1
|
Cho HJ, Jung HJ. Cyclophilin A knockdown inhibits the proliferation and metastatic ability of AGS gastric cancer stem cells by downregulating CD147/STAT3/AKT/ERK and epithelial‑mesenchymal transition. Mol Med Rep 2025; 31:14. [PMID: 39513611 PMCID: PMC11551680 DOI: 10.3892/mmr.2024.13379] [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: 09/06/2024] [Accepted: 10/17/2024] [Indexed: 11/15/2024] Open
Abstract
Gastric cancer stem cells (GCSCs) contribute to the challenging aspects of gastric cancer, such as progression, metastasis, treatment resistance and recurrence. Inhibitors targeting cyclophilin A (CypA) have shown potential in curtailing GCSC growth. Building upon this, the current study delved deeper into understanding the functional role of CypA in controlling the proliferation and metastatic capabilities of GCSCs, employing CypA‑specific small interfering RNA. The results revealed that knockdown of CypA led to significant suppression of the growth and tumorsphere‑forming capacity of GCSCs derived from AGS cells. This effect was mediated by arresting the cell cycle at the G0/G1 and S phases, and promoting apoptosis. Furthermore, silencing of CypA exerted inhibitory effects on the migration and invasion of AGS GCSCs by modulating the process of epithelial‑mesenchymal transition. Notably, the observed antiproliferative and antimetastatic effects of CypA knockdown were associated with the downregulation of critical regulators of gastric cancer stemness, such as CD44, CD133, aldehyde dehydrogenase 1 family member A1, NANOG, OCT4 and SOX2. This regulation occurred through inactivation of the CD147/STAT3/AKT/ERK signaling pathway. Additionally, CypA knockdown effectively curbed in vivo tumor growth of AGS GCSCs in a chorioallantoic membrane assay using chick embryos. These findings underscore the critical role of CypA in promoting the proliferation and metastasis of GCSCs, highlighting its potential as an effective therapeutic target for eradicating GCSCs and improving gastric cancer treatment outcomes.
Collapse
Affiliation(s)
- Hee Jeong Cho
- Department of Life Science and Biochemical Engineering, Graduate School, Sun Moon University, Asan, Chungcheongnam 31460, Republic of Korea
| | - Hye Jin Jung
- Department of Life Science and Biochemical Engineering, Graduate School, Sun Moon University, Asan, Chungcheongnam 31460, Republic of Korea
- Department of Pharmaceutical Engineering and Biotechnology, Sun Moon University, Asan, Chungcheongnam 31460, Republic of Korea
| |
Collapse
|
3
|
Wang K, Chen X, Lin P, Wu J, Huang Q, Chen Z, Tian J, Wang H, Tian Y, Shi M, Qian M, Hui B, Zhu Y, Li L, Yao R, Bian H, Zhu P, Chen R, Chen L. CD147-K148me2-Driven Tumor Cell-Macrophage Crosstalk Provokes NSCLC Immunosuppression via the CCL5/CCR5 Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400611. [PMID: 38873823 PMCID: PMC11304266 DOI: 10.1002/advs.202400611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/15/2024] [Indexed: 06/15/2024]
Abstract
Immunosuppression is a major hallmark of tumor progression in non-small cell lung cancer (NSCLC). Cluster of differentiation 147 (CD147), an important pro-tumorigenic factor, is closely linked to NSCLC immunosuppression. However, the role of CD147 di-methylation in the immunosuppressive tumor microenvironment (TME) remains unclear. Here, di-methylation of CD147 at Lys148 (CD147-K148me2) is identified as a common post-translational modification (PTM) in NSCLC that is significantly associated with unsatisfying survival outcomes among NSCLC sufferers, especially those in the advanced stages of the disease. The methyltransferase NSD2 catalyzes CD147 to generate CD147-K148me2. Further analysis demonstrates that CD147-K148me2 reestablishes the immunosuppressive TME and promotes NSCLC progression. Mechanistically, this modification promotes the interaction between cyclophilin A (CyPA) and CD147, and in turn, increases CCL5 gene transcription by activating p38-ZBTB32 signaling, leading to increased NSCLC cell-derived CCL5 secretion. Subsequently, CD147-K148me2-mediated CCL5 upregulation facilitates M2-like tumor-associated macrophage (TAM) infiltration in NSCLC tissues via CCL5/CCR5 axis-dependent intercellular crosstalk between tumor cells and macrophages, which is inhibited by blocking CD147-K148me2 with the targeted antibody 12C8. Overall, this study reveals the role of CD147-K148me2-driven intercellular crosstalk in the development of NSCLC immunosuppression, and provides a potential interventional strategy for PTM-targeted NSCLC therapy.
