1
|
Mahmoudi A, Butler AE, Orekhov AN, Jamialahmadi T, Sahebkar A. Statins as a Potential Treatment for Non-alcoholic Fatty Liver Disease: Target Deconvolution using Protein-protein Interaction Network Analysis. Curr Med Chem 2025; 32:1355-1377. [PMID: 37644746 DOI: 10.2174/0929867331666230829164832] [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: 02/14/2023] [Revised: 06/28/2023] [Accepted: 07/18/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND The hallmark of non-alcoholic fatty liver disease (NAFLD) is aberrant buildup of triglycerides (TGs) in hepatocytes. Many genes promote NAFLD development. Using bioinformatics tools, we investigated the possible effect of statins on genes involved in NAFLD progression. METHODS Protein interactions of statins and NAFLD were searched in gene-drug and gene-disease databases. A Protein-Protein interaction (PPI) network was constructed to find hub genes and Molecular Complex Detection (MCODE) of NAFLD-related genes. Shared protein targets between protein targets of statins and NAFLD-associated genes were identified. Next, targets of each statin were assayed with all modular clusters in the MCODEs related to NAFLD. Biological process and pathway enrichment analysis for shared proteins was performed. RESULTS Screening protein targets for conventional statins and curated NAFLD-related genes identified 343 protein targets and 70 genes, respectively. A Venn diagram of NAFLD-related genes and protein targets of statins showed 24 shared proteins. The biological pathways on KEGG enrichment associated with the 24 shared protein sets were evaluated and included cytokine-cytokine receptor interaction, adipocytokine, PPAR, TNF and AMPK signaling pathways. Gene Ontology analysis showed major involvement in lipid metabolic process regulation and inflammatory response. PPI network analysis of 70 protein targets indicated 13 hub genes (PPARA, IL4, CAT, LEP, SREBF1, PRKCA, CYP2E1, NFE2L2, PTEN, NR1H4, ADIPOQ, GSTP1 and TGFB1). Comparing all seven statins with the three MCODE clusterings and 13 hub genes revealed that simvastatin as the most associated statin with NAFLD. CONCLUSION Simvastatin has the most impact on NAFLD-related genes versus other statins.
Collapse
Affiliation(s)
- Ali Mahmoudi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alexandra E Butler
- Department of Medical Sciences, Royal College of Surgeons in Ireland Bahrain, Adliya, Bahrain
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow 125315, Russia
- Institute for Atherosclerosis Research, Osennyaya Street 4-1-207, Moscow 121609, Russia
| | - Tannaz Jamialahmadi
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
2
|
Liu JC, Lei SY, Zhang DH, He QY, Sun YY, Zhu HJ, Qu Y, Zhou SY, Yang Y, Li C, Guo ZN. The pleiotropic effects of statins: a comprehensive exploration of neurovascular unit modulation and blood-brain barrier protection. Mol Med 2024; 30:256. [PMID: 39707228 DOI: 10.1186/s10020-024-01025-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024] Open
Abstract
The blood-brain barrier (BBB) is the most central component of the neurovascular unit (NVU) and is crucial for the maintenance of the internal environment of the central nervous system and the regulation of homeostasis. A multitude of neuroprotective agents have been developed to exert neuroprotective effects and improve the prognosis of patients with ischemic stroke. These agents have been designed to maintain integrity and promote BBB repair. Statins are widely used as pharmacological agents for the treatment and prevention of ischemic stroke, making them a cornerstone in the pharmacological armamentarium for this condition. The primary mechanism of action is the reduction of serum cholesterol through the inhibition of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase, which results in a decrease in low-density lipoprotein cholesterol (LDL-C) and an increase in cholesterol clearance. Nevertheless, basic and clinical research has indicated that statins may exert additional pleiotropic effects beyond LDL-C reduction. Previous studies on ischemic stroke have demonstrated that statins can enhance neurological function, reduce inflammation, and promote angiogenic and synaptic processes following ischemic stroke. The BBB has been increasingly recognized for its role in the development and progression of ischemic stroke. Statins have also been found to play a potential BBB protective role by affecting members of the NVU. This review aimed to provide a comprehensive theoretical basis for the clinical application of statins by systematically detailing how statins influence the BBB, particularly focusing on the regulation of the function of each member of the NVU.
Collapse
Affiliation(s)
- Jia-Cheng Liu
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Shuang-Yin Lei
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Dian-Hui Zhang
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Qian-Yan He
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Ying-Ying Sun
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Hong-Jing Zhu
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Yang Qu
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Sheng-Yu Zhou
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Yi Yang
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Xinmin Street 1#, Changchun, 130021, China
| | - Chao Li
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China.
| | - Zhen-Ni Guo
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Xinmin Street 1#, Changchun, 130021, China.
- Neuroscience Research Center, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China.
| |
Collapse
|
3
|
Karimi Jirandehi A, Asgari R, Keshavarz Shahbaz S, Rezaei N. Nanomedicine marvels: crafting the future of cancer therapy with innovative statin nano-formulation strategies. NANOSCALE ADVANCES 2024:d4na00808a. [PMID: 39478996 PMCID: PMC11515941 DOI: 10.1039/d4na00808a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 10/11/2024] [Indexed: 11/02/2024]
Abstract
Statins, traditionally used for managing hyperlipidemia and cardiovascular diseases, have garnered significant interest for their potential anti-cancer properties. Research indicates that statins can inhibit critical processes in cancer development, such as apoptosis, angiogenesis, and metastasis. Despite their promising anti-cancer effects, the clinical application of statins in oncology has been hampered by their inherent low solubility and bioavailability. These pharmacokinetic challenges can be effectively addressed through the use of nano-based drug delivery systems. Nano-formulations enhance the delivery and therapeutic efficacy of statins by improving their solubility, stability, and targeting ability, thus maximizing their concentration within the tumor microenvironment and minimizing systemic side effects. This review delves into the potential of nanoparticles as carriers for statins in cancer therapy. It explores the mechanisms by which statins exert their anti-cancer effects, such as through the inhibition of the mevalonate pathway, modulation of immune responses, and induction of apoptosis. Furthermore, the review examines the development of various statin-loaded nano-formulations, highlighting their advantages over conventional formulations. The novelty of this review lies in its focus on recent advancements in nanoformulations that enhance statin delivery to the tumor microenvironment. By discussing the current advancements and prospects of statin nano-formulations, this review aims to provide a comprehensive understanding of how these innovative strategies can improve cancer treatment outcomes and enhance the quality of life for patients. The integration of nanotechnology with statin therapy offers a novel approach to overcoming existing therapeutic limitations and paving the way for more effective and safer cancer treatments.
Collapse
Affiliation(s)
- Ashkan Karimi Jirandehi
- Student Research Committee, School of Medicine, Qazvin University of Medical Sciences Qazvin Iran
- USERN Office, Qazvin University of Medical Science Qazvin Iran
| | - Reza Asgari
- Student Research Committee, School of Medicine, Qazvin University of Medical Sciences Qazvin Iran
- USERN Office, Qazvin University of Medical Science Qazvin Iran
| | - Sanaz Keshavarz Shahbaz
- Cellular and Molecular Research Center, Research Institute for Prevention of Noncommunicable Disease, Qazvin University of Medical Sciences Qazvin Iran
- USERN Office, Qazvin University of Medical Science Qazvin Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science Tehran Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN) Tehran Iran
| |
Collapse
|
4
|
Liu XY, Chen B, Zhang R, Zhang MQ, Ma YY, Han Y, Jiang JD, Zhang JP. Atorvastatin-induced intracerebral hemorrhage is inhibited by berberine in zebrafish. J Appl Toxicol 2024; 44:1198-1213. [PMID: 38639436 DOI: 10.1002/jat.4614] [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: 01/17/2024] [Revised: 03/18/2024] [Accepted: 04/02/2024] [Indexed: 04/20/2024]
Abstract
Intracerebral hemorrhage (ICH), for which there are currently no effective preventive or treatment methods, has a very high fatality rate. Statins, such as atorvastatin (ATV), are the first-line drugs for regulating blood lipids and treating hyperlipidemia-related cardiovascular diseases. However, ATV-associated ICH has been reported, although its incidence is rare. In this study, we aimed to investigate the protective action and mechanisms of berberine (BBR) against ATV-induced brain hemorrhage. We established an ICH model in zebrafish induced by ATV (2 μM) and demonstrated the effects of BBR (10, 50, and 100 μM) on ICH via protecting the vascular network using hemocyte staining and three transgenic zebrafish. BBR was found to reduce brain inflammation and locomotion injury in ICH-zebrafish. Mechanism research showed that ATV increased the levels of VE-cadherin and occludin proteins but disturbed their localization at the cell membrane by abnormal phosphorylation, which decreased the number of intercellular junctions between vascular endothelial cells (VECs), disrupting the integrity of vascular walls. BBR reversed the effects of ATV by promoting autophagic degradation of phosphorylated VE-cadherin and occludin in ATV-induced VECs examined by co-immunoprecipitation (co-IP). These findings provide crucial insights into understanding the BBR mechanisms involved in the maintenance of vascular integrity and in mitigating adverse reactions to ATV.
Collapse
Affiliation(s)
- Xin-Yan Liu
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Chen
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rui Zhang
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Miao-Qing Zhang
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan-Yuan Ma
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Han
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian-Dong Jiang
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing-Pu Zhang
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
5
|
Zhao Y, Wang Y, Ren J, Gong W, Nie X, Peng Y, Li J, Duan C. Atorvastatin causes developmental and behavioral toxicity in yellowstripe goby (Mugilogobius chulae) embryos/larvae via disrupting lipid metabolism and autophagy processes. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 271:106909. [PMID: 38593744 DOI: 10.1016/j.aquatox.2024.106909] [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: 01/19/2024] [Revised: 03/25/2024] [Accepted: 04/05/2024] [Indexed: 04/11/2024]
Abstract
Atorvastatin (ATV) is one of the most commonly prescribed lipid-lowering drugs detected frequently in the environment due to its high use and low degradation rate. However, the toxic effects of residual ATV in the aquatic environment on non-target organisms and its toxic mechanisms are still largely unknown. In the present study, embryos of a native estuarine benthic fish, Mugilogobius chulae, were employed to investigate the developmental and behavioral toxic effects of ATV including environmentally relevant concentrations. The aim of this study was to provide a scientific basis for ecological risk assessment of ATV in the aquatic environment by investigating the changes of biological endpoints at multiple levels in M. chulae embryos/larvae. The results showed that ATV had significantly lethal and teratogenic effects on M. chulae embryos/larvae and caused abnormal changes in developmental parameters including hatch rate, body length, heart rate, and spontaneous movement. ATV exposure caused oxidative stress in M. chulae embryos/larvae subsequently inhibited autophagy and activated apoptosis, leading to abnormal developmental processes and behavioral changes in M. chulae embryos/larvae. The disruptions of lipid metabolism, autophagy, and apoptosis in M. chulae embryos/larvae caused by ATV exposure may pose a potential ecological risk at the population level.
Collapse
Affiliation(s)
- Yufei Zhao
- Department of Ecology, Jinan University, Guangzhou, 510632, China
| | - Yimeng Wang
- Department of Ecology, Jinan University, Guangzhou, 510632, China; Guangdong Laboratory Animals Monitoring Institute, Guangzhou, 510663, China
| | - Jinzhi Ren
- Department of Ecology, Jinan University, Guangzhou, 510632, China
| | - Weibo Gong
- Department of Ecology, Jinan University, Guangzhou, 510632, China
| | - Xiangping Nie
- Department of Ecology, Jinan University, Guangzhou, 510632, China.
| | - Ying Peng
- Research and Development Center for Watershed Environmental Eco-Engineering, Beijing Normal University, Zhuhai, 519087, China
| | - Jianjun Li
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, 510663, China
| | - Chunni Duan
- Department of Ecology, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
6
|
Zhao Y, Duan C, Zhang H, Gong W, Wang Y, Ren J, Nie X, Li J. Response of lipid metabolism, energy supply, and cell fate in yellowstripe goby (Mugilogobius chulae) exposed to environmentally relevant concentrations atorvastatin. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 341:122991. [PMID: 37995957 DOI: 10.1016/j.envpol.2023.122991] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/07/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023]
Abstract
The usage of typical pharmaceuticals and personal care products (PPCPs) such as cardiovascular and lipid-modulating drugs in clinical care accounts for the largest share of pharmaceutical consumption in most countries. Atorvastatin (ATV), one of the most commonly used lipid-lowering drugs, is frequently detected with lower concentrations in aquatic environments owing to its wide application, low removal, and degradation rates. However, the adverse effects of ATV on non-target aquatic organisms, especially the molecular mechanisms behind the toxic effects, still remain unclear. Therefore, this study investigated the potentially toxic effects of ATV exposure (including environmental concentrations) on yellowstripe goby (Mugilogobius chulae) and addressed the multi-dimensional responses. The results showed that ATV caused typical hepatotoxicity to M. chulae. ATV interfered with lipid metabolism by blocking fatty acid β-oxidation and led to the over-consumption of lipids. Thus, the exposed organism was obliged to alter the energy supply patterns and substrates utilization pathways to keep the normal energy supply. In addition, the higher concentration of ATV exposure caused oxidative stress to the organism. Subsequently, M. chulae triggered the autophagy and apoptosis processes with the help of key stress-related transcriptional regulators FOXOs and Sestrins to degrade the damaged organelles and proteins to maintain intracellular homeostasis.
