1
|
Fang C, He X, Tang F, Wang Z, Pan C, Zhang Q, Wu J, Wang Q, Liu D, Zhang Y. Where lung cancer and tuberculosis intersect: recent advances. Front Immunol 2025; 16:1561719. [PMID: 40242762 PMCID: PMC11999974 DOI: 10.3389/fimmu.2025.1561719] [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: 01/16/2025] [Accepted: 03/12/2025] [Indexed: 04/18/2025] Open
Abstract
Lung cancer (LC) and tuberculosis (TB) represent two major global public health issues. Prior evidence has suggested a link between TB infection and an increased risk of LC. As advancements in LC treatment have led to extended survival rates for LC patients, the co-occurrence of TB and LC has grown more prevalent and poses novel clinical challenges. The intricate molecular mechanisms connecting TB and LC are closely intertwined and many issues remain to be addressed. This review focuses on resemblance between the immunosuppression in tumor and granuloma microenvironments, exploring immunometabolism, cell plasticity, inflammatory signaling pathways, microbiomics, and up-to-date information derived from spatial multi-omics between TB and LC. Furthermore, we outline immunization-related molecular mechanisms underlying these two diseases and propose future research directions. By discussing recent advances and potential targets, this review aims to establish a foundation for developing future therapeutic strategies targeting LC with concurrent TB infection.
Collapse
Affiliation(s)
- Chunju Fang
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Xuanlu He
- School of Clinical Medicine, Zunyi Medical University, Zunyi, China
| | - Fei Tang
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Zi Wang
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Cong Pan
- School of Biological Sciences, Guizhou Education University, Guiyang, China
- Translational Medicine Research Center, eBond Pharmaceutical Technology Co., Ltd., Chengdu, China
| | - Qi Zhang
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Jing Wu
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Qinglan Wang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Daishun Liu
- Department of Respiratory and Critical Care Medicine, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Yu Zhang
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
- National Health Commission Key Laboratory of Pulmonary Immune-Related Diseases, Guizhou Provincial People’s Hospital, Guiyang, China
| |
Collapse
|
2
|
Bohórquez JA, Jagannath C, Xu H, Wang X, Yi G. T Cell Responses during Human Immunodeficiency Virus/ Mycobacterium tuberculosis Coinfection. Vaccines (Basel) 2024; 12:901. [PMID: 39204027 PMCID: PMC11358969 DOI: 10.3390/vaccines12080901] [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: 07/12/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
Coinfection with Mycobacterium tuberculosis (Mtb) and the human immunodeficiency virus (HIV) is a significant public health concern. Individuals infected with Mtb who acquire HIV are approximately 16 times more likely to develop active tuberculosis. T cells play an important role as both targets for HIV infection and mediators of the immune response against both pathogens. This review aims to synthesize the current literature and provide insights into the effects of HIV/Mtb coinfection on T cell populations and their contributions to immunity. Evidence from multiple in vitro and in vivo studies demonstrates that T helper responses are severely compromised during coinfection, leading to impaired cytotoxic responses. Moreover, HIV's targeting of Mtb-specific cells, including those within granulomas, offers an explanation for the severe progression of the disease. Herein, we discuss the patterns of differentiation, exhaustion, and transcriptomic changes in T cells during coinfection, as well as the metabolic adaptations that are necessary for T cell maintenance and functionality. This review highlights the interconnectedness of the immune response and the pathogenesis of HIV/Mtb coinfection.
Collapse
Affiliation(s)
- José Alejandro Bohórquez
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA;
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Center for Infectious Diseases and Translational Medicine, Houston Methodist Research Institute, Houston, TX 77030, USA;
| | - Huanbin Xu
- Tulane National Primate Research Center, Tulane University School of Medicine, Tulane University, Covington, LA 70112, USA; (H.X.); (X.W.)
| | - Xiaolei Wang
- Tulane National Primate Research Center, Tulane University School of Medicine, Tulane University, Covington, LA 70112, USA; (H.X.); (X.W.)
| | - Guohua Yi
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA;
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA
- Department of Medicine, The University of Texas at Tyler School of Medicine, Tyler, TX 75708, USA
| |
Collapse
|
3
|
López-González JA, Martínez-Soto JM, Avila-Cervantes C, Mata-Pineda AL, Álvarez-Hernández G, Álvarez-Meza JB, Bolado-Martínez E, Candia-Plata MDC. Evaluation of Systemic Inflammation Before and After Standard Anti-tuberculosis Treatment in Patients With Active Pulmonary Tuberculosis and Diabetes Mellitus. Cureus 2024; 16:e55391. [PMID: 38562330 PMCID: PMC10984244 DOI: 10.7759/cureus.55391] [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] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
Background Diabetes mellitus (DM) is a common comorbidity of active pulmonary tuberculosis (APTB) that increases the risk of treatment failure during anti-tuberculosis chemotherapy. Evaluating systemic inflammatory response could help determine differences in response to treatment between APTB patients and those with APTB and DM. Methodology To explore changes in systemic inflammation, measured by a set of inflammatory mediators in subjects with APTB and TBDM before and after six months of anti-tuberculosis chemotherapy, 30 APTB and nine TBDM subjects underwent cytokine testing, including interleukin (IL)-6, IL-8, IL-10, interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α), and transforming growth factor-beta 1 (TGF-β1) by enzyme-linked immunosorbent assay, C-reactive protein by nephelometry, and sialic acid by colorimetric assay at baseline and following six months of standard anti-tuberculosis treatment. Sputum smear microscopy or molecular biology (Xpert MTB/RIF) was used for diagnosis, and sputum smear microscopy was performed monthly during the treatment of the patient with pulmonary tuberculosis to evaluate his evolution. Principal component analysis examined changes in the inflammatory status. Results Both groups showed negative sputum smear microscopy in the sixth month after starting anti-tuberculosis chemotherapy. TGF-β1 was found to be significantly higher in subjects with TBDM before treatment compared to APTB patients (p<0.001), and systemic inflammation continued only in TBDM subjects after treatment (accumulation and persistence of inflammatory mediators like IL-6, IL-8, IL-10, IFN-γ, TNF-α, TGF-β1, C-reactive protein, and sialic acid in blood). On the other hand, the mediators IFN-γ, C-reactive protein, and total sialic acid were found to be most influential in distinguishing pre- and post-treatment inflammatory response in subjects with APTB without DM. Conclusions Inflammatory mediators analyzed in combination, including IFN-γ, CRP, and total sialic acid, may be useful in evaluating the systemic inflammatory response in subjects with APTB and TBDM before and after anti-tuberculosis treatment. Determining these mediators revealed persistent systemic inflammation in TBDM subjects after six months of standard tuberculosis treatment, despite negative sputum smear microscopy results and good glycemic control. This suggests a need for inflammation-modulating therapies during tuberculosis control. Finally, monitoring sputum smear microscopy results alongside the determination of proposed inflammatory mediators (IFN-γ, CRP, and total sialic acid) are effective in evaluating the response to anti-tuberculosis treatment in APTB subjects without DM, warranting further investigation.
Collapse
|
4
|
Huang T, He J, Zhou X, Pan H, He F, Du A, Yu B, Jiang N, Li X, Yuan K, Wang Z. Discovering common pathogenetic processes between COVID-19 and tuberculosis by bioinformatics and system biology approach. Front Cell Infect Microbiol 2023; 13:1280223. [PMID: 38162574 PMCID: PMC10757339 DOI: 10.3389/fcimb.2023.1280223] [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: 08/19/2023] [Accepted: 11/07/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction The coronavirus disease 2019 (COVID-19) pandemic, stemming from the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has persistently threatened the global health system. Meanwhile, tuberculosis (TB) caused by Mycobacterium tuberculosis (M. tuberculosis) still continues to be endemic in various regions of the world. There is a certain degree of similarity between the clinical features of COVID-19 and TB, but the underlying common pathogenetic processes between COVID-19 and TB are not well understood. Methods To elucidate the common pathogenetic processes between COVID-19 and TB, we implemented bioinformatics and systematic research to obtain shared pathways and molecular biomarkers. Here, the RNA-seq datasets (GSE196822 and GSE126614) are used to extract shared differentially expressed genes (DEGs) of COVID-19 and TB. The common DEGs were used to identify common pathways, hub genes, transcriptional regulatory networks, and potential drugs. Results A total of 96 common DEGs were selected for subsequent analyses. Functional enrichment analyses showed that viral genome replication and immune-related pathways collectively contributed to the development and progression of TB and COVID-19. Based on the protein-protein interaction (PPI) network analysis, we identified 10 hub genes, including IFI44L, ISG15, MX1, IFI44, OASL, RSAD2, GBP1, OAS1, IFI6, and HERC5. Subsequently, the transcription factor (TF)-gene interaction and microRNA (miRNA)-gene coregulatory network identified 61 TFs and 29 miRNAs. Notably, we identified 10 potential drugs to treat TB and COVID-19, namely suloctidil, prenylamine, acetohexamide, terfenadine, prochlorperazine, 3'-azido-3'-deoxythymidine, chlorophyllin, etoposide, clioquinol, and propofol. Conclusion This research provides novel strategies and valuable references for the treatment of tuberculosis and COVID-19.
Collapse
Affiliation(s)
- Tengda Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jinyi He
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyi Zhou
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hongyuan Pan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Fang He
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Ao Du
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Bingxuan Yu
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Nan Jiang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoquan Li
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Kefei Yuan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhen Wang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Ashenafi S, Loreti MG, Bekele A, Aseffa G, Amogne W, Kassa E, Aderaye G, Brighenti S. Inflammatory immune profiles associated with disease severity in pulmonary tuberculosis patients with moderate to severe clinical TB or anemia. Front Immunol 2023; 14:1296501. [PMID: 38162636 PMCID: PMC10756900 DOI: 10.3389/fimmu.2023.1296501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024] Open
Abstract
Background Immune control of Mycobacterium tuberculosis (Mtb) infection is largely influenced by the extensive disease heterogeneity that is typical for tuberculosis (TB). In this study, the peripheral inflammatory immune profile of different sub-groups of pulmonary TB patients was explored based on clinical disease severity, anemia of chronic disease, or the radiological extent of lung disease. Methods Plasma samples were obtained from n=107 patients with active pulmonary TB at the time of diagnosis and after start of standard chemotherapy. A composite clinical TB symptoms score, blood hemoglobin status and chest X-ray imaging were used to sub-group TB patients into 1.) mild and moderate-severe clinical TB, 2.) anemic and non-anemic TB, or 3.) limited and extensive lung involvement. Plasma levels of biomarkers associated with inflammation pathways were assessed using a Bio-Plex Magpix 37-multiplex assay. In parallel, Th1/Th2 cytokines were quantified with a 27-multiplex in matched plasma and cell culture supernatants from whole blood stimulated with M. tuberculosis-antigens using the QuantiFERON-TB Gold assay. Results Clinical TB disease severity correlated with low blood hemoglobin levels and anemia but not with radiological findings in this study cohort. Multiplex protein analyses revealed that distinct clusters of inflammation markers and cytokines separated the different TB disease sub-groups with variable efficacy. Several top-ranked markers overlapped, while other markers were unique with regards to their importance to differentiate the TB disease severity groups. A distinct immune response profile defined by elevated levels of BAFF, LIGHT, sTNF-R1 and 2, IP-10, osteopontin, chitinase-3-like protein 1, and IFNα2 and IL-8, were most effective in separating TB patients with different clinical disease severity and were also promising candidates for treatment monitoring. TB patients with mild disease displayed immune polarization towards mixed Th1/Th2 responses, while pro-inflammatory and B cell stimulating cytokines as well as immunomodulatory mediators predominated in moderate-severe TB disease and anemia of TB. Conclusions Our data demonstrated that clinical disease severity in TB is associated with anemia and distinct inflammatory immune profiles. These results contribute to the understanding of immunopathology in pulmonary TB and define top-ranked inflammatory mediators as biomarkers of disease severity and treatment prognosis.
Collapse
Affiliation(s)
- Senait Ashenafi
- Department of Pathology, School of Medicine, College of Health Sciences, Tikur Anbessa Specialized Hospital and Addis Ababa University, Addis Ababa, Ethiopia
- Department of Medicine Huddinge, Center for Infectious Medicine (CIM), ANA Futura, Karolinska Institutet, Stockholm, Sweden
| | - Marco Giulio Loreti
- Department of Medicine Huddinge, Center for Infectious Medicine (CIM), ANA Futura, Karolinska Institutet, Stockholm, Sweden
| | - Amsalu Bekele
- Department of Internal Medicine, School of Medicine, College of Health Sciences, Tikur Anbessa Specialized Hospital and Addis Ababa University, Addis Ababa, Ethiopia
| | - Getachew Aseffa
- Department of Radiology, School of Medicine, College of Health Sciences, Tikur Anbessa Specialized Hospital and Addis Ababa University, Addis Ababa, Ethiopia
| | - Wondwossen Amogne
- Department of Internal Medicine, School of Medicine, College of Health Sciences, Tikur Anbessa Specialized Hospital and Addis Ababa University, Addis Ababa, Ethiopia
| | - Endale Kassa
- Department of Internal Medicine, School of Medicine, College of Health Sciences, Tikur Anbessa Specialized Hospital and Addis Ababa University, Addis Ababa, Ethiopia
| | - Getachew Aderaye
- Department of Internal Medicine, School of Medicine, College of Health Sciences, Tikur Anbessa Specialized Hospital and Addis Ababa University, Addis Ababa, Ethiopia
| | - Susanna Brighenti
- Department of Medicine Huddinge, Center for Infectious Medicine (CIM), ANA Futura, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
6
|
Kazemi S, Mirzaei R, Karampoor S, Hosseini-Fard SR, Ahmadyousefi Y, Soltanian AR, Keramat F, Saidijam M, Alikhani MY. Circular RNAs in tuberculosis: From mechanism of action to potential diagnostic biomarker. Microb Pathog 2023; 185:106459. [PMID: 37995882 DOI: 10.1016/j.micpath.2023.106459] [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: 05/25/2023] [Revised: 10/01/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (M. tuberculosis), continues to be a major global health concern. Understanding the molecular intricacies of TB pathogenesis is crucial for developing effective diagnostic and therapeutic approaches. Circular RNAs (circRNAs), a class of single-stranded RNA molecules characterized by covalently closed loops, have recently emerged as potential diagnostic biomarkers in various diseases. CircRNAs have been demonstrated to modulate the host's immunological responses against TB, specifically by reducing monocyte apoptosis, augmenting autophagy, and facilitating macrophage polarization. This review comprehensively explores the roles and mechanisms of circRNAs in TB pathogenesis. We also discuss the growing body of evidence supporting their utility as promising diagnostic biomarkers for TB. By bridging the gap between fundamental circRNA biology and TB diagnostics, this review offers insights into the exciting potential of circRNAs in combatting this infectious disease.
