1
|
Gao F, Ma Y, Yu C, Duan Q. miR-125b-5p regulates FFA-induced hepatic steatosis in L02 cells by targeting estrogen-related receptor alpha. Gene 2025; 959:149419. [PMID: 40113187 DOI: 10.1016/j.gene.2025.149419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/02/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND & AIMS NAFLD is a global and complex liver disease caused by multiple factors. Intrahepatocellular steatosis is the primary prerequisite for the occurrence and development of NAFLD. It has been shown that miR-125b-5p is highly correlated with NAFLD, and ESRRA is a factor that regulates lipid metabolism. The purpose of our study is to investigate whether miR-125b-5p regulates FFA-induced steatosis in L02 cells by targeting ESRRA. APPROACHES AND RESULTS Estrogen-related receptor alpha (ESRRA) was identified as a direct target of miR-125b-5p through database prediction and a dual-luciferase reporter gene assay. L02 cells were induced with free fatty acids (OA:PA, 2:1) at concentrations of 0.3 mM, 0.6 mM, 0.9 mM, 1.2 mM and 1.5 mM for 24 h, 48 h and 72 h, respectively. The degree of hepatocyte steatosis and triglyceride content were separately manifested by oil red O staining and colorimetric method. Cell viability per group was detected by CCK-8 assay. Eventually, 0.9 mM and 24 h were screened out as the optimal concentration and time for establishing the in-vitro model of hepatic steatosis. Followingly, miR-125b-5p and ESRRA were knocked down by transient transfection. We monitored the expressions of lipid metabolism factors SREBP-1c, ACC1 and FAS and determine triglyceride content within the cells per group. The data showed that knockdown of ESRRA led to down-regulation of the expressions of SREBP-1, ACC1, FAS and triglyceride content. Meanwhile, knockdown of ESRRA and miR-125b-5p resulted that the expressions of ESRRA, SREBP-1, ACC1, FAS and triglyceride content rebounded. CONCLUSIONS MiR-125b-5p down-regulates the expressions of lipid metabolism-related factors by negatively regulating ESRRA, thereby improving hepatic steatosis.
Collapse
Affiliation(s)
- Fen Gao
- Gansu University of Chinese Medicine, Gansu 730000, China.
| | - Yanhua Ma
- Gansu University of Chinese Medicine, Gansu 730000, China.
| | - Chun Yu
- Gansu University of Chinese Medicine, Gansu 730000, China
| | | |
Collapse
|
2
|
Liu X, Lu J, Ni X, He Y, Wang J, Deng Z, Zhang G, Shi T, Chen W. FASN promotes lipid metabolism and progression in colorectal cancer via the SP1/PLA2G4B axis. Cell Death Discov 2025; 11:122. [PMID: 40148316 PMCID: PMC11950308 DOI: 10.1038/s41420-025-02409-9] [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: 11/09/2024] [Revised: 02/20/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Abnormal metabolic reprogramming is essential for tumorigenesis, metastasis, and the regulation of immune responses. Fatty acid synthase (FASN), a key enzyme in lipid metabolism, plays a crucial role in these processes. However, the relationship between FASN-mediated lipid reprogramming and the immune response in colorectal cancer (CRC) remains unclear. The present study demonstrated that FASN expression is elevated in CRC tissues and is significantly associated with poor prognosis. Functional experiments revealed that FASN promotes proliferation, migration, invasion, and phosphatidylcholine (PC) production in CRC cells. Additionally, in vivo experiments revealed that FASN knockdown significantly inhibits tumor growth and the spread of CRC cells to the lungs. Mechanistically, FASN, which is upregulated in CRC tissues, drives cancer cell proliferation, metastasis, and PC metabolism through the SP1/PLA2G4B axis, subsequently suppressing the antitumor response of natural killer (NK) cells in a PC-dependent manner. These findings provide new insights into lipid metabolism and the immunobiology of CRC, suggesting potential targets for the treatment and prevention of CRC. Schematic diagram showing the mechanism by which FASN promotes cancer cell proliferation, metastasis, and PC metabolism in CRC via the SP1/PLA2G4B axis, subsequently suppressing the antitumor response of NK cells in a PC-dependent manner. FFA free fatty acid, LPA lysophosphatidic acid, PA phosphatidate, DAG diglyceride, PC phosphatidylcholine, LPC lysophosphatidylcholine, CE cholesterol ester, TAG triacylglycerol.
Collapse
Affiliation(s)
- Xin Liu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Jiachun Lu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Xiangyu Ni
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Yuxin He
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Jiayu Wang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Zilin Deng
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Guangbo Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Weichang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China.
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China.
| |
Collapse
|
3
|
Mao X, Huang L, Liu X, Lin X, Wu Q, Wang X, Ren Y, Ma J, Zhang M, Lin Y, Ralser DJ, Mustea A, Chen G, Sun P. High glucose levels promote glycolysis and cholesterol synthesis via ERRα and suppress the autophagy-lysosomal pathway in endometrial cancer. Cell Death Dis 2025; 16:182. [PMID: 40097416 PMCID: PMC11914573 DOI: 10.1038/s41419-025-07499-y] [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: 09/11/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025]
Abstract
Endometrial cancer (EC) patients with Diabetes Mellitus (DM) always have a poor prognosis. Estrogen-related receptor α (ERRα) is known as the metabolic-related prognostic factor for EC. However, the mechanism linking glycolipid metabolism dysfunction mediated by ERRα to poor prognosis of EC with DM is still unclear. In vitro, high-glucose (HG) levels showed enhancement of ERRα expression, cell proliferation, and inhibition of the autophagic lysosomes and apoptosis by flow cytometry analysis, transmission electron microscopy, and CCK-8 assays. Mechanistically, lose-and-gain function assay, DNA sequencing, and CO-IP revealed HG increased ERRα expression to promote the transcription of HK2 and HMGCS1, which were the key rate-limiting enzyme of glycolysis-cholesterol synthesis and their metabolites suppressed the autophagy-lysosomal pathway in an ERRα-dependent manner. Furthermore, CO-IP and molecular dynamics simulation uncovered the protein residues (ARG 769HK2 vs. ARG 313HMGCS1) of HK2 and HMGCS1 could bind to p62 to form stable protein complexes involved in the autophagy-lysosomal pathway. In EC tissue from patients with comorbid DM, ERRα was significantly higher expressed compared to EC tissue from patients without evidence for DM (p < 0.05). The 3D EC organoid model with HG stimulation showed that the cell viability of XCT790 + carboplatin treatment was similar to that of metformin+carboplatin treatment, while the obviously bigger volume of organoids was more visible in the metformin+carboplatin group, indicating the therapy of XCT790 + carboplatin had the better inhibition of EC organoids with the same carboplatin dose. Besides insights into the interaction of HG and the autophagy-lysosomal pathway via ERRα, our present study points out the potential benefit of targeting ERRα in patients with EC with dysregulation of glucose and cholesterol metabolism.
Collapse
Affiliation(s)
- Xiaodan Mao
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Lixiang Huang
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Xianhua Liu
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
- Pathology Department, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, China
| | - Xite Lin
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Qibin Wu
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Xinrui Wang
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350013, China
| | - Yuan Ren
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Jincheng Ma
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Maotong Zhang
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Yao Lin
- Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350001, China
| | - Damian J Ralser
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Alexander Mustea
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Gang Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, Fujian, China.
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China.
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China.
| |
Collapse
|
4
|
Deng X, Liu H, Zhao W, Wu R, Chen K, Li Q, Yazlık MO, Özkan H, Ren J, Zhang J, Liu S, Mei L, Li S, Zhan J, Hu B. Expression of AMPK and PLIN2 in the regulation of lipid metabolism and oxidative stress in bitches with open cervix pyometra. BMC Vet Res 2025; 21:164. [PMID: 40082867 PMCID: PMC11905669 DOI: 10.1186/s12917-025-04622-1] [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: 12/29/2024] [Accepted: 02/24/2025] [Indexed: 03/16/2025] Open
Abstract
The pathogenesis of canine pyometra is multifactorial, involving hormonal imbalances, aberrant immune responses, and metabolic dysregulation includes lipid metabolism and oxidative stress. This study focuses on lipid metabolism and oxidative stress, revealing the key regulatory role of AMPK and PLIN2 in canine pyometra. Bitches with open cervix pyometra (n:8) and healthy bitches undergoing elective ovariohysterectomy (n:4) were enrolled to the study. In experiment one, the serum and tissue levels of Malondialdehyde (MDA) and Superoxide Dismutase (SOD) activity were assessed. Additionally, uterine histopathological analysis, AMPK and PLIN2 expressions were determined through immunohistochemistry. Furthermore, inflammation, oxidative stress, and lipid metabolism-related factors were evaluated using Western blot analysis. In experiment two, primary cell cultures were prepared from healthy uterine endometrial cells of the dogs in control group. Cultured canine endometrial epithelial cells were treated with lipopolysaccharide (LPS) along with oleic acid (OA) to induce an inflammatory response. Tissue and serum MDA and SOD levels were greater in dogs with pyometra. Accumulated lipid droplets were observed in the uterine tissue of bitches with pyometra. The phosphorylation of AMPK and the expression of PLIN2 significantly increased in the pyometra group. The expression of related lipid synthesis proteins such as ACC1, FASN, SREBP-1c, and PLIN2 was upregulated, while PPARα and PGC1α were downregulated in bitches with pyometra. In experiment two, activation of AMPK and PLIN2 not only restores the expression of PGC1α, but also effectively alleviates inflammation and oxidative stress. The role of lipid metabolism and oxidative stress in canine pyometra is elucidated, thus contributing to the pathogenesis of pyometra in dogs.