Collapse
Affiliation(s)
- Yufei Zhao
- Department of Ecology, Jinan University, Guangzhou, 510632, China
| | - Chunni Duan
- Department of Ecology, Jinan University, Guangzhou, 510632, China
| | - Huiyu Zhang
- Department of Ecology, Jinan University, Guangzhou, 510632, China
| | - Weibo Gong
- Department of Ecology, Jinan University, Guangzhou, 510632, China
| | - Yimeng Wang
- Department of Ecology, Jinan University, Guangzhou, 510632, China
| | - Jinzhi Ren
- Department of Ecology, Jinan University, Guangzhou, 510632, China
| | - Xiangping Nie
- Department of Ecology, Jinan University, Guangzhou, 510632, China.
| | - Jianjun Li
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, 510663, China
| |
Collapse
|
7
|
Roth L, Dogan S, Tuna BG, Aranyi T, Benitez S, Borrell-Pages M, Bozaykut P, De Meyer GRY, Duca L, Durmus N, Fonseca D, Fraenkel E, Gillery P, Giudici A, Jaisson S, Johansson M, Julve J, Lucas-Herald AK, Martinet W, Maurice P, McDonnell BJ, Ozbek EN, Pucci G, Pugh CJA, Rochfort KD, Roks AJM, Rotllan N, Shadiow J, Sohrabi Y, Spronck B, Szeri F, Terentes-Printzios D, Tunc Aydin E, Tura-Ceide O, Ucar E, Yetik-Anacak G. Pharmacological modulation of vascular ageing: A review from VascAgeNet. Ageing Res Rev 2023; 92:102122. [PMID: 37956927 DOI: 10.1016/j.arr.2023.102122] [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: 07/05/2023] [Revised: 10/27/2023] [Accepted: 11/09/2023] [Indexed: 11/20/2023]
Abstract
Vascular ageing, characterized by structural and functional changes in blood vessels of which arterial stiffness and endothelial dysfunction are key components, is associated with increased risk of cardiovascular and other age-related diseases. As the global population continues to age, understanding the underlying mechanisms and developing effective therapeutic interventions to mitigate vascular ageing becomes crucial for improving cardiovascular health outcomes. Therefore, this review provides an overview of the current knowledge on pharmacological modulation of vascular ageing, highlighting key strategies and promising therapeutic targets. Several molecular pathways have been identified as central players in vascular ageing, including oxidative stress and inflammation, the renin-angiotensin-aldosterone system, cellular senescence, macroautophagy, extracellular matrix remodelling, calcification, and gasotransmitter-related signalling. Pharmacological and dietary interventions targeting these pathways have shown potential in ameliorating age-related vascular changes. Nevertheless, the development and application of drugs targeting vascular ageing is complicated by various inherent challenges and limitations, such as certain preclinical methodological considerations, interactions with exercise training and sex/gender-related differences, which should be taken into account. Overall, pharmacological modulation of endothelial dysfunction and arterial stiffness as hallmarks of vascular ageing, holds great promise for improving cardiovascular health in the ageing population. Nonetheless, further research is needed to fully elucidate the underlying mechanisms and optimize the efficacy and safety of these interventions for clinical translation.
Collapse
Affiliation(s)
- Lynn Roth
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.
| | - Soner Dogan
- Department of Medical Biology, School of Medicine, Yeditepe University, Istanbul, Turkiye
| | - Bilge Guvenc Tuna
- Department of Biophysics, School of Medicine, Yeditepe University, Istanbul, Turkiye
| | - Tamas Aranyi
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary; Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Sonia Benitez
- CIBER de Diabetes y enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Cardiovascular Biochemistry, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Maria Borrell-Pages
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - Perinur Bozaykut
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkiye
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Laurent Duca
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling", Université de Reims Champagne Ardenne (URCA), Reims, France
| | - Nergiz Durmus
- Department of Pharmacology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkiye
| | - Diogo Fonseca
- Laboratory of Pharmacology and Pharmaceutical Care, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Emil Fraenkel
- 1st Department of Internal Medicine, University Hospital, Pavol Jozef Šafárik University of Košice, Košice, Slovakia
| | - Philippe Gillery
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling", Université de Reims Champagne Ardenne (URCA), Reims, France; Laboratoire de Biochimie-Pharmacologie-Toxicologie, Centre Hospitalier et Universitaire de Reims, Reims, France
| | - Alessandro Giudici
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Maastricht University, the Netherlands; GROW School for Oncology and Reproduction, Maastricht University, the Netherlands
| | - Stéphane Jaisson
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling", Université de Reims Champagne Ardenne (URCA), Reims, France; Laboratoire de Biochimie-Pharmacologie-Toxicologie, Centre Hospitalier et Universitaire de Reims, Reims, France
| | | | - Josep Julve
- CIBER de Diabetes y enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Endocrinology, Diabetes and Nutrition group, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | | | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Pascal Maurice
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling", Université de Reims Champagne Ardenne (URCA), Reims, France
| | - Barry J McDonnell
- Centre for Cardiovascular Health and Ageing, Cardiff Metropolitan University, Cardiff, UK
| | - Emine Nur Ozbek
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir, Turkiye
| | - Giacomo Pucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Christopher J A Pugh
- Centre for Cardiovascular Health and Ageing, Cardiff Metropolitan University, Cardiff, UK
| | - Keith D Rochfort
- School of Nursing, Psychotherapy, and Community Health, Dublin City University, Dublin, Ireland
| | - Anton J M Roks
- Department of Internal Medicine, Division of Vascular Disease and Pharmacology, Erasmus Medical Center, Erasmus University, Rotterdam, the Netherlands
| | - Noemi Rotllan
- CIBER de Diabetes y enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Pathophysiology of lipid-related diseases, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - James Shadiow
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Yahya Sohrabi
- Molecular Cardiology, Dept. of Cardiology I - Coronary and Peripheral Vascular Disease, University Hospital Münster, Westfälische Wilhelms-Universität, 48149 Münster, Germany; Department of Medical Genetics, Third Faculty of Medicine, Charles University, 100 00 Prague, Czechia
| | - Bart Spronck
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Maastricht University, the Netherlands; Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Australia
| | - Flora Szeri
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Dimitrios Terentes-Printzios
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Elif Tunc Aydin
- Department of Cardiology, Hospital of Ataturk Training and Research Hospital, Katip Celebi University, Izmir, Turkiye
| | - Olga Tura-Ceide
- Biomedical Research Institute-IDIBGI, Girona, Spain; Department of Pulmonary Medicine, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS); University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias, Madrid, Spain
| | - Eda Ucar
- Department of Biophysics, School of Medicine, Yeditepe University, Istanbul, Turkiye
| | - Gunay Yetik-Anacak
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir, Turkiye; Department of Pharmacology, Faculty of Pharmacy, Acıbadem Mehmet Aydinlar University, Istanbul, Turkiye.
| |
Collapse
|
8
|
Wang D, Dai S, Lou D, Wang T, Wang S, Zheng Z. Association between statins exposure and risk of skin cancer: an updated meta-analysis. Int J Dermatol 2023; 62:1332-1344. [PMID: 37681467 DOI: 10.1111/ijd.16816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 08/05/2023] [Accepted: 08/09/2023] [Indexed: 09/09/2023]
Abstract
This study aimed to investigate the relationship between statin (lipophilic statin and hydrophilic statin) exposure and the risk of skin cancer. The incidence of skin cancer under statin exposure was used as the primary outcome, and the relevant studies were screened from Web of Science, PubMed, Cochrane Library, and EBSCO electronic database until September 2022. Ten observational studies and two randomized controlled trials (RCTs) were included. The statistical results indicated that in lipophilic statins, the exposed group had a higher risk of skin cancer than the non-exposed group (OR: 1.09, P = 0.003). However, compared with the non-exposed group, there was no significant difference between hydrophilic statins exposure and the incidence of skin cancer (OR: 1.02, P = 0.341). Further subgroup analysis of the subtypes of statins revealed that compared with the non-exposed group, exposure to lovastatin (OR: 1.18, P = 0.048) or simvastatin (OR: 1.11, P < 0.001) was a risk factor for skin cancer. Besides, subgroup analysis based on the subtypes of skin cancer demonstrated that the risks of melanoma (OR: 1.13, P = 0.009), basal cell carcinoma (BCC) (OR: 1.05, P = 0.036), and squamous cell carcinoma (SCC) (OR: 1.13, P = 0.026) under lipophilic statin exposure were significantly higher than those in the non-exposed group. On the contrary, compared with the non-exposed group, the risk of BCC was significantly reduced under the exposure of hydrophilic statins (OR: 0.93, P = 0.031). This study showed that the relationship between statin exposure and skin cancer risk was affected by the subtypes of statins and skin cancer subtypes.
Collapse
Affiliation(s)
- Dongying Wang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Senjie Dai
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Dandi Lou
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Tianyue Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shihui Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Zhen Zheng
- Department of Respiratory Medicine, Ningbo Yinzhou No. 2 Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
9
|
Lee YCG, Chou FN, Tung SY, Chou HC, Ko TL, Fann YC, Juan SH. Tumoricidal Activity of Simvastatin in Synergy with RhoA Inactivation in Antimigration of Clear Cell Renal Cell Carcinoma Cells. Int J Mol Sci 2023; 24:ijms24119738. [PMID: 37298689 DOI: 10.3390/ijms24119738] [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: 03/25/2023] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
Among kidney cancers, clear cell renal cell carcinoma (ccRCC) has the highest incidence rate in adults. The survival rate of patients diagnosed as having metastatic ccRCC drastically declines even with intensive treatment. We examined the efficacy of simvastatin, a lipid-lowering drug with reduced mevalonate synthesis, in ccRCC treatment. Simvastatin was found to reduce cell viability and increase autophagy induction and apoptosis. In addition, it reduced cell metastasis and lipid accumulation, the target proteins of which can be reversed through mevalonate supplementation. Moreover, simvastatin suppressed cholesterol synthesis and protein prenylation that is essential for RhoA activation. Simvastatin might also reduce cancer metastasis by suppressing the RhoA pathway. A gene set enrichment analysis (GSEA) of the human ccRCC GSE53757 data set revealed that the RhoA and lipogenesis pathways are activated. In simvastatin-treated ccRCC cells, although RhoA was upregulated, it was mainly restrained in the cytosolic fraction and concomitantly reduced Rho-associated protein kinase activity. RhoA upregulation might be a negative feedback effect owing to the loss of RhoA activity caused by simvastatin, which can be restored by mevalonate. RhoA inactivation by simvastatin was correlated with decreased cell metastasis in the transwell assay, which was mimicked in dominantly negative RhoA-overexpressing cells. Thus, owing to the increased RhoA activation and cell metastasis in the human ccRCC dataset analysis, simvastatin-mediated Rho inactivation might serve as a therapeutic target for ccRCC patients. Altogether, simvastatin suppressed the cell viability and metastasis of ccRCC cells; thus, it is a potentially effective ccRCC adjunct therapy after clinical validation for ccRCC treatment.
Collapse
Affiliation(s)
- Yuan-Chii Gladys Lee
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Fang-Ning Chou
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Szu-Yu Tung
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsiu-Chu Chou
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Tsui-Ling Ko
- College of Science, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
| | - Yang C Fann
- Intramural IT and Bioinformatics Program, Division of Intramural, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shu-Hui Juan
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
10
|
Jahangiri L. Metastasis in Neuroblastoma and Its Link to Autophagy. Life (Basel) 2023; 13:life13030818. [PMID: 36983973 PMCID: PMC10056181 DOI: 10.3390/life13030818] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Neuroblastoma is a paediatric malignancy originating from the neural crest that commonly occurs in the abdomen and adrenal gland, leading to cancer-related deaths in children. Distant metastasis can be encountered at diagnosis in greater than half of these neuroblastoma patients. Autophagy, a self-degradative process, plays a key role in stress-related responses and the survival of cells and has been studied in neuroblastoma. Accordingly, in the early stages of metastasis, autophagy may suppress cancer cell invasion and migration, while its role may be reversed in later stages, and it may facilitate metastasis by enhancing cancer cell survival. To that end, a body of literature has revealed the mechanistic link between autophagy and metastasis in neuroblastoma in multiple steps of the metastatic cascade, including cancer cell invasion and migration, anoikis resistance, cancer cell dormancy, micrometastasis, and metastatic outbreak. This review aims to take a step forward and discuss the significance of multiple molecular players and compounds that may link autophagy to metastasis and map their function to various metastatic steps in neuroblastoma.
Collapse
Affiliation(s)
- Leila Jahangiri
- School of Science and Technology, Nottingham Trent University, Clifton Site, Nottingham NG11 8NS, UK
- Division of Cellular and Molecular Pathology, Department of Pathology, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
11
|
Li RX, Chen LY, Limbu SM, Yao B, Qian YF, Zhou WH, Chen LQ, Qiao F, Zhang ML, Du ZY, Luo Y. Atorvastatin remodels lipid distribution between liver and adipose tissues through blocking lipoprotein efflux in fish. Am J Physiol Regul Integr Comp Physiol 2023; 324:R281-R292. [PMID: 36572553 DOI: 10.1152/ajpregu.00222.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The regulation of cholesterol metabolism in fish is still unclear. Statins play important roles in promoting cholesterol metabolism development in mammals. However, studies on the role of statins in cholesterol metabolism in fish are currently limited. The present study evaluated the effects of statins on cholesterol metabolism in fish. Nile tilapia (Oreochromis niloticus) were fed on control diets supplemented with three atorvastatin levels (0, 12, and 24 mg/kg diet, ATV0, ATV12, and ATV24, respectively) for 4 wk. Intriguingly, the results showed that both atorvastatin treatments increased hepatic cholesterol and triglyceride contents mainly through inhibiting bile acid synthesis and efflux, and compensatorily enhancing cholesterol synthesis in fish liver (P < 0.05). Moreover, atorvastatin treatment significantly inhibited hepatic very-low-density lipoprotein (VLDL) assembly and thus decreased serum VLDL content (P < 0.05). However, fish treated with atorvastatin significantly reduced cholesterol and triglycerides contents in adipose tissue (P < 0.05). Further molecular analysis showed that atorvastatin treatment promoted cholesterol synthesis and lipogenesis pathways, but inhibited lipid catabolism and low-density lipoprotein (LDL) uptake in the adipose tissue of fish (P < 0.05). In general, atorvastatin induced the remodeling of lipid distribution between liver and adipose tissues through blocking VLDL efflux from the liver to adipose tissue of fish. Our results provide a novel regulatory pattern of cholesterol metabolism response caused by atorvastatin in fish, which is distinct from mammals: cholesterol inhibition by atorvastatin activates hepatic cholesterol synthesis and inhibits its efflux to maintain cholesterol homeostasis, consequently reduces cholesterol storage in fish adipose tissue.