Collapse
Affiliation(s)
- Sima Kazemi
- Infectious Disease Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Reza Hosseini-Fard
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yaghoub Ahmadyousefi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran; Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Reza Soltanian
- Department of Biostatistics and Epidemiology, School of Public Health, Hamadan University of Medical Sciences, Iran
| | - Fariba Keramat
- Infectious Disease Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Massoud Saidijam
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Yousef Alikhani
- Infectious Disease Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
7
|
Ashenafi S, Brighenti S. Reinventing the human tuberculosis (TB) granuloma: Learning from the cancer field. Front Immunol 2022; 13:1059725. [PMID: 36591229 PMCID: PMC9797505 DOI: 10.3389/fimmu.2022.1059725] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Tuberculosis (TB) remains one of the deadliest infectious diseases in the world and every 20 seconds a person dies from TB. An important attribute of human TB is induction of a granulomatous inflammation that creates a dynamic range of local microenvironments in infected organs, where the immune responses may be considerably different compared to the systemic circulation. New and improved technologies for in situ quantification and multimodal imaging of mRNA transcripts and protein expression at the single-cell level have enabled significantly improved insights into the local TB granuloma microenvironment. Here, we review the most recent data on regulation of immunity in the TB granuloma with an enhanced focus on selected in situ studies that enable spatial mapping of immune cell phenotypes and functions. We take advantage of the conceptual framework of the cancer-immunity cycle to speculate how local T cell responses may be enhanced in the granuloma microenvironment at the site of Mycobacterium tuberculosis infection. This includes an exploratory definition of "hot", immune-inflamed, and "cold", immune-excluded TB granulomas that does not refer to the level of bacterial replication or metabolic activity, but to the relative infiltration of T cells into the infected lesions. Finally, we reflect on the current knowledge and controversy related to reactivation of active TB in cancer patients treated with immune checkpoint inhibitors such as PD-1/PD-L1 and CTLA-4. An understanding of the underlying mechanisms involved in the induction and maintenance or disruption of immunoregulation in the TB granuloma microenvironment may provide new avenues for host-directed therapies that can support standard antibiotic treatment of persistent TB disease.
Collapse
Affiliation(s)
- Senait Ashenafi
- Department of Medicine Huddinge, Center for Infectious Medicine (CIM), Karolinska Institutet, ANA Futura, Huddinge, Sweden,Department of Pathology, School of Medicine, College of Health Sciences, Tikur Anbessa Specialized Hospital and Addis Ababa University, Addis Ababa, Ethiopia
| | - Susanna Brighenti
- Department of Medicine Huddinge, Center for Infectious Medicine (CIM), Karolinska Institutet, ANA Futura, Huddinge, Sweden,*Correspondence: Susanna Brighenti,
| |
Collapse
|
8
|
Shah T, Shah Z, Yasmeen N, Baloch Z, Xia X. Pathogenesis of SARS-CoV-2 and Mycobacterium tuberculosis Coinfection. Front Immunol 2022; 13:909011. [PMID: 35784278 PMCID: PMC9246416 DOI: 10.3389/fimmu.2022.909011] [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: 03/31/2022] [Accepted: 05/23/2022] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease-2019 (COVID-19), caused by SARS-CoV-2, is an infectious disease that poses severe threats to global public health and significant economic losses. The COVID-19 global burden is rapidly increasing, with over 246.53 million COVID-19 cases and 49.97 million deaths reported in the WHO 2021 report. People with compromised immunity, such as tuberculosis (TB) patients, are highly exposed to severe COVID-19. Both COVID-19 and TB diseases spread primarily through respiratory droplets from an infected person to a healthy person, which may cause pneumonia and cytokine storms, leading to severe respiratory disorders. The COVID-19-TB coinfection could be fatal, exacerbating the current COVID-19 pandemic apart from cellular immune deficiency, coagulation activation, myocardial infarction, and other organ dysfunction. This study aimed to assess the pathogenesis of SARS-CoV-2-Mycobacterium tuberculosis coinfections. We provide a brief overview of COVID19-TB coinfection and discuss SARS-CoV-2 host cellular receptors and pathogenesis. In addition, we discuss M. tuberculosis host cellular receptors and pathogenesis. Moreover, we highlight the impact of SARS-CoV-2 on TB patients and the pathological pathways that connect SARS-CoV-2 and M. tuberculosis infection. Further, we discuss the impact of BCG vaccination on SARS-CoV-2 cases coinfected with M. tuberculosis, as well as the diagnostic challenges associated with the coinfection.
Collapse
Affiliation(s)
- Taif Shah
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Zahir Shah
- College of Veterinary Sciences, The University of Agriculture Peshawar, Peshawar, Pakistan
| | - Nafeesa Yasmeen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zulqarnain Baloch
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Xueshan Xia
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
9
|
Kathamuthu GR, Sridhar R, Baskaran D, Babu S. Dominant expansion of CD4+, CD8+ T and NK cells expressing Th1/Tc1/Type 1 cytokines in culture-positive lymph node tuberculosis. PLoS One 2022; 17:e0269109. [PMID: 35617254 PMCID: PMC9135291 DOI: 10.1371/journal.pone.0269109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 05/15/2022] [Indexed: 01/11/2023] Open
Abstract
Lymph node culture-positive tuberculosis (LNTB+) is associated with increased mycobacterial antigen-induced pro-inflammatory cytokine production compared to LN culture-negative tuberculosis (LNTB-). However, the frequencies of CD4+, CD8+ T cells and NK cells expressing Th1/Tc1/Type 1 (IFNγ, TNFα, IL-2), Th17/Tc17/Type 17 (IL-17A, IL-17F, IL-22) cytokines and cytotoxic (perforin [PFN], granzyme [GZE] B, CD107a) markers in LNTB+ and LNTB- individuals are not known. Thus, we have studied the unstimulated (UNS) and mycobacterial antigen-induced frequencies of CD4+, CD8+ T and NK cells expressing Th1, Th17 cytokines and cytotoxic markers using flow cytometry. The frequencies of CD4+, CD8+ T and NK cells expressing cytokines and cytotoxic markers were not significantly different between LNTB+ and LNTB- individuals in UNS condition. In contrast, upon Mtb antigen stimulation, LNTB+ individuals are associated with significantly increased frequencies of CD4+ T cells (PPD [IFNγ, TNFα], ESAT-6 PP [IFNγ, TNFα], CFP-10 PP [IFNγ, TNFα, IL-2]), CD8+ T cells (PPD [IFNγ], ESAT-6 PP [IFNγ], CFP-10 PP [TNFα]) and NK cells (PPD [IFNγ, TNFα], ESAT-6 PP [IFNγ, TNFα], CFP-10 PP [TNFα]) expressing Th1/Tc1/Type 1, but not Th17/Tc17/Type 17 cytokines and cytotoxic markers compared to LNTB- individuals. LNTB+ individuals did not show any significant alterations in the frequencies of CD4+, CD8+ T cells and NK cells expressing cytokines and cytotoxic markers compared to LNTB- individuals upon HIV Gag PP and P/I antigen stimulation. Increased frequencies of CD4+, CD8+ T and NK cells expressing Th1/Tc1/Type 1 cytokines among the LNTB+ group indicates that the presence of mycobacteria plays a dominant role in the activation of key correlates of immune protection or induces higher immunopathology.
Collapse
Affiliation(s)
- Gokul Raj Kathamuthu
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai, India
- National Institute for Research in Tuberculosis (NIRT), Chennai, India
- * E-mail:
| | | | - Dhanaraj Baskaran
- National Institute for Research in Tuberculosis (NIRT), Chennai, India
| | - Subash Babu
- National Institutes of Health-NIRT-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, United States of America
| |
Collapse
|
10
|
Ashenafi S, Muvva JR, Mily A, Snäll J, Zewdie M, Chanyalew M, Rehn A, Rahman S, Aseffa G, Bekele A, Aderaye G, Lema B, Svensson M, Brighenti S. Immunosuppressive Features of the Microenvironment in Lymph Nodes Granulomas from Tuberculosis and HIV-Co-Infected Patients. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:653-670. [PMID: 35092727 PMCID: PMC9302207 DOI: 10.1016/j.ajpath.2021.12.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/07/2021] [Accepted: 12/22/2021] [Indexed: 11/30/2022]
Abstract
Tuberculosis (TB) and HIV co-infection claims many lives every year. This study assessed immune responses in Mycobacterium tuberculosis-infected lymph node tissues from HIV-negative and HIV-positive patients compared with the peripheral circulation with a focus on myeloid cells and the cell-signaling enzymes, inducible nitric oxide synthase, and arginase (Arg)-1. Methods included immunohistochemistry or confocal microscopy and computerized image analyses, quantitative real-time PCR, multiplex Luminex, and flow cytometry. These findings indicate enhanced chronic inflammation and immune activation in TB/HIV co-infection but also enhanced immunosuppressive responses. Poorly formed necrotic TB granulomas with a high expression of M. tuberculosis antigens were elevated in TB/HIV-co-infected lymph nodes, and inducible nitric oxide synthase and Arg-1 expression was significantly higher in TB/HIV-co-infected compared with HIV-negative TB or control tissues. High Arg-1 expression was found in myeloid cells with a phenotype characteristic of myeloid-derived suppressor cells (MDCS) that were particularly abundant in TB/HIV-co-infected tissues. Accordingly, Lin-/HLA-DRlow/int/CD33+/CD11b+/CD15+ granulocytic myeloid-derived suppressor cells were significantly elevated in blood samples from TB/HIV-co-infected patients. CD15+ myeloid-derived suppressor cells correlated with plasma HIV viral load and M. tuberculosis antigen load in tissue but were inversely associated with peripheral CD4 T-cells counts. Enhanced chronic inflammation driven by M. tuberculosis and HIV co-infection may promote Arg-1-expressing MDSCs at the site of infection thereby advancing TB disease progression.
Collapse
Affiliation(s)
- Senait Ashenafi
- Center for Infectious Medicine, Department of Medicine, ANA Futura, Karolinska Institutet, Huddinge, Sweden; Department of Pathology, Tikur Anbessa Specialized Hospital and Addis Ababa University, College of Health Sciences, Addis Ababa, Ethiopia
| | - Jagadeeswara Rao Muvva
- Center for Infectious Medicine, Department of Medicine, ANA Futura, Karolinska Institutet, Huddinge, Sweden
| | - Akhirunnesa Mily
- Center for Infectious Medicine, Department of Medicine, ANA Futura, Karolinska Institutet, Huddinge, Sweden
| | - Johanna Snäll
- Center for Infectious Medicine, Department of Medicine, ANA Futura, Karolinska Institutet, Huddinge, Sweden
| | - Martha Zewdie
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | | | - Anders Rehn
- Center for Infectious Medicine, Department of Medicine, ANA Futura, Karolinska Institutet, Huddinge, Sweden
| | - Sayma Rahman
- Center for Infectious Medicine, Department of Medicine, ANA Futura, Karolinska Institutet, Huddinge, Sweden
| | - Getachew Aseffa
- Department of Radiology, Tikur Anbessa Specialized Hospital and Addis Ababa University, College of Health Sciences, Addis Ababa, Ethiopia
| | - Amsalu Bekele
- Department of Internal Medicine, Tikur Anbessa Specialized Hospital and Addis Ababa University, College of Health Sciences, Addis Ababa, Ethiopia
| | - Getachew Aderaye
- Department of Internal Medicine, Tikur Anbessa Specialized Hospital and Addis Ababa University, College of Health Sciences, Addis Ababa, Ethiopia
| | - Beede Lema
- Department of Surgery, Tikur Anbessa Specialized Hospital and Addis Ababa University, College of Health Sciences, Addis Ababa, Ethiopia
| | - Mattias Svensson
- Center for Infectious Medicine, Department of Medicine, ANA Futura, Karolinska Institutet, Huddinge, Sweden
| | - Susanna Brighenti
- Center for Infectious Medicine, Department of Medicine, ANA Futura, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
11
|
Xavier MB, Pires CAA, Gomes CMDC, Rodrigues GF, Xavier DP, de Brito JAGDSM, Corbett CEP. The expression of FOXP3 in lesions of several forms of leprosy in patients co-infected with HIV. PLoS Negl Trop Dis 2021; 15:e0009887. [PMID: 34748560 PMCID: PMC8601607 DOI: 10.1371/journal.pntd.0009887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/18/2021] [Accepted: 10/08/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Brazil remains endemic for infection by the human immunodeficiency virus (HIV) and leprosy, having a major impact on public health and the life quality of affected patients. Although the relevance of this co-infection is recognized, several aspects, such as the immune response, are not yet fully understood. The objective of this study was to investigate the expression of FOXP3+ Treg cells in leprosy skin lesions and to correlate their clinical forms, laboratory characteristics (CD4, CD8, and CV), and the immune reconstitution syndrome in HIV-leprosy co-infection. METHODOLOGY/PRINCIPAL FINDINGS An observational, cross-sectional, and analytical study was carried out comparing four groups of patients: those with concomitant diagnosis of leprosy and HIV infection without a leprosy reaction, those with leprosy and HIV co-infection patients with a reverse reaction (RR), those with leprosy without HIV and without reaction, and those with leprosywithout HIV and with RR. The patients were diagnosed at a dermatology outpatient clinic located in Belém, Pará, Brazil, from 2003 to 2017. In the sample studied, there was a positive correlation between FOXP3+ cell density and viral load, negative correlation with blood CD4+ (not statistically significant), significant positive correlation in CD8 count in patients with leprosy reaction, and positive relationship in patients with IRIS. The density of cells expressing FOXP3 was higher in the BL/LL forms in patients without HIV, although the difference was not statistically significant. However, the cell mean was higher in the TT/BT forms in patients co-infected with leprosy and HIV, showing contradictory results. CONCLUSIONS/SIGNIFICANCE These findings support that higher activity of the HIV may stimulate or result in a higher expression of FOXP3-Tregs and that they may be involved in active immunosuppression observed at the infection site at the tissue level. This supports the need to expand studies on FOXP3+ Treg cells in co-infected patients.
