1
|
Yang C, Li Z, Tian K, Meng X, Wang X, Song D, Wang X, Xu T, Sun P, Zhong J, Song Y, Ma W, Liu Y, Yu D, Shen R, Jiang C, Cai J. LncRNA-Mediated TPI1 and PKM2 Promote Self-Renewal and Chemoresistance in GBM. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2402600. [PMID: 39342418 DOI: 10.1002/advs.202402600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 09/15/2024] [Indexed: 10/01/2024]
Abstract
Temozolomide (TMZ) resistance is one of the major reasons for poor prognosis in patients with glioblastoma (GBM). Long noncoding RNAs (lncRNAs) are involved in multiple biological processes, including TMZ resistance. Linc00942 is a potential regulator of TMZ sensitivity in GBM cells is shown previously. However, the underlying mechanism of TMZ resistance induced by Linc00942 is unknown. In this study, the sequence of Linc00942 by rapid amplification of cDNA ends assay in TMZ-resistant GBM cells is identified and confirmed that Linc00942 contributes to self-renewal and TMZ resistance in GBM cells. Chromatin isolation by RNA purification followed by mass spectrometry (ChIRP-MS) and followed by Western blotting (ChIRP-WB) assays shows that Linc00492 interacted with TPI1 and PKM2, subsequently promoting their phosphorylation, dimerization, and nuclear translocation. The interaction of Linc00942 with TPI1 and PKM2 leads to increased acetylation of H3K4 and activation of the STAT3/P300 axis, resulting in the marked transcriptional activation of SOX9. Moreover, the knockdown of SOX9 reversed TMZ resistance induced by Linc00492 both in vitro and in vivo. In summary, Linc00942 strongly promotes SOX9 expression by interacting with TPI1 and PKM2 is found, thereby driving self-renewal and TMZ resistance in GBM cells. These findings suggest potential combined therapeutic strategies to overcome TMZ resistance in patients with GBM.
Collapse
Affiliation(s)
- Changxiao Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Ziwei Li
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Kaifu Tian
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Xiangqi Meng
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Xinyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Dan Song
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Xuan Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Tianye Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Penggang Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Junzhe Zhong
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Yu Song
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Wenbin Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Yuxiang Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Daohan Yu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Ruofei Shen
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Chuanlu Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
- The Sixth Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Jinquan Cai
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| |
Collapse
|
2
|
Yang JC, Zhao J, Chen YH, Wang R, Rong Z, Wang SY, Wu YM, Wang HN, Yang L, Liu R. miR-29a-5p rescues depressive-like behaviors in a CUMS-induced mouse model by facilitating microglia M2-polarization in the prefrontal cortex via TMEM33 suppression. J Affect Disord 2024; 360:188-197. [PMID: 38821373 DOI: 10.1016/j.jad.2024.05.156] [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: 09/05/2023] [Revised: 05/20/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
BACKGROUND Depression accounts for a high proportion of neuropsychiatric disorders and is associated with abnormal states of neurons in specific brain regions. Microglia play a pivotal role in the inflammatory state during depression development; however, the exact mechanism underlying chronic mood states remains unknown. Thus, the present study aimed to determine whether microRNAs (miRNAs) alleviate stress-induced depression-like behavior in mice by regulating the expression levels of their target genes, explore the role of neuroinflammation induced by microglial activation in the pathogenesis and progression of depression, and determine whether the role of the miR-29a-5p/transmembrane protein 33 (TMEM33) axis. METHODS In this study, chronic unpredictable mild stress (CUMS) mouse depression model, various behavioral tests, western blotting, dual-luciferase reporter assay, enzyme-linked immunosorbent assay, real-time quantitative reverse transcription PCR, immunofluorescence and lentivirus-mediated gene transfer were used. RESULTS After exposure to the CUMS paradigm, miR-29a-5p was significantly down-regulated. This downregulation subsequently promoted the polarization of microglia M1 by upregulating the expression of TMEM33, resulting in enhanced inflammatory chemokines affecting neurons. Conversely, the upregulation of miR-29a-5p within the prefrontal cortex (PFC) suppressed TMEM33 expression, facilitated microglia M2-polarization, and ameliorated depressive-like behavior. LIMITATIONS Only rodent models of depression were used, and human samples were not included. CONCLUSIONS The results of this study suggest that miR-29a-5p deficits within the PFC mediate microglial anomalies and contribute to depressive-like behaviors. miR-29a-5p and TMEM33 may, therefore, serve as potential therapeutic targets for the treatment of depression.
Collapse
Affiliation(s)
- Jing-Cheng Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, Shaanxi Province, China
| | - Jun Zhao
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, Shaanxi Province, China
| | - Yi-Huan Chen
- Department of Psychiatry, Xijing Hospital, Air Force Medical University, Xi'an 710032, Shaanxi Province, China
| | - Rui Wang
- Department of Military Medical Center, Tangdu Hospital, Air Force Medical University, Xi'an 710038, Shaanxi Province, China
| | - Zheng Rong
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, Shaanxi Province, China
| | - Sai-Ying Wang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, Shaanxi Province, China
| | - Yu-Mei Wu
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, Shaanxi Province, China
| | - Hua-Ning Wang
- Department of Psychiatry, Xijing Hospital, Air Force Medical University, Xi'an 710032, Shaanxi Province, China.
| | - Le Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, Shaanxi Province, China.
| | - Rui Liu
- Department of Rehabilitation, Tangdu Hospital, Air Force Medical University, Xi'an 710038, Shaanxi Province, China.
| |
Collapse
|
3
|
Jin Q, Qi D, Zhang M, Qu H, Dong Y, Sun M, Quan C. CLDN6 inhibits breast cancer growth and metastasis through SREBP1-mediated RAS palmitoylation. Cell Mol Biol Lett 2024; 29:112. [PMID: 39169280 PMCID: PMC11337767 DOI: 10.1186/s11658-024-00629-y] [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: 02/04/2024] [Accepted: 08/08/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Breast cancer (BC) ranks as the third most fatal malignant tumor worldwide, with a strong reliance on fatty acid metabolism. CLDN6, a candidate BC suppressor gene, was previously identified as a regulator of fatty acid biosynthesis; however, the underlying mechanism remains elusive. In this research, we aim to clarify the specific mechanism through which CLDN6 modulates fatty acid anabolism and its impact on BC growth and metastasis. METHODS Cell function assays, tumor xenograft mouse models, and lung metastasis mouse models were conducted to evaluate BC growth and metastasis. Human palmitic acid assay, triglyceride assay, Nile red staining, and oil red O staining were employed to investigate fatty acid anabolism. Reverse transcription polymerase chain reaction (RT-PCR), western blot, immunohistochemistry (IHC) assay, nuclear fractionation, immunofluorescence (IF), immunoprecipitation and acyl-biotin exchange (IP-ABE), chromatin immunoprecipitation (ChIP), dual luciferase reporter assay, and co-immunoprecipitation (Co-IP) were applied to elucidate the underlying molecular mechanism. Moreover, tissue microarrays of BC were analyzed to explore the clinical implications. RESULTS We identified that CLDN6 inhibited BC growth and metastasis by impeding RAS palmitoylation both in vitro and in vivo. We proposed a unique theory suggesting that CLDN6 suppressed RAS palmitoylation through SREBP1-modulated de novo palmitic acid synthesis. Mechanistically, CLDN6 interacted with MAGI2 to prevent KLF5 from entering the nucleus, thereby restraining SREBF1 transcription. The downregulation of SREBP1 reduced de novo palmitic acid synthesis, hindering RAS palmitoylation and subsequent endosomal sorting complex required for transport (ESCRT)-mediated plasma membrane localization required for RAS oncogenic activation. Besides, targeting inhibition of RAS palmitoylation synergized with CLDN6 to repress BC progression. CONCLUSIONS Our findings provide compelling evidence that CLDN6 suppresses the palmitic acid-induced RAS palmitoylation through the MAGI2/KLF5/SREBP1 axis, thereby impeding BC malignant progression. These results propose a new insight that monitoring CLDN6 expression alongside targeting inhibition of palmitic acid-mediated palmitoylation could be a viable strategy for treating oncogenic RAS-driven BC.
