1
|
Toh J, Reitsma AJ, Tajima T, Younes SF, Ezeiruaku C, Jenkins KC, Peña JK, Zhao S, Wang X, Lee EYZ, Glass MC, Kalesinskas L, Ganesan A, Liang I, Pai JA, Harden JT, Vallania F, Vizcarra EA, Bhagat G, Craig FE, Swerdlow SH, Morscio J, Dierickx D, Tousseyn T, Satpathy AT, Krams SM, Natkunam Y, Khatri P, Martinez OM. Multi-modal analysis reveals tumor and immune features distinguishing EBV-positive and EBV-negative post-transplant lymphoproliferative disorders. Cell Rep Med 2024; 5:101851. [PMID: 39657667 PMCID: PMC11722118 DOI: 10.1016/j.xcrm.2024.101851] [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: 07/31/2023] [Revised: 03/09/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024]
Abstract
The oncogenic Epstein-Barr virus (EBV) can drive tumorigenesis with disrupted host immunity, causing malignancies including post-transplant lymphoproliferative disorders (PTLDs). PTLD can also arise in the absence of EBV, but the biological differences underlying EBV(+) and EBV(-) B cell PTLD and the associated host-EBV-tumor interactions remain poorly understood. Here, we reveal the core differences between EBV(+) and EBV(-) PTLD, characterized by increased expression of genes related to immune processes or DNA interactions, respectively, and the augmented ability of EBV(+) PTLD B cells to modulate the tumor microenvironment through elaboration of monocyte-attracting cytokines/chemokines. We create a reference resource of proteins distinguishing EBV(+) B lymphoma cells from EBV(-) B lymphoma including the immunomodulatory molecules CD300a and CD24, respectively. Moreover, we show that CD300a is essential for maximal survival of EBV(+) PTLD B lymphoma cells. Our comprehensive multi-modal analyses uncover the biological underpinnings of PTLD and offer opportunities for precision therapies.
Collapse
Affiliation(s)
- Jiaying Toh
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA; PhD Program in Immunology, Stanford University School of Medicine, Stanford, CA, USA; Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA; Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Andrea J Reitsma
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| | - Tetsuya Tajima
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| | - Sheren F Younes
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Chimere Ezeiruaku
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| | - Kayla C Jenkins
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| | - Josselyn K Peña
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA; PhD Program in Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Shuchun Zhao
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Xi Wang
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| | - Esmond Y Z Lee
- PhD Program in Stem Cell and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Marla C Glass
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| | - Laurynas Kalesinskas
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA; Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA, USA; PhD Program in Biomedical Informatics, Stanford University School of Medicine, Stanford, CA, USA
| | - Ananthakrishnan Ganesan
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA; Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA, USA; Institute for Computational and Mathematical Engineering, School of Engineering, Stanford University, Stanford, CA, USA
| | - Irene Liang
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| | - Joy A Pai
- PhD Program in Immunology, Stanford University School of Medicine, Stanford, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - James T Harden
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA; PhD Program in Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Francesco Vallania
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA; Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Edward A Vizcarra
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| | - Govind Bhagat
- Department of Pathology, Columbia University, New York, NY, USA
| | - Fiona E Craig
- Laboratory of Medicine and Pathology, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Steven H Swerdlow
- Division of Hematopathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Julie Morscio
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven, Leuven, Belgium
| | - Daan Dierickx
- Department of Hematology, University Hospitals Leuven, and the Laboratory for Experimental Hematology, Department of Oncology, University of Leuven, Leuven, Belgium
| | - Thomas Tousseyn
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven, Leuven, Belgium; Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Ansuman T Satpathy
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Stanford Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sheri M Krams
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA; Stanford Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yasodha Natkunam
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Purvesh Khatri
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA; Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA, USA; Stanford Immunology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Olivia M Martinez
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA; Stanford Immunology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
2
|
Althaus J, Nilius-Eliliwi V, Maghnouj A, Döring S, Schroers R, Hudecek M, Hahn SA, Mika T. Cytotoxicity of CD19-CAR-NK92 cells is primarily mediated via perforin/granzyme pathway. Cancer Immunol Immunother 2023:10.1007/s00262-023-03443-1. [PMID: 37052701 PMCID: PMC10361870 DOI: 10.1007/s00262-023-03443-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 03/28/2023] [Indexed: 04/14/2023]
Abstract
Chimeric antigen receptors (CARs) have improved cancer immunotherapy in recent years. Immune cells, such as Natural killer cells (NK-cells) or T cells, are used as effector cells in CAR-therapy. NK92-cells, a cell line with known cytotoxic activity, are of particular interest in CAR-therapy since culturing conditions are simple and anti-tumor efficacy combined with a manageable safety profile was proven in clinical trials. The major pathways of immune effector cells, including NK92-cells, to mediate cytotoxicity, are the perforin/granzyme and the death-receptor pathway. Detailed knowledge of CAR-effector cells' cytotoxic mechanisms is essential to unravel resistance mechanisms, which potentially arise by resistance against apoptosis-inducing signaling. Since mutations in apoptosis pathways are frequent in lymphoma, the impact on CAR-mediated cytotoxicity is of clinical interest. In this study, knockout models of CD19-CAR-NK92 cells were designed, to investigate cytotoxic pathways in vitro. Knockout of perforin 1 (Prf1) and subsequent abrogation of the perforin/granzyme pathway dramatically reduced the cytotoxicity of CD19-CAR-NK92 cells. In contrast, knockout of FasL and inhibition of TRAIL (tumor necrosis factor-related apoptosis-inducing ligands) did not impair cytotoxicity in most conditions. In conclusion, these results indicate the perforin/granzyme pathway as the major pathway to mediate cytotoxicity in CD19-CAR-NK92 cells.
Collapse
Affiliation(s)
- Jonas Althaus
- Department of Molecular Gastrointestinal Oncology, Ruhr University Bochum, Bochum, Germany
| | - Verena Nilius-Eliliwi
- Department of Molecular Gastrointestinal Oncology, Ruhr University Bochum, Bochum, Germany
- Department of Medicine, Hematology and Oncology, Knappschaftskrankenhaus Bochum, Ruhr University Bochum, In der Schornau 23-25, D-44892, Bochum, Germany
| | - Abdelouahid Maghnouj
- Department of Molecular Gastrointestinal Oncology, Ruhr University Bochum, Bochum, Germany
| | - Sascha Döring
- Department of Molecular Gastrointestinal Oncology, Ruhr University Bochum, Bochum, Germany
| | - Roland Schroers
- Department of Medicine, Hematology and Oncology, Knappschaftskrankenhaus Bochum, Ruhr University Bochum, In der Schornau 23-25, D-44892, Bochum, Germany
| | - Michael Hudecek
- Department of Internal Medicine 2, University Hospital of Würzburg, Würzburg, Germany
| | - Stephan A Hahn
- Department of Molecular Gastrointestinal Oncology, Ruhr University Bochum, Bochum, Germany
| | - Thomas Mika
- Department of Molecular Gastrointestinal Oncology, Ruhr University Bochum, Bochum, Germany.
