1
|
Dong X, Nie C, Yin Y, Xu H, Zhang Y, Han L, Xu C. Role of B7H4 and Fas in regulation of trophoblast cells and development of pre-eclampsia: a cross-sectional study. J OBSTET GYNAECOL 2025; 45:2452840. [PMID: 39889763 DOI: 10.1080/01443615.2025.2452840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 01/05/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND This study aimed to investigate the role of B7H4 and Fas in pre-eclampsia (PE) occurrence and development and reveal its potential mechanisms. METHODS Thirty healthy individuals and 60 patients with PE were enrolled in the study. In addition, the clinical characteristics of all participants were collected, including age, gestational weeks at delivery, gestational time, number of births, systolic blood pressure, diastolic blood pressure and foetal weight. The chi-square test was used to evaluate differences in clinical characteristics between the high- and low-expression groups. The expression levels of B7H4 and Fas were analysed using western blotting and real-time quantitative polymerase chain reaction (RT-qPCR). The upstream regulators of B7H4 in trophoblasts were predicted and estimated using a luciferase reporter assay. The proliferation and motility of trophoblasts were assessed using CCK8 and transwell assays, respectively. RESULTS B7H4 and Fas were upregulated in PE (p < 0.05) and showed diagnostic potential with insufficient sensitivity and specificity [B7H4: area under curve (AUC) = 0.790, sensitivity = 65%, specificity = 83.33%; Fas: AUC = 0.717, sensitivity = 68.34%, specificity = 73.33%]. B7H4 and Fas were closely associated with increased blood pressure in patients with PE (p < 0.05), and the combination of B7H4 and Fas increased the diagnostic efficacy (AUC = 0.864), sensitivity (73.33%) and specificity (86.67%). In trophoblast cells, miR-4319 negatively regulated B7H4 and Fas expression as well as cell proliferation, migration and invasion (p < 0.05). Overexpression of B7H4 alleviated the inhibitory effects of miR-4319, which were reversed by Fas knockdown (p < 0.05). CONCLUSIONS miR-4319 mediates the progression of trophoblast progression by negatively regulating the expression of B7H4 and Fas. Therefore, B7H4 and Fas may serve as potential biomarkers for the prediction and treatment of PE.
Collapse
Affiliation(s)
- Xiujuan Dong
- Department of Obstetrics, Sunshine Union Hospital, Weifang, China
| | - Cuicui Nie
- Department of Obstetrics, Sunshine Union Hospital, Weifang, China
| | - Yanli Yin
- Department of Obstetrics, Sunshine Union Hospital, Weifang, China
| | - Haixia Xu
- Department of Obstetrics, Sunshine Union Hospital, Weifang, China
| | - Yanxia Zhang
- Department of Obstetrics, Sunshine Union Hospital, Weifang, China
| | - Lihua Han
- Department of Obstetrics, Sunshine Union Hospital, Weifang, China
| | - Congcong Xu
- Department of Obstetrics, Sunshine Union Hospital, Weifang, China
| |
Collapse
|
2
|
Silveira DA, Ribeiro FM, Simão ÉM, Mattos VLD, Góes EG. Expression of genes and pathways associated with the B7-CD28 superfamily in response to irradiation of blood cells using 137Cs. Int J Radiat Biol 2020; 97:149-155. [PMID: 33253600 DOI: 10.1080/09553002.2021.1857454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
PURPOSE DNA damage is one of the main consequences of exposure to ionizing irradiation (IR). Recent studies indicate that IR can modulate the expression of immune system-related genes. However, the effects of IR on the expression of genes and pathways of the B7-CD28 superfamily remain poorly defined. The aim of this study was to evaluate the modulation of genes and pathways related to the B7-CD28 superfamily in response to IR. MATERIALS AND METHODS In this study, we used transcriptome data available from the Gene Expression Omnibus (GEO) database to investigate the modulation of the response of genes and pathways of samples of human peripheral blood irradiated with doses of 150, 300, and 600 cGy. The data were obtained at 6 and 24 h after irradiation. The relationship between genes and pathways was established through the Reactome database. The behavior of these pathways was analyzed using mathematical methods based on relative activity and diversity. Analysis of variance (ANOVA) followed by multiple comparisons tests (Bonferroni and Tamhanes) was used to identify differentially expressed genes. Data on transcriptomes were analyzed through ViaComplex V.1.0 and IBM SPSS Statistics 22. RESULTS For the pathways investigated in this study, we observed that the effects produced by these doses significantly modified the behavior of five pathways associated with the immune system. Also, the dose of 300 cGy might trigger signaling for the activation of T cells through the negative regulation (p < .05) of the co-inhibitory PDCD1LG2 gene. Positive regulation caused by 300 cGy (p < .05) of the CD80 receptor was observed by us, which might be related to a stimulatory signal. According to our findings, this dose induced the production of cytokines and genes that are associated with the activation and differentiation of T cells. CONCLUSIONS Our findings indicate that the irradiation modulated the organization of the biological system, suggesting that 300 cGy is more efficient in activating the immune system.
Collapse
Affiliation(s)
- Daner A Silveira
- Institute of Mathematics, Statistics and Physics, Federal University of Rio Grande, Rio Grande, Brazil
| | - Fernanda M Ribeiro
- Institute of Mathematics, Statistics and Physics, Federal University of Rio Grande, Rio Grande, Brazil
| | - Éder M Simão
- Nanoscience Graduate Program, Franciscan University, Santa Maria, Brazil
| | - Viviane L D Mattos
- Institute of Mathematics, Statistics and Physics, Federal University of Rio Grande, Rio Grande, Brazil
| | - Evamberto G Góes
- Institute of Mathematics, Statistics and Physics, Federal University of Rio Grande, Rio Grande, Brazil
| |
Collapse
|
3
|
Wang JY, Wang WP. B7-H4, a promising target for immunotherapy. Cell Immunol 2019; 347:104008. [PMID: 31733822 DOI: 10.1016/j.cellimm.2019.104008] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/23/2019] [Accepted: 11/02/2019] [Indexed: 02/07/2023]
Abstract
The coinhibitory molecule B7-H4, an important member of the B7 family, is abnormally expressed in tumors, inflammation and autoimmune diseases. B7-H4 negatively regulates T cell immune response and promotes immune escape by inhibiting the proliferation, cytokine secretion, and cell cycle of T cells. Moreover, B7-H4 plays an extremely important role in tumorigenesis and tumor development including cell proliferation, invasion, metastasis, anti-apoptosis, etc. In addition, B7-H4 has the other biological functions, such as protection against type 1 diabetes (T1D) and islet cell transplantation. Therefore, B7-H4 has been identified as a novel marker or a therapeutic target for the treatment of tumors, inflammation, autoimmune diseases, and organ transplantation. Here, we summarized the expression profiles, physiological and pathological functions, and regulatory mechanisms of B7-H4, the signaling pathways involved, as well as B7-H4-based immunotherapy.