Collapse
Affiliation(s)
- Marília Brasil Xavier
- Research Laboratory in Tropical Dermatology and Endemic Diseases of the Nucleus of Tropical Medicine/Federal University of Pará (NMT/UFPA), Belém, Pará, Brazil
- Research Laboratory on Skin diseases of Sanitary Interest, Biological and Health Sciences Center, State University of Pará (UEPA), Belém, Pará, Brazil
| | - Carla Andréa Avelar Pires
- Research Laboratory in Tropical Dermatology and Endemic Diseases of the Nucleus of Tropical Medicine/Federal University of Pará (NMT/UFPA), Belém, Pará, Brazil
- Research Laboratory on Skin diseases of Sanitary Interest, Biological and Health Sciences Center, State University of Pará (UEPA), Belém, Pará, Brazil
| | - Cláudia Maria de Castro Gomes
- Laboratory of Pathology of Infectious Diseases, Department of Pathology, Medical School, São Paulo University, São Paulo, Brazil
| | - Gabriela Fernandes Rodrigues
- Laboratory of Pathology of Infectious Diseases, Department of Pathology, Medical School, São Paulo University, São Paulo, Brazil
| | - Débora Pinheiro Xavier
- Research Laboratory in Tropical Dermatology and Endemic Diseases of the Nucleus of Tropical Medicine/Federal University of Pará (NMT/UFPA), Belém, Pará, Brazil
| | | | - Carlos Eduardo Pereira Corbett
- Laboratory of Pathology of Infectious Diseases, Department of Pathology, Medical School, São Paulo University, São Paulo, Brazil
| |
Collapse
|
12
|
Pan L, Chen X, Liu X, Qiu W, Liu Y, Jiang W, Zheng Y, Mou Y, Xu W, Li X, Ge H, Zhu H. Innate lymphoid cells exhibited IL-17-expressing phenotype in active tuberculosis disease. BMC Pulm Med 2021; 21:318. [PMID: 34641843 PMCID: PMC8513179 DOI: 10.1186/s12890-021-01678-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 09/21/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Innate lymphoid cells (ILCs), as an important group of innate immunity, could respond rapidly to Mycobacterium tuberculosis (Mtb) infection. In this research, we studied the phenotypic changes of circulatory ILCs in active tuberculosis (TB) disease. METHODS We recruited 40 patients with active Mtb infection (TB group) and 41 healthy subjects (NC group), and collected their clinical information and peripheral blood. Circulating ILCs, ILC subsets, dendritic cells (DCs), macrophages, and the production of cytokines in ILCs were tested by flow cytometry (FCM). Enzyme-linked immunosorbent assay (ELISA) was used to detect plasma IL-23. RESULTS Compared with healthy control, total ILCs (0.73% vs. 0.42%, P = 0.0019), ILC1 (0.55% vs. 0.31%, P = 0.0024) and CD117+ ILC2 (0.02% vs. 0.01%, P = 0.0267) were upregulated in TB group. The total IL-17+ lymphocytes were elevated (3.83% vs. 1.76%, P = 0.0006) while the IL-22+ lymphocytes remained unchanged. Within ILC subsets, ILC3, CD117+ ILC2 and ILC1 in TB group all expressed increased IL-17 (15.15% vs. 4.55%, 19.01% vs. 4.57%, 8.79% vs. 3.87%, P < 0.0001) but similar IL-22 comparing with healthy control. TB group had more plasma IL-23 than NC group (7.551 vs. 5.564 pg/mL, P = 0.0557). Plasma IL-23 in TB group was positively correlated to IL-17+ ILC3 (r = 0.4435, P = 0.0141), IL-17+CD117+ ILC2 (r = 0.5385, P = 0.0021) and IL-17+ ILC1(r = 0.3719, P = 0.0430). TB group also had elevated DCs (9.35% vs. 6.49%, P < 0.0001) while macrophages remained unchanged. Within TB group, higher proportion of IL-17+ ILCs was related to severer inflammatory status and poorer clinical condition. CONCLUSIONS In active TB disease, circulatory ILCs were upregulated and exhibited IL-17-expressing phenotype. This may expand the understanding of immune reaction to Mtb infection.
Collapse
Affiliation(s)
- Linyue Pan
- Department of Respiratory and Critical Care Medicine, The Affiliated Huadong Hospital of Fudan University, 221 West Yan'an Road, Shanghai, 200040, China.,Department of Respiratory and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoli Chen
- Department of Tuberculosis, the Sixth People's Hospital of Nantong, Jiangsu, China
| | - Xuanqi Liu
- Department of Respiratory and Critical Care Medicine, The Affiliated Huadong Hospital of Fudan University, 221 West Yan'an Road, Shanghai, 200040, China
| | - Wenjia Qiu
- Department of Respiratory and Critical Care Medicine, The Affiliated Huadong Hospital of Fudan University, 221 West Yan'an Road, Shanghai, 200040, China
| | - Yunhuan Liu
- Department of Respiratory and Critical Care Medicine, The Affiliated Huadong Hospital of Fudan University, 221 West Yan'an Road, Shanghai, 200040, China
| | - Weiping Jiang
- Department of Respiratory and Critical Care Medicine, The Affiliated Huadong Hospital of Fudan University, 221 West Yan'an Road, Shanghai, 200040, China
| | - Yang Zheng
- Department of Respiratory and Critical Care Medicine, The Affiliated Huadong Hospital of Fudan University, 221 West Yan'an Road, Shanghai, 200040, China
| | - Yan Mou
- Department of Respiratory and Critical Care Medicine, The Affiliated Huadong Hospital of Fudan University, 221 West Yan'an Road, Shanghai, 200040, China
| | - Wei Xu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiangyang Li
- Department of Respiratory and Critical Care Medicine, The Affiliated Huadong Hospital of Fudan University, 221 West Yan'an Road, Shanghai, 200040, China
| | - Haiyan Ge
- Department of Respiratory and Critical Care Medicine, The Affiliated Huadong Hospital of Fudan University, 221 West Yan'an Road, Shanghai, 200040, China.
| | - Huili Zhu
- Department of Respiratory and Critical Care Medicine, The Affiliated Huadong Hospital of Fudan University, 221 West Yan'an Road, Shanghai, 200040, China
| |
Collapse
|
13
|
Tiwari M, Verma M, Singh P, Bharti D. Role of Fok I VDR polymorphism in TB risk assessment; A Study in Central India population. Meta Gene 2021. [DOI: 10.1016/j.mgene.2021.100896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
14
|
Morgan J, Muskat K, Tippalagama R, Sette A, Burel J, Lindestam Arlehamn CS. Classical CD4 T cells as the cornerstone of antimycobacterial immunity. Immunol Rev 2021; 301:10-29. [PMID: 33751597 PMCID: PMC8252593 DOI: 10.1111/imr.12963] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/11/2021] [Accepted: 02/13/2021] [Indexed: 12/13/2022]
Abstract
Tuberculosis is a significant health problem without an effective vaccine to combat it. A thorough understanding of the immune response and correlates of protection is needed to develop a more efficient vaccine. The immune response against Mycobacterium tuberculosis (Mtb) is complex and involves all aspects of the immune system, however, the optimal protective, non‐pathogenic T cell response against Mtb is still elusive. This review will focus on discussing CD4 T cell immunity against mycobacteria and its importance in Mtb infection with a primary focus on human studies. We will in particular discuss the large heterogeneity of immune cell subsets that have been revealed by recent immunological investigations at an unprecedented level of detail. These studies have identified specific classical CD4 T cell subsets important for immune responses against Mtb in various states of infection. We further discuss the functional attributes that have been linked to the various subsets such as upregulation of activation markers and cytokine production. Another important topic to be considered is the antigenic targets of Mtb‐specific immune responses, and how antigen reactivity is influenced by both disease state and environmental exposure(s). These are key points for both vaccines and immune diagnostics development. Ultimately, these factors are holistically considered in the definition and investigations of what are the correlates on protection and resolution of disease.
Collapse
Affiliation(s)
- Jeffrey Morgan
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Kaylin Muskat
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Rashmi Tippalagama
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Julie Burel
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA, USA
| | | |
Collapse
|
15
|
Barreto-Duarte B, Sterling TR, Fiske CT, Almeida A, Nochowicz CH, Smith RM, Barnett L, Warren C, Blackman A, Lapa e Silva JR, Andrade BB, Kalams SA. Increased Frequency of Memory CD4+ T-Cell Responses in Individuals With Previously Treated Extrapulmonary Tuberculosis. Front Immunol 2020; 11:605338. [PMID: 33391271 PMCID: PMC7774017 DOI: 10.3389/fimmu.2020.605338] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/13/2020] [Indexed: 11/13/2022] Open
Abstract
Extrapulmonary TB (EPTB) occurs with increased frequency in persons with underlying immunodeficiency. Even after recovery from acute illness, differences in immune phenotype and activation persist. Studies defining characteristics of immune responses after recovery from extrapulmonary TB may provide insights into factors that increase TB risk. We performed two case-control studies (in the United States and Brazil) among HIV-seronegative adults with previous EPTB (n = 9; 25), previous pulmonary TB (n = 7; 25), latent M. tuberculosis (Mtb) infection (n = 11; 25), and uninfected TB contacts (n = 10; 25). We assessed the frequency of dual CD4+ interferon-γ and tumor necrosis factor-α responses after stimulation with overlapping Mtb peptides from ESAT-6 or CFP-10, or gamma-irradiated Mtb H37Rv, proliferative responses to Mtb antigens, T-regulatory cell (Treg) frequency and phenotype. In both study populations, individuals with prior EPTB had the highest frequency of intracellular cytokine-producing cells in response to Mtb antigens (p < 0.05; p <.0001). Persons with prior EPTB in Brazil had the highest levels of CD4 proliferation to Mtb antigens (p < 0.0001), and the highest expression of CD39 on Tregs (p < 0.0001). Individuals with treated EPTB maintained high frequencies of Mtb-specific memory responses and active Treg cells, suggesting that susceptibility to EPTB occurs despite the ability to develop and maintain enhanced adaptive immune responses.
Collapse
Affiliation(s)
- Beatriz Barreto-Duarte
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Universidade Salvador (UNIFACS), Laureate Universities, Salvador, Brazil
- Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Timothy R. Sterling
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Tuberculosis Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Christina T. Fiske
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Tuberculosis Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Alexandre Almeida
- Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cynthia H. Nochowicz
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Tuberculosis Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Rita M. Smith
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Tuberculosis Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Louise Barnett
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Tuberculosis Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Christian Warren
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Tuberculosis Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Amondrea Blackman
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Tuberculosis Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | | | - Bruno B. Andrade
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Universidade Salvador (UNIFACS), Laureate Universities, Salvador, Brazil
- Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Brazil
- Curso de Medicina, Centro Universitário Faculdades de Tecnologia e Ciências (UniFTC), Salvador, Brazil
- Curso de Medicina, Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil
| | - Spyros A. Kalams
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Tuberculosis Center, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
16
|
Mily A, Sarker P, Taznin I, Hossain D, Haq MA, Kamal SMM, Agerberth B, Brighenti S, Raqib R. Slow radiological improvement and persistent low-grade inflammation after chemotherapy in tuberculosis patients with type 2 diabetes. BMC Infect Dis 2020; 20:933. [PMID: 33287713 PMCID: PMC7722325 DOI: 10.1186/s12879-020-05473-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 10/05/2020] [Indexed: 11/10/2022] Open
Abstract
Background Diabetes mellitus type 2 (DM) may impede immune responses in tuberculosis (TB) and thus contribute to enhanced disease severity. In this study, we aimed to evaluate DM-mediated alterations in clinical, radiological and immunological outcomes in TB disease. Methods Newly diagnosed pulmonary TB patients with or without DM (TB n = 40; TB-DM n = 40) were recruited in Dhaka, Bangladesh. Clinical symptoms, sputum smear and culture conversion as well as chest radiography were assessed. Peripheral blood and sputum samples were collected at the time of diagnosis (baseline) and after 1, 2 and 6 months of standard anti-TB treatment. Blood samples were also obtained from healthy controls (n = 20). mRNA expression of inflammatory markers in blood and sputum samples were quantified using real-time PCR. Results The majority of TB-DM patients had poor glycemic control (HbA1c > 8%) and displayed elevated pulmonary pathology (P = 0.039) particularly in the middle (P < 0.004) and lower lung zones (P < 0.02) throughout the treatment period. However, reduction of clinical symptoms and time to sputum smear and culture conversion did not differ between the groups. Transcripts levels of the pro-inflammatory cytokines IL-1β (P = 0.003 at month-1 and P = 0.045 at month-2) and TNF-α (P = 0.005 at month-1) and the anti-inflammatory cytokine IL-10 (P = 0.005 at month-2) were higher in peripheral blood after anti-TB treatment in TB-DM compared to TB patients. Conversely in sputum, TB-DM patients had reduced CD4 (P < 0.009 at month-1) and IL-10 (P = 0.005 at month-1 and P = 0.006 at month-2) transcripts, whereas CD8 was elevated (P = 0.016 at month-2). At 1- and 2-month post-treatment, sputum IL-10 transcripts were inversely correlated with fasting blood glucose and HbA1c levels in all patients. Conclusion Insufficient up-regulation of IL-10 in the lung may fuel persistent local inflammation thereby promoting lung pathology in TB-DM patients with poorly controlled DM.
