenesis as a life-threatening disease. Therefore, it may support effective planning and implementation of epigenetics approaches for introducing proper therapies or effective vaccines.
Collapse
Affiliation(s)
- Shadi Abbasnia
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hiva Sharebiani
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saman Soleimanpour
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arman Mosavat
- Blood Borne Infections Research Center, Academic Center for Education, Culture, and Research (ACECR), Razavi Khorasan, Mashhad, Iran
| | - Seyed Abdolrahim Rezaee
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
25
|
Wei J, Ning H, Ramos‐Espinosa O, Eickhoff CS, Hou R, Wang Q, Fu M, Liu EY, Fan D, Hoft DF, Liu J. Tristetraprolin mediates immune evasion of mycobacterial infection in macrophages. FASEB Bioadv 2024; 6:249-262. [PMID: 39114448 PMCID: PMC11301268 DOI: 10.1096/fba.2024-00022] [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: 01/31/2024] [Revised: 05/15/2024] [Accepted: 05/30/2024] [Indexed: 08/10/2024] Open
Abstract
Immune evasion of Mycobacterium tuberculosis (Mtb) facilitates intracellular bacterial growth. The mechanisms of immune evasion, however, are still not fully understood. In this study, we reveal that tristetraprolin (TTP), one of the best characterized RNA-binding proteins controlling the stability of targeted mRNAs, mediates innate immune evasion of mycobacteria. We found that TTP knockout mice displayed reduced bacterial burden in the early stage after Mtb aerosol challenge. Macrophages deficient in TTP also showed an inhibition in intracellular mycobacterial growth. Live mycobacteria induced TTP protein expression in macrophages, which was blocked by the mTOR inhibitor rapamycin. Rapamycin and AZD8055 specifically blocked 4EBP1 phosphorylation in infected macrophages and suppressed intracellular BCG growth. Rapamycin promoted TTP protein degradation through the ubiquitination pathway, whereas the proteasome inhibitor MG-132 blocked rapamycin function and thus stabilized TTP protein. TTP induction suppressed the expression of iNOS/TNF-α/IL-12/IL-23, and weakened protective immune responses in macrophages, whereas rapamycin enhanced the bactericidal effects through TTP inhibition. Moreover, blocking TTP binding increased the expression of TNF-α and iNOS and suppressed intracellular mycobacterial growth. Overall, our study reveals a novel role for RNA-binding protein TTP in Mtb immune evasion mechanisms and provides a potential target for host-directed therapy against tuberculosis (TB).
Collapse
Affiliation(s)
- Jiawei Wei
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal MedicineSaint Louis University School of Medicine, Saint Louis UniversitySt. LouisMissouriUSA
| | - Huan Ning
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal MedicineSaint Louis University School of Medicine, Saint Louis UniversitySt. LouisMissouriUSA
| | - Octavio Ramos‐Espinosa
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal MedicineSaint Louis University School of Medicine, Saint Louis UniversitySt. LouisMissouriUSA
| | - Christopher S. Eickhoff
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal MedicineSaint Louis University School of Medicine, Saint Louis UniversitySt. LouisMissouriUSA
| | - Rong Hou
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal MedicineSaint Louis University School of Medicine, Saint Louis UniversitySt. LouisMissouriUSA
| | - Qinghong Wang
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal MedicineSaint Louis University School of Medicine, Saint Louis UniversitySt. LouisMissouriUSA
| | - Mingui Fu
- Shock/Trauma Research Center, Department of Basic Medical Science, School of MedicineUniversity of Missouri‐Kansas CityKansas CityMissouriUSA
| | - Ethan Y. Liu
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal MedicineSaint Louis University School of Medicine, Saint Louis UniversitySt. LouisMissouriUSA
| | - Daping Fan
- Department of Cell Biology and AnatomyUniversity of South Carolina School of MedicineColumbiaSouth CarolinaUSA
| | - Daniel F. Hoft
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal MedicineSaint Louis University School of Medicine, Saint Louis UniversitySt. LouisMissouriUSA
| | - Jianguo Liu
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal MedicineSaint Louis University School of Medicine, Saint Louis UniversitySt. LouisMissouriUSA
| |
Collapse
|
26
|
Reid VA, Ramos EI, Veerapandian R, Carmona A, Gadad SS, Dhandayuthapani S. Differential Expression of lncRNAs in HIV Patients with TB and HIV-TB with Anti-Retroviral Treatment. Noncoding RNA 2024; 10:40. [PMID: 39051374 PMCID: PMC11270221 DOI: 10.3390/ncrna10040040] [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: 05/21/2024] [Revised: 06/26/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024] Open
Abstract
Tuberculosis (TB) is the leading cause of death among people with HIV-1 infection. To improve the diagnosis and treatment of HIV-TB patients, it is important to understand the mechanisms underlying these conditions. Here, we used an integrated genomics approach to analyze and determine the lncRNAs that are dysregulated in HIV-TB patients and HIV-TB patients undergoing anti-retroviral therapy (ART) using a dataset available in the public domain. The analyses focused on the portion of the genome transcribed into non-coding transcripts, which historically have been poorly studied and received less focus. This revealed that Mtb infection in HIV prominently up-regulates the expression of long non-coding RNA (lncRNA) genes DAAM2-AS1, COL4A2-AS1, LINC00599, AC008592.1, and CLRN1-AS1 and down-regulates the expression of lncRNAs AC111000.4, AC100803.3, AC016168.2, AC245100.7, and LINC02073. It also revealed that ART down-regulates the expression of some lncRNA genes (COL4A2-AS1, AC079210.1, MFA-AS1, and LINC01993) that are highly up-regulated in HIV-TB patients. Furthermore, the interrogation of the genomic regions that are associated with regulated lncRNAs showed enrichment for biological processes linked to immune pathways in TB-infected conditions. However, intriguingly, TB patients treated with ART showed completely opposite and non-overlapping pathways. Our findings suggest that lncRNAs could be used to identify critical diagnostic, prognostic, and treatment targets for HIV-TB patients.
Collapse
Affiliation(s)
- Victoria A. Reid
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (V.A.R.); (R.V.); (A.C.)
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA;
| | - Enrique I. Ramos
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA;
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Raja Veerapandian
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (V.A.R.); (R.V.); (A.C.)
| | - Areanna Carmona
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (V.A.R.); (R.V.); (A.C.)
| | - Shrikanth S. Gadad
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (V.A.R.); (R.V.); (A.C.)
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA;
- Frederick L. Francis Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Subramanian Dhandayuthapani
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (V.A.R.); (R.V.); (A.C.)
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA;
- Frederick L. Francis Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| |
Collapse
|
27
|
Lai R, Williams T, Rakib T, Lee J, Behar SM. Heterogeneity in lung macrophage control of Mycobacterium tuberculosis is modulated by T cells. Nat Commun 2024; 15:5710. [PMID: 38977711 PMCID: PMC11231272 DOI: 10.1038/s41467-024-48515-7] [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: 10/16/2023] [Accepted: 05/03/2024] [Indexed: 07/10/2024] Open
Abstract
Following Mycobacterium tuberculosis infection, alveolar macrophages are initially infected but ineffectively restrict bacterial replication. The distribution of M. tuberculosis among different cell types in the lung changes with the onset of T cell immunity when the dominant infected cellular niche shifts from alveolar to monocyte-derived macrophages (MDM). We hypothesize that changes in bacterial distribution among different cell types is driven by differences in T cell recognition of infected cells and their subsequent activation of antimicrobial effector mechanisms. We show that CD4 and CD8 T cells efficiently eliminate M. tuberculosis infection in alveolar macrophages, but they have less impact on suppressing infection in MDM, which may be a bacterial niche. Importantly, CD4 T cell responses enhance MDM recruitment to the lung. Thus, the outcome of infection depends on the interaction between the T cell subset and the infected cell; both contribute to the resolution and persistence of the infection.
