1
|
Lee S, Kim JH, Jang IH, Jo S, Lee SY, Oh SC, Kim SM, Kong L, Ko J, Kim TD. Harnessing B7-H6 for Anticancer Immunotherapy: Expression, Pathways, and Therapeutic Strategies. Int J Mol Sci 2024; 25:10326. [PMID: 39408655 PMCID: PMC11476788 DOI: 10.3390/ijms251910326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Cancer therapies have evolved from traditional chemotherapy to more precise molecular-targeted immunotherapies, which have been associated with improved side effects and outcomes. These modern strategies rely on cancer-specific biomarkers that differentiate malignant from normal cells. The B7 family of immune checkpoint molecules is crucial for cancer immune evasion and a prime therapeutic target. B7-H6, a recently identified member of the B7 family, has emerged as a promising therapeutic target. Unlike other B7 proteins, B7-H6 is not expressed in healthy tissues but is upregulated in several cancers. It binds to NKp30, activating natural killer (NK) cells and triggering immune responses against cancer cells. This review explores the expression of B7-H6 in different cancers, the factors that regulate its expression, and its intrinsic and extrinsic pathways. Additionally, we discuss potential anticancer therapies targeting B7-H6, highlighting its significance in advancing precision medicine. Understanding the role of B7-H6 in cancer immunity may inform the development of appropriate therapies that exploit its cancer-specific expression.
Collapse
Affiliation(s)
- Sunyoung Lee
- Center for Cell and Gene Therapy, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; (S.L.); (J.H.K.); (I.-H.J.); (S.J.); (S.Y.L.); (S.-C.O.); (S.-M.K.); (L.K.)
- Division of Life Sciences, Korea University, Seoul 02841, Republic of Korea;
| | - Ji Hyun Kim
- Center for Cell and Gene Therapy, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; (S.L.); (J.H.K.); (I.-H.J.); (S.J.); (S.Y.L.); (S.-C.O.); (S.-M.K.); (L.K.)
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - In-Hwan Jang
- Center for Cell and Gene Therapy, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; (S.L.); (J.H.K.); (I.-H.J.); (S.J.); (S.Y.L.); (S.-C.O.); (S.-M.K.); (L.K.)
| | - Seona Jo
- Center for Cell and Gene Therapy, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; (S.L.); (J.H.K.); (I.-H.J.); (S.J.); (S.Y.L.); (S.-C.O.); (S.-M.K.); (L.K.)
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Soo Yun Lee
- Center for Cell and Gene Therapy, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; (S.L.); (J.H.K.); (I.-H.J.); (S.J.); (S.Y.L.); (S.-C.O.); (S.-M.K.); (L.K.)
| | - Se-Chan Oh
- Center for Cell and Gene Therapy, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; (S.L.); (J.H.K.); (I.-H.J.); (S.J.); (S.Y.L.); (S.-C.O.); (S.-M.K.); (L.K.)
| | - Seok-Min Kim
- Center for Cell and Gene Therapy, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; (S.L.); (J.H.K.); (I.-H.J.); (S.J.); (S.Y.L.); (S.-C.O.); (S.-M.K.); (L.K.)
| | - Lingzu Kong
- Center for Cell and Gene Therapy, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; (S.L.); (J.H.K.); (I.-H.J.); (S.J.); (S.Y.L.); (S.-C.O.); (S.-M.K.); (L.K.)
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Jesang Ko
- Division of Life Sciences, Korea University, Seoul 02841, Republic of Korea;
| | - Tae-Don Kim
- Center for Cell and Gene Therapy, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; (S.L.); (J.H.K.); (I.-H.J.); (S.J.); (S.Y.L.); (S.-C.O.); (S.-M.K.); (L.K.)
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| |
Collapse
|
2
|
Bahojb Mahdavi SZ, Pouladi N, Amini M, Baradaran B, Najafi S, Vaghef Mehrabani S, Yari A, Ghobadi Alamdari S, Mokhtarzadeh AA. Let-7a-3p overexpression increases chemosensitivity to carmustine and synergistically promotes autophagy and suppresses cell survival in U87MG glioblastoma cancer cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6903-6918. [PMID: 38587542 DOI: 10.1007/s00210-024-03060-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/18/2024] [Indexed: 04/09/2024]
Abstract
In terms of primary brain tumors, glioblastoma is one of the most aggressive and common brain tumors. The high resistance of glioblastoma to chemotherapy has made it vital to find alternative treatments and biological mechanisms to reduce the survival of cancer cells. Given that, the objective of the present research was to explore the potential of let-7a-3p when used in combination with carmustine in human glioblastoma cancer cells. Based on previous studies, the expression of let-7a is downregulated in the U87MG cell line. Let-7a-3p transfected into U87MG glioblastoma cells. Cell viability of the cells was assessed by MTT assay. The apoptotic induction in U87MG cancerous cells was determined through the utilization of DAPI and Annexin V/PI staining techniques. Moreover, the induction of autophagy and cell cycle arrest was evaluated by flow cytometry. Furthermore, cell migration was evaluated by the wound healing assay while colony formation assay was conducted to evaluate colony formation. Also, the expression of the relevant genes was evaluated using qRT-PCR. Transfection of let-7a-3p mimic in U87MG cells increased the expression of the miRNA and also increased the sensitivity of U87MG cells to carmustine. Let-7a-3p and carmustine induced sub-G1 and S phase cell cycle arrest, respectively. Combination treatment of let-7a-3p and carmustine synergistically increased arrested cells and induced apoptosis through regulating involved genes including P53, caspase-3, Bcl-2, and Bax. Combined treatment with let-7a-3p and carmustine also induced autophagy and increased the expression of the ATG5 and Beclin 1 (ATG6). Furthermore, let-7a-3p combined with carmustine inhibited cell migration via decreasing the expression of MMP-2. Moreover, the combination therapy decreased the ability of U87MG to form colonies through downregulating CD-44. In conclusion, our work suggests that combining let-7a-3p replacement therapy with carmustine treatment could be considered a promising strategy in treatment and can increase efficiency of glioblastoma chemotherapy.
Collapse
Affiliation(s)
- Seyedeh Zahra Bahojb Mahdavi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Nasser Pouladi
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shiva Vaghef Mehrabani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Amirhossein Yari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sania Ghobadi Alamdari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Cell and Molecular Biology, Faculty of Basic Science, University of Maragheh, Maragheh, Iran
| | | |
Collapse
|
3
|
Chen X, Cui Y, Zou L. Treatment advances in high-grade gliomas. Front Oncol 2024; 14:1287725. [PMID: 38660136 PMCID: PMC11039916 DOI: 10.3389/fonc.2024.1287725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
High-grade gliomas (HGG) pose significant challenges in modern tumour therapy due to the distinct biological properties and limitations of the blood-brain barrier. This review discusses recent advancements in HGG treatment, particularly in the context of immunotherapy and cellular therapy. Initially, treatment strategies focus on targeting tumour cells guided by the molecular characteristics of various gliomas, encompassing chemotherapy, radiotherapy and targeted therapy for enhanced precision. Additionally, technological enhancements are augmenting traditional treatment modalities. Furthermore, immunotherapy, emphasising comprehensive tumour management, has gained widespread attention. Immune checkpoint inhibitors, vaccines and CAR-T cells exhibit promising efficacy against recurrent HGG. Moreover, emerging therapies such as tumour treating fields (TTFields) offer additional treatment avenues for patients with HGG. The combination of diverse treatments holds promise for improving the prognosis of HGG, particularly in cases of recurrence.
