1
|
Li Y, Pan AP, Yu AY. Recent Progression of Pathogenesis and Treatment for Diabetic Cataracts. Semin Ophthalmol 2024:1-8. [PMID: 39530428 DOI: 10.1080/08820538.2024.2427789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/22/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Background: Cataracts are still the main cause of blindness worldwide. The incidence of cataracts in the diabetic population is 3-5 times higher than in healthy people. With the increasing incidence of diabetes and the development of aging, as well as the higher risk of surgical and postoperative complications of diabetic patients undergoing surgery, it is still necessary to study the occurrence and development mechanism of diabetic cataracts as well as potential therapeutic targets and therapeutic drugs.Methods: A retrospective review of the literature from PubMed (2017-2024).Results: We summarized the the current literature on the molecular mechanism and prevention and treatment of diabetic cataracts.Conclusions: The aqueous humor metabolism changes, oxidative stress, reactive oxygen species generation increase, the conversion of polyol pathway, as well as non-coding RNA expression changes play important roles in diabetic cataract and these processes is closely linked with each other. Inhibitors or drugs target to these processes, such as aldose reductase inhibitors, antioxidants, natural flavonoid compounds, as well as nanotechnology-based therapeutic product, have shown promising prospects in the prevention and treatment of diabetic cataract.
Collapse
Affiliation(s)
- Yike Li
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang P. R. China
- National Clinical Research Center for Ocular Diseases, Wenzhou, Zhejiang, P. R. China
| | - An-Peng Pan
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang P. R. China
- National Clinical Research Center for Ocular Diseases, Wenzhou, Zhejiang, P. R. China
| | - A-Yong Yu
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang P. R. China
- National Clinical Research Center for Ocular Diseases, Wenzhou, Zhejiang, P. R. China
| |
Collapse
|
2
|
Lee HJ, Chae CW, Han HJ. Enhancing the therapeutic efficacy of mesenchymal stem cell transplantation in diabetes: Amelioration of mitochondrial dysfunction-induced senescence. Biomed Pharmacother 2023; 168:115759. [PMID: 37865993 DOI: 10.1016/j.biopha.2023.115759] [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: 08/25/2023] [Revised: 10/09/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023] Open
Abstract
Mesenchymal stem cell (MSC) transplantation offers significant potential for the treatment of diabetes mellitus (DM) and its complications. However, hyperglycemic conditions can induce senescence and dysfunction in both transplanted and resident MSCs, thereby limiting their therapeutic potential. Mitochondrial dysfunction and oxidative stress are key contributors to this process in MSCs exposed to hyperglycemia. As such, strategies aimed at mitigating mitochondrial dysfunction could enhance the therapeutic efficacy of MSC transplantation in DM. In this review, we provide an updated overview of how mitochondrial dysfunction mediates MSC senescence. We present experimental evidence for the molecular mechanisms behind high glucose-induced mitochondrial dysfunction in MSCs, which include impairment of mitochondrial biogenesis, mitochondrial calcium regulation, the mitochondrial antioxidant system, mitochondrial fusion-fission dynamics, mitophagy, and intercellular mitochondrial transfer. Furthermore, we propose potential pharmacological candidates that could improve the efficacy of MSC transplantation by enhancing mitochondrial function in patients with DM and related complications.
Collapse
Affiliation(s)
- Hyun Jik Lee
- Laboratory of Veterinary Physiology, College of Veterinary Medicine and Veterinary Medicine Center, Chungbuk National University, Cheongju 28644, Republic of South Korea; Institute for Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju 28644, Republic of South Korea
| | - Chang Woo Chae
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 Four Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul 08826, Republic of Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 Four Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
3
|
Potz BA, Sabe SA, Scrimgeour LA, Sabe AA, Harris DD, Abid MR, Clements RT, Sellke FW. Calpain inhibition decreases oxidative stress via mitochondrial regulation in a swine model of chronic myocardial ischemia. Free Radic Biol Med 2023; 208:700-707. [PMID: 37748718 PMCID: PMC10598262 DOI: 10.1016/j.freeradbiomed.2023.09.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/18/2023] [Accepted: 09/23/2023] [Indexed: 09/27/2023]
Abstract
INTRODUCTION Calpain overexpression is implicated in mitochondrial damage leading to tissue oxidative stress and myocardial ischemic injury. The aim of this study was to determine the effects of calpain inhibition (CI) on mitochondrial impairment and oxidative stress in a swine model of chronic myocardial ischemia and metabolic syndrome. METHODS Yorkshire swine were fed a high-fat diet for 4 weeks to induce metabolic syndrome then underwent placement of an ameroid constrictor to the left circumflex artery. Three weeks later, animals received: no drug (control, "CON"; n= 7); a low-dose calpain inhibitor (0.12 mg/kg; "LCI", n= 7); or high-dose calpain inhibitor (0.25 mg/kg; "HCI", n=7). Treatment continued for 5 weeks, followed by tissue harvest. Cardiac tissue was assayed for protein carbonyl content, as well as antioxidant and mitochondrial protein expression. Reactive oxygen species (ROS) and mitochondrial respiration was measured in H9c2 cells following exposure to normoxia or hypoxia (1%) for 24 h with or without CI. RESULTS In ischemic myocardial tissue, CI was associated with decreased total oxidative stress compared to control. CI was also associated with increased expression of mitochondrial proteins superoxide dismutase 1, SDHA, and pyruvate dehydrogenase compared to control. 100 nM of calpain inhibitor decreased ROS levels and respiration in both normoxic and hypoxic H9c2 cardiomyoblasts. CONCLUSIONS In the setting of metabolic syndrome, CI improves oxidative stress in chronically ischemic myocardial tissue. Decreased oxidative stress may be via modulation of mitochondrial proteins involved in free radical scavenging and production.
Collapse
Affiliation(s)
- Brittany A Potz
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, USA
| | - Sharif A Sabe
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, USA
| | - Laura A Scrimgeour
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, USA
| | - Ashraf A Sabe
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, USA
| | - Dwight D Harris
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, USA
| | - M Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, USA
| | - Richard T Clements
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, USA
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, USA.
| |
Collapse
|
4
|
Blue EE, White JJ, Dush MK, Gordon WW, Wyatt BH, White P, Marvin CT, Helle E, Ojala T, Priest JR, Jenkins MM, Almli LM, Reefhuis J, Pangilinan F, Brody LC, McBride KL, Garg V, Shaw GM, Romitti PA, Nembhard WN, Browne ML, Werler MM, Kay DM, Mital S, Chong JX, Nascone-Yoder NM, Bamshad MJ. Rare variants in CAPN2 increase risk for isolated hypoplastic left heart syndrome. HGG ADVANCES 2023; 4:100232. [PMID: 37663545 PMCID: PMC10474499 DOI: 10.1016/j.xhgg.2023.100232] [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: 04/24/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023] Open
Abstract
Hypoplastic left heart syndrome (HLHS) is a severe congenital heart defect (CHD) characterized by hypoplasia of the left ventricle and aorta along with stenosis or atresia of the aortic and mitral valves. HLHS represents only ∼4%-8% of all CHDs but accounts for ∼25% of deaths. HLHS is an isolated defect (i.e., iHLHS) in 70% of families, the vast majority of which are simplex. Despite intense investigation, the genetic basis of iHLHS remains largely unknown. We performed exome sequencing on 331 families with iHLHS aggregated from four independent cohorts. A Mendelian-model-based analysis demonstrated that iHLHS was not due to single, large-effect alleles in genes previously reported to underlie iHLHS or CHD in >90% of families in this cohort. Gene-based association testing identified increased risk for iHLHS associated with variation in CAPN2 (p = 1.8 × 10-5), encoding a protein involved in functional adhesion. Functional validation studies in a vertebrate animal model (Xenopus laevis) confirmed CAPN2 is essential for cardiac ventricle morphogenesis and that in vivo loss of calpain function causes hypoplastic ventricle phenotypes and suggest that human CAPN2707C>T and CAPN21112C>T variants, each found in multiple individuals with iHLHS, are hypomorphic alleles. Collectively, our findings show that iHLHS is typically not a Mendelian condition, demonstrate that CAPN2 variants increase risk of iHLHS, and identify a novel pathway involved in HLHS pathogenesis.
Collapse
Affiliation(s)
- Elizabeth E. Blue
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | | | - Michael K. Dush
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA
| | - William W. Gordon
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Brent H. Wyatt
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA
| | - Peter White
- Institute for Genomic Medicine, Nationwide Children’s Hospital, and Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Colby T. Marvin
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Emmi Helle
- New Children’s Hospital and Pediatric Research Center, Helsinki University Hospital, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tiina Ojala
- New Children’s Hospital and Pediatric Research Center, Helsinki University Hospital, Helsinki, Finland
| | - James R. Priest
- Stanford University School of Medicine, Lucile Packard Children’s Hospital, Stanford, CA, USA
| | - Mary M. Jenkins
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Lynn M. Almli
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jennita Reefhuis
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Faith Pangilinan
- Genetics and Environment Interaction Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lawrence C. Brody
- Genetics and Environment Interaction Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kim L. McBride
- Center for Cardiovascular Research, Nationwide Children’s Hospital, and Division of Genetic and Genomic Medicine, Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Vidu Garg
- Center for Cardiovascular Research and The Heart Center, Nationwide Children’s Hospital, and Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Gary M. Shaw
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Paul A. Romitti
- Department of Epidemiology, College of Public Health, The University of Iowa, Iowa City, IA, USA
| | | | - Marilyn L. Browne
- Birth Defects Registry, New York State Department of Health, Albany, NY, USA
- Department of Epidemiology and Biostatistics, School of Public Health, University at Albany, Rensselaer, NY, USA
| | - Martha M. Werler
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Denise M. Kay
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - National Birth Defects Prevention Study
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Invitae, San Francisco, CA, USA
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Institute for Genomic Medicine, Nationwide Children’s Hospital, and Department of Pediatrics, The Ohio State University, Columbus, OH, USA
- New Children’s Hospital and Pediatric Research Center, Helsinki University Hospital, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Stanford University School of Medicine, Lucile Packard Children’s Hospital, Stanford, CA, USA
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA, USA
- Genetics and Environment Interaction Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
- Center for Cardiovascular Research, Nationwide Children’s Hospital, and Division of Genetic and Genomic Medicine, Department of Pediatrics, The Ohio State University, Columbus, OH, USA
- Center for Cardiovascular Research and The Heart Center, Nationwide Children’s Hospital, and Department of Pediatrics, The Ohio State University, Columbus, OH, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Epidemiology, College of Public Health, The University of Iowa, Iowa City, IA, USA
- University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Birth Defects Registry, New York State Department of Health, Albany, NY, USA
- Department of Epidemiology and Biostatistics, School of Public Health, University at Albany, Rensselaer, NY, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY, USA
- Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - University of Washington Center for Mendelian Genomics
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Invitae, San Francisco, CA, USA
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Institute for Genomic Medicine, Nationwide Children’s Hospital, and Department of Pediatrics, The Ohio State University, Columbus, OH, USA
- New Children’s Hospital and Pediatric Research Center, Helsinki University Hospital, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Stanford University School of Medicine, Lucile Packard Children’s Hospital, Stanford, CA, USA
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA, USA
- Genetics and Environment Interaction Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
- Center for Cardiovascular Research, Nationwide Children’s Hospital, and Division of Genetic and Genomic Medicine, Department of Pediatrics, The Ohio State University, Columbus, OH, USA
- Center for Cardiovascular Research and The Heart Center, Nationwide Children’s Hospital, and Department of Pediatrics, The Ohio State University, Columbus, OH, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Epidemiology, College of Public Health, The University of Iowa, Iowa City, IA, USA
- University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Birth Defects Registry, New York State Department of Health, Albany, NY, USA
- Department of Epidemiology and Biostatistics, School of Public Health, University at Albany, Rensselaer, NY, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY, USA
- Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Seema Mital
- Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Jessica X. Chong
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Michael J. Bamshad
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| |
Collapse
|
5
|
Huang YP, Huang WW, Tsai KF, Shiao LR, Yang ZH, Tseng SY, Lin YH, Chen CY, Chan P, Leung YM. CDN1163, a SERCA activator, causes intracellular Ca 2+ leak, mitochondrial hyperpolarization and cell cycle arrest in mouse neuronal N2A cells. Neurotoxicology 2023; 98:9-15. [PMID: 37429421 DOI: 10.1016/j.neuro.2023.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 06/11/2023] [Accepted: 07/07/2023] [Indexed: 07/12/2023]
Abstract
OBJECTIVE Activity or expression of sarcoplasmic/endoplasmic reticulum Ca2+ ATPase (SERCA) is diminished in some disease states such as cardiac failure and diabetes mellitus. A newly developed activator of SERCA, CDN1163, reportedly rescued or alleviated pathological conditions attributed to dysfunctional SERCA. We examined whether CDN1163 could relieve mouse neuronal N2A cell growth inhibition caused by cyclopiazonic acid (CPA, SERCA inhibitor). We also examined how CDN1163 affected cytosolic Ca2+, mitochondrial Ca2+ and mitochondrial membrane potential. METHODS Cell viability was measured by MTT assay and trypan blue exclusion test. Cytosolic Ca2+, mitochondrial Ca2+ and mitochondrial membrane potential were measured using fura 2, Rhod-2 and JC-1, respectively, as fluorescent probes. RESULTS CDN1163 (10 μM) itself suppressed cell proliferation, and did not alleviate CPA's inhibitory effect (and vice versa). Cell cycle was arrested at the G1 phase after CDN1163 treatment. CDN1163 treatment caused a slow yet persistent cytosolic [Ca2+] elevation partly due to Ca2+ release from an internal store other than the CPA-sensitive endoplasmic reticulum (ER). Treatment with CDN1163 for 3 h raised mitochondrial Ca2+ level and such increase was suppressed by MCU-i4 (an inhibitor of mitochondria Ca2+ uniporter, MCU), suggesting Ca2+ entered the mitochondrial matrix through MCU. Treatment of cells with CDN1163 up to 2 days resulted in mitochondrial hyperpolarization. CONCLUSION CDN1163 caused internal Ca2+ leak, cytosolic Ca2+ overload, mitochondrial Ca2+ elevation and hyperpolarization, cell cycle arrest and cell growth inhibition.
