1
|
Nainu F, Ophinni Y, Shiratsuchi A, Nakanishi Y. Apoptosis and Phagocytosis as Antiviral Mechanisms. Subcell Biochem 2023; 106:77-112. [PMID: 38159224 DOI: 10.1007/978-3-031-40086-5_3] [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: 01/03/2024]
Abstract
Viruses are infectious entities that make use of the replication machinery of their hosts to produce more progenies, causing disease and sometimes death. To counter viral infection, metazoan hosts are equipped with various defense mechanisms, from the rapid-evoking innate immune responses to the most advanced adaptive immune responses. Previous research demonstrated that cells in fruit flies and mice infected with Drosophila C virus and influenza, respectively, undergo apoptosis, which triggers the engulfment of apoptotic virus-infected cells by phagocytes. This process involves the recognition of eat-me signals on the surface of virus-infected cells by receptors of specialized phagocytes, such as macrophages and neutrophils in mice and hemocytes in fruit flies, to facilitate the phagocytic elimination of virus-infected cells. Inhibition of phagocytosis led to severe pathologies and death in both species, indicating that apoptosis-dependent phagocytosis of virus-infected cells is a conserved antiviral mechanism in multicellular organisms. Indeed, our understanding of the mechanisms underlying apoptosis-dependent phagocytosis of virus-infected cells has shed a new perspective on how hosts defend themselves against viral infection. This chapter explores the mechanisms of this process and its potential for developing new treatments for viral diseases.
Collapse
Affiliation(s)
- Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia.
| | - Youdiil Ophinni
- Division of Clinical Virology, Center for Infectious Diseases, Kobe University Graduate School of Medicine, Kobe, Japan
- Laboratory of Host Defense, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Akiko Shiratsuchi
- Center for Medical Education, Sapporo Medical University, Sapporo, Japan
- Division of Biological Function and Regulation, Graduate School of Medicine, Sapporo Medical University, Sapporo, Japan
| | | |
Collapse
|
2
|
Catalán D, Mansilla MA, Ferrier A, Soto L, Oleinika K, Aguillón JC, Aravena O. Immunosuppressive Mechanisms of Regulatory B Cells. Front Immunol 2021; 12:611795. [PMID: 33995344 PMCID: PMC8118522 DOI: 10.3389/fimmu.2021.611795] [Citation(s) in RCA: 184] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/19/2021] [Indexed: 12/12/2022] Open
Abstract
Regulatory B cells (Bregs) is a term that encompasses all B cells that act to suppress immune responses. Bregs contribute to the maintenance of tolerance, limiting ongoing immune responses and reestablishing immune homeostasis. The important role of Bregs in restraining the pathology associated with exacerbated inflammatory responses in autoimmunity and graft rejection has been consistently demonstrated, while more recent studies have suggested a role for this population in other immune-related conditions, such as infections, allergy, cancer, and chronic metabolic diseases. Initial studies identified IL-10 as the hallmark of Breg function; nevertheless, the past decade has seen the discovery of other molecules utilized by human and murine B cells to regulate immune responses. This new arsenal includes other anti-inflammatory cytokines such IL-35 and TGF-β, as well as cell surface proteins like CD1d and PD-L1. In this review, we examine the main suppressive mechanisms employed by these novel Breg populations. We also discuss recent evidence that helps to unravel previously unknown aspects of the phenotype, development, activation, and function of IL-10-producing Bregs, incorporating an overview on those questions that remain obscure.
Collapse
Affiliation(s)
- Diego Catalán
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile.,Instituto Milenio en Inmunología e Inmunoterapia, Santiago, Chile
| | - Miguel Andrés Mansilla
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Ashley Ferrier
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile.,Instituto Milenio en Inmunología e Inmunoterapia, Santiago, Chile
| | - Lilian Soto
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile.,Unidad de Dolor, Hospital Clínico, Universidad de Chile (HCUCH), Santiago, Chile
| | | | - Juan Carlos Aguillón
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Octavio Aravena
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| |
Collapse
|
3
|
Zheng X, Wang J, Zhang X, Fu Y, Peng Q, Lu J, Wei L, Li Z, Liu C, Wu Y, Yan Q, Ma J. RNA m 6 A methylation regulates virus-host interaction and EBNA2 expression during Epstein-Barr virus infection. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:351-362. [PMID: 33434416 PMCID: PMC8127537 DOI: 10.1002/iid3.396] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/24/2020] [Accepted: 11/25/2020] [Indexed: 12/19/2022]
Abstract
Introduction N6‐methyladenosine (m6A) is the most prevalent modification that occurs in messenger RNA (mRNA), affecting mRNA splicing, translation, and stability. This modification is reversible, and its related biological functions are mediated by “writers,” “erasers,” and “readers.” The field of viral epitranscriptomics and the role of m6A modification in virus–host interaction have attracted much attention recently. When Epstein–Barr virus (EBV) infects a human B lymphocyte, it goes through three phases: the pre‐latent phase, latent phase, and lytic phase. Little is known about the viral and cellular m6A epitranscriptomes in EBV infection, especially in the pre‐latent phase during de novo infection. Methods Methylated RNA immunoprecipitation sequencing (MeRIP‐seq) and MeRIP‐RT‐qPCR were used to determine the m6A‐modified transcripts during de novo EBV infection. RIP assay was used to confirm the binding of EBNA2 and m6A readers. Quantitative reverse‐transcription polymerase chain reaction (RT‐qPCR) and Western blot analysis were performed to test the effect of m6A on the host and viral gene expression. Results Here, we provided mechanistic insights by examining the viral and cellular m6A epitranscriptomes during de novo EBV infection, which is in the pre‐latent phase. EBV EBNA2 and BHRF1 were highly m6A‐modified upon EBV infection. Knockdown of METTL3 (a “writer”) decreased EBNA2 expression levels. The emergent m6A modifications induced by EBV infection preferentially distributed in 3ʹ untranslated regions of cellular transcripts, while the lost m6A modifications induced by EBV infection preferentially distributed in coding sequence regions of mRNAs. EBV infection could influence the host cellular m6A epitranscriptome. Conclusions These results reveal the critical role of m6A modification in the process of de novo EBV infection.
Collapse
Affiliation(s)
- Xiang Zheng
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Cancer Research Institute, Department of Microbiology, Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, China.,Department of Pathology, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha, Hunan, China
| | - Jia Wang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Cancer Research Institute, Department of Microbiology, Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha, Hunan, China.,Department of Immunology, Changzhi Medical College, Changzhi, Shanxi, China
| | - Xiaoyue Zhang
- Cancer Research Institute, Department of Microbiology, Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha, Hunan, China
| | - Yuxin Fu
- Cancer Research Institute, Department of Microbiology, Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Qiu Peng
- Cancer Research Institute, Department of Microbiology, Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha, Hunan, China
| | - Jianhong Lu
- Cancer Research Institute, Department of Microbiology, Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Lingyu Wei
- Cancer Research Institute, Department of Microbiology, Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha, Hunan, China
| | - Zhengshuo Li
- Cancer Research Institute, Department of Microbiology, Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha, Hunan, China
| | - Can Liu
- Cancer Research Institute, Department of Microbiology, Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha, Hunan, China
| | - Yangge Wu
- Department of Pathology, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha, Hunan, China
| | - Qun Yan
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jian Ma
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Cancer Research Institute, Department of Microbiology, Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha, Hunan, China
| |
Collapse
|
4
|
Eiza N, Zuckerman E, Carlebach M, Rainis T, Goldberg Y, Vadasz Z. Increased killer B cells in chronic HCV infection may lead to autoimmunity and increased viral load. Clin Exp Immunol 2019; 193:183-193. [PMID: 29665000 DOI: 10.1111/cei.13139] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2018] [Indexed: 01/20/2023] Open
Abstract
Regulatory B (Breg ) cells are characterized by various membrane markers and the secretion of different inhibitory cytokines. A new subset of Breg cells was identified as CD5hi Fas-ligand (FasL)hi . Their main reported role is to suppress anti-viral and anti-tumour immune responses, and, hence they have been dubbed 'killer' B cells. In this study, we aim to assess the role of these cells in chronic hepatitis C virus (HCV) infection, and determine if they contribute to the increased viral load and persistence of HCV and its related autoimmunity. (i) FasL expression on CD5hi B cells is increased significantly in HCV-infected patients compared to healthy individuals [28·06 ± 6·71 mean fluorescence intensity (MFI) ± standard error of the mean (s.e.m.), median = 27·9 versus 10·87 ± 3·97 MFI ± s.e.m., median = 10·3, respectively, P < 0·0001]. (ii) Killer B cells from HCV patients increased autologous CD4+ T cell apoptosis compared to the apoptosis in healthy individuals [39·17% ± 7·18% mean ± standard deviation (s.d.), median = 39·6 versus 25·92 ± 8·65%, mean ± s.d., median = 24·1%, P < 0·0001, respectively]. A similar increase was observed in CD8+ T cell apoptosis (54·67 ± 15·49% mean ± s.d., median = 57·3 versus 21·07% ± 7·4%, mean ± s.d., median = 20%, P = 0·0006, respectively). (iii) By neutralizing FasL with monoclonal anti-FasL antibodies, we have shown that the induction of apoptosis by killer B cells is FasL-dependent. (iv) Increased expression of FasL on CD5hi B cells is correlated positively with an increased viral load and the presence of anti-nuclear antibodies and rheumatoid factor in HCV. This is the first study in which killer B cells have been suggested to play a pathogenic role in HCV. They seem to be involved in HCV's ability to escape efficient immune responses.
