51
|
Tran AC, Diogo GR, Paul MJ, Copland A, Hart P, Mehta N, Irvine EB, Mussá T, Drake PMW, Ivanyi J, Alter G, Reljic R. Mucosal Therapy of Multi-Drug Resistant Tuberculosis With IgA and Interferon-γ. Front Immunol 2020; 11:582833. [PMID: 33193394 PMCID: PMC7606302 DOI: 10.3389/fimmu.2020.582833] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/29/2020] [Indexed: 12/31/2022] Open
Abstract
New evidence has been emerging that antibodies can be protective in various experimental models of tuberculosis. Here, we report on protection against multidrug-resistant Mycobacterium tuberculosis (MDR-TB) infection using a combination of the human monoclonal IgA 2E9 antibody against the alpha-crystallin (Acr, HspX) antigen and mouse interferon-gamma in mice transgenic for the human IgA receptor, CD89. The effect of the combined mucosal IgA and IFN-γ; treatment was strongest (50-fold reduction) when therapy was applied at the time of infection, but a statistically significant reduction of lung bacterial load was observed even when the therapy was initiated once the infection had already been established. The protection involving enhanced phagocytosis and then neutrophil mediated killing of infected cells was IgA isotype mediated, because treatment with an IgG version of 2E9 antibody was not effective in human IgG receptor CD64 transgenic mice. The Acr antigen specificity of IgA antibodies for protection in humans has been indicated by their elevated serum levels in latent tuberculosis unlike the lack of IgA antibodies against the virulence-associated MPT64 antigen. Our results represent the first evidence for potential translation of mucosal immunotherapy for the management of MDR-TB.
Collapse
Affiliation(s)
- Andy C Tran
- Institute for Infection and Immunity, St. George's University, London, United Kingdom
| | - Gil R Diogo
- Institute for Infection and Immunity, St. George's University, London, United Kingdom
| | - Matthew J Paul
- Institute for Infection and Immunity, St. George's University, London, United Kingdom
| | - Alastair Copland
- Institute for Infection and Immunity, St. George's University, London, United Kingdom
| | - Peter Hart
- Institute for Infection and Immunity, St. George's University, London, United Kingdom
| | - Nickita Mehta
- Ragon Institute, Harvard, Cambridge, MA, United States
| | | | - Tufária Mussá
- Department of Microbiology, Faculty of Medicine, Eduardo Mondlane University, Maputo, Mozambique.,Departamento de Plataformas Tecnológicas em Saúde, Instituto Nacional de Saúde, Maputo, Mozambique
| | - Pascal M W Drake
- Institute for Infection and Immunity, St. George's University, London, United Kingdom
| | - Juraj Ivanyi
- Departamento de Plataformas Tecnológicas em Saúde, Instituto Nacional de Saúde, Maputo, Mozambique.,Guy's Campus of King's College London, London, United Kingdom
| | - Galit Alter
- Ragon Institute, Harvard, Cambridge, MA, United States
| | - Rajko Reljic
- Institute for Infection and Immunity, St. George's University, London, United Kingdom
| |
Collapse
|
52
|
Arora SK, Naqvi N, Alam A, Ahmad J, Alsati BS, Sheikh JA, Kumar P, Mitra DK, Rahman SA, Hasnain SE, Ehtesham NZ. Mycobacterium smegmatis Bacteria Expressing Mycobacterium tuberculosis-Specific Rv1954A Induce Macrophage Activation and Modulate the Immune Response. Front Cell Infect Microbiol 2020; 10:564565. [PMID: 33163415 PMCID: PMC7583720 DOI: 10.3389/fcimb.2020.564565] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/28/2020] [Indexed: 12/14/2022] Open
Abstract
Mycobacterium tuberculosis (M. tb), the intracellular pathogen causing tuberculosis, has developed mechanisms that endow infectivity and allow it to modulate host immune response for its survival. Genomic and proteomic analyses of non-pathogenic and pathogenic mycobacteria showed presence of genes and proteins that are specific to M. tb. In silico studies predicted that M.tb Rv1954A is a hypothetical secretory protein that exhibits intrinsically disordered regions and possess B cell/T cell epitopes. Treatment of macrophages with Rv1954A led to TLR4-mediated activation with concomitant increase in secretion of pro-inflammatory cytokines, IL-12 and TNF-α. In vitro studies showed that rRv1954A protein or Rv1954A knock-in M. smegmatis (Ms_Rv1954A) activates macrophages by enhancing the expression of CD80 and CD86. An upregulation in the expression of CD40 and MHC I/II was noted in the presence of Rv1954A, pointing to its role in enhancing the association of APCs with T cells and in the modulation of antigen presentation, respectively. Ms_Rv1954A showed increased infectivity, induction of ROS and RNS, and apoptosis in RAW264.7 macrophage cells. Rv1954A imparted protection against oxidative and nitrosative stress, thereby enhancing the survival of Ms_Rv1954A inside macrophages. Mice immunized with Ms_Rv1954A showed that splenomegaly and primed splenocytes restimulated with Rv1954A elicited a Th1 response. Infection of Ms_Rv1954A in mice through intratracheal instillation leads to enhanced infiltration of lymphocytes in the lungs without formation of granuloma. While Rv1954A is immunogenic, it did not cause adverse pathology. Purified Rv1954A or Rv1954A knock-in M. smegmatis (Ms_Rv1954A) elicited a nearly two-fold higher titer of IgG response in mice, and PTB patients possess a higher IgG titer against Rv1954A, also pointing to its utility as a diagnostic marker for TB. The observed modulation of innate and adaptive immunity renders Rv1954A a vital protein in the pathophysiology of this pathogen.
Collapse
Affiliation(s)
- Simran Kaur Arora
- Indian Council of Medical Research (ICMR)-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India.,Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India
| | - Nilofer Naqvi
- Indian Council of Medical Research (ICMR)-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| | - Anwar Alam
- Indian Council of Medical Research (ICMR)-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| | - Javeed Ahmad
- Indian Council of Medical Research (ICMR)-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| | - Basma Saud Alsati
- Indian Council of Medical Research (ICMR)-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| | | | - Prabin Kumar
- Department of Transplant Immunology and Immunogenetics, All India Institute of Medical Sciences, New Delhi, India
| | - Dipendra Kumar Mitra
- Department of Transplant Immunology and Immunogenetics, All India Institute of Medical Sciences, New Delhi, India
| | | | - Seyed Ehtesham Hasnain
- Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India.,Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Hyderabad, India
| | - Nasreen Zafar Ehtesham
- Indian Council of Medical Research (ICMR)-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| |
Collapse
|
53
|
Ong CWM, Migliori GB, Raviglione M, MacGregor-Skinner G, Sotgiu G, Alffenaar JW, Tiberi S, Adlhoch C, Alonzi T, Archuleta S, Brusin S, Cambau E, Capobianchi MR, Castilletti C, Centis R, Cirillo DM, D'Ambrosio L, Delogu G, Esposito SMR, Figueroa J, Friedland JS, Ho BCH, Ippolito G, Jankovic M, Kim HY, Rosales Klintz S, Ködmön C, Lalle E, Leo YS, Leung CC, Märtson AG, Melazzini MG, Najafi Fard S, Penttinen P, Petrone L, Petruccioli E, Pontali E, Saderi L, Santin M, Spanevello A, van Crevel R, van der Werf MJ, Visca D, Viveiros M, Zellweger JP, Zumla A, Goletti D. Epidemic and pandemic viral infections: impact on tuberculosis and the lung: A consensus by the World Association for Infectious Diseases and Immunological Disorders (WAidid), Global Tuberculosis Network (GTN), and members of the European Society of Clinical Microbiology and Infectious Diseases Study Group for Mycobacterial Infections (ESGMYC). Eur Respir J 2020; 56:2001727. [PMID: 32586885 PMCID: PMC7527651 DOI: 10.1183/13993003.01727-2020] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 06/12/2020] [Indexed: 01/08/2023]
Abstract
Major epidemics, including some that qualify as pandemics, such as severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), HIV, influenza A (H1N1)pdm/09 and most recently COVID-19, affect the lung. Tuberculosis (TB) remains the top infectious disease killer, but apart from syndemic TB/HIV little is known regarding the interaction of viral epidemics and pandemics with TB. The aim of this consensus-based document is to describe the effects of viral infections resulting in epidemics and pandemics that affect the lung (MERS, SARS, HIV, influenza A (H1N1)pdm/09 and COVID-19) and their interactions with TB. A search of the scientific literature was performed. A writing committee of international experts including the European Centre for Disease Prevention and Control Public Health Emergency (ECDC PHE) team, the World Association for Infectious Diseases and Immunological Disorders (WAidid), the Global Tuberculosis Network (GTN), and members of the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Mycobacterial Infections (ESGMYC) was established. Consensus was achieved after multiple rounds of revisions between the writing committee and a larger expert group. A Delphi process involving the core group of authors (excluding the ECDC PHE team) identified the areas requiring review/consensus, followed by a second round to refine the definitive consensus elements. The epidemiology and immunology of these viral infections and their interactions with TB are discussed with implications for diagnosis, treatment and prevention of airborne infections (infection control, viral containment and workplace safety). This consensus document represents a rapid and comprehensive summary on what is known on the topic.
Collapse
Affiliation(s)
- Catherine Wei Min Ong
- Dept of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore
- These authors contributed equally
- Members of ESGMYC
| | - Giovanni Battista Migliori
- Servizio di Epidemiologia Clinica delle Malattie Respiratorie, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
- These authors contributed equally
| | - Mario Raviglione
- Centre for Multidisciplinary Research in Health Science, University of Milan, Milan, Italy
- Global Studies Institute, University of Geneva, Geneva, Switzerland
| | | | - Giovanni Sotgiu
- Clinical Epidemiology and Medical Statistics Unit, Dept of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Jan-Willem Alffenaar
- Sydney Pharmacy School, University of Sydney, Sydney, Australia
- Westmead Hospital, Sydney, Australia
- Marie Bashir Institute of Infectious Diseases and Biosecurity, University of Sydney, Sydney, Australia
- Members of ESGMYC
| | - Simon Tiberi
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Division of Infection, Royal London Hospital, Barts Health NHS Trust, London, UK
- Members of ESGMYC
| | - Cornelia Adlhoch
- Public Health Emergency Team, European Centre for Disease Prevention and Control, Stockholm, Sweden
- European Centre for Disease Prevention and Control Public Health Emergency team co-authors
| | - Tonino Alonzi
- Translational Research Unit, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Sophia Archuleta
- Dept of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sergio Brusin
- Public Health Emergency Team, European Centre for Disease Prevention and Control, Stockholm, Sweden
- European Centre for Disease Prevention and Control Public Health Emergency team co-authors
| | - Emmanuelle Cambau
- AP-HP-Lariboisiere, Bacteriologie, Laboratory Associated to the National Reference Centre for Mycobacteria, IAME UMR1137, INSERM, University of Paris, Paris, France
- Members of ESGMYC
| | - Maria Rosaria Capobianchi
- Laboratory of Virology, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Concetta Castilletti
- Laboratory of Virology, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Rosella Centis
- Servizio di Epidemiologia Clinica delle Malattie Respiratorie, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
| | - Daniela M Cirillo
- Emerging Bacterial Pathogens Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Members of ESGMYC
| | | | - Giovanni Delogu
- Università Cattolica Sacro Cuore, Roma, Italy
- Mater Olbia Hospital, Olbia, Italy
- Members of ESGMYC
| | - Susanna M R Esposito
- Pediatric Clinic, Pietro Barilla Children's Hospital, University of Parma, Parma, Italy
| | | | - Jon S Friedland
- St George's, University of London, London, UK
- Members of ESGMYC
| | - Benjamin Choon Heng Ho
- Tuberculosis Control Unit, Dept of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore
| | - Giuseppe Ippolito
- Scientific Direction, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Mateja Jankovic
- School of Medicine, University of Zagreb and Clinic for Respiratory Diseases, University Hospital Center Zagreb, Zagreb, Croatia
- Members of ESGMYC
| | - Hannah Yejin Kim
- Sydney Pharmacy School, University of Sydney, Sydney, Australia
- Westmead Hospital, Sydney, Australia
- Marie Bashir Institute of Infectious Diseases and Biosecurity, University of Sydney, Sydney, Australia
| | - Senia Rosales Klintz
- Public Health Emergency Team, European Centre for Disease Prevention and Control, Stockholm, Sweden
- European Centre for Disease Prevention and Control Public Health Emergency team co-authors
| | - Csaba Ködmön
- Public Health Emergency Team, European Centre for Disease Prevention and Control, Stockholm, Sweden
- European Centre for Disease Prevention and Control Public Health Emergency team co-authors
| | - Eleonora Lalle
- Laboratory of Virology, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Yee Sin Leo
- National Centre for Infectious Diseases, Singapore
| | - Chi-Chiu Leung
- Hong Kong Tuberculosis, Chest and Heart Diseases Association, Wanchai, Hong Kong, China
| | - Anne-Grete Märtson
- Dept of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Saeid Najafi Fard
- Translational Research Unit, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Pasi Penttinen
- Public Health Emergency Team, European Centre for Disease Prevention and Control, Stockholm, Sweden
- European Centre for Disease Prevention and Control Public Health Emergency team co-authors
| | - Linda Petrone
- Translational Research Unit, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Elisa Petruccioli
- Translational Research Unit, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | | | - Laura Saderi
- Clinical Epidemiology and Medical Statistics Unit, Dept of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Miguel Santin
- Dept of Infectious Diseases, Bellvitge University Hospital-Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Dept of Clinical Science, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Members of ESGMYC
| | - Antonio Spanevello
- Division of Pulmonary Rehabilitation, Istituti Clinici Scientifici Maugeri, IRCCS, Tradate, Italy
- Dept of Medicine and Surgery, Respiratory Diseases, University of Insubria, Varese-Como, Italy
| | - Reinout van Crevel
- Radboudumc Center for Infectious Diseases, Radboud Institute for Health Sciences, Radboudumc, Nijmegen, The Netherlands
- Centre for Tropical Medicine and Global Health, Nuffield Dept of Medicine, University of Oxford, Oxford, UK
- Members of ESGMYC
| | - Marieke J van der Werf
- Public Health Emergency Team, European Centre for Disease Prevention and Control, Stockholm, Sweden
- European Centre for Disease Prevention and Control Public Health Emergency team co-authors
| | - Dina Visca
- Division of Pulmonary Rehabilitation, Istituti Clinici Scientifici Maugeri, IRCCS, Tradate, Italy
- Dept of Medicine and Surgery, Respiratory Diseases, University of Insubria, Varese-Como, Italy
| | - Miguel Viveiros
- Global Health and Tropical Medicine, Institute of Hygiene and Tropical Medicine, NOVA University of Lisbon, Lisbon, Portugal
- Members of ESGMYC
| | | | - Alimuddin Zumla
- Dept of Infection, Division of Infection and Immunity, University College London and NIHR Biomedical Research Centre, UCL Hospitals NHS Foundation Trust, London, UK
| | - Delia Goletti
- Translational Research Unit, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
- Saint Camillus International University of Health and Medical Sciences, Rome, Italy
- Members of ESGMYC
| |
Collapse
|
54
|
Wang H, Yan S, Liu H, Li L, Song J, Wang G, Wang H, Wu Y, Shao Z, Fu R. Infection risk in autoimmune hematological disorders with low-dose rituximab treatment. J Clin Lab Anal 2020; 34:e23455. [PMID: 32794271 PMCID: PMC7595891 DOI: 10.1002/jcla.23455] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/07/2020] [Accepted: 06/12/2020] [Indexed: 12/12/2022] Open
Abstract
Background Rituximab has been widely used in many autoimmune diseases. Aim To evaluate the infection risk of rituximab in autoimmune hematological disorders. Methods Retrospectively studied and compared the clinical data of 89 patients in our hospital who used low‐dose rituximab (group R) or pulse cyclophosphamide (group C) for their refractory/relapsed autoimmune hematological diseases from January 2011 to January 2017. The kinds of their diseases included autoimmune hemolytic disease (AIHA), Evans syndrome, and idiopathic thrombocytopenic purpura (ITP). All patients chose either rituximab treatment or cyclophosphamide treatment on their own considerations. Findings The median follow‐up time was six months in group R and four months in group C. After treatments, the patients in group R showed higher white blood cell (WBC) count and neutrophil count than group C (P = .020, P = .037). CD20‐positive B cells in group R remained at a very low level after rituximab treatment and need about 15 months to return to normal level, which was longer than group C (six months). The incidence of infection in these two groups has no significant difference, which was 34.7% (17/30) in group R and 32.5% (13/28) in group C (P = .976). Tuberculosis infections after rituximab treatment were found in three patients for the first time. Conclusion The G‐CSF, nadir WBC count, and IgA level were protective factors of infection during rituximab treatment. Low‐dose rituximab therapy in autoimmune hematological diseases does not increase infection risk compared with cyclophosphamide.
