1
|
Xu T, Xiao W, Li W, Xu X, Zhang H, Zhang X. Exploring the causal relationship between immune factors and chondrosarcoma: a Mendelian randomization study. Discov Oncol 2025; 16:801. [PMID: 40382743 PMCID: PMC12086138 DOI: 10.1007/s12672-025-02654-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 05/09/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Previous studies have investigated the potential role of immune factors in chondrosarcoma (CHS). However, the causal relationship is unknown. METHODS A two-sample Mendelian randomization (MR) was used to explore potential correlations between 731 immunocyte phenotypes, 91 inflammatory proteins, and CHS. The data were derived from published summary statistics of genome-wide association studies. Inverse-variance weighted was employed as the primary method. Furthermore, a range of analytical methods, including MR-Egger, weighted mode, and weighted median was used to enhance the robustness of the results. A two-step MR was used to assess the mediating effects of inflammatory proteins. Subsequently, sensitivity and MR Steiger directionality tests were performed. RESULTS MR analyses showed that 12 immunocyte phenotypes were positively correlated with CHS (P < 0.05, OR > 1), and 11 immunocyte phenotypes were negatively correlated with CHS (P < 0.05, OR < 1). Five inflammatory proteins were positively associated with CHS (P < 0.05, OR > 1). No heterogeneous or horizontal pleiotropy was found. The MR Steiger analysis found no statistically significant evidence of reverse causation. Mediation analysis did not identify any potential mediating effects. CONCLUSION Our study underscores the pivotal role of immune factors in CHS and offers insights that can inform future research.
Collapse
Affiliation(s)
- Taichuan Xu
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Wentao Xiao
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Wenjie Li
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Xianfa Xu
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Haiwen Zhang
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Xian Zhang
- Department of Spine, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, 214072, Jiangsu, China.
| |
Collapse
|
2
|
Wu H, Liu J, Zhang XH, Jin S, Li P, Liu H, Zhao L, Wang J, Zhao S, Tian HD, Lai JR, Hao Y, Liu GR, Hou K, Yan M, Liu SL, Pang D. The combination of flaxseed lignans and PD-1/ PD-L1 inhibitor inhibits breast cancer growth via modulating gut microbiome and host immunity. Drug Resist Updat 2025; 80:101222. [PMID: 40048957 DOI: 10.1016/j.drup.2025.101222] [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: 12/29/2024] [Revised: 02/22/2025] [Accepted: 02/22/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Patients with breast cancer (BC) who benefit from the PD-1/PD-L1 inhibitor (PDi) is limited, necessitating novel strategies to improve immunotherapy efficacy of BC. Here we aimed to investigate the inhibitory effects of flaxseed lignans (FL) on the biological behaviors of BC and evaluate the roles of FL in enhancing the anticancer effects of PDi. METHODS HPLC was used to detect the content of enterolactone (ENL), the bacterial transformation product of FL. Transcript sequencing was performed and identified CD38 as a downstream target gene of ENL. CD38-overexpressing cells were constructed and cell proliferation, colony formation, wound healing and transwell assays were used to assess the function of ENL/CD38 axis on BC cells in vitro. Multiplexed immunohistochemistry (mIHC) and CyTOF were used to detect the changes of the tumor immune microenvironment (TIM). 16S rDNA sequencing was used to explore the changes of gut microbiota in mice. A series of in vivo experiments were conducted to investigate the anticancer effects and mechanisms of FL and PDi. RESULTS FL was converted to ENL by gut microbiota and FL administration inhibited the progression of BC. ENL inhibited the malignant behaviors of BC by downregulating CD38, a key gene associated with immunosuppression and PD-1/PD-L1 blockade resistance. The mIHC assay revealed that FL administration enhanced CD3+, CD4+ and CD8+ cells and reduced F4/80+ cells in TIM. CyTOF confirmed the regulatory effects of FL and FL in combination with PDi (FLcPDi) on TIM. In addition, 16S rDNA analysis demonstrated that FLcPDi treatment significantly elevated the abundance of Akkermansia and, importantly, Akkermansia administration enhanced the response to PDi in mice treated with antibiotics. CONCLUSIONS The FL/ENL/CD38 axis inhibited BC progression. FL enhanced the anticancer effects of PDi by modulating gut microbiota and host immunity.
Collapse
Affiliation(s)
- Hao Wu
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Jiena Liu
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Xing-Hua Zhang
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Shengye Jin
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ping Li
- The Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Huidi Liu
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Liuying Zhao
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jianyu Wang
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shilu Zhao
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hong-Da Tian
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Jin-Ru Lai
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Yi Hao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Gui-Rong Liu
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Kaijian Hou
- School of Public Health, Shantou University, Shantou, China; Longhu People's Hospital, Shantou, China.
| | - Meisi Yan
- Department of Pathology, Harbin Medical University, Harbin, China.
| | - Shu-Lin Liu
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China; Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada.
| | - Da Pang
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
3
|
Lv R, Wang D, Wang T, Li R, Zhuang A. Causality between gut microbiota, immune cells, and breast cancer: Mendelian randomization analysis. Medicine (Baltimore) 2024; 103:e40815. [PMID: 39654239 PMCID: PMC11630993 DOI: 10.1097/md.0000000000040815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 12/12/2024] Open
Abstract
The association between gut microbiota (GM) and breast cancer (BC) has been studied. Nevertheless, the causal relationship between them and the potential mediating factors have not been clearly defined. Therefore, in this study, Mendelian randomization analysis (MR) was employed to explore the causal relationship between 473 GM and BC, as well as the mediating effect of potential immune cells. In this investigation, we availed ourselves of the publicly accessible summary statistics from the genome-wide association study to undertake two-sample and reverse Mendelian randomization analyses on GM and BC, with the intention of clarifying the causal association between GM and BC. Subsequently, through the application of the two-step Mendelian randomization analysis, it was revealed that the relationship between GM and BC was mediated by immune cells. The stability of the research outcomes was verified via sensitivity analysis. Mendelian randomization analysis elucidated the protective impacts of 8 genera on BC (such as Phylum Actinobacteriota, Species Bacteroides A plebeius A, Species Bifidobacterium adolescentis, Species CAG-841 sp002479075, Family Fibrobacteraceae, Order Fibrobacterales, Class Fibrobacteria, and Species Phascolarctobacterium sp003150755). Additionally, there are 23 immune cell traits related to BC. Our research findings showed that the species Megamonas funiformis was associated with an increased risk of BC, and 11.20% of this effect was mediated by CD38 on IgD+ CD24-. Likewise, HLA DR on CD33br HLA DR+ CD14- mediated the causal relationship between Species Prevotellamassilia and BC, having a mediating ratio of 7.89%. This study clarifies a potential causal relationship between GM, immune cells, and BC and provides genetic evidence for this causal connection. It offers research directions for the subsequent prevention and treatment of BC through the interaction between GM and immune cells, and provides a reference for future mechanistic and clinical studies in this field.
Collapse
Affiliation(s)
- Rui Lv
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Danyan Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhuji Second People’s Hospital, Zhuji, China
| | - Tengyue Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Rongqun Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Aiwen Zhuang
- Institute of TCM Literature and Information, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| |
Collapse
|
4
|
Liang L, Yue C, Li W, Tang J, He Q, Zeng F, Cao J, Liu S, Chen Y, Li X, Zhou Y. CD38 symmetric dimethyl site R58 promotes malignant tumor cell immune escape by regulating the cAMP-GSK3β-PD-L1 axis. Heliyon 2024; 10:e37958. [PMID: 39386836 PMCID: PMC11462232 DOI: 10.1016/j.heliyon.2024.e37958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/02/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024] Open
Abstract
In recent years, immunotherapy has emerged as an effective approach for treating tumors, with programmed cell death ligand 1 (PD-L1)/programmed cell death protein-1 (PD-1) immune checkpoint blockade (ICB) being a promising strategy. However, suboptimal therapeutic efficacy limits its clinical benefit. Understanding the regulation mechanism of PD-L1 expression is crucial for improving anti-PD-L1/PD-1 therapy and developing more effective tumor immunotherapy. Previous studies have revealed that resistance to PD-L1/PD-1 blockade therapy arises from the upregulation of CD38 on tumor cells induced by ATRA and IFN-β, which mediates the inhibition of CD8+ T cell function through adenosine receptor signaling, thereby promoting immune evasion.Yet, the precise role of CD38 in regulating PD-L1 on malignant tumor cells and its impact on CD8+ T cells through PD-L1 remain unclear. Here, we demonstrate that CD38 is highly expressed in malignant tumors (lung cancer, nasopharyngeal carcinoma, cervical cancer) and upregulates PD-L1 protein expression, impairing CD8+ T cell function. Mechanistically, CD38 phosphorylates GSK3β via the adenosine-activated cAMP-PKA signaling pathway, leading to GSK3β inactivation and enhanced PD-L1 stability and expression, facilitating tumor immune escape. Furthermore, we identify PRMT5 as a novel CD38-interacting molecule that symmetrically dimethylates CD38 arginine position 58, augmenting PD-L1 stability and expression through the ADO-cAMP-GSK3β signaling axis. This inhibits CD8+ T cell-mediated tumor cell killing, enabling tumor cells to evade immune surveillance. Our findings suggest that targeting the CD38 R58 site offers a new avenue for enhancing anti-PD-L1/PD-1 therapy efficacy in tumor treatment.