Collapse
Affiliation(s)
- N Eiza
- Division of Allergy and Clinical Immunology, Bnai Zion Medical Center, Haifa, Israel
| | - E Zuckerman
- Unit of Hepatology, Carmel Medical Center, Haifa, Israel
| | - M Carlebach
- Division of Gastroenterology, Bnai Zion Medical Center, Haifa, Israel.,Faculty of Medicine, Technion, Haifa, Israel
| | - T Rainis
- Division of Gastroenterology, Bnai Zion Medical Center, Haifa, Israel.,Faculty of Medicine, Technion, Haifa, Israel
| | - Y Goldberg
- Department of Statistics, University of Haifa, Haifa, Israel
| | - Z Vadasz
- Division of Allergy and Clinical Immunology, Bnai Zion Medical Center, Haifa, Israel
| |
Collapse
|
5
|
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
|
6
|
Virus Infection and Death Receptor-Mediated Apoptosis. Viruses 2017; 9:v9110316. [PMID: 29077026 PMCID: PMC5707523 DOI: 10.3390/v9110316] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/23/2017] [Accepted: 10/25/2017] [Indexed: 02/07/2023] Open
Abstract
Virus infection can trigger extrinsic apoptosis. Cell-surface death receptors of the tumor necrosis factor family mediate this process. They either assist persistent viral infection or elicit the elimination of infected cells by the host. Death receptor-mediated apoptosis plays an important role in viral pathogenesis and the host antiviral response. Many viruses have acquired the capability to subvert death receptor-mediated apoptosis and evade the host immune response, mainly by virally encoded gene products that suppress death receptor-mediated apoptosis. In this review, we summarize the current information on virus infection and death receptor-mediated apoptosis, particularly focusing on the viral proteins that modulate death receptor-mediated apoptosis.
Collapse
|
7
|
Yamada A, Arakaki R, Saito M, Kudo Y, Ishimaru N. Dual Role of Fas/FasL-Mediated Signal in Peripheral Immune Tolerance. Front Immunol 2017; 8:403. [PMID: 28424702 PMCID: PMC5380675 DOI: 10.3389/fimmu.2017.00403] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 03/21/2017] [Indexed: 12/20/2022] Open
Abstract
Fas-mediated apoptosis contributes to physiological and pathological cellular processes, such as differentiation and survival. In particular, the roles of Fas in immune cells are complex and critical for the maintenance of immune tolerance. The precise pathways and unique functions associated with Fas/FasL-mediated signaling in the immune system are known. The dual character of Fas/FasL-mediated immune regulation that induces beneficial or harmful effects is associated with the onset or development of immune disorders. Studies on mutations in genes encoding Fas and FasL gene of humans and mice contributed to our understanding of the pathogenesis of autoimmune diseases. Here, we review the opposing functions of Fas/FasL-mediated signaling, bilateral effects of Fas/FasL on in immune cells, and complex pathogenesis of autoimmunity mediated by Fas/FasL.
Collapse
Affiliation(s)
- Akiko Yamada
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Rieko Arakaki
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Masako Saito
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yasusei Kudo
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| |
Collapse
|
8
|
Wang J, Wu J, Yi L, Hou Z, Li W. Pathological analysis, detection of antigens, FasL expression analysis and leucocytes survival analysis in tilapia (Oreochromis niloticus) after infection with green fluorescent protein labeled Streptococcus agalactiae. FISH & SHELLFISH IMMUNOLOGY 2017; 62:86-95. [PMID: 28063953 DOI: 10.1016/j.fsi.2017.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 01/02/2017] [Accepted: 01/03/2017] [Indexed: 06/06/2023]
Abstract
The pathogenesis of Streptococcus agalactiae infection in tilapia has not been fully described. To understand this, we investigated the clinic-pathological features of acute experimental septicemia in tilapia (Oreochromis niloticus) after receiving an intra-peritoneal injection with S. agalactiae THN-1901GFP. Immunohistochemistry and sections of pathological tissues were used to estimate the level of damage in the head-kidney, liver, spleen and trunk-kidney. The expression of FasL was analyzed by western blotting in these samples based on their damage levels. Leucocytes were isolated from the head-kidney and incubated with S. agalactiae THN-1901GFP. Then, phagocytosis, programmed cell death and the expression of FasL were analyzed. The infected tissues showed varying degrees of necrosis and histolysis. The serous membrane of the intestine was dissolved by S. agalactiae THN-1901GFP. Antigens of S. agalactiae THN-1901GFP accumulated in different parts of the infected organs. In the head-kidney and spleen, the expression of FasL was up-regulated in parallel with increased tissue damage. After being incubated with S. agalactiae THN-1901GFP, the phagocytic capacity and ability were both very high and the expression of FasL remained high in leucocytes. S. agalactiae THN-1901GFP was able to survive for a long period of time after being engulfed by phagocytic cells. These findings offer insight into the pathogenesis of S. agalactiae infection in tilapia.
Collapse
Affiliation(s)
- Jingyuan Wang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, PR China
| | - Jinying Wu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, PR China.
| | - Liyuan Yi
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, PR China
| | - Zengxin Hou
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, PR China
| | - Wensheng Li
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, PR China
| |
Collapse
|
9
|
Dunham J, van Driel N, Eggen BJ, Paul C, 't Hart BA, Laman JD, Kap YS. Analysis of the cross-talk of Epstein-Barr virus-infected B cells with T cells in the marmoset. Clin Transl Immunology 2017; 6:e127. [PMID: 28243437 PMCID: PMC5311918 DOI: 10.1038/cti.2017.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 02/06/2023] Open
Abstract
Despite the well-known association of Epstein–Barr virus (EBV), a lymphocryptovirus (LCV), with multiple sclerosis, a clear pathogenic role for disease progression has not been established. The translationally relevant experimental autoimmune encephalomyelitis (EAE) model in marmoset monkeys revealed that LCV-infected B cells have a central pathogenic role in the activation of T cells that drive EAE progression. We hypothesized that LCV-infected B cells induce T-cell functions relevant for EAE progression. In the current study, we examined the ex vivo cross-talk between lymph node mononuclear cells (MNCs) from EAE marmosets and (semi-) autologous EBV-infected B-lymphoblastoid cell lines (B-LCLs). Results presented here demonstrate that infection with EBV B95-8 has a strong impact on gene expression profile of marmoset B cells, particularly those involved with antigen processing and presentation or co-stimulation to T cells. At the cellular level, we observed that MNC co-culture with B-LCLs induced decrease of CCR7 expression on T cells from EAE responder marmosets, but not in EAE monkeys without clinically evident disease. B-LCL interaction with T cells also resulted in significant loss of CD27 expression and reduced expression of IL-23R and CCR6, which coincided with enhanced IL-17A production. These results highlight the profound impact that EBV-infected B-LCL cells can have on second and third co-stimulatory signals involved in (autoreactive) T-cell activation.
