1
|
Peregrino ES, Castañeda-Casimiro J, Vázquez-Flores L, Estrada-Parra S, Wong-Baeza C, Serafín-López J, Wong-Baeza I. The Role of Bacterial Extracellular Vesicles in the Immune Response to Pathogens, and Therapeutic Opportunities. Int J Mol Sci 2024; 25:6210. [PMID: 38892397 PMCID: PMC11172497 DOI: 10.3390/ijms25116210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Pathogenic bacteria have several mechanisms to evade the host's immune response and achieve an efficient infection. Bacterial extracellular vesicles (EVs) are a relevant cellular communication mechanism, since they can interact with other bacterial cells and with host cells. In this review, we focus on the EVs produced by some World Health Organization (WHO) priority Gram-negative and Gram-positive pathogenic bacteria; by spore-producing bacteria; by Mycobacterium tuberculosis (a bacteria with a complex cell wall); and by Treponema pallidum (a bacteria without lipopolysaccharide). We describe the classification and the general properties of bacterial EVs, their role during bacterial infections and their effects on the host immune response. Bacterial EVs contain pathogen-associated molecular patterns that activate innate immune receptors, which leads to cytokine production and inflammation, but they also contain antigens that induce the activation of B and T cell responses. Understanding the many effects of bacterial EVs on the host's immune response can yield new insights on the pathogenesis of clinically important infections, but it can also lead to the development of EV-based diagnostic and therapeutic strategies. In addition, since EVs are efficient activators of both the innate and the adaptive immune responses, they constitute a promising platform for vaccine development.
Collapse
Affiliation(s)
- Eliud S. Peregrino
- Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico; (E.S.P.); (J.C.-C.)
| | - Jessica Castañeda-Casimiro
- Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico; (E.S.P.); (J.C.-C.)
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico; (S.E.-P.); (J.S.-L.)
| | - Luis Vázquez-Flores
- Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico; (L.V.-F.); (C.W.-B.)
| | - Sergio Estrada-Parra
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico; (S.E.-P.); (J.S.-L.)
| | - Carlos Wong-Baeza
- Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico; (L.V.-F.); (C.W.-B.)
| | - Jeanet Serafín-López
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico; (S.E.-P.); (J.S.-L.)
| | - Isabel Wong-Baeza
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico; (S.E.-P.); (J.S.-L.)
| |
Collapse
|
2
|
Zhao L, Fan K, Sun X, Li W, Qin F, Shi L, Gao F, Zheng C. Host-directed therapy against mycobacterium tuberculosis infections with diabetes mellitus. Front Immunol 2024; 14:1305325. [PMID: 38259491 PMCID: PMC10800548 DOI: 10.3389/fimmu.2023.1305325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Tuberculosis (TB) is caused by the bacterial pathogen Mycobacterium tuberculosis (MTB) and is one of the principal reasons for mortality and morbidity worldwide. Currently, recommended anti-tuberculosis drugs include isoniazid, rifampicin, ethambutol, and pyrazinamide. TB treatment is lengthy and inflicted with severe side-effects, including reduced patient compliance with treatment and promotion of drug-resistant strains. TB is also prone to other concomitant diseases such as diabetes and HIV. These drug-resistant and complex co-morbid characteristics increase the complexity of treating MTB. Host-directed therapy (HDT), which effectively eliminates MTB and minimizes inflammatory tissue damage, primarily by targeting the immune system, is currently an attractive complementary approach. The drugs used for HDT are repositioned drugs in actual clinical practice with relative safety and efficacy assurance. HDT is a potentially effective therapeutic intervention for the treatment of MTB and diabetic MTB, and can compensate for the shortcomings of current TB therapies, including the reduction of drug resistance and modulation of immune response. Here, we summarize the state-of-the-art roles and mechanisms of HDT in immune modulation and treatment of MTB, with a special focus on the role of HDT in diabetic MTB, to emphasize the potential of HDT in controlling MTB infection.
Collapse
Affiliation(s)
- Li Zhao
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Ke Fan
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Xuezhi Sun
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Wei Li
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Fenfen Qin
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Liwen Shi
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Feng Gao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chunlan Zheng
- Department of Tuberculosis III, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| |
Collapse
|
3
|
Kim GB, Kim S, Hwang YH, Kim S, Lee I, Kim SA, Goo J, Yang Y, Jeong C, Nam GH, Kim IS. Harnessing Oncolytic Extracellular Vesicles for Tumor Cell-Preferential Cytoplasmic Delivery of Misfolded Proteins for Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2300527. [PMID: 37226374 DOI: 10.1002/smll.202300527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 05/12/2023] [Indexed: 05/26/2023]
Abstract
In this study, extracellular vesicles (EVs) are reimagined as more than just a cellular waste disposal system and are repurposed for cancer immunotherapy. Potent oncolytic EVs (bRSVF-EVs) loaded with misfolded proteins (MPs) are engineered, which are typically considered cellular debris. By impairing lysosomal function using bafilomycin A1 and expressing the respiratory syncytial virus F protein, a viral fusogen, MPs are successfully loaded into the EVs expressing RSVF. bRSVF-EVs preferentially transplant a xenogeneic antigen onto cancer cell membranes in a nucleolin-dependent manner, triggering an innate immune response. Furthermore, bRSVF-EV-mediated direct delivery of MPs into the cancer cell cytoplasm initiates endoplasmic reticulum stress and immunogenic cell death (ICD). This mechanism of action leads to substantial antitumor immune responses in murine tumor models. Importantly, when combined with PD-1 blockade, bRSVF-EV treatment elicits robust antitumor immunity, resulting in prolonged survival and complete remission in some cases. Overall, the findings demonstrate that utilizing tumor-targeting oncolytic EVs for direct cytoplasmic delivery of MPs to induce ICD in cancer cells represents a promising approach for enhancing durable antitumor immunity.
Collapse
Affiliation(s)
- Gi Beom Kim
- SHIFTBIO.INC, Seoul, 02751, Republic of Korea
| | | | - Yeong Ha Hwang
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Seohyun Kim
- SHIFTBIO.INC, Seoul, 02751, Republic of Korea
| | - Inkyu Lee
- SHIFTBIO.INC, Seoul, 02751, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Seong A Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jiyoung Goo
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KHU-KIST Department of Converging Science and Technology, Kyunghee University, Seoul, 02447, Republic of Korea
| | - Yoosoo Yang
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Cherlhyun Jeong
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KHU-KIST Department of Converging Science and Technology, Kyunghee University, Seoul, 02447, Republic of Korea
| | - Gi-Hoon Nam
- SHIFTBIO.INC, Seoul, 02751, Republic of Korea
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - In-San Kim
- SHIFTBIO.INC, Seoul, 02751, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| |
Collapse
|
4
|
Diatlova A, Linkova N, Lavrova A, Zinchenko Y, Medvedev D, Krasichkov A, Polyakova V, Yablonskiy P. Molecular Markers of Early Immune Response in Tuberculosis: Prospects of Application in Predictive Medicine. Int J Mol Sci 2023; 24:13261. [PMID: 37686061 PMCID: PMC10487556 DOI: 10.3390/ijms241713261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Tuberculosis (TB) remains an important public health problem and one of the leading causes of death. Individuals with latent tuberculosis infection (LTBI) have an increased risk of developing active TB. The problem of the diagnosis of the various stages of TB and the identification of infected patients in the early stages has not yet been solved. The existing tests (the tuberculin skin test and the interferon-gamma release assay) are useful to distinguish between active and latent infections. But these tests cannot be used to predict the development of active TB in individuals with LTBI. The purpose of this review was to analyze the extant data of the interaction of M. tuberculosis with immune cells and identify molecular predictive markers and markers of the early stages of TB. An analysis of more than 90 sources from the literature allowed us to determine various subpopulations of immune cells involved in the pathogenesis of TB, namely, macrophages, dendritic cells, B lymphocytes, T helper cells, cytotoxic T lymphocytes, and NK cells. The key molecular markers of the immune response to M. tuberculosis are cytokines (IL-1β, IL-6, IL-8, IL-10, IL-12, IL-17, IL-22b, IFNɣ, TNFa, and TGFß), matrix metalloproteinases (MMP-1, MMP-3, and MMP-9), and their inhibitors (TIMP-1, TIMP-2, TIMP-3, and TIMP-4). It is supposed that these molecules could be used as biomarkers to characterize different stages of TB infection, to evaluate the effectiveness of its treatment, and as targets of pharmacotherapy.