Collapse
Affiliation(s)
- Rocky Lai
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| | - Travis Williams
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| | - Tasfia Rakib
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jinhee Lee
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| | - Samuel M Behar
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
28
|
Rabe H, Lönnermark E, Johansson E, Gilljam M, Jönsson B. In vitro stimulation with nontuberculous mycobacteria induced a stronger cytokine response in leukocytes isolated from individuals with latent tuberculosis compared to those isolated from active tuberculosis or cystic fibrosis patients. Tuberculosis (Edinb) 2024; 147:102504. [PMID: 38522174 DOI: 10.1016/j.tube.2024.102504] [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: 12/19/2023] [Revised: 03/08/2024] [Accepted: 03/17/2024] [Indexed: 03/26/2024]
Abstract
Mycobacterium tuberculosis and opportunistic environmental non-tuberculous mycobacteria (NTM) can cause severe infection. Why latent tuberculosis infection advances to active disease, and why some individuals with cystic fibrosis (CF) develop pulmonary infections with NTM is still poorly understood. The aim of this study was to investigate the effector function of peripheral blood mononuclear cells (PBMC) from individuals with active or latent tuberculosis, individuals with CF with or without pulmonary NTM-infection and healthy controls, by measuring cytokine response to in vitro stimulation with different species of NTMs. The cytokine concentrations of IL-17A, IL-22, IL-23, IL-10, IL12p70 and IFN-γ were measured in PBMC-culture supernatants after stimulation with NTMs. PBMCs from individuals with latent tuberculosis infection showed strong IL-17A, IL-22, and IFN-γ responses compared to individuals with active tuberculosis or CF. IL-10 production was low in both tuberculosis groups compared to the CF groups and controls. This study suggests that IL-17A and IL-22 might be important to keep tuberculosis in a latent phase and that individuals with CF with an ongoing NTM infection seem to have a low cytokine response.
Collapse
Affiliation(s)
- Hardis Rabe
- Unit of Biological Function, Research Institutes of Sweden (RISE AB), Gothenburg, Sweden; Institute of Biomedicine, Department of infectious diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Elisabeth Lönnermark
- Institute of Biomedicine, Department of infectious diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Infectious Diseases, Gothenburg, Sweden.
| | - Ewa Johansson
- Institute of Biomedicine, Department of infectious diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Microbiology, Gothenburg, Sweden.
| | - Marita Gilljam
- Region Västra Götaland, Sahlgrenska University Hospital, Respiratory Medicine, Department of Internal Medicine and Clinical Nutrition, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg CF Centre, Adult Clinic, Gothenburg, Sweden.
| | - Bodil Jönsson
- Institute of Biomedicine, Department of infectious diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Microbiology, Gothenburg, Sweden.
| |
Collapse
|
29
|
Maio M, Barros J, Joly M, Vahlas Z, Marín Franco JL, Genoula M, Monard SC, Vecchione MB, Fuentes F, Gonzalez Polo V, Quiroga MF, Vermeulen M, Vu Manh TP, Argüello RJ, Inwentarz S, Musella R, Ciallella L, González Montaner P, Palmero D, Lugo Villarino G, Sasiain MDC, Neyrolles O, Vérollet C, Balboa L. Elevated glycolytic metabolism of monocytes limits the generation of HIF1A-driven migratory dendritic cells in tuberculosis. eLife 2024; 12:RP89319. [PMID: 38922679 PMCID: PMC11208050 DOI: 10.7554/elife.89319] [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: 06/27/2024] Open
Abstract
During tuberculosis (TB), migration of dendritic cells (DCs) from the site of infection to the draining lymph nodes is known to be impaired, hindering the rapid development of protective T-cell-mediated immunity. However, the mechanisms involved in the delayed migration of DCs during TB are still poorly defined. Here, we found that infection of DCs with Mycobacterium tuberculosis (Mtb) triggers HIF1A-mediated aerobic glycolysis in a TLR2-dependent manner, and that this metabolic profile is essential for DC migration. In particular, the lactate dehydrogenase inhibitor oxamate and the HIF1A inhibitor PX-478 abrogated Mtb-induced DC migration in vitro to the lymphoid tissue-specific chemokine CCL21, and in vivo to lymph nodes in mice. Strikingly, we found that although monocytes from TB patients are inherently biased toward glycolysis metabolism, they differentiate into poorly glycolytic and poorly migratory DCs compared with healthy subjects. Taken together, these data suggest that because of their preexisting glycolytic state, circulating monocytes from TB patients are refractory to differentiation into migratory DCs, which may explain the delayed migration of these cells during the disease and opens avenues for host-directed therapies for TB.
Collapse
Grants
- PICT-2019-01044 Agencia Nacional de Promoción de la Investigación, el Desarrollo Tecnológico y la Innovación
- PICT-2020-00501 Agencia Nacional de Promoción Científica y Tecnológica
- 11220200100299CO Consejo Nacional de Investigaciones Científicas y Técnicas
- ANRS2018-02 Agence Nationale de Recherches sur le Sida et les Hépatites Virales
- ECTZ 118551/118554 Agence Nationale de Recherches sur le Sida et les Hépatites Virales
- ECTZ 205320/305352 Agence Nationale de Recherches sur le Sida et les Hépatites Virales
- ECTZ103104 Agence Nationale de Recherches sur le Sida et les Hépatites Virales
- ECTZ101971 Agence Nationale de Recherches sur le Sida et les Hépatites Virales
- ANR-20-CE14-0028 Agence Nationale de la Recherche
- MAT-PI-17493-A-04 Inserm Transfert
- CONICET The Argentinean National Council of Scientific and Technical Investigations
- PIP 11220200100299CO The Argentinean National Council of Scientific and Technical Investigations
- ANRS2018-02 The Centre National de la Recherche Scientifique, Université Paul Sabatier, the Agence Nationale de Recherche sur le Sida et les hépatites virales (ANRS)
- ECTZ 118551/118554 The Centre National de la Recherche Scientifique, Université Paul Sabatier, the Agence Nationale de Recherche sur le Sida et les hépatites virales (ANRS)
- ECTZ 205320/305352 The Centre National de la Recherche Scientifique, Université Paul Sabatier, the Agence Nationale de Recherche sur le Sida et les hépatites virales (ANRS)
- ANRS ECTZ103104 The Centre National de la Recherche Scientifique, Université Paul Sabatier, the Agence Nationale de Recherche sur le Sida et les hépatites virales (ANRS)
- ECTZ101971 The Centre National de la Recherche Scientifique, Université Paul Sabatier, the Agence Nationale de Recherche sur le Sida et les hépatites virales (ANRS)
- ANR-20-CE14-0028 The French ANR JCJC-Epic-SCENITH
- MAT-PI-17493-A-04 CoPoC Inserm-transfert
Collapse
Affiliation(s)
- Mariano Maio
- Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de MedicinaBuenos AiresArgentina
- International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina / International Research Project ToulouseToulouseFrance
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Universidad de Buenos AiresBuenos AiresArgentina
| | - Joaquina Barros
- Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de MedicinaBuenos AiresArgentina
- International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina / International Research Project ToulouseToulouseFrance
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Universidad de Buenos AiresBuenos AiresArgentina
| | - Marine Joly
- International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina / International Research Project ToulouseToulouseFrance
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPSToulouseFrance
| | - Zoi Vahlas
- International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina / International Research Project ToulouseToulouseFrance
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPSToulouseFrance
| | - José Luis Marín Franco
- Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de MedicinaBuenos AiresArgentina
- International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina / International Research Project ToulouseToulouseFrance
| | - Melanie Genoula
- Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de MedicinaBuenos AiresArgentina
- International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina / International Research Project ToulouseToulouseFrance
| | - Sarah C Monard
- International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina / International Research Project ToulouseToulouseFrance
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPSToulouseFrance
| | - María Belén Vecchione
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Universidad de Buenos AiresBuenos AiresArgentina
| | - Federico Fuentes
- Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de MedicinaBuenos AiresArgentina
| | - Virginia Gonzalez Polo
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Universidad de Buenos AiresBuenos AiresArgentina
| | - María Florencia Quiroga
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Universidad de Buenos AiresBuenos AiresArgentina
| | - Mónica Vermeulen
- Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de MedicinaBuenos AiresArgentina
| | - Thien-Phong Vu Manh
- Aix Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-LuminyMarseilleFrance
| | - Rafael J Argüello
- Aix Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-LuminyMarseilleFrance
| | - Sandra Inwentarz
- Instituto Prof. Dr. Raúl Vaccarezza and Hospital de Infecciosas Dr. F.J. MuñizBuenos AiresArgentina
| | - Rosa Musella
- Instituto Prof. Dr. Raúl Vaccarezza and Hospital de Infecciosas Dr. F.J. MuñizBuenos AiresArgentina
| | - Lorena Ciallella
- Instituto Prof. Dr. Raúl Vaccarezza and Hospital de Infecciosas Dr. F.J. MuñizBuenos AiresArgentina
| | - Pablo González Montaner
- Instituto Prof. Dr. Raúl Vaccarezza and Hospital de Infecciosas Dr. F.J. MuñizBuenos AiresArgentina
| | - Domingo Palmero
- Instituto Prof. Dr. Raúl Vaccarezza and Hospital de Infecciosas Dr. F.J. MuñizBuenos AiresArgentina
| | - Geanncarlo Lugo Villarino