Collapse
Affiliation(s)
- Jordon Dunham
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands; Department of Neuroscience, University Groningen, University Medical Center, Groningen, The Netherlands
| | - Nikki van Driel
- Department of Immunobiology, Biomedical Primate Research Centre , Rijswijk, The Netherlands
| | - Bart Jl Eggen
- Department of Neuroscience, University Groningen, University Medical Center , Groningen, The Netherlands
| | - Chaitali Paul
- Department of Neuroscience, University Groningen, University Medical Center , Groningen, The Netherlands
| | - Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands; Department of Neuroscience, University Groningen, University Medical Center, Groningen, The Netherlands
| | - Jon D Laman
- Department of Neuroscience, University Groningen, University Medical Center , Groningen, The Netherlands
| | - Yolanda S Kap
- Department of Immunobiology, Biomedical Primate Research Centre , Rijswijk, The Netherlands
| |
Collapse
|
10
|
Sale S, Carone AE, Fumi M, Pancione Y, Rocco V. Detection of Apoptotic Lymphocytes Through Sysmex XN-1000 As a Diagnostic Marker for Mononucleosis Syndrome. J Clin Lab Anal 2016; 30:779-93. [PMID: 27094572 DOI: 10.1002/jcla.21938] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 12/28/2015] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND The infectious mononucleosis (IM) includes clue elements, apoptotic and atypical lymphocytes. In IM, the evaluation of dot plot provided by Sysmex XN-1000 analyzer revealed a stretched lymphocytic cluster, white cell differential channel (WDF), on cytogram. METHODS In this study, we analyzed 698 samples that include 39 IM, 76 chronic lymphoproliferative disorders, 25 nonclonal lymphocytosis, and 40 healthy donors. Five hundred eighteen samples with other diseases or interference were evaluated. The algorithm was validated on 40,000 files that were received from internal database of Sysmex-Dasit. RESULTS The analysis of flow cytometry standard (FCS) files in WDF channel and presumed apoptotic lymphocytes counts on side scatter/forward scatter (SSC/FSC) and SSC/SFL (where SFL is side fluorescence) dot plot revealed excellent correlation among apoptotic cells on peripheral blood smear (R(2) = 0.79 and 0.75). There was a variation of positional parameters in lymphocyte clusters WX, WY, and WZ. If WX-SSC > 500 and WY-SFL > 1,000 and WZ-FSC > 700, specificity equals to 99% and sensitivity equals to 100%. If nucleated red blood cell (NRBC) <0.03 × 10(3) /μl, specificity equals to 100%. In received files, positives were 1% adding the simultaneous presence of a percentage of events in the two gates relating to the apoptotic lymphocytes of 1.2% of WBC. CONCLUSION On Sysmex XN-1000, dot-plot observation allowed immediate detection of IM. Meanwhile, an algorithm based on the parameters on these plots can be calculated with excellent performance.
Collapse
Affiliation(s)
- Silvia Sale
- Clinical Pathology Laboratory, A.O.R.N. "G. Rummo,", Benevento, Italy
| | | | - Maurizio Fumi
- Clinical Pathology Laboratory, A.O.R.N. "G. Rummo,", Benevento, Italy
| | - Ylenia Pancione
- Clinical Pathology Laboratory, A.O.R.N. "G. Rummo,", Benevento, Italy
| | - Vincenzo Rocco
- Clinical Pathology Laboratory, A.O.R.N. "G. Rummo,", Benevento, Italy.
| |
Collapse
|
11
|
Lundy SK, Klinker MW, Fox DA. Killer B lymphocytes and their fas ligand positive exosomes as inducers of immune tolerance. Front Immunol 2015; 6:122. [PMID: 25852690 PMCID: PMC4367442 DOI: 10.3389/fimmu.2015.00122] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 03/04/2015] [Indexed: 01/31/2023] Open
Abstract
Induction of immune tolerance is a key process by which the immune system is educated to modulate reactions against benign stimuli such as self-antigens and commensal microbes. Understanding and harnessing the natural mechanisms of immune tolerance may become an increasingly useful strategy for treating many types of allergic and autoimmune diseases, as well as for improving the acceptance of solid organ transplants. Our laboratory and others have been interested in the natural ability of some B lymphocytes to express the death-inducing molecule Fas ligand (FasL), and their ability to kill T helper (TH) lymphocytes. We have recently shown that experimental transformation of human B cells by a non-replicative variant of Epstein-Barr virus (EBV) consistently resulted in high expression of functional FasL protein. The production and release of FasL+ exosomes that co-expressed major histocompatibility complex (MHC) class II molecules and had the capacity to kill antigen-specific TH cells was also observed. Several lines of evidence indicate that FasL+ B cells and FasL+MHCII+ exosomes have important roles in natural immune tolerance and have a great deal of therapeutic potential. Taken together, these findings suggest that EBV-immortalized human B lymphoblastoid cell lines could be used as cellular factories for FasL+ exosomes, which would be employed to therapeutically establish and/or regain immune tolerance toward specific antigens. The goals of this review are to summarize current knowledge of the roles of FasL+ B cells and exosomes in immune regulation, and to suggest methods of manipulating killer B cells and FasL+ exosomes for clinical purposes.
Collapse
Affiliation(s)
- Steven K Lundy
- Department of Internal Medicine-Rheumatology, University of Michigan Medical School , Ann Arbor, MI , USA ; Graduate Training Program in Immunology, University of Michigan Medical School , Ann Arbor, MI , USA
| | - Matthew W Klinker
- Graduate Training Program in Immunology, University of Michigan Medical School , Ann Arbor, MI , USA
| | - David A Fox
- Department of Internal Medicine-Rheumatology, University of Michigan Medical School , Ann Arbor, MI , USA ; Graduate Training Program in Immunology, University of Michigan Medical School , Ann Arbor, MI , USA
| |
Collapse
|
12
|
Rincón-Arévalo H, Sanchez-Parra CC, Castaño D, Yassin L, Vásquez G. Regulatory B Cells and Mechanisms. Int Rev Immunol 2015; 35:156-76. [PMID: 25793964 DOI: 10.3109/08830185.2015.1015719] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Regulatory B cells have gained prominence in their role as modulators of the immune response against tumors, infectious diseases, and autoimmune diseases, such as systemic lupus erythematosus, rheumatoid arthritis, and multiple sclerosis, among others. The concept of regulatory B cells has been strongly associated with interleukin (IL)-10 production; however, there is growing evidence that supports the existence of other regulatory mechanisms, such as the production of transforming growth factor β (TGF-β), induced cell death of effector T cells, and the induction of CD4(+)CD25(-)Foxp3(+) regulatory T cells. The regulatory function of B cells has been associated with the presence and activation of molecules such as CD40, CD19, CD1d, and BCR. Alterations in signaling by any of these pathways leads to a marked defect in regulatory B cells and to increased clinical symptoms and proinflammatory signs, both in murine models and in autoimmune diseases in humans. B cells mainly exert their regulatory effect through the inhibition of proliferation and production of proinflammatory mediators, such as TNF-α, IFN-γ, and IL-17 by CD4(+) T cells. A better understanding of how regulatory B cells function will offer new perspectives with regard to the treatment of various human diseases.
Collapse
Affiliation(s)
- Héctor Rincón-Arévalo
- a Grupo de Inmunología Celular e InmunogenéInstituto de Investigaciones Médicas, Facultad de Medicina , Universidad de Antioquia Medellín , Colombia
| | - Claudia C Sanchez-Parra
- a Grupo de Inmunología Celular e InmunogenéInstituto de Investigaciones Médicas, Facultad de Medicina , Universidad de Antioquia Medellín , Colombia
| | - Diana Castaño
- a Grupo de Inmunología Celular e InmunogenéInstituto de Investigaciones Médicas, Facultad de Medicina , Universidad de Antioquia Medellín , Colombia
| | - Lina Yassin
- a Grupo de Inmunología Celular e InmunogenéInstituto de Investigaciones Médicas, Facultad de Medicina , Universidad de Antioquia Medellín , Colombia.,b Grupo de Ciencias Básicas, Facultad de Medicina , Universidad CES , Medellín , Colombia
| | - Gloria Vásquez
- a Grupo de Inmunología Celular e InmunogenéInstituto de Investigaciones Médicas, Facultad de Medicina , Universidad de Antioquia Medellín , Colombia
| |
Collapse
|
13
|
Peptides designed to spatially depict the Epstein-Barr virus major virion glycoprotein gp350 neutralization epitope elicit antibodies that block virus-neutralizing antibody 72A1 interaction with the native gp350 molecule. J Virol 2015; 89:4932-41. [PMID: 25694592 DOI: 10.1128/jvi.03269-14] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 02/10/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Epstein-Barr virus (EBV) is the etiologic agent of infectious mononucleosis and the root cause of B-cell lymphoproliferative disease in individuals with a weakened immune system, as well as a principal cofactor in nasopharyngeal carcinoma, various lymphomas, and other cancers. The EBV major virion surface glycoprotein gp350 is viewed as the best vaccine candidate to prevent infectious mononucleosis in healthy EBV-naive persons and EBV-related cancers in at-risk individuals. Previous epitope mapping of gp350 revealed only one dominant neutralizing epitope, which has been shown to be the target of the monoclonal antibody 72A1. Computer modeling of the 72A1 antibody interaction with the gp350 amino terminus was used to identify gp350 amino acids that could form strong ionic, electrostatic, or hydrogen bonds with the 72A1 antibody. Peptide DDRTTLQLAQNPVYIPETYPYIKWDN (designated peptide 2) and peptide GSAKPGNGSYFASVKTEMLGNEID (designated peptide 3) were designed to spatially represent the gp350 amino acids predicted to interact with the 72A1 antibody paratope. Peptide 2 bound to the 72A1 antibody and blocked 72A1 antibody recognition of the native gp350 molecule. Peptide 2 and peptide 3 were recognized by human IgG and shown to elicit murine antibodies that could target gp350 and block its recognition by the 72A1 antibody. This work provides a structural mapping of the interaction between the EBV-neutralizing antibody 72A1 and the major virion surface protein gp350. gp350 mimetic peptides that spatially depict the EBV-neutralizing epitope would be useful as a vaccine to focus the immune system exclusively to this important virus epitope. IMPORTANCE The production of virus-neutralizing antibodies targeting the Epstein-Barr virus (EBV) major surface glycoprotein gp350 is important for the prevention of infectious mononucleosis and EBV-related cancers. The data presented here provide the first in silico map of the gp350 interaction with a virus-blocking monoclonal antibody. Immunization with gp350 peptides identified by in silico mapping generated antibodies that cross-react with the EBV gp350 molecule and block recognition of the gp350 molecule by a virus-neutralizing antibody. Through its ability to focus the immune system exclusively on the gp350 sequence important for viral entry, these peptides may form the basis of an EBV vaccine candidate. This strategy would sidestep the production of other irrelevant gp350 antibodies that divert the immune system from generating a protective antiviral response or that impede access to the virus-blocking epitope by protective antibodies.
