1
|
Yu J, Li Y, Hu J, Wang Y. Interleukin-33 induces angiogenesis after myocardial infarction via AKT/eNOS signaling pathway. Int Immunopharmacol 2024; 143:113433. [PMID: 39486188 DOI: 10.1016/j.intimp.2024.113433] [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: 08/08/2024] [Revised: 10/06/2024] [Accepted: 10/15/2024] [Indexed: 11/04/2024]
Abstract
Myocardial infarction (MI) is one of the leading causes of mortality and morbidity worldwide. MI-damaged vascular structures are difficult to completely restore due to the heart's low regenerative capacity. Given interleukin-33 (IL-33) as a potent endothelial activator promoting angiogenesis, this study investigated the role of IL-33 in angiogenesis and cardiac repair after MI. A mouse model of MI was established. IL-33 improved cardiac function and induced an increase in vascular density after MI. Besides, IL-33 promoted human endothelial cells proliferation, migration, and differentiation under both normoxic and hypoxic conditions, consistently with increased angiogenesis in vivo. Mechanistic studies demonstrated that IL-33 could promote angiogenesis by activating eNOS and AKT, and stimulating NO production in vivo and in vitro. Given that injection of exogenous IL-33 induced an inflammatory response, we employed a multifunctional biomimetic nanoparticle drug delivery system to deliver IL-33, thereby enhancing its targeting to the heart for fibrotic therapy and reducing inflammation. In conclusion, our results indicate that IL-33 promotes endothelial angiogenesis after MI through AKT/eNOS/NO signaling pathway. PM&EM/IL-33 nanoparticles may hold promising therapeutic potential for protecting cardiac ischemic injury and mitigating inflammation.
Collapse
Affiliation(s)
- Jiaqi Yu
- Beijing Anzhen Hospital, Capital Medical University, Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Centre for Cardiovascular Disorders, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China; Beijing Institute of Heart, Lung and Blood Vessel Disease, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China.
| | - Yuyu Li
- Beijing Anzhen Hospital, Capital Medical University, Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Centre for Cardiovascular Disorders, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China; Beijing Institute of Heart, Lung and Blood Vessel Disease, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Jiaxin Hu
- Cardiovascular Disease Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi, Hubei, China; Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi 445000, China
| | - Yuan Wang
- Beijing Anzhen Hospital, Capital Medical University, Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Centre for Cardiovascular Disorders, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China; Beijing Institute of Heart, Lung and Blood Vessel Disease, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China.
| |
Collapse
|
2
|
Matboli M, Al-Amodi HS, Khaled A, Khaled R, Roushdy MMS, Ali M, Diab GI, Elnagar MF, Elmansy RA, TAhmed HH, Ahmed EME, Elzoghby DMA, M.Kamel HF, Farag MF, ELsawi HA, Farid LM, Abouelkhair MB, Habib EK, Fikry H, Saleh LA, Aboughaleb IH. Comprehensive machine learning models for predicting therapeutic targets in type 2 diabetes utilizing molecular and biochemical features in rats. Front Endocrinol (Lausanne) 2024; 15:1384984. [PMID: 38854687 PMCID: PMC11157016 DOI: 10.3389/fendo.2024.1384984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 05/03/2024] [Indexed: 06/11/2024] Open
Abstract
Introduction With the increasing prevalence of type 2 diabetes mellitus (T2DM), there is an urgent need to discover effective therapeutic targets for this complex condition. Coding and non-coding RNAs, with traditional biochemical parameters, have shown promise as viable targets for therapy. Machine learning (ML) techniques have emerged as powerful tools for predicting drug responses. Method In this study, we developed an ML-based model to identify the most influential features for drug response in the treatment of type 2 diabetes using three medicinal plant-based drugs (Rosavin, Caffeic acid, and Isorhamnetin), and a probiotics drug (Z-biotic), at different doses. A hundred rats were randomly assigned to ten groups, including a normal group, a streptozotocin-induced diabetic group, and eight treated groups. Serum samples were collected for biochemical analysis, while liver tissues (L) and adipose tissues (A) underwent histopathological examination and molecular biomarker extraction using quantitative PCR. Utilizing five machine learning algorithms, we integrated 32 molecular features and 12 biochemical features to select the most predictive targets for each model and the combined model. Results and discussion Our results indicated that high doses of the selected drugs effectively mitigated liver inflammation, reduced insulin resistance, and improved lipid profiles and renal function biomarkers. The machine learning model identified 13 molecular features, 10 biochemical features, and 20 combined features with an accuracy of 80% and AUC (0.894, 0.93, and 0.896), respectively. This study presents an ML model that accurately identifies effective therapeutic targets implicated in the molecular pathways associated with T2DM pathogenesis.
Collapse
Affiliation(s)
- Marwa Matboli
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hiba S. Al-Amodi
- Biochemistry Department, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Abdelrahman Khaled
- Bioinformatics Group, Center of Informatics Sciences (CIS), School of Information Technology and Computer Sciences, Nile University, Giza, Egypt
| | - Radwa Khaled
- Biotechnology/Biomolecular Chemistry Department, Faculty of Science, Cairo University, Cairo, Egypt
- Medicinal Biochemistry and Molecular Biology Department, Modern University for Technology and Information, Cairo, Egypt
| | - Marian M. S. Roushdy
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Marwa Ali
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | | | - Rasha A. Elmansy
- Anatomy Unit, Department of Basic Medical Sciences, College of Medicine and Medical Sciences, Qassim University, Buraydah, Saudi Arabia
- Department of Anatomy and Cell Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hagir H. TAhmed
- Anatomy Unit, Department of Basic Medical Sciences, College of Medicine and Medical Sciences, AlNeelain University, Khartoum, Sudan
| | - Enshrah M. E. Ahmed
- Pathology Unit, Department of Basic Medical Sciences, College of Medicine and Medical Sciences, Gassim University, Buraydah, Saudi Arabia
| | | | - Hala F. M.Kamel
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
- Biochemistry Department, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mohamed F. Farag
- Medical Physiology Department, Armed Forces College of Medicine, Cairo, Egypt
| | - Hind A. ELsawi
- Department of Internal Medicine, Badr University in Cairo, Badr, Egypt
| | - Laila M. Farid
- Pathology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Eman K. Habib
- Department of Anatomy and Cell Biology, Faculty of Medicine, Galala University, Attaka, Suez Governorate, Egypt
| | - Heba Fikry
- Department of Histology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Lobna A. Saleh
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | |
Collapse
|
3
|
Mone P, Agyapong ED, Morciano G, Jankauskas SS, De Luca A, Varzideh F, Pinton P, Santulli G. Dysfunctional mitochondria elicit bioenergetic decline in the aged heart. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:13. [PMID: 39015481 PMCID: PMC11250775 DOI: 10.20517/jca.2023.50] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Aging represents a complex biological progression affecting the entire body, marked by a gradual decline in tissue function, rendering organs more susceptible to stress and diseases. The human heart holds significant importance in this context, as its aging process poses life-threatening risks. It entails macroscopic morphological shifts and biochemical changes that collectively contribute to diminished cardiac function. Among the numerous pivotal factors in aging, mitochondria play a critical role, intersecting with various molecular pathways and housing several aging-related agents. In this comprehensive review, we provide an updated overview of the functional role of mitochondria in cardiac aging.
Collapse
Affiliation(s)
- Pasquale Mone
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Medicine and Health Sciences, University of Molise, Campobasso 86100, Italy
| | - Esther Densu Agyapong
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Medical Sciences, University of Ferrara, Ferrara 44121, Italy
| | - Giampaolo Morciano
- Department of Medical Sciences, University of Ferrara, Ferrara 44121, Italy
- Maria Cecilia Hospital, GVM Care & Research, Cotignola 48033, Italy
| | - Stanislovas S. Jankauskas
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Antonio De Luca
- Department of Mental and Physical Health and Preventive Medicine, Vanvitelli University, Naples 80100, Italy
| | - Fahimeh Varzideh
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, Ferrara 44121, Italy
- Maria Cecilia Hospital, GVM Care & Research, Cotignola 48033, Italy
| | - Gaetano Santulli
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Medicine and Health Sciences, University of Molise, Campobasso 86100, Italy
- Department of Advanced Biomedical Sciences, “Federico II” University, International Translational Research and Medical Education (ITME) Consortium, Academic Research Unit, Naples 80131, Italy
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
4
|
Samidurai A, Olex AL, Ockaili R, Kraskauskas D, Roh SK, Kukreja RC, Das A. Integrated Analysis of lncRNA-miRNA-mRNA Regulatory Network in Rapamycin-Induced Cardioprotection against Ischemia/Reperfusion Injury in Diabetic Rabbits. Cells 2023; 12:2820. [PMID: 38132140 PMCID: PMC10742118 DOI: 10.3390/cells12242820] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
The inhibition of mammalian target of rapamycin (mTOR) with rapamycin (RAPA) provides protection against myocardial ischemia/reperfusion (I/R) injury in diabetes. Since interactions between transcripts, including long non-coding RNA (lncRNA), microRNA(miRNA) and mRNA, regulate the pathophysiology of disease, we performed unbiased miRarray profiling in the heart of diabetic rabbits following I/R injury with/without RAPA treatment to identify differentially expressed (DE) miRNAs and their predicted targets of lncRNAs/mRNAs. Results showed that among the total of 806 unique miRNAs targets, 194 miRNAs were DE after I/R in diabetic rabbits. Specifically, eight miRNAs, including miR-199a-5p, miR-154-5p, miR-543-3p, miR-379-3p, miR-379-5p, miR-299-5p, miR-140-3p, and miR-497-5p, were upregulated and 10 miRNAs, including miR-1-3p, miR-1b, miR-29b-3p, miR-29c-3p, miR-30e-3p, miR-133c, miR-196c-3p, miR-322-5p, miR-499-5p, and miR-672-5p, were significantly downregulated after I/R injury. Interestingly, RAPA treatment significantly reversed these changes in miRNAs. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis indicated the participation of miRNAs in the regulation of several signaling pathways related to I/R injury, including MAPK signaling and apoptosis. Furthermore, in diabetic hearts, the expression of lncRNAs, HOTAIR, and GAS5 were induced after I/R injury, but RAPA suppressed these lncRNAs. In contrast, MALAT1 was significantly reduced following I/R injury, with the increased expression of miR-199a-5p and suppression of its target, the anti-apoptotic protein Bcl-2. RAPA recovered MALAT1 expression with its sponging effect on miR-199-5p and restoration of Bcl-2 expression. The identification of novel targets from the transcriptome analysis in RAPA-treated diabetic hearts could potentially lead to the development of new therapeutic strategies for diabetic patients with myocardial infarction.
Collapse
Affiliation(s)
- Arun Samidurai
- Division of Cardiology, Pauley Heart Center, Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (A.S.); (R.O.); (D.K.); (S.K.R.)
| | - Amy L. Olex
- Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Ramzi Ockaili
- Division of Cardiology, Pauley Heart Center, Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (A.S.); (R.O.); (D.K.); (S.K.R.)
| | - Donatas Kraskauskas
- Division of Cardiology, Pauley Heart Center, Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (A.S.); (R.O.); (D.K.); (S.K.R.)
| | - Sean K. Roh
- Division of Cardiology, Pauley Heart Center, Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (A.S.); (R.O.); (D.K.); (S.K.R.)
| | - Rakesh C. Kukreja
- Division of Cardiology, Pauley Heart Center, Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (A.S.); (R.O.); (D.K.); (S.K.R.)
| | - Anindita Das
- Division of Cardiology, Pauley Heart Center, Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (A.S.); (R.O.); (D.K.); (S.K.R.)
| |
Collapse
|
5
|
Takenaka M, Kodama M, Murayama T, Ishigami-Yuasa M, Mori S, Ishida R, Suzuki J, Kanemaru K, Sugihara M, Iino M, Miura A, Nishio H, Morimoto S, Kagechika H, Sakurai T, Kurebayashi N. Screening for Novel Type 2 Ryanodine Receptor Inhibitors by Endoplasmic Reticulum Ca 2+ Monitoring. Mol Pharmacol 2023; 104:275-286. [PMID: 37678938 DOI: 10.1124/molpharm.123.000720] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/21/2023] [Accepted: 08/29/2023] [Indexed: 09/09/2023] Open
Abstract
Type 2 ryanodine receptor (RyR2) is a Ca2+ release channel on the endoplasmic (ER)/sarcoplasmic reticulum that plays a central role in the excitation-contraction coupling in the heart. Hyperactivity of RyR2 has been linked to ventricular arrhythmias in patients with catecholaminergic polymorphic ventricular tachycardia and heart failure, where spontaneous Ca2+ release via hyperactivated RyR2 depolarizes diastolic membrane potential to induce triggered activity. In such cases, drugs that suppress RyR2 activity are expected to prevent the arrhythmias, but there is no clinically available RyR2 inhibitors at present. In this study, we searched for RyR2 inhibitors from a well-characterized compound library using a recently developed ER Ca2+-based assay, where the inhibition of RyR2 activity was detected by the increase in ER Ca2+ signals from R-CEPIA1er, a genetically encoded ER Ca2+ indicator, in RyR2-expressing HEK293 cells. By screening 1535 compounds in the library, we identified three compounds (chloroxylenol, methyl orsellinate, and riluzole) that greatly increased the ER Ca2+ signal. All of the three compounds suppressed spontaneous Ca2+ oscillations in RyR2-expressing HEK293 cells and correspondingly reduced the Ca2+-dependent [3H]ryanodine binding activity. In cardiomyocytes from RyR2-mutant mice, the three compounds effectively suppressed abnormal Ca2+ waves without substantial effects on the action-potential-induced Ca2+ transients. These results confirm that ER Ca2+-based screening is useful for identifying modulators of ER Ca2+ release channels and suggest that RyR2 inhibitors have potential to be developed as a new category of antiarrhythmic drugs. SIGNIFICANCE STATEMENT: We successfully identified three compounds having RyR2 inhibitory action from a well-characterized compound library using an endoplasmic reticulum Ca2+-based assay, and demonstrated that these compounds suppressed arrhythmogenic Ca2+ wave generation without substantially affecting physiological action-potential induced Ca2+ transients in cardiomyocytes. This study will facilitate the development of RyR2-specific inhibitors as a potential new class of drugs for life-threatening arrhythmias induced by hyperactivation of RyR2.