Collapse
Affiliation(s)
- Yi-Ping Huang
- Department of Physiology, China Medical University, Taichung 40402, Taiwan
| | - Wen-Wei Huang
- Department of Internal Medicine, Gastroenterology and Hepatology Section, An Nan Hospital, China Medical University, Tainan, Taiwan
| | - Kun-Feng Tsai
- Department of Internal Medicine, Gastroenterology and Hepatology Section, An Nan Hospital, China Medical University, Tainan, Taiwan; Department of Medical Sciences Industry, Chang Jung Christian University, Tainan, Taiwan
| | - Lian-Ru Shiao
- Department of Physiology, China Medical University, Taichung 40402, Taiwan
| | - Zih-He Yang
- Department of Physiology, China Medical University, Taichung 40402, Taiwan
| | - Shao-Yun Tseng
- Department of Physiology, China Medical University, Taichung 40402, Taiwan
| | - Yu-Hsien Lin
- Department of Physiology, China Medical University, Taichung 40402, Taiwan
| | - Cing-Yu Chen
- Department of Cosmetic Science, Providence University, Taichung, Taiwan; School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Paul Chan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yuk-Man Leung
- Department of Physiology, China Medical University, Taichung 40402, Taiwan.
| |
Collapse
|
6
|
Zhao M, Li N, Zhou H. SGLT1: A Potential Drug Target for Cardiovascular Disease. Drug Des Devel Ther 2023; 17:2011-2023. [PMID: 37435096 PMCID: PMC10332373 DOI: 10.2147/dddt.s418321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 06/29/2023] [Indexed: 07/13/2023] Open
Abstract
SGLT1 and SGLT2 are the two main members of the sodium-glucose cotransporters (SGLTs), which are mainly responsible for glucose reabsorption in the body. In recent years, many large clinical trials have shown that SGLT2 inhibitors have cardiovascular protection for diabetic and non-diabetic patients independent of lowering blood glucose. However, SGLT2 was barely detected in the hearts of humans and animals, while SGLT1 was highly expressed in myocardium. As SGLT2 inhibitors also have a moderate inhibitory effect on SGLT1, the cardiovascular protection of SGLT2 inhibitors may be due to SGLT1 inhibition. SGLT1 expression is associated with pathological processes such as cardiac oxidative stress, inflammation, fibrosis, and cell apoptosis, as well as mitochondrial dysfunction. The purpose of this review is to summarize the protective effects of SGLT1 inhibition on hearts in various cell types, including cardiomyocytes, endothelial cells, and fibroblasts in preclinical studies, and to highlight the underlying molecular mechanisms of protection against cardiovascular diseases. Selective SGLT1 inhibitors could be considered a class of drugs for cardiac-specific therapy in the future.
Collapse
Affiliation(s)
- Mengnan Zhao
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People’s Republic of China
| | - Na Li
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People’s Republic of China
| | - Hong Zhou
- Department of Endocrinology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People’s Republic of China
| |
Collapse
|
7
|
Lu SY, Tsai BCK, Van Thao D, Lai CH, Chen MYC, Kuo WW, Kuo CH, Lin KH, Hsieh DJY, Huang CY. Cardiac-specific overexpression of insulin-like growth factor II receptor-α interferes with the regulation of calcium homeostasis in the heart under hyperglycemic conditions. Mol Biol Rep 2023; 50:4329-4338. [PMID: 36928640 DOI: 10.1007/s11033-023-08327-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/09/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND Diabetic cardiomyopathy is a progressive disease caused by inexplicit mechanisms, and a novel factor, insulin-like growth factor II receptor-α (IGF-IIRα), may contribute to aggravating its pathogenesis. We hypothesized that IGF-IIRα could intensify diabetic heart injury. METHODS AND RESULTS To demonstrate the potential role of IGF-IIRα in the diabetic heart, we used (SD-TG [IGF-IIRα]) transgenic rat model with cardiac-specific overexpression of IGF-IIRα, along with H9c2 cells, to study the effects of IGF-IIRα in the heart under hyperglycemic conditions. IGF-IIRα was found to remodel calcium homeostasis and intracellular Ca2+ overload-induced autophagy disturbance in the heart during diabetes. IGF-IIRα overexpression induced intracellular Ca2+ alteration by downregulating phosphorylated phospholamban/sarcoplasmic/endoplasmic reticulum calcium-ATPase 2a (PLB/SERCA2a), resulting in the suppression of Ca2+ uptake into the endoplasmic reticulum. Additionally, IGF-IIRα itself contributed to Ca2+ withdrawal from the endoplasmic reticulum by increasing the expression of CaMKIIδ in the active form. Furthermore, alterations in Ca2+ homeostasis significantly dysregulated autophagy in the heart during diabetes. CONCLUSIONS Our study reveals the novel role of IGF-IIRα in regulating cardiac intracellular Ca2+ homeostasis and its related autophagy interference, which contribute to the development of diabetic cardiomyopathy. In future, the present study findings have implications in the development of appropriate therapy to reduce diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Shang-Yeh Lu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Division of Cardiovascular Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
| | - Bruce Chi-Kang Tsai
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Dao Van Thao
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chin-Hu Lai
- Division of Cardiovascular Surgery, Department of Surgery, Taichung Armed Force General Hospital, Taichung, Taiwan
- National Defense Medical Center, Taipei, Taiwan
| | | | - Wei-Wen Kuo
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
- Ph.D. Program for Biotechnology Industry, China Medical University, Taichung, Taiwan
| | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
- Department of Kinesiology and Health Science, College of William and Mary, Williamsburg, VA, USA
| | - Kuan-Ho Lin
- Division of Cardiovascular Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
- Department of Emergency Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Dennis Jine-Yuan Hsieh
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan.
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan.
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan.
| |
Collapse
|
8
|
de Souza EG, Peixoto JVC, Rank C, Petterle RR, Fogaça RTH, Wolska BM, Dias FAL. Effects of High-Intensity Interval Training and Continuous Training on Exercise Capacity, Heart Rate Variability and Isolated Hearts in Diabetic Rats. Arq Bras Cardiol 2022; 120:e20220396. [PMID: 36629606 PMCID: PMC9833297 DOI: 10.36660/abc.20220396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 09/21/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND High-intensity interval training (HIIT) has been suggested as an alternative for continuous training (CT) in people with diabetes mellitus (DM) due to its short duration and potential to improve adherence to exercise. However, data on its impact on heart rate variability (HRV) are scarce. OBJECTIVES To assess and compare the effects of HIIT and CT on exercise capacity, HRV and isolated hearts in diabetic rats. METHODS DM (intravenous streptozotocin, 45 mg.kg -1 ) and control (C) animals performed 20 sessions (5 days/week, 50 min, for 4 weeks) of CT on a treadmill (70% of maximal exercise capacity) or HIIT (cycles of 1:1min at 50% and 90% of maximal exercise capacity). HRV was assessed by continuous electrocardiogram, and cardiac function assessed in isolated perfused hearts. For data analysis, we used the framework of the multivariate covariance generalized linear model or one-way ANOVA followed by Tukey's test, considering p<0.05 as significant. RESULTS Higher exercise capacity (m/min) was achieved in HIIT (DM-HIIT: 36.5 [IQR 30.0-41.3]; C-HIIT: 41.5 [37.8-44.5], both n=10) compared to CT (DM-CT: 29.0 [23.8-33.0]; C-CT: 32.0 [29.5-37.0], both n=10) (p<0.001). Heart rate (bpm) was lower in DM compared to controls (p<0.001) both in vivo (DM-HIIT:348±51, C-HIIT:441±66, DM-CT:361±70, C-CT:437±38) and in isolated hearts. There were no differences in HRV between the groups. Maximum and minimal dP/dt were reduced in DM, except +dP/dt in DM-HIIT vs. C-HIIT (mean difference: 595.5±250.3, p=0.190). CONCLUSION Short-term HIIT promotes greater improvement in exercise performance compared to CT, including in DM, without causing significant changes in HRV.
Collapse
Affiliation(s)
- Eduardo Gomes de Souza
- Universidade Federal do ParanáDepartamento de FisiologiaCuritibaPRBrasil Universidade Federal do Paraná – Departamento de Fisiologia , Curitiba , PR – Brasil
| | - João Victor Capelli Peixoto
- Universidade Federal do ParanáDepartamento de FisiologiaCuritibaPRBrasil Universidade Federal do Paraná – Departamento de Fisiologia , Curitiba , PR – Brasil
| | - Claucio Rank
- Universidade Federal do ParanáDepartamento de FisiologiaCuritibaPRBrasil Universidade Federal do Paraná – Departamento de Fisiologia , Curitiba , PR – Brasil
| | - Ricardo Rasmussen Petterle
- Universidade Federal do ParanáDepartamento de Medicina IntegradaCuritibaPRBrasil Universidade Federal do Paraná – Departamento de Medicina Integrada , Curitiba , PR – Brasil
| | - Rosalvo Tadeu Hochmuller Fogaça
- Universidade Federal do ParanáDepartamento de FisiologiaCuritibaPRBrasil Universidade Federal do Paraná – Departamento de Fisiologia , Curitiba , PR – Brasil
| | - Beata Maria Wolska
- University of Illinois at ChicagoChicagoIllinoisEUA University of Illinois at Chicago – Medicine, Physiology and Biophysics, Chicago , Illinois – EUA
| | - Fernando Augusto Lavezzo Dias
- Universidade Federal do ParanáDepartamento de FisiologiaCuritibaPRBrasil Universidade Federal do Paraná – Departamento de Fisiologia , Curitiba , PR – Brasil
| |
Collapse
|
9
|
Feng Y, Qin R, Xu L, Ma X, Ding D, Li S, Chen L, Liu Y, Sun W, Chen H. Ion drugs for precise orthotopic tumor management by in situ the generation of toxic ion and drug pools. Am J Cancer Res 2022; 12:734-746. [PMID: 34976210 PMCID: PMC8692900 DOI: 10.7150/thno.66468] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/21/2021] [Indexed: 11/05/2022] Open
Abstract
Background: Asymmetric intracellular and extracellular ionic gradients are critical to the survivability of mammalian cells. Given the importance of manganese (Mn2+), calcium (Ca2+), and bicarbonate (HCO3 -) ions, any alteration of the ion-content balance could induce a series of cellular responses. HCO3 - plays an indispensable role for Mn-mediated Fenton-like reaction, but this is difficult to achieve because bicarbonates are tightly regulated by live cells, and are limited in anticancer efficacy. Methods: A responsive and biodegradable biomineral, Mn-doped calcium carbonate integrated with dexamethasone phosphate (DEX) (Mn:CaCO3-DEX), was reported to enable synergistic amplification of tumor oxidative stress, reduce inflammation, and induce Ca-overload cell apoptosis by elevating the intracellular and extracellular ionic gradients. Results: Under the acidic environment in tumor region, the ions (Mn2+, CO3 2-, Ca2+) were released by the degradation of Mn:CaCO3-DEX and then escalated oxidative stresses by triggering a HCO3 --indispensable Mn-based Fenton-like reaction and breaking Ca2+ ion homeostasis to cause oxidative stress in cells and calcification. The released anti-inflammatory and antitumor drug, DEX, could alleviate the inflammatory environment. The investigations in vitro and in vivo demonstrated that the synergistic oncotherapy could effectively inhibit the growth of subcutaneous tumors and orthotopic liver tumors. Notably, normal cells showed greater tolerance of the synergistic influences. Conclusion: As an ion drug, Mn:CaCO3-DEX is an excellent potential diagnostic agent for precise orthotopic tumor management by the generation in situ of toxic ion and drug pools in the environment of tumor region, with synergistic effects of enhanced chemodynamic therapy, calcification, and anti-inflammation effects.
Collapse
Affiliation(s)
- Yushuo Feng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Ruixue Qin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Lihua Xu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xiaoqian Ma
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Dandan Ding
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Shi Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Lei Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yaqing Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Wenjing Sun
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hongmin Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| |
Collapse
|
10
|
Calpain-Mediated Mitochondrial Damage: An Emerging Mechanism Contributing to Cardiac Disease. Cells 2021; 10:cells10082024. [PMID: 34440793 PMCID: PMC8392834 DOI: 10.3390/cells10082024] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/19/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022] Open
Abstract
Calpains belong to the family of calcium-dependent cysteine proteases expressed ubiquitously in mammals and many other organisms. Activation of calpain is observed in diseased hearts and is implicated in cardiac cell death, hypertrophy, fibrosis, and inflammation. However, the underlying mechanisms remain incompletely understood. Recent studies have revealed that calpains target and impair mitochondria in cardiac disease. The objective of this review is to discuss the role of calpains in mediating mitochondrial damage and the underlying mechanisms, and to evaluate whether targeted inhibition of mitochondrial calpain is a potential strategy in treating cardiac disease. We expect to describe the wealth of new evidence surrounding calpain-mediated mitochondrial damage to facilitate future mechanistic studies and therapy development for cardiac disease.
Collapse
|
11
|
Activation of Ca 2+/Calmodulin-Dependent Protein Kinase II (CaMKII) with Lidocaine Provokes Pyroptosis of Glioblastoma Cells. Bull Exp Biol Med 2021; 171:297-304. [PMID: 34302204 DOI: 10.1007/s10517-021-05216-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Indexed: 10/20/2022]
Abstract
The study examines the problem whether pyroptosis of U87-MG glioblastoma cells can result from activation of Ca2+/calmodulin-dependent protein kinase II (CaMKII) by a local anesthetic. Glioblastoma cells exposed to various concentrations of typical local anesthetic lidocaine demonstrated augmented cytosolic flux of Ca2+, while suppression of CaMKII expression with the corresponding siRNA significantly inhibited this effect in cells treated with 2 mM lidocaine. Lidocaine up-regulated the expression of mRNA caspase-3 and gasdermin GSDME proteins, whereas silencing of CaMKII gene with siRNA significantly moderated this effect. In addition, lidocaine inhibited proliferation of U87-MG cells, and this effect was prevented by silencing CaMKII gene. Thus, lidocaine activated protein kinase CaMKII, which phosphorylated TRPV1 ion channels and induced calcium overload of U87-MG glioblastoma cells, thereby provoking their pyroptosis.
Collapse
|
12
|
Maiuolo J, Carresi C, Gliozzi M, Musolino V, Scarano F, Coppoletta AR, Guarnieri L, Nucera S, Scicchitano M, Bosco F, Ruga S, Zito MC, Macri R, Cardamone A, Serra M, Mollace R, Tavernese A, Mollace V. Effects of Bergamot Polyphenols on Mitochondrial Dysfunction and Sarcoplasmic Reticulum Stress in Diabetic Cardiomyopathy. Nutrients 2021; 13:nu13072476. [PMID: 34371986 PMCID: PMC8308586 DOI: 10.3390/nu13072476] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/18/2021] [Accepted: 07/18/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular disease is the leading cause of death and disability in the Western world. In order to safeguard the structure and the functionality of the myocardium, it is extremely important to adequately support the cardiomyocytes. Two cellular organelles of cardiomyocytes are essential for cell survival and to ensure proper functioning of the myocardium: mitochondria and the sarcoplasmic reticulum. Mitochondria are responsible for the energy metabolism of the myocardium, and regulate the processes that can lead to cell death. The sarcoplasmic reticulum preserves the physiological concentration of the calcium ion, and triggers processes to protect the structural and functional integrity of the proteins. The alterations of these organelles can damage myocardial functioning. A proper nutritional balance regarding the intake of macronutrients and micronutrients leads to a significant improvement in the symptoms and consequences of heart disease. In particular, the Mediterranean diet, characterized by a high consumption of plant-based foods, small quantities of red meat, and high quantities of olive oil, reduces and improves the pathological condition of patients with heart failure. In addition, nutritional support and nutraceutical supplementation in patients who develop heart failure can contribute to the protection of the failing myocardium. Since polyphenols have numerous beneficial properties, including anti-inflammatory and antioxidant properties, this review gathers what is known about the beneficial effects of polyphenol-rich bergamot fruit on the cardiovascular system. In particular, the role of bergamot polyphenols in mitochondrial and sarcoplasmic dysfunctions in diabetic cardiomyopathy is reported.