Collapse
Affiliation(s)
- Xin Deng
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, Sichuan, 611130, PR China
| | - Hui Liu
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Wei Zhao
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, Sichuan, 611130, PR China
| | - Rui Wu
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Kuo Chen
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Qing Li
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, Sichuan, 611130, PR China
| | - Murat Onur Yazlık
- Department of Obstetrics and Gynecology, Ankara University Faculty of Veterinary Medicine, Ankara, 06070, Turkey
| | - Hüseyin Özkan
- Faculty of Veterinary Medicine, Department of Genetics, Hatay Mustafa Kemal University, Hatay, 31060, Turkey
| | - Jingyuan Ren
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, Sichuan, 611130, PR China
| | - Jiacheng Zhang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Shiyi Liu
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, Sichuan, 611130, PR China
| | - Ling Mei
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, Sichuan, 611130, PR China
| | - Shangfeng Li
- Zhi Pet Animal Hospital, Chengdu, Sichuan, 611830, PR China
| | - Jiasui Zhan
- Department of Forest Mycology and Plant Pathology, Swedish University of Agricultural Sciences, Uppsala, SE-75007, Sweden
| | - Binhong Hu
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, Sichuan, 611130, PR China.
- Department of Forest Mycology and Plant Pathology, Swedish University of Agricultural Sciences, Uppsala, SE-75007, Sweden.
- Zhi Pet Animal Hospital, Chengdu, Sichuan, 611830, PR China.
| |
Collapse
|
5
|
Wang J, Hu X, Li Y, Li S, Wang T, Wang D, Gao Y, Wang Q, Zhou J, Wan C. Impaired lipid homeostasis and elevated lipid oxidation of erythrocyte membrane in adolescent depression. Redox Biol 2025; 80:103491. [PMID: 39809016 PMCID: PMC11780951 DOI: 10.1016/j.redox.2025.103491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
Adolescent depression is a globally concerned mental health issue, the pathophysiological mechanisms of which remain elusive. Membrane lipids play a crucial role in brain development and function, potentially serving as a crossroad for the abnormalities in neurotransmitters, neuroendocrine, inflammation, oxidative stress, and energy metabolism observed in depressed adolescents. The primary aim of this study was to investigate the erythrocyte membrane lipid profile in adolescent depression. A total of 2838 erythrocyte membrane lipids were detected and quantified in 81 adolescents with depression and 67 matched healthy adolescents using ultra-high performance liquid chromatography-mass spectrometry. Depressed adolescents exhibited significantly different membrane lipid characteristics compared to healthy controls. Specifically, the levels of cholesterol, sphingomyelins, and ceramides were increased, while ether lipids were decreased in patients. Moreover, the patients showed reduced polyunsaturated fatty acids and elevated lipophilic index in membrane, suggesting diminished membrane fluidity. The higher oxidized membrane lipids and plasma malondialdehyde were observed in adolescent depression, indicating the presence of oxidative stress. Importantly, membrane lipid damage was associated with more severe depressive symptoms and worse cognitive function in patients. In addition, reduced polyunsaturated fatty acids and membrane fluidity may be partly responsible for the blunted niacin skin flushing response found in depressed adolescents. In conclusion, our results reveal impaired erythrocyte membrane lipid homeostasis in adolescents with depression, which may implicate membrane dysfunction in the brain. These findings offer new insights into the underlying molecular mechanisms of adolescent depression, highlighting the potential of counteracting membrane damage as a promising avenue for future therapeutic interventions.
Collapse
Affiliation(s)
- Jinfeng Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China.
| | - Xiaowen Hu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China.
| | - Ya Li
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang, Shandong, China.
| | - Shuhui Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China.
| | - Tianqi Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China.
| | - Dandan Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China.
| | - Yan Gao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China.
| | - Qian Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China.
| | - Jiansong Zhou
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Chunling Wan
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China; Shanghai Mental Health Center, Shanghai Key Laboratory of Psychiatry Disorders, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
6
|
Jiang M, Xu L, Lin W, Liu W, Zhang Y, Wang H, Zhao L. LncRNA CRCMSL interferes in phospholipid unsaturation to suppress colorectal cancer progression via reducing membrane fluidity. J Adv Res 2025:S2090-1232(25)00075-X. [PMID: 39921055 DOI: 10.1016/j.jare.2025.02.003] [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: 04/20/2024] [Revised: 11/18/2024] [Accepted: 02/04/2025] [Indexed: 02/10/2025] Open
Abstract
INTRODUCTION Reprogrammed metabolism is an important basis of colorectal cancer (CRC) progression; however, its mechanisms remain unclear. This study illustrated a novel mechanism for long noncoding RNA (lncRNA) CRCMSL in CRC, which was identified as a CRC suppressor in our previous study. OBJECTIVE To investigate whether CRCMSL suppresses colorectal cancer by interfering in lipid metabolism. METHODS Potential functions of CRCMSL were predicted by GSEA, which led to lipidomics. Ferroptosis process in CRC were evaluated by protein markers, probe-reported lipid peroxidation signals and transmission electron microscopy. Order and fluidity of phospholipid bilayers were detected by Laurdan generalized polarization (GP) assays and fluorescence recovery after photobleaching (FRAP) assays, respectively. RNA pull-down and RIP assays were performed to explore the target of CRCMSL. qPCR, western blot and enzyme activity detections were used to explore the effects of CRCMSL on the target. Orthotopic and subcutaneous xenografts in nude mice were used to validate efficacy of CRC in vivo. RESULTS CRCMSL-knockdown upregulated lipid synthesis and remodeled fatty acyl chains in phospholipids, inspiring studies on ferroptosis and phospholipid bilayers. CRCMSL-mediated biological processes and behaviors were restored by stearoyl-CoA desaturase (SCD), a key enzyme for the synthesis of monounsaturated fatty acids (MUFAs), suggesting that CRCMSL promotes ferroptosis and reduces membrane fluidity by interfering in phospholipid unsaturation. The target of CRCMSL in fatty acid metabolism is acetyl-CoA carboxylase 1 (ACC1), a key enzyme for de novo fatty acid synthesis. CRCMSL promoted ACC1 phosphorylation to restrict its activity. Firsocostat, an ACC oral inhibitor ND630, is a potential drug for CRC treatment in combination with CRCMSL. CONCLUSION Our study illustrated a novel mechanism of CRCMSL-ACC1 axis-associated fatty acid metabolism in CRC progression, providing laboratory evidence for the development of targeted therapies for patients with advanced CRC.
Collapse
Affiliation(s)
- Muhong Jiang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Department of Pathology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China.
| | - Lijun Xu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Department of Pathology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China.
| | - Wandie Lin
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Weiwei Liu
- Department of Pathology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China.
| | - Yujie Zhang
- Department of Pathology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China.
| | - Hui Wang
- Department of Medical Oncology, Affiliated Tumour Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Liang Zhao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Department of Pathology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China.
| |
Collapse
|
7
|
Ma Y, Wu F, Yu Z, Yang L. Evaluating the association between lipidome and female reproductive diseases through comprehensive Mendelian randomization analyses. Sci Rep 2025; 15:2448. [PMID: 39828767 PMCID: PMC11743779 DOI: 10.1038/s41598-025-86794-2] [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: 07/02/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025] Open
Abstract
This study aimed to assess the causal relationship between lipidome and female reproductive diseases (FRDs) using an advanced series of Mendelian randomization (MR) methods. This study utilized genome-wide association study (GWAS) summary statistics encompassing 179 lipidomes and six prevalent FRDs, namely polycystic ovary syndrome (PCOS), endometriosis, uterine fibroid, female infertility, uterine endometrial cancer, and ovarian cancer. The two-sample MR (TSMR) approach was employed to investigate the causal relationships, with further validation using false discovery rate (FDR) and multivariable MR (MVMR) methods. Subsequently, a range of comprehensive evaluations were performed, including sensitivity analysis, mediation MR analysis, reverse MR analysis, and steiger test. Examining 179 lipidome traits as exposures and 6 FRDs as outcomes, this study identified significant causal effects of 56 lipids on FRDs. Following multiple testing correction and MVMR validation, sphingomyelin (d38:2) was found to have a protective effect against PCOS (β = -0.104, 95% CI: -0.199 ~ -0.010, P = 0.031). Phosphatidylcholine (18:0_22:6) was associated with a decreased risk of developing uterine fibroid (β = -0.111, 95% CI: -0.201~ -0.021, P = 0.016), and sterol ester (27:1/20:3) showed significance in uterine endometrial cancer (β = -0.248, 95% CI: -0.443 ~ -0.053, P = 0.013). Conversely, phosphatidylethanolamine (18:2_0:0) was associated with increased risk of endometriosis (β = 0.183, 95% CI: 0.015 ~ 0.350, P = 0.033), while sterol ester (27:1/18:1) posed a risk influence on uterine fibroid (β = 1.007, 95% CI: 0.925 ~ 1.089, P < 0.001), and phosphatidylcholine (16:0_22:6) on uterine endometrial cancer (β = 0.229, 95% CI: 0.039 ~ 0.420, P = 0.018). Furthermore, it was determined that the causal associations between these lipidome profiles and FRDs were independent of BMI, obesity, diabetes, smoking, alcohol use, physical activity, inflammation, depression, waist-hip ratio, vitamin D, dehydroepiandrosterone sulphate, sex hormone binding globulin, and testosterone levels. Most outcomes passed consistent tests without evidence of heterogeneity, pleiotropy, or reverse causality. The results indicated a close association between specific lipidomes, particularly sphingomyelin, lysophosphatidylethanolamine, cholesterol ester, and phosphatidylcholines, with FRDs. These lipid species may potentially serve as biomarkers and future drug targets for the treatment of FRDs.