- Department of Medicine, Hematology and Oncology, Knappschaftskrankenhaus Bochum, Ruhr University Bochum, In der Schornau 23-25, D-44892, Bochum, Germany.
| |
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: 34] [Impact Index Per Article: 8.5] [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
|
Chang LY, Li Y, Kaplan DE. Endotoxemia contributes to CD27+ memory B-cell apoptosis via enhanced sensitivity to Fas ligation in patients with Cirrhosis. Sci Rep 2016; 6:36862. [PMID: 27857173 PMCID: PMC5114671 DOI: 10.1038/srep36862] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 10/20/2016] [Indexed: 12/25/2022] Open
Abstract
Peripheral CD27+ memory B-cells become quantitatively reduced and dysfunctional in patients with cirrhosis through poorly characterized mechanisms. We hypothesized that the disappearance of CD27+ memory B-cells results from enhanced sensitivity to apoptosis caused by exposure to gut microbial translocation products. Using isolated naïve and memory B-cells from patients with cirrhosis and age-matched controls, ex vivo and activation-induced sensitivity to Fas-mediated apoptosis was assessed under relevant experimental conditions. We observed differential expression of CD95(Fas) in CD27+ B-cells from cirrhotic patients that was inversely correlated with peripheral CD27+ B-cell frequency. While memory B-cells from cirrhotic patients were resistant to Fas-mediated apoptosis ex vivo, Toll-like receptor 4(TLR4)-ligation restored Fas-sensitivity. Sensitivity to Fas-mediated apoptosis could be transferred to healthy donor memory B-cells by co-culturing these cells with plasma from cirrhotic patients, a sensitivity partially mediated by Fas and TLR4 signaling, and partially rescued via B-cell receptor crosslinking. We conclude that peripheral CD27+ memory B-cells in cirrhosis exhibit increased sensitivity to Fas-induced apoptosis in an activation-dependent manner to which endotoxin contributes, associated with reduced frequency of circulating memory B-cells. Destruction of this critical cell subset may contribute to the cirrhotic immunodeficiency state and heightened risk of systemic infections in advanced liver disease.
Collapse
Affiliation(s)
- Li-Yuan Chang
- Medicine and Research Services, Corporal Michael J. Crescenz VA Medical Center, 3900 Woodland Avenue, Philadelphia, PA 19104, USA.,Division of Gastroenterology, Department of Medicine, University of Pennsylvania, 9th floor BRB, 421 Curie Blvd, Philadelphia, PA 19104, USA
| | - Yonghai Li
- Medicine and Research Services, Corporal Michael J. Crescenz VA Medical Center, 3900 Woodland Avenue, Philadelphia, PA 19104, USA.,Division of Gastroenterology, Department of Medicine, University of Pennsylvania, 9th floor BRB, 421 Curie Blvd, Philadelphia, PA 19104, USA
| | - David E Kaplan
- Medicine and Research Services, Corporal Michael J. Crescenz VA Medical Center, 3900 Woodland Avenue, Philadelphia, PA 19104, USA.,Division of Gastroenterology, Department of Medicine, University of Pennsylvania, 9th floor BRB, 421 Curie Blvd, Philadelphia, PA 19104, USA
| |
Collapse
|
5
|
CD95 Signaling Inhibits B Cell Receptor-Mediated Gammaherpesvirus Replication in Apoptosis-Resistant B Lymphoma Cells. J Virol 2016; 90:9782-9796. [PMID: 27558422 DOI: 10.1128/jvi.00668-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 08/13/2016] [Indexed: 11/20/2022] Open
Abstract
While CD95 is an apoptosis-inducing receptor and has emerged as a potential anticancer therapy target, mounting evidence shows that CD95 is also emerging as a tumor promoter by activating nonapoptotic signaling pathways. Gammaherpesviral infection is closely associated with lymphoproliferative diseases, including B cell lymphomas. The nonapoptotic function of CD95 in gammaherpesvirus-associated lymphomas is largely unknown. Here, we show that stimulation of CD95 agonist antibody drives the majority of sensitive gammaherpesvirus-transformed B cells to undergo caspase-dependent apoptosis and promotes the survival and proliferation of a subpopulation of apoptosis-resistant B cells. Surprisingly, CD95-mediated nonapoptotic signaling induced beta interferon (IFN-β) expression and correlatively inhibited B cell receptor (BCR)-mediated gammaherpesviral replication in the apoptosis-resistant lymphoma cells without influencing BCR signaling. Further analysis showed that IFN-β alone or synergizing with CD95 blocked the activation of lytic switch proteins and the gene expression of gammaherpesviruses. Our findings indicate that, independent of its apoptotic activity, CD95 signaling activity plays an important role in blocking viral replication in apoptosis-resistant, gammaherpesvirus-associated B lymphoma cells, suggesting a novel mechanism that indicates how host CD95 prototype death receptor controls the life cycle of gammaherpesviruses independent of its apoptotic activity. IMPORTANCE Gammaherpesviruses are closely associated with lymphoid malignancies and other cancers. Viral replication and persistence strategies leading to cancer involve the activation of antiapoptotic and proliferation programs, as well as evasion of the host immune response. Here, we provide evidence that the stimulation of CD95 agonist antibody, mimicking one of the major mechanisms of cytotoxic T cell killing, inhibits B cell receptor-mediated gammaherpesviral replication in CD95 apoptosis-resistant lymphoma cells. CD95-induced type I interferon (IFN-β) contributes to the inhibition of gammaherpesviral replication. This finding sheds new light on the CD95 nonapoptotic function and provides a novel mechanism for gammaherpesviruses that helps them to escape host immune surveillance.
Collapse
|
6
|
Park GB, Kim D, Park SJ, Lee HK, Kim JH, Kim YS, Park SG, Choi IH, Yoon SH, Lee YJ, Paeng S, Hur DY. Pre-stimulation of CD81 expression by resting B cells increases proliferation following EBV infection, but the overexpression of CD81 induces the apoptosis of EBV-transformed B cells. Int J Mol Med 2015; 36:1464-78. [PMID: 26498453 PMCID: PMC4678167 DOI: 10.3892/ijmm.2015.2372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 09/23/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C virus (HCV) E2 protein binds to CD81, which is a component of the B cell co-stimulatory complex. The E2-CD81 interaction leads to B cell proliferation, protein tyrosine phosphorylation and to the hypermutation of immunoglobulin genes. Epidemiological studies have reported a high prevalence of B cell non-Hodgkin lymphoma (NHL) in HCV-positive patients, suggesting a potential association between HCV and Epstein-Barr virus (EBV) in the genesis of B lymphocyte proliferative disorders. In the present study, in order to investigate the association between EBV and HCV in B cells, we created an in vitro EBV-induced B cell transformation model. CD81 was gradually overexpressed during transformation by EBV. B cells isolated from HCV-positive patients grew more rapidly and clumped together earlier than B cells isolated from healthy donors following EBV infection. Pre-stimulation of CD81 expressed by resting B cells with anti-CD81 monoclonal antibody (mAb) or HCV E2 accelerated the generation of lymphoblastoid cell lines (LCLs) by EBV infection. These cells proliferated prominently through the early expression of interleukin-10 and intracellular latent membrane protein (LMP)-l. By contrast, the overexpression of CD81 on EBV-transformed B cells by anti-CD81 mAb or HCV E2 protein induced apoptosis through reactive oxygen species (ROS)-mediated mitochondrial dysfunction. These results suggest that the engagement of CD81 expressed by B cells has differential effects on B cell fate (proliferation or apoptosis) according to EBV infection and the expression level of CD81.