Collapse
Affiliation(s)
- Qiu Jin
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, China
| | - Da Qi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, China
| | - Mingzi Zhang
- The Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience Keck School of Medicine of the University of Southern California, 1501 San Pablo Street, Los Angeles, 90033, CA, US
| | - Huinan Qu
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, China
| | - Yuan Dong
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, China
| | - Minghao Sun
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, China
| | - Chengshi Quan
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, China.
| |
Collapse
|
4
|
Liu Y, Zhao Y, Song H, Li Y, Liu Z, Ye Z, Zhao J, Wu Y, Tang J, Yao M. Metabolic reprogramming in tumor immune microenvironment: Impact on immune cell function and therapeutic implications. Cancer Lett 2024; 597:217076. [PMID: 38906524 DOI: 10.1016/j.canlet.2024.217076] [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: 03/18/2024] [Revised: 05/23/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
Understanding of the metabolic reprogramming has revolutionized our insights into tumor progression and potential treatment. This review concentrates on the aberrant metabolic pathways in cancer cells within the tumor microenvironment (TME). Cancer cells differ from normal cells in their metabolic processing of glucose, amino acids, and lipids in order to adapt to heightened biosynthetic and energy needs. These metabolic shifts, which crucially alter lactic acid, amino acid and lipid metabolism, affect not only tumor cell proliferation but also TME dynamics. This review also explores the reprogramming of various immune cells in the TME. From a therapeutic standpoint, targeting these metabolic alterations represents a novel cancer treatment strategy. This review also discusses approaches targeting the regulation of metabolism of different nutrients in tumor cells and influencing the tumor microenvironment to enhance the immune response. In summary, this review summarizes metabolic reprogramming in cancer and its potential as a target for innovative therapeutic strategies, offering fresh perspectives on cancer treatment.
Collapse
Affiliation(s)
- Yuqiang Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Thoracic Surgery and Oncology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Yu Zhao
- Department of Thoracic Surgery, Sheng Jing Hospital, China Medical University, Shenyang, Liaoning, 110000, China
| | - Huisheng Song
- Affiliated Qingyuan Hospital, Guangzhou Medica University, Qingyuan People's Hospital, Qingyuan, Guangdong, 511500, China
| | - Yunting Li
- Department of Pediatrics, Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Zihao Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Thoracic Surgery and Oncology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Zhiming Ye
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Thoracic Surgery and Oncology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Jianzhu Zhao
- Department of oncology, Sheng Jing Hospital, China Medical University, Shenyang, Liaoning, 110000, China
| | - Yuzheng Wu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Thoracic Surgery and Oncology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Jun Tang
- Department of Thoracic Surgery, Sheng Jing Hospital, China Medical University, Shenyang, Liaoning, 110000, China.
| | - Maojin Yao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Thoracic Surgery and Oncology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China.
| |
Collapse
|
5
|
Zhang H, Liu Y, Zhang K, Hong Z, Liu Z, Liu Z, Li G, Xu Y, Pi J, Fu J, Xu Y. Understanding the Transcription Factor NFE2L1/NRF1 from the Perspective of Hallmarks of Cancer. Antioxidants (Basel) 2024; 13:758. [PMID: 39061827 PMCID: PMC11274343 DOI: 10.3390/antiox13070758] [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: 04/14/2024] [Revised: 06/15/2024] [Accepted: 06/17/2024] [Indexed: 07/28/2024] Open
Abstract
Cancer cells subvert multiple properties of normal cells, including escaping strict cell cycle regulation, gaining resistance to cell death, and remodeling the tumor microenvironment. The hallmarks of cancer have recently been updated and summarized. Nuclear factor erythroid 2-related factor 1 (NFE2L1, also named NRF1) belongs to the cap'n'collar (CNC) basic-region leucine zipper (bZIP) family. It acts as a transcription factor and is indispensable for maintaining both cellular homoeostasis and organ integrity during development and growth, as well as adaptive responses to pathophysiological stressors. In addition, NFE2L1 mediates the proteasome bounce-back effect in the clinical proteasome inhibitor therapy of neuroblastoma, multiple myeloma, and triple-negative breast cancer, which quickly induces proteasome inhibitor resistance. Recent studies have shown that NFE2L1 mediates cell proliferation and metabolic reprogramming in various cancer cell lines. We combined the framework provided by "hallmarks of cancer" with recent research on NFE2L1 to summarize the role and mechanism of NFE2L1 in cancer. These ongoing efforts aim to contribute to the development of potential novel cancer therapies that target the NFE2L1 pathway and its activity.
Collapse
Affiliation(s)
- Haomeng Zhang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital, China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang 110001, China
| | - Yong Liu
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Ke Zhang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital, China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang 110001, China
| | - Zhixuan Hong
- Department of Nutrition and Food Hygiene, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Zongfeng Liu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital, China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang 110001, China
| | - Zhe Liu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital, China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang 110001, China
| | - Guichen Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital, China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang 110001, China
| | - Yuanyuan Xu
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
- Laboratory of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
- Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Jingbo Pi
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
- Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Jingqi Fu
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
- Department of Nutrition and Food Hygiene, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Yuanhong Xu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital, China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang 110001, China
| |
Collapse
|
6
|
Cheng Y, He J, Zuo B, He Y. Role of lipid metabolism in hepatocellular carcinoma. Discov Oncol 2024; 15:206. [PMID: 38833109 DOI: 10.1007/s12672-024-01069-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 05/28/2024] [Indexed: 06/06/2024] Open
Abstract
Hepatocellular carcinoma (HCC), an aggressive malignancy with a dismal prognosis, poses a significant public health challenge. Recent research has highlighted the crucial role of lipid metabolism in HCC development, with enhanced lipid synthesis and uptake contributing to the rapid proliferation and tumorigenesis of cancer cells. Lipids, primarily synthesized and utilized in the liver, play a critical role in the pathological progression of various cancers, particularly HCC. Cancer cells undergo metabolic reprogramming, an essential adaptation to the tumor microenvironment (TME), with fatty acid metabolism emerging as a key player in this process. This review delves into intricate interplay between HCC and lipid metabolism, focusing on four key areas: de novo lipogenesis, fatty acid oxidation, dysregulated lipid metabolism of immune cells in the TME, and therapeutic strategies targeting fatty acid metabolism for HCC treatment.
Collapse
Affiliation(s)
- Yulin Cheng
- MOE Engineering Center of Hematological Disease, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Cyrus Tang Hematology Center, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Jun He
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Bin Zuo
- MOE Engineering Center of Hematological Disease, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Cyrus Tang Hematology Center, Soochow University, Suzhou, Jiangsu, 215006, China
| | - Yang He
- MOE Engineering Center of Hematological Disease, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Cyrus Tang Hematology Center, Soochow University, Suzhou, Jiangsu, 215006, China.