Collapse
Affiliation(s)
- Xi Chen
- Department of Radiotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yi Cui
- State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Liqun Zou
- Department of Medical Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Kamali AN, Bautista JM, Eisenhut M, Hamedifar H. Immune checkpoints and cancer immunotherapies: insights into newly potential receptors and ligands. Ther Adv Vaccines Immunother 2023; 11:25151355231192043. [PMID: 37662491 PMCID: PMC10469281 DOI: 10.1177/25151355231192043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 07/14/2023] [Indexed: 09/05/2023] Open
Abstract
Checkpoint markers and immune checkpoint inhibitors have been increasingly identified and developed as potential immunotherapeutic targets in various human cancers. Despite valuable efforts to discover novel immune checkpoints and their ligands, the precise roles of their therapeutic functions, as well as the broad identification of their counterpart receptors, remain to be addressed. In this context, it has been suggested that various putative checkpoint receptors can be induced upon activation. In the tumor microenvironment, T cells, as crucial immune response against malignant diseases as well as other immune central effector cells, such as natural killer cells, are regulated via co-stimulatory or co-inhibitory signals from immune or tumor cells. Studies have shown that exposure of T cells to tumor antigens upregulates the expression of inhibitory checkpoint receptors, leading to T-cell dysfunction or exhaustion. Although targeting immune checkpoint regulators has shown relative clinical efficacy in some tumor types, most trials in the field of cancer immunotherapies have revealed unsatisfactory results due to de novo or adaptive resistance in cancer patients. To overcome these obstacles, combinational therapies with newly discovered inhibitory molecules or combined blockage of several checkpoints provide a rationale for further research. Moreover, precise identification of their receptors counterparts at crucial checkpoints is likely to promise effective therapies. In this review, we examine the prospects for the application of newly emerging checkpoints, such as T-cell immunoglobulin and mucin domain 3, lymphocyte activation gene-3, T-cell immunoreceptor with Ig and ITIM domains (TIGIT), V-domain Ig suppressor of T-cell activation (VISTA), new B7 family proteins, and B- and T-cell lymphocyte attenuator, in association with immunotherapy of malignancies. In addition, their clinical and biological significance is discussed, including their expression in various human cancers, along with their roles in T-cell-mediated immune responses.
Collapse
Affiliation(s)
- Ali N. Kamali
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Simin Dasht Industrial Area, Karaj, Iran
- CinnaGen Research and Production Co., Alborz 3165933155, Iran
| | - José M. Bautista
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain
- Research Institute Hospital 12 de Octubre, Madrid, Spain
| | - Michael Eisenhut
- Department of Pediatrics, Luton and Dunstable University Hospital NHS Foundation Trust, Luton, UK
| | - Haleh Hamedifar
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
- CinnaGen Research and Production Co., Alborz, Iran
| |
Collapse
|
5
|
Allahyarzadeh Khiabani N, Amin Doustvandi M, Mohammadnejad F, Salmani Hassan Kohal E, Boushehri N, Jafarlou M, Baradaran B. Combination of B7H6-siRNA and temozolomide synergistically reduces stemness and migration properties of glioblastoma cancer cells. Exp Cell Res 2023:113667. [PMID: 37247720 DOI: 10.1016/j.yexcr.2023.113667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 05/20/2023] [Accepted: 05/25/2023] [Indexed: 05/31/2023]
Abstract
Glioblastoma multiforme (GBM) is among the malignant brain tumors of the central nervous system (CNS). The survival of this disease is about 14 months after diagnosis. To date, temozolomide is known as first-line therapy for glioma. Drug resistance and severe side effects against this drug are important obstacles to the effective treatment of this cancer. Small interfering RNA (siRNA) can adjust the expression of several genes and is used as a new method of gene therapy. Recent studies have shown that siRNAs can increase the sensitivity of cancer cells to chemotherapy drugs. This study aimed to understand the potential role and molecular mechanism of the combination therapy of B7H6-siRNA and temozolomide in glioblastoma cancer. U87 cells were treated with B7H6-siRNA and temozolomide, separately and in combination. Cell viability, stemness, cell migration, and apoptosis were measured. The results of this work presented the synergistic effect of B7H6-siRNA and temozolomide in inhibiting the cancerous features of the U87 cell line. Down-regulating B7H6-siRNA expression inhibited the cell viability of U87 glioblastoma cancer cells and increased their sensitivity to temozolomide. In addition, a noteworthy decrease in cell migration ability and stemness, an increase in apoptosis were observed in the combined groups compared to B7H6-siRNA and temozolomide individually. According to the results, a combination of B7H6-siRNA and temozolomide can be a promising strategy in glioblastoma cancer therapy.
Collapse
Affiliation(s)
- Nadia Allahyarzadeh Khiabani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Genetics, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Mohammad Amin Doustvandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Neda Boushehri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Jafarlou
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
6
|
Mohammadi A, Najafi S, Amini M, Baradaran B, Firouzamandi M. B7H6 silencing increases chemosensitivity to dacarbazine and suppresses cell survival and migration in cutaneous melanoma. Melanoma Res 2023; 33:173-183. [PMID: 37053079 DOI: 10.1097/cmr.0000000000000890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Cutaneous melanoma (CM) is a highly metastatic cancer whose incidence rate is heightening worldwide. B7H6, as one of the co-stimulatory ligands of the B7 family, is expressed in malignant cells, involved in tumorigenesis. This study aimed to investigate the significance of B7H6 in CM cell chemosensitivity and metastatic ability. A375 CM cells were transfected with B7H6-siRNA and treated with dacarbazine individually or combined. The MTT assay to estimate half-maximal inhibitory concentration of dacarbazine and cell viability, the apoptotic induction using Annexin V/PI, cell cycle progression via flow cytometry, and wound healing assay for determining the migration ability of cells and assessing the clonogenic potential of A375 cells were executed. Functional analyses were performed to evaluate changes in A375 cells. The results illustrated that B7H6 suppression significantly increased the chemosensitivity of A375 cells to dacarbazine. Apoptosis induction by dacarbazine was enhanced after B7H6 knockdown through modulating Caspase-3, Bax, and Bcl-2 mRNA levels. Western blotting indicated enhancement of cleaved caspase-3 protein expression in treatment groups. A375 cells were arrested at the sub-G1 and S phases when using B7H6-siRNA and dacarbazine. B7H6 suppression combined with dacarbazine restrained cell migration through suppression of matrix metalloproteinase (MMP) expression, including MMP2, MMP3, and MMP9. In addition, the clonogenic ability of A375 cells was decreased by downregulating Sox2, Nanog, and CD44 mRNA levels. A visible decrement in STAT3 protein expression was observed in the combination group. Hence, our findings revealed that B7H6 knockdown with dacarbazine could be a promising treatment approach for cutaneous melanoma.
Collapse
Affiliation(s)
- Alaleh Mohammadi
- Department of Pathobiology, Faculty of Veterinary Medicine, University of Tabriz
- Immunology Research Center
| | | | | | - Behzad Baradaran
- Immunology Research Center
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
7
|
Comprehensive characterization of B7 family members in NSCLC and identification of its regulatory network. Sci Rep 2023; 13:4311. [PMID: 36922519 PMCID: PMC10017798 DOI: 10.1038/s41598-022-26776-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 12/20/2022] [Indexed: 03/17/2023] Open
Abstract
B7 family members act as co-stimulatory or co-inhibitory molecules in the adaptive immune system. Thisstudy aimed to investigate the dysregulation, prognostic value and regulatory network of B7 family members in non-small cell lung cancer (NSCLC). Data for lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) patients were extracted from public databases. Patient prognosis was determined by Kaplan-Meier analysis. The downstream signaling pathways of B7 family were identified via GO and KEGG analysis. The key B7 related genes were selected by network, correlation and functional annotation analysis. Most B7 family members were dysregulated in LUAD and LUSC. The expression of B7-1/2/H3 and B7-H5 were significantly associated with overall survival in LUAD and LUSC, respectively. The major pathway affected by B7 family was the EGFR tyrosine kinase inhibitor resistance and ErbB signaling pathway. MAPK1, MAPK3 and MAP2K1 were pivotal B7 related genes in both LUAD and LUSC. This study reveals an overall dysregulation of B7 family members in NSCLC and highlights the potential of combination use of tyrosine kinase inhibitors or MEK/ERK inhibitors with B7 member blockade for NSCLC treatment.
