1
|
Xiang Y, Liu X, Sun Q, Liao K, Liu X, Zhao Z, Feng L, Liu Y, Wang B. The development of cancers research based on mitochondrial heat shock protein 90. Front Oncol 2023; 13:1296456. [PMID: 38098505 PMCID: PMC10720920 DOI: 10.3389/fonc.2023.1296456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/23/2023] [Indexed: 12/17/2023] Open
Abstract
Mitochondrial heat shock protein 90 (mtHsp90), including Tumor necrosis factor receptor-associated protein 1 (TRAP1) and Hsp90 translocated from cytoplasm, modulating cellular metabolism and signaling pathways by altering the conformation, activity, and stability of numerous client proteins, and is highly expressed in tumors. mtHsp90 inhibition results in the destabilization and eventual degradation of its client proteins, leading to interference with various tumor-related pathways and efficient control of cancer cell development. Among these compounds, gamitrinib, a specific mtHsp90 inhibitor, has demonstrated its safety and efficacy in several preclinical investigations and is currently undergoing evaluation in clinical trials. This review aims to provide a comprehensive overview of the present knowledge pertaining to mtHsp90, encompassing its structure and function. Moreover, our main emphasis is on the development of mtHsp90 inhibitors for various cancer therapies, to present a thorough overview of the recent pre-clinical and clinical advancements in this field.
Collapse
Affiliation(s)
- Yuchu Xiang
- West China Hospital of Sichuan University, Sichuan University, Chengdu, China
| | - Xudong Liu
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Qi Sun
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, Centre for Safe Medication Practice and Research, The University of Hong Kong, Pok Fu Lam, Hong Kong SAR, China
| | - Kuo Liao
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Xiaohan Liu
- Multiscale Research Institute of Complex Systems, Fudan University, Shanghai, China
| | - Zihui Zhao
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lishuang Feng
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yan Liu
- Department of Organ Transplantation, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Bo Wang
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Urology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| |
Collapse
|
2
|
Shen L, Chen YL, Huang CC, Shyu YC, Seftor REB, Seftor EA, Hendrix MJC, Chien DS, Chu YW. CVM-1118 (foslinanib), a 2-phenyl-4-quinolone derivative, promotes apoptosis and inhibits vasculogenic mimicry via targeting TRAP1. Pathol Oncol Res 2023; 29:1611038. [PMID: 37351538 PMCID: PMC10283505 DOI: 10.3389/pore.2023.1611038] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 05/19/2023] [Indexed: 06/24/2023]
Abstract
CVM-1118 (foslinanib) is a phosphoric ester compound selected from 2-phenyl-4-quinolone derivatives. The NCI 60 cancer panel screening showed CVM-1125, the major active metabolite of CVM-1118, to exhibit growth inhibitory and cytotoxic effects at nanomolar range. CVM-1118 possesses multiple bioactivities, including inducing cellular apoptosis, cell cycle arrest at G2/M, as well as inhibiting vasculogenic mimicry (VM) formation. The TNF receptor associated protein 1 (TRAP1) was identified as the binding target of CVM-1125 using nematic protein organization technique (NPOT) interactome analysis. Further studies demonstrated CVM-1125 reduced the protein level of TRAP1 and impeded its downstream signaling by reduction of cellular succinate levels and destabilization of HIF-1α. The pharmacogenomic biomarkers associated with CVM-1118 were also examined by Whole Genome CRISPR Knock-Out Screening. Two hits (STK11 and NF2) were confirmed with higher sensitivity to the drug in cell knock-down experiments. Biological assays indicate that the mechanism of action of CVM-1118 is via targeting TRAP1 to induce mitochondrial apoptosis, suppress tumor cell growth, and inhibit vasculogenic mimicry formation. Most importantly, the loss-of-function mutations of STK11 and NF2 are potential biomarkers of CVM-1118 which can be applied in the selection of cancer patients for CVM-1118 treatment. CVM-1118 is currently in its Phase 2a clinical development.
Collapse
Affiliation(s)
| | | | | | - Yu-Chiau Shyu
- Community Medicine Research Center, Chang Gung Memorial Hospital Keelung Branch, Keelung, Taiwan
- Department of Nursing, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | | | - Elisabeth A. Seftor
- Department of Biology, Shepherd University, Shepherdstown, WV, United States
| | - Mary J. C. Hendrix
- Department of Biology, Shepherd University, Shepherdstown, WV, United States
| | | | | |
Collapse
|
3
|
Endometriosis Stem Cells as a Possible Main Target for Carcinogenesis of Endometriosis-Associated Ovarian Cancer (EAOC). Cancers (Basel) 2022; 15:cancers15010111. [PMID: 36612107 PMCID: PMC9817684 DOI: 10.3390/cancers15010111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Endometriosis is a serious recurrent disease impairing the quality of life and fertility, and being a risk for some histologic types of ovarian cancer defined as endometriosis-associated ovarian cancers (EAOC). The presence of stem cells in the endometriotic foci could account for the proliferative, migrative and angiogenic activity of the lesions. Their phenotype and sources have been described. The similarly disturbed expression of several genes, miRNAs, galectins and chaperones has been observed both in endometriotic lesions and in ovarian or endometrial cancer. The importance of stem cells for nascence and sustain of malignant tumors is commonly appreciated. Although the proposed mechanisms promoting carcinogenesis leading from endometriosis into the EAOC are not completely known, they have been discussed in several articles. However, the role of endometriosis stem cells (ESCs) has not been discussed in this context. Here, we postulate that ESCs may be a main target for the carcinogenesis of EAOC and present the possible sequence of events resulting finally in the development of EAOC.
Collapse
|
4
|
Ferreira-Gonzalez S, Man TY, Esser H, Aird R, Kilpatrick AM, Rodrigo-Torres D, Younger N, Campana L, Gadd VL, Dwyer B, Aleksieva N, Boulter L, Macmillan MT, Wang Y, Mylonas KJ, Ferenbach DA, Kendall TJ, Lu WY, Acosta JC, Kurian D, O'Neill S, Oniscu GC, Banales JM, Krimpenfort PJ, Forbes SJ. Senolytic treatment preserves biliary regenerative capacity lost through cellular senescence during cold storage. Sci Transl Med 2022; 14:eabj4375. [PMID: 36475903 DOI: 10.1126/scitranslmed.abj4375] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Liver transplantation is the only curative option for patients with end-stage liver disease. Despite improvements in surgical techniques, nonanastomotic strictures (characterized by the progressive loss of biliary tract architecture) continue to occur after liver transplantation, negatively affecting liver function and frequently leading to graft loss and retransplantation. To study the biological effects of organ preservation before liver transplantation, we generated murine models that recapitulate liver procurement and static cold storage. In these models, we explored the response of cholangiocytes and hepatocytes to cold storage, focusing on responses that affect liver regeneration, including DNA damage, apoptosis, and cellular senescence. We show that biliary senescence was induced during organ retrieval and exacerbated during static cold storage, resulting in impaired biliary regeneration. We identified decoy receptor 2 (DCR2)-dependent responses in cholangiocytes and hepatocytes, which differentially affected the outcome of those populations during cold storage. Moreover, CRISPR-mediated DCR2 knockdown in vitro increased cholangiocyte proliferation and decreased cellular senescence but had the opposite effect in hepatocytes. Using the p21KO model to inhibit senescence onset, we showed that biliary tract architecture was better preserved during cold storage. Similar results were achieved by administering senolytic ABT737 to mice before procurement. Last, we perfused senolytics into discarded human donor livers and showed that biliary architecture and regenerative capacities were better preserved. Our results indicate that cholangiocytes are susceptible to senescence and identify the use of senolytics and the combination of senotherapies and machine-perfusion preservation to prevent this phenotype and reduce the incidence of biliary injury after transplantation.
