1
|
Niu Z, Zhang Y, Wang Y, Liu D, Wang J, Shi T, Xu X, Li L. MTFR2 promotes endometrial carcinoma cell proliferation and growth via the miR-132-3p/PI3K/Akt signaling pathway. Front Med (Lausanne) 2025; 11:1505071. [PMID: 40129972 PMCID: PMC11931630 DOI: 10.3389/fmed.2024.1505071] [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: 10/02/2024] [Accepted: 11/25/2024] [Indexed: 03/26/2025] Open
Abstract
Objective Understanding the mechanisms underlying endometrial cancer progression is crucial for the development of effective targeted therapies. In this study, we investigated the role of MTFR2 in endometrial cancer cell. Methods The expression of MTFR2 in endometrial cancer was analyzed using The Cancer Genome Atlas (TCGA) dataset and detected in endometrial cancer tissues and cells, respectively. Gain-of-function and loss-of-function approaches were utilized to investigate the impact of MTFR2 on endometrial cancer cell proliferation and tumorigenesis in both in vitro and in vivo settings. Computational tools were employed to predict microRNAs (miRNAs) that potentially regulate MTFR2, and these predictions were experimentally validated. Results The expression of MTFR2 is enhanced in endometrial carcinoma, and it is positively correlated with the poor prognosis of patients. Functional studies show that MTFR2 promoted the proliferation, migration and invasion of endometrial cancer cells. Bioinformatics analysis and luciferase assays identified that MTFR2 is a potential target of miR-132-3p, and transfection with miR-132-3p mimics attenuated the MTFR2-induced activation of the PI3K/Akt pathway. Conclusion Our findings highlight the critical role of MTFR2 in promoting endometrial cancer cell proliferation and growth through the miR-132-3p/PI3K/Akt signaling pathway. Targeting this signaling axis may offer potential therapeutic strategies for endometrial cancer treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lei Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
2
|
Liu Y, Wang H, Zhang S, Peng N, Hai S, Zhao H, Liu J, Liu W. The role of mitochondrial biogenesis, mitochondrial dynamics and mitophagy in gastrointestinal tumors. Cancer Cell Int 2025; 25:46. [PMID: 39955547 PMCID: PMC11829463 DOI: 10.1186/s12935-025-03685-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 02/09/2025] [Indexed: 02/17/2025] Open
Abstract
Gastrointestinal tumors remain the leading causes of cancer-related deaths, and their morbidity and mortality remain high, which imposes a great socio-economic burden globally. Mitochondrial homeostasis depend on proper function and interaction of mitochondrial biogenesis, mitochondrial dynamics (fission and fusion) and mitophagy. Recent studies have demonstrated close implication of mitochondrial homeostasis in gastrointestinal tumorigenesis and development. In this review, we summarized the research progress on gastrointestinal tumors and mitochondrial quality control, as well as the underlying molecular mechanisms. It is anticipated that the comprehensive understanding of mitochondrial homeostasis in gastrointestinal carcinogenesis would benefit the application of mitochondria-targeted therapies for gastrointestinal tumors in future.
Collapse
Affiliation(s)
- Yihong Liu
- Department of Gastroenterology, the First Affiliated Hospital of China Medical University, 155# North Nanjing Street, Heping District, Shenyang City, 110001, Liaoning Province, China
| | - Hao Wang
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shen Zhang
- Department of Gastroenterology, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Na Peng
- Department of Gastroenterology, the First Affiliated Hospital of China Medical University, 155# North Nanjing Street, Heping District, Shenyang City, 110001, Liaoning Province, China
| | - Shuangshuang Hai
- Department of Gastroenterology, the First Affiliated Hospital of China Medical University, 155# North Nanjing Street, Heping District, Shenyang City, 110001, Liaoning Province, China
| | - Haibo Zhao
- Department of Gastroenterology, the First Affiliated Hospital of China Medical University, 155# North Nanjing Street, Heping District, Shenyang City, 110001, Liaoning Province, China
| | - Jingwei Liu
- Department of Anus and Intestine Surgery, the First Affiliated Hospital of China Medical University, 155# North Nanjing Street, Heping District, Shenyang City, 110001, Liaoning Province, China.
| | - Weixin Liu
- Department of Gastroenterology, the First Affiliated Hospital of China Medical University, 155# North Nanjing Street, Heping District, Shenyang City, 110001, Liaoning Province, China.