Collapse
|
8
|
Fan J, Wang Y, Liang X, Peng Y, Li S, Li X, Zhou F, Li Y. B7-H6 enhances F-actin rearrangement by targeting c-MYC activation to promote medulloblastoma migration and invasion. Med Oncol 2023; 40:85. [PMID: 36692844 DOI: 10.1007/s12032-023-01947-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 01/08/2023] [Indexed: 01/25/2023]
Abstract
Medulloblastoma (MB) is children's most common primary malignant primitive neuro-ectodermal tumor. Group 3 MB showed a higher propensity to metastasis, which is molecularly characterized by c-MYC gene amplification. The activation of c-MYC promotes the remodeling of the F-actin cytoskeleton to enhance metastasis. The B7 homologue 6 (B7-H6) is associated with the manifold essential hallmarks of tumorigenesis. In this study, we will explore whether B7-H6 regulates the reorganization of F-actin by elevating the c-MYC expression to promote metastasis. The Daoy cell line was used to act as the cell model of medulloblastoma. Small interfering RNA and the plasmid were used to downregulate and upregulate the expression of B7-H6 in Daoy cells. Transwell assays with/without the matrigel matrix were used to detect migration and invasion of Daoy cells. Western blots were used to detect the expression of related proteins. Immunofluorescence staining was used to observe the impact of B7-H6 on the c-MYC /F-actin axis. B7-H6 improved migration and invasion in the Daoy cell line. B7-H6 enhanced the rearrangement of F-actin and activated the expression of MMP-9 and MMP-2. B7-H6 promoted the remodeling of F-actin by targeting c-MYC activation to reinforce migration and invasion. B7-H6 acts as a promoter of migration and invasion in medulloblastoma by activating the c-MYC /F-actin axis.
Collapse
Affiliation(s)
- Jianing Fan
- School of Medicine, Chongqing University, Chongqing, China.,Department of Pathology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Yangyang Wang
- Bioengineering College of Chongqing University, Chongqing, China.,Department of Pathology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Xiao Liang
- School of Medicine, Chongqing University, Chongqing, China.,Department of Pathology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Yan Peng
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Shijie Li
- Department of Pathology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Xiaoju Li
- Department of Pathology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Fanlin Zhou
- Department of Pathology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Yu Li
- School of Medicine, Chongqing University, Chongqing, China. .,Department of Pathology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China.
| |
Collapse
|
9
|
Wang Y, Li M, Wang G, Wu H. Role of B7 family members in glioma: Promising new targets for tumor immunotherapy. Front Oncol 2023; 12:1091383. [PMID: 36741734 PMCID: PMC9890054 DOI: 10.3389/fonc.2022.1091383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/15/2022] [Indexed: 01/19/2023] Open
Abstract
Glioma, is a representative type of intracranial tumor among adults, usually has a weak prognosis and limited treatment options. Traditional therapies, including surgery, chemotherapy, and radiotherapy, have had little impact on patient survival time. Immunotherapies designed to target the programmed cell death protein 1 (PD-1)/programmed death ligand 1 (PD-L1) signaling pathway have successfully treated various human cancers, informing the development of similar therapies for glioma. However, anti-PD-L1 response rates remain limited in glioma patients. Thus, exploring novel checkpoints targeting additional immunomodulatory pathways for activating durable antitumor immune responses and improving glioma outcomes is needed. Researchers have identified other B7 family checkpoint molecules, including PD-L2, B7-H2, B7-H3, B7-H4, and B7-H6. The current review article evaluates the expression of all 10 reported members of the B7 family in human glioma using The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) data, as well as summarizes studies evaluating the clinical meanings and functions of B7 family molecules in gliomas. B7 family checkpoints may contribute to different immunotherapeutic management options for glioma patients.
Collapse
Affiliation(s)
- Yan Wang
- Department of Radiation Oncology, Third People’s Hospital of Zhengzhou, Zhengzhou, Henan, China
| | - Mengxi Li
- Department of Radiation Oncology, Third People’s Hospital of Zhengzhou, Zhengzhou, Henan, China
| | - Gang Wang
- Department of Radiation Oncology, Third People’s Hospital of Zhengzhou, Zhengzhou, Henan, China,*Correspondence: Gang Wang, ; Hui Wu,
| | - Hui Wu
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China,*Correspondence: Gang Wang, ; Hui Wu,
| |
Collapse
|
10
|
Zhang X, Zhao L, Zhang H, Zhang Y, Ju H, Wang X, Ren H, Zhu X, Dong Y. The immunosuppressive microenvironment and immunotherapy in human glioblastoma. Front Immunol 2022; 13:1003651. [PMID: 36466873 PMCID: PMC9712217 DOI: 10.3389/fimmu.2022.1003651] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/01/2022] [Indexed: 08/09/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most malignant intracranial tumor in adults, characterized by extensive infiltrative growth, high vascularization, and resistance to multiple therapeutic approaches. Among the many factors affecting the therapeutic effect, the immunosuppressive GBM microenvironment that is created by cells and associated molecules via complex mechanisms plays a particularly important role in facilitating evasion of the tumor from the immune response. Accumulating evidence is also revealing a close association of the gut microbiota with the challenges in the treatment of GBM. The gut microbiota establishes a connection with the central nervous system through bidirectional signals of the gut-brain axis, thus affecting the occurrence and development of GBM. In this review, we discuss the key immunosuppressive components in the tumor microenvironment, along with the regulatory mechanism of the gut microbiota involved in immunity and metabolism in the GBM microenvironment. Lastly, we concentrate on the immunotherapeutic strategies currently under investigation, which hold promise to overcome the hurdles of the immunosuppressive tumor microenvironment and improve the therapeutic outcome for patients with GBM.
Collapse
Affiliation(s)
- Xuehua Zhang
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Leilei Zhao
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - He Zhang
- Department of Immunology, Qiqihar Medical University, Qiqihar, China
| | - Yurui Zhang
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Huanyu Ju
- Department of Immunology, Harbin Medical University, Harbin, China
| | - Xiaoyu Wang
- Department of Neurology, Hongda Hospital, Jinxiang, China
| | - Huan Ren
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xiao Zhu
- School of Computer and Control Engineering, Yantai University, Yantai, China
| | - Yucui Dong
- Department of Immunology, Binzhou Medical University, Yantai, China
| |
Collapse
|
11
|
Chen Y, Zheng A, Zhang Y, Xiao M, Zhao Y, Wu X, Li M, Du F, Chen Y, Chen M, Li W, Li X, Sun Y, Gu L, Xiao Z, Shen J. Dysregulation of B7 family and its association with tumor microenvironment in uveal melanoma. Front Immunol 2022; 13:1026076. [PMID: 36311731 PMCID: PMC9615147 DOI: 10.3389/fimmu.2022.1026076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/20/2022] [Indexed: 11/25/2022] Open
Abstract
Background Uveal melanoma (UVM) is the most common primary intraocular malignancy in adults with a poor prognosis. B7 family is an important modulator of the immune response. However, its dysregulation and underlying molecular mechanism in UVM still remains unclear. Methods Data were derived from TCGA and GEO databases. The prognosis was analyzed by Kaplan-Meier curve. The ESTIMATE algorithm, CIBERSORT algorithm, and TIMER database were used to demonstrate the correlation between B7 family and tumor immune microenvironment in UVM. Single-cell RNA sequencing was used to detect the expression levels of the B7 family in different cell types of UVM. UVM was classified into different types by consistent clustering. Enrichment analysis revealed downstream signaling pathways of the B7 family. The interaction between different cell types was visualized by cell chat. Results The expression level of B7 family in UVM was significantly dysregulated and negatively correlated with methylation level. The expression of B7 family was associated with prognosis and immune infiltration, and B7 family plays an important role in the tumor microenvironment (TME). B7 family members were highly expressed in monocytes/macrophages of UVM compared with other cell types. Immune response and visual perception were the main functions affected by B7 family. The result of cell chat showed that the interaction between photoreceptor cells and immune-related cells was mainly generated by HLA-C-CD8A. CABP4, KCNJ10 and RORB had the strongest correlation with HLA-C-CD8A, and their high expression was significantly correlated with poor prognosis. CABP4 and RORB were specifically expressed in photoreceptor cells. Conclusions Dysregulation of the B7 family in UVM is associated with poor prognosis and affects the tumor immune microenvironment. CABP4 and RORB can serve as potential therapeutic targets for UVM, which can be regulated by the B7 family to affect the visual perception and immune response function of the eye, thus influencing the prognosis of UVM.