Collapse
Affiliation(s)
- Ye Ma
- Department of Anesthesiology, The Sixth Medical Center of PLA General Hospital, Beijing, 100048, China
| | - Fang Wu
- Senior Department of Otolaryngology-Head & Neck Surgery, The Sixth Medical Center of PLA General Hospital, Beijing, 100048, China
| | - Zeming Yu
- Department of Anesthesiology, The Sixth Medical Center of PLA General Hospital, Beijing, 100048, China
| | - Lu Yang
- Department of Anesthesiology, The Sixth Medical Center of PLA General Hospital, Beijing, 100048, China.
| |
Collapse
|
8
|
Lou Y, Jiang F, Guan J. The effect of lipidomes on the risk of endometrioid endometrial cancer: a Mendelian randomization study. Front Oncol 2024; 14:1436955. [PMID: 39493450 PMCID: PMC11527595 DOI: 10.3389/fonc.2024.1436955] [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: 05/22/2024] [Accepted: 10/01/2024] [Indexed: 11/05/2024] Open
Abstract
Objective This study aimed to explore the potential effects between various human plasma lipidomes and endometrioid endometrial cancer (EEC) by using Mendelian randomization (MR) methods. Methods This study designated a total of 179 human plasma lipidomes from the genome-wide association study (GWAS) database as the exposure variable. An EEC-related dataset from the GWAS (GCST006465) served as the outcome variable. MR analyses used the inverse variance-weighted method (IVW), MR-Egger, weighted median, simple mode, and weighted mode methods for regression calculations, accounting for possible biases induced by linkage disequilibrium and weak instrument variables. Any lipidomes failing to pass heterogeneity and horizontal pleiotropy tests were deemed to lack significant causal impact on the outcome. Results The results of IVW analysis disclosed that a variety of human plasma lipidomes (n = 15) exhibited a significant causal effect on EEC (p < 0.05). A subset of these lipidomes (n = 13) passed heterogeneity and horizontal pleiotropy tests, which demonstrated consistent and viable causal effects (p < 0.05) including glycerophospholipids, glycerolipids, and sterols. Specifically, phosphatidylcholine (odds ratio [OR]: 1.065-1.129, p < 0.05) exhibited a significant positive causal effect on the occurrence of EEC. Conversely, sterol ester (OR = 0.936, p = 0.007), diacylglycerol (OR = 0.914, p = 0.036), phosphatidylcholine (OR: 0.903-0.927, p < 0.05), phosphatidylethanolamine (OR = 0.907, p = 0.046) and triacylglycerol (OR: 0.880-0.924, p < 0.05) showed a notable negative causal association with EEC, suggesting their inhibitory effects on the EEC occurrence. Conclusions The study revealed that human plasma lipidomes have complex impacts on EEC through Mendelian randomization. This indicated that the diversity of structural changes in lipidomes could show different effects on subtypes and then affect EEC occurrence. Although these lipids had the potential to be promising biomarkers, they needed to be further clinically validated nevertheless.
Collapse
Affiliation(s)
- Yaochen Lou
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Feng Jiang
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Jun Guan
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
9
|
Al Mamun A, Geng P, Wang S, Shao C. Role of Pyroptosis in Endometrial Cancer and Its Therapeutic Regulation. J Inflamm Res 2024; 17:7037-7056. [PMID: 39377044 PMCID: PMC11457779 DOI: 10.2147/jir.s486878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 09/21/2024] [Indexed: 10/09/2024] Open
Abstract
Pyroptosis is an inflammatory cell death induced by inflammasomes that release several pro-inflammatory mediators such as interleukin-18 (IL-18) and interleukin-1β (IL-1β). Pyroptosis, a type of programmed cell death, has recently received increased interest both as a therapeutic and immunological mechanism. Numerous studies have provided substantial evidence supporting the involvement of inflammasomes and pyroptosis in a variety of pathological conditions including cancers, nerve damage, inflammatory diseases and metabolic conditions. Researchers have demonstrated that dysregulation of pyroptosis and inflammasomes contribute to the progression of endometriosis and gynecological malignancies. Current research also indicates that inflammasome and pyroptosis-dependent signaling pathways may further induce the progression of endometrial cancer (EC). More specifically, dysregulation of NLR family pyrin domain 3 (NLRP3) and caspase-1-dependent pyroptosis play a contributory role in the pathogenesis and development of EC. Therefore, pyroptosis-regulated protein gasdermin D (GSDMD) may be an independent prognostic biomarker for the detection of EC. This review presents the molecular mechanisms of pyroptosis-dependent signaling pathways and their contributory role and function in advancing EC. Moreover, this review offers new insights into potential future applications and innovative approaches in utilizing pyroptosis to develop effective anti-cancer therapies.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
| | - Peiwu Geng
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| | - Shuanghu Wang
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| | - Chuxiao Shao
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| |
Collapse
|
10
|
Losby M, Hayes M, Valfort A, Sopariwala DH, Sanders R, Walker JK, Xu W, Narkar VA, Zhang L, Billon C, Burris TP. The Estrogen Receptor-Related Orphan Receptors Regulate Autophagy through TFEB. Mol Pharmacol 2024; 106:164-172. [PMID: 39168657 PMCID: PMC11413914 DOI: 10.1124/molpharm.124.000889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024] Open
Abstract
Autophagy is an essential self-degradative and recycling mechanism that maintains cellular homeostasis. Estrogen receptor-related orphan receptors (ERRs) are fundamental in regulating cardiac metabolism and function. Previously, we showed that ERR agonists improve cardiac function in models of heart failure and induce autophagy. Here, we characterized a mechanism by which ERRs induce the autophagy pathway in cardiomyocytes. Transcription factor EB (TFEB) is a master regulator of the autophagy-lysosome pathway and has been shown to be crucial regulator of genes that control autophagy. We discovered that TFEB is a direct ERR target gene whose expression is induced by ERR agonists. Activation of ERR results in increased TFEB expression in both neonatal rat ventricular myocytes and C2C12 myoblasts. An ERR-dependent increase in TFEB expression results in increased expression of an array of TFEB target genes, which are critical for the stimulation of autophagy. Pharmacologically targeting ERR is a promising potential method for the treatment of many diseases where stimulation of autophagy may be therapeutic, including heart failure. SIGNIFICANCE STATEMENT: Estrogen receptor-related receptor agonists function as exercise mimetics and also display efficacy in animal models of metabolic disease, obesity, and heart failure.
Collapse
Affiliation(s)
- McKenna Losby
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| | - Matthew Hayes
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| | - Aurore Valfort
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| | - Danesh H Sopariwala
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| | - Ryan Sanders
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| | - John K Walker
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| | - Weiyi Xu
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| | - Vihang A Narkar
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| | - Lilei Zhang
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| | - Cyrielle Billon
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| | - Thomas P Burris
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis (M.L.); Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida (M.H., A.V., R.S., T.P.B.); University of Florida Genetics Institute, Gainesville, Florida (T.P.B.); Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, (D.H.S., V.A.N.); Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri (J.K.W.); Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX (W.X., L.Z.); and Center for Clinical Pharmacology, St Louis College of Pharmacy, University of Health Sciences and Pharmacy, St. Louis MO (C.B.)
| |
Collapse
|
11
|
Tong X, Cui Y. Mendelian randomization analysis of the causal relationship between serum metabolites and thoracic aortic aneurysm. Medicine (Baltimore) 2024; 103:e39686. [PMID: 39287234 PMCID: PMC11404878 DOI: 10.1097/md.0000000000039686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/15/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024] Open
Abstract
Thoracic aortic aneurysm (TAA) is associated with changes in the levels of metabolites; however, the exact causal relationships remain unclear. Identifying this complex relationship may provide new insights into the pathogenesis of TAA. We used genome-wide association studies to investigate the relationship between metabolites and TAA in this study. A total of 1400 serum metabolites were investigated for their potential causal effects on the risk of TAA. We performed bidirectional and 2-sample Mendelian randomization (MR) analysis using 5 MR tests: MR-Egger, weighted mode, weighted median, inverse variance weighted (IVW), and simple mode. We also performed sensitivity analysis to verify our findings, including heterogeneity analysis using IVW and MR-Egger tests and pleiotropy analysis using the MR-Egger test. Multiple metabolites were identified as having a causal effect on the risk of TAA, particularly those related to lipid metabolites; the top 2 risk factors identified using the IVW test were 3-carboxy-4-methyl-5-pentyl-2-furanpropionate (P = .019) and 5alpha-androstan-3alpha,17alpha-diol (P = .021), whereas the 2 top protective factors were 1-stearoyl-2-docosahexaenoyl-gpc (P = .023) and 1-oleoyl-2-docosahexaenoyl-GPC (P = .005). Sensitivity analysis verified the lack of heterogeneity (P = .499, .584, .232, and .624, respectively; IVW test) or pleiotropy (P = .621, .483, .598, and .916, respectively; Egger test). Our study provides new evidence of a causal relationship between metabolites and the risk of TAA, thus providing new insights into the pathogenesis of this disease. These findings suggest a promising approach for metabolite-based therapeutic interventions.
Collapse
Affiliation(s)
- Xiaoshan Tong
- Department of Cardiac Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Yu Cui
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
12
|
Lin X, Chen T, Wang L, Ren Y, Lin W, Mao X, Sun P. A metabolic-inflammatory-nutritional score (MINS) is associated with lymph node metastasis and prognostic stratification for endometrial cancer patients. Int J Med Sci 2024; 21:2379-2389. [PMID: 39310257 PMCID: PMC11413899 DOI: 10.7150/ijms.96179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/16/2024] [Indexed: 09/25/2024] Open
Abstract
Objective: This study aims to propose a personalized cancer prediction model based on the metabolic-inflammatory-nutritional score (MINS) for predicting lymph node metastasis (LNM) in endometrial cancer (EC) and validated prediction of survival probability in patients with a family history of Lynch syndrome-associated cancers (LSAC). Methods: A total of 676 patients diagnosed with EC were enrolled in this study. We calculated the optimal cutoff value using restricted cubic splines (RCS) analysis or the mean value. Our feature selection process for constructing the MINS involved using the LASSO regression model. MINS were evaluated for LNM using logistic regression analysis. To assess the prognostic value of the MINS, we generated survival curves using the Kaplan-Meier method with a log-rank test. Furthermore, we constructed a nomogram to validate the prognostic significance of the MINS. The predictive accuracy of nomogram was evaluated using the concordance index (C-index) and calibration plot. Results: LNM risk was associated with family history of LSAC and MINS group (all adjusted p<0.05). Patients in the high-risk MINS group or patients with a family history of LSAC exhibited poorer overall survival (p=0.038, p=0.001, respectively). Additionally, a nomogram was demonstrated effective predictive performance with a C-index of 0.778 (95% CI: 0.725-0.832). Conclusion: Preoperative MINS has been determined to be associated with the risk of LNM in EC patients. Utilizing MINS as a basis, the development of a prognostic nomogram holds promise as an effective tool for risk stratification in clinical settings among EC patients with a family history of LSAC.