Collapse
Affiliation(s)
- Rui-Xin Li
- LANEH, School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| | - Ling-Yun Chen
- LANEH, School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| | - Samwel M Limbu
- Department of Aquaculture Technology, School of Aquatic Sciences and Fisheries Technology, University of Dar es Salaam, Dar es Salaam, Tanzania
| | - Bing Yao
- LANEH, School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| | - Yi-Fan Qian
- LANEH, School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| | - Wen-Hao Zhou
- LANEH, School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| | - Li-Qiao Chen
- LANEH, School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| | - Fang Qiao
- LANEH, School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| | - Mei-Ling Zhang
- LANEH, School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| | - Zhen-Yu Du
- LANEH, School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| | - Yuan Luo
- LANEH, School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| |
Collapse
|
12
|
Atorvastatin induces downregulation of matrix metalloproteinase-2/9 in MDA-MB-231 triple negative breast cancer cells. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 40:22. [PMID: 36445561 DOI: 10.1007/s12032-022-01880-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 10/29/2022] [Indexed: 11/30/2022]
Abstract
Matrix metalloproteinases (MMPs) are a family of endopeptidases, mainly responsible of extracellular tissue remodeling. Abundant expression of MMPs leads to a number of tumorigenic processes including proliferation, angiogenesis, metastasis and invasion. Therefore, suppressing MMP expression is particularly important in cancer. Atorvastatin is a member of statin family, with cholesterol-lowering properties. Recently, it has emerged as a potential anticancer agent. Multiple researchers have reported promising results of atorvastatin use in cancer therapies. However, its effect on the expression of matrix metalloproteinases in breast cancer is unknown. In the present study, we have confirmed the apoptotic activity of atorvastatin on highly metastatic MDA-MB-231 triple negative breast cancer cells and investigated the gene expression of MMP-2/9. In this regard, MTT analysis was performed to evaluate cytotoxicity. Apoptotic activity was assessed by Annexin V binding and multicaspase assays. Western blot analysis was used to detect the apoptosis-related proteins. RT-PCR analysis was performed to evaluate the mRNA expression levels of MMP-2/9. Results indicated that atorvastatin reduces cell viability significantly at 5 µM after 48 h of treatment (p < 0.0001). It also induces caspase-dependent apoptosis, alters the expression of Bax and Bcl-2 in favour of apoptosis and stimulates cell cycle arrest at S phase (p < 0.05). Moreover, atorvastatin downregulates the mRNA expression of MMP-2 and MMP-9 significantly (p < 0.05). In conclusion, these results demonstrate for the first time that atorvastatin inhibits MMP-2 and MMP-9 gene expression in MDA-MB-231 cells, in addition to inducing caspase-dependent apoptosis.
Collapse
|
13
|
Maxwell ZA, Suazo KF, Brown HM, Distefano MD, Arriaga EA. Combining Isoprenoid Probes with Antibody Markers for Mass Cytometric Analysis of Prenylation in Single Cells. Anal Chem 2022; 94:11521-11528. [PMID: 35952372 PMCID: PMC9441216 DOI: 10.1021/acs.analchem.2c01509] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Protein prenylation is an essential post-translational modification that plays a key role in facilitating protein localization. Aberrations in protein prenylation have been indicated in multiple disease pathologies including progeria, some forms of cancer, and Alzheimer's disease. While there are single-cell methods to study prenylation, these methods cannot simultaneously assess prenylation and other cellular changes in the complex cell environment. Here, we report a novel method to monitor, at the single-cell level, prenylation and expression of autophagy markers. An isoprenoid analogue containing a terminal alkyne, substrate of prenylation enzymes, was metabolically incorporated into cells in culture. Treatment with a terbium reporter containing an azide functional group, followed by copper-catalyzed azide-alkyne cycloaddition, covalently attached terbium ions to prenylated proteins within cells. In addition, simultaneous treatment with a holmium-containing analogue of the reporter, without an azide functional group, was used to correct for non-specific retention at the single-cell level. This procedure was compatible with other mass cytometric sample preparation steps that use metal-tagged antibodies. We demonstrate that this method reports changes in levels of prenylation in competitive and inhibitor assays, while tracking autophagy molecular markers with metal-tagged antibodies. The method reported here makes it possible to track prenylation along with other molecular pathways in single cells of complex systems, which is essential to elucidate the role of this post-translational modification in disease, cell response to pharmacological treatments, and aging.
Collapse
|
14
|
Farnesyl diphosphate synthase regulated endothelial proliferation and autophagy during rat pulmonary arterial hypertension induced by monocrotaline. Mol Med 2022; 28:94. [PMID: 35962329 PMCID: PMC9373289 DOI: 10.1186/s10020-022-00511-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 07/13/2022] [Indexed: 11/10/2022] Open
Abstract
Background The proliferation ability and autophagy level of pulmonary artery endothelial cells (PAECs) play an important role in promoting the development of pulmonary artery hypertension (PAH), and there is still no effective treatment for PAH. Farnesyl diphosphate synthase (FDPS) is a key enzyme in the mevalonate pathway. The intermediate metabolites of this pathway are closely related to the activity of autophagy-associated small G proteins, including Ras-related C3 botulinum toxin substrate 1 (Rac1). Studies have shown that the mevalonate pathway affects the activation levels of different small G proteins, autophagy signaling pathways, vascular endothelial function, and so on. However, the exact relationship between them is still unclear in PAH. Method In vitro, western blotting and mRFP-GFP-LC3 puncta formation assays were used to observe the expression of FDPS and the level of autophagy in PAECs treated with monocrotaline pyrrole (MCTP). In addition, cell proliferation and migration assays were used to assess the effect of FDPS on endothelial function, and Rac1 activity assays were used to evaluate the effect of Rac1 activation on PAEC autophagy via the PI3K/AKT/mTOR signaling pathway. In vivo, the right heart catheterization method, hematoxylin and eosin (H&E) staining and western blotting were used to determine the effect of FDPS on PAEC autophagy and monocrotaline (MCT)-induced PAH. Results We show that the expression of FDPS is increased in the PAH module in vitro and in vivo, concomitant with the induction of autophagy and the activation of Rac1. Our data demonstrate that inhibition of FDPS ameliorates endothelial function and decreases MCT-induced autophagy levels. Mechanistically, we found that FDPS promotes autophagy, Rac1 activity and endothelial disfunction through the PI3K/AKT/mTOR signaling pathway. Conclusion Our study suggests that FDPS contributes to active small G protein-induced autophagy during MCT-induced PAH, which may serve as a potential therapeutic target against PAH. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00511-7.
Collapse
|
15
|
Behrouj H, Vakili O, Sadeghdoust A, Aligolighasemabadi N, Khalili P, Zamani M, Mokarram P. Epigenetic regulation of autophagy in coronavirus disease 2019 (COVID-19). Biochem Biophys Rep 2022; 30:101264. [PMID: 35469237 PMCID: PMC9021360 DOI: 10.1016/j.bbrep.2022.101264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 11/22/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has become the most serious global public health issue in the past two years, requiring effective therapeutic strategies. This viral infection is a contagious disease caused by new coronaviruses (nCoVs), also called severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Autophagy, as a highly conserved catabolic recycling process, plays a significant role in the growth and replication of coronaviruses (CoVs). Therefore, there is great interest in understanding the mechanisms that underlie autophagy modulation. The modulation of autophagy is a very complex and multifactorial process, which includes different epigenetic alterations, such as histone modifications and DNA methylation. These mechanisms are also known to be involved in SARS-CoV-2 replication. Thus, molecular understanding of the epigenetic pathways linked with autophagy and COVID-19, could provide novel therapeutic targets for COVID-19 eradication. In this context, the current review highlights the role of epigenetic regulation of autophagy in controlling COVID-19, focusing on the potential therapeutic implications.
Collapse
Affiliation(s)
- Hamid Behrouj
- Behbahan Faculty of Medical Sciences, Behbahan, Iran
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Adel Sadeghdoust
- Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Neda Aligolighasemabadi
- Department of Internal Medicine, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Parnian Khalili
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mozhdeh Zamani
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pooneh Mokarram
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Iran
| |
Collapse
|
16
|
Novel Effects of Statins on Cancer via Autophagy. Pharmaceuticals (Basel) 2022; 15:ph15060648. [PMID: 35745567 PMCID: PMC9228383 DOI: 10.3390/ph15060648] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 02/05/2023] Open
Abstract
Cancer is one of the main causes of death globally. Most of the molecular mechanisms underlying cancer are marked by complex aberrations that activate the critical cell-signaling pathways that play a pivotal role in cell metabolism, tumor development, cytoskeletal reorganization, and metastasis. The phosphatidylinositol 3-kinase/protein kinase-B/mammalian target of the rapamycin (PI3K/AKT/mTOR) pathway is one of the main signaling pathways involved in carcinogenesis and metastasis. Autophagy, a cellular pathway that delivers cytoplasmic components to lysosomes for degradation, plays a dual role in cancer, as either a tumor promoter or a tumor suppressor, depending on the stage of the carcinogenesis. Statins are the group of drugs of choice to lower the level of low-density lipoprotein (LDL) cholesterol in the blood. Experimental and clinical data suggest the potential of statins in the treatment of cancer. In vitro and in vivo studies have demonstrated the molecular mechanisms through which statins inhibit the proliferation and metastasis of cancer cells in different types of cancer. The anticancer properties of statins have been shown to result in the suppression of tumor growth, the induction of apoptosis, and autophagy. This literature review shows the dual role of the autophagic process in cancer and the latest scientific evidence related to the inducing effect exerted by statins on autophagy, which could explain their anticancer potential.
Collapse
|
17
|
Goenka L, Dubashi B, Selvarajan S, Ganesan P. Use of "Repurposed" Drugs in the Treatment of Epithelial Ovarian Cancer: A Systematic Review. Am J Clin Oncol 2022; 45:168-174. [PMID: 35320817 DOI: 10.1097/coc.0000000000000900] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Epithelial ovarian cancer has poor outcomes with standard therapy and limited options for treatment of recurrent disease. This systematic review summarizes the data on the clinical use of repurposed drugs. We searched for clinical studies using "repurposed" agents for the treatment of ovarian cancer in the following databases: PubMed, clinicaltrials.gov, Clinical Trial Registry of India, European Clinical Trials Registry, and Chinese Clinical Trial Registry. We excluded reviews, preclinical studies, and non-English language studies. We assessed the quality of included studies. The following agents/class of agents were included: statins, hydroxychloroquine, metformin, itraconazole, nonsteroidal anti-inflammatory drugs, vitamin D, proton pump inhibitors, beta-blockers, and sodium valproate. Only one randomized controlled trial investigated metformin, which found no benefit of metformin. However, this had a high risk of bias (no details of randomization). Among the observational studies, 70% were of high quality (Newcastle-Ottawa scale ≥7). Clinical benefit was seen for itraconazole, beta-blockers, metformin, statins, and proton pump inhibitors. Though multiple studies aim to repurpose agents in epithelial ovarian cancer, the most published literature is observational, and none are practice-changing. Given the solid preclinical data regarding the anticancer efficacy of these agents, well-designed clinical trials are urgently required.
Collapse
|
18
|
Silva RCMC, Ribeiro JS, da Silva GPD, da Costa LJ, Travassos LH. Autophagy Modulators in Coronavirus Diseases: A Double Strike in Viral Burden and Inflammation. Front Cell Infect Microbiol 2022; 12:845368. [PMID: 35433503 PMCID: PMC9010404 DOI: 10.3389/fcimb.2022.845368] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/02/2022] [Indexed: 12/12/2022] Open
Abstract
Coronaviruses are the etiologic agents of several diseases. Coronaviruses of critical medical importance are characterized by highly inflammatory pathophysiology, involving severe pulmonary impairment and infection of multiple cell types within the body. Here, we discuss the interplay between coronaviruses and autophagy regarding virus life cycle, cell resistance, and inflammation, highlighting distinct mechanisms by which autophagy restrains inflammatory responses, especially those involved in coronavirus pathogenesis. We also address different autophagy modulators available and the rationale for drug repurposing as an attractive adjunctive therapy. We focused on pharmaceuticals being tested in clinical trials with distinct mechanisms but with autophagy as a common target. These autophagy modulators act in cell resistance to virus infection and immunomodulation, providing a double-strike to prevent or treat severe disease development and death from coronaviruses diseases.