Collapse
Affiliation(s)
- Lin Liang
- Breast Cancer Center, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China
| | - Chunxue Yue
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China
| | - Wentao Li
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China
| | - Jingqiong Tang
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Qian He
- Department of Radiation Oncology, Hunan Cancer Hospital & the Afliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Feng Zeng
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China
| | - Jiaying Cao
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China
| | - Siyi Liu
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China
| | - Yan Chen
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China
| | - Xin Li
- Breast Cancer Center, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Yanhong Zhou
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
5
|
Li L, Zhou X, Liu W, Chen Z, Xiao X, Deng G. Supplementation with NAD+ and its precursors: A rescue of female reproductive diseases. Biochem Biophys Rep 2024; 38:101715. [PMID: 38698835 PMCID: PMC11063225 DOI: 10.1016/j.bbrep.2024.101715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/14/2024] [Accepted: 04/17/2024] [Indexed: 05/05/2024] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential coenzyme involved in many pathophysiological processes. Supplementation with NAD+ and its precursors has been demonstrated as an emerging therapeutic strategy for the diseases. NAD+ also plays an important role in the reproductive system. Here, we summarize the function of NAD+ in various reproductive diseases and review the application of NAD+ and its precursors in the preservation of reproductive capacity and the prevention of embryonic malformations. It is shown that NAD+ shows good promise as a therapeutic approach for saving reproductive capacity.
Collapse
Affiliation(s)
- Lan Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Xin Zhou
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Wene Liu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Zhen Chen
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Xiaoqin Xiao
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Guiming Deng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| |
Collapse
|
6
|
Wang X, Li H, Chen H, Fang K, Chang X. Overexpression of circulating CD38+ NK cells in colorectal cancer was associated with lymph node metastasis and poor prognosis. Front Oncol 2024; 14:1309785. [PMID: 38463232 PMCID: PMC10921414 DOI: 10.3389/fonc.2024.1309785] [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: 10/08/2023] [Accepted: 01/31/2024] [Indexed: 03/12/2024] Open
Abstract
Introduction Lymph node metastasis (LNM) is a critical prognostic factor for colorectal cancer (CRC). Due to the potential influence of immune system on CRC progression, investigation into lymphocyte subsets as clinical markers has gained attention. The objective of this study was to assess the capability of lymphocyte subsets in evaluating the lymph node status and prognosis of CRC. Methods Lymphocyte subsets, including T cells (CD3+), natural killer cells (NK, CD3- CD56+), natural killer-like T cells (NK-like T, CD3+ CD56+), CD38+ NK cells (CD3- CD56+ CD38+) and CD38+ NK-like T cells (CD3+ CD56+ CD38+), were detected by flow cytometry. Univariate and multivariate analyses were used to assess the risk factors of LNM. The prognostic role of parameters was evaluated by survival analysis. Results The proportion of CD38+ NK cells within the NK cell population was significantly higher in LNM-positive patients (p <0.0001). However, no significant differences were observed in the proportions of other lymphocyte subsets. Poorer histologic grade (odds ratio [OR] =4.76, p =0.03), lymphovascular invasion (LVI) (OR =22.38, p <0.01), and CD38+ NK cells (high) (OR =4.54, p <0.01) were identified as independent risk factors for LNM. Furthermore, high proportion of CD38+ NK cells was associated with poor prognosis of CRC patients (HR=2.37, p =0.03). Conclusions It was demonstrated that the proportion of CD38+ NK cells was a marker overexpressed in LNM-positive patients compared with LNM-negative patients. Moreover, an elevated proportion of CD38+ NK cells is a risk factor for LNM and poor prognosis in CRC.
Collapse
Affiliation(s)
- Xueling Wang
- Center for Clinical Research, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haoran Li
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Huixian Chen
- Center for Clinical Research, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Kehua Fang
- Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaotian Chang
- Center for Clinical Research, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
7
|
Paulus A, Malavasi F, Chanan-Khan A. CD38 as a multifaceted immunotherapeutic target in CLL. Leuk Lymphoma 2022; 63:2265-2275. [DOI: 10.1080/10428194.2022.2090551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Affiliation(s)
- Aneel Paulus
- Department of Hematology-Oncology, Mayo Clinic Florida, Jacksonville, FL, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Fabio Malavasi
- Dipartimento Scienze Mediche, Università di Torino, Torino, Italy
- Fondazione Ricerca Molinette ONLUS, Università di Torino, Torino, Italy
| | - Asher Chanan-Khan
- Department of Hematology-Oncology, Mayo Clinic Florida, Jacksonville, FL, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
8
|
CD38: An important regulator of T cell function. Biomed Pharmacother 2022; 153:113395. [PMID: 35834988 DOI: 10.1016/j.biopha.2022.113395] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/03/2022] [Accepted: 07/07/2022] [Indexed: 11/23/2022] Open
Abstract
Cluster of differentiation 38 (CD38) is a multifunctional extracellular enzyme on the cell surface with NADase and cyclase activities. CD38 is not only expressed in human immune cells, such as lymphocytes and plasma cells, but also is abnormally expressed in a variety of tumor cells, which is closely related to the occurrence and development of tumors. T cells are one of the important immune cells in the body. As NAD consuming enzymes, CD38, ART2, SIRT1 and PARP1 are closely related to the number and function of T cells. CD38 may also influence the activity of ART2, SIRT1 and PARP1 through the CD38-NAD+ axis to indirectly affect the number and function of T cells. Thus, CD38-NAD+ axis has a profound effect on T cell activity. In this paper, we reviewed the role and mechanism of CD38+ CD4+ T cells / CD38+ CD8+ T cells in cellular immunity and the effects of the CD38-NAD+ axis on T cell activity. We also summarized the relationship between the CD38 expression level on T cell surface and disease prediction and prognosis, the effects of anti-CD38 monoclonal antibodies on T cell activity and function, and the role of anti-CD38 chimeric antigen receptor (CAR) T cell therapy in tumor immunity. This will provide an important theoretical basis for a comprehensive understanding of the relationship between CD38 and T cells.
Collapse
|
9
|
Barrientos-Robledo SG, Cebada-Ruiz JA, Rodríguez-Alba JC, Baltierra-Uribe SL, Díaz Y Orea MA, Romero-Ramírez H. CD38 a biomarker and therapeutic target in non-hematopoietic tumors. Biomark Med 2022; 16:387-400. [PMID: 35195042 DOI: 10.2217/bmm-2021-0575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The type II transmembrane glycoprotein CD38 has recently been implicated in regulating metabolism and the pathogenesis of multiple conditions, including aging, inflammation and cancer. CD38 is overexpressed in several tumor cells and microenvironment tumoral cells, associated to migration, angiogenesis, cell invasion and progression of the disease. Thus, CD38 has been used as a progression marker for different cancer types as well as in immunotherapy. This review focuses on describing the involvement of CD38 in various non-hematopoietic cancers.