Collapse
Affiliation(s)
- Xite Lin
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, Fuzhou 350001, China
| | - Tianai Chen
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, Fuzhou 350001, China
| | - Liang Wang
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, Fuzhou 350001, China
- Department of Obstetrics and Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China
| | - Yuan Ren
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, Fuzhou 350001, China
| | - Wenyu Lin
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, Fuzhou 350001, China
| | - Xiaodan Mao
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, Fuzhou 350001, China
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, Fuzhou 350001, China
- Department of Obstetrics and Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China
| |
Collapse
|
13
|
Yang Z, Chen J, Wen M, Lei J, Zeng M, Li S, Long Y, Zhou Z, Wang C. Genetic association of lipids and lipid-lowering drug target genes with Endometrial carcinoma: a drug target Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1446457. [PMID: 39193372 PMCID: PMC11347345 DOI: 10.3389/fendo.2024.1446457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Aberrant lipid metabolism is intricately linked to the development of endometrial cancer, and statin lipid-lowering medications are regarded as promising adjunctive therapies for future management of this malignancy. This study employed Mendelian randomization (MR) to explore the causal association between lipid traits and endometrial cancer while assessing the potential impact of drug targets on lower lipids on endometrial cancer. METHOD Two-sample Mendelian randomization was employed to probe the causal association between lipid traits and endometrial carcinoma. Drug-target Mendelian randomization was also utilized to identify potential drug-target genes for managing endometrial carcinoma. In instances where lipid-mediated effects through particular drug targets were notable, the impacts of these drug targets on endometrial carcinoma risk factors were investigated to bolster the findings. RESULT No causal association between genetically predicted lipid traits (LDL-C, TG, TC, and HDL-C) and EC was found in two-sample Mendelian randomization. In drug target Mendelian randomization, genetic modeling of apolipoprotein B (APOB) (OR [95%CI]=0.31, [0.16-0.60]; p=4.73e-04) and cholesteryl ester transfer protein (CETP) (OR [95%CI]=1.83, [1.38-2.43]; p=2.91e-05) genetic mimicry was associated with non-endometrioid carcinoma. CONCLUSION The results of our MR study revealed no causal association between genetically predicted lipid traits (LDL-C, TG, TC, and HDL-C) and EC. Among the six lipid-lowering drug targets, we observed a significant association between lower predicted APOB levels and higher CETP levels with an increased risk of endometrioid carcinoma. These findings provide novel insights into the importance of lipid regulation in individuals with endometrial carcinoma, warranting further clinical validation and mechanistic investigations.
Collapse
Affiliation(s)
- Zhehan Yang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Junpan Chen
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Minghao Wen
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Jiayuan Lei
- The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, China
| | - Ming Zeng
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Sichen Li
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yao Long
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiyi Zhou
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Chunyan Wang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
14
|
Passaponti S, Manzan Martins C, Cresti L, Romagnoli R, Paulesu L, Ietta F, Ermini L. Lysosome dynamics during human endometrial stromal cells decidualization: effect of para-nonylphenol. Am J Physiol Cell Physiol 2024; 327:C113-C121. [PMID: 38738312 DOI: 10.1152/ajpcell.00604.2023] [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: 11/08/2023] [Revised: 04/11/2024] [Accepted: 04/28/2024] [Indexed: 05/14/2024]
Abstract
During the process of decidualization, the stromal cells of the endometrium change dynamically to create a favorable environment for embryo implantation. Lysosome activity has often been associated with physiological changes in the endometrium during the preimplantation period and early pregnancy. In this study, the effect of para-nonylphenol (p-NP), an endocrine disruptor, on human immortalized endometrial stromal cells (tHESCs) was investigated. After exposure to p-NP (1 nM and 1 pM), the cells were examined for the decidualization markers connexin-43, insulin like growth factor binding protein 1 (IGFBP1), and prolactin. In addition, the effect of p-NP on lysosome biogenesis and exocytosis was investigated by examining the expression and localization of the transcription factor EB (TFEB) and that of the lysosomal-associated membrane protein 1 (LAMP-1). Finally, we evaluated the effect of p-NP on extracellular matrix (ECM) remodeling using a fibronectin assay. Our results showed that p-NP reduced the expression of prolactin protein, increased the nuclear localization of TFEB, and induced the increase and translocation of the lysosomal protein LAMP-1 to the membrane of tHESCs. The data indicate an impairment of decidualization and suggest an increase in lysosomal biogenesis and exocytosis, which is supported by the higher release of active cathepsin D by tHESCs. Given the importance of cathepsins in the processing and degradation of the ECM during trophoblast invasiveness and migration into the decidua, our results appear to be clear evidence of the negative effects of p-NP on endometrial processes that are fundamental to reproductive success and the establishment of pregnancy.NEW & NOTEWORTHY Endocrine disruptors, such as para-nonylphenol, affect the decidualization of human endometrial stromal cells with an impact on decidualization itself, lysosome biogenesis and exocytosis, and extracellular matrix remodeling. All these alterations may negatively impact embryo implantation with the success of reproduction and the establishment of pregnancy.
Collapse
Affiliation(s)
- Sofia Passaponti
- Department of Life Sciences, University of Siena, Siena, Italy
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | | | - Laura Cresti
- Department of Life Sciences, University of Siena, Siena, Italy
| | | | - Luana Paulesu
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Francesca Ietta
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Leonardo Ermini
- Department of Life Sciences, University of Siena, Siena, Italy
| |
Collapse
|
15
|
Zhao Z, Wang J, Kong W, Newton MA, Burkett WC, Sun W, Buckingham L, O’Donnell J, Suo H, Deng B, Shen X, Zhang X, Hao T, Zhou C, Bae-Jump VL. Palmitic Acid Exerts Anti-Tumorigenic Activities by Modulating Cellular Stress and Lipid Droplet Formation in Endometrial Cancer. Biomolecules 2024; 14:601. [PMID: 38786008 PMCID: PMC11117634 DOI: 10.3390/biom14050601] [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: 02/26/2024] [Revised: 04/24/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
Epidemiological and clinical evidence have extensively documented the role of obesity in the development of endometrial cancer. However, the effect of fatty acids on cell growth in endometrial cancer has not been widely studied. Here, we reported that palmitic acid significantly inhibited cell proliferation of endometrial cancer cells and primary cultures of endometrial cancer and reduced tumor growth in a transgenic mouse model of endometrial cancer, in parallel with increased cellular stress and apoptosis and decreased cellular adhesion and invasion. Inhibition of cellular stress by N-acetyl-L-cysteine effectively reversed the effects of palmitic acid on cell proliferation, apoptosis, and invasive capacity in endometrial cancer cells. Palmitic acid increased the intracellular formation of lipid droplets in a time- and dose-dependent manner. Depletion of lipid droplets by blocking DGAT1 and DGAT2 effectively increased the ability of palmitic acid to inhibit cell proliferation and induce cleaved caspase 3 activity. Collectively, this study provides new insight into the effect of palmitic acid on cell proliferation and invasion and the formation of lipid droplets that may have potential clinical relevance in the treatment of obesity-driven endometrial cancer.
Collapse
Affiliation(s)
- Ziyi Zhao
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100026, China; (Z.Z.); (J.W.); (W.K.); (H.S.); (B.D.); (X.Z.)
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.A.N.); (W.C.B.); (W.S.); (L.B.); (T.H.)
| | - Jiandong Wang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100026, China; (Z.Z.); (J.W.); (W.K.); (H.S.); (B.D.); (X.Z.)
| | - Weimin Kong
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100026, China; (Z.Z.); (J.W.); (W.K.); (H.S.); (B.D.); (X.Z.)
| | - Meredith A. Newton
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.A.N.); (W.C.B.); (W.S.); (L.B.); (T.H.)
| | - Wesley C. Burkett
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.A.N.); (W.C.B.); (W.S.); (L.B.); (T.H.)
| | - Wenchuan Sun
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.A.N.); (W.C.B.); (W.S.); (L.B.); (T.H.)
| | - Lindsey Buckingham
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.A.N.); (W.C.B.); (W.S.); (L.B.); (T.H.)
| | - Jillian O’Donnell
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.A.N.); (W.C.B.); (W.S.); (L.B.); (T.H.)
| | - Hongyan Suo
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100026, China; (Z.Z.); (J.W.); (W.K.); (H.S.); (B.D.); (X.Z.)
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.A.N.); (W.C.B.); (W.S.); (L.B.); (T.H.)
| | - Boer Deng
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100026, China; (Z.Z.); (J.W.); (W.K.); (H.S.); (B.D.); (X.Z.)
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.A.N.); (W.C.B.); (W.S.); (L.B.); (T.H.)
| | - Xiaochang Shen
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100026, China; (Z.Z.); (J.W.); (W.K.); (H.S.); (B.D.); (X.Z.)
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.A.N.); (W.C.B.); (W.S.); (L.B.); (T.H.)
| | - Xin Zhang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100026, China; (Z.Z.); (J.W.); (W.K.); (H.S.); (B.D.); (X.Z.)
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.A.N.); (W.C.B.); (W.S.); (L.B.); (T.H.)
| | - Tianran Hao
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.A.N.); (W.C.B.); (W.S.); (L.B.); (T.H.)
| | - Chunxiao Zhou
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.A.N.); (W.C.B.); (W.S.); (L.B.); (T.H.)
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Victoria L. Bae-Jump
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.A.N.); (W.C.B.); (W.S.); (L.B.); (T.H.)