- International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina / International Research Project ToulouseToulouseFrance
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPSToulouseFrance
| | - María del Carmen Sasiain
- Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de MedicinaBuenos AiresArgentina
- International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina / International Research Project ToulouseToulouseFrance
| | - Olivier Neyrolles
- International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina / International Research Project ToulouseToulouseFrance
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPSToulouseFrance
| | - Christel Vérollet
- International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina / International Research Project ToulouseToulouseFrance
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPSToulouseFrance
| | - Luciana Balboa
- Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de MedicinaBuenos AiresArgentina
- International Associated Laboratory (LIA) CNRS IM-TB/HIV (1167), Buenos Aires, Argentina / International Research Project ToulouseToulouseFrance
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Universidad de Buenos AiresBuenos AiresArgentina
| |
Collapse
|
30
|
Dintwe OB, Ballweber Fleming L, Voillet V, McNevin J, Seese A, Naidoo A, Omarjee S, Bekker LG, Kublin JG, De Rosa SC, Newell EW, Fiore-Gartland A, Andersen-Nissen E, McElrath MJ. Adolescent BCG revaccination induces a phenotypic shift in CD4 + T cell responses to Mycobacterium tuberculosis. Nat Commun 2024; 15:5191. [PMID: 38890283 PMCID: PMC11189459 DOI: 10.1038/s41467-024-49050-1] [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: 04/17/2023] [Accepted: 05/15/2024] [Indexed: 06/20/2024] Open
Abstract
A recent clinical trial demonstrated that Bacille Calmette-Guérin (BCG) revaccination of adolescents reduced the risk of sustained infection with Mycobacterium tuberculosis (M.tb). In a companion phase 1b trial, HVTN 602/Aeras A-042, we characterize in-depth the cellular responses to BCG revaccination or to a H4:IC31 vaccine boost to identify T cell subsets that could be responsible for the protection observed. High-dimensional clustering analysis of cells profiled using a 26-color flow cytometric panel show marked increases in five effector memory CD4+ T cell subpopulations (TEM) after BCG revaccination, two of which are highly polyfunctional. CITE-Seq single-cell analysis shows that the activated subsets include an abundant cluster of Th1 cells with migratory potential. Additionally, a small cluster of Th17 TEM cells induced by BCG revaccination expresses high levels of CD103; these may represent recirculating tissue-resident memory cells that could provide pulmonary immune protection. Together, these results identify unique populations of CD4+ T cells with potential to be immune correlates of protection conferred by BCG revaccination.
Collapse
Affiliation(s)
- One B Dintwe
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Cape Town HVTN Immunology Laboratory, Hutchinson Centre Research Institute of South Africa, Cape Town, South Africa
| | | | - Valentin Voillet
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Cape Town HVTN Immunology Laboratory, Hutchinson Centre Research Institute of South Africa, Cape Town, South Africa
| | - John McNevin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Aaron Seese
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Anneta Naidoo
- Cape Town HVTN Immunology Laboratory, Hutchinson Centre Research Institute of South Africa, Cape Town, South Africa
| | - Saleha Omarjee
- Cape Town HVTN Immunology Laboratory, Hutchinson Centre Research Institute of South Africa, Cape Town, South Africa
| | - Linda-Gail Bekker
- The Desmond Tutu HIV Centre, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - James G Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Stephen C De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Evan W Newell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Andrew Fiore-Gartland
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Erica Andersen-Nissen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Cape Town HVTN Immunology Laboratory, Hutchinson Centre Research Institute of South Africa, Cape Town, South Africa.
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
31
|
Østergaard AA, Feddersen S, Barnkob MB, Lynggaard RB, Karstoft ACA, Borup M, Titlestad IL, Jensen TT, Hilberg O, Wejse C, Bjerrum S, Blaabjerg M, Assing K, Johansen IS. Whole-blood culture-derived cytokine combinations for the diagnosis of tuberculosis. Front Immunol 2024; 15:1397941. [PMID: 38933274 PMCID: PMC11199390 DOI: 10.3389/fimmu.2024.1397941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/07/2024] [Indexed: 06/28/2024] Open
Abstract
Introduction The diagnosis of tuberculosis (TB) disease and TB infection (TBI) remains a challenge, and there is a need for non-invasive and blood-based methods to differentiate TB from conditions mimicking TB (CMTB), TBI, and healthy controls (HC). We aimed to determine whether combination of cytokines and established biomarkers could discriminate between 1) TB and CMTB 2) TB and TBI 3) TBI and HC. Methods We used hemoglobin, total white blood cell count, neutrophils, monocytes, C-reactive protein, and ten Meso Scale Discovery analyzed cytokines (interleukin (IL)-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12p70, IL-13, interferon (IFN)-ɣ, and tumor necrosis factor (TNF)-α) in TruCulture whole blood tubes stimulated by lipopolysaccharides (LPS), zymosan (ZYM), anti-CD3/28 (CD3), and unstimulated (Null) to develop three index tests able to differentiate TB from CMTB and TBI, and TBI from HC. Results In 52 persons with CMTB (n=9), TB (n=23), TBI (n=10), and HC (n=10), a combination of cytokines (LPS-IFN-ɣ, ZYM-IFN-ɣ, ZYM-TNF-α, ZYM-IL-1β, LPS-IL-4, and ZYM-IL-6) and neutrophil count could differentiate TB from CMTB with a sensitivity of 52.2% (95% CI: 30.9%-73.4%) and a specificity of 100 % (66.4%-100%). Null- IFN-ɣ, Null-IL-8, CD3-IL-6, CD3-IL-8, CD3-IL-13, and ZYM IL-1b discriminated TB from TBI with a sensitivity of 73.9% (56.5% - 91.3%) and a specificity of 100% (69.2-100). Cytokines and established biomarkers failed to differentiate TBI from HC with ≥ 98% specificity. Discussion Selected cytokines may serve as blood-based add-on tests to detect TB in a low-endemic setting, although these results need to be validated.
Collapse
Affiliation(s)
- Anne Ahrens Østergaard
- Research Unit of Infectious Diseases, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark
| | - Søren Feddersen
- Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
- Clinical Biochemistry, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Mike B. Barnkob
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
| | | | - Amanda Cecilie Annie Karstoft
- Research Unit of Infectious Diseases, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Maria Borup
- Department of Respiratory Medicine, Odense University Hospital, Odense, Denmark
| | - Ingrid Louise Titlestad
- Department of Respiratory Medicine, Odense University Hospital, Odense, Denmark
- Odense Respiratory Research Unit (ODIN), Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | | | - Ole Hilberg
- Department of Medicine, Vejle Hospital, Hospital Lillebælt, Vejle, Denmark
| | - Christian Wejse
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Center for Global Health (GloHAU), Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Stephanie Bjerrum
- Research Unit of Infectious Diseases, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Infectious Diseases, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Morten Blaabjerg
- Research Unit of Neurology, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Neurology, Odense University Hospital, Odense, Denmark
| | - Kristian Assing
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
- Research Unit of Clinical Immunology, Department of Clinical Research, Odense, Denmark
| | - Isik Somuncu Johansen
- Research Unit of Infectious Diseases, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark
| |
Collapse
|
32
|
Rungelrath V, Ahmed M, Hicks L, Miller SM, Ryter KT, Montgomery K, Ettenger G, Riffey A, Abdelwahab WM, Khader SA, Evans JT. Vaccination with Mincle agonist UM-1098 and mycobacterial antigens induces protective Th1 and Th17 responses. NPJ Vaccines 2024; 9:100. [PMID: 38844494 PMCID: PMC11156909 DOI: 10.1038/s41541-024-00897-x] [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: 01/31/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is one of the top infectious killers in the world. The only licensed vaccine against TB, Bacille Calmette-Guérin (BCG), provides variable protection against pulmonary TB, especially in adults. Hence, novel TB vaccine approaches are urgently needed. Both Th1 and Th17 responses are necessary for protection against TB, yet effective adjuvants and vaccine delivery systems for inducing robust Th1 and Th17 immunity are lacking. Herein we describe a synthetic Mincle agonist, UM-1098, and a silica nanoparticle delivery system that drives Th1/Th17 responses to Mtb antigens. Stimulation of human peripheral blood mononuclear cells (hPBMCs) with UM-1098 induced high levels of Th17 polarizing cytokines IL-6, IL-1β, IL-23 as well as IL-12p70, IL-4 and TNF-α in vitro. PBMCs from both C57BL/6 and BALB/c mice responded with a similar cytokine pattern in vitro and in vivo. Importantly, intramuscular (I.M.) vaccination with UM-1098-adjuvanted TB antigen M72 resulted in significantly higher antigen-specific IFN-γ and IL-17A levels in C57BL/6 wt mice than Mincle KO mice. Vaccination of C57BL/6 wt mice with immunodominant Mtb antigens ESAT6/Ag85B or M72 resulted in predominantly Th1 and Th17 responses and induced antigen-specific serum antibodies. Notably, in a virulent Mtb challenge model, vaccination with UM-1098 adjuvanted ESAT6/Ag85B or M72 significantly reduced lung bacterial burden when compared with unvaccinated mice and protection occurred in the absence of pulmonary inflammation. These data demonstrate that the synthetic Mincle agonist UM-1098 induces strong Th1 and Th17 immunity after vaccination with Mtb antigens and provides protection against Mtb infection in mice.
