1
|
Zhang X, Li E, Kuang Y, Gai Y, Feng Y, Huang Y, Wei Z, Niu J, Yu S, Yang Z, Zhang Q, Sai B, Zhu Y. MTCH2 regulates NRF2-mediated RRM1 expression to promote melanoma proliferation and dacarbazine insensitivity. Cell Death Dis 2025; 16:268. [PMID: 40204724 PMCID: PMC11982210 DOI: 10.1038/s41419-025-07618-9] [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: 09/26/2024] [Revised: 03/09/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
Melanoma is among the 10 most prevalent malignant tumors, posing a significant threat to human health. A detailed understanding of the molecular mechanisms driving its progression is crucial for advancing treatment strategies and outcomes. Based on bioinformatic analysis and experimental validation, this study identified mitochondrial carrier homolog 2 (MTCH2) as a key regulator of melanoma proliferation. Mechanistically, MTCH2 enhanced the expression and nuclear translocation of nuclear factor (erythroid-derived-2)-like 2 (NRF2), which up-regulated ribonucleotide reductase subunit M1 (RRM1) expression, thereby promoting melanoma cell proliferation. Targeting RRM1 in combination with dacarbazine significantly inhibited tumor growth in nude mouse xenograft models. These findings elucidate a mechanistic link between MTCH2 and the NRF2-RRM1 axis in melanoma proliferation and highlight potential therapeutic targets for intervention.
Collapse
Affiliation(s)
- Xuedan Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Enjiang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Yingmin Kuang
- Department of Organ Transplantation, First Affiliated Hospital of Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Yanlong Gai
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Yu Feng
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Yu Huang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Zhenyan Wei
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Junzi Niu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Song Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Zhe Yang
- Department of Pathology, First Affiliated Hospital of Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Qiao Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Buqing Sai
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.
| | - Yuechun Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.
| |
Collapse
|
2
|
Liu H, Tan S, Zhao Z, Tang X, Li Z, Qi J. METTL3/YTDHF1 Stabilizes MTCH2 mRNA to Regulate Ferroptosis in Glioma Cells. FRONT BIOSCI-LANDMRK 2025; 30:25718. [PMID: 40018930 DOI: 10.31083/fbl25718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 12/08/2024] [Accepted: 12/12/2024] [Indexed: 03/01/2025]
Abstract
BACKGROUND Gliomas are aggressive brain tumors known for their poor prognosis and resistance to standard treatment options. Ferroptosis is an iron-dependent form of regulated cell death that has emerged as a promising target for cancer treatment. This study examined how the methyltransferase-like 3/YTH domain family protein 1 (METTL3/YTHDF1) axis influences ferroptosis and glioma progression by stabilizing mitochondrial carrier homolog 2 (MTCH2) messenger RNA (mRNA). METHODS MTCH2 expression in glioma tissues and cell lines was evaluated through quantitative real-time polymerase chain reaction (PCR) and western blot analyses. To assess the effects of MTCH2 knockdown and overexpression on glioma cell functions, we performed a series of functional assays, including cell viability, colony formation, and measurements of lipid reactive oxygen species (lipid ROS) and malondialdehyde (MDA) levels. Additionally, we conducted RNA immunoprecipitation (RIP) and RNA stability assays to explore the underlying mechanisms governing the interaction between METTL3, YTHDF1, and the stability of MTCH2 mRNA. RESULTS MTCH2 was significantly upregulated in glioma tissues and cell lines. Silencing of MTCH2 resulted in decreased glioma cell proliferation and induced ferroptosis, as evidenced by increased lipid peroxidation and ROS accumulation. Conversely, overexpression of MTCH2 enhanced glioma cell survival and reduced ferroptosis. METTL3-mediated N6-methyladenosine (m6A) modification enhanced MTCH2 mRNA stability by enabling YTHDF1 to bind and protect the modified mRNA from degradation. CONCLUSION The METTL3/YTHDF1/MTCH2 axis plays a critical role in glioma progression by inhibiting ferroptosis and promoting tumor cell survival. Targeting this pathway may provide a new and effective treatment strategy for glioma patients.
Collapse
Affiliation(s)
- Hongjun Liu
- Department of Neurosurgery, The Affiliated Hospital of North Sichuan Medical College, 637000 Nanchong, Sichuan, China
| | - Shasha Tan
- Department of Neurosurgery, The Affiliated Hospital of North Sichuan Medical College, 637000 Nanchong, Sichuan, China
| | - Zhenyu Zhao
- Department of Neurosurgery, The Affiliated Hospital of North Sichuan Medical College, 637000 Nanchong, Sichuan, China
| | - Xiaoping Tang
- Department of Neurosurgery, The Affiliated Hospital of North Sichuan Medical College, 637000 Nanchong, Sichuan, China
| | - Zhou Li
- Department of Neurosurgery, The Affiliated Nanchong Central Hospital of North Sichuan Medical College, 637000 Nanchong, Sichuan, China
| | - Jian Qi
- Department of Neurosurgery, The Affiliated Hospital of North Sichuan Medical College, 637000 Nanchong, Sichuan, China
| |
Collapse
|
3
|
Zhao Y, Wu S, Cao G, Song P, Lan CG, Zhang L, Sang YH. Mitochondrial carrier homolog 2 is important for mitochondrial functionality and non-small cell lung cancer cell growth. Cell Death Dis 2025; 16:95. [PMID: 39948081 PMCID: PMC11825924 DOI: 10.1038/s41419-025-07419-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 01/13/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025]
Abstract