Collapse
Affiliation(s)
- Yao Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Pidu District People’s Hospital, Chengdu, Sichuan, China
| | - Anfu Zheng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yao Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Mintao Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Meijuan Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Wanping Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Xiaobing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yuhong Sun
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Li Gu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Zhangang Xiao
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: Jing Shen, ; Zhangang Xiao,
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
- *Correspondence: Jing Shen, ; Zhangang Xiao,
| |
Collapse
|
12
|
Targeted Therapy of B7 Family Checkpoints as an Innovative Approach to Overcome Cancer Therapy Resistance: A Review from Chemotherapy to Immunotherapy. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27113545. [PMID: 35684481 PMCID: PMC9182385 DOI: 10.3390/molecules27113545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 11/17/2022]
Abstract
It is estimated that there were 18.1 million cancer cases worldwide in 2018, with about 9 million deaths. Proper diagnosis of cancer is essential for its effective treatment because each type of cancer requires a specific treatment procedure. Cancer therapy includes one or more approaches such as surgery, radiotherapy, chemotherapy, and immunotherapy. In recent years, immunotherapy has received much attention and immune checkpoint molecules have been used to treat several cancers. These molecules are involved in regulating the activity of T lymphocytes. Accumulated evidence shows that targeting immune checkpoint regulators like PD-1/PD-L1 and CTLA-4 are significantly useful in treating cancers. According to studies, these molecules also have pivotal roles in the chemoresistance of cancer cells. Considering these findings, the combination of immunotherapy and chemotherapy can help to treat cancer with a more efficient approach. Among immune checkpoint molecules, the B7 family checkpoints have been studied in various cancer types such as breast cancer, myeloma, and lymphoma. In these cancers, they cause the cells to become resistant to the chemotherapeutic agents. Discovering the exact signaling pathways and selective targeting of these checkpoint molecules may provide a promising avenue to overcome cancer development and therapy resistance. Highlights: (1) The development of resistance to cancer chemotherapy or immunotherapy is the main obstacle to improving the outcome of these anti-cancer therapies. (2) Recent investigations have described the involvement of immune checkpoint molecules in the development of cancer therapy resistance. (3) In the present study, the molecular participation of the B7 immune checkpoint family in anticancer therapies has been highlighted. (4) Targeting these immune checkpoint molecules may be considered an efficient approach to overcoming this obstacle.
Collapse
|
13
|
Peng G, Liu Y, Yang C, Shen C. MicroRNA-25 promotes cell proliferation, migration and invasion in glioma by directly targeting cell adhesion molecule 2. Exp Ther Med 2021; 23:16. [PMID: 34815768 PMCID: PMC8593921 DOI: 10.3892/etm.2021.10938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 08/19/2020] [Indexed: 12/11/2022] Open
Abstract
Numerous microRNAs (miRNAs/miRs) have been demonstrated to serve oncogenic or suppressive roles in glioma. Exploration of the underlying molecular mechanism of miRNAs in the development and progression of glioma is beneficial for the identification of novel therapeutic targets. In the present study, the function of miR-25 in glioma progression, as well as its underlying mechanism, were investigated. It was determined that miR-25 was significantly upregulated in glioma tissues and cell lines compared with normal brain tissues and cells, respectively. Furthermore, high expression levels of miR-25 were associated with an advanced clinical stage. The knockdown of miR-25 expression significantly reduced glioma cell proliferation, migration and invasion. Cell adhesion molecule 2 (CADM2) was identified as a direct target of miR-25 in glioma cells. Moreover, CADM2 expression level was significantly downregulated and inversely correlated with miR-25 expression level in glioma tissues, indicating that the expression of CADM2 was negatively regulated by miR-25. The inhibition of CADM2 expression counteracted the effects on glioma cell proliferation, migration and invasion caused by miR-25 downregulation. Furthermore, CADM2 knockdown considerably promoted the proliferation and migration of glioma cells. In summary, the present study demonstrated that miR-25 was significantly upregulated in glioma and that it promoted glioma cell proliferation, migration and invasion, at least partially, by directly targeting CADM2. These findings expanded the understanding of the molecular mechanism that underlies glioma progression.
Collapse
Affiliation(s)
- Gang Peng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yi Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Chenxing Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Chenfu Shen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
14
|
Bolandi N, Derakhshani A, Hemmat N, Baghbanzadeh A, Asadzadeh Z, Afrashteh Nour M, Brunetti O, Bernardini R, Silvestris N, Baradaran B. The Positive and Negative Immunoregulatory Role of B7 Family: Promising Novel Targets in Gastric Cancer Treatment. Int J Mol Sci 2021; 22:ijms221910719. [PMID: 34639059 PMCID: PMC8509619 DOI: 10.3390/ijms221910719] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 09/28/2021] [Accepted: 10/01/2021] [Indexed: 12/30/2022] Open
Abstract
Gastric cancer (GC), with a heterogeneous nature, is the third leading cause of death worldwide. Over the past few decades, stable reductions in the incidence of GC have been observed. However, due to the poor response to common treatments and late diagnosis, this cancer is still considered one of the lethal cancers. Emerging methods such as immunotherapy with immune checkpoint inhibitors (ICIs) have transformed the landscape of treatment for GC patients. There are presently eleven known members of the B7 family as immune checkpoint molecules: B7-1 (CD80), B7-2 (CD86), B7-H1 (PD-L1, CD274), B7-DC (PDCD1LG2, PD-L2, CD273), B7-H2 (B7RP1, ICOS-L, CD275), B7-H3 (CD276), B7-H4 (B7x, B7S1, Vtcn1), B7-H5 (VISTA, Gi24, DD1α, Dies1 SISP1), B7-H6 (NCR3LG1), B7-H7 (HHLA2), and Ig-like domain-containing receptor 2 (ILDR2). Interaction of the B7 family of immune-regulatory ligands with the corresponding receptors resulted in the induction and inhibition of T cell responses by sending co-stimulatory and co-inhibitory signals, respectively. Manipulation of the signals provided by the B7 family has significant potential in the management of GC.
Collapse
Affiliation(s)
- Nadia Bolandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia 571478334, Iran
| | - Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy
| | - Nima Hemmat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
| | - Mina Afrashteh Nour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia 571478334, Iran
| | - Oronzo Brunetti
- Medical Oncology Unit—IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
| | - Renato Bernardini
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95100 Catania, Italy;
| | - Nicola Silvestris
- Medical Oncology Unit—IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
- Department of Biomedical Sciences and Human Oncology (DIMO), University of Bari, 70124 Bari, Italy
- Correspondence: (N.S.); (B.B.); Tel.: +98-413-3371440 (B.B.); Fax: +98-413-3371311 (B.B.)
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 516615731, Iran
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran
- Correspondence: (N.S.); (B.B.); Tel.: +98-413-3371440 (B.B.); Fax: +98-413-3371311 (B.B.)
| |
Collapse
|
15
|
Dowlati A, Chan T. Pursuing Immunotherapeutic Targets in SCLC. J Thorac Oncol 2021; 16:1056-1057. [PMID: 34154789 DOI: 10.1016/j.jtho.2021.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 10/21/2022]
Affiliation(s)
- Afshin Dowlati
- University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, Ohio.
| | - Timothy Chan
- Center for Immunotherapy & Precision Immuno-Oncology, Cleveland Clinic Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
16
|
Liu X, Guo R, Xu Y. B7-H6 as a Diagnostic Biomarker for Cervical Squamous Cell Carcinoma. Genet Test Mol Biomarkers 2021; 25:463-470. [PMID: 34280008 DOI: 10.1089/gtmb.2020.0313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background: B7-H6, a newly discovered member of the immunoglobulin superfamily, exerts antitumor effects by binding to NKP30 receptor on natural killer cells; it has important clinical implications. Cell surface ectodomain shedding of B7-H6 generates soluble B7-H6 (sB7-H6), which is highly expressed and serves as a valuable biomarker in multiple tumors, but the clinical significance and diagnostic value of B7-H6 in cervical squamous cell carcinoma (CSCC) remains unclear. Objective: To assess the expression and diagnostic value of B7-H6 in CSCC. Methods: In this study, 69 cervical specimens were analyzed for B7-H6 expression: 25 paired CSCC tissues were examined using quantitative real-time polymerase chain reaction, and 24 paraffin-embedded CSCC tissues and 20 normal tissues were analyzed immunohistochemically. Furthermore, plasma samples from 30 CSCC patients and 24 healthy controls were examined using ELISA. Results: B7-H6 mRNA and protein levels were significantly higher in CSCC tissues than in adjacent normal cervical tissues (p < 0.05). Immunohistochemical analysis revealed that high B7-H6 expression correlated with stromal invasion (p = 0.043), lymphovascular space involvement (p = 0.005), lymph node metastasis (p = 0.019), and International Federation of Gynecology and Obstetrics (FIGO) stage (p = 0.002). Moreover, ELISA results demonstrated that the sB7-H6 concentration in peripheral blood was higher in CSCC patients than in healthy controls (p < 0.0001). Notably, at the optimal cutoff point of 0.076 ng/mL, sB7-H6 showed 93.3% sensitivity and 62.5% specificity in the discrimination of CSCC patients from healthy controls. Conclusions: B7-H6 mRNA and protein levels are markedly increased in CSCC tissues and peripheral blood samples, and the B7-H6 level can be used as a biomarker for predicting the severity of CSCC disease and discriminating CSCC patients from healthy controls.