Collapse
Affiliation(s)
- Susana G Barrientos-Robledo
- Laboratorio de Inmunología Experimental, Benemérita Universidad Autónoma de Puebla, Facultad de Medicina, Puebla, Mexico
| | - Jorge A Cebada-Ruiz
- Laboratorio de Inmunología Experimental, Benemérita Universidad Autónoma de Puebla, Facultad de Medicina, Puebla, Mexico
| | - Juan C Rodríguez-Alba
- Unidad de Citometría de Flujo, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Veracruz, Mexico
| | - Shantal L Baltierra-Uribe
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Maria A Díaz Y Orea
- Laboratorio de Inmunología Experimental, Benemérita Universidad Autónoma de Puebla, Facultad de Medicina, Puebla, Mexico
| | - Héctor Romero-Ramírez
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados, Mexico City, Mexico
| |
Collapse
|
10
|
Duan Y, Chen R, Huang Y, Meng X, Chen J, Liao C, Tang Y, Zhou C, Gao X, Sun J. Tuning the ignition of CAR: optimizing the affinity of scFv to improve CAR-T therapy. Cell Mol Life Sci 2021; 79:14. [PMID: 34966954 PMCID: PMC11073403 DOI: 10.1007/s00018-021-04089-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/06/2021] [Accepted: 12/10/2021] [Indexed: 10/19/2022]
Abstract
How single-chain variable fragments (scFvs) affect the functions of chimeric antigen receptors (CARs) has not been well studied. Here, the components of CAR with an emphasis on scFv were described, and then several methods to measure scFv affinity were discussed. Next, scFv optimization studies for CD19, CD38, HER2, GD2 or EGFR were overviewed, showing that tuning the affinity of scFv could alleviate the on-target/off-tumor toxicity. The affinities of scFvs for different antigens were also summarized to designate a relatively optimal working range for CAR design. Last, a synthetic biology approach utilizing a low-affinity synthetic Notch (synNotch) receptor to achieve ultrasensitivity of antigen-density discrimination and murine models to assay the on-target/off-tumor toxicity of CARs were highlighted. Thus, this review provides preliminary guidelines of choosing the right scFvs for CARs.
Collapse
Affiliation(s)
- Yanting Duan
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, China
| | - Ruoqi Chen
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, China
| | - Yanjie Huang
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Xianhui Meng
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, China
| | - Jiangqing Chen
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, China
| | - Chan Liao
- Department of Hematology-Oncology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yongmin Tang
- Department of Hematology-Oncology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chun Zhou
- School of Public Health, and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaofei Gao
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Jie Sun
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, China.
| |
Collapse
|
11
|
Lu X, Song X, Hao X, Liu X, Zhang X, Yuan N, Ma H, Zhang Z. miR-186-3p attenuates the tumorigenesis of cervical cancer via targeting insulin-like growth factor 1 to suppress PI3K-Akt signaling pathway. Bioengineered 2021; 12:7079-7092. [PMID: 34551673 PMCID: PMC8806770 DOI: 10.1080/21655979.2021.1977053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
miR-186-3p acts as a tumor suppressor in various cancers. This study aimed to explore the expression levels of miR-186-3p and its role in cervical cancer. We analyzed the effects of miR-186-3p and insulin-like growth factor 1 (IGF1) on the proliferation, invasion, and apoptosis of cervical cancer cells in vitro by regulating the PI3K/Akt signaling pathway. In cervical cancer tissues and cells, miR-186-3p was downregulated, and IGF1 was upregulated. In addition, miR-186-3p inhibited cell proliferation and invasion and enhanced apoptosis of cervical cancer cells. Moreover, our results showed that miR-186-3p inversely regulated the mRNA expression of IGF1 through direct contact. Knockdown of IGF1 reversed the results of miR-186-3p inhibitor in cervical cancer cells. In addition, the PI3K/Akt signaling pathway was activated by the miR-186-3p inhibitor, although partially arrested by IGF1 knockdown. The PI3K/Akt signaling pathway inhibitor suppressed miR-186-3p inhibitor-stimulated cell proliferation in cervical cancer. In conclusion, miR-186-3p inhibits tumorigenesis of cervical cancer by repressing IGF1, which inactivates the PI3K/Akt signaling pathway, implicating miR-186-3p as a potential new target for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Xiurong Lu
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Xiao Song
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Xiaohui Hao
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Xiaoyu Liu
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Xianyu Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Na Yuan
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Huan Ma
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Zhilin Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| |
Collapse
|
12
|
Ding Z, He Y, Fu Y, Zhu N, Zhao M, Song Y, Huang X, Chen S, Yang Y, Zhang C, Hu Q, Ni Y, Ding L. CD38 Multi-Functionality in Oral Squamous Cell Carcinoma: Prognostic Implications, Immune Balance, and Immune Checkpoint. Front Oncol 2021; 11:687430. [PMID: 34211854 PMCID: PMC8239289 DOI: 10.3389/fonc.2021.687430] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/27/2021] [Indexed: 01/09/2023] Open
Abstract
Background CD38 belongs to the ribosyl cyclase family and is expressed on various hematological cells and involved in immunosuppression and tumor promotion. Although targeting CD38 antibodies has been approved for treatment of multiple myeloma, the function of CD38 in solid tumor, oral squamous cell carcinoma (OSCC) etc., has not been investigated. Methods This retrospective study included 92 OSCC samples and analyzed the spatial distribution of CD38 by immunohistochemistry (IHC). The values of diagnosis and prognosis of CD38 were evaluated. Additionally, 53 OSCC preoperative peripheral blood samples were used to be analyzed by flow cytometry. Tumor Immune Estimation Resource (TIMER) and cBioPortal databases were used to study CD38 level in various tumors and its correlation with tumor immune microenvironment in head and neck squamous cell carcinoma (HNSCC). Results CD38 ubiquitously presented in tumor cells (TCs), fibroblast-like cells (FLCs), and tumor-infiltrating lymphocytes (TILs). Patients with highly expressed CD38 in TCs (CD38TCs) had higher TNM stage and risk of lymph node metastasis. Upregulation of CD38 in FLCs (CD38FLCs) was significantly associated with poor WPOI. Escalated CD38 in TILs (CD38TILs) led to higher Ki-67 level of tumor cells. Moreover, patients with enhanced CD38TCs were susceptible to postoperative metastasis occurrence, and those with highly expressed CD38TILs independently predicted shorter overall and disease-free survival. Strikingly, patients with highly expressed CD38TILs, but not CD38TCs and CD38FLCs, had significantly lower CD3+CD4+ T cells and higher ratio of CD3-CD16+CD56+NK cells. The imbalance of immune system is attributed to dysregulated immune checkpoint molecules (VISTA, PD-1, LAG-3, CTLA-4, TIGIT, GITR) as well as particular immune cell subsets, which were positively correlated with CD38 expression in HNSCC. Conclusion CD38 is a poor prognostic biomarker for OSCC patients and plays a vital role in governing immune microenvironment and circulating lymphocyte homeostasis. Co-expression between CD38 and immune checkpoint molecules provides new insight into immune checkpoint therapy.
Collapse
Affiliation(s)
- Zhuang Ding
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yijia He
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yong Fu
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Nisha Zhu
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Mengxiang Zhao
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yuxian Song
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiaofeng Huang
- Department of Oral Pathology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Sheng Chen
- Department of Oral Pathology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yan Yang
- Department of Oral Pathology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Caihong Zhang
- Research Institute of Superconductor Electronics, School of Electronic Science and Engineering, Nanjing University, Nanjing, China
| | - Qingang Hu
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yanhong Ni
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Liang Ding
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
13
|
CD38 and Regulation of the Immune Response Cells in Cancer. JOURNAL OF ONCOLOGY 2021; 2021:6630295. [PMID: 33727923 PMCID: PMC7936891 DOI: 10.1155/2021/6630295] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/12/2021] [Accepted: 02/17/2021] [Indexed: 12/26/2022]
Abstract
Cancer is a leading cause of death worldwide. Understanding the functional mechanisms associated with metabolic reprogramming, which is a typical feature of cancer cells, is key to effective therapy. CD38, primarily a NAD + glycohydrolase and ADPR cyclase, is a multifunctional transmembrane protein whose abnormal overexpression in a variety of tumor types is associated with cancer progression. It is linked to VEGFR2 mediated angiogenesis and immune suppression as it favors the recruitment of suppressive immune cells like Tregs and myeloid-derived suppressor cells, thus helping immune escape. CD38 is expressed in M1 macrophages and in neutrophil and T cell-mediated immune response and is associated with IFNγ-mediated suppressor activity of immune responses. Targeting CD38 with anti-CD38 monoclonal antibodies in hematological malignancies has shown excellent results. Bearing that in mind, targeting CD38 in other nonhematological cancer types, especially carcinomas, which are of epithelial origin with specific anti-CD38 antibodies alone or in combination with immunomodulatory drugs, is an interesting option that deserves profound consideration.