Collapse
Affiliation(s)
- Mai Takenaka
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Masami Kodama
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Takashi Murayama
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Mari Ishigami-Yuasa
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Shuichi Mori
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Ryosuke Ishida
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Junji Suzuki
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Kazunori Kanemaru
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Masami Sugihara
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Masamitsu Iino
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Aya Miura
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Hajime Nishio
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Sachio Morimoto
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Hiroyuki Kagechika
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Takashi Sakurai
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| | - Nagomi Kurebayashi
- Department of Cellular and Molecular Pharmacology (M.T., M.K., T.M., T.S., N.K.) and Department of Clinical Laboratory Medicine (M.S.), Juntendo University Graduate School of Medicine, Tokyo, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan (M.I.-Y., Sh.M., R.I., H.K.); Department of Physiology, University of California San Francisco, San Francisco, California (J.S.); Department of Physiology, Nihon University School of Medicine, Tokyo, Japan (K.K., M.I.); Department of Legal Medicine, Hyogo Medical University, Nishinomiya, Japan (A.M., H.N.); and Department of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan (Sa.M.)
| |
Collapse
|
6
|
Ramos-Mondragón R, Lozhkin A, Vendrov AE, Runge MS, Isom LL, Madamanchi NR. NADPH Oxidases and Oxidative Stress in the Pathogenesis of Atrial Fibrillation. Antioxidants (Basel) 2023; 12:1833. [PMID: 37891912 PMCID: PMC10604902 DOI: 10.3390/antiox12101833] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Atrial fibrillation (AF) is the most common type of cardiac arrhythmia and its prevalence increases with age. The irregular and rapid contraction of the atria can lead to ineffective blood pumping, local blood stasis, blood clots, ischemic stroke, and heart failure. NADPH oxidases (NOX) and mitochondria are the main sources of reactive oxygen species in the heart, and dysregulated activation of NOX and mitochondrial dysfunction are associated with AF pathogenesis. NOX- and mitochondria-derived oxidative stress contribute to the onset of paroxysmal AF by inducing electrophysiological changes in atrial myocytes and structural remodeling in the atria. Because high atrial activity causes cardiac myocytes to expend extremely high energy to maintain excitation-contraction coupling during persistent AF, mitochondria, the primary energy source, undergo metabolic stress, affecting their morphology, Ca2+ handling, and ATP generation. In this review, we discuss the role of oxidative stress in activating AF-triggered activities, regulating intracellular Ca2+ handling, and functional and anatomical reentry mechanisms, all of which are associated with AF initiation, perpetuation, and progression. Changes in the extracellular matrix, inflammation, ion channel expression and function, myofibril structure, and mitochondrial function occur during the early transitional stages of AF, opening a window of opportunity to target NOX and mitochondria-derived oxidative stress using isoform-specific NOX inhibitors and mitochondrial ROS scavengers, as well as drugs that improve mitochondrial dynamics and metabolism to treat persistent AF and its transition to permanent AF.
Collapse
Affiliation(s)
- Roberto Ramos-Mondragón
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Drive, 2301 Medical Science Research Building III, Ann Arbor, MI 48109, USA; (R.R.-M.); (L.L.I.)
| | - Andrey Lozhkin
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Aleksandr E. Vendrov
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Marschall S. Runge
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Lori L. Isom
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Drive, 2301 Medical Science Research Building III, Ann Arbor, MI 48109, USA; (R.R.-M.); (L.L.I.)
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nageswara R. Madamanchi
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| |
Collapse
|
7
|
Dridi H, Santulli G, Bahlouli L, Miotto MC, Weninger G, Marks AR. Mitochondrial Calcium Overload Plays a Causal Role in Oxidative Stress in the Failing Heart. Biomolecules 2023; 13:1409. [PMID: 37759809 PMCID: PMC10527470 DOI: 10.3390/biom13091409] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/13/2023] [Accepted: 09/17/2023] [Indexed: 09/29/2023] Open
Abstract
Heart failure is a serious global health challenge, affecting more than 6.2 million people in the United States and is projected to reach over 8 million by 2030. Independent of etiology, failing hearts share common features, including defective calcium (Ca2+) handling, mitochondrial Ca2+ overload, and oxidative stress. In cardiomyocytes, Ca2+ not only regulates excitation-contraction coupling, but also mitochondrial metabolism and oxidative stress signaling, thereby controlling the function and actual destiny of the cell. Understanding the mechanisms of mitochondrial Ca2+ uptake and the molecular pathways involved in the regulation of increased mitochondrial Ca2+ influx is an ongoing challenge in order to identify novel therapeutic targets to alleviate the burden of heart failure. In this review, we discuss the mechanisms underlying altered mitochondrial Ca2+ handling in heart failure and the potential therapeutic strategies.
Collapse
Affiliation(s)
- Haikel Dridi
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY 10032, USA; (L.B.); (M.C.M.); (G.W.); (A.R.M.)
| | - Gaetano Santulli
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA;
| | - Laith Bahlouli
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY 10032, USA; (L.B.); (M.C.M.); (G.W.); (A.R.M.)
| | - Marco C. Miotto
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY 10032, USA; (L.B.); (M.C.M.); (G.W.); (A.R.M.)
| | - Gunnar Weninger
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY 10032, USA; (L.B.); (M.C.M.); (G.W.); (A.R.M.)
| | - Andrew R. Marks
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians & Surgeons, New York, NY 10032, USA; (L.B.); (M.C.M.); (G.W.); (A.R.M.)
| |
Collapse
|
8
|
Salemi S, Zamanian MY, Giménez‐Llort L, Jalali Z, Mahmoodi M, Golmohammadi M, Kaeidi A, Taghipour Z, Khademalhosseini M, Modanloo M, Hajizadehi MR. Distinct signatures on d-galactose-induced aging and preventive/protective potency of two low-dose vitamin D supplementation regimens on working memory, muscular damage, cardiac and cerebral oxidative stress, and SIRT1 and calstabin2 downregulation. Food Sci Nutr 2023; 11:5050-5062. [PMID: 37701236 PMCID: PMC10494626 DOI: 10.1002/fsn3.3422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/21/2023] [Accepted: 04/22/2023] [Indexed: 09/14/2023] Open
Abstract
Chronic administration of d-galactose (d-gal) in rodents reproduces the overproduction of reactive oxygen species of physiological aging. The present research shows for the first time distinct signatures on d-gal-induced aging (500 mg/kg, 6 weeks) and the preventive and protective potential of two vitamin D (50 IU) supplementation regimens (pre-induction and simultaneous, respectively) in two vital organs (heart and brain). d-gal-induced notorious alterations in working memory, a strong increase in brain malondialdehyde (MDA) oxidative levels, and strong downregulation of sirtuin 1 (SIRT1) in the heart and hippocampus and of calstabin2 in the heart. Cardiac and brain superoxide dismutase (SOD) and glutathione peroxidase (GPx) enzymatic antioxidant capacities were damaged, brain calstabin2 was downregulated, and neuropathology was observed. Heart damage also included a moderate increase in MDA levels, serologic lactate dehydrogenase (LDH), total creatine kinase (CK) activities, and histopathological alterations. The used dose of vitamin D was enough to prevent cognitive impairment, avoid muscular damage, hamper cardiac and cerebral oxidative stress, and SIRT1 and calstabin2 downregulation. Most importantly, the potencies of the two preventive schedules depended on the tissue and level of study. The pre-induction schedule prevented d-gal-induced aging by 1 order of magnitude higher than simultaneous administration in all the variables studied except for SIRT1, whose strong downregulation induced by d-gal was equally prevented by both schedules. The benefits of vitamin D for oxidative stress were stronger in the brain than in the heart. Brain MDA levels were more sensitive to damage, while SOD and GPx antioxidant enzymatic activities were in the heart. In this order, the magnitude of SOD, MDA, and GPx oxidative stress markers was sensitive to prevention. In summary, the results unveiled distinct aging induction, preventive signatures, and sensitivity of markers depending on different levels of study and tissues, which are relevant from a mechanistic view and in the design of targeted interventions.
Collapse
Affiliation(s)
- Sahar Salemi
- Department of Biochemistry, School of MedicineRafsanjan University of Medical SciencesRafsanjanIran
| | - Mohammad Yasin Zamanian
- Neurophysiology Research CenterHamadan University of Medical SciencesHamadanIran
- Department of Pharmacology and Toxicology, School of PharmacyHamadan University of Medical SciencesHamadanIran
| | - Lydia Giménez‐Llort
- Institute of Neuroscience & Department of Psychiatry and Forensic MedicineUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Zahra Jalali
- Department of Biochemistry, School of MedicineRafsanjan University of Medical SciencesRafsanjanIran
| | - Mehdi Mahmoodi
- Department of Clinical Biochemistry, Afzalipoor Faculty of MedicineKerman University of Medical SciencesKermanIran
| | | | - Ayat Kaeidi
- Department of Physiology, School of MedicineRafsanjan University of Medical SciencesRafsanjanIran
| | - Zahra Taghipour
- Department of Anatomy, School of MedicineRafsanjan University of Medical SciencesRafsanjanIran
| | - Morteza Khademalhosseini
- Department of Pathology, School of MedicineRafsanjan University of Medical SciencesRafsanjanIran
| | - Mona Modanloo
- Pharmaceutical Sciences Research CenterMazandaran University of Medical SciencesSariIran
| | - Mohammad Reza Hajizadehi
- Department of Biochemistry, School of MedicineRafsanjan University of Medical SciencesRafsanjanIran
| |
Collapse
|
9
|
Walweel K, Beard N, van Helden DF, Laver DR. Dantrolene inhibition of ryanodine channels (RyR2) in artificial lipid bilayers depends on FKBP12.6. J Gen Physiol 2023; 155:e202213277. [PMID: 37279522 PMCID: PMC10244881 DOI: 10.1085/jgp.202213277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 03/18/2023] [Accepted: 05/22/2023] [Indexed: 06/08/2023] Open
Abstract
Dantrolene is a neutral hydantoin that is clinically used as a skeletal muscle relaxant to prevent overactivation of the skeletal muscle calcium release channel (RyR1) in response to volatile anesthetics. Dantrolene has aroused considerable recent interest as a lead compound for stabilizing calcium release due to overactive cardiac calcium release channels (RyR2) in heart failure. Previously, we found that dantrolene produces up to a 45% inhibition RyR2 with an IC50 of 160 nM, and that this inhibition requires the physiological association between RyR2 and CaM. In this study, we tested the hypothesis that dantrolene inhibition of RyR2 in the presence of CaM is modulated by RyR2 phosphorylation at S2808 and S2814. Phosphorylation was altered by incubations with either exogenous phosphatase (PP1) or kinases; PKA to phosphorylate S2808 or endogenous CaMKII to phosphorylate S2814. We found that PKA caused selective dissociation of FKBP12.6 from the RyR2 complex and a loss of dantrolene inhibition. Rapamycin-induced FKBP12.6 dissociation from RyR2 also resulted in the loss of dantrolene inhibition. Subsequent incubations of RyR2 with exogenous FKBP12.6 reinstated dantrolene inhibition. These findings indicate that the inhibitory action of dantrolene on RyR2 depends on RyR2 association with FKBP12.6 in addition to CaM as previously found.
Collapse
Affiliation(s)
- Kafa Walweel
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, Australia
| | - Nicole Beard
- Faculty of Science and Technology, University of Canberra, Bruce, Australia
| | - Dirk F. van Helden
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, Australia
| | - Derek R. Laver
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, Australia
| |
Collapse
|
10
|
Jankauskas SS, Mone P, Avvisato R, Varzideh F, De Gennaro S, Salemme L, Macina G, Kansakar U, Cioppa A, Frullone S, Gambardella J, Di Mauro M, Tesorio T, Santulli G. miR-181c targets Parkin and SMAD7 in human cardiac fibroblasts: Validation of differential microRNA expression in patients with diabetes and heart failure with preserved ejection fraction. Mech Ageing Dev 2023; 212:111818. [PMID: 37116731 DOI: 10.1016/j.mad.2023.111818] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/15/2023] [Accepted: 04/19/2023] [Indexed: 04/30/2023]
Abstract
BACKGROUND Cardiac fibrosis represents a key feature in the pathophysiology of heart failure with preserved ejection fraction (HFpEF), a condition highly prevalent amongst geriatric patients, especially if diabetic. The microRNA miR-181c has been shown to be associated with the response to exercise training in HFpEF patients and has been also linked to diabetic cardiovascular complications. However, the underlying mechanisms have not been fully elucidated. OBJECTIVE To measure circulating miR-181c in elderly patients with HFpEF and DM and identify gene targets pathophysiologically relevant in HFpEF. METHODS We quantified circulating miR-181c in frail older adults with a confirmed diagnosis of HFpEF and diabetes, and, as control, we enrolled age-matched subjects without HFpEF and without diabetes. We validated in human cardiac fibroblasts the molecular mechanisms linking miR-181c to a pro-fibrotic response. RESULTS 51 frail patients were included (34 patients with diabetes and HFpEF and 17 age-matched controls. We observed that miR-181c was significantly upregulated (p<0.0001) in HFpEF patients vs controls. We confirmed in vitro that miR-181c is targeting PRKN and SMAD7. CONCLUSIONS We demonstrate that miR-181c levels are significantly increased in frail elderly adults with diabetes and HFpEF and that miR-181c targets PRKN and SMAD7 in human cardiac fibroblasts.