Collapse
Affiliation(s)
- Sofia Ferreira-Gonzalez
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Tak Yung Man
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Hannah Esser
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
- Department of Visceral, Transplant and Thoracic Surgery, Centre of Operative Medicine, Innsbruck Medical University, Anichstrasse 35, Innsbruck 6020, Austria
| | - Rhona Aird
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Alastair M Kilpatrick
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Daniel Rodrigo-Torres
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Nicholas Younger
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Lara Campana
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Victoria L Gadd
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Benjamin Dwyer
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Niya Aleksieva
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Luke Boulter
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Mark T Macmillan
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Yinmiao Wang
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Katie J Mylonas
- Centre for Inflammation Research (CIR), University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - David A Ferenbach
- Centre for Inflammation Research (CIR), University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Timothy J Kendall
- Centre for Inflammation Research (CIR), University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Wei-Yu Lu
- Centre for Inflammation Research (CIR), University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Juan Carlos Acosta
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Cancer, University of Edinburgh, Crewe Road, Edinburgh EH4 2XR, UK
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria-SODERCAN, C/ Albert Einstein 22, Santander, 39011, Spain
| | - Dominic Kurian
- Proteomic and Metabolomics Unit, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Stephen O'Neill
- Department of Transplant Surgery, Belfast City Hospital, 51 Lisburn Road, Belfast BT9 7AB, UK
- Centre for Public Health, Queen's University Belfast, Institute of Clinical Science, Block A, Royal Victoria Hospital, Belfast BT12 6BA, UK
| | - Gabriel C Oniscu
- Edinburgh Transplant Centre, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh EH16 4SA, UK
- Department of Clinical Surgery, University of Edinburgh, 51 Little France Crescent, Edinburgh EH16 4SA, UK
| | - Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBERehd, Ikerbasque, San Sebastian 20014, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31009 Pamplona, Spain
| | | | - Stuart J Forbes
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| |
Collapse
|
5
|
Dabravolski SA, Sukhorukov VN, Kalmykov VA, Orekhov NA, Grechko AV, Orekhov AN. Heat Shock Protein 90 as Therapeutic Target for CVDs and Heart Ageing. Int J Mol Sci 2022; 23:ijms23020649. [PMID: 35054835 PMCID: PMC8775949 DOI: 10.3390/ijms23020649] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death globally, representing approximately 32% of all deaths worldwide. Molecular chaperones are involved in heart protection against stresses and age-mediated accumulation of toxic misfolded proteins by regulation of the protein synthesis/degradation balance and refolding of misfolded proteins, thus supporting the high metabolic demand of the heart cells. Heat shock protein 90 (HSP90) is one of the main cardioprotective chaperones, represented by cytosolic HSP90a and HSP90b, mitochondrial TRAP1 and ER-localised Grp94 isoforms. Currently, the main way to study the functional role of HSPs is the application of HSP inhibitors, which could have a different way of action. In this review, we discussed the recently investigated role of HSP90 proteins in cardioprotection, atherosclerosis, CVDs development and the involvements of HSP90 clients in the activation of different molecular pathways and signalling mechanisms, related to heart ageing.
Collapse
Affiliation(s)
- Siarhei A. Dabravolski
- Department of Clinical Diagnostics, Vitebsk State Academy of Veterinary Medicine [UO VGAVM], 7/11 Dovatora Str., 210026 Vitebsk, Belarus
- Correspondence:
| | - Vasily N. Sukhorukov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, AP Avtsyn Research Institute of Human Morphology, 3 Tsyurupy Str., 117418 Moscow, Russia; (V.N.S.); (V.A.K.)
- Laboratory of Medical Genetics, Russian Medical Research Center of Cardiology, Institute of Experimental Cardiology, 15-a 3-rd Cherepkovskaya Str., 121552 Moscow, Russia
| | - Vladislav A. Kalmykov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, AP Avtsyn Research Institute of Human Morphology, 3 Tsyurupy Str., 117418 Moscow, Russia; (V.N.S.); (V.A.K.)
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
| | - Nikolay A. Orekhov
- Institute for Atherosclerosis Research, 4-1-207 Osennyaya Str., 121609 Moscow, Russia; (N.A.O.); (A.N.O.)
| | - Andrey V. Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 14-3 Solyanka Str., 109240 Moscow, Russia;
| | - Alexander N. Orekhov
- Institute for Atherosclerosis Research, 4-1-207 Osennyaya Str., 121609 Moscow, Russia; (N.A.O.); (A.N.O.)
| |
Collapse
|
6
|
Wang N, Zhu P, Huang R, Sun L, Dong D, Gao Y. Suppressing TRAP1 sensitizes glioblastoma multiforme cells to temozolomide. Exp Ther Med 2021; 22:1246. [PMID: 34539842 PMCID: PMC8438667 DOI: 10.3892/etm.2021.10681] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 03/11/2021] [Indexed: 12/11/2022] Open
Abstract
Glioma is a common malignant tumor of the central nervous system, accounting for ~50% of intracranial tumors. The current standard therapy for glioma is surgical resection followed by postoperative adjuvant radiotherapy and temozolomide (TMZ) chemotherapy. However, resistance to TMZ is one of the factors affecting prognosis. It has been reported that TNF receptor-associated protein 1 (TRAP1) is overexpressed in numerous types of tumor and that interfering with its function may abrogate chemotherapy resistance. TRAP1 inhibitor Gamitrinib triphenylphosphonium (G-TPP) and shRNA were used in the present study to suppress the function of this molecule in glioblastoma multiforme (GBM) cell lines. MTT assay was performed to evaluate the combined effect of G-TPP and TMZ treatment. To investigate the underlying mechanism responsible for this combined effect, the mitochondrial unfolded protein response (mtUPR), mitophagy, mitochondrial fusion and reactive oxygen species (ROS) were quantified using western blotting and immunofluorescence techniques. TMZ treatment induced apoptosis in GBM cells by activating the p53 pathway, whilst simultaneously downregulating mitophagy and enhancing mitochondrial fusion. The latter may occur in order to compensate for the defect caused by downregulated mitophagy. Suppressing the function of TRAP1 disturbed this compensatory mechanism by inducing mtUPR, which resulted in a burst of ROS formation and sensitized the GBM cells to the effects of TMZ treatment. Thus, suppressing the function of TRAP1 sensitized GBM cells to TMZ lysis by inducing mtUPR and the subsequent ROS burst. TRAP1 is therefore considered to be a promising target for GBM therapy.
Collapse
Affiliation(s)
- Nan Wang
- Department of Neurosurgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Peining Zhu
- Department of Neurosurgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Renxuan Huang
- Department of Neurosurgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Liankun Sun
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Delu Dong
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Yufei Gao
- Department of Neurosurgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130000, P.R. China
| |
Collapse
|
7
|
Yang S, Xiao H, Cao L. Recent advances in heat shock proteins in cancer diagnosis, prognosis, metabolism and treatment. Biomed Pharmacother 2021; 142:112074. [PMID: 34426258 DOI: 10.1016/j.biopha.2021.112074] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023] Open
Abstract
Heat shock proteins (HSPs) are a group of proteins, also known as molecular chaperones, which participate in protein folding and maturation in response to stresses or high temperature. According to their molecular weights, mammalian HSPs are classified into HSP27, HSP40, HSP60, HSP70, HSP90, and large HSPs. Previous studies have revealed that HSPs play important roles in oncogenesis and malignant progression because they can modulate all six hallmark traits of cancer. Because of this, HSPs have been propelled into the spotlight as biomarkers for cancer diagnosis and prognosis, as well as an exciting anticancer drug target. However, the relationship between the expression level of HSPs and their activity and cancer diagnosis, prognosis, metabolism and treatment is not clear and has not been completely established. Herein, this review summarizes and discusses recent advances and perspectives in major HSPs as biomarkers for cancer diagnosis, as regulators for cancer metabolism or as therapeutic targets for cancer therapy, which may provide new directions to improve the accuracy of cancer diagnosis and develop more effective and safer anticancer therapeutics.