Collapse
Affiliation(s)
- Akhirunnesa Mily
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, ANA Futura, Karolinska Institutet, Stockholm, Sweden.,Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Protim Sarker
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Inin Taznin
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Delwar Hossain
- Respiratory Medicine, Bangladesh Institute of Research and Rehabilitation in Diabetes, Endocrine and Metabolic Disorders, Dhaka, Bangladesh
| | - Md Ahsanul Haq
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - S M Mostofa Kamal
- National Institute of the Diseases of the Chest and Hospital, Dhaka, Bangladesh
| | - Birgitta Agerberth
- Clinical Microbiology, Department of Laboratory Medicine (Labmed), ANA Futura, Karolinska Institutet, Stockholm, Sweden
| | - Susanna Brighenti
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, ANA Futura, Karolinska Institutet, Stockholm, Sweden
| | - Rubhana Raqib
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh.
| |
Collapse
|
17
|
Tapela K, Ochieng’ Olwal C, Quaye O. Parallels in the pathogenesis of SARS-CoV-2 and M. tuberculosis: a synergistic or antagonistic alliance? Future Microbiol 2020; 15:1691-1695. [PMID: 33404259 PMCID: PMC7789731 DOI: 10.2217/fmb-2020-0179] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/08/2020] [Indexed: 01/12/2023] Open
Affiliation(s)
- Kesego Tapela
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
- Department of Biochemistry, Cell & Molecular Biology, College of Basic & Applied Sciences, University of Ghana, Accra, Ghana
| | - Charles Ochieng’ Olwal
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
- Department of Biochemistry, Cell & Molecular Biology, College of Basic & Applied Sciences, University of Ghana, Accra, Ghana
| | - Osbourne Quaye
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
- Department of Biochemistry, Cell & Molecular Biology, College of Basic & Applied Sciences, University of Ghana, Accra, Ghana
| |
Collapse
|
18
|
Diverse immune environments in human lung tuberculosis granulomas assessed by quantitative multiplexed immunofluorescence. Mod Pathol 2020; 33:2507-2519. [PMID: 32591586 DOI: 10.1038/s41379-020-0600-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/05/2020] [Accepted: 06/05/2020] [Indexed: 01/07/2023]
Abstract
The precise nature of the local immune responses in lung tuberculosis (TB) granulomas requires a comprehensive understanding of their environmental complexities. At its most basic level, a granuloma is a compact, organized immune aggregate of macrophages surrounded by myeloid, B and T cells. We established two complementary multiplex immunolabeling panels to simultaneously evaluate the myeloid and lymphocytic contexture of 14 human lung TB granulomas in formalin-fixed paraffin-embedded tissue samples. We observed diverse CD3+ and CD8+ T-cell and CD20+ B lymphocyte compositions of the granuloma immune environment and a relatively homogeneous distribution of all myeloid cells. We also found significant associations between CD8+ T-cell densities and the myeloid marker CD11b and phagocytic cell marker CD68. In addition, significantly more CD68+ macrophages and CD8+ T cells were found in Mycobacterium tuberculosis-infected granulomas, as detected by Ziehl-Neelsen staining. FOXP3 expression was predominately found in a small subset of CD4+ T cells in different granulomas. As the success or failure of each granuloma is determined by the immune response within that granuloma at a local and not a systemic level, we attempted to identify the presence of reactive T cells based on expression of the T-cell activation marker CD137 (4-1BB) and programmed cell death-1 (PD-1). Only a small fraction of the CD4+ and CD8+ T cells expressed PD-1. CD137 expression was found only in a very small fraction of the CD4+ T cells in two granulomas. Our results also showed that multinucleated giant cells showed strong PD-L1 but not CTLA-4 membrane staining. This study offers new insights into the heterogeneity of immune cell infiltration in lung TB granulomas, suggesting that each TB granuloma represents a unique immune environment that might be independently influenced by the local adaptive immune response, bacterial state, and overall host disease status.
Collapse
|
19
|
Berry SB, Haack AJ, Theberge AB, Brighenti S, Svensson M. Host and Pathogen Communication in the Respiratory Tract: Mechanisms and Models of a Complex Signaling Microenvironment. Front Med (Lausanne) 2020; 7:537. [PMID: 33015094 PMCID: PMC7511576 DOI: 10.3389/fmed.2020.00537] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 07/29/2020] [Indexed: 01/15/2023] Open
Abstract
Chronic lung diseases are a leading cause of morbidity and mortality across the globe, encompassing a diverse range of conditions from infections with pathogenic microorganisms to underlying genetic disorders. The respiratory tract represents an active interface with the external environment having the primary immune function of resisting pathogen intrusion and maintaining homeostasis in response to the myriad of stimuli encountered within its microenvironment. To perform these vital functions and prevent lung disorders, a chemical and biological cross-talk occurs in the complex milieu of the lung that mediates and regulates the numerous cellular processes contributing to lung health. In this review, we will focus on the role of cross-talk in chronic lung infections, and discuss how different cell types and signaling pathways contribute to the chronicity of infection(s) and prevent effective immune clearance of pathogens. In the lung microenvironment, pathogens have developed the capacity to evade mucosal immunity using different mechanisms or virulence factors, leading to colonization and infection of the host; such mechanisms include the release of soluble and volatile factors, as well as contact dependent (juxtracrine) interactions. We explore the diverse modes of communication between the host and pathogen in the lung tissue milieu in the context of chronic lung infections. Lastly, we review current methods and approaches used to model and study these host-pathogen interactions in vitro, and the role of these technological platforms in advancing our knowledge about chronic lung diseases.
Collapse
Affiliation(s)
- Samuel B. Berry
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Amanda J. Haack
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | | | - Susanna Brighenti
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Mattias Svensson
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
20
|
Abstract
Lymph nodes, particularly thoracic lymph nodes, are among the most common sites of extrapulmonary tuberculosis (TB). However, Mycobacterium tuberculosis (Mtb) infection in these organs is understudied. Aside from being sites of initiation of the adaptive immune system, lymph nodes also serve as niches of Mtb growth and persistence. Mtb infection results in granuloma formation that disrupts and—if it becomes large enough—replaces the normal architecture of the lymph node that is vital to its function. In preclinical models, successful TB vaccines appear to prevent spread of Mtb from the lungs to the lymph nodes. Reactivation of latent TB can start in the lymph nodes resulting in dissemination of the bacteria to the lungs and other organs. Involvement of the lymph nodes may improve Bacille Calmette-Guerin (BCG) vaccine efficacy. Lastly, drug penetration to the lymph nodes is poor compared to blood, lung tissue, and lung granulomas. Future studies on evaluating the efficacy of vaccines and anti-TB drug treatments should include consideration of the effects on thoracic lymph nodes and not just the lungs.
Collapse
Affiliation(s)
- Sharie Keanne C. Ganchua
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Alexander G. White
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Edwin C. Klein
- Division of Laboratory Animal Resources, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - JoAnne L. Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
21
|
Rakshit S, Ahmed A, Adiga V, Sundararaj BK, Sahoo PN, Kenneth J, D’Souza G, Bonam W, Johnson C, Franken KL, Ottenhoff TH, Finak G, Gottardo R, Stuart KD, De Rosa SC, McElrath MJ, Vyakarnam A. BCG revaccination boosts adaptive polyfunctional Th1/Th17 and innate effectors in IGRA+ and IGRA- Indian adults. JCI Insight 2019; 4:130540. [PMID: 31743110 PMCID: PMC6975271 DOI: 10.1172/jci.insight.130540] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 11/13/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUNDBacille Calmette-Guérin (BCG) vaccine is protective against Tuberculosis (TB) in children, but its efficacy wanes with age. Consequently, determining if BCG revaccination augments anti-TB immunity in young adults in TB endemic regions is vital.METHODSTwo hundred healthy adults, BCG vaccinated at birth, were tested for their IFN-γ release assay (IGRA) status. Of these, 28 IGRA+ and 30 IGRA- were BCG revaccinated, and 24 IGRA+ and 23 IGRA- subjects served as unvaccinated controls. T and innate cell responses to mycobacterial antigens were analyzed by 14-color flow cytometry over 34 weeks.RESULTSIFN-γ and/or IL-2 Ag85A- and BCG-specific CD4+ and CD8+ T cell responses were boosted by revacciantion at 4 and 34 weeks, respectively, and were > 2-fold higher in IGRA+ compared with IGRA- vaccinees. Polyfunctional Ag85A, BCG, and mycobacterium tuberculosis (Mtb) latency Ag-specific (LTAg-specific) CD4+ T cells expressing up to 8 cytokines were also significantly enhanced in both IGRA+ and IGRA- vaccinees relative to unvaccinated controls, most markedly in IGRA+ vaccinees. A focused analysis of Th17 responses revealed expansion of Ag85A-, BCG-, and LTAg-specific total IL-17A+,IL-17F+,IL-22+, and IL-10+ CD4+ T cell effectors in both IGRA+ and IGRA- subjects. Also, innate IFN-γ+ NK/γδ/NKT cell responses were higher in both IGRA+ and IGRA- vaccinees compared with controls. This is the first evidence to our knowledge that BCG revaccination significantly boosts antimycobacterial Th1/Th17 responses in IGRA+ and IGRA- subjects.CONCLUSIONThese data show that BCG revaccination is immunogenic in IGRA- and IGRA+ subjects, implying that Mtb preinfection in IGRA+ subjects does not impact immunogenicity. This has implications for public health and vaccine development strategies.FUNDINGThis work was funded principally by DBT-NIH (BT/MB/Indo-US/HIPC/2013).
Collapse
Affiliation(s)
- Srabanti Rakshit
- Laboratory of Immunology of HIV-TB Co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Asma Ahmed
- Laboratory of Immunology of HIV-TB Co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Vasista Adiga
- Laboratory of Immunology of HIV-TB Co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Bharath K. Sundararaj
- Laboratory of Immunology of HIV-TB Co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Pravat Nalini Sahoo
- Laboratory of Immunology of HIV-TB Co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - John Kenneth
- Department of Infectious Diseases and
- Department of Pulmonary Medicine, St. John’s Research Institute, Bangalore, India
| | - George D’Souza
- Department of Infectious Diseases and
- Department of Pulmonary Medicine, St. John’s Research Institute, Bangalore, India
| | | | | | - Kees L.M.C. Franken
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Tom H.M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Greg Finak
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | - Stephen C. De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Laboratory Medicine and
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Annapurna Vyakarnam
- Laboratory of Immunology of HIV-TB Co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, Guy’s Hospital, King’s College London, London, United Kingdom
| |
Collapse
|
22
|
Carow B, Hauling T, Qian X, Kramnik I, Nilsson M, Rottenberg ME. Spatial and temporal localization of immune transcripts defines hallmarks and diversity in the tuberculosis granuloma. Nat Commun 2019; 10:1823. [PMID: 31015452 PMCID: PMC6479067 DOI: 10.1038/s41467-019-09816-4] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 04/02/2019] [Indexed: 01/04/2023] Open
Abstract
Granulomas are the pathological hallmark of tuberculosis (TB) and the niche where bacilli can grow and disseminate or the immunological microenvironment in which host cells interact to prevent bacterial dissemination. Here we show 34 immune transcripts align to the morphology of lung sections from Mycobacterium tuberculosis-infected mice at cellular resolution. Colocalizing transcript networks at <10 μm in C57BL/6 mouse granulomas increase complexity with time after infection. B-cell clusters develop late after infection. Transcripts from activated macrophages are enriched at subcellular distances from M. tuberculosis. Encapsulated C3HeB/FeJ granulomas show necrotic centers with transcripts associated with immunosuppression (Foxp3, Il10), whereas those in the granuloma rims associate with activated T cells and macrophages. We see highly diverse networks with common interactors in similar lesions. Different immune landscapes of M. tuberculosis granulomas depending on the time after infection, the histopathological features of the lesion, and the proximity to bacteria are here defined.