Collapse
Affiliation(s)
- Ke Wang
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Xiaohong Chen
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Peng Lin
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Jiao Wu
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Qiang Huang
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
- School of MedicineShanghai UniversityShanghai200444China
| | - Zhi‐Nan Chen
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Jiale Tian
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Hao Wang
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Ye Tian
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Mingyan Shi
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Meirui Qian
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Bengang Hui
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
- Department of Thoracic Surgery of Tangdu HospitalFourth Military Medical UniversityXi'an710038China
| | - Yumeng Zhu
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Ling Li
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Rui Yao
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Huijie Bian
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Ping Zhu
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Ruo Chen
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Liang Chen
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
- School of MedicineShanghai UniversityShanghai200444China
| |
Collapse
|
4
|
Wang G, Liu X, Liu H, Zhang X, Shao Y, Jia X. A novel necroptosis related gene signature and regulatory network for overall survival prediction in lung adenocarcinoma. Sci Rep 2023; 13:15345. [PMID: 37714937 PMCID: PMC10504370 DOI: 10.1038/s41598-023-41998-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 09/04/2023] [Indexed: 09/17/2023] Open
Abstract
We downloaded the mRNA expression profiles of patients with LUAD and corresponding clinical data from The Cancer Genome Atlas (TCGA) database and used the Least Absolute Shrinkage and Selection Operator Cox regression model to construct a multigene signature in the TCGA cohort, which was validated with patient data from the GEO cohort. Results showed differences in the expression levels of 120 necroptosis-related genes between normal and tumor tissues. An eight-gene signature (CYLD, FADD, H2AX, RBCK1, PPIA, PPID, VDAC1, and VDAC2) was constructed through univariate Cox regression, and patients were divided into two risk groups. The overall survival of patients in the high-risk group was significantly lower than of the patients in the low-risk group in the TCGA and GEO cohorts, indicating that the signature has a good predictive effect. The time-ROC curves revealed that the signature had a reliable predictive role in both the TCGA and GEO cohorts. Enrichment analysis showed that differential genes in the risk subgroups were associated with tumor immunity and antitumor drug sensitivity. We then constructed an mRNA-miRNA-lncRNA regulatory network, which identified lncRNA AL590666. 2/let-7c-5p/PPIA as a regulatory axis for LUAD. Real-time quantitative PCR (RT-qPCR) was used to validate the expression of the 8-gene signature. In conclusion, necroptosis-related genes are important factors for predicting the prognosis of LUAD and potential therapeutic targets.
Collapse
Affiliation(s)
- Guoyu Wang
- Department of Traditional Chinese Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xue Liu
- Department of Respiration, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huaman Liu
- Department of General Medicine, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinyue Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yumeng Shao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinhua Jia
- Department of Respiration, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
5
|
Lan T, Zhao Y, Du Y, Ma C, Wang R, Zhang Q, Wang S, Wei W, Yuan H, Huang Q. Fabrication of a Novel Au Star@AgAu Yolk-Shell Nanostructure for Ovarian Cancer Early Diagnosis and Targeted Therapy. Int J Nanomedicine 2023; 18:3813-3824. [PMID: 37457800 PMCID: PMC10348339 DOI: 10.2147/ijn.s413457] [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: 03/21/2023] [Accepted: 06/22/2023] [Indexed: 07/18/2023] Open
Abstract
Purpose A novel CYPA-targeted, SiO2 encapsulated Au star@AgAu yolk-shell nanostructure (YSNS) was synthesized and used for ovarian cancer early diagnosis and therapy. Methods Diverse spectroscopic and microscopic methods were utilized to investigate the pattern of the yolk-shell nanostructure. In addition, in vitro and in vivo experiments were carried out. Results It can be found that the ratio of HAuCl4 and AgNO3 played a critical role in the constitution of the yolk-shell nanostructure. The as-prepared yolk-shell nanostructure showed excellent SERS performance, which could be utilized as SERS substrate for specific sensitivity analysis of ovarian cancer markers cyclophilin A (CYPA) with detectable limit of 7.76*10-10 μg/mL. In addition, the as-prepared yolk-shell nanostructure possessed outstanding photothermal performance, which could be used as photothermal agent for ovarian cancer therapy. Experiments in vitro and in vivo proved that the as-prepared yolk-shell nanostructures are ideal candidate for early diagnosis and therapy for ovarian cancer in one platform. Conclusion This work holds promise to offer a new method for the detection and therapy of ovarian cancer in the early stage.