Collapse
Affiliation(s)
- Jia-Yu Wang
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Wei-Peng Wang
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China.
| |
Collapse
|
4
|
Chapoval AI, Chapoval SP, Shcherbakova NS, Shcherbakov DN. Immune Checkpoints of the B7 Family. Part 1. General Characteristics and First Representatives: B7-1, B7-2, B7-H1, B7-H2, and B7-DC. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2019. [DOI: 10.1134/s1068162019040101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
5
|
Jiang Y, Cai G, Lin J, Zhang J, Bo Z, Li Y, Wang C, Tong Y. B7-H4 is highly expressed in aggressive Epstein-Barr virus positive diffuse large B-cell lymphoma and inhibits apoptosis through upregulating Erk1/2 and Akt signalling pathways. Infect Agent Cancer 2019; 14:20. [PMID: 31406503 PMCID: PMC6686556 DOI: 10.1186/s13027-019-0234-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/22/2019] [Indexed: 11/22/2022] Open
Abstract
Background B7-H4 is among the B7 family members which may serve as a new targetable immune checkpoint molecule. It was reported that high level of serum B7-H4 level may be correlated with lymphoma. Nevertheless, the role of B7-H4 in Epstein-Barr Virus-Positive diffuse large B cell lymphoma (EBV+DLBCL) has not been addressed although it has been suggested that B7-H4 could promote tumor growth and metastatic progression in certain cancers. Methods Between January 2005 and November 2017 at the department of Hematology, Shanghai Jiao Tong University School of Medicine affiliated Shanghai General Hospital 260 DLBCL samples were analyzed for EBV-encoded small RNA (EBV-EBER) by in situ hybridization. The expression level of B7-H4 in DLBCL tumor tissue was evaluated by immunohistochemistry. Furthermore, the role of B7-H4 in DLBCL was further investigated in DLBCL cell line. Results EBV+DLBCL patients suffered from markedly lower overall survival (OS) and progression-free survival (PFS) rates in our study. We showed that B7-H4 was significantly overexpressed in 16 EBV+-subgroup cases out of 260 DLBCL patients. We further found that EBV infection in lymphoblast cells led to enhanced expression of B7-H4 followed by increased cell viability and reduced apoptosis. In contrast, inhibition of B7-H4 simultaneously impaired cell viability and induced apoptosis. Mechanistically, inhibiting B7-H4 resulted in decreased phosphorylation Erk 1/2 and Akt. Conclusion Our study reveals a critical role of B7-H4 in EBV+DLBCL development by regulating cell survival and apoptosis through the Erk and Akt signalling pathways. Targetting B7-H4 may be promising in the therapy of EBV+DLBCL.
Collapse
Affiliation(s)
- Ying Jiang
- 1Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080 People's Republic of China
| | - Gangli Cai
- Department of Hematology, JinHua Hospital of TCM, 439 Shuangxi West Road, Jinhua, 321017 People's Republic of China
| | - Jun Lin
- 3Department of Pathology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080 People's Republic of China
| | - Jing Zhang
- 4Department of Integrated Therapy, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032 People's Republic of China
| | - Zhilei Bo
- 1Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080 People's Republic of China
| | - Ying Li
- 1Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080 People's Republic of China
| | - Chun Wang
- 1Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080 People's Republic of China
| | - Yin Tong
- 1Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080 People's Republic of China
| |
Collapse
|
6
|
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
|
7
|
Leibler C, Thiolat A, Elsner RA, El Karoui K, Samson C, Grimbert P. Costimulatory blockade molecules and B-cell-mediated immune response: current knowledge and perspectives. Kidney Int 2019; 95:774-786. [PMID: 30711200 DOI: 10.1016/j.kint.2018.10.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/17/2018] [Accepted: 10/18/2018] [Indexed: 12/12/2022]
Abstract
There is an urgent need for therapeutic agents that target humoral alloimmunity in solid organ transplantation. This includes sensitized patients with preformed donor-specific human leukocyte antigen antibodies and patients who develop de novo donor-specific antibodies, both of which are associated with acute and chronic antibody-mediated rejection and allograft loss. In the last decade, both experimental and clinical studies highlighted the major impact of costimulation molecules in the control of immune responses both in the field of transplantation and autoimmune disease. Although these molecules have been initially developed to control the early steps of T-cell activation, recent evidence also supports their influence at several steps of the humoral response. In this review, we aim to provide an overview of the current knowledge of the effects of costimulatory blockade agents on humoral responses in both autoimmune and allogeneic contexts. We first present the effects of costimulatory molecules on the different steps of alloantibody production. We then summarize mechanisms and clinical results observed using cytotoxic T lymphocyte antigen-4 (CTLA4)-Ig molecules both in transplantation and autoimmunity. Finally, we present the potential interest and implications of other costimulatory family members as therapeutic targets, with emphasis on combinatorial approaches, for the optimal control of the alloantigen-specific humoral response.