Collapse
Affiliation(s)
- Xuejing Liu
- Department of Gynecology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Ruimeng Guo
- Department of Gynecology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yanying Xu
- Department of Gynecology, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
17
|
Guo R, Liu G, Li C, Liu X, Xu Y, Yang W, Wang F. B7 homolog 6 promotes the progression of cervical cancer. Exp Ther Med 2021; 22:774. [PMID: 34055073 PMCID: PMC8145428 DOI: 10.3892/etm.2021.10206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 04/12/2021] [Indexed: 12/16/2022] Open
Abstract
B7 homolog 6 (B7-H6) was recently discovered to act as a co-stimulatory molecule. In particular, the expression of B7-H6 has been found to play an important biological role in several types of tumors. The aim of the present study was to determine the role of B7-H6 in cervical cancer. Immunohistochemistry was used to analyze the expression levels of B7-H6 in cervical precancerous and cancerous tissues. Furthermore, the expression of B7-H6 was knocked down in HeLa cells using short hairpin RNA and the effects of B7-H6 on HeLa cell proliferation, migration and invasion were determined using Cell Counting Kit-8, colony formation, wound healing and Transwell invasion assays, respectively. In addition, flow cytometry was used to analyze the levels of cell apoptosis and the cell cycle distribution. The results of the immunohistochemical staining revealed that the expression levels of B7-H6 were upregulated in cervical lesions. Furthermore, the expression levels of B7-H6 were positively associated with the clinical stage of the cervical lesions. B7-H6 knockdown suppressed the invasive, migratory and proliferative abilities of HeLa cells, and promoted G1 cell cycle arrest and apoptosis. In conclusion, the findings of the present study suggested that B7-H6 may serve as a novel oncogene and may hold promise as a potential therapeutic target for cervical cancer.
Collapse
Affiliation(s)
- Ruimeng Guo
- Department of Gynecology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Guoyan Liu
- Department of Gynecology, The General Hospital of Tianjin Medical University, Tianjin 300053, P.R. China
| | - Changying Li
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Xuejing Liu
- Department of Gynecology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Yanying Xu
- Department of Gynecology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Weina Yang
- Department of Gynecology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Fang Wang
- Department of Gynecology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| |
Collapse
|
18
|
Baragaño Raneros A, Rodriguez RM, Bernardo Flórez A, Palomo P, Colado E, Minguela A, Suárez Álvarez B, López-Larrea C. Bromodomain protein BRD4 is an epigenetic activator of B7-H6 expression in acute myeloid leukemia. Oncoimmunology 2021; 10:1897294. [PMID: 33796404 PMCID: PMC8007156 DOI: 10.1080/2162402x.2021.1897294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 02/24/2021] [Indexed: 12/27/2022] Open
Abstract
B7-H6, a ligand for the NK activating receptor NKp30, has been identified as a biomarker of poor prognosis in several solid cancers. However, little is known about the role of B7-H6 and the mechanisms that control its expression in acute myeloid leukemia (AML). Epigenome modulation, including epigenomic reader dysregulation, is one of the hallmarks of AML. Bromodomain-containing protein 4 (BRD4), the best-known member of the BET family of epigenetic readers, is overexpressed in AML cells and regulates the transcription of genes involved in the pathogenesis of AML, as MYC oncogene. Here, we analyze the role of BRD4 in regulating B7-H6 in AML cells. Results demonstrated that the specific inhibition of BRD4 drastically reduces the expression of B7-H6 in AML cells. Histone acetylation mediated by CBP30/P300 facilitates the binding of BRD4 to the B7-H6 promoter, which recruits the P-TEFb elongation factor that phosphorylates RNA polymerase II, thereby activating B7-H6 transcription. BRD4 also co-bounded with JMJD6 at the distal enhancer of the B7-H6 gene. Metabolic modulation with metformin modifies the acetylation pattern in the B7-H6 promoter, impairing BRD4 binding, thereby inhibiting B7-H6 expression. B7-H6 knockdown induces the apoptosis in HEL-R cell line. Moreover, a high level of B7-H6 expression in AML patients is related to increased BRD4 levels, myelodysplastic-derived AML, and del5q, the two latter being associated with poor prognosis. Our data show that BRD4 is a positive regulator of the pro-tumorigenic molecule B7-H6 and that the blockage of the B7-H6 is a potential therapeutic target for the treatment of AML.
Collapse
Affiliation(s)
- Aroa Baragaño Raneros
- Translation Immunology Laboratory, Instituto De Investigación Biosanitaria Del Principado De Asturias-ISPA, Oviedo, Spain
| | - Ramon M Rodriguez
- Translation Immunology Laboratory, Instituto De Investigación Biosanitaria Del Principado De Asturias-ISPA, Oviedo, Spain
| | - Aida Bernardo Flórez
- Translation Immunology Laboratory, Instituto De Investigación Biosanitaria Del Principado De Asturias-ISPA, Oviedo, Spain
| | - Pilar Palomo
- Translation Immunology Laboratory, Instituto De Investigación Biosanitaria Del Principado De Asturias-ISPA, Oviedo, Spain
- Department of Hematology, Hospital Universitario Central De Asturias, Oviedo, Spain
| | - Enrique Colado
- Department of Hematology, Hospital Universitario Central De Asturias, Oviedo, Spain
- Department of Laboratory Medicine, Hospital Universitario Central De Asturias, Oviedo, Spain
| | - Alfredo Minguela
- Immunology Service, Instituto Murciano De Investigación Biosanitaria (IMIB), Hospital Clínico Universitario Virgen De La Arrixaca, Murcia, Spain
| | - Beatriz Suárez Álvarez
- Translation Immunology Laboratory, Instituto De Investigación Biosanitaria Del Principado De Asturias-ISPA, Oviedo, Spain
| | - Carlos López-Larrea
- Translation Immunology Laboratory, Instituto De Investigación Biosanitaria Del Principado De Asturias-ISPA, Oviedo, Spain
- Department of Immunology, Hospital Universitario Central De Asturias, Oviedo, Spain
| |
Collapse
|
19
|
Banu N, Riera-Leal A, Haramati J, Ortiz-Lazareno PC, Panikar SS, Bastidas-Ramirez BE, Gutierrez-Silerio GY, Solorzano-Ibarra F, Tellez-Bañuelos MC, Gutierrez-Franco J, Bueno-Topete MR, Pereira-Suarez AL, Del Toro-Arreola S. B7-H6, an immunoligand for the natural killer cell activating receptor NKp30, reveals inhibitory effects on cell proliferation and migration, but not apoptosis, in cervical cancer derived-cell lines. BMC Cancer 2020; 20:1083. [PMID: 33172426 PMCID: PMC7654602 DOI: 10.1186/s12885-020-07608-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 10/31/2020] [Indexed: 12/20/2022] Open
Abstract
Background Although great progress has been made in treatment regimens, cervical cancer remains as one of the most common cancer in women worldwide. Studies focusing on molecules that regulate carcinogenesis may provide potential therapeutic strategies for cervical cancer. B7-H6, an activating immunoligand expressed by several tumor cells, is known to activate NK cell-mediated cytotoxicity once engaged with its natural receptor NKp30. However, the opposite, that is, the effects in the tumor cell triggered by B7-H6 after interacting with NKp30 has not yet been well explored. Methods In this study, we evaluated the surface expression of B7-H6 by flow cytometry. Later, we stimulated B7-H6 positive cervical cancer derived-cell lines (HeLa and SiHa) with recombinant soluble NKp30 (sNKp30) protein and evaluated biological effects using the impedance RTCA system for cell proliferation, the scratch method for cell migration, and flow cytometry for apoptosis. Cellular localization of B7-H6 was determined using confocal microscopy. Results Notably, we observed that the addition of sNKp30 to the cervical cancer cell lines decreased tumor cell proliferation and migration rate, but had no effect on apoptosis. We also found that B7-H6 is selectively maintained in tumor cell lines, and that efforts to sort and purify B7-H6 negative or positive cells were futile, as negative cells, when cultured, regained the expression of B7-H6 and B7-H6 positive cells, when sorted and cultivated, lost a percentage of B7-H6 expression. Conclusions Our results suggest that B7-H6 has an important, as of yet undescribed, role in the biology of the cervical tumor cells themselves, suggesting that this protein might be a promising target for anti-tumor therapy in the future.