Collapse
|
14
|
Klinker MW, Lizzio V, Reed TJ, Fox DA, Lundy SK. Human B Cell-Derived Lymphoblastoid Cell Lines Constitutively Produce Fas Ligand and Secrete MHCII(+)FasL(+) Killer Exosomes. Front Immunol 2014; 5:144. [PMID: 24765093 PMCID: PMC3980107 DOI: 10.3389/fimmu.2014.00144] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 03/20/2014] [Indexed: 12/18/2022] Open
Abstract
Immune suppression mediated by exosomes is an emerging concept with potentially immense utility for immunotherapy in a variety of inflammatory contexts, including allogeneic transplantation. Exosomes containing the apoptosis-inducing molecule Fas ligand (FasL) have demonstrated efficacy in inhibiting antigen-specific immune responses upon adoptive transfer in animal models. We report here that a very high frequency of human B cell-derived lymphoblastoid cell lines (LCL) constitutively produce MHCII+FasL+ exosomes that can induce apoptosis in CD4+ T cells. All LCL tested for this study (>20 independent cell lines) showed robust expression of FasL, but had no detectable FasL on the cell surface. Given this intracellular sequestration, we hypothesized that FasL in LCL was retained in the secretory lysosome and secreted via exosomes. Indeed, we found both MHCII and FasL proteins present in LCL-derived exosomes, and using a bead-based exosome capture assay demonstrated the presence of MHCII+FasL+ exosomes among those secreted by LCL. Using two independent experimental approaches, we demonstrated that LCL-derived exosomes were capable of inducing antigen-specific apoptosis in autologous CD4+ T cells. These results suggest that LCL-derived exosomes may present a realistic source of immunosuppressive exosomes that could reduce or eliminate T cell-mediated responses against donor-derived antigens in transplant recipients.
Collapse
Affiliation(s)
- Matthew W Klinker
- Graduate Program in Immunology, University of Michigan , Ann Arbor, MI , USA ; Division of Rheumatology, Department of Internal Medicine, University of Michigan , Ann Arbor, MI , USA
| | - Vincent Lizzio
- Division of Rheumatology, Department of Internal Medicine, University of Michigan , Ann Arbor, MI , USA
| | - Tamra J Reed
- Division of Rheumatology, Department of Internal Medicine, University of Michigan , Ann Arbor, MI , USA
| | - David A Fox
- Graduate Program in Immunology, University of Michigan , Ann Arbor, MI , USA ; Division of Rheumatology, Department of Internal Medicine, University of Michigan , Ann Arbor, MI , USA
| | - Steven K Lundy
- Graduate Program in Immunology, University of Michigan , Ann Arbor, MI , USA ; Division of Rheumatology, Department of Internal Medicine, University of Michigan , Ann Arbor, MI , USA
| |
Collapse
|
15
|
|
16
|
|
17
|
Klinker MW, Lundy SK. Multiple mechanisms of immune suppression by B lymphocytes. Mol Med 2012; 18:123-37. [PMID: 22033729 PMCID: PMC3276396 DOI: 10.2119/molmed.2011.00333] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 10/21/2011] [Indexed: 12/20/2022] Open
Abstract
Suppression of the immune system after the resolution of infection or inflammation is an important process that limits immune-mediated pathogenesis and autoimmunity. Several mechanisms of immune suppression have received a great deal of attention in the past three decades. These include mechanisms related to suppressive cytokines, interleukin (IL)-10 and transforming growth factor (TGF)-β, produced by regulatory cells, and mechanisms related to apoptosis mediated by death ligands, Fas ligand (FasL) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), expressed by killer or cytotoxic cells. Despite many lines of evidence supporting an important role for B lymphocytes as both regulatory and killer cells in many inflammatory settings, relatively little attention has been given to understanding the biology of these cells, their relative importance or their usefulness as therapeutic targets. This review is intended to give an overview of the major mechanisms of immunosuppression used by B lymphocytes during both normal and inflammatory contexts. The more recent discoveries of expression of granzyme B, programmed death 1 ligand 2 (PD-L2) and regulatory antibody production by B cells as well as the interactions of regulatory and killer B cells with regulatory T cells, natural killer T (NKT) cells and other cell populations are discussed. In addition, new evidence on the basis of independent characterizations of regulatory and killer CD5(+) B cells point toward the concept of a multipotent suppressor B cell with seemingly high therapeutic potential.
Collapse
Affiliation(s)
- Matthew W Klinker
- Department of Internal Medicine, Division of Rheumatology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Steven K Lundy
- Department of Internal Medicine, Division of Rheumatology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| |
Collapse
|
18
|
Bockstal V, Guirnalda P, Caljon G, Goenka R, Telfer JC, Frenkel D, Radwanska M, Magez S, Black SJ. T. brucei infection reduces B lymphopoiesis in bone marrow and truncates compensatory splenic lymphopoiesis through transitional B-cell apoptosis. PLoS Pathog 2011; 7:e1002089. [PMID: 21738467 PMCID: PMC3128123 DOI: 10.1371/journal.ppat.1002089] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Accepted: 04/12/2011] [Indexed: 12/31/2022] Open
Abstract
African trypanosomes of the Trypanosoma brucei species are extracellular protozoan parasites that cause the deadly disease African trypanosomiasis in humans and contribute to the animal counterpart, Nagana. Trypanosome clearance from the bloodstream is mediated by antibodies specific for their Variant Surface Glycoprotein (VSG) coat antigens. However, T. brucei infection induces polyclonal B cell activation, B cell clonal exhaustion, sustained depletion of mature splenic Marginal Zone B (MZB) and Follicular B (FoB) cells, and destruction of the B-cell memory compartment. To determine how trypanosome infection compromises the humoral immune defense system we used a C57BL/6 T. brucei AnTat 1.1 mouse model and multicolor flow cytometry to document B cell development and maturation during infection. Our results show a more than 95% reduction in B cell precursor numbers from the CLP, pre-pro-B, pro-B, pre-B and immature B cell stages in the bone marrow. In the spleen, T. brucei induces extramedullary B lymphopoiesis as evidenced by significant increases in HSC-LMPP, CLP, pre-pro-B, pro-B and pre-B cell populations. However, final B cell maturation is abrogated by infection-induced apoptosis of transitional B cells of both the T1 and T2 populations which is not uniquely dependent on TNF-, Fas-, or prostaglandin-dependent death pathways. Results obtained from ex vivo co-cultures of living bloodstream form trypanosomes and splenocytes demonstrate that trypanosome surface coat-dependent contact with T1/2 B cells triggers their deletion. We conclude that infection-induced and possibly parasite-contact dependent deletion of transitional B cells prevents replenishment of mature B cell compartments during infection thus contributing to a loss of the host's capacity to sustain antibody responses against recurring parasitemic waves. African trypanosomiasis caused by Trypanosoma brucei species is fatal in both humans and animals and cannot be combated by vaccination because of extensive parasite antigenic variation. Effective trypanosome control and clearance from the bloodstream involves the action of antibodies specific for the parasite's highly diverse variable surface glycoprotein antigens. However, experimental infections in mice have shown that trypanosomiasis elicits a rapid process of B cell exhaustion and loss of protective antibody responses. Indeed, both marginal zone B cells, the first line of defense against blood-borne pathogens like T. brucei parasites, and follicular B cells, which are the major source for developing high-affinity antibody-producing plasma cells and memory B cells, become depleted during infection. In addition, existing B-cell memory, both against parasite antigens and non related pathogens, is destroyed early on in infection. Here, we demonstrate that during infection, B cell development is decreased in the bone marrow and early B cell development is taken over by the spleen. However, full maturation of developing B cells is abrogated by the occurrence of transitional B cell apoptosis. This impairs the replenishment of the mature marginal zone and follicular B cell pools and prevents the buildup of protective immunity against successive parasitemic waves.