Collapse
|
14
|
Gao L, Li J, He J, Liang L, He Z, Yue C, Jin X, Luo G, Zhou Y. CD90 affects the biological behavior and energy metabolism level of gastric cancer cells by targeting the PI3K/AKT/HIF-1α signaling pathway. Oncol Lett 2021; 21:191. [PMID: 33574930 DOI: 10.3892/ol.2021.12451] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 09/08/2020] [Indexed: 02/07/2023] Open
Abstract
CD90, also known as Thy-1 cell surface antigen, is located on human chromosome 11q23.3, and encodes a glycosylphosphatidylinositol-linked cell surface glycoprotein. CD90 serves a key role in malignancy by regulating cell proliferation, metastasis and angiogenesis. Gastric cancer is one of the most common types of malignancy. Patients with advanced gastric cancer have a poor prognosis. CD90 plays a key role in the occurrence and progression of gastric cancer. However, the molecular mechanism of CD90 in gastric cancer is currently unclear. In order to identify the molecular mechanism by which CD90 affects the biological behavior and energy metabolism of gastric cancer cells, the present study used Cell Counting Kit-8 assays, lactate concentration determination and ATP content determination. The results demonstrated that CD90 promotes proliferation and inhibits senescence in gastric cancer cells. In addition, CD90 enhanced the invasion and migration abilities of AGS gastric cancer cells. Overexpression of CD90 resulted in the accumulation of intracellular lactic acid in AGS cells. CD90 upregulated lactate dehydrogenase levels and increased the NADPH/NADP+ ratio in AGS cells. CD90 overexpression decreased the ATP concentration in AGS cells. PI3K, PDK1, phosphorylated-AKT-Ser473, HIF-1α, MDM2 and SIRT1 levels were upregulated in CD90-overexpressing AGS cells, compared with AGS cells transfected with the empty vector. In contrast, PTEN, p53, SIRT2, SIRT3 and SIRT6 were downregulated. The results indicate that CD90 affects the biological behavior and levels of energy metabolism of gastric cancer cells by targeting the PI3K/AKT/HIF-1α signaling pathway.
Collapse
Affiliation(s)
- Lu Gao
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China.,Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410078, P.R. China
| | - Jun Li
- Department of Nursing, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Junyu He
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410078, P.R. China
| | - Lin Liang
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410078, P.R. China
| | - Zhengxi He
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China
| | - Chunxue Yue
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410078, P.R. China
| | - Xi Jin
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410078, P.R. China
| | - Gengqiu Luo
- Department of Pathology, Xiangya Hospital, Basic School of Medicine, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yanhong Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China.,Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410078, P.R. China
| |
Collapse
|
15
|
Abstract
CD38 is a transmembrane glycoprotein that is widely expressed in a variety of human tissues and cells, especially those in the immune system. CD38 protein was previously considered as a cell activation marker, and today monoclonal antibodies targeting CD38 have witnessed great achievements in multiple myeloma and promoted researchers to conduct research on other tumors. In this review, we provide a wide-ranging review of the biology and function of the human molecule outside the field of myeloma. We focus mainly on current research findings to summarize and update the findings gathered from diverse areas of study. Based on these findings, we attempt to extend the role of CD38 in the context of therapy of solid tumors and expand the role of the molecule from a simple marker to an immunomodulator.
Collapse
Affiliation(s)
- Yanli Li
- Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, PR China
| | - Rui Yang
- Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, PR China
| | - Limo Chen
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009 USA
| | - Sufang Wu
- Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, PR China
| |
Collapse
|
16
|
Kiselevskiy M, Shubina I, Chikileva I, Sitdikova S, Samoylenko I, Anisimova N, Kirgizov K, Suleimanova A, Gorbunova T, Varfolomeeva S. Immune Pathogenesis of COVID-19 Intoxication: Storm or Silence? Pharmaceuticals (Basel) 2020; 13:E166. [PMID: 32722596 PMCID: PMC7465708 DOI: 10.3390/ph13080166] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 01/08/2023] Open
Abstract
Dysregulation of the immune system undoubtedly plays an important and, perhaps, determining role in the COVID-19 pathogenesis. While the main treatment of the COVID-19 intoxication is focused on neutralizing the excessive inflammatory response, it is worth considering an equally significant problem of the immunosuppressive conditions including immuno-paralysis, which lead to the secondary infection. Therefore, choosing a treatment strategy for the immune-mediated complications of coronavirus infection, one has to pass between Scylla and Charybdis, so that, in the fight against the "cytokine storm," it is vital not to miss the point of the immune silence that turns into immuno-paralysis.
Collapse
Affiliation(s)
- Mikhail Kiselevskiy
- FSBI N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 24 Kashirskoye sh., 115548 Moscow, Russia; (I.C.); (S.S.); (I.S.); (N.A.); (K.K.); (A.S.); (T.G.); (S.V.)
| | - Irina Shubina
- FSBI N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 24 Kashirskoye sh., 115548 Moscow, Russia; (I.C.); (S.S.); (I.S.); (N.A.); (K.K.); (A.S.); (T.G.); (S.V.)
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Liao S, Liang L, Yue C, He J, He Z, Jin X, Luo G, Zhou Y. CD38 is involved in cell energy metabolism via activating the PI3K/AKT/mTOR signaling pathway in cervical cancer cells. Int J Oncol 2020; 57:338-354. [PMID: 32319590 DOI: 10.3892/ijo.2020.5040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 02/14/2020] [Indexed: 11/06/2022] Open
Abstract
In contrast to normal cells, cancer cells typically undergo metabolic reprogramming. Studies have shown that oncogenes play an important role in this metabolic reprogramming. CD38 is a multifunctional transmembrane protein that is expressed abnormally in a variety of tumor types. To investigate the effect and possible mechanism of CD38 in cervical cancer cells and to provide a new therapeutic target for the treatment of cervical cancer, the present study identified that CD38 is involved in regulating cell metabolism in cervical cancer cells. Liquid chromatography‑tandem mass spectrometry and bioinformatic analyses revealed that differentially abundant proteins in CD38‑overexpressed cervical cancer cells (CaSki‑CD38 and HeLa‑CD38) are predominantly involved in glycolytic pathways, oxidative phosphorylation and the NAD/NADH metabolic process. Further experiments using an ATP test kit and lactate test kit revealed that CD38 promotes glucose consumption, increases lactate accumulation and increases ATP production. In addition, CD38 increases the phosphorylation of phosphatidylserine/threonine kinase (AKT), mechanistic target of rapamycin (mTOR) and phosphatidylinositol‑4,5‑bisphosphate 3‑kinase (PI3K), which play a key role in tumor metabolism. Furthermore, it was found that the energy metabolism of cervical cancer cells was inhibited following treatment with the mTOR inhibitor rapamycin. In conclusion, the results of the present study suggested that CD38 regulates the metabolism of cervical cancer cells by regulating the PI3K/AKT/mTOR pathway, which may be a candidate target for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Shan Liao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Lin Liang
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410078, P.R. China
| | - Chunxue Yue
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410078, P.R. China
| | - Junyu He
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410078, P.R. China
| | - Zhengxi He
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410078, P.R. China
| | - Xi Jin
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410078, P.R. China
| | - Gengqiu Luo
- Department of Pathology, Xiangya Hospital, Basic School of Medicine, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yanhong Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
18
|
The Good, the Bad and the Unknown of CD38 in the Metabolic Microenvironment and Immune Cell Functionality of Solid Tumors. Cells 2019; 9:cells9010052. [PMID: 31878283 PMCID: PMC7016859 DOI: 10.3390/cells9010052] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 12/06/2019] [Accepted: 12/18/2019] [Indexed: 02/08/2023] Open
Abstract
The regulation of the immune microenvironment within solid tumors has received increasing attention with the development and clinical success of immune checkpoint blockade therapies, such as those that target the PD-1/PD-L1 axis. The metabolic microenvironment within solid tumors has proven to be an important regulator of both the natural suppression of immune cell functionality and the de novo or acquired resistance to immunotherapy. Enzymatic proteins that generate immunosuppressive metabolites like adenosine are thus attractive targets to couple with immunotherapies to improve clinical efficacy. CD38 is one such enzyme. While the role of CD38 in hematological malignancies has been extensively studied, the impact of CD38 expression within solid tumors is largely unknown, though most current data indicate an immunosuppressive role for CD38. However, CD38 is far from a simple enzyme, and there are several remaining questions that require further study. To effectively treat solid tumors, we must learn as much about this multifaceted protein as possible—i.e., which infiltrating immune cell types express CD38 for functional activities, the most effective CD38 inhibitor(s) to employ, and the influence of other similarly functioning enzymes that may also contribute towards an immunosuppressive microenvironment. Gathering knowledge such as this will allow for intelligent targeting of CD38, the reinvigoration of immune functionality and, ultimately, tumor elimination.