Discovering new molecular targets for non-small cell lung cancer (NSCLC) is critically important. Enhanced mitochondrial function can promote NSCLC progression by enabling metabolic reprogramming, resistance to apoptosis, and increased cell proliferation. Mitochondrial carrier homolog 2 (MTCH2), located in the outer mitochondrial membrane, is pivotal in regulating mitochondrial activities. This study examines MTCH2 expression and its functional role in NSCLC. Bioinformatic analysis showed that MTCH2 is overexpressed in NSCLC tissues, correlating with poor prognosis and other key clinical parameters of the patients. In addition, single-cell sequencing data revealed higher MTCH2 expression levels in cancer cells of NSCLC tumor mass. Moreover, MTCH2 is also upregulated in locally-treated NSCLC tissues and multiple primary/established human NSCLC cells. In various NSCLC cells, silencing MTCH2 via targeted shRNA or knockout (KO) using the CRISPR/Cas9 method significantly hindered cell proliferation, migration and invasion, while inducing apoptosis. MTCH2 knockdown or KO robustly impaired mitochondrial function, as indicated by reduced mitochondrial respiration, decreased complex I activity, lower ATP levels, lower mitochondrial membrane potential (mitochondrial depolarization), and increased reactive oxygen species (ROS) production. Conversely, ectopic overexpression of MTCH2 in primary NSCLC cells enhanced mitochondrial complex I activity and ATP production, promoting cell proliferation and migration. In vivo, the intratumoral injection of MTCH2 shRNA adeno-associated virus (aav) impeded the growth of subcutaneous xenografts of primary NSCLC cells in nude mice. In MTCH2 shRNA aav-injected NSCLC xenograft tissues, there was decreases in MTCH2 expression, mitochondrial complex I activity, ATP content, and the glutathione (GSH)/glutathione disulfide (GSSG) ratio, but increase in thiobarbituric acid reactive substances (TBAR) activity. Additionally, MTCH2 silencing led to reduced Ki-67 staining but increased apoptosis in NSCLC xenografts. Collectively, these findings demonstrate that overexpressed MTCH2 promotes NSCLC cell growth potentially through the maintenance of mitochondrial hyper-function, highlighting MTCH2 as a novel and promising therapeutic target for treating this disease.
Collapse
Affiliation(s)
- Yong Zhao
- Department of Thoracic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Siyang Wu
- Respiratory Intensive Care Unit, Affiliated Hospital of YouJiang Medical University for Nationalities, Baise, China
| | - Guohong Cao
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Peidong Song
- Department of Cardiothoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Chang-Gong Lan
- Guangxi Key Laboratory of basic and translational research of Bone and Joint Degenerative Diseases, Guangxi Biomedical Materials Engineering Research Center for Bone and Joint Degenerative Diseases, Baise, China.
- Department of Orthopaedics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China.
| | - Lin Zhang
- Department of Thoracic Surgery, Suzhou Ninth People's Hospital Affiliated to Soochow University, Suzhou, China.
| | - Yong-Hua Sang
- Department of Cardiothoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
4
|
Liu Y, Chen A, Wu Y, Ni J, Wang R, Mao Y, Sun N, Mi Y. Identification of mitochondrial carrier homolog 2 as an important therapeutic target of castration-resistant prostate cancer. Cell Death Dis 2025; 16:70. [PMID: 39910035 PMCID: PMC11799199 DOI: 10.1038/s41419-025-07406-5] [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: 09/21/2024] [Revised: 01/12/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025]
Abstract
We here investigate the expression of the mitochondrial carrier homolog 2 (MTCH2) and its potential function in castration-resistant prostate cancer (CRPC). Bioinformatic analyses reveal that MTCH2 overexpression is associated with critical clinical parameters of prostate cancer. Single-cell sequencing data indicate elevated MTCH2 expression in the prostate cancer epithelium. MTCH2 is also upregulated in locally treated CRPC tissue and various primary human CRPC cells. Using genetic silencing via shRNA and knockout (KO) through the CRISPR-sgRNA approach, we showed that the depletion of MTCH2 impaired mitochondrial function, resulting in a reduced oxygen consumption rate, diminished complex I activity, and decreased ATP levels, mitochondrial depolarization, and increased reactive oxygen species production in primary CRPC cells. The silencing or KO of MTCH2 significantly inhibited cell viability, proliferation, and migration, together with a marked increase in apoptosis in the primary CRPC cells. In contrast, ectopic expression of MTCH2 provided CRPC cells with pro-tumorigenic properties, enhancing ATP production and promoting cell proliferation and migration. MTCH2 silencing also markedly inhibited the growth of subcutaneous xenografts of the primary CRPC cells in nude mice. The MTCH2-silenced xenografts exhibited increased apoptosis, elevated lipid peroxidation, and decreased ATP levels. These results provide new insights into the role of MTCH2 in supporting mitochondrial function and CRPC progression.
Collapse
Affiliation(s)
- Yankui Liu
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Anjie Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yufan Wu
- Department of Urology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, China
| | - Jiang Ni
- Department of Pharmacy, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Rong Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yong Mao
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China.
| | - Ning Sun
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.
| | - Yuanyuan Mi
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China.