Collapse
Affiliation(s)
- Viki Bockstal
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts, United States of America
- Laboratory for Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Molecular and Cellular Interactions, VIB, Brussels, Belgium
| | - Patrick Guirnalda
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts, United States of America
| | - Guy Caljon
- Laboratory for Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Molecular and Cellular Interactions, VIB, Brussels, Belgium
- Unit of Veterinary Protozoology, Institute of Tropical Medicine, Antwerp, Belgium
| | - Radhika Goenka
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts, United States of America
| | - Janice C. Telfer
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts, United States of America
| | - Deborah Frenkel
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts, United States of America
| | | | - Stefan Magez
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts, United States of America
- Laboratory for Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Molecular and Cellular Interactions, VIB, Brussels, Belgium
- * E-mail: (SJB); (SM)
| | - Samuel J. Black
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts, United States of America
- * E-mail: (SJB); (SM)
| |
Collapse
|
19
|
Lettau M, Paulsen M, Schmidt H, Janssen O. Insights into the molecular regulation of FasL (CD178) biology. Eur J Cell Biol 2010; 90:456-66. [PMID: 21126798 DOI: 10.1016/j.ejcb.2010.10.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Revised: 10/07/2010] [Accepted: 10/08/2010] [Indexed: 01/26/2023] Open
Abstract
Fas ligand (FasL, CD95L, APO-1L, CD178, TNFSF6, APT1LG1) is the key death factor of receptor-triggered programmed cell death in immune cells. FasL/Fas-dependent apoptosis plays a pivotal role in activation-induced cell death, termination of immune responses, elimination of autoreactive cells, cytotoxic effector function of T and NK cells, and the establishment of immune privilege. Deregulation or functional impairment of FasL threatens the maintenance of immune homeostasis and defense and results in severe autoimmunity. In addition, FasL has been implicated as an accessory or costimulatory receptor in T cell activation. The molecular mechanisms underlying this reverse signaling capacity are, however, poorly understood and still controversially discussed. Many aspects of FasL biology have been ascribed to selective protein-protein interactions mediated by a unique polyproline region located in the membrane-proximal intracellular part of FasL. Over the past decade, we and others identified a large number of putative FasL-interacting molecules that bind to this polyproline stretch via Src homology 3 or WW domains. Individual interactions were analyzed in more detail and turned out to be crucial for the lysosomal storage, the transport and the surface appearance of the death factor and potentially also for reverse signaling. This review summarizes the work in the framework of the Collaborative Research Consortium 415 (CRC 415) and provides facts and hypotheses about FasL-interacting proteins and their potential role in FasL biology.
Collapse
Affiliation(s)
- Marcus Lettau
- Christian-Albrechts-University, Institute of Immunology, D-24105 Kiel, Germany.
| | | | | | | |
Collapse
|
20
|
Orr MK, Burnside JS, Phillips CA, Philip R, Dombrowski KE, Wright SE. MHC-Restricted Presentation of a Single Repeat of MUC1 Mucin. Immunol Invest 2009; 36:271-83. [PMID: 17558710 DOI: 10.1080/08820130601187384] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Major histocompatibility complex (MHC) restriction of antigen presentation of a single mucin1 (MUC1) variable number of tandem repeats peptide (VNTR1) was examined by generating cytotoxic T lymphocytes (CTL) derived from peripheral blood mononuclear cells (PBMC) stimulated with a single repeat MUC1 peptide presented by allogeneic (MHC-independent) or autologous (MHC-dependent) Epstein-Barr Virus (EBV) immortalized B lymphocytes. The ability to generate greater CTL activity against MUC1-expressing tumor cells by stimulation with autologous versus allogeneic EBV-B supports the hypothesis that presentation of a single repeat of MUC1 peptide is MHC-restricted (MHC-dependent).
Collapse
Affiliation(s)
- Melody K Orr
- Department of Internal Medicine and Women's Health and Research Institute, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, USA
| | | | | | | | | | | |
Collapse
|
21
|
Lundy SK. Killer B lymphocytes: the evidence and the potential. Inflamm Res 2009; 58:345-57. [PMID: 19262989 DOI: 10.1007/s00011-009-0014-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Accepted: 12/04/2008] [Indexed: 12/12/2022] Open
Abstract
Immune regulation plays a critical role in controlling potentially dangerous inflammation and maintaining health. The Fas ligand/Fas receptor axis has been studied extensively as a mechanism of killing T cells and other cells during infections, autoimmunity, and cancer. FasL expression has been primarily attributed to activated T cells and NK cells. Evidence has emerged that B lymphocytes can express FasL and other death-inducing ligands, and can mediate cell death under many circumstances. Among B cell subsets, the expression of both Fas ligand and IL-10 is highest on the CD5(+) B cell population, suggesting that CD5(+) B cells may have a specialized regulatory function. The relevance of killer B cells to normal immune regulation, disease pathogenesis, and inflammation is discussed.
Collapse
Affiliation(s)
- Steven K Lundy
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA.
| |
Collapse
|
22
|
Miyara M, Amoura Z, Piette JC, Gorochov G. Cellules T régulatrices et lupus érythémateux systémique. Rev Med Interne 2008; 29:691-5. [DOI: 10.1016/j.revmed.2008.04.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Accepted: 04/18/2008] [Indexed: 12/13/2022]
|
23
|
Rich RF, Cook WJ, Green WR. Spontaneous in vivo retrovirus-infected T and B cells, but not dendritic cells, mediate antigen-specific Fas ligand/Fas-dependent apoptosis of anti-retroviral CTL. Virology 2005; 346:287-300. [PMID: 16337984 DOI: 10.1016/j.virol.2005.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2005] [Revised: 07/14/2005] [Accepted: 10/12/2005] [Indexed: 12/14/2022]
Abstract
C57BL/6 (H-2b), but not spontaneous virus-expressing AKR.H-2b congenic, mice generate retrovirus-specific CD8+ CTL responses to the immunodominant Kb-restricted epitope, KSPWFTTL. AKR.H-2b non-responsiveness is mediated by a peripheral tolerance mechanism. When co-cultured with primed B6 antiviral pCTL, AKR.H-2b splenocytes are recognized by the antiviral TcR as "veto" cells, which inhibit by an exquisitely virus-specific, MHC-restricted, veto cell FasL/responder T cell Fas, mediated apoptotic mechanism. Here, AKR.H-2b thymus, lymph node, and bone marrow cells are also shown to inhibit antiviral CTL generation. Purified AKR.H-2b CD4+ and CD8+ T cells, and B cells, served effectively as FasL-dependent veto cells. In contrast, AKR.H-2b dendritic cells (DC) did not efficiently veto antiviral CTL responses, despite expressing sufficient MHC class I/viral peptide complexes for TcR recognition. AKR.H-2b DC also expressed FasL mRNA and cell surface protein, albeit at a lower level than AKR.H-2b T and B cells. These findings suggest a fail-safe escape mechanism by virus-infected cells for escape from CTL-mediated immunity.
Collapse
Affiliation(s)
- Robert F Rich
- Department of Microbiology and Immunology and the Norris Cotton Cancer Center, Dartmouth Medical School, 1 Medical Center Drive, Borwell 603 West, Lebanon, New Hampshire 03756, USA
| | | | | |
Collapse
|
24
|
Bonardelle D, Benihoud K, Kiger N, Bobé P. B lymphocytes mediate Fas-dependent cytotoxicity in MRL/lpr mice. J Leukoc Biol 2005; 78:1052-9. [PMID: 16204618 DOI: 10.1189/jlb.0904536] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The Fas/Fas ligand (FasL) pathway is one of the two major effector mechanisms of T cell-mediated cytotoxicity. To prevent nonspecific killing by lymphoid cells, FasL expression on the cell surface of immune effector cells is strictly regulated. However, MRL/lpr autoimmune-prone mice massively overexpress FasL on their T lymphocytes, which render them able to kill Fas+ targets in vitro and in vivo. It is surprising that we show in the present work that B lymphocytes purified from MRL/lpr spleen cells express FasL to the same extent as T cells at the mRNA and protein level. These B cells are potent cytotoxic effectors against Fas+ but not Fas- targets. The B lymphocyte effectors were used ex vivo without any in vitro activation by B cell stimuli. Furthermore, we found that MRL/lpr B lymphocytes have the same cytotoxic potential as natural killer cells, which have been characterized as potent, Fas-mediated, cytotoxic effectors. The level of membrane-anchored FasL increases with the size of the B cell and cell-surface activation marker CD69 expression, indicating that the expression of FasL is up-regulated in parallel with the activation state of the B cell. The activated B cell population contained the major cytotoxic activity, and a minor part was associated with CD138/Syndecan-1+ plasma cells.