Collapse
Affiliation(s)
- Claire Leibler
- Service de Néphrologie et Transplantation, Pôle Cancérologie-Immunité-Transplantation-Infectiologie, Paris-Est Creteil, France; Institut National de la Santé et de la Recherch Médicale, U955, Equipe 21 and Université Paris-Est, Créteil, France; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Allan Thiolat
- Institut National de la Santé et de la Recherch Médicale, U955, Equipe 21 and Université Paris-Est, Créteil, France
| | - Rebecca A Elsner
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Khalil El Karoui
- Service de Néphrologie et Transplantation, Pôle Cancérologie-Immunité-Transplantation-Infectiologie, Paris-Est Creteil, France; Institut National de la Santé et de la Recherch Médicale, U955, Equipe 21 and Université Paris-Est, Créteil, France
| | - Chloe Samson
- Institut National de la Santé et de la Recherch Médicale, U955, Equipe 21 and Université Paris-Est, Créteil, France
| | - Philippe Grimbert
- Service de Néphrologie et Transplantation, Pôle Cancérologie-Immunité-Transplantation-Infectiologie, Paris-Est Creteil, France; Institut National de la Santé et de la Recherch Médicale, U955, Equipe 21 and Université Paris-Est, Créteil, France.
| |
Collapse
|
8
|
Xue Q, Yin Z, Varshithreddy N, Liang HS, Wang MY, Dong WL, Zhang X, Gu Y, Fang Q. The immunomodulatory function of human amniotic fluid stromal cells on B lymphocytes. JOURNAL OF NEURORESTORATOLOGY 2018. [DOI: 10.26599/jnr.2018.9040010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
9
|
|
10
|
Xie N, Cai JB, Zhang L, Zhang PF, Shen YH, Yang X, Lu JC, Gao DM, Kang Q, Liu LX, Zhang C, Huang XY, Zou H, Zhang XY, Song ZJ, Sun HX, Fu BM, Ke AW, Shi GM. Upregulation of B7-H4 promotes tumor progression of intrahepatic cholangiocarcinoma. Cell Death Dis 2017; 8:3205. [PMID: 29235470 PMCID: PMC5870586 DOI: 10.1038/s41419-017-0015-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 09/23/2017] [Accepted: 09/25/2017] [Indexed: 12/12/2022]
Abstract
Recent reports show that B7-H4 is highly expressed in a variety of tumor cells, functions as a negative regulator of T cells and then promotes tumor progression. However, its expression and role in intrahepatic cholangiocarcinoma (ICC) remain unclear. In present study, B7-H4 expression in ICC and peritumoral tissues was determined at the level of mRNA and protein, and its bioactivity in ICC cells was studied after modification of B7-H4 expression. Then, the mechanism related to tumor progression induced by B7-H4 expression in ICC cells was explored. Finally, clinical significance of B7-H4 expression in ICC patients was further analyzed. The results showed that B7-H4 expression in ICC was much higher than that in peritumoral tissues at the level of both mRNA and protein. The high level of B7-H4 in ICC cells induced epithelial-to-mesenchymal transitions and promoted invasion and metastasis of tumor cells through activation of ERK1/2 signaling. The elevated B7-H4 expression was associated with the downregulated Bax, upregulated Bcl-2 expression, and activation of caspase-3. Clinically, high B7-H4 expression in tumor samples was significantly related to malignant phenotype, such as lymph node metastasis, high tumor stage, and poor differentiation. ICC patients with high expression of B7-H4 had shorter overall survival (OS) and disease-free survival. Moreover, the B7-H4 expression was an independent prognostic factor for predicting OS and tumor recurrence of ICC patients after operation. In conclusion, high expression of B7-H4 promotes tumor progression of ICC and may be a novel therapeutic target for ICC patients.
Collapse
Affiliation(s)
- Nan Xie
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry, Department of Liver Surgery and Liver Transplant of Zhongshan Hospital, Liver Cancer Institute of Fudan University, Fudan University, Shanghai, 200032, China
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650101, China
| | - Jia-Bin Cai
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry, Department of Liver Surgery and Liver Transplant of Zhongshan Hospital, Liver Cancer Institute of Fudan University, Fudan University, Shanghai, 200032, China
| | - Lu Zhang
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry, Department of Liver Surgery and Liver Transplant of Zhongshan Hospital, Liver Cancer Institute of Fudan University, Fudan University, Shanghai, 200032, China
| | - Peng-Fei Zhang
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry, Department of Liver Surgery and Liver Transplant of Zhongshan Hospital, Liver Cancer Institute of Fudan University, Fudan University, Shanghai, 200032, China
| | - Ying-Hao Shen
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry, Department of Liver Surgery and Liver Transplant of Zhongshan Hospital, Liver Cancer Institute of Fudan University, Fudan University, Shanghai, 200032, China
| | - Xuan Yang
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry, Department of Liver Surgery and Liver Transplant of Zhongshan Hospital, Liver Cancer Institute of Fudan University, Fudan University, Shanghai, 200032, China
| | - Jia-Cheng Lu
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry, Department of Liver Surgery and Liver Transplant of Zhongshan Hospital, Liver Cancer Institute of Fudan University, Fudan University, Shanghai, 200032, China
| | - Dong-Mei Gao
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry, Department of Liver Surgery and Liver Transplant of Zhongshan Hospital, Liver Cancer Institute of Fudan University, Fudan University, Shanghai, 200032, China
| | - Qiang Kang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650101, China
| | - Li-Xin Liu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650101, China
| | - Chi Zhang
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry, Department of Liver Surgery and Liver Transplant of Zhongshan Hospital, Liver Cancer Institute of Fudan University, Fudan University, Shanghai, 200032, China
| | - Xiao-Yong Huang
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry, Department of Liver Surgery and Liver Transplant of Zhongshan Hospital, Liver Cancer Institute of Fudan University, Fudan University, Shanghai, 200032, China
| | - Hao Zou
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650101, China
| | - Xin-Yu Zhang
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry, Department of Liver Surgery and Liver Transplant of Zhongshan Hospital, Liver Cancer Institute of Fudan University, Fudan University, Shanghai, 200032, China
| | - Zheng-Ji Song
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, 157 Jin Bi Road, Kunming, Yunnan, 650032, China
| | - Hai-Xiang Sun
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry, Department of Liver Surgery and Liver Transplant of Zhongshan Hospital, Liver Cancer Institute of Fudan University, Fudan University, Shanghai, 200032, China
| | - Bi-Mang Fu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650101, China.