Collapse
Affiliation(s)
- Honglei Wang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Siyang Yan
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Hui Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Lijuan Li
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jia Song
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Guojin Wang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Huaquan Wang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuhong Wu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zonghong Shao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
55
|
Chen C, Xu H, Peng Y, Luo H, Huang GX, Wu XJ, Dai YC, Luo HL, Zhang JA, Zheng BY, Zhang XN, Chen ZW, Xu JF. Elevation in the counts of IL-35-producing B cells infiltrating into lung tissue in mycobacterial infection is associated with the downregulation of Th1/Th17 and upregulation of Foxp3 +Treg. Sci Rep 2020; 10:13212. [PMID: 32764544 PMCID: PMC7411070 DOI: 10.1038/s41598-020-69984-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 06/01/2020] [Indexed: 12/19/2022] Open
Abstract
IL-35 is an anti-inflammatory cytokine and is thought to be produced by regulatory T (Treg) cells. A previous study found that IL-35 was upregulated in the serum of patients with active tuberculosis (ATB), and IL-35-producing B cells infiltrated to tuberculous granuloma of patients with ATB. Purified B cells from such patients generated more IL-35 after stimulation by antigens of Mycobacterium tuberculosis and secreted more IL-10. However, the function and the underlying mechanisms of IL-35-producing B cells in TB progression have not been investigated. The present study found that the expression of mRNA of IL-35 subsets Ebi3 and p35 was elevated in mononuclear cells from peripheral blood, spleen, bone marrow, and lung tissue in a mouse model infected with Mycobacterium bovis BCG, as tested by real-time polymerase chain reaction. Accordingly, the flow cytometry analysis showed that the counts of a subset of IL-35+ B cells were elevated in the circulating blood and in the spleen, bone marrow, and lung tissue in BCG-infected mice, whereas anti-TB therapy reduced IL-35-producing B cells. Interestingly, BCG infection could drive the infiltration of IL-35-producing B cells into the lung tissue, and the elevated counts of IL-35-producing B cells positively correlated with the bacterial load in the lungs. Importantly, the injection of exogenous IL-35 stimulated the elevation in the counts of IL-35-producing B cells and was associated with the downregulation of Th1/Th17 and upregulation of Foxp3+Treg.The study showed that a subset of IL-35-producing B cells might take part in the downregulation of immune response in mycobacterial infection.
Collapse
Affiliation(s)
- Chen Chen
- Department of Clinical Immunology, Institute of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, China
- Molecular Diagnostic Center, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Huan Xu
- Department of Clinical Immunology, Institute of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, China
| | - Ying Peng
- Department of Clinical Immunology, Institute of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, China
| | - Hong Luo
- Department of Clinical Immunology, Institute of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, China
| | - Gui-Xian Huang
- Department of Clinical Immunology, Institute of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, China
| | - Xian-Jin Wu
- Department of Clinical Laboratory, Huizhou Municipal Central Hospital, No. 41 North Eling Road, Huizhou, 516001, China
| | - You-Chao Dai
- Department of Clinical Immunology, Institute of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, China
| | - Hou-Long Luo
- Department of Clinical Immunology, Institute of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, China
| | - Jun-Ai Zhang
- Department of Clinical Immunology, Institute of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, China
| | - Bi-Ying Zheng
- Department of Clinical Immunology, Institute of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, China
| | - Xiang-Ning Zhang
- Department of Pathophysiology, Basic Medical School, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, China
| | - Zheng W Chen
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL, USA
| | - Jun-Fa Xu
- Department of Clinical Immunology, Institute of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, No. 1 Xincheng Road, Dongguan, 523808, China.
| |
Collapse
|
56
|
Arora SK, Alam A, Naqvi N, Ahmad J, Sheikh JA, Rahman SA, Hasnain SE, Ehtesham NZ. Immunodominant Mycobacterium tuberculosis Protein Rv1507A Elicits Th1 Response and Modulates Host Macrophage Effector Functions. Front Immunol 2020; 11:1199. [PMID: 32793184 PMCID: PMC7385400 DOI: 10.3389/fimmu.2020.01199] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 05/14/2020] [Indexed: 12/12/2022] Open
Abstract
Mycobacterium tuberculosis (M. tb) persists as latent infection in nearly a quarter of the global population and remains the leading cause of death among infectious diseases. While BCG is the only vaccine for TB, its inability to provide complete protection makes it imperative to engineer BCG such that it expresses immunodominant antigens that can enhance its protective potential. In-silico comparative genomic analysis of Mycobacterium species identified M. tb Rv1507A as a “signature protein” found exclusively in M. tb. In-vitro (cell lines) and in-vivo experiments carried out in mice, using purified recombinant Rv1507A revealed it to be a pro-inflammatory molecule, eliciting significantly high levels of IL-6, TNF-α, and IL-12. There was increased expression of activation markers CD69, CD80, CD86, antigen presentation molecules (MHC I/MHCII), and associated Th1 type of immune response. Rv1507A knocked-in M. smegmatis also induced significantly higher pro-inflammatory Th1 response and higher survivability under stress conditions, both in-vitro (macrophage RAW264.7 cells) and in-vivo (mice). Sera derived from human TB patients showed significantly enhanced B-cell response against M. tb Rv1507A. The ability of M. tb Rv1507A to induce immuno-modulatory effect, B cell response, and significant memory response, renders it a putative vaccine candidate that demands further exploration.
Collapse
Affiliation(s)
- Simran Kaur Arora
- Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India.,ICMR-National Institute of Pathology, New Delhi, India
| | - Anwar Alam
- ICMR-National Institute of Pathology, New Delhi, India
| | - Nilofer Naqvi
- ICMR-National Institute of Pathology, New Delhi, India
| | - Javeed Ahmad
- ICMR-National Institute of Pathology, New Delhi, India
| | | | | | - Seyed Ehtesham Hasnain
- Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India.,Dr. Reddy's Institute of Life Sciences, Hyderabad, India
| | | |
Collapse
|
57
|
Moore DK, Leisching GR, Snyders CI, Gutschmidt A, van Rensburg IC, Loxton AG. Immunoglobulin profile and B-cell frequencies are altered with changes in the cellular microenvironment independent of the stimulation conditions. IMMUNITY INFLAMMATION AND DISEASE 2020; 8:458-467. [PMID: 32639690 PMCID: PMC7416019 DOI: 10.1002/iid3.328] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/13/2020] [Accepted: 06/21/2020] [Indexed: 12/19/2022]
Abstract
Introduction B‐cells are essential in the defense against Mycobacterium tuberculosis. Studies on isolated cells may not accurately reflect the responses that occur in vivo due to the presence of other cells. This study elucidated the influence of microenvironment complexity on B‐cell polarization and function in the context of tuberculosis disease. Methods B‐cell function was tested in whole blood, peripheral blood mononuclear cells (PBMCs), and as isolated cells. The different fractions were stimulated and the B‐cell phenotype and immunoglobulin profiles analyzed. Results The immunoglobulin profile and developmental B‐cell frequencies varied for each of the investigated sample types, while in an isolated cellular environment, secretion of immunoglobulin isotypes immunoglobulin A (IgA), IgG2, and IgG3 was hampered. The differences in the immunoglobulin profile highlight the importance of cell‐cell communication for B‐cell activation. Furthermore, a decrease in marginal zone B‐cell frequencies and an increase in T1 B‐cells was observed following cell isolation, indicating impaired B‐cell development in response to in vitro antigenic stimulation in isolation. Conclusion Our results suggest that humoral B‐cell function and development was impaired likely due to a lack of costimulatory signals from other cell types. Thus, B‐cell function should ideally be studied in a PBMC or whole blood fraction.
Collapse
Affiliation(s)
- Dannielle K Moore
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Gina R Leisching
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Candice I Snyders
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Andrea Gutschmidt
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Ilana C van Rensburg
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Andre G Loxton
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
58
|
Dos Santos DCM, Lovero KL, Schmidt CM, Barros ACMW, Quintanilha AP, Barbosa AP, Pone MVS, Pone SM, Araujo JM, de Paula Martins C, Cosme EM, Dourado de Oliveira TR, Miceli AL, Vieira ML, Queiroz A, Coca Velarde LG, Kritski A, de Fátima Pombo March M, Maria de Azevedo Sias S, SantÀAnna CC, Riley LW, Araújo Cardoso CA. Serological biomarkers for monitoring response to treatment of pulmonary and extrapulmonary tuberculosis in children and adolescents. Tuberculosis (Edinb) 2020; 123:101960. [PMID: 32741536 PMCID: PMC7436889 DOI: 10.1016/j.tube.2020.101960] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 02/17/2020] [Accepted: 05/26/2020] [Indexed: 01/19/2023]
Abstract
Key measures to halt the spread of tuberculosis (TB) include early diagnosis, effective treatment, and monitoring disease management. We sought to evaluate the use of serum immunoglobulin levels against antigens present in cell envelope of Mycobacterium tuberculosis to monitor TB treatment response in children and adolescents with pulmonary (PTB) or extrapulmonary TB (EPTB). Blood samples were collected prior to and one, two, and six months following treatment initiation. Serum immunoglobulin levels against cardiolipin, sulfatide, mycolic acid and Mce1A protein were measured by ELISA. Serum from 53 TB patients and 12 healthy participants were analyzed. After six months of successful treatment, there was a significant decrease (p < 0.0001) in IgM levels against cardiolipin, sulfatide, mycolic acid and Mce1A protein and IgG levels against Mce1A protein when compared to baseline immunoglobulin levels. There was no significant variation in antibody levels during follow-up between participants with PTB and EPTB, confirmed and unconfirmed TB diagnosis, and HIV infection status. Antibody levels in control participants without TB did not decrease during follow-up. These results suggest that immunoglobulin responses to mycobacterial cell wall products may be a useful tool to monitor treatment response in children and adolescents with PTB or EPTB.