Collapse
Affiliation(s)
- Shuxian Yang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing 100193, China; Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China
| | - Haiyan Xiao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing 100193, China; Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China
| | - Li Cao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing 100193, China; Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
| |
Collapse
|
8
|
Staňková P, Kučera O, Peterová E, Elkalaf M, Rychtrmoc D, Melek J, Podhola M, Zubáňová V, Červinková Z. Western Diet Decreases the Liver Mitochondrial Oxidative Flux of Succinate: Insight from a Murine NAFLD Model. Int J Mol Sci 2021; 22:6908. [PMID: 34199098 PMCID: PMC8268937 DOI: 10.3390/ijms22136908] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/20/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
Mitochondria play an essential role in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). Previously, we found that succinate-activated respiration was the most affected mitochondrial parameter in mice with mild NAFLD. In this study, we focused on the role of succinate dehydrogenase (SDH) in NAFLD pathogenesis. To induce the progression of NAFLD to nonalcoholic steatohepatitis (NASH), C57BL/6J mice were fed a Western-style diet (WD) or control diet for 30 weeks. NAFLD severity was evaluated histologically and the expression of selected proteins and genes was assessed. Mitochondrial respiration was measured by high-resolution respirometry. Liver redox status was assessed using glutathione, malondialdehyde, and mitochondrial production of reactive oxygen species (ROS). Metabolomic analysis was performed by GC/MS. WD consumption for 30 weeks led to reduced succinate-activated respiration. We also observed decreased SDH activity, decreased expression of the SDH activator sirtuin 3, decreased gene expression of SDH subunits, and increased levels of hepatic succinate, an important signaling molecule. Succinate receptor 1 (SUCNR1) gene and protein expression were reduced in the livers of WD-fed mice. We did not observe signs of oxidative damage compared to the control group. The changes observed in WD-fed mice appear to be adaptive to prevent mitochondrial respiratory chain overload and massive ROS production.
Collapse
Affiliation(s)
- Pavla Staňková
- Department of Physiology, Faculty of Medicine in Hradec Králové, Charles University, Šimkova 870, 500 03 Hradec Králové, Czech Republic; (P.S.); (E.P.); (M.E.); (D.R.); (J.M.); (V.Z.); (Z.Č.)
| | - Otto Kučera
- Department of Physiology, Faculty of Medicine in Hradec Králové, Charles University, Šimkova 870, 500 03 Hradec Králové, Czech Republic; (P.S.); (E.P.); (M.E.); (D.R.); (J.M.); (V.Z.); (Z.Č.)
| | - Eva Peterová
- Department of Physiology, Faculty of Medicine in Hradec Králové, Charles University, Šimkova 870, 500 03 Hradec Králové, Czech Republic; (P.S.); (E.P.); (M.E.); (D.R.); (J.M.); (V.Z.); (Z.Č.)
- Department of Medical Biochemistry, Faculty of Medicine in Hradec Králové, Charles University, Šimkova 870, 500 03 Hradec Králové, Czech Republic
| | - Moustafa Elkalaf
- Department of Physiology, Faculty of Medicine in Hradec Králové, Charles University, Šimkova 870, 500 03 Hradec Králové, Czech Republic; (P.S.); (E.P.); (M.E.); (D.R.); (J.M.); (V.Z.); (Z.Č.)
- Department of Pathophysiology, Third Faculty of Medicine, Charles University Prague, Ruská 87, 100 00 Prague, Czech Republic
| | - David Rychtrmoc
- Department of Physiology, Faculty of Medicine in Hradec Králové, Charles University, Šimkova 870, 500 03 Hradec Králové, Czech Republic; (P.S.); (E.P.); (M.E.); (D.R.); (J.M.); (V.Z.); (Z.Č.)
| | - Jan Melek
- Department of Physiology, Faculty of Medicine in Hradec Králové, Charles University, Šimkova 870, 500 03 Hradec Králové, Czech Republic; (P.S.); (E.P.); (M.E.); (D.R.); (J.M.); (V.Z.); (Z.Č.)
| | - Miroslav Podhola
- The Fingerland Department of Pathology, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, Sokolská 581, 500 05 Hradec Králové, Czech Republic;
| | - Veronika Zubáňová
- Department of Physiology, Faculty of Medicine in Hradec Králové, Charles University, Šimkova 870, 500 03 Hradec Králové, Czech Republic; (P.S.); (E.P.); (M.E.); (D.R.); (J.M.); (V.Z.); (Z.Č.)
- Department of Clinical Biochemistry and Diagnostics, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, Sokolská 581, 500 05 Hradec Králové, Czech Republic
| | - Zuzana Červinková
- Department of Physiology, Faculty of Medicine in Hradec Králové, Charles University, Šimkova 870, 500 03 Hradec Králové, Czech Republic; (P.S.); (E.P.); (M.E.); (D.R.); (J.M.); (V.Z.); (Z.Č.)
| |
Collapse
|
9
|
Kielbik M, Szulc-Kielbik I, Klink M. Calreticulin-Multifunctional Chaperone in Immunogenic Cell Death: Potential Significance as a Prognostic Biomarker in Ovarian Cancer Patients. Cells 2021; 10:130. [PMID: 33440842 PMCID: PMC7827772 DOI: 10.3390/cells10010130] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 12/11/2022] Open
Abstract
Immunogenic cell death (ICD) is a type of death, which has the hallmarks of necroptosis and apoptosis, and is best characterized in malignant diseases. Chemotherapeutics, radiotherapy and photodynamic therapy induce intracellular stress response pathways in tumor cells, leading to a secretion of various factors belonging to a family of damage-associated molecular patterns molecules, capable of inducing the adaptive immune response. One of them is calreticulin (CRT), an endoplasmic reticulum-associated chaperone. Its presence on the surface of dying tumor cells serves as an "eat me" signal for antigen presenting cells (APC). Engulfment of tumor cells by APCs results in the presentation of tumor's antigens to cytotoxic T-cells and production of cytokines/chemokines, which activate immune cells responsible for tumor cells killing. Thus, the development of ICD and the expression of CRT can help standard therapy to eradicate tumor cells. Here, we review the physiological functions of CRT and its involvement in the ICD appearance in malignant disease. Moreover, we also focus on the ability of various anti-cancer drugs to induce expression of surface CRT on ovarian cancer cells. The second aim of this work is to discuss and summarize the prognostic/predictive value of CRT in ovarian cancer patients.