| |
Collapse
|
3
|
Bao Z, Yang M, Guo Y, Ge Q, Zhang H. MTFR2 accelerates hepatocellular carcinoma mediated by metabolic reprogramming via the Akt signaling pathway. Cell Signal 2024; 123:111366. [PMID: 39182591 DOI: 10.1016/j.cellsig.2024.111366] [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: 04/28/2024] [Revised: 08/13/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
Metabolic reprogramming has recently been identified as a hallmark of malignancies. The shift from oxidative phosphorylation to glycolysis in hepatocellular carcinoma (HCC) meets the demands of rapid cell growth and provides a microenvironment for tumor progression. This study sought to uncover the function and mechanism of MTFR2 in the metabolic reprogramming of HCC. Elevated MTFR2 expression was associated with poor patient prognosis. Downregulation of MTFR2 blocked malignant behaviors, epithelial-to-mesenchymal transition (EMT), and glycolysis in HCC cells. Nuclear transcription factor Y subunit gamma (NFYC) was also associated with poor patient prognosis, and NFYC bound to the promoter of MTFR2 to activate transcription and promote Akt signaling. The repressive effects of NFYC knockdown on EMT and glycolysis in HCC cells were compromised by MTFR2 overexpression, elicited through the activation of the Akt signaling. Knockdown of NFYC slowed the growth and intrahepatic metastasis in vivo, which was reversed by MTFR2 overexpression. In conclusion, our work shows that activation of MTFR2 by the transcription factor NFYC promotes Akt signaling, thereby potentiating metabolic reprogramming in HCC development. Targeting the NFYC/MTFR2/Akt axis may represent a therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Zhongming Bao
- Department of Hepatobiliary Surgery, Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huaiyin 223300, Jiangsu, PR China
| | - Ming Yang
- Department of Hepatobiliary Surgery, Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huaiyin 223300, Jiangsu, PR China
| | - Yunhu Guo
- Department of Hepatobiliary Surgery, Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huaiyin 223300, Jiangsu, PR China
| | - Qi Ge
- School of Biological Science and Food Engineering, Chuzhou University, Chuzhou 239000, Anhui, PR China.
| | - Huaguo Zhang
- Department of Hepatobiliary Surgery, Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huaiyin 223300, Jiangsu, PR China.
| |
Collapse
|
4
|
Li K, Yang B, Du Y, Ding Y, Shen S, Sun Z, Liu Y, Wang Y, Cao S, Ren W, Wang X, Li M, Zhang Y, Wu J, Zheng W, Yan W, Li L. The HOXC10/NOD1/ERK axis drives osteolytic bone metastasis of pan-KRAS-mutant lung cancer. Bone Res 2024; 12:47. [PMID: 39191757 PMCID: PMC11349752 DOI: 10.1038/s41413-024-00350-8] [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: 12/07/2023] [Revised: 06/09/2024] [Accepted: 07/12/2024] [Indexed: 08/29/2024] Open
Abstract
While KRAS mutation is the leading cause of low survival rates in lung cancer bone metastasis patients, effective treatments are still lacking. Here, we identified homeobox C10 (HOXC10) as a lynchpin in pan-KRAS-mutant lung cancer bone metastasis. Through RNA-seq approach and patient tissue studies, we demonstrated that HOXC10 expression was dramatically increased. Genetic depletion of HOXC10 preferentially impeded cell proliferation and migration in vitro. The bioluminescence imaging and micro-CT results demonstrated that inhibition of HOXC10 significantly reduced bone metastasis of KRAS-mutant lung cancer in vivo. Mechanistically, the transcription factor HOXC10 activated NOD1/ERK signaling pathway to reprogram epithelial-mesenchymal transition (EMT) and bone microenvironment by activating the NOD1 promoter. Strikingly, inhibition of HOXC10 in combination with STAT3 inhibitor was effective against KRAS-mutant lung cancer bone metastasis by triggering ferroptosis. Taken together, these findings reveal that HOXC10 effectively alleviates pan-KRAS-mutant lung cancer with bone metastasis in the NOD1/ERK axis-dependent manner, and support further development of an effective combinatorial strategy for this kind of disease.