Collapse
Affiliation(s)
- Viktoria Rungelrath
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Mushtaq Ahmed
- Department of Microbiology, University of Chicago, 920 E. 58th St., Chicago, IL, 60637, USA
| | - Linda Hicks
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Shannon M Miller
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Kendal T Ryter
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Kyle Montgomery
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - George Ettenger
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Alexander Riffey
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Walid M Abdelwahab
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Shabaana Abdul Khader
- Department of Microbiology, University of Chicago, 920 E. 58th St., Chicago, IL, 60637, USA
| | - Jay T Evans
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA.
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA.
| |
Collapse
|
33
|
Khanna H, Gupta S, Sheikh Y. Cell-Mediated Immune Response Against Mycobacterium tuberculosis and Its Potential Therapeutic Impact. J Interferon Cytokine Res 2024; 44:244-259. [PMID: 38607324 DOI: 10.1089/jir.2024.0030] [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] [Indexed: 04/13/2024] Open
Abstract
Cell-mediated immune response is critical for Mycobacterium tuberculosis (M.tb) control. Understanding of pathophysiology and role played by different cell mediators is essential for vaccine development and better management of patients with M.tb. A complex array of cytokines and chemokines are involved in the immune response against M.tb; however, their relative contribution in protection remains to be further explored. The purpose of this review is to summarize the current understanding regarding the cytokine and chemokine profiles in M.tb infection in order to assist research in the field to pursue new direction in prevention and control. We have also summarized recent findings on vaccine trials that have been developed and or are under trials that are targeting these molecules.
Collapse
Affiliation(s)
- Harshika Khanna
- Department of Pediatrics, King George's Medical University, Lucknow, India
| | | | - Yasmeen Sheikh
- Department of Pediatrics, King George's Medical University, Lucknow, India
| |
Collapse
|
34
|
Tsareva A, Shelyakin PV, Shagina IA, Myshkin MY, Merzlyak EM, Kriukova VV, Apt AS, Linge IA, Chudakov DM, Britanova OV. Aberrant adaptive immune response underlies genetic susceptibility to tuberculosis. Front Immunol 2024; 15:1380971. [PMID: 38799462 PMCID: PMC11116662 DOI: 10.3389/fimmu.2024.1380971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/11/2024] [Indexed: 05/29/2024] Open
Abstract
Mycobacterium tuberculosis (Mtb) remains a major threat worldwide, although only a fraction of infected individuals develops tuberculosis (TB). TB susceptibility is shaped by multiple genetic factors, and we performed comparative immunological analysis of two mouse strains to uncover relevant mechanisms underlying susceptibility and resistance. C57BL/6 mice are relatively TB-resistant, whereas I/St mice are prone to develop severe TB, partly due to the MHC-II allelic variant that shapes suboptimal CD4+ T cell receptor repertoire. We investigated the repertoires of lung-infiltrating helper T cells and B cells at the progressed stage in both strains. We found that lung CD4+ T cell repertoires of infected C57BL/6 but not I/St mice contained convergent TCR clusters with functionally confirmed Mtb specificity. Transcriptomic analysis revealed a more prominent Th1 signature in C57BL/6, and expression of pro-inflammatory IL-16 in I/St lung-infiltrating helper T cells. The two strains also showed distinct Th2 signatures. Furthermore, the humoral response of I/St mice was delayed, less focused, and dominated by IgG/IgM isotypes, whereas C57BL/6 mice generated more Mtb antigen-focused IgA response. We conclude that the inability of I/St mice to produce a timely and efficient anti-Mtb adaptive immune responses arises from a suboptimal helper T cell landscape that also impacts the humoral response, leading to diffuse inflammation and severe disease.
Collapse
Affiliation(s)
- Anastasiia Tsareva
- Precision Oncology Division, Boston Gene Laboratory, Waltham, MA, United States
| | - Pavel V. Shelyakin
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Abu Dhabi Stem Cells Center, Abu Dhabi, United Arab Emirates
| | - Irina A. Shagina
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Mikhail Yu. Myshkin
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Ekaterina M. Merzlyak
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Valeriia V. Kriukova
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Alexander S. Apt
- Laboratory for Immunogenetics, Central Tuberculosis Research Institute, Moscow, Russia
| | - Irina A. Linge
- Laboratory for Immunogenetics, Central Tuberculosis Research Institute, Moscow, Russia
| | - Dmitriy M. Chudakov
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Abu Dhabi Stem Cells Center, Abu Dhabi, United Arab Emirates
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- Central European Institute of Technology, Masaryk University, Brno, Czechia
| | - Olga V. Britanova
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of Genomics of Adaptive Immunity, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| |
Collapse
|
35
|
Kim ES, Islam J, Lee HJ, Seong SY, Youn JI, Kwon BS, Kim SJ, Lee JH. Myeloid-derived suppressor cells in pleural effusion as a diagnostic marker for early discrimination of pulmonary tuberculosis from pneumonia. Front Immunol 2024; 15:1390327. [PMID: 38742106 PMCID: PMC11089152 DOI: 10.3389/fimmu.2024.1390327] [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: 02/23/2024] [Accepted: 04/17/2024] [Indexed: 05/16/2024] Open
Abstract
INTRODUCTION Tuberculous pleural effusion (TPE) stands as one of the primary forms of extrapulmonary tuberculosis (TB) and frequently manifests in regions with a high prevalence of TB, consequently being a notable cause of pleural effusion in such areas. However, the differentiation between TPE and parapneumonic pleural effusion (PPE) presents diagnostic complexities. This study aimed to evaluate the potential of myeloid-derived suppressor cells (MDSCs) in the pleural fluid as a potential diagnostic marker for distinguishing between TPE and PPE. METHODS Adult patients, aged 18 years or older, who presented to the emergency room of a tertiary referral hospital and received a first-time diagnosis of pleural effusion, were prospectively enrolled in the study. Various immune cell populations, including T cells, B cells, natural killer (NK) cells, and MDSCs, were analyzed in both pleural fluid and peripheral blood samples. RESULTS In pleural fluid, the frequency of lymphocytes, including T, B, and NK cells, was notably higher in TPE compared to PPE. Conversely, the frequency of polymorphonuclear (PMN)-MDSCs was significantly higher in PPE. Notably, compared to traditional markers such as the neutrophil-to-lymphocyte ratio and adenosine deaminase level, the frequency of PMN-MDSCs emerged as a more effective discriminator between PPE and TPE. PMN-MDSCs demonstrated superior positive and negative predictive values and exhibited a higher area under the curve in the receiver operating characteristic curve analysis. PMN-MDSCs in pleural effusion increased the levels of reactive oxygen species and suppressed the production of interferon-gamma from T cells following nonspecific stimulation. These findings suggest that MDSC-mediated immune suppression may contribute to the pathology of both TPE and PPE. DISCUSSION The frequency of PMN-MDSCs in pleural fluid is a clinically useful indicator for distinguishing between TPE and PPE.