Collapse
Affiliation(s)
- Michal Kielbik
- Institute of Medical Biology, Polish Academy of Sciences, 106 Lodowa Str., 93-232 Lodz, Poland; (I.S.-K.); (M.K.)
| | | | | |
Collapse
|
10
|
Lofaro FD, Boraldi F, Garcia-Fernandez M, Estrella L, Valdivielso P, Quaglino D. Relationship Between Mitochondrial Structure and Bioenergetics in Pseudoxanthoma elasticum Dermal Fibroblasts. Front Cell Dev Biol 2020; 8:610266. [PMID: 33392199 PMCID: PMC7773789 DOI: 10.3389/fcell.2020.610266] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
Pseudoxanthoma elasticum (PXE) is a genetic disease considered as a paradigm of ectopic mineralization disorders, being characterized by multisystem clinical manifestations due to progressive calcification of skin, eyes, and the cardiovascular system, resembling an age-related phenotype. Although fibroblasts do not express the pathogenic ABCC6 gene, nevertheless these cells are still under investigation because they regulate connective tissue homeostasis, generating the "arena" where cells and extracellular matrix components can promote pathologic calcification and where activation of pro-osteogenic factors can be associated to pathways involving mitochondrial metabolism. The aim of the present study was to integrate structural and bioenergenetic features to deeply investigate mitochondria from control and from PXE fibroblasts cultured in standard conditions and to explore the role of mitochondria in the development of the PXE fibroblasts' pathologic phenotype. Proteomic, biochemical, and morphological data provide new evidence that in basal culture conditions (1) the protein profile of PXE mitochondria reveals a number of differentially expressed proteins, suggesting changes in redox balance, oxidative phosphorylation, and calcium homeostasis in addition to modified structure and organization, (2) measure of oxygen consumption indicates that the PXE mitochondria have a low ability to cope with a sudden increased need for ATP via oxidative phosphorylation, (3) mitochondrial membranes are highly polarized in PXE fibroblasts, and this condition contributes to increased reactive oxygen species levels, (4) ultrastructural alterations in PXE mitochondria are associated with functional changes, and (5) PXE fibroblasts exhibit a more abundant, branched, and interconnected mitochondrial network compared to control cells, indicating that fusion prevail over fission events. In summary, the present study demonstrates that mitochondria are modified in PXE fibroblasts. Since mitochondria are key players in the development of the aging process, fibroblasts cultured from aged individuals or aged in vitro are more prone to calcify, and in PXE, calcified tissues remind features of premature aging syndromes; it can be hypothesized that mitochondria represent a common link contributing to the development of ectopic calcification in aging and in diseases. Therefore, ameliorating mitochondrial functions and cell metabolism could open new strategies to positively regulate a number of signaling pathways associated to pathologic calcification.
Collapse
Affiliation(s)
| | - Federica Boraldi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Maria Garcia-Fernandez
- Department of Human Physiology, Biomedical Research Institute of Málaga, University of Malaga, Málaga, Spain
| | - Lara Estrella
- Department of Human Physiology, Biomedical Research Institute of Málaga, University of Malaga, Málaga, Spain
| | - Pedro Valdivielso
- Department of Medicine and Dermatology, Instituto de Investigación Biomédica de Málaga, University of Malaga, Málaga, Spain
- Internal Medicine Unit, Hospital Virgen de la Victoria, Málaga, Spain
| | - Daniela Quaglino
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
11
|
Molecular Chaperones: Molecular Assembly Line Brings Metabolism and Immunity in Shape. Metabolites 2020; 10:metabo10100394. [PMID: 33023034 PMCID: PMC7600384 DOI: 10.3390/metabo10100394] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022] Open
Abstract
Molecular chaperones are a set of conserved proteins that have evolved to assist the folding of many newly synthesized proteins by preventing their misfolding under conditions such as elevated temperatures, hypoxia, acidosis and nutrient deprivation. Molecular chaperones belong to the heat shock protein (HSP) family. They have been identified as important participants in immune functions including antigen presentation, immunostimulation and immunomodulation, and play crucial roles in metabolic rewiring and epigenetic circuits. Growing evidence has accumulated to indicate that metabolic pathways and their metabolites influence the function of immune cells and can alter transcriptional activity through epigenetic modification of (de)methylation and (de)acetylation. However, whether molecular chaperones can regulate metabolic programs to influence immune activity is still largely unclear. In this review, we discuss the available data on the biological function of molecular chaperones to immune responses during inflammation, with a specific focus on the interplay between molecular chaperones and metabolic pathways that drive immune cell fate and function.
Collapse
|
12
|
Dutta Gupta S, Pan CH. Recent update on discovery and development of Hsp90 inhibitors as senolytic agents. Int J Biol Macromol 2020; 161:1086-1098. [DOI: 10.1016/j.ijbiomac.2020.06.115] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/22/2020] [Accepted: 06/11/2020] [Indexed: 02/06/2023]
|
13
|
Reuter T, Vorwerk S, Liss V, Chao TC, Hensel M, Hansmeier N. Proteomic Analysis of Salmonella-modified Membranes Reveals Adaptations to Macrophage Hosts. Mol Cell Proteomics 2020; 19:900-912. [PMID: 32102972 PMCID: PMC7196581 DOI: 10.1074/mcp.ra119.001841] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/24/2020] [Indexed: 01/19/2023] Open
Abstract
Systemic infection and proliferation of intracellular pathogens require the biogenesis of a growth-stimulating compartment. The gastrointestinal pathogen Salmonella enterica commonly forms highly dynamic and extensive tubular membrane compartments built from Salmonella-modified membranes (SMMs) in diverse host cells. Although the general mechanism involved in the formation of replication-permissive compartments of S. enterica is well researched, much less is known regarding specific adaptations to different host cell types. Using an affinity-based proteome approach, we explored the composition of SMMs in murine macrophages. The systematic characterization provides a broader landscape of host players to the maturation of Salmonella-containing compartments and reveals core host elements targeted by Salmonella in macrophages as well as epithelial cells. However, we also identified subtle host specific adaptations. Some of these observations, such as the differential involvement of the COPII system, Rab GTPases 2A, 8B, 11 and ER transport proteins Sec61 and Sec22B may explain cell line-dependent variations in the pathophysiology of Salmonella infections. In summary, our system-wide approach demonstrates a hitherto underappreciated impact of the host cell type in the formation of intracellular compartments by Salmonella.
Collapse
Affiliation(s)
- Tatjana Reuter
- CellNanOs - Center for Cellular Nanoanalytics Osnabrück, School of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Stephanie Vorwerk
- CellNanOs - Center for Cellular Nanoanalytics Osnabrück, School of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Viktoria Liss
- Division of Microbiology, School of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Tzu-Chiao Chao
- Institute of Environmental Change and Society, Department of Biology, University of Regina, Regina, Canada
| | - Michael Hensel
- Division of Microbiology, School of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany; CellNanOs - Center for Cellular Nanoanalytics Osnabrück, School of Biology/Chemistry, University of Osnabrück, Osnabrück, Germany.
| | - Nicole Hansmeier
- Department of Biology, Faculty of Science, Luther College at University of Regina, Regina, Canada.
| |
Collapse
|
14
|
Molecular Chaperones in Cancer Stem Cells: Determinants of Stemness and Potential Targets for Antitumor Therapy. Cells 2020; 9:cells9040892. [PMID: 32268506 PMCID: PMC7226806 DOI: 10.3390/cells9040892] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/30/2020] [Accepted: 04/03/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer stem cells (CSCs) are a great challenge in the fight against cancer because these self-renewing tumorigenic cell fractions are thought to be responsible for metastasis dissemination and cases of tumor recurrence. In comparison with non-stem cancer cells, CSCs are known to be more resistant to chemotherapy, radiotherapy, and immunotherapy. Elucidation of mechanisms and factors that promote the emergence and existence of CSCs and their high resistance to cytotoxic treatments would help to develop effective CSC-targeting therapeutics. The present review is dedicated to the implication of molecular chaperones (protein regulators of polypeptide chain folding) in both the formation/maintenance of the CSC phenotype and cytoprotective machinery allowing CSCs to survive after drug or radiation exposure and evade immune attack. The major cellular chaperones, namely heat shock proteins (HSP90, HSP70, HSP40, HSP27), glucose-regulated proteins (GRP94, GRP78, GRP75), tumor necrosis factor receptor-associated protein 1 (TRAP1), peptidyl-prolyl isomerases, protein disulfide isomerases, calreticulin, and also a transcription heat shock factor 1 (HSF1) initiating HSP gene expression are here considered as determinants of the cancer cell stemness and potential targets for a therapeutic attack on CSCs. Various approaches and agents are discussed that may be used for inhibiting the chaperone-dependent development/manifestations of cancer cell stemness.