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
16
|
Zhuang W, Liu C, Hong Y, Zheng Y, Huang M, Tang H, Zhao L, Huang Z, Tu M, Yu L, Chen J, Zhang Y, Chen X, Lin F, Gao Q, Yu C, Huang Y. Tumor-suppressive miR-4732-3p is sorted into fucosylated exosome by hnRNPK to avoid the inhibition of lung cancer progression. J Exp Clin Cancer Res 2024; 43:123. [PMID: 38654325 PMCID: PMC11036635 DOI: 10.1186/s13046-024-03048-1] [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: 12/19/2023] [Accepted: 04/16/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Aberrant fucosylation observed in cancer cells contributes to an augmented release of fucosylated exosomes into the bloodstream, where miRNAs including miR-4732-3p hold promise as potential tumor biomarkers in our pilot study. However, the mechanisms underlying the sorting of miR-4732-3p into fucosylated exosomes during lung cancer progression remain poorly understood. METHODS A fucose-captured strategy based on lentil lectin-magnetic beads was utilized to isolate fucosylated exosomes and evaluate the efficiency for capturing tumor-derived exosomes using nanoparticle tracking analysis (NTA). Fluorescence in situ hybridization (FISH) and qRT-PCR were performed to determine the levels of miR-4732-3p in non-small cell lung cancer (NSCLC) tissue samples. A co-culture system was established to assess the release of miRNA via exosomes from NSCLC cells. RNA immunoprecipitation (RIP) and miRNA pull-down were applied to validate the interaction between miR-4732-3p and heterogeneous nuclear ribonucleoprotein K (hnRNPK) protein. Cell functional assays, cell derived xenograft, dual-luciferase reporter experiments, and western blot were applied to examine the effects of miR-4732-3p on MFSD12 and its downstream signaling pathways, and the impact of hnRNPK in NSCLC. RESULTS We enriched exosomes derived from NSCLC cells using the fucose-captured strategy and detected a significant upregulation of miR-4732-3p in fucosylated exosomes present in the serum, while its expression declined in NSCLC tissues. miR-4732-3p functioned as a tumor suppressor in NSCLC by targeting 3'UTR of MFSD12, thereby inhibiting AKT/p21 signaling pathway to induce cell cycle arrest in G2/M phase. NSCLC cells preferentially released miR-4732-3p via exosomes instead of retaining them intracellularly, which was facilitated by the interaction of miR-4732-3p with hnRNPK protein for selective sorting into fucosylated exosomes. Moreover, knockdown of hnRNPK suppressed NSCLC cell proliferation, with the elevated levels of miR-4732-3p in NSCLC tissues but the decreased expression in serum fucosylated exosomes. CONCLUSIONS NSCLC cells escape suppressive effects of miR-4732-3p through hnRNPK-mediated sorting of them into fucosylated exosomes, thus supporting cell malignant properties and promoting NSCLC progression. Our study provides a promising biomarker for NSCLC and opens a novel avenue for NSCLC therapy by targeting hnRNPK to prevent the "exosome escape" of tumor-suppressive miR-4732-3p from NSCLC cells.
Collapse
Affiliation(s)
- Wanzhen Zhuang
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Chengxiu Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, 361102, China
- Institute of Future Technology, Beijing Hotgen Biotech Co., Ltd, Beijing, 102600, China
| | - Yilin Hong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Yue Zheng
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Minjian Huang
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, 350001, China
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Haijun Tang
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, 350001, China
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Lilan Zhao
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, 350001, China
- Department of Thoracic Surgery, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Zhixin Huang
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
- Integrated Chinese and Western Medicine College, Fujian University of Traditional Chinese Medicine, Fuzhou, 350108, China
| | - Mingshu Tu
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Lili Yu
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Jianlin Chen
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Yi Zhang
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Xiongfeng Chen
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, 350001, China
- Department of Scientific Research, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Fan Lin
- Department of Geriatric Medicine, Fujian Provincial Hospital, Fuzhou, 350001, China
- Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Qi Gao
- Institute of Future Technology, Beijing Hotgen Biotech Co., Ltd, Beijing, 102600, China
| | - Chundong Yu
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, 350001, China.
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, 361102, China.
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou, 350001, China.
| | - Yi Huang
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, 350001, China.
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China.
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou, 350001, China.
- Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Fuzhou, 350001, China.
- Central Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China.
- Fujian Provincial Key Laboratory of Critical Care Medicine, Fujian Provincial Key Laboratory of Cardiovascular Disease, Fuzhou, 350001, China.
| |
Collapse
|
17
|
Benabdelkamel H, Jaber MA, Akkour K, AlMalki RH, Alfadda AA, Masood A, Joy SS, Alhalal H, Alwehaibi MA, Arafah M, Alshehri E, Abdel Rahman AM. Metabolomic Profiling of Blood Plasma in Females with Hyperplasia and Endometrial Cancer. Metabolites 2024; 14:109. [PMID: 38393001 PMCID: PMC10890097 DOI: 10.3390/metabo14020109] [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: 12/26/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Uterine cancer is the most prevalent gynecologic malignancy in women worldwide. Endometrial cancer (EC) has an 81% five-year survival rate, depending on disease stage and time of diagnosis. While endometrial cancer is largely treatable when detected early, no established screening techniques are available in clinical practice. As a result, one of the most significant issues in the medical field is the development of novel ways for early cancer identification, which could boost treatment success rates. Liquid chromatography-high-resolution mass spectrometry (LC-HRMS)-based metabolomics was employed to explore the metabolomic markers and pathways unique to this cancer type and link them to the benign endometrial hyperplasia that may progress to cancer in 5% to 25% of patients. The study involved 59 postmenopausal participants, 20 with EC type 1, 20 with benign hyperplasia, and 19 healthy participants. Metabolite distribution changes were analyzed, and 338 of these features were dysregulated and significant. The first two main components, PC1 and PC2, were responsible for 11.5% and 12.2% of the total metabolites, respectively. Compared with the control group (CO), EC samples had 203 differentially expressed metabolites (180 upregulated and 23 downregulated); in hyperplasia (HP), 157 metabolites were dysregulated (127 upregulated and 30 downregulated) compared to the CO group while 21 metabolites exhibited differential regulation (16 upregulated and 5 downregulated) in EC plasma samples compared to the HP group. Hyperplasia samples exhibited similar metabolic changes to those reported in cancer, except for alterations in triglyceride levels, 7a,12 b-dihydroxy-5b-Cholan-24-oic acid, and Hept-2-enedioyl carnitine levels. The metabolites N-heptanoyl glycine and -(Methylthio)-2,3-isopentyl phosphate and formimino glutamic acid can be specific markers for hyperplasia conditions and dimethyl phosphatidyl ethanolamine and 8-isoprostaglandin E2 can be specific markers for EC conditions. Metabolic activities rely on mitochondrial oxidative phosphorylation for energy generation. The changes in metabolites identified in our study indicate that endometrial cancer cells adopt alternative strategies to increase energy production to meet the energy demand, thereby supporting proliferation.
Collapse
Affiliation(s)
- Hicham Benabdelkamel
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Malak A Jaber
- Pharmaceutical Medicinal Chemistry & Pharmacognosy, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman 1196, Jordan
| | - Khalid Akkour
- Obstetrics and Gynecology Department, College of Medicine, King Saud University Medical City, King Saud University, Riyadh 11461, Saudi Arabia
| | - Reem H AlMalki
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11461, Saudi Arabia
| | - Assim A Alfadda
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
- Department of Medicine, College of Medicine, King Saud University Medical City, King Saud University, Riyadh 11461, Saudi Arabia
| | - Afshan Masood
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Salini Scaria Joy
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Hani Alhalal
- Obstetrics and Gynecology Department, College of Medicine, King Saud University Medical City, King Saud University, Riyadh 11461, Saudi Arabia
| | - Moudi A Alwehaibi
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Maria Arafah
- Department of Pathology, College of Medicine, King Saud University Medical City, King Saud University, Riyadh 11461, Saudi Arabia
| | - Eman Alshehri
- Obstetrics and Gynecology Department, College of Medicine, King Saud University Medical City, King Saud University, Riyadh 11461, Saudi Arabia
| | - Anas M Abdel Rahman
- Metabolomics Section, Department of Clinical Genomics, Center for Genome Medicine, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh 11211, Saudi Arabia
| |
Collapse
|
18
|
Vanacker JM, Forcet C. ERRα: unraveling its role as a key player in cell migration. Oncogene 2024; 43:379-387. [PMID: 38129506 DOI: 10.1038/s41388-023-02899-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/31/2023] [Accepted: 11/14/2023] [Indexed: 12/23/2023]
Abstract
Cell migration is essential throughout the life of multicellular organisms, and largely depends on the spatial and temporal regulation of cytoskeletal dynamics, cell adhesion and signal transduction. Interestingly, Estrogen-related receptor alpha (ERRα) has been identified as a major regulator of cell migration in both physiological and pathological conditions. ERRα is an orphan member of the nuclear hormone receptor superfamily of transcription factors and displays many biological functions. ERRα is a global regulator of energy metabolism, and it is also highly involved in bone homeostasis, development, differentiation, immunity and cancer progression. Importantly, in some instances, the regulation of these biological processes relies on the ability to orchestrate cell movements. Therefore, this review describes how ERRα-mediated cell migration contributes not only to tissue homeostasis but also to tumorigenesis and metastasis, and highlights the molecular and cellular mechanisms by which ERRα finely controls the cell migratory potential.