Collapse
|
25
|
Aksu G, Bayram N, Ulger Z, Ozturk C, Ozyurek RA, Bakiler RA, Kutukculer N. Inverse relationship between the ratio of ICAM-1 expressing lymphocytes and serum TGF-β1 concentrations in acute rheumatic fever. J Autoimmun 2005; 25:141-9. [PMID: 16023325 DOI: 10.1016/j.jaut.2005.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2005] [Revised: 05/25/2005] [Accepted: 05/25/2005] [Indexed: 11/22/2022]
Abstract
Autoimmunity in acute rheumatic fever (ARF) is triggered by group-A beta hemolytic streptococci (GAS). Although most of the recent work has focused on the major impact of lymphocytes, the exact immunopathogenesis is still unresolved. Regulation of self-tolerance in response to GAS has been investigated in various animal experiments. This study aimed to associate the ratio of lymphocytes bearing adhesion/costimulatory molecules, Bcl-2/CD95 and serum TGF-beta1 concentrations with clinical stages of ARF. Thirty-five patients were assigned according to the clinical stages. Bcl-2 expression on CD19+ and CD3+ lymphocytes was similar within patient groups and controls. CD62p expression was higher in patients with carditis. The ratio of ICAM-1 bearing lymphocytes was significantly different between patient groups and controls and was increased through acute to remission stages longitudinally. In contrast, a gradual and significant decrease in TGF-beta1 concentrations was observed longitudinally from acute to chronic stages. A positive correlation has been documented between ICAM-1+ lymphocyte ratios and Fas+ cytotoxic T cell ratios supported by a prominent increase in CD95+ T cells. These data draw our attention to the role of ICAM-1, Fas and TGF-beta1 in ARF pathogenesis through the perspective of self-tolerance in a clinical setting.
Collapse
Affiliation(s)
- Guzide Aksu
- Ege University School of Medicine, Department of Pediatric Immunology, 80. Sok. 27/11, 35100 Bornova Izmir, Turkey.
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Rabies virus (RABV) is a pathogen well-adapted to the nervous system, where it infects neurons. RABV is transmitted by the bite of an infected animal. It enters the nervous system via a motor neuron through the neuromuscular junction, or via a sensory nerve through nerve spindles. It then travels from one neuron to the next, along the spinal cord to the brain and the salivary glands. The virions are then excreted in the saliva of the animal and can be transmitted to another host by bite. Thus preservation of neuronal network integrity is crucial for the virus to be transmitted. Successful invasion of the nervous system by RABV seems to be the result of a subversive strategy based on the survival of infected neurons. This strategy includes protection against virus-mediated apoptosis and destruction of T cells that invade the CNS in response to infection.
Collapse
Affiliation(s)
- M Lafon
- Unité de Neuroimmunologie Virale, Département de Neuroscience, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris Cedex 15, France.
| |
Collapse
|
27
|
Kotsiopriftis M, Tanner JE, Alfieri C. Heat shock protein 90 expression in Epstein-Barr virus-infected B cells promotes gammadelta T-cell proliferation in vitro. J Virol 2005; 79:7255-61. [PMID: 15890964 PMCID: PMC1112101 DOI: 10.1128/jvi.79.11.7255-7261.2005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The aim of this study was to elucidate the in vitro response of gammadelta T cells to Epstein-Barr virus (EBV)-infected B cells and to determine whether EBV-induced heat shock proteins (HSPs) might serve as gammadelta T-cell stimulants. Cytofluorometric analysis revealed HSP90 cell surface expression in 12% of the EBV-immortalized B-cell population in all four of the B-cell lines tested. HSP27, HSP60, and HSP70 were not detected on the cell surface by cytofluorometry in these same B-cell lines. HSP90 and HSP60, but not HSP70 or HSP27, were detected on the cell surface after 125I cell surface labeling and immunoprecipitation with anti-human HSP monoclonal antibodies. In vitro kinetic studies indicated that gammadelta T cells increased at least twofold by day 11 postinfection in cultures of EBV-seronegative peripheral blood lymphocytes infected with EBV, whereas percentages of alphabeta T cells in these same cultures either decreased slightly or remained relatively unchanged in response to EBV infection. Addition of anti-human HSP90 monoclonal antibody to the EBV-infected lymphocyte cultures inhibited gammadelta T-cell expansion by 92%. The inhibition of gammadelta T-cell expansion by anti-HSP90 antibody was reversed upon treatment with exogenous HSP90. Taken together, these results indicate that HSP90 played an important role in the stimulation of gammadelta T cells during EBV infection of B cells in vitro and may serve as an important immunomodulator of gammadelta T cells during acute EBV infection.
Collapse
Affiliation(s)
- Maria Kotsiopriftis
- Sainte-Justine Hospital Research Center, 3175 Côte Sainte-Catherine Road, Montréal (Québec), Canada H3T 1C5
| | | | | |
Collapse
|
28
|
Petrovas C, Mueller YM, Katsikis PD. Apoptosis of HIV-specific CD8+ T cells: an HIV evasion strategy. Cell Death Differ 2005; 12 Suppl 1:859-70. [PMID: 15818412 DOI: 10.1038/sj.cdd.4401595] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- C Petrovas
- Department of Microbiology and Immunology, and Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | | | | |
Collapse
|
29
|
Abstract
Fas ligand (FasL) is a type II transmembrane protein that plays a critical role in immune homeostasis by binding to its receptor Fas (CD95) and inducing apoptosis. Fas/FasL dysregulation contributes to infectious disease pathogenesis. Microorganisms may inhibit Fas signal transduction to prolong intracellular survival and prevent killing by immune effector cells. FasL may be upregulated in directly infected cells to enhance killing of responding immune cells and facilitate immune evasion. The host response to infection may aim to induce apoptosis in directly infected cells, but immune cells that target directly infected cells can induce Fas-mediated apoptosis of uninfected bystander cells. FasL also contributes to the generation and regulation of the inflammatory response in infection. The multiple roles of FasL in infectious disease pathogenesis are discussed in the context of viral, bacterial and parasitic infections.
Collapse
Affiliation(s)
- D H Dockrell
- Division of Genomic Medicine, University of Sheffield, School of Medicine and Biomedical Sciences, Sheffield, UK.
| |
Collapse
|
30
|
Uda H, Mima T, Yamaguchi N, Katada Y, Fukuda M, Fujii N, Nakamura K, Saiki O. Expansion of a CD28-intermediate subset among CD8 T cells in patients with infectious mononucleosis. J Virol 2002; 76:6602-8. [PMID: 12050373 PMCID: PMC136291 DOI: 10.1128/jvi.76.13.6602-6608.2002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Infectious mononucleosis (IM) is an acute sporadic infection that usually affects young adults, and during infection a massive expansion of CD8 T cells is generally considered to occur. However, CD28 expression of the expanded cells has not been characterized. When peripheral blood mononuclear cells of acute IM (AIM) patients were analyzed by flow cytometry, a continuous spectrum of CD28 intensity ranging from negative to high, which could be separated into CD28 negative, intermediate (int), and positive, was seen for CD8 T cells. We studied 26 IM patients who were diagnosed on the basis of standard methods and found that all patients had the continuous CD28 spectrum. CD28 is a costimulatory molecule on T cells, and its expression is associated with the subdivision of CD8 cells into cytotoxic (CD28-positive) and suppressor (CD28-negative) T cells. After 24 h of ex vivo culturing, however, the continuous spectrum was found to consist of only CD28-positive and CD28-negative CD8 T cells, because the CD28-int cells had disappeared due to apoptosis. The CD28-int T cells have several cytotoxic functions, suggesting that CD28-int T cells are effectors. Examination of other costimulatory markers in AIM patients showed that CD80 and CD152 were not affected. In patients with other viral infections, such as measles or rubella, however, the continuous spectrum was not detected. These results suggest that there is an unusual CD28 expression pattern in patients with AIM, namely, the presence of a functional CD28-int subset among CD8 T cells. These findings are of special importance for clarifying the defense mechanism against Epstein-Barr virus infection, and the role of CD28 molecules in humans and should also be helpful for the diagnosis of AIM.