Collapse
Affiliation(s)
- Kun Li
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Health Science Center, East China Normal University, Shanghai, 200241, China
- Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing, 401120, China
| | - Bo Yang
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yingying Du
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yi Ding
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Shihui Shen
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
- Joint Center for Translational Medicine, Shanghai Fifth People's Hospital, Fudan University and School of Life Science, East China Normal University, Shanghai, 200240, China
| | - Zhengwang Sun
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Yun Liu
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yuhan Wang
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Siyuan Cao
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Wenjie Ren
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xiangyu Wang
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Mengjuan Li
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yunpeng Zhang
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Juan Wu
- Department of Pharmacy The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Wei Zheng
- Orthopaedic Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
- Department of Orthopedics, General Hospital of Western Theater Command, Chengdu, 610000, China.
- College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China.
| | - Wangjun Yan
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Lei Li
- Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing, 401120, China.
- School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Joint Center for Translational Medicine, Shanghai Fifth People's Hospital, Fudan University and School of Life Science, East China Normal University, Shanghai, 200240, China.
| |
Collapse
|
5
|
Wu Z, Xiao C, Li F, Huang W, You F, Li X. Mitochondrial fusion-fission dynamics and its involvement in colorectal cancer. Mol Oncol 2024; 18:1058-1075. [PMID: 38158734 PMCID: PMC11076987 DOI: 10.1002/1878-0261.13578] [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: 08/15/2023] [Revised: 10/21/2023] [Accepted: 12/28/2023] [Indexed: 01/03/2024] Open
Abstract
The incidence and mortality rates of colorectal cancer have elevated its status as a significant public health concern. Recent research has elucidated the crucial role of mitochondrial fusion-fission dynamics in the initiation and progression of colorectal cancer. Elevated mitochondrial fission or fusion activity can contribute to the metabolic reprogramming of tumor cells, thereby activating oncogenic pathways that drive cell proliferation, invasion, migration, and drug resistance. Nevertheless, excessive mitochondrial fission can induce apoptosis, whereas moderate mitochondrial fusion can protect cells from oxidative stress. This imbalance in mitochondrial dynamics can exert dual roles as both promoters and inhibitors of colorectal cancer progression. This review provides an in-depth analysis of the fusion-fission dynamics and the underlying pathological mechanisms in colorectal cancer cells. Additionally, it offers partial insights into the mitochondrial kinetics in colorectal cancer-associated cells, such as immune and endothelial cells. This review is aimed at identifying key molecular events involved in colorectal cancer progression and highlighting the potential of mitochondrial dynamic proteins as emerging targets for pharmacological intervention.
Collapse
Affiliation(s)
- Zihong Wu
- Hospital of Chengdu University of Traditional Chinese MedicineChina
| | - Chong Xiao
- Hospital of Chengdu University of Traditional Chinese MedicineChina
- Oncology Teaching and Research DepartmentChengdu University of Traditional Chinese MedicineChina
| | - Fang Li
- Hospital of Chengdu University of Traditional Chinese MedicineChina
| | - Wenbo Huang
- Hospital of Chengdu University of Traditional Chinese MedicineChina
| | - Fengming You
- Hospital of Chengdu University of Traditional Chinese MedicineChina
- Institute of OncologyChengdu University of Traditional Chinese MedicineChina
| | - Xueke Li
- Hospital of Chengdu University of Traditional Chinese MedicineChina
- Oncology Teaching and Research DepartmentChengdu University of Traditional Chinese MedicineChina
| |
Collapse
|
6
|