| |
Collapse
|
5
|
Ahmed A, Iaconisi GN, Di Molfetta D, Coppola V, Caponio A, Singh A, Bibi A, Capobianco L, Palmieri L, Dolce V, Fiermonte G. The Role of Mitochondrial Solute Carriers SLC25 in Cancer Metabolic Reprogramming: Current Insights and Future Perspectives. Int J Mol Sci 2024; 26:92. [PMID: 39795950 PMCID: PMC11719790 DOI: 10.3390/ijms26010092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/23/2024] [Accepted: 12/23/2024] [Indexed: 01/30/2025] Open
Abstract
Cancer cells undergo remarkable metabolic changes to meet their high energetic and biosynthetic demands. The Warburg effect is the most well-characterized metabolic alteration, driving cancer cells to catabolize glucose through aerobic glycolysis to promote proliferation. Another prominent metabolic hallmark of cancer cells is their increased reliance on glutamine to replenish tricarboxylic acid (TCA) cycle intermediates essential for ATP production, aspartate and fatty acid synthesis, and maintaining redox homeostasis. In this context, mitochondria, which are primarily used to maintain energy homeostasis and support balanced biosynthesis in normal cells, become central organelles for fulfilling the heightened biosynthetic and energetic demands of proliferating cancer cells. Mitochondrial coordination and metabolite exchange with other cellular compartments are crucial. The human SLC25 mitochondrial carrier family, comprising 53 members, plays a pivotal role in transporting TCA intermediates, amino acids, vitamins, nucleotides, and cofactors across the inner mitochondrial membrane, thereby facilitating this cross-talk. Numerous studies have demonstrated that mitochondrial carriers are altered in cancer cells, actively contributing to tumorigenesis. This review comprehensively discusses the role of SLC25 carriers in cancer pathogenesis and metabolic reprogramming based on current experimental evidence. It also highlights the research gaps that need to be addressed in future studies. Understanding the involvement of these carriers in tumorigenesis may provide valuable novel targets for drug development.
Collapse
Affiliation(s)
- Amer Ahmed
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy; (A.A.); (D.D.M.); (A.C.); (A.S.); (L.P.)
| | - Giorgia Natalia Iaconisi
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (G.N.I.); (L.C.)
| | - Daria Di Molfetta
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy; (A.A.); (D.D.M.); (A.C.); (A.S.); (L.P.)
| | - Vincenzo Coppola
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH 43210, USA;
| | - Antonello Caponio
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy; (A.A.); (D.D.M.); (A.C.); (A.S.); (L.P.)
| | - Ansu Singh
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy; (A.A.); (D.D.M.); (A.C.); (A.S.); (L.P.)
| | - Aasia Bibi
- Department of Translational Biomedicine and Neuroscience, University of Bari, 70125 Bari, Italy;
| | - Loredana Capobianco
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (G.N.I.); (L.C.)
| | - Luigi Palmieri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy; (A.A.); (D.D.M.); (A.C.); (A.S.); (L.P.)
| | - Vincenza Dolce
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Giuseppe Fiermonte
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy; (A.A.); (D.D.M.); (A.C.); (A.S.); (L.P.)
| |
Collapse
|
6
|
Sun G, Song Y, Li C, Sun B, Li C, Sun J, Xiao P, Zhang Z. MTCH2 promotes the malignant progression of ovarian cancer through the upregulation of AIMP2 expression levels, mitochondrial dysfunction and by mediating energy metabolism. Oncol Lett 2024; 28:492. [PMID: 39185493 PMCID: PMC11342418 DOI: 10.3892/ol.2024.14625] [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: 02/21/2023] [Accepted: 07/11/2024] [Indexed: 08/27/2024] Open
Abstract
Ovarian cancer (OC) is a gynecological malignancy that ranks among the most common female cancers worldwide and notably reduces a patient's quality of life. Mitochondrial carrier homology 2 (MTCH2) is a mitochondrial outer membrane protein that serves a regulatory role in mitochondrial metabolism and cell death. The precise contribution and underlying molecular pathways of MTCH2 in the context of OC development is currently unclear. The present study aimed to investigate the roles of MTCH2 in the energy metabolism, cell proliferation and metastatic potential of OC cells and evaluate the regulatory relationship between MTCH2, aminoacyl transfer RNA synthetase-interacting multifunctional protein 2 (AIMP2) and claudin-3. An analysis of 67 patients with high-grade serous OC demonstrated increased expression levels of MTCH2, AIMP2 and claudin-3 in OC tumor tissue samples compared with in corresponding normal tissues adjacent to OC tissue samples. MTCH2 overexpression was significantly associated with the International Federation of Gynecology and Obstetrics stage and tumor differentiation of the OC tumor samples. In vitro experiments using the SK-OV-3 OC cell line demonstrated that MTCH2 exerts a regulatory effect on the cell proliferation, invasion and migratory capabilities of these cells. Knockdown of MTCH2 reduced ATP production, induced mitochondrial dysfunction and promoted cytoskeleton remodeling and apoptosis in SK-OV-3 OC cells. In addition, MTCH2 knockdown downregulated the expression levels of both claudin-3 and AIMP2 proteins. Knockdown of AIMP2 inhibited the regulatory effect of MTCH2. Co-immunoprecipitation experiments demonstrated that MTCH2 interacts with AIMP2 and claudin-3. The present study provides novel insights into the treatment of OC metastasis, as MTCH2 was demonstrated to serve roles in the progression of OC cells through the regulation of claudin-3 via AIMP2, which could provide novel insights into the treatment of ovarian cancer metastasis.