Collapse
Affiliation(s)
- Danielle C M Dos Santos
- Departamento de Patologia, Faculdade de Medicina, Universidade Federal Fluminense, Niterói, Brazil; Laboratório Multiusuário de Apoio à Pesquisa Em Nefrologia e Ciências Médicas, Faculdade de Medicina, Universidade Federal Fluminense, Niterói, Brazil
| | - Kathryn L Lovero
- New York State Psychiatric Institute, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, 1051 Riverside Drive #24, New York, NY, 10032, USA
| | - Christiane M Schmidt
- Laboratório Multiusuário de Apoio à Pesquisa Em Nefrologia e Ciências Médicas, Faculdade de Medicina, Universidade Federal Fluminense, Niterói, Brazil; Departamento Materno-Infantil, Faculdade de Medicina, Universidade Federal Fluminense, Niterói, Brazil
| | - Ana Cláudia M W Barros
- Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Fiocruz, Rio de Janeiro, Brazil
| | | | - Ana Paula Barbosa
- Programa de Controle de Tuberculose da Secretária de Saúde de São Gonçalo, RJ, Brazil
| | - Marcos V S Pone
- Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Fiocruz, Rio de Janeiro, Brazil
| | - Sheila M Pone
- Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Fiocruz, Rio de Janeiro, Brazil
| | | | - Camila de Paula Martins
- Laboratório Multiusuário de Apoio à Pesquisa Em Nefrologia e Ciências Médicas, Faculdade de Medicina, Universidade Federal Fluminense, Niterói, Brazil
| | - Estela Magalhães Cosme
- Departamento Materno-Infantil, Faculdade de Medicina, Universidade Federal Fluminense, Niterói, Brazil
| | | | - Ana Lúcia Miceli
- Programa de Controle de Tuberculose da Secretária de Saúde de Duque de Caxias, RJ, Brazil
| | - Maria Luíza Vieira
- Programa de Controle de Tuberculose da Secretária de Saúde de Duque de Caxias, RJ, Brazil
| | | | | | - Afranio Kritski
- Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maria de Fátima Pombo March
- Instituto de Puericultura e Pediatria Martagão Gesteira, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Clemax C SantÀAnna
- Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Instituto de Puericultura e Pediatria Martagão Gesteira, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lee W Riley
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, United States
| | - Claudete A Araújo Cardoso
- Laboratório Multiusuário de Apoio à Pesquisa Em Nefrologia e Ciências Médicas, Faculdade de Medicina, Universidade Federal Fluminense, Niterói, Brazil; Departamento Materno-Infantil, Faculdade de Medicina, Universidade Federal Fluminense, Niterói, Brazil.
| |
Collapse
|
59
|
Davis JS, Ferreira D, Paige E, Gedye C, Boyle M. Infectious Complications of Biological and Small Molecule Targeted Immunomodulatory Therapies. Clin Microbiol Rev 2020; 33:e00035-19. [PMID: 32522746 PMCID: PMC7289788 DOI: 10.1128/cmr.00035-19] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The past 2 decades have seen a revolution in our approach to therapeutic immunosuppression. We have moved from relying on broadly active traditional medications, such as prednisolone or methotrexate, toward more specific agents that often target a single receptor, cytokine, or cell type, using monoclonal antibodies, fusion proteins, or targeted small molecules. This change has transformed the treatment of many conditions, including rheumatoid arthritis, cancers, asthma, and inflammatory bowel disease, but along with the benefits have come risks. Contrary to the hope that these more specific agents would have minimal and predictable infectious sequelae, infectious complications have emerged as a major stumbling block for many of these agents. Furthermore, the growing number and complexity of available biologic agents makes it difficult for clinicians to maintain current knowledge, and most review articles focus on a particular target disease or class of agent. In this article, we review the current state of knowledge about infectious complications of biologic and small molecule immunomodulatory agents, aiming to create a single resource relevant to a broad range of clinicians and researchers. For each of 19 classes of agent, we discuss the mechanism of action, the risk and types of infectious complications, and recommendations for prevention of infection.
Collapse
Affiliation(s)
- Joshua S Davis
- Department of Infectious Diseases and Immunology, John Hunter Hospital, Newcastle, NSW, Australia
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia
| | - David Ferreira
- School of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Emma Paige
- Department of Infectious Diseases, Alfred Hospital, Melbourne, VIC, Australia
| | - Craig Gedye
- School of Medicine, University of New South Wales, Sydney, NSW, Australia
- Department of Oncology, Calvary Mater Hospital, Newcastle, NSW, Australia
| | - Michael Boyle
- Department of Infectious Diseases and Immunology, John Hunter Hospital, Newcastle, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
60
|
Bright MR, Curtis N, Messina NL. The role of antibodies in Bacille Calmette Guérin-mediated immune responses and protection against tuberculosis in humans: A systematic review. Tuberculosis (Edinb) 2020; 131:101947. [PMID: 33691988 DOI: 10.1016/j.tube.2020.101947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 05/02/2020] [Accepted: 05/11/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND The mechanisms underlying Bacille Calmette-Guérin (BCG) vaccine's protective effects against tuberculosis (TB) are incompletely understood but are proposed to involve a predominantly cell-mediated process. However, there is increasing evidence for the involvement of antibodies in the control of Mycobacteria tuberculosis and in the immune response to BCG. METHODS We did a systematic review of studies investigating anti-BCG antibodies in individuals with active or latent TB, and in the response to BCG vaccination. RESULTS Of 1417 articles screened, 70 were relevant, comprising 52 investigating anti-BCG antibodies in TB and 18 investigating the anti-BCG antibody response to BCG-vaccination. Individuals with active TB have higher levels of anti-BCG antibodies compared with individuals with latent TB or healthy individuals. Antibodies to BCG are present after BCG vaccination. There is some evidence for the in utero transfer of maternal anti-BCG antibodies to infants. CONCLUSIONS BCG vaccination induces a humoral response. Antibodies targeted against BCG and its antigens may play a role in protection against active TB.
Collapse
Affiliation(s)
- Matthew R Bright
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia; Murdoch Children's Research Institute, Parkville, Victoria, Australia; Infectious Diseases Unit, Royal Children's Hospital Melbourne, Parkville, Victoria, Australia.
| | - Nicole L Messina
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia; Murdoch Children's Research Institute, Parkville, Victoria, Australia.
| |
Collapse
|
61
|
Bucsan AN, Mehra S, Khader SA, Kaushal D. The current state of animal models and genomic approaches towards identifying and validating molecular determinants of Mycobacterium tuberculosis infection and tuberculosis disease. Pathog Dis 2020; 77:5543892. [PMID: 31381766 PMCID: PMC6687098 DOI: 10.1093/femspd/ftz037] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 07/25/2019] [Indexed: 12/31/2022] Open
Abstract
Animal models are important in understanding both the pathogenesis of and immunity to tuberculosis (TB). Unfortunately, we are beginning to understand that no animal model perfectly recapitulates the human TB syndrome, which encompasses numerous different stages. Furthermore, Mycobacterium tuberculosis infection is a very heterogeneous event at both the levels of pathogenesis and immunity. This review seeks to establish the current understanding of TB pathogenesis and immunity, as validated in the animal models of TB in active use today. We especially focus on the use of modern genomic approaches in these models to determine the mechanism and the role of specific molecular pathways. Animal models have significantly enhanced our understanding of TB. Incorporation of contemporary technologies such as single cell transcriptomics, high-parameter flow cytometric immune profiling, proteomics, proteomic flow cytometry and immunocytometry into the animal models in use will further enhance our understanding of TB and facilitate the development of treatment and vaccination strategies.
Collapse
Affiliation(s)
- Allison N Bucsan
- Tulane Center for Tuberculosis Research, Covington, LA, USA.,Tulane National Primate Research Center, Covington, LA, USA
| | - Smriti Mehra
- Tulane National Primate Research Center, Covington, LA, USA
| | | | - Deepak Kaushal
- Tulane Center for Tuberculosis Research, Covington, LA, USA.,Tulane National Primate Research Center, Covington, LA, USA.,Southwest National Primate Research Center, San Antonio, TX, USA.,Texas Biomedical Research Institute, San Antonio, TX, USA
| |
Collapse
|
62
|
Choreño-Parra JA, Weinstein LI, Yunis EJ, Zúñiga J, Hernández-Pando R. Thinking Outside the Box: Innate- and B Cell-Memory Responses as Novel Protective Mechanisms Against Tuberculosis. Front Immunol 2020; 11:226. [PMID: 32117325 PMCID: PMC7034257 DOI: 10.3389/fimmu.2020.00226] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 01/28/2020] [Indexed: 12/31/2022] Open
Abstract
Tuberculosis (TB) is currently the deadliest infectious disease worldwide. Failure to create a highly effective vaccine has limited the control of the TB epidemic. Historically, the vaccine field has relied on the paradigm that IFN-γ-mediated CD4+ T cell memory responses are the principal correlate of protection in TB. Nonetheless, the demonstration that other cellular subsets offer protective memory responses against Mycobacterium tuberculosis (Mtb) is emerging. Among these are memory-like features of macrophages, myeloid cell precursors, natural killer (NK) cells, and innate lymphoid cells (ILCs). Additionally, the dynamics of B cell memory responses have been recently characterized at different stages of the clinical spectrum of Mtb infection, suggesting a role for B cells in human TB. A better understanding of the immune mechanisms underlying such responses is crucial to better comprehend protective immunity in TB. Furthermore, targeting immune compartments other than CD4+ T cells in TB vaccine strategies may benefit a significant proportion of patients co-infected with Mtb and the human immunodeficiency virus (HIV). Here, we summarize the memory responses of innate immune cells and B cells against Mtb and propose them as novel correlates of protection that could be harnessed in future vaccine development programs.
Collapse
Affiliation(s)
- José Alberto Choreño-Parra
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico.,Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - León Islas Weinstein
- Section of Experimental Pathology, Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Edmond J Yunis
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA, United States.,Department of Pathology, Harvard Medical School, Boston, MA, United States
| | - Joaquín Zúñiga
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico.,Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Rogelio Hernández-Pando
- Section of Experimental Pathology, Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
63
|
Lyashchenko KP, Vordermeier HM, Waters WR. Memory B cells and tuberculosis. Vet Immunol Immunopathol 2020; 221:110016. [PMID: 32050091 DOI: 10.1016/j.vetimm.2020.110016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/30/2019] [Accepted: 01/29/2020] [Indexed: 02/09/2023]
Abstract
Immunological memory is a central feature of adaptive immunity. Memory B cells are generated upon stimulation with antigen presented by follicular dendritic cells in the peripheral lymphoid tissues. This process typically involves class-switch recombination and somatic hypermutation and it can be dependent or independent on germinal centers or T cell help. The mature B cell memory pool is generally characterized by remarkable heterogeneity of functionally and phenotypically distinct sub-populations supporting multi-layer immune plasticity. Memory B cells found in human patients infected with Mycobacterium tuberculosis include IgD+ CD27+ and IgM+ CD27+ subsets. In addition, expansion of atypical memory B cells characterized by the lack of CD27 expression and by inability to respond to antigen-induced re-activation is documented in human tuberculosis. These functionally impaired memory B cells are believed to have adverse effects on host immunity. Human and animal studies demonstrate recruitment of antigen-activated B cells to the infection sites and their presence in lung granulomas where proliferating B cells are organized into discrete clusters resembling germinal centers of secondary lymphoid organs. Cattle studies show development of IgM+, IgG+, and IgA+ memory B cells in M. bovis infection with the ability to rapidly differentiate into antibody-producing plasma cells upon antigen re-exposure. This review discusses recent advances in research on generation, re-activation, heterogeneity, and immunobiological functions of memory B cells in tuberculosis. The role of memory B cells in post-skin test recall antibody responses in bovine tuberculosis and implications for development of improved immunodiagnostics are also reviewed.
Collapse
Affiliation(s)
| | - H Martin Vordermeier
- Tuberculosis Research Group, Animal and Plant Health Agency, Addlestone, United Kingdom; Institute for Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, United Kingdom
| | - W Ray Waters
- National Animal Disease Center, Agricultural Research Service, US Department of Agriculture, Ames, IA, USA
| |
Collapse
|
64
|
Screening for pulmonary tuberculosis in high-risk groups of diabetic patients. Int J Infect Dis 2020; 93:84-89. [PMID: 31978585 DOI: 10.1016/j.ijid.2020.01.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/09/2019] [Accepted: 01/14/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The double burden of diabetes mellitus (DM) and tuberculosis (TB) has attracted increasing attention, because DM not only increases the risk of active TB but also affects treatment outcomes. Screening for TB among diabetic patients has been recommended, but requires real-world evidence by considering its cost-effectiveness, cost-utility ratio, and cost-benefit ratio. METHODS A screening program was conducted in Jiangyin City of Jiangsu Province, China. A total of 14 869 diabetic patients received regular physical examinations for three consecutive years and were followed for the diagnosis of TB. The cost of screening and the effectiveness, utility, and social benefits attributed to the program were evaluated. In addition, a matched case-control study was conducted and the nomogram was used to identify high-risk groups that could be the target population for screening. RESULTS Among the 14 869 diabetic patients who participated in this screening program, 22 were diagnosed with TB, resulting in an incremental cost-effectiveness ratio (ICER) of 83 910 CNY per disability-adjusted life-year (DALY) gained and a cost-benefit ratio of 0.50. If the screening program was limited to high-risk diabetic patients by considering body mass index (BMI), fasting blood glucose (FBG), and triglycerides, the ICER decreased to 34 303 CNY per DALY gained and the cost-benefit ratio increased to 1.22. CONCLUSIONS Screening for TB using regular chest X-ray examinations is feasible but not economical in areas with a low incidence of TB. It is recommended that diabetic patients with a low BMI, high FBG, and low triglycerides are selected as subjects for TB screening.