Collapse
Affiliation(s)
- Jessica Maiuolo
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Cristina Carresi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Micaela Gliozzi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Vincenzo Musolino
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Federica Scarano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Anna Rita Coppoletta
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Lorenza Guarnieri
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Saverio Nucera
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Miriam Scicchitano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Francesca Bosco
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Stefano Ruga
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Maria Caterina Zito
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Roberta Macri
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Antonio Cardamone
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Maria Serra
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Rocco Mollace
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- IRCCS San Raffaele, Via di Valcannuta 247, 00133 Rome, Italy
| | - Annamaria Tavernese
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Vincenzo Mollace
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario di Germaneto, 88100 Catanzaro, Italy; (J.M.); (C.C.); (M.G.); (V.M.); (F.S.); (A.R.C.); (L.G.); (S.N.); (M.S.); (F.B.); (S.R.); (M.C.Z.); (R.M.); (A.C.); (M.S.); (R.M.); (A.T.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
- IRCCS San Raffaele, Via di Valcannuta 247, 00133 Rome, Italy
- Correspondence: ; Tel.: +39-327-475-8006
| |
Collapse
|
13
|
Dai SH, Li YW, Hong QX, Su T, Xu SY. Exaggerated activities of TRPM7 underlie bupivacaine-induced neurotoxicity in the SH-SY5Y cells preconditioned with high glucose. J Biochem Mol Toxicol 2021; 35:e22826. [PMID: 34060177 DOI: 10.1002/jbt.22826] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 02/28/2021] [Accepted: 05/18/2021] [Indexed: 11/12/2022]
Abstract
Hyperglycemia is considered a risk factor for the enhancement of local anesthetic-induced neurotoxicity. Transient receptor potential melastatin 7 (TRPM7), a kinase-coupled cation channel, has been implicated in a variety of neuropathological processes, including intracellular calcium disturbance and high glucose-induced neuropathy. In this study, we investigated whether TRPM7-related pathophysiology is involved in bupivacaine-induced neurotoxicity in SH-SY5Y cells and how hyperglycemia acts as a risk factor. For initial neurotoxicity evaluation, it was confirmed that cell damage and apoptosis induced by acute exposure to bupivacaine were dependent on its concentration and glucose preconditioning. High glucose preconditioning facilitated the bupivacaine-induced fast and temporary rise in intracellular free calcium concentration ([Ca2+ ]i ), which was attributed to both calcium influx through TRPM7 and calcium store release. Additionally, bupivacaine was shown to increase TRPM7-like currents, particularly in cells preconditioned with high glucose. Bupivacaine-induced neurotoxicity in hyperglycemia was correlated with extracellular signal-regulated kinase (ERK), but not protein kinase B (AKT) activation. Inhibition of TRPM7 and ERK activity alleviates bupivacaine neurotoxicity. These results suggest that therapeutically targeting TRPM7-related pathophysiological changes could be a potential strategy for treating local anesthetic-induced neurotoxicity exacerbated by hyperglycemia.
Collapse
Affiliation(s)
- Shuang-Hua Dai
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Ya-Wen Li
- Department of Anesthesiology, Shenzhen Maternal and Child Health, Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Qing-Xiong Hong
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Tao Su
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shi-Yuan Xu
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Aboalgasm H, Ballo R, Mkatazo T, Gwanyanya A. Hyperglycaemia-Induced Contractile Dysfunction and Apoptosis in Cardiomyocyte-Like Pulsatile Cells Derived from Mouse Embryonic Stem Cells. Cardiovasc Toxicol 2021; 21:695-709. [PMID: 33983555 DOI: 10.1007/s12012-021-09660-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/07/2021] [Indexed: 10/21/2022]
Abstract
Hyperglycaemia, a key metabolic abnormality in diabetes mellitus, is implicated in pathological cardiogenesis during embryological development. However, the underlying mechanisms and potential therapeutic targets remain unknown. We, therefore, studied the effect of hyperglycaemia on mouse embryonic stem cell (mESC) cardiac differentiation. The mESCs were differentiated via embryoid body (EB) formation and cultured under conditions with baseline (25 mM) or high (50 mM) glucose. Time-lapse microscopy images of pulsatile mESCs and Ca2+ transients were recorded. Biomarkers of cellular changes were detected using immunocytochemistry, terminal deoxynucleotidyl transferase dUTP nick-end labelling (TUNEL) assay, and Western blot analyses. Differentiated, spontaneously beating mESCs stained positive for cardiac troponin T, α-actinin 2, myosin heavy chain, and connexin 43. Hyperglycaemia decreased the EB diameter and number of beating EBs as well as the cellular amplitude of contraction, the Ca2+ transient, and the contractile response to caffeine (1 mM), but had no effect on the expression of the sarco-endoplasmic reticulum calcium transport ATPase 2 (SERCA 2). Furthermore, hyperglycaemia decreased the expression of B cell lymphoma 2 (Bcl-2) and increased the expression of cytoplasmic cytochrome c and the number of TUNEL-positive cells, but had no effect on the expression of one of the mitochondrial fusion regulatory proteins, optic atrophy protein 1 (OPA1). Overall, hyperglycaemia suppressed the mESC cardiomyocyte-like differentiation and induced contractile dysfunction. The results are consistent with mechanisms involving abnormal Ca2+ handling and mitochondrial-dependent apoptosis, factors which represent potential therapeutic targets in developmental diabetic cardiac disease.
Collapse
Affiliation(s)
- Hamida Aboalgasm
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925, South Africa
| | - Robea Ballo
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925, South Africa
| | - Thulisa Mkatazo
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925, South Africa
| | - Asfree Gwanyanya
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, 7925, South Africa.
| |
Collapse
|
15
|
Palomer X, Aguilar-Recarte D, García R, Nistal JF, Vázquez-Carrera M. Sirtuins: To Be or Not To Be in Diabetic Cardiomyopathy. Trends Mol Med 2021; 27:554-571. [PMID: 33839024 DOI: 10.1016/j.molmed.2021.03.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/25/2021] [Accepted: 03/10/2021] [Indexed: 12/16/2022]
Abstract
Diabetic cardiomyopathy is the leading cause of death among people with diabetes. Despite its severity and poor prognosis, there are currently no approved specific drugs to prevent or even treat diabetic cardiomyopathy. There is a need to understand the pathogenic mechanisms underlying the development of diabetic cardiomyopathy to design new therapeutic strategies. These mechanisms are complex and intricate and include metabolic dysregulation, inflammation, oxidative stress, fibrosis, and apoptosis. Sirtuins, a group of deacetylase enzymes, play an important role in all these processes and are, therefore, potential molecular targets for treating this disease. In this review, we discuss the role of sirtuins in the heart, focusing on their contribution to the pathogenesis of diabetic cardiomyopathy and how their modulation could be therapeutically useful.
Collapse
Affiliation(s)
- Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB); and Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Pediatric Research Institute - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - David Aguilar-Recarte
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB); and Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Pediatric Research Institute - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Raquel García
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Santander, Spain; Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - J Francisco Nistal
- Servicio de Cirugía Cardiovascular, Hospital Universitario Marqués de Valdecilla, Santander, Spain; Departamento de Ciencias Médicas y Quirúrgicas, Facultad de Medicina, Universidad de Cantabria, Santander, Spain; Instituto de Investigación Marqués de Valdecilla (IDIVAL); Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Santander, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB); and Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Pediatric Research Institute - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain.
| |
Collapse
|
16
|
Preventing Myocardial Injury Following Non-Cardiac Surgery: A Potential Role for Preoperative Antioxidant Therapy with Ubiquinone. Antioxidants (Basel) 2021; 10:antiox10020276. [PMID: 33579045 PMCID: PMC7916807 DOI: 10.3390/antiox10020276] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/05/2021] [Accepted: 02/05/2021] [Indexed: 02/07/2023] Open
Abstract
Over 240 million non-cardiac operations occur each year and are associated with a 15-20% incidence of adverse perioperative cardiovascular events. Unfortunately, preoperative therapies that have been useful for chronic ischemic heart diseases, such as coronary artery revascularization, antiplatelet agents, and beta-blockers have failed to improve outcomes. In a pre-clinical swine model of ischemic heart disease, we showed that daily administration of ubiquinone (coenzyme Q10, CoQ10) enhances the antioxidant status of mitochondria within chronically ischemic heart tissue, potentially via a PGC1α-dependent mechanism. In a randomized controlled trial, among high-risk patients undergoing elective vascular surgery, we showed that NT Pro-BNP levels are an important means of risk-stratification during the perioperative period and can be lowered with administration of CoQ10 (400 mg/day) for 3 days prior to surgery. The review provides background information for the role of oxidant stress and inflammation during high-risk operations and the potential novel application of ubiquinone as a preoperative antioxidant therapy that might reduce perioperative adverse cardiovascular outcomes.
Collapse
|
17
|
Matsumoto N, Omagari D, Ushikoshi-Nakayama R, Yamazaki T, Inoue H, Saito I. Hyperglycemia Induces Generation of Reactive Oxygen Species and Accelerates Apoptotic Cell Death in Salivary Gland Cells. Pathobiology 2021; 88:234-241. [PMID: 33556940 DOI: 10.1159/000512639] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/27/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Type-2 diabetes mellitus (T2DM) is associated with several systemic vascular symptoms and xerostomia. It is considered that hyperglycemia-induced polyuria and dehydration cause decreased body-water volume, leading to decreased saliva secretion and, ultimately, xerostomia. In T2DM, increased production of reactive oxygen species (ROS) causes tissue damage to vascular endothelial cells as well as epithelial tissue, including pancreas and cornea. Hence, a similar phenomenon may occur in other tissues and glands in a hyperglycemic environment. METHODS Salivary gland tissue injury was examined, using T2DM model mouse (db/db). Transferase-mediated dUTP nick-end labeling (TUNEL) was conducted to evaluate tissue injury. The levels of malondialdehyde (MDA) and 8-hydroxy-2'-deoxyguanosine, Bax/Bcl-2 ratio were measured as indicator of oxidative stress. Moreover, in vitro ROS production and cell injury was evaluated by mouse salivary gland-derived normal cells under high-glucose condition culture. RESULTS In vivo and in vitro analysis showed a higher percentage of TUNEL-positive cells and higher levels of MDA and 8-hydroxy-2'-deoxyguanosine in salivary gland tissue of db/db mice. This suggests damage of saliva secretion-associated lipids and DNA by hyperglycemic-induced oxidative stress. To analyze the mechanism by which hyperglycemia promotes ROS production, mouse salivary gland-derived cells were isolated. The cell culture with high-glucose medium enhanced ROS production and promotes apoptotic and necrotic cell death. CONCLUSION These findings suggest a novel mechanism whereby hyperglycemic-induced ROS production promotes salivary gland injury, resulting in hyposalivation.
Collapse
Affiliation(s)
- Naoyuki Matsumoto
- Department of Pathology, Tsurumi University School of Dental Medicine, Yokohama, Japan,
| | - Daisuke Omagari
- Department of Pathology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | | | - Tomoe Yamazaki
- Department of Pathology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Hiroko Inoue
- Department of Pharmaceutical Sciences, Nihon Pharmaceutical University, Saitama, Japan
| | - Ichiro Saito
- Department of Pathology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| |
Collapse
|
18
|
Yang Y, Du J, Xu R, Shen Y, Yang D, Li D, Hu H, Pei H, Yang Y. Melatonin alleviates angiotensin-II-induced cardiac hypertrophy via activating MICU1 pathway. Aging (Albany NY) 2020; 13:493-515. [PMID: 33259334 PMCID: PMC7834983 DOI: 10.18632/aging.202159] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 08/19/2020] [Indexed: 01/19/2023]
Abstract
Mitochondrial calcium uptake 1 (MICU1) is a pivotal molecule in maintaining mitochondrial homeostasis under stress conditions. However, it is unclear whether MICU1 attenuates mitochondrial stress in angiotensin II (Ang-II)-induced cardiac hypertrophy or if it has a role in the function of melatonin. Here, small-interfering RNAs against MICU1 or adenovirus-based plasmids encoding MICU1 were delivered into left ventricles of mice or incubated with neonatal murine ventricular myocytes (NMVMs) for 48 h. MICU1 expression was depressed in hypertrophic myocardia and MICU1 knockdown aggravated Ang-II-induced cardiac hypertrophy in vivo and in vitro. In contrast, MICU1 upregulation decreased cardiomyocyte susceptibility to hypertrophic stress. Ang-II administration, particularly in NMVMs with MICU1 knockdown, led to significantly increased reactive oxygen species (ROS) overload, altered mitochondrial morphology, and suppressed mitochondrial function, all of which were reversed by MICU1 supplementation. Moreover, peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α)/MICU1 expression in hypertrophic myocardia increased with melatonin. Melatonin ameliorated excessive ROS generation, promoted mitochondrial function, and attenuated cardiac hypertrophy in control but not MICU1 knockdown NMVMs or mice. Collectively, our results demonstrate that MICU1 attenuates Ang-II-induced cardiac hypertrophy by inhibiting mitochondria-derived oxidative stress. MICU1 activation may be the mechanism underlying melatonin-induced protection against myocardial hypertrophy.
Collapse
Affiliation(s)
- Yi Yang
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Jin Du
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Rui Xu
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Yang Shen
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Dachun Yang
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - De Li
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Houxiang Hu
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Haifeng Pei
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Yongjian Yang
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| |
Collapse
|
19
|
Aluksanasuwan S, Plumworasawat S, Malaitad T, Chaiyarit S, Thongboonkerd V. High glucose induces phosphorylation and oxidation of mitochondrial proteins in renal tubular cells: A proteomics approach. Sci Rep 2020; 10:5843. [PMID: 32246012 PMCID: PMC7125224 DOI: 10.1038/s41598-020-62665-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 03/18/2020] [Indexed: 12/11/2022] Open
Abstract
Mitochondrial dysfunction has been thought to play roles in the pathogenesis of diabetic nephropathy (DN). However, precise mechanisms underlying mitochondrial dysfunction in DN remained unclear. Herein, mitochondria were isolated from renal tubular cells after exposure to normal glucose (5.5 mM glucose), high glucose (25 mM glucose), or osmotic control (5.5 mM glucose + 19.5 mM mannitol) for 96 h. Comparative proteomic analysis revealed six differentially expressed proteins among groups that were subsequently identified by tandem mass spectrometry (nanoLC-ESI-ETD MS/MS) and confirmed by Western blotting. Several various types of post-translational modifications (PTMs) were identified in all of these identified proteins. Interestingly, phosphorylation and oxidation were most abundant in mitochondrial proteins whose levels were exclusively increased in high glucose condition. The high glucose-induced increases in phosphorylation and oxidation of mitochondrial proteins were successfully confirmed by various assays including MS/MS analyses. Moreover, high glucose also increased levels of phosphorylated ezrin, intracellular ATP and ROS, all of which could be abolished by a p38 MAPK inhibitor (SB239063), implicating a role of p38 MAPK-mediated phosphorylation in high glucose-induced mitochondrial dysfunction. These data indicate that phosphorylation and oxidation of mitochondrial proteins are, at least in part, involved in mitochondrial dysfunction in renal tubular cells during DN.