- MOH Key Lab of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
7
|
Zhao W, Cai Z, Zhang J, Zhang X, Yu B, Fu X, Zhang T, Hu J, Shao Y, Gu Y. PKM2 promotes myoblast growth and inosine monophosphate-specific deposition in Jingyuan chicken. Res Vet Sci 2024; 173:105275. [PMID: 38678847 DOI: 10.1016/j.rvsc.2024.105275] [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: 02/08/2024] [Revised: 04/08/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
Inosine monophosphate (IMP) is widely regarded as an important indicator for evaluating the flavour of poultry meat. However, little is known about the molecular mechanisms affecting the specific deposition of IMP. In this study, we functionally verified PKM2 (Pyruvate kinase M2), a candidate gene related to IMP synthesis, in order to reveal the important role of PKM2 in meat flavour and muscle development of Jingyuan chickens. The results showed that the IMP content in breast muscle of Jingyuan chickens was negatively correlated with PKM2 mRNA expression (r = -0.1710), while the IMP content in leg muscle was significantly positively correlated with PKM2 mRNA expression (r = 0.7350) (P < 0.05). During myogenesis, PKM2 promoted the proliferation rate of myoblasts and the expression of proliferation marker genes, inhibited the apoptosis rate and the expression of apoptosis marker genes, and decreased the expression of differentiation marker genes. Up-regulation of PKM2 enhanced the expression of key genes in the purine metabolic pathway and the de novo synthesis pathway of IMP, and suppressed the expression of key genes in the salvage pathway. ELISA assays showed that PKM2 decreased IMP and hypoxanthine (HX) contents, while adenosine triphosphate (ATP) and uric acid (UA) contents were clearly elevated. In summary, these studies revealed that PKM2 regulates myogenesis and specific deposition of IMP, which can be used to improve the quality of Jingyuan chicken meat.
Collapse
Affiliation(s)
- Wei Zhao
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Zhengyun Cai
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Juan Zhang
- College of Animal Science and Technology, Ningxia University, Yinchuan, China.
| | - Xinyu Zhang
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Baojun Yu
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Xi Fu
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Tong Zhang
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Jiahuan Hu
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Yandi Shao
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Yaling Gu
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| |
Collapse
|
8
|
Zhang S, Kim EJ, Huang J, Liu P, Donahue K, Wang Q, Wang Y, Mcilwain S, Xie L, Chen X, Li L, Xu W. NEAT1 repression by MED12 creates chemosensitivity in p53 wild-type breast cancer cells. FEBS J 2024; 291:1909-1924. [PMID: 38380720 PMCID: PMC11068489 DOI: 10.1111/febs.17097] [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: 07/09/2023] [Revised: 12/05/2023] [Accepted: 02/09/2024] [Indexed: 02/22/2024]
Abstract
Breast cancer is often treated with chemotherapy. However, the development of chemoresistance results in treatment failure. Long non-coding RNA nuclear paraspeckle assembly transcript 1 (NEAT1) has been shown to contribute to chemoresistance in breast cancer cells. In studying the transcriptional regulation of NEAT1 using multi-omics approaches, we showed that NEAT1 is up-regulated by 5-fluorouracil in breast cancer cells with wild-type cellular tumor antigen p53 but not in mutant-p53-expressing breast cancer cells. The regulation of NEAT1 involves mediator complex subunit 12 (MED12)-mediated repression of histone acetylation marks at the promoter region of NEAT1. Knockdown of MED12 but not coactivator-associated arginine methyltransferase 1 (CARM1) induced histone acetylation at the NEAT1 promoter, leading to elevated NEAT1 mRNAs, resulting in a chemoresistant phenotype. The MED12-dependent regulation of NEAT1 differs between wild-type and mutant p53-expressing cells. MED12 depletion led to increased expression of NEAT1 in a wild-type p53 cell line, but decreased expression in a mutant p53 cell line. Chemoresistance caused by MED12 depletion can be partially rescued by NEAT1 knockdown in p53 wild-type cells. Collectively, our study reveals a novel mechanism of chemoresistance dependent on MED12 transcriptional regulation of NEAT1 in p53 wild-type breast cancer cells.
Collapse
Affiliation(s)
- Shengjie Zhang
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA
- Present Address: Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Eui-Jun Kim
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Junfeng Huang
- School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
| | - Peng Liu
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53706, USA
- Department of Biostatistics and Medical Informatics, Carbone Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Kristine Donahue
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Qinchuan Wang
- Department of Surgical Oncology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310018, China
| | - Yidan Wang
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sean Mcilwain
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53706, USA
| | - Ling Xie
- Department of Biochemistry & Biophysics, Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Xian Chen
- Department of Biochemistry & Biophysics, Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53706, USA
| | - Wei Xu
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
9
|
Upadhyay S, Khan S, Hassan MI. Exploring the diverse role of pyruvate kinase M2 in cancer: Navigating beyond glycolysis and the Warburg effect. Biochim Biophys Acta Rev Cancer 2024; 1879:189089. [PMID: 38458358 DOI: 10.1016/j.bbcan.2024.189089] [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: 12/20/2023] [Revised: 02/25/2024] [Accepted: 03/03/2024] [Indexed: 03/10/2024]
Abstract
Pyruvate Kinase M2, a key enzyme in glycolysis, has garnered significant attention in cancer research due to its pivotal role in the metabolic reprogramming of cancer cells. Originally identified for its association with the Warburg effect, PKM2 has emerged as a multifaceted player in cancer biology. The functioning of PKM2 is intricately regulated at multiple levels, including controlling the gene expression via various transcription factors and non-coding RNAs, as well as adding post-translational modifications that confer distinct functions to the protein. Here, we explore the diverse functions of PKM2, encompassing newly emerging roles in non-glycolytic metabolic regulation, immunomodulation, inflammation, DNA repair and mRNA processing, beyond its canonical role in glycolysis. The ever-expanding list of its functions has recently grown to include roles in subcellular compartments such as the mitochondria and extracellular milieu as well, all of which make PKM2 an attractive drug target in the pursuit of therapeutics for cancer.
Collapse
Affiliation(s)
- Saurabh Upadhyay
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Shumayila Khan
- International Health Division, Indian Council of Medical Research, Ansari Nagar, New Delhi 110029, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India.
| |
Collapse
|
10
|
Kapoor S, Kalmegh V, Kumar H, Mandoli A, Shard A. Rare diseases and pyruvate kinase M2: a promising therapeutic connection. Drug Discov Today 2024; 29:103949. [PMID: 38492882 DOI: 10.1016/j.drudis.2024.103949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Pyruvate kinase M2 (PKM2) is a key glycolytic enzyme that regulates proliferating cell metabolism. The role of PKM2 in common diseases has been well established, but its role in rare diseases (RDs) is less understood. Over the past few years, PKM2 has emerged as a crucial player in RDs, including, neoplastic, respiratory, metabolic, and neurological disorders. Herein, we summarize recent findings and developments highlighting PKM2 as an emerging key player in RDs. We also discuss the current status of PKM2 modulation in RDs with particular emphasis on preclinical and clinical studies in addition to current challenges in the field.
Collapse
Affiliation(s)
- Saumya Kapoor
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India
| | - Vaishnavi Kalmegh
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, NIPER-A, Gandhinagar, Gujarat, India.
| | - Amit Mandoli
- Department of Biotechnology, NIPER-A, Gandhinagar, Gujarat, India.
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India.
| |
Collapse
|
11
|
Zou Y, Zhang Y, Li M, Cao K, Song C, Zhang Z, Cai K, Geng D, Chen S, Wu Y, Zhang N, Sun G, Wang J, Zhang Y, Sun Y. Regulation of lipid metabolism by E3 ubiquitin ligases in lipid-associated metabolic diseases. Int J Biol Macromol 2024; 265:130961. [PMID: 38508558 DOI: 10.1016/j.ijbiomac.2024.130961] [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: 07/25/2023] [Revised: 03/10/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024]
Abstract
Previous studies have progressively elucidated the involvement of E3 ubiquitin (Ub) ligases in regulating lipid metabolism. Ubiquitination, facilitated by E3 Ub ligases, modifies critical enzymes in lipid metabolism, enabling them to respond to specific signals. In this review, we aim to present a comprehensive analysis of the role of E3 Ub ligases in lipid metabolism, which includes lipid synthesis and lipolysis, and their influence on cellular lipid homeostasis through the modulation of lipid uptake and efflux. Furthermore, it explores how the ubiquitination process governs the degradation or activation of pivotal enzymes, thereby regulating lipid metabolism at the transcriptional level. Perturbations in lipid metabolism have been implicated in various diseases, including hepatic lipid metabolism disorders, atherosclerosis, diabetes, and cancer. Therefore, this review focuses on the association between E3 Ub ligases and lipid metabolism in lipid-related diseases, highlighting enzymes critically involved in lipid synthesis and catabolism, transcriptional regulators, lipid uptake translocators, and transporters. Overall, this review aims to identify gaps in current knowledge, highlight areas requiring further research, offer potential targeted therapeutic approaches, and provide a comprehensive outlook on clinical conditions associated with lipid metabolic diseases.