Collapse
Affiliation(s)
- Berit Carow
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Thomas Hauling
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, 171 65, Solna, Sweden
| | - Xiaoyan Qian
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, 171 65, Solna, Sweden
| | - Igor Kramnik
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, 02118, USA
| | - Mats Nilsson
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, 171 65, Solna, Sweden
| | - Martin E Rottenberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden.
| |
Collapse
|
23
|
Lydiawati E, Sirithida C, Vannda S, Vortey H, Ratana H, Listiawan MY, Agusni I. LEPROSY AND HUMAN IMMUNODEFICIENCY VIRUS COINFECTION: A RARE CASE REPORT. INDONESIAN JOURNAL OF TROPICAL AND INFECTIOUS DISEASE 2019. [DOI: 10.20473/ijtid.v7i4.8869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Leprosy, or Hansen disease, is a chronic infectious disease caused by Mycobacterium leprae which is associated with inflammation that may damage the skin and the peripheral nerves. Leprosy remains an important public health problem in Southeast Asia, America, and Africa. It has been speculated that, as with TB, HIV infection may exacerbate leprosy lesions and/or lead to increased susceptibility to leprosy. We report the case of leprosy and HIV co-infection and reveals its clinical manifestation. A 34-year-old female came to outpatient clinic complaining of rednessplaque on her face of 2-months duration. It was also accompanied with thick sensation but without itchy or burning sensation. We found thick erythematous plaque with sharp margin and hypoesthesia on her face and body. There were no madarosis, saddle nose, lagophthalmos, nor sign of neuritis. The slit-skin smear revealed BI 1+ globi and MI 2%. From laboratory examination we found CBC was within normal limit, IgM anti PGL-1 titer was 1265 u/mL and IgG anti PGL-1 was 834 u/mL Both histological examination on her ear lobe and extremity revealed that was similar to the lesion of leprosy. The detection of HIV antibody was positive with CD4 count on 325 cells/μL. We treat her with MDT for MB leprosy along with ART (Tenofovir, Lamivudine, and Efavirenz). After 6-months follow-up we observed no progression of the lesions though the slit-skin smear after completing 6 months of therapy become negative. M. leprae does not seem to accelerate the decline of immune function when associated with HIV infection. HIV infection does not seem to affect the clinical classification and progression of leprosy. Treatment of the HIV-leprosy co-infected patient consists of the combination of ARTs and anti-leprosy agents. So that, the treatment of leprosy and HIV co-infection does not differ from that of a seronegative leprosy patient.
Collapse
|
24
|
Satti I, McShane H. Current approaches toward identifying a correlate of immune protection from tuberculosis. Expert Rev Vaccines 2018; 18:43-59. [PMID: 30466332 DOI: 10.1080/14760584.2019.1552140] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Mycobacterium tuberculosis kills more people than any other pathogen. Vaccination is the most cost-effective control measure for any infectious disease. Development of an effective vaccine against tuberculosis is hindered by the uncertain predictive value of preclinical animal models, incomplete understanding of protective immunity and lack of validated immune correlates of protection (COP). AREAS COVERED Here we review what is known about protective immunity against M.tb, the preclinical and clinical cohorts that can be utilized to identify COP, and COP that have been identified to date. EXPERT COMMENTARY The identification of COP would allow the rational design and development of vaccine candidates which can then be optimized and prioritized based on the induction of these immune responses. Once validated in field efficacy trials, such COP could potentially facilitate the development and licensure of vaccines, in combination with human efficacy data. The identification and validation of COP would represent a very significant advance in TB vaccine development. Every opportunity to collect samples and cohorts on which to cross-validate pre-existing COP and identify novel COP should be exploited. Furthermore, global cooperation and collaboration on such samples will ensure that the utility of such precious samples is fully exploited.
Collapse
Affiliation(s)
- Iman Satti
- a Jenner Institute, Nuffield Department of Medicine , University of Oxford , Oxford , UK
| | - Helen McShane
- a Jenner Institute, Nuffield Department of Medicine , University of Oxford , Oxford , UK
| |
Collapse
|
25
|
Abstract
Protective immunity in tuberculosis (TB) is subject of debate in the TB research community, as this is key to fully understand TB pathogenesis and to develop new promising tools for TB diagnosis and prognosis as well as a more efficient TB vaccine. IFN-γ producing CD4+ T cells are key in TB control, but may not be sufficient to provide protection. Additional subsets have been identified that contribute to protection such as multifunctional and cytolytic T-cell subsets, including classical and nonclassical T cells as well as novel innate immune cell subsets resulting from trained immunity. However, to define protective immune responses against TB, the complexity of balancing TB immunity also has to be considered. In this review, insights into effector cell immunity and how this is modulated by regulatory cells, associated comorbidities and the host microbiome, is discussed. We systematically map how different suppressive immune cell subsets may affect effector cell responses at the local site of infection. We also dissect how common comorbidities such as HIV, helminths and diabetes may bias protective TB immunity towards pathogenic and regulatory responses. Finally, also the composition and diversity of the microbiome in the lung and gut could affect host TB immunity. Understanding these various aspects of the immunological balance in the human host is fundamental to prevent TB infection and disease.
Collapse
Affiliation(s)
- Susanna Brighenti
- Karolinska Institutet, Department of Medicine, Center for Infectious Medicine (CIM), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Simone A. Joosten
- Leiden University Medical Center, Department of Infectious Diseases, Leiden, The Netherlands
| |
Collapse
|
26
|
Jeyanathan M, Yao Y, Afkhami S, Smaill F, Xing Z. New Tuberculosis Vaccine Strategies: Taking Aim at Un-Natural Immunity. Trends Immunol 2018; 39:419-433. [DOI: 10.1016/j.it.2018.01.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 01/02/2018] [Accepted: 01/16/2018] [Indexed: 12/13/2022]
|
27
|
Parasa VR, Muvva JR, Rose JF, Braian C, Brighenti S, Lerm M. Inhibition of Tissue Matrix Metalloproteinases Interferes with Mycobacterium tuberculosis-Induced Granuloma Formation and Reduces Bacterial Load in a Human Lung Tissue Model. Front Microbiol 2017; 8:2370. [PMID: 29259583 PMCID: PMC5723394 DOI: 10.3389/fmicb.2017.02370] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/16/2017] [Indexed: 12/30/2022] Open
Abstract
Granulomas are hallmarks of pulmonary tuberculosis (TB) and traditionally viewed as host-protective structures. However, recent evidence suggest that Mycobacterium tuberculosis (Mtb) uses its virulence factors to stimulate the formation of granuloma. In the present study, we investigated the contribution of matrix metalloproteinases (MMPs), host enzymes that cause degradation of the extracellular matrix, to granuloma formation and bacterial load in Mtb-infected tissue. To this end, we used our lung tissue model for TB, which is based on human lung-derived cells and primary human monocyte-derived macrophages. Global inhibition of MMPs in the Mtb-infected tissue model reduced both granuloma formation and bacterial load. The infection caused upregulation of a set of MMPs (MMP1, 3, 9, and 12), and this finding could be validated in lung biopsies from patients with non-cavitary TB. Data from this study indicate that MMP activation contributes to early TB granuloma formation, suggesting that host-directed, MMP-targeted intervention could be considered as adjunct therapy to TB treatment.
Collapse
Affiliation(s)
- Venkata R Parasa
- Division of Medical Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | | | - Jeronimo F Rose
- Division of Medical Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Clara Braian
- Division of Medical Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Susanna Brighenti
- Center for Infectious Medicine, Karolinska Institute, Stockholm, Sweden
| | - Maria Lerm
- Division of Medical Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
28
|
Parasa VR, Forsslund H, Enger T, Lorenz D, Kullberg S, Eklund A, Sköld M, Wahlström J, Grunewald J, Brighenti S. Enhanced CD8 + cytolytic T cell responses in the peripheral circulation of patients with sarcoidosis and non-Löfgren's disease. Respir Med 2017; 138S:S38-S44. [PMID: 29055517 DOI: 10.1016/j.rmed.2017.10.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 10/06/2017] [Accepted: 10/09/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND The role of CD4+ T cells in the immunopathogenesis of pulmonary sarcoidosis is well-established, while less is known about the phenotype and function of CD8+ cytolytic T cells (CTLs). METHODS CD8+ CTLs were explored in peripheral blood and bronchoalveolar lavage (BAL) samples obtained from up to 25 patients with sarcoidosis and 25 healthy controls. The proportion of CTLs was assessed by the expression of cytolytic effector molecules perforin, granzyme B and granulysin in CD8+ T cells, using flow cytometry. Cytolytic function in blood lymphocytes was assessed using a standard 51Cr-release assay. Patients with Löfgren´s syndrome (LS) and an acute disease onset, were compared to non-LS patients with an insidious onset. RESULTS Higher proportions of peripheral CD8+ CTLs expressing perforin and granzyme B were observed in sarcoidosis compared to healthy controls. Blood CTLs from non-LS patients had significantly higher expression of perforin, granzyme B and granulysin compared to matched BAL, while LS patients maintained lower levels of effector molecules in both compartments. Mitogen-stimulated peripheral lymphocytes from sarcoidosis patients, particularly from the non-LS group, showed a higher target cell lysis compared to controls. CONCLUSION These results demonstrated enhanced peripheral CD8+ CTL responses in sarcoidosis, especially in non-LS patients who have an increased risk of chronic disease. Further comprehensive clinical studies are warranted to increase our understanding of CD8+ CTL responses in sarcoidosis.
Collapse
Affiliation(s)
- Venkata Ramanarao Parasa
- Karolinska Institutet, Center for Infectious Medicine, Department of Medicine Huddinge, Stockholm, Sweden
| | - Helena Forsslund
- Karolinska Institutet, Respiratory Medicine Unit, Department of Medicine Solna, Stockholm, Sweden
| | - Tobias Enger
- Karolinska Institutet, Respiratory Medicine Unit, Department of Medicine Solna, Stockholm, Sweden
| | - Daniel Lorenz
- Karolinska Institutet, Respiratory Medicine Unit, Department of Medicine Solna, Stockholm, Sweden
| | - Susanna Kullberg
- Karolinska Institutet, Respiratory Medicine Unit, Department of Medicine Solna, Stockholm, Sweden
| | - Anders Eklund
- Karolinska Institutet, Respiratory Medicine Unit, Department of Medicine Solna, Stockholm, Sweden
| | - Magnus Sköld
- Karolinska Institutet, Respiratory Medicine Unit, Department of Medicine Solna, Stockholm, Sweden
| | - Jan Wahlström
- Karolinska Institutet, Respiratory Medicine Unit, Department of Medicine Solna, Stockholm, Sweden
| | - Johan Grunewald
- Karolinska Institutet, Respiratory Medicine Unit, Department of Medicine Solna, Stockholm, Sweden
| | - Susanna Brighenti
- Karolinska Institutet, Center for Infectious Medicine, Department of Medicine Huddinge, Stockholm, Sweden.
| |
Collapse
|
29
|
Modulation of PD-L1 and CD8 Activity in Idiopathic and Infectious Chronic Inflammatory Conditions. Appl Immunohistochem Mol Morphol 2017; 25:100-109. [PMID: 27438510 DOI: 10.1097/pai.0000000000000298] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Programmed death-ligand 1 (PD-L1) can reduce the immune response by inhibiting CD8 T-cell proliferation and cytotoxic activity. We studied a series of human viral (molloscum, human papillomavirus, herpes simplex, cytomegalovirus, Epstein-Barr virus, smallpox) and bacterial infections (Helicobacter pylori) for the in situ expression of PD-L1, mononuclear cell infiltration, and CD8 activity and compared this to noninfectious idiopathic inflammatory conditions to better define which immune responses may be more highly correlated with an infectious agent. Each viral and bacterial infection showed an increased PD-L1 expression that was most prominent in the mononuclear cell/CD8+ infiltrate surrounding the infection. However, the CD8 cells were mostly quiescent as evidenced by the low Ki67 index and minimal granzyme expression. Using a melanoma mouse model, acute reovirus infection increased PD-L1 expression, but decreased CD8 cytotoxic activity and Treg (FOXP3) cell numbers. In comparison, idiopathic noninfectious chronic inflammatory processes including lichen sclerosis, eczema, Sjogren's disease, and ulcerative colitis showed a comparable strong PD-L1 expression in the mononuclear cell infiltrates but much greater Treg infiltration. However, this strong immunosuppressor profile was ineffective as evidenced by strong CD8 proliferation and granzyme expression. These data suggest that viral and bacterial infections induce a PD-L1 response that, unlike noninfectious chronic inflammatory conditions, dampens the activity of the recruited CD8 cells which, in turn, may enhance the ability of anti-PD-L1 therapy to eliminate the infectious agent.
Collapse
|
30
|
Kinnear C, Hoal EG, Schurz H, van Helden PD, Möller M. The role of human host genetics in tuberculosis resistance. Expert Rev Respir Med 2017; 11:721-737. [PMID: 28703045 DOI: 10.1080/17476348.2017.1354700] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Tuberculosis (TB) remains a public health problem: the latest estimate of new incident cases per year is a staggering 10.4 million. Despite this overwhelming number, the majority of the immunocompetent population can control infection with Mycobacterium tuberculosis. The human genome underlies the immune response and contributes to the outcome of TB infection. Areas covered: Investigations of TB resistance in the general population have closely mirrored those of other infectious diseases and initially involved epidemiological observations. Linkage and association studies, including studies of VDR, SLC11A1 and HLA-DRB1 followed. Genome-wide association studies of common variants, not necessarily sufficient for disease, became possible after technological advancements. Other approaches involved the identification of those individuals with rare disease-causing mutations that strongly predispose to TB, epistasis and the role of ethnicity in disease. Despite these efforts, infection outcome, on an individual basis, cannot yet be predicted. Expert commentary: The early identification of future disease progressors is necessary to stem the TB epidemic. Human genetics may contribute to this endeavour and could in future suggest pathways to target for disease prevention. This will however require concerted efforts to establish large, well-phenotyped cohorts from different ethnicities, improved genomic resources and a better understanding of the human genome architecture.