Collapse
Affiliation(s)
- Hiroshi Uda
- Department of Clinical Investigation, Osaka-Minami National Hospital, Kido-Higashi Cho 2-1, Kawachi-Nagano City, Osaka 586-8521, Japan
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Zheng BJ, Ng SP, Chua DTT, Sham JST, Kwong DLW, Lam CK, Ng MH. Peripheral gamma delta T-cell deficit in nasopharyngeal carcinoma. Int J Cancer 2002; 99:213-7. [PMID: 11979436 DOI: 10.1002/ijc.10326] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Previous studies identified CD56(+) and CD56(-) subsets of peripheral gamma delta T cells from healthy donors. Both subsets responded to stimulation by a myeloma cell line, XG-7 and undergo vigorous ex vivo expansion in the presence of exogenous IL-2. They are cytotoxic for different tumor targets including nasopharyngeal carcinoma, but they differ from one another in that the CD56(-) subset has an additional growth requirement for IL-7 and exhibited greater cytotoxicity against nasopharyngeal carcinoma (NPC) targets. These immune cells were further shown to retard tumor growth in a nude mice NPC model. To assess if these immune cells might contribute to host defense against NPC, we compared gamma delta T-cell status of NPC patients with healthy donors and survivors who had been in clinical remission of the cancer. It was found that peripheral gamma delta T cells of patients were impaired in their response to the stimulatory effects of XG-7 and exhibited weak or essentially no cytotoxicity for the NPC targets. The deficits were present in early and advanced stages of the cancer but were restored among survivors after successful treatment of the cancer. These findings support a role for peripheral gamma delta T cells in host defense against NPC. It was noted that these immune cells comprise less than 5% of peripheral blood monocytic cells and hence it was not surprising that this component of host defense was breached early in the development of the cancer.
Collapse
Affiliation(s)
- Bo Jian Zheng
- Department of Microbiology, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
32
|
Zuñiga E, Motran CC, Montes CL, Yagita H, Gruppi A. Trypanosoma cruzi infection selectively renders parasite-specific IgG+ B lymphocytes susceptible to Fas/Fas ligand-mediated fratricide. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:3965-73. [PMID: 11937553 DOI: 10.4049/jimmunol.168.8.3965] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The control of B cell expansion has been thought to be solely regulated by T lymphocytes. We show in this study that Trypanosoma cruzi infection induces up-regulation of both Fas and Fas ligand (FasL) molecules on B cells and renders them susceptible to B cell-B cell killing (referred to as fratricide throughout this paper) mediated via Fas/FasL. Moreover, by in vivo administration of anti-FasL blocking mAb we demonstrate that Fas-mediated B cell apoptosis is an ongoing process during this parasitic infection. We also provide evidence that B cells that have switched to IgG isotype are the preferential targets of B cell fratricide. More strikingly, this death pathway selectively affects IgG(+) B cells reactive to parasite but not self Ags. Parasite-specific but not self-reactive B cells triggered during this response are rescued after either in vitro or in vivo FasL blockade. Fratricide among parasite-specific IgG(+) B lymphocytes could impair the immune control of T. cruzi and possibly other chronic protozoan parasites. Our results raise the possibility that the blockade of Fas/FasL interaction in the B cell compartment of T. cruzi-infected mice may provide a means for enhancing antiparasitic humoral immune response without affecting host tolerance.
Collapse
Affiliation(s)
- Elina Zuñiga
- Department of Clinical Biochemistry, Faculty of Chemical Science, National University of Cordoba, Cordoba, Argentina
| | | | | | | | | |
Collapse
|
33
|
Goltz M, Ericsson T, Patience C, Huang CA, Noack S, Sachs DH, Ehlers B. Sequence analysis of the genome of porcine lymphotropic herpesvirus 1 and gene expression during posttransplant lymphoproliferative disease of pigs. Virology 2002; 294:383-93. [PMID: 12009880 DOI: 10.1006/viro.2002.1390] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The porcine lymphotropic herpesvirus 1 (PLHV-1), the first gammaherpesvirus of pigs, has been detected at a high prevalence in healthy pig populations. A porcine gammaherpesvirus has also been detected at high copy numbers in animals suffering from posttransplant lymphoproliferative disease (PTLD). While human PTLD is a EBV-associated complication following clinical transplantation, porcine PTLD is a disease recently described in pigs undergoing experimental allogeneic hematopoietic stem cell transplantation. Here we demonstrate that PLHV-1 and the virus present in porcine PTLD are indistinguishable, and present the characterization of 73 kbp of the genome of PLHV-1. We identified homologs of cellular genes, including a putative G protein-coupled receptor (GCR) as well as a viral homolog of the bcl-2 oncogene (v-bcl-2) and show significant transcription of these genes as well as of several other PLHV-1 genes in lymph nodes of a PTLD-affected pig. These data indicate that PLHV-1 is active during PTLD and may be involved in the etiology of this lymphoproliferative disease.
Collapse
Affiliation(s)
- Michael Goltz
- Robert Koch-Institut, Nordufer 20, Berlin, 13353, Germany.
| | | | | | | | | | | | | |
Collapse
|
34
|
Mustafa T, Bjune TG, Jonsson R, Pando RH, Nilsen R. Increased expression of fas ligand in human tuberculosis and leprosy lesions: a potential novel mechanism of immune evasion in mycobacterial infection. Scand J Immunol 2001; 54:630-9. [PMID: 11902340 DOI: 10.1046/j.1365-3083.2001.01020.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
To study the location and mechanism of apoptosis within the human tuberculosis (TB) and leprosy lesions, parallel sections were analyzed for mycobacterial antigens (M.Ag), Fas ligand (FasL), Fas, CD68 and Mac387 by immunohistochemistry, and apoptotic cells by the terminal deoxynucleotidyl-transferase-mediated dUTP-digoxigenin nick end labelling method. Cutaneous leishmaniasis and foreign body granulomas were analyzed for comparison. The heavily infected macrophages in multibacillary TB and leprosy granulomas very strongly expressed FasL, indicating that a mycobacterial infection can induce an increased expression of FasL in a population of infected macrophages, which may protect them from the attack of Fas-expressing lymphocytes. However, macrophages with high levels of leishmania amastigotes did not selectively express FasL, suggesting that this phenomenon is specific for the mycobacteria. Interestingly, in the well-formed TB granulomas, 84% of the multinucleated giant cells strongly expressed FasL. The expression of Fas was weak (34%) or absent. A higher number (33%) of epithelioid cells expressed FasL than Fas (23%). Lymphocytes were scanty among the epithelioid cells. The frequency of apoptotic cells was higher in the epithelioid cells (0.25%) than the mononuclear cells in the mantle zone (0.14%). Thus, the epithelioid cells and the multinucleated giant cells by virtue of the increased expression of FasL may make these granulomas an immune privileged site for mycobacteria.
Collapse
Affiliation(s)
- T Mustafa
- Centre for International Health, Department of Odontology, University of Bergen, Norway.
| | | | | | | | | |
Collapse
|
35
|
Mueller YM, De Rosa SC, Hutton JA, Witek J, Roederer M, Altman JD, Katsikis PD. Increased CD95/Fas-induced apoptosis of HIV-specific CD8(+) T cells. Immunity 2001; 15:871-82. [PMID: 11754810 DOI: 10.1016/s1074-7613(01)00246-1] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Why HIV-specific CD8(+) T cells ultimately fail to clear or control HIV infection is not known. We show here that HIV-specific CD8(+) T cells exhibit increased sensitivity to CD95/Fas-induced apoptosis. This apoptosis is 3-fold higher compared to CMV-specific CD8(+) T cells from the same patients. HIV-specific CD8(+) T cells express the CD45RA(-)CD62L(-) but lack the CD45RA(+)CD62L(-) T cell effector memory (T(EM)) phenotype. This skewing is not found in CMV- and EBV-specific CD8(+) T cells in HIV-infected individuals. CD95/Fas-induced apoptosis is much higher in the CD45RA(-)CD62L(-) T(EM) cells. However, cytotoxicity and IFNgamma production by HIV-specific CD8(+) T cells is not impaired. Our data suggest that the survival and differentiation of HIV-specific CD8(+) T cells may be compromised by CD95/Fas apoptosis induced by FasL-expressing HIV-infected cells.
Collapse
Affiliation(s)
- Y M Mueller
- Department of Microbiology and Immunology, MCP Hahnemann University, Philadelphia, Pennsylvania 19129, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Kin Y, Shibuya M, Maru Y. A suppressive effect of an Epstein-Barr virus-immortalized cell line on leukemic cells. Clin Biochem 2001; 34:507-10. [PMID: 11676982 DOI: 10.1016/s0009-9120(01)00240-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
MESH Headings
- Aged
- Antigens, CD/biosynthesis
- Antigens, Differentiation/biosynthesis
- Blotting, Western
- Cell Division
- Cell Line
- Chromosome Aberrations
- Coculture Techniques
- Fatal Outcome
- Female
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Philadelphia Chromosome
- Precipitin Tests
- Reverse Transcriptase Polymerase Chain Reaction
- Vincristine/therapeutic use
Collapse
Affiliation(s)
- Y Kin
- Department of Genetics, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108, Japan
| | | | | |
Collapse
|
37
|
Tanner JE, Alfieri C. The Epstein-Barr virus and post-transplant lymphoproliferative disease: interplay of immunosuppression, EBV, and the immune system in disease pathogenesis. Transpl Infect Dis 2001; 3:60-9. [PMID: 11395971 DOI: 10.1034/j.1399-3062.2001.003002060.x] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Transplant patients are at particular risk for developing post-transplant lymphoproliferative disease (PTLD) following administration of immunosuppressive therapy. In many cases the PTLD lesions express Epstein-Barr virus (EBV) latent and lytic genes as well as elevated levels of host cytokines. An outline of the potential contributions of EBV, host cytokines and T cells, and the immunosuppressive cyclosporine A, tacrolimus, and anti-CD3 antibody in the mechanism and pathogenesis of this disease is presented and discussed.