Collapse
Affiliation(s)
- Guangyu Sun
- Department of Gynecology, Cangzhou People's Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Yanmin Song
- Department of Gynecology, Cangzhou People's Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Congxian Li
- Department of Gynecology, Cangzhou People's Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Bo Sun
- Department of Gynecology, Cangzhou People's Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Chengcheng Li
- Department of Gynecology, Cangzhou People's Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Jinbao Sun
- Department of Gynecology, Cangzhou People's Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Ping Xiao
- Department of Gynecology, Cangzhou People's Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Zhengmao Zhang
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Hebei Cancer Hospital, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
7
|
Yu J, Xue Y. Preparation of transferrin-targeted temozolomide nano-micelles and their anti-glioma effect. PeerJ 2024; 12:e17979. [PMID: 39285923 PMCID: PMC11404485 DOI: 10.7717/peerj.17979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/06/2024] [Indexed: 09/19/2024] Open
Abstract
Objectives This study aims to develop brain-targeted temozolomide (TMZ) nanograins using the biodegradable polymer material PEG-PLA as a carrier. The model drug TMZ was encapsulated within the polymer using targeted nanotechnology. Key characteristics such as appearance, particle size, size distribution, drug loading capacity, in vitro release rate, stability, and anti-tumor effects were systematically evaluated through in vitro experiments. Methods Transmission electron microscopy (TEM) and Malvern size analyzer were employed to observe the morphological and particle size features of the TMZ nanospheres at various time points to assess stability. The effects of TMZ nanograins on glioma cell viability and apoptosis were evaluated using MTT assays and flow cytometry. Results The targeted TMZ nano-micelles were successfully synthesized. After loading and targeted modifications, the particle size increased from 50.7 to 190 nm, indicating successful encapsulation of TMZ. The average particle size of the nano-micelles remained stable around 145 ± 10 nm at 1 day, 15 days, and 30 days post-preparation. The release rate of the nano-micelles was monitored at 2 h, 12 h, 24 h, and 48 h post-dialysis, ultimately reaching 95.8%. Compared to TMZ alone, the TMZ-loaded PEG-PLA nano-micelles exhibited enhanced cytotoxicity and apoptosis in glioma cells. This was accompanied by increased mitochondrial membrane potential and reactive oxygen species (ROS) levels following treatment with the TMZ nano-micelles. Conclusions TMZ-loaded nano-micelles demonstrated a gradual release profile and significantly enhanced inhibitory effects on human glioma U251 cells compared to TMZ alone. The findings suggest that TMZ-loaded PEG-PLA nano-micelles may offer a more effective therapeutic approach for glioma treatment.
Collapse
Affiliation(s)
- Jun Yu
- Department of Neurosurgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yonghua Xue
- Department of Neurosurgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
8
|
Peng X, Yang Y, Hou R, Zhang L, Shen C, Yang X, Luo Z, Yin Z, Cao Y. MTCH2 in Metabolic Diseases, Neurodegenerative Diseases, Cancers, Embryonic Development and Reproduction. Drug Des Devel Ther 2024; 18:2203-2213. [PMID: 38882047 PMCID: PMC11180440 DOI: 10.2147/dddt.s460448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/21/2024] [Indexed: 06/18/2024] Open
Abstract
Mitochondrial carrier homolog 2 (MTCH2) is a member of the solute carrier 25 family, located on the outer mitochondrial membrane. MTCH2 was first identified in 2000. The development in MTCH2 research is rapidly increasing. The most well-known role of MTCH2 is linking to the pro-apoptosis BID to facilitate mitochondrial apoptosis. Genetic variants in MTCH2 have been investigated for their association with metabolic and neurodegenerative diseases, however, no intervention or therapeutic suggestions were provided. Recent studies revealed the physiological and pathological function of MTCH2 in metabolic diseases, neurodegenerative diseases, cancers, embryonic development and reproduction via regulating mitochondrial apoptosis, metabolic shift between glycolysis and oxidative phosphorylation, mitochondrial fusion/fission, epithelial-mesenchymal transition, etc. This review endeavors to assess a total of 131 published articles to summarise the structure and physiological/pathological role of MTCH2, which has not previously been conducted. This review concludes that MTCH2 plays a crucial role in metabolic diseases, neurodegenerative diseases, cancers, embryonic development and reproduction, and the predominant molecular mechanism is regulation of mitochondrial function. This review gives a comprehensive state of current knowledgement on MTCH2, which will promote the therapeutic research of MTCH2.
Collapse
Affiliation(s)
- Xiaoqing Peng
- School of Pharmacy, Anhui Medical University, Hefei, People’s Republic of China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Hefei, Anhui, People’s Republic of China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Key National Health Commission Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, People’s Republic of China
| | - Yuanyuan Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Key National Health Commission Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, People’s Republic of China
| | - Ruirui Hou
- School of Pharmacy, Anhui Medical University, Hefei, People’s Republic of China
| | - Longbiao Zhang
- School of Pharmacy, Anhui Medical University, Hefei, People’s Republic of China
| | - Can Shen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Xiaoyan Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Zhigang Luo
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Zongzhi Yin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Key National Health Commission Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, People’s Republic of China
| | - Yunxia Cao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Key National Health Commission Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, People’s Republic of China
| |
Collapse
|
9
|
Jiang W, Miao Y, Xing X, Liu S, Xing W, Qian F. MTCH2 stimulates cellular proliferation and cycles via PI3K/Akt pathway in breast cancer. Heliyon 2024; 10:e28172. [PMID: 38560664 PMCID: PMC10979243 DOI: 10.1016/j.heliyon.2024.e28172] [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: 04/06/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024] Open
Abstract