Collapse
Affiliation(s)
- Craig Kinnear
- a SAMRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical TB Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences , Stellenbosch University , Cape Town , South Africa
| | - Eileen G Hoal
- a SAMRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical TB Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences , Stellenbosch University , Cape Town , South Africa
| | - Haiko Schurz
- a SAMRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical TB Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences , Stellenbosch University , Cape Town , South Africa
| | - Paul D van Helden
- a SAMRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical TB Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences , Stellenbosch University , Cape Town , South Africa
| | - Marlo Möller
- a SAMRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical TB Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences , Stellenbosch University , Cape Town , South Africa
| |
Collapse
|
31
|
Abstract
Immunity against Mycobacterium tuberculosis requires a balance between adaptive immune responses to constrain bacterial replication and the prevention of potentially damaging immune activation. Regulatory T (Treg) cells express the transcription factor Foxp3+ and constitute an essential counterbalance of inflammatory Th1 responses and are required to maintain immune homeostasis. The first reports describing the presence of Foxp3-expressing CD4+ Treg cells in tuberculosis (TB) emerged in 2006. Different Treg cell subsets, most likely specialized for different tissues and microenvironments, have been shown to expand in both human TB and animal models of TB. Recently, additional functional roles for Treg cells have been demonstrated during different stages and spectrums of TB disease. Foxp3+ regulatory cells can quickly expand during early infection and impede the onset of cellular immunity and persist during chronic TB infection. Increased frequencies of Treg cells have been associated with a detrimental outcome of active TB, and may be dependent on the M. tuberculosis strain, animal model, local environment, and the stage of infection. Some investigations also suggest that Treg cells are required together with effector T cell responses to obtain reduced pathology and sterilizing immunity. In this review, we will first provide an overview of the regulatory cells and mechanisms that control immune homeostasis. Then, we will review what is known about the phenotype and function of Treg cells from studies in human TB and experimental animal models of TB. We will discuss the potential role of Treg cells in the progression of TB disease and the relevance of this knowledge for future efforts to prevent, modulate, and treat TB.
Collapse
|
32
|
Busch M, Herzmann C, Kallert S, Zimmermann A, Höfer C, Mayer D, Zenk SF, Muche R, Lange C, Bloom BR, Modlin RL, Stenger S. Lipoarabinomannan-Responsive Polycytotoxic T Cells Are Associated with Protection in Human Tuberculosis. Am J Respir Crit Care Med 2017; 194:345-55. [PMID: 26882070 DOI: 10.1164/rccm.201509-1746oc] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE The development of host-targeted, prophylactic, and therapeutic interventions against tuberculosis requires a better understanding of the immune mechanisms that determine the outcome of infection with Mycobacterium tuberculosis. OBJECTIVES To identify T-cell-dependent mechanisms that are protective in tuberculosis. METHODS Multicolor flow cytometry, cell sorting and growth inhibition assays were employed to compare the frequency, phenotype and function of T lymphocytes from bronchoalveolar lavage or the peripheral blood. MEASUREMENTS AND MAIN RESULTS At two independent study sites, bronchoalveolar lavage cells from donors with latent tuberculosis infection limited the growth of virulent Mycobacterium tuberculosis more efficiently than those in patients who developed disease. Unconventional, glycolipid-responsive T cells contributed to reduced mycobacterial growth because antibodies to CD1b inhibited this effect by 55%. Lipoarabinomannan was the most potent mycobacterial lipid antigen (activation of 1.3% T lymphocytes) and activated CD1b-restricted T cells that limited bacterial growth. A subset of IFN-γ-producing lipoarabinomannan-responsive T cells coexpressed the cytotoxic molecules perforin, granulysin, and granzyme B, which we termed polycytotoxic T cells. Taking advantage of two well-defined cohorts of subjects latently infected with Mycobacterium tuberculosis or patients who developed active disease after infection, we found a correlation between the frequency of polycytotoxic T cells and the ability to control infection (latent tuberculosis infection, 62%; posttuberculosis patients, 26%). CONCLUSIONS Our data define an unconventional CD8(+) T-cell subset (polycytotoxic T cells) that is based on antigen recognition and function. The results link clinical and mechanistic evidence that glycolipid-responsive, polycytotoxic T cells contribute to protection against tuberculosis.
Collapse
Affiliation(s)
- Martin Busch
- 1 Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Christian Herzmann
- 2 Division of Clinical Infectious Diseases, German Center for Infection Research, Borstel, Germany
| | - Stephanie Kallert
- 1 Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Andreas Zimmermann
- 1 Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Christoph Höfer
- 1 Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Daniel Mayer
- 1 Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Sebastian F Zenk
- 1 Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Rainer Muche
- 3 Institute for Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Christoph Lange
- 2 Division of Clinical Infectious Diseases, German Center for Infection Research, Borstel, Germany
| | - Barry R Bloom
- 4 Harvard School of Public Health, Boston, Massachusetts; and
| | - Robert L Modlin
- 5 Division of Dermatology, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Steffen Stenger
- 1 Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | | |
Collapse
|
33
|
Tuberculous Lymphadenitis Is Associated with Enhanced Baseline and Antigen-Specific Induction of Type 1 and Type 17 Cytokines and Reduced Interleukin-1β (IL-1β) and IL-18 at the Site of Infection. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00045-17. [PMID: 28249842 DOI: 10.1128/cvi.00045-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 02/18/2017] [Indexed: 01/21/2023]
Abstract
Tuberculous lymphadenitis (TBL) is characterized by an expansion of Th1 and Th17 cells with altered serum levels of proinflammatory cytokines. However, the cytokine profile at the site of infection, i.e., the affected lymph nodes, has not been examined in detail. To estimate the baseline and mycobacterial antigen-stimulated concentrations of type 1, type 17, and other proinflammatory cytokines in patients with TBL (n = 14), we examined both the baseline and the antigen-specific concentrations of these cytokines before and after chemotherapy and compared them with those in individuals with pulmonary tuberculosis (PTB) (n = 14). In addition, we also compared the cytokine responses in whole blood and those in the lymph nodes of TBL individuals. We observed significantly enhanced baseline and antigen-specific levels of type 1 cytokines (gamma interferon [IFN-γ] and tumor necrosis factor alpha [TNF-α]) and a type 17 cytokine (interleukin-17 [IL-17]) and significantly diminished baseline and antigen-specific levels of proinflammatory cytokines (IL-1β and IL-18) in the whole blood of TBL individuals compared to those in the whole blood of PTB individuals. Moreover, we also observed a pattern of baseline and antigen-specific cytokine production at the site of infection (lymph node) similar to that in the whole blood of TBL individuals. Following standard antituberculosis (anti-TB) treatment, we observed alterations in the baseline and/or antigen-specific levels of IFN-γ, TNF-α, IL-1β, and IL-18. TBL is therefore characterized by enhanced baseline and antigen-specific production of type 1 and type 17 cytokines and reduced baseline and antigen-specific production of IL-1β and IL-18 at the site of infection.
Collapse
|
34
|
Valentini D, Rao M, Rane L, Rahman S, Axelsson-Robertson R, Heuchel R, Löhr M, Hoft D, Brighenti S, Zumla A, Maeurer M. Peptide microarray-based characterization of antibody responses to host proteins after bacille Calmette-Guérin vaccination. Int J Infect Dis 2017; 56:140-154. [PMID: 28161459 DOI: 10.1016/j.ijid.2017.01.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/19/2017] [Accepted: 01/22/2017] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Bacille Calmette-Guérin (BCG) is the world's most widely distributed vaccine, used against tuberculosis (TB), in cancer immunotherapy, and in autoimmune diseases due to its immunomodulatory properties. To date, the effect of BCG vaccination on antibody responses to host proteins has not been reported. High-content peptide microarrays (HCPM) offer a unique opportunity to gauge specific humoral immune responses. METHODS The sera of BCG-vaccinated healthy adults were tested on a human HCPM platform (4953 randomly selected epitopes of human proteins) to detect specific immunoglobulin gamma (IgG) responses. Samples were obtained at 56, 112, and 252 days after vaccination. Immunohistology was performed on lymph node tissue from patients with TB lymphadenitis. Results were analysed with a combination of existing and novel statistical methods. RESULTS IgG recognition of host peptides exhibited a peak at day 56 post BCG vaccination in all study subjects tested, which diminished over time. Primarily, IgG responses exhibited increased reactivity to ion transporters (sodium, calcium channels), cytokine receptors (interleukin 2 receptor β (IL2Rβ), fibroblast growth factor receptor 1 (FGFR1)), other cell surface receptors (inositol, somatostatin, angiopoeitin), ribonucleoprotein, and enzymes (tyrosine kinases, phospholipase) on day 56. There was decreased IgG reactivity to transforming growth factor-beta type 1 receptor (TGFβR1) and, in agreement with the peptide microarray findings, immunohistochemical analysis of TB-infected lymph node samples revealed an overexpression of TGFβR in granulomatous lesions. Moreover, the vesicular monoamine transporter (VMAT2) showed increased reactivity on days 112 and 252, but not on day 56 post-vaccination. IgG to interleukin 4 receptor (IL4R) showed increased reactivity at 112 days post-vaccination, while IgG to IL2Rβ and FGFR1 showed decreased reactivity on days 112 and 252 as compared to day 56 post BCG vaccination. CONCLUSIONS BCG vaccination modifies the host's immune landscape after 56 days, but this imprint changes over time. This may influence the establishment of immunological memory in BCG-vaccinated individuals.
Collapse
Affiliation(s)
- Davide Valentini
- Centre for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden; Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm, Sweden
| | - Martin Rao
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm, Sweden
| | - Lalit Rane
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm, Sweden
| | - Sayma Rahman
- Center for Infectious Medicine (CIM), Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rebecca Axelsson-Robertson
- Centre for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden; Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm, Sweden
| | - Rainer Heuchel
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Matthias Löhr
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Daniel Hoft
- Division of Immunobiology, Departments of Internal Medicine and Molecular Microbiology, Saint Louis University Medical Centre, Saint Louis, Missouri, USA
| | - Susanna Brighenti
- Center for Infectious Medicine (CIM), Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Alimuddin Zumla
- Centre for Clinical Microbiology, Division of Infection and Immunity, University College London, and NIHR Biomedical Research Centre, UCL Hospitals NHS Foundation Trust, London, UK
| | - Markus Maeurer
- Centre for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden; Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
35
|
Abstract
Peptide-specific conventional T cells have been major targets for designing most antimycobacterial vaccines. Immune responses mediated by conventional T cells exhibit a delayed onset upon primary infection and are highly variable in different human populations. In contrast, innate-like T cells quickly respond to pathogens and display effector functions without undergoing extensive clonal expansion. Specifically, the activation of innate-like T cells depends on the promiscuous interaction of highly conserved antigen-presenting molecules, non-peptidic antigens, and likely semi-invariant T cell receptors. In antimicrobial immune responses, mucosal-associated invariant T cells are activated by riboflavin precursor metabolites presented by major histocompatibility complex-related protein I, while lipid-specific T cells including natural killer T cells are activated by lipid metabolites presented by CD1 proteins. Multiple innate-like T cell subsets have been shown to be protective or responsive in mycobacterial infections. Through rapid cytokine secretion, innate-like T cells function in early defense and memory response, offering novel advantages over conventional T cells in the design of anti-tuberculosis strategies.
Collapse
Affiliation(s)
- Shouxiong Huang
- Department of Environmental Health, University of Cincinnati College of Medicine , Cincinnati, OH , USA
| |
Collapse
|
36
|
Javan MR, Jalali nezhad AA, Shahraki S, Safa A, Aali H, Kiani Z. Cross-talk between the Immune System and Tuberculosis Pathogenesis; a Review with Emphasis on the Immune Based Treatment. INTERNATIONAL JOURNAL OF BASIC SCIENCE IN MEDICINE 2016. [DOI: 10.15171/ijbsm.2016.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
|
37
|
Cao Y, Wang X, Cao Z, Cheng X. Vitamin D receptor gene FokI polymorphisms and tuberculosis susceptibility: a meta-analysis. Arch Med Sci 2016; 12:1118-1134. [PMID: 27695504 PMCID: PMC5016579 DOI: 10.5114/aoms.2016.60092] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 05/27/2015] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION The association between FokI polymorphism of vitamin D receptor (VDR) and tuberculosis (TB) susceptibility has been investigated previously; however, the results were inconsistent and conflicting. In the present study, a meta-analysis was performed to assess the relationship between VDR FokI gene polymorphism and the risk of TB. MATERIAL AND METHODS Databases including PubMed and Embase were searched for genetic association studies of FokI polymorphism of vitamin D receptor (VDR) and TB. Data were extracted by two independent authors and the pooled odds ratio (OR) with 95% confidence interval (CI) was calculated to assess the strength of the association between VDR FokI gene polymorphism and TB risk. Meta-regression and subgroup analyses were performed to identify the source of heterogeneity. RESULTS Thirty-four studies with a total of 5669 cases and 6525 controls were reviewed in the present meta-analysis. A statistically significant correlation was found between VDR FokI gene polymorphism and increased TB risk in two comparison models: the homozygote model (ff vs. FF: OR = 1.37, 95% CI: 1.17-1.60; Pheterogeneity = 0.001) and the recessive model (ff vs. Ff + FF: OR = 1.32, 95% CI: 1.14-1.52; Pheterogeneity = 0.006). Meta-regression found no source contributing to heterogeneity. However, sub-group analyses revealed that there was a statistically increased TB risk in the East and Southeast Asian population. CONCLUSIONS Synthesis of the available studies suggests that homozygosity for the FokI polymorphism of the VDR gene might be associated with an increased TB risk, especially in the East and Southeast Asian population. Additional well-designed, larger-scale epidemiological studies among different ethnicities are needed.