| | - Ai-Wu Ke
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry, Department of Liver Surgery and Liver Transplant of Zhongshan Hospital, Liver Cancer Institute of Fudan University, Fudan University, Shanghai, 200032, China.
| | - Guo-Ming Shi
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry, Department of Liver Surgery and Liver Transplant of Zhongshan Hospital, Liver Cancer Institute of Fudan University, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
11
|
Abstract
Observations noting the presence of white blood cell infiltrates within tumors date back more than a century, however the cellular and molecular mechanisms regulating tumor immunity continue to be elucidated. The recent successful use of monoclonal antibodies to block immune regulatory pathways to enhance tumor-specific immune responses for the treatment of cancer has encouraged the identification of additional immune regulatory receptor/ligand pathways. Over the past several years, a growing body of data has identified B7-H4 (VTCN1/B7x/B7S1) as a potential therapeutic target for the treatment of cancer. The potential clinical significance of B7-H4 is supported by the high levels of B7-H4 expression found in numerous tumor tissues and correlation of the level of expression on tumor cells with adverse clinical and pathologic features, including tumor aggressiveness. The biological activity of B7-H4 has been associated with decreased inflammatory CD4+ T-cell responses and a correlation between B7-H4-expressing tumor-associated macrophages and FoxP3+ regulatory T cells (Tregs) within the tumor microenvironment. Since B7-H4 is expressed on tumor cells and tumor-associated macrophages in various cancer types, therapeutic blockade of B7-H4 could favorably alter the tumor microenvironment allowing for antigen-specific clearance tumor cells. The present review highlights the therapeutic potential of targeting B7-H4.
Collapse
Affiliation(s)
- Joseph R Podojil
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
12
|
Hong B, Qian Y, Zhang H, Sang YW, Cheng LF, Wang Q, Gao S, Zheng M, Yao HP. Expression of B7-H4 and hepatitis B virus X in hepatitis B virus-related hepatocellular carcinoma. World J Gastroenterol 2016; 22:4538-4546. [PMID: 27182163 PMCID: PMC4858635 DOI: 10.3748/wjg.v22.i18.4538] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 02/27/2016] [Accepted: 03/18/2016] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression and clinical significance of B7-H4 and hepatitis B virus X (HBx) protein in hepatitis B virus-related hepatocellular carcinoma (HBV-HCC).
METHODS: The expression of B7-H4 in the human HCC cell lines HepG2 and HepG2.2.15 were detected by western blot, flow cytometry, and immunofluorescence. The expression of B7-H4 and HBx in 83 HBV-HCC was detected by immunohistochemistry, and the relationship with clinicopathological features was analyzed. Paraffin sections were generated from 83 HBV-HCC patients (22 females and 61 males) enrolled in this study. The age of these patients ranged from 35 to 77 years, with an average of 52.5 ± 11.3 years. All experiments were approved by the Ethics Committees of the Second Affiliated Hospital, Zhejiang University School of Medicine.
RESULTS: B7-H4 was significantly upregulated in HepG2.2.15 cells compared to HepG2 cells. Specifically, the protein expression of B7-H4 in the lysates of HepG2 cells was more than that in HepG2.2.15 cells. In addition, HBx was expressed only in HepG2.2.15 cells. Similar data were obtained by flow cytometry. The positive rates of B7-H4 and HBx in the tissues of 83 HBV-HCC patients were 68.67% (57/83) and 59.04% (49/83), respectively. The expression of HBx was correlated with tumor node metastases (TNM) stage, and the expression of B7-H4 was positively correlated with HBx (rs = 0.388; P < 0.01). The expression level of B7-H4 in HBx-positive HBV-HCC tissues was substantially higher than that in HBx-negative HBV-HCC tissues. The expression level of B7H4 was negatively related to tumor TNM stage.
CONCLUSION: Higher expression of HBx and B7-H4 was correlated with tumor progression of HBV-HCC, suggesting that B7-H4 may be involved in facilitating HBV-related hepatocarcinogenesis.
Collapse
|
13
|
B7-H4 downregulation induces mitochondrial dysfunction and enhances doxorubicin sensitivity via the cAMP/CREB/PGC1-α signaling pathway in HeLa cells. Pflugers Arch 2015; 466:2323-38. [PMID: 24658911 DOI: 10.1007/s00424-014-1493-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 02/26/2014] [Accepted: 02/26/2014] [Indexed: 12/15/2022]
Abstract
B7-H4 is a B7 family coregulatory protein that inhibits T cell-mediated immunity. B7-H4 is overexpressed in various cancers; however, the functional role of B7-H4 in cancer metabolism is poorly understood. Because mitochondria play pivotal roles in development, proliferation, and death of cancer cells, we investigated molecular and functional alterations of mitochondria in B7-H4-depleted HeLa cells. In a human study, overexpression of B7-H4 was confirmed in the cervices of adenocarcinoma patients (n = 3) compared to noncancer patients (n = 3). In the cell line model, B7-H4 depletion was performed by transfection with small interfering RNA (siRNA). B7-H4 depletion suppressed oxygen consumption rate, ATP production, and mitochondrial membrane potential and mass and increased reactive oxygen species production. In particular, electron transport complex III activity was significantly impaired in siB7-H4-treated cells. Coincidently, depletion of B7-H4 suppressed major mitochondrial regulators (peroxisome proliferator-activated receptor gamma coactivator 1-alpha [PGC1-α] and mitochondrial transcription factor A), a component of oxidative phosphorylation (ubiquinol-cytochrome c reductase core protein 1), and an antiapoptosis protein (Bcl-XL). Mitochondrial dysfunction in siRNA-treated cells significantly augmented oxidative stress, which strongly activated the JNK/P38/caspase axis in the presence of doxorubicin, resulting in increased apoptotic cell death. Investigating the mechanism of B7-H4-mediated mitochondrial modulation, we found that B7-H4 depletion significantly downregulated the cAMP/cAMP response element-binding protein/PGC1-α signaling pathway. Based on these findings, we conclude that B7-H4 has a role in the regulation of mitochondrial function, which is closely related to cancer cell physiology and drug sensitivity.