Collapse
|
65
|
Tavassol ZH, Aziziraftar SK, Behrouzi A, Ghazanfari M, Masoumi M, Fateh A, Vaziri F, Siadat SD. Evaluation of Mycobacterium kansasii extracellular vesicles role in BALB/c mice immune modulatory. Int J Mycobacteriol 2020; 9:58-61. [PMID: 32474490 DOI: 10.4103/ijmy.ijmy_212_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Mycobacterium kansasii as a nontuberculosis mycobacteria, naturally release extracellular vesicles (EVs) with widespread utilities. The aim of the present study was the extraction and biological evaluation of M. kansasii EV and its role in BALB/c mice immune modulatory by considering EVs medical usage specificities. METHOD Density gradient ultracentrifugation method was used to EVs extraction from standard species of M. kansasii. Biologic validation of EVs has been performed by physicochemical experiments. Immunization has been done by subcutaneous injection to BALB/c mice, then spleen cell isolation and lymphocyte transformation test and eventually ELISA cytokine assays were made for interleukin-10 (IL-10) and interferon-gamma (IFN-γ). UNLABELLED IBM SPSS version 22 software (SPSS. Inc., Chicago, IL, USA) was used for the data calculation. The evaluation of variables was conducted using one sample t-test. RESULTS Physicochemical experiment results contribute that extracted EVs have intransitive capability to use in immunization schedule. Finally, ELISA test results showed that EVs induced IL-10 production, but have no effect on IFN-γ. CONCLUSIONS In this current study, EVs were prepared in high-quality composition. The results of cytokine assay revealed that the extracted EVs have anti-inflammatory property. Accordingly, this macromolecule can be used as immune modulatory agents to prevent severe immune reactions, especially in lungs disorders.
Collapse
Affiliation(s)
- Zahra Hoseini Tavassol
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | | | - Ava Behrouzi
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Morteza Ghazanfari
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Morteza Masoumi
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Aboulfazl Fateh
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Farzam Vaziri
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
66
|
Rajamanickam A, Munisankar S, Dolla CK, Babu S. Undernutrition is associated with perturbations in T cell-, B cell-, monocyte- and dendritic cell- subsets in latent Mycobacterium tuberculosis infection. PLoS One 2019; 14:e0225611. [PMID: 31821327 PMCID: PMC6903744 DOI: 10.1371/journal.pone.0225611] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 11/07/2019] [Indexed: 12/11/2022] Open
Abstract
Undernutrition, as described by low body mass index (BMI), is a foremost risk factor for the progression of active Tuberculosis (TB). Undernutrition is also known to impact the baseline frequencies of innate and adaptive immune cells in animal models. To verify whether undernutrition has any influence on the baseline frequencies of immune cells in latent Mycobacterium tuberculosis infection (LTBI), we examined the frequencies of T cell-, B cell, monocyte- and dendritic cell (DC)- subsets in individuals with LTBI and low BMI (LBMI) and contrasted them with LTBI and normal BMI (NBMI) groups. LBMI was characterized by decreased frequencies and absolute cell counts of T cells, B cells and NK cells in comparison with NBMI. LBMI individuals demonstrated significantly enhanced frequencies of naïve and effector CD4+ and CD8+ T cells and significantly decreased frequencies of central memory, effector memory CD4+ and CD8+ T cells and regulatory T cells. Among B cell subsets, LBMI individuals demonstrated significantly diminished frequencies of naïve, immature, classical memory, activated memory, atypical memory and plasma cells. In addition, LBMI individuals showed significantly decreased frequencies of classical monocytes, myeloid DCs and plasmacytoid DCs and significantly increased frequencies of intermediate and non-classical monocytes and myeloid derived suppressor cells. BMI exhibited a positive correlation with B cell and NK cell counts. Our data, therefore, demonstrates that coexistent undernutrition in LTBI is characterized by the occurrence of a significant modulation in the frequency of innate and adaptive immune cell subsets.
Collapse
Affiliation(s)
- Anuradha Rajamanickam
- National Institute of Health-NIRT-International Center for Excellence in Research, Chennai, India
- * E-mail:
| | - Saravanan Munisankar
- National Institute of Health-NIRT-International Center for Excellence in Research, Chennai, India
| | | | - Subash Babu
- National Institute 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
|
67
|
Serum anti-Mce1A immunoglobulin detection as a tool for differential diagnosis of tuberculosis and latent tuberculosis infection in children and adolescents. Tuberculosis (Edinb) 2019; 120:101893. [PMID: 32090854 DOI: 10.1016/j.tube.2019.101893] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 11/28/2019] [Accepted: 12/01/2019] [Indexed: 11/21/2022]
|
68
|
Biffar L, Blunt L, Atkins W, Anderson P, Holder T, Xing Z, Vordermeier M, McShane H, Villarreal-Ramos B. Evaluating the sensitivity of the bovine BCG challenge model using a prime boost Ad85A vaccine regimen. Vaccine 2019; 38:1241-1248. [PMID: 31759733 DOI: 10.1016/j.vaccine.2019.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/30/2019] [Accepted: 11/04/2019] [Indexed: 10/25/2022]
Abstract
In the absence of biomarkers of protective immunity, newly developed vaccines against bovine tuberculosis need to be evaluated in virulent Mycobacterium bovis challenge experiments, which require the use of expensive and highly in demand Biological Safety Level 3 (BSL3) animal facilities. The recently developed bovine BCG challenge model offers a cheaper and faster way to test new vaccine candidates and additionally reduces the severity of the challenge compared to virulent M. bovis challenge in line with the remits of the NC3Rs. In this work we sought to establish the sensitivity of the BCG challenge model by testing a prime boost vaccine regimen that previously increased protection over BCG alone against M. bovis challenge. All animals, except the control group, were vaccinated subcutaneously with BCG Danish, and half of those were then boosted with a recombinant adenoviral vector expressing Antigen 85A, Ad85A. All animals were challenged with BCG Tokyo into the prescapular lymph node and the bacterial load within the lymph nodes was established. All vaccinated animals, independent of the vaccination regimen, cleared BCG significantly faster from the lymph node than control animals, suggesting a protective effect. There was however, no difference between the BCG and the BCG-Ad85A regimens. Additionally, we analysed humoral and cellular immune responses taken prior to challenge for possible predictors of protection. Cultured ELISpot identified significantly higher IFN-ɣ responses in protected vaccinated animals, relative to controls, but not in unprotected vaccinated animals. Furthermore, a trend for protected animals to produce more IFN-ɣ by quantitative PCR and intracellular staining was observed. Thus, this model can also be an attractive alternative to M. bovis challenge models for the discovery of protective biomarkers.
Collapse
Affiliation(s)
- Lucia Biffar
- Jenner Institute Oxford, Old Road Campus Research Build, Roosevelt Dr, Oxford OX3 7DQ, UK; TB Immunology and Vaccinology Team, Department of Bacteriology, Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK
| | - Laura Blunt
- TB Immunology and Vaccinology Team, Department of Bacteriology, Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK
| | - William Atkins
- TB Immunology and Vaccinology Team, Department of Bacteriology, Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK
| | - Paul Anderson
- TB Immunology and Vaccinology Team, Department of Bacteriology, Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK
| | - Tom Holder
- TB Immunology and Vaccinology Team, Department of Bacteriology, Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK
| | - Zhou Xing
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Martin Vordermeier
- TB Immunology and Vaccinology Team, Department of Bacteriology, Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK; Institute of Biological, Environmental & Rural Sciences (IBERS), Aberystwyth University, Penglais, Aberystwyth, Ceredigion SY23 3DA, UK
| | - Helen McShane
- Jenner Institute Oxford, Old Road Campus Research Build, Roosevelt Dr, Oxford OX3 7DQ, UK
| | - Bernardo Villarreal-Ramos
- TB Immunology and Vaccinology Team, Department of Bacteriology, Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK; Institute of Biological, Environmental & Rural Sciences (IBERS), Aberystwyth University, Penglais, Aberystwyth, Ceredigion SY23 3DA, UK.
| |
Collapse
|
69
|
Purification and Characterisation of Badger IgA and Its Detection in the Context of Tuberculosis. Vet Sci 2019; 6:vetsci6040089. [PMID: 31684068 PMCID: PMC6958344 DOI: 10.3390/vetsci6040089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/28/2019] [Accepted: 10/31/2019] [Indexed: 11/16/2022] Open
Abstract
European badgers are a wildlife reservoir of bovine tuberculosis in parts of Great Britain. Accurate diagnosis of tuberculosis in badgers is important for the development of strategies for the control of the disease. Sensitive serological tests for badger TB are needed for reasons such as cost and simplicity. Assay of mucosal IgA could be useful for diagnosing respiratory pathogens such as Mycobacterium bovis and for monitoring the response to mucosal vaccination. To develop an IgA assay, we purified secretory IgA from badger bile, identifying secretory component (SC), heavy chain (HC) and light chain (LC), at 66, 46 and 27 Kda, respectively, on the basis of size comparison with other species. Monoclonal antibodies (mAbs) were generated to purified IgA. We selected two for ELISA development. The detection limit of the IgA-specific mAbs was found to be approximately 20 ng/mL when titrated against purified badger bile. One monoclonal antibody specific for badger IgA was used to detect IgA in serum and tracheal aspirate with specificity to an immunodominant antigen of M. bovis. An M. bovis infection dose-dependent IgA response was observed in experimentally infected badgers. IgA was also detected by immunohistochemistry in the lungs of bTB-infected badgers. With further characterisation, these represent new reagents for the study of the IgA response in badgers.
Collapse
|
70
|
Yu X, Feng J, Huang L, Gao H, Liu J, Bai S, Wu B, Xie J. Molecular Basis Underlying Host Immunity Subversion by Mycobacterium tuberculosis PE/PPE Family Molecules. DNA Cell Biol 2019; 38:1178-1187. [PMID: 31580738 DOI: 10.1089/dna.2019.4852] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mycobacterium tuberculosis proline-glutamic acid (PE)/proline-proline-glutamic acid (PPE) family proteins, with >160 members, are crucial for virulence, cell wall, host cell fate, host Th1/Th2 balance, and CD8+ T cell recognition. Ca2+ signaling is involved in PE/PPE protein-mediated host-pathogen interaction. PE/PPE proteins also function in heme utilization and nitric oxide production. PE/PPE family proteins are intensively pursued as diagnosis biomarkers and vaccine components.
Collapse
Affiliation(s)
- Xiaowen Yu
- Chongqing Key Laboratory of Traditional Chinese Medicine to Prevent and Treat Autoimmune Diseases, Chongqing, P.R. China
| | - Jing Feng
- Chongqing Key Laboratory of Traditional Chinese Medicine to Prevent and Treat Autoimmune Diseases, Chongqing, P.R. China
| | - Lu Huang
- Department of Pathology, Xinqiao Hospital, Army Medical University, Chongqing, P.R. China
| | - Hongyan Gao
- Chongqing Key Laboratory of Traditional Chinese Medicine to Prevent and Treat Autoimmune Diseases, Chongqing, P.R. China
| | - Jinkun Liu
- Chongqing Key Laboratory of Traditional Chinese Medicine to Prevent and Treat Autoimmune Diseases, Chongqing, P.R. China
| | - Shutong Bai
- Chongqing Key Laboratory of Traditional Chinese Medicine to Prevent and Treat Autoimmune Diseases, Chongqing, P.R. China
| | - Bin Wu
- Chongqing Key Laboratory of Traditional Chinese Medicine to Prevent and Treat Autoimmune Diseases, Chongqing, P.R. China
| | - Jianping Xie
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, P.R. China
| |
Collapse
|
71
|
Bénard A, Sakwa I, Schierloh P, Colom A, Mercier I, Tailleux L, Jouneau L, Boudinot P, Al-Saati T, Lang R, Rehwinkel J, Loxton AG, Kaufmann SHE, Anton-Leberre V, O'Garra A, Sasiain MDC, Gicquel B, Fillatreau S, Neyrolles O, Hudrisier D. B Cells Producing Type I IFN Modulate Macrophage Polarization in Tuberculosis. Am J Respir Crit Care Med 2019; 197:801-813. [PMID: 29161093 DOI: 10.1164/rccm.201707-1475oc] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
RATIONALE In addition to their well-known function as antibody-producing cells, B lymphocytes can markedly influence the course of infectious or noninfectious diseases via antibody-independent mechanisms. In tuberculosis (TB), B cells accumulate in lungs, yet their functional contribution to the host response remains poorly understood. OBJECTIVES To document the role of B cells in TB in an unbiased manner. METHODS We generated the transcriptome of B cells isolated from Mycobacterium tuberculosis (Mtb)-infected mice and validated the identified key pathways using in vitro and in vivo assays. The obtained data were substantiated using B cells from pleural effusion of patients with TB. MEASUREMENTS AND MAIN RESULTS B cells isolated from Mtb-infected mice displayed a STAT1 (signal transducer and activator of transcription 1)-centered signature, suggesting a role for IFNs in B-cell response to infection. B cells stimulated in vitro with Mtb produced type I IFN, via a mechanism involving the innate sensor STING (stimulator of interferon genes), and antagonized by MyD88 (myeloid differentiation primary response 88) signaling. In vivo, B cells expressed type I IFN in the lungs of Mtb-infected mice and, of clinical relevance, in pleural fluid from patients with TB. Type I IFN expression by B cells induced an altered polarization of macrophages toward a regulatory/antiinflammatory profile in vitro. In vivo, increased provision of type I IFN by B cells in a murine model of B cell-restricted Myd88 deficiency correlated with an enhanced accumulation of regulatory/antiinflammatory macrophages in Mtb-infected lungs. CONCLUSIONS Type I IFN produced by Mtb-stimulated B cells favors macrophage polarization toward a regulatory/antiinflammatory phenotype during Mtb infection.