The MTCH2 protein is located on the mitochondrial outer membrane and regulates mitochondria-related cell death. This study set out to investigate the role of MTCH2 in the underlying pathophysiological mechanisms of breast cancer (BC). MTCH2 expression levels in BC were analyzed using bioinformatics prior to verification by cell lines in vitro. Experiments of over-expression and siRNA-mediated knockdown of MTCH2 were conducted to assess its biological functions, including its effects on cellular proliferation and cycle progression. Xenografts were utilised for in vivo study and signaling pathway alterations were examined to identify the mechanisms driven by MTCH2 in BC proliferation and cell-cycle regulation. MTCH2 was up-regulated in BC and correlated with patients' overall survival. Over-expression of MTCH2 promoted cellular proliferation and cycle progression, while silencing MTCH2 had the opposite effect. Xenograft experiments were utilised to confirm the in vitro cellular findings and it was identified that the PI3K/Akt signaling pathway was activated by MTCH2 over-expression and suppressed by its silencing. Moreover, the activation of IGF-1R rescued cellular growth and cycle arrest induced by MTCH2-silencing. Overall, this study reveals that expression of MTCH2 in BC is upregulated and potentiates cellular proliferation and cycle progression via the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Wenying Jiang
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
- Department of Breast Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Yuxia Miao
- Department of Echocardiography, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Xiaoxiao Xing
- Department of Ultrasound Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Shuiqing Liu
- Department of Ultrasound Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Wei Xing
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Feng Qian
- Department of Ultrasound Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
- Department of Ultrasonography, People’s Hospital of Ziyang County, Ankang, 725399, China
| |
Collapse
|
10
|
Zhang JW, Huang LY, Li YN, Tian Y, Yu J, Wang XF. Mitochondrial carrier homolog 2 increases malignant phenotype of human gastric epithelial cells and promotes proliferation, invasion, and migration of gastric cancer cells. World J Gastrointest Oncol 2024; 16:991-1005. [PMID: 38577443 PMCID: PMC10989370 DOI: 10.4251/wjgo.v16.i3.991] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/21/2023] [Accepted: 01/19/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND The precise role of mitochondrial carrier homolog 2 (MTCH2) in promoting malignancy in gastric mucosal cells and its involvement in gastric cancer cell metastasis have not been fully elucidated. AIM To determine the role of MTCH2 in gastric cancer. METHODS We collected 65 samples of poorly differentiated gastric cancer tissue and adjacent tissues, constructed MTCH2-overexpressing and MTCH2-knockdown cell models, and evaluated the proliferation, migration, and invasion of human gastric epithelial cells (GES-1) and human gastric cancer cells (AGS) cells. The mitochondrial membrane potential (MMP), mitochondrial permeability transformation pore (mPTP) and ATP fluorescence probe were used to detect mitochondrial function. Mitochondrial function and ATP synthase protein levels were detected via Western blotting. RESULTS The expression of MTCH2 and ATP2A2 in gastric cancer tissues was significantly greater than that in adjacent tissues. Overexpression of MTCH2 promoted colony formation, invasion, migration, MMP expression and ATP production in GES-1 and AGS cells while upregulating ATP2A2 expression and inhibiting cell apoptosis; knockdown of MTCH2 had the opposite effect, promoting overactivation of the mPTP and promoting apoptosis. CONCLUSION MTCH2 can increase the malignant phenotype of GES-1 cells and promote the proliferation, invasion, and migration of gastric cancer cells by regulating mitochondrial function, providing a basis for targeted therapy for gastric cancer cells.
Collapse
Affiliation(s)
- Jing-Wen Zhang
- School of Basic Medical Science, NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ling-Yan Huang
- Department of Pathology, General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ya-Ning Li
- School of Basic Medical Science, NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ying Tian
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Jia Yu
- School of Basic Medical Science, NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Xiao-Fei Wang
- Department of Pathology, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, Hebei Province, China
| |
Collapse
|
11
|
Zheng X, Chu B. The biology of mitochondrial carrier homolog 2. Mitochondrion 2024; 75:101837. [PMID: 38158152 DOI: 10.1016/j.mito.2023.101837] [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: 08/08/2023] [Revised: 12/24/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
The mitochondrial carrier system is in charge of small molecule transport between the mitochondria and the cytoplasm as well as being an integral portion of the core mitochondrial function. One member of the mitochondrial carrier family of proteins, mitochondrial carrier homolog 2 (MTCH2), is characterized as a critical mitochondrial outer membrane protein insertase participating in mitochondrial homeostasis. Accumulating evidence demonstrate that MTCH2 is integrally linked to cell death and mitochondrial metabolism, and its genetic alterations cause a variety of disease phenotypes, ranging from obesity, Alzheimer's disease, and tumor. To provide a comprehensive insight into the current understanding of MTCH2, we present a detailed description of the physiopathological functions of MTCH2, ranging from apoptosis, mitochondrial dynamics, and metabolic homeostasis regulation. Moreover, we summarized the impact of MTCH2 in human diseases, and highlighted tumors, to assess the role of MTCH2 mutations or variable expression on pathogenesis and target therapeutic options.
Collapse
Affiliation(s)
- Xiaohe Zheng
- Department of Pathology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, China
| | - Binxiang Chu
- Department of Orthopedic, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, China.
| |
Collapse
|
12
|
Mao M, Yuan Q, Xia X, Cui Y, Chen M, Yang W. Integrative analysis defines DDIT3 amplification as a correlative and essential factor for glioma malignancy. Am J Cancer Res 2023; 13:5418-5430. [PMID: 38058808 PMCID: PMC10695819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 10/14/2023] [Indexed: 12/08/2023] Open
Abstract
Glioma, particularly glioblastoma multiforme (GBM), is a highly aggressive and lethal primary brain tumor with poor prognosis. Metabolic reprogramming and endoplasmic reticulum (ER) stress are two crucial factors contributing to glioma pathogenesis. However, the intricate coordination between these processes remains incompletely understood. Here, we conducted an integrative analysis to elucidate the nodal role of DNA Damage Inducible Transcript 3 (DDIT3) to couple metabolisms and stress responses in glioma. We demonstrated a positive association between DDIT3 amplification/enhanced expression with glioma malignancy, indicating its potential as a novel biomarker for prognosis and treatment stratification. Genomic and transcriptomic analyses further revealed the involvement of DDIT3 enhancement in glioma progression. Moreover, immune infiltration analysis showed that distinct DDIT3 expression groups had different immune microenvironment. Finally, in vitro validations confirmed the impact of DDIT3 on proliferation and migration of glioma cells. Our findings provide novel insights into the complex interplay between metabolic reprogramming and ER stress, and defines DDIT3 as a promising therapeutic target in glioma.