Collapse
Affiliation(s)
- Stanislovas S Jankauskas
- Department of Medicine, Einstein Institute for Aging Research, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Pasquale Mone
- Department of Medicine, Einstein Institute for Aging Research, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA; ASL Avellino, Avellino, 83100, Italy
| | - Roberta Avvisato
- Department of Medicine, Einstein Institute for Aging Research, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Fahimeh Varzideh
- Department of Medicine, Einstein Institute for Aging Research, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA
| | | | - Luigi Salemme
- Casa di Cura "Montevergine", Mercogliano (Avellino), 83013, Italy
| | | | - Urna Kansakar
- Department of Medicine, Einstein Institute for Aging Research, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Angelo Cioppa
- Casa di Cura "Montevergine", Mercogliano (Avellino), 83013, Italy
| | | | - Jessica Gambardella
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA
| | | | - Tullio Tesorio
- Casa di Cura "Montevergine", Mercogliano (Avellino), 83013, Italy
| | - Gaetano Santulli
- Department of Medicine, Einstein Institute for Aging Research, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA; Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA.
| |
Collapse
|
11
|
Mone P, Varzideh F, Jankauskas SS, Pansini A, Lombardi A, Frullone S, Santulli G. SGLT2 Inhibition via Empagliflozin Improves Endothelial Function and Reduces Mitochondrial Oxidative Stress: Insights From Frail Hypertensive and Diabetic Patients. Hypertension 2022; 79:1633-1643. [PMID: 35703100 PMCID: PMC9642044 DOI: 10.1161/hypertensionaha.122.19586] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/01/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Frailty is a multidimensional condition often diagnosed in older adults with hypertension and diabetes, and both these conditions are associated with endothelial dysfunction and oxidative stress. We investigated the functional role of the SGLT2 (sodium glucose cotransporter 2) inhibitor empagliflozin in frail diabetic and hypertensive older adults. METHODS We studied the effects of empagliflozin in consecutive hypertensive and diabetic older patients with frailty presenting at the ASL (local health unit of the Italian Ministry of Health) of Avellino, Italy, from March 2021 to January 2022. Moreover, we performed in vitro experiments in human endothelial cells to measure cell viability, permeability, mitochondrial Ca2+, and oxidative stress. RESULTS We evaluated 407 patients; 325 frail elders with diabetes successfully completed the study. We propensity-score matched 75 patients treated with empagliflozin and 75 with no empagliflozin. We observed a correlation between glycemia and Montreal Cognitive Assessment (MoCA) score and between glycemia and 5-meter gait speed (5mGS). At 3-month follow-up, we detected a significant improvement in the MoCA score and in the 5mGS in patients receiving empagliflozin compared with non-treated subjects. Mechanistically, we demonstrate that empagliflozin significantly reduces mitochondrial Ca2+ overload and reactive oxygen species production triggered by high glucose in human endothelial cells, attenuates cellular permeability, and improves cell viability in response to oxidative stress. CONCLUSIONS Taken together, our data indicate that empagliflozin reduces frailty in diabetic and hypertensive patients, most likely by decreasing the mitochondrial generation of reactive oxygen species in endothelial cells.
Collapse
Affiliation(s)
- Pasquale Mone
- Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, New York, NY
- Asl Avellino
| | - Fahimeh Varzideh
- Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, New York, NY
| | | | | | - Angela Lombardi
- Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, New York, NY
| | | | - Gaetano Santulli
- Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, New York, NY
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY
| |
Collapse
|
12
|
Dridi H, Forrester F, Umanskaya A, Xie W, Reiken S, Lacampagne A, Marks A. Role of oxidation of excitation-contraction coupling machinery in age-dependent loss of muscle function in C. elegans. eLife 2022; 11:75529. [PMID: 35506650 PMCID: PMC9113742 DOI: 10.7554/elife.75529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
Age-dependent loss of body wall muscle function and impaired locomotion occur within 2 weeks in C. elegans; however, the underlying mechanism has not been fully elucidated. In humans, age-dependent loss of muscle function occurs at about 80 years of age and has been linked to dysfunction of ryanodine receptor (RyR)/intracellular calcium (Ca2+) release channels on the sarcoplasmic reticulum (SR). Mammalian skeletal muscle RyR1 channels undergo age-related remodeling due to oxidative overload, leading to loss of the stabilizing subunit calstabin1 (FKBP12) from the channel macromolecular complex. This destabilizes the closed state of the channel resulting in intracellular Ca2+ leak, reduced muscle function, and impaired exercise capacity. We now show that the C. elegans RyR homolog, UNC-68, exhibits a remarkable degree of evolutionary conservation with mammalian RyR channels and similar age-dependent dysfunction. Like RyR1 in mammals UNC-68 encodes a protein that comprises a macromolecular complex which includes the calstabin1 homolog FKB-2 and is immunoreactive with antibodies raised against the RyR1 complex. Further, as in aged mammals, UNC-68 is oxidized and depleted of FKB-2 in an age-dependent manner, resulting in 'leaky' channels, depleted SR Ca2+ stores, reduced body wall muscle Ca2+ transients, and age-dependent muscle weakness. FKB-2 (ok3007)-deficient worms exhibit reduced exercise capacity. Pharmacologically induced oxidization of UNC-68 and depletion of FKB-2 from the channel independently caused reduced body wall muscle Ca2+ transients. Preventing FKB-2 depletion from the UNC-68 macromolecular complex using the Rycal drug S107 improved muscle Ca2+ transients and function. Taken together, these data suggest that UNC-68 oxidation plays a role in age-dependent loss of muscle function. Remarkably, this age-dependent loss of muscle function induced by oxidative overload, which takes ~2 years in mice and ~80 years in humans, occurs in less than 2-3 weeks in C. elegans, suggesting that reduced antioxidant capacity may contribute to the differences in life span amongst species.
Collapse
Affiliation(s)
- Haikel Dridi
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| | - Frances Forrester
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| | - Alisa Umanskaya
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| | - Wenjun Xie
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| | - Steven Reiken
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| | - Alain Lacampagne
- U1046, Montpellier University, INSERM, CNRS, Montpellier, France
| | - Andrew Marks
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, United States
| |
Collapse
|
13
|
Dridi H, Santulli G, Gambardella J, Jankauskas SS, Yuan Q, Yang J, Reiken S, Wang X, Wronska A, Liu X, Lacampagne A, Marks AR. IP3 receptor orchestrates maladaptive vascular responses in heart failure. J Clin Invest 2022; 132:e152859. [PMID: 35166236 PMCID: PMC8843748 DOI: 10.1172/jci152859] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 12/21/2021] [Indexed: 12/02/2022] Open
Abstract
Patients with heart failure (HF) have augmented vascular tone, which increases cardiac workload, impairing ventricular output and promoting further myocardial dysfunction. The molecular mechanisms underlying the maladaptive vascular responses observed in HF are not fully understood. Vascular smooth muscle cells (VSMCs) control vasoconstriction via a Ca2+-dependent process, in which the type 1 inositol 1,4,5-trisphosphate receptor (IP3R1) on the sarcoplasmic reticulum (SR) plays a major role. To dissect the mechanistic contribution of intracellular Ca2+ release to the increased vascular tone observed in HF, we analyzed the remodeling of IP3R1 in aortic tissues from patients with HF and from controls. VSMC IP3R1 channels from patients with HF and HF mice were hyperphosphorylated by both serine and tyrosine kinases. VSMCs isolated from IP3R1VSMC-/- mice exhibited blunted Ca2+ responses to angiotensin II (ATII) and norepinephrine compared with control VSMCs. IP3R1VSMC-/- mice displayed significantly reduced responses to ATII, both in vivo and ex vivo. HF IP3R1VSMC-/- mice developed significantly less afterload compared with HF IP3R1fl/fl mice and exhibited significantly attenuated progression toward decompensated HF and reduced interstitial fibrosis. Ca2+-dependent phosphorylation of the MLC by MLCK activated VSMC contraction. MLC phosphorylation was markedly increased in VSMCs from patients with HF and HF mice but reduced in VSMCs from HF IP3R1VSMC-/- mice and HF WT mice treated with ML-7. Taken together, our data indicate that VSMC IP3R1 is a major effector of increased vascular tone, which contributes to increased cardiac afterload and decompensation in HF.
Collapse
MESH Headings
- Animals
- Calcium Signaling
- Heart Failure/genetics
- Heart Failure/metabolism
- Heart Failure/physiopathology
- Humans
- Inositol 1,4,5-Trisphosphate Receptors/genetics
- Inositol 1,4,5-Trisphosphate Receptors/metabolism
- Mice
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Vasoconstriction
Collapse
Affiliation(s)
- Haikel Dridi
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Gaetano Santulli
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
- Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, New York, New York, USA
- Department of Molecular Pharmacology, Einstein-Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Neuroimmunology and Inflammation, Albert Einstein College of Medicine, New York, New York, USA
| | - Jessica Gambardella
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
- Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, New York, New York, USA
- International Translational Research and Medical Education (ITME) Consortium, Department of Advanced Biomedical Science, “Federico II” University, Naples, Italy
| | - Stanislovas S. Jankauskas
- Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, New York, New York, USA
- Department of Molecular Pharmacology, Einstein-Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Neuroimmunology and Inflammation, Albert Einstein College of Medicine, New York, New York, USA
| | - Qi Yuan
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Jingyi Yang
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Steven Reiken
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Xujun Wang
- Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, New York, New York, USA
- Department of Molecular Pharmacology, Einstein-Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Neuroimmunology and Inflammation, Albert Einstein College of Medicine, New York, New York, USA
| | - Anetta Wronska
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Xiaoping Liu
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Alain Lacampagne
- PhyMedExp, University of Montpellier, CNRS, INSERM, CHRU Montpellier, Montpellier, France
| | - Andrew R. Marks
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
14
|
Mone P, de Donato A, Varzideh F, Kansakar U, Jankauskas SS, Pansini A, Santulli G. Functional role of miR-34a in diabetes and frailty. FRONTIERS IN AGING 2022; 3:949924. [PMID: 35923683 PMCID: PMC9340262 DOI: 10.3389/fragi.2022.949924] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 06/29/2022] [Indexed: 01/05/2023]
Abstract
Emerging evidence has shown that microRNAs (miRNAs) play critical role in the pathogenesis of several disorders. In the present minireview, we focus our attention on the functional role of a specific miRNA, namely miR-34a, in the pathophysiology of frailty and diabetes mellitus. Based on the current literature, we speculate that this miRNA may serve as a potential biomarker of frailty in diabetic older adults. Additionally, its actions on oxidative stress might represent a druggable target to obtain new potentials treatments.
Collapse
Affiliation(s)
- Pasquale Mone
- Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States
- ASL Avellino, Avellino, Italy
- *Correspondence: Pasquale Mone, ;,
| | | | - Fahimeh Varzideh
- Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States
| | - Urna Kansakar
- Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States
| | - Stanislovas S. Jankauskas
- Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States
| | | | - Gaetano Santulli
- Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States
- Department of Molecular Pharmacology, Einstein Institute for Aging Research, Einstein-Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
15
|
Yuan Q, Dridi H, Clarke OB, Reiken S, Melville Z, Wronska A, Kushnir A, Zalk R, Sittenfeld L, Marks AR. RyR1-related myopathy mutations in ATP and calcium binding sites impair channel regulation. Acta Neuropathol Commun 2021; 9:186. [PMID: 34809703 PMCID: PMC8609856 DOI: 10.1186/s40478-021-01287-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 10/31/2021] [Indexed: 11/10/2022] Open
Abstract
The type 1 ryanodine receptor (RyR1) is an intracellular calcium (Ca2+) release channel on the sarcoplasmic/endoplasmic reticulum that is required for skeletal muscle contraction. RyR1 channel activity is modulated by ligands, including the activators Ca2+ and ATP. Patients with inherited mutations in RyR1 may exhibit muscle weakness as part of a heterogeneous, complex disorder known as RYR1-related myopathy (RYR1-RM) or more recently termed RYR1-related disorders (RYR1-RD). Guided by high-resolution structures of skeletal muscle RyR1, obtained using cryogenic electron microscopy, we introduced mutations into putative Ca2+ and ATP binding sites and studied the function of the resulting mutant channels. These mutations confirmed the functional significance of the Ca2+ and ATP binding sites identified by structural studies based on the effects on channel regulation. Under normal conditions, Ca2+ activates RyR1 at low concentrations (µM) and inhibits it at high concentrations (mM). Mutations in the Ca2+-binding site impaired both activating and inhibitory regulation of the channel, suggesting a single site for both high and low affinity Ca2+-dependent regulation of RyR1 function. Mutation of residues that interact with the adenine ring of ATP abrogated ATP binding to the channel, whereas mutating residues that interact with the triphosphate tail only affected the degree of activation. In addition, patients with mutations at the Ca2+ or ATP binding sites suffer from muscle weakness, therefore impaired RyR1 channel regulation by either Ca2+ or ATP may contribute to the pathophysiology of RYR1-RM in some patients.
Collapse
|
16
|
Tang XH, Gambardella J, Jankauskas S, Wang X, Santulli G, Gudas LJ, Levi R. A Retinoic Acid Receptor β 2 Agonist Improves Cardiac Function in a Heart Failure Model. J Pharmacol Exp Ther 2021; 379:182-190. [PMID: 34389654 PMCID: PMC8626778 DOI: 10.1124/jpet.121.000806] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/10/2021] [Indexed: 12/22/2022] Open
Abstract
We previously demonstrated that the selective retinoic acid receptor (RAR) β 2 agonist AC261066 reduces oxidative stress in an ex vivo murine model of ischemia/reperfusion. We hypothesized that by decreasing oxidative stress and consequent fibrogenesis, AC261066 could attenuate the development of contractile dysfunction in post-ischemic heart failure (HF). We tested this hypothesis in vivo using an established murine model of myocardial infarction (MI), obtained by permanent occlusion of the left anterior descending coronary artery. Treating mice with AC261066 in drinking water significantly attenuated the post-MI deterioration of echocardiographic indices of cardiac function, diminished remodeling, and reduced oxidative stress, as evidenced by a decrease in malondialdehyde level and p38 mitogen-activated protein kinase expression in cardiomyocytes. The effects of AC261066 were also associated with a decrease in interstitial fibrosis, as shown by a marked reduction in collagen deposition and α-smooth muscle actin expression. In cardiac murine fibroblasts subjected to hypoxia, AC261066 reversed hypoxia-induced decreases in superoxide dismutase 2 and angiopoietin-like 4 transcriptional levels as well as the increase in NADPH oxidase 2 mRNA, demonstrating that the post-MI cardioprotective effects of AC261066 are associated with an action at the fibroblast level. Thus, AC261066 alleviates post-MI cardiac dysfunction by modulating a set of genes involved in the oxidant/antioxidant balance. These AC261066 responsive genes diminish interstitial fibrogenesis and remodeling. Since MI is a recognized major cause of HF, our data identify RARβ 2 as a potential pharmacological target in the treatment of HF. SIGNIFICANCE STATEMENT: A previous report showed that the selective retinoic acid receptor (RAR) β 2 agonist AC261066 reduces oxidative stress in an ex vivo murine model of ischemia/reperfusion. This study shows that AC261066 attenuates the development of contractile dysfunction and maladaptive remodeling in post-ischemic heart failure (HF) by modulating a set of genes involved in oxidant/antioxidant balance. Since myocardial infarction is a recognized major cause of HF, these data identify RARβ 2 as a potential pharmacological target in the treatment of HF.