Collapse
Affiliation(s)
- Alan Bénard
- 1 Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier, Toulouse, France.,2 International Associated Laboratory CNRS "IM-TB/HIV (Immunometabolism and Macrophages in Tuberculosis/Human Immunodeficiency Virus-1 Co-infection)," Toulouse, France, and Buenos Aires, Argentina.,3 Department of Surgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - Imme Sakwa
- 4 Deutsches Rheuma-Forschungszentrum, a Leibniz Institute, Berlin, Germany
| | - Pablo Schierloh
- 2 International Associated Laboratory CNRS "IM-TB/HIV (Immunometabolism and Macrophages in Tuberculosis/Human Immunodeficiency Virus-1 Co-infection)," Toulouse, France, and Buenos Aires, Argentina.,5 Instituto de Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas, Academia Nacional de Medicina, Pacheco de Melo, Buenos Aires, Argentina
| | - André Colom
- 1 Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier, Toulouse, France.,2 International Associated Laboratory CNRS "IM-TB/HIV (Immunometabolism and Macrophages in Tuberculosis/Human Immunodeficiency Virus-1 Co-infection)," Toulouse, France, and Buenos Aires, Argentina
| | - Ingrid Mercier
- 1 Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier, Toulouse, France.,2 International Associated Laboratory CNRS "IM-TB/HIV (Immunometabolism and Macrophages in Tuberculosis/Human Immunodeficiency Virus-1 Co-infection)," Toulouse, France, and Buenos Aires, Argentina.,6 Laboratoire d'Ingénierie des Systèmes Biologiques et des Procédés, Université de Toulouse, CNRS, Institut National de la Recherche Agronomique (INRA), Institut National des Sciences Appliquées, Toulouse, France
| | - Ludovic Tailleux
- 7 Unit of Mycobacterial Genetics and.,8 Unit for Integrated Mycobacterial Pathogenomics, Institut Pasteur, Paris, France
| | - Luc Jouneau
- 9 Virologie et Immunologie Moléculaires, INRA, Jouy-en-Josas, France
| | - Pierre Boudinot
- 9 Virologie et Immunologie Moléculaires, INRA, Jouy-en-Josas, France
| | - Talal Al-Saati
- 10 Institut National de la Santé et de la Recherche Médicale (INSERM)/Université Paul Sabatier/École Nationale Vétérinaire de Toulouse/Centre Régional d'Exploration Fonctionnelle et Ressources Expérimentales, Service d'Histopathologie, Centre Hospitalier Universitaire, Purpan, Toulouse, France
| | - Roland Lang
- 11 Institute of Clinical Microbiology, Immunology, and Hygiene, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Jan Rehwinkel
- 12 Radcliffe Department of Medicine, Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Andre G Loxton
- 13 South African Medical Research Council Centre for Tuberculosis Research, Department of Science and Technology/National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Stellenbosch University, Cape Town, South Africa
| | - Stefan H E Kaufmann
- 14 Department of Immunology, Max Planck Institute of Infection Biology, Berlin, Germany
| | - Véronique Anton-Leberre
- 6 Laboratoire d'Ingénierie des Systèmes Biologiques et des Procédés, Université de Toulouse, CNRS, Institut National de la Recherche Agronomique (INRA), Institut National des Sciences Appliquées, Toulouse, France
| | - Anne O'Garra
- 15 Division of Immunoregulation, Medical Research Council, National Institute for Medical Research, London, United Kingdom.,16 National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Maria Del Carmen Sasiain
- 2 International Associated Laboratory CNRS "IM-TB/HIV (Immunometabolism and Macrophages in Tuberculosis/Human Immunodeficiency Virus-1 Co-infection)," Toulouse, France, and Buenos Aires, Argentina.,5 Instituto de Medicina Experimental-Consejo Nacional de Investigaciones Científicas y Técnicas, Academia Nacional de Medicina, Pacheco de Melo, Buenos Aires, Argentina
| | - Brigitte Gicquel
- 9 Virologie et Immunologie Moléculaires, INRA, Jouy-en-Josas, France
| | - Simon Fillatreau
- 4 Deutsches Rheuma-Forschungszentrum, a Leibniz Institute, Berlin, Germany.,17 Institut Necker-Enfants Malades, INSERM U1151-CNRS Unité Mixte de Recherche 8253, Paris, France.,18 Université Paris Descartes, Sorbonne Paris Cité, Paris, France; and.,19 Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants Malades, Paris, France
| | - Olivier Neyrolles
- 1 Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier, Toulouse, France.,2 International Associated Laboratory CNRS "IM-TB/HIV (Immunometabolism and Macrophages in Tuberculosis/Human Immunodeficiency Virus-1 Co-infection)," Toulouse, France, and Buenos Aires, Argentina
| | - Denis Hudrisier
- 1 Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier, Toulouse, France.,2 International Associated Laboratory CNRS "IM-TB/HIV (Immunometabolism and Macrophages in Tuberculosis/Human Immunodeficiency Virus-1 Co-infection)," Toulouse, France, and Buenos Aires, Argentina
| |
Collapse
|
72
|
Ernst JD. Float Like Bacilli, STING Like a B: Type I Interferons in Tuberculosis. Am J Respir Crit Care Med 2019; 197:706-707. [PMID: 29262261 DOI: 10.1164/rccm.201712-2368ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Joel D Ernst
- 1 Division of Infectious Diseases and Immunology New York University School of Medicine New York, New York
| |
Collapse
|
73
|
Wang S, Chang E, Byanyima P, Huang P, Sanyu I, Musisi E, Sessolo A, Davis JL, Worodria W, Huang L, Lin J. Association between common telomere length genetic variants and telomere length in an African population and impacts of HIV and TB. J Hum Genet 2019; 64:1033-1040. [PMID: 31388112 DOI: 10.1038/s10038-019-0646-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 07/14/2019] [Accepted: 07/15/2019] [Indexed: 01/29/2023]
Abstract
Prior studies in predominantly European (Caucasian) populations have discovered common genetic variants (single nucleotide polymorphisms, SNPs) associated with leukocyte telomere length (LTL), but whether these same variants affect LTL in non-Caucasian populations are largely unknown. We investigated whether six genetic variants previously associated with LTL (TERC (rs10936599), TERT (rs2736100), NAF1 (7675998), OBFC1 (rs9420907), ZNF208 (rs8105767), and RTEL1 (rs755017)) are correlated with telomere length (TL) in peripheral blood mononuclear cells (PBMCs) in a cohort of Africans living with and without HIV and undergoing evaluation for tuberculosis (TB). We found OBFC1 and the genetic sum score of the effect alleles across all six loci to be associated with shorter TL (adjusted for age, gender, HIV status, and smoking pack-years (p < 0.02 for both OBFC1 and the genetic sum score). In an analysis stratified by HIV status, the genetic sum score is associated with LTL in both groups with and without HIV. On the contrary, a stratified analysis according to TB status revealed that in the TB-positive subgroup, the genetic sum score is not associated with LTL, whereas the relationship remains in the TB-negative subgroup. The different impacts of HIV and TB on the association between the genetic sum score and LTL indicate different modes of modification and suggest that the results found in this cohort with HIV and TB participants may not be applied to the African general population. Future studies need to carefully consider these confounding factors.
Collapse
Affiliation(s)
- Stephanie Wang
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Emily Chang
- HIV, Infectious Diseases, and Global Medicine Division, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | | | - Peter Huang
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Ingvar Sanyu
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Emmanuel Musisi
- Infectious Diseases Research Collaboration, Kampala, Uganda.,Department of Biochemistry, College of Natural Sciences, Makerere University, Kampala, Uganda
| | - Abdul Sessolo
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - J Lucian Davis
- Epidemiology of Microbial Diseases, Yale School of Public Health and Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - William Worodria
- Infectious Diseases Research Collaboration, Kampala, Uganda.,Makerere University-University of California San Francisco (MU-UCSF) Research Collaboration, Kampala, Uganda
| | - Laurence Huang
- HIV, Infectious Diseases, and Global Medicine Division, Department of Medicine, University of California San Francisco, San Francisco, CA, USA.,Makerere University-University of California San Francisco (MU-UCSF) Research Collaboration, Kampala, Uganda.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Jue Lin
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA.
| | | |
Collapse
|
74
|
Dai YC, Wang WD, Zhang JA, Chen C, Luo HL, Xu H, Peng Y, Luo H, Yang XR, Chen X, Wu XJ, Chen GH, Chen ZW, Xu JF. MTB driven B cells producing IL-35 and secreting high level of IL-10 in the patients with active pulmonary tuberculosis. Mol Immunol 2019; 112:175-181. [PMID: 31170628 DOI: 10.1016/j.molimm.2019.05.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/09/2019] [Accepted: 05/24/2019] [Indexed: 01/04/2023]
Abstract
Regulatory B cells (Bregs) have critical roles as a negative regulator of immunity, mainly due to the fact that it secrets high a level of interleukin 10 (IL-10). Recently, a new subset of Bregs was identified as a key source of IL-35, which is an immunosuppressive cytokine and conventionally thought to be secreted by regulatory T cells (Tregs). Our previous study showed that the level of IL-35 in serum was elevated in the patients with active tuberculosis (ATB). However, none of the studies reported that IL-35 is secreted by B cells in ATB patients. In the current study, we found that the mRNA expressions of the both subunits (p35 and Ebi3) of IL-35 by circulating B cells were increased in ATB patients. By using immunohistochemistry and immunofluorescence staining, we found a subset of B cells infiltrated into the tuberculous granuloma of ATB patients also expressed IL-35. Moreover, Mycobacterium tuberculosis (MTB) lysate stimulation assay also demonstrated higher levels of IL-35 were exerted by MTB lysate within purified B cells from healthy control group (HC). Flow cytometry analysis further showed that the IL-35-producing B cells from ATB patients produced a higher level of IL-10. Taken together, IL-35-producing B cells may play a regulatory role during MTB infection by producing IL-10.
Collapse
Affiliation(s)
- You-Chao Dai
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical University, Dongguan, 523808, China; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, China.
| | - Wan-Dang Wang
- Department of Clinical medicine laboratory, Affiliated Xiaolan Hospital, Southern Medical University, China.
| | - Jun-Ai Zhang
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical University, Dongguan, 523808, China; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, China.
| | - Chen Chen
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical University, Dongguan, 523808, China; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, China.
| | - Hou-Long Luo
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical University, Dongguan, 523808, China; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, China.
| | - Huan Xu
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical University, Dongguan, 523808, China; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, China.
| | - Ying Peng
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical University, Dongguan, 523808, China; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, China.
| | - Hong Luo
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical University, Dongguan, 523808, China; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, China.
| | - Xu-Ran Yang
- Department of Clinical medicine laboratory, Affiliated Xiaolan Hospital, Southern Medical University, China.
| | - Xinchun Chen
- Guangdong Key Laboratory for Emerging Infectious Diseases, Shenzhen Third People's Hospital, Guangdong Medical University, Shenzhen, 518020, China.
| | - Xian-Jin Wu
- Department of Clinical Laboratory, Huizhou Municipal Central Hospital, China.
| | - Guang-Hui Chen
- Department of Clinical medicine laboratory, Affiliated Xiaolan Hospital, Southern Medical University, China.
| | - Zheng W Chen
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine, USA.
| | - Jun-Fa Xu
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical University, Dongguan, 523808, China; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
75
|
Loxton AG. Bcells and their regulatory functions during Tuberculosis: Latency and active disease. Mol Immunol 2019; 111:145-151. [PMID: 31054408 DOI: 10.1016/j.molimm.2019.04.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 03/20/2019] [Accepted: 04/23/2019] [Indexed: 12/24/2022]
Abstract
Tuberculosis (TB) is a global epidemic with devastating consequences. Emerging evidence suggests that B-cells have the ability to modulate the immune response and understanding these roles during Mycobacterium tuberculosis (M.tb) infection can help to find new strategies to treat TB. The immune system of individuals with pulmonary TB form granulomas in the lung which controls the infection by inhibiting the M.tb growth and acts as a physical barrier. Thereafter, surviving M.tb become dormant and in most cases the host's immunity prevents TB reactivation. B-cells execute several immunological functions and are regarded as protective regulators of immune responses by antibody and cytokine production, as well as presenting antigen. Some of these B-cells, or regulatory B-cells, have been shown to express death-inducing ligands, such as Fas ligand (FasL). This expression and binding to the Fas receptor leads to apoptosis, a major immune regulation mechanism, in addition to the ability to induce T-cell tolerance. Here, I discuss the relevance of B-cells, in particular their non-humoral functions by addressing their regulatory properties during M.tb infection.
Collapse
Affiliation(s)
- Andre G Loxton
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241 Cape Town, 8000, South Africa.
| |
Collapse
|
76
|
Chen S, Sandford S, Kirman JR, Rehm BHA. Innovative antigen carrier system for the development of tuberculosis vaccines. FASEB J 2019; 33:7505-7518. [PMID: 30870010 DOI: 10.1096/fj.201802501rr] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A major obstacle to tuberculosis (TB)-subunit-vaccine development has been the induction of inadequate levels of protective immunity due to the limited breadth of antigen in vaccine preparations. In this study, immunogenic mycobacterial fusion peptides Ag85B-TB10.4 and Ag85B-TB10.4-Rv2660c were covalently displayed on the surface of self-assembled polyester particles. This study investigated whether polyester particles displaying mycobacterial antigens could provide augmented immunogenicity (i.e., offer an innovative vaccine formulation) when compared with free soluble antigens. Herein, polyester particle-based particulate vaccines were produced in an endotoxin-free Escherichia coli strain and emulsified with the adjuvant dimethyl dioctadecyl ammonium bromide. C57BL/6 mice were used to study the immunogenicity of formulated particulate vaccines. The result of humoral immunity showed the antibodies only interacted with target antigens and not with PhaC and the background proteins of the production host. The analysis of T helper 1 cellular immunity indicated that a relatively strong production of cellular immunity biomarkers, IFN-γ and IL-17A cytokines, was induced by particulate vaccines when compared with the respective soluble controls. This study demonstrated that polyester particles have the potential to perform as a mycobacterial antigen-delivery agent to induce augmented antigen-specific immune responses in contrast to free soluble vaccines.-Chen, S., Sandford, S., Kirman, J. R., Rehm, B. H. A. Innovative antigen carrier system for the development of tuberculosis vaccines.