Collapse
Affiliation(s)
- Jean-Marc Vanacker
- Centre de Recherche en Cancérologie de Lyon, CNRS UMR5286, Inserm U1052, Université de Lyon, Lyon, France
| | - Christelle Forcet
- Institut de Génomique Fonctionnelle de Lyon, UMR5242, Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique, Université Claude Bernard-Lyon 1, Lyon, France.
| |
Collapse
|
19
|
Ma J, Liu D, Mao X, Huang L, Ren Y, Xu X, Huang X, Deng C, Shi F, Sun P. Enhanced Diagnostic Efficiency of Endometrial Carcinogenesis and Progression in Women with Abnormal Uterine Bleeding through Peripheral Blood Cytokine Testing: A Multicenter Retrospective Cohort Study. Int J Med Sci 2024; 21:601-611. [PMID: 38464838 PMCID: PMC10920852 DOI: 10.7150/ijms.91506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/09/2024] [Indexed: 03/12/2024] Open
Abstract
Objective: This study aimed to evaluate the role of plasma cytokine detection in endometrial cancer screening and tumor progression assessment in patients with abnormal uterine bleeding. Methods: In this multicenter retrospective cohort study of 287 patients with abnormal uterine bleeding, comprehensive clinical information and laboratory assessments, including cytokines, routine blood tests, and tumor markers, were performed. Associations between the clinical indicators and endometrial carcinogenesis/progression were evaluated. The independent risk factors for endometrial cancer and endometrial cancer with deep myometrial invasion were analyzed using multivariate binary logistic regression. Additionally, a diagnostic model was used to evaluate the predictive efficacy of these identified risk factors. Results: In patients with abnormal uterine bleeding, low IL-4 and high IL-8 levels were independent risk factors for endometrial cancer (p < 0.05). Combining IL-4, IL-8, CA125, and menopausal status improved the accuracy of assessing endometrial cancer risk. The area under curve of the model is 0.816. High IL-6 and IL-8 levels were independent risk factors for deep myometrial invasion in patients with endometrial cancer (p < 0.05). Similarly, combining IL-6, IL-8, and Monocyte counts enhanced the accuracy of assessing endometrial cancer risk with deep myometrial invasion. The area under curve of the model is 0.753. Conclusions: Cytokines such as IL-4, IL-8, and IL-6 can serve as markers for monitoring endometrial cancer and its progression in women with abnormal uterine bleeding.
Collapse
Affiliation(s)
- Jincheng Ma
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
| | - Dabin Liu
- Department of Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Xiaodan Mao
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
| | - Leyi Huang
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
| | - Yuan Ren
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
| | - Xiaozhen Xu
- Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, Fujian, China
| | - Xiaoli Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, Fujian, China
- Department of Obstetrics and Gynecology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, Fujian, China
| | - Caiping Deng
- The Second Hospital of Nanping City, Nanping 354200, Fujian, China
| | - Feifeng Shi
- Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, Fujian, China
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
- Department of Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China
| |
Collapse
|
20
|
Li Z, Zhang W, Guo S, Qi G, Huang J, Gao J, Zhao J, Kang L, Li Q. A Review of Advances in Mitochondrial Research in Cancer. Cancer Control 2024; 31:10732748241299072. [PMID: 39487853 PMCID: PMC11531673 DOI: 10.1177/10732748241299072] [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: 07/19/2024] [Revised: 09/28/2024] [Accepted: 10/23/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Abnormalities in mitochondrial structure or function are closely related to the development of malignant tumors. Mitochondrial metabolic reprogramming provides precursor substances and energy for the vital activities of tumor cells, so that cancer cells can rapidly adapt to the unfavorable environment of hypoxia and nutrient deficiency. Mitochondria can enable tumor cells to gain the ability to proliferate, escape immune responses, and develop drug resistance by altering constitutive junctions, oxidative phosphorylation, oxidative stress, and mitochondrial subcellular relocalization. This greatly reduces the rate of effective clinical control of tumors. PURPOSE Explore the major role of mitochondria in cancer, as well as targeted mitochondrial therapies and mitochondria-associated markers. CONCLUSIONS This review provides a comprehensive analysis of the various aspects of mitochondrial aberrations and addresses drugs that target mitochondrial therapy, providing a basis for clinical mitochondria-targeted anti-tumor therapy.
Collapse
Affiliation(s)
- Zhiru Li
- Graduate School, North China University of Science and Technology, Tangshan, China
- The Fourth Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Wu Zhang
- Center of Treatment of Myasthenia Gravis, People’s Hospital of Shijiazhuang Affiliated to Hebei Medical, Shijiazhuang, China
| | - Shaowei Guo
- The Fourth Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Guoyan Qi
- Center of Treatment of Myasthenia Gravis, People’s Hospital of Shijiazhuang Affiliated to Hebei Medical, Shijiazhuang, China
| | - Jiandi Huang
- The Fourth Department of Oncology, Hebei General Hospital, Shijiazhuang, China
- Graduate School, Hebei North University, Zhangjiakou, China
| | - Jin Gao
- Department of Thyroid & Breast Surgery Ward, Hebei General Hospital, Shijiazhuang, China
| | - Jing Zhao
- The Sixth Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Lin Kang
- Department of Pathology, Hebei General Hospital, Shijiazhuang, China
| | - Qingxia Li
- The Fourth Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
21
|
Su P, Mao X, Ma J, Huang L, Yu L, Tang S, Zhuang M, Lu Z, Osafo KS, Ren Y, Wang X, Lin X, Huang L, Huang X, Braicu EI, Sehouli J, Sun P. ERRα promotes glycolytic metabolism and targets the NLRP3/caspase-1/GSDMD pathway to regulate pyroptosis in endometrial cancer. J Exp Clin Cancer Res 2023; 42:274. [PMID: 37864196 PMCID: PMC10588109 DOI: 10.1186/s13046-023-02834-7] [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: 06/06/2023] [Accepted: 09/15/2023] [Indexed: 10/22/2023] Open
Abstract
BACKGROUND Tumor cells can resist chemotherapy-induced pyroptosis through glycolytic reprogramming. Estrogen-related receptor alpha (ERRα) is a central regulator of cellular energy metabolism associated with poor cancer prognosis. Herein, we refine the oncogenic role of ERRα in the pyroptosis pathway and glycolytic metabolism. METHODS The interaction between ERRα and HIF-1α was verified using co-immunoprecipitation. The transcriptional binding sites of ERRα and NLRP3 were confirmed using dual-luciferase reporter assay and cleavage under targets and tagmentation (CUT&Tag). Flow cytometry, transmission electron microscopy, scanning electron microscopy, cell mito stress test, and extracellular acidification rate analysis were performed to investigate the effects of ERRα on the pyroptosis pathway and glycolytic metabolism. The results of these experiments were further confirmed in endometrial cancer (EC)-derived organoids and nude mice. In addition, the expression of ERRα-related pyroptosis genes was analyzed using The Cancer Genome Atlas and Gene Expression Omnibus database. RESULTS Triggered by a hypoxic microenvironment, highly expressed ERRα could bind to the promoter of NLRP3 and inhibit caspase-1/GSDMD signaling, which reduced inflammasome activation and increased pyroptosis resistance, thereby resulting in the resistance of cancer cells to cisplatin. Moreover, ERRα activated glycolytic rate-limiting enzyme to bridge glycolytic metabolism and pyroptosis in EC. This phenomenon was further confirmed in EC-derived organoids and nude mice. CUT & Tag sequencing and The Cancer Genome Atlas database analysis showed that ERRα participated in glycolysis and programmed cell death, which resulted in EC progression. CONCLUSIONS ERRα inhibits pyroptosis in an NLRP3-dependent manner and induces glycolytic metabolism, resulting in cisplatin resistance in EC cells.
Collapse
Affiliation(s)
- Pingping Su
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Xiaodan Mao
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Jincheng Ma
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Lixiang Huang
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Lirui Yu
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Shuting Tang
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Mingzhi Zhuang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, Fujian, China
| | - Zhonglei Lu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, Fujian, China
| | - Kelvin Stefan Osafo
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Yuan Ren
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Xinrui Wang
- Medical Research Center, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
| | - Xite Lin
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Leyi Huang
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Xiaoli Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Fujian Medical University, FuzhouFujian, 350005, China
| | - Elena Ioana Braicu
- Department of Gynecology and Obstetrics, Charité Virchow University Hospital, Augustenberger Platz1, 13353, Berlin, Germany
| | - Jalid Sehouli
- Department of Gynecology and Obstetrics, Charité Virchow University Hospital, Augustenberger Platz1, 13353, Berlin, Germany
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China.
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China.
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China.
- National Key Clinical Specialty Construction Program of China (Gynecology), Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
22
|
Zhang L, Li Z, Zhang L, Qin Y, Yu D. Dissecting the multifaced function of transcription factor EB (TFEB) in human diseases: From molecular mechanism to pharmacological modulation. Biochem Pharmacol 2023; 215:115698. [PMID: 37482200 DOI: 10.1016/j.bcp.2023.115698] [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: 05/09/2023] [Revised: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
The transcription factor EB (TFEB) is a transcription factor of the MiT/TFE family that translocations from the cytoplasm to the nucleus in response to various stimuli, including lysosomal stress and nutrient starvation. By activating genes involved in lysosomal function, autophagy, and lipid metabolism, TFEB plays a crucial role in maintaining cellular homeostasis. Dysregulation of TFEB has been implicated in various diseases, including cancer, neurodegenerative diseases, metabolic diseases, cardiovascular diseases, infectious diseases, and inflammatory diseases. Therefore, modulating TFEB activity with agonists or inhibitors may have therapeutic potential. In this review, we reviewed the recently discovered regulatory mechanisms of TFEB and their impact on human diseases. Additionally, we also summarize the existing TFEB inhibitors and agonists (targeted and non-targeted) and discuss unresolved issues and future research directions in the field. In summary, this review sheds light on the crucial role of TFEB, which may pave the way for its translation from basic research to practical applications, bringing us closer to realizing the full potential of TFEB in various fields.
Collapse
Affiliation(s)
- Lijuan Zhang
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Zhijia Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| | - Yuan Qin
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China; Medical Research Center, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China.
| | - Dongke Yu
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China.
| |
Collapse
|
23
|
Schüler-Toprak S, Skrzypczak M, Gründker C, Ortmann O, Treeck O. Role of Estrogen Receptor β, G-Protein Coupled Estrogen Receptor and Estrogen-Related Receptors in Endometrial and Ovarian Cancer. Cancers (Basel) 2023; 15:2845. [PMID: 37345182 DOI: 10.3390/cancers15102845] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/08/2023] [Accepted: 05/16/2023] [Indexed: 06/23/2023] Open
Abstract
Ovarian and endometrial cancers are affected by estrogens and their receptors. It has been long known that in different types of cancers, estrogens activate tumor cell proliferation via estrogen receptor α (ERα). In contrast, the role of ERs discovered later, including ERβ and G-protein-coupled ER (GPER1), in cancer is less well understood, but the current state of knowledge indicates them to have a considerable impact on both cancer development and progression. Moreover, estrogen related receptors (ERRs) have been reported to affect pathobiology of many tumor types. This article provides a summary and update of the current findings on the role of ERβ, GPER1, and ERRs in ovarian and endometrial cancer. For this purpose, original research articles on the role of ERβ, GPER1, and ERRs in ovarian and endometrial cancers listed in the PubMed database have been reviewed.