Collapse
|
15
|
Pant K, Richard S, Peixoto E, Gradilone SA. Role of Glucose Metabolism Reprogramming in the Pathogenesis of Cholangiocarcinoma. Front Med (Lausanne) 2020; 7:113. [PMID: 32318579 PMCID: PMC7146077 DOI: 10.3389/fmed.2020.00113] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 03/12/2020] [Indexed: 12/21/2022] Open
Abstract
Cholangiocarcinoma (CCA) is one of the most lethal cancers, and its rate of occurrence is increasing annually. The diagnoses of CCA patients remain elusive due to the lack of early symptoms and is misdiagnosed as HCC in a considerable percentage of patients. It is crucial to explore the underlying mechanisms of CCA carcinogenesis and development to find out specific biomarkers for early diagnosis of CCA and new promising therapeutic targets. In recent times, the reprogramming of tumor cells metabolism has been recognized as a hallmark of cancer. The modification from the oxidative phosphorylation metabolic pathway to the glycolysis pathway in CCA meets the demands of cancer cell proliferation and provides a favorable environment for tumor development. The alteration of metabolic programming in cancer cells is complex and may occur via mutations and epigenetic modifications within oncogenes, tumor suppressor genes, signaling pathways, and glycolytic enzymes. Herein we review the altered metabolism in cancer and the signaling pathways involved in this phenomena as they may affect CCA development. Understanding the regulatory pathways of glucose metabolism such as Akt/mTOR, HIF1α, and cMyc in CCA may further develop our knowledge of this devastating disease and may offer relevant information in the exploration of new diagnostic biomarkers and targeted therapeutic approaches for CCA.
Collapse
Affiliation(s)
- Kishor Pant
- The Hormel Institute, University of Minnesota, Austin, MN, United States
| | - Seth Richard
- The Hormel Institute, University of Minnesota, Austin, MN, United States
| | - Estanislao Peixoto
- The Hormel Institute, University of Minnesota, Austin, MN, United States
| | - Sergio A Gradilone
- The Hormel Institute, University of Minnesota, Austin, MN, United States.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
16
|
Khare T, Khare S, Angdisen JJ, Zhang Q, Stuckel A, Mooney BP, Ridenhour SE, Gitan RS, Hammoud GM, Ibdah JA. Defects in long-chain 3-hydroxy acyl-CoA dehydrogenase lead to hepatocellular carcinoma: A novel etiology of hepatocellular carcinoma. Int J Cancer 2020; 147:1461-1473. [PMID: 32115688 DOI: 10.1002/ijc.32943] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 01/02/2020] [Accepted: 02/17/2020] [Indexed: 12/14/2022]
Abstract
The incidence of both nonalcoholic fatty liver disease (NAFLD) and hepatocellular carcinoma (HCC) have been increasing at an alarming rate. Little is known about NAFLD without cirrhosis as a risk for HCC. Here we report, for the first time, generation of a mouse model with a defect in long-chain 3-hydoxy acyl-CoA dehydrogenase (LCHAD). The LCHAD exon 15 deletion was embryonic lethal to the homozygous mice whereas heterozygous mice (HT) develop significant hepatic steatosis starting at young age (3 months old) and HCC at older age (>13 months old) without any evidence of fibrosis or cirrhosis. None of the wild-type (WT) mice developed steatosis and HCC (n = 39), whereas HT-LCHAD mice (n = 41) showed steatosis and ~20% (8/41) developed liver masses with histological features of HCC. Proteomic analysis of liver tissues from WT-mice and HT-mice with no signs of HCC was conducted. Proteins with significant changes in abundance were identified by mass spectrometry. Abundance of 24 proteins was significantly different (p < 0.01) between WT and HT-LCHAD mice. The proteins found to vary in abundance are associated with different cellular response processes ranging from intermediary metabolism of carbohydrate, protein and lipid to oxidative stress, signal transduction and the process of tumorigenesis. Protein expression pattern of the HT-LCHAD mouse liver indicates predisposition to HCC and suggests that impaired hepatic mitochondrial fatty acid oxidation plays an important role in the development and progression of HCC. To assess the implication of these studies in human disease, we demonstrated significant downregulation of HADHA transcripts in HCC patients.
Collapse
Affiliation(s)
- Tripti Khare
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, USA
| | - Sharad Khare
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, USA.,Harry S Truman Veterans' Hospital, Columbia, MO, USA
| | - Jerry J Angdisen
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, USA
| | - Qiong Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, USA
| | - Alexei Stuckel
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, USA
| | - Brian P Mooney
- Gehrke Proteomics center, University of Missouri, Columbia, MO, USA
| | - Suzanne E Ridenhour
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, USA
| | - Raad S Gitan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, USA
| | - Ghassan M Hammoud
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, USA
| | - Jamal A Ibdah
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, USA.,Harry S Truman Veterans' Hospital, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| |
Collapse
|
17
|
El Rayes SM, Aboelmagd A, Gomaa MS, Fathalla W, Ali IAI, Pottoo FH, Khan FA. Newly synthesized 3-(4-chloro-phenyl)-3-hydroxy-2,2-dimethyl-propionic acid methyl ester derivatives selectively inhibit the proliferation of colon cancer cells. RSC Adv 2020; 10:8825-8841. [PMID: 35496560 PMCID: PMC9049988 DOI: 10.1039/c9ra10950a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 02/21/2020] [Indexed: 11/28/2022] Open
Abstract
A series of 24 compounds were synthesized based on structure modification of the model methyl-3-(4-chlorophenyl)-3-hydroxy-2,2-dimethylpropanoate as potent HDACIs. Saponification and hydrazinolysis of the model ester afforded the corresponding acid and hydrazide, respectively. The model ester was transformed into the corresponding trichloroacetimidate or acetate by the reaction with trichloroacetonitrile and acetic anhydride, respectively. N-Alkyl-3-(4-chlorophenyl)-3-hydroxy-2,2-dimethylpropan-amides and methyl-2-[(3-(4-chlorophenyl)-3-hydroxy-2,2-dimethylpropanoyl)amino] alkanoates were obtained by the reaction of corresponding acid or hydrazide with amines and amino acid esters via DCC and azide coupling methods. Methyl-3-aryl-3-(4-chlorophenyl)-2,2-dimethylpropanoates were obtained in good yields and short reaction time from the corresponding trichloroacetimidate or acetate by the reaction with C-active nucleophiles in the presence of TMSOTf (0.1 eq.%) via C–C bond formation. The antiproliferative and apoptotic activity were further studied with molecular docking. The 48 post-treatments showed that out of 24 compounds, 12 compounds showed inhibitory actions on HCT-116 cells, we have calculated the inhibitory action (IC50) of these compounds on HCT-116 and we have found that the IC50 values were in between 0.12 mg mL−1 to 0.81 mg mL−1. The compounds (7a & 7g) showed highest inhibitory activity (0.12 mg mL−1), whereas compound 7d showed the lowest inhibitory activity (0.81 mg mL−1). We have also examined inhibitory action on normal and non-cancerous cells (HEK-293 cells) and confirmed that action of these compounds was specific to cancerous cells. The cancerous cells were also examined for nuclear disintegration through staining with DAPI, (4′,6-diamidino-2-phenylindole) is a blue-fluorescent DNA stain, and we have found that there was loss of DAPI staining in the compound treated cancerous cells. The compounds were found to potentially act through the HSP90 and TRAP1 mediated signaling pathway. Compounds 7a and 7g showed the highest selectivity to TRAP1 which explained its superior activity. A series of 24 compounds were synthesized based on structure modification of the model methyl-3-(4-chlorophenyl)-3-hydroxy-2,2-dimethylpropanoate as potent HDACIs.![]()