Collapse
Affiliation(s)
- Ga Bin Park
- Department of Anatomy and Research Center for Tumor Immunology, Inje University College of Medicine, Busan 614-735, Republic of Korea
| | - Daejin Kim
- Department of Anatomy and Research Center for Tumor Immunology, Inje University College of Medicine, Busan 614-735, Republic of Korea
| | - Sung Jae Park
- Department of Internal Medicine, Inje University Busan Paik Hospital, Busan 614-735, Republic of Korea
| | - Hyun-Kyung Lee
- Department of Internal Medicine, Inje University Busan Paik Hospital, Busan 614-735, Republic of Korea
| | - Ji Hyun Kim
- Department of Internal Medicine, Inje University Busan Paik Hospital, Busan 614-735, Republic of Korea
| | - Yeong Seok Kim
- Department of Anatomy and Research Center for Tumor Immunology, Inje University College of Medicine, Busan 614-735, Republic of Korea
| | - Sae-Gwang Park
- Department of Microbiology, Inje University College of Medicine, Busan 614-735, Republic of Korea
| | - In-Hak Choi
- Department of Microbiology, Inje University College of Medicine, Busan 614-735, Republic of Korea
| | - Sung Ho Yoon
- Department of Plastic Surgery, Inje University Haeundae Paik Hospital, Busan 614-735, Republic of Korea
| | - Youn Jae Lee
- Department of Internal Medicine, Inje University Busan Paik Hospital, Busan 614-735, Republic of Korea
| | - Sunghwa Paeng
- Department of Neurosurgery, Inje University Busan Paik Hospital, Busan 614-735, Republic of Korea
| | - Dae Young Hur
- Department of Anatomy and Research Center for Tumor Immunology, Inje University College of Medicine, Busan 614-735, Republic of Korea
| |
Collapse
|
7
|
Hatton OL, Harris-Arnold A, Schaffert S, Krams SM, Martinez OM. The interplay between Epstein-Barr virus and B lymphocytes: implications for infection, immunity, and disease. Immunol Res 2015; 58:268-76. [PMID: 24619311 DOI: 10.1007/s12026-014-8496-1] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human B cells are the primary targets of Epstein-Barr virus (EBV) infection. In most cases, EBV infection is asymptomatic because of a highly effective host immune response, but some individuals develop self-limiting infectious mononucleosis, while others develop EBV-associated lymphoid or epithelial malignancies. The viral and immune factors that determine the outcome of infection are not understood. The EBV life cycle includes a lytic phase, culminating in the production of new viral particles, and a latent phase, during which the virus remains largely silent for the lifetime of the host in memory B cells. Thus, in healthy individuals, there is a tightly orchestrated interplay between EBV and the host that allows the virus to persist. To promote viral persistence, EBV has evolved a variety of strategies to modulate the host immune response including inhibition of immune cell function, blunting of apoptotic pathways, and interfering with antigen processing and presentation pathways. In this article, we focus on mechanisms by which dysregulation of the host B cell and immune modulation by the virus can contribute to development of EBV+ B cell lymphomas.
Collapse
Affiliation(s)
- Olivia L Hatton
- Program in Immunology and Department of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | | | | | | | | |
Collapse
|
8
|
Berkova Z, Wang S, Ao X, Wise JF, Braun FK, Rezaeian AH, Sehgal L, Goldenberg DM, Samaniego F. CD74 interferes with the expression of fas receptor on the surface of lymphoma cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014; 33:80. [PMID: 25304249 PMCID: PMC4210479 DOI: 10.1186/s13046-014-0080-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 09/22/2014] [Indexed: 01/21/2023]
Abstract
BACKGROUND Resistance to Fas-mediated apoptosis limits the efficacy of currently available chemotherapy regimens. We identified CD74, which is known to be overexpressed in hematological malignancies, as one of the factors interfering with Fas-mediated apoptosis. METHODS CD74 expression was suppressed in human B-lymphoma cell lines, BJAB and Raji, by either transduction with lentivirus particles or transfection with episomal vector, both encoding CD74-specific shRNAs or non-target shRNA. Effect of CD74 expression on Fas signaling was evaluated by comparing survival of mice hydrodynamically transfected with vector encoding full-length CD74 or empty vector. Sensitivity of cells with suppressed CD74 expression to FasL, edelfosine, doxorubicin, and a humanized CD74-specific antibody, milatuzumab, was evaluated by flow cytometry and compared to control cells. Fas signaling in response to FasL stimulation and the expression of Fas signaling components were evaluated by Western blot. Surface expression of Fas was detected by flow cytometry. RESULTS We determined that cells with suppressed CD74 are more sensitive to FasL-induced apoptosis and Fas signaling-dependent chemotherapies, edelfosine and doxorubicin, than control CD74-expressing cells. On the other hand, expression of full-length CD74 in livers protected the mice from a lethal challenge with agonistic anti-Fas antibody Jo2. A detailed analysis of Fas signaling in cells lacking CD74 and control cells revealed increased cleavage/activation of pro-caspase-8 and corresponding enhancement of caspase-3 activation in the absence of CD74, suggesting that CD74 affects the immediate early steps in Fas signaling at the plasma membrane. Cells with suppressed CD74 expression showed increased staining of Fas receptor on their surface. Pre-treatment with milatuzumab sensitized BJAB cells to Fas-mediated apoptosis. CONCLUSION We anticipate that specific targeting of the CD74 on the cell surface will sensitize CD74-expressing cancer cells to Fas-mediated apoptosis, and thus will increase effectiveness of chemotherapy regimens for hematological malignancies.