Collapse
Affiliation(s)
- Xiao-Han Tang
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| | - Jessica Gambardella
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| | - Stanislovas Jankauskas
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| | - Xujun Wang
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| | - Gaetano Santulli
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| | - Lorraine J Gudas
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| | - Roberto Levi
- Department of Pharmacology, Weill Cornell Medicine, New York, New York (X.-H.T., L.J.G., R.L.); Departments of Medicine (Cardiology) and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (J.G., S.J., X.W., G.S.)
| |
Collapse
|
17
|
Di Fonso A, Pietrangelo L, D’Onofrio L, Michelucci A, Boncompagni S, Protasi F. Ageing Causes Ultrastructural Modification to Calcium Release Units and Mitochondria in Cardiomyocytes. Int J Mol Sci 2021; 22:8364. [PMID: 34445071 PMCID: PMC8395047 DOI: 10.3390/ijms22168364] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/20/2021] [Accepted: 07/28/2021] [Indexed: 12/12/2022] Open
Abstract
Ageing is associated with an increase in the incidence of heart failure, even if the existence of a real age-related cardiomyopathy remains controversial. Effective contraction and relaxation of cardiomyocytes depend on efficient production of ATP (handled by mitochondria) and on proper Ca2+ supply to myofibrils during excitation-contraction (EC) coupling (handled by Ca2+ release units, CRUs). Here, we analyzed mitochondria and CRUs in hearts of adult (4 months old) and aged (≥24 months old) mice. Analysis by confocal and electron microscopy (CM and EM, respectively) revealed an age-related loss of proper organization and disposition of both mitochondria and EC coupling units: (a) mitochondria are improperly disposed and often damaged (percentage of severely damaged mitochondria: adults 3.5 ± 1.1%; aged 16.5 ± 3.5%); (b) CRUs that are often misoriented (longitudinal) and/or misplaced from the correct position at the Z line. Immunolabeling with antibodies that mark either the SR or T-tubules indicates that in aged cardiomyocytes the sarcotubular system displays an extensive disarray. This disarray could be in part caused by the decreased expression of Cav-3 and JP-2 detected by western blot (WB), two proteins involved in formation of T-tubules and in docking SR to T-tubules in dyads. By WB analysis, we also detected increased levels of 3-NT in whole hearts homogenates of aged mice, a product of nitration of protein tyrosine residues, recognized as marker of oxidative stress. Finally, a detailed EM analysis of CRUs (formed by association of SR with T-tubules) points to ultrastructural modifications, i.e., a decrease in their frequency (adult: 5.1 ± 0.5; aged: 3.9 ± 0.4 n./50 μm2) and size (adult: 362 ± 40 nm; aged: 254 ± 60 nm). The changes in morphology and disposition of mitochondria and CRUs highlighted by our results may underlie an inefficient supply of Ca2+ ions and ATP to the contractile elements, and possibly contribute to cardiac dysfunction in ageing.
Collapse
Affiliation(s)
- Alessia Di Fonso
- CAST, Center for Advanced Studies and Technology, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy; (A.D.F.); (A.M.); (S.B.); (F.P.)
- DMSI, Department of Medicine and Aging Sciences, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy
| | - Laura Pietrangelo
- CAST, Center for Advanced Studies and Technology, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy; (A.D.F.); (A.M.); (S.B.); (F.P.)
- DMSI, Department of Medicine and Aging Sciences, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy
| | - Laura D’Onofrio
- IZSAM, Istituto Zooprofilattico Sperimentale dell’Abruzzo e del Molise G. Caporale of Teramo, 64100 Teramo, Italy;
| | - Antonio Michelucci
- CAST, Center for Advanced Studies and Technology, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy; (A.D.F.); (A.M.); (S.B.); (F.P.)
| | - Simona Boncompagni
- CAST, Center for Advanced Studies and Technology, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy; (A.D.F.); (A.M.); (S.B.); (F.P.)
- DNICS, Department of Neuroscience, Imaging and Clinical Sciences, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy
| | - Feliciano Protasi
- CAST, Center for Advanced Studies and Technology, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy; (A.D.F.); (A.M.); (S.B.); (F.P.)
- DMSI, Department of Medicine and Aging Sciences, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
18
|
Abstract
Ion exchange between intracellular and extracellular spaces is the basic mechanism for controlling cell metabolism and signal transduction. This process is mediated by ion channels and transporters on the plasma membrane, or intracellular membranes that surround various organelles, in response to environmental stimuli. Macroautophagy (hereafter referred to as autophagy) is one of the lysosomal-dependent degradation pathways that maintains homeostasis through the degradation and recycling of cellular components (e.g., dysfunctional proteins and damaged organelles). Although autophagy-related (ATG) proteins play a central role in regulating the formation of autophagy-related member structures (e.g., phagophores, autophagosomes, and autolysosomes), the autophagic process also involves changes in expression and function of ion channels and transporters. Here we discuss current knowledge of the mechanisms that regulate autophagy in mammalian cells, with special attention to the ion channels and transporters. We also highlight prospects for the development of drugs targeting ion channels and transporters in autophagy.
Collapse
Affiliation(s)
- Ruoxi Zhang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
19
|
Matarese A, Gambardella J, Sardu C, Santulli G. miR-98 Regulates TMPRSS2 Expression in Human Endothelial Cells: Key Implications for COVID-19. Biomedicines 2020; 8:462. [PMID: 33143053 PMCID: PMC7693865 DOI: 10.3390/biomedicines8110462] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 10/25/2020] [Accepted: 10/29/2020] [Indexed: 12/11/2022] Open
Abstract
The two main co-factors needed by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to enter human cells are angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2). Here, we focused on the study of microRNAs that specifically target TMPRSS2. Through a bioinformatic approach, we identified miR-98-5p as a suitable candidate. Since we and others have shown that endothelial cells play a pivotal role in the pathogenesis of the coronavirus disease 2019 (COVID-19), we mechanistically validated miR-98-5p as a regulator of TMPRSS2 transcription in two different human endothelial cell types, derived from the lung and from the umbilical vein. Taken together, our findings indicate that TMPRSS2 represents a valid target in COVID-19 treatment, which may be achieved by specific non-coding-RNA approaches.
Collapse
Affiliation(s)
- Alessandro Matarese
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (A.M.); (J.G.)
- AORN “Antonio Cardarelli”, 80100 Naples, Italy
| | - Jessica Gambardella
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (A.M.); (J.G.)
- Department of Advanced Biomedical Science, “Federico II” University, and International Translational Research and Medical Education Consortium (ITME), 80131 Naples, Italy
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
- Department of Medical Sciences, International University of Health and Medical Sciences “S. Camillo”, 00131 Rome, Italy
| | - Gaetano Santulli
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; (A.M.); (J.G.)
- Department of Advanced Biomedical Science, “Federico II” University, and International Translational Research and Medical Education Consortium (ITME), 80131 Naples, Italy
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
20
|
RyR1-targeted drug discovery pipeline integrating FRET-based high-throughput screening and human myofiber dynamic Ca 2+ assays. Sci Rep 2020; 10:1791. [PMID: 32019969 PMCID: PMC7000700 DOI: 10.1038/s41598-020-58461-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/13/2020] [Indexed: 11/18/2022] Open
Abstract
Elevated cytoplasmic [Ca2+] is characteristic in severe skeletal and cardiac myopathies, diabetes, and neurodegeneration, and partly results from increased Ca2+ leak from sarcoplasmic reticulum stores via dysregulated ryanodine receptor (RyR) channels. Consequently, RyR is recognized as a high-value target for drug discovery to treat such pathologies. Using a FRET-based high-throughput screening assay that we previously reported, we identified small-molecule compounds that modulate the skeletal muscle channel isoform (RyR1) interaction with calmodulin and FK506 binding protein 12.6. Two such compounds, chloroxine and myricetin, increase FRET and inhibit [3H]ryanodine binding to RyR1 at nanomolar Ca2+. Both compounds also decrease RyR1 Ca2+ leak in human skinned skeletal muscle fibers. Furthermore, we identified compound concentrations that reduced leak by > 50% but only slightly affected Ca2+ release in excitation-contraction coupling, which is essential for normal muscle contraction. This report demonstrates a pipeline that effectively filters small-molecule RyR1 modulators towards clinical relevance.
Collapse
|
21
|
The Interplay between Ca 2+ Signaling Pathways and Neurodegeneration. Int J Mol Sci 2019; 20:ijms20236004. [PMID: 31795242 PMCID: PMC6928941 DOI: 10.3390/ijms20236004] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/18/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022] Open
Abstract
Calcium (Ca2+) homeostasis is essential for cell maintenance since this ion participates in many physiological processes. For example, the spatial and temporal organization of Ca2+ signaling in the central nervous system is fundamental for neurotransmission, where local changes in cytosolic Ca2+ concentration are needed to transmit information from neuron to neuron, between neurons and glia, and even regulating local blood flow according to the required activity. However, under pathological conditions, Ca2+ homeostasis is altered, with increased cytoplasmic Ca2+ concentrations leading to the activation of proteases, lipases, and nucleases. This review aimed to highlight the role of Ca2+ signaling in neurodegenerative disease-related apoptosis, where the regulation of intracellular Ca2+ homeostasis depends on coordinated interactions between the endoplasmic reticulum, mitochondria, and lysosomes, as well as specific transport mechanisms. In neurodegenerative diseases, alterations-increased oxidative stress, energy metabolism alterations, and protein aggregation have been identified. The aggregation of α-synuclein, β-amyloid peptide (Aβ), and huntingtin all adversely affect Ca2+ homeostasis. Due to the mounting evidence for the relevance of Ca2+ signaling in neuroprotection, we would focus on the expression and function of Ca2+ signaling-related proteins, in terms of the effects on autophagy regulation and the onset and progression of neurodegenerative diseases.
Collapse
|
22
|
Sheng Z, Xu Y, Li F, Wang S, Huang T, Lu P. CSN5 attenuates Ang II-induced cardiac hypertrophy through stabilizing LKB1. Exp Cell Res 2019; 376:11-17. [PMID: 30710502 DOI: 10.1016/j.yexcr.2019.01.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/07/2019] [Accepted: 01/29/2019] [Indexed: 12/18/2022]
Abstract
CSN5 is a critical subunit of the COP9 signalosome (CSN) and has been involved in various cellular processes, but little is known about the role of CSN5 in cardiac disease. In the present study, we found that the expression of CSN5 was increased in Angiotensin II (Ang II)-induced cardiac hypertrophic mice hearts and Ang II-treated cardiomyocytes. We also observed that overexpression of CSN5 significantly inhibited Ang II-induced cardiac hypertrophy, whereas CSN5 silence exhibited the opposite phenotypes. Further investigation demonstrated that CSN5 maintained the activity of AMP-activated protein kinase (AMPK) in cardiomyocyte by enhancement of LKB1. Mechanistically, we found that CSN5 directly interacted and deubiquitinated LKB1 for its stabilization in cardiomyocytes. Finally, our results demonstrated that the anti-hypertrophic effect of CSN5 was partially dependent on stabilization of LKB1. Collectively, these findings suggested that strategies based on activation of CSN5/LKB1 axis might be promising in the treatment of hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Zhiyong Sheng
- Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Yun Xu
- Department of emergency, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Fuxin Li
- Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Shu Wang
- Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Tieqiu Huang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Peng Lu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
23
|
Li Z, Zhang X, Guo Z, Zhong Y, Wang P, Li J, Li Z, Liu P. SIRT6 Suppresses NFATc4 Expression and Activation in Cardiomyocyte Hypertrophy. Front Pharmacol 2019; 9:1519. [PMID: 30670969 PMCID: PMC6331469 DOI: 10.3389/fphar.2018.01519] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 12/11/2018] [Indexed: 12/15/2022] Open
Abstract
NFATc4, a member from the Nuclear Factor of Activated T cells (NFATs) transcription factor family, plays a pivotal role in the development of cardiac hypertrophy. NFATc4 is dephosphorylated by calcineurin and translocated from the cytoplasm to the nucleus to regulate the expression of hypertrophic genes, like brain natriuretic polypeptide (BNP). The present study identified SIRT6, an important subtype of NAD+ dependent class III histone deacetylase, to be a negative regulator of NFATc4 in cardiomyocyte hypertrophy. In phenylephrine (PE)-induced hypertrophic cardiomyocyte model, overexpression of SIRT6 by adenovirus infection or by plasmid transfection repressed the protein and mRNA expressions of NFATc4, elevated its phosphorylation level, prevented its nuclear accumulation, subsequently suppressed its transcriptional activity and downregulated its target gene BNP. By contrast, mutant of SIRT6 without deacetylase activity (H133Y) did not demonstrate these effects, suggesting that the inhibitory effect of SIRT6 on NFATc4 was dependent on its deacetylase activity. Moreover, the effect of SIRT6 overexpression on repressing BNP expression was reversed by NFATc4 replenishment, whereas the effect of SIRT6 deficiency on upregulating BNP was recovered by NFATc4 silencing. Mechanistically, interactions between SIRT6 and NFATc4 might possibly facilitate the deacetylation of NFATc4 by SIRT6, thereby preventing the activation of NFATc4. In conclusion, the present study reveals that SIRT6 suppresses the expression and activation of NFATc4. These findings provide more evidences of the anti-hypertrophic effect of SIRT6 and suggest SIRT6 as a potential therapeutic target for cardiac hypertrophy.
Collapse
Affiliation(s)
- Zhenzhen Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiaoying Zhang
- Department of Pharmacology, School of Medicine, Xizang Minzu University, Shaanxi, China
| | - Zhen Guo
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yao Zhong
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,Department of Cardiology, Third People's Hospital of Dongguan, Dongguan, China
| | - Panxia Wang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jingyan Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhuoming Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Peiqing Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
24
|
Chen H, Ji Y, Yan X, Su G, Chen L, Xiao J. Berberine attenuates apoptosis in rat retinal Müller cells stimulated with high glucose via enhancing autophagy and the AMPK/mTOR signaling. Biomed Pharmacother 2018; 108:1201-1207. [PMID: 30372821 DOI: 10.1016/j.biopha.2018.09.140] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 09/26/2018] [Accepted: 09/26/2018] [Indexed: 11/30/2022] Open
Abstract
Berberine (BBR) has beneficial effects on diabetes and the multiple complications of diabetes due to its anti-apoptotic activity; however, the effect of BBR on diabetic retinopathy and its mechanism of action have not been clarified. The present study investigated the effect of BBR on Müller cells stimulated with high glucose (HG). Primary retinal Müller cells were incubated with high glucose to induce cell apoptosis; cells were pretreated with the AMPK inhibitor compound C and the AMPK activator AICAR to further explore the role of the AMPK/mTOR signaling pathway in the anti-apoptotic action of BBR. Immunofluorescence was used to measure apoptosis and autophagy. Western blot analysis was employed to determine the levels of p-AMPK and p-mTOR, as well as apoptosis-related proteins and autophagy-related proteins in Müller cells. Our results showed that BBR attenuated apoptosis, up regulated Bcl-2 and down regulated Bax and caspase-3 expression; enhanced the formation of autophagy, elevated the expression of Beclin-1 and LC3II and activated the AMPK/mTOR signaling pathway in Müller cells under high glucose conditions compared with the control group. The effect of BBR was partly blocked by compound C and strengthened by AICAR. BBR may have therapeutic potential to protect Müller cells from high-glucose-inducing apoptosis through enhancing autophagy and activating the AMPK/mTOR signaling pathway.