Collapse
Affiliation(s)
- Siripat Aluksanasuwan
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Sirikanya Plumworasawat
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Thanyalak Malaitad
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Sakdithep Chaiyarit
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
20
|
Brennan S, Chen S, Makwana S, Martin CA, Sims MW, Alonazi ASA, Willets JM, Squire IB, Rainbow RD. A novel form of glycolytic metabolism-dependent cardioprotection revealed by PKCα and β inhibition. J Physiol 2019; 597:4481-4501. [PMID: 31241168 DOI: 10.1113/jp278332] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 06/24/2019] [Indexed: 01/21/2023] Open
Abstract
KEY POINTS Acute hyperglycaemia at the time of a heart attack worsens the outcome for the patient. Acute hyperglycaemia is not limited to diabetic patients and can be due to a stress response in non-diabetics. This study suggests that the damaging cardiac effects of hyperglycaemia can be reversed by selective PKC inhibition. If PKCα/β isoforms are inhibited, then high glucose itself becomes protective against ischaemic damage. Selective PKC inhibition may therefore be a useful therapeutic tool to limit the damage that can occur during a heart attack by stress-induced hyperglycaemia. ABSTRACT Hyperglycaemia has a powerful association with adverse prognosis for patients with acute coronary syndromes (ACS). Previous work shows that high glucose prevents ischaemic preconditioning and causes electrical and mechanical disruption via protein kinase C α/β (PKCα/β) activation. The present study aimed to: (i) determine whether the adverse clinical association of hyperglycaemia in ACS can be replicated in preclinical cellular models of ACS and (ii) determine the importance of PKCα/β activation to the deleterious effect of glucose. Freshly isolated rat, guinea pig or rabbit cardiomyocytes were exposed to simulated ischaemia after incubation in the presence of normal (5 mm) or high (20 mm) glucose in the absence or presence of small molecule or tat-peptide-linked PKCαβ inhibitors. In each of the four conditions, the following hallmarks of cardioprotection were recorded using electrophysiology or fluorescence imaging: cardiomyocyte contraction and survival, action potential stability and time to failure, intracellular calcium and ATP, mitochondrial depolarization, ischaemia-sensitive leak current, and time to Kir 6.2 opening. High glucose alone resulted in decreased cardiomyocyte contraction and survival; however, it also imparted cardioprotection in the presence of PKCα/β inhibitors. This cardioprotective phenotype displayed improvements in all of the measured parameters and decreased myocardium damage during whole heart coronary ligation experiments. High glucose is deleterious to cellular and whole-heart models of simulated ischaemia, in keeping with the clinical association of hyperglycaemia with an adverse outcome in ACS. PKCαβ inhibition revealed high glucose to show a cardioprotective phenotype in this setting. The results of the present study suggest the potential for the therapeutic application of PKCαβ inhibition in ACS associated with hyperglycaemia.
Collapse
Affiliation(s)
- Sean Brennan
- Department of Cardiovascular Sciences, University of Leicester, Glenfield General Hospital, Leicester, UK
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Shen Chen
- Department of Cardiovascular Sciences, University of Leicester, Glenfield General Hospital, Leicester, UK
| | - Samir Makwana
- Department of Cardiovascular Sciences, University of Leicester, Glenfield General Hospital, Leicester, UK
| | - Christopher A Martin
- Department of Cardiovascular Sciences, University of Leicester, Glenfield General Hospital, Leicester, UK
| | - Mark W Sims
- Department of Cardiovascular Sciences, University of Leicester, Glenfield General Hospital, Leicester, UK
| | - Asma S A Alonazi
- Department of Molecular and Cellular Biology, University of Leicester, Leicester, UK
- Department of Pharmacology and Toxicology, Pharmacy College, King Saud University, Riyadh, Saudi Arabia
| | - Jonathan M Willets
- Department of Molecular and Cellular Biology, University of Leicester, Leicester, UK
| | - Iain B Squire
- Department of Cardiovascular Sciences, University of Leicester, Glenfield General Hospital, Leicester, UK
- Leicester NIHR Biomedical Research Centre, Glenfield General Hospital, Leicester, UK
| | - Richard D Rainbow
- Department of Cardiovascular Sciences, University of Leicester, Glenfield General Hospital, Leicester, UK
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
21
|
Davargaon RS, Sambe AD, Muthangi V V S. Trolox prevents high glucose-induced apoptosis in rat myocardial H9c2 cells by regulating GLUT-4 and antioxidant defense mechanism. IUBMB Life 2019; 71:1876-1895. [PMID: 31359611 DOI: 10.1002/iub.2133] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 07/01/2019] [Indexed: 12/12/2022]
Abstract
Redox imbalance due to hyperglycemia is a causative factor for an increased generation of reactive oxygen species (ROS) that leads to mitochondrial dysfunction and the release of cytochrome-c. The aim of the present study is to elucidate the functional role of oxidative stress (OS) in the induction of apoptosis in H9c2 cells in the hyperglycemic state through glucose transporter-4 (GLUT-4) regulation and antioxidant status. H9c2 cells were incubated with 15, 24, and 33 mM glucose for 24, 48, and 72 hr to induce hyperglycemic stress. Hyperglycemic episodes have significantly influenced GLUT-4 mRNA regulation, depleted glutathione (GSH) and its associated enzymes, reduced cellular antioxidant enzymes (AOEs), caused nuclear condensation, and induced apoptosis by activating caspase-9 and 3 and annexin V binding in a concentration and duration-dependent manner. Trolox pretreatment significantly enhanced the GLUT-4 mRNA and antioxidant defense mechanism, suppressed nuclear condensation, and prevented cytochrome-c release, thereby reducing mitochondrial-dependent apoptosis. The present study shows that the toxic effect of high glucose is significantly regulated and that OS induction can be prevented through a water-soluble vitamin E analog "Trolox" treatment.
Collapse
Affiliation(s)
| | - Asha Devi Sambe
- Laboratory of Gerontology, Department of Zoology, J.B. Campus, Bangalore University, Bengaluru, Karnataka, India
| | | |
Collapse
|
22
|
Yin MS, Zhang YC, Xu SH, Liu JJ, Sun XH, Liang C, Wang Y, Li J, Wang FW, Wang QL, Mu YL. Puerarin prevents diabetic cardiomyopathy in vivo and in vitro by inhibition of inflammation. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2019; 21:476-493. [PMID: 29322879 DOI: 10.1080/10286020.2017.1405941] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 11/13/2017] [Indexed: 06/07/2023]
Abstract
Diabetic cardiomyopathy (DCM) is one of the chief diabetes mellitus complications. Inflammation factors may be one reason for the damage from DM. The purpose of this research is to study the potential protective effects of puerarin on DM and the possible mechanisms of action related to NF-κB signal pathway. Following administration of puerarin to the disease model rat, several changes were observed including the changes of serum biochemical index, improved diastolic dysfunction, and enhanced endogenous antioxidant enzymes activities, further NF-κB signaling activation. Puerarin showed cardio-protective effects on DCM by inhibiting inflammation, and it might be a potential candidate for the treatment of DCM.
Collapse
Affiliation(s)
- Mao-Shan Yin
- a Center for Drug Evaluation , Food and Drug Administration , Beijing 100038 , China
- b Key Laboratory for Rare Disease of Shandong Province, Department of Pharmacology , Institute of Materia Medica, Shandong Academy of Medical Sciences , Jinan , China
| | - Yi-Chi Zhang
- c Department of Pharmacy , Jinan Children's Hospital , Jinan 250022 , China
| | - Shu-Hong Xu
- d State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica , Chinese Academy of Medical Sciences & Peking Union Medical College , Beijing 100050 , China
| | - Jing-Jing Liu
- b Key Laboratory for Rare Disease of Shandong Province, Department of Pharmacology , Institute of Materia Medica, Shandong Academy of Medical Sciences , Jinan , China
| | - Xiao-Hui Sun
- b Key Laboratory for Rare Disease of Shandong Province, Department of Pharmacology , Institute of Materia Medica, Shandong Academy of Medical Sciences , Jinan , China
| | - Chen Liang
- b Key Laboratory for Rare Disease of Shandong Province, Department of Pharmacology , Institute of Materia Medica, Shandong Academy of Medical Sciences , Jinan , China
| | - Yan Wang
- b Key Laboratory for Rare Disease of Shandong Province, Department of Pharmacology , Institute of Materia Medica, Shandong Academy of Medical Sciences , Jinan , China
| | - Jie Li
- b Key Laboratory for Rare Disease of Shandong Province, Department of Pharmacology , Institute of Materia Medica, Shandong Academy of Medical Sciences , Jinan , China
| | - Fu-Wen Wang
- b Key Laboratory for Rare Disease of Shandong Province, Department of Pharmacology , Institute of Materia Medica, Shandong Academy of Medical Sciences , Jinan , China
| | - Qing-Li Wang
- a Center for Drug Evaluation , Food and Drug Administration , Beijing 100038 , China
| | - Yan-Ling Mu
- b Key Laboratory for Rare Disease of Shandong Province, Department of Pharmacology , Institute of Materia Medica, Shandong Academy of Medical Sciences , Jinan , China
| |
Collapse
|
23
|
Naz H, Tarique M, Ahamad S, Alajmi MF, Hussain A, Rehman MT, Luqman S, Hassan MI. Hesperidin-CAMKIV interaction and its impact on cell proliferation and apoptosis in the human hepatic carcinoma and neuroblastoma cells. J Cell Biochem 2019; 120:15119-15130. [PMID: 31021496 DOI: 10.1002/jcb.28774] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/08/2019] [Accepted: 01/14/2019] [Indexed: 01/11/2023]
Abstract
Calcium/calmodulin-dependent protein kinase IV (CAMKIV) is a key regulatory molecule of cell signaling, and thereby controls its growth and proliferation, including expression of certain genes. The overexpression of CAMKIV is directly associated with the development of different types of cancers. Hesperidin is abundantly found in citrus fruits and exhibits wide range of pharmacological activities including anti-inflammatory, antibacterial and anticancerous effects. We have investigated binding mechanism of hesperidin with the CAMKIV using molecular docking methods followed by fluorescence quenching and isothermal titration calorimetric assays. An appreciable binding affinity of hesperidin was observed with CAMKIV during fluorescence quenching and isothermal titration calorimetric studies. Efficacy of hesperidin to inhibit the growth of human hepatic carcinoma (HepG2) and neuroblastoma (SH-SY5Y) cancer cell lines were investigated. Hesperidin has significantly reduced the proliferation of HepG2 and SH-SY5Y cells and induces apoptosis by activating the caspase-3-dependent intrinsic pathway through the upregulation of proapoptotic Bax protein. Hesperidin treatment reduces the mitochondrial membrane potential of HepG2 and SH-SY5Y cells. All these observations clearly anticipated hesperidin a potent inhibitor of CAMKIV which may be further exploited a newer therapeutic approach for the management of different cancer types.
Collapse
Affiliation(s)
- Huma Naz
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Mohd Tarique
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Shahzaib Ahamad
- Department of Biotechnology, College of Engineering & Technology, IFTM University, Delhi Road, Moradabad, India
| | - Mohamed F Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Md Tabish Rehman
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Suaib Luqman
- Molecular Bioprospection Department of Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
24
|
Zuo L, Zhu Y, Hu L, Liu Y, Wang Y, Hu Y, Wang H, Pan X, Li K, Du N, Huang Y. PI3-kinase/Akt pathway-regulated membrane transportation of acid-sensing ion channel 1a/Calcium ion influx/endoplasmic reticulum stress activation on PDGF-induced HSC Activation. J Cell Mol Med 2019; 23:3940-3950. [PMID: 30938088 PMCID: PMC6533492 DOI: 10.1111/jcmm.14275] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 01/14/2019] [Accepted: 01/27/2019] [Indexed: 12/15/2022] Open
Abstract
Acid-sensing ion channel 1a (ASIC1a) allows Na+ and Ca2+ flow into cells. It is expressed during inflammation, in tumour and ischaemic tissue, in the central nervous system and non-neuronal injury environments. Endoplasmic reticulum stress (ERS) is caused by the accumulation of misfolded proteins that interferes with intracellular calcium homoeostasis. Our recent reports showed ASIC1a and ERS are involved in liver fibrosis progression, particularly in hepatic stellate cell (HSC) activation. In this study, we investigated the roles of ASIC1a and ERS in activated HSC. We found that ASIC1a and ERS-related proteins were up-regulated in carbon tetrachloride (CCl4 )-induced fibrotic mouse liver tissues, and in patient liver tissues with hepatocellular carcinoma with severe liver fibrosis. The results show silencing ASIC1a reduced the expression of ERS-related biomarkers GRP78, Caspase12 and IREI-XBP1. And, ERS inhibition by 4-PBA down-regulated the high expression of ASIC1a induced by PDGF, suggesting an interactive relationship. In PDGF-induced HSCs, ASIC1a was activated and migrated to the cell membrane, leading to extracellular calcium influx and ERS, which was mediated by PI3K/AKT pathway. Our work shows PDGF-activated ASIC1a via the PI3K/AKT pathway, induced ERS and promoted liver fibrosis progression.