Collapse
Affiliation(s)
- Zuzana Berkova
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| | - Shu Wang
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| | - Xue Ao
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| | - Jillian F Wise
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| | - Frank K Braun
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| | - Abdol H Rezaeian
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| | - Lalit Sehgal
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| | - David M Goldenberg
- Immunomedics, Inc., Morris Plains, NJ, 07950, USA. .,Center for Molecular Medicine and Immunology, Garden State Cancer Center, Morris Plains, NJ, 07950, USA.
| | - Felipe Samaniego
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| |
Collapse
|
9
|
PARK GABIN, KIM YEONGSEOK, LEE HYUNKYUNG, CHO DAEHO, KIM DAEJIN, HUR DAEYOUNG. CD80 (B7.1) and CD86 (B7.2) induce EBV-transformed B cell apoptosis through the Fas/FasL pathway. Int J Oncol 2013; 43:1531-40. [DOI: 10.3892/ijo.2013.2091] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Accepted: 08/16/2013] [Indexed: 11/06/2022] Open
|
10
|
Hatton O, Lambert SL, Phillips LK, Vaysberg M, Natkunam Y, Esquivel CO, Krams SM, Martinez OM. Syk-induced phosphatidylinositol-3-kinase activation in Epstein-Barr virus posttransplant lymphoproliferative disorder. Am J Transplant 2013; 13:883-890. [PMID: 23398911 PMCID: PMC4008139 DOI: 10.1111/ajt.12137] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 11/19/2012] [Accepted: 12/06/2012] [Indexed: 01/25/2023]
Abstract
Posttransplant lymphoproliferative disorder (PTLD)-associated Epstein-Barr virus (EBV)+ B cell lymphomas are serious complications of solid organ and bone marrow transplantation. The EBV protein LMP2a, a B cell receptor (BCR) mimic, provides survival signals to virally infected cells through Syk tyrosine kinase. Therefore, we explored whether Syk inhibition is a viable therapeutic strategy for EBV-associated PTLD. We have shown that R406, the active metabolite of the Syk inhibitor fostamatinib, induces apoptosis and cell cycle arrest while decreasing downstream phosphatidylinositol-3'-kinase (PI3K)/Akt signaling in EBV+ B cell lymphoma PTLD lines in vitro. However, Syk inhibition did not inhibit or delay the in vivo growth of solid tumors established from EBV-infected B cell lines. Instead, we observed tumor growth in adjacent inguinal lymph nodes exclusively in fostamatinib-treated animals. In contrast, direct inhibition of PI3K/Akt significantly reduced tumor burden in a xenogeneic mouse model of PTLD without evidence of tumor growth in adjacent inguinal lymph nodes. Taken together, our data indicate that Syk activates PI3K/Akt signaling which is required for survival of EBV+ B cell lymphomas. PI3K/Akt signaling may be a promising therapeutic target for PTLD, and other EBV-associated malignancies.
Collapse
Affiliation(s)
- O. Hatton
- Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Surgery / Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - S. L. Lambert
- Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Surgery / Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - L. K. Phillips
- Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Surgery / Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - M. Vaysberg
- Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Surgery / Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Y. Natkunam
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - C. O. Esquivel
- Department of Surgery / Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - S. M. Krams
- Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Surgery / Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - O. M. Martinez
- Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA,Department of Surgery / Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
11
|
Daniluk U, Kerros C, Tao RH, Wise JF, Ao X, Berkova Z, Samaniego F. The peptide derived from the Ig-like domain of human herpesvirus 8 K1 protein induces death in hematological cancer cells. J Exp Clin Cancer Res 2012; 31:69. [PMID: 22929310 PMCID: PMC3517441 DOI: 10.1186/1756-9966-31-69] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 08/17/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Although significant progress has been made in the treatment of lymphomas, many lymphomas exhibit resistance to cell death, suggesting a defective Fas signaling, which remains poorly understood. We previously reported that cells expressing the K1 protein of human herpesvirus 8 (HHV-8) resist death through the complex formation of the Ig-like domain of K1 with Fas. Recently, we investigated whether peptides derived from the Ig-like domain of the K1 protein may affect cell death. METHODS K1 positive and negative cell lines were incubated with the K1-derived peptides, and cell death (apoptotic and necrotic) was assessed by flow cytometry and LDH assay. Activation of caspases was assessed by fluorometric assay and flow cytometry. Fas receptor-independent, peptide-mediated cell killing was tested in the Fas-resistant Daudi cell line and Jurkat cell clones deficient in caspase-8 and FADD functionality. Activation of TNF receptors I and II was blocked by pre-incubation with corresponding blocking antibodies. The effect of the K1 peptide in vivo was tested in a mouse xenograft model. RESULTS We observed that the peptide S20-3 enhanced cell death in K1-positive BJAB cells and HHV-8 positive primary effusion lymphoma (PEL) cell lines. Similar effects of this peptide were observed in B-cell lymphoma and T-lymphoblastic leukemia cells without K1 expression but not in normal human peripheral blood mononuclear cells. A single intratumoral injection of the S20-3 peptide decreased the growth of Jurkat xenografts in SCID mice. The mechanism of tumor cell death induced by the S20-3 peptide was associated with activation of caspases, but this activity was only partially inhibited by the pan-caspase inhibitor z-VAD. Furthermore, the K1 peptide also killed Fas-resistant Daudi cells, and this killing effect was inhibited by pre-incubation of cells with antibodies blocking TNFRI. CONCLUSION Taken together, these findings indicate that the S20-3 peptide can selectively induce the death of malignant hematological cell lines by Fas- and/or TNFRI-dependent mechanisms, suggesting the K1-derived peptide or peptidomimetic may have promising therapeutic potential for the treatment of hematological cancers.
Collapse
Affiliation(s)
- Urszula Daniluk
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | | | | | | | | | | | | |
Collapse
|
12
|
Yang D, Torres CM, Bardhan K, Zimmerman M, McGaha TL, Liu K. Decitabine and vorinostat cooperate to sensitize colon carcinoma cells to Fas ligand-induced apoptosis in vitro and tumor suppression in vivo. THE JOURNAL OF IMMUNOLOGY 2012; 188:4441-9. [PMID: 22461695 DOI: 10.4049/jimmunol.1103035] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The death receptor Fas and its physiological ligand (FasL) regulate apoptosis of cancerous cells, thereby functioning as a critical component of the host cancer immunosurveillance system. To evade Fas-mediated apoptosis, cancer cells often downregulate Fas to acquire an apoptosis-resistant phenotype, which is a hallmark of metastatic human colorectal cancer. Therefore, targeting Fas resistance is of critical importance in Fas-based cancer therapy and immunotherapy. In this study, we demonstrated that epigenetic inhibitors decitabine and vorinostat cooperate to upregulate Fas expression in metastatic human colon carcinoma cells. Decitabine also upregulates BNIP3 and Bik expression, whereas vorinostat decreased Bcl-x(L) expression. Altered expression of Fas, BNIP3, Bik, and Bcl-x(L) resulted in effective sensitization of the metastatic human colon carcinoma cells to FasL-induced apoptosis. Using an experimental metastasis mouse model, we further demonstrated that decitabine and vorinostat cooperate to suppress colon carcinoma metastasis. Analysis of tumor-bearing lung tissues revealed that a large portion of tumor-infiltrating CD8(+) T cells are FasL(+), and decitabine and vorinostat-mediated tumor-suppression efficacy was significantly decreased in Fas(gld) mice compared with wild-type mice, suggesting a critical role for FasL in decitabine and vorinostat-mediated tumor suppression in vivo. Consistent with their function in apoptosis sensitization, decitabine and vorinostat significantly increased the efficacy of CTL adoptive transfer immunotherapy in an experimental metastasis mouse model. Thus, our data suggest that combined modalities of chemotherapy to sensitize the tumor cell to Fas-mediated apoptosis and CTL immunotherapy is an effective approach for the suppression of colon cancer metastasis.