Collapse
Affiliation(s)
- Yuanming Zou
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China; Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Mohan Li
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Kexin Cao
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Chunyu Song
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Zhaobo Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Kexin Cai
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Danxi Geng
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Shuxian Chen
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yanjiao Wu
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China; Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110001, Liaoning Province, People's Republic of China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, 77 Puhe Road, Shenbei New District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Guozhe Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Jing Wang
- Department of Hematology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Yixiao Zhang
- Department of Urology Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China; Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110001, Liaoning Province, People's Republic of China; Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| |
Collapse
|
12
|
Kamil M, Kina UY, Atmaca HN, Unal S, Deveci G, Burak P, Aly ASI. Endoplasmic reticulum localized TMEM33 domain-containing protein is crucial for all life cycle stages of the malaria parasite. Mol Microbiol 2024; 121:767-780. [PMID: 38238886 DOI: 10.1111/mmi.15228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 12/14/2023] [Accepted: 01/06/2024] [Indexed: 04/19/2024]
Abstract
Endoplasmic reticulum (ER) plays a pivotal role in the regulation of stress responses in multiple eukaryotic cells. However, little is known about the effector mechanisms that regulate stress responses in ER of the malaria parasite. Herein, we aimed to identify the importance of a transmembrane protein 33 (TMEM33)-domain-containing protein in life cycle of the rodent malaria parasite Plasmodium berghei. TMEM33 is an ER membrane-resident protein that is involved in regulating stress responses in various eukaryotic cells. A C-terminal tagged TMEM33 was localized in the ER throughout the blood and mosquito stages of development. Targeted deletion of TMEM33 confirmed its importance for asexual blood stages and ookinete development, in addition to its essential role for sporozoite infectivity in the mammalian host. Pilot scale analysis shows that the loss of TMEM33 results in the initiation of ER stress response and induction of autophagy. Our findings conclude an important role of TMEM33 in the development of all life cycle stages of the malaria parasite, which indicates its potential as an antimalarial target.
Collapse
Affiliation(s)
- Mohd Kamil
- Aly Lab, Department of Microbiology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Istanbul, Turkey
- Center for Global Infectious Disease Research, Seattle Childrens Research Institute, Seattle, Washington, USA
| | - Umit Yasar Kina
- Aly Lab, Department of Microbiology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Istanbul, Turkey
| | - Habibe Nur Atmaca
- Aly Lab, Department of Microbiology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Istanbul, Turkey
| | - Sinem Unal
- Aly Lab, Department of Microbiology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Istanbul, Turkey
| | - Gozde Deveci
- Aly Lab, Department of Microbiology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Istanbul, Turkey
| | - Pinar Burak
- Aly Lab, Department of Microbiology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Istanbul, Turkey
| | - Ahmed S I Aly
- Aly Lab, Department of Microbiology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Istanbul, Turkey
- Department of Biotechnology, School of Science and Engineering, Al Akhawayn University, Ifrane, Morocco
| |
Collapse
|
13
|
Wang M, Li Y, Li S, Wang T, Wang M, Wu H, Zhang M, Luo S, Zhao C, Li Q, Cheng H. Cinobufacini injection delays hepatocellular carcinoma progression by regulating lipid metabolism via SREBP1 signaling pathway and affecting macrophage polarization. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117472. [PMID: 37995825 DOI: 10.1016/j.jep.2023.117472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 11/05/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cinobufacini injection, an aqueous extract of the toad, is a commonly used anti-tumor animal herbal medicine in clinical practice. It has the effects of detoxifying, reducing swelling, and relieving pain. AIMS OF THE STUDY To investigate the effects of Cinobufacini injection on hepatocellular carcinoma progression by regulating lipid metabolism and macrophage polarization in the tumor microenvironment and to identify the potential molecular mechanisms. MATERIALS AND METHODS To establish the axillary transplantation tumor model of hepatocellular carcinoma Hepa1-6 in C57BL/6 mice, and to evaluate the inhibitory effect of Cinobufacini injection on hepatocellular carcinoma in vivo as well as drug delivery security. Combined metabolomics and transcriptomics analysis of the effect of Cinobufagin Injection on tumor microenvironment. An in vitro mouse co-culture model of peritoneal macrophages and Hepa1-6 cells was established to research the effects of Cinobufacini injection on macrophage polarization, hepatocellular carcinoma cell growth, migration, and changes in lipid metabolism. Cinobufacini injection inhibition of the AMPK/SREBP1/FASN signaling pathway regulating cholesterol metabolism and affecting macrophage polarization was examined using qRT-PCR, lentiviral transfection, immunofluorescence, and Western blot. RESULT In vivo experiments demonstrated that Cinobufacini injection treatment significantly inhibited the growth of Hepa1-6 hepatomas, along with a reduction in cholesterol content and a decrease in the percentage of M2 macrophages in tumor tissue. In vitro, we found that Cinobufacini injection inhibits IL-4-induced M2 macrophage polarization, reduces the cholesterol content of Hepa1-6 cells in a co-culture system, and inhibits the promotion of hepatocellular carcinoma cells by M2 macrophages. In addition, successful overexpression of SREBP1 in Hepa1-6 cells showed more pronounced cellular activity whereas Cinobufacini injection inhibited this change and reduced intracellular lipid levels. CONCLUSION Cinobufacini injection inhibits cholesterol synthesis within the tumor microenvironment via the AMPK/SERBP1/FASN signaling pathway, which in turn blocks the M2 polarization of macrophages, leading to the weakening of hepatocellular carcinoma growth and migration, and the promotion of its apoptosis. Our findings provide an important Introduction to understanding the molecular mechanism of Cinobufacini injection's anticancer activity and provide reliable theoretical and experimental support for its clinical application.
Collapse
Affiliation(s)
- Meng Wang
- Anhui University of Chinese Medicine, Key Laboratory of Xin'an Medicine, China, The Functional Activity and Resource Utilization on Edible and Medicinal Fungi Joint Laboratory of Anhui Province, Hefei, 230038, China
| | - Yueyue Li
- Anhui University of Chinese Medicine, Key Laboratory of Xin'an Medicine, China, The Functional Activity and Resource Utilization on Edible and Medicinal Fungi Joint Laboratory of Anhui Province, Hefei, 230038, China
| | - Shanshan Li
- Anhui University of Chinese Medicine, Key Laboratory of Xin'an Medicine, China, The Functional Activity and Resource Utilization on Edible and Medicinal Fungi Joint Laboratory of Anhui Province, Hefei, 230038, China
| | - Ting Wang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230000, China
| | - Manman Wang
- Anhui University of Chinese Medicine, Key Laboratory of Xin'an Medicine, China, The Functional Activity and Resource Utilization on Edible and Medicinal Fungi Joint Laboratory of Anhui Province, Hefei, 230038, China
| | - Huan Wu
- Anhui University of Chinese Medicine, Key Laboratory of Xin'an Medicine, China, The Functional Activity and Resource Utilization on Edible and Medicinal Fungi Joint Laboratory of Anhui Province, Hefei, 230038, China
| | - Mei Zhang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230000, China
| | - Shengyong Luo
- Anhui Academy of Medical Sciences, Hefei, 230061, China
| | - Cheng Zhao
- Anqing Shihua Hospital of Nanjing Drum Tower Hospital Group, Anqing, 264000, China
| | - Qinglin Li
- Anhui University of Chinese Medicine, Key Laboratory of Xin'an Medicine, China, The Functional Activity and Resource Utilization on Edible and Medicinal Fungi Joint Laboratory of Anhui Province, Hefei, 230038, China.