Collapse
Affiliation(s)
- Longquan Zuo
- Department of Pharmacy, Hospital of Armed Police of Anhui Province, Hefei, China
| | - Yueqin Zhu
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases, Anhui Medical University, Hefei, China
| | - Lili Hu
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases, Anhui Medical University, Hefei, China
| | - Yanyi Liu
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases, Anhui Medical University, Hefei, China
| | - Yinghong Wang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yamin Hu
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases, Anhui Medical University, Hefei, China
| | - Huan Wang
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases, Anhui Medical University, Hefei, China
| | - Xuesheng Pan
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases, Anhui Medical University, Hefei, China
| | - Kuayue Li
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases, Anhui Medical University, Hefei, China
| | - Na Du
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases, Anhui Medical University, Hefei, China
| | - Yan Huang
- School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases, Anhui Medical University, Hefei, China
| |
Collapse
|
25
|
Salin Raj P, Swapna SUS, Raghu KG. High glucose induced calcium overload via impairment of SERCA/PLN pathway and mitochondrial dysfunction leads to oxidative stress in H9c2 cells and amelioration with ferulic acid. Fundam Clin Pharmacol 2019; 33:412-425. [PMID: 30739350 DOI: 10.1111/fcp.12452] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 01/15/2019] [Accepted: 01/31/2019] [Indexed: 01/05/2023]
Abstract
Oxidative stress and associated complications are the major pathological concerns of diabetic cardiomyopathy (DC). We aim to elucidate the mechanisms by which high glucose (HG) induced alteration in calcium homeostasis and evaluation of the beneficial effect of two concentrations (10 and 25 μm) of ferulic acid (FA). HG was induced in H9c2 cardiomyoblast by treating with glucose (33 mm) for 48 h, and FA was co-treated. Intracellular calcium ([Ca2+ ]i) overload was found increased significantly with HG. For elucidation of mechanism, the SERCA pathway and mitochondrial integrity (transmembrane potential and permeability transition pore) were explored. Then, we assessed oxidative stress, and cell injury with brain natriuretic peptide (BNP), atrial natriuretic peptide (ANP), and lactate dehydrogenase (LDH) release. HG caused significant [Ca2+ ]i overload through downregulation of SERCA2/1, pPLN, and pPKA C-α; and upregulation of PLN and PKA C-α and alteration in the integrity of mitochondria with HG. The [Ca2+ ]i overload in turn caused oxidative stress via generation of reactive oxygen species, lipid peroxidation, and protein carbonylation. This resulted in cell injury which was evident with significant release of BNP, ANP, and LDH. FA co-treatment was effective to mitigate all pathological changes caused by HG. From the overall results, we conclude that [Ca2+ ]i overload via SERCA pathway and altered mitochondrial integrity is the main cause for oxidative stress during HG. Based on our result, we report that FA could be an attractive nutraceutical for DC.
Collapse
Affiliation(s)
- Palayyan Salin Raj
- Biochemistry and Molecular Mechanism Laboratory, Agro-Processing and Technology Division, CSIR - National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, 695019, Kerala, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus Ghaziabad, Uttar Pradesh, 201 002, India
| | - Sasi U S Swapna
- Biochemistry and Molecular Mechanism Laboratory, Agro-Processing and Technology Division, CSIR - National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, 695019, Kerala, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus Ghaziabad, Uttar Pradesh, 201 002, India
| | - Kozhiparambil G Raghu
- Biochemistry and Molecular Mechanism Laboratory, Agro-Processing and Technology Division, CSIR - National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, 695019, Kerala, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus Ghaziabad, Uttar Pradesh, 201 002, India
| |
Collapse
|
26
|
Tang YW, Shi CJ, Yang HL, Cai P, Liu QH, Yang XL, Kong LY, Wang XB. Synthesis and evaluation of isoprenylation-resveratrol dimer derivatives against Alzheimer's disease. Eur J Med Chem 2019; 163:307-319. [PMID: 30529634 DOI: 10.1016/j.ejmech.2018.11.040] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/16/2018] [Accepted: 11/16/2018] [Indexed: 01/09/2023]
Abstract
A series of resveratrol dimer derivatives against Alzheimer's disease (AD) was obtained by structural modification and transformation using resveratrol as substrate. Biological analysis revealed that these derivatives had moderate inhibitory activity against human monoamine oxidase B (hMAO-B). In particular, 3 and 7 showed the better inhibitory activity for hMAO-B (IC50 = 3.91 ± 0.23 μM, 0.90 ± 0.01 μM) respectively. Compound 3 (IC50 = 46.95 ± 0.21 μM for DPPH, 1.43 and 1.74 trolox equivalent by ABTS and FRAP method respectively), and 7 (IC50 = 35.33 ± 0.15 μM for DPPH, 1.70 and 1.97 trolox equivalent by ABTS method and FRAP method respectively) have excellent antioxidant effects. Cellular assay shown that 3 and 7 had lower toxicity and were resistant to neurotoxicity induced by oxidative toxins (H2O2, rotenone and oligomycin-A). More importantly, the selected compounds have neuroprotective effects against ROS generation, H2O2-induced apoptosis and a significant in vitro anti-inflammatory activity. The results of the parallel artificial membrane permeability assay for blood-brain barrier indicated that 3 and 7 would be predominant to cross the blood-brain barrier. In this study, mouse microglia BV2 cells were used to establish cell oxidative stress injury model with H2O2 and to explore the protective effect and mechanism of 3 and 7. In general, 3 and 7 can be considered candidates for potential treatment of AD.
Collapse
Affiliation(s)
- Yan-Wei Tang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Cun-Jian Shi
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Hua-Li Yang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Pei Cai
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qiao-Hong Liu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xue-Lian Yang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Ling-Yi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Xiao-Bing Wang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
27
|
In Vivo Monitoring of Calpain Activity by Forster Resonance Energy Transfer. Methods Mol Biol 2019. [PMID: 30617795 DOI: 10.1007/978-1-4939-8988-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Calpains are a 15-member class of calcium-activated nonlysosomal neutral proteases. They are involved in many cellular processes and are highly upregulated in pathological conditions. Some are ubiquitously expressed (CAPN1, CAPN2, CAPN4, CAPN5, CAPN7, and CAPN10), but others are thought to be localized in specific tissues. The monitoring of in vivo calpain activity is required for physiological, pathological, and therapeutic evaluations. This past decade, a tool for monitoring calpain activity in such conditions was developed using Forster resonance energy transfer (FRET). Studies showed that the level of calpain activity correlates with a decrease in FRET between the two fluorescent proteins. This chapter describes the methodologies from the design of the construct to the imaging procedure and analysis to evaluate ubiquitous calpain activity in vivo.
Collapse
|
28
|
Ahmad S, Masjoan Juncos JX, Ahmad A, Zaky A, Wei CC, Bradley WE, Zafar I, Powell P, Mariappan N, Vetal N, Louch WE, Ford DA, Doran SF, Matalon S, Dell'Italia LJ. Bromine inhalation mimics ischemia-reperfusion cardiomyocyte injury and calpain activation in rats. Am J Physiol Heart Circ Physiol 2018; 316:H212-H223. [PMID: 30379573 DOI: 10.1152/ajpheart.00652.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Halogens are widely used, highly toxic chemicals that pose a potential threat to humans because of their abundance. Halogens such as bromine (Br2) cause severe pulmonary and systemic injuries; however, the mechanisms of their toxicity are largely unknown. Here, we demonstrated that Br2 and reactive brominated species produced in the lung and released in blood reach the heart and cause acute cardiac ultrastructural damage and dysfunction in rats. Br2-induced cardiac damage was demonstrated by acute (3-24 h) increases in circulating troponin I, heart-type fatty acid-binding protein, and NH2-terminal pro-brain natriuretic peptide. Transmission electron microscopy demonstrated acute (3-24 h) cardiac contraction band necrosis, disruption of z-disks, and mitochondrial swelling and disorganization. Echocardiography and hemodynamic analysis revealed left ventricular (LV) systolic and diastolic dysfunction at 7 days. Plasma and LV tissue had increased levels of brominated fatty acids. 2-Bromohexadecanal (Br-HDA) injected into the LV cavity of a normal rat caused acute LV enlargement with extensive disruption of the sarcomeric architecture and mitochondrial damage. There was extensive infiltration of neutrophils and increased myeloperoxidase levels in the hearts of Br2- or Br2 reactant-exposed rats. Increased bromination of sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) and increased phosphalamban after Br2 inhalation decreased cardiac SERCA activity by 70%. SERCA inactivation was accompanied by increased Ca2+-sensitive LV calpain activity. The calpain-specific inhibitor MDL28170 administered within 1 h after exposure significantly decreased calpain activity and acute mortality. Bromine inhalation and formation of reactive brominated species caused acute cardiac injury and myocardial damage that can lead to heart failure. NEW & NOTEWORTHY The present study defines left ventricular systolic and diastolic dysfunction due to cardiac injury after bromine (Br2) inhalation. A calpain-dependent mechanism was identified as a potential mediator of cardiac ultrastructure damage. This study not only highlights the importance of monitoring acute cardiac symptoms in victims of Br2 exposure but also defines calpains as a potential target to treat Br2-induced toxicity.
Collapse
Affiliation(s)
- Shama Ahmad
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Juan Xavier Masjoan Juncos
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Aftab Ahmad
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Ahmed Zaky
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Chih-Chang Wei
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama.,Department of Veterans Affairs Medical Center , Birmingham, Alabama
| | - Wayne E Bradley
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama.,Department of Veterans Affairs Medical Center , Birmingham, Alabama
| | - Iram Zafar
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Pamela Powell
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama.,Department of Veterans Affairs Medical Center , Birmingham, Alabama
| | - Nithya Mariappan
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Nilam Vetal
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo , Oslo , Norway.,KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - David A Ford
- Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University , St. Louis, Missouri
| | - Stephen F Doran
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Sadis Matalon
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Louis J Dell'Italia
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama.,Department of Veterans Affairs Medical Center , Birmingham, Alabama
| |
Collapse
|
29
|
Ng KM, Lau YM, Dhandhania V, Cai ZJ, Lee YK, Lai WH, Tse HF, Siu CW. Empagliflozin Ammeliorates High Glucose Induced-Cardiac Dysfuntion in Human iPSC-Derived Cardiomyocytes. Sci Rep 2018; 8:14872. [PMID: 30291295 PMCID: PMC6173708 DOI: 10.1038/s41598-018-33293-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 09/03/2018] [Indexed: 12/12/2022] Open
Abstract
Empagliflozin, a sodium-glucose co-transporter (SGLT) inhibitor, reduces heart failure and sudden cardiac death but the underlying mechanisms remain elusive. In cardiomyocytes, SGLT1 and SGLT2 expression is upregulated in diabetes mellitus, heart failure, and myocardial infarction. We hypothesise that empagliflozin exerts direct effects on cardiomyocytes that attenuate diabetic cardiomyopathy. To test this hypothesis, cardiomyocytes derived from human induced pluripotent stem cells (hiPSCs) were used to test the potential effects of empagliflozin on neutralization of cardiac dysfunction induced by diabetic-like cultures. Our results indicated that insulin-free high glucose culture significantly increased the size of and NPPB, SGLT1 and SGLT2 expression of hiPSC-derived cardiomyocytes. In addition, high glucose-treated hiPSC-derived cardiomyocytes exhibited reduced contractility regardless of the increased calcium transient capacity. Interestingly, application of empagliflozin before or after high glucose treatment effectively reduced the high glucose-induced cardiac abnormalities. Since application of empagliflozin did not significantly alter viability or glycolytic capacity of the hiPSC-derived cardiomyocytes, it is plausible that empagliflozin exerts its effects via the down-regulation of SGLT1, SGLT2 and GLUT1 expression. These observations provide supportive evidence that may help explain its unexpected benefit observed in the EMPA-REG trial.
Collapse
Affiliation(s)
- Kwong-Man Ng
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, Hong Kong, China
| | - Yee-Man Lau
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, Hong Kong, China
| | - Vidhu Dhandhania
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, Hong Kong, China
| | - Zhu-Jun Cai
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, Hong Kong, China
| | - Yee-Ki Lee
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, Hong Kong, China
| | - Wing-Hon Lai
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, Hong Kong, China
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, Hong Kong, China
| | - Chung-Wah Siu
- Cardiology Division, Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, Hong Kong, China.
| |
Collapse
|
30
|
Edhager AV, Povlsen JA, Løfgren B, Bøtker HE, Palmfeldt J. Proteomics of the Rat Myocardium during Development of Type 2 Diabetes Mellitus Reveals Progressive Alterations in Major Metabolic Pathways. J Proteome Res 2018; 17:2521-2532. [PMID: 29847139 DOI: 10.1021/acs.jproteome.8b00276] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Congestive heart failure and poor clinical outcome after myocardial infarction are known complications in patients with type-2 diabetes mellitus (T2DM). Protein alterations may be involved in the mechanisms underlying these disarrays in the diabetic heart. Here we map proteins involved in intracellular metabolic pathways in the Zucker diabetic fatty rat heart as T2DM develops using MS based proteomics. The prediabetic state only induced minor pathway changes, whereas onset and late T2DM caused pronounced perturbations. Two actin-associated proteins, ARPC2 and TPM3, were up-regulated at the prediabetic state indicating increased actin dynamics. All differentially regulated proteins involved in fatty acid metabolism, both peroxisomal and mitochondrial, were up-regulated at late T2DM, whereas enzymes of branched chain amino acid degradation were all down-regulated. At both onset and late T2DM, two members of the serine protease inhibitor superfamily, SERPINA3K and SERPINA3L, were down-regulated. Furthermore, we found alterations in proteins involved in clearance of advanced glycation end-products and lipotoxicity, DCXR and CBR1, at both onset and late T2DM. These proteins deserve elucidation with regard to their role in T2DM pathogenesis and their respective role in the deterioration of the diabetic heart. Data are available via ProteomeXchange with identifiers PXD009538, PXD009554, and PXD009555.
Collapse
Affiliation(s)
- Anders Valdemar Edhager
- Research Unit for Molecular Medicine, Department of Clinical Medicine , Aarhus University and Aarhus University Hospital , 8200 , Aarhus N , Denmark
| | | | - Bo Løfgren
- Department of Cardiology , Aarhus University Hospital , 8200 , Aarhus N , Denmark.,Institute for Experimental Clinical Research , Aarhus University , 8000 , Aarhus C , Denmark
| | - Hans Erik Bøtker
- Department of Cardiology , Aarhus University Hospital , 8200 , Aarhus N , Denmark
| | - Johan Palmfeldt
- Research Unit for Molecular Medicine, Department of Clinical Medicine , Aarhus University and Aarhus University Hospital , 8200 , Aarhus N , Denmark
| |
Collapse
|
31
|
Guo Z, Liu N, Chen L, Zhao X, Li MR. Independent roles of CGRP in cardioprotection and hemodynamic regulation in ischemic postconditioning. Eur J Pharmacol 2018; 828:18-25. [DOI: 10.1016/j.ejphar.2018.03.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 03/19/2018] [Accepted: 03/19/2018] [Indexed: 11/26/2022]
|
32
|
Panahi G, Pasalar P, Zare M, Rizzuto R, Meshkani R. MCU-knockdown attenuates high glucose-induced inflammation through regulating MAPKs/NF-κB pathways and ROS production in HepG2 cells. PLoS One 2018; 13:e0196580. [PMID: 29709004 PMCID: PMC5927441 DOI: 10.1371/journal.pone.0196580] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 04/16/2018] [Indexed: 01/25/2023] Open
Abstract
Mitochondrial Ca2+ is a key regulator of organelle physiology and the excessive increase in mitochondrial calcium is associated with the oxidative stress. In the present study, we investigated the molecular mechanisms linking mitochondrial calcium to inflammatory and coagulative responses in hepatocytes exposed to high glucose (HG) (33mM glucose). Treatment of HepG2 cells with HG for 24 h induced insulin resistance, as demonstrated by an impairment of insulin-stimulated Akt phosphorylation. HepG2 treatment with HG led to an increase in mitochondrial Ca2+ uptake, while cytosolic calcium remained unchanged. Inhibition of MCU by lentiviral-mediated shRNA prevented mitochondrial calcium uptake and downregulated the inflammatory (TNF-α, IL-6) and coagulative (PAI-1 and FGA) mRNA expression in HepG2 cells exposed to HG. The protection from HG-induced inflammation by MCU inhibition was accompanied by a decrease in the generation of reactive oxygen species (ROS). Importantly, MCU inhibition in HepG2 cells abrogated the phosphorylation of p38, JNK and IKKα/IKKβ in HG treated cells. Taken together, these data suggest that MCU inhibition may represent a promising therapy for prevention of deleterious effects of obesity and metabolic diseases.