Collapse
|
19
|
Duenas-Gonzalez A, Gonzalez-Fierro A. Pharmacodynamics of current and emerging treatments for cervical cancer. Expert Opin Drug Metab Toxicol 2019; 15:671-682. [DOI: 10.1080/17425255.2019.1648431] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Alfonso Duenas-Gonzalez
- Unit of Biomedical Research on Cancer, Instituto de Investigaciones Biomédicas UNAM/Instituto Nacional de Cancerología, Mexico City, Mexico
| | | |
Collapse
|
20
|
Human papillomavirus 16 infection alters the Toll-like receptors and downstream signaling cascade: A plausible early event in cervical squamous cell carcinoma development. Gynecol Oncol 2019; 155:151-160. [PMID: 31375269 DOI: 10.1016/j.ygyno.2019.07.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 07/19/2019] [Accepted: 07/22/2019] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Toll-like receptors constitute an important component of innate immune mechanism. HPV is a known etiological factor of cervical cancer and is known to interfere with the expression of TLRs and downstream signaling pathway. It remains poorly understood whether HPV modulates the expression of TLRs. Hence, understanding HPV mediated immune alterations might aid in identifying novel therapeutic targets. The aim was to study the relative gene expression of TLRs & downstream signaling pathway in cervical carcinoma. METHODS Cervical squamous cell carcinoma (CSCC) and normal cervical tissues were obtained. Subsequent to HPV genotyping, mRNA expression profiling using PCR Array was performed. Protein expression of relevant genes with western blot was studied. Levels of cytokines in cervicovaginal washes were estimated using a Luminex multiplex platform. RESULTS All cases of cervical cancer were HR-HPV positive and predominant subtype was HPV16 (71.1%). Significant TLR4 upregulation and TLR2,7 downregulation were observed in HR-HPV infected cervix. TLR4,7 demonstrated low expression in CSCC. Molecules from cancer allied pathways; RELA, AKT, CDKN2A, and MDM2 demonstrated upregulation in CSCC. Protein expression data corroborated with gene expression profile. A diminished level of Th1 cytokines TNF-α, IFN-ɣ, IL-17, and IL-12 was observed in CSCC. Significantly increased levels of IL-1β, IL-6 and IL-2 were detected in HR-HPV infected cervix. Kaplan Meier curve demonstrated high TLR4 and low TLR7 expression was associated with poor prognosis. CONCLUSION The study demonstrates the HPV mediated dampening of the innate immune response in CSCC and provides support for exploring potential TLR2, 7 agonists as an adjunct therapy in CSCC patients.
Collapse
|
21
|
Wu S, Li W, Wu Z, Cheng T, Wang P, Li N, Liang X, Chi M, Zhang S, Ma Y, Li Y, Chai L. TNFAIP8 promotes cisplatin resistance in cervical carcinoma cells by inhibiting cellular apoptosis. Oncol Lett 2019; 17:4667-4674. [PMID: 30944654 PMCID: PMC6444441 DOI: 10.3892/ol.2019.10076] [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: 05/19/2018] [Accepted: 01/21/2019] [Indexed: 12/16/2022] Open
Abstract
Cervical cancer is the second most prevalent malignant tumor in women worldwide. Failure of successful treatment is most prevalent in patients with the metastatic disease and the chemotherapy refractory disease. Tumor necrosis factor α-induced protein 8 (TNFAIP8) serves as an anti-apoptotic and pro-oncogenic protein, and is associated with cancer progression and poor prognosis in a number of different cancer types. However, the physiological and pathophysiological roles of TNFAIP8 in cervical carcinogenesis and development remain poorly understood. In the present study, it was demonstrated that TNFAIP8 protein expression levels were significantly increased in cervical cancer tissues compared with the non-tumor adjacent tissues using immunohistochemistry. Additionally, it was demonstrated that TNFAIP8 overexpression is associated with cisplatin resistance. Furthermore, depletion of TNFAIP8 impaired HeLa cell proliferation and viability in vitro, improved cisplatin sensitivity, and promoted cisplatin-induced cellular apoptosis and death. Subsequent mechanistic analysis demonstrated that TNFAIP8 silencing promoted caspase-8/-3 activation and p38 phosphorylation in HeLa cells treated with cisplatin, whereas apoptosis regulator B-cell lymphoma-2 expression was inhibited with TNFAIP8-silenced HeLa cells following treatment with cisplatin. These data suggested that TNFAIP8 serves as an anti-apoptotic protein against cisplatin-induced cell death, which eventually leads to chemotherapeutic drug-treatment failure. Therefore, the present data suggested that TNFAIP8 may be a promising therapeutic target for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Suxia Wu
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, P.R. China
- Department of Pathology, Henan University School of Basic Medical Sciences, Kaifeng, Henan 475004, P.R. China
| | - Weihua Li
- Clinical Laboratory, The First Affiliated Hospital of Henan University, Kaifeng, Henan 475001, P.R. China
| | - Zhenghui Wu
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, P.R. China
- Department of Immunology, Henan University School of Basic Medical Sciences, Kaifeng, Henan 475004, P.R. China
| | - Tianran Cheng
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Ping Wang
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, P.R. China
- Department of Immunology, Henan University School of Basic Medical Sciences, Kaifeng, Henan 475004, P.R. China
| | - Na Li
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, P.R. China
- Department of Immunology, Henan University School of Basic Medical Sciences, Kaifeng, Henan 475004, P.R. China
| | - Xiaonan Liang
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, P.R. China
- Department of Immunology, Henan University School of Basic Medical Sciences, Kaifeng, Henan 475004, P.R. China
| | - Mengmeng Chi
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Shuman Zhang
- Department of Gynaecology and Obstetrics, Affiliated Huaihe Hospital of Henan University, Kaifeng, Henan 475000, P.R. China
| | - Yuanfang Ma
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, P.R. China
- Department of Immunology, Henan University School of Basic Medical Sciences, Kaifeng, Henan 475004, P.R. China
| | - Yanyun Li
- Department of Gynaecology and Obstetrics, Affiliated Huaihe Hospital of Henan University, Kaifeng, Henan 475000, P.R. China
| | - Lihui Chai
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, P.R. China
- Department of Immunology, Henan University School of Basic Medical Sciences, Kaifeng, Henan 475004, P.R. China
| |
Collapse
|
22
|
Chen X, Xiong D, Ye L, Yang H, Mei S, Wu J, Chen S, Mi R. SPP1 inhibition improves the cisplatin chemo-sensitivity of cervical cancer cell lines. Cancer Chemother Pharmacol 2019; 83:603-613. [PMID: 30627777 DOI: 10.1007/s00280-018-3759-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 12/15/2018] [Indexed: 01/01/2023]
Abstract
PURPOSE Cisplatin (DDP)-based chemotherapy is a standard strategy for cervical cancer, while chemoresistance remains a huge challenge. In the present study, we aimed to explore the effects of SPP1 on the proliferation and apoptosis rate of the HeLa cervical cancer cell line with cisplatin (DDP) resistance. METHODS Microarray analysis was employed to select differentially expressed genes in cervical cancer tissues and adjacent tissues. Then, we established a DDP-resistant HeLa cell line (res-HeLa). Western blotting was used to detect SPP1 expression in both tissue and cells. After the transfection with si-SPP1 and pcDNA3.1-SPP1, colony formation and MTT assays were applied to detect cell proliferation changes. Flow cytometry was employed to detect the cell apoptosis rate. Western blotting was performed to verify the activation of PI3K/Akt signal pathway proteins related to DDP resistance. RESULTS SPP1 was overexpressed in cervical cancer tissues and cell lines. Compared to normal HeLa cells, expression of SPP1 was significantly enhanced in res-HeLa cells. SPP1 knockdown resulted in repressed proliferation and enhanced apoptosis of res-HeLa cells, which could be reversed by SPP1 overexpression in HeLa cells. Additionally, downregulation of SPP1 improved the DDP sensitivity of HeLa by inhibiting the PI3K/Akt signaling pathway. CONCLUSION SPP1 inhibition could suppress proliferation, induce apoptosis and increase the DDP chemo-sensitivity of HeLa cells.