Collapse
Affiliation(s)
- Ting Lan
- Medical Technology School of Xuzhou Medical University, Xuzhou City, Jiangsu, 221000, People’s Republic of China
| | - Yang Zhao
- Medical Technology School of Xuzhou Medical University, Xuzhou City, Jiangsu, 221000, People’s Republic of China
- Public Experimental Research Center of Xuzhou Medical University, Xuzhou City, Jiangsu, 221004, People’s Republic of China
| | - Yu Du
- Medical Technology School of Xuzhou Medical University, Xuzhou City, Jiangsu, 221000, People’s Republic of China
- Xuzhou Center for Disease Control and Prevention, Xuzhou City, Jiangsu, 221006, People’s Republic of China
| | - Chunyi Ma
- Medical Technology School of Xuzhou Medical University, Xuzhou City, Jiangsu, 221000, People’s Republic of China
| | - Rui Wang
- Medical Technology School of Xuzhou Medical University, Xuzhou City, Jiangsu, 221000, People’s Republic of China
| | - Qianlei Zhang
- Medical Technology School of Xuzhou Medical University, Xuzhou City, Jiangsu, 221000, People’s Republic of China
| | - Shanshan Wang
- Medical Technology School of Xuzhou Medical University, Xuzhou City, Jiangsu, 221000, People’s Republic of China
| | - Wenxian Wei
- Testing Center, Yangzhou University, Yangzhou City, Jiangsu, 225009, People’s Republic of China
| | - Honghua Yuan
- Public Experimental Research Center of Xuzhou Medical University, Xuzhou City, Jiangsu, 221004, People’s Republic of China
| | - Qingli Huang
- Medical Technology School of Xuzhou Medical University, Xuzhou City, Jiangsu, 221000, People’s Republic of China
- Public Experimental Research Center of Xuzhou Medical University, Xuzhou City, Jiangsu, 221004, People’s Republic of China
| |
Collapse
|
6
|
Xin S, Liu L, Li Y, Yang J, Zuo L, Cao P, Yan Q, Li S, Yang L, Cui T, Lu J. Cyclophilin A binds to AKT1 and facilitates the tumorigenicity of Epstein-Barr virus by mediating the activation of AKT/mTOR/NF-κB positive feedback loop. Virol Sin 2022; 37:913-921. [PMID: 36075565 PMCID: PMC9797372 DOI: 10.1016/j.virs.2022.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 08/31/2022] [Indexed: 01/19/2023] Open
Abstract
The AKT/mTOR and NF-κB signalings are crucial pathways activated in cancers including nasopharyngeal carcinoma (NPC), which is prevalent in southern China and closely related to Epstein-Barr virus (EBV) infection. How these master pathways are persistently activated in EBV-associated NPC remains to be investigated. Here we demonstrated that EBV-encoded latent membrane protein 1 (LMP1) promoted cyclophilin A (CYPA) expression through the activation of NF-κB. The depletion of CYPA suppressed cell proliferation and facilitated apoptosis. CYPA was able to bind to AKT1, thus activating AKT/mTOR/NF-κB signaling cascade. Moreover, the use of mTOR inhibitor, rapamycin, subverted the activation of the positive feedback loop, NF-κB/CYPA/AKT/mTOR. It is reasonable that LMP1 expression derived from initial viral infection is enough to assure the constant potentiation of AKT/mTOR and NF-κB signalings. This may partly explain the fact that EBV serves as a tumor-promoting factor with minimal expression of the viral oncoprotein LMP1 in malignancies. Our findings provide new insight into the understanding of causative role of EBV in tumorigenicity during latent infection.
Collapse
Affiliation(s)
- Shuyu Xin
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China,Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, China,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, 410078, China,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, China,Department of Hematology, National Clinical Research Center for Geriatric Disorders, Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410080, China
| | - Lingzhi Liu
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China,Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, China,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, 410078, China,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, China,Department of Hematology, National Clinical Research Center for Geriatric Disorders, Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410080, China
| | - Yanling Li
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China,Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, China,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, 410078, China
| | - Jing Yang
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, 410078, China
| | - Lielian Zuo
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, 410078, China
| | - Pengfei Cao
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, 410078, China,Department of Hematology, National Clinical Research Center for Geriatric Disorders, Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410080, China
| | - Qijia Yan
- Department of Hematology, National Clinical Research Center for Geriatric Disorders, Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410080, China
| | - Shen Li
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China,Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, China,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, 410078, China
| | - Li Yang
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China,Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, China,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, 410078, China
| | - Taimei Cui
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, China
| | - Jianhong Lu
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China,Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, China,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, 410078, China,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, China,Department of Hematology, National Clinical Research Center for Geriatric Disorders, Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410080, China,Corresponding author.