| | - Hui Cheng
- Anhui University of Chinese Medicine, Key Laboratory of Xin'an Medicine, China, The Functional Activity and Resource Utilization on Edible and Medicinal Fungi Joint Laboratory of Anhui Province, Hefei, 230038, China.
| |
Collapse
|
14
|
Guerrero-Ochoa P, Rodríguez-Zapater S, Anel A, Esteban LM, Camón-Fernández A, Espilez-Ortiz R, Gil-Sanz MJ, Borque-Fernando Á. Prostate Cancer and the Mevalonate Pathway. Int J Mol Sci 2024; 25:2152. [PMID: 38396837 PMCID: PMC10888820 DOI: 10.3390/ijms25042152] [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: 01/10/2024] [Revised: 02/04/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Antineoplastic therapies for prostate cancer (PCa) have traditionally centered around the androgen receptor (AR) pathway, which has demonstrated a significant role in oncogenesis. Nevertheless, it is becoming progressively apparent that therapeutic strategies must diversify their focus due to the emergence of resistance mechanisms that the tumor employs when subjected to monomolecular treatments. This review illustrates how the dysregulation of the lipid metabolic pathway constitutes a survival strategy adopted by tumors to evade eradication efforts. Integrating this aspect into oncological management could prove valuable in combating PCa.
Collapse
Affiliation(s)
- Patricia Guerrero-Ochoa
- Health Research Institute of Aragon Foundation, 50009 Zaragoza, Spain; (P.G.-O.); (A.C.-F.); (R.E.-O.); (M.J.G.-S.)
| | - Sergio Rodríguez-Zapater
- Minimally Invasive Research Group (GITMI), Faculty of Veterinary Medicine, University of Zaragoza, 50009 Zaragoza, Spain;
| | - Alberto Anel
- Department of Biochemistry and Molecular and Cellular Biology, Faculty of Sciences, University of Zaragoza, 50009 Zaragoza, Spain;
| | - Luis Mariano Esteban
- Department of Applied Mathematics, Escuela Universitaria Politécnica de La Almunia, Institute for Biocomputation and Physic of Complex Systems, Universidad de Zaragoza, 50100 La Almunia de Doña Godina, Spain
| | - Alejandro Camón-Fernández
- Health Research Institute of Aragon Foundation, 50009 Zaragoza, Spain; (P.G.-O.); (A.C.-F.); (R.E.-O.); (M.J.G.-S.)
| | - Raquel Espilez-Ortiz
- Health Research Institute of Aragon Foundation, 50009 Zaragoza, Spain; (P.G.-O.); (A.C.-F.); (R.E.-O.); (M.J.G.-S.)
- Department of Urology, Miguel Servet University Hospital, 50009 Zaragoza, Spain
- Area of Urology, Department of Surgery, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| | - María Jesús Gil-Sanz
- Health Research Institute of Aragon Foundation, 50009 Zaragoza, Spain; (P.G.-O.); (A.C.-F.); (R.E.-O.); (M.J.G.-S.)
- Department of Urology, Miguel Servet University Hospital, 50009 Zaragoza, Spain
| | - Ángel Borque-Fernando
- Health Research Institute of Aragon Foundation, 50009 Zaragoza, Spain; (P.G.-O.); (A.C.-F.); (R.E.-O.); (M.J.G.-S.)
- Department of Applied Mathematics, Escuela Universitaria Politécnica de La Almunia, Institute for Biocomputation and Physic of Complex Systems, Universidad de Zaragoza, 50100 La Almunia de Doña Godina, Spain
- Department of Urology, Miguel Servet University Hospital, 50009 Zaragoza, Spain
- Area of Urology, Department of Surgery, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| |
Collapse
|
15
|
Ma M, Yu Q, Delafield DG, Cui Y, Li Z, Li M, Wu W, Shi X, Westmark PR, Gutierrez A, Ma G, Gao A, Xu M, Xu W, Westmark CJ, Li L. On-Tissue Spatial Proteomics Integrating MALDI-MS Imaging with Shotgun Proteomics Reveals Soy Consumption-Induced Protein Changes in a Fragile X Syndrome Mouse Model. ACS Chem Neurosci 2024; 15:119-133. [PMID: 38109073 PMCID: PMC11127747 DOI: 10.1021/acschemneuro.3c00497] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023] Open
Abstract
Fragile X syndrome (FXS), the leading cause of inherited intellectual disability and autism, is caused by the transcriptional silencing of the FMR1 gene, which encodes the fragile X messenger ribonucleoprotein (FMRP). FMRP interacts with numerous brain mRNAs that are involved in synaptic plasticity and implicated in autism spectrum disorders. Our published studies indicate that single-source, soy-based diets are associated with increased seizures and autism. Thus, there is an acute need for an unbiased protein marker identification in FXS in response to soy consumption. Herein, we present a spatial proteomics approach integrating mass spectrometry imaging with label-free proteomics in the FXS mouse model to map the spatial distribution and quantify levels of proteins in the hippocampus and hypothalamus brain regions. In total, 1250 unique peptides were spatially resolved, demonstrating the diverse array of peptidomes present in the tissue slices and the broad coverage of the strategy. A group of proteins that are known to be involved in glycolysis, synaptic transmission, and coexpression network analysis suggest a significant association between soy proteins and metabolic and synaptic processes in the Fmr1KO brain. Ultimately, this spatial proteomics work represents a crucial step toward identifying potential candidate protein markers and novel therapeutic targets for FXS.
Collapse
Affiliation(s)
- Min Ma
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Qinying Yu
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Daniel G. Delafield
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Yusi Cui
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Zihui Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Miyang Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Wenxin Wu
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Xudong Shi
- Division of Otolaryngology, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Pamela R. Westmark
- Department of Neurology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Alejandra Gutierrez
- Department of Neurology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
- Molecular Environmental Toxicology Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Gui Ma
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Ang Gao
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Meng Xu
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Wei Xu
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Cara J. Westmark
- Department of Neurology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
- Molecular Environmental Toxicology Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States
- Lachman Institute for Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
16
|
Zhang Y, Wu MJ, Lu WC, Li YC, Chang CJ, Yang JY. Metabolic switch regulates lineage plasticity and induces synthetic lethality in triple-negative breast cancer. Cell Metab 2024; 36:193-208.e8. [PMID: 38171333 DOI: 10.1016/j.cmet.2023.12.003] [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: 01/31/2023] [Revised: 06/23/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024]
Abstract
Metabolic reprogramming is key for cancer development, yet the mechanism that sustains triple-negative breast cancer (TNBC) cell growth despite deficient pyruvate kinase M2 (PKM2) and tumor glycolysis remains to be determined. Here, we find that deficiency in tumor glycolysis activates a metabolic switch from glycolysis to fatty acid β-oxidation (FAO) to fuel TNBC growth. We show that, in TNBC cells, PKM2 directly interacts with histone methyltransferase EZH2 to coordinately mediate epigenetic silencing of a carnitine transporter, SLC16A9. Inhibition of PKM2 leads to impaired EZH2 recruitment to SLC16A9, and in turn de-represses SLC16A9 expression to increase intracellular carnitine influx, programming TNBC cells to an FAO-dependent and luminal-like cell state. Together, these findings reveal a new metabolic switch that drives TNBC from a metabolically heterogeneous-lineage plastic cell state to an FAO-dependent-lineage committed cell state, where dual targeting of EZH2 and FAO induces potent synthetic lethality in TNBC.