Collapse
|
14
|
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
|
15
|
Frey AB. Suppression of T cell responses in the tumor microenvironment. Vaccine 2015; 33:7393-7400. [PMID: 26403368 DOI: 10.1016/j.vaccine.2015.08.096] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 07/06/2015] [Accepted: 08/05/2015] [Indexed: 12/29/2022]
Abstract
The immune system recognizes protein antigens expressed in transformed cells evidenced by accumulation of antigen-specific T cells in tumor and tumor draining lymph nodes. However, despite demonstrable immune response, cancers grow progressively suggesting that priming of antitumor immunity is insufficiently vigorous or that antitumor immunity is suppressed, or both. Compared to virus infection, antitumor T cells are low abundance that likely contributes to tumor escape and enhancement of priming is a long-sought goal of experimental vaccination therapy. Furthermore, patient treatment with antigen-specific T cells can in some cases overcome deficient priming and cause tumor regression supporting the notion that low numbers of T cells permits tumor outgrowth. However, tumor-induced suppression of antitumor immune response is now recognized as a significant factor contributing to cancer growth and reversal of the inhibitory influences within the tumor microenvironment is a major research objective. Multiple cell types and factors can inhibit T cell functions in tumors and may be grouped in two general classes: T cell intrinsic and T cell extrinsic. T cell intrinsic factors are exemplified by T cell expression of cell surface inhibitory signaling receptors that, after contact with cells expressing a cognate ligand, inactivate proximal T Cell Receptor-mediated signal transduction therein rendering T cells dysfunctional. T cell extrinsic factors are more diverse in nature and are produced by tumors and various non-tumor cells in the tumor microenvironment. These include proteins secreted by tumor or stromal cells, highly reactive soluble oxygen and nitrogen species, cytokines, chemokines, gangliosides, and toxic metabolites. These factors may restrict T cell entrance into the tumor parenchyma, cause inactivation of effector phase T cell functions, or induce T cell apoptosis ultimately causing diminished cancer elimination. Here, we review the contributions of inhibitory factors to tumor T cell dysfunction leading to tumor escape.
Collapse
Affiliation(s)
- Alan B Frey
- Department of Cell Biology, Perlmutter Cancer Center, New York University Langone School of Medicine, 550 First Avenue, New York, NY 10016, USA.
| |
Collapse
|
16
|
Kim D, Park GB, Hur DY. Apoptotic signaling through reactive oxygen species in cancer cells. World J Immunol 2014; 4:158-173. [DOI: 10.5411/wji.v4.i3.158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 06/03/2014] [Accepted: 10/16/2014] [Indexed: 02/05/2023] Open
Abstract
Reactive oxygen species (ROS) take part in diverse biological processes like cell growth, programmed cell death, cell senescence, and maintenance of the transformed state through regulation of signal transduction. Cancer cells adapt to new higher ROS circumstance. Sometimes, ROS induce cancer cell proliferation. Meanwhile, elevated ROS render cancer cells vulnerable to oxidative stress-induced cell death. However, this prominent character of cancer cells allows acquiring a resistance to oxidative stress conditions relative to normal cells. Activated signaling pathways that increase the level of intracellular ROS in cancer cells not only render up-regulation of several genes involved in cellular proliferation and evasion of apoptosis but also cause cancer cells and cancer stem cells to develop a high metabolic rate. In over the past several decades, many studies have indicated that ROS play a critical role as the secondary messenger of tumorigenesis and metastasis in cancer from both in vitro and in vivo. Here we summarize the role of ROS and anti-oxidants in contributing to or preventing cancer. In addition, we review the activated signaling pathways that make cancer cells susceptible to death.
Collapse
|
17
|
Guo G, Cao D, Xu H, Ruan Z, Fei L, Xie Z, Wu Y, Chen Y. The characteristic expression of B7-H3 and B7-H4 in liver biopsies from patients with HBV-related acute-on-chronic liver failure. Pathol Int 2014; 62:665-74. [PMID: 23005593 DOI: 10.1111/j.1440-1827.2012.02856.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Hepatitis B virus (HBV) is a major public health problem, and HBV-related acute-on-chronic liver failure (HBV-ACLF) has an extremely poor prognosis due to a lack of effective treatments. B7-H3 and B7-H4 are two novel members of the B7 superfamily that are actively involved in regulating the pathogenesis of infectious diseases. However, the intrahepatic expression of both members in HBV-ACLF patients has yet to be described. In this study, we analyzed the expression of B7-H3 and B7-H4 in HBV-ACLF biopsies by immunohistochemistry. Our results showed that both members were observed in all HBV-ACLF samples, and their expression was chiefly observed on infiltrating inflammatory cells and the damaged bile ducts. Immunofluorescence double staining showed that B7-H4 was expressed chiefly on CD3(+) T cells, CD68(+) macrophages, CK-18(+) bile ducts, and CD31(+) endothelial cells, while B7-H3 was found on all cell types detected. The expression of the programmed death (PD)-1 ligands, PD-L1 and PD-L2, was also detected in these liver tissues and they were found to be co-expressed with B7-H3 and B7-H4. These results suggest that the B7-family signaling is most likely to affect the pathogenesis of this disease, and a clear understanding of their functional roles may further elucidate the disease process.
Collapse
Affiliation(s)
- Guoning Guo
- Department of Emergency, South-West Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | |
Collapse
|
18
|
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
|
19
|
Abstract
B7-H4 is a ligand in the B7 costimulatory family, executing suppressive function on the immune system in many diseases, such as cancer, allograft rejection, and autoimmune diseases. The receptor for this molecule has yet to be clarified. The engagement of B7-H4 inhibits proliferation of immune cells by stopping the cell cycle at the G0/G1 phase and leads to apoptosis via the Fas/FasL pathway consequently accelerating tumor progression and alleviating allograft rejection. The pathogenic role of B7-H4 in tumors has been widely established, but few studies have focused on its function in other disorders. Here, we review recent advances in our understanding of B7-H4 biology in disease settings other than tumors and document the beneficial values to treat those diseases by targeting this molecule and related signaling pathways.