Collapse
Affiliation(s)
- J E Tanner
- Department of Pediatrics, Children's Hospital of Eastern Ontario, University of Ottawa Medical School, Ottawa, Ontario, Canada
| | | |
Collapse
|
38
|
Abstract
Apoptosis is a physiological process critical for tissue homeostasis. It is essential for the regulation of immune responses. A series of molecules transduce apoptoic signals and induce the characteristic morphological appearances of apoptotic cells. Infectious diseases modulate apoptosis and this contributes to disease pathogenesis. Infection with HIV results in enhanced levels of CD4 T-lymphocyte apoptosis in both directly infected cells and in uninfected bystander cells. A variety of HIV proteins including gp120 contribute to this process. A number of different pathways induce HIV-associated CD4 T-lymphocyte apoptosis and apoptosis of uninfected bystander cells is particularly associated with increased susceptibility to Fas. Other viruses including hepatitis viruses and the human herpesviruses also modulate apoptosis. Bacterial infection induces apoptosis which is frequently mediated by the direct activation of caspases in the absence of death receptor ligation. Bacterial induction of apoptosis may either be due to bacterial factors such as the invasin IpaB of Shigella flexneri or be the result of host immune responses which control infection as demonstrated in infections due to Mycobacterium spp. Apoptosis may be modulated by therapeutic strategies, such as antiretroviral therapy, and an improved understanding of infection-associated apoptosis modulation will aid the design of novel therapeutic approaches to control infectious diseases.
Collapse
Affiliation(s)
- D H Dockrell
- Division of Genomic Medicine, University of Sheffield Medical School, Department of Infectious Diseases, Royal Hallamshire Hospital, Sheffield, UK.
| |
Collapse
|
39
|
Khorana AA, Rosenblatt JD, Young FM. Immunopathogenesis of HIV and HTLV-1 infection: mechanisms for lymphomagenesis. Cancer Treat Res 2001; 104:19-74. [PMID: 11191127 DOI: 10.1007/978-1-4615-1601-9_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2023]
Affiliation(s)
- A A Khorana
- Cancer Center and Hematology-Oncology Unit, University of Rochester Medical Center, Rochester, New York, USA
| | | | | |
Collapse
|
40
|
Lundy SK, Lerman SP, Boros DL. Soluble egg antigen-stimulated T helper lymphocyte apoptosis and evidence for cell death mediated by FasL(+) T and B cells during murine Schistosoma mansoni infection. Infect Immun 2001; 69:271-80. [PMID: 11119515 PMCID: PMC97881 DOI: 10.1128/iai.69.1.271-280.2001] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Granuloma formation around schistosomal eggs is induced by soluble egg antigens (SEA) and mediated by the activity of CD4(+) Th lymphocytes and their cytokines. Regulation of the inflammatory Th cell response during infection is still insufficiently understood. The hypothesis of this study was that activation-induced cell death (AICD) of CD4(+) T cells is involved in the immune inflammatory response. This study investigated the dynamics of splenic and granuloma CD4(+) Th cell apoptosis and Fas ligand (FasL) expression during the acute and chronic stages of murine schistosomal infection. Enhanced apoptosis of freshly isolated CD4(+) Th lymphocytes commenced after egg deposition and persisted during the peak and modulated phases of granuloma formation. After oviposition, CD4(+), CD8(+), and CD19(+) splenocytes and granuloma cells expressed elevated levels of FasL but FasL expression declined during the downmodulated stage of infection. In culture, SEA induced splenic and granuloma CD4(+) T-cell apoptosis and stimulated expression of FasL on splenic but not granuloma CD4(+) T cells, CD8(+) T cells, and CD19(+) B cells. SEA-stimulated splenocytes and granuloma cells preferentially lysed a Fas-transfected target cell line. Depletion of B cells from SEA-stimulated splenic cultures decreased CD4(+) T cell apoptosis. Coculture of purified splenic B cells with CD4(+) T cells and adoptive transfer of purified B cells indicated that antigen-stimulated B cells can kill CD4(+) Th cells. However, CD4(+) T cells were the dominant mediators of apoptosis in the granuloma. This study indicates that AICD is involved in the apoptosis of CD4(+) T cells during schistosomal infection.
Collapse
Affiliation(s)
- S K Lundy
- Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | | | |
Collapse
|
41
|
Sasaki Y, Ami Y, Nakasone T, Shinohara K, Takahashi E, Ando S, Someya K, Suzaki Y, Honda M. Induction of CD95 ligand expression on T lymphocytes and B lymphocytes and its contribution to apoptosis of CD95-up-regulated CD4+ T lymphocytes in macaques by infection with a pathogenic simian/human immunodeficiency virus. Clin Exp Immunol 2000; 122:381-9. [PMID: 11122244 PMCID: PMC1905812 DOI: 10.1046/j.1365-2249.2000.01327.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Using an established SIV/HIV-C2/1-infected cynomolgus monkey model displaying stable CD4+ T cell depletion, the kinetics of apoptosis and the levels of expression of CD95 membrane-associated CD95L on lymphocytes were investigated to test the involvement of the CD95/CD95L system in CD4+ T lymphocyte loss in vivo. Rapid depletion of CD4+ T cells occurred up to 2 weeks after infection, with chronic CD4+ T lymphopenia thereafter. During the initial CD4+ T cell loss, which was accompanied by viraemia, about 90% of the peripheral CD4+ T cell subset underwent spontaneous apoptotic cell death during 24 h of culture. Increased expression of CD95 was observed on both CD4+ and CD8+ T cell subsets, with CD95 expression on CD8+ cells declining rapidly, but high CD95 expression being maintained on CD4+ cells. Since CD95L was expressed on CD8+ T cells, B cells and to a lesser extent on CD4+ T cells, this suggests that CD95-mediated apoptosis might be controlled in an autocrine/paracrine fashion.
Collapse
Affiliation(s)
- Y Sasaki
- Department of Safety Research on Biologics, National Institute of Infectious Diseases, Tokyo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Cinatl J, Blaheta R, Bittoova M, Scholz M, Margraf S, Vogel JU, Cinatl J, Doerr HW. Decreased neutrophil adhesion to human cytomegalovirus-infected retinal pigment epithelial cells is mediated by virus-induced up-regulation of Fas ligand independent of neutrophil apoptosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:4405-13. [PMID: 11035078 DOI: 10.4049/jimmunol.165.8.4405] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Human CMV (HCMV) retinitis frequently leads to blindness in iatrogenically immunosuppressed patients and in the end stage of AIDS. Despite the general proinflammatory potential of HCMV, virus infection is associated with a rather mild cellular inflammatory response in the retina. To investigate this phenomenon, the influence of HCMV (strains AD169 or Hi91) infection on C-X-C chemokine secretion, ICAM-1 expression, and neutrophil recruitment in cultured human retinal pigment epithelial (RPE) cells was studied. Supernatants from infected cultures contained enhanced levels of IL-8 and melanoma growth-stimulating activity/Gro alpha and induced neutrophil chemotaxis compared with supernatants from uninfected RPE cells. Despite HCMV-induced ICAM-1 expression on RPE cells, binding of activated neutrophils to HCMV-infected RPE cells and subsequent transepithelial penetration were significantly reduced. Reduced neutrophil adhesion to infected RPE cells correlated with HCMV-induced up-regulation of constitutive Fas ligand (FasL) expression. Functional blocking of FasL on RPE cells with the neutralizing mAbs NOK-1 and NOK-2 or of the Fas receptor on neutrophils with mAbB-D29 prevented the HCMV-induced impairment of neutrophil/RPE interactions. Fas-FasL-dependent impairment of neutrophil binding had occurred by 10 min after neutrophil/RPE coculture without apoptotic signs. Neutrophil apoptosis was first detected after 4 h. Treatment of neutrophils with a specific inhibitor of caspase-8 suppressed apoptosis, whereas it did not prevent impaired neutrophil binding to infected RPE. The current results suggest a novel role for FasL in the RPE regulation of neutrophil binding. This may be an important feature of virus escape mechanisms and for sustaining the immune-privileged character of the retina during HCMV ocular infection.
Collapse
Affiliation(s)
- J Cinatl
- Zentrum der Hygiene, Institut für Medizinische Virologie, Klinikum der Johann Wolfgang Goethe Universität, Frankfurt am Main, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
|