Collapse
Affiliation(s)
- Shuxiong Chen
- Institute of Fundamental Sciences, Massey University Manawatu, Palmerston North, New Zealand
| | - Sarah Sandford
- Microbiology and Immunology Department, Otago University, Dunedin, New Zealand; and
| | - Joanna R Kirman
- Microbiology and Immunology Department, Otago University, Dunedin, New Zealand; and
| | - Bernd H A Rehm
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan Campus, Brisbane, Queensland, Australia
| |
Collapse
|
77
|
Counoupas C, Triccas JA, Britton WJ. Deciphering protective immunity against tuberculosis: implications for vaccine development. Expert Rev Vaccines 2019; 18:353-364. [PMID: 30793629 DOI: 10.1080/14760584.2019.1585246] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The development of more effective tuberculosis (TB) vaccines is essential for the global control of TB. Recently, there have been major advances in the field, but an important hindrance remains the lack of correlates of protection against TB. This requires each vaccine candidate to undergo clinical efficacy trials based on data from animal protection studies, but the results from animal models do not necessarily predict efficacy in humans. AREAS COVERED In this review we summarize our current knowledge of immune mechanisms that may contribute to protective immunity against TB following vaccination and relate these to protective efficacy in animal models and recent clinical trials. Although some initial trials did not reproduce protection against TB in humans, recent trials have demonstrated promising efficacy for three vaccine approaches. EXPERT OPINION Although CD4+ T lymphocytes are essential for protection against TB, there is no clear correlation between conventional CD4+ or CD8+ T cell responses and protective efficacy of TB vaccines. Recent attention has focused on other immune responses, including donor unrestricted T cells, B lymphocytes, and antibodies. Prospective studies on samples from vaccinated individuals protected in recent trials will allow evaluation of these alternative immune mechanisms as potential correlates of protection.
Collapse
Affiliation(s)
- Claudio Counoupas
- a Tuberculosis Research Program Centenary Institute , The University of Sydney , Camperdown , NSW , Australia.,b The University of Sydney , Central Clinical School Faculty of Medicine and Health , Sydney , NSW , Australia
| | - James A Triccas
- a Tuberculosis Research Program Centenary Institute , The University of Sydney , Camperdown , NSW , Australia
| | - Warwick J Britton
- a Tuberculosis Research Program Centenary Institute , The University of Sydney , Camperdown , NSW , Australia.,b The University of Sydney , Central Clinical School Faculty of Medicine and Health , Sydney , NSW , Australia
| |
Collapse
|
78
|
Konečný P, Ehrlich R, Gulumian M, Jacobs M. Immunity to the Dual Threat of Silica Exposure and Mycobacterium tuberculosis. Front Immunol 2019; 9:3069. [PMID: 30687311 PMCID: PMC6334662 DOI: 10.3389/fimmu.2018.03069] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 12/11/2018] [Indexed: 01/28/2023] Open
Abstract
Exposure to silica and the consequent development of silicosis are well-known health problems in countries with mining and other dust producing industries. Apart from its direct fibrotic effect on lung tissue, chronic and immunomodulatory character of silica causes susceptibility to tuberculosis (TB) leading to a significantly higher TB incidence in silica-exposed populations. The presence of silica particles in the lung and silicosis may facilitate initiation of tuberculous infection and progression to active TB, and exacerbate the course and outcome of TB, including prognosis and survival. However, the exact mechanisms of the involvement of silica in the pathological processes during mycobacterial infection are not yet fully understood. In this review, we focus on the host's immunological response to both silica and Mycobacterium tuberculosis, on agents of innate and adaptive immunity, and particularly on silica-induced immunological modifications in co-exposure that influence disease pathogenesis. We review what is known about the impact of silica and Mycobacterium tuberculosis or their co-exposure on the host's immune system, especially an impact that goes beyond an exclusive focus on macrophages as the first line of the defense. In both silicosis and TB, acquired immunity plays a major role in the restriction and/or elimination of pathogenic agents. Further research is needed to determine the effects of silica in adaptive immunity and in the pathogenesis of TB.
Collapse
Affiliation(s)
- Petr Konečný
- Centre for Environmental and Occupational Health, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa.,Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Rodney Ehrlich
- Centre for Environmental and Occupational Health, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Mary Gulumian
- National Health Laboratory Service, Department of Toxicology and Biochemistry, National Institute for Occupational Health, Johannesburg, South Africa.,Division of Molecular Medicine and Haematology, University of the Witwatersrand, Johannesburg, South Africa.,National Health Laboratory Service, Johannesburg, South Africa
| | - Muazzam Jacobs
- Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,National Health Laboratory Service, Johannesburg, South Africa.,Immunology of Infectious Disease Research Unit, South African Medical Research Council, Cape Town, South Africa
| |
Collapse
|
79
|
Abstract
Mycobacterium tuberculosis is the leading cause of death worldwide from a single bacterial pathogen. The World Health Organization estimates that annually 1 million children have tuberculosis (TB) disease and many more harbor a latent form. Accurate estimates are hindered by under-recognition and challenges in diagnosis. To date, an accurate diagnostic test to confirm TB in children does not exist. Treatment is lengthy but outcomes are generally favorable with timely initiation. With the End TB Strategy, there is an urgent need for improved diagnostics and treatment to prevent the unnecessary morbidity and mortality from TB in children.
Collapse
Affiliation(s)
- Tania A Thomas
- Division of Infectious Diseases and International Health, University of Virginia, PO Box 801340, Charlottesville, VA 22908-1340, USA.
| |
Collapse
|
80
|
Yuan C, Qu ZL, Tang XL, Liu Q, Luo W, Huang C, Pan Q, Zhang XL. Mycobacterium tuberculosis Mannose-Capped Lipoarabinomannan Induces IL-10-Producing B Cells and Hinders CD4 +Th1 Immunity. iScience 2018; 11:13-30. [PMID: 30572206 PMCID: PMC6299163 DOI: 10.1016/j.isci.2018.11.039] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/08/2018] [Accepted: 11/28/2018] [Indexed: 12/26/2022] Open
Abstract
The importance of Th1/interferon (IFN)-γ-mediated responses in mycobacterial infection has been well established. However, little is known about B cell-mediated immunity during Mycobacterium tuberculosis (Mtb) infection. Interleukin (IL)-10-producing B cells (B10 cells), a subset of B regulatory cells (Bregs), are implicated in modulating the immune response. Herein, we found that B10 cells were significantly increased in patients with tuberculosis. Furthermore, mannose-capped lipoarabinomannan (ManLAM), a major surface lipoglycan component from Mtb, induced a significant increase in B10 cells, which enriched in CD5+ B1a B cells. ManLAM induced IL-10 production mainly by activating MyD88/PI3K/AKT/Ap-1 and K63-linked ubiquitination of NF-κB essential modulator/nuclear factor kappa-light-chain-enhancer of activated B cells signaling pathways in B cells via Toll-like receptor 2. IL-10 production by ManLAM-treated B cells further inhibited CD4+ Th1 polarization, leading to increased susceptibility to mycobacterial infection compared with ManLAM-treated IL-10−/− B group. Thus, we report a new immunoregulation mechanism in which Mtb ManLAM-induced B10 cells negatively regulate host anti-TB cellular immunity. Mtb mannose-capped lipoarabinomannan (ManLAM) induces IL-10 production in B cells ManLAM-induced B10 cells enrich in CD5+ B1a B cells ManLAM binding with TLR2 triggers MyD88 signaling pathways of B cells ManLAM-induced B10 cells hinder CD4+Th1 immunity during Mtb infection in mice
Collapse
Affiliation(s)
- Chunhui Yuan
- State Key Laboratory of Virology and Medical Research Institute, Hubei Province Key Laboratory of Allergy and Immunology and Department of Immunology, Wuhan University School of Basic Medical Sciences, Wuchang, Wuhan 430071, China; Department of Laboratory Medicine, Wuhan Children's Hospital, Huazhong University of Science and Technology, Jiangan, Wuhan 430015, China
| | - Zi-Lu Qu
- State Key Laboratory of Virology and Medical Research Institute, Hubei Province Key Laboratory of Allergy and Immunology and Department of Immunology, Wuhan University School of Basic Medical Sciences, Wuchang, Wuhan 430071, China
| | - Xiao-Lei Tang
- State Key Laboratory of Virology and Medical Research Institute, Hubei Province Key Laboratory of Allergy and Immunology and Department of Immunology, Wuhan University School of Basic Medical Sciences, Wuchang, Wuhan 430071, China
| | - Qi Liu
- State Key Laboratory of Virology and Medical Research Institute, Hubei Province Key Laboratory of Allergy and Immunology and Department of Immunology, Wuhan University School of Basic Medical Sciences, Wuchang, Wuhan 430071, China
| | - Wei Luo
- State Key Laboratory of Virology and Medical Research Institute, Hubei Province Key Laboratory of Allergy and Immunology and Department of Immunology, Wuhan University School of Basic Medical Sciences, Wuchang, Wuhan 430071, China
| | - Chun Huang
- State Key Laboratory of Virology and Medical Research Institute, Hubei Province Key Laboratory of Allergy and Immunology and Department of Immunology, Wuhan University School of Basic Medical Sciences, Wuchang, Wuhan 430071, China
| | - Qin Pan
- State Key Laboratory of Virology and Medical Research Institute, Hubei Province Key Laboratory of Allergy and Immunology and Department of Immunology, Wuhan University School of Basic Medical Sciences, Wuchang, Wuhan 430071, China.
| | - Xiao-Lian Zhang
- State Key Laboratory of Virology and Medical Research Institute, Hubei Province Key Laboratory of Allergy and Immunology and Department of Immunology, Wuhan University School of Basic Medical Sciences, Wuchang, Wuhan 430071, China.
| |
Collapse
|
81
|
Wu S, Shi H, Cao N, Ye L, Yu C, Zheng L. The correlation of immunologic derangement and juvenile recurrent parotitis: an investigation of the laboratory immunological observation. Acta Otolaryngol 2018; 138:1112-1116. [PMID: 30702022 DOI: 10.1080/00016489.2018.1515498] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND Juvenile recurrent parotitis (JRP) is defined as recurrent parotid inflammation, generally associated with nonobstructive sialectasis of the parotid gland. In addition, the etiology remains unclear, probably immunologically mediated. AIM The purposes of the present study were to report the relationship between JRP and immune function from the measurement of the JRP patients' immunoglobulins and T-lymphocyte subset. METHODS Immunologic assay from 2014 to 2017 of 100 children diagnosed with JRP at Shanghai Ninth Hospital compared with the 100 normal children by age. RESULTS The CD4 level of JRP children aged >6 years was significant lower than the one of JRP preschool children (p < .05), while the IgG level was significant higher than the one of the JRP preschool children (p < .05). In comparison with the normal children, the value of CD8 T cells, immunoglobulin G (IgG), immunoglobulin E (IgE), immunoglobulin A (IgA) and C3 (p < .01) of JRP children was significant higher, while the value of CD4 T cells was lower (p < .01) in spite of age. What is more, the value of CD8 T cells of JRP preschool children was much significant higher than the one of the normal preschool children (p < .01). CONCLUSION The immune function of JRP patients may become disorder: the suppression cellular immune function and inadequate humoral immune expression.
Collapse
Affiliation(s)
- Shufeng Wu
- Department of Oral Surgery, Shanghai 9th Peoples Hospital Affiliated to Shanghai, Jiaotong University School of Medicine, Shanghai, China
| | - Huan Shi
- Department of Oral Surgery, Shanghai 9th Peoples Hospital Affiliated to Shanghai, Jiaotong University School of Medicine, Shanghai, China
| | - Ningning Cao
- Department of Oral Surgery, Shanghai 9th Peoples Hospital Affiliated to Shanghai, Jiaotong University School of Medicine, Shanghai, China
| | - Lei Ye
- Department of Oral Surgery, Shanghai 9th Peoples Hospital Affiliated to Shanghai, Jiaotong University School of Medicine, Shanghai, China
| | - Chuangqi Yu
- Department of Oral Surgery, Shanghai 9th Peoples Hospital Affiliated to Shanghai, Jiaotong University School of Medicine, Shanghai, China
| | - Lingyan Zheng
- Department of Oral Surgery, Shanghai 9th Peoples Hospital Affiliated to Shanghai, Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
82
|
Bekale RB, Du Plessis SM, Hsu NJ, Sharma JR, Sampson SL, Jacobs M, Meyer M, Morse GD, Dube A. Mycobacterium Tuberculosis and Interactions with the Host Immune System: Opportunities for Nanoparticle Based Immunotherapeutics and Vaccines. Pharm Res 2018; 36:8. [PMID: 30411187 PMCID: PMC6362825 DOI: 10.1007/s11095-018-2528-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 10/17/2018] [Indexed: 02/06/2023]
Abstract
Tuberculosis (TB) caused by Mycobacterium tuberculosis remains a deadly infectious disease. The thin pipeline of new drugs for TB, the ineffectiveness in adults of the only vaccine available, i.e. the Bacillus Calmette-Guerin vaccine, and increasing global antimicrobial resistance, has reinvigorated interest in immunotherapies. Nanoparticles (NPs) potentiate the effect of immune modulating compounds (IMC), enabling cell targeting, improved transfection of antigens, enhanced compound stability and provide opportunities for synergistic action, via delivery of multiple IMCs. In this review we describe work performed in the application of NPs towards achieving immune modulation for TB treatment and vaccination. Firstly, we present a comprehensive review of M. tuberculosis and how the bacterium modulates the host immune system. We find that current work suggest great promise of NP based immunotherapeutics as novel treatments and vaccination systems. There is need to intensify research efforts in this field, and rationally design novel NP immunotherapeutics based on current knowledge of the mycobacteriology and immune escape mechanisms employed by M. tuberculosis.