Collapse
Affiliation(s)
- Han Chen
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Yingshi Ji
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Xin Yan
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; General Hospital of Fushun Mining Bureau, Fushun, 113008, China
| | - Guanfang Su
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Li Chen
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; School of Nursing, Jilin University, Changchun, 130021, China
| | - Jun Xiao
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China.
| |
Collapse
|
25
|
An Intervention Target for Myocardial Fibrosis: Autophagy. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6215916. [PMID: 29850542 PMCID: PMC5911341 DOI: 10.1155/2018/6215916] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 02/07/2018] [Accepted: 02/19/2018] [Indexed: 02/07/2023]
Abstract
Myocardial fibrosis (MF) is the result of metabolic imbalance of collagen synthesis and metabolism, which is widespread in various cardiovascular diseases. Autophagy is a lysosomal degradation pathway which is highly conserved. In recent years, research on autophagy has been increasing and the researchers have also become cumulatively aware of the specified association between autophagy and MF. This review highlights the role of autophagy in MF and the potential effects through the administration of medicine.
Collapse
|
26
|
Gambardella J, Trimarco B, Iaccarino G, Santulli G. New Insights in Cardiac Calcium Handling and Excitation-Contraction Coupling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1067:373-385. [PMID: 28956314 PMCID: PMC5889357 DOI: 10.1007/5584_2017_106] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Excitation-contraction (EC) coupling denotes the conversion of electric stimulus in mechanic output in contractile cells. Several studies have demonstrated that calcium (Ca2+) plays a pivotal role in this process. Here we present a comprehensive and updated description of the main systems involved in cardiac Ca2+ handling that ensure a functional EC coupling and their pathological alterations, mainly related to heart failure.
Collapse
Affiliation(s)
- Jessica Gambardella
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
- Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, Fisciano, Italy
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Guido Iaccarino
- Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, Fisciano, Italy
| | - Gaetano Santulli
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy.
- Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave, Forch 525, 10461, New York, NY, USA.
| |
Collapse
|
27
|
Santulli G, Lewis D, des Georges A, Marks AR, Frank J. Ryanodine Receptor Structure and Function in Health and Disease. Subcell Biochem 2018; 87:329-352. [PMID: 29464565 PMCID: PMC5936639 DOI: 10.1007/978-981-10-7757-9_11] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ryanodine receptors (RyRs) are ubiquitous intracellular calcium (Ca2+) release channels required for the function of many organs including heart and skeletal muscle, synaptic transmission in the brain, pancreatic beta cell function, and vascular tone. In disease, defective function of RyRs due either to stress (hyperadrenergic and/or oxidative overload) or genetic mutations can render the channels leaky to Ca2+ and promote defective disease-causing signals as observed in heat failure, muscular dystrophy, diabetes mellitus, and neurodegerative disease. RyRs are massive structures comprising the largest known ion channel-bearing macromolecular complex and exceeding 3 million Daltons in molecular weight. RyRs mediate the rapid release of Ca2+ from the endoplasmic/sarcoplasmic reticulum (ER/SR) to stimulate cellular functions through Ca2+-dependent processes. Recent advances in single-particle cryogenic electron microscopy (cryo-EM) have enabled the determination of atomic-level structures for RyR for the first time. These structures have illuminated the mechanisms by which these critical ion channels function and interact with regulatory ligands. In the present chapter we discuss the structure, functional elements, gating and activation mechanisms of RyRs in normal and disease states.
Collapse
Affiliation(s)
- Gaetano Santulli
- The Wu Center for Molecular Cardiology, Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY, USA
- The Wilf Family Cardiovascular Research Institute and the Einstein-Mount Sinai Diabetes Research Center, Department of Medicine, Albert Einstein College of Medicine - Montefiore University Hospital, New York, NY, USA
| | - Daniel Lewis
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Amedee des Georges
- Advanced Science Research Center at the Graduate Center of the City University of New York, New York, NY, USA
- Department of Chemistry & Biochemistry, City College of New York, New York, NY, USA
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY, USA
| | - Andrew R Marks
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
- Department of Medicine, Columbia University, New York, NY, USA
| | - Joachim Frank
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA.
- Department of Biological Sciences, Columbia University, New York, NY, USA.
| |
Collapse
|
28
|
Kondratskyi A, Kondratska K, Skryma R, Klionsky DJ, Prevarskaya N. Ion channels in the regulation of autophagy. Autophagy 2017; 14:3-21. [PMID: 28980859 PMCID: PMC5846505 DOI: 10.1080/15548627.2017.1384887] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 09/07/2017] [Accepted: 09/21/2017] [Indexed: 12/18/2022] Open
Abstract
Autophagy is a cellular process in which the cell degrades and recycles its own constituents. Given the crucial role of autophagy in physiology, deregulation of autophagic machinery is associated with various diseases. Hence, a thorough understanding of autophagy regulatory mechanisms is crucially important for the elaboration of efficient treatments for different diseases. Recently, ion channels, mediating ion fluxes across cellular membranes, have emerged as important regulators of both basal and induced autophagy. However, the mechanisms by which specific ion channels regulate autophagy are still poorly understood, thus underscoring the need for further research in this field. Here we discuss the involvement of major types of ion channels in autophagy regulation.
Collapse
Affiliation(s)
- Artem Kondratskyi
- Inserm, U-1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, University of Lille 1, Villeneuve d'Ascq, France
| | - Kateryna Kondratska
- Inserm, U-1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, University of Lille 1, Villeneuve d'Ascq, France
| | - Roman Skryma
- Inserm, U-1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, University of Lille 1, Villeneuve d'Ascq, France
| | - Daniel J. Klionsky
- Life Sciences Institute, and Department of Molecular, Cellular and Developmental Biology; University of Michigan, Ann Arbor, MI, USA
| | - Natalia Prevarskaya
- Inserm, U-1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, University of Lille 1, Villeneuve d'Ascq, France
| |
Collapse
|
29
|
Li S, Liu C, Gu L, Wang L, Shang Y, Liu Q, Wan J, Shi J, Wang F, Xu Z, Ji G, Li W. Autophagy protects cardiomyocytes from the myocardial ischaemia-reperfusion injury through the clearance of CLP36. Open Biol 2017; 6:rsob.160177. [PMID: 27512143 PMCID: PMC5008017 DOI: 10.1098/rsob.160177] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 07/08/2016] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of the death worldwide. An increasing number of studies have found that autophagy is involved in the progression or prevention of CVD. However, the precise mechanism of autophagy in CVD, especially the myocardial ischaemia-reperfusion injury (MI/R injury), is unclear and controversial. Here, we show that the cardiomyocyte-specific disruption of autophagy by conditional knockout of Atg7 leads to severe contractile dysfunction, myofibrillar disarray and vacuolar cardiomyocytes. A negative cytoskeleton organization regulator, CLP36, was found to be accumulated in Atg7-deficient cardiomyocytes. The cardiomyocyte-specific knockout of Atg7 aggravates the MI/R injury with cardiac hypertrophy, contractile dysfunction, myofibrillar disarray and severe cardiac fibrosis, most probably due to CLP36 accumulation in cardiomyocytes. Altogether, this work reveals autophagy may protect cardiomyocytes from the MI/R injury through the clearance of CLP36, and these findings define a novel relationship between autophagy and the regulation of stress fibre in heart.
Collapse
Affiliation(s)
- Shiguo Li
- Department of Radiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Chao Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Lei Gu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Lina Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Yongliang Shang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Qiong Liu
- Department of Radiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Junyi Wan
- Department of Radiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Jian Shi
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Fang Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Zhiliang Xu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Guangju Ji
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
30
|
Lombardi A, Trimarco B, Iaccarino G, Santulli G. Impaired mitochondrial calcium uptake caused by tacrolimus underlies beta-cell failure. Cell Commun Signal 2017; 15:47. [PMID: 29132395 PMCID: PMC5684747 DOI: 10.1186/s12964-017-0203-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 11/02/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND One of the most common side effects of the immunosuppressive drug tacrolimus (FK506) is the increased risk of new-onset diabetes mellitus. However, the molecular mechanisms underlying this association have not been fully clarified. METHODS We studied the effects of the therapeutic dose of tacrolimus on mitochondrial fitness in beta-cells. RESULTS We demonstrate that tacrolimus impairs glucose-stimulated insulin secretion (GSIS) in beta-cells through a previously unidentified mechanism. Indeed, tacrolimus causes a decrease in mitochondrial Ca2+ uptake, accompanied by altered mitochondrial respiration and reduced ATP production, eventually leading to impaired GSIS. CONCLUSION Our observations individuate a new fundamental mechanism responsible for the augmented incidence of diabetes following tacrolimus treatment. Indeed, this drug alters Ca2+ fluxes in mitochondria, thereby compromising metabolism-secretion coupling in beta-cells.
Collapse
Affiliation(s)
- Angela Lombardi
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, "Federico II" University of Naples, Naples, Italy
| | - Guido Iaccarino
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana", University of Salerno, Fisciano, Italy
| | - Gaetano Santulli
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA.
- Department of Advanced Biomedical Sciences, "Federico II" University of Naples, Naples, Italy.
| |
Collapse
|
31
|
Zhao YT, Guo YB, Gu L, Fan XX, Yang HQ, Chen Z, Zhou P, Yuan Q, Ji GJ, Wang SQ. Sensitized signalling between L-type Ca2+ channels and ryanodine receptors in the absence or inhibition of FKBP12.6 in cardiomyocytes. Cardiovasc Res 2017; 113:332-342. [PMID: 28077437 DOI: 10.1093/cvr/cvw247] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Accepted: 12/03/2016] [Indexed: 12/19/2022] Open
Abstract
Aims The heart contraction is controlled by the Ca2+-induced Ca2+ release (CICR) between L-type Ca2+ channels and ryanodine receptors (RyRs). The FK506-binding protein FKBP12.6 binds to RyR subunits, but its role in stabilizing RyR function has been debated for long. Recent reports of high-resolution RyR structure show that the HD2 domain that binds to the SPRY2 domain of neighbouring subunit in FKBP-bound RyR1 is detached and invisible in FKBP-null RyR2. The present study was to test the consequence of FKBP12.6 absence on the in situ activation of RyR2. Methods and results Using whole-cell patch-clamp combined with confocal imaging, we applied a near threshold depolarization to activate a very small fraction of LCCs, which in turn activated RyR Ca2+ sparks stochastically. FKBP12.6-knockout and FK506/rapamycin treatments increased spark frequency and LCC-RyR coupling fidelity without altering LCC open probability. Neither FK506 nor rapamycin further altered LCC-RyR coupling fidelity in FKBP12.6-knockout cells. In loose-seal patch-clamp experiments, the LCC-RyR signalling kinetics, indexed by the delay for a LCC sparklet to trigger a RyR spark, was accelerated after FKBP12.6 knockout and FK506/rapamycin treatments. These results demonstrated that RyRs became more sensitive to Ca2+ triggers without FKBP12.6. Isoproterenol (1 μM) further accelerated the LCC-RyR signalling in FKBP12.6-knockout cells. The synergistic sensitization of RyRs by catecholaminergic signalling and FKBP12.6 dysfunction destabilized the CICR system, leading to chaotic Ca2+ waves and ventricular arrhythmias. Conclusion FKBP12.6 keeps the RyRs from over-sensitization, stabilizes the potentially regenerative CICR system, and thus may suppress the life-threatening arrhythmogenesis.
Collapse
Affiliation(s)
- Yan-Ting Zhao
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, 5 Yiheyuan Road, Beijing 100871, China
| | - Yun-Bo Guo
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, 5 Yiheyuan Road, Beijing 100871, China
| | - Lei Gu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China
| | - Xue-Xin Fan
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, 5 Yiheyuan Road, Beijing 100871, China
| | - Hua-Qian Yang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, 5 Yiheyuan Road, Beijing 100871, China
| | - Zheng Chen
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China
| | - Peng Zhou
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, 5 Yiheyuan Road, Beijing 100871, China
| | - Qi Yuan
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China
| | - Guang-Ju Ji
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China
| | - Shi-Qiang Wang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, 5 Yiheyuan Road, Beijing 100871, China
| |
Collapse
|
32
|
Lu L, Guo J, Hua Y, Huang K, Magaye R, Cornell J, Kelly DJ, Reid C, Liew D, Zhou Y, Chen A, Xiao W, Fu Q, Wang BH. Cardiac fibrosis in the ageing heart: Contributors and mechanisms. Clin Exp Pharmacol Physiol 2017; 44 Suppl 1:55-63. [PMID: 28316086 DOI: 10.1111/1440-1681.12753] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 03/09/2017] [Accepted: 03/12/2017] [Indexed: 01/30/2023]
Abstract
Cardiac fibrosis refers to an excessive deposition of extracellular matrix (ECM) in cardiac tissue. Fibrotic tissue is stiffer and less compliant, resulting in subsequent cardiac dysfunction and heart failure. Cardiac fibrosis in the ageing heart may involve activation of fibrogenic signalling and inhibition of anti-fibrotic signalling, leading to an imbalance of ECM turnover. Excessive accumulation of ECM such as collagen in older patients contributes to progressive ventricular dysfunction. Overexpression of collagen is derived from various sources, including higher levels of fibrogenic growth factors, proliferation of fibroblasts and cellular transdifferentiation. These may be triggered by factors, such as oxidative stress, inflammation, hypertension, cellular senescence and cell death, contributing to age-related fibrotic cardiac remodelling. In this review, we will discuss the fibrogenic contributors in age-related cardiac fibrosis, and the potential mechanisms by which fibrogenic processes can be interrupted for therapeutic intent.