Collapse
Affiliation(s)
- Nehla Banu
- Instituto de Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Sierra Mojada # 950, Colonia Independencia, CP, 44340, Guadalajara, Jalisco, Mexico.,Laboratorio de Inmunología, Departamento de Fisiología, CUCS, Universidad de Guadalajara, Guadalajara, Mexico
| | - Annie Riera-Leal
- Institute for Regenerative Cures, Department of Dermatology, University of California-Davis, Davis, USA
| | - Jesse Haramati
- Laboratorio de Inmunobiología, Departamento de Biología Celular y Molecular, CUCBA, Universidad de Guadalajara, Guadalajara, Mexico
| | | | - Sandeep Surendra Panikar
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Blanca Estela Bastidas-Ramirez
- Instituto de Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Sierra Mojada # 950, Colonia Independencia, CP, 44340, Guadalajara, Jalisco, Mexico
| | - Gloria Yareli Gutierrez-Silerio
- Instituto de Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Sierra Mojada # 950, Colonia Independencia, CP, 44340, Guadalajara, Jalisco, Mexico.,Laboratorio de Inmunología, Departamento de Fisiología, CUCS, Universidad de Guadalajara, Guadalajara, Mexico
| | - Fabiola Solorzano-Ibarra
- Instituto de Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Sierra Mojada # 950, Colonia Independencia, CP, 44340, Guadalajara, Jalisco, Mexico
| | - Martha Cecilia Tellez-Bañuelos
- Laboratorio de Inmunobiología, Departamento de Biología Celular y Molecular, CUCBA, Universidad de Guadalajara, Guadalajara, Mexico
| | - Jorge Gutierrez-Franco
- Unidad Académica de Ciencias Químico Biológicas y Farmacéuticas, Universidad Autónoma de Nayarit, Tepic, Mexico
| | - Miriam Ruth Bueno-Topete
- Instituto de Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Sierra Mojada # 950, Colonia Independencia, CP, 44340, Guadalajara, Jalisco, Mexico
| | - Ana Laura Pereira-Suarez
- Laboratorio de Inmunología, Departamento de Fisiología, CUCS, Universidad de Guadalajara, Guadalajara, Mexico
| | - Susana Del Toro-Arreola
- Instituto de Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Sierra Mojada # 950, Colonia Independencia, CP, 44340, Guadalajara, Jalisco, Mexico. .,Laboratorio de Inmunología, Departamento de Fisiología, CUCS, Universidad de Guadalajara, Guadalajara, Mexico.
| |
Collapse
|
20
|
Anti-fouling SERS-based immunosensor for point-of-care detection of the B7–H6 tumor biomarker in cervical cancer patient serum. Anal Chim Acta 2020; 1138:110-122. [DOI: 10.1016/j.aca.2020.09.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 12/13/2022]
|
21
|
Yang S, Yuan L, Wang Y, Zhu M, Wang J, Ke X. B7-H6 Promotes Cell Proliferation, Migration and Invasion of Non-Hodgkin Lymphoma via Ras/MEK/ERK Pathway Based on Quantitative Phosphoproteomics Data. Onco Targets Ther 2020; 13:5795-5805. [PMID: 32606790 PMCID: PMC7308182 DOI: 10.2147/ott.s257512] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 05/28/2020] [Indexed: 01/01/2023] Open
Abstract
Purpose B7 homologue 6 (B7-H6) has been found at an up-regulated level in multiple cancer cells and identified to be positively correlated with inferior clinical features. In non-Hodgkin lymphoma (NHL), however, the roles of B7-H6 and the underlying mechanism of action remain unclear. Through in vivo and in vitro experiments, the aim of this study was to explore the regulatory mechanism of B7-H6 in NHL in order to provide new therapeutic strategies that can potentially be applied in clinical practice. Methods The expression of B7-H6 in T-lymphoblastic lymphoma (TLBL), diffuse large B cell lymphoma (DLBCL) and lymph node reactive hyperplasia (LRH) tissues were compared by immunohistochemistry. A total of 10 NHL cell lines were screened by Western blot to evaluate the expression of B7-H6. The effects of B7-H6 knockdown on cell proliferation, migration and invasion of NHL cells were studied in vivo using a transplanted tumor mice model, and in vitro by Cell Counting Kit-8 (CCK-8) and Transwell assays. Quantitative phosphoproteomics was performed to identify the changes of protein phosphorylation and related pathways affected by B7-H6. The effects of B7-H6 on NHL were validated via B7-H6 overexpression and pathway inhibitor assays. Results The expression levels of B7-H6 in NHL cell lines, and TLBL and DLBCL tissues were significantly increased compared with those in the control groups. Inhibition of cell proliferation, migration and invasion was observed in Jurkat and Raji cells with B7-H6 knockdown. The ability of B7-H6 in promoting tumorigenesis was further validated by in vivo experiments. In addition, Ras and HIF-1 signaling pathways were shown to be significantly affected by B7-H6 through quantitative phosphorylation proteomics analysis. Ras/MEK/ERK pathway was verified to be significantly inhibited after B7-H6 knockdown by Western blot analysis. Strikingly, MEK inhibitor AZD8330 was found to have the ability to sufficiently inhibit Ras/MEK/ERK pathway, partially reverse cell proliferation and completely reverse cell migration and invasion induced by B7-H6. Conclusion B7-H6 promotes cell proliferation, migration and invasion in NHL via Ras/MEK/ERK pathway. Hence, B7-H6 or Ras/MEK/ERK pathway targeting may be used as potential therapeutics for treating NHL.
Collapse
Affiliation(s)
- Siyuan Yang
- Department of Hematology and Lymphoma Research Center, Peking University Third Hospital, Beijing 100191, People's Republic of China
| | - Lei Yuan
- Department of Hematology and Lymphoma Research Center, Peking University Third Hospital, Beijing 100191, People's Republic of China
| | - Yanfang Wang
- Department of Hematology and Lymphoma Research Center, Peking University Third Hospital, Beijing 100191, People's Republic of China
| | - Mingxia Zhu
- Department of Hematology and Lymphoma Research Center, Peking University Third Hospital, Beijing 100191, People's Republic of China
| | - Jing Wang
- Department of Hematology and Lymphoma Research Center, Peking University Third Hospital, Beijing 100191, People's Republic of China
| | - Xiaoyan Ke
- Department of Hematology and Lymphoma Research Center, Peking University Third Hospital, Beijing 100191, People's Republic of China
| |
Collapse
|
22
|
Gutierrez-Silerio GY, Franco-Topete RA, Haramati J, Navarrete-Medina EM, Gutierrez-Franco J, Bueno-Topete MR, Bastidas-Ramirez BE, Ramos-Marquez ME, Del Toro-Arreola S. Positive staining of the immunoligand B7-H6 in abnormal/transformed keratinocytes consistently accompanies the progression of cervical cancer. BMC Immunol 2020; 21:9. [PMID: 32138659 PMCID: PMC7059382 DOI: 10.1186/s12865-020-0341-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/26/2020] [Indexed: 12/26/2022] Open
Abstract
Background B7-H6 has been revealed as an endogenous immunoligand expressed in a variety of tumors, but not expressed in healthy tissues. Heretofore, no studies have been reported describing B7-H6 in women with cervical cancer. To investigate this question, our present study was conducted. Results This retrospective study comprised a total of 62 paraffinized cervical biopsies, which were distributed in five groups: low-grade squamous intraepithelial lesions (LSIL), high-grade squamous intraepithelial lesions (HSIL), squamous cervical carcinoma (SCC), uterine cervical adenocarcinoma (UCAC), and a group of cervicitis (as a control for non-abnormal/non-transformed cells). Cervical sections were stained by immunohistochemistry to explore the expression of B7-H6, which was reported according to the immunoreactive score (IRS) system. We observed a complete lack of B7-H6 in LSIL abnormal epithelial cells. Interestingly, B7-H6 began to be seen in HSIL abnormal epithelial cells; more than half of this group had B7-H6 positive cells, with staining characterized by a cytoplasmic and membranous pattern. B7-H6 in the SCC group was also seen in the majority of the sections, showing the same cytoplasmic and membranous pattern. Strong evidence of B7-H6 was notably found in UCAC tumor columnar cells (in 100% of the specimens, also with cytoplasmic and membranous pattern). Moreover, consistent B7-H6 staining was observed in infiltrating plasma cells in all groups. Conclusions B7-H6 IRS positively correlated with disease stage in the development of cervical cancer; additionally, B7-H6 scores were found to be even higher in the more aggressive uterine cervical adenocarcinoma, suggesting a possible future therapeutic target for this cancer type.