Collapse
Affiliation(s)
- Raymonde B Bekale
- Discipline of Pharmaceutics, School of Pharmacy, University of the Western Cape, Cape Town, South Africa
| | - Su-Mari Du Plessis
- NRF-DST Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Nai-Jen Hsu
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Jyoti R Sharma
- National Health Laboratory Service, Johannesburg, South Africa
| | - Samantha L Sampson
- NRF-DST Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Muazzam Jacobs
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- National Health Laboratory Service, Johannesburg, South Africa
- Immunology of Infectious Disease Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Mervin Meyer
- DST/Mintek Nanotechnology Innovation Centre (NIC), Biolabels Unit, Department of Biotechnology, University of the Western Cape (UWC), Cape Town, South Africa
| | - Gene D Morse
- AIDS Clinical Trials Group Pharmacology Specialty Laboratory, New York State Center of Excellence in Bioinformatics and Life Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Admire Dube
- Discipline of Pharmaceutics, School of Pharmacy, University of the Western Cape, Cape Town, South Africa.
| |
Collapse
|
83
|
Dyatlov AV, Apt AS, Linge IA. B lymphocytes in anti-mycobacterial immune responses: Pathogenesis or protection? Tuberculosis (Edinb) 2018; 114:1-8. [PMID: 30711147 DOI: 10.1016/j.tube.2018.10.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/12/2018] [Accepted: 10/23/2018] [Indexed: 12/12/2022]
Abstract
The role of B cells and antibodies in tuberculosis (TB) immunity, protection and pathogenesis remain contradictory. The presence of organized B cell follicles close to active TB lesions in the lung tissue raises the question about the role of these cells in local host-pathogen interactions. In this short review, we summarize the state of our knowledge concerning phenotypes of B cells populating tuberculous lungs, their secretory activity, interactions with other immune cells and possible involvement in protective vs. pathogenic TB immunity.
Collapse
Affiliation(s)
- Alexander V Dyatlov
- Laboratory for Immunogenetics, Central Institute for Tuberculosis, Moscow, Russia
| | - Alexander S Apt
- Laboratory for Immunogenetics, Central Institute for Tuberculosis, Moscow, Russia; Department of Immunology, School of Biology, M. V. Lomonosov Moscow State University, Russia.
| | - Irina A Linge
- Laboratory for Immunogenetics, Central Institute for Tuberculosis, Moscow, Russia
| |
Collapse
|
84
|
López V, Alberdi P, Fuente JDL. Common Strategies, Different Mechanisms to Infect the Host: Anaplasma and Mycobacterium. Tuberculosis (Edinb) 2018. [DOI: 10.5772/intechopen.71535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
85
|
Chen S, Sandford S, Kirman J, Rehm BHA. Design of Bacterial Inclusion Bodies as Antigen Carrier Systems. ACTA ACUST UNITED AC 2018. [DOI: 10.1002/adbi.201800118] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Shuxiong Chen
- Institute of Fundamental Sciences; Massey University Manawatu; Palmerston North 4474 New Zealand
| | - Sarah Sandford
- Microbiology and Immunology Department; Otago University; Dunedin 9054 New Zealand
| | - Joanna Kirman
- Microbiology and Immunology Department; Otago University; Dunedin 9054 New Zealand
| | - Bernd H. A. Rehm
- Centre for Cell Factories and Biopolymers; Griffith Institute for Drug Discovery; Griffith University Nathan Campus; Brisbane 4111 Australia
| |
Collapse
|
86
|
Mycobacterium indicus pranii protein MIP_05962 induces Th1 cell mediated immune response in mice. Int J Med Microbiol 2018; 308:1000-1008. [PMID: 30190103 DOI: 10.1016/j.ijmm.2018.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 08/15/2018] [Accepted: 08/27/2018] [Indexed: 01/25/2023] Open
Abstract
Utility of Mycobacterium indicus pranii (MIP) as a multistage vaccine against mycobacterial infections demands identification of its protective antigens. We explored antigenicity and immunogenicity of a candidate protein MIP_05962 that depicts homology to HSP18 of M. leprae and antigen1 of Mycobacterium tuberculosis. This protein elicited substantial antibody response in immunized mice along with modulation of cellular immune response towards protective Th1 type. Both CD4+ and CD8+ subsets from immunized mice produced hallmark protective cytokines, IFN-γ, TNF-α and IL-2. This protein also enhanced the CD4+ effector memory that could act as first line of defence during infections. These results point to MIP_05962 as a protective antigen that contributes, in conjunction with others, to the protective immunity of this live vaccine candidate.
Collapse
|
87
|
Bai X, Aerts SL, Verma D, Ordway DJ, Chan ED. Epidemiologic Evidence of and Potential Mechanisms by Which Second-Hand Smoke Causes Predisposition to Latent and Active Tuberculosis. Immune Netw 2018; 18:e22. [PMID: 29984040 PMCID: PMC6026693 DOI: 10.4110/in.2018.18.e22] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/15/2018] [Accepted: 06/16/2018] [Indexed: 12/13/2022] Open
Abstract
Many studies have linked cigarette smoke (CS) exposure and tuberculosis (TB) infection and disease although much fewer have studied second-hand smoke (SHS) exposure. Our goal is to review the epidemiologic link between SHS and TB as well as to summarize the effects SHS and direct CS on various immune cells relevant for TB. PubMed searches were performed using the key words "tuberculosis" with "cigarette," "tobacco," or "second-hand smoke." The bibliography of relevant papers were examined for additional relevant publications. Relatively few studies associate SHS exposure with TB infection and active disease. Both SHS and direct CS can alter various components of host immunity resulting in increased vulnerability to TB. While the epidemiologic link of these 2 health maladies is robust, more definitive, mechanistic studies are required to prove that SHS and direct CS actually cause increased susceptibility to TB.
Collapse
Affiliation(s)
- Xiyuan Bai
- Department of Medicine, Denver Veterans Affairs Medical Center, University of Colorado Anschutz Medical Center, Denver, CO 80045, USA
- Department of Medicine and Office of Academic Affairs, National Jewish Health, Denver, CO 80206, USA
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Denver, CO 80045, USA
| | - Shanae L. Aerts
- Department of Medicine and Office of Academic Affairs, National Jewish Health, Denver, CO 80206, USA
| | - Deepshikha Verma
- Department of Microbiology, Immunology, and Pathology, Mycobacteria Research Laboratories, Colorado State University, Fort Collins, CO 80523, USA
| | - Diane J. Ordway
- Department of Microbiology, Immunology, and Pathology, Mycobacteria Research Laboratories, Colorado State University, Fort Collins, CO 80523, USA
| | - Edward D. Chan
- Department of Medicine, Denver Veterans Affairs Medical Center, University of Colorado Anschutz Medical Center, Denver, CO 80045, USA
- Department of Medicine and Office of Academic Affairs, National Jewish Health, Denver, CO 80206, USA
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Denver, CO 80045, USA
| |
Collapse
|
88
|
Choudhary A, Patel D, Honnen W, Lai Z, Prattipati RS, Zheng RB, Hsueh YC, Gennaro ML, Lardizabal A, Restrepo BI, Garcia-Viveros M, Joe M, Bai Y, Shen K, Sahloul K, Spencer JS, Chatterjee D, Broger T, Lowary TL, Pinter A. Characterization of the Antigenic Heterogeneity of Lipoarabinomannan, the Major Surface Glycolipid of Mycobacterium tuberculosis, and Complexity of Antibody Specificities toward This Antigen. THE JOURNAL OF IMMUNOLOGY 2018; 200:3053-3066. [PMID: 29610143 PMCID: PMC5911930 DOI: 10.4049/jimmunol.1701673] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 02/16/2018] [Indexed: 12/17/2022]
Abstract
Lipoarabinomannan (LAM), the major antigenic glycolipid of Mycobacterium tuberculosis, is an important immunodiagnostic target for detecting tuberculosis (TB) infection in HIV-1–coinfected patients, and is believed to mediate a number of functions that promote infection and disease development. To probe the human humoral response against LAM during TB infection, several novel LAM-specific human mAbs were molecularly cloned from memory B cells isolated from infected patients and grown in vitro. The fine epitope specificities of these Abs, along with those of a panel of previously described murine and phage-derived LAM-specific mAbs, were mapped using binding assays against LAM Ags from several mycobacterial species and a panel of synthetic glycans and glycoconjugates that represented diverse carbohydrate structures present in LAM. Multiple reactivity patterns were seen that differed in their specificity for LAM from different species, as well as in their dependence on arabinofuranoside branching and nature of capping at the nonreducing termini. Competition studies with mAbs and soluble glycans further defined these epitope specificities and guided the design of highly sensitive immunodetection assays capable of detecting LAM in urine of TB patients, even in the absence of HIV-1 coinfection. These results highlighted the complexity of the antigenic structure of LAM and the diversity of the natural Ab response against this target. The information and novel reagents described in this study will allow further optimization of diagnostic assays for LAM and may facilitate the development of potential immunotherapeutic approaches to inhibit the functional activities of specific structural motifs in LAM.
Collapse
Affiliation(s)
- Alok Choudhary
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103
| | - Deendayal Patel
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103
| | - William Honnen
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103
| | - Zhong Lai
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103
| | - Raja Sekhar Prattipati
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103
| | - Ruixiang Blake Zheng
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Ying-Chao Hsueh
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103
| | - Maria Laura Gennaro
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103
| | - Alfred Lardizabal
- Global Tuberculosis Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103
| | - Blanca I Restrepo
- University of Texas Health Science Center at Houston, School of Public Health at Brownsville, Brownsville, TX 78520
| | | | - Maju Joe
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Yu Bai
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Ke Shen
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Kamar Sahloul
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - John S Spencer
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO 80523; and
| | - Delphi Chatterjee
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO 80523; and
| | - Tobias Broger
- Foundation for Innovative New Diagnostics, Geneva 1202, Switzerland
| | - Todd L Lowary
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Abraham Pinter
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103;
| |
Collapse
|
89
|
Novel Immunoinformatics Approaches to Design Multi-epitope Subunit Vaccine for Malaria by Investigating Anopheles Salivary Protein. Sci Rep 2018; 8:1125. [PMID: 29348555 PMCID: PMC5773588 DOI: 10.1038/s41598-018-19456-1] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 01/02/2018] [Indexed: 12/19/2022] Open
Abstract
Malaria fever has been pervasive for quite a while in tropical developing regions causing high morbidity and mortality. The causal organism is a protozoan parasite of genus Plasmodium which spreads to the human host by the bite of hitherto infected female Anopheles mosquito. In the course of biting, a salivary protein of Anopheles helps in blood feeding behavior and having the ability to elicit the host immune response. This study represents a series of immunoinformatics approaches to design multi-epitope subunit vaccine using Anopheles mosquito salivary proteins. Designed subunit vaccine was evaluated for its immunogenicity, allergenicity and physiochemical parameters. To enhance the stability of vaccine protein, disulfide engineering was performed in a region of high mobility. Codon adaptation and in silico cloning was also performed to ensure the higher expression of designed subunit vaccine in E. coli K12 expression system. Finally, molecular docking and simulation study was performed for the vaccine protein and TLR-4 receptor, to determine the binding free energy and complex stability. Moreover, the designed subunit vaccine was found to induce anti-salivary immunity which may have the ability to prevent the entry of Plasmodium sporozoites into the human host.
Collapse
|
90
|
Sun ET, Xia D, Li BH, Ma J, Dong YY, Ding SS, Chen BF, Wen YF. Association of Immune Factors with Drug-Resistant Tuberculosis: A Case-Control Study. Med Sci Monit 2017; 23:5330-5336. [PMID: 29118314 PMCID: PMC5691569 DOI: 10.12659/msm.904309] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background Presently, studies of factors associated with drug-resistant tuberculosis (TB) focus on patients’ socio-demographic characteristics and living habits, to the exclusion of biochemical indicators, especially immune factors. This study was carried out to determine whether immune factors are associated with drug-resistant TB. Material/Methods A total of 227 drug-resistant pulmonary TB patients and 225 drug-susceptible pulmonary TB patients were enrolled in this study. Information on socio-demographic characteristics and biochemical indicators were obtained through their clinical records. Non-conditional logistic regression was used to analyze the association of these indicators with drug-resistant TB. Results There were significant differences in re-treatment, marital status, alanine aminotransferase (ALT), blood uric acid (BUA), carcino-embryonic antigen (CEA), T-spot, and CD3 and CD4 counts between the 2 groups. In multivariable analysis, re-treatment [Odds Ratio (OR)=5.290, 95% Confidence Interval [CI]=2.652–10.551); CD3 (OR=1.034, 95% CI=1.001–1.068); CD4 (OR=1.035, 95% CI =1.001–1.070) and IgM (OR=1.845, 95% CI=1.153–2.952) were associated with drug-resistant TB. Conclusions These results suggest the need for greater attention to re-treatment cases and immune function when treating drug-resistant TB.