Collapse
Affiliation(s)
- Xing Chen
- Department of Obstetrics and Gynecology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Linhai, 317000, Zhejiang, China
| | - Dongsheng Xiong
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China
| | - Liya Ye
- Department of Obstetrics and Gynecology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Linhai, 317000, Zhejiang, China
| | - Huichun Yang
- Department of Obstetrics and Gynecology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Linhai, 317000, Zhejiang, China
| | - Shuangshuang Mei
- Department of Obstetrics and Gynecology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Linhai, 317000, Zhejiang, China
| | - Jinhong Wu
- Department of Obstetrics and Gynecology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Linhai, 317000, Zhejiang, China
| | - Shanshan Chen
- Department of Obstetrics and Gynecology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Linhai, 317000, Zhejiang, China
| | - Ruoran Mi
- Department of Obstetrics and Gynecology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
23
|
Ge Y, Long Y, Xiao S, Liang L, He Z, Yue C, Wei X, Zhou Y. CD38 affects the biological behavior and energy metabolism of nasopharyngeal carcinoma cells. Int J Oncol 2018; 54:585-599. [PMID: 30535454 PMCID: PMC6317656 DOI: 10.3892/ijo.2018.4651] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/12/2018] [Indexed: 02/07/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is the most common malignant tumor type in Southern China and South-East Asia. Cluster of differentiation (CD)38 is highly expressed in the human immune system and participates in the activation of T, natural killer and plasma cells mediated by CD2 and CD3 through synergistic action. CD38 is a type II transmembrane glycoprotein, which was observed to mediate diverse activities, including signal transduction, cell adhesion and cyclic ADP-ribose synthesis. However, the significance of CD38 in NPC biological behavior and cellular energy metabolism has not been examined. In order to elucidate the effect of CD38 on the biological behavior of NPC cells, stable CD38-overexpressed NPC cell lines were established. It was demonstrated that CD38 promoted NPC cell proliferation with Cell Counting Kit-8 and colony formation assays. It was also indicated that CD38 inhibited cell senescence, and promoted cell metastasis. Furthermore, it was determined that CD38 promoted the conversion of cells to the S phase and decreased the content of reactive oxygen species and Ca2+. Additionally, cell metabolism assays demonstrated that CD38 increased the concentration of ATP, lactic acid, cyclic adenosine monophosphate and human ADP/acrp30 concentration in NPC cells. To investigate the possible mechanism, bioinformatics analysis and mass spectrometry technology was used to determine the most notably changing molecule and signaling pathways, and it was determined and verified that CD38 regulated the metabolic-associated signaling pathways associated with tumor protein 53, hypoxia inducible factor-1α and sirtuin 1. The present results indicated that CD38 may serve a carcinogenic role in NPC by regulating metabolic-associated signaling pathways.
Collapse
Affiliation(s)
- Yanshan Ge
- Department of Oncology, Hunan Provincial Tumor Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Yuehua Long
- Department of Oncology, Hunan Provincial Tumor Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Songshu Xiao
- Department of Gynecology and Obstetrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410078, P.R. China
| | - Lin Liang
- Department of Oncology, Hunan Provincial Tumor Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Zhengxi He
- Department of Oncology, Hunan Provincial Tumor Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Chunxue Yue
- Department of Oncology, Hunan Provincial Tumor Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Xiong Wei
- Department of Oncology, Hunan Provincial Tumor Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Yanhong Zhou
- Department of Oncology, Hunan Provincial Tumor Hospital, Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| |
Collapse
|
24
|
Liao S, Xiao S, Chen H, Zhang M, Chen Z, Long Y, Gao L, He J, Ge Y, Yi W, Wu M, Li G, Zhou Y. The receptor for activated protein kinase C promotes cell growth, invasion and migration in cervical cancer. Int J Oncol 2017; 51:1497-1507. [PMID: 29048616 PMCID: PMC5642390 DOI: 10.3892/ijo.2017.4137] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 09/21/2017] [Indexed: 01/21/2023] Open
Abstract
Cervical cancer is one of the most common malignant tumors in women all over the world. However, the exact etiology of cervical cancer remains unclear. The receptor for activated protein kinase C (RACK1) is reported to be involved in tumorigenesis and tumor progression. Besides, the prognostic value of RACK1 in several kinds of tumors has been identified. However, there are limited studies on the functional role of RACK1 in cervical cancer. In this study, we tested the expression level of RACK1 by immunohistochemistry and western blot technologies and find that it is upregulated in cervical cancer. Colony formation and CCK8 assays indicate that RACK1 promotes cell proliferation in CaSki cervical cancer cells. While the silence of RACK1 decreases the cell proliferation in CCK8 analysis. β-galactosidase staining suggests that RACK1 decreases cell senescence in cervical cancer cells. Invasion and migration assay show that RACK1 promotes the invasion and migration of cervical cancer cells. Also, when RACK1 was silenced, it exerts the opposite result. Furthermore, the mRNA expression levels of MMP‑3, MMP‑9 and MMP‑10 were upregulated in RACK1‑overexpressed CaSki cells by qPCR analysis. RACK1 also induces S phase accumulation in cell cycle analysis and suppresses cell apoptosis in cervical cancer cells. Flow cytometry analysis of mitochondria functions suggests that RACK1 increases the mitochondrial membrane potential (Δψm) levels to prevent mitochondrial apoptosis in cervical cancer cells. To explore the possible mechanism of RACK1, we tested and found that RACK1 upregulates the expression of NF-κB, cyclin D1 and CDK4 and downregulates the expression of p53, p38, p21 and STAT1 in cervical cancer cells. These results suggest that RACK1 promotes cell growth and invasion and inhibits the senescence and apoptosis in cervical cancer cells probably by affecting the p53 pathway.