Collapse
Affiliation(s)
- Yingsheng Zhang
- Department of Medicine and Biological Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA 90048, USA.
| | - Meng-Ju Wu
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Departments of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Wan-Chi Lu
- Institute of Biochemistry and Molecular Biology, China Medical University, Taichung 406040, Taiwan
| | - Yi-Chuan Li
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung 406040, Taiwan; Department of Biological Science and Technology, China Medical University, Taichung 406040, Taiwan
| | - Chun Ju Chang
- Institute of Biochemistry and Molecular Biology, China Medical University, Taichung 406040, Taiwan; Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung 406040, Taiwan.
| | - Jer-Yen Yang
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung 406040, Taiwan; Department of Biological Science and Technology, China Medical University, Taichung 406040, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406040, Taiwan.
| |
Collapse
|
17
|
Guo L, Wang L, Qin G, Zhang J, Peng J, Li L, Chen X, Wang D, Qiu J, Wang E. M-type pyruvate kinase 2 (PKM2) tetramerization alleviates the progression of right ventricle failure by regulating oxidative stress and mitochondrial dynamics. J Transl Med 2023; 21:888. [PMID: 38062516 PMCID: PMC10702013 DOI: 10.1186/s12967-023-04780-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Right ventricle failure (RVF) is a progressive heart disease that has yet to be fully understood at the molecular level. Elevated M-type pyruvate kinase 2 (PKM2) tetramerization alleviates heart failure, but detailed molecular mechanisms remain unclear. OBJECTIVE We observed changes in PKM2 tetramerization levels during the progression of right heart failure and in vitro cardiomyocyte hypertrophy and explored the causal relationship between altered PKM2 tetramerization and the imbalance of redox homeostasis in cardiomyocytes, as well as its underlying mechanisms. Ultimately, our goal was to propose rational intervention strategies for the treatment of RVF. METHOD We established RVF in Sprague Dawley (SD) rats by intraperitoneal injection of monocrotaline (MCT). The pulmonary artery pressure and right heart function of rats were assessed using transthoracic echocardiography combined with right heart catheterization. TEPP-46 was used both in vivo and in vitro to promote PKM2 tetramerization. RESULTS We observed that oxidative stress and mitochondrial disorganization were associated with increased apoptosis in the right ventricular tissue of RVF rats. Quantitative proteomics revealed that PKM2 was upregulated during RVF and negatively correlated with the cardiac function. Facilitating PKM2 tetramerization promoted mitochondrial network formation and alleviated oxidative stress and apoptosis during cardiomyocyte hypertrophy. Moreover, enhancing PKM2 tetramer formation improved cardiac mitochondrial morphology, mitigated oxidative stress and alleviated heart failure. CONCLUSION Disruption of PKM2 tetramerization contributed to RVF by inducing mitochondrial fragmentation, accumulating ROS, and finally promoted the progression of cardiomyocyte apoptosis. Facilitating PKM2 tetramerization holds potential as a promising therapeutic approach for RVF.
Collapse
Affiliation(s)
- Lizhe Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Lu Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Gang Qin
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Junjie Zhang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Jin Peng
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Longyan Li
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiang Chen
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Dandan Wang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, China
| | - Jian Qiu
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, China.
- Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.
| | - E Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, China.
| |
Collapse
|
18
|
Liu K, Hu S, Qiu L, Wang M, Zhang Z, Sun G, Zhang Y. Nrf1 is not a direct target gene of SREBP1, albeit both are integrated into the rapamycin-responsive regulatory network in human hepatoma cells. PLoS One 2023; 18:e0294508. [PMID: 38011090 PMCID: PMC10681226 DOI: 10.1371/journal.pone.0294508] [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: 02/14/2023] [Accepted: 11/02/2023] [Indexed: 11/29/2023] Open
Abstract
The essential role of protein degradation by ubiquitin-proteasome system is exerted primarily for maintaining cellular protein homeostasis. The transcriptional activation of proteasomal genes by mTORC1 signaling depends on Nrf1, but whether this process is directly via SREBP1 remains elusive. In this study, our experiment evidence revealed that Nrf1 is not a direct target of SREBP1, although both are involved in the rapamycin-responsive regulatory networks. Closely scrutinizing two distinct transcriptomic datasets unraveled no significant changes in transcriptional expression of Nrf1 and almost all proteasomal subunits in either siSREBP2-silencing cells or SREBP1-∕-MEFs, when compared to equivalent controls. However, distinct upstream signaling to Nrf1 dislocation by p97 and its processing by DDI1/2, along with downstream proteasomal expression, may be monitored by mTOR signaling, to various certain extents, depending on distinct experimental settings in different types of cells. Our further evidence has been obtained from DDI1-∕-(DDI2insC) cells, demonstrating that putative effects of mTOR on the rapamycin-responsive signaling to Nrf1 and proteasomes may also be executed partially through a DDI1/2-independent mechanism, albeit the detailed regulatory events remain to be determined.
Collapse
Affiliation(s)
- Keli Liu
- Bioengineering College, Chongqing University, Shapingba District, Chongqing, China
- Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, Jiangjin District, Chongqing, China
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, Shapingba District, Chongqing, China
| | - Shaofan Hu
- Bioengineering College, Chongqing University, Shapingba District, Chongqing, China
- Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, Jiangjin District, Chongqing, China
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, Shapingba District, Chongqing, China
| | - Lu Qiu
- Bioengineering College, Chongqing University, Shapingba District, Chongqing, China
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, Shapingba District, Chongqing, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Meng Wang
- Bioengineering College, Chongqing University, Shapingba District, Chongqing, China
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, Shapingba District, Chongqing, China
| | - Zhengwen Zhang
- Laboratory of Neuroscience, Institute of Cognitive Neuroscience and School of Pharmacy, University College London, London, England, United Kingdom
| | - Guiyin Sun
- Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, Jiangjin District, Chongqing, China
| | - Yiguo Zhang
- Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, Jiangjin District, Chongqing, China
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, Shapingba District, Chongqing, China
| |
Collapse
|
19
|
Bobotis BC, Braniff O, Gargus M, Akinluyi ET, Awogbindin IO, Tremblay MÈ. Sex differences of microglia in the healthy brain from embryonic development to adulthood and across lifestyle influences. Brain Res Bull 2023; 202:110752. [PMID: 37652267 DOI: 10.1016/j.brainresbull.2023.110752] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/15/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
Microglia, the central nervous system innate immune cells, play a critical role in maintaining a homeostatic environment in the brain throughout life. These cells exhibit an impressive range of functions and characteristics that help to ensure proper functioning of the brain. Notably, microglia can present differences in their genetic and physical traits, which can be influenced by a range of factors, including age, environmental exposures, disease, and sex. Remarkably, microglia have been found to express receptors for sex hormones, suggesting that these hormones may play a role in modulating microglial behavior and potentially contribute to sex differences. Additionally, sex-chromosomal factors were shown to impact microglial genetics and functioning. In this review, we will examine how microglial responses in homeostasis are impacted by their interaction with sex hormones and sex chromosomes. Specifically, our investigation will focus on examining this interaction from embryonic development to adulthood, and the influence of lifestyle elements on various microglial features, including density and distribution, morphology, transcriptome, and proteome.