Collapse
Affiliation(s)
- En-Tao Sun
- School of Public Health, Wannan Medical College, Wuhu, Anhui, China (mainland)
| | - Dan Xia
- School of Public Health, Wannan Medical College, Wuhu, Anhui, China (mainland)
| | - Ben-He Li
- School of Public Health, Wannan Medical College, Wuhu, Anhui, China (mainland)
| | - Jun Ma
- School of Public Health, Wannan Medical College, Wuhu, Anhui, China (mainland)
| | - Yuan-Yuan Dong
- School of Public Health, Wannan Medical College, Wuhu, Anhui, China (mainland)
| | - Shu-Shu Ding
- School of Public Health, Wannan Medical College, Wuhu, Anhui, China (mainland)
| | - Bai-Feng Chen
- School of Public Health, Wannan Medical College, Wuhu, Anhui, China (mainland)
| | - Yu-Feng Wen
- School of Public Health, Wannan Medical College, Wuhu, Anhui, China (mainland)
| |
Collapse
|
91
|
Jang JY, Baek KJ, Choi Y, Ji S. Relatively low invasive capacity of Porphyromonas gingivalis strains into human gingival fibroblasts in vitro. Arch Oral Biol 2017; 83:265-271. [DOI: 10.1016/j.archoralbio.2017.08.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/09/2017] [Accepted: 08/10/2017] [Indexed: 01/28/2023]
|
92
|
Abstract
Mycobacterium tuberculosis is the leading cause of death worldwide from a single bacterial pathogen. The World Health Organization estimates that annually 1 million children have tuberculosis (TB) disease and many more harbor a latent form. Accurate estimates are hindered by under-recognition and challenges in diagnosis. To date, an accurate diagnostic test to confirm TB in children does not exist. Treatment is lengthy but outcomes are generally favorable with timely initiation. With the End TB Strategy, there is an urgent need for improved diagnostics and treatment to prevent the unnecessary morbidity and mortality from TB in children.
Collapse
Affiliation(s)
- Tania A Thomas
- Division of Infectious Diseases and International Health, University of Virginia, PO Box 801340, Charlottesville, VA 22908-1340, USA.
| |
Collapse
|
93
|
Abstract
Immunology is a central theme when it comes to tuberculosis (TB). The outcome of human infection with Mycobacterium tuberculosis is dependent on the ability of the immune response to clear or contain the infection. In cases where this fails, the bacterium replicates, disseminates within the host, and elicits a pathologic inflammatory response, and disease ensues. Clinical presentation of TB disease is remarkably heterogeneous, and the disease phenotype is largely dependent on host immune status. Onward transmission of M. tuberculosis to new susceptible hosts is thought to depend on an excessive inflammatory response causing a breakdown of the lung matrix and formation of lung cavities. But this varies in cases of underlying immunological dysfunction: for example, HIV-1 infection is associated with less cavitation, while diabetes mellitus comorbidity is associated with increased cavitation and risk of transmission. In compliance with the central theme of immunology in tuberculosis, we rely on detection of an adaptive immune response, in the form of interferon-gamma release assays or tuberculin skin tests, to diagnose infection with M. tuberculosis. Here we review the immunology of TB in the human host, focusing on cellular and humoral adaptive immunity as well as key features of innate immune responses and the underlying immunological dysfunction which associates with human TB risk factors. Our review is restricted to human immunology, and we highlight distinctions from the immunological dogma originating from animal models of TB, which pervade the field.
Collapse
|
94
|
Moliva JI, Turner J, Torrelles JB. Immune Responses to Bacillus Calmette-Guérin Vaccination: Why Do They Fail to Protect against Mycobacterium tuberculosis? Front Immunol 2017; 8:407. [PMID: 28424703 PMCID: PMC5380737 DOI: 10.3389/fimmu.2017.00407] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 03/22/2017] [Indexed: 12/11/2022] Open
Abstract
Mycobacterium tuberculosis (M.tb), the causative agent of tuberculosis (TB), is the current leading cause of death due to a single infectious organism. Although curable, the broad emergence of multi-, extensive-, extreme-, and total-drug resistant strains of M.tb has hindered eradication efforts of this pathogen. Furthermore, computational models predict a quarter of the world’s population is infected with M.tb in a latent state, effectively serving as the largest reservoir for any human pathogen with the ability to cause significant morbidity and mortality. The World Health Organization has prioritized new strategies for improved vaccination programs; however, the lack of understanding of mycobacterial immunity has made it difficult to develop new successful vaccines. Currently, Mycobacterium bovis bacillus Calmette–Guérin (BCG) is the only vaccine approved for use to prevent TB. BCG is highly efficacious at preventing meningeal and miliary TB, but is at best 60% effective against the development of pulmonary TB in adults and wanes as we age. In this review, we provide a detailed summary on the innate immune response of macrophages, dendritic cells, and neutrophils in response to BCG vaccination. Additionally, we discuss adaptive immune responses generated by BCG vaccination, emphasizing their specific contributions to mycobacterial immunity. The success of future vaccines against TB will directly depend on our understanding of mycobacterial immunity.
Collapse
Affiliation(s)
- Juan I Moliva
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Joanne Turner
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA.,Center for Microbial Interface Biology, The Ohio State University, Columbus, OH, USA
| | - Jordi B Torrelles
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA.,Center for Microbial Interface Biology, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
95
|
Alvarez N, Serpa D, Kadir R, Tirado Y, Borrero R, Fernández S, Cabrera R, Valdes Y, Zayas C, Acevedo R, Izquierdo L, Sarmiento ME, Norazmi MN, Pérez JL, Acosta A. Specific and cross-reactive immune response against Mycobacterium tuberculosis antigens in mice immunized with proteoliposomes from Mycobacterium bovis BCG. Asian Pac J Trop Biomed 2017. [DOI: 10.1016/j.apjtb.2016.12.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
96
|
Evaluation of the Antibody in Lymphocyte Supernatant Assay to Detect Active Tuberculosis. PLoS One 2017; 12:e0169118. [PMID: 28085899 PMCID: PMC5234774 DOI: 10.1371/journal.pone.0169118] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 12/12/2016] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND We aimed to evaluate the antibody in lymphocyte supernatant (ALS) assay as a biomarker to diagnose tuberculosis among adults from Tanzania with and without HIV. METHODS Adults admitted with suspicion for tuberculosis had sputa obtained for GeneXpert MTB/RIF, acid-fast bacilli smear and mycobacterial culture; blood was obtained prior to treatment initiation and after 4 weeks. Adults hospitalized with non-infectious conditions served as controls. Peripheral blood mononuclear cells were cultured unstimulated for 72 hours. Anti-mycobacterial antibodies were measured from culture supernatants by ELISA, using BCG vaccine as the coating antigen. Median ALS responses were compared between cases and controls at baseline and between cases over time. RESULTS Of 97 TB cases, 85 were microbiologically confirmed and 12 were clinically diagnosed. Median ALS responses from TB cases (0.366 OD from confirmed cases and 0.285 from clinical cases) were higher compared to controls (0.085, p<0.001). ALS responses did not differ based on HIV status, CD4 count or sputum smear status. Over time, the median ALS values declined significantly (0.357 at baseline; 0.198 after 4-weeks, p<0.001). CONCLUSIONS Robust ALS responses were mounted by patients with TB regardless of HIV status, CD4 count, or low sputum bacillary burden, potentially conferring a unique niche for this immunologic biomarker for TB.
Collapse
|
97
|
Linge I, Dyatlov A, Kondratieva E, Avdienko V, Apt A, Kondratieva T. B-lymphocytes forming follicle-like structures in the lung tissue of tuberculosis-infected mice: Dynamics, phenotypes and functional activity. Tuberculosis (Edinb) 2016; 102:16-23. [PMID: 28061947 DOI: 10.1016/j.tube.2016.11.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/09/2016] [Accepted: 11/16/2016] [Indexed: 12/13/2022]
Abstract
During tuberculosis (TB) infection, B cells form follicles in close vicinity of lung granuloma. We assessed the dynamics of follicle formation, surface phenotypes and functional activity of lung B cells during TB course in genetically susceptible mice. The follicles appeared early post infection and peaked at weeks 7-8. Lung B cells resembled classical B2 cells (CD19+IgMloIgDhiCD1d-CD21/35intCD5-CD11b-CD43-), but differed from them by the absence of B2 marker CD23. Lung B-cells constitutively expressed MHC II molecules, presented mycobacterial antigens to immune CD4+ T-cells and produced high amounts of IL-6 and IL-11, but no classical type 1 (TNF-α, IFN-γ), or anti-inflammatory (IL-10, TGF-β) cytokines. The total antibody response in tuberculous lung showed almost no specificity to mycobacteria. A panel of monoclonal antibodies obtained from lung B cells contained only few clones with reactivity to mycobacteria. Our results suggest that anti-TB B cell response in the lung has clear pathological and doubtful protective role.
Collapse
Affiliation(s)
- Irina Linge
- Department of Immunology, Central Institute for Tuberculosis, Moscow, Russia
| | - Alexander Dyatlov
- Department of Immunology, Central Institute for Tuberculosis, Moscow, Russia
| | - Elena Kondratieva
- Department of Immunology, Central Institute for Tuberculosis, Moscow, Russia
| | - Vadim Avdienko
- Department of Immunology, Central Institute for Tuberculosis, Moscow, Russia
| | - Alexander Apt
- Department of Immunology, Central Institute for Tuberculosis, Moscow, Russia; Department of Immunology, School of Biology, Moscow State University, Moscow, Russia.
| | - Tatiana Kondratieva
- Department of Immunology, Central Institute for Tuberculosis, Moscow, Russia
| |
Collapse
|
98
|
TB trifusion antigen adsorbed on calcium phosphate nanoparticles stimulates strong cellular immunity in mice. BIOTECHNOL BIOPROC E 2016. [DOI: 10.1007/s12257-016-0326-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
99
|
CD4+ T-cell-independent mechanisms suppress reactivation of latent tuberculosis in a macaque model of HIV coinfection. Proc Natl Acad Sci U S A 2016; 113:E5636-44. [PMID: 27601645 DOI: 10.1073/pnas.1611987113] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The synergy between Mycobacterium tuberculosis (Mtb) and HIV in coinfected patients has profoundly impacted global mortality because of tuberculosis (TB) and AIDS. HIV significantly increases rates of reactivation of latent TB infection (LTBI) to active disease, with the decline in CD4(+) T cells believed to be the major causality. In this study, nonhuman primates were coinfected with Mtb and simian immunodeficiency virus (SIV), recapitulating human coinfection. A majority of animals exhibited rapid reactivation of Mtb replication, progressing to disseminated TB and increased SIV-associated pathology. Although a severe loss of pulmonary CD4(+) T cells was observed in all coinfected macaques, a subpopulation of the animals was still able to prevent reactivation and maintain LTBI. Investigation of pulmonary immune responses and pathology in this cohort demonstrated that increased CD8(+) memory T-cell proliferation, higher granzyme B production, and expanded B-cell follicles correlated with protection from reactivation. Our findings reveal mechanisms that control SIV- and TB-associated pathology. These CD4-independent protective immune responses warrant further studies in HIV coinfected humans able to control their TB infection. Moreover, these findings will provide insight into natural immunity to Mtb and will guide development of novel vaccine strategies and immunotherapies.
Collapse
|
100
|
Jacobs AJ, Mongkolsapaya J, Screaton GR, McShane H, Wilkinson RJ. Antibodies and tuberculosis. Tuberculosis (Edinb) 2016; 101:102-113. [PMID: 27865379 PMCID: PMC5120988 DOI: 10.1016/j.tube.2016.08.001] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 07/19/2016] [Accepted: 08/04/2016] [Indexed: 12/16/2022]
Abstract
Tuberculosis (TB) remains a major public health problem internationally, causing 9.6 million new cases and 1.5 million deaths worldwide in 2014. The Bacillus Calmette-Guérin vaccine is the only licensed vaccine against TB, but its protective effect does not extend to controlling the development of infectious pulmonary disease in adults. The development of a more effective vaccine against TB is therefore a pressing need for global health. Although it is established that cell-mediated immunity is necessary for the control of latent infection, the presupposition that such immunity is sufficient for vaccine-induced protection has recently been challenged. A greater understanding of protective immunity against TB is required to guide future vaccine strategies against TB. In contrast to cell-mediated immunity, the human antibody response against M.tb is conventionally thought to exert little immune control over the course of infection. Humoral responses are prominent during active TB disease, and have even been postulated to contribute to immunopathology. However, there is evidence to suggest that specific antibodies may limit the dissemination of M.tb, and potentially also play a role in prevention of infection via mucosal immunity. Further, antibodies are now understood to confer protection against a range of intracellular pathogens by modulating immunity via Fc-receptor mediated phagocytosis. In this review, we will explore the evidence that antibody-mediated immunity could be reconsidered in the search for new vaccine strategies against TB.
Collapse
Affiliation(s)
- Ashley J Jacobs
- Department of Medicine, Imperial College London, W2 1PG, United Kingdom; Clinical Infectious Diseases Research Initiative and Department of Medicine, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Observatory 7925, South Africa.
| | | | - Gavin R Screaton
- Department of Medicine, Imperial College London, W2 1PG, United Kingdom
| | - Helen McShane
- The Jenner Institute, University of Oxford, OX3 7DQ, United Kingdom
| | - Robert J Wilkinson
- Department of Medicine, Imperial College London, W2 1PG, United Kingdom; Clinical Infectious Diseases Research Initiative and Department of Medicine, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Observatory 7925, South Africa; The Francis Crick Institute, London NW1 2AT, United Kingdom
| |
Collapse
|