Collapse
Affiliation(s)
- Mengqian Mao
- Department of Neurosurgery, West China Hospital, Sichuan UniversityChengdu 610041, Sichuan, China
| | - Qiuyun Yuan
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan UniversityChengdu 610041, Sichuan, China
| | - Xiaoqiang Xia
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan UniversityChengdu 610041, Sichuan, China
| | - Yiyuan Cui
- Department of Neurosurgery, West China Hospital, Sichuan UniversityChengdu 610041, Sichuan, China
| | - Mina Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan UniversityChengdu 610041, Sichuan, China
| | - Wanchun Yang
- Department of Neurosurgery, West China Hospital, Sichuan UniversityChengdu 610041, Sichuan, China
| |
Collapse
|
13
|
Abdulla MH, Alzailai AA, Vaali-Mohammed MA, Ahmad R, Fatima S, Zubaidi A, Traiki TB, Mahmood A, Hamoud Alrashoudi R, Khan Z. The platinum coordination complex inhibits cell invasion-migration and epithelial-to-mesenchymal transition by altering the TGF-β-SMAD pathway in colorectal cancer. Front Pharmacol 2023; 14:1178190. [PMID: 38027033 PMCID: PMC10679924 DOI: 10.3389/fphar.2023.1178190] [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: 03/02/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction: There is a steady increase in colorectal cancer (CRC) incidences worldwide; at diagnosis, about 20 percent of cases show metastases. The transforming growth factor-beta (TGF-β) signaling pathway is one of the critical pathways that influence the expression of cadherins allowing the epithelial-to-mesenchymal transition (EMT), which is involved in the progression of the normal colorectal epithelium to adenoma and metastatic carcinoma. The current study aimed to investigate the impact of a novel coordination complex of platinum (salicylaldiminato) PT(II) complex with dimethyl propylene linkage (PT-complex) on TGF-β and EMT markers involved in the invasion and migration of the human HT-29 and SW620 CRC cell lines. Methods: Functional study and wound healing assay showed PT-complex significantly reduced cell motility and the migration and invasion of CRC cell lines compared to the untreated control. Western blot performed in the presence and absence of TGF-β demonstrated that PT-complex significantly regulated the TGF-β-mediated altered expressions of EMT markers. Results and Discussion: PT-complex attenuated the migration and invasion by upregulating the protein expression of EMT-suppressing factor E-cadherin and suppressing EMT-inducing factors such as N-Cadherin and Vimentin. Moreover, PT-complex significantly suppressed the activation of SMAD3 in both CRC cell lines. Further, the microarray data analysis revealed differential expression of genes related to invasion and migration. In conclusion, besides displaying antiproliferative activity, the PT complex can decrease the metastasis of CRC cell lines by modulating TGF-β-regulated EMT markers. These findings provide new insight into TGF-β/SMAD signaling as the molecular mechanism involved in the antitumoral properties of novel PT-complex.
Collapse
Affiliation(s)
- Maha-Hamadien Abdulla
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Aminah Ahmad Alzailai
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Mansoor-Ali Vaali-Mohammed
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Rehan Ahmad
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Sabiha Fatima
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed Zubaidi
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Thamer bin Traiki
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Amer Mahmood
- Stem Cell Unit, Department of Anatomy, King Saud University College of Medicine, Riyadh, Saudi Arabia
| | - Reem Hamoud Alrashoudi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Zahid Khan
- Genome Research Chair, Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
14
|
Qi X, Yuan Q, Xia X, Li W, Cao M, Yang W. Clinical and molecular analysis of cilia-associated gene signature for prognostic prediction in glioma. J Cancer Res Clin Oncol 2023; 149:11443-11455. [PMID: 37386136 DOI: 10.1007/s00432-023-05022-4] [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: 05/09/2023] [Accepted: 06/23/2023] [Indexed: 07/01/2023]
Abstract
PURPOSE Glioma is a highly malignant and unfavorable cancer in the brain. Recent evidence highlights the vital role of cilia-related pathways as novel regulators of glioma development. However, the prognostic potential of ciliary pathways in glioma is still ambiguous. In this study, we aim to construct a gene signature using cilia-related genes to facilitate the prognostication of glioma. METHODS A multi-stage approach was employed to build the ciliary gene signature for prognostication of glioma. The strategy involved the implementation of univariate, LASSO, and stepwise multivariate Cox regression analyses based on TCGA cohort, followed by independent validation in CGGA and REMBRANDT cohort. The study further revealed molecular differences at the genomic, transcriptomic, and proteomic levels between distinct groups. RESULTS A prognostic tool utilizing a 9-gene signature based on ciliary pathways was developed to assess the clinical outcomes of glioma patients. The risk scores generated by the signature demonstrated a negative correlation with patient survival rates. The validation of the signature in an independent cohort reinforced its prognostic capabilities. In-depth analysis uncovered distinctive molecular characteristics at the genomic, transcriptomic, and protein-interactive levels in the high- and low-risk groups. Furthermore, the gene signature was able to predict the sensitivity of glioma patients to conventional chemotherapeutic drugs. CONCLUSION This study has established the utility of a ciliary gene signature as a reliable prognostic predictor of glioma patient survival. Findings not only enhance our comprehension of the intricate molecular mechanisms of cilia pathways in glioma, but also hold significant clinical implications in directing chemotherapeutic strategies.