Collapse
Affiliation(s)
- Rafael Cardoso Maciel Costa Silva
- Laboratório de Imunoreceptores e Sinalização Celular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jhones Sousa Ribeiro
- Laboratório de Imunoreceptores e Sinalização Celular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gustavo Peixoto Duarte da Silva
- Laboratório de Genética e Imunologia das Infecções Virais, Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana Jesus da Costa
- Laboratório de Genética e Imunologia das Infecções Virais, Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Holanda Travassos
- Laboratório de Imunoreceptores e Sinalização Celular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
19
|
Eshraghi M, Ahmadi M, Afshar S, Lorzadeh S, Adlimoghaddam A, Rezvani Jalal N, West R, Dastghaib S, Igder S, Torshizi SRN, Mahmoodzadeh A, Mokarram P, Madrakian T, Albensi BC, Łos MJ, Ghavami S, Pecic S. Enhancing autophagy in Alzheimer's disease through drug repositioning. Pharmacol Ther 2022; 237:108171. [PMID: 35304223 DOI: 10.1016/j.pharmthera.2022.108171] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/18/2022] [Accepted: 03/08/2022] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is one of the biggest human health threats due to increases in aging of the global population. Unfortunately, drugs for treating AD have been largely ineffective. Interestingly, downregulation of macroautophagy (autophagy) plays an essential role in AD pathogenesis. Therefore, targeting autophagy has drawn considerable attention as a therapeutic approach for the treatment of AD. However, developing new therapeutics is time-consuming and requires huge investments. One of the strategies currently under consideration for many diseases is "drug repositioning" or "drug repurposing". In this comprehensive review, we have provided an overview of the impact of autophagy on AD pathophysiology, reviewed the therapeutics that upregulate autophagy and are currently used in the treatment of other diseases, including cancers, and evaluated their repurposing as a possible treatment option for AD. In addition, we discussed the potential of applying nano-drug delivery to neurodegenerative diseases, such as AD, to overcome the challenge of crossing the blood brain barrier and specifically target molecules/pathways of interest with minimal side effects.
Collapse
Affiliation(s)
- Mehdi Eshraghi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada
| | - Mazaher Ahmadi
- Faculty of Chemistry, Bu-Ali Sina University, Hamedan, Iran; Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeid Afshar
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Shahrokh Lorzadeh
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada
| | - Aida Adlimoghaddam
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; St. Boniface Hospital Albrechtsen Research Centre, Division of Neurodegenerative Disorders, Winnipeg, MB R2H2A6, Canada
| | | | - Ryan West
- Department of Chemistry and Biochemistry, California State University, Fullerton, United States of America
| | - Sanaz Dastghaib
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz Iran
| | - Somayeh Igder
- Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Amir Mahmoodzadeh
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Pooneh Mokarram
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tayyebeh Madrakian
- Faculty of Chemistry, Bu-Ali Sina University, Hamedan, Iran; Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Benedict C Albensi
- St. Boniface Hospital Albrechtsen Research Centre, Division of Neurodegenerative Disorders, Winnipeg, MB R2H2A6, Canada; Nova Southeastern Univ. College of Pharmacy, Davie, FL, United States of America; University of Manitoba, College of Medicine, Winnipeg, MB R3E 0V9, Canada
| | - Marek J Łos
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 0V9, Canada; Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada; Faculty of Medicine in Zabrze, University of Technology in Katowice, Academia of Silesia, 41-800 Zabrze, Poland
| | - Stevan Pecic
- Department of Chemistry and Biochemistry, California State University, Fullerton, United States of America.
| |
Collapse
|
20
|
Fluvastatin sensitizes pancreatic cancer cells toward radiation therapy and suppresses radiation- and/or TGF-β-induced tumor-associated fibrosis. J Transl Med 2022; 102:298-311. [PMID: 34773069 DOI: 10.1038/s41374-021-00690-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 11/08/2022] Open
Abstract
Pancreatic cancer (PC) is highly resistant to chemo and radiotherapy. Radiation-induced fibrosis (RIF) is a major cause of clinical concern for various malignancies, including PC. In this study, we aimed to evaluate the radiosensitizing and anti-RIF potential of fluvastatin in PC. Short-term viability and clonogenic survival assays were used to evaluate the radiosensitizing potential of fluvastatin in multiple human and murine PC cell lines. The expression of different proteins was analyzed to understand the mechanisms of fluvastatin-mediated radiosensitization of PC cells and its anti-RIF effects in both mouse and human pancreatic stellate cells (PSCs). Finally, these effects of fluvastatin and/or radiation were assessed in an immune-competent syngeneic murine model of PC. Fluvastatin radiosensitized multiple PC cell lines, as well as radioresistant cell lines in vitro, by inhibiting radiation-induced DNA damage repair response. Nonmalignant cells, such as PSCs and NIH3T3 cells, were less sensitive to fluvastatin-mediated radiosensitization than PC cells. Interestingly, fluvastatin suppressed radiation and/or TGF-β-induced activation of PSCs, as well as the fibrogenic properties of these cells in vitro. Fluvastatin considerably augmented the antitumor effect of external radiation therapy and also suppressed intra-tumor RIF in vivo. These findings suggested that along with radiation, fluvastatin co-treatment may be a potential therapeutic approach against PC.
Collapse
|
21
|
Taruselli MT, Kolawole EM, Qayum AA, Haque TT, Caslin HL, Abebayehu D, Kee SA, Dailey JM, Jackson KG, Burchett JR, Spence AJ, Pondicherry N, Barnstein BO, Gomez G, Straus DB, Ryan JJ. Fluvastatin enhances IL-33-mediated mast cell IL-6 and TNF production. Cell Immunol 2022; 371:104457. [PMID: 34883342 PMCID: PMC8782378 DOI: 10.1016/j.cellimm.2021.104457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 01/03/2023]
Abstract
Statins are HMG-CoA reductase inhibitors prescribed for lowering cholesterol. They can also inhibit inflammatory responses by suppressing isoprenylation of small G proteins. Consistent with this, we previously found that fluvastatin suppresses IgE-mediated mast cell function. However, some studies have found that statins induced pro-inflammatory cytokines in macrophages and NK cells. In contrast to IgE signaling, we show that fluvastatin augments IL-33-induced TNF and IL-6 production by mast cells. This effect required the key mast cell growth factor, stem cell factor (SCF). Treatment of IL-33-activated mast cells with mevalonic acid or isoprenoids reduced fluvastatin effects, suggesting fluvastatin acts at least partly by reducing isoprenoid production. Fluvastatin also enhanced IL-33-induced NF-κB transcriptional activity and promoted neutrophilic peritonitis in vivo, a response requiring mast cell activation. Other statins tested did not enhance IL-33 responsiveness. Therefore, this work supports observations of unexpected pro-inflammatory effects of some statins and suggests mechanisms by which this may occur. Because statins are candidates for repurposing in inflammatory disorders, our work emphasizes the importance of understanding the pleiotropic and possible unexpected effects of these drugs.
Collapse
Affiliation(s)
- Marcela T Taruselli
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, United States
| | | | - Amina Abdul Qayum
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Tamara T Haque
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Heather L Caslin
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, United States
| | - Daniel Abebayehu
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Sydney A Kee
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, United States
| | - Jordan M Dailey
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Kaitlyn G Jackson
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Jason R Burchett
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, United States
| | - Andrew J Spence
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, United States
| | - Neha Pondicherry
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, United States
| | - Brian O Barnstein
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, United States
| | - Gregorio Gomez
- University of Houston College of Medicine, Department of Biomedical Sciences, Houston, TX 77204, United States
| | - David B Straus
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, United States
| | - John J Ryan
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, United States.
| |
Collapse
|
22
|
Argininosuccinate lyase is a metabolic vulnerability in breast development and cancer. NPJ Syst Biol Appl 2021; 7:36. [PMID: 34535676 PMCID: PMC8448827 DOI: 10.1038/s41540-021-00195-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 08/09/2021] [Indexed: 02/07/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is fundamental to both normal tissue development and cancer progression. We hypothesized that EMT plasticity defines a range of metabolic phenotypes and that individual breast epithelial metabolic phenotypes are likely to fall within this phenotypic landscape. To determine EMT metabolic phenotypes, the metabolism of EMT was described within genome-scale metabolic models (GSMMs) using either transcriptomic or proteomic data from the breast epithelial EMT cell culture model D492. The ability of the different data types to describe breast epithelial metabolism was assessed using constraint-based modeling which was subsequently verified using 13C isotope tracer analysis. The application of proteomic data to GSMMs provided relatively higher accuracy in flux predictions compared to the transcriptomic data. Furthermore, the proteomic GSMMs predicted altered cholesterol metabolism and increased dependency on argininosuccinate lyase (ASL) following EMT which were confirmed in vitro using drug assays and siRNA knockdown experiments. The successful verification of the proteomic GSMMs afforded iBreast2886, a breast GSMM that encompasses the metabolic plasticity of EMT as defined by the D492 EMT cell culture model. Analysis of breast tumor proteomic data using iBreast2886 identified vulnerabilities within arginine metabolism that allowed prognostic discrimination of breast cancer patients on a subtype-specific level. Taken together, we demonstrate that the metabolic reconstruction iBreast2886 formalizes the metabolism of breast epithelial cell development and can be utilized as a tool for the functional interpretation of high throughput clinical data.
Collapse
|
23
|
Gupta P, Kumar N, Garg M. Emerging roles of autophagy in the development and treatment of urothelial carcinoma of the bladder. Expert Opin Ther Targets 2021; 25:787-797. [PMID: 34636265 DOI: 10.1080/14728222.2021.1992384] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 10/08/2021] [Indexed: 01/07/2023]
Abstract
INTRODUCTION High recurrence rates, frequent surveillance strategies, and current multidisciplinary treatment approaches make urothelial carcinoma of bladder (UCB) one of the most expensive cancers to clinically manage. Recent studies have demonstrated a role for autophagy in bladder tumorigenesis. It serves as a tumor suppressor by maintaining genomic integrity and preventing tumor proliferation during initial stages of tumor development. Nevertheless, once established, cancer cells may utilize protective autophagy to endure cellular stress and survive in the adverse environment. Its excessive stimulation supports cancer cells' resistance to therapeutic modalities. AREAS COVERED PubMed and Google Scholar electronic databases were searched for recently published studies. This review summarizes emerging roles of autophagy in development/progression of UCB and treatment resistance and explores novel therapeutic targets for prevention of cancer invasion, metastatic spread', and disease relapse. EXPERT OPINION The development of novel therapies via targeting of autophagy may augment current treatment regimens and improve clinical outcomes. Synthetic compounds or plant-based metabolites are reported to enhance cancer therapies by modulating autophagic flux. Successful autophagy-focused therapeutic intervention requires a mechanistic understanding of autophagic effects on tumor initiation and progression and the development of efficient biomarkers to monitor it in tumor tissues.
Collapse
Affiliation(s)
- Pratishtha Gupta
- Department of Biochemistry, University of Lucknow, Lucknow, India
| | - Niraj Kumar
- Translational Health Science and Technology Institute (THSTI), Faridabad, India
| | - Minal Garg
- Department of Biochemistry, University of Lucknow, Lucknow, India
| |
Collapse
|
24
|
Xie L, Zhu G, Shang J, Chen X, Zhang C, Ji X, Zhang Q, Wei Y. An overview on the biological activity and anti-cancer mechanism of lovastatin. Cell Signal 2021; 87:110122. [PMID: 34438015 DOI: 10.1016/j.cellsig.2021.110122] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 02/07/2023]
Abstract
Lovastatin, a secondary metabolite isolated from fungi, is often used as a representative drug to reduce blood lipid concentration and treat hypercholesterolemia. Its structure is similar to that of HMG-CoA. Lovastatin inhibits the binding of the substrate to HMG-CoA reductase, and strongly competes with HMG-CoA reductase (HMGR), thereby exerting a hypolipidemic effect. Further, its safety has been confirmed in vivo and in vitro. Lovastatin also has anti-inflammatory, anti-cancer, and neuroprotective effects. Therefore, the biological activity of lovastatin, especially its anti-cancer effect, has garnered research attention. Several in vitro studies have confirmed that lovastatin has a significant inhibitory effect on cancer cell viability in a variety of cancers (such as breast, liver, cervical, lung, and colon cancer). At the same time, lovastatin can also increase the sensitivity of some types of cancer cells to chemotherapeutic drugs and strengthen their therapeutic effect. Lovastatin inhibits cell proliferation and regulates cancer cell signaling pathways, thereby inducing apoptosis and cell cycle arrest. This article reviews the structure, biosynthetic pathways, and applications of lovastatin, focusing on the anti-cancer effects and mechanisms of action.
Collapse
Affiliation(s)
- Liguo Xie
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China.
| | - Guodong Zhu
- Yunnan Minzu University, Library, Kunming 650500, China.
| | - Junjie Shang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China.
| | - Xuemei Chen
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China.
| | - Chunting Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China.
| | - Xiuling Ji
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China.
| | - Qi Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China.
| | - Yunlin Wei
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China.
| |
Collapse
|
25
|
Huang P, Zhu S, Liang X, Zhang Q, Liu C, Song L. Revisiting Lung Cancer Metastasis: Insight From the Functions of Long Non-coding RNAs. Technol Cancer Res Treat 2021; 20:15330338211038488. [PMID: 34431723 PMCID: PMC8392855 DOI: 10.1177/15330338211038488] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Globally, lung cancer is the most common cause of cancer-related deaths. After
diagnosis at all stages, <7% of patients survive for 10 years. Thus,
diagnosis at later stages and the lack of effective and personalized drugs
reflect a significant need to better understand the mechanisms underpinning lung
cancer progression. Metastasis should be responsible for the high lethality and
recurrence rates seen in lung cancer. Metastasis depends on multiple crucial
steps, including epithelial–mesenchymal transition, vascular remodeling, and
colonization. Therefore, in-depth investigations of metastatic molecular
mechanisms can provide valuable insights for lung cancer treatment. Recently,
long noncoding RNAs (lncRNAs) have attracted considerable attention owing to
their complex roles in cancer progression. In lung cancer, multiple lncRNAs have
been reported to regulate metastasis. In this review, we highlight the major
molecular mechanisms underlying lncRNA-mediated regulation of lung cancer
metastasis, including (1) lncRNAs acting as competing endogenous RNAs, (2)
lncRNAs regulating the transduction of several signal pathways, and (3) lncRNA
coordination with enhancer of zeste homolog 2. Thus, lncRNAs appear to execute
their functions on lung cancer metastasis by regulating angiogenesis, autophagy,
aerobic glycolysis, and immune escape. However, more comprehensive studies are
required to characterize these lncRNA regulatory networks in lung cancer
metastasis, which can provide promising and innovative novel therapeutic
strategies to combat this disease.