Collapse
Affiliation(s)
- Samir M. El Rayes
- Department of Chemistry
- Faculty of Science
- Suez Canal University
- Ismailia
- Egypt
| | - Ahmed Aboelmagd
- Department of Chemistry
- Faculty of Science
- Suez Canal University
- Ismailia
- Egypt
| | - Mohamed S. Gomaa
- Department of Pharmaceutical
- College of Clinical Pharmacy
- Imam Abdulrahman Bin Faisal University
- Dammam 31441
- Kingdom of Saudi Arabia
| | - Walid Fathalla
- Department of Physics and Math
- Faculty of Engineering
- Port-Said University
- Port-Said
- Egypt
| | - Ibrahim A. I. Ali
- Department of Chemistry
- Faculty of Science
- Suez Canal University
- Ismailia
- Egypt
| | - Faheem H. Pottoo
- Department of Pharmacology
- College of Clinical Pharmacy
- Imam Abdulrahman Bin Faisal University
- Dammam 31441
- Kingdom of Saudi Arabia
| | - Firdos Alam Khan
- Department of Stem Cell Research
- Institute of Research and Medical Consultations (IRMC)
- Imam Abdulrahman Bin Faisal University
- Dammam 31441
- Saudi Arabia
| |
Collapse
|
18
|
Biebl MM, Buchner J. Structure, Function, and Regulation of the Hsp90 Machinery. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a034017. [PMID: 30745292 DOI: 10.1101/cshperspect.a034017] [Citation(s) in RCA: 206] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Heat shock protein 90 (Hsp90) is a molecular chaperone involved in the maturation of a plethora of substrates ("clients"), including protein kinases, transcription factors, and E3 ubiquitin ligases, positioning Hsp90 as a central regulator of cellular proteostasis. Hsp90 undergoes large conformational changes during its ATPase cycle. The processing of clients by cytosolic Hsp90 is assisted by a cohort of cochaperones that affect client recruitment, Hsp90 ATPase function or conformational rearrangements in Hsp90. Because of the importance of Hsp90 in regulating central cellular pathways, strategies for the pharmacological inhibition of the Hsp90 machinery in diseases such as cancer and neurodegeneration are being developed. In this review, we summarize recent structural and mechanistic progress in defining the function of organelle-specific and cytosolic Hsp90, including the impact of individual cochaperones on the maturation of specific clients and complexes with clients as well as ways of exploiting Hsp90 as a drug target.
Collapse
Affiliation(s)
- Maximilian M Biebl
- Center for Integrated Protein Science, Department of Chemistry, Technische Universität München, D-85748 Garching, Germany
| | - Johannes Buchner
- Center for Integrated Protein Science, Department of Chemistry, Technische Universität München, D-85748 Garching, Germany
| |
Collapse
|
19
|
Lettini G, Lepore S, Crispo F, Sisinni L, Esposito F, Landriscina M. Heat shock proteins in cancer stem cell maintenance: A potential therapeutic target? Histol Histopathol 2019; 35:25-37. [PMID: 31322279 DOI: 10.14670/hh-18-153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer stem cells (CSCs) are a subpopulation of tumor cells with unlimited self-renewal capability, multilineage differentiation potential and long-term tumor repopulation capacity. CSCs reside in anatomically distinct regions within the tumor microenvironment, called niches, and this favors the maintenance of CSC properties and preserves their phenotypic plasticity. Indeed, CSCs are characterized by a flexible state based on their capacity to interconvert between a differentiated and a stem-like phenotype, and this depends on the activation of adaptive mechanisms in response to different environmental conditions. Heat Shock Proteins (HSPs) are molecular chaperones, upregulated upon cell exposure to several stress conditions and are responsible for normal maturation, localization and activity of intra and extracellular proteins. Noteworthy, HSPs play a central role in several cellular processes involved in tumor initiation and progression (i.e. cell viability, resistance to apoptosis, stress conditions and drug therapy, EMT, bioenergetics, invasiveness, metastasis formation) and, thus, are widely considered potential molecular targets. Furthermore, much evidence suggests a key regulatory function for HSPs in CSC maintenance and their upregulation has been proposed as a mechanism used by CSCs to adapt to unfavorable environmental conditions, such as nutrient deprivation, hypoxia, inflammation. This review discusses the relevance of HSPs in CSC biology, highlighting their role as novel potential molecular targets to develop anticancer strategies aimed at CSC targeting.
Collapse
Affiliation(s)
- Giacomo Lettini
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Silvia Lepore
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Fabiana Crispo
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Lorenza Sisinni
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Franca Esposito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy.
| | - Matteo Landriscina
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy.,Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.
| |
Collapse
|
20
|
Hunt RJ, Bateman JM. Mitochondrial retrograde signaling in the nervous system. FEBS Lett 2017; 592:663-678. [PMID: 29086414 DOI: 10.1002/1873-3468.12890] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/16/2017] [Accepted: 10/20/2017] [Indexed: 01/12/2023]
Abstract
Mitochondria generate the majority of cellular ATP and are essential for neuronal function. Loss of mitochondrial activity leads to primary mitochondrial diseases and may contribute to neurodegenerative diseases such as Alzheimer's and Parkinson's disease. Mitochondria communicate with the cell through mitochondrial retrograde signaling pathways. These signaling pathways are triggered by mitochondrial dysfunction and allow the organelle to control nuclear gene transcription. Neuronal mitochondrial retrograde signaling pathways have been identified in disease model systems and targeted to restore neuronal function and prevent neurodegeneration. In this review, we describe yeast and mammalian cellular models that have paved the way in the investigation of mitochondrial retrograde mechanisms. We then discuss the evidence for retrograde signaling in neurons and our current knowledge of retrograde signaling mechanisms in neuronal model systems. We argue that targeting mitochondrial retrograde pathways has the potential to lead to novel treatments for neurological diseases.
Collapse
Affiliation(s)
- Rachel J Hunt
- Wolfson Centre for Age-Related Diseases, King's College London, UK
| | - Joseph M Bateman
- Wolfson Centre for Age-Related Diseases, King's College London, UK
| |
Collapse
|
21
|
Sisinni L, Maddalena F, Condelli V, Pannone G, Simeon V, Li Bergolis V, Lopes E, Piscazzi A, Matassa DS, Mazzoccoli C, Nozza F, Lettini G, Amoroso MR, Bufo P, Esposito F, Landriscina M. TRAP1 controls cell cycle G2-M transition through the regulation of CDK1 and MAD2 expression/ubiquitination. J Pathol 2017; 243:123-134. [PMID: 28678347 DOI: 10.1002/path.4936] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 05/23/2017] [Accepted: 06/21/2017] [Indexed: 12/16/2022]
Abstract
Regulation of tumour cell proliferation by molecular chaperones is still a complex issue. Here, the role of the HSP90 molecular chaperone TRAP1 in cell cycle regulation was investigated in a wide range of human breast, colorectal, and lung carcinoma cell lines, and tumour specimens. TRAP1 modulates the expression and/or the ubiquitination of key cell cycle regulators through a dual mechanism: (i) transcriptional regulation of CDK1, CYCLIN B1, and MAD2, as suggested by gene expression profiling of TRAP1-silenced breast carcinoma cells; and (ii) post-transcriptional quality control of CDK1 and MAD2, being the ubiquitination of these two proteins enhanced upon TRAP1 down-regulation. Mechanistically, TRAP1 quality control on CDK1 is crucial for its regulation of mitotic entry, since TRAP1 interacts with CDK1 and prevents CDK1 ubiquitination in cooperation with the proteasome regulatory particle TBP7, this representing the limiting factor in TRAP1 regulation of the G2-M transition. Indeed, TRAP1 silencing results in enhanced CDK1 ubiquitination, lack of nuclear translocation of CDK1/cyclin B1 complex, and increased MAD2 degradation, whereas CDK1 forced up-regulation partially rescues low cyclin B1 and MAD2 levels and G2-M transit in a TRAP1-poor background. Consistently, the CDK1 inhibitor RO-3306 is less active in a TRAP1-high background. Finally, a significant correlation was observed between TRAP1 and Ki67, CDK1 and/or MAD2 expression in breast, colorectal, and lung human tumour specimens. This study represents the first evidence that TRAP1 is relevant in the control of the complex machinery that governs cell cycle progression and mitotic entry and provides a strong rationale to regard TRAP1 as a biomarker to select tumours with deregulated cell cycle progression and thus likely poorly responsive to novel cell cycle inhibitors. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Lorenza Sisinni
- Laboratory of Pre-clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Francesca Maddalena
- Laboratory of Pre-clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Valentina Condelli
- Laboratory of Pre-clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Giuseppe Pannone
- Anatomic Pathology Unit, Department of Clinic and Experimental Medicine, University of Foggia, Italy
| | - Vittorio Simeon
- Laboratory of Pre-clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Valeria Li Bergolis
- Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, Italy
| | - Elvira Lopes
- Laboratory of Pre-clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Annamaria Piscazzi
- Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, Italy
| | - Danilo Swann Matassa
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Italy
| | - Carmela Mazzoccoli
- Laboratory of Pre-clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Filomena Nozza
- Laboratory of Pre-clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Giacomo Lettini
- Laboratory of Pre-clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Maria Rosaria Amoroso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Italy
| | - Pantaleo Bufo
- Anatomic Pathology Unit, Department of Clinic and Experimental Medicine, University of Foggia, Italy
| | - Franca Esposito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Italy
| | - Matteo Landriscina
- Laboratory of Pre-clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy.,Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, Italy
| |
Collapse
|
22
|
Lettini G, Maddalena F, Sisinni L, Condelli V, Matassa DS, Costi MP, Simoni D, Esposito F, Landriscina M. TRAP1: a viable therapeutic target for future cancer treatments? Expert Opin Ther Targets 2017; 21:805-815. [PMID: 28664757 DOI: 10.1080/14728222.2017.1349755] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION HSP90 molecular chaperones (i.e., HSP90α, HSP90β, GRP94 and TRAP1) are potential therapeutic targets to design novel anticancer agents. However, despite numerous designed HSP90 inhibitors, most of them have failed due to unfavorable toxicity profiles and lack of specificity toward different HSP90 paralogs. Indeed, a major limitation in this field is the high structural homology between different HSP90 chaperones, which significantly limits our capacity to design paralog-specific inhibitors. Area covered: This review examines the relevance of TRAP1 in tumor development and progression, with an emphasis on its oncogenic/oncosuppressive role in specific human malignancies and its multifaceted and context-dependent functions in cancer cells. Herein, we discuss the rationale for considering TRAP1 as a potential molecular target and the strategies used to date, to achieve its compartmentalized inhibition directly in mitochondria. Expert opinion: TRAP1 targeting may represent a promising strategy for cancer therapy, based on the increasing and compelling evidence supporting TRAP1 involvement in human carcinogenesis. However, considering the complexity of TRAP1 biology, future strategies of drug discovery need to improve selectivity and specificity toward TRAP1 respect to other HSP90 paralogs. The characterization of specific human malignancies suitable for TRAP1 targeting is also mandatory.
Collapse
Affiliation(s)
- Giacomo Lettini
- a Laboratory of Pre-Clinical and Translational Research , IRCCS, Referral Cancer Center of Basilicata , Rionero in Vulture , Italy
| | - Francesca Maddalena
- a Laboratory of Pre-Clinical and Translational Research , IRCCS, Referral Cancer Center of Basilicata , Rionero in Vulture , Italy
| | - Lorenza Sisinni
- a Laboratory of Pre-Clinical and Translational Research , IRCCS, Referral Cancer Center of Basilicata , Rionero in Vulture , Italy
| | - Valentina Condelli
- a Laboratory of Pre-Clinical and Translational Research , IRCCS, Referral Cancer Center of Basilicata , Rionero in Vulture , Italy
| | - Danilo Swann Matassa
- b Department of Molecular Medicine and Medical Biotechnology , University of Naples Federico II , Napoli , Italy
| | - Maria Paola Costi
- c Department of Life Sciences , University of Modena and Reggio Emilia , Modena , Italy
| | - Daniele Simoni
- d Department of Chemical and Pharmaceutical Sciences , University of Ferrara , Ferrara , Italy
| | - Franca Esposito
- b Department of Molecular Medicine and Medical Biotechnology , University of Naples Federico II , Napoli , Italy
| | - Matteo Landriscina
- a Laboratory of Pre-Clinical and Translational Research , IRCCS, Referral Cancer Center of Basilicata , Rionero in Vulture , Italy.,e Medical Oncology Unit, Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy
| |
Collapse
|
23
|
Chen JF, Wu QS, Xie YX, Si BL, Yang PP, Wang WY, Hua Q, He Q. TRAP1 ameliorates renal tubulointerstitial fibrosis in mice with unilateral ureteral obstruction by protecting renal tubular epithelial cell mitochondria. FASEB J 2017; 31:4503-4514. [PMID: 28710113 DOI: 10.1096/fj.201700283r] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 06/12/2017] [Indexed: 12/12/2022]
Abstract
Mitochondrial dysfunction causes renal tubular epithelial cell injury and promotes cell apoptosis and renal tubulointerstitial fibrosis (TIF) progression. TNF receptor-associated protein 1 (TRAP1) is a molecular chaperone protein that is localized in mitochondria. It plays an important role in cell apoptosis; however, its functional mechanism in TIF remains unclear. In this study, we observed the effects of TRAP1 in renal tubular epithelial cell mitochondria in mice with unilateral ureteral obstruction and its function in cell apoptosis and TIF. Results show that TRAP1 could protect the mitochondrial structure in renal tubular epithelial cells; maintain the levels of mitochondrial membrane potential, ATP, and mitochondrial DNA copy number; inhibit reactive oxygen species production; stabilize the expression of the mitochondrial inner membrane protein mitofilin; reduce renal tubular epithelial cell apoptosis; and inhibit TIF. These results provide new theoretical foundations for additional understanding of the antifibrotic mechanism of TRAP1 in the kidney.-Chen, J.-F., Wu, Q.-S., Xie, Y.-X., Si, B.-L., Yang, P.-P., Wang, W.-Y., Hua, Q., He, Q. TRAP1 ameliorates renal tubulointerstitial fibrosis in mice with unilateral ureteral obstruction by protecting renal tubular epithelial cell mitochondria.
Collapse
Affiliation(s)
- Jun-Feng Chen
- Division of Hemodialysis, Nanjing First Hospital, Nanjing Medical University, Nanjing, China;
| | - Qi-Shun Wu
- Division of Nephrology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Yu-Xian Xie
- Division of Nephrology, People's Hospital of Suzhou High-Tech District, Jiangsu University, Suzhou, China
| | - Bo-Lin Si
- Division of Nephrology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Ping-Ping Yang
- Division of Nephrology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Wen-Yan Wang
- Division of Nephrology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Qin Hua
- Division of Nephrology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Qing He
- Division of Hemodialysis, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
24
|
Zhao X, Sun K, Lan Z, Song W, Cheng L, Chi W, Chen J, Huo Y, Xu L, Liu X, Deng H, Siegenthaler JA, Chen L. Tenofovir and adefovir down-regulate mitochondrial chaperone TRAP1 and succinate dehydrogenase subunit B to metabolically reprogram glucose metabolism and induce nephrotoxicity. Sci Rep 2017; 7:46344. [PMID: 28397817 PMCID: PMC5387747 DOI: 10.1038/srep46344] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 03/16/2017] [Indexed: 02/05/2023] Open
Abstract
Despite the therapeutic success of tenofovir (TFV) for treatment of HIV-1 infection, numerous cases of nephrotoxicity have been reported. Mitochondrial toxicity has been purported as the major target of TFV-associated renal tubulopathy but the underlying molecular mechanism remains unclear. In this report, we use metabolomics and proteomics with HK-2 cells and animal models to dissect the molecular pathways underlying nephropathy caused by TFV and its more toxic analog, adefovir (ADV). Proteomic analysis shows that mitochondrial chaperone TRAP1 and mtDNA replicating protein SSBP1 were significantly down-regulated in TFV and ADV treated HK-2 cells compared with controls. Transmission electron microscopy (TEM) revealed that TFV and ADV-treated HK-2 cells had accumulated glycogen, a phenotype that was also observed in mice treated with TFV and ADV. Analysis of the proteins in TCA cycle showed succinate dehydrogenase subunit B (SDHB) was nearly depleted in glucose oxidative phosphorylation pathway however certain enzymes in the glycolysis and glycogen synthesis pathway had elevated expression in TFV and ADV-treated HK-2 cells. These results suggest that TFV and ADV may cause mitochondrial dysfunction in renal tubular cells and reprogramming of glucose metabolism. The resulting glycogen accumulation may partially contribute to TFV and ADV induced renal dysfunction.
Collapse
Affiliation(s)
- Xinbin Zhao
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Kun Sun
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Zhou Lan
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Wenxin Song
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Lili Cheng
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Wenna Chi
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
- Collaborative Innovation Center for Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Jing Chen
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Yi Huo
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Lina Xu
- Technology Center for Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xiaohui Liu
- Technology Center for Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Julie A. Siegenthaler
- Department of Pediatrics, Denver-Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
| | - Ligong Chen
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
- Collaborative Innovation Center for Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
25
|
Mitochondrial complex I inhibition triggers a mitophagy-dependent ROS increase leading to necroptosis and ferroptosis in melanoma cells. Cell Death Dis 2017; 8:e2716. [PMID: 28358377 PMCID: PMC5386536 DOI: 10.1038/cddis.2017.133] [Citation(s) in RCA: 417] [Impact Index Per Article: 52.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 02/23/2017] [Accepted: 02/28/2017] [Indexed: 12/21/2022]
Abstract
Inhibition of complex I (CI) of the mitochondrial respiratory chain by BAY 87-2243 (‘BAY') triggers death of BRAFV600E melanoma cell lines and inhibits in vivo tumor growth. Here we studied the mechanism by which this inhibition induces melanoma cell death. BAY treatment depolarized the mitochondrial membrane potential (Δψ), increased cellular ROS levels, stimulated lipid peroxidation and reduced glutathione levels. These effects were paralleled by increased opening of the mitochondrial permeability transition pore (mPTP) and stimulation of autophagosome formation and mitophagy. BAY-induced cell death was not due to glucose shortage and inhibited by the antioxidant α-tocopherol and the mPTP inhibitor cyclosporin A. Tumor necrosis factor receptor-associated protein 1 (TRAP1) overexpression in BAY-treated cells lowered ROS levels and inhibited mPTP opening and cell death, whereas the latter was potentiated by TRAP1 knockdown. Knockdown of autophagy-related 5 (ATG5) inhibited the BAY-stimulated autophagosome formation, cellular ROS increase and cell death. Knockdown of phosphatase and tensin homolog-induced putative kinase 1 (PINK1) inhibited the BAY-induced Δψ depolarization, mitophagy stimulation, ROS increase and cell death. Dynamin-related protein 1 (Drp1) knockdown induced mitochondrial filamentation and inhibited BAY-induced cell death. The latter was insensitive to the pancaspase inhibitor z-VAD-FMK, but reduced by necroptosis inhibitors (necrostatin-1, necrostatin-1s)) and knockdown of key necroptosis proteins (receptor-interacting serine/threonine-protein kinase 1 (RIPK1) and mixed lineage kinase domain-like (MLKL)). BAY-induced cell death was also reduced by the ferroptosis inhibitor ferrostatin-1 and overexpression of the ferroptosis-inhibiting protein glutathione peroxidase 4 (GPX4). This overexpression also inhibited the BAY-induced ROS increase and lipid peroxidation. Conversely, GPX4 knockdown potentiated BAY-induced cell death. We propose a chain of events in which: (i) CI inhibition induces mPTP opening and Δψ depolarization, that (ii) stimulate autophagosome formation, mitophagy and an associated ROS increase, leading to (iii) activation of combined necroptotic/ferroptotic cell death.
Collapse
|
26
|
Masgras I, Sanchez-Martin C, Colombo G, Rasola A. The Chaperone TRAP1 As a Modulator of the Mitochondrial Adaptations in Cancer Cells. Front Oncol 2017; 7:58. [PMID: 28405578 PMCID: PMC5370238 DOI: 10.3389/fonc.2017.00058] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 03/15/2017] [Indexed: 12/18/2022] Open
Abstract
Mitochondria can receive, integrate, and transmit a variety of signals to shape many biochemical activities of the cell. In the process of tumor onset and growth, mitochondria contribute to the capability of cells of escaping death insults, handling changes in ROS levels, rewiring metabolism, and reprograming gene expression. Therefore, mitochondria can tune the bioenergetic and anabolic needs of neoplastic cells in a rapid and flexible way, and these adaptations are required for cell survival and proliferation in the fluctuating environment of a rapidly growing tumor mass. The molecular bases of pro-neoplastic mitochondrial adaptations are complex and only partially understood. Recently, the mitochondrial molecular chaperone TRAP1 (tumor necrosis factor receptor associated protein 1) was identified as a key regulator of mitochondrial bioenergetics in tumor cells, with a profound impact on neoplastic growth. In this review, we analyze these findings and discuss the possibility that targeting TRAP1 constitutes a new antitumor approach.
Collapse
Affiliation(s)
- Ionica Masgras
- Dipartimento di Scienze Biomediche, Istituto di Neuroscienze, Consiglio Nazionale delle Ricerche (CNR), Università di Padova , Padova , Italy
| | - Carlos Sanchez-Martin
- Dipartimento di Scienze Biomediche, Istituto di Neuroscienze, Consiglio Nazionale delle Ricerche (CNR), Università di Padova , Padova , Italy
| | - Giorgio Colombo
- Istituto di Chimica del Riconoscimento Molecolare, Consiglio Nazionale delle Ricerche (CNR) , Milano , Italy
| | - Andrea Rasola
- Dipartimento di Scienze Biomediche, Istituto di Neuroscienze, Consiglio Nazionale delle Ricerche (CNR), Università di Padova , Padova , Italy
| |
Collapse
|