Collapse
|
20
|
Ceeraz S, Nowak EC, Noelle RJ. B7 family checkpoint regulators in immune regulation and disease. Trends Immunol 2013; 34:556-63. [PMID: 23954143 PMCID: PMC3821798 DOI: 10.1016/j.it.2013.07.003] [Citation(s) in RCA: 235] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 07/19/2013] [Accepted: 07/19/2013] [Indexed: 12/14/2022]
Abstract
Fine-tuning the immune response and maintaining tolerance to self-antigens involves a complex network of co-stimulatory and co-inhibitory molecules. The recent FDA approval of ipilimumab, a monoclonal antibody blocking cytotoxic T lymphocyte antigen (CTLA)-4, demonstrates the impact of checkpoint regulators in disease. This is reinforced by ongoing clinical trials targeting not only CTLA-4, but also the programmed death (PD)-1 and B7-H4 pathways in various disease states. Recently, two new B7 family inhibitory ligands, V-domain Ig suppressor of T cell activation (VISTA) and B7-H6 were identified. Here, we review recent understanding of B7 family members and their concerted regulation of the immune response to either self or foreign pathogens. We also discuss clinical developments in targeting these pathways in different disease settings, and introduce VISTA as a putative therapeutic target.
Collapse
Affiliation(s)
- Sabrina Ceeraz
- Geisel School of Medicine at Dartmouth, Department of Microbiology and Immunology, Norris Cotton Cancer Centre, 1 Medical Center Drive, Lebanon, New Hampshire 03756, USA
| | | | | |
Collapse
|
21
|
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
|
22
|
Leung J, Suh WK. Host B7-H4 Regulates Antitumor T Cell Responses through Inhibition of Myeloid-Derived Suppressor Cells in a 4T1 Tumor Transplantation Model. THE JOURNAL OF IMMUNOLOGY 2013; 190:6651-61. [DOI: 10.4049/jimmunol.1201242] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
23
|
Chen Y, Guo G, Guo S, Shimoda S, Shroyer KR, Tang Y, Wu Y. Intracellular B7-H4 suppresses bile duct epithelial cell apoptosis in human primary biliary cirrhosis. Inflammation 2012; 34:688-97. [PMID: 21120594 DOI: 10.1007/s10753-010-9280-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The expression and function of B7-H4, a recently identified co-inhibitory molecule of the B7 superfamily, in the pathogenesis of primary biliary cirrhosis (PBC) is still unclear. Here the expression of B7-H4 in sections from PBC patients (n = 16) was examined by immunohistochemistry and it was detected in primary bile duct epithelial cells (BECs) which were isolated from PBC patients by flow cytometry (FACs). Moreover, we also analyzed BECs-associated B7-H4 function through knock-down of its expression via RNA interference (RNAi) in vitro. Immunohistochemistry and FACs evidenced that the expression of B7-H4 was restricted in the cytoplasm of BECs from PBC patients, while it was completely absent in normal liver tissues. The cytoplasmic B7-H4 gene was cloned, and sequenced analysis showed it was encoded by the same gene to the membrane B7-H4. Interesting, silencing B7-H4 by specific RNAi resulted in enhanced FasL expression and BEC apoptosis. Conversely, interruption of Fas\FasL interaction with using FasL blocking antibodies (clone 4H9) reversed cell apoptosis. Our results suggested that the intracellular B7-H4 appears to prevent Fas/FasL-mediated BEC apoptosis during the progression of PBC, and indicates B7-H4 is a possible target for therapeutic intervention of this disease.
Collapse
Affiliation(s)
- Yongwen Chen
- Institute of Immunology, PLA, Third Military Medical University, Chongqing, 400038, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
24
|
Wilcox RA, Ansell SM, Lim MS, Zou W, Chen L. The B7 homologues and their receptors in hematologic malignancies. Eur J Haematol 2012; 88:465-75. [PMID: 22372959 DOI: 10.1111/j.1600-0609.2012.01766.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The B7 homologues and their receptors regulate both peripheral tolerance and adaptive immunity. This field is rapidly evolving as new ligands and receptors are being identified. Much of the work supporting their role in the regulation of host anti-tumor immunity has been derived from experimental models and clinical trials in solid malignancies. However, a growing body of evidence demonstrates that the B7-H family has important immunologic and non-immunologic functions in a variety of hematologic malignancies. Herein, we will review recent evidence that supports the therapeutic targeting of the B7 homologues in hematologic malignancies.
Collapse
Affiliation(s)
- Ryan A Wilcox
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI 48109-5948, USA.
| | | | | | | | | |
Collapse
|
25
|
He C, Qiao H, Jiang H, Sun X. The inhibitory role of b7-h4 in antitumor immunity: association with cancer progression and survival. Clin Dev Immunol 2011; 2011:695834. [PMID: 22013483 PMCID: PMC3195678 DOI: 10.1155/2011/695834] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 08/08/2011] [Indexed: 01/05/2023]
Abstract
B7-H4 is one of the most recently identified members of B7 superfamily of costimulatory molecules serving as an inhibitory modulator of T-cell response. B7-H4 is broadly expressed in human peripheral tissues and inducibly expressed in immune cells. The expression of B7-H4 has been observed in various types of human cancer tissues, and its soluble form has been detected in blood samples from cancer patients. However, its precise physiological role is still elusive, as its receptor has not been identified and the expression levels are not consistent. This paper summarizes the pertinent data on the inhibitory role of B7-H4 in antitumor immunity and its association with cancer progression and survival in human patients. The paper also discusses the clinical significance of investigating B7-H4 as potential markers for cancer diagnosis and prognosis, and as therapeutic targets.