Collapse
Affiliation(s)
- Anastasiia Diatlova
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2–4, 191036 St. Petersburg, Russia
| | - Natalia Linkova
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2–4, 191036 St. Petersburg, Russia
- Biogerontology Department, St. Petersburg Institute of Bioregulation and Gerontology, Dynamo pr., 3, 197110 St. Petersburg, Russia
| | - Anastasia Lavrova
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2–4, 191036 St. Petersburg, Russia
- Department of Hospital Surgery, Faculty of Medicine, St. Petersburg State University, University Embankment, 7–9, 199034 St. Petersburg, Russia
| | - Yulia Zinchenko
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2–4, 191036 St. Petersburg, Russia
| | - Dmitrii Medvedev
- Biogerontology Department, St. Petersburg Institute of Bioregulation and Gerontology, Dynamo pr., 3, 197110 St. Petersburg, Russia
| | - Alexandr Krasichkov
- Department of Radio Engineering Systems, Electrotechnical University “LETI”, Prof. Popova Street 5F, 197022 St. Petersburg, Russia
| | - Victoria Polyakova
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2–4, 191036 St. Petersburg, Russia
| | - Piotr Yablonskiy
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2–4, 191036 St. Petersburg, Russia
- Department of Hospital Surgery, Faculty of Medicine, St. Petersburg State University, University Embankment, 7–9, 199034 St. Petersburg, Russia
| |
Collapse
|
5
|
Kayongo A, Nyiro B, Siddharthan T, Kirenga B, Checkley W, Lutaakome Joloba M, Ellner J, Salgame P. Mechanisms of lung damage in tuberculosis: implications for chronic obstructive pulmonary disease. Front Cell Infect Microbiol 2023; 13:1146571. [PMID: 37415827 PMCID: PMC10320222 DOI: 10.3389/fcimb.2023.1146571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 06/05/2023] [Indexed: 07/08/2023] Open
Abstract
Pulmonary tuberculosis is increasingly recognized as a risk factor for COPD. Severe lung function impairment has been reported in post-TB patients. Despite increasing evidence to support the association between TB and COPD, only a few studies describe the immunological basis of COPD among TB patients following successful treatment completion. In this review, we draw on well-elaborated Mycobacterium tuberculosis-induced immune mechanisms in the lungs to highlight shared mechanisms for COPD pathogenesis in the setting of tuberculosis disease. We further examine how such mechanisms could be exploited to guide COPD therapeutics.
Collapse
Affiliation(s)
- Alex Kayongo
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
- Makerere University College of Health Sciences, Lung Institute, Makerere University, Kampala, Uganda
| | - Brian Nyiro
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Trishul Siddharthan
- Division of Pulmonary and Critical Care Medicine, University of Miami, Miami, FL, United States
| | - Bruce Kirenga
- Makerere University College of Health Sciences, Lung Institute, Makerere University, Kampala, Uganda
| | - William Checkley
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, United States
- Center for Global Non-Communicable Disease Research and Training, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Moses Lutaakome Joloba
- Makerere University College of Health Sciences, Lung Institute, Makerere University, Kampala, Uganda
| | - Jerrold Ellner
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Padmini Salgame
- Department of Medicine, Center for Emerging Pathogens, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
6
|
Brito C, Lourenço C, Magalhães J, Reis S, Borges M. Nanoparticles as a Delivery System of Antigens for the Development of an Effective Vaccine against Toxoplasma gondii. Vaccines (Basel) 2023; 11:vaccines11040733. [PMID: 37112645 PMCID: PMC10142924 DOI: 10.3390/vaccines11040733] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/10/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Nanoparticles include particles ranging in size from nanometers to micrometers, whose physicochemical characteristics are optimized to make them appropriate delivery vehicles for drugs or immunogens important in the fight and/or prevention of infectious diseases. There has been a rise in the use of nanoparticles in preventive vaccine formulations as immunostimulatory adjuvants, and as vehicles for immunogen delivery to target immune cells. Toxoplasma is important worldwide, and may cause human toxoplasmosis. In immunocompetent hosts, infection is usually asymptomatic, but in immunocompromised patients it can cause serious neurological and ocular consequences, such as encephalitis and retinochoroiditis. Primary infection during pregnancy may cause abortion or congenital toxoplasmosis. Currently, there is no effective human vaccine against this disease. Evidence has emerged from several experimental studies testing nanovaccines showing them to be promising tools in the prevention of experimental toxoplasmosis. For the present study, a literature review was carried out on articles published over the last 10 years through the PubMed database, pertaining to in vivo experimental models of T. gondii infection where nanovaccines were tested and protection and immune responses evaluated. This review aims to highlight the way forward in the search for an effective vaccine for toxoplasmosis.
Collapse
|
7
|
Meng C, Liu J, Kang X, Xu Z, Xu S, Li X, Pan Z, Chen X, Jiao X. Discrepancy in Response of Mouse Dendritic Cells against BCG: Weak Immune Effects of Plasmacytoid Dendritic Cells Compared to Classical Dendritic Cells despite the Uptake of Bacilli. Trop Med Infect Dis 2023; 8:tropicalmed8030140. [PMID: 36977141 PMCID: PMC10057906 DOI: 10.3390/tropicalmed8030140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/25/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023] Open
Abstract
Tuberculosis (TB), a zoonosis characterized by chronic respiratory infections, is mainly caused by Mycobacterium tuberculosis and is associated with one of the heaviest disease burdens in the world. Dendritic cells (DCs) play a key role and act as a bridge between innate and adaptive immune responses against TB. DCs are divided into distinct subsets. Currently, the response of DCs to mycobacterial infections is poorly understood. Herein, we aimed to evaluate the responses of splenic conventional DCs (cDC) and plasmacytoid DCs (pDC), subsets to Bacillus Calmette–Guérin (BCG) infection in mice. Splenic pDC had a significantly higher infection rate and intracellular bacterial count than cDC and the CD8+ and CD8− cDC subsets after BCG infection. However, the expression levels of CD40, CD80, CD86, and MHC-II molecules were significantly upregulated in splenic cDC and the CD8 cDC subsets compared to pDC during BCG infection. Splenic cDC had a higher expression of IFN-γ and IL-12p70 than pDC, whereas pDC had higher levels of TNF-α and MCP-1 than cDC in mice infected with BCG. At early stages of immunization with BCG containing the Ag85A protein, splenic cDC and pDC could present the Ag85A peptide to a specific T hybridoma; however, cDC had a stronger antigen presenting activity than pDC. In summary, splenic cDC and pDC extensively participate in mouse immune responses against BCG infection in vivo. Although pDC had a higher BCG uptake, cDC induced stronger immunological effects, including activation and maturation, cytokine production, and antigen presentation.
Collapse
Affiliation(s)
- Chuang Meng
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Jun Liu
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
| | - Xilong Kang
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Zhengzhong Xu
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Shuangyuan Xu
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
| | - Xin Li
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Zhiming Pan
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Xiang Chen
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Correspondence: (X.C.); (X.J.)
| | - Xinan Jiao
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Correspondence: (X.C.); (X.J.)
| |
Collapse
|
8
|
Larsen SE, Williams BD, Rais M, Coler RN, Baldwin SL. It Takes a Village: The Multifaceted Immune Response to Mycobacterium tuberculosis Infection and Vaccine-Induced Immunity. Front Immunol 2022; 13:840225. [PMID: 35359957 PMCID: PMC8960931 DOI: 10.3389/fimmu.2022.840225] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/08/2022] [Indexed: 11/18/2022] Open
Abstract
Despite co-evolving with humans for centuries and being intensely studied for decades, the immune correlates of protection against Mycobacterium tuberculosis (Mtb) have yet to be fully defined. This lapse in understanding is a major lag in the pipeline for evaluating and advancing efficacious vaccine candidates. While CD4+ T helper 1 (TH1) pro-inflammatory responses have a significant role in controlling Mtb infection, the historically narrow focus on this cell population may have eclipsed the characterization of other requisite arms of the immune system. Over the last decade, the tuberculosis (TB) research community has intentionally and intensely increased the breadth of investigation of other immune players. Here, we review mechanistic preclinical studies as well as clinical anecdotes that suggest the degree to which different cell types, such as NK cells, CD8+ T cells, γ δ T cells, and B cells, influence infection or disease prevention. Additionally, we categorically outline the observed role each major cell type plays in vaccine-induced immunity, including Mycobacterium bovis bacillus Calmette-Guérin (BCG). Novel vaccine candidates advancing through either the preclinical or clinical pipeline leverage different platforms (e.g., protein + adjuvant, vector-based, nucleic acid-based) to purposefully elicit complex immune responses, and we review those design rationales and results to date. The better we as a community understand the essential composition, magnitude, timing, and trafficking of immune responses against Mtb, the closer we are to reducing the severe disease burden and toll on human health inflicted by TB globally.