Collapse
Affiliation(s)
- Ghodratollah Panahi
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, I.R Iran
| | - Parvin Pasalar
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, I.R Iran
| | - Mina Zare
- Recombinant Protein Laboratory, Department of Biochemistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Reza Meshkani
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, I.R Iran
- * E-mail:
| |
Collapse
|
33
|
Lee HY, Ryu GH, Choi WY, Yang WS, Lee HW, Ma CJ. Protective Effect of Water Extracted Spirulina maxima on Glutamate-induced Neuronal Cell Death in Mouse Hippocampal HT22 Cell. Pharmacogn Mag 2018; 14:242-247. [PMID: 29720839 PMCID: PMC5909323 DOI: 10.4103/pm.pm_191_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/05/2017] [Indexed: 12/20/2022] Open
Abstract
Introduction: Spirulina maxima was used as important nutritional source in the Aztec civilization because it is rich in proteins and vitamins. It contains various antioxidants such as phycocyanin and flavonoids. Based on abundant antioxidants, S. maxima is known to possess anti-inflammatory effect, especially on neuronal cells. Materials and Methods: S. maxima was extracted in water and contain of phycocyanin was identified by high-performance liquid chromatography. Cell viability test was performed with treatment of S. maxima extract. After, oxidative stress-related mechanisms were evaluated by detecting the accumulation of reactive oxygen species (ROS) and Ca2+ influx, and decrease of mitochondrial membrane potential (MMP) level. Then, the glutathione (GSH) related assays were conducted. Results: The water extracted S. maxima exerted the neuroprotective activity by attenuating the ROS and Ca2+ formation, maintaining the MMP level, and protecting the activity of the antioxidant enzymes by increasing reduced GSH against oxidative stress compared to control. Conclusion: The results suggested that water extracted S. maxima showed powerful neuroprotective effect through the mechanism related to antioxidant activity, able to preventing the radical-mediated cell death. SUMMARY Water extracted Spirulina maxima contains C-phycocyanin Water extracted Spirulina maxima exerts neuroprotective effect on HT22 cell To investigate the protective mechanisms, reactive oxygen species, Ca2+, mitochondrial membrane potential, Glutathione-related assays were performed.
Abbreviations used: ROS: Reactive oxygen species; MMP: Mitochondrial membrane potential; GSH: Glutathione; GSSG: Glutathione disulfide, oxidized glutathione; GPx: Glutathione peroxidase; GR: Glutathione reductase; DMEM: Dulbecco's modified Eagle's medium; FBS: Fetal bovine serum; DCF-DA: 2',7'-dichlorofluorescein diacetate; PBS: Phosphate buffered serum; Rho 123: Rhodamine 123; NADPH: Nicotinamide adenine dinucleotide phosphate; DTNB: 5,5'-dithiobis-2-nitrobenzoic acid, Ellman's reagent; GSSG-R: Glutathione disulfide reductase; MTT: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; DMSO: Dimethyl sulfoxide; HPLC: High-performance liquid chromatography.
Collapse
Affiliation(s)
- Hyeon Yong Lee
- Department of Teaics, Seowon University, Musimseo-ro, Seowon-gu, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Ga Hee Ryu
- Department of Medical Biomaterials Engineering, College of Biomedical Science, Kangwon National University, Gangwondaehak-gil, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Woon Yong Choi
- Department of Medical Biomaterials Engineering, College of Biomedical Science, Kangwon National University, Gangwondaehak-gil, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Woo Seung Yang
- Department of Medical Biomaterials Engineering, College of Biomedical Science, Kangwon National University, Gangwondaehak-gil, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Hyeon Woo Lee
- Department of Medical Biomaterials Engineering, College of Biomedical Science, Kangwon National University, Gangwondaehak-gil, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Choong Je Ma
- Department of Medical Biomaterials Engineering, College of Biomedical Science, Kangwon National University, Gangwondaehak-gil, Chuncheon-si, Gangwon-do, Republic of Korea.,Institute of Bioscience and Biotechnology, Kangwon National University, Gangwondaehak-gil, Chuncheon-si, Gangwon-do, Republic of Korea
| |
Collapse
|
34
|
Jadaun P, Yadav D, Bisen PS. Spirulina platensis prevents high glucose-induced oxidative stress mitochondrial damage mediated apoptosis in cardiomyoblasts. Cytotechnology 2018; 70:523-536. [PMID: 28702859 PMCID: PMC5851949 DOI: 10.1007/s10616-017-0121-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 07/05/2017] [Indexed: 12/26/2022] Open
Abstract
The current study was undertaken to study the effect of Spirulina platensis (Spirulina) extract on enhanced oxidative stress during high glucose induced cell death in H9c2 cells. H9c2 cultured under high glucose (33 mM) conditions resulted in a noteworthy increase in oxidative stress (free radical species) accompanied by loss of mitochondrial membrane potential, release of cytochrome c, increase in caspase activity and pro-apoptotic protein (Bax). Spirulina extract (1 μg/mL), considerably inhibited increased ROS and RNS levels, reduction in cytochrome c release, raise in mitochondrial membrane potential, decreased the over expression of proapoptotic protein Bax and suppressed the Bax/Bcl2 ratio with induced apoptosis without affecting cell viability. Overall results suggest that Spirulina extract plays preventing role against enhanced oxidative stress during high glucose induced apoptosis in cardiomyoblasts as well as related dysfunction in H9c2 cells.
Collapse
Affiliation(s)
- Pratiksha Jadaun
- School of Studies in Biotechnology, Jiwaji University, Gwalior, 474011 MP India
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Kangwon-Do 712-749 Korea
| | - Prakash Singh Bisen
- School of Studies in Biotechnology, Jiwaji University, Gwalior, 474011 India
| |
Collapse
|
35
|
Liu T, Chen XM, Sun JY, Jiang XS, Wu Y, Yang S, Huang HZ, Ruan XZ, Du XG. Palmitic Acid-Induced Podocyte Apoptosis via the Reactive Oxygen Species-Dependent Mitochondrial Pathway. Kidney Blood Press Res 2018; 43:206-219. [PMID: 29490300 DOI: 10.1159/000487673] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 02/15/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Chronic kidney disease (CKD) is often accompanied by hyperlipidemia, which accelerates progression of the disease. Podocyte injury can lead to dysfunction of the glomerular filtration barrier, which is associated with proteinuria, a risk marker for the progression of CKD. Our previous studies demonstrated that palmitic acid (PA) can induce podocyte apoptosis; however, the underlying mechanisms are unclear. In the present study, we investigated the specific molecular mechanisms of PA-induced apoptosis in cultured podocytes. METHODS We cultured mouse podocytes and treated them with PA. Then, cell viability was measured using the Cell Counting Kit-8 colorimetric assay, lipid uptake was assessed by Oil Red O staining and boron-dipyrromethene staining, apoptosis was measured by flow cytometry, mitochondrial injury was assessed by JC-1 staining and transmission electron microscopy, and mitochondrial production of reactive oxygen species (ROS) was evaluated by fluorescence microscopy using the MitoSOX Red reagent. The effects of PA on the mitochondria-mediated caspase activation pathway were investigated by examining the expression of caspase-8, cleaved caspase-9, cleaved caspase-3, cleaved poly (ADP-ribose) polymerase (PARP), B-cell lymphoma 2 (Bcl-2), Bax, Bid, cytochrome c, and Fas-associated protein with death domain (FADD) using western blotting. The translocation of Bax and cytochrome c were detected by immunofluorescence. RESULTS PA treatment significantly increased lipid accumulation and induced podocyte apoptosis. We investigated whether the two primary apoptosis signaling pathways (death receptor-mediated pathway and mitochondria-mediated pathway) were involved in the execution of PA-induced podocyte apoptosis, and found that the levels of FADD, caspase-8, and Bid did not significantly change during this process. Meanwhile, PA treatment induced an increase in Bax protein expression and a decrease in Bcl-2 protein expression, with Bax translocation to the mitochondria. Furthermore, PA treatment induced mitochondrial impairment, and triggered the release of cytochrome c from the mitochondria to cytosol, with a concomitant dose-dependent increase in the levels of cleaved caspase-9, cleaved caspase-3, and PARP. Meanwhile, PA treatment increased mitochondrial production of ROS, and the mitochondria-targeted antioxidant mitoTEMPO significantly ameliorated PA-induced podocyte apoptosis. CONCLUSION Our findings indicated that PA induced caspase-dependent podocyte apoptosis through the mitochondrial pathway, and mitochondrial ROS production participated in this process, thus potentially contributing to podocyte injury.
Collapse
Affiliation(s)
- Ting Liu
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xue-Mei Chen
- Emergency Department, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ji-Ye Sun
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xu-Shun Jiang
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Wu
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shan Yang
- Emergency Department, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hui-Zhe Huang
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiong-Zhong Ruan
- Centre for Nephrology, Royal Free and University College Medical School, University College London, Royal Free Campus, London, United Kingdom.,Centre for Lipid Research, Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Xiao-Gang Du
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Disease, Chongqing, China
| |
Collapse
|
36
|
Chang G, Liu J, Qin S, Jiang Y, Zhang P, Yu H, Lu K, Zhang N, Cao L, Wang Y, Li Y, Zhang D. Cardioprotection by exenatide: A novel mechanism via improving mitochondrial function involving the GLP-1 receptor/cAMP/PKA pathway. Int J Mol Med 2017; 41:1693-1703. [PMID: 29286061 DOI: 10.3892/ijmm.2017.3318] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 11/24/2017] [Indexed: 11/06/2022] Open
Abstract
Accumulating evidence suggests that glucagon-like peptide-1 (GLP-1) and its analogues exert cardioprotective effects via modulating cardiomyocyte metabolism. Mitochondria play a pivotal role in the regulation of cell metabolism. It was hypothesized that treatment with exenatide, a GLP-1 analogue, may exert cardioprotective effects by improving mitochondrial function in an in vitro model of hypoxia/reoxygenation (H/R). H9c2 cells were employed to establish an in vitro model of H/R. Exenatide was added to the cells for 30 min prior to exposure to hypoxia. The GLP-1 receptor antagonist exendin‑(9‑39), the cyclic adenosine monophosphate (cAMP) inhibitor Rp-cAMPS and the protein kinase A (PKA) inhibitor H-89 were added to the cells for 10 min prior to treatment with exenatide. The release of lactate dehydrogenase (LDH) and creatine kinase-MB (CK-MB) and cardiomyocyte apoptosis were evaluated. The characteristics of mitochondrial morphology and functions, including ATP synthesis, membrane potential (ΔΨm), mitochondrial permeability transition pore (mPTP), mitochondrial ATPase activity and oxidative stress, were determined. the mitochondrial uncoupling protein-3 (UCP-3) and nuclear respiratory factor-1 (Nrf-1) were also investigated by western blot analysis. Exenatide pretreatment significantly decreased LDH and CK-MB release and cardiomyocyte apoptosis in H9c2 cells subjected to H/R. More importantly, to the best of our knowledge, this is the first report of exenatide pretreatment decreasing mitochondrial abnormalities and reducing oxidative stress, while enhancing ATP synthesis, mitochondrial ATPase activity and ΔΨm in H9c2 cells subjected to H/R. Exenatide pretreatment also decreased mitochondrial calcium overload and inhibited the opening of mPTP in H9c2 cells subjected to H/R. Furthermore, exenatide pretreatment upregulated UCP-3 and Nrf-1 expression in H9c2 cells subjected to H/R. However, the abovementioned observed effects of exenatide were all abolished when exenatide was co-administered with exendin‑(9‑39), Rp-cAMPS and̸or H-89. Therefore, the GLP-1 analogue exenatide was found to exert cardioprotective effects in an in vitro model of H/R, and this cardioprotection may be attributed to the improvement of mitochondrial function. These effects are most likely associated with the activation of the GLP-1 receptor/cAMP/PKA signaling pathway.
Collapse
Affiliation(s)
- Guanglei Chang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, Sichuan 400016, P.R. China
| | - Jian Liu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, Sichuan 400016, P.R. China
| | - Shu Qin
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, Sichuan 400016, P.R. China
| | - Youqin Jiang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, Sichuan 400016, P.R. China
| | - Peng Zhang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, Sichuan 400016, P.R. China
| | - Hui Yu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, Sichuan 400016, P.R. China
| | - Kai Lu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, Sichuan 400016, P.R. China
| | - Nan Zhang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, Sichuan 400016, P.R. China
| | - Li Cao
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, Sichuan 400016, P.R. China
| | - Ying Wang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, Sichuan 400016, P.R. China
| | - Yong Li
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Dongying Zhang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, Sichuan 400016, P.R. China
| |
Collapse
|
37
|
Qu Y, Liu Y, Zhu Y, Chen L, Sun W, Zhu Y. Epoxyeicosatrienoic Acid Inhibits the Apoptosis of Cerebral Microvascular Smooth Muscle Cells by Oxygen Glucose Deprivation via Targeting the JNK/c-Jun and mTOR Signaling Pathways. Mol Cells 2017; 40:837-846. [PMID: 29081082 PMCID: PMC5712513 DOI: 10.14348/molcells.2017.0084] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/25/2017] [Accepted: 10/10/2017] [Indexed: 02/07/2023] Open
Abstract
As a component of the neurovascular unit, cerebral smooth muscle cells (CSMCs) are an important mediator in the development of cerebral vascular diseases such as stroke. Epoxyeicosatrienoic acids (EETs) are the products of arachidonic acid catalyzed by cytochrome P450 epoxygenase. EETs are shown to exert neuroprotective effects. In this article, the role of EET in the growth and apoptosis of CSMCs and the underlying mechanisms under oxygen glucose deprivation (OGD) conditions were addressed. The viability of CMSCs was decreased significantly in the OGD group, while different subtypes of EETs, especially 14,15-EET, could increase the viability of CSMCs under OGD conditions. RAPA (serine/threonine kinase Mammalian Target of Rapamycin), a specific mTOR inhibitor, could elevate the level of oxygen free radicals in CSMCs as well as the anti-apoptotic effects of 14,15-EET under OGD conditions. However, SP600125, a specific JNK (c-Jun N-terminal protein kinase) pathway inhibitor, could attenuate oxygen free radicals levels in CSMCs as well as the anti-apoptotic effects of 14,15-EET under OGD conditions. These results strongly suggest that EETs exert protective functions during the growth and apoptosis of CSMCs, via the JNK/c-Jun and mTOR signaling pathways in vitro. We are the first to disclose the beneficial roles and underlying mechanism of 14,15-EET in CSMC under OGD conditions.