Collapse
Affiliation(s)
- Xin Qi
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Qiuyun Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoqiang Xia
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wenhao Li
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Muqing Cao
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Wanchun Yang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
15
|
Niyazi M, Han L, Husaiyin S, Aishanjiang A, Guo M, Muhaimati G, Rozi H, Sun H, Lu J, Ma C, Rouzi N, Liu X, Zhu K. Analysis of somatic mutations and key driving factors of cervical cancer progression. Open Med (Wars) 2023; 18:20230759. [PMID: 37533736 PMCID: PMC10390753 DOI: 10.1515/med-2023-0759] [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: 01/11/2023] [Revised: 05/29/2023] [Accepted: 06/26/2023] [Indexed: 08/04/2023] Open
Abstract
We investigated the somatic mutations and key driving factors of cervical cancer by whole exome sequencing . We found 22,183 somatic single nucleotide variations (SNVs) in 52 paired samples. Somatic SNVs in cervical cancer were significantly higher than those in high-grade intraepithelial lesion and low-grade squamous intraepithelial lesion groups (P < 0.05). C → T/G accounted for 44.12% of base substitution. Copy number variation (false discovery rate < 0.05) was found in 57 chromosome regions. The three regions with significant differences between cervical cancer and non-cervical cancer groups were 1q21.1, 3q26.33, and 13q33.1, covering genes related to tumor proliferation, differentiation, and apoptosis. The frequency of human papillomavirus (HPV) insertion/integration and the number of "tCw" mutations in the cervical cancer group were significantly higher than those in the non-cervical cancer group (P < 0.05). The total number of mutations was positively correlated with the number of "tCw" mutations (R 2 = 0.7967). HPV insertion/integration (OR = 2.302, CI = 1.523-3.589, P = 0.0005), APOBEC enrichment (OR = 17.875, CI = 2.117-150.937, P = 0.001), and HLA-B*39 in HLA-I (OR = 6.435, CI = 0.823-48.919, P = 0.0042) were risk factors for cervical cancer, while HLA-DQB1*05 in HLA-II was a protective factor (OR = 0.426, CI = 0.197-0.910, P = 0.032). Conclusively, HPV insertion/integration, APOBEC mutagenesis, and HLA polymorphisms are high-risk factors for cervical cancer and may be causes of genome instability and somatic mutations. This study provides experimental data for revealing the molecular mechanism of cervical cancer.
Collapse
Affiliation(s)
- Mayinuer Niyazi
- Department of Obstetrics and Gynecology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi830001, China
| | - Lili Han
- Department of Obstetrics and Gynecology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi830001, China
| | - Sulaiya Husaiyin
- Department of Obstetrics and Gynecology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi830001, China
| | - Ayimila Aishanjiang
- Department of Obstetrics and Gynecology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi830001, China
| | - Min Guo
- Department of Obstetrics and Gynecology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi830001, China
| | - Gulibanu Muhaimati
- Department of Obstetrics and Gynecology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi830001, China
| | - Hankez Rozi
- Department of Obstetrics and Gynecology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi830001, China
| | - Haiyan Sun
- Department of Obstetrics and Gynecology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi830001, China
| | - Jing Lu
- Department of Obstetrics and Gynecology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi830001, China
| | - Chunhua Ma
- Department of Obstetrics and Gynecology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi830001, China
| | - Nuermangul Rouzi
- Department of Obstetrics and Gynecology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi830001, China
| | - Xiaowan Liu
- Department of Obstetrics and Gynecology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi830001, China
| | - Kaichun Zhu
- Department of Obstetrics and Gynecology, People’s Hospital of Xinjiang Uygur Autonomous Region, No. 91, Tianchi Road, Tianshan District, Urumqi830001, China
| |
Collapse
|
16
|
Li R, He H, He X. APOC1 promotes the progression of osteosarcoma by binding to MTCH2. Exp Ther Med 2023; 25:163. [PMID: 36911382 PMCID: PMC9996334 DOI: 10.3892/etm.2023.11862] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 01/06/2023] [Indexed: 02/25/2023] Open
Abstract
Osteosarcoma is the most prevalent primary malignant bone cancer worldwide. Apolipoprotein C1 (APOC1) and mitochondrial carrier homolog 2 (MTCH2) have been identified to be upregulated during the oncogenesis and metastasis of osteosarcoma. The aim of the present study was to explore the role of APOC1 in osteosarcoma progression and the mechanisms associated with MTCH2. APOC1 and MTCH2 expression in osteosarcoma cells was assessed by reverse transcription-quantitative PCR and western blotting. Then, APOC1 was silenced to detect its effect on cell viability, proliferation and apoptosis using Cell Counting Kit-8, a colony formation assay and TUNEL staining, respectively. Transwell and wound healing assays were used to evaluate cell invasion and migration. The interaction between APOC1 and MTCH2 as predicted by the Biological General Repository for Interaction Datasets and the Search Tool for the Retrieval of Interacting Genes/Proteins databases was verified by co-immunoprecipitation assay. Subsequently, rescue experiments were performed to analyze the regulatory effects of APOC1 on MTCH2 in the biological behavior and Warburg effect of osteosarcoma cells. Significantly upregulated APOC1 and MTCH2 expression was found in osteosarcoma SAOS-2 cells. APOC1 silencing attenuated cell viability, inhibited proliferation and promoted cell apoptosis, coupled with the decreased Bcl-2 expression and increased Bax and cleaved-caspase 3 expression. The invasive and migratory capacities of SAOS-2 cells were also suppressed following APOC1 knockdown. Moreover, APOC1 was confirmed to interact with MTCH2 in osteosarcoma cells. MTCH2 upregulation inhibited the impacts of APOC1 deletion on the malignant behavior of osteosarcoma cells. APOC1 silencing-induced oxidative phosphorylation elevation and Warburg effect decrease were partially restored by MTCH2 upregulation. In sum, APOC1 promoted progression of osteosarcoma by binding to MTCH2, suggesting that targeting the APOC1/MTCH2 axis may be a potential treatment of osteosarcoma.