Collapse
Affiliation(s)
- Sasha E. Larsen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Brittany D. Williams
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,Department of Global Health, University of Washington, Seattle, WA, United States
| | - Maham Rais
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Rhea N. Coler
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,Department of Global Health, University of Washington, Seattle, WA, United States,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| | - Susan L. Baldwin
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,*Correspondence: Susan L. Baldwin,
| |
Collapse
|
9
|
Zhu W, Lv J, Xie X, Tian C, Liu J, Zhou H, Sun C, Li J, Hu Z, Li X. The oncolytic virus VT09X optimizes immune checkpoint therapy in low immunogenic melanoma. Immunol Lett 2021; 241:15-22. [PMID: 34774916 DOI: 10.1016/j.imlet.2021.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 10/26/2021] [Accepted: 11/09/2021] [Indexed: 12/14/2022]
Abstract
Tumors with a low level of pre-existing immune cell infiltration respond poorly to immune checkpoint therapies. Oncolytic viruses optimize immunotherapies by modulating the tumor microenvironment and affecting multiple steps in the cancer-immunity cycle, making them an attractive agent for combination strategies. We engineered an HSV-1-based oncolytic virus and investigated its antitumor effects in combination with the marketed PD-1 antibody Keytruda (pembrolizumab) in hPD-1 knock-in mice bearing non-immunogenic B16-F10 melanoma. Our results showed enhanced CD8+ and CD4+ T cell infiltration, IFN-γ secretion and PD-L1 expression in tumors, subsequently leading to the prolonged overall survival of mice. Systemic changes in lymphocyte cell proportions were also observed in the peripheral blood. In summary, these findings provide evidence that oncolytic viruses can be engineered as a potential platform for combination therapies, especially to treat tumors that are poorly responsive to immune checkpoint therapy.
Collapse
Affiliation(s)
- Wei Zhu
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China
| | - Jingwen Lv
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China
| | - Xin Xie
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China
| | - Chao Tian
- Beijing WellGene Company, Ltd, Beijing 100085, China
| | - Jiajia Liu
- Beijing WellGene Company, Ltd, Beijing 100085, China
| | - Hua Zhou
- Beijing WellGene Company, Ltd, Beijing 100085, China
| | - Chunyang Sun
- Beijing WellGene Company, Ltd, Beijing 100085, China
| | - Jingfeng Li
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China; Beijing WellGene Company, Ltd, Beijing 100085, China
| | - Zongfeng Hu
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China
| | - Xiaopeng Li
- School of Pharmacy, Yantai University, Yantai 264005, Shandong, China; Beijing WellGene Company, Ltd, Beijing 100085, China.
| |
Collapse
|
10
|
Using oncolytic viruses to ignite the tumour immune microenvironment in bladder cancer. Nat Rev Urol 2021; 18:543-555. [PMID: 34183833 DOI: 10.1038/s41585-021-00483-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2021] [Indexed: 02/06/2023]
Abstract
The advent of immune checkpoint inhibition (ICI) has transformed the treatment paradigm for bladder cancer. However, despite the success of ICI in other tumour types, the majority of ICI-treated patients with bladder cancer failed to respond. The lack of efficacy in some patients could be attributed to a paucity of pre-existing immune reactive cells within the tumour immune microenvironment, which limits the beneficial effects of ICI. In this setting, strategies to attract lymphocytes before implementation of ICI could be helpful. Oncolytic virotherapy is thought to induce the release of damage-associated molecular patterns, eliciting a pro-inflammatory cytokine cascade and stimulating the activation of the innate immune system. Concurrently, oncolytic virotherapy-induced oncolysis leads to further release of neoantigens and subsequent epitope spreading, culminating in a robust, tumour-specific adaptive immune response. Combination therapy using oncolytic virotherapy with ICI has proven successful in a number of preclinical studies and is beginning to enter clinical trials for the treatment of both non-muscle-invasive and muscle-invasive bladder cancer. In this context, understanding of the mechanisms underpinning oncolytic virotherapy and its potential synergism with ICI will enable clinicians to effectively deploy oncolytic virotherapy, either as monotherapy or as combination therapy in the different clinical stages of bladder cancer.
Collapse
|
11
|
Lin SC, Lin CC, Li S, Lin WY, Lehman CW, Bracci NR, Tsai SW. Alleviation of Collagen-Induced Arthritis by Crotonoside through Modulation of Dendritic Cell Differentiation and Activation. PLANTS 2020; 9:plants9111535. [PMID: 33182776 PMCID: PMC7698099 DOI: 10.3390/plants9111535] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/09/2020] [Accepted: 11/09/2020] [Indexed: 12/22/2022]
Abstract
Crotonoside, a guanosine analog originally isolated from Croton tiglium, is reported to be a potent tyrosine kinase inhibitor with immunosuppressive effects on immune cells. Due to its potential immunotherapeutic effects, we aimed to evaluate the anti-arthritic activity of crotonoside and explore its immunomodulatory properties in alleviating the severity of arthritic symptoms. To this end, we implemented the treatment of crotonoside on collagen-induced arthritic (CIA) DBA/1 mice and investigated its underlying mechanisms towards pathogenic dendritic cells (DCs). Our results suggest that crotonoside treatment remarkably improved clinical arthritic symptoms in this CIA mouse model as indicated by decreased pro-inflammatory cytokine production in the serum and suppressed expression of co-stimulatory molecules, CD40, CD80, and MHC class II, on CD11c+ DCs from the CIA mouse spleens. Additionally, crotonoside treatment significantly reduced the infiltration of CD11c+ DCs into the synovial tissues. Our in vitro study further demonstrated that bone marrow-derived DCs (BMDCs) exhibited lower yield in numbers and expressed lower levels of CD40, CD80, and MHC-II when incubated with crotonoside. Furthermore, LPS-stimulated mature DCs exhibited limited capability to prime antigen-specific CD4+ and T-cell proliferation, cytokine secretions, and co-stimulatory molecule expressions when treated with crotonoside. Our pioneer study highlights the immunotherapeutic role of crotonoside in the alleviation of the CIA via modulation of pathogenic DCs, thus creating possible applications of crotonoside as an immunosuppressive agent that could be utilized and further explored in treating autoimmune disorders in the future.
Collapse
Affiliation(s)
- Shih-Chao Lin
- Bachelor Degree Program in Marine Biotechnology, National Taiwan Ocean University, Keelung 202301, Taiwan;
| | - Chi-Chien Lin
- Institute of Biomedical Science, The iEGG and Animal Biotechnology Center, National Chung-Hsing University, Taichung 402204, Taiwan; (C.-C.L.); (W.-Y.L.)
- Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40705, Taiwan
- Department of Pharmacology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Shiming Li
- Hubei Key Laboratory for Processing and Application of Catalytic Materials, College of Chemistry and Chemical Engineering, Huanggang Normal University, Hubei 438000, China;
| | - Wan-Yi Lin
- Institute of Biomedical Science, The iEGG and Animal Biotechnology Center, National Chung-Hsing University, Taichung 402204, Taiwan; (C.-C.L.); (W.-Y.L.)
| | - Caitlin W. Lehman
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24063, USA; (C.W.L.); (N.R.B.)
| | - Nicole R. Bracci
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24063, USA; (C.W.L.); (N.R.B.)
| | - Sen-Wei Tsai
- Department of Physical Medicine and Rehabilitation, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 427, Taiwan
- Department of Physical Medicine and Rehabilitation, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
- Correspondence:
| |
Collapse
|
12
|
Wu H, Xu Z, Wang Z, Ren Z, Li L, Ruan Y. Dendritic cells with METTL3 gene knockdown exhibit immature properties and prolong allograft survival. Genes Immun 2020; 21:193-202. [PMID: 32457372 DOI: 10.1038/s41435-020-0099-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 04/12/2020] [Accepted: 04/15/2020] [Indexed: 12/18/2022]
Abstract
Maturation of dendritic cells (DCs) initiates adaptive immune responses and thereby provokes allograft rejection. Here, this study aimed to explore the effect of Methyltransferase-like protein 3 (METTL3) silencing on DC function and the role of METTL3-silencing donor DCs in the immune response after mouse heart transplantation. Bone marrow-derived DCs from donor BALB/c mice were infected with lentiviruses expressing METTL3-specific short hairpin RNA (LV-METTL3 shRNA) to silence METTL3. Then METTL3-silencing DCs were treated with lipopolysaccharide (LPS) for another 48 h to induce DC maturation. Recipient C57BL/6 mice were injected with phosphate-buffered saline (PBS), immature DCs, and METTL3 shRNA-DCs prior to the cardiac transplantation involving the transfer of hearts from donor BALB/c mice to recipient C57BL/6 mice. In vitro we demonstrated that METTL3-silencing DCs had lower expression of MHCII, costimulatory molecules (CD80, CD86), and DC-related cytokines (IFN-γ, IL-12) as well as lower ability to activate T-cell proliferation, which were consistent with the characteristics of tolerogenic DCs. In vivo we found that METTL3-silencing donor DCs induced immune tolerance after mouse heart transplantation and prolonged the allograft survival, which might be associated with Th1/Th2 immune deviation. In summary, METTL3-silencing DCs exhibit immature properties and prolong allograft survival.