Collapse
Affiliation(s)
- Shan Liao
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital
- The Key Laboratory of Carcinogenesis of The Chinese Ministry of Health, Xiangya Hospital
| | - Songshu Xiao
- Department of Gynecology and Obstetrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan
| | - Hongxiang Chen
- The Gynecology Department, People's Hospital of Xinjiang
| | - Manying Zhang
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital
| | - Zhifang Chen
- The Gynecology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumchi, Xinjiang, P.R. China
| | - Yuehua Long
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital
| | - Lu Gao
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital
| | - Junyu He
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital
| | - Yanshan Ge
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital
| | - Wei Yi
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital
| | - Minghua Wu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital
| | - Guiyuan Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital
- The Key Laboratory of Carcinogenesis of The Chinese Ministry of Health, Xiangya Hospital
| | - Yanhong Zhou
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital
- The Key Laboratory of Carcinogenesis of The Chinese Ministry of Health, Xiangya Hospital
| |
Collapse
|
25
|
Liao S, Xiao S, Chen H, Zhang M, Chen Z, Long Y, Gao L, Zhu G, He J, Peng S, Xiong W, Zeng Z, Li Z, Zhou M, Li X, Ma J, Wu M, Xiang J, Li G, Zhou Y. CD38 enhances the proliferation and inhibits the apoptosis of cervical cancer cells by affecting the mitochondria functions. Mol Carcinog 2017; 56:2245-2257. [PMID: 28544069 DOI: 10.1002/mc.22677] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 03/28/2017] [Accepted: 05/20/2017] [Indexed: 01/10/2023]
Abstract
Cervical cancer is one of the most common malignant tumors in women all over the world. The exact mechanism of occurrence and development of cervical cancer has not been fully elucidated. CD38 is a type II transmembrane glycoprotein, which was found to mediate diverse activities, including signal transduction, cell adhesion, and cyclic ADP-ribose synthesis. Here, we reported that CD38 promoted cell proliferation and inhibited cell apoptosis in cervical cancer cells by affecting the mitochondria functions. We established stable cervical cancer cell lines with CD38 over-expressed. CCK8 assay and colony formation assay indicated that CD38 promoted cervical cancer cell proliferation. Nude mouse tumorigenicity assay showed that CD38 significantly promotes tumor growth in vivo. CD38 also induced S phase accumulation in cell cycle analysis and suppressed cell apoptosis in cervical cancer cells. Meanwhile, flow cytometry analysis of mitochondria functions suggested that CD38 decreased intracellular Ca2+ levels in cervical cancer cells and CD38 was involved in down-regulation of ROS levels and prevented mitochondrial apoptosis in cervical cancer cells. The percentage of cells with loss of mitochondrial membrane potential (Δψm) in CD38-overexpressed cervical cancer cells was less than control groups. Furthermore, we found an up-regulation of MDM2, cyclinA1, CDK4, cyclinD1, NF-kB P65, c-rel, and a downregulation of P53, P21, and P38 by Western blot analysis. These results indicated that CD38 enhanced the proliferation and inhibited the apoptosis of cervical cancer cells by affecting the mitochondria functions.
Collapse
Affiliation(s)
- Shan Liao
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Songshu Xiao
- Department of Gynecology and Obstetrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongxiang Chen
- The Gynecology Department, People's Hospital of Xinjiang, Urumchi, Xinjiang, China
| | - Manying Zhang
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Zhifang Chen
- The Gynecology Department, The First Affiliated Hospital of Xinjiang Medical University, Urumchi, Xinjiang, China
| | - Yuehua Long
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Lu Gao
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Guangchao Zhu
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Junyu He
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Shuping Peng
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Zheng Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Ming Zhou
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xiaoling Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Jian Ma
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Minghua Wu
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Juanjuan Xiang
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Guiyuan Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yanhong Zhou
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
26
|
Bahrami A, Hasanzadeh M, Hassanian SM, ShahidSales S, Ghayour-Mobarhan M, Ferns GA, Avan A. The Potential Value of the PI3K/Akt/mTOR Signaling Pathway for Assessing Prognosis in Cervical Cancer and as a Target for Therapy. J Cell Biochem 2017; 118:4163-4169. [PMID: 28475243 DOI: 10.1002/jcb.26118] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/04/2017] [Indexed: 12/31/2022]
Abstract
Cervical cancer is a common gynecological cancer and a leading cause of cancer-related death in women globally. There is a need for the identification of prognostic and predictive biomarker for risk stratification. The phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway is often dysregulated in cervical cancer, indicating that it may be a potential therapeutic target in the treatment of this malignancy, and could perhaps be used as a novel biomarker in the assessment of risk of developing cervical cancer. We aimed to provide an overview of the potential applications of the PI3K/Akt/mTOR pathway as biomarker for risk stratification, in predicting the prognosis of cervical cancer, and for developing new therapeutic approaches in patients with cervical cancer. J. Cell. Biochem. 118: 4163-4169, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Afsane Bahrami
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Malihe Hasanzadeh
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Gynecology Oncology, Woman Health Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Majid Ghayour-Mobarhan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
27
|
Jiang Y, Xu H, Wang J. Alantolactone induces apoptosis of human cervical cancer cells via reactive oxygen species generation, glutathione depletion and inhibition of the Bcl-2/Bax signaling pathway. Oncol Lett 2016; 11:4203-4207. [PMID: 27313767 DOI: 10.3892/ol.2016.4511] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 12/17/2015] [Indexed: 01/06/2023] Open
Abstract
Alantolactone is the active ingredient in frankincense, and is extracted from the dry root of elecampane. It has a wide variety of uses, including as an insect repellent, antibacterial, antidiuretic, analgesic and anticancer agent. In addition, alantolactone induces apoptosis of human cervical cancer cells, however, its mechanism of action remains to be elucidated. Therefore, the present study investigated whether alantolactone was able to induce apoptosis of human cervical cancer cells, and its potential mechanisms of action were analyzed. Treatment of HeLa cells with alantolactone (0, 10, 20, 30, 40, 50 and 60 µM) for 12 h significantly inhibited growth in a dose-dependent manner. Cells treated with 30 µM of alantolactone for 0, 3, 6 and 12 h demonstrated marked induction of apoptosis in a time-dependent manner. Treatment of HeLa cells with 30 µM of alantolactone for 0, 3, 6 and 12 h significantly induced the generation of reactive oxygen species (ROS) and inhibited glutathione (GSH) production in HeLa cells in a dose-dependent manner. Alantolactone additionally markedly inhibited the Bcl-2/Bax signaling pathway in HeLa cells. Therefore, administration of alantolactone induced apoptosis of human cervical cancer cells via ROS generation, GSH depletion and inhibition of the Bcl-2/Bax signaling pathway.
Collapse
Affiliation(s)
- Yan Jiang
- Department of Gynecological Oncology, Anhui Cancer Hospital, Hefei, Anhui 230031, P.R. China
| | - Hanjie Xu
- Department of Gynecology and Obstetrics, Anhui Cancer Hospital, Hefei, Anhui 230031, P.R. China
| | - Jiafei Wang
- Department of Gynecological Oncology, Anhui Cancer Hospital, Hefei, Anhui 230031, P.R. China
| |
Collapse
|
28
|
PEI ZHEN, ZHU GUANGCHAO, HUO XIAOLEI, GAO LU, LIAO SHAN, HE JUNYU, LONG YUEHUA, YI HONG, XIAO SONGSHU, YI WEI, CHEN PAN, LI XIAOLING, LI GUIYUAN, ZHOU YANHONG. CD24 promotes the proliferation and inhibits the apoptosis of cervical cancer cells in vitro. Oncol Rep 2015; 35:1593-601. [DOI: 10.3892/or.2015.4521] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 10/22/2015] [Indexed: 11/05/2022] Open
|
29
|
Zhong G, Chen X, Fang X, Wang D, Xie M, Chen Q. Fra-1 is upregulated in lung cancer tissues and inhibits the apoptosis of lung cancer cells by the P53 signaling pathway. Oncol Rep 2015; 35:447-53. [PMID: 26549498 DOI: 10.3892/or.2015.4395] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/23/2015] [Indexed: 11/05/2022] Open
Abstract
Fos-related antigen-1 (Fra-1) is a member of the activator protein-1 transcription factor superfamily. It plays important roles in oncogenesis in various types of malignancies. Herein, we investigated the expression of Fra-1 in lung cancer tissues by qPCR, immunohistochemistry, and western blot technologies. The results showed that Fra-1 was overexpressed in the lung cancer tissues when compared with the level in the adjacent non-cancerous tissues. To explore the possible mechanism of Fra-1 in lung cancer, we elucidated the effect of Fra-1 on the apoptosis of lung cancer H460 cells, and found that the rate of cell apoptosis was decreased in the H460/Fra-1 cells compared with the H460 or H460/vector cells. Cell apoptosis is closely related with a reduction in mitochondrial membrane potential (ΔΨm) and an increase in intracellular reactive oxygen species (ROS) and calcium ion (Ca2+) concentrations. Our results showed that overexpression of Fra-1 in the lung cancer H460 cells, led to an increase in ΔΨm and and a decrease in intracellular ROS and Ca2+ concentrations. Furthermore, we found that Fra-1 was correlated with dysregulation of the P53 signaling pathway in lung cancer tissues in vitro. At the same time, we found that Fra-1 overexpression affected the expression of MDM2 and P53 in vivo. In summary, our results suggest that Fra-1 is upregulated in lung cancer tissues and functions by affecting the P53 signaling pathway in lung cancer.