Collapse
Affiliation(s)
- Dafeng Yang
- Department of Biochemistry and Molecular Biology, Georgia Health Sciences University, Augusta, GA 30912, USA
| | | | | | | | | | | |
Collapse
|
13
|
Hatton O, Phillips LK, Vaysberg M, Hurwich J, Krams SM, Martinez OM. Syk activation of phosphatidylinositol 3-kinase/Akt prevents HtrA2-dependent loss of X-linked inhibitor of apoptosis protein (XIAP) to promote survival of Epstein-Barr virus+ (EBV+) B cell lymphomas. J Biol Chem 2011; 286:37368-78. [PMID: 21908615 DOI: 10.1074/jbc.m111.255125] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
B cell lymphoma survival requires tonic or ligand-independent signals through activation of Syk by the B cell receptor. The Epstein-Barr virus (EBV) protein latent membrane 2a (LMP2a), a mimic of the B cell receptor, provides constitutive survival signals for latently infected cells through Syk activation; however, the precise downstream mechanisms coordinating this survival response in EBV+ B cell lymphomas remain to be elucidated. Herein, we assess the mechanism of Syk survival signaling in EBV+ B cell lymphomas from post-transplant lymphoproliferative disorder (PTLD) to discover virally controlled therapeutic targets involved in lymphomagenesis and tumor progression. Using small molecule inhibition and siRNA strategies, we show that Syk inhibition reduces proliferation and induces apoptosis of PTLD-derived EBV+ B cell lines. Syk inhibition also reduces autocrine IL-10 production. Although Syk inhibition attenuates signaling through both the PI3K/Akt and Erk pathways, only PI3K/Akt inhibition causes apoptosis of PTLD-derived cell lines. Loss of the endogenous caspase inhibitor XIAP is observed after Syk or PI3K/Akt inhibition. The loss of XIAP and apoptosis that results from Syk or PI3K/Akt inhibition is reversed by inhibition of the mitochondrial protease HtrA2. Thus, Syk drives EBV+ B cell lymphoma survival through PI3K/Akt activation, which prevents the HtrA2-dependent loss of XIAP. Syk, Akt, and XIAP antagonists may present potential new therapeutic strategies for PTLD through targeting of EBV-driven survival signals.
Collapse
Affiliation(s)
- Olivia Hatton
- Program in Immunology, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | | | | | |
Collapse
|
14
|
Srivastava S, Macaubas C, Deshpande C, Alexander HC, Chang SY, Sun Y, Park JL, Lee T, Begovich A, Mellins ED. Monocytes are resistant to apoptosis in systemic juvenile idiopathic arthritis. Clin Immunol 2010; 136:257-68. [PMID: 20462799 PMCID: PMC2902595 DOI: 10.1016/j.clim.2010.04.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 04/06/2010] [Accepted: 04/07/2010] [Indexed: 11/16/2022]
Abstract
We investigated whether circulating monocytes from patients with systemic juvenile idiopathic arthritis (SJIA) are resistant to apoptosis and which apoptotic pathway(s) may mediate this resistance. A microarray analysis of peripheral blood mononuclear cells (PBMC) of SJIA samples and RT-PCR analysis of isolated monocytes showed that monocytes from active SJIA patients express transcripts that imply resistance to apoptosis. SJIA monocytes incubated in low serum show reduced annexin binding and diminished FasL up-regulation compared to controls. SJIA monocytes are less susceptible to anti-Fas-induced apoptosis and, upon activation of the mitochondrial pathway with staurosporine, show diminished Bid cleavage and Bcl-w down-regulation compared to controls. Exposure to SJIA plasma reduces responses to apoptotic triggers in normal monocytes. Thus, SJIA monocytes are resistant to apoptosis due to alterations in both the extrinsic and intrinsic apoptosis pathways, and circulating factors associated with active SJIA may confer this phenotype.
Collapse
Affiliation(s)
| | - Claudia Macaubas
- Department of Pediatrics, Program in Immunology, Stanford University
| | - Chetan Deshpande
- Department of Pediatrics, Program in Immunology, Stanford University
| | | | | | - Yue Sun
- Department of Pediatrics, Program in Immunology, Stanford University
| | - Jane L. Park
- Department of Pediatrics, Program in Immunology, Stanford University
- Division of Pediatric Rheumatology, Department of Pediatrics, Stanford University
| | - Tzielan Lee
- Division of Pediatric Rheumatology, Department of Pediatrics, Stanford University
| | | | | |
Collapse
|
15
|
Watanabe KI, Ambekar C, Wang H, Ciccolini A, Schimmer AD, Dror Y. SBDS-deficiency results in specific hypersensitivity to Fas stimulation and accumulation of Fas at the plasma membrane. Apoptosis 2009; 14:77-89. [PMID: 19009351 DOI: 10.1007/s10495-008-0275-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Shwachman-Diamond syndrome (SDS) is an inherited disorder characterized by reduced cellularity in the bone marrow and exocrine pancreas. Most patients have mutations in the SBDS gene, whose functions are unknown. We previously showed that cells deficient in the SBDS protein are characterized by accelerated apoptosis and Fas hypersensitivity, suggesting that the protein might play an important role in Fas-mediated apoptosis. To study the mechanism of Fas hypersensitivity, we compared shRNA-mediated SBDS-knockdown HeLa cells and SDS marrow CD34+ cells for their sensitivity to several groups of apoptosis inducers. Marked hypersensitivity was noticed in response to Fas stimulation, but not to tumor necrosis factor-alpha, DNA-damaging agents, transcription inhibition or protein synthesis inhibition. To identify the Fas signaling factors that cause hypersensitivity, we analyzed the expression of the pathway's proteins. We found that Fas accumulated at the plasma membrane in SBDS-knockdown cells with corresponding expression of Fas transcript 1, the main Fas transcript which contains both the transmembrane domain and the death domain. However, the total levels of Fas protein and mRNA were comparable to controls, and Fas internalization occurred normally. Expression of FADD, caspase-8 and -3 were not elevated and the pathway inhibitors: ERK, c-FLIP and XIAP were not decreased. These results suggest that SBDS loss results in abnormal accumulation of Fas at the plasma membrane, where it sensitizes the cells to stimulation by Fas ligand.