Collapse
Affiliation(s)
- Hongbing Wu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University Hubei Cardiovascular Medicine Clinical Research Center Hubei Key Laboratory of Cardiology, 430060, Wuhan, Hubei, China
| | - Zhaojia Xu
- Department of Critical Care Medicine, Jin Yin-tan Hospital, Wuhan, Hubei, China
| | - Zhiwei Wang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University Hubei Cardiovascular Medicine Clinical Research Center Hubei Key Laboratory of Cardiology, 430060, Wuhan, Hubei, China.
| | - Zongli Ren
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University Hubei Cardiovascular Medicine Clinical Research Center Hubei Key Laboratory of Cardiology, 430060, Wuhan, Hubei, China
| | - Luocheng Li
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University Hubei Cardiovascular Medicine Clinical Research Center Hubei Key Laboratory of Cardiology, 430060, Wuhan, Hubei, China
| | - Yongle Ruan
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University Hubei Cardiovascular Medicine Clinical Research Center Hubei Key Laboratory of Cardiology, 430060, Wuhan, Hubei, China
| |
Collapse
|
13
|
Cai X, Ge N, Rong R, Lu Y, Zhang J, Xu J. High expression of BTLA and B7-H4 on the surface of myeloid dendritic cells has a negative regulatory effect on their anti-tuberculosis immunity activity in pleural tuberculosis patients. Tuberculosis (Edinb) 2019; 119:101877. [PMID: 31677529 DOI: 10.1016/j.tube.2019.101877] [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: 03/26/2019] [Revised: 09/19/2019] [Accepted: 10/09/2019] [Indexed: 10/25/2022]
Abstract
To investigate the effects of the surface markers B- and T-lymphocyte attenuator (BTLA) and B7 homologous body 4 (B7-H4) on expression of CD83, and Human Leukocyte Antigen-DR isotype (HLA-DR) that can activate dendritic cells (DCs). Flow cytometry was used to detect the co-expression of BTLA and B7-H4 on myeloid DCs (mDCs) in peripheral blood (PB) and pleural effusions (PE) in 15 volunteers and 20 tuberculous pleurisy (TP) patients. Co-expression of BTLA and B7-H4 (double positive (DP)) mDCs in PB and PE of TP patients were enhanced. The proportion of DP mDC in PB decreased markedly after 2 weeks treatment, but was still greater than in controls. Expression of CD83 and HLA-DR on DP mDCs was higher than on BTLA and B7-H4 double negative (DN) expressing mDCs in PB of different TP groups. Expression of CD83 on DP mDCs in PB and PE of TP patients was greater than that of controls. Expression of HLA-DR on DP mDCs in TP patient PB was lower than in TP PE and controls. In pleural tuberculosis (TB) patients, high expression of BTLA and B7-H4 promoted a high level of CD83 and HLA-DR, which had a negative regulatory effect on mDCs on anti-TB immunity.
Collapse
Affiliation(s)
- Xiaozhen Cai
- Department of Respiratory Medicine, Dongguan Houjie Hospital Affiliated to Guangdong Medical University, Dongguan, 523945, China.
| | - Nanhai Ge
- Department of Respiratory Medicine, Dongguan Houjie Hospital Affiliated to Guangdong Medical University, Dongguan, 523945, China
| | - Rong Rong
- Department of Respiratory Medicine, Dongguan Houjie Hospital Affiliated to Guangdong Medical University, Dongguan, 523945, China
| | - Yuanbin Lu
- Institute of Laboratory Medicine, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Junai Zhang
- Institute of Laboratory Medicine, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Junfa Xu
- Institute of Laboratory Medicine, Guangdong Medical University, Dongguan, 523808, Guangdong, China.
| |
Collapse
|
14
|
Platelets Enhance Dendritic Cell Responses against Staphylococcus aureus through CD40-CD40L. Infect Immun 2018; 86:IAI.00186-18. [PMID: 29914928 DOI: 10.1128/iai.00186-18] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/12/2018] [Indexed: 12/24/2022] Open
Abstract
Staphylococcus aureus is a major human pathogen that can cause mild to severe life-threatening infections in many tissues and organs. Platelets are known to participate in protection against S. aureus by direct killing and by enhancing the activities of neutrophils and macrophages in clearing S. aureus infection. Platelets have also been shown to induce monocyte differentiation into dendritic cells and to enhance activation of dendritic cells. Therefore, in the present study, we explored the role of platelets in enhancing bone marrow-derived dendritic cell (BMDC) function against S. aureus We observed a significant increase in dendritic cell phagocytosis and intracellular killing of a methicillin-resistant Staphylococcus aureus (MRSA) strain (USA300) by thrombin-activated platelets or their releasates. Enhancement of bacterial uptake and killing by DCs is mediated by platelet-derived CD40L. Coculture of USA300 and BMDCs in the presence of thrombin-activated platelet releasates invokes upregulation of the maturation marker CD80 on DCs and enhanced production of the proinflammatory cytokines tumor necrosis factor alpha (TNF-α), interleukin 12 (IL-12), and IL-6. Overall, these observations support our hypothesis that platelets play a critical role in the host defense against S. aureus infection. Platelets stimulate DCs, leading to direct killing of S. aureus and enhanced DC maturation, potentially leading to adaptive immune responses against S. aureus.
Collapse
|
15
|
|
16
|
Allen RP, Bolandparvaz A, Ma JA, Manickam VA, Lewis JS. Latent, Immunosuppressive Nature of Poly(lactic- co-glycolic acid) Microparticles. ACS Biomater Sci Eng 2018; 4:900-918. [PMID: 30555893 PMCID: PMC6290919 DOI: 10.1021/acsbiomaterials.7b00831] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Use of biomaterials to spatiotemporally control the activation of immune cells is at the forefront of biomedical engineering research. As more biomaterial strategies are employed for immunomodulation, understanding the immunogenicity of biodegradable materials and their byproducts is paramount in tailoring systems for immune activation or suppression. Poly(D,L-lactic-co-glycolic acid) (PLGA), one of the most commonly studied polymers in tissue engineering and drug delivery, has been previously described on one hand as an immune adjuvant, and on the other as a nonactivating material. In this study, the effect of PLGA microparticles (MPs) on the maturation status of murine bone marrow-derived dendritic cells (DCs), the primary initiators of adaptive immunity, was investigated to decipher the immunomodulatory properties of this biomaterial. Treatment of bone marrow-derived DCs from C57BL/6 mice with PLGA MPs led to a time dependent decrease in the maturation level of these cells, as quantified by decreased expression of the positive stimulatory molecules MHCII, CD80, and CD86 as well as the ability to resist maturation following challenge with lipopolysaccharide (LPS). Moreover, this immunosuppression was dependent on the molecular weight of the PLGA used to fabricate the MPs, as higher molecular weight polymers required longer incubation to produce comparable dampening of maturation molecules. These phenomena were correlated to an increase in lactic acid both intracellularly and extracellularly during DC/PLGA MP coculture, which is postulated to be the primary agent behind the observed immune inhibition. This hypothesis is supported by our results demonstrating that resistance to LPS stimulation may be due to the ability of PLGA MP-derived lactic acid to inhibit the phosphorylation of TAK1 and therefore prevent NF-κB activation. This work is significant as it begins to elucidate how PLGA, a prominent biomaterial with broad applications ranging from tissue engineering to pharmaceutics, could modulate the local immune environment and offers insight on engineering PLGA to exploit its evolving immunogenicity.