Collapse
Affiliation(s)
- Lu Lu
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Vic., Australia.,School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jingbin Guo
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Vic., Australia.,Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yue Hua
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Vic., Australia.,School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Kevin Huang
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Vic., Australia
| | - Ruth Magaye
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Vic., Australia
| | - Jake Cornell
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Vic., Australia
| | - Darren J Kelly
- Department of Medicine, St Vincent's Hospital, University of Melbourne, Melbourne, Vic., Australia
| | - Christopher Reid
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Vic., Australia.,NHMRC Cardiovascular Centre of Research Excellence, School of Public Health, Curtin University, Perth, WA, Australia
| | - Danny Liew
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Vic., Australia
| | - Yingchun Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Aihua Chen
- Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Xiao
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Vic., Australia
| | - Qiang Fu
- Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Bing Hui Wang
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Vic., Australia
| |
Collapse
|
33
|
Santulli G, Nakashima R, Yuan Q, Marks AR. Intracellular calcium release channels: an update. J Physiol 2017; 595:3041-3051. [PMID: 28303572 PMCID: PMC5430224 DOI: 10.1113/jp272781] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 02/20/2017] [Indexed: 12/19/2022] Open
Abstract
Ryanodine receptors (RyRs) and inositol 1,4,5-trisphosphate receptors (IP3 Rs) are calcium (Ca2+ ) release channels on the endo/sarcoplasmic reticulum (ER/SR). Here we summarize the latest advances in the field, describing the recently discovered mechanistic roles of intracellular Ca2+ release channels in the regulation of mitochondrial fitness and endothelial function, providing novel therapeutic options for the treatment of heart failure, hypertension, and diabetes mellitus.
Collapse
Affiliation(s)
- Gaetano Santulli
- The Wu Center for Molecular CardiologyColumbia UniversityNew YorkNYUSA
- Department of Physiology and Cellular BiophysicsCollege of Physicians and SurgeonsColumbia University Medical CenterNew YorkNYUSA
| | - Ryutaro Nakashima
- The Wu Center for Molecular CardiologyColumbia UniversityNew YorkNYUSA
- Department of Physiology and Cellular BiophysicsCollege of Physicians and SurgeonsColumbia University Medical CenterNew YorkNYUSA
| | - Qi Yuan
- The Wu Center for Molecular CardiologyColumbia UniversityNew YorkNYUSA
- Department of Physiology and Cellular BiophysicsCollege of Physicians and SurgeonsColumbia University Medical CenterNew YorkNYUSA
| | - Andrew R. Marks
- The Wu Center for Molecular CardiologyColumbia UniversityNew YorkNYUSA
- Department of Physiology and Cellular BiophysicsCollege of Physicians and SurgeonsColumbia University Medical CenterNew YorkNYUSA
- Department of MedicineColumbia UniversityNew YorkNYUSA
| |
Collapse
|
34
|
Chang Y, Li Y, Guo X, Li T, Chen Y, Dai D, Sun Y. The association of ideal cardiovascular health and left ventricle hypertrophy in rural population of northeast China: A cross-sectional observational study. Medicine (Baltimore) 2017; 96:e6050. [PMID: 28178152 PMCID: PMC5313009 DOI: 10.1097/md.0000000000006050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
In 2010, the American Heart Association (AHA) published a new concept "ideal cardiovascular health" (CVH), which consisted of 4 behaviors (smoking, body mass index [BMI], physical activity, and diet score) and 3 health factors (total cholesterol [TC], blood pressure [BP], and fasting plasma glucose [FPG]). This study was aimed to investigate the association between CVH with left ventricle hypertrophy (LVH) in a rural general population.From January 2012 to August 2013, we conducted this cross-sectional study using a multi-stage cluster sampling method. A representative sample of individuals who were at 35 years or older was selected. All the 7 CVH metrics were estimated for ideal, intermediate, and poor levels. LVH was accessed by echocardiography and classified into concentric remodeling, concentric LVH, and eccentric LVH. The association between CVH and LVH was determined.The final data were obtained from 10,684 adults (5497 men and 5187 women) in the rural areas of northeast China. Overall, the prevalence rates of concentric remodeling, concentric LVH, and eccentric LVH were 5.1%, 4.9%, and 12.8%, respectively. The prevalence of concentric/eccentric LVH was inversely related to the numbers of ideal CVH metrics. Multivariate logistic regression analysis indicated that only poor BP was associated with concentric remodeling among the 7 CVH metrics; poor BP was highly associated with concentric LVH (OR: 8.49; 95% CI: 4.59-15.7); poor BMI was highly associated with eccentric LVH (OR: 5.87; 95% CI: 4.83-7.14). Compared to subjects with 5 to 7 ideal CVH metrics, subjects with 4, 3, 2, 1, and 0 ideal CVH metrics had an increased risk for both concentric and eccentric LVH in a number-dependent manner. The subjects with poor CVH status had a 5.90-fold higher risk of developing concentric LVH and a 3.24-fold higher risk of developing eccentric LVH, compared to subjects with ideal-intermediate CVH.Our study found that an inversely gradient relationship existed between the prevalence of concentric/eccentric LVH with the numbers of ideal CVH metrics. Although not all the 7 CVH metrics were associated with LVH, the components of CVH metrics carried a synergistic effect beyond the risk related to the component alone.
Collapse
Affiliation(s)
| | | | | | - Tan Li
- Department of Cardiovascular Ultrasound, the First Hospital of China Medical University, Shenyang, People's Republic of China
| | | | | | | |
Collapse
|
35
|
Santulli G, Lewis DR, Marks AR. Physiology and pathophysiology of excitation-contraction coupling: the functional role of ryanodine receptor. J Muscle Res Cell Motil 2017; 38:37-45. [PMID: 28653141 PMCID: PMC5813681 DOI: 10.1007/s10974-017-9470-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/06/2017] [Indexed: 12/21/2022]
Abstract
Calcium (Ca2+) release from intracellular stores plays a key role in the regulation of skeletal muscle contraction. The type 1 ryanodine receptors (RyR1) is the major Ca2+ release channel on the sarcoplasmic reticulum (SR) of myocytes in skeletal muscle and is required for excitation-contraction (E-C) coupling. This article explores the role of RyR1 in skeletal muscle physiology and pathophysiology.
Collapse
Affiliation(s)
- Gaetano Santulli
- The Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University Medical Center, Columbia University, New York, NY, USA
| | - Daniel R Lewis
- The Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University Medical Center, Columbia University, New York, NY, USA
| | - Andrew R Marks
- The Wu Center for Molecular Cardiology, Columbia University, New York, NY, USA.
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University Medical Center, Columbia University, New York, NY, USA.
- Department of Medicine, Columbia University, New York, NY, USA.
| |
Collapse
|
36
|
Abu-Omar N, Das J, Szeto V, Feng ZP. Neuronal Ryanodine Receptors in Development and Aging. Mol Neurobiol 2017; 55:1183-1192. [DOI: 10.1007/s12035-016-0375-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 12/28/2016] [Indexed: 01/09/2023]
|
37
|
Husser D, Ueberham L, Dinov B, Kosiuk J, Kornej J, Hindricks G, Shoemaker MB, Roden DM, Bollmann A, Büttner P. Genomic contributors to atrial electroanatomical remodeling and atrial fibrillation progression: Pathway enrichment analysis of GWAS data. Sci Rep 2016; 6:36630. [PMID: 27857207 PMCID: PMC5114680 DOI: 10.1038/srep36630] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 10/11/2016] [Indexed: 12/18/2022] Open
Abstract
In atrial fibrillation (AF), left atrial diameter (LAD) and low voltage area (LVA) are intermediate phenotypes that are associated with AF type and progression. In this study, we tested the hypothesis, that these phenotypes share common, genetically-determined pathways using pathway enrichment analysis of GWAS data. Samples from 660 patients with paroxysmal (n = 370) or persistent AF (n = 290) were genotyped for ~1,000,000 SNPs. SNPs found significantly associated with LAD, LVA or AF type were used for gene-based association tests in a systematic biological Knowledge-based mining system for Genome-wide Genetic studies (KGG). Associated genes were tested for pathway enrichment using two enrichment tools (WebGestalt and GATHER) and the databases provided by Kyoto Encyclopedia of Genes and Genomes. The calcium signaling pathway (hsa04020) was the only pathway that reached statistical significance for LAD and LVA in both enrichment tools and was also significantly associated with AF type. Within this pathway, there were 39 genes (i.e. CACNA1C, RyR2) that were associated with LAD, LVA and AF type. In conclusion, there is a genomic contribution to electroanatomical remodeling (LAD, LVA) and AF type via the calcium signaling pathway. Future and larger studies are necessary to replicate and apply these findings.
Collapse
Affiliation(s)
- Daniela Husser
- Department of Electrophysiology, Heart Center Leipzig, Leipzig University, Germany
| | - Laura Ueberham
- Department of Electrophysiology, Heart Center Leipzig, Leipzig University, Germany
| | - Borislav Dinov
- Department of Electrophysiology, Heart Center Leipzig, Leipzig University, Germany
| | - Jedrzej Kosiuk
- Department of Electrophysiology, Heart Center Leipzig, Leipzig University, Germany
| | - Jelena Kornej
- Department of Electrophysiology, Heart Center Leipzig, Leipzig University, Germany
| | - Gerhard Hindricks
- Department of Electrophysiology, Heart Center Leipzig, Leipzig University, Germany
| | | | - Dan M Roden
- Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Andreas Bollmann
- Department of Electrophysiology, Heart Center Leipzig, Leipzig University, Germany
| | - Petra Büttner
- Department of Electrophysiology, Heart Center Leipzig, Leipzig University, Germany
| |
Collapse
|
38
|
Santulli G, Iaccarino G. Adrenergic signaling in heart failure and cardiovascular aging. Maturitas 2016; 93:65-72. [PMID: 27062709 PMCID: PMC5036981 DOI: 10.1016/j.maturitas.2016.03.022] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 03/23/2016] [Accepted: 03/25/2016] [Indexed: 12/15/2022]
Abstract
Both cardiovascular disease and aging are associated with changes in the sympathetic nervous system. Indeed, mounting evidence indicates that adrenergic receptors are functionally involved in numerous processes underlying both aging and cardiovascular disorders, in particular heart failure. This article will review the pathophysiological role of the sympathetic nervous system in heart failure and cardiovascular aging.
Collapse
Affiliation(s)
- Gaetano Santulli
- College of Physicians & Surgeons, Columbia University Medical Center, New York, NY, USA.
| | - Guido Iaccarino
- Division of Internal Medicine, Department of Medicine and Surgery, University of Salerno, Italy.
| |
Collapse
|
39
|
Yuan Q, Yang J, Santulli G, Reiken SR, Wronska A, Kim MM, Osborne BW, Lacampagne A, Yin Y, Marks AR. Maintenance of normal blood pressure is dependent on IP3R1-mediated regulation of eNOS. Proc Natl Acad Sci U S A 2016; 113:8532-8537. [PMID: 27402766 PMCID: PMC4968706 DOI: 10.1073/pnas.1608859113] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Endothelial cells (ECs) are critical mediators of blood pressure (BP) regulation, primarily via the generation and release of vasorelaxants, including nitric oxide (NO). NO is produced in ECs by endothelial NO synthase (eNOS), which is activated by both calcium (Ca(2+))-dependent and independent pathways. Here, we report that intracellular Ca(2+) release from the endoplasmic reticulum (ER) via inositol 1,4,5-trisphosphate receptor (IP3R) is required for Ca(2+)-dependent eNOS activation. EC-specific type 1 1,4,5-trisphosphate receptor knockout (IP3R1(-/-)) mice are hypertensive and display blunted vasodilation in response to acetylcholine (ACh). Moreover, eNOS activity is reduced in both isolated IP3R1-deficient murine ECs and human ECs following IP3R1 knockdown. IP3R1 is upstream of calcineurin, a Ca(2+)/calmodulin-activated serine/threonine protein phosphatase. We show here that the calcineurin/nuclear factor of activated T cells (NFAT) pathway is less active and eNOS levels are decreased in IP3R1-deficient ECs. Furthermore, the calcineurin inhibitor cyclosporin A, whose use has been associated with the development of hypertension, reduces eNOS activity and vasodilation following ACh stimulation. Our results demonstrate that IP3R1 plays a crucial role in the EC-mediated vasorelaxation and the maintenance of normal BP.
Collapse
Affiliation(s)
- Qi Yuan
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032; Wu Center for Molecular Cardiology, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032
| | - Jingyi Yang
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032; Wu Center for Molecular Cardiology, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032; Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Gaetano Santulli
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032; Wu Center for Molecular Cardiology, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032;
| | - Steven R Reiken
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032; Wu Center for Molecular Cardiology, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032
| | - Anetta Wronska
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032; Wu Center for Molecular Cardiology, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032
| | - Mindy M Kim
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032
| | - Brent W Osborne
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032
| | - Alain Lacampagne
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032; INSERM U1046, CNRS UMR-9214, Université de Montpellier, 34295 Montpellier, France
| | - Yuxin Yin
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China;
| | - Andrew R Marks
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032; Wu Center for Molecular Cardiology, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032; Department of Medicine, Columbia University, New York, NY 10032
| |
Collapse
|
40
|
Lu J, Sun D, Liu Z, Li M, Hong H, Liu C, Gao S, Li H, Cai Y, Chen S, Li Z, Ye J, Liu P. SIRT6 suppresses isoproterenol-induced cardiac hypertrophy through activation of autophagy. Transl Res 2016; 172:96-112.e6. [PMID: 27016702 DOI: 10.1016/j.trsl.2016.03.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Revised: 01/25/2016] [Accepted: 03/02/2016] [Indexed: 11/25/2022]
Abstract
Reduction in autophagy has been reported to contribute to the pathogenesis of cardiac hypertrophy. However, the molecular pathways leading to impaired autophagy at the presence of hypertrophic stimuli remain to be elucidated. The present study aimed to investigate the role of sirtuin 6 (SIRT6), a sirtuin family member, in regulating cardiomyocyte autophagy, and its implication in prevention of cardiac hypertrophy. Primary neonatal rat cardiomyocytes (NRCMs) or Sprague-Dawley (SD) rats were submitted to isoproterenol (ISO) treatment, and then the hypertrophic responses and changes in autophagy activity were measured. The influence of SIRT6 on autophagy was observed in cultured NRCMs with gain- and loss-of-function approaches to regulate SIRT6 expression, and further confirmed in vivo by intramyocardial delivery of an adenovirus vector encoding SIRT6 cDNA. In addition, the involvement of SIRT6-mediated autophagy in attenuation of cardiomyocyte hypertrophy induced by ISO was determined basing on genetic or pharmaceutical disruption of autophagy, and the underlying mechanism was preliminarily explored. ISO-caused cardiac hypertrophy accompanying with a significant decrease in autophagy activity. SIRT6 overexpression enhanced autophagy in NRCMs and in rat hearts, whereas knockdown of SIRT6 by RNA interference led to suppression of cardiomyocyte autophagy. Furthermore, the protective effect of SIRT6 against ISO-stimulated hypertrophy was associated with induction of autophagy. SIRT6 promoted nuclear retention of forkhead box O3 transcription factor possibly via attenuating Akt signaling, which was responsible for autophagy activation. Our findings revealed that SIRT6 positively regulates autophagy in cardiomyocytes, which may help to ameliorate ISO-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- Jing Lu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Duanping Sun
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Zhiping Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Min Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Huiqi Hong
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Cui Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Si Gao
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Hong Li
- Department of Biochemistry and Molecular Biology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Yi Cai
- Guangzhou Research Institute of Snake Venom, Guangzhou Medical College, Guangzhou, Guangdong, P.R. China
| | - Shaorui Chen
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Zhuoming Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Jiantao Ye
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China.
| | - Peiqing Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China.
| |
Collapse
|
41
|
Wang J, Tan X, Yang Q, Zeng X, Zhou Y, Luo W, Lin X, Song L, Cai J, Wang T, Wu X. Inhibition of autophagy promotes apoptosis and enhances anticancer efficacy of adriamycin via augmented ROS generation in prostate cancer cells. Int J Biochem Cell Biol 2016; 77:80-90. [PMID: 27247025 DOI: 10.1016/j.biocel.2016.05.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 05/07/2016] [Accepted: 05/27/2016] [Indexed: 12/14/2022]
Abstract
The interplay between autophagy and apoptosis response to chemotherapy is still a subject of intense debate in recent years. More efforts have focused on the regulation effects of apoptosis on autophagy, whereas how autophagy affects apoptosis remains poorly understood. In this study performed on prostate cancer cells, we investigated the role of autophagy in adriamycin-induced apoptosis, as well as the mechanisms mediating the effects of autophagy on apoptosis response to adriamycin (ADM). The results show that ADM not only inhibited cell viability and enhanced apoptosis, but also promoted autophagy via PI3K/Akt(T308)/mTOR signal pathway. Inhibition of autophagy by either pharmacological inhibitor chloroquine (CQ) or RNA interference of Atg5 increased ADM-induced apoptosis and enhanced the chemosensitivity of prostate cancer cells. Moreover, blockade of autophagy augmented reactive oxygen species (ROS) generation induced by ADM. Scavenging of ROS by antioxidant N-acetyl-cysteine (NAC) reversed the strengthened effects of CQ on ADM-induced apoptosis and rescued the cells from apoptosis. The results identified ROS as a potential mediator directing the modulation effects of the protective autophagy on apoptosis response to ADM. Suppression of the protective autophagy might provide a promising strategy to increase the anticancer efficacy of agents in the treatment of prostate cancer.
Collapse
Affiliation(s)
- Jizhong Wang
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou 510632, China
| | - Xiangpeng Tan
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou 510632, China
| | - Qi Yang
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou 510632, China
| | - Xiangfeng Zeng
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou 510632, China
| | - Yuying Zhou
- Department of Cell biology, Jinan University, Guangzhou 510632, China
| | - Wu Luo
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou 510632, China
| | - Xiaomian Lin
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou 510632, China
| | - Li Song
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou 510632, China
| | - Jialong Cai
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou 510632, China
| | - Tianxiang Wang
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou 510632, China
| | - Xiaoping Wu
- Institute of Tissue Transplantation and Immunology, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, and Key Laboratory of Molecule Immunology and Antibody Engineering of Guangdong Province, Jinan University, Guangzhou 510632, China; School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
42
|
Exchange of chemical signals between cardiac cells. Fundamental role on cell communication and metabolic cooperation. Exp Cell Res 2016; 346:130-6. [PMID: 27237090 DOI: 10.1016/j.yexcr.2016.05.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 05/09/2016] [Accepted: 05/10/2016] [Indexed: 01/14/2023]
Abstract
The exchange of chemical signals between cardiac cells and its relevance for cell communication and metabolic cooperation was reviewed. The role of gap junctions on the transfer of chemical information was discussed as well as the different factors involved in its regulation including changes in cell volume, high glucose, activation of the renin angiotensin aldosterone system including the intracrine effect of renin and angiotensin II on chemical coupling and cardiac energetics. Finally, the possible role of epigenetic changes of the renin angiotensin aldosterone system (RAAS) on the expression of components of the RAAS was discussed. The evidence available leads to the conception of the heart as a metabolic syncytium in which glucose as well nucleotides and hormones can flow from cell-to-cell though gap junctions, providing a new vision of how alterations in metabolic cooperation can induce cardiac diseases. These findings represent a stimulus for future research in this important area of cardiac physiology and pathology.
Collapse
|
43
|
Ji W, Wei S, Hao P, Xing J, Yuan Q, Wang J, Xu F, Chen Y. Aldehyde Dehydrogenase 2 Has Cardioprotective Effects on Myocardial Ischaemia/Reperfusion Injury via Suppressing Mitophagy. Front Pharmacol 2016; 7:101. [PMID: 27148058 PMCID: PMC4838626 DOI: 10.3389/fphar.2016.00101] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 04/04/2016] [Indexed: 12/20/2022] Open
Abstract
Mitophagy, a selective form of autophagy, is excessively activated in myocardial ischemia/reperfusion (I/R). The study investigated whether aldehyde dehydrogenase 2 (ALDH2) exerted its cardioprotective effect by regulating mitophagy. Myocardial infarct size and apoptosis after I/R in rats were ameliorated by Alda-1, an ALDH2 activator, and aggravated by ALDH2 inhibition. Both in I/R rats and hypoxia/reoxygenation H9C2 cells, ALDH2 activation suppressed phosphatase and tensin homolog-induced putative kinase 1 (PINK1)/Parkin expression, regulating mitophagy, by preventing 4-hydroxynonenal, reactive oxygen species and mitochondrial superoxide accumulation. Furthermore, the effect was enhanced by ALDH2 inhibition. Thus, ALDH2 may protect hearts against I/R injury by suppressing PINK1/Parkin-dependent mitophagy.
Collapse
Affiliation(s)
- Wenqing Ji
- Department of Emergency, Qilu Hospital, Shandong UniversityJinan, China; Chest Pain Center, Qilu Hospital, Shandong UniversityJinan, China; Institute of Emergency and Critical Care Medicine, Shandong UniversityJinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong UniversityJinan, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong UniversityJinan, China
| | - Shujian Wei
- Department of Emergency, Qilu Hospital, Shandong UniversityJinan, China; Chest Pain Center, Qilu Hospital, Shandong UniversityJinan, China; Institute of Emergency and Critical Care Medicine, Shandong UniversityJinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong UniversityJinan, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong UniversityJinan, China
| | - Panpan Hao
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University Jinan, China
| | - Junhui Xing
- Department of Emergency, Qilu Hospital, Shandong UniversityJinan, China; Chest Pain Center, Qilu Hospital, Shandong UniversityJinan, China
| | - Qiuhuan Yuan
- Department of Emergency, Qilu Hospital, Shandong UniversityJinan, China; Chest Pain Center, Qilu Hospital, Shandong UniversityJinan, China; Institute of Emergency and Critical Care Medicine, Shandong UniversityJinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong UniversityJinan, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong UniversityJinan, China
| | - Jiali Wang
- Department of Emergency, Qilu Hospital, Shandong UniversityJinan, China; Chest Pain Center, Qilu Hospital, Shandong UniversityJinan, China; Institute of Emergency and Critical Care Medicine, Shandong UniversityJinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong UniversityJinan, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong UniversityJinan, China
| | - Feng Xu
- Department of Emergency, Qilu Hospital, Shandong UniversityJinan, China; Chest Pain Center, Qilu Hospital, Shandong UniversityJinan, China; Institute of Emergency and Critical Care Medicine, Shandong UniversityJinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong UniversityJinan, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong UniversityJinan, China
| | - Yuguo Chen
- Department of Emergency, Qilu Hospital, Shandong UniversityJinan, China; Chest Pain Center, Qilu Hospital, Shandong UniversityJinan, China; Institute of Emergency and Critical Care Medicine, Shandong UniversityJinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital, Shandong UniversityJinan, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong UniversityJinan, China
| |
Collapse
|
44
|
Calstabin 2: An important regulator for learning and memory in mice. Sci Rep 2016; 6:21087. [PMID: 26888649 PMCID: PMC4758079 DOI: 10.1038/srep21087] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 01/14/2016] [Indexed: 12/20/2022] Open
Abstract
Calstabin2, also named FK506 binding protein 12.6 (FKBP12.6), is a subunit of ryanodine receptor subtype 2 (RyR2) macromolecular complex, which is an intracellular calcium channel and abundant in the brain. Previous studies identified a role of leaky neuronal RyR2 in posttraumatic stress disorder (PTSD). However, the functional role of Calstabin2 in the cognitive function remains unclear. Herein, we used a mouse model of genetic deletion of Calstabin2 to investigate the function of Calstabin2 in cognitive dysfunction. We found that Calstabin2 knockout (KO) mice showed significantly reduced performance in Morris Water Maze (MWM), long-term memory (LTM) contextual fear testing, and rotarod test when compared to wild type (WT) littermates. Indeed, genetic deletion of Calstabin2 reduced long-term potentiation (LTP) at the hippocampal CA3-CA1 connection, increased membrane excitability, and induced RyR2 leak. Finally, we demonstrated that the increase in cytoplasmic calcium activated Ca(2+) dependent potassium currents and led to neuronal apoptosis in KO hippocampal neurons. Thus, these results suggest that neuronal RyR2 Ca(2+) leak due to Calstabin2 deletion contributes to learning deficiency and memory impairment.
Collapse
|
45
|
Santulli G, Borras C, Bousquet J, Calzà L, Cano A, Illario M, Franceschi C, Liotta G, Maggio M, Molloy WD, Montuori N, O’Caoimh R, Orfila F, Rauter AP, Santoro A, Iaccarino G. Models for preclinical studies in aging-related disorders: One is not for all. Transl Med UniSa 2016; 13:4-12. [PMID: 27042427 PMCID: PMC4811343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2022] Open
Abstract
Preclinical studies are essentially based on animal models of a particular disease. The primary purpose of preclinical efficacy studies is to support generalization of treatment-effect relationships to human subjects. Researchers aim to demonstrate a causal relationship between an investigational agent and a disease-related phenotype in such models. Numerous factors can muddle reliable inferences about such cause-effect relationships, including biased outcome assessment due to experimenter expectations. For instance, responses in a particular inbred mouse might be specific to the strain, limiting generalizability. Selecting well-justified and widely acknowledged model systems represents the best start in designing preclinical studies, especially to overcome any potential bias related to the model itself. This is particularly true in the research that focuses on aging, which carries unique challenges, mainly attributable to the fact that our already long lifespan makes designing experiments that use people as subjects extremely difficult and largely impractical.
Collapse
Affiliation(s)
- Gaetano Santulli
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center; College of Physicians & Surgeons, New York, USA;,Corresponding authors: Guido Iaccarino, MD, PhD; ; Gaetano Santulli, MD, PhD;
| | - Consuelo Borras
- Department of Physiology, University of Valencia /INCLIVA, Valencia, Spain
| | - Jean Bousquet
- MACVIA-LR, European Innovation Partnership on Active and Healthy Ageing Reference Site, University Hospital of Montpellier, France;,INSERM, VIMA : Ageing and chronic diseases. Epidemiological and public health approaches, Paris, France,Université Versailles St-Quentin-en-Yvelines, France
| | - Laura Calzà
- Health Sciences and Technologies - Interdepartmental Center for Industrial Research (HST-ICIR) University of Bologna
| | - Antonio Cano
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia/INCLIVA, Valencia, Spain
| | - Maddalena Illario
- Department of Translational Medical Sciences, Federico II University, and R&D Unit, Federico II University Hospital
| | - Claudio Franceschi
- IRCCS Institute of Neurological Sciences, Bologna;,National Research Council of Italy, CNR, Institute for Organic Synthesis and Photoreactivity (ISOF) and Institute of Molecular Genetics, Bologna, Italy;,Dept. of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Giuseppe Liotta
- Department of Biomedicine and Prevention “Tor Vergata” University of Rome, Italy
| | - Marcello Maggio
- Department of Clinical and Experimental Medicine, University of Parma; University Hospital of Parma
| | - William D. Molloy
- Centre for Gerontology and Rehabilitation, University College Cork, Ireland
| | - Nunzia Montuori
- Department of Translational Medical Sciences, Federico II University, and R&D Unit, Federico II University Hospital
| | - Rónán O’Caoimh
- Centre for Gerontology and Rehabilitation, University College Cork, Ireland;,Health Research Board, Clinical Research Facility Galway, National University of Ireland, Galway, Ireland
| | - Francesc Orfila
- Institut Universitari d’Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Barcelona, Spain
| | - Amelia P. Rauter
- Departamento de Quimica e Bioquímica, Universidade de Lisboa, Portugal
| | - Aurelia Santoro
- Dept. of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Guido Iaccarino
- Department of Medicine and Surgery, University of Salerno, Italy.,Corresponding authors: Guido Iaccarino, MD, PhD; ; Gaetano Santulli, MD, PhD;
| |
Collapse
|
46
|
Kallikrein-related peptidase 8 is expressed in myocardium and induces cardiac hypertrophy. Sci Rep 2016; 7:20024. [PMID: 26823023 PMCID: PMC4731818 DOI: 10.1038/srep20024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 12/22/2015] [Indexed: 12/13/2022] Open
Abstract
The tissue kallikrein-related peptidase family (KLK) is a group of trypsin- and chymotrypsin-like serine proteases that share a similar homology to parent tissue kallikrein (KLK1). KLK1 is identified in heart and has anti-hypertrophic effects. However, whether other KLK family members play a role in regulating cardiac function remains unknown. In the present study, we demonstrated for the first time that KLK8 was expressed in myocardium. KLK8 expression was upregulated in left ventricle of cardiac hypertrophy models. Both intra-cardiac adenovirus-mediated and transgenic-mediated KLK8 overexpression led to cardiac hypertrophy in vivo. In primary neonatal rat cardiomyocytes, KLK8 knockdown inhibited phenylephrine (PE)-induced cardiomyocyte hypertrophy, whereas KLK8 overexpression promoted cardiomyocyte hypertrophy via a serine protease activity-dependent but kinin receptor-independent pathway. KLK8 overexpression increased epidermal growth factor (EGF) production, which was blocked by the inhibitors of serine protease. EGF receptor (EGFR) antagonist and EGFR knockdown reversed the hypertrophy induced by KLK8 overexpression. KLK8-induced cardiomyocyte hypertrophy was also significantly decreased by blocking the protease-activated receptor 1 (PAR1) or PAR2 pathway. Our data suggest that KLK8 may promote cardiomyocyte hypertrophy through EGF signaling- and PARs-dependent but a kinin receptor-independent pathway. It is implied that different KLK family members can subtly regulate cardiac function and remodeling.
Collapse
|
47
|
Ock S, Lee WS, Ahn J, Kim HM, Kang H, Kim HS, Jo D, Abel ED, Lee TJ, Kim J. Deletion of IGF-1 Receptors in Cardiomyocytes Attenuates Cardiac Aging in Male Mice. Endocrinology 2016; 157:336-45. [PMID: 26469138 PMCID: PMC4701888 DOI: 10.1210/en.2015-1709] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IGF-1 receptor (IGF-1R) signaling is implicated in cardiac hypertrophy and longevity. However, the role of IGF-1R in age-related cardiac remodeling is only partially understood. We therefore sought to determine whether the deletion of the IGF-1R in cardiomyocytes might delay the development of aging-associated myocardial pathologies by examining 2-year-old male cardiomyocyte-specific IGF-1R knockout (CIGF1RKO) mice. Aging was associated with the induction of IGF-1R expression in hearts. Cardiomyocytes hypertrophied with age in wild-type (WT) mice. In contrast, the cardiac hypertrophic response associated with aging was blunted in CIGF1RKO mice. Concomitantly, fibrosis was reduced in aged CIGF1RKO compared with aged WT hearts. Expression of proinflammatory cytokines such as IL-1α, IL-1β, IL-6, and receptor activator of nuclear factor-κB ligand was increased in aged WT hearts, but this increase was attenuated in aged CIGF1RKO hearts. Phosphorylation of Akt was increased in aged WT, but not in aged CIGF1RKO, hearts. In cultured cardiomyocytes, IGF-1 induced senescence as demonstrated by increased senescence-associated β-galactosidase staining, and a phosphoinositide 3-kinase inhibitor inhibited this effect. Furthermore, inhibition of phosphoinositide 3-kinase significantly prevented the increase in IL-1α, IL-1β, receptor activator of nuclear factor-κB ligand, and p21 protein expression by IGF-1. These data reveal an essential role for the IGF-1-IGF-1R-Akt pathway in mediating cardiomyocyte senescence.
Collapse
MESH Headings
- Aging
- Animals
- Biomarkers/metabolism
- Cardiomegaly/immunology
- Cardiomegaly/metabolism
- Cardiomegaly/pathology
- Cardiomegaly/prevention & control
- Cells, Cultured
- Cellular Senescence/drug effects
- Cytokines/antagonists & inhibitors
- Cytokines/genetics
- Cytokines/metabolism
- Enzyme Inhibitors/pharmacology
- Fibrosis
- Gene Expression Regulation, Developmental/drug effects
- Heart Ventricles/drug effects
- Heart Ventricles/immunology
- Heart Ventricles/metabolism
- Heart Ventricles/pathology
- Insulin-Like Growth Factor I/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/immunology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Phosphatidylinositol 3-Kinase/metabolism
- Phosphoinositide-3 Kinase Inhibitors
- Phosphorylation/drug effects
- Protein Processing, Post-Translational/drug effects
- Proto-Oncogene Proteins c-akt/metabolism
- Receptor, IGF Type 1/agonists
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Signal Transduction/drug effects
- Ventricular Remodeling/drug effects
Collapse
Affiliation(s)
- Sangmi Ock
- Division of Endocrinology and Metabolism (S.O., J.A., H.M.K., J.K.), Department of Internal Medicine, Division of Cardiology (W.S.L.), Department of Internal Medicine, and Departments of Anesthesiology (H.K.) and Pathology (T.J.L.), College of Medicine, Chung-Ang University, Seoul, 156-755, Korea; Department of Biochemistry (H.-S.K.), College of Medicine, The Catholic University of Korea, Seoul, 110-758, Korea; Department of Surgery (D.J.), Vanderbilt University School of Medicine, Nashville, Tennessee 37232; and Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism (D.A.), University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - Wang Soo Lee
- Division of Endocrinology and Metabolism (S.O., J.A., H.M.K., J.K.), Department of Internal Medicine, Division of Cardiology (W.S.L.), Department of Internal Medicine, and Departments of Anesthesiology (H.K.) and Pathology (T.J.L.), College of Medicine, Chung-Ang University, Seoul, 156-755, Korea; Department of Biochemistry (H.-S.K.), College of Medicine, The Catholic University of Korea, Seoul, 110-758, Korea; Department of Surgery (D.J.), Vanderbilt University School of Medicine, Nashville, Tennessee 37232; and Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism (D.A.), University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - Jihyun Ahn
- Division of Endocrinology and Metabolism (S.O., J.A., H.M.K., J.K.), Department of Internal Medicine, Division of Cardiology (W.S.L.), Department of Internal Medicine, and Departments of Anesthesiology (H.K.) and Pathology (T.J.L.), College of Medicine, Chung-Ang University, Seoul, 156-755, Korea; Department of Biochemistry (H.-S.K.), College of Medicine, The Catholic University of Korea, Seoul, 110-758, Korea; Department of Surgery (D.J.), Vanderbilt University School of Medicine, Nashville, Tennessee 37232; and Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism (D.A.), University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - Hyun Min Kim
- Division of Endocrinology and Metabolism (S.O., J.A., H.M.K., J.K.), Department of Internal Medicine, Division of Cardiology (W.S.L.), Department of Internal Medicine, and Departments of Anesthesiology (H.K.) and Pathology (T.J.L.), College of Medicine, Chung-Ang University, Seoul, 156-755, Korea; Department of Biochemistry (H.-S.K.), College of Medicine, The Catholic University of Korea, Seoul, 110-758, Korea; Department of Surgery (D.J.), Vanderbilt University School of Medicine, Nashville, Tennessee 37232; and Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism (D.A.), University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - Hyun Kang
- Division of Endocrinology and Metabolism (S.O., J.A., H.M.K., J.K.), Department of Internal Medicine, Division of Cardiology (W.S.L.), Department of Internal Medicine, and Departments of Anesthesiology (H.K.) and Pathology (T.J.L.), College of Medicine, Chung-Ang University, Seoul, 156-755, Korea; Department of Biochemistry (H.-S.K.), College of Medicine, The Catholic University of Korea, Seoul, 110-758, Korea; Department of Surgery (D.J.), Vanderbilt University School of Medicine, Nashville, Tennessee 37232; and Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism (D.A.), University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - Ho-Shik Kim
- Division of Endocrinology and Metabolism (S.O., J.A., H.M.K., J.K.), Department of Internal Medicine, Division of Cardiology (W.S.L.), Department of Internal Medicine, and Departments of Anesthesiology (H.K.) and Pathology (T.J.L.), College of Medicine, Chung-Ang University, Seoul, 156-755, Korea; Department of Biochemistry (H.-S.K.), College of Medicine, The Catholic University of Korea, Seoul, 110-758, Korea; Department of Surgery (D.J.), Vanderbilt University School of Medicine, Nashville, Tennessee 37232; and Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism (D.A.), University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - Daewoong Jo
- Division of Endocrinology and Metabolism (S.O., J.A., H.M.K., J.K.), Department of Internal Medicine, Division of Cardiology (W.S.L.), Department of Internal Medicine, and Departments of Anesthesiology (H.K.) and Pathology (T.J.L.), College of Medicine, Chung-Ang University, Seoul, 156-755, Korea; Department of Biochemistry (H.-S.K.), College of Medicine, The Catholic University of Korea, Seoul, 110-758, Korea; Department of Surgery (D.J.), Vanderbilt University School of Medicine, Nashville, Tennessee 37232; and Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism (D.A.), University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - E Dale Abel
- Division of Endocrinology and Metabolism (S.O., J.A., H.M.K., J.K.), Department of Internal Medicine, Division of Cardiology (W.S.L.), Department of Internal Medicine, and Departments of Anesthesiology (H.K.) and Pathology (T.J.L.), College of Medicine, Chung-Ang University, Seoul, 156-755, Korea; Department of Biochemistry (H.-S.K.), College of Medicine, The Catholic University of Korea, Seoul, 110-758, Korea; Department of Surgery (D.J.), Vanderbilt University School of Medicine, Nashville, Tennessee 37232; and Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism (D.A.), University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - Tae Jin Lee
- Division of Endocrinology and Metabolism (S.O., J.A., H.M.K., J.K.), Department of Internal Medicine, Division of Cardiology (W.S.L.), Department of Internal Medicine, and Departments of Anesthesiology (H.K.) and Pathology (T.J.L.), College of Medicine, Chung-Ang University, Seoul, 156-755, Korea; Department of Biochemistry (H.-S.K.), College of Medicine, The Catholic University of Korea, Seoul, 110-758, Korea; Department of Surgery (D.J.), Vanderbilt University School of Medicine, Nashville, Tennessee 37232; and Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism (D.A.), University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - Jaetaek Kim
- Division of Endocrinology and Metabolism (S.O., J.A., H.M.K., J.K.), Department of Internal Medicine, Division of Cardiology (W.S.L.), Department of Internal Medicine, and Departments of Anesthesiology (H.K.) and Pathology (T.J.L.), College of Medicine, Chung-Ang University, Seoul, 156-755, Korea; Department of Biochemistry (H.-S.K.), College of Medicine, The Catholic University of Korea, Seoul, 110-758, Korea; Department of Surgery (D.J.), Vanderbilt University School of Medicine, Nashville, Tennessee 37232; and Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism (D.A.), University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| |
Collapse
|
48
|
Zheng J, Zhai K, Chen Y, Zhang X, Miao L, Wei B, Ji G. Nitric oxide mediates stretch-induced Ca2+ oscillation in smooth muscle. J Cell Sci 2016; 129:2430-7. [DOI: 10.1242/jcs.180638] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 03/23/2016] [Indexed: 11/20/2022] Open
Abstract
The stretching of smooth muscle tissue modulates contraction via augmentation of Ca2+ transients, but the mechanism underlying stretch-induced Ca2+ transients is still unknown. We found that mechanical stretching and maintenance of mouse urinary bladder smooth muscle strips and single myocytes at the initial length of 30% and 18%, respectively, resulted in Ca2+ oscillations. Experiments indicated that mechanical stretching remarkably increases the production of nitric oxide (NO) as well as the amplitude and duration of muscle contraction. Stretch-induced Ca2+ oscillations and contractility increases were completely abolished by NO inhibitor L-NAME or eNOS gene inactivation. Moreover, exposure of eNOS knockout myocytes to exogenous NO donor induced Ca2+ oscillations. The stretch-induced Ca2+ oscillations were greatly inhibited by selective IP3R inhibitor xestospongin C and partially inhibited by ryanodine. Moreover, the stretch-induced Ca2+ oscillations were also suppressed by LY294002, but not by the soluble guanylyl cyclase (sGC) inhibitor ODQ. These results suggest that myocytes stretching and maintenance at a certain length resulted in Ca2+ oscillations that is NO dependent and sGC/cGMP independent and results from the activation of PI(3)K in smooth muscle.
Collapse
Affiliation(s)
- Ji Zheng
- Urological Surgery Research Institute, Southwest Hospital, Third Military Medical University, Gao Tanyan Rd. 30, Chongqing 400038, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Rd, Beijing 100101, China
| | - Kui Zhai
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Rd, Beijing 100101, China
| | - Yingxiao Chen
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Rd, Beijing 100101, China
| | - Xu Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Rd, Beijing 100101, China
| | - Lin Miao
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Rd, Beijing 100101, China
| | - Bin Wei
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, United States
| | - Guangju Ji
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Rd, Beijing 100101, China
| |
Collapse
|
49
|
Liu XP, Gao H, Huang XY, Chen YF, Feng XJ, He YH, Li ZM, Liu PQ. Peroxisome proliferator-activated receptor gamma coactivator 1 alpha protects cardiomyocytes from hypertrophy by suppressing calcineurin-nuclear factor of activated T cells c4 signaling pathway. Transl Res 2015; 166:459-473.e3. [PMID: 26118953 DOI: 10.1016/j.trsl.2015.06.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 05/21/2015] [Accepted: 06/02/2015] [Indexed: 01/11/2023]
Abstract
Peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α) is a crucial coregulator interacting with multiple transcriptional factors in the regulation of cardiac hypertrophy. The present study revealed that PGC-1α protected cardiomyocytes from hypertrophy by suppressing calcineurin-nuclear factor of activated T cells c4 (NFATc4) signaling pathway. Overexpression of PGC-1α by adenovirus infection prevented the increased protein and messenger RNA expression of NFATc4 in phenylephrine (PE)-treated hypertrophic cardiomyocytes, whereas knockdown of PGC-1α by RNA silencing augmented the expression of NFATc4. An interaction between PGC-1α and NFATc4 was observed in both the cytoplasm and nucleus of neonatal rat cardiomyocytes. Adenovirus PGC-1α prevented the nuclear import of NFATc4 and increased its phosphorylation level of NFATc4, probably through repressing the expression and activity of calcineurin and interfering with the interaction between calcineurin and NFATc4. On the contrary, PGC-1α silencing aggravated PE-induced calcineurin activation, NFATc4 dephosphorylation, and nuclear translocation. Moreover, the binding activity and transcription activity of NFATc4 to DNA promoter of brain natriuretic peptide were abrogated by PGC-1α overexpression but were enhanced by PGC-1α knockdown. The effect of PGC-1α on suppressing the calcinuerin-NFATc4 signaling pathway might at least partially contribute to the protective effect of PGC-1α on cardiomyocyte hypertrophy. These findings provide novel insights into the role of PGC-1α in regulation of cardiac hypertrophy.
Collapse
Affiliation(s)
- Xue-Ping Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou, PR China
| | - Hui Gao
- Department of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou, PR China; Department of Pharmacology, School of Medicine, Jishou University, Jishou, PR China
| | - Xiao-Yang Huang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou, PR China
| | - Yan-Fang Chen
- Department of Pharmacy, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Xiao-Jun Feng
- Department of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou, PR China
| | - Yan-Hong He
- Department of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou, PR China
| | - Zhuo-Ming Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou, PR China.
| | - Pei