Collapse
Affiliation(s)
- Eun Sun Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
- Hospital Medicine Center, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jahirul Islam
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hee-Jae Lee
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Republic of Korea
| | - Seung-Yong Seong
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Shaperon Inc., Republic of Korea
| | - Je-In Youn
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, Republic of Korea
- SG Medical, 3-11, Ogeum-ro 13-gil, Songpa-gu, Seoul, Republic of Korea
| | - Byoung Soo Kwon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
- Division of Pulmonary and Critical Care Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Se Joong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Sheikh Khalifa Specialty Hospital, Ras Al Khaimah, United Arab Emirates
| | - Jae-Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
- Division of Pulmonary and Critical Care Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| |
Collapse
|
36
|
Yasuda I, Saludar NRD, Sayo AR, Suzuki S, Yokoyama A, Ozeki Y, Kobayashi H, Nishiyama A, Matsumoto S, Cox SE, Tanaka T, Yamashita Y. Evaluation of cytokine profiles related to Mycobacterium tuberculosis latent antigens using a whole-blood assay in the Philippines. Front Immunol 2024; 15:1330796. [PMID: 38665909 PMCID: PMC11044679 DOI: 10.3389/fimmu.2024.1330796] [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: 10/31/2023] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Introduction There is no useful method to discriminate between latent tuberculosis infection (LTBI) and active pulmonary tuberculosis (PTB). This study aimed to investigate the potential of cytokine profiles to discriminate between LTBI and active PTB using whole-blood stimulation with Mycobacterium tuberculosis (MTB) antigens, including latency-associated antigens. Materials and methods Patients with active PTB, household contacts of active PTB patients and community exposure subjects were recruited in Manila, the Philippines. Peripheral blood was collected from the participants and used for whole-blood stimulation (WBS) with either the early secretory antigenic target and the 10-kDa culture filtrate protein (ESAT-6/CFP-10), Rv3879c or latency-associated MTB antigens, including mycobacterial DNA-binding protein 1 (MDP-1), α-crystallin (Acr) and heparin-binding hemagglutinin (HBHA). Multiple cytokine concentrations were analyzed using the Bio-Plex™ multiplex cytokine assay. Results A total of 78 participants consisting of 15 active PTB patients, 48 household contacts and 15 community exposure subjects were eligible. The MDP-1-specific IFN-γ level in the active PTB group was significantly lower than that in the household contact group (p < 0.001) and the community exposure group (p < 0.001). The Acr-specific TNF-α and IL-10 levels in the active PTB group were significantly higher than those in the household contact (TNF-α; p = 0.001, IL-10; p = 0.001) and community exposure (TNF-α; p < 0.001, IL-10; p = 0.01) groups. However, there was no significant difference in the ESAT-6/CFP-10-specific IFN-γ levels among the groups. Conclusion The patterns of cytokine profiles induced by latency-associated MTB antigens using WBS have the potential to discriminate between LTBI and active PTB. In particular, combinations of IFN-γ and MDP-1, TNF-α and Acr, and IL-10 and Acr are promising. This study provides the first demonstration of the utility of MDP-1-specific cytokine responses in WBS.
Collapse
Affiliation(s)
- Ikkoh Yasuda
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
- Department of General Internal Medicine and Clinical Infectious Diseases, Fukushima Medical University, Fukushima, Japan
- Department of General Internal Medicine and Infectious Diseases, Kita-Fukushima Medical Center, Fukushima, Japan
| | | | | | - Shuichi Suzuki
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| | - Akira Yokoyama
- Department of Bacteriology, Niigata University Graduate School of Medicine, Niigata, Japan
- Department of Respiratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Yuriko Ozeki
- Department of Bacteriology, Niigata University Graduate School of Medicine, Niigata, Japan
| | - Haruka Kobayashi
- Department of Bacteriology, Niigata University Graduate School of Medicine, Niigata, Japan
| | - Akihito Nishiyama
- Department of Bacteriology, Niigata University Graduate School of Medicine, Niigata, Japan
| | - Sohkichi Matsumoto
- Department of Bacteriology, Niigata University Graduate School of Medicine, Niigata, Japan
- Department of Medical Microbiology, Universitas Airlangga, Faculty of Medicine, Surabaya, Indonesia
- Division of Research Aids, Hokkaido University Institute for Vaccine Research & Development, Sapporo, Japan
| | - Sharon E. Cox
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Takeshi Tanaka
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
- Department of Infectious Diseases, Nagasaki University Hospital, Nagasaki, Japan
- Infection Control and Education Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Yoshiro Yamashita
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
- Department of Respiratory Medicine, Shunkaikai Inoue Hospital, Nagasaki, Japan
| |
Collapse
|
37
|
Rees CE, Swift BM, Haldar P. State-of-the-art detection of Mycobacterium tuberculosis in blood during tuberculosis infection using phage technology. Int J Infect Dis 2024; 141S:106991. [PMID: 38447755 DOI: 10.1016/j.ijid.2024.106991] [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: 02/01/2024] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/08/2024] Open
Abstract
Tuberculosis (TB), an aerosol-transmitted infection caused by Mycobacterium tuberculosis (Mtb), remains the commonest cause of death globally, from an infectious bacterial disease. Nine years on from the launch of the World Health Organization (WHO)'s END-TB strategy, disease incidence rates are stubbornly unchanged [1]. While this represents, in part, a reversal of improving trends caused by the COVID-19 pandemic, it also reflects the fragility and inadequacy of healthcare systems to sustain TB control [2]. Although multifactorial, a key reason for this is the ineffectiveness of existing clinical tools to meet the two key objectives of the END-TB strategy-(i) early diagnosis and treatment of TB disease (to limit onward transmission); and (ii) disease prevention through screening for asymptomatic TB infection (TBI). Meeting both objectives will rely on the development of new biomarkers with high accuracy, but the global nature of the TB problem also requires that new tests are rapid, low cost and can be measured in patients by sampling from universally accessible sites. In this review, we will present the accumulating evidence for circulating Mtb in both TB disease and asymptomatic TBI and discuss the potential utility of novel bacteriophage-based technology for blood-based detection of Mtb.
Collapse
Affiliation(s)
| | - Benjamin Mc Swift
- Royal Veterinary College, Department of Pathobiology and Population Sciences, Herts, UK
| | - Pranabashis Haldar
- NIHR Leicester Biomedical Research Centre, Department of Respiratory Sciences, University of Leicester, Leicester, UK.
| |
Collapse
|
38
|
Kwon KW, Choi HG, Choi HH, Choi E, Kim H, Kim HJ, Shin SJ. Immunogenicity and protective efficacy of RipA, a peptidoglycan hydrolase, against Mycobacterium tuberculosis Beijing outbreak strains. Vaccine 2024; 42:1941-1952. [PMID: 38368223 DOI: 10.1016/j.vaccine.2024.02.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 12/11/2023] [Accepted: 02/11/2024] [Indexed: 02/19/2024]
Abstract
Given that individuals with latent tuberculosis (TB) infection represent the major reservoir of TB infection, latency-associated antigens may be promising options for development of improved multi-antigenic TB subunit vaccine. Thus, we selected RipA, a peptidoglycan hydrolase required for efficient cell division of Mycobacterium tuberculosis (Mtb), as vaccine candidate. We found that RipA elicited activation of dendritic cells (DCs) by induction of phenotypic maturation, increased production of inflammatory cytokines, and prompt stimulation of MAPK and NF-κB signaling pathways. In addition, RipA-treated DCs promoted Th1-polarzied immune responses of naïve CD4+ T cells with increased proliferation and activated T cells from Mtb-infected mice, which conferred enhanced control of mycobacterial growth inside macrophages. Moreover, mice immunized with RipA formulated in GLA-SE adjuvant displayed remarkable generation of Ag-specific polyfunctional CD4+ T cells in both lung and spleen. Following an either conventional or ultra-low dose aerosol challenges with 2 Mtb Beijing clinical strains, RipA/GLA-SE-immunization was not inferior to BCG by mediating protection as single Ag. Collectively, our findings highlighted that RipA could be a novel candidate as a component of multi-antigenic TB subunit vaccines.