Collapse
Affiliation(s)
- Guangwei Zhong
- Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xi Chen
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xia Fang
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Dongsheng Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Mingxuan Xie
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Qiong Chen
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
30
|
He J, Zhu G, Gao L, Chen P, Long Y, Liao S, Yi H, Yi W, Pei Z, Wu M, Li X, Xiang J, Peng S, Ma J, Zhou M, Xiong W, Zeng Z, Xiang B, Tang K, Cao L, Li G, Zhou Y. Fra-1 is upregulated in gastric cancer tissues and affects the PI3K/Akt and p53 signaling pathway in gastric cancer. Int J Oncol 2015; 47:1725-34. [PMID: 26330014 DOI: 10.3892/ijo.2015.3146] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 07/30/2015] [Indexed: 11/06/2022] Open
Abstract
Gastric cancer is an aggressive disease that continues to have a daunting impact on global health. Fra-1 (FOSL1) plays important roles in oncogenesis in various malignancies. We investigated the expression of Fra-1 in gastric cancer (GC) tissues by qPCR, immunohistochemistry (IHC) and western blot technologies. The results showed that Fra-1 was overexpressed in gastric cancer tissues compared with the adjacent non‑cancerous tissues. To explore the possible mechanism of Fra-1 in GC, we elucidated the effect of Fra-1 in the apoptosis and cell cycle of gastric cancer cells, AGS, and found that a considerable decrease in apoptotic cells and increase of S phase rate were observed for AGS cells with Fra-1 overexpession. We identified and confirmed that Fra-1 affected the expression level of CTTN and EZR in vitro through LC-MS/MS analyses and western blot technology. Furthermore, we found that Fra-1 was correlated with dysregulation PI3K/Akt and p53 signaling pathway in gastric cancer tissues in vitro. Moreover, we found that Fra-1 overexpression affected the expression of PI3K, Akt, MDM2 and p53 in vivo. In summary, our results suggest that Fra-1 is upregulated in gastric cancer tissues and plays its function by affecting the PI3K/Akt and p53 signaling pathway in gastric cancer.
Collapse
Affiliation(s)
- Junyu He
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Guangchao Zhu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Lu Gao
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Pan Chen
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Yuehua Long
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Shan Liao
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Hong Yi
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Wei Yi
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Zhen Pei
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Minghua Wu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Xiaoling Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Juanjuan Xiang
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Shuping Peng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Jian Ma
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Ming Zhou
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Wei Xiong
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Zhaoyang Zeng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Bo Xiang
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Ke Tang
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Li Cao
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Guiyuan Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Yanhong Zhou
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| |
Collapse
|
31
|
Zhu GC, Gao L, He J, Long Y, Liao S, Wang H, Li X, Yi W, Pei Z, Wu M, Xiang J, Peng S, Ma J, Zhou M, Zeng Z, Xiang B, Xiong W, Tang K, Cao L, Li X, Li G, Zhou Y. CD90 is upregulated in gastric cancer tissues and inhibits gastric cancer cell apoptosis by modulating the expression level of SPARC protein. Oncol Rep 2015; 34:2497-506. [PMID: 26329007 DOI: 10.3892/or.2015.4243] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 07/24/2015] [Indexed: 01/08/2023] Open
Abstract
Cluster of differentiation 90 (CD90) (Thy-1) plays important roles in the oncogenesis in various types of malignancies. In the present study, we investigated the expression of CD90 in gastric cancer (GC) tissues by q-PCR, immunohistochemistry (IHC), and western blot technologies. The results showed that CD90 was overexpressed in gastric cancer tissues compared with the level in the adjacent non‑cancerous tissues. To explore the possible mechanism of CD90 in GC, we elucidated the effect of CD90 on the apoptosis of AGS gastric cancer cells, and found that a considerable decrease in apoptotic cells was observed for AGS cells with CD90 overexpression. Meanwhile, the rate of apoptotic cells was increased in the AGS cells with CD90 interference (siCD90) compared with that in the AGS cells. Cell apoptosis is closely related to a reduction in mitochondrial membrane potential (ΔΨm) and an increase in intracellular reactive oxygen species (ROS) and calcium ion (Ca2+) concentrations. Our results showed that overexpression of CD90 in the AGS gastric cancer cells led to an increase in ΔΨm and a decrease in intracellular ROS and Ca2+ concentrations. At the same time, siCD90 reduced ΔΨm and the increase in intracellular ROS and Ca2+ concentrations. Furthermore, we identified and confirmed that CD90 functions by modulating the expression level of secreted protein, acidic, cysteine‑rich (osteonectin) (SPARC) in vitro through LC‑MS/MS analyses and western blot technology. In summary, our results suggest that CD90 is upregulated in gastric cancer and inhibits gastric cancer cell apoptosis by modulating the expression level of SPARC protein.
Collapse
Affiliation(s)
- Guang Chao Zhu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Lu Gao
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Junyu He
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Yuehua Long
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Shan Liao
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Haiyun Wang
- School of Life Science and Technology, Tongji University, Shanghai 200092, P.R. China
| | - Xujuan Li
- School of Life Science and Technology, Tongji University, Shanghai 200092, P.R. China
| | - Wei Yi
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Zhen Pei
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Minghua Wu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Juanjuan Xiang
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Shuping Peng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Jian Ma
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Ming Zhou
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Zhaoyang Zeng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Bo Xiang
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Wei Xiong
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Ke Tang
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Li Cao
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Xiaoling Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Guiyuan Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Yanhong Zhou
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| |
Collapse
|
32
|
Xiao S, Zhou Y, Yi W, Luo G, Jiang B, Tian Q, Li Y, Xue M. Fra-1 is downregulated in cervical cancer tissues and promotes cervical cancer cell apoptosis by p53 signaling pathway in vitro. Int J Oncol 2015; 46:1677-84. [PMID: 25651840 DOI: 10.3892/ijo.2015.2873] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/22/2015] [Indexed: 11/05/2022] Open
Abstract
Cervical cancer is a potentially preventable disease; however, it is the third most commonly diagnosed cancer and the fourth leading cause of cancer deaths in women worldwide. Cervical cancer is thought to develop through a multistep process involving virus, tumor suppressor genes, proto-oncogenes and immunological factors. It is known that human papillomavirus (HPV) infection is necessary but insufficient to cause malignancy. At present, the etiology of cervical carcinoma remains poorly understood. In this study, we found that the expression of FOS-like antigen-1 (Fra-1) gene was downregulated in cervical cancer compared with the adjacent non-cancerous tissues by RT-qPCR, immunohistochemistry (IHC) and western blotting techniques. To uncover the effect of Fra-1 on cervical cancer, we tested and confirmed that Fra-1 significantly inhibited the proliferation of HeLa cells by MMT assays in vitro. At the same time, overexpression of Fra-1 promoted apoptosis of HeLa cells. To explore the possible mechanism of Fra-1 in cervical cancer, we tested the expression levels of key molecules in p53 signaling pathway by western blotting technology. The results showed that p53 was downregulated in cervical cancer compared with the adjacent non-cancerous tissues, but MDM2 proto-oncogene, E3 ubiquitin protein ligase (MDM2) was upregulated in cervical cancer. In vitro, the p53 was upregulated and MDM2 was downregulated in HeLa cells with Fra-1 overexpression. In summary, our results suggested that Fra-1 expression is low in cervical cancer tissues and promotes apoptosis of cervical cancer cells by p53 signaling pathway.
Collapse
Affiliation(s)
- Songshu Xiao
- Department of Gynecology and Obstetrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Yanhong Zhou
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Wei Yi
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Guijuan Luo
- Department of Gynecology and Obstetrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Bin Jiang
- Department of Gynecology and Obstetrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Qi Tian
- Department of Gynecology and Obstetrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Yueran Li
- Department of Gynecology and Obstetrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Min Xue
- Department of Gynecology and Obstetrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|