Collapse
Affiliation(s)
- Riley P. Allen
- Department of Biomedical Engineering, University of California Davis, 1 Shields Avenue, Davis, California 95616, United States
| | - Amir Bolandparvaz
- Department of Biomedical Engineering, University of California Davis, 1 Shields Avenue, Davis, California 95616, United States
| | - Jeffrey A. Ma
- Department of Biomedical Engineering, University of California Davis, 1 Shields Avenue, Davis, California 95616, United States
| | - Vishal A. Manickam
- Department of Biomedical Engineering, University of California Davis, 1 Shields Avenue, Davis, California 95616, United States
| | - Jamal S. Lewis
- Department of Biomedical Engineering, University of California Davis, 1 Shields Avenue, Davis, California 95616, United States
| |
Collapse
|
17
|
Lu YB, Xiao DQ, Liang KD, Zhang JA, Wang WD, Yu SY, Zheng BY, Gao YC, Dai YC, Jia Y, Chen C, Zhuang ZG, Wang X, Fu XX, Zhou Y, Zhong J, Chen ZW, Xu JF. Profiling dendritic cell subsets in the patients with active pulmonary tuberculosis. Mol Immunol 2017; 91:86-96. [PMID: 28889065 DOI: 10.1016/j.molimm.2017.08.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/30/2017] [Accepted: 08/14/2017] [Indexed: 02/07/2023]
Abstract
Dendritic cell (DC) plays an important role in the immune response against pulmonary tuberculosis. However, the phenotypic profile of DC subsets in peripheral blood in individuals with active pulmonary tuberculosis (APT) is still inconclusive. Here, we demonstrated that the absolute numbers of total DC (tDC), myeloid DC (mDC) and plasmacytoid DC (pDC) in individuals with APT were decreased compared to healthy controls (HCs). The decreased number of DCs, especially of pDC, seems to be a useful diagnostic marker of APT. Meanwhile, the number of DCs was associated with the prolonged/complicated TB, ATD treatment effect and lymphocyte immune reactions, as manifested that relapsed APT patients with a higher number of tDC and lower number of pDC compared to newly diagnosed patients. Interestingly, mDC from APT patients displayed high expressions of CD83 and CCR7, but pDC displayed low expressions of CD83 and CCR7. Moreover, DCs from APT patients expressed lower levels of HLA-DR and CD80, but expressed a higher level of CD86 than those from HCs. However, the antigen uptake capacity of DC subsets was not different between APT and HCs, despite the antigen uptake capacity of pDC was much lower than that of mDC in both APT patients and HCs. Our data represent a systematic profile of DC subsets in the blood of APT patients, and would represent a useful biomarker for APT.
Collapse
Affiliation(s)
- Yuan-Bin Lu
- Institute of Laboratory Medicine, Guangdong Medical University, No. 1 Xincheng Road, Dongguan 523808, China; Department of Laboratory Medicine, Dongguan 5th Hospital, Dongguan 523000, China
| | - De-Qian Xiao
- Institute of Laboratory Medicine, Guangdong Medical University, No. 1 Xincheng Road, Dongguan 523808, China
| | - Kui-Di Liang
- Department of Respiration, Dongguan 6th Hospital, Dongguan 523000, China
| | - Jun-Ai Zhang
- Institute of Laboratory Medicine, Guangdong Medical University, No. 1 Xincheng Road, Dongguan 523808, China; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, No. 1 Xincheng Road, Dongguan 523808, China
| | - Wan-Dang Wang
- Institute of Laboratory Medicine, Guangdong Medical University, No. 1 Xincheng Road, Dongguan 523808, China; Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL, USA
| | - Shi-Yan Yu
- Institute of Laboratory Medicine, Guangdong Medical University, No. 1 Xincheng Road, Dongguan 523808, China; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, No. 1 Xincheng Road, Dongguan 523808, China
| | - Bi-Ying Zheng
- Institute of Laboratory Medicine, Guangdong Medical University, No. 1 Xincheng Road, Dongguan 523808, China
| | - Yu-Chi Gao
- Institute of Laboratory Medicine, Guangdong Medical University, No. 1 Xincheng Road, Dongguan 523808, China; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, No. 1 Xincheng Road, Dongguan 523808, China
| | - You-Chao Dai
- Institute of Laboratory Medicine, Guangdong Medical University, No. 1 Xincheng Road, Dongguan 523808, China; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, No. 1 Xincheng Road, Dongguan 523808, China
| | - Yan Jia
- Institute of Laboratory Medicine, Guangdong Medical University, No. 1 Xincheng Road, Dongguan 523808, China; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, No. 1 Xincheng Road, Dongguan 523808, China
| | - Chen Chen
- Institute of Laboratory Medicine, Guangdong Medical University, No. 1 Xincheng Road, Dongguan 523808, China; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, No. 1 Xincheng Road, Dongguan 523808, China
| | - Ze-Gang Zhuang
- Institute of Laboratory Medicine, Guangdong Medical University, No. 1 Xincheng Road, Dongguan 523808, China; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, No. 1 Xincheng Road, Dongguan 523808, China
| | - Xin Wang
- Institute of Laboratory Medicine, Guangdong Medical University, No. 1 Xincheng Road, Dongguan 523808, China; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, No. 1 Xincheng Road, Dongguan 523808, China
| | - Xiao-Xia Fu
- Institute of Laboratory Medicine, Guangdong Medical University, No. 1 Xincheng Road, Dongguan 523808, China
| | - Yong Zhou
- Department of Laboratory Medicine, Dongguan 5th Hospital, Dongguan 523000, China
| | - Jixin Zhong
- Cardiovascular Research Institute, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA
| | - 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
- Institute of Laboratory Medicine, Guangdong Medical University, No. 1 Xincheng Road, Dongguan 523808, China; Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, No. 1 Xincheng Road, Dongguan 523808, China.
| |
Collapse
|
18
|
Gao Y, Xu X, Feng J, Ma Y, Zheng D, Meng Y, Shan F. Effects of interleukin-1 receptor-associated kinase 1 RNA interference in dendritic cells on inflammatory cytokine release and T-cell proliferation. Mol Med Rep 2016; 14:5685-5692. [DOI: 10.3892/mmr.2016.5946] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 09/14/2016] [Indexed: 11/06/2022] Open
|
19
|
Meng C, Wang X, Xu Z, Hu M, Liu J, Pan Z, Chen X, Jiao X. Murine Flt3 ligand-generated plasmacytoid and conventional dendritic cells display functional differentiation in activation, inflammation, and antigen presentation during BCG infection in vitro. In Vitro Cell Dev Biol Anim 2016; 53:67-76. [PMID: 27496194 DOI: 10.1007/s11626-016-0076-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 07/22/2016] [Indexed: 12/31/2022]
Abstract
Dendritic cells (DCs) are composed of distinct subsets. Their immunologic functions (especially in pathogenic infection, such as with mycobacteria) are poorly understood, largely because of their rarity and difficulty of preparation. We used the murine Fms-like tyrosine kinase 3 (Flt3) ligand to generate conventional DCs (FL-cDCs) and plasmacytoid DCs (FL-pDCs) and further evaluated their immunological responses to bacillus Calmette-Guérin (BCG) infection in vitro. BCG cells were observed inside both FL-cDCs and FL-pDCs by confocal microscopy, as confirmed by flow cytometric analysis showing a low infection rate of approximately 6 %, which was similar to in vivo results. The CD40, CD80, CD86, and MHC-II proteins were significantly upregulated in both FL-cDCs and -pDCs beginning at 4 h post-BCG exposure. FL-pDCs secreted TNF-α and IL-6 earlier and at significantly higher levels in the first 12 h following infection, but demonstrated delayed and weak activation and maturation compared to FL-cDCs. Although both subsets proved capable of presenting a mycobacterial antigen, FL-pDCs exhibited weaker activity in this respect than did FL-cDCs. In summary, the existence of FL-generated cDCs and pDCs imply functional differentiation in activation, inflammation, and antigen presentation, although both cells types participated extensively in the immune response to BCG infection.