| |
Collapse
|
7
|
Han JM, Jung HJ. Cyclophilin A/CD147 Interaction: A Promising Target for Anticancer Therapy. Int J Mol Sci 2022; 23:ijms23169341. [PMID: 36012604 PMCID: PMC9408992 DOI: 10.3390/ijms23169341] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/12/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Cyclophilin A (CypA), which has peptidyl-prolyl cis-trans isomerase (PPIase) activity, regulates multiple functions of cells by binding to its extracellular receptor CD147. The CypA/CD147 interaction plays a crucial role in the progression of several diseases, including inflammatory diseases, coronavirus infection, and cancer, by activating CD147-mediated intracellular downstream signaling pathways. Many studies have identified CypA and CD147 as potential therapeutic targets for cancer. Their overexpression promotes growth, metastasis, therapeutic resistance, and the stem-like properties of cancer cells and is related to the poor prognosis of patients with cancer. This review aims to understand the biology and interaction of CypA and CD147 and to review the roles of the CypA/CD147 interaction in cancer pathology and the therapeutic potential of targeting the CypA/CD147 axis. To validate the clinical significance of the CypA/CD147 interaction, we analyzed the expression levels of PPIA and BSG genes encoding CypA and CD147, respectively, in a wide range of tumor types using The Cancer Genome Atlas (TCGA) database. We observed a significant association between PPIA/BSG overexpression and poor prognosis, such as a low survival rate and high cancer stage, in several tumor types. Furthermore, the expression of PPIA and BSG was positively correlated in many cancers. Therefore, this review supports the hypothesis that targeting the CypA/CD147 interaction may improve treatment outcomes for patients with cancer.
Collapse
Affiliation(s)
- Jang Mi Han
- Department of Life Science and Biochemical Engineering, Graduate School, Sun Moon University, Asan 31460, Korea
| | - Hye Jin Jung
- Department of Life Science and Biochemical Engineering, Graduate School, Sun Moon University, Asan 31460, Korea
- Department of Pharmaceutical Engineering and Biotechnology, Sun Moon University, Asan 31460, Korea
- Genome-Based BioIT Convergence Institute, Sun Moon University, Asan 31460, Korea
- Correspondence: ; Tel.: +82-41-530-2354; Fax: +82-41-530-2939
| |
Collapse
|
8
|
Li Y, Yang L. Cyclophilin A represses reactive oxygen species generation and death of hypoxic non-small-cell lung cancer cells by degrading thioredoxin-interacting protein. Cell Cycle 2022; 21:1996-2007. [PMID: 35579671 DOI: 10.1080/15384101.2022.2078615] [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: 11/03/2022] Open
Abstract
Cyclophilin A (cypA) is overexpressed in many types of carcinomas, including non-small-cell lung cancer (NSCLC). However, the effect of anoxia, a critical feature of the carcinoma cell microenvironment, on cypA expression in NSCLC is unknown. Here, formaldehyde-fixed and paraffin-embedded samples were collected from 60 subjects with NSCLC. The protein expression levels of cypA and hypoxia-inducible factor-1α (HIF-1α) were evaluated using immunohistochemistry. Kaplan-Meier analysis showed that subjects with high cypA expression had remarkably shorter progression-free survival than those with low cypA expression. Furthermore, cypA expression levels were significantly related to HIF-1α expression levels (Spearman's correlation=0.34, P<0.0001). To further assess the effect of cypA, an anoxic carcinoma cell model was established. CypA expression was remarkably upregulated in H1299 and A549 cell lines under hypoxic conditions. Overexpression of cypA restored hypoxia-impaired cell growth and prevented reactive oxygen species (ROS) production and cell death in hypoxic A549 and H1299 cells. However, these phenotypes were not altered by the inactive R55A mutant of cypA. Mechanistic studies demonstrated that cypA can bind to and degrade the tumor suppressor protein TXNIP in H1299 and A549 cells. Restored TXNIP expression in cypA-overexpressed and hypoxic NSCLC cells led to increased ROS levels and apoptotic cell numbers and decreased cell growth compared with cypA-overexpressed and hypoxic NSCLC cells. These findings indicate that anoxia results in an increase in cypA expression in NSCLC. Additionally, cypA served as an oncogene during hypoxia by interacting with TXNIP.
Collapse
Affiliation(s)
- Yang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi 710061, P.R. China
| | - Lan Yang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|