Collapse
Affiliation(s)
- Renjie Li
- School of Nursing, Sun Yat-Sen University, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Huixian He
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xinxin He
- School of Medicine, Foshan University, Foshan, Guangdong 528000, P.R. China
| |
Collapse
|
17
|
Khan I, Işık EB, Mahfooz S, Khan AM, Hatiboglu MA. Identification of Genetic Alterations in Rapid Progressive Glioblastoma by Use of Whole Exome Sequencing. Diagnostics (Basel) 2023; 13:diagnostics13061017. [PMID: 36980325 PMCID: PMC10047503 DOI: 10.3390/diagnostics13061017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Glioblastoma poses an inevitable threat to patients despite aggressive therapy regimes. It displays a great level of molecular heterogeneity and numerous substitutions in several genes have been documented. Next-generation sequencing techniques have identified various molecular signatures that have led to a better understanding of the molecular pathogenesis of glioblastoma. In this limited study, we sought to identify genetic variants in a small number of rare patients with aggressive glioblastoma. METHODS Five tumor tissue samples were isolated from four patients with rapidly growing glioblastoma. Genomic DNA was isolated and whole exome sequencing was used to study protein-coding regions. Generated FASTQ files were analyzed and variants were called for each sample. Variants were prioritized with different approaches and functional annotation was applied for the detrimental variants. RESULTS A total of 49,780 somatic variants were identified in the five glioblastoma samples studied, with the majority as missense substitutions. The top ten genes with the highest number of substitutions were MUC3A, MUC4, MUC6, OR4C5, PDE4DIP, AHNAK2, OR4C3, ZNF806, TTN, and RP1L1. Notably, variant prioritization after annotation indicated that the MTCH2 (Chr11: 47647265 A>G) gene sequence change was putative deleterious in all of the aggressive tumor samples. CONCLUSION The MTCH2 (Chr11: 47647265 A>G) gene substitution was identified as putative deleterious in highly aggressive glioblastomas, which merits further investigation. Moreover, a high tumor mutation burden was observed, with a signature of the highest substitutions in MUC3A, MUC4, MUC6, OR4C5, PDE4DIP, AHNAK2, OR4C3, ZNF806, TTN, and RP1L1 genes. The findings provide critical, initial data for the further rational design of genetic screening and diagnostic approaches against aggressive glioblastoma.
Collapse
Affiliation(s)
- Imran Khan
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Beykoz, 34820 Istanbul, Turkey
| | - Esra Büşra Işık
- Department of Microbiology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Beykoz, 34820 Istanbul, Turkey
| | - Sadaf Mahfooz
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Beykoz, 34820 Istanbul, Turkey
| | - Asif M Khan
- Department of Microbiology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Beykoz, 34820 Istanbul, Turkey
- Centre for Bioinformatics, School of Data Sciences, Perdana University, Damansara Heights, Kuala Lumpur 50490, Malaysia
| | - Mustafa Aziz Hatiboglu
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Beykoz, 34820 Istanbul, Turkey
- Department of Neurosurgery, Bezmialem Vakif University Medical School, Vatan Street, Fatih, 34093 Istanbul, Turkey
| |
Collapse
|
18
|
Green A, Hossain T, Eckmann DM. Mitochondrial dynamics involves molecular and mechanical events in motility, fusion and fission. Front Cell Dev Biol 2022; 10:1010232. [PMID: 36340034 PMCID: PMC9626967 DOI: 10.3389/fcell.2022.1010232] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/06/2022] [Indexed: 11/13/2022] Open
Abstract
Mitochondria are cell organelles that play pivotal roles in maintaining cell survival, cellular metabolic homeostasis, and cell death. Mitochondria are highly dynamic entities which undergo fusion and fission, and have been shown to be very motile in vivo in neurons and in vitro in multiple cell lines. Fusion and fission are essential for maintaining mitochondrial homeostasis through control of morphology, content exchange, inheritance of mitochondria, maintenance of mitochondrial DNA, and removal of damaged mitochondria by autophagy. Mitochondrial motility occurs through mechanical and molecular mechanisms which translocate mitochondria to sites of high energy demand. Motility also plays an important role in intracellular signaling. Here, we review key features that mediate mitochondrial dynamics and explore methods to advance the study of mitochondrial motility as well as mitochondrial dynamics-related diseases and mitochondrial-targeted therapeutics.
Collapse
Affiliation(s)
- Adam Green
- Department of Anesthesiology, The Ohio State University, Columbus, OH, United States
| | - Tanvir Hossain
- Department of Anesthesiology, The Ohio State University, Columbus, OH, United States
| | - David M. Eckmann
- Department of Anesthesiology, The Ohio State University, Columbus, OH, United States
- Center for Medical and Engineering Innovation, The Ohio State University, Columbus, OH, United States
- *Correspondence: David M. Eckmann,
| |
Collapse
|
19
|
Teng XQ, Qu J, Li GH, Zhuang HH, Qu Q. Small Interfering RNA for Gliomas Treatment: Overcoming Hurdles in Delivery. Front Cell Dev Biol 2022; 10:824299. [PMID: 35874843 PMCID: PMC9304887 DOI: 10.3389/fcell.2022.824299] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Gliomas are central nervous system tumors originating from glial cells, whose incidence and mortality rise in coming years. The current treatment of gliomas is surgery combined with chemotherapy or radiotherapy. However, developing therapeutic resistance is one of the significant challenges. Recent research suggested that small interfering RNA (siRNA) has excellent potential as a therapeutic to silence genes that are significantly involved in the manipulation of gliomas’ malignant phenotypes, including proliferation, invasion, metastasis, therapy resistance, and immune escape. However, it is challenging to deliver the naked siRNA to the action site in the cells of target tissues. Therefore, it is urgent to develop delivery strategies to transport siRNA to achieve the optimal silencing effect of the target gene. However, there is no systematic discussion about siRNAs’ clinical potential and delivery strategies in gliomas. This review mainly discusses siRNAs’ delivery strategies, especially nanotechnology-based delivery systems, as a potential glioma therapy. Moreover, we envisage the future orientation and challenges in translating these findings into clinical applications.
Collapse
Affiliation(s)
- Xin-Qi Teng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Pharmacy, The Second Xiangya Hospital, Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Jian Qu
- Department of Pharmacy, The Second Xiangya Hospital, Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Guo-Hua Li
- Department of Pharmacy, The Second Xiangya Hospital, Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Hai-Hui Zhuang
- Department of Pharmacy, The Second Xiangya Hospital, Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Qiang Qu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Qiang Qu,
| |
Collapse
|