Collapse
Affiliation(s)
| | - Olivia Braniff
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Makenna Gargus
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Elizabeth Toyin Akinluyi
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Department of Pharmacology and Therapeutics, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Ifeoluwa Oluleke Awogbindin
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Neuroimmunology Group, Molecular Drug Metabolism and Toxicology Laboratory, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Neurosciences Axis, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada; Department of Molecular Medicine, Université Laval, Québec, QC, Canada; Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada.
| |
Collapse
|
20
|
Zhang H, Wang J, Yang J, He Q, Guan S, Qiao M, Zhao J, Wang X. TMEM33 as a Prognostic Biomarker of Cervical Cancer and Its Correlation with Immune Infiltration. Mediators Inflamm 2023; 2023:5542181. [PMID: 37273452 PMCID: PMC10239303 DOI: 10.1155/2023/5542181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/12/2023] [Accepted: 05/15/2023] [Indexed: 06/06/2023] Open
Abstract
In women all over the world, cervical cancer (CC) ranks as the fourth most common form of cancer to be diagnosed. It was previously reported that transmembrane protein 33(TMEM33) could report a poor prognosis in several cancers. The current study is aimed at investigating the potential prognostic value of TMEM33 and its relevance to the tumor microenvironment in CC in a comprehensive manner. In this study, CC specimens presented noticeably higher TMEM33 expression level in comparison to nontumor specimens. In pan-cancer assays, it was found that TMEM33 was present at a high level in many different kinds of tumors. We found that patients with CC patients who had a high TMEM33 expression presented worse overall survival (OS) and disease-free survival (DFS) relative to patients who had a low TMEM33 expression. According to the results of a multivariate analysis, a high level of TMEM33 expression can significantly and independently predict the prognosis of CC. The levels of TMEM33 were found to have a negative correlation with resting dendritic cells, resting mast cells, plasma cells, T cells CD8, T cells regulatory, and regulatory T cells. Finally, we confirmed that TMEM33 was overexpressed in CC cells, and its knockdown distinctly suppressed the proliferation and invasion of CC cells. Overall, we provided evidences that TMEM33 could be used as a potential biomarker to assess the prognosis and the level of immune infiltration in CC.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Ultrasound, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, China
| | - Jun Wang
- Department of Ultrasound, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, China
| | - Ji Yang
- Department of Ultrasound, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, China
| | - Qingwen He
- Department of Ultrasound, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, China
| | - Sanli Guan
- Department of Ultrasound, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, China
| | - Minxia Qiao
- Department of Ultrasound, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, China
| | - Jian Zhao
- Department of Ultrasound, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, China
| | - Xiu Wang
- Department of Ultrasound, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, China
| |
Collapse
|
21
|
Li Y, Wu S, Zhao X, Hao S, Li F, Wang Y, Liu B, Zhang D, Wang Y, Zhou H. Key events in cancer: Dysregulation of SREBPs. Front Pharmacol 2023; 14:1130747. [PMID: 36969840 PMCID: PMC10030587 DOI: 10.3389/fphar.2023.1130747] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
Lipid metabolism reprogramming is an important hallmark of tumor progression. Cancer cells require high levels of lipid synthesis and uptake not only to support their continued replication, invasion, metastasis, and survival but also to participate in the formation of biological membranes and signaling molecules. Sterol regulatory element binding proteins (SREBPs) are core transcription factors that control lipid metabolism and the expression of important genes for lipid synthesis and uptake. A growing number of studies have shown that SREBPs are significantly upregulated in human cancers and serve as intermediaries providing a mechanistic link between lipid metabolism reprogramming and malignancy. Different subcellular localizations, including endoplasmic reticulum, Golgi, and nucleus, play an indispensable role in regulating the cleavage maturation and activity of SREBPs. In this review, we focus on the relationship between aberrant regulation of SREBPs activity in three organelles and tumor progression. Because blocking the regulation of lipid synthesis by SREBPs has gradually become an important part of tumor therapy, this review also summarizes and analyzes several current mainstream strategies.
Collapse
Affiliation(s)
- Yunkuo Li
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Shouwang Wu
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Xiaodong Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Shiming Hao
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Faping Li
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Yuxiong Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Bin Liu
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Difei Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
- *Correspondence: Yishu Wang, Honglan Zhou,
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Yishu Wang, Honglan Zhou,
| |
Collapse
|
22
|
Zhang Q, Zhang J, Yao A, Tian X, Han Z, Yuan Y, Tao K, Yang X. OTUB2 promotes the progression of endometrial cancer by regulating the PKM2-mediated PI3K/AKT signaling pathway. Cell Biol Int 2023; 47:428-438. [PMID: 36316812 DOI: 10.1002/cbin.11950] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/27/2022] [Accepted: 10/01/2022] [Indexed: 11/07/2022]
Abstract
Endometrial carcinoma (EC) morbidity and mortality have been increasing in recent years. Otubain 2 (OTUB2) was shown to be upregulated in EC patients, so the aim of this study was to explore the role of OTUB2 in EC. Cell Counting Kit-8 (CCK-8), colony formation, enzyme-linked immunosorbent assay, the wound healing assay, and Transwell invasion assays were used to investigate the specific role of OTUB2 in EC tumorigenesis. Real-time polymerase chain reaction and western blot analysis were used to detect the expression of OTUB2 in EC tissues and cells. OTUB2 is upregulated in EC patients and cell lines and is associated with a poor prognosis. The overexpression of OTUB2 promoted glycolysis and induced the proliferation, migration, and invasion of endometrial cancer cells. The silencing of OTUB2 had the opposite effect. In addition, the silencing of OTUB2 significantly suppressed the expression levels of PKM2. Importantly, inhibition of the PKM2/PI3K/AKT signaling pathway significantly reversed the promoting effect of OTUB2 overexpression on EC. OTUB2 regulated the proliferation and invasion of EC cells by regulating the PKM2/PI3K/AKT signaling pathway. OTUB2 may serve as a potential prognostic and therapeutic target in EC.
Collapse
Affiliation(s)
- Qian Zhang
- Department of The First of Internal Medicine, Shaanxi Provincial Cancer Hospital, Xi'an, Shannxi, People's Republic of China
| | - Jing Zhang
- Department of The Fourth of Gynecologic Oncology, Shaanxi Provincial Cancer Hospital, Xi'an, Shannxi, People's Republic of China
| | - Anmei Yao
- Department of The Second of Gynecologic Oncology, Shaanxi Provincial Cancer Hospital, Xi'an, Shannxi, People's Republic of China
| | - Xiaofei Tian
- Department of The Second of Gynecologic Oncology, Shaanxi Provincial Cancer Hospital, Xi'an, Shannxi, People's Republic of China
| | - Zhihong Han
- Department of The Second of Gynecologic Oncology, Shaanxi Provincial Cancer Hospital, Xi'an, Shannxi, People's Republic of China
| | - Yuan Yuan
- Department of The Second of Gynecologic Oncology, Shaanxi Provincial Cancer Hospital, Xi'an, Shannxi, People's Republic of China
| | - Kai Tao
- Department of The Second of Gynecologic Oncology, Shaanxi Provincial Cancer Hospital, Xi'an, Shannxi, People's Republic of China
| | - Xuemei Yang
- Department of The Second of Gynecologic Oncology, Shaanxi Provincial Cancer Hospital, Xi'an, Shannxi, People's Republic of China
| |
Collapse
|
23
|
Tsai PL, Cameron CJF, Forni MF, Wasko RR, Naughton BS, Horsley V, Gerstein MB, Schlieker C. Dynamic quality control machinery that operates across compartmental borders mediates the degradation of mammalian nuclear membrane proteins. Cell Rep 2022; 41:111675. [PMID: 36417855 PMCID: PMC9827541 DOI: 10.1016/j.celrep.2022.111675] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/15/2022] [Accepted: 10/26/2022] [Indexed: 11/23/2022] Open
Abstract
Many human diseases are caused by mutations in nuclear envelope (NE) proteins. How protein homeostasis and disease etiology are interconnected at the NE is poorly understood. Specifically, the identity of local ubiquitin ligases that facilitate ubiquitin-proteasome-dependent NE protein turnover is presently unknown. Here, we employ a short-lived, Lamin B receptor disease variant as a model substrate in a genetic screen to uncover key elements of NE protein turnover. We identify the ubiquitin-conjugating enzymes (E2s) Ube2G2 and Ube2D3, the membrane-resident ubiquitin ligases (E3s) RNF5 and HRD1, and the poorly understood protein TMEM33. RNF5, but not HRD1, requires TMEM33 both for efficient biosynthesis and function. Once synthesized, RNF5 responds dynamically to increased substrate levels at the NE by departing from the endoplasmic reticulum, where HRD1 remains confined. Thus, mammalian protein quality control machinery partitions between distinct cellular compartments to address locally changing substrate loads, establishing a robust cellular quality control system.