Collapse
Affiliation(s)
- Ken-Ichiro Watanabe
- Cell Biology Program, Research Institute, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | | | | | | | | | | |
Collapse
|
16
|
Song H, Park G, Kim YS, Hur I, Kim H, Ryu JW, Lee HK, Cho DH, Choi IH, Lee WJ, Hur DY. B7-H4 reverse signaling induces the apoptosis of EBV-transformed B cells through Fas ligand up-regulation. Cancer Lett 2008; 266:227-37. [PMID: 18417276 DOI: 10.1016/j.canlet.2008.02.067] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2007] [Revised: 02/25/2008] [Accepted: 02/26/2008] [Indexed: 12/18/2022]
Abstract
B7-H4 has an inhibitory effect on immune responses via the down-regulation of T cell-mediated immunity, but how the engagement of B7-H4 molecules by counter molecules affects the signaling mechanism of the B7-H4-expressing cells is poorly defined. In this study, we found that B7-H4 expression was enhanced on B cells infected with Epstein-Barr virus (EBV) and that triggering of these molecules induced apoptosis of EBV-transformed B cells. Engagement of B7-H4 initially increased intracellular level of ROS, which then induced the expression of FasL. Engagement of B7-H4 subsequently provoked Fas-mediated and caspase-dependent apoptosis in association with cytochrome c and AIF, and EndoG was released from the mitochondria on EBV-transformed B cells. These results suggest that B7-H4 may be a potential therapeutic target for EBV involved malignancy diseases.
Collapse
Affiliation(s)
- Hyunkeun Song
- Department of Anatomy and Tumor Immunology, Inje University College of Medicine, 633-165 Kaekum-2-dong, Jin-gu, Busan 614-735, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Kim YS, Park GB, Song HK, Hur I, Lee HK, Kang JS, Hahm E, Lee WJ, Hur DY. Cross-linking of CD54 on Burkitt Lymphoma Cell Line Raji and Ramos Induces FasL Expression by Reactive Oxygen Species and Apoptosis of Adjacent Cells in Fas/FasL Interaction. J Immunother 2007; 30:727-39. [PMID: 17893565 DOI: 10.1097/cji.0b013e31814a69fa] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
CD54 is a cell surface adhesive glycoprotein, which is expressed in most cells. Interaction between CD54 and its ligands is involved in several cellular events including activation, proliferation, and cell death and also cell-to-cell adhesion. In the present study, we found that cross-linking of CD54 on Burkitt lymphoma cell lines, Raji and Ramos, induced apoptosis. We investigated that cross-linking of CD54 on Raji and Ramos using immobilized anti-CD54 mAb (clone 6.5B5) leads to apoptosis. CD54-induced apoptosis took place in association with an increase of intracellular reactive oxygen species (ROS) and a loss of the mitochondrial membrane potential and also the activation of caspases 3 and 9, resulting in the degradation of the proteolytic poly (ADP-ribose) polymerase. Pretreatment of each N-acetyl cystein and N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone (a broad caspase inhibitor) blocked apoptosis. Cross-linking of CD54 immediately induced expression of fasL, which was inhibited by pretreatment of N-acetyl cystein. NOK-1 (antagonistic anti-fasL), ZB4 (antagonistic anti-fas), and N-benzyloxycarbonyl-Ile-Glu-Thr-Asp-fluoromethylketon (caspase 8 inhibitor) effectively rescued cells from apoptosis via adjacent fas-fasL interaction but did not block ROS generation. Taken together, it is concluded that engagement of CD54 on B lymphoma cell lines by anti-CD54 mAb may trigger fasL expression through ROS generation and may subsequently induce apoptosis in adjacent fas-fasL interaction.
Collapse
Affiliation(s)
- Yeong Seok Kim
- Department of Anatomy and Research Center for Women's Disease, Inje University Busan Paik Hospital, Busan, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Epstein-Barr virus (EBV) infection is linked to approximately 90% of B-cell lymphomas associated with posttransplant lymphoproliferative disease (PTLD), a serious complication for immunosuppressed transplant recipients. In this paper, we review the myriad ways by which EBV can inadvertently drive the genesis and persistence of B-cell lymphomas, particularly when the antiviral immune response is compromised. Probing the basic mechanisms by which EBV infection proceeds and contributes to malignancy in such cases will hopefully improve our understanding and treatment of PTLD and other EBV-associated malignancies.
Collapse
Affiliation(s)
- Andrew L Snow
- Program in Immunology, Stanford University School of Medicine, Stanford, California, USA
| | | |
Collapse
|
19
|
Snow AL, Lambert SL, Natkunam Y, Esquivel CO, Krams SM, Martinez OM. EBV can protect latently infected B cell lymphomas from death receptor-induced apoptosis. THE JOURNAL OF IMMUNOLOGY 2006; 177:3283-93. [PMID: 16920969 DOI: 10.4049/jimmunol.177.5.3283] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The relationship between EBV infection and sensitivity to death receptor (DR)-induced apoptosis is poorly understood. Using EBV- and EBV+ BJAB cells, we provide the first evidence that EBV can protect latently infected B cell lymphomas from apoptosis triggered through Fas or TRAIL receptors. Caspase 8 activation was impaired and cellular FLIP recruitment was enriched in death-inducing signaling complexes formed in EBV-infected BJAB cells relative to parent BJAB cells. Furthermore, latent membrane protein 1 expression alone could reduce caspase activation and confer partial resistance to DR apoptosis in BJAB cells. This protective effect was dependent on C-terminal activating region 2-driven NF-kappaB activation, which in turn up-regulated cellular FLIP expression in latent membrane protein 1+ BJAB cells. Thus, the ability of latent EBV to block DR apoptosis may help to ensure the survival of host cells during B cell differentiation, and contribute to the development of B cell lymphomas, especially in immunocompromised individuals.
Collapse
Affiliation(s)
- Andrew L Snow
- Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | |
Collapse
|
20
|
Kim YS, Park GB, Choi YM, Kwon OS, Song HK, Kang JS, Kim YI, Lee WJ, Hur DY. Ligation of centrocyte/centroblast marker 1 on Epstein-Barr virus--transformed B lymphocytes induces cell death in a reactive oxygen species--dependent manner. Hum Immunol 2006; 67:795-807. [PMID: 17055356 DOI: 10.1016/j.humimm.2006.07.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2006] [Revised: 06/11/2006] [Accepted: 07/11/2006] [Indexed: 12/12/2022]
Abstract
After primary infection of B cells with Epstein-Barr virus (EBV), infected B cells express several viral homologs of human genes that promote activation (LMP1 and CD40) or survival (BHRF and BCL2). EBV-infected B cells also express germinal center phenotype markers, such as CD77, PNA, CD95, and CD38. This transformation of B cells by EBV infection resembles normal B-cell activation and differentiation arising in the germinal center. In the present study, we found that EBV-transformed B cells expressed centrocyte/centroblast marker 1 (CM1), a possible marker of GC B cells and an inducer of their apoptosis. Moreover, ligation of CM1 on EBV-transformed B cells by immobilized anti-CM1 monoclonal antibody induced cell death. The ligation of CM1 immediately increased the generation of intracellular reactive oxygen species (ROS) and disrupted the mitochondrial membrane potential. Pretreatment with N-acetyl cystein (an ROS inhibitor) almost completely blocked this cell death, but Z-VAD-fmk (a caspase inhibitor) did not. We further investigated whether apoptosis-inducing factor (AIF) and endonuclease G (EndoG), which are both related to caspase-independent cell death, would be translocated to the nucleus during the ligation of CM1. We found that AIF and EndoG were released to the cytosplam but not translocated to the nucleus. Moreover, cytochalasin D, a cytoskeleton disruptor, rescued the cells from CM1-mediated cell death and blocked ROS generation. Therefore, it is conceivable that CM1 signaling might provoke cytoskeleton polymerization and trigger ROS generation. Taking these observations together, we conclude that the ligation of CM1 on EBV-transformed B cells can cause cell death via the ROS produced by F-actin polymerization in a caspase-independent manner, although this cell death might be unrelated to AIF and EndoG release from the mitochondria.