Collapse
Affiliation(s)
- Kee Woong Kwon
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea; Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul 03722, South Korea; Department of Microbiology, College of Medicine, Gyeongsang National University, Jinju 52727, South Korea
| | - Han-Gyu Choi
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, South Korea
| | - Hong-Hee Choi
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Eunsol Choi
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Hagyu Kim
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Hwa-Jung Kim
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea; Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul 03722, South Korea.
| |
Collapse
|
39
|
Zeng L, Ma X, Qu M, Tang M, Li H, Lei C, Ji J, Li H. Immunogenicity and protective efficacy of Ag85A and truncation of PstS1 fusion protein vaccines against tuberculosis. Heliyon 2024; 10:e27034. [PMID: 38463854 PMCID: PMC10920368 DOI: 10.1016/j.heliyon.2024.e27034] [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: 04/06/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/12/2024] Open
Abstract
Tuberculosis (TB) is an important public health problem, and the One Health approach is essential for controlling zoonotic tuberculosis. Therefore, a rationally designed and more effective TB vaccine is urgently needed. To enhance vaccine efficacy, it is important to design vaccine candidates that stimulate both cellular and humoral immunity against TB. In this study, we fused the secreted protein Ag85A as the T cell antigen with truncated forms of the mycobacterial cell wall protein PstS1 with B cell epitopes to generate vaccine candidates, Ag85A-tnPstS1 (AP1, AP2, and AP3), and tested their immunogenicity and protective efficacy in mice. The three vaccine candidates induced a significant increase in the levels of T cell-related cytokines such as IFN-γ and IL-17, and AP1 and AP2 can induce more balanced Th1/Th2 responses than AP3. Strong humoral immune responses were also observed in which the production of IgG antibodies including its subclasses IgG1, IgG2c, and IgG3 was tremendously stimulated. AP1 and AP2 induced early antibody responses and more IgG3 isotype antibodies than AP3. Importantly, the mice immunised with the subunit vaccine candidates, particularly AP1 and AP2, had lower bacterial burdens than the control mice. Moreover, the serum from immunised mice can enhance phagocytosis and phagosome-lysosome fusion in macrophages, which can help to eradicate intracellular bacteria. These results indicate that the subunit vaccines Ag85A-tnPstS1 can be promising vaccine candidates for tuberculosis prevention.
Collapse
Affiliation(s)
- Lingyuan Zeng
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Xiuling Ma
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Mengjin Qu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Minghui Tang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Huoming Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Chengrui Lei
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Jiahong Ji
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Hao Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| |
Collapse
|
40
|
Krmeská V, Shen L, Nylén S, Wowk PF, Rothfuchs AG. BCG infection dose guides dendritic cell migration and T cell priming in the draining lymph node. Scand J Immunol 2024; 99:e13342. [PMID: 38441294 DOI: 10.1111/sji.13342] [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: 05/26/2023] [Revised: 10/11/2023] [Accepted: 11/09/2023] [Indexed: 03/07/2024]
Abstract
In contrast to delayed-type hypersensitivity (DTH) and other hallmark reactions of cell-mediated immunity that correlate with vaccine-mediated protection against Mycobacterium tuberculosis, the contribution of vaccine dose on responses that emerge early after infection in the skin with Bacille Calmette-Guérin (BCG) is not well understood. We used a mouse model of BCG skin infection to study the effect of BCG dose on the relocation of skin Dendritic cells (DCs) to draining lymph node (DLN). Mycobacterium antigen 85B-specific CD4+ P25 T cell-receptor transgenic (P25 TCRTg) cells were used to probe priming to BCG in DLN. DC migration and T cell priming were studied across BCG inocula that varied up to 100-fold (104 to 106 Colony-forming units-CFUs). In line with earlier results in guinea pigs, DTH reaction in our model correlated with BCG dose. Importantly, priming of P25 TCRTg cells in DLN also escalated in a dose-dependent manner, peaking at day 6 after infection. Similar dose-escalation effects were seen for DC migration from infected skin and the accompanying transport of BCG to the DLN. BCG-triggered upregulation of co-stimulatory molecules on migratory DCs was restricted to the first 24 hour after infection and was independent of BCG dose over a 10-fold range (105 to 106 CFUs). The dose seemed to be a determinant of the number of total skin DCs that move to the DLN. In summary, our results support the use of higher BCG doses to detect robust DC migration and T cell priming.
Collapse
Affiliation(s)
- Veronika Krmeská
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Lei Shen
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Susanne Nylén
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Pryscilla Fanini Wowk
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (ICC/Fiocruz-PR), Curitiba, Brazil
| | | |
Collapse
|
41
|
Ruelas Castillo J, Neupane P, Karanika S, Krug S, Quijada D, Garcia A, Ayeh S, Yilma A, Costa DL, Sher A, Fotouhi N, Serbina N, Karakousis PC. The heme oxygenase-1 metalloporphyrin inhibitor stannsoporfin enhances the bactericidal activity of a novel regimen for multidrug-resistant tuberculosis in a murine model. Antimicrob Agents Chemother 2024; 68:e0104323. [PMID: 38132181 PMCID: PMC10848751 DOI: 10.1128/aac.01043-23] [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: 08/10/2023] [Accepted: 11/17/2023] [Indexed: 12/23/2023] Open
Abstract
Multidrug-resistant (MDR) Mycobacterium tuberculosis (Mtb) poses significant challenges to global tuberculosis (TB) control efforts. Host-directed therapies (HDTs) offer a novel approach to TB treatment by enhancing immune-mediated clearance of Mtb. Prior preclinical studies found that the inhibition of heme oxygenase-1 (HO-1), an enzyme involved in heme metabolism, with tin-protoporphyrin IX (SnPP) significantly reduced mouse lung bacillary burden when co-administered with the first-line antitubercular regimen. Here, we evaluated the adjunctive HDT activity of a novel HO-1 inhibitor, stannsoporfin (SnMP), in combination with a novel MDR-TB regimen comprising a next-generation diarylquinoline, TBAJ-876 (S), pretomanid (Pa), and a new oxazolidinone, TBI-223 (O) (collectively, SPaO), in Mtb-infected BALB/c mice. After 4 weeks of treatment, SPaO + SnMP 5mg/kg reduced mean lung bacillary burden by an additional 0.69 log10 (P = 0.01) relative to SPaO alone. As early as 2 weeks post-treatment initiation, SnMP adjunctive therapy differentially altered the expression of pro-inflammatory cytokine genes and CD38, a marker of M1 macrophages. Next, we evaluated the sterilizing potential of SnMP adjunctive therapy in a mouse model of microbiological relapse. After 6 weeks of treatment, SPaO + SnMP 10mg/kg reduced lung bacterial burdens to 0.71 ± 0.23 log10 colony-forming units (CFUs), a 0.78 log-fold greater decrease in lung CFU compared to SpaO alone (P = 0.005). However, adjunctive SnMP did not reduce microbiological relapse rates after 5 or 6 weeks of treatment. SnMP was well tolerated and did not significantly alter gross or histological lung pathology. SnMP is a promising HDT candidate requiring further study in combination with regimens for drug-resistant TB.
Collapse
Affiliation(s)
- Jennie Ruelas Castillo
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Pranita Neupane
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Styliani Karanika
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Stefanie Krug
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Darla Quijada
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Garcia
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Samuel Ayeh
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Addis Yilma
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Diego L. Costa
- Departmento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | - Petros C. Karakousis
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
42
|
Ghoshal A, Verma A, Bhaskar A, Dwivedi VP. The uncharted territory of host-pathogen interaction in tuberculosis. Front Immunol 2024; 15:1339467. [PMID: 38312835 PMCID: PMC10834760 DOI: 10.3389/fimmu.2024.1339467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/03/2024] [Indexed: 02/06/2024] Open
Abstract
Mycobacterium tuberculosis (M.tb) effectively manipulates the host processes to establish the deadly respiratory disease, Tuberculosis (TB). M.tb has developed key mechanisms to disrupt the host cell health to combat immune responses and replicate efficaciously. M.tb antigens such as ESAT-6, 19kDa lipoprotein, Hip1, and Hsp70 destroy the integrity of cell organelles (Mitochondria, Endoplasmic Reticulum, Nucleus, Phagosomes) or delay innate/adaptive cell responses. This is followed by the induction of cellular stress responses in the host. Such cells can either undergo various cell death processes such as apoptosis or necrosis, or mount effective immune responses to clear the invading pathogen. Further, to combat the infection progression, the host secretes extracellular vesicles such as exosomes to initiate immune signaling. The exosomes can contain M.tb as well as host cell-derived peptides that can act as a double-edged sword in the immune signaling event. The host-symbiont microbiota produces various metabolites that are beneficial for maintaining healthy tissue microenvironment. In juxtaposition to the above-mentioned mechanisms, M.tb dysregulates the gut and respiratory microbiome to support its replication and dissemination process. The above-mentioned interconnected host cellular processes of Immunometabolism, Cellular stress, Host Microbiome, and Extracellular vesicles are less explored in the realm of exploration of novel Host-directed therapies for TB. Therefore, this review highlights the intertwined host cellular processes to control M.tb survival and showcases the important factors that can be targeted for designing efficacious therapy.