Collapse
Affiliation(s)
- Gloria Yareli Gutierrez-Silerio
- Instituto de Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Sierra Mojada # 950, Colonia Independencia, CP 44340, Guadalajara, Jalisco, Mexico.,Instituto Transdisciplinar de Investigación y Servicios, CUCEI, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Ramon Antonio Franco-Topete
- Laboratorio de Patología, Departamento de Microbiología y Patología, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico.,Departamento de Anatomía Patológica, Nuevo Hospital Civil de Guadalajara "Dr. Juan I. Menchaca", Guadalajara, Jalisco, Mexico
| | - Jesse Haramati
- Laboratorio de Inmunobiología, Departamento de Biología Celular y Molecular, CUCBA, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Eduardo Miguel Navarrete-Medina
- Laboratorio de Patología, Departamento de Microbiología y Patología, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico.,Departamento de Anatomía Patológica, Nuevo Hospital Civil de Guadalajara "Dr. Juan I. Menchaca", Guadalajara, Jalisco, Mexico
| | - Jorge Gutierrez-Franco
- Unidad Académica de Ciencias Químico Biológicas y Farmacéuticas, Universidad Autónoma de Nayarit, Tepic, Nayarit, Mexico
| | - Miriam Ruth Bueno-Topete
- Instituto de Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Sierra Mojada # 950, Colonia Independencia, CP 44340, Guadalajara, Jalisco, Mexico.,Instituto Transdisciplinar de Investigación y Servicios, CUCEI, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Blanca Estela Bastidas-Ramirez
- Instituto de Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Sierra Mojada # 950, Colonia Independencia, CP 44340, Guadalajara, Jalisco, Mexico.,Instituto Transdisciplinar de Investigación y Servicios, CUCEI, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Martha Eloisa Ramos-Marquez
- Instituto de Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Sierra Mojada # 950, Colonia Independencia, CP 44340, Guadalajara, Jalisco, Mexico
| | - Susana Del Toro-Arreola
- Instituto de Enfermedades Crónico Degenerativas, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Sierra Mojada # 950, Colonia Independencia, CP 44340, Guadalajara, Jalisco, Mexico. .,Instituto Transdisciplinar de Investigación y Servicios, CUCEI, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico. .,Laboratorio de Inmunología, Departamento de Fisiología, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico.
| |
Collapse
|
23
|
Hu Y, Zeng T, Xiao Z, Hu Q, Li Y, Tan X, Yue H, Wang W, Tan H, Zou J. Immunological role and underlying mechanisms of B7-H6 in tumorigenesis. Clin Chim Acta 2020; 502:191-198. [PMID: 31904350 DOI: 10.1016/j.cca.2019.12.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/31/2019] [Accepted: 12/31/2019] [Indexed: 02/07/2023]
Abstract
B7 homolog 6 (B7-H6) has been identified as involved in tumorigenesis. Elucidating its role and potential mechanism of action is essential for understanding tumorigenesis and the potential development of an effective clinical strategy. Abnormal overexpression of B7-H6 in various types of tumors was reported to be linked with poor prognosis. B7-H6 suppresses the initiation of the "caspase cascade" and induces anti-apoptosis by STAT3 pathway activation to provoke tumorigenesis. B7-H6 facilitates tumor proliferation and cell cycle progression by regulating apoptosis suppressors. B7-H6 induces cellular cytotoxicity, secretion of TNF-α and IFN-γ and B7-H6-specific BiTE triggers T cells to accelerate tumorigenesis. B7-H6 induces abnormal immunological progression by HER2-scFv mediated ADCC and NKp30 immune escape to promote tumorigenesis. B7-H6 promotes tumorigenesis via apoptosis inhibition, proliferation and immunological progression. B7-H6 may a valuable potential biomarker and therapeutic strategy for diagnostics, prognostics and treatment in cancer.
Collapse
Affiliation(s)
- Yuxuan Hu
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, PR China
| | - Tian Zeng
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, PR China
| | - Zheng Xiao
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, PR China
| | - Qihao Hu
- Cardiothoracic Surgery, The Second Affiliated Hospital, University of South China, Hengyang, Hunan 421001, PR China
| | - Yukun Li
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, PR China
| | - Xiongjin Tan
- The Second Department of Orthopaedic, 922 Hospital of PLA, Hengyang, Hunan 410011, PR China
| | - Haiyan Yue
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, PR China; Department of Pathology, The Central Hospital of Shaoyang, Shaoyang, Hunan 422000, PR China
| | - Wensong Wang
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, PR China
| | - Hui Tan
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, PR China.
| | - Juan Zou
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, PR China.
| |
Collapse
|
24
|
Zhou H, Dong J, Guo L, Wang X, Wang K, Cai X, Yang S. The prognostic value of B7-H6 in esophageal squamous cell carcinoma. Sci Rep 2019; 9:18122. [PMID: 31792298 PMCID: PMC6889130 DOI: 10.1038/s41598-019-54731-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 11/18/2019] [Indexed: 02/07/2023] Open
Abstract
B7-H6, a member of the B7 family molecules, participates in the clearance of tumor cells by binding to NKp30 on NK cells. B7-H6 expression level in esophageal squamous cell carcinoma (ESCC) and the clinical value remain unknown. The goal of this study was to determine the expression of B7-H6 in ESCC and further explore its clinical significance. We retrospectively collected the clinical data of 145 patients diagnosed with ESCC between January 2007 and December 2008. The expression of B7-H6 of the pathological tissue samples was detected by immunohistochemistry. The chi-square (χ2) test was used to analyse the relationships of B7-H6 and clinicopathological characteristics. Survival and hazard functions were estimated using the Kaplan-Meier method, and survival between groups was compared using the two-sided log-rank test. The Cox proportional hazards regression model was used to adjust for the risk factors related to overall survival (OS). 133/145 (91.72%) of the ESCC tissue samples exhibited B7-H6 expression. The expression level of B7-H6 was correlated with T stage (P = 0.036) and lymphatic metastasis status (P = 0.044). High B7-H6 expression (P = 0.003) was associated with a significantly worse OS than low B7-H6 expression. Multivariate Cox proportional hazards regression analysis demonstrated that tumour size (P = 0.021), B7-H6 expression (P = 0.025) and lymphatic metastasis status (P = 0.049) were independent prognostic factors of OS for ESCC. Collectively, our findings suggest that B7-H6 is widely expressed in ESCC samples. And B7-H6 may represent a predictor of poor prognosis for ESCC.