Collapse
Affiliation(s)
- Changjun He
- The Hepatosplenic Surgery Center, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Haiquan Qiao
- The Hepatosplenic Surgery Center, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Hongchi Jiang
- The Hepatosplenic Surgery Center, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xueying Sun
- The Hepatosplenic Surgery Center, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1005, New Zealand
| |
Collapse
|
26
|
Guo G, Shang Y, Zhu G, Bao X, Xu S, Chen Y. The expression and distribution of immunomodulatory proteins B7-H1, B7-DC, B7-H3, and B7-H4 in rheumatoid synovium. Clin Rheumatol 2011; 31:271-81. [PMID: 21833687 DOI: 10.1007/s10067-011-1815-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 06/08/2011] [Accepted: 07/12/2011] [Indexed: 12/31/2022]
Abstract
CD28/B7 signals have been shown to have the capacity to regulate T cell activation and participate in regulating the development of rheumatoid arthritis (RA). However, the expression and anatomical distribution of some members of the B7 superfamily including B7-H1, B7-DC, B7-H3 and B7-H4 in RA synovium is still unclear. We analyzed the expression of these molecules in synovial tissues from RA patients. Immunohistochemistry showed that all of these molecules were observed in synovium. On the cellular level, all of them were found on cell membrane and in cytoplasma. The expression of B7-DC and B7-H3 was major on capillaries, synovicytes and infiltrated inflammatory cells in the lining layer, while B7-H1 and B7-H4 were detected in some inflammatory cells residing in the sublining and lining layer. Fluorescent dual staining indicated that all these molecules were principally associated with CD31(+) endothelial cells and CD68(+) macrophages. In addition, B7-H1 and B7-H3 were also observed on CD3(+) T cells (including CD4(+) and CD8(+) T cells). Interestingly, B7-H1/B7-H4, B7-H3/B7-DC were co-expressed on the same cells. The characteristic expression and distribution of these molecules in synovium indicated that they probably have different effects during the progress of RA, and a clear understanding of their functional roles may further elucidate the pathogenesis of this disease.
Collapse
Affiliation(s)
- Guoning Guo
- Department of Emergency, South-West Hospital, Third Military Medical University, Chongqing, 400038, China
| | | | | | | | | | | |
Collapse
|
27
|
Chen LJ, Sun J, Wu HY, Zhou SM, Tan Y, Tan M, Shan BE, Lu BF, Zhang XG. B7-H4 expression associates with cancer progression and predicts patient's survival in human esophageal squamous cell carcinoma. Cancer Immunol Immunother 2011; 60:1047-55. [PMID: 21519829 PMCID: PMC11029661 DOI: 10.1007/s00262-011-1017-3] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 04/07/2011] [Indexed: 12/12/2022]
Abstract
A retrospective cohort study including 112 patients suffering from esophageal squamous cell carcinoma (ESCC) was performed to investigate the expression of B7-H4 in ESCC and determine its association with patient's clinicopathological parameters and survival. Expression levels of B7-H4 on tumor cells and densities of tumor infiltrating lymphocytes (TILs) in the surgical specimens of ESCC tissues were characterized using immunohistochemical assays. Uni- and multivariate analyses were performed to evaluate the prognostic value of B7-H4 expression levels and densities of TILs in tumor sections. Positive B7-H4 immunostaining was observed in 107 of 112 (95.5%) of ESCC tissue sections. We further divided all patients into two major subgroups, a lower B7-H4 expression group with 46 patients and a higher B7-H4 expression group with 66 patients. We found that expression levels of B7-H4 on tumor cells were significantly correlated with patient's gender (P = 0.0288), distant metastasis (P = 0.0500), and TNM stage (P = 0.0258). Moreover, tumor cell B7-H4 expression was inversely correlated with densities of CD3(+) T cells in tumor nest (P = 0.0424) and CD8(+) T cells in tumor stroma (P = 0.0229). The overall survival rate of the patients with higher B7-H4 expression was significantly worse than that of the patients with lower B7-H4 expression (P = 0.0105, Hazard Ratio: 1.854, 95%CI:1.152-2.902). Markers of cell-mediated immune responses such as CD3, CD8, and T-bet were associated with better patient survival. The present study demonstrated that B7-H4 expression in human ESCC is associated with cancer progression, reduced tumor immunosurveillance and worse patient outcomes. B7-H4 can serve as a novel prognostic predictor for human ESCC and a potential target for the immune therapy against this malignancy.
Collapse
Affiliation(s)
- Lu-jun Chen
- Institute of Medical Biotechnology, Medical College of Suzhou University, 708 Renmin Road, 215007 Suzhou, Jiangsu China
| | - Jing Sun
- Institute of Medical Biotechnology, Medical College of Suzhou University, 708 Renmin Road, 215007 Suzhou, Jiangsu China
| | - Hong-ya Wu
- Clinical Immunology Laboratory, The First Affiliated Hospital of Suzhou University, Suzhou, Jiangsu China
| | - Shu-ming Zhou
- Department of Tumor Biological Treatment, Yixing Tumor Hospital, Wuxi, Jiangsu China
| | - Yan Tan
- Department of Pathology, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu China
| | - Ming Tan
- Department of Tumor Biological Treatment, Yixing Tumor Hospital, Wuxi, Jiangsu China
| | - Bao-en Shan
- Tumor Research Institute, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei China
| | - Bin-feng Lu
- Department of Immunology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213 USA
| | - Xue-guang Zhang
- Institute of Medical Biotechnology, Medical College of Suzhou University, 708 Renmin Road, 215007 Suzhou, Jiangsu China
| |
Collapse
|
28
|
Quandt D, Fiedler E, Boettcher D, Marsch WC, Seliger B. B7-h4 expression in human melanoma: its association with patients' survival and antitumor immune response. Clin Cancer Res 2011; 17:3100-11. [PMID: 21378130 DOI: 10.1158/1078-0432.ccr-10-2268] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Cancers have developed a number of strategies to escape immune responses including the differential expression of costimulatory molecules of the B7 family. B7-H3 and B7-H4 have recently been described in different tumor entities but the relevance for melanoma has not yet been studied so far. EXPERIMENTAL DESIGN Using immunohistochemistry, B7-H3 and B7-H4 expression was studied on 29 melanoma lesions. Survival curves and log-rank tests were used to test the association of protein expression with survival. Cell lines were evaluated for B7-H3 and B7-H4 expression by PCR and flow cytometry. Functional T-cell-tumor coculture assays were carried out with in vitro generated tumor transfectants. RESULTS B7-H3 and B7-H4 expression was detected in primary tumor lesions (29 of 29 and 28 of 29) and in metastases (28 of 29 and 26 of 29). The numbers of CD68(+) macrophages were significantly lower in patients with low B7-H4 expression, whereas CD8(+) T-cell infiltrates were independent of expression levels. Furthermore, a survival benefit for patients with B7-H4 low expressing melanoma was found, whereas B7-H3 was not associated with any clinical parameter. All 23 melanoma cell lines analyzed expressed B7-H3 and B7-H4 mRNA and protein, but B7-H4 was restricted to intracellular compartments. On silencing of B7-H3 by specific shRNA tumor-associated antigen-specific T cell responses were unaltered. Overexpression of B7-H4 on melanoma cells did not alter the cytotoxicity of different CD8(+) effector cells, but drastically inhibited cytokine production. CONCLUSIONS Our study provides for the first time evidence of B7-H4 expression on melanoma cells as a mechanism controlling tumor immunity which is associated with patients' survival.