Collapse
Affiliation(s)
- Yan Cao
- Key Laboratory of Tuberculosis Prevention and Treatment, and Beijing Key Laboratory of New Techniques for Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis, 309th Hospital, Beijing, China
| | - Xinjing Wang
- Key Laboratory of Tuberculosis Prevention and Treatment, and Beijing Key Laboratory of New Techniques for Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis, 309th Hospital, Beijing, China
| | - Zhihong Cao
- Key Laboratory of Tuberculosis Prevention and Treatment, and Beijing Key Laboratory of New Techniques for Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis, 309th Hospital, Beijing, China
| | - Xiaoxing Cheng
- Key Laboratory of Tuberculosis Prevention and Treatment, and Beijing Key Laboratory of New Techniques for Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis, 309th Hospital, Beijing, China
| |
Collapse
|
38
|
Host-directed therapies for antimicrobial resistant respiratory tract infections. Curr Opin Pulm Med 2016; 22:203-11. [DOI: 10.1097/mcp.0000000000000271] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
39
|
Abstract
Adaptive immunity towards tuberculosis (TB) has been extensively studied for many years. In addition, in recent years the profound contribution of innate immunity to host defence against this disease has become evident. The discovery of pattern recognition receptors, which allow innate immunity to tailor its response to different infectious agents, has challenged the view that this arm of immunity is nonspecific. Evidence is now accumulating that innate immunity can remember a previous exposure to a microorganism and respond differently during a second exposure. Although the specificity and memory of innate immunity cannot compete with the highly sophisticated adaptive immune response, its contribution to host defence against infection and to vaccine-induced immunity should not be underestimated and needs to be explored. Here, we present the concept of trained immunity and discuss how this may contribute to new avenues for control of TB.
Collapse
Affiliation(s)
- M Lerm
- Division of Microbiology and Molecular Medicine, Faculty of Medicine and Health Sciences, Linköping, Sweden
| | - M G Netea
- Radboud Institute for Molecular Life Sciences, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
40
|
Abstract
T regulatory cells (Treg) constitute a specialized subset of T cells that play a pivotal role in preventing the occurrence of autoimmune diseases by suppressing deleterious activities of immune cells. Contrarily, they can have adverse effect on immune response against infectious diseases where Treg weaken the host immunity leading to enhanced microbial load and thereby increase in severity of the disease. Here, we have attempted to review plethora of information documenting prevalence of Treg in tuberculosis (TB) and their involvement in progression and immunopathogenesis of the disease. Further, we have laid emphasis on the possible use of Treg as a biomarker for determining the TB treatment efficacy. Also, we have discussed the probable contribution of Treg in dampening the efficacy of BCG, the anti-TB vaccine. Finally, we have speculated some of the possible strategies which might be explored by exploiting Treg for enhancing the efficacy of TB management.
Collapse
|
41
|
Axelsson-Robertson R, Rao M, Loxton AG, Walzl G, Bates M, Zumla A, Maeurer M. Frequency of Mycobacterium tuberculosis-specific CD8+ T-cells in the course of anti-tuberculosis treatment. Int J Infect Dis 2016; 32:23-9. [PMID: 25809751 DOI: 10.1016/j.ijid.2015.01.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 01/16/2015] [Accepted: 01/16/2015] [Indexed: 12/30/2022] Open
Abstract
Anti-tuberculosis drug treatment is known to affect the number, phenotype, and effector functionality of antigen-specific T-cells. In order to objectively gauge Mycobacterium tuberculosis (MTB)-specific CD8+ T-cells at the single-cell level, we developed soluble major histocompatibility complex (MHC) class I multimers/peptide multimers, which allow analysis of antigen-specific T-cells without ex vivo manipulation or functional tests. We constructed 38 MHC class I multimers covering some of the most frequent MHC class I alleles (HLA-A*02:01, A*24:02, A*30:01, A*30:02, A*68:01, B*58:01, and C*07:01) pertinent to a South African or Zambian population, and presenting the following MTB-derived peptides: the early expressed secreted antigens TB10.4 (Rv0288), Ag85B (Rv1886c), and ESAT-6 (Rv3875), as well as intracellular enzymes, i.e., glycosyltransferase 1 (Rv2957), glycosyltransferase 2 (Rv2958c), and cyclopropane fatty acid synthase (Rv0447c). Anti-TB treatment appeared to impact on the frequency of multimer-positive CD8+ T-cells, with a general decrease after 6 months of therapy. Also, a reduction in the total central memory CD8+ T-cell frequencies, as well as the antigen-specific compartment in CD45RA-CCR7+ T-cells was observed. We discuss our findings on the basis of differential dynamics of MTB-specific T-cell frequencies, impact of MTB antigen load on T-cell phenotype, and antigen-specific T-cell responses in tuberculosis.
Collapse
Affiliation(s)
- Rebecca Axelsson-Robertson
- Centre for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden
| | - Martin Rao
- Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine (LABMED), Karolinska Institutet, Hälsovägen F79, Karolinska University Hospital Huddinge Campus, SE14186, Stockholm, Sweden
| | - Andre G Loxton
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research and MRC Centre for Molecular and Cellular Biology, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Health Sciences, Stellenbosch University, South Africa
| | - Gerhard Walzl
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research and MRC Centre for Molecular and Cellular Biology, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Health Sciences, Stellenbosch University, South Africa
| | - Matthew Bates
- Division of Infection and Immunity, University College London, London, UK; UNZA-UCLMs Research and Training Project, University Teaching Hospital, Lusaka, Zambia
| | - Alimuddin Zumla
- Division of Infection and Immunity, University College London, London, UK; UNZA-UCLMs Research and Training Project, University Teaching Hospital, Lusaka, Zambia; NIHR Biomaedical Research Centre at University College London Hospitals, London, UK
| | - Markus Maeurer
- Centre for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden; Division of Therapeutic Immunology (TIM), Department of Laboratory Medicine (LABMED), Karolinska Institutet, Hälsovägen F79, Karolinska University Hospital Huddinge Campus, SE14186, Stockholm, Sweden.
| |
Collapse
|
42
|
Zenk SF, Vollmer M, Schercher E, Kallert S, Kubis J, Stenger S. Hypoxia promotes Mycobacterium tuberculosis-specific up-regulation of granulysin in human T cells. Med Microbiol Immunol 2015; 205:219-29. [PMID: 26613797 DOI: 10.1007/s00430-015-0442-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 11/16/2015] [Indexed: 12/18/2022]
Abstract
Oxygen tension affects local immune responses in inflammation and infection. In tuberculosis mycobacteria avoid hypoxic areas and preferentially persist and reactivate in the oxygen-rich apex of the lung. Oxygen restriction activates antimicrobial effector mechanisms in macrophages and restricts growth of intracellular Mycobacterium tuberculosis (M.Tb). The effect of oxygen restriction on T cell-mediated antimicrobial effector mechanisms is unknown. Therefore we determined the influence of hypoxia on the expression of granulysin, an antimicrobial peptide of lymphocytes. Hypoxia increased the antigen-specific up-regulation of granulysin mRNA and protein in human CD4(+) and CD8(+) T lymphocytes. This observation was functionally relevant, because oxygen restriction supported the growth-limiting effect of antigen-specific T cells against virulent M.Tb residing in primary human macrophages. Our results provide evidence that oxygen restriction promotes the expression of granulysin and suggest that this effect-in conjunction with additional T cell-mediated immune responses-supports protection against mycobacteria. The therapeutic modulation of oxygen availability may offer a new strategy for the host-directed therapy of infectious diseases with intracellular pathogens.
Collapse
Affiliation(s)
- Sebastian F Zenk
- Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Michael Vollmer
- Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Esra Schercher
- Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Stephanie Kallert
- Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Jan Kubis
- Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Steffen Stenger
- Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany.
| |
Collapse
|
43
|
Jasenosky LD, Scriba TJ, Hanekom WA, Goldfeld AE. T cells and adaptive immunity to Mycobacterium tuberculosis in humans. Immunol Rev 2015; 264:74-87. [PMID: 25703553 DOI: 10.1111/imr.12274] [Citation(s) in RCA: 243] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The adaptive immune response mediated by T cells is critical for control of Mycobacterium tuberculosis (M. tuberculosis) infection in humans. However, the M. tuberculosis antigens and host T-cell responses that are required for an effective adaptive immune response to M. tuberculosis infection are yet to be defined. Here, we review recent findings on CD4(+) and CD8(+) T-cell responses to M. tuberculosis infection and examine the roles of distinct M. tuberculosis-specific T-cell subsets in control of de novo and latent M. tuberculosis infection, and in the evolution of T-cell immunity to M. tuberculosis in response to tuberculosis treatment. In addition, we discuss recent studies that elucidate aspects of M. tuberculosis-specific adaptive immunity during human immunodeficiency virus co-infection and summarize recent findings from vaccine trials that provide insight into effective adaptive immune responses to M. tuberculosis infection.
Collapse
Affiliation(s)
- Luke D Jasenosky
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | |
Collapse
|
44
|
Abstract
Our understanding of the host-pathogen relationship in tuberculosis (TB) can help guide drug discovery in at least two ways. First, the recognition that host immunopathology affects lesional TB drug distribution means that pharmacokinetic evaluation of drug candidates needs to move beyond measurements of drug levels in blood, whole lungs, or alveolar epithelial lining fluid to include measurements in specific types of lesions. Second, by restricting the replication of M. tuberculosis (Mtb) subpopulations in latent TB infection and in active disease, the host immune response puts Mtb into a state associated with phenotypic tolerance to TB drugs selected for their activity against replicating Mtb. This has spurred a major effort to conduct high throughput screens in vitro for compounds that can kill Mtb when it is replicating slowly if at all. Each condition used in vitro to slow Mtb's replication and thereby model the phenotypically drug-tolerant state has advantages and disadvantages. Lead candidates emerging from such in vitro studies face daunting challenges in the design of proof-of-concept studies in animal models. Moreover, some non-replicating subpopulations of Mtb fail to resume replication when plated on agar, although their viability is demonstrable by other means. There is as yet no widely replicated assay in which to screen compounds for their ability to kill this 'viable but non-culturable' subpopulation. Despite these hurdles, drugs that can kill slowly replicating or non-replicating Mtb may offer our best hope for treatment-shortening combination chemotherapy of TB.
Collapse
Affiliation(s)
- Carl Nathan
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY, USA
| | | |
Collapse
|
45
|
Liaquat A, Iram S, Hussain S, Yusuf NW, Azeem H. Concomitant presence of culture-proven active pulmonary tuberculosis in patients with chronic obstructive pulmonary disease - A hospital based study. Pak J Med Sci 2015; 31:1344-8. [PMID: 26870094 PMCID: PMC4744279 DOI: 10.12669/pjms.316.8166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 05/08/2015] [Accepted: 09/03/2015] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE To find out the prevalence of concomitant active pulmonary Tuberculosis (TB) in patients of Chronic Obstructive Pulmonary Disease (COPD) using the gold standard liquid and solid culture media for the detection of acid fast bacillus. METHODS Eighty clinically and radiologically diagnosed cases of COPD of any severity, ≥40 years of age with no previous history of anti-tuberculous therapy were selected from department of Pulmonology, Jinnah Hospital, Lahore. Detailed demographic profile, clinical symptomatology and history of smoking were recorded. Sputum samples of these patients were subjected to ZiehlNeelsen (ZN) stain and culture on Lowenstein-Jensen (L.J) medium and Mycobacterium Growth Indicator Tube (MGIT) for the detection and isolation of Mycobacteriumtuberculosis (MTB). RESULTS Out of 80 COPD patients, 6 (7.5%) were culture positive for acid fast bacillus consistent with active tuberculous infection. The concomitance was more prevalent in elderly, male, smokers. MGIT was a more sensitive and a rapid technique to detect the presence of mycobacterium as compared to LJ culture media and ZN stain. CONCLUSION The prevalence of active TB in COPD patients was 7.5%. Detection was improved when liquid culture media was employed for the detection of acid fast bacillus. Regular monitoring and screening of patients with COPD for PTB should be routinely carried out in susceptible cohort to avoid cross spreading of infection and appropriate management.
Collapse
Affiliation(s)
- Aneela Liaquat
- Aneela Liaquat, M.Phil Microbiology. Govt. College University Lahore, Pakistan
| | - Shagufta Iram
- Shagufta Iram, M.Phil Microbiology, Assistant Professor, Dept. of Pathology, Allama Iqbal Medical College, Lahore, Pakistan
| | - Shahida Hussain
- Shahida Hussain, M.Phil Biotechnology, Dept. of Pathology, Allama Iqbal Medical College, Lahore, Pakistan
| | - Noshin Wasim Yusuf
- Noshin Wasim Yusuf, MBBS, MRC.Path, FRC.Path (UK), Professor of Pathology, Rashid Lateef Medical College, Lahore, Pakistan
| | - Hassan Azeem
- Hassan Azeem, MBBS, Dept. of Pulmonology. Jinnah Hospital Lahore, Pakistan
| |
Collapse
|
46
|
Braian C, Svensson M, Brighenti S, Lerm M, Parasa VR. A 3D Human Lung Tissue Model for Functional Studies on Mycobacterium tuberculosis Infection. J Vis Exp 2015. [PMID: 26485646 PMCID: PMC4692636 DOI: 10.3791/53084] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Tuberculosis (TB) still holds a major threat to the health of people worldwide, and there is a need for cost-efficient but reliable models to help us understand the disease mechanisms and advance the discoveries of new treatment options. In vitro cell cultures of monolayers or co-cultures lack the three-dimensional (3D) environment and tissue responses. Herein, we describe an innovative in vitro model of a human lung tissue, which holds promise to be an effective tool for studying the complex events that occur during infection with Mycobacterium tuberculosis (M. tuberculosis). The 3D tissue model consists of tissue-specific epithelial cells and fibroblasts, which are cultured in a matrix of collagen on top of a porous membrane. Upon air exposure, the epithelial cells stratify and secrete mucus at the apical side. By introducing human primary macrophages infected with M. tuberculosis to the tissue model, we have shown that immune cells migrate into the infected-tissue and form early stages of TB granuloma. These structures recapitulate the distinct feature of human TB, the granuloma, which is fundamentally different or not commonly observed in widely used experimental animal models. This organotypic culture method enables the 3D visualization and robust quantitative analysis that provides pivotal information on spatial and temporal features of host cell-pathogen interactions. Taken together, the lung tissue model provides a physiologically relevant tissue micro-environment for studies on TB. Thus, the lung tissue model has potential implications for both basic mechanistic and applied studies. Importantly, the model allows addition or manipulation of individual cell types, which thereby widens its use for modelling a variety of infectious diseases that affect the lungs.