Collapse
Affiliation(s)
- Susanne Schüler-Toprak
- Department of Obstetrics and Gynecology, University Medical Center Regensburg, Caritas-Hospital St. Josef, 93053 Regensburg, Germany
| | - Maciej Skrzypczak
- Second Department of Gynecology, Medical University of Lublin, 20-954 Lublin, Poland
| | - Carsten Gründker
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Olaf Ortmann
- Department of Obstetrics and Gynecology, University Medical Center Regensburg, Caritas-Hospital St. Josef, 93053 Regensburg, Germany
| | - Oliver Treeck
- Department of Obstetrics and Gynecology, University Medical Center Regensburg, Caritas-Hospital St. Josef, 93053 Regensburg, Germany
| |
Collapse
|
24
|
Géhin C, Fowler SJ, Trivedi DK. Chewing the fat: How lipidomics is changing our understanding of human health and disease in 2022. ANALYTICAL SCIENCE ADVANCES 2023; 4:104-131. [PMID: 38715925 PMCID: PMC10989624 DOI: 10.1002/ansa.202300009] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/18/2023] [Accepted: 04/18/2023] [Indexed: 11/17/2024]
Abstract
Lipids are biological molecules that play vital roles in all living organisms. They perform many cellular functions, such as 1) forming cellular and subcellular membranes, 2) storing and using energy, and 3) serving as chemical messengers during intra- and inter-cellular signal transduction. The large-scale study of the pathways and networks of cellular lipids in biological systems is called "lipidomics" and is one of the fastest-growing omics technologies of the last two decades. With state-of-the-art mass spectrometry instrumentation and sophisticated data handling, clinical studies show how human lipid composition changes in health and disease, thereby making it a valuable medium to collect for clinical applications, such as disease diagnostics, therapeutic decision-making, and drug development. This review gives a comprehensive overview of current workflows used in clinical research, from sample collection and preparation to data and clinical interpretations. This is followed by an appraisal of applications in 2022 and a perspective on the exciting future of clinical lipidomics.
Collapse
Affiliation(s)
- Caroline Géhin
- Manchester Institute of Biotechnology, Department of ChemistryUniversity of ManchesterManchesterUK
| | - Stephen J. Fowler
- Department of Respiratory MedicineManchester University Hospitals NHS Foundation TrustManchesterUK
- School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
- NIHR Manchester Biomedical Research CentreManchester University Hospitals NHS Foundation TrustManchesterUK
| | - Drupad K. Trivedi
- Manchester Institute of Biotechnology, Department of ChemistryUniversity of ManchesterManchesterUK
| |
Collapse
|
25
|
Li J, Yang H, Zhang L, Zhang S, Dai Y. Metabolic reprogramming and interventions in endometrial carcinoma. Biomed Pharmacother 2023; 161:114526. [PMID: 36933381 DOI: 10.1016/j.biopha.2023.114526] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/18/2023] Open
Abstract
Cancer cells are usually featured by metabolic adaptations that facilitate their growth, invasion, and metastasis. Thus, reprogramming of intracellular energy metabolism is currently one of the hotspots in the field of cancer research. Whereas aerobic glycolysis (known as the Warburg effect) has long been considered a dominant form of energy metabolism in cancer cells, emerging evidence indicates that other metabolic forms, especially oxidative phosphorylation (OXPHOS), may play a critical role at least in some types of cancer. Of note, women with metabolic syndromes (MetS), including obesity, hyperglycemia, dyslipidemia, and hypertension, have an increased risk of developing endometrial carcinoma (EC), suggesting a close link between metabolism and EC. Interestingly, the metabolic preferences vary among EC cell types, particularly cancer stem cells and chemotherapy-resistant cells. Currently, it is commonly accepted that glycolysis is the main energy provider in EC cells, while OXPHOS is reduced or impaired. Moreover, agents specifically targeting the glycolysis and/or OXPHOS pathways can inhibit tumor cell growth and promote chemosensitization. For example, metformin and weight control not only reduce the incidence of EC but also improve the prognosis of EC patients. In this review, we comprehensively overview the current in-depth understanding of the relationship between metabolism and EC and provide up-to-date insights into the development of novel therapies targeting energy metabolism for auxiliary treatment in combination with chemotherapy for EC, especially those resistant to conventional chemotherapy.
Collapse
Affiliation(s)
- Jiajia Li
- The Laboratory of Cancer Precision Medicine, the First Hospital of Jilin University, Changchun, Jilin 130061, China; Department of Gynecologic Oncology, Gynecology and Obstetrics Center, the First Hospital of Jilin University, Changchun, Jilin 130012, China
| | - Hongmei Yang
- Department of Critical Care Medicine, the First Hospital of Jilin University, Changchun, Jilin 130012, China
| | - Lingyi Zhang
- Department of Gynecology and Obstetrics, the Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Songling Zhang
- Department of Gynecologic Oncology, Gynecology and Obstetrics Center, the First Hospital of Jilin University, Changchun, Jilin 130012, China.
| | - Yun Dai
- The Laboratory of Cancer Precision Medicine, the First Hospital of Jilin University, Changchun, Jilin 130061, China.
| |
Collapse
|
26
|
ERRα Up-Regulates Invadopodia Formation by Targeting HMGCS1 to Promote Endometrial Cancer Invasion and Metastasis. Int J Mol Sci 2023; 24:ijms24044010. [PMID: 36835419 PMCID: PMC9964422 DOI: 10.3390/ijms24044010] [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: 12/13/2022] [Revised: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Estrogen-related receptor alpha (ERRα) plays an important role in endometrial cancer (EC) progression. However, the biological roles of ERRα in EC invasion and metastasis are not clear. This study aimed to investigate the role of ERRα and 3-hydroxy-3-methylglutaryl-CoA synthase 1 (HMGCS1) in regulating intracellular cholesterol metabolism to promote EC progression. ERRα and HMGCS1 interactions were detected by co-immunoprecipitation, and the effects of ERRα/HMGCS1 on the metastasis of EC were investigated by wound-healing and transwell chamber invasion assays. Cellular cholesterol content was measured to verify the relationship between ERRα and cellular cholesterol metabolism. Additionally, immunohistochemistry was performed to confirm that ERRα and HMGCS1 were related to EC progression. Furthermore, the mechanism was investigated using loss-of-function and gain-of-function assays or treatment with simvastatin. High expression levels of ERRα and HMGCS1 promoted intracellular cholesterol metabolism for invadopodia formation. Moreover, inhibiting ERRα and HMGCS1 expression significantly weakened the malignant progression of EC in vitro and in vivo. Our functional analysis showed that ERRα promoted EC invasion and metastasis through the HMGCS1-mediated intracellular cholesterol metabolism pathway, which was dependent on the epithelial-mesenchymal transition pathway. Our findings suggest that ERRα and HMGCS1 are potential targets to suppress EC progression.
Collapse
|
27
|
Wang S, Hao HF, Jiao YN, Fu JL, Guo ZW, Guo Y, Yuan Y, Li PP, Han SY. Dandelion extract inhibits triple-negative breast cancer cell proliferation by interfering with glycerophospholipids and unsaturated fatty acids metabolism. Front Pharmacol 2022; 13:942996. [PMID: 36147318 PMCID: PMC9486077 DOI: 10.3389/fphar.2022.942996] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/17/2022] [Indexed: 11/23/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype with limited treatment options and a poor prognosis. TNBC exists widely reprogrammed lipid metabolism, and its metabolic-associated proteins and oncometabolites are promising as potential therapeutic targets. Dandelion (Taraxacum mongolicum) is a classical herbal medicine used to treat breast diseases based on traditional Chinese medicine theory and was reported to have antitumor effects and lipid regulatory capacities. Our previous study showed that dandelion extract was effective against TNBC. However, whether dandelion extract could regulate the lipid metabolisms of TNBC and exert its antitumor effects via interfering with lipids metabolism remained unclear. In this study, an integrated approach combined with network pharmacology and multi-omics techniques (including proteomics, metabolomics, and lipidomics) was performed to investigate the potential regulatory mechanisms of dandelion extract against TNBC. We first determined the antitumor effects of dandelion extract in vitro and in vivo. Then, network pharmacology analysis speculated the antitumor effects involving various metabolic processes, and the multi-omics results of the cells, tumor tissues, and plasma revealed the changes in the metabolites and metabolic-associated proteins after dandelion extract treatment. The alteration of glycerophospholipids and unsaturated fatty acids were the most remarkable types of metabolites. Therefore, the metabolism of glycerophospholipids and unsaturated fatty acids, and their corresponding proteins CHKA and FADS2, were considered the primary regulatory pathways and biomarkers of dandelion extract against TNBC. Subsequently, experimental validation showed that dandelion extract decreased CHKA expression, leading to the inhibition of the PI3K/AKT pathway and its downstream targets, SREBP and FADS2. Finally, the molecular docking simulation suggested that picrasinoside F and luteolin in dandelion extract had the most highly binding scores with CHKA, indicating they may be the potential CHKA inhibitors to regulate glycerophospholipids metabolisms of TNBC. In conclusion, we confirmed the antitumor effects of dandelion extract against TNBC cells in vitro and demonstrated that dandelion extract could interfere with glycerophospholipids and unsaturated fatty acids metabolism via downregulating the CHKA expression and inhibiting PI3K/AKT/SREBP/FADS2 axis.