Collapse
Affiliation(s)
- Peng Huang
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
| | - Shaomi Zhu
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
| | - Xin Liang
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
| | - Qinxiu Zhang
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
| | - Chi Liu
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
| | - Linjiang Song
- Reproductive & Women-Children Hospital, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, P.R. China
| |
Collapse
|
26
|
Almasi S, Crawford Parks TE, Ravel-Chapuis A, MacKenzie A, Côté J, Cowan KN, Jasmin BJ. Differential regulation of autophagy by STAU1 in alveolar rhabdomyosarcoma and non-transformed skeletal muscle cells. Cell Oncol (Dordr) 2021; 44:851-870. [PMID: 33899158 DOI: 10.1007/s13402-021-00607-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2021] [Indexed: 11/30/2022] Open
Abstract
PURPOSE Recent work has highlighted the therapeutic potential of targeting autophagy to modulate cell survival in a variety of diseases including cancer. Recently, we found that the RNA-binding protein Staufen1 (STAU1) is highly expressed in alveolar rhabdomyosarcoma (ARMS) and that this abnormal expression promotes tumorigenesis. Here, we asked whether STAU1 is involved in the regulation of autophagy in ARMS cells. METHODS We assessed the impact of STAU1 expression modulation in ARMS cell lines (RH30 and RH41), non-transformed skeletal muscle cells (C2C12) and STAU1-transgenic mice using complementary techniques. RESULTS We found that STAU1 silencing reduces autophagy in the ARMS cell lines RH30 and RH41, while increasing their apoptosis. Mechanistically, this inhibitory effect was found to be caused by a direct negative impact of STAU1 depletion on the stability of Beclin-1 (BECN1) and ATG16L1 mRNAs, as well as by an indirect inhibition of JNK signaling via increased expression of Dual specificity phosphatase 8 (DUSP8). Pharmacological activation of JNK or expression silencing of DUSP8 was sufficient to restore autophagy in STAU1-depleted cells. By contrast, we found that STAU1 downregulation in non-transformed skeletal muscle cells activates autophagy in a mTOR-dependent manner, without promoting apoptosis. A similar effect was observed in skeletal muscles obtained from STAU1-overexpressing transgenic mice. CONCLUSIONS Together, our data indicate an effect of STAU1 on autophagy regulation in ARMS cells and its differential role in non-transformed skeletal muscle cells. Our findings suggest a cancer-specific potential of targeting STAU1 for the treatment of ARMS.
Collapse
Affiliation(s)
- Shekoufeh Almasi
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada.,The Eric J. Poulin Centre for Neuromuscular Diseases, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Tara E Crawford Parks
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada.,The Eric J. Poulin Centre for Neuromuscular Diseases, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Aymeric Ravel-Chapuis
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada.,The Eric J. Poulin Centre for Neuromuscular Diseases, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Alex MacKenzie
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada.,The Eric J. Poulin Centre for Neuromuscular Diseases, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Division of Endocrinology, Department of Paediatric, CHEO, University of Ottawa, Ottawa, ON, Canada
| | - Jocelyn Côté
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada.,The Eric J. Poulin Centre for Neuromuscular Diseases, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Kyle N Cowan
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada.,Division of Paediatric Surgery, Department of Surgery, CHEO, University of Ottawa, Ottawa, ON, Canada.,Molecular Biomedicine Program, CHEO, Ottawa, ON, Canada
| | - Bernard J Jasmin
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada. .,The Eric J. Poulin Centre for Neuromuscular Diseases, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
27
|
Lunder M, Janić M, Šabovič M. Treating Arterial Ageing in Patients with Diabetes: From Mechanisms to Effective Drugs. Int J Mol Sci 2021; 22:ijms22062796. [PMID: 33801956 PMCID: PMC8001638 DOI: 10.3390/ijms22062796] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 12/17/2022] Open
Abstract
Diabetes mellitus is a major healthcare problem. It is not only characterized by hyperglycemia and chronic complications, but in longer lasting diabetes and a longer living population, it is also associated with accelerated arterial ageing, which importantly contributes to cardiovascular complications. The accelerated arterial ageing in patients with diabetes should be considered separately from arterial ageing in patients without diabetes. Basic and clinical research have allowed better insight into the mechanisms of arterial ageing. In a simplified mechanistic way, it could be considered that the three tightly connected cornerstone characteristics of arterial ageing in patients with diabetes are: phenotypic presentation as endothelial dysfunction and arterial stiffness, and the underlying basic ageing-facilitating mechanism represented as the impaired expression of genetic longevity pathways. Currently, specific drugs for preventing/treating arterial ageing are not available. Therefore, we aimed to review the capacity of available drugs, particularly antidiabetic drugs, to interfere with the arterial ageing process. In the near future, these characteristics could help to guide therapy in patients with diabetes. Overall, it appears that arterial ageing could become a new target in diabetes. The expanding knowledge regarding the capability of antidiabetic drugs and other available drugs to inhibit/delay arterial aging is therefore essential.
Collapse
Affiliation(s)
- Mojca Lunder
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, Zaloška cesta 7, SI-1000 Ljubljana, Slovenia; (M.L.); (M.J.)
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - Miodrag Janić
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, Zaloška cesta 7, SI-1000 Ljubljana, Slovenia; (M.L.); (M.J.)
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - Mišo Šabovič
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
- Department of Vascular Diseases, University Medical Centre Ljubljana, Zaloška cesta 7, SI-1000 Ljubljana, Slovenia
- Correspondence: ; Tel.: +386-15228032; Fax: +386-15228070
| |
Collapse
|
28
|
Feltrin S, Ravera F, Traversone N, Ferrando L, Bedognetti D, Ballestrero A, Zoppoli G. Sterol synthesis pathway inhibition as a target for cancer treatment. Cancer Lett 2020; 493:19-30. [DOI: 10.1016/j.canlet.2020.07.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 07/05/2020] [Accepted: 07/09/2020] [Indexed: 12/21/2022]
|
29
|
Rodrigues‐Diez RR, Tejera‐Muñoz A, Marquez‐Exposito L, Rayego‐Mateos S, Santos Sanchez L, Marchant V, Tejedor Santamaria L, Ramos AM, Ortiz A, Egido J, Ruiz‐Ortega M. Statins: Could an old friend help in the fight against COVID-19? Br J Pharmacol 2020; 177:4873-4886. [PMID: 32562276 PMCID: PMC7323198 DOI: 10.1111/bph.15166] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/01/2020] [Accepted: 06/10/2020] [Indexed: 12/21/2022] Open
Abstract
The COVID-19 pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has overwhelmed healthcare systems requiring the rapid development of treatments, at least, to reduce COVID-19 severity. Drug repurposing offers a fast track. Here, we discuss the potential beneficial effects of statins in COVID-19 patients based on evidence that they may target virus receptors, replication, degradation, and downstream responses in infected cells, addressing both basic research and epidemiological information. Briefly, statins could modulate virus entry, acting on the SARS-CoV-2 receptors, ACE2 and CD147, and/or lipid rafts engagement. Statins, by inducing autophagy activation, could regulate virus replication or degradation, exerting protective effects. The well-known anti-inflammatory properties of statins, by blocking several molecular mechanisms, including NF-κB and NLRP3 inflammasomes, could limit the "cytokine storm" in severe COVID-19 patients which is linked to fatal outcome. Finally, statin moderation of coagulation response activation may also contribute to improving COVID-19 outcomes. LINKED ARTICLES: This article is part of a themed issue on The Pharmacology of COVID-19. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.21/issuetoc.
Collapse
Affiliation(s)
- Raul R. Rodrigues‐Diez
- Cellular and Molecular Biology in Renal and Vascular Pathology LaboratoryFundación Instituto de Investigación Sanitaria‐Fundación Jiménez Díaz‐Universidad Autónoma MadridMadridSpain
- Red de Investigación Renal (REDINREN)Instituto de Salud Carlos IIIMadridSpain
| | - Antonio Tejera‐Muñoz
- Cellular and Molecular Biology in Renal and Vascular Pathology LaboratoryFundación Instituto de Investigación Sanitaria‐Fundación Jiménez Díaz‐Universidad Autónoma MadridMadridSpain
- Red de Investigación Renal (REDINREN)Instituto de Salud Carlos IIIMadridSpain
| | - Laura Marquez‐Exposito
- Cellular and Molecular Biology in Renal and Vascular Pathology LaboratoryFundación Instituto de Investigación Sanitaria‐Fundación Jiménez Díaz‐Universidad Autónoma MadridMadridSpain
- Red de Investigación Renal (REDINREN)Instituto de Salud Carlos IIIMadridSpain
| | - Sandra Rayego‐Mateos
- Red de Investigación Renal (REDINREN)Instituto de Salud Carlos IIIMadridSpain
- GE‐06 Pathophysiology of Renal and Vascular Damage Laboratory, Maimonides Biomedical Research Institute of Cordoba (IMIBIC)University of CórdobaCórdobaSpain
| | - Laura Santos Sanchez
- Cellular and Molecular Biology in Renal and Vascular Pathology LaboratoryFundación Instituto de Investigación Sanitaria‐Fundación Jiménez Díaz‐Universidad Autónoma MadridMadridSpain
- Red de Investigación Renal (REDINREN)Instituto de Salud Carlos IIIMadridSpain
| | - Vanessa Marchant
- Cellular and Molecular Biology in Renal and Vascular Pathology LaboratoryFundación Instituto de Investigación Sanitaria‐Fundación Jiménez Díaz‐Universidad Autónoma MadridMadridSpain
- Red de Investigación Renal (REDINREN)Instituto de Salud Carlos IIIMadridSpain
| | - Lucía Tejedor Santamaria
- Cellular and Molecular Biology in Renal and Vascular Pathology LaboratoryFundación Instituto de Investigación Sanitaria‐Fundación Jiménez Díaz‐Universidad Autónoma MadridMadridSpain
- Red de Investigación Renal (REDINREN)Instituto de Salud Carlos IIIMadridSpain
| | - Adrian M. Ramos
- Red de Investigación Renal (REDINREN)Instituto de Salud Carlos IIIMadridSpain
- Laboratory of Nephrology and HypertensionFundación Instituto de Investigación Sanitaria‐Fundación Jiménez Díaz‐Universidad Autónoma MadridMadridSpain
| | - Alberto Ortiz
- Red de Investigación Renal (REDINREN)Instituto de Salud Carlos IIIMadridSpain
- Laboratory of Nephrology and HypertensionFundación Instituto de Investigación Sanitaria‐Fundación Jiménez Díaz‐Universidad Autónoma MadridMadridSpain
| | - Jesus Egido
- Renal, Vascular and Diabetes Research LaboratoryFundación Instituto de Investigación Sanitaria‐Fundación Jiménez Díaz Universidad AutónomaMadridSpain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)MadridSpain
| | - Marta Ruiz‐Ortega
- Cellular and Molecular Biology in Renal and Vascular Pathology LaboratoryFundación Instituto de Investigación Sanitaria‐Fundación Jiménez Díaz‐Universidad Autónoma MadridMadridSpain
- Red de Investigación Renal (REDINREN)Instituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
30
|
El-Ashmawy NE, Al-Ashmawy GM, Amr EA, Khedr EG. Inhibition of lovastatin- and docosahexaenoic acid-initiated autophagy in triple negative breast cancer reverted resistance and enhanced cytotoxicity. Life Sci 2020; 259:118212. [PMID: 32768581 DOI: 10.1016/j.lfs.2020.118212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 07/26/2020] [Accepted: 08/03/2020] [Indexed: 12/17/2022]
Abstract
AIMS Autophagy plays a complex role in breast cancer by suppressing or improving the efficiency of treatment. Triple-negative breast cancer (TNBC) cell line (MDA-MB-231) is associated with aggressive response and developing therapy resistance. MDA-MB-231 cells depend on autophagy for survival. Also, the potential benefits of autophagy inhibition in ameliorating developed chemotherapy resistance towards MDA-MB-231 remains to be elucidated. Despite showing anti-tumorigenic activities, the use of lovastatin and docosahexaenoic acid (DHA) for treating different types of cancers is still limited. We aimed to investigate the protective effect of autophagy inhibition by chloroquine (CQ) in MDA-MB-231 cells resistance treated with lovastatin or DHA. MAIN METHODS MDA-MB-231 cells were treated with 30 μM lovastatin and/or 100 μM DHA for 48 h plus 20 μM CQ. Autophagic flux was assessed in association with the expression of multidrug resistance gene 1 (MDR1), transforming growth factor beta 1 gene (TGF-β1), and autophagy-related 7 gene (ATG7). KEY FINDINGS Both drugs exhibited dose-dependent cytotoxicity, enhanced the autophagic flux represented by increased LC3BII protein concentration and decreased p62 protein concentration, and up-regulated the expression of MDR1, TGF-β1, and ATG7 genes. CQ addition enhanced the cytotoxicity of drugs and inhibited the autophagic flux which is detected by higher levels of LC3BII and p62 correlated with the reverted MDR1, TGF-β1 and ATG7 genes expression. SIGNIFICANCE Autophagy inhibition by CQ showed an ameliorative effect on lovastatin- and DHA-induced resistance and enhanced their cytotoxicity, providing a promising strategy in breast cancer therapy.