Collapse
Affiliation(s)
| | | | | | - Ved Prakash Dwivedi
- Immunobiology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| |
Collapse
|
43
|
McIntyre S, Warner J, Rush C, Vanderven HA. Antibodies as clinical tools for tuberculosis. Front Immunol 2023; 14:1278947. [PMID: 38162666 PMCID: PMC10755875 DOI: 10.3389/fimmu.2023.1278947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Tuberculosis (TB) is a leading cause of morbidity and mortality worldwide. Global research efforts to improve TB control are hindered by insufficient understanding of the role that antibodies play in protective immunity and pathogenesis. This impacts knowledge of rational and optimal vaccine design, appropriate diagnostic biomarkers, and development of therapeutics. Traditional approaches for the prevention and diagnosis of TB may be less efficacious in high prevalence, remote, and resource-poor settings. An improved understanding of the immune response to the causative agent of TB, Mycobacterium tuberculosis (Mtb), will be crucial for developing better vaccines, therapeutics, and diagnostics. While memory CD4+ T cells and cells and cytokine interferon gamma (IFN-g) have been the main identified correlates of protection in TB, mounting evidence suggests that other types of immunity may also have important roles. TB serology has identified antibodies and functional characteristics that may help diagnose Mtb infection and distinguish between different TB disease states. To date, no serological tests meet the World Health Organization (WHO) requirements for TB diagnosis, but multiplex assays show promise for improving the sensitivity and specificity of TB serodiagnosis. Monoclonal antibody (mAb) therapies and serum passive infusion studies in murine models of TB have also demonstrated some protective outcomes. However, animal models that better reflect the human immune response to Mtb are necessary to fully assess the clinical utility of antibody-based TB prophylactics and therapeutics. Candidate TB vaccines are not designed to elicit an Mtb-specific antibody response, but evidence suggests BCG and novel TB vaccines may induce protective Mtb antibodies. The potential of the humoral immune response in TB monitoring and control is being investigated and these studies provide important insight into the functional role of antibody-mediated immunity against TB. In this review, we describe the current state of development of antibody-based clinical tools for TB, with a focus on diagnostic, therapeutic, and vaccine-based applications.
Collapse
Affiliation(s)
- Sophie McIntyre
- Biomedical Sciences and Molecular Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Douglas, QLD, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Douglas, QLD, Australia
| | - Jeffrey Warner
- Biomedical Sciences and Molecular Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Douglas, QLD, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Douglas, QLD, Australia
| | - Catherine Rush
- Biomedical Sciences and Molecular Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Douglas, QLD, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Douglas, QLD, Australia
| | - Hillary A. Vanderven
- Biomedical Sciences and Molecular Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Douglas, QLD, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Douglas, QLD, Australia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
44
|
García-Bengoa M, Vergara EJ, Tran AC, Bossi L, Cooper AM, Pearl JE, Mussá T, von Köckritz-Blickwede M, Singh M, Reljic R. Immunogenicity of PE18, PE31, and PPE26 proteins from Mycobacterium tuberculosis in humans and mice. Front Immunol 2023; 14:1307429. [PMID: 38124744 PMCID: PMC10730732 DOI: 10.3389/fimmu.2023.1307429] [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: 10/04/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
Introduction The large family of PE and PPE proteins accounts for as much as 10% of the genome of Mycobacterium tuberculosis. In this study, we explored the immunogenicity of three proteins from this family, PE18, PE31, and PPE26, in humans and mice. Methods The investigation involved analyzing the immunoreactivity of the selected proteins using sera from TB patients, IGRA-positive household contacts, and IGRA-negative BCG vaccinated healthy donors from the TB endemic country Mozambique. Antigen-recall responses were examined in PBMC from these groups, including the evaluation of cellular responses in healthy unexposed individuals. Moreover, systemic priming and intranasal boosting with each protein, combined with the Quil-A adjuvant, were conducted in mice. Results We found that all three proteins are immunoreactive with sera from TB patients, IGRA-positive household contacts, and IGRA-negative BCG vaccinated healthy controls. Likewise, antigen-recall responses were induced in PBMC from all groups, and the proteins stimulated proliferation of peripheral blood mononuclear cells from healthy unexposed individuals. In mice, all three antigens induced IgG antibody responses in sera and predominantly IgG, rather than IgA, responses in bronchoalveolar lavage. Additionally, CD4+ and CD8+ effector memory T cell responses were observed in the spleen, with PE18 demonstrating the ability to induce tissue-resident memory T cells in the lungs. Discussion Having demonstrated immunogenicity in both humans and mice, the protective capacity of these antigens was evaluated by challenging immunized mice with low-dose aerosol of Mycobacterium tuberculosis H37Rv. The in vitro Mycobacterial Growth Inhibition Assay (MGIA) and assessment of viable bacteria in the lung did not demonstrate any ability of the vaccination protocol to reduce bacterial growth. We therefore concluded that these three specific PE/PPE proteins, while immunogenic in both humans and mice, were unable to confer protective immunity under these conditions.
Collapse
Affiliation(s)
- María García-Bengoa
- Institute of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Center for Emerging Infections and Zoonosis (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
- LIONEX Diagnostics and Therapeutics GmbH, Braunschweig, Germany
| | - Emil Joseph Vergara
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| | - Andy C. Tran
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| | - Lorenzo Bossi
- Immunxperts SA, a Q² Solutions Company, Gosselies, Belgium
| | - Andrea M. Cooper
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - John E. Pearl
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Tufária Mussá
- Department of Microbiology, Faculty of Medicine, Eduardo Mondlane University, Maputo, Mozambique
| | - Maren von Köckritz-Blickwede
- Institute of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Center for Emerging Infections and Zoonosis (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Mahavir Singh
- LIONEX Diagnostics and Therapeutics GmbH, Braunschweig, Germany
| | - Rajko Reljic
- Institute for Infection and Immunity, St. George’s University of London, London, United Kingdom
| |
Collapse
|
45
|
Lai R, Williams T, Rakib T, Lee J, Behar SM. Heterogeneity in lung macrophage control of Mycobacterium tuberculosis is determined by T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.29.569283. [PMID: 38076803 PMCID: PMC10705395 DOI: 10.1101/2023.11.29.569283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Following Mycobacterium tuberculosis infection, alveolar macrophages are initially infected but ineffectively restrict bacterial replication. The distribution of M. tuberculosis among different cell types in the lung changes with the onset of T cell immunity when the dominant infected cellular niche shifts from alveolar to monocyte-derived macrophages (MDM). We hypothesize that changes in bacterial distribution among different cell types is driven by differences in T cell recognition of infected cells and their subsequent activation of antimicrobial effector mechanisms. We show that CD4 and CD8 T cells efficiently eliminate M. tuberculosis infection in alveolar macrophages, but they have less impact on suppressing infection in MDM, which may be a bacterial niche. Importantly, CD4 T cell responses enhance MDM recruitment to the lung. Thus, the outcome of infection depends on the interaction between the T cell subset and the infected cell; both contribute to the resolution and persistence of the infection.
Collapse
Affiliation(s)
- Rocky Lai
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Travis Williams
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Tasfia Rakib
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Jinhee Lee
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Samuel M. Behar
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
46
|
Li Z, Peng C, Chen L, Wang P, Wang F. Construction and Immunogenicity Evaluation of Recombinant Bacillus subtilis Expressing HA1 Protein of H9N2 Avian Influenza Virus. Curr Microbiol 2023; 81:25. [PMID: 38040977 DOI: 10.1007/s00284-023-03548-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/31/2023] [Indexed: 12/03/2023]
Abstract
The H9N2 subtype of the avian influenza virus (AIV) is one of the main subtypes of low pathogenic AIV, and it seriously affects the poultry breeding industry. Currently, vaccination is still one of China's main strategies for controlling H9N2 avian influenza. In this study, we selected MW548848.1 on the current popular main branch h9.4.2.5 as the reference strain, and we optimized the amino acid sequence of HA1 to make it suitable for expression in Bacillus subtilis. The B. subtilis expression vector showed good safety and stress resistance; therefore, this study constructed a recombinant B. subtilis expressing H9N2 HA1 protein and evaluated its immunogenicity in mice. The following results were obtained: the sIgA level of HA1 protein in small intestine fluid and the IgG level of PHT43-HA1/B. subtilis in serum were significantly improved (P < 0.01); PHT43-HA1/B. subtilis can cause a special immune response in mice; and cytokine detection interferon-gamma (IFN-γ) (P < 0.05) and Interleukin 2 (IL-2) (P < 0.01) expressions significantly increased. Additionally, the study found that PHT43-HA1/B. subtilis can alleviate the attack of H9N2 AIV in the spleen, lungs, and small intestine of mice. This study was the first to use an oral recombinant B. subtilis-HA1 vaccine candidate, and it provides theoretical data and technical reference for the creation of a new live vector vaccine against H9N2 AIV.