Collapse
MESH Headings
- Acetylcysteine/pharmacology
- Actins/metabolism
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Antigens, Differentiation, B-Lymphocyte/immunology
- Antigens, Differentiation, B-Lymphocyte/physiology
- Antioxidants/pharmacology
- Apoptosis/drug effects
- Apoptosis/physiology
- Apoptosis Inducing Factor/metabolism
- B-Lymphocytes/metabolism
- B-Lymphocytes/physiology
- B-Lymphocytes/virology
- Caspase Inhibitors
- Caspases/metabolism
- Cell Nucleus/metabolism
- Cell Transformation, Viral
- Cells, Cultured
- Cytochalasin D/pharmacology
- Cytochromes c/metabolism
- Endodeoxyribonucleases/metabolism
- Herpesvirus 4, Human
- Humans
- Membrane Potential, Mitochondrial/drug effects
- Membrane Potential, Mitochondrial/immunology
- Mitochondria/metabolism
- Phosphatidylserines/metabolism
- Reactive Oxygen Species/antagonists & inhibitors
- Reactive Oxygen Species/metabolism
Collapse
Affiliation(s)
- Yeong Seok Kim
- Department of Anatomy and Research Center for Women's Disease, Inje University College of Medicine, Busan, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Snow AL, Vaysberg M, Krams SM, Martinez OM. EBV B lymphoma cell lines from patients with post-transplant lymphoproliferative disease are resistant to TRAIL-induced apoptosis. Am J Transplant 2006; 6:976-85. [PMID: 16611333 DOI: 10.1111/j.1600-6143.2006.01295.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Lymphomas associated with post-transplant lymphoproliferative disease (PTLD) represent a significant complication of immunosuppression in transplant recipients. In immunocompetent individuals, EBV-specific cytotoxic T lymphocytes (CTL) prevent the outgrowth of activated B lymphoblasts through apoptosis induction. Soluble versions of TNF-related apoptosis-inducing ligand/Apo2 ligand (TRAIL) can induce apoptosis in numerous tumor cell types. Given the therapeutic potential of TRAIL, we examined the sensitivity of EBV+ spontaneous lymphoblastoid cell lines (SLCL) derived from patients with PTLD to treatment with soluble TRAIL. Despite abundant expression of TRAIL receptors (TRAIL-R), resistance to TRAIL-induced apoptosis was observed in all SLCL examined. This resistance could not be overcome by concomitant treatment with several pharmacological agents. Unlike BJAB positive control cells, for each SLCL tested, cleavage and activation of caspase 8 was inhibited due to failed recruitment of FADD and caspase 8 to TRAIL receptors upon stimulation. Further indicative of a proximal defect, TRAIL receptor aggregation could not be detected on the cell surface of SLCL following ligand engagement. These results suggest that the use of TRAIL for eliminating PTLD-associated tumors may be of limited clinical utility, and illustrate another mechanism by which EBV+ B lymphoma cells can evade tumor surveillance at the level of death receptor signaling.
Collapse
Affiliation(s)
- A L Snow
- Program in Immunology, Department of Surgery, Stanford University School of Medicine, Stanford, California 94305-5492, USA
| | | | | | | |
Collapse
|
22
|
Jeon YK, Kim H, Park SO, Choi HY, Kim YA, Park SS, Kim JE, Kim YN, Kim CW. Resistance to Fas-mediated apoptosis is restored by cycloheximide through the downregulation of cellular FLIPL in NK/T-cell lymphoma. J Transl Med 2005; 85:874-84. [PMID: 15924153 DOI: 10.1038/labinvest.3700291] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Extranodal NK/T-cell lymphoma (NKTL), nasal type, is a highly aggressive neoplasm and is strongly associated with Epstein-Barr virus (EBV). In this study, we demonstrate that EBV-positive NKTL cell lines, namely, Hank-1, NK-YS, and NK-L, are resistant to Fas-mediated apoptosis induced by anti-Fas antibodies despite high levels of Fas surface expression and no mutation in the Fas gene. Fas stimulation of Hank-1 and NK-YS cells showed little processing of caspase 8, caspase 3, or bid, although the proximal signaling molecules of the death-inducing signaling complex, namely, Fas, Fas-associated protein with a death domain, caspase 8, and bid were present in these cells. Consistent with previous reports on the hypermethylation of death associated protein (DAP) kinase in NKTLs, the promoter of DAP kinase was methylated and its mRNA not detected in Hank-1 cells. However, the restoration of DAP kinase expression by 5-aza-2'-deoxycytidine did not sensitize Hank-1 to Fas-mediated apoptosis, indicating that DAP kinase deficiency does not contribute to resistance to Fas-mediated apoptosis. Since etoposide-induced apoptosis involved caspase 3 activation in Hank-1 and NK-YS cells, the caspase 3-dependent apoptotic machinery appears to be intact. Interestingly, cotreatment of Hank-1 with cycloheximide, a protein synthesis inhibitor, markedly sensitized cells to Fas-mediated apoptosis along with caspase 8 activation and c-FLIP(L) (cellular FLICE inhibitory protein long form) downregulation. Moreover, immunohistochemistry on paraffin-embedded tissue revealed c-FLIP expression in 39% (14 of 36) of NKTL patients. Taken together, these findings indicate that c-FLIP(L)-mediated resistance to Fas contributes to the development and progression of NKTLs. This study also suggests that agents capable of downregulating c-FLIP(L) could be used to treat NKTL.
Collapse
Affiliation(s)
- Yoon Kyung Jeon
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Zhang W, Ding EX, Wang Q, Zhu DQ, He J, Li YL, Wang YH. Fas ligand expression in colon cancer: A possible mechanism of tumor immune privilege. World J Gastroenterol 2005; 11:3632-5. [PMID: 15962391 PMCID: PMC4315977 DOI: 10.3748/wjg.v11.i23.3632] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To detect the expression of Fas ligand (FasL) in colon cancer tissues and cell lines and analyze the function of FasL-expressing colon cancer cells in inducing Fas-sensitive T lymphocyte apoptosis.
METHODS: Ninety surgically resected colon cancer tissues and 15 hepatic metastasis specimens were investigated by immunohistochemical method with normal colon mucosa and colon adenoma as control. The relationship between FasL expression and pathologic features was also analyzed. FasL expression of 4 colon cancer cell lines, SW620, Lovo, LS-174T and SW1116, were detected by Western blotting assay. The function of FasL expressed on colon cancer cells was determined by coculture assay with Jurkat T lymphocytes, the apoptotic rate of which was detected by flow cytometry assay.