Collapse
Affiliation(s)
- Clara Braian
- Department of Clinical and Experimental Medicine, Linköping University
| | | | | | - Maria Lerm
- Department of Clinical and Experimental Medicine, Linköping University;
| | - Venkata R Parasa
- Department of Clinical and Experimental Medicine, Linköping University; Department of Medicine, Karolinska Institute
| |
Collapse
|
47
|
Subbian S, Tsenova L, Kim MJ, Wainwright HC, Visser A, Bandyopadhyay N, Bader JS, Karakousis PC, Murrmann GB, Bekker LG, Russell DG, Kaplan G. Lesion-Specific Immune Response in Granulomas of Patients with Pulmonary Tuberculosis: A Pilot Study. PLoS One 2015; 10:e0132249. [PMID: 26133981 PMCID: PMC4489805 DOI: 10.1371/journal.pone.0132249] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 06/11/2015] [Indexed: 01/20/2023] Open
Abstract
The formation and maintenance of granulomas is central to the host response to Mycobacterium tuberculosis (Mtb) infection. It is widely accepted that the lungs of patients with tuberculosis (TB) usually contain multiple infection foci, and that the granulomas evolve and differentiate independently, resulting in considerable heterogeneity. Although gene expression profiles of human blood cells have been proposed as biomarkers of Mtb infection and/or active disease, the immune profiles of discrete lesion types has not been studied extensively. Using histology, immunopathology and genome-wide transcriptome analysis, we explored the immunological profile of human lung TB granulomas. We show that although the different granulomas share core similarities in their immunological/inflammatory characteristics, they also exhibit significant divergence. Despite similar numbers of CD68+ macrophages in the different lesions, the extent of immune reactivity, as determined by the density of CD3+ T cells in the macrophage rich areas, and the extent of fibrosis, shows considerable variation. Both quantitative and qualitative differences among significantly differentially expressed genes (SDEG) were noted in each of the lesion types studied. Further, network/pathway analysis of SDEG revealed differential regulation of inflammatory response, immune cell trafficking, and cell mediated immune response in the different lesions. Our data highlight the formidable challenges facing ongoing efforts to identify peripheral blood biomarkers due to the diversity of lesion types and complexity of local immune responses in the lung.
Collapse
MESH Headings
- Cellular Microenvironment
- Fibrosis
- Gene Expression Profiling
- Granuloma, Respiratory Tract/genetics
- Granuloma, Respiratory Tract/immunology
- Granuloma, Respiratory Tract/pathology
- Humans
- Inflammation
- Interleukin-7/physiology
- Lung/pathology
- Lymphocyte Activation
- Macrophages/immunology
- Necrosis
- Pilot Projects
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- RNA, Messenger/isolation & purification
- Receptors, Calcitriol/physiology
- STAT1 Transcription Factor/physiology
- Signal Transduction
- T-Lymphocyte Subsets/immunology
- Transcriptome
- Tuberculosis, Multidrug-Resistant/genetics
- Tuberculosis, Multidrug-Resistant/immunology
- Tuberculosis, Multidrug-Resistant/pathology
- Tuberculosis, Pulmonary/genetics
- Tuberculosis, Pulmonary/immunology
- Tuberculosis, Pulmonary/pathology
Collapse
Affiliation(s)
- Selvakumar Subbian
- Laboratory of Mycobacterial Immunity and Pathogenesis, Public Health Research Institute (PHRI), Rutgers Biomedical and Health Sciences, Rutgers, The State University of New Jersey, Newark, New Jersey, United States of America
- * E-mail:
| | - Liana Tsenova
- Laboratory of Mycobacterial Immunity and Pathogenesis, Public Health Research Institute (PHRI), Rutgers Biomedical and Health Sciences, Rutgers, The State University of New Jersey, Newark, New Jersey, United States of America
- Department of Biological Sciences, NYC College of Technology, Brooklyn, New York, United States of America
| | - Mi-Jeong Kim
- Department of Immunobiology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Helen C. Wainwright
- Division of Anatomical Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Annalie Visser
- Division of Anatomical Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Nirmalya Bandyopadhyay
- Department of Biomedical Engineering, High-Throughput Biology Center and Institute of Computational Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Joel S. Bader
- Department of Biomedical Engineering, High-Throughput Biology Center and Institute of Computational Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Petros C. Karakousis
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine and Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Gabriele B. Murrmann
- Department of General and Thoracic Surgery, Medisch Centrum Leeuwarden, Leeuwarden, The Netherlands
| | - Linda-Gail Bekker
- The Desmond Tutu HIV Centre, Institute of Infectious Disease and Molecular Medicine and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - David G. Russell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Gilla Kaplan
- Bill and Melinda Gates Foundation, Seattle, Washington, United States of America
| |
Collapse
|
48
|
Suarez GV, Angerami MT, Vecchione MB, Laufer N, Turk G, Ruiz MJ, Mesch V, Fabre B, Maidana P, Ameri D, Cahn P, Sued O, Salomón H, Bottasso OA, Quiroga MF. HIV-TB coinfection impairs CD8(+) T-cell differentiation and function while dehydroepiandrosterone improves cytotoxic antitubercular immune responses. Eur J Immunol 2015; 45:2529-41. [PMID: 26047476 DOI: 10.1002/eji.201545545] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 05/08/2015] [Accepted: 06/01/2015] [Indexed: 12/14/2022]
Abstract
Tuberculosis (TB) is the leading cause of death among HIV-positive patients. The decreasing frequencies of terminal effector (TTE ) CD8(+) T cells may increase reactivation risk in persons latently infected with Mycobacterium tuberculosis (Mtb). We have previously shown that dehydroepiandrosterone (DHEA) increases the protective antitubercular immune responses in HIV-TB patients. Here, we aimed to study Mtb-specific cytotoxicity, IFN-γ secretion, memory status of CD8(+) T cells, and their modulation by DHEA during HIV-TB coinfection. CD8(+) T cells from HIV-TB patients showed a more differentiated phenotype with diminished naïve and higher effector memory and TTE T-cell frequencies compared to healthy donors both in total and Mtb-specific CD8(+) T cells. Notably, CD8(+) T cells from HIV-TB patients displayed higher Terminal Effector (TTE ) CD45RA(dim) proportions with lower CD45RA expression levels, suggesting a not fully differentiated phenotype. Also, PD-1 expression levels on CD8(+) T cells from HIV-TB patients increased although restricted to the CD27(+) population. Interestingly, DHEA plasma levels positively correlated with TTE in CD8(+) T cells and in vitro DHEA treatment enhanced Mtb-specific cytotoxic responses and terminal differentiation in CD8(+) T cells from HIV-TB patients. Our data suggest that HIV-TB coinfection promotes a deficient CD8(+) T-cell differentiation, whereas DHEA may contribute to improving antitubercular immunity by enhancing CD8(+) T-cell functions during HIV-TB coinfection.
Collapse
Affiliation(s)
- Guadalupe V Suarez
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Universidad de Buenos Aires - CONICET, Argentina
| | - Matías T Angerami
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Universidad de Buenos Aires - CONICET, Argentina
| | - María B Vecchione
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Universidad de Buenos Aires - CONICET, Argentina
| | - Natalia Laufer
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Universidad de Buenos Aires - CONICET, Argentina.,Hospital Juan A. Fernández, Buenos Aires, Argentina
| | - Gabriela Turk
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Universidad de Buenos Aires - CONICET, Argentina
| | - Maria J Ruiz
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Universidad de Buenos Aires - CONICET, Argentina
| | - Viviana Mesch
- Departamento de Bioquímica Clínica, INFIBIOC, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina
| | - Bibiana Fabre
- Departamento de Bioquímica Clínica, INFIBIOC, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina
| | - Patricia Maidana
- Departamento de Bioquímica Clínica, INFIBIOC, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina
| | - Diego Ameri
- Hospital Juan A. Fernández, Buenos Aires, Argentina
| | - Pedro Cahn
- Hospital Juan A. Fernández, Buenos Aires, Argentina.,Área de Investigaciones Médicas, Fundación Huésped, Buenos Aires, Argentina
| | - Omar Sued
- Área de Investigaciones Médicas, Fundación Huésped, Buenos Aires, Argentina
| | - Horacio Salomón
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Universidad de Buenos Aires - CONICET, Argentina
| | - Oscar A Bottasso
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), CONICET-UNR, Rosario, Santa Fe, Argentina
| | - María F Quiroga
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Universidad de Buenos Aires - CONICET, Argentina
| |
Collapse
|
49
|
Ahuja V, Subodh S, Tuteja A, Mishra V, Garg SK, Gupta N, Makharia G, Acharya SK. Genome-wide gene expression analysis for target genes to differentiate patients with intestinal tuberculosis and Crohn's disease and discriminative value of FOXP3 mRNA expression. Gastroenterol Rep (Oxf) 2015; 4:59-67. [PMID: 25969456 PMCID: PMC4760064 DOI: 10.1093/gastro/gov015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/18/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND AIMS Crohn's disease (CD) and intestinal tuberculosis (ITB) are both chronic granulomatous conditions with similar phenotypic presentations. Hence, there is need for a biomarker to differentiate between both these two diseases. This study aimed at genome-wide gene expression analysis of colonic biopsies from confirmed cases of ITB and CD in comparison with controls. To evaluate the role of T regulatory cells, forkhead box P3 (FOXP3) mRNA expression was quantified in serum as well as in colonic biopsies from patients with ITB and with the controls. METHODS Paired samples, including serum and colonic biopsies, were taken from 33 study subjects (CD, ITB and controls), and total RNA was extracted. Human whole genome gene expression microarray analysis was performed using the Illumina HumanWG-6 BeadChip Kit with six total RNA samples of the three groups in duplicates. Real-time PCR for FOXP3 mRNA expression was analyzed in serum samples and colonic biopsy samples (4-CD, 5-ITB, 4-controls). RESULTS In CD and ITB there was 1.5-fold upregulation of 92 and 382 genes and 1.5-fold downregulation of 91 and 256 genes, respectively. Peroxisome proliferators via the PPARγ pathway were most significantly downregulated (P < 0.005) in CD. Additionally, the IL4/5/6 signaling pathways and Toll-like receptor signaling pathway were identified as significantly differentially regulated (P < 0.005) at > 2-fold change. In ITB, the complement activation pathway, specifically the classical pathway, was the most significantly upregulated. FOXP3 mRNA expression was significantly elevated in colonic biopsies obtained from ITB patients as compared with CD cases (4.70 ± 2.21 vs 1.48 ± 0.31, P = 0.016). CONCLUSIONS FOXP3 mRNA expression in colonic mucosa could be a discriminatory marker between ITB and CD. Upregulation of the complement activation pathway in ITB suggests that pathogenetic mechanisms for ITB are similar to those of pulmonary tuberculosis. In CD, downregulation of PPARγ was seen in colonic tissue, suggesting that restoration of PPARγ-dependent anti-microbial barrier function may be a therapeutic target.
Collapse
Affiliation(s)
- Vineet Ahuja
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, India, and
| | - Swati Subodh
- The Centre for Genomic Application (An IGIB-IMM collaboration), New Delhi, India
| | - Amit Tuteja
- The Centre for Genomic Application (An IGIB-IMM collaboration), New Delhi, India
| | - Veena Mishra
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, India, and
| | - Sushil Kumar Garg
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, India, and
| | - Neha Gupta
- The Centre for Genomic Application (An IGIB-IMM collaboration), New Delhi, India
| | - Govind Makharia
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, India, and
| | - S K Acharya
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, India, and
| |
Collapse
|
50
|
Boer MC, Joosten SA, Ottenhoff THM. Regulatory T-Cells at the Interface between Human Host and Pathogens in Infectious Diseases and Vaccination. Front Immunol 2015; 6:217. [PMID: 26029205 PMCID: PMC4426762 DOI: 10.3389/fimmu.2015.00217] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 04/20/2015] [Indexed: 12/20/2022] Open
Abstract
Regulatory T-cells (Tregs) act at the interface of host and pathogen interactions in human infectious diseases. Tregs are induced by a wide range of pathogens, but distinct effects of Tregs have been demonstrated for different pathogens and in different stages of infection. Moreover, Tregs that are induced by a specific pathogen may non-specifically suppress immunity against other microbes and parasites. Thus, Treg effects need to be assessed not only in homologous but also in heterologous infections and vaccinations. Though Tregs protect the human host against excessive inflammation, they probably also increase the risk of pathogen persistence and chronic disease, and the possibility of disease reactivation later in life. Mycobacterium leprae and Mycobacterium tuberculosis, causing leprosy and tuberculosis, respectively, are among the most ancient microbes known to mankind, and are master manipulators of the immune system toward tolerance and pathogen persistence. The majority of mycobacterial infections occur in settings co-endemic for viral, parasitic, and (other) bacterial coinfections. In this paper, we discuss recent insights in the activation and activity of Tregs in human infectious diseases, with emphasis on early, late, and non-specific effects in disease, coinfections, and vaccination. We highlight mycobacterial infections as important models of modulation of host responses and vaccine-induced immunity by Tregs.
Collapse
Affiliation(s)
- Mardi C Boer
- Department of Infectious Diseases, Leiden University Medical Center , Leiden , Netherlands
| | - Simone A Joosten
- Department of Infectious Diseases, Leiden University Medical Center , Leiden , Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center , Leiden , Netherlands
| |
Collapse
|