Collapse
Affiliation(s)
- Dagmar Quandt
- Martin Luther University Halle-Wittenberg, Institute of Medical Immunology, Halle, Germany
| | | | | | | | | |
Collapse
|
29
|
Petroff MG, Perchellet A. B7 family molecules as regulators of the maternal immune system in pregnancy. Am J Reprod Immunol 2010; 63:506-19. [PMID: 20384620 DOI: 10.1111/j.1600-0897.2010.00841.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Placental and fetal growth and development are associated with chronic exposure of the maternal immune system to fetally derived, paternally inherited antigens. Because maternal lymphocytes are aware of fetal antigens, active tolerance mechanisms are required to ensure unperturbed progression of pregnancy and delivery of a healthy newborn. These mechanisms of tolerance may include deletion, receptor downregulation, and anergy of fetal antigen-specific cells in lymphoid tissues, as well as regulation at the maternal-fetal interface by a variety of locally expressed immunoregulatory molecules. The B7 family of costimulatory molecules comprises one group of immunoregulatory molecules present in the decidua and placenta. B7 family members mediate both inhibitory and stimulatory effects on T-cell activation and effector functions and may play a critical role in maintaining tolerance to the fetus. Here, we review the known functions of the B7 family proteins in pregnancy.
Collapse
Affiliation(s)
- Margaret G Petroff
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | | |
Collapse
|
30
|
Park GB, Song H, Kim YS, Sung M, Ryu JW, Lee HK, Cho DH, Kim D, Lee WJ, Hur DY. Cell cycle arrest induced by engagement of B7-H4 on Epstein-Barr virus-positive B-cell lymphoma cell lines. Immunology 2010; 128:360-8. [PMID: 20067536 DOI: 10.1111/j.1365-2567.2009.03111.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
B7-H4 is a recently discovered B7 family member that has inhibitory effects on T-cell immunity. However, the reverse signalling mechanism of the B7-H4-expressing cells remains unclear. Previous work has shown that B7-H4 expression was enhanced on B cells following Epstein-Barr virus (EBV) infection, and engagement of cell-surface-expressed B7-H4 induces cell death of EBV-transformed B cells. Here we found that B7-H4 was constitutively expressed on EBV-positive lymphoma cells, Raji and IM-9 cells, but was not expressed on EBV-negative lymphoma cells (Ramos). Engagement of B7-H4 significantly reduced cell growth of Raji and IM-9 cells and resulted in cell cycle arrest at G0-G1 phase in a dose- and time-dependent manner. To clarify the mechanism of cell cycle arrest via activation of B7-H4, cell cycle regulatory factors were examined by reverse transcription-polymerase chain reaction and immunoblotting. We found that B7-H4 triggered down-regulation of CDK4/6 and up-regulation of p21 expression at both protein and RNA levels. Furthermore, CDK2 and cyclin E/D expression was down-regulated by B7-H4 triggering. Additionally, the down-regulation of phospho-AKT and phospho-cyclin E were clearly detected in B7-H4-activated Raji cells, but the phosphorylation of p53 was constitutively maintained. These results indicate that B7-H4-mediated signalling on EBV-positive B-cell lymphoma cells modulates the cell cycle through down-regulation of the AKT pathway. Consequently, B7-H4 may be a new potential target for use in EBV-positive lymphoma therapy.
Collapse
Affiliation(s)
- Ga Bin Park
- Department of Anatomy and Tumour Immunology, Inje University College of Medicine, Busan, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
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
|
32
|
Seliger B, Marincola FM, Ferrone S, Abken H. The complex role of B7 molecules in tumor immunology. Trends Mol Med 2008; 14:550-9. [PMID: 18986838 DOI: 10.1016/j.molmed.2008.09.010] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2008] [Revised: 09/29/2008] [Accepted: 09/29/2008] [Indexed: 12/31/2022]
Abstract
T-cell activation requires the interaction of the T-cell receptor with a cognate major histocompatibility complex (MHC)-peptide complex. Initiated by antigen engagement, the adaptive immune response is orchestrated by a complex balance between stimulatory and inhibitory signals that are predominantly controlled by members of the B7 family. Here, we review the current knowledge on B7 family members concerning their constitutive and regulated expression, modulation of the immune response and their role in the evasion of host immune surveillance. We also discuss recent therapeutic strategies that aim to improve immune-cell recognition of tumors and induce tolerance to autoreactive immune responses in normal tissues by manipulating B7 functions.
Collapse
Affiliation(s)
- Barbara Seliger
- Martin Luther University, Institute of Medical Immunology, 06112 Halle, Germany.
| | | | | | | |
Collapse
|