Collapse
Affiliation(s)
- Youyang Qu
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, P. R.
China
| | - Yu Liu
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, P. R.
China
| | - Yanmei Zhu
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, P. R.
China
| | - Li Chen
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, P. R.
China
| | - Wei Sun
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, P. R.
China
| | - Yulan Zhu
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, P. R.
China
| |
Collapse
|
38
|
Guo Y, Zhuang X, Huang Z, Zou J, Yang D, Hu X, Du Z, Wang L, Liao X. Klotho protects the heart from hyperglycemia-induced injury by inactivating ROS and NF-κB-mediated inflammation both in vitro and in vivo. Biochim Biophys Acta Mol Basis Dis 2017; 1864:238-251. [PMID: 28982613 DOI: 10.1016/j.bbadis.2017.09.029] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 09/10/2017] [Accepted: 09/28/2017] [Indexed: 12/23/2022]
Abstract
Cardiac inflammation and oxidative stress play a key role in the pathogenesis of diabetic cardiomyopathy (DCM). The anti-aging protein Klotho has been found to protect cells from inflammation and oxidative stress. The current study aimed to explore the cardioprotective effects of Klotho on DCM and the underlying mechanisms. H9c2 cells and neonatal cardiomyocytes were incubated with 33mM glucose in the presence or absence of Klotho. Klotho pretreatment effectively inhibited high glucose-induced inflammation, ROS generation, apoptosis, mitochondrial dysfunction, fibrosis and hypertrophy in both H9c2 cells and neonatal cardiomyocytes. In STZ-induced type 1 diabetic mice, intraperitoneal injection of Klotho at 0.01mg/kg per 48h for 3months completely suppressed cardiac inflammatory cytokines and oxidative stress and prevented cardiac cell death and remodeling, which subsequently improved cardiac dysfunction without affecting hyperglycemia. This study revealed that Klotho may exert its protective effects by augmenting nuclear factor erythroid 2-related factor 2 (Nrf2) expression and inactivating nuclear factor κB (NF-κB) activation both in vitro and in vivo. Thus, this work demonstrated for the first time that the anti-aging protein Klotho may be a potential therapeutic agent to treat DCM by inhibiting oxidative stress and inflammation. We also demonstrated the critical roles of the Nrf2 and NF-κB pathways in diabetes-stimulated cardiac injuries and indicated that they may be key therapeutic targets for diabetic complications.
Collapse
Affiliation(s)
- Yue Guo
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China; Key Laboratory of Assisted Circulation, Ministry of Health, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, PR China
| | - Xiaodong Zhuang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China
| | - Zena Huang
- Department of Critical Care Medicine and Emergency, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
| | - Jing Zou
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, PR China
| | - Daya Yang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China
| | - Xun Hu
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China
| | - Zhimin Du
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China
| | - Lichun Wang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China; Key Laboratory of Assisted Circulation, Ministry of Health, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, PR China.
| | - Xinxue Liao
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China; Key Laboratory of Assisted Circulation, Ministry of Health, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, PR China.
| |
Collapse
|
39
|
Naz H, Tarique M, Khan P, Luqman S, Ahamad S, Islam A, Ahmad F, Hassan MI. Evidence of vanillin binding to CAMKIV explains the anti-cancer mechanism in human hepatic carcinoma and neuroblastoma cells. Mol Cell Biochem 2017; 438:35-45. [PMID: 28744811 DOI: 10.1007/s11010-017-3111-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/01/2017] [Indexed: 12/21/2022]
Abstract
Human calcium/calmodulin-dependent protein kinase IV (CAMKIV) is a member of Ser/Thr kinase family, and is associated with different types of cancer and neurodegenerative diseases. Vanillin is a natural compound, a primary component of the extract of the vanilla bean which possesses varieties of pharmacological features including anti-oxidant, anti-inflammatory, anti-bacterial and anti-tumor. Here, we have investigated the binding mechanism and affinity of vanillin to the CAMKIV which is being considered as a potential drug target for cancer and neurodegenerative diseases. We found that vanillin binds strongly to the active site cavity of CAMKIV and stabilized by a large number of non-covalent interactions. We explored the utility of vanillin as anti-cancer agent and found that it inhibits the proliferation of human hepatocyte carcinoma (HepG2) and neuroblastoma (SH-SY5Y) cells in a dose-dependent manner. Furthermore, vanillin treatment resulted into the significant reduction in the mitochondrial membrane depolarization and ROS production that eventually leads to apoptosis in HepG2 and SH-SY5Y cancer cells. These findings may offer a novel therapeutic approach by targeting the CAMKIV using natural product and its derivative with a minimal side effect.
Collapse
Affiliation(s)
- Huma Naz
- Centre for Interdisciplinary Research in Basic Sciences,Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Mohd Tarique
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Parvez Khan
- Centre for Interdisciplinary Research in Basic Sciences,Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Suaib Luqman
- CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, 226015, India
| | - Shahzaib Ahamad
- Department of Biotechnology, College of Engineering & Technology, IFTM University, Lodhipur-Rajput, Delhi Road, Moradabad, India
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences,Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Faizan Ahmad
- Centre for Interdisciplinary Research in Basic Sciences,Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences,Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India.
| |
Collapse
|
40
|
Wang H, Zhai N, Chen Y, Xu H, Huang K. Cadmium induces Ca 2+ mediated, calpain-1/caspase-3-dependent apoptosis in primary cultured rat proximal tubular cells. J Inorg Biochem 2017; 172:16-22. [DOI: 10.1016/j.jinorgbio.2017.04.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 04/06/2017] [Accepted: 04/08/2017] [Indexed: 01/15/2023]
|
41
|
Huang X, Liao W, Huang Y, Jiang M, Chen J, Wang M, Lin H, Guan S, Liu J. Neuroprotective effect of dual specificity phosphatase 6 against glutamate-induced cytotoxicity in mouse hippocampal neurons. Biomed Pharmacother 2017; 91:385-392. [PMID: 28475917 DOI: 10.1016/j.biopha.2017.04.096] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 04/01/2017] [Accepted: 04/20/2017] [Indexed: 12/31/2022] Open
Abstract
Dual specificity phosphatase 6 (DUSP6), a member of the dual specificity protein phosphatase subfamily, can inactivate ERK1/2. However, its possible role in glutamate-induced oxidative cytotoxicity effects is not clear.Here, we aimed to investigate whether DUSP6 was neuroprotective against glutamate-induced cytotoxicity in HT22 mouse hippocampal cells and primary cultured hippocampal neurons (pc-HNeu). HT22 and pc-HNeu cells were treated with varying concentrations of glutamate (from 0.05mM to 5.0mM) and DUSP6 protein expression were detected by western blotting. DUSP6-overexpressing HT22 and pc-HNeu cells were generated by transfection with DUSP6-overexpressing plasmid. The effects of DUSP6 overexpression on glutamate-induced cytotoxicity, cell death, cell apoptosis, and cell autophagy were determined by cell proliferation assays, flow cytometry, transmission electron microscopy, and western blotting. Glutamate treatment from 0.5mM to 5.0mM downregulated DUSP6 protein expression in both HT22 and pc-HNeu cells. DUSP6 overexpression ameliorated glutamate-induced cell death, apoptosis, and autophagy in both HT22 and pc-HNeu cells. Furthermore, ERK1/2 phosphorylation was decreased by DUSP6 overexpression. In conclusion, DUSP6 has neuroprotective effects against glutamate-induced cytotoxicity in HT22 and pc-HNeu cells. Targeting DUSP6 may be a useful strategy to prevent neuronal death in neurodegenerative diseases including AD.
Collapse
Affiliation(s)
- Xiaoyun Huang
- Department of Neurology, The Affiliated Houjie Hospital, Guangdong Medical University, 21 Hetian Road, Dongguan, 523945, PR China
| | - Wang Liao
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou, 510120, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou 510080, PR China; Laboratory of RNA and Major Diseases of Brain and Heart, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou, 510120, PR China
| | - Yihong Huang
- Department of Neurology, The Affiliated Houjie Hospital, Guangdong Medical University, 21 Hetian Road, Dongguan, 523945, PR China
| | - Mujun Jiang
- Department of Neurology, The First Affiliated Hospital, Bengbu Medical College, 287 Changhuai Road, Bengbu, Anhui 233004, PR China
| | - Jianjun Chen
- Department of Neurology, The Affiliated Houjie Hospital, Guangdong Medical University, 21 Hetian Road, Dongguan, 523945, PR China
| | - Mingxia Wang
- Department of Neurology, The Affiliated Houjie Hospital, Guangdong Medical University, 21 Hetian Road, Dongguan, 523945, PR China
| | - Han Lin
- Department of Neurology, The Affiliated Houjie Hospital, Guangdong Medical University, 21 Hetian Road, Dongguan, 523945, PR China
| | - Shaobing Guan
- Department of Neurology, The Affiliated Houjie Hospital, Guangdong Medical University, 21 Hetian Road, Dongguan, 523945, PR China
| | - Jun Liu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou, 510120, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou 510080, PR China; Laboratory of RNA and Major Diseases of Brain and Heart, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou, 510120, PR China.
| |
Collapse
|
42
|
Chen M, Zhou B, Zhong P, Rajamanickam V, Dai X, Karvannan K, Zhou H, Zhang X, Liang G. Increased Intracellular Reactive Oxygen Species Mediates the Anti-Cancer Effects of WZ35 via Activating Mitochondrial Apoptosis Pathway in Prostate Cancer Cells. Prostate 2017; 77:489-504. [PMID: 27990666 DOI: 10.1002/pros.23287] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/18/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND The limited treatment option for recurrent prostate cancer and eventual resistant to conventional chemotherapy drugs has fueled continued interest in finding new anti-neoplastic agents. WZ35, a chemical analog of curcumin, had been demonstrated to have high chemical stability and potential anticancer effects in gastric cancer cells. The present study aimed to investigate the anti-prostate cancer effects of WZ35 in vitro and in vivo as well as the underlying mechanism. METHODS Two prostate cancer cell lines RM-1 and DU145 were utilized to test the anti-cancer effects of WZ35 and the underlying mechanism. MTT assay was used to assess the cytotoxic effect of WZ35. Cell cycle distribution, apoptosis, alteration of ROS, and [Ca2+ ]i level were evaluated using flow cytometry. Western blotting assay was applied to measure the levels of proteins associated with apoptosis and cell cycle. Immunofluorescence staining and Electron micrographs were used to evaluate activation of mitochondrial apoptosis pathway. Tumor models in nude mice were induced by injection of RM-1 prostate cancer cells to test the in vivo anticancer action of WZ35. RESULTS Our results showed that WZ35 treatment induced loss of cell viability, cell apoptosis, and G2/M cycle arrest in both RM-1 and DU145 cells, coupled with ROS overproduction, intracellular calcium surge, and activation of mitochondrial apoptosis pathway in RM-1 cells. Interestingly, all above changes induced by WZ35 were completely reversed by ROS blockage. In addition, prevention of [Ca2+ ]i elevation by BAPTA/AM also inhibited activation of mitochondrial apoptosis pathway induced by WZ35. In vivo studies, WZ35 treatment significantly inhibited RM-1 homograft tumor growth along with increased ROS accumulation, mitochondrial disruption, and cell apoptosis in tumor tissues. CONCLUSIONS In conclusion, this work provides a novel anticancer candidate for the treatment of prostate cancer and demonstrated that increased ROS mediate the anti-cancer effects of WZ35 via activating mitochondrial apoptosis pathway. Importantly, this work also reveals that targeting ROS generation might be an effective strategy in human androgen-resistant prostate cancer treatment. Prostate 77:489-504, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Minxiao Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical Universtiy, Wenzhou, Zhejiang, China
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bin Zhou
- The Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Peng Zhong
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical Universtiy, Wenzhou, Zhejiang, China
| | - Vinothkumar Rajamanickam
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical Universtiy, Wenzhou, Zhejiang, China
| | - Xuanxuan Dai
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kanchana Karvannan
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical Universtiy, Wenzhou, Zhejiang, China
| | - Huiping Zhou
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical Universtiy, Wenzhou, Zhejiang, China
| | - Xiuhua Zhang
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical Universtiy, Wenzhou, Zhejiang, China
| |
Collapse
|
43
|
Chen HM, Luo H, Zeng WB, Liu B, Huang JC, Liu M, Zeng YJ, Zheng Q, Li JQ, Sun XG, Zhou YC. Salvianolic acid B attenuates oxidized low-density lipoprotein-induced endothelial cell apoptosis through inhibition of oxidative stress, p53, and caspase-3 pathways. Chin J Integr Med 2017:10.1007/s11655-016-2645-4. [PMID: 28116660 DOI: 10.1007/s11655-016-2645-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To investigate the effect of salvianolic acid B (Sal B) on oxidized low-density lipoprotein (ox-LDL)-induced human umbilical vein endothelial cells (HUVECs) apoptosis and the possible mechanism. METHODS HUVECs were divided into 6 groups, including control group, ox-LDL group, vitamin C group (positive control), and 5, 10 and 20 μg/mL Sal B groups. Cell viability of HUVECs was determined by 3-(4,5-dimethylthiazol- 2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The anti-apoptotic effect of Sal B was tested by Hoechst 33258 staining and Annexin V/propidium iodide flflow cytometry analysis. Apoptosis-related genes (p53, Bcl-2 and Bax) expression and caspase-3 activity were also determined. Oxidative stress markers malondialdehyde (MDA) and superoxide dismutase (SOD) were determined by the corresponding kits. RESULTS In HUVECs, ox-LDL signifificantly reduced cell viability and induced apoptosis (P<0.05 or P<0.01), however, Sal B diminished the effects of ox-LDL in a dose-dependent manner (P<0.05). Moreover, 10 and 20 μg/mL Sal B reduced the expression levels of p53, increased the Bcl-2/Bax ratio and inhibited the caspase-3 activity in ox-LDL-treated HUVECs (P<0.05). In addition, 5, 10 and 20 μg/mL Sal B signifificantly enhanced the activity of SOD, while decreased the level of MDA in the HUVECs which treated with ox-LDL (P<0.05). CONCLUSION Sal B exhibited anti-apoptotic effects in ox-LDL-induced endothelial cell injury by suppressing oxidative stress, p53, and caspase-3.