Collapse
Affiliation(s)
- Shan Wang
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital and Institute, Beijing, China
| | - Hui-feng Hao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yan-na Jiao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jia-lei Fu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zheng-wang Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yang Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yuan Yuan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital and Institute, Beijing, China
| | - Ping-ping Li
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital and Institute, Beijing, China
- *Correspondence: Shu-yan Han, ; Ping-ping Li,
| | - Shu-yan Han
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital and Institute, Beijing, China
- *Correspondence: Shu-yan Han, ; Ping-ping Li,
| |
Collapse
|
28
|
The Regulatory Role of Lipid Metabolism in Endometrial Cancer. JOURNAL OF ONCOLOGY 2022; 2022:6458877. [PMID: 36072980 PMCID: PMC9444396 DOI: 10.1155/2022/6458877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/12/2022] [Indexed: 12/24/2022]
Abstract
Endometrial cancer is the 6th most common carcinoma as well as the 2nd most common malignancy worldwide in women. It is closely related to fat content, and dyslipidemia is among the most significant metabolic changes in this cancer. Therefore, further understanding of the regulation mechanism in lipid metabolism of endometrial cancer is conducive to the development of better therapeutic strategies and methods. Here, we systematically review the signaling pathways that regulate lipid metabolism in endometrial cancer and the research progress of drugs and targeted therapies that act on lipid metabolism by retrieving relevant articles. The underlying mechanism of occurrence and development of endometrial cancer is relatively clear and comprehensively reviewed here. But following more research studies will help to illuminate more specific regulatory roles of lipid metabolism in endometrial cancer and explore new possible mechanisms, prognostic and therapeutic targets, and subsequent drugs. Our review will provide a full view for the following investigation of lipid metabolism in endometrial cancer.
Collapse
|
29
|
Astanina E, Bussolino F, Doronzo G. Transcription factor EB controls both motogenic and mitogenic cell activities. FEBS Lett 2022; 596:1973-1980. [PMID: 35781277 DOI: 10.1002/1873-3468.14442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/09/2022] [Accepted: 06/26/2022] [Indexed: 11/10/2022]
Abstract
Transcription factor EB (TFEB) belongs to the microphthalmia family of bHLH-leucine zipper transcription factors and was first identified as an oncogene in a subset of renal cell carcinomas. In addition to exhibiting oncogenic activity, TFEB coordinates genetic programs connected with the cellular response to stress conditions, including roles in lysosome biogenesis, autophagy, modulation of metabolism. As is the case for other transcription factors, the activities of TFEB are not limited to a specific cellular condition such as the response to stress and recent findings indicate that TFEB has more widespread functions. Here, we review the emerging roles of TFEB in regulating cellular proliferation and motility. The well-established and emerging roles of TFEB suggest that this protein serves as a hub of signalling networks involved in many non-communicable diseases, such as cancer, ischaemic diseases and immune disorders, drug resistance mechanisms, and tissue generation.
Collapse
Affiliation(s)
- Elena Astanina
- Department of Oncology, University of Torino (IT).,Candiolo Cancer Institute-IRCCS-FPO, Candiolo (Torino) (IT)
| | - Federico Bussolino
- Department of Oncology, University of Torino (IT).,Candiolo Cancer Institute-IRCCS-FPO, Candiolo (Torino) (IT)
| | - Gabriella Doronzo
- Department of Oncology, University of Torino (IT).,Candiolo Cancer Institute-IRCCS-FPO, Candiolo (Torino) (IT)
| |
Collapse
|
30
|
Su P, Yu L, Mao X, Sun P. Role of HIF-1α/ERRα in Enhancing Cancer Cell Metabolism and Promoting Resistance of Endometrial Cancer Cells to Pyroptosis. Front Oncol 2022; 12:881252. [PMID: 35800058 PMCID: PMC9253301 DOI: 10.3389/fonc.2022.881252] [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: 02/22/2022] [Accepted: 05/25/2022] [Indexed: 12/24/2022] Open
Abstract
Oxygen is critical to energy metabolism, and tumors are often characterized by a hypoxic microenvironment. Owing to the high metabolic energy demand of malignant tumor cells, their survival is promoted by metabolic reprogramming in the hypoxic microenvironment, which can confer tumor cell resistance to pyroptosis. Pyroptosis resistance can inhibit anti-tumor immunity and promote the development of malignant tumors. Hypoxia inducible factor-1α (HIF-1α) is a key regulator of metabolic reprogramming in tumor cells, and estrogen-related receptor α (ERRα) plays a key role in regulating cellular energy metabolism. Therefore, the close interaction between HIF-1α and ERRα influences the metabolic and functional changes in cancer cells. In this review, we summarize the reprogramming of tumor metabolism involving HIF-1α/ERRα. We review our understanding of the role of HIF-1α/ERRα in promoting tumor growth adaptation and pyroptosis resistance, emphasize its key role in energy homeostasis, and explore the regulation of HIF-1α/ERRα in preventing and/or treating endometrial carcinoma patients. This review provides a new perspective for the study of the molecular mechanisms of metabolic changes in tumor progression.
Collapse
Affiliation(s)
- Pingping Su
- Laboratory of Gynecological Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Lirui Yu
- Laboratory of Gynecological Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiaodan Mao
- Laboratory of Gynecological Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Women and Children’s Critical Diseases Research, Fuzhou, China
| | - Pengming Sun
- Laboratory of Gynecological Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Women and Children’s Critical Diseases Research, Fuzhou, China
- *Correspondence: Pengming Sun,
| |
Collapse
|
31
|
Yu J, Yao HW, Liu TT, Wang D, Shi JH, Yuan GW, Ma S, Wu LY. Comprehensive Analysis and Experimental Validation of a Novel Estrogen/Progesterone-Related Prognostic Signature for Endometrial Cancer. J Pers Med 2022; 12:jpm12060914. [PMID: 35743699 PMCID: PMC9225250 DOI: 10.3390/jpm12060914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/16/2022] [Accepted: 05/23/2022] [Indexed: 12/04/2022] Open
Abstract
Estrogen and progesterone are the major determinants of the occurrence and development of endometrial cancer (EC), which is one of the most common gynecological cancers worldwide. Our purpose was to develop a novel estrogen/progesterone-related gene signature to better predict the prognosis of EC and help discover effective therapeutic strategies. We downloaded the clinical and RNA-seq data of 397 EC patients from The Cancer Genome Atlas (TCGA) database. The “limma” R package was used to screen for estrogen/progesterone-related differentially expressed genes (DEGs) between EC and normal tissues. Univariate and multivariate Cox proportional hazards regression analyses were applied to identify these DEGs that were associated with prognosis; then, a novel estrogen/progesterone-related prognostic signature comprising CDC25B, GNG3, ITIH3, PRXL2A and SDHB was established. The Kaplan–Meier (KM) survival analysis showed that the low-risk group identified by this signature had significantly longer overall survival (OS) than the high-risk group; the receiver operating characteristic (ROC) and risk distribution curves suggested this signature was an accurate predictor independent of risk factors. A nomogram incorporating the signature risk score and stage was constructed, and the calibration plot suggested it could accurately predict the survival rate. Compared with normal tissues, tumor tissues had increased mRNA levels of GNG3 and PRXL2A and a reduced mRNA level of ITIH3. The knockdown of PRXL2A and GNG3 significantly inhibited the proliferation and colony formation of Ishikawa and AN3CA cells, while the inhibition of PRXL2A expression suppressed xenograft growth. In this study, five estrogen/progesterone-related genes were identified and incorporated into a novel signature, which provided a new classification tool for improved risk assessment and potential molecular targets for EC therapies.
Collapse
Affiliation(s)
- Jing Yu
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (J.Y.); (H.-W.Y.); (G.-W.Y.)
| | - Hong-Wen Yao
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (J.Y.); (H.-W.Y.); (G.-W.Y.)
| | - Ting-Ting Liu
- Department of Blood Grouping, Beijing Red Cross Blood Center, Beijing 100088, China;
| | - Di Wang
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China;
| | - Jian-Hong Shi
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China;
| | - Guang-Wen Yuan
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (J.Y.); (H.-W.Y.); (G.-W.Y.)
| | - Sai Ma
- Department of Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
- Correspondence: (S.M.); (L.-Y.W.)
| | - Ling-Ying Wu
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; (J.Y.); (H.-W.Y.); (G.-W.Y.)
- Correspondence: (S.M.); (L.-Y.W.)
| |
Collapse
|
32
|
Li M, Jiang H, Chen S, Ma Y. GATA binding protein 1 recruits histone deacetylase 2 to the promoter region of nuclear receptor binding protein 2 to affect the tumor microenvironment and malignancy of thyroid carcinoma. Bioengineered 2022; 13:11320-11341. [PMID: 35491849 PMCID: PMC9278442 DOI: 10.1080/21655979.2022.2068921] [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] [Indexed: 12/24/2022] Open
Abstract
The tumor microenvironment (TME) and activated angiogenesis in thyroid carcinoma (TC) are critical for tumor growth and metastasis. Nuclear receptor binding protein 2 (NRBP2) has been suggested as a tumor suppressor. This study examines the function of NRBP2 in the progression of TC and the regulatory mechanism. By analyzing bioinformatic tools including GSE165724 dataset and the Cancer Genome Atlas system, we predicted NRBP2 as a poorly expressed gene in TC. Decreased NRBP2 expression was detected in TC tumor tissues and cells. Poor expression of NRBP2 was linked to unfavorable prognosis of patients. GATA binding protein 1 (GATA1) was found as a negative regulator of NRBP2. It recruited histone deacetylase2 (HDAC2) to the NRBP2 promoter to trigger histone deacetylation. NRBP2 overexpression suppressed growth of TC cells, and it reduced expression of TME markers, M2 polarization of macrophages, and angiogenesis in TC. Similar results were reproduced in vivo in nude mice. However, the anti-oncogenic roles of NRBP2 were blocked after further overexpression of GATA1 or HDAC2. In summary, this study demonstrates that GATA1 recruits HDAC2 to the NRBP2 promoter and enhances the TME and angiogenesis in TC cells.
Collapse
Affiliation(s)
- Mengyuan Li
- Department of Ultrasound, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, P.R. China
| | - Hongwei Jiang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, P.R. China
| | - Shengjiang Chen
- Department of Ultrasound, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, P.R. China
| | - Yujin Ma
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, P.R. China
| |
Collapse
|