Collapse
Affiliation(s)
- Zixuan Li
- Department of Veterinary Public Health, College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, Shandong, China
| | - Chong Peng
- Department of Veterinary Public Health, College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, Shandong, China
| | - Lijun Chen
- Department of Veterinary Public Health, College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, Shandong, China
| | - Peng Wang
- Department of Veterinary Public Health, College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| | - Fangkun Wang
- Department of Veterinary Public Health, College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China.
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, Shandong, China.
| |
Collapse
|
47
|
Khan S, Ahmad F, Ansari MI, Ashfaque M, Islam MH, Khubaib M. Toxin-Antitoxin system of Mycobacterium tuberculosis: Roles beyond stress sensor and growth regulator. Tuberculosis (Edinb) 2023; 143:102395. [PMID: 37722233 DOI: 10.1016/j.tube.2023.102395] [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: 04/14/2023] [Revised: 07/15/2023] [Accepted: 08/10/2023] [Indexed: 09/20/2023]
Abstract
The advent of effective drug regimen and BCG vaccine has significantly decreased the rate of morbidity and mortality of TB. However, lengthy treatment and slower recovery rate, as well as reactivation of the disease with the emergence of multi-drug, extensively-drug, and totally-drug resistance strains, pose a serious concern. The complexities associated are due to the highly evolved and complex nature of the bacterium itself. One of the unique features of Mycobacterium tuberculosis [M.tb] is that it has undergone reductive evolution while maintaining and amplified a few gene families. One of the critical gene family involved in the virulence and pathogenesis is the Toxin-Antitoxin system. These families are believed to harbor virulence signature and are strongly associated with various stress adaptations and pathogenesis. The M.tb TA systems are linked with growth regulation machinery during various environmental stresses. The genes of TA systems are differentially expressed in the host during an active infection, oxidative stress, low pH stress, and starvation, which essentially indicate their role beyond growth regulators. Here in this review, we have discussed different roles of TA gene families in various stresses and their prospective role at the host-pathogen interface, which could be exploited to understand the M.tb associated pathomechanisms better and further designing the new strategies against the pathogen.
Collapse
Affiliation(s)
- Saima Khan
- Department of Biosciences, Integral University, Lucknow, India
| | - Firoz Ahmad
- Department of Biosciences, Integral University, Lucknow, India
| | | | | | | | - Mohd Khubaib
- Department of Biosciences, Integral University, Lucknow, India.
| |
Collapse
|
48
|
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] [Download PDF] [Figures] [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
|
49
|
Castillo JR, Neupane P, Karanika S, Krug S, Quijada D, Garcia A, Ayeh S, Yilma A, Costa DL, Sher A, Fotouhi N, Serbina N, Karakousis PC. The heme oxygenase-1 metalloporphyrin inhibitor stannsoporfin enhances the bactericidal activity of a novel regimen for multidrug-resistant tuberculosis in a murine model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.09.552716. [PMID: 37609351 PMCID: PMC10441415 DOI: 10.1101/2023.08.09.552716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Multidrug-resistant (MDR) Mycobacterium tuberculosis (Mtb) poses significant challenges to global tuberculosis (TB) control efforts. Host-directed therapies (HDT) offer a novel approach for TB treatment by enhancing immune-mediated clearance of Mtb. Prior preclinical studies found that inhibition of heme oxygenase-1 (HO-1), an enzyme involved in heme metabolism, with tin-protoporphyrin IX (SnPP) significantly reduced mouse lung bacillary burden when co-administered with the first-line antitubercular regimen. Here we evaluated the adjunctive HDT activity of a novel HO-1 inhibitor, stannsoporfin (SnMP), in combination with a novel MDR-TB regimen comprising a next-generation diarylquinoline, TBAJ-876 (S), pretomanid (Pa), and a new oxazolidinone, TBI-223 (O) (collectively, SPaO) in Mtb-infected BALB/c mice. After 4 weeks of treatment, SPaO + SnMP 5 mg/kg reduced mean lung bacillary burden by an additional 0.69 log10 (P=0.01) relative to SPaO alone. As early as 2 weeks post-treatment initiation, SnMP adjunctive therapy differentially altered the expression of pro-inflammatory cytokine genes, and CD38, a marker of M1 macrophages. Next, we evaluated the sterilizing potential of SnMP adjunctive therapy in a mouse model of microbiological relapse. After 6 weeks of treatment, SPaO + SnMP 10 mg/kg reduced lung bacterial burdens to 0.71 ± 0.23 log10 CFU, a 0.78 log-fold greater decrease in lung CFU compared to SpaO alone (P=0.005). However, adjunctive SnMP did not reduce microbiological relapse rates after 5 or 6 weeks of treatment. SnMP was well tolerated and did not significantly alter gross or histological lung pathology. SnMP is a promising HDT candidate requiring further study in combination with regimens for drug-resistant TB.
Collapse
Affiliation(s)
- Jennie Ruelas Castillo
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pranita Neupane
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Styliani Karanika
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stefanie Krug
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Darla Quijada
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew Garcia
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Samuel Ayeh
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Addis Yilma
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Diego L. Costa
- Departmento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Petros C. Karakousis
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
50
|
Fan X, Zhao X, Wang R, Li M, Luan X, Wang R, Wan K, Liu H. A novel multistage antigens ERA005f confer protection against Mycobacterium tuberculosis by driving Th-1 and Th-17 type T cell immune responses. Front Immunol 2023; 14:1276887. [PMID: 38022539 PMCID: PMC10662081 DOI: 10.3389/fimmu.2023.1276887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Tuberculosis (TB) is a major threat to human health. In 2021, TB was the second leading cause of death after COVID-19 among infectious diseases. The Bacillus Calmette-Guérin vaccine (BCG), the only licensed TB vaccine, is ineffective against adult TB. Therefore, there is an urgent need to develop new effective vaccines. Methods In this study, we developed a novel multistage subunit vaccine (ERA005f) comprising various proteins expressed in metabolic states, based on three immunodominant antigens (ESAT-6, Rv2628, and Ag85B). We utilized the E. coli prokaryotic expression system to express ERA005f and subsequently purified the protein using nickel affinity chromatography and anion exchange. Immunogenicity and protective efficacy of ERA005f and ERA005m were evaluated in BALB/c mice. Results ERA005f was consistently expressed as an inclusion body in a prokaryotic expression system, and a highly pure form of the protein was successfully obtained. Both ERA005f and ERA005m significantly improved IgG titers in the serum. In addition, mice immunized with ERA005f and ERA005m generated higher titers of antigen-specific IgG2a than the other groups. Elispot results showed that, compared with other groups, ERA005f increased the numbers of IFN-γ-secreting and IL-4-secreting T cells, especially the number of IFN-γ-secreting T cells. Meanwhile, ERA005f induced a higher number of IFN-γ+ T lymphocytes than ERA005m did. In addition, ERA005f improved the expression of cytokines, including IFN-γ, IL-12p70, TNF-α, IL-17, and GM-CSF and so on. Importantly, both ERA005f and ERA005m significantly inhibited the growth of Mtb. Conclusion The novel multistage antigen ERA005f elicited a strong antigen-specific humoral response and Th-1 and Th-17 cell-mediated immunity in mice. Meanwhile, it can effectively inhibit H37Rv growth in vitro, and represents a correlate of protection in vivo, indicating that ERA005f may exhibit excellent protective efficacy against Mycobacterium tuberculosis H37Rv infection. Our study suggests that ERA005f has the potential to be a promising multistage tuberculosis vaccine candidate.
Collapse
Affiliation(s)
- Xueting Fan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiuqin Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ruibai Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Machao Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiuli Luan
- Department of Tuberculosis, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Ruihuan Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Kanglin Wan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Haican Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|