Collapse
Affiliation(s)
- Hong-Mei Chen
- Key Laboratory of Molecular Biology, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
- Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hao Luo
- Key Laboratory of Molecular Biology, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Wen-Bi Zeng
- Key Laboratory of Molecular Biology, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Bin Liu
- Key Laboratory of Molecular Biology, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jia-Cheng Huang
- The Fifth Department of Internal Medicine, Dongguan Hospital of Traditional Chinese Medicine, Dongguan, Guangdong Province, 523000, China
| | - Min Liu
- Department of Parasitology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yan-Jin Zeng
- The Fifth Department of Internal Medicine, Dongguan Hospital of Traditional Chinese Medicine, Dongguan, Guangdong Province, 523000, China
| | - Qiang Zheng
- Department of Traditional Chinese Medicine, Shantou Central Hospital, Shantou, Guangdong Province, 515031, China
| | - Ji-Qiang Li
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Xue-Gang Sun
- Key Laboratory of Molecular Biology, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Ying-Chun Zhou
- Key Laboratory of Molecular Biology, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
- Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
44
|
Epoxyeicosatrienoic Acid as Therapy for Diabetic and Ischemic Cardiomyopathy. Trends Pharmacol Sci 2016; 37:945-962. [DOI: 10.1016/j.tips.2016.08.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/12/2016] [Accepted: 08/17/2016] [Indexed: 12/19/2022]
|
45
|
Lee DS, Jeong GS. Butein provides neuroprotective and anti-neuroinflammatory effects through Nrf2/ARE-dependent haem oxygenase 1 expression by activating the PI3K/Akt pathway. Br J Pharmacol 2016; 173:2894-909. [PMID: 27465039 PMCID: PMC5055139 DOI: 10.1111/bph.13569] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 07/12/2016] [Accepted: 07/16/2016] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Butein, 3,4,2',4'-tetrahydroxychalcone, has various pharmacological effects. However, no study has demonstrated the specific neurobiological mechanisms of the effects of butein in neuronal cells. The present study examined the role of butein as an antioxidative and anti-inflammatory inducer of haem oxygenase 1 (HO1) in mouse hippocampal HT22, BV2 microglial and primary mouse hippocampus neurons. EXPERIMENTAL APPROACH We investigated the neuroprotective effects of butein on glutamate-induced HT22 cell and primary mouse hippocampal neuron death and its anti-neuroinflammatory effects on LPS-induced activation of BV2 cells. We elucidated the underlying mechanisms by assessing the involvement of NF-κB, HO1, nuclear factor-E2-related factor 2 (Nrf2) and Akt signalling. KEY RESULTS Butein decreased cellular oxidative injury and the production of ROS in glutamate-treated HT22 cells and primary mouse hippocampal neurons. Furthermore, butein suppressed LPS-induced pro-inflammatory enzymes and mediators in BV2 microglia. Butein inhibited IL-6, IL-1β and TNF-α production and mRNA expression. In addition, butein decreased NO and PGE2 production and inducible NOS and COX-2 expression through the NF-κB signalling pathway. Butein up-regulated Nrf2/ARE-mediated HO1 expression through the PI3K/Akt pathway and this was positively associated with its cytoprotective effects and anti-neuroinflammatory actions. CONCLUSION AND IMPLICATIONS Our results indicate that butein effectively prevents glutamate-induced oxidative damage and LPS-induced activation and that the induction of HO1 by butein through the PI3K/Akt pathway and Nrf2 activation appears to play a pivotal role in its effects on neuronal cells. Our results provide evidence for the neuroprotective properties of butein.
Collapse
Affiliation(s)
- Dong-Sung Lee
- College of Pharmacy, Chosun University, Gwangju, Korea
| | | |
Collapse
|
46
|
Yan X, Xun M, Li J, Wu L, Dou X, Zheng J. Activation of Na+/K+-ATPase attenuates high glucose-induced H9c2 cell apoptosis via suppressing ROS accumulation and MAPKs activities by DRm217. Acta Biochim Biophys Sin (Shanghai) 2016; 48:883-893. [PMID: 27563007 DOI: 10.1093/abbs/gmw079] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 05/23/2016] [Indexed: 01/08/2023] Open
Abstract
Hyperglycemia is one of the major factors responsible for the myocardial apoptosis and dysfunction in diabetes. Many studies have proved that there is a close relationship between decreased Na+/K+-ATPase activity and diabetic cardiomyopathy. However, the effect of directly activated Na+/K+-ATPase on high glucose-induced myocardial injury is still unknown. Here we found that DRm217, a Na+/K+-ATPase's DR-region specific monoclonal antibody and direct activator, could prevent high glucose-induced H9c2 cell injury, reactive oxygen species (ROS) release, and mitochondrial dysfunction. High glucose-treatment decreased Na+/K+-ATPase activity and increased intracellular Ca2+ level, whereas DRm217 increased Na+/K+-ATPase activity and alleviated Ca2+ overload. Inhibition of Ca2+ overload or closing sodium calcium exchanger (NCX channel) could reverse high glucose-induced ROS increasing and cell injury. In addition, DRm217 could significantly attenuate high glucose-induced p38, JNK and ERK1/2 phosphorylation, which were involved in high glucose-induced cell injury and ROS accumulation. Our findings suggest that DRm217 may protect against the deleterious effects of high glucose in the heart. Prevention of high glucose-induced myocardial cell injury by specific Na+/K+-ATPase activator may be an attractive therapeutic option.
Collapse
Affiliation(s)
- Xiaofei Yan
- Department of Biochemistry and Molecular Biology, Medical College of Xi'an Jiaotong University, Xi'an 710061, China
| | - Meng Xun
- Department of Immunology and Microbiology, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jing Li
- Department of Biochemistry and Molecular Biology, Medical College of Xi'an Jiaotong University, Xi'an 710061, China
| | - Litao Wu
- Department of Biochemistry and Molecular Biology, Medical College of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaojuan Dou
- Department of Biochemistry and Molecular Biology, Medical College of Xi'an Jiaotong University, Xi'an 710061, China
| | - Jin Zheng
- Hospital of Nephrology, First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
47
|
Diaz-Juarez J, Suarez J, Cividini F, Scott BT, Diemer T, Dai A, Dillmann WH. Expression of the mitochondrial calcium uniporter in cardiac myocytes improves impaired mitochondrial calcium handling and metabolism in simulated hyperglycemia. Am J Physiol Cell Physiol 2016; 311:C1005-C1013. [PMID: 27681178 DOI: 10.1152/ajpcell.00236.2016] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 09/23/2016] [Indexed: 12/21/2022]
Abstract
Diabetic cardiomyopathy is associated with metabolic changes, including decreased glucose oxidation (Gox) and increased fatty acid oxidation (FAox), which result in cardiac energetic deficiency. Diabetic hyperglycemia is a pathophysiological mechanism that triggers multiple maladaptive phenomena. The mitochondrial Ca2+ uniporter (MCU) is the channel responsible for Ca2+ uptake in mitochondria, and free mitochondrial Ca2+ concentration ([Ca2+]m) regulates mitochondrial metabolism. Experiments with cardiac myocytes (CM) exposed to simulated hyperglycemia revealed reduced [Ca2+]m and MCU protein levels. Therefore, we investigated whether returning [Ca2+]m to normal levels in CM by MCU expression could lead to normalization of Gox and FAox with no detrimental effects. Mouse neonatal CM were exposed for 72 h to normal glucose [5.5 mM glucose + 19.5 mM mannitol (NG)], high glucose [25 mM glucose (HG)], or HG + adenoviral MCU expression. Gox and FAox, [Ca2+]m, MCU levels, pyruvate dehydrogenase (PDH) activity, oxidative stress, mitochondrial membrane potential, and apoptosis were assessed. [Ca2+]m and MCU protein levels were reduced after 72 h of HG. Gox was decreased and FAox was increased in HG, PDH activity was decreased, phosphorylated PDH levels were increased, and mitochondrial membrane potential was reduced. MCU expression returned these parameters toward NG levels. Moreover, increased oxidative stress and apoptosis were reduced in HG by MCU expression. We also observed reduced MCU protein levels and [Ca2+]m in hearts from type 1 diabetic mice. Thus we conclude that HG-induced metabolic alterations can be reversed by restoration of MCU levels, resulting in return of [Ca2+]m to normal levels.
Collapse
Affiliation(s)
- Julieta Diaz-Juarez
- Department of Medicine, University of California, San Diego, La Jolla, California; and.,Department of Pharmacology, Instituto Nacional de Cardiología Ignacio Chávez, Tlalpan, México, Mexico
| | - Jorge Suarez
- Department of Medicine, University of California, San Diego, La Jolla, California; and
| | - Federico Cividini
- Department of Medicine, University of California, San Diego, La Jolla, California; and
| | - Brian T Scott
- Department of Medicine, University of California, San Diego, La Jolla, California; and
| | - Tanja Diemer
- Department of Medicine, University of California, San Diego, La Jolla, California; and
| | - Anzhi Dai
- Department of Medicine, University of California, San Diego, La Jolla, California; and
| | - Wolfgang H Dillmann
- Department of Medicine, University of California, San Diego, La Jolla, California; and
| |
Collapse
|
48
|
Suwanjang W, Prachayasittikul S, Prachayasittikul V. Effect of 8-hydroxyquinoline and derivatives on human neuroblastoma SH-SY5Y cells under high glucose. PeerJ 2016; 4:e2389. [PMID: 27635352 PMCID: PMC5012261 DOI: 10.7717/peerj.2389] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 07/29/2016] [Indexed: 12/21/2022] Open
Abstract
8-Hydroxyquinoline and derivatives exhibit multifunctional properties, including antioxidant, antineurodegenerative, anticancer, anti-inflammatory and antidiabetic activities. In biological systems, elevation of intracellular calcium can cause calpain activation, leading to cell death. Here, the effect of 8-hydroxyquinoline and derivatives (5-chloro-7-iodo-8-hydroxyquinoline or clioquinol and 8-hydroxy-5-nitroquinoline or nitroxoline) on calpain-dependent (calpain-calpastatin) pathways in human neuroblastoma (SH-SY5Y) cells was investigated. 8-Hydroxyquinoline and derivatives ameliorated high glucose toxicity in SH-SY5Y cells. The investigated compounds, particularly clioquinol, attenuated the increased expression of calpain, even under high-glucose conditions. 8-Hydroxyquinoline and derivatives thus adversely affected the promotion of neuronal cell death by high glucose via the calpain-calpastatin signaling pathways. These findings support the beneficial effects of 8-hydroxyquinolines for further therapeutic development.
Collapse
Affiliation(s)
- Wilasinee Suwanjang
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University , Bangkok , Thailand
| | - Supaluk Prachayasittikul
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University , Bangkok , Thailand
| | - Virapong Prachayasittikul
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University , Bangkok , Thailand
| |
Collapse
|
49
|
Zaman JAB, Harling L, Ashrafian H, Darzi A, Gooderham N, Athanasiou T, Peters NS. Post-operative atrial fibrillation is associated with a pre-existing structural and electrical substrate in human right atrial myocardium. Int J Cardiol 2016; 220:580-8. [PMID: 27390994 PMCID: PMC4994770 DOI: 10.1016/j.ijcard.2016.06.249] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 06/01/2016] [Accepted: 06/26/2016] [Indexed: 12/19/2022]
Abstract
Background Post-operative atrial fibrillation (POAF) is a major health economic burden. However, the precise mechanisms in POAF remain unclear. In other forms of AF, sites of high dominant frequency (DF) in sinus rhythm (SR) may harbour ‘AF nests’. We studied AF inducibility in relation to substrate changes using epicardial electrograms and cardiomyocyte calcium handling in the atria of AF naïve patients. Method Bipolar electrograms were recorded from the lateral right atrial (RA) wall in 34 patients undergoing coronary surgery using a high-density array in sinus rhythm (NSR). RA burst pacing at 200/500/1000 ms cycle lengths (CL) was performed, recording episodes of AF > 30 s. Co-localised RA tissue was snap frozen for RNA and protein extraction. Results Electrograms prolonged during AF (76.64 ± 29.35 ms) vs. NSR/pacing (p < 0.001). Compared to NSR, electrogram amplitude was reduced during AF and during pacing at 200 ms CL (p < 0.001). Electrogram DF was significantly lower in AF (75.87 ± 23.63 Hz) vs. NSR (89.33 ± 25.99 Hz) (p < 0.05), and NSR DF higher in AF inducible patients at the site of AF initiation (p < 0.05). Structurally, POAF atrial myocardium demonstrated reduced sarcolipin gene (p = 0.0080) and protein (p = 0.0242) expression vs. NSR. Phospholamban gene and protein expression was unchanged. SERCA2a protein expression remained unchanged, but MYH6 (p = 0.0297) and SERCA2A (p = 0.0343) gene expression was reduced in POAF. Conclusions Human atrial electrograms prolong and reduce in amplitude in induced peri-operative AF vs. NSR or pacing. In those sustaining AF, high DF sites in NSR may indicate ‘AF nests’. This electrical remodelling is accompanied by structural remodelling with altered expression of cardiomyocyte calcium handling detectable before POAF. These novel upstream substrate changes offer a novel mechanism and manifestation of human POAF.
Collapse
Affiliation(s)
- Junaid A B Zaman
- Myocardial Function, National Heart & Lung Institute, Hammersmith Hospital, Imperial College London, UK; Cardiovascular Medicine, Stanford University, Palo Alto, USA
| | - Leanne Harling
- Myocardial Function, National Heart & Lung Institute, Hammersmith Hospital, Imperial College London, UK; Department of Surgery and Cancer, St Mary's Hospital, Imperial College London, UK; Department of Biomolecular Medicine, South Kensington Campus, Imperial College London, UK
| | - Hutan Ashrafian
- Department of Surgery and Cancer, St Mary's Hospital, Imperial College London, UK; Department of Biomolecular Medicine, South Kensington Campus, Imperial College London, UK
| | - Ara Darzi
- Myocardial Function, National Heart & Lung Institute, Hammersmith Hospital, Imperial College London, UK
| | - Nigel Gooderham
- Department of Biomolecular Medicine, South Kensington Campus, Imperial College London, UK
| | - Thanos Athanasiou
- Myocardial Function, National Heart & Lung Institute, Hammersmith Hospital, Imperial College London, UK; Department of Surgery and Cancer, St Mary's Hospital, Imperial College London, UK.
| | - Nicholas S Peters
- Myocardial Function, National Heart & Lung Institute, Hammersmith Hospital, Imperial College London, UK
| |
Collapse
|
50
|
Novel triterpenoid AECHL-1 induces apoptosis in breast cancer cells by perturbing the mitochondria–endoplasmic reticulum interactions and targeting diverse apoptotic pathways. Biochim Biophys Acta Gen Subj 2016; 1860:1056-70. [DOI: 10.1016/j.bbagen.2016.02.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 01/29/2016] [Accepted: 02/02/2016] [Indexed: 12/18/2022]
|