Collapse
Affiliation(s)
- Nahla E El-Ashmawy
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Egypt, Postal code: 31527.
| | - Ghada M Al-Ashmawy
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Egypt, Postal code: 31527.
| | - Eman A Amr
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Egypt, Postal code: 31527.
| | - Eman G Khedr
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Egypt, Postal code: 31527.
| |
Collapse
|
31
|
Paez PA, Kolawole M, Taruselli MT, Ajith S, Dailey JM, Kee SA, Haque TT, Barnstein BO, McLeod JJA, Caslin HL, Kiwanuka KN, Fukuoka Y, Le QT, Schwartz LB, Straus DB, Gewirtz DA, Martin RK, Ryan JJ. Fluvastatin Induces Apoptosis in Primary and Transformed Mast Cells. J Pharmacol Exp Ther 2020; 374:104-112. [PMID: 32434944 PMCID: PMC7306917 DOI: 10.1124/jpet.119.264234] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 04/01/2020] [Indexed: 12/18/2022] Open
Abstract
Statin drugs are widely employed in the clinic to reduce serum cholesterol. Because of their hydroxymethylglutaryl coenzyme A reductase antagonism, statins also reduce isoprenyl lipids necessary for the membrane anchorage and signaling of small G-proteins in the Ras superfamily. We previously found that statins suppress immunoglobulin E (IgE)-mediated mast cell activation, suggesting these drugs might be useful in treating allergic disease. Although IgE-induced function is critical to allergic inflammation, mast cell proliferation and survival also impact atopic disease and mast cell neoplasia. In this study, we describe fluvastatin-mediated apoptosis in primary and transformed mast cells. An IC50 was achieved between 0.8 and 3.5 μM in both cell types, concentrations similar to the reported fluvastatin serum Cmax value. Apoptosis was correlated with reduced stem cell factor (SCF)-mediated signal transduction, mitochondrial dysfunction, and caspase activation. Complementing these data, we found that p53 deficiency or Bcl-2 overexpression reduced fluvastatin-induced apoptosis. We also noted evidence of cytoprotective autophagy in primary mast cells treated with fluvastatin. Finally, we found that intraperitoneal fluvastatin treatment reduced peritoneal mast cell numbers in vivo These findings offer insight into the mechanisms of mast cell survival and support the possible utility of statins in mast cell-associated allergic and neoplastic diseases. SIGNIFICANCE STATEMENT: Fluvastatin, a statin drug used to lower cholesterol, induces apoptosis in primary and transformed mast cells by antagonizing protein isoprenylation, effectively inhibiting stem cell factor (SCF)-induced survival signals. This drug may be an effective means of suppressing mast cell survival.
Collapse
Affiliation(s)
- Patrick A Paez
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Motunrayo Kolawole
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Marcela T Taruselli
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Siddarth Ajith
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Jordan M Dailey
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Sydney A Kee
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Tamara T Haque
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Brian O Barnstein
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Jamie Josephine Avila McLeod
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Heather L Caslin
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Kasalina N Kiwanuka
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Yoshihiro Fukuoka
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Quang T Le
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Lawrence B Schwartz
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - David B Straus
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - David A Gewirtz
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - Rebecca K Martin
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| | - John J Ryan
- Departments of Biology (P.A.P., E.M.K., M.T.T., S.A., J.M.D., S.A.K., T.T.H., B.O.B., J.J.A.M., H.L.C., K.N.K., Y.F., D. B.S., J.J.R.), Internal Medicine (Q.T.L., L.B.S.), Pharmacology and Toxicology (D.A.G.), and Microbiology and Immunology (R.K.M.), Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
32
|
Elsayed I, El-Dahmy RM, El-Emam SZ, Elshafeey AH, El Gawad NAA, El-Gazayerly ON. Response surface optimization of biocompatible elastic nanovesicles loaded with rosuvastatin calcium: enhanced bioavailability and anticancer efficacy. Drug Deliv Transl Res 2020; 10:1459-1475. [PMID: 32394333 DOI: 10.1007/s13346-020-00761-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Statins are mainly used for the treatment of hyperlipidemia, but recently, their anticancer role was extremely investigated. The goal of this study was to statistically optimize novel elastic nanovesicles containing rosuvastatin calcium to improve its transdermal permeability, bioavailability, and anticancer effect. The elastic nanovesicles were composed of Tween® 80, cetyl alcohol, and clove oil. The nanodispersions were investigated for their entrapment efficiency, particle size, zeta potential, polydispersity index, and elasticity. The optimized elastic nanovesicular dispersion is composed of 20% cetyl alcohol, 53.47% Tween 80, and 26.53% clove oil. Carboxy methylcellulose was utilized to convert the optimized elastic nanovesicular dispersion into elastic nanovesicular gels. Both the optimized dispersion and the optimized gel (containing 2% w/v carboxymethylcellulose) were subjected to in vitro release study, scanning and transmission electron microscopy, histopathological evaluation, and ex vivo permeation. The cell viability assay of the optimized gel on MCF-7 and Hela cell lines showed significant antiproliferative and potent cytotoxic effects when compared to the drug gel. Moreover, the optimized gel accomplished a significant increase in rosuvastatin bioavailability upon comparison with the drug gel. The optimized gel could be considered as a promising nanocarrier for statins transdermal delivery to increase their systemic bioavailability and anticancer effect. Graphical abstract.
Collapse
|
33
|
Roedig H, Damiescu R, Zeng-Brouwers J, Kutija I, Trebicka J, Wygrecka M, Schaefer L. Danger matrix molecules orchestrate CD14/CD44 signaling in cancer development. Semin Cancer Biol 2020; 62:31-47. [PMID: 31412297 DOI: 10.1016/j.semcancer.2019.07.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 02/06/2023]
Abstract
The tumor matrix together with inflammation and autophagy are crucial regulators of cancer development. Embedded in the tumor stroma are numerous proteoglycans which, in their soluble form, act as danger-associated molecular patterns (DAMPs). By interacting with innate immune receptors, the Toll-like receptors (TLRs), DAMPs autonomously trigger aseptic inflammation and can regulate autophagy. Biglycan, a known danger proteoglycan, can regulate the cross-talk between inflammation and autophagy by evoking a switch between pro-inflammatory CD14 and pro-autophagic CD44 co-receptors for TLRs. Thus, these novel mechanistic insights provide some explanation for the plethora of reports indicating that the same matrix-derived DAMP acts either as a promoter or suppressor of tumor growth. In this review we will summarize and critically discuss the role of the matrix-derived DAMPs biglycan, hyaluronan, and versican in regulating the TLR-, CD14- and CD44-signaling dialogue between inflammation and autophagy with particular emphasis on cancer development.
Collapse
Affiliation(s)
- Heiko Roedig
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Goethe University, Frankfurt am Main, Germany
| | - Roxana Damiescu
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Goethe University, Frankfurt am Main, Germany
| | - Jinyang Zeng-Brouwers
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Goethe University, Frankfurt am Main, Germany
| | - Iva Kutija
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Goethe University, Frankfurt am Main, Germany
| | - Jonel Trebicka
- Translational Hepatology, Department of Internal Medicine I, University Clinic Frankfurt, Germany
| | - Malgorzata Wygrecka
- Department of Biochemistry, Faculty of Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Liliana Schaefer
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Goethe University, Frankfurt am Main, Germany.
| |
Collapse
|
34
|
Schmukler E, Pinkas‐Kramarski R. Autophagy induction in the treatment of Alzheimer's disease. Drug Dev Res 2020; 81:184-193. [DOI: 10.1002/ddr.21605] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/05/2019] [Accepted: 09/10/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Eran Schmukler
- Department of NeurobiologySchool of Neurobiology, Biochemistry and Biophysics, the George S. Wise Faculty of Life Sciences, Tel‐Aviv University Ramat‐Aviv Israel
| | - Ronit Pinkas‐Kramarski
- Department of NeurobiologySchool of Neurobiology, Biochemistry and Biophysics, the George S. Wise Faculty of Life Sciences, Tel‐Aviv University Ramat‐Aviv Israel
| |
Collapse
|
35
|
Hong J, Bang M. Anti-inflammatory Strategies for Schizophrenia: A Review of Evidence for Therapeutic Applications and Drug Repurposing. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2020; 18:10-24. [PMID: 31958901 PMCID: PMC7006977 DOI: 10.9758/cpn.2020.18.1.10] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 09/12/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023]
Abstract
Schizophrenia is a debilitating psychiatric disorder with a substantial socioeconomic and humanistic burden. Currently available treatment strategies mostly rely on antipsychotic drugs, which block dopaminergic effects in the mesolimbic pathway of the brain. Although antipsychotic drugs help relieve psychotic symptoms, a definitive cure for schizophrenia has yet to be achieved. Recent advances in neuroinflammation research suggest that proinflammatory processes in the brain could cause alterations in neurobehavioral development and increase vulnerability to schizophrenia. With a growing need for novel strategies in the treatment of schizophrenia, it would be meaningful to review the current evidence supporting the therapeutic potential of anti-inflammatory strategies. This review details the key findings of clinical trials that investigate the efficacy of anti-inflammatory agents as adjuvants to antipsychotic treatment. We further discuss the possibilities of repurposing anti-inflammatory agents and developing novel strategies for the treatment of schizophrenia.
Collapse
Affiliation(s)
- Jonghee Hong
- CHA University School of Medicine, Seongnam, Korea
| | - Minji Bang
- Department of Psychiatry, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea
| |
Collapse
|
36
|
Hsieh C, Li C, Hsu C, Chen H, Chen Y, Liu Y, Liu Y, Kuo H, Liu P. Mitochondrial protection by simvastatin against angiotensin II-mediated heart failure. Br J Pharmacol 2019; 176:3791-3804. [PMID: 31265743 PMCID: PMC6780047 DOI: 10.1111/bph.14781] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 06/04/2019] [Accepted: 06/06/2019] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Mitochondrial dysfunction plays a role in the progression of cardiovascular diseases including heart failure. 3-Hydroxy-3-methylglutaryl-CoA reductase inhibitors (statins), which inhibit ROS synthesis, show cardioprotective effects in chronic heart failure. However, the beneficial role of statins in mitochondrial protection in heart failure remains unclear. EXPERIMENTAL APPROACH Rats were treated with angiotensin II (1.5 mg·kg-1 ·day-1 ) or co-administered simvastatin (oral, 10 mg·kg-1 ) for 14 days; and then administration was stopped for the following 14 days. Cardiac structure/function was examined by wheat germ agglutinin staining and echocardiography. Mitochondrial morphology and the numbers of lipid droplets, lysosomes, autophagosomes, and mitophagosomes were determined by transmission electron microscopy. Human cardiomyocytes were stimulated, and intracellular ROS and mitochondrial membrane potential (ΔΨm ) changes were measured by flow cytometry and JC-1 staining, respectively. Autophagy and mitophagy-related and mitochondria-regulated apoptotic proteins were identified by immunohistochemistry and western blotting. KEY RESULTS Simvastatin significantly reduced ROS production and attenuated the disruption of ΔΨm . Simvastatin induced the accumulation of lipid droplets to provide energy for maintaining mitochondrial function, promoted autophagy and mitophagy, and inhibited mitochondria-mediated apoptosis. These findings suggest that mitochondrial protection mediated by simvastatin plays a therapeutic role in heart failure prevention by modulating antioxidant status and promoting energy supplies for autophagy and mitophagy to inhibit mitochondrial damage and cardiomyocyte apoptosis. CONCLUSION AND IMPLICATIONS Mitochondria play a key role in mediating heart failure progression. Simvastatin attenuated heart failure, induced by angiotensin II, via mitochondrial protection and might provide a new therapy to prevent heart failure.
Collapse
Affiliation(s)
- Chong‐Chao Hsieh
- Graduate Institute of Clinical Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Division of Cardiovascular Surgery, Department of SurgeryKaohsiung Medical University HospitalKaohsiungTaiwan
| | - Chia‐Yang Li
- Graduate Institute of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Center for Infectious Disease and Cancer ResearchKaohsiung Medical UniversityKaohsiungTaiwan
| | - Chih‐Hsin Hsu
- Department of Internal MedicineCheng Kung University HospitalTainanTaiwan
| | - Hsiu‐Lin Chen
- Department of Respiratory Therapy, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
| | - Yung‐Hsiang Chen
- Graduate Institute of Integrated Medicine, College of Chinese MedicineChina Medical UniversityTaichungTaiwan
- Department of Psychology, College of Medical and Health ScienceAsia UniversityTaichungTaiwan
| | - Yu‐Peng Liu
- Graduate Institute of Clinical Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
| | - Yu‐Ru Liu
- Department of Respiratory Therapy, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
| | - Hsuan‐Fu Kuo
- Graduate Institute of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Department of Internal Medicine, Kaohsiung Municipal Ta‐Tung HospitalKaohsiung Medical UniversityKaohsiungTaiwan
| | - Po‐Len Liu
- Department of Respiratory Therapy, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Regenerative Medicine and Cell Therapy Research CenterKaohsiung Medical UniversityKaohsiungTaiwan
| |
Collapse
|
37
|
Meske V, Albert F, Gerstenberg S, Kallwellis K, Ohm TG. NPC1-deficient neurons are selectively vulnerable for statin treatment. Neuropharmacology 2019; 151:159-170. [DOI: 10.1016/j.neuropharm.2019.04.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 03/07/2019] [Accepted: 04/10/2019] [Indexed: 10/27/2022]
|
38
|
Guo GF, Wang YX, Zhang YJ, Chen XX, Lu JB, Wang HH, Jiang C, Qiu HQ, Xia LP. Predictive and prognostic implications of 4E-BP1, Beclin-1, and LC3 for cetuximab treatment combined with chemotherapy in advanced colorectal cancer with wild-type KRAS: Analysis from real-world data. World J Gastroenterol 2019; 25:1840-1853. [PMID: 31057298 PMCID: PMC6478617 DOI: 10.3748/wjg.v25.i15.1840] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/20/2019] [Accepted: 03/25/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the main causes of cancer-related deaths in China and around the world. Advanced CRC (ACRC) patients suffer from a low cure rate though treated with targeted therapies. The response rate is about 50% to chemotherapy and cetuximab, a monoclonal antibody targeting epidermal growth factor receptor (EGFR) and used for ACRC with wild-type KRAS. It is important to identify more predictors of cetuximab efficacy to further improve precise treatment. Autophagy, showing a key role in the cancer progression, is influenced by the EGFR pathway. Whether autophagy can predict cetuximab efficacy in ACRC is an interesting topic.