Collapse
Affiliation(s)
- Huan Zhou
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Jun Dong
- Department of VIP Region, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Liyi Guo
- Department of Radiotherapy, Huiyang Hospital Affiliated Southern Medical University, Huizhou, Guangdong, China
| | - Xicheng Wang
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Kailin Wang
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Xiuyu Cai
- Department of VIP Region, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China.
| | - Shu Yang
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China.
| |
Collapse
|
25
|
Chapoval AI, Chapoval SP, Shcherbakova NS, Shcherbakov DN. Immune Checkpoints of the B7 Family. Part 2. Representatives of the B7 Family B7-H3, B7-H4, B7-H5, B7-H6, B7-H7, and ILDR2 and Their Receptors. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2019. [DOI: 10.1134/s1068162019050091] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
26
|
Zhang Y, Zhao Z, Li S, Dong L, Li Y, Mao Y, Liang Y, Tao Y, Ma J. Inhibition of miR‑214 attenuates the migration and invasion of triple‑negative breast cancer cells. Mol Med Rep 2019; 19:4035-4042. [PMID: 30942417 PMCID: PMC6471216 DOI: 10.3892/mmr.2019.10112] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 11/21/2018] [Indexed: 12/26/2022] Open
Abstract
Triple‑negative breast cancer (TNBC) is a subtype of breast cancer. MicroRNA (miR)‑214 is closely associated with controlling the development of tumor cells; therefore, in the present study, the target gene and effects of miR‑214 on TNBC cells were explored. Luciferase activity was examined by luciferase reporter assay. The viability, invasion and migration of MDA‑MB‑231 TNBC cells were measured using Cell Counting kit‑8, Transwell and wound‑healing assays, respectively. The expression levels of various factors were determined using reverse transcription‑quantitative polymerase chain reaction and western blotting. The results demonstrated that the expression levels of miR‑214 were higher and the levels of α1‑antitrypsin (α1‑AT) were lower in TNBC tissues compared with in normal tissues. Subsequently, α1‑AT was revealed to be a target of miR‑214. Furthermore, inhibition of miR‑214 decreased cell viability, invasion and migration, enhanced the expression of E‑cadherin and tissue inhibitor of metalloproteinases‑2, and reduced the expression of metastatic tumour antigen 1 and matrix metalloproteinase‑2. Inhibition of miR‑214 also significantly downregulated the phosphorylation of protein kinase B (Akt) and mammalian target of rapamycin (mTOR), and markedly downregulated that of phosphoinositide 3‑kinase (PI3K); however, the expression levels of total PI3K, Akt and mTOR remained stable in all groups. Taken together, these findings indicated that α1‑AT may be a target of miR‑214. Downregulation of miR‑214 markedly suppressed the viability, migration and invasion of MDA‑MB‑231 cells, and inhibited the PI3K/Akt/mTOR pathway. These findings suggested that miR‑214 targeting α1‑AT may be a potential mechanism underlying TNBC development.
Collapse
Affiliation(s)
- Yi Zhang
- Thyroid-Breast Surgery Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Zhijing Zhao
- Thyroid-Breast Surgery Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Siqi Li
- Thyroid-Breast Surgery Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Liying Dong
- Thyroid-Breast Surgery Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Yan Li
- Thyroid-Breast Surgery Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Ying Mao
- Thyroid-Breast Surgery Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Ying Liang
- Thyroid-Breast Surgery Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Yun Tao
- Clinical Skill Center, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Junfeng Ma
- Thyroid-Breast Surgery Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| |
Collapse
|
27
|
Rap2B promotes cell adhesion, proliferation, migration and invasion of human glioma. J Neurooncol 2019; 143:221-229. [PMID: 30997639 DOI: 10.1007/s11060-019-03163-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/02/2019] [Indexed: 01/08/2023]
Abstract
PURPOSE Rap2B, a member of the GTP-binding proteins, is generally up-regulated in numerous types of tumors. Nevertheless, the influence and regulatory mechanisms of Rap2B in gliomas are still not corroborated. Therefore, we analyzed the expression of Rap2B in glioma tissues and cells, and researched its significance in adhesion, proliferation, migration and invasion of the glioma cell line. METHODS We analyzed the expression of Rap2B in different pathologic grades of glioma tissues by tissue microarray and immunohistochemistry. We assessed the expression of Rap2B in glioma tissue and non-tumor tissue by Western blot. And the expression of Rap2b protein in glioma cells and normal human astrocytes (NHA) was detected by Western blot. In addition, we disclosed the effect of Rap2B knockdown on cell adhesion, proliferation, migration and invasion by using cell attachment assay, CCK-8 assay, cell migration assay and Wound Healing assay, cell invasion assay, respectively. Western blot was used to detect the changes of expression level of NF-kB, MMP-2 and MMP-9 protein when downregulated the expression of Rap2B. RESULTS The tissue microarray immunohistochemical results of glioma showed that the expression of Rap2B had no significant correlations between Rap2B expression and the clinicopathologic variables, including patient age (P = 0.352), gender (P = 0.858), WHO Grade (P = 0.693) and histology type (P = 0.877). Western blot analysis showed that the glioma tissue had a dramatically increase of Rap2B expression compared with the non-tumor tissues (P < 0.01). And the expression of Rap2B was markedly up-regulated in all 5 glioma cell lines compared with that in normal human astrocytes (NHA) (P < 0.01). We found that the ability of adhesion, proliferation, migration and invasion of glioma cells were significantly decreased after downregulated Rap2B expression compared with the control group (P < 0.05). In addition, Western blot results showed that the expression levels of NF-kB, MMP-2 and MMP-9 in the interference group were significantly lower than those in the negative control group (P < 0.05). CONCLUSIONS Rap2B expression is up-regulated in glioma tissues and glioma cell lines. Knockdown of Rap2B inhibits glioma cells' adhesion and proliferation in vitro. Knockdown of Rap2B inhibits glioma cells' migration in vitro. Knockdown of Rap2B inhibits glioma cells' invasion and MMPs activity through NF-kB pathway.
Collapse
|
28
|
Chen L, Feng J, Xu B, Zhou Y, Zheng X, Wu C, Jiang J. B7-H6 expression in human hepatocellular carcinoma and its clinical significance [corrected]. Cancer Cell Int 2018; 18:126. [PMID: 30186042 PMCID: PMC6122564 DOI: 10.1186/s12935-018-0627-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 08/29/2018] [Indexed: 12/25/2022] Open
Abstract
Background Recent studies have suggested that B7-H6, a new member of the B7 family of ligands, not only is a crucial regulator of NK cell-mediated immune responses but also has important clinical implications due to its abnormal expression in many human cancers. We have previously reported that higher B7-H6 expression levels in ovarian cancer tissues are positively correlated with tumor metastasis and cancer progression. To date, the expression of B7-H6 in human hepatocellular carcinoma (HCC) and the clinical significance of B7-H6 expression still remain elusive. Methods In the present study, the expression level of B7-H6 was examined in both HCC tissues and HCC cell lines (HepG2 and SMMC-7721). And the clinical significance of B7-H6 was analyzed as well. Results Our results revealed that B7-H6 was expressed abnormally in HCC tissues, which was greatly related to tumor size. The TCGA data also showed that the B7-H6 mRNA expression level was significantly negatively correlated with the survival of HCC patients. Next, to investigate the functions of B7-H6 in HCC, we successfully constructed B7-H6 knockdown expression human HCC cell lines using the RNA interference technology. Our studies showed that reduced expression of B7-H6 in HepG2 and SMMC-7721 cells significantly attenuated cell proliferation as well as cell migration and invasion. Besides, depletion of B7-H6 greatly induced cell cycle arrest at G1 phase. And also B7-H6 knockdown in HCC cell lines dramatically decreased the C-myc, C-fos and Cyclin-D1 expression. Conclusions Our present findings suggested that B7-H6 played an important role in oncogenesis of HCC on cellular level, and B7-H6 could be employed to develop immunotherapeutic approaches targeting this malignancy.
Collapse
Affiliation(s)
- Lujun Chen
- 1Department of Tumor Biological Treatment and Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China.,2Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China.,3Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - Jun Feng
- 1Department of Tumor Biological Treatment and Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China.,2Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China.,3Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - Bin Xu
- 1Department of Tumor Biological Treatment and Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China.,2Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China.,3Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - You Zhou
- 1Department of Tumor Biological Treatment and Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China.,2Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China.,3Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - Xiao Zheng
- 1Department of Tumor Biological Treatment and Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China.,2Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China.,3Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - Changping Wu
- 1Department of Tumor Biological Treatment and Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China.,2Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China.,3Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - Jingting Jiang
- 1Department of Tumor Biological Treatment and Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China.,2Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China.,3Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| |
Collapse
|