RESULTS: Fifty-six (62.22%) cases of all the 90 colon cancer tissues and all (100%) the liver metastasis specimens expressed FasL, significantly higher than normal colon mucosa and colonic adenoma. Higher expression of FasL was found in more advanced stage of colon cancer and in cancer tissues with lymphatic or hepatic metastasis. All the colon cancer cell lines were found to express FasL. After coculture with the SW1116 cells for 24 h with an effector: target ratio 10:1, the rate of apoptosis of Jurkat cells rose from 1.9% to 21.0%.
CONCLUSION: The expression of FasL is upregulated in colon cancer and the functionally expressed FasL can induce apoptosis of Fas-expressing T lymphocytes.
Collapse
Affiliation(s)
- Wei Zhang
- Department of General Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 20003, China.
| | | | | | | | | | | | | |
Collapse
|
24
|
Schmitz I, Weyd H, Krueger A, Baumann S, Fas SC, Krammer PH, Kirchhoff S. Resistance of short term activated T cells to CD95-mediated apoptosis correlates with de novo protein synthesis of c-FLIPshort. THE JOURNAL OF IMMUNOLOGY 2004; 172:2194-200. [PMID: 14764686 DOI: 10.4049/jimmunol.172.4.2194] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In the early phase of an immune response, T cells are activated and acquire effector functions. Whereas these short term activated T cells are resistant to CD95-mediated apoptosis, activated T cells in prolonged culture are readily sensitive, leading to activation-induced cell death and termination of the immune response. The translation inhibitor, cycloheximide, partially overcomes the apoptosis resistance of short term activated primary human T cells. Using this model we show in this study that sensitization of T cells to apoptosis occurs upstream of mitochondria. Neither death-inducing signaling complex formation nor expression of Bcl-2 proteins is altered in sensitized T cells. Although the caspase-8 inhibitor c-FLIP(long) was only slightly down-regulated in sensitized T cells, c-FLIP(short) became almost undetectable. This correlated with caspase-8 activation and apoptosis. These data suggest that c-FLIP(short), rather than c-FLIP(long), confers resistance of T cells to CD95-mediated apoptosis in the context of immune responses.
Collapse
Affiliation(s)
- Ingo Schmitz
- Tumor Immunology Program, German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|
25
|
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
|
26
|
Smolewski P, Darzynkiewicz Z, Robak T. Caspase-mediated cell death in hematological malignancies: theoretical considerations, methods of assessment, and clinical implications. Leuk Lymphoma 2003; 44:1089-104. [PMID: 12916860 DOI: 10.1080/1042819031000077007] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Apoptosis, the caspase-mediated cell death, plays an important role in the etiology, pathogenesis and therapy of a variety of diseases. Abnormalities of apoptosis regulation, resulting in either its inhibition or enhancement, play a key role in the development of various malignant hematological disorders. Several routine and new therapeutic strategies in Oncohematology are based on apoptosis modulation. Cytotoxic effects of most antineoplastic drugs are based on induction of apoptosis. The accurate estimate of incidence of apoptosis, therefore, is of importance in Oncohematology. In this review we provide an overview of the methods designed to measure the incidence of apoptosis, including the recently developed assays that are based on detection of caspases activation. We also review recent findings on the role of caspase-mediated cell death in hematological malignancies and discuss their clinical implications, including new therapeutical strategies that evolve from these findings.
Collapse
Affiliation(s)
- Piotr Smolewski
- Department of Hematology, Medical University of Lodz, Copernicus Hospital, ul. Ciokowskiego 2, 93-510 Lodz, Poland.
| | | | | |
Collapse
|
27
|
Kim YJ, Brutkiewicz RR, Broxmeyer HE. Role of 4-1BB (CD137) in the functional activation of cord blood CD28(-)CD8(+) T cells. Blood 2002; 100:3253-60. [PMID: 12384425 DOI: 10.1182/blood-2001-11-0136] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The CD28(-) subset of CD8(+) T cells is associated with cytotoxic T lymphocyte (CTL) effector function. We investigated a potential role for 4-1BB, a costimulatory molecule structurally related to members of the tumor necrosis factor (TNF) receptor family, in the generation and functional activation of CD28(-) CTLs by using human cord blood (CB) cells composed exclusively of naive CD8(+) T cells with few or no CD28(-) CTLs. The 4-1BB was induced preferentially on the CB CD28(-)CD8(+) T cells when CD28 down-regulation was induced by interleukin 15 (IL-15) and IL-12 stimulation. Anti-4-1BB costimulation induced dramatic phenotypic changes in the CD28(-) CTLs, including restoration of CD28 expression as well as that of memory markers such as CD45RO and CC chemokine receptor 6 (CCR6). Anti-4-1BB costimulation also promoted long-term survival of CD28(-) CTLs, which were sensitive to activation-induced cell death upon anti-CD3 stimulation. The memory-type CD28(+) CTLs induced by anti-4-1BB costimulation acquired a greatly enhanced content of granzyme B, a cytolytic mediator, and enhanced cytotoxic activity as compared with CD28(-) CTLs. Strong cytotoxicity of memory-type CTLs to a 4-1BB ligand-expressing Epstein-Barr virus (EBV)-transformed B-cell line was almost completely abrogated by 4-1BB-Fc, a soluble form of 4-1BB, suggesting involvement of 4-1BB in cytolytic processes. Taken all together, our results suggest that 4-1BB plays a role in the differentiation of effector memory CTLs.
Collapse
MESH Headings
- Antigens, CD
- CD28 Antigens/analysis
- CD8 Antigens/analysis
- Cell Differentiation
- Cell Line, Transformed
- Cell Survival
- Cytotoxicity, Immunologic
- Fetal Blood/cytology
- Fetal Blood/immunology
- Gene Expression Regulation/drug effects
- Granzymes
- Herpesvirus 4, Human/physiology
- Humans
- Immunologic Memory
- Infant, Newborn
- Interleukin-12/pharmacology
- Interleukin-15/pharmacology
- K562 Cells
- Lymphocyte Activation/drug effects
- Lymphocyte Activation/physiology
- Receptors, Nerve Growth Factor/biosynthesis
- Receptors, Nerve Growth Factor/genetics
- Receptors, Nerve Growth Factor/physiology
- Receptors, Tumor Necrosis Factor/biosynthesis
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/physiology
- Serine Endopeptidases/analysis
- T-Lymphocyte Subsets/drug effects
- T-Lymphocyte Subsets/enzymology
- T-Lymphocyte Subsets/immunology
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/enzymology
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Necrosis Factor Receptor Superfamily, Member 9
Collapse
Affiliation(s)
- Young-June Kim
- Department of Microbiology and Immunology, Walther Oncology Center, Indiana University School of Medicine, Indianapolis, IN 46202-5254, USA
| | | | | |
Collapse
|