AIM To investigate the effect of autophagy on the efficacy of cetuximab in colon cancer cells and ACRC patients with wild-type KRAS.
METHODS ACRC patients treated with cetuximab plus chemotherapy, with detailed data and tumor tissue, at Sun Yat-sen University Cancer Center from January 1, 2005, to October 1, 2015, were studied. Expression of autophagy-related proteins [Beclin1, microtubule-associated protein 1A/B-light chain 3 (LC3), and 4E-binding protein 1 (4E-BP1)] was examined by Western blot in CRC cells and by immunohistochemistry in cancerous and normal tissues. The effect of autophagy on cetuximab-treated cancer cells was confirmed by MTT assay. The associations between Beclin1, LC3, and 4E-BP1 expression in tumor tissue and the efficacy of cetuximab-based therapy were analyzed.
RESULTS In CACO-2 cells exposed to cetuximab, LC3 and 4E-BP1 were upregulated, and P62 was downregulated. Autophagosome formation was observed, and autophagy increased the efficacy of cetuximab. In 68 ACRC patients, immunohistochemistry showed that Beclin1 levels were significantly correlated with those of LC3 (0.657, P < 0.001) and 4E-BP1 (0.211, P = 0.042) in ACRC tissues. LC3 was significantly overexpressed in tumor tissues compared to normal tissues (P < 0.001). In 45 patients with wild-type KRAS, the expression levels of these three proteins were not related to progression-free survival; however, the expression levels of Beclin1 (P = 0.010) and 4E-BP1 (P = 0.005), pathological grade (P = 0.002), and T stage (P = 0.004) were independent prognostic factors for overall survival (OS).
CONCLUSION The effect of cetuximab on colon cancer cells might be improved by autophagy. LC3 is overexpressed in tumor tissues, and Beclin1 and 4E-BP1 could be significant predictors of OS in ACRC patients treated with cetuximab.
Collapse
Affiliation(s)
- Gui-Fang Guo
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
| | - Yi-Xing Wang
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
| | - Yi-Jun Zhang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
- Pathology Department, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Xiu-Xing Chen
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
| | - Jia-Bin Lu
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
- Pathology Department, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Hao-Hua Wang
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
| | - Chang Jiang
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
| | - Hui-Quan Qiu
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
| | - Liang-Ping Xia
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
| |
Collapse
|
39
|
Kao LT, Hung SH, Kao PF, Liu JC, Lin HC. Inverse association between statin use and head and neck cancer: Population-based case-control study in Han population. Head Neck 2019; 41:1193-1198. [PMID: 30809863 DOI: 10.1002/hed.25501] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/16/2018] [Accepted: 08/17/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND This case-control study aimed to find the relationship between prior statin use and head and neck cancer occurrence using a large population-based database. METHODS This study used claims data from the Taiwan Longitudinal Health Insurance Database. We included 5515 patients with head and neck cancer as cases and 5515 propensity score-matched patients without head and neck cancer as controls. Conditional logistic regressions were performed to investigate the relationship between head and neck cancer and prior statin exposure. RESULTS Of the 11 030 total sampled patients, 16.95% had previously received prescriptions for statins. In addition, statin exposure was found in 15.99% of cases and 17.91% of controls. The logistic regression also revealed that the adjusted odds ratio of prior statin exposure for cases was 0.86 (95% confidence interval: 0.77-0.95) compared to propensity score-matched controls. CONCLUSION This study found an inverse association between statin usage and head and neck cancer occurrence.
Collapse
Affiliation(s)
- Li-Ting Kao
- Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan.,Sleep Research Center, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Pharmacy Practice, Tri-Service General Hospital, Taipei, Taiwan
| | - Shih-Han Hung
- Department of Otolaryngology, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Otolaryngology, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Pai-Feng Kao
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ju-Chi Liu
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Cardiovascular Medicine, Department of Medicine, Medical University Hospital--Shuang Ho Hospital, Taipei, Taiwan
| | - Herng-Ching Lin
- Sleep Research Center, Taipei Medical University Hospital, Taipei, Taiwan.,School of Health Care Administration, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
40
|
Tetrandrine-Induced Autophagy in MDA-MB-231 Triple-Negative Breast Cancer Cell through the Inhibition of PI3K/AKT/mTOR Signaling. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:7517431. [PMID: 30713576 PMCID: PMC6332987 DOI: 10.1155/2019/7517431] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 11/24/2018] [Accepted: 12/03/2018] [Indexed: 12/13/2022]
Abstract
The present study examined the effects of tetrandrine suppressing proliferation, targeting LC3, p62, and Beclin-1 autophagy genes by inhibiting PI3K/AKT/mTOR signaling in Triple-negative breast cancer (TNBC) MDA-MB-231 cell. Cell viability and apoptosis were evaluated by MTT and Annexin-V/PI double staining. Cytotoxicity was determined with LDH assay. Western Blot and Immunofluorescence were used to measure the protein levels of p62/SQSTM1, Beclin1, LC3-II/LC3-I, and PTEN/PI3K/AKT/mTOR signaling. Results showed that tetrandrine inhibited the MDA-MB-231 cell proliferation and induced the apoptosis. Tetrandrine at doses of 12.8, 16.1, and 25.7μmol/L showed significant cytotoxicity on MDA-MB-231 cells (p<0.01). Tetrandrine induced MDA-MB-231 cell autophagy by decreasing p62/SQSTM1 expression, improving the expression of Beclin1 and LC3-II/LC3-I (p<0.01), inhibiting the PI3K/AKT /mTOR pathway by downregulating the expression of p-AKT ser473/AKT, p-PI3K/PI3K p110α, and p-mTOR ser2448/mTOR and upregulating PTEN expression. These findings revealed that tetrandrine could suppress proliferation and induce autophagy in MDA-MB-231 cell by inhibiting the PI3K/AKT/mTOR pathway and might be a promising anti-triple-negative breast cancer drug.
Collapse
|
41
|
Hu MB, Zhang JW, Gao JB, Qi YW, Gao Y, Xu L, Ma Y, Wei ZZ. Atorvastatin induces autophagy in MDA-MB-231 breast cancer cells. Ultrastruct Pathol 2018; 42:409-415. [PMID: 30300062 DOI: 10.1080/01913123.2018.1522406] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This article explores the effects of atorvastatin on cultured breast cancer cells. Our experiment demonstrated that atorvastatin triggered autophagy and inhibited proliferation in breast cancer cells. A CCK8 assay indicated that atorvastatin can inhibit the activity of MDA-MB-231 breast cancer cells. Western blotting results showed that atorvastatin increased the conversion of light chain 3 (LC3)-I to LC3-phosphatidylethanolamine conjugate (LC3-II). Confocal microscopy was used to reveal the appearance of a punctate structure in the cytoplasm, and electron microscopy was used to reveal the formation of double-membrane autophagosome. In conclusion, our study showed that atorvastatin may affect MDA-MB-231 breast cancer cells by inducing autophagy.
Collapse
Affiliation(s)
- Ming-Bai Hu
- a Department of Breast and Thyroid Surgery, Zhongnan Hospital, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center , Wuhan University , Wuhan , Hubei , China
| | - Jing-Wei Zhang
- a Department of Breast and Thyroid Surgery, Zhongnan Hospital, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center , Wuhan University , Wuhan , Hubei , China
| | - Jing-Bo Gao
- a Department of Breast and Thyroid Surgery, Zhongnan Hospital, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center , Wuhan University , Wuhan , Hubei , China
| | - Yu-Wen Qi
- a Department of Breast and Thyroid Surgery, Zhongnan Hospital, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center , Wuhan University , Wuhan , Hubei , China
| | - Yang Gao
- a Department of Breast and Thyroid Surgery, Zhongnan Hospital, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center , Wuhan University , Wuhan , Hubei , China
| | - Liu Xu
- b Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences , Wuhan University , Wuhan , Hubei , China
| | - Yanbing Ma
- b Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences , Wuhan University , Wuhan , Hubei , China
| | - Zheng-Zhuan Wei
- a Department of Breast and Thyroid Surgery, Zhongnan Hospital, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center , Wuhan University , Wuhan , Hubei , China
| |
Collapse
|
42
|
Chen YA, Shih HW, Lin YC, Hsu HY, Wu TF, Tsai CH, Wu CL, Wu HY, Hsieh JT, Tang CH, Lai CH. Simvastatin Sensitizes Radioresistant Prostate Cancer Cells by Compromising DNA Double-Strand Break Repair. Front Pharmacol 2018; 9:600. [PMID: 29950990 PMCID: PMC6008406 DOI: 10.3389/fphar.2018.00600] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/18/2018] [Indexed: 12/17/2022] Open
Abstract
Prostate cancer (PCa) is one of the most prevalent male cancers in western world. Radiation therapy (RT) is commonly used to treat PCa patients. However, a certain proportion of patients develop radioresistant PCa cells, which results in metastatic disease. Statins, which inhibit 3-hydroxy-3-methyl glutaryl coenzyme A (HMG-CoA) reductase, are commonly used to treat hypercholesterolemia, exhibiting beneficial effects on cardiovascular diseases and on several types of cancers, including PCa. However, the mechanistic details and crosstalk between statins and RT in PCa cells remain unknown. In this study, radioresistant DOC-2/DAB2 interactive protein (DAB2IP)-deficient PCa cells were used to evaluate whether simvastatin could enhance the effect of ionizing radiation (IR). The crucial molecules that associated with simvastatin elevated radiosensitivity in PCa cells were explored. Our results demonstrated that a combination treatment with simvastatin and IR synergistically induced apoptosis of radioresistant PCa cells. In addition, simvastatin appeared to compromise DNA double-strand breaks repair by activating the expressions of histone 2A family member X (γ-H2AX) and phospho-checkpoint kinase 1 (p-CHK1), suggesting an underlying mechanism for this radiosensitization of PCa cells. These findings reveal that simvastatin may be a potent therapeutic agent for co-treatment with radiation to overcome radioresistance in PCa cells.
Collapse
Affiliation(s)
- Yu-An Chen
- Graduate Institute of Basic Medical Science, School of Medicine, China Medical University, Taichung, Taiwan
| | - Hua-Wei Shih
- Graduate Institute of Basic Medical Science, School of Medicine, China Medical University, Taichung, Taiwan
| | - Yi-Chun Lin
- Graduate Institute of Basic Medical Science, School of Medicine, China Medical University, Taichung, Taiwan
| | - Hui-Ying Hsu
- Graduate Institute of Basic Medical Science, School of Medicine, China Medical University, Taichung, Taiwan
| | - Tsu-Fang Wu
- Department of Applied Cosmetology, Hung Kuang University, Taichung, Taiwan
| | - Chen-Han Tsai
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Lin Wu
- Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Molecular Infectious Disease Research Center, Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Hui-Yu Wu
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, School of Medicine, China Medical University, Taichung, Taiwan.,Graduate Institute of Biomedical Sciences, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chih-Ho Lai
- Graduate Institute of Basic Medical Science, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Molecular Infectious Disease Research Center, Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Nursing, Asia University, Taichung, Taiwan
| |
Collapse
|
43
|
Cheng C, Geng F, Cheng X, Guo D. Lipid metabolism reprogramming and its potential targets in cancer. Cancer Commun (Lond) 2018; 38:27. [PMID: 29784041 PMCID: PMC5993136 DOI: 10.1186/s40880-018-0301-4] [Citation(s) in RCA: 520] [Impact Index Per Article: 74.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/12/2018] [Indexed: 12/13/2022] Open
Abstract
Reprogramming of lipid metabolism is a newly recognized hallmark of malignancy. Increased lipid uptake, storage and lipogenesis occur in a variety of cancers and contribute to rapid tumor growth. Lipids constitute the basic structure of membranes and also function as signaling molecules and energy sources. Sterol regulatory element-binding proteins (SREBPs), a family of membrane-bound transcription factors in the endoplasmic reticulum, play a central role in the regulation of lipid metabolism. Recent studies have revealed that SREBPs are highly up-regulated in various cancers and promote tumor growth. SREBP cleavage-activating protein is a key transporter in the trafficking and activation of SREBPs as well as a critical glucose sensor, thus linking glucose metabolism and de novo lipid synthesis. Targeting altered lipid metabolic pathways has become a promising anti-cancer strategy. This review summarizes recent progress in our understanding of lipid metabolism regulation in malignancy, and highlights potential molecular targets and their inhibitors for cancer treatment.