Collapse
Affiliation(s)
- Pei-Ling Tsai
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Christopher J F Cameron
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA; Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Maria Fernanda Forni
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA
| | - Renee R Wasko
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA
| | - Brigitte S Naughton
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Valerie Horsley
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA
| | - Mark B Gerstein
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA; Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06511, USA; Department of Computer Science, Yale University, New Haven, CT 06511, USA; Department of Statistics and Data Science, Yale University, New Haven, CT 06511, USA
| | - Christian Schlieker
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA; Department of Cell Biology, Yale School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
24
|
Chen H, Zhao X, Li Y, Zhang S, Wang Y, Wang L, Ma W. High Expression of TMEM33 Predicts Poor Prognosis and Promotes Cell Proliferation in Cervical Cancer. Front Genet 2022; 13:908807. [PMID: 35832191 PMCID: PMC9271802 DOI: 10.3389/fgene.2022.908807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/07/2022] [Indexed: 01/22/2023] Open
Abstract
Background: The prognosis of patients with advanced cervical cancer remains unsatisfactory. A study indicated that transmembrane protein 33 (TMEM33) was implicated in tumor recurrence, while its role in cervical cancer has not been elucidated. Methods: TMEM33 expression in cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC) was primarily screened in The Cancer Genome Atlas (TCGA), and further validated in Gene Expression Omnibus (GEO) database. The Kaplan–Meier plotter analysis and Cox regression were constructed to evaluate the prognostic value of TMEM33 in CESC. Functional enrichment analysis was performed with GO, KEGG and GSEA tools. CCK-8 assay and colony formation assay were performed to investigate the carcinogenesis role of TMEM33 in cervical cancer cell proliferation. Results: TMEM33 expression was significantly elevated in CESC compared with normal tissues. High expression of TMEM33 was associated with poor prognostic clinical characteristics in CESC patients. KM-plotter analysis revealed that patients with increased TMEM33 had shorter overall survival (OS), progress free interval (PFI), and disease specific survival (DSS). Moreover, Multivariate Cox analysis confirmed that high TMEM33 expression was an independent risk factor for OS in patients with CESC. TMEM33 was associated with immune infiltrates, and its expression was correlated with tumorigenesis-related genes RNF4, OCIAD1, TMED5, DHX15, MED28 and LETM1. More importantly, knockdown of TMEM33 in cervical cancer cells decreased the expression of those genes and inhibited cell proliferation. Conclusion: Increased TMEM33 in cervical cancer can serve as an independent prognostic marker and might play a role in tumorigenesis by promoting cell proliferation.
Collapse
Affiliation(s)
- Hanxiang Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong LaiBo Biotechnology Co., Ltd., Jinan, China
| | - Xia Zhao
- Department of Clinical Laboratory, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Yongqing Li
- Department of Clinical Laboratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | | | - Yunshan Wang
- Medical Research and Laboratory Diagnostic Center, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lili Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- *Correspondence: Lili Wang, ; Wanshan Ma,
| | - Wanshan Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- *Correspondence: Lili Wang, ; Wanshan Ma,
| |
Collapse
|
25
|
Yang Y, Li M, Ma Y, Ye W, Si Y, Zheng X, Liu H, Cheng L, Zhang L, Zhang H, Zhang X, Lei Y, Shen L, Zhang F, Ma H. LncRNA NEAT1 Potentiates SREBP2 Activity to Promote Inflammatory Macrophage Activation and Limit Hantaan Virus Propagation. Front Microbiol 2022; 13:849020. [PMID: 35495674 PMCID: PMC9044491 DOI: 10.3389/fmicb.2022.849020] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/10/2022] [Indexed: 11/17/2022] Open
Abstract
As the global prototypical zoonotic hantavirus, Hantaan virus (HTNV) is prevalent in Asia and is the leading causative agent of severe hemorrhagic fever with renal syndrome (HFRS), which has profound morbidity and mortality. Macrophages are crucial components of the host innate immune system and serve as the first line of defense against HTNV infection. Previous studies indicated that the viral replication efficiency in macrophages determines hantavirus pathogenicity, but it remains unknown which factor manipulates the macrophage activation pattern and the virus-host interaction process. Here, we performed the transcriptomic analysis of HTNV-infected mouse bone marrow-derived macrophages and identified the long noncoding RNA (lncRNA) nuclear enriched abundant transcript 1 (NEAT1), especially the isoform NEAT1-2, as one of the lncRNAs that is differentially expressed at the early phase. Based on coculture experiments, we revealed that silencing NEAT1-2 hinders inflammatory macrophage activation and facilitates HTNV propagation, while enhancing NEAT1-2 transcription effectively restrains viral replication. Furthermore, sterol response element binding factor-2 (SREBP2), which controls the cholesterol metabolism process, was found to stimulate macrophages by promoting the production of multiple inflammatory cytokines upon HTNV infection. NEAT1-2 could potentiate SREBP2 activity by upregulating Srebf1 expression and interacting with SREBP2, thus stimulating inflammatory macrophages and limiting HTNV propagation. More importantly, we demonstrated that the NEAT1-2 expression level in patient monocytes was negatively correlated with viral load and HFRS disease progression. Our results identified a function and mechanism of action for the lncRNA NEAT1 in heightening SREBP2-mediated macrophage activation to restrain hantaviral propagation and revealed the association of NEAT1 with HFRS severity.
Collapse
Affiliation(s)
- Yongheng Yang
- College of Life Sciences, Northwest University, Xi'an, China.,Department of Microbiology, School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Mengyun Li
- College of Life Sciences, Northwest University, Xi'an, China.,Department of Microbiology, School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Yongtao Ma
- Department of Emergency, Children's Hospital of Kaifeng City, Kaifeng, China
| | - Wei Ye
- Department of Microbiology, School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Yue Si
- Department of Microbiology, School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Xuyang Zheng
- Department of Microbiology, School of Basic Medicine, The Fourth Military Medical University, Xi'an, China.,Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - He Liu
- Department of Microbiology, School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Linfeng Cheng
- Department of Microbiology, School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Liang Zhang
- Department of Microbiology, School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Hui Zhang
- Department of Microbiology, School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Xijing Zhang
- Department of Anesthesiology and Critical Care Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yingfeng Lei
- Department of Microbiology, School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Lixin Shen
- College of Life Sciences, Northwest University, Xi'an, China
| | - Fanglin Zhang
- Department of Microbiology, School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Hongwei Ma
- Department of Microbiology, School of Basic Medicine, The Fourth Military Medical University, Xi'an, China.,Department of Anesthesiology and Critical Care Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
26
|
Khateb A, Ronai ZA. Lipid metabolism: new twists to the Yin and Yang of PKM2 in cancer. EMBO J 2021; 40:e109683. [PMID: 34642948 DOI: 10.15252/embj.2021109683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 11/09/2022] Open
Abstract
While canonical and non-canonical functions of pyruvate kinase M2 (PKM2) are recognized to mediate often-opposing roles in cancer, its contribution to cellular and systemic fatty acid homeostasis remains poorly understood. A new study by Liu et al (2021) uncovers ER transmembrane protein TMEM33 as a novel target of PKM2, which is essential for regulation of cancer cell cholesterol metabolism. These findings highlight the diversity of tissue-specific functions of PKM2 and potential implications for cancer treatment.
Collapse
Affiliation(s)
- Ali Khateb
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Ze'ev A Ronai
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|