Wei L, Feng Z, Dou Q, Tan L, Zhao X, Hao B. Dysregulation of MTFR2, ATP5IF1 and BAK1 in Sertoli cells relates to idiopathic non-obstructive azoospermia via inhibiting mitochondrial fission and inducing mitochondrial dysfunction†. Biol Reprod 2024; 110:408-418. [PMID: 37903059 DOI: 10.1093/biolre/ioad150] [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: 07/14/2023] [Revised: 09/28/2023] [Accepted: 10/24/2023] [Indexed: 11/01/2023] Open
Abstract
Non-obstructive azoospermia affects more than 10% of infertile men with over 70% patients are idiopathic with uncharacterized molecular mechanisms, which is referred as idiopathic non-obstructive azoospermia. In this study, we checked the morphology of Sertoli cell mitochondria in testis biopsies from patients with idiopathic non-obstructive azoospermia and patients with obstructive azoospermia who have normal spermiogenesis. The expression of 104 genes controlling mitochondria fission and fusion were analyzed in three gene expression datasets including a total of 60 patients with non-obstructive azoospermia. The levels of 7 candidate genes were detected in testis biopsies from 38 patients with idiopathic non-obstructive azoospermia and 24 patients with obstructive azoospermia who have normal spermatogenesis by RT-qPCR. Cell viability, apoptosis, mitochondria membrane potential, adenosine triphosphate production, oxygen consumption, and mitochondria morphology were examined in primary human Sertoli cells. Mouse spermatogonial stem cells were used to detect the cell supporting capacity of Sertoli cells. We observed that patients with idiopathic non-obstructive azoospermia had elongated mitochondria. MTFR2 and ATP5IF1 were downregulated, whereas BAK1 was upregulated in idiopathic non-obstructive azoospermia testis and Sertoli cells. Sertoli cells from patients with idiopathic non-obstructive azoospermia had reduced viability, mitochondria membrane potential, adenosine triphosphate production, oxygen consumption rate, glycolysis and increased apoptosis. Knockdown MTFR2 in Sertoli cells increased the mitochondria size. Knockdown ATP5IF1 did not change mitochondrial morphology but increased adenosine triphosphate hydrolysis. Overexpression of BAK1 reduced membrane potential and upregulated cell apoptosis. The dysregulation of all these three genes contributed to the dysfunction of Sertoli cells, which provides a clue for idiopathic non-obstructive azoospermia treatment.
Collapse
Affiliation(s)
- Lei Wei
- Reproductive Medical Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zonggang Feng
- Reproductive Medical Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qian Dou
- Reproductive Medical Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li Tan
- Reproductive Medical Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xinghua Zhao
- Department of Urology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bin Hao
- Department of Urology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
7
|
Wu Z, Xiao C, Long J, Huang W, You F, Li X. Mitochondrial dynamics and colorectal cancer biology: mechanisms and potential targets. Cell Commun Signal 2024; 22:91. [PMID: 38302953 PMCID: PMC10835948 DOI: 10.1186/s12964-024-01490-4] [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: 08/22/2023] [Accepted: 01/11/2024] [Indexed: 02/03/2024] Open
Abstract
Colorectal cancer (CRC) is a significant public health concern, and its development is associated with mitochondrial dysfunction. Mitochondria can adapt to the high metabolic demands of cancer cells owing to their plasticity and dynamic nature. The fusion-fission dynamics of mitochondria play a crucial role in signal transduction and metabolic functions of CRC cells. Enhanced mitochondrial fission promotes the metabolic reprogramming of CRC cells, leading to cell proliferation, metastasis, and chemoresistance. Excessive fission can also trigger mitochondria-mediated apoptosis. In contrast, excessive mitochondrial fusion leads to adenosine triphosphate (ATP) overproduction and abnormal tumor proliferation, whereas moderate fusion protects intestinal epithelial cells from oxidative stress-induced mitochondrial damage, thus preventing colitis-associated cancer (CAC). Therefore, an imbalance in mitochondrial dynamics can either promote or inhibit CRC progression. This review provides an overview of the mechanism underlying mitochondrial fusion-fission dynamics and their impact on CRC biology. This revealed the dual role of mitochondrial fusion-fission dynamics in CRC development and identified potential drug targets. Additionally, this study partially explored mitochondrial dynamics in immune and vascular endothelial cells in the tumor microenvironment, suggesting promising prospects for targeting key fusion/fission effector proteins against CRC.