Collapse
Affiliation(s)
- Chunming Cheng
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, OH, 43210, USA
| | - Feng Geng
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, OH, 43210, USA
| | - Xiang Cheng
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, OH, 43210, USA
| | - Deliang Guo
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, OH, 43210, USA.
| |
Collapse
|
44
|
Ma H, Li X, Wang J, Hornicek F, Garbutt C, Chang X, Duan Z. Expression and Clinical Implication of Autophagy-Associated Protein p62 in Osteosarcoma. Oncology 2018; 95:52-60. [DOI: 10.1159/000487437] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 01/25/2018] [Indexed: 12/31/2022]
|
45
|
Liu Y, Yang H, Jia G, Li L, Chen H, Bi J, Wang C. The Synergistic Neuroprotective Effects of Combined Rosuvastatin and Resveratrol Pretreatment against Cerebral Ischemia/Reperfusion Injury. J Stroke Cerebrovasc Dis 2018. [PMID: 29525080 DOI: 10.1016/j.jstrokecerebrovasdis.2018.01.033] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND It is well accepted that both rosuvastatin and resveratrol exert neuroprotective effects on cerebral ischemia/reperfusion injury through some common pathways. Resveratrol has also been demonstrated to protect against cerebral ischemia/reperfusion injury through enhancing autophagy. Thus, we hypothesized that combined rosuvastatin and resveratrol pretreatment had synergistic effects on cerebral ischemia/reperfusion injury. MATERIALS AND METHODS Adult male Sprague Dawley rats receiving middle cerebral artery occlusion surgery as animal model of cerebral ischemia/reperfusion injury were randomly assigned to 4 groups: control, resveratrol alone pretreatment, rosuvastatin alone pretreatment, and combined rosuvastatin and resveratrol pretreatment. Rosuvastatin (10 mg/kg) or resveratrol (50 mg/kg) was administrated once a day for 7 days before cerebral ischemia onset. RESULTS We found that combined rosuvastatin and resveratrol pretreatment not only significantly decreased the neurologic defective score, cerebral infarct volume, the levels of caspase-3, and Interleukin-1β (IL-1β) but also significantly increased the ratios of Bcl-2/Bax and LC3II/LC3I, as well as the level of Becline-1, compared with resveratrol alone or rosuvastatin alone pretreatment group. Rosuvastatin alone pretreatment significantly increased the ratio of LC3II/LC3I and the level of Beclin-1. However, there were no significant differences in the neurologic defective score, cerebral infarct volume, the levels of caspase-3, IL-1β, and Beclin-1, and the ratios of Bcl-2/Bax and LC3II/LC3I between resveratrol pretreatment group and rosuvastatin pretreatment group. CONCLUSIONS Synergistically enhanced antiapoptosis, anti-inflammation, and autophagy activation might be responsible for the synergistic neuroprotective effects of combining rosuvastatin with resveratrol on cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Ying Liu
- Department of Neurology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - HongNa Yang
- Department of Critical-care Medicine, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - GuoYong Jia
- Department of Neurology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - Lan Li
- Department of Neurology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - Hui Chen
- Department of Neurology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - JianZhong Bi
- Department of Neurology Medicine, Second Hospital of Shandong University, Shandong University, Jinan, Shandong, China.
| | - CuiLan Wang
- Department of Neurology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, China; Brain Science Research Institute, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
46
|
Zhang Z, Liu T, Yu M, Li K, Li W. The plant alkaloid tetrandrine inhibits metastasis via autophagy-dependent Wnt/β-catenin and metastatic tumor antigen 1 signaling in human liver cancer cells. J Exp Clin Cancer Res 2018; 37:7. [PMID: 29334999 PMCID: PMC5769468 DOI: 10.1186/s13046-018-0678-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 01/08/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Tetrandrine is a bisbenzylisoquinoline alkaloid isolated from the Chinese medicinal herb Stephania tetrandra S. Moore. We previously demonstrated that tetrandrine exhibits potent antitumor effects in many types of cancer cells. In this study, we investigated the effects of tetrandrine on human hepatocellular carcinoma (HCC) metastasis. METHODS The invasion and migration effects were evaluated via wound healing and transwell assays. Immunofluorescence and western blotting analyses were used to investigate the levels of epithelial-mesenchymal transition (EMT)-related protein. A metastasis model was established to investigate the inhibitory effect of tetrandrine on hepatocellular carcinoma metastasis in vivo. RESULTS Tetrandrine inhibits HCC invasion and migration by preventing cell EMT. The underlying mechanism was closely associated with tetrandrine-induced human liver cell autophagy, which inhibits Wnt/β-catenin pathway activity and decreases metastatic tumor antigen 1 (MTA1) expression to modulate cancer cell metastasis. CONCLUSION Our findings demonstrate, for the first time, that tetrandrine plays a significant role in the inhibition of human hepatocellular carcinoma metastasis and provide novel insights into the application of tetrandrine in clinical HCC treatment.
Collapse
Affiliation(s)
- Zhenxing Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072 People’s Republic of China
| | - Ting Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072 People’s Republic of China
| | - Man Yu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072 People’s Republic of China
| | - Kangdi Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072 People’s Republic of China
| | - Wenhua Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072 People’s Republic of China
| |
Collapse
|
47
|
Efficacy and Mechanism of Preoperative Simvastatin Therapy on Myocardial Protection after Extracorporeal Circulation. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6082430. [PMID: 29250545 PMCID: PMC5698794 DOI: 10.1155/2017/6082430] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 09/08/2017] [Accepted: 09/26/2017] [Indexed: 02/01/2023]
Abstract
Background Cardiopulmonary bypass (CPB) causes systemic inflammatory response and ischemia-reperfusion (IR) injury. Objective To investigate the effect and mechanism of simvastatin on myocardial injury in cardiac valve surgery with CPB. Methods One hundred thirty patients were randomly assigned to the statin group (n = 65) or control group (n = 65). Simvastatin was administered preoperatively and postoperatively. Duration of intensive care unit stay, duration of assisted ventilation, and left ventricular ejection fraction were recorded. Plasma was analysed for troponin T (cTnT), isoenzyme of creatine kinase (CK-MB), tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), and interleukin-8 (IL-8). Ultrastructure of the myocardium and autophagosomes were observed. Beclin-1, LC3-II/I, P62, AMPK, and the phosphorylation of AMPK in cardiomyocytes were detected. Results Simvastatin significantly reduced the duration of assisted ventilation (P = 0.030) and ejection fraction was significantly higher in the statin group (P = 0.024). Simvastatin significantly reduced the levels of cTnT, CK-MB, TNF-α, IL-6, and IL-8 (P < 0.05), reduced the expression of LC3-II/LC3-I and Beclin 1, and increased the expression of phosphorylation of AMPK. Simvastatin reduced the generation of autophagosomes and the ultrastructural injuries to myocardium. Conclusion Perioperative statin therapy reduced myocardial injury by regulating myocardial autophagy and activating the phosphorylation of AMPK. The registration number of this study is ChiCTR-TRC-14005164.
Collapse
|
48
|
Reilly JE, Neighbors JD, Hohl RJ. Targeting protein geranylgeranylation slows tumor development in a murine model of prostate cancer metastasis. Cancer Biol Ther 2017; 18:872-882. [PMID: 27624889 PMCID: PMC5710670 DOI: 10.1080/15384047.2016.1219817] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 07/11/2016] [Accepted: 07/29/2016] [Indexed: 12/23/2022] Open
Abstract
The isoprenoid biosynthetic pathway (IBP) plays a critical role in providing substrates and enzymes necessary for the post-translational modification and thus activation of a number of proteins involved in prostate cancer metastasis. Previous work by our lab found novel compound disodium [(6Z,11E,15E)-9-[bis(sodiooxy)phosphoryl]-17-hydroxy-2,6,12,16-tetramethyheptadeca-2,6,11,15-tetraen-9-yl]phosphonate (GGOHBP), which inhibits the IBP enzyme geranylgeranyl diphosphate synthase (GGDPS), reduced protein geranylgeranylation without altering protein farnesylation. This activity significantly reduced adrenal gland tumor burden in a murine model of human prostate cancer metastasis which relied on treatment of established disease. The present study determined the ability of GGDPS inhibition to slow the development of prostate cancer metastasis in a preventative murine model. Using tail vein injection of human derived PC-3 prostate cancer cells 4 d after initiating daily GGOHBP or vehicle treatments, we found GGOHBP significantly reduced whole body tumor burden, significantly slowed the development of tumors, and prolonged overall survival as compared to vehicle treated animals. The observed reduction in soft tissue tumor burden corresponded to a biochemical reduction in Rap1A geranylgeranylation, which for prostate cancer is important in its own merit and which serves as a surrogate marker for Rho family, i.e. Rac, protein modification. This effect was present in all treated mice pointing to strong target engagement, which was not observed in non-tumor burdened tissues or control mice. Our findings reiterate a role for protein geranylgeranylation in the development of prostate cancer metastasis in vivo.
Collapse
Affiliation(s)
| | | | - Raymond J. Hohl
- Department of Pharmacology, University of Iowa, Iowa City, IA, USA
- Departments of Internal Medicine and Pharmacology, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
49
|
Lin Z, Zhang Z, Jiang X, Kou X, Bao Y, Liu H, Sun F, Ling S, Qin N, Jiang L, Yang Y. Mevastatin blockade of autolysosome maturation stimulates LBH589-induced cell death in triple-negative breast cancer cells. Oncotarget 2017; 8:17833-17848. [PMID: 28147319 PMCID: PMC5392290 DOI: 10.18632/oncotarget.14868] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 01/11/2017] [Indexed: 12/14/2022] Open
Abstract
Histone deacetylase inhibitors (HDACi) are promising anti-cancer agents, and combining a HDACi with other agents is an attractive therapeutic strategy in solid tumors. We report here that mevastatin increases HDACi LBH589-induced cell death in triple-negative breast cancer (TNBC) cells. Combination treatment inhibited autophagic flux by preventing Vps34/Beclin 1 complex formation and downregulating prenylated Rab7, an active form of the small GTPase necessary for autophagosome-lysosome fusion. This means that co-treatment with mevastatin and LBH589 activated LKB1/AMPK signaling and subsequently inhibited mTOR. Co-treatment also led to cell cycle arrest in G2/M phase and induced corresponding expression changes of proteins regulating the cell cycle. Co-treatment also increased apoptosis both in vitro and in vivo, and reduced tumor volumes in xenografted mice. Our results indicate that disruption of autophagosome-lysosome fusion likely underlies mevastatin-LBH589 synergistic anticancer effects. This study confirms the synergistic efficacy of, and demonstrates a potential therapeutic role for mevastatin plus LBH589 in targeting aggressive TNBC, and presents a novel therapeutic strategy for further clinical study. Further screening for novel autophagy modulators could be an efficient approach to enhance HDACi-induced cell death in solid tumors.
Collapse
Affiliation(s)
- Zhaohu Lin
- Department of Pharmacology and Biochemistry, School of Pharmacy Fudan University, Shanghai 201203, China.,Chemical Biology, Roche Pharmaceutical Research and Early Development, Roche Innovation Center Shanghai, Shanghai 201203, China
| | - Zhuqing Zhang
- Department of Pharmacology and Biochemistry, School of Pharmacy Fudan University, Shanghai 201203, China
| | - Xiaoxiao Jiang
- Department of Pharmacology and Biochemistry, School of Pharmacy Fudan University, Shanghai 201203, China
| | - Xinhui Kou
- Department of Pharmacology and Biochemistry, School of Pharmacy Fudan University, Shanghai 201203, China
| | - Yong Bao
- Department of Pharmacology and Biochemistry, School of Pharmacy Fudan University, Shanghai 201203, China
| | - Huijuan Liu
- Department of Pharmacology and Biochemistry, School of Pharmacy Fudan University, Shanghai 201203, China
| | - Fanghui Sun
- Department of Pharmacology and Biochemistry, School of Pharmacy Fudan University, Shanghai 201203, China
| | - Shuang Ling
- Interdisciplinary Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ning Qin
- Chemical Biology, Roche Pharmaceutical Research and Early Development, Roche Innovation Center Shanghai, Shanghai 201203, China
| | - Lan Jiang
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA
| | - Yonghua Yang
- Department of Pharmacology and Biochemistry, School of Pharmacy Fudan University, Shanghai 201203, China
| |
Collapse
|
50
|
Simakova MN, Bisen S, Dopico AM, Bukiya AN. Statin therapy exacerbates alcohol-induced constriction of cerebral arteries via modulation of ethanol-induced BK channel inhibition in vascular smooth muscle. Biochem Pharmacol 2017; 145:81-93. [PMID: 28865873 DOI: 10.1016/j.bcp.2017.08.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/28/2017] [Indexed: 12/26/2022]
Abstract
Statins constitute the most commonly prescribed drugs to decrease cholesterol (CLR). CLR is an important modulator of alcohol-induced cerebral artery constriction (AICAC). Using rats on a high CLR diet (2% CLR) we set to determine whether atorvastatin administration (10mg/kg daily for 18-23weeks) modified AICAC. Middle cerebral arteries were pressurized in vitro at 60mmHg and AICAC was evoked by 50mM ethanol, that is within the range of blood alcohol detected in humans following moderate-to-heavy drinking. AICAC was evident in high CLR+atorvastatin group but not in high CLR diet+placebo. Statin exacerbation of AICAC persisted in de-endothelialized arteries, and was blunted by CLR enrichment in vitro. Fluorescence imaging of filipin-stained arteries showed that atorvastatin decreased vascular smooth muscle (VSM) CLR when compared to placebo, this difference being reduced by CLR enrichment in vitro. Voltage- and calcium-gated potassium channels of large conductance (BK) are known VSM targets of ethanol, with their beta1 subunit being necessary for ethanol-induced channel inhibition and resulting AICAC. Ethanol-induced BK inhibition in excised membrane patches from freshly isolated myocytes was exacerbated in the high CLR diet+atorvastatin group when compared to high CLR diet+placebo. Unexpectedly, atorvastatin decreased the amount and function of BK beta1 subunit as documented by immunofluorescence imaging and functional patch-clamp studies. Atorvastatin exacerbation of ethanol-induced BK inhibition disappeared upon artery CLR enrichment in vitro. Our study demonstrates for the first time statin's ability to exacerbate the vascular effect of a widely consumed drug of abuse, this exacerbation being driven by statin modulation of ethanol-induced BK channel inhibition in the VSM via CLR-mediated mechanism.
Collapse
Affiliation(s)
- Maria N Simakova
- Dept. Pharmacology, University of Tennessee HSC, Memphis, TN 38103, United States
| | - Shivantika Bisen
- Dept. Pharmacology, University of Tennessee HSC, Memphis, TN 38103, United States
| | - Alex M Dopico
- Dept. Pharmacology, University of Tennessee HSC, Memphis, TN 38103, United States
| | - Anna N Bukiya
- Dept. Pharmacology, University of Tennessee HSC, Memphis, TN 38103, United States.
| |
Collapse
|