Collapse
Affiliation(s)
- Chuang Meng
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiaoyan Wang
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
| | - Zhengzhong Xu
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
| | - Maozhi Hu
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China.,Testing Center, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jiaying Liu
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
| | - Zhiming Pan
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiang Chen
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China.
| | - Xinan Jiao
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
20
|
Parida SK, Poiret T, Zhenjiang L, Meng Q, Heyckendorf J, Lange C, Ambati AS, Rao MV, Valentini D, Ferrara G, Rangelova E, Dodoo E, Zumla A, Maeurer M. T-Cell Therapy: Options for Infectious Diseases. Clin Infect Dis 2016; 61Suppl 3:S217-24. [PMID: 26409284 PMCID: PMC4583575 DOI: 10.1093/cid/civ615] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The emergence of drug-resistant tuberculosis is challenging tuberculosis control worldwide. In the absence of an effective vaccine to prevent primary infection with Mycobacterium tuberculosis and tuberculosis disease, host-directed therapies may offer therapeutic options, particularly for patients with multidrug-resistant and extensively drug-resistant tuberculosis where prognosis is often limited. CD8+ and CD4+ T cells mediate antigen-specific adaptive cellular immune responses. Their use in precision immunotherapy in clinical conditions, especially in treating cancer as well as for prevention of life-threatening viral infections in allogeneic transplant recipients, demonstrated safety and clinical efficacy. We review key achievements in T-cell therapy, including the use of recombinant immune recognition molecules (eg, T-cell receptors and CD19 chimeric antigen receptors), and discuss its potential in the clinical management of patients with drug-resistant and refractory tuberculosis failing conventional therapy.
Collapse
Affiliation(s)
- Shreemanta K Parida
- Therapeutic Immunology Division, Department of Laboratory Medicine, Karolinska Institutet
| | - Thomas Poiret
- Therapeutic Immunology Division, Department of Laboratory Medicine, Karolinska Institutet Centre for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Solna, Sweden
| | - Liu Zhenjiang
- Therapeutic Immunology Division, Department of Laboratory Medicine, Karolinska Institutet
| | - Qingda Meng
- Therapeutic Immunology Division, Department of Laboratory Medicine, Karolinska Institutet
| | - Jan Heyckendorf
- Division of Clinical Infectious Diseases, German Center for Infection Research, Research Center Borstel
| | - Christoph Lange
- Division of Clinical Infectious Diseases, German Center for Infection Research, Research Center Borstel International Health/Infectious Diseases, University of Lübeck, Germany Department of Medicine, Karolinska Institutet
| | - Aditya S Ambati
- Therapeutic Immunology Division, Department of Laboratory Medicine, Karolinska Institutet Centre for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Solna, Sweden Department of Medicine, Karolinska Institutet
| | - Martin V Rao
- Therapeutic Immunology Division, Department of Laboratory Medicine, Karolinska Institutet
| | - Davide Valentini
- Therapeutic Immunology Division, Department of Laboratory Medicine, Karolinska Institutet Centre for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Solna, Sweden
| | | | - Elena Rangelova
- Pancreatic Surgery Unit, Division of Surgery, Department of Clinical Science, Intervention and Technology
| | - Ernest Dodoo
- Therapeutic Immunology Division, Department of Laboratory Medicine, Karolinska Institutet Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden
| | - Alimuddin Zumla
- Department of Infection, Division of Infection and Immunity, Centre for Clinical Microbiology, University College London National Institute for Health Research Biomedical Research Centre, University College London Hospitals, United Kingdom
| | - Markus Maeurer
- Therapeutic Immunology Division, Department of Laboratory Medicine, Karolinska Institutet Centre for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
21
|
Cruz-Aguilar M, Castillo-Rodal AI, Schcolnik-Cabrera A, Bonifaz LC, Molina G, López-Vidal Y. TLR4 and DC-SIGN receptors recognized Mycobacterium scrofulaceum promoting semi-activated phenotype on bone marrow dendritic cells. Tuberculosis (Edinb) 2016; 99:31-40. [PMID: 27450002 DOI: 10.1016/j.tube.2016.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 02/18/2016] [Accepted: 04/05/2016] [Indexed: 01/25/2023]
Abstract
Nontuberculous mycobacteria (NTM) are recognized as emerging pathogens and their immune regulatory mechanisms are not well described yet. From them, Mycobacterium avium is known to be a weak activator of dendritic cells (DCs) that impairs the response induced by BCG vaccine. However, whether other NTM such as Mycobacterium scrofulaceum may modulate the activation of DCs, has not been extensively studied. Here, we exposed bone marrow-derived DCs (BMDCs) to M. scrofulaceum and we analyzed the effect on the activation of DCs. We found that M. scrofulaceum has a comparable ability to induce a semi-mature DC phenotype, which was produced by its interaction with DC-SIGN and TLR4 receptors in a synergic effect. BMDCs exposed to M. scrofulaceum showed high expression of PD-L2 and production of IL-10, as well as low levels of co-stimulatory molecules and pro-inflammatory cytokines. In addition to immunophenotype induced on DCs, changes in morphology, re-organization of cytoskeleton and decreased migratory capacity are consistent with a semi-mature phenotype. However, unlike other pathogenic mycobacteria, the DC-semi-mature phenotype induced by M. scrofulaceum was reversed after re-exposure to BCG, suggesting that modulation mechanisms of DC-activation used by M. scrofulaceum are different to other known pathogenic mycobacteria. This is the first report about the immunophenotypic characterization of DC stimulated by M. scrofulaceum.
Collapse
Affiliation(s)
- Marisa Cruz-Aguilar
- Programa de Inmunología Molecular Microbiana, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico.
| | - Antonia I Castillo-Rodal
- Programa de Inmunología Molecular Microbiana, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico.
| | - Alejandro Schcolnik-Cabrera
- Programa de Inmunología Molecular Microbiana, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico.
| | - Laura C Bonifaz
- Unidad de Investigación Médica en Inmunoquímica, Instituto Mexicano del Seguro Social, México, DF, Mexico.
| | - Gabriela Molina
- Programa de Inmunología Molecular Microbiana, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico.
| | - Yolanda López-Vidal
- Programa de Inmunología Molecular Microbiana, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico.
| |
Collapse
|
22
|
Braga AF, Moretto DF, Gigliotti P, Peruchi M, Vilani-Moreno FR, Campanelli AP, Latini ACP, Iyer A, Das PK, Souza VNBD. Activation and cytokine profile of monocyte derived dendritic cells in leprosy: in vitro stimulation by sonicated Mycobacterium leprae induces decreased level of IL-12p70 in lepromatous leprosy. Mem Inst Oswaldo Cruz 2015. [PMID: 26222022 PMCID: PMC4569830 DOI: 10.1590/0074-02760140230] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Dendritic cells (DCs) play a pivotal role in the connection of innate and adaptive immunity of hosts to mycobacterial infection. Studies on the interaction of monocyte-derived DCs (MO-DCs) using Mycobacterium leprae in leprosy patients are rare. The present study demonstrated that the differentiation of MOs to DCs was similar in all forms of leprosy compared to normal healthy individuals. In vitro stimulation of immature MO-DCs with sonicated M. leprae induced variable degrees of DC maturation as determined by the increased expression of HLA-DR, CD40, CD80 and CD86, but not CD83, in all studied groups. The production of different cytokines by the MO-DCs appeared similar in all of the studied groups under similar conditions. However, the production of interleukin (IL)-12p70 by MO-DCs from lepromatous (LL) leprosy patients after in vitro stimulation with M. leprae was lower than tuberculoid leprosy patients and healthy individuals, even after CD40 ligation with CD40 ligand-transfected cells. The present cumulative findings suggest that the MO-DCs of LL patients are generally a weak producer of IL-12p70 despite the moderate activating properties ofM. leprae. These results may explain the poor M. leprae-specific cell-mediated immunity in the LL type of leprosy.
Collapse
Affiliation(s)
- André Flores Braga
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista, Botucatu, SP, BR
| | | | - Patrícia Gigliotti
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista, Botucatu, SP, BR
| | | | | | - Ana Paula Campanelli
- Departamento de Microbiologia e Imunologia, Faculdade de Odontologia de Bauru, Universidade de São Paulo, Bauru, SP, BR
| | | | - Anand Iyer
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, North Holland, NL
| | | | | |
Collapse
|
23
|
Neisseria gonorrhoeae Modulates Immunity by Polarizing Human Macrophages to a M2 Profile. PLoS One 2015; 10:e0130713. [PMID: 26125939 PMCID: PMC4488386 DOI: 10.1371/journal.pone.0130713] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 05/22/2015] [Indexed: 01/15/2023] Open
Abstract
Current data suggest that Neisseria gonorrhoeae is able to suppress the protective immune response at different levels, such as B and T lymphocytes and antigen-presenting cells. The present report is focused on gonococcus evasion mechanism on macrophages (MФ) and its impact in the subsequent immune response. In response to various signals MФ may undergo classical-M1 (M1-MФ) or alternative-M2 (M2-MФ) activation. Until now there are no reports of the gonococcus effects on human MФ polarization. We assessed the phagocytic ability of monocyte-derived MФ (MDM) upon gonococcal infection by immunofluorescence and gentamicin protection experiments. Then, we evaluated cytokine profile and M1/M2 specific-surface markers on MФ challenged with N. gonorrhoeae and their proliferative effect on T cells. Our findings lead us to suggest N. gonorrhoeae stimulates a M2-MФ phenotype in which some of the M2b and none of the M1-MФ-associated markers are induced. Interestingly, N. gonorrhoeae exposure leads to upregulation of a Programmed Death Ligand 1 (PD-L1), widely known as an immunosuppressive molecule. Moreover, functional results showed that N. gonorrhoeae-treated MФ are unable to induce proliferation of human T-cells, suggesting a more likely regulatory phenotype. Taken together, our data show that N. gonorroheae interferes with MФ polarization. This study has important implications for understanding the mechanisms of clearance versus long-term persistence of N. gonorroheae infection and might be applicable for the development of new therapeutic strategies.
Collapse
|
24
|
Francisco NM, Hsu NJ, Keeton R, Randall P, Sebesho B, Allie N, Govender D, Quesniaux V, Ryffel B, Kellaway L, Jacobs M. TNF-dependent regulation and activation of innate immune cells are essential for host protection against cerebral tuberculosis. J Neuroinflammation 2015; 12:125. [PMID: 26112704 PMCID: PMC4488051 DOI: 10.1186/s12974-015-0345-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 06/16/2015] [Indexed: 11/10/2022] Open
Abstract
Background Tuberculosis (TB) affects one third of the global population, and TB of the central nervous system (CNS-TB) is the most severe form of tuberculosis which often associates with high mortality. The pro-inflammatory cytokine tumour necrosis factor (TNF) plays a critical role in the initial and long-term host immune protection against Mycobacterium tuberculosis (M. tuberculosis) which involves the activation of innate immune cells and structure maintenance of granulomas. However, the contribution of TNF, in particular neuron-derived TNF, in the control of cerebral M. tuberculosis infection and its protective immune responses in the CNS were not clear. Methods We generated neuron-specific TNF-deficient (NsTNF−/−) mice and compared outcomes of disease against TNFf/f control and global TNF−/− mice. Mycobacterial burden in brains, lungs and spleens were compared, and cerebral pathology and cellular contributions analysed by microscopy and flow cytometry after M. tuberculosis infection. Activation of innate immune cells was measured by flow cytometry and cell function assessed by cytokine and chemokine quantification using enzyme-linked immunosorbent assay (ELISA). Results Intracerebral M. tuberculosis infection of TNF−/− mice rendered animals highly susceptible, accompanied by uncontrolled bacilli replication and eventual mortality. In contrast, NsTNF−/− mice were resistant to infection and presented with a phenotype similar to that in TNFf/f control mice. Impaired immunity in TNF−/− mice was associated with altered cytokine and chemokine synthesis in the brain and characterised by a reduced number of activated innate immune cells. Brain pathology reflected enhanced inflammation dominated by neutrophil influx. Conclusion Our data show that neuron-derived TNF has a limited role in immune responses, but overall TNF production is necessary for protective immunity against CNS-TB.
Collapse
Affiliation(s)
- Ngiambudulu M Francisco
- Division of Immunology, Department of Clinical Laboratory Science, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925, South Africa.
| | - Nai-Jen Hsu
- Division of Immunology, Department of Clinical Laboratory Science, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925, South Africa.
| | - Roanne Keeton
- Division of Immunology, Department of Clinical Laboratory Science, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925, South Africa.
| | - Philippa Randall
- Division of Immunology, Department of Clinical Laboratory Science, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925, South Africa.
| | - Boipelo Sebesho
- Division of Immunology, Department of Clinical Laboratory Science, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925, South Africa.
| | - Nasiema Allie
- Division of Immunology, Department of Clinical Laboratory Science, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925, South Africa. .,Division for Postgraduate Studies, University of the Western Cape, Bellville, South Africa.
| | - Dhirendra Govender
- Division of Anatomical Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa. .,National Health Laboratory Service, Johannesburg, South Africa.
| | - Valerie Quesniaux
- Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans, France. .,CNRS UMR7355, Orleans, France.
| | - Bernhard Ryffel
- Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans, France. .,CNRS UMR7355, Orleans, France.
| | - Lauriston Kellaway
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| | - Muazzam Jacobs
- Division of Immunology, Department of Clinical Laboratory Science, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925, South Africa. .,National Health Laboratory Service, Johannesburg, South Africa.
| |
Collapse
|
25
|
Mendum TA, Wu H, Kierzek AM, Stewart GR. Lipid metabolism and Type VII secretion systems dominate the genome scale virulence profile of Mycobacterium tuberculosis in human dendritic cells. BMC Genomics 2015; 16:372. [PMID: 25956932 PMCID: PMC4425887 DOI: 10.1186/s12864-015-1569-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 04/23/2015] [Indexed: 12/20/2022] Open
Abstract
Background Mycobacterium tuberculosis continues to kill more people than any other bacterium. Although its archetypal host cell is the macrophage, it also enters, and survives within, dendritic cells (DCs). By modulating the behaviour of the DC, M. tuberculosis is able to manipulate the host’s immune response and establish an infection. To identify the M. tuberculosis genes required for survival within DCs we infected primary human DCs with an M. tuberculosis transposon library and identified mutations with a reduced ability to survive. Results Parallel sequencing of the transposon inserts of the surviving mutants identified a large number of genes as being required for optimal intracellular fitness in DCs. Loci whose mutation attenuated intracellular survival included those involved in synthesising cell wall lipids, not only the well-established virulence factors, pDIM and cord factor, but also sulfolipids and PGL, which have not previously been identified as having a direct virulence role in cells. Other attenuated loci included the secretion systems ESX-1, ESX-2 and ESX-4, alongside many PPE genes, implicating a role for ESX-5. In contrast the canonical ESAT-6 family of ESX substrates did not have intra-DC fitness costs suggesting an alternative ESX-1 associated virulence mechanism. With the aid of a gene-nutrient interaction model, metabolic processes such as cholesterol side chain catabolism, nitrate reductase and cysteine-methionine metabolism were also identified as important for survival in DCs. Conclusion We conclude that many of the virulence factors required for survival in DC are shared with macrophages, but that survival in DCs also requires several additional functions, such as cysteine-methionine metabolism, PGLs, sulfolipids, ESX systems and PPE genes. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1569-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tom A Mendum
- Department of Microbial and Cellular Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK.
| | - Huihai Wu
- Department of Microbial and Cellular Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK.
| | - Andrzej M Kierzek
- Department of Microbial and Cellular Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK.
| | - Graham R Stewart
- Department of Microbial and Cellular Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK.
| |
Collapse
|
26
|
Hodgson K, Morris J, Bridson T, Govan B, Rush C, Ketheesan N. Immunological mechanisms contributing to the double burden of diabetes and intracellular bacterial infections. Immunology 2015; 144:171-85. [PMID: 25262977 DOI: 10.1111/imm.12394] [Citation(s) in RCA: 243] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Revised: 08/12/2014] [Accepted: 08/19/2014] [Indexed: 12/11/2022] Open
Abstract
Diabetes has been recognized as an important risk factor for a variety of intracellular bacterial infections, but research into the dysregulated immune mechanisms contributing to the impaired host-pathogen interactions is in its infancy. Diabetes is characterized by a chronic state of low-grade inflammation due to activation of pro-inflammatory mediators and increased formation of advanced glycation end products. Increased oxidative stress also exacerbates the chronic inflammatory processes observed in diabetes. The reduced phagocytic and antibacterial activity of neutrophils and macrophages provides an intracellular niche for the pathogen to replicate. Phagocytic and antibacterial dysfunction may be mediated directly through altered glucose metabolism and oxidative stress. Furthermore, impaired activation of natural killer cells contributes to decreased levels of interferon-γ, required for promoting macrophage antibacterial mechanisms. Together with impaired dendritic cell function, this impedes timely activation of adaptive immune responses. Increased intracellular oxidation of antigen-presenting cells in individuals with diabetes alters the cytokine profile generated and the subsequent balance of T-cell immunity. The establishment of acute intracellular bacterial infections in the diabetic host is associated with impaired T-cell-mediated immune responses. Concomitant to the greater intracellular bacterial burden and potential cumulative effect of chronic inflammatory processes, late hyper-inflammatory cytokine responses are often observed in individuals with diabetes, contributing to systemic pathology. The convergence of intracellular bacterial infections and diabetes poses new challenges for immunologists, providing the impetus for multidisciplinary research.
Collapse
Affiliation(s)
- Kelly Hodgson
- Infectious Diseases and Immunopathogenesis Research Group, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Qld, Australia
| | | | | | | | | | | |
Collapse
|
27
|
Roberts LL, Robinson CM. Mycobacterium tuberculosis infection of human dendritic cells decreases integrin expression, adhesion and migration to chemokines. Immunology 2014; 141:39-51. [PMID: 23981064 DOI: 10.1111/imm.12164] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 08/19/2013] [Accepted: 08/20/2013] [Indexed: 12/25/2022] Open
Abstract
Tuberculosis (TB) remains a major global health problem accounting for millions of deaths annually. Approximately one-third of the world's population is infected with the causative agent Mycobacterium tuberculosis. The onset of an adaptive immune response to M. tuberculosis is delayed compared with other microbial infections. This delay permits bacterial growth and dissemination. The precise mechanism(s) responsible for this delay have remained obscure. T-cell activation is preceded by dendritic cell (DC) migration from infected lungs to local lymph nodes and synapsis with T cells. We hypothesized that M. tuberculosis may impede the ability of DCs to reach lymph nodes and initiate an adaptive immune response. We used primary human DCs to determine the effect of M. tuberculosis on expression of heterodimeric integrins involved in cellular adhesion and migration. We also evaluated the ability of infected DCs to adhere to and migrate through lung endothelial cells, which is necessary to reach lymph nodes. We show by flow cytometry and confocal microscopy that M. tuberculosis-infected DCs exhibit a significant reduction in surface expression of the β(2) (CD18) integrin. Distribution of integrin β(2) is also markedly altered in M. tuberculosis-infected DCs. A corresponding reduction in the αL (CD11a) and αM (CD11b) subunits that associate with integrin β(2) was also observed. Consistent with reduced integrin surface expression, we show a significant reduction in adherence to lung endothelial cell monolayers and migration towards lymphatic chemokines when DCs are infected with M. tuberculosis. These findings suggest that M. tuberculosis modulates DC adhesion and migration to increase the time required to initiate an adaptive immune response.
Collapse
Affiliation(s)
- Lawton L Roberts
- Department of Pathology, Microbiology, & Immunology, University of South Carolina School of Medicine, Columbia, SC, USA
| | | |
Collapse
|
28
|
Abdul Hafid SR, Chakravarthi S, Nesaretnam K, Radhakrishnan AK. Tocotrienol-adjuvanted dendritic cells inhibit tumor growth and metastasis: a murine model of breast cancer. PLoS One 2013; 8:e74753. [PMID: 24069344 PMCID: PMC3777960 DOI: 10.1371/journal.pone.0074753] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 08/06/2013] [Indexed: 11/23/2022] Open
Abstract
Tocotrienol-rich fraction (TRF) from palm oil is reported to possess anti-cancer and immune-enhancing effects. In this study, TRF supplementation was used as an adjuvant to enhance the anti-cancer effects of dendritic cells (DC)-based cancer vaccine in a syngeneic mouse model of breast cancer. Female BALB/c mice were inoculated with 4T1 cells in mammary pad to induce tumor. When the tumor was palpable, the mice in the experimental groups were injected subcutaneously with DC-pulsed with tumor lysate (TL) from 4T1 cells (DC+TL) once a week for three weeks and fed daily with 1 mg TRF or vehicle. Control mice received unpulsed DC and were fed with vehicle. The combined therapy of using DC+TL injections and TRF supplementation (DC+TL+TRF) inhibited (p<0.05) tumor growth and metastasis. Splenocytes from the DC+TL+TRF group cultured with mitomycin-C (MMC)-treated 4T1 cells produced higher (p<0.05) levels of IFN-γ and IL-12. The cytotoxic T-lymphocyte (CTL) assay also showed enhanced tumor-specific killing (p<0.05) by CD8(+) T-lymphocytes isolated from mice in the DC+TL+TRF group. This study shows that TRF has the potential to be used as an adjuvant to enhance effectiveness of DC-based vaccines.
Collapse
Affiliation(s)
- Sitti Rahma Abdul Hafid
- Pathology Division, Faculty of Medicine and Health, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
- Nutrition Unit, Malaysian Palm Oil Board, Bandar Baru Bangi, Selangor, Malaysia
| | - Srikumar Chakravarthi
- Pathology Division, Faculty of Medicine and Health, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | | | - Ammu Kutty Radhakrishnan
- Pathology Division, Faculty of Medicine and Health, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| |
Collapse
|
29
|
Abstract
The immune response against Mycobacterium tuberculosis is multifactorial, involving a network of innate and adaptive immune responses. Characterization of the immune response, a clear understanding of the dynamics and interplay of different arms of the immune response are critical to allow the development of better tools for combating tuberculosis. Dendritic cells (DCs) are one of the key cells in bridging innate and adaptive immune response through their significant role in capturing, processing and presenting antigens. The outcome of interaction of M. tuberculosis with DCs is not fully understood and the available reports are contradictory were some findings reported that DCs strengthen the cellular immune response against mycobacterium infection whereas others reported M. tuberculosis impairs the function of DCs were infected DCs are poor stimulators of M. tuberculosis Ag-specific CD4 T cells. Other studies showed that the outcome depends on M. tuberculosis strain type and type of receptor on DCs during recognition. In this review I shall highlight the recent findings in the outcome of interaction of Mycobacterium tuberculosis with DCs.
Collapse
Affiliation(s)
- Adane Mihret
- Armauer Hansen Research Institute; Addis Ababa, Ethopia.
| |
Collapse
|
30
|
Li W, Deng G, Li M, Liu X, Wang Y. Roles of Mucosal Immunity against Mycobacterium tuberculosis Infection. Tuberc Res Treat 2012; 2012:791728. [PMID: 23213508 PMCID: PMC3504404 DOI: 10.1155/2012/791728] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 09/12/2012] [Accepted: 09/27/2012] [Indexed: 12/21/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), is one of the world's leading infectious causes of morbidity and mortality. As a mucosal-transmitted pathogen, Mtb infects humans and animals mainly through the mucosal tissue of the respiratory tract. Apart from providing a physical barrier against the invasion of pathogen, the major function of the respiratory mucosa may be to serve as the inductive sites to initiate mucosal immune responses and sequentially provide the first line of defense for the host to defend against this pathogen. A large body of studies in the animals and humans have demonstrated that the mucosal immune system, rather than the systemic immune system, plays fundamental roles in the host's defense against Mtb infection. Therefore, the development of new vaccines and novel delivery routes capable of directly inducing respiratory mucosal immunity is emphasized for achieving enhanced protection from Mtb infection. In this paper, we outline the current state of knowledge regarding the mucosal immunity against Mtb infection, including the development of TB vaccines, and respiratory delivery routes to enhance mucosal immunity are discussed.
Collapse
Affiliation(s)
- Wu Li
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, Ningxia 750021, China
- College of Life Science, Ningxia University, 539 W. Helanshan Road, Xixia District, Yinchuan, Ningxia 750021, China
| | - Guangcun Deng
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, Ningxia 750021, China
- College of Life Science, Ningxia University, 539 W. Helanshan Road, Xixia District, Yinchuan, Ningxia 750021, China
| | - Min Li
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, Ningxia 750021, China
- College of Life Science, Ningxia University, 539 W. Helanshan Road, Xixia District, Yinchuan, Ningxia 750021, China
| | - Xiaoming Liu
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, Ningxia 750021, China
- College of Life Science, Ningxia University, 539 W. Helanshan Road, Xixia District, Yinchuan, Ningxia 750021, China
| | - Yujiong Wang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, Ningxia 750021, China
- College of Life Science, Ningxia University, 539 W. Helanshan Road, Xixia District, Yinchuan, Ningxia 750021, China
| |
Collapse
|