Collapse
Affiliation(s)
- Zihong Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Chong Xiao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
- Oncology Teaching and Research Department of Chengdu, University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Jing Long
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Wenbo Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Fengming You
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
- Institute of Oncology, Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Xueke Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
- Oncology Teaching and Research Department of Chengdu, University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
8
|
Liu S, Tang S, Yang G, Li Q. Lysine Demethylase 1B Promotes Tear Secretion Disorder in Sjogren's Syndrome by Regulating the PAX6/CLU Axis. J Mol Neurosci 2023; 73:28-38. [PMID: 36542318 DOI: 10.1007/s12031-022-02094-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022]
Abstract
The impacts of lysine demethylase 1B (KDM1B) have been probed in multiple diseases, but the effects of KDM1B on SS remained obscure. The study aimed to unravel the efficiency of KDM1B on SS progression via the paired box 6 (PAX6)/clusterin (CLU) axis. NODB10. H2b mice were selected to establish the SS model. KDM1B, Pax6, and CLU expression in SS mice was assessed. Adeno-associated viruses carrying KDM1B, Pax6, and CLU were injected into the SS mice to detect tear secretion, epithelium corneal fluorescein staining scores, and levels of specific markers of lacrimal gland epithelial cells, neurotransmitter receptors that induce secretion from the lacrimal gland, and genes encoding normal tear components. The relation among KDM1B, Pax6, and CLU was examined. The rescue experiments were conducted for verifying the interaction among KDM1B, Pax6, and CLU. KDM1B expression was elevated, while Pax6 and CLU levels were decreased in the lacrimal gland tissues of SS mouse models. KDM1B decrement and Pax6 augmentation improved tear secretion, reduced corneal fluorescein staining score, decreased levels of specific markers of lacrimal gland epithelial cells, and increased levels of neurotransmitter receptors that induce secretion from the lacrimal gland and genes encoding normal tear components. KDM1D suppressed Pax6 expression by mediating H3K4me2 demethylation. Pax6 promoted the expression of CLU at the transcriptional level by binding to the CLU promoter. Silencing of Pax6 or CLU could reverse the effects of KDM1B reduction on improving the tear secretion disorder of SS mice. Silencing KDM1B mitigates the tear secretion disorder of SS mice via modulating the Pax6/CLU axis.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Ophthalmology, Beijing Jishuitan Hospital, Beijing, 100096, China.
| | - Shaohua Tang
- Department of Ophthalmology, Beijing Jishuitan Hospital, Beijing, 100096, China
| | - Guang Yang
- Department of Ophthalmology, Beijing Jishuitan Hospital, Beijing, 100096, China
| | - Qingnan Li
- Department of Ophthalmology, Beijing Jishuitan Hospital, Beijing, 100096, China
| |
Collapse
|
9
|
Lian Z, Pang P, Zhu Y, Du W, Zhou J. Prognostic Value and Potential Mechanism of MTFR2 in Lung Adenocarcinoma. Front Oncol 2022; 12:832517. [PMID: 35600359 PMCID: PMC9117628 DOI: 10.3389/fonc.2022.832517] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 04/01/2022] [Indexed: 12/24/2022] Open
Abstract
Mitochondrial fission regulator 2 (MTFR2) belongs to the MTFR1 family, which plays a crucial role in regulating oxidative phosphorylation. Recent studies indicate that it also participates in cancer carcinogenesis and development; however, the clinical significance of MTFR2 in lung adenocarcinoma has not been fully confirmed. Our current study investigated the relationships between clinical characteristics and MTFR2 expression based on The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GSE31210) dataset, and clinical histopathological sample cohort. In addition, Kaplan–Meier and Cox regression analyses were additionally performed to evaluate the association between MTFR2 expression and patient survival. Gene set enrichment analysis (GESA) was conducted to spot possible pathways associated with MTFR2. Moreover, a single-sample GESA (ssGESA) was performed to evaluate the association between MTFR2 expression and immune cell infiltration. Cell colony formation assay, CCK-8 assay, cell cycle assay, and transwell assay were performed to verify the cell proliferation, migration, and invasion abilities after interfering with MTFR2 in lung cancer cells. Western blot assay was applied to identify the underlying protein levels. The results indicated that the elevated MTFR2 expression in lung adenocarcinoma samples correlated with T stage (P < 0.001), N stage (P = 0.005), M stage (P = 0.015), pathological stage (P = 0.002), and TP53 status (P < 0.001). Patients with a higher MTFR2 expression correlated with poorer overall survival (P < 0.01) and progression-free survival (P = 0.002). Knockdown of MTFR2 inhibited cell proliferation, migration, and invasion via AKT-cyclin D1 signaling and EMT pathways. Moreover, MTFR2 expression significantly positively correlated with Th2 cells (P < 0.001). Taken together, MTFR2 could serve as a novel prognostic indicator and therapeutic target for lung adenocarcinoma.
Collapse
Affiliation(s)
- Zengzhi Lian
- Department of Pulmonary and Critical Care Medicine, Taicang Affiliated Hospital of Soochow University, Suzhou, China
| | - Pei Pang
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yan Zhu
- Department of Emergency and Critical Care Medicine, Changzheng Hospital of Second Military Medical University, Shanghai, China
| | - Wenwen Du
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jintao Zhou
- Department of Pulmonary and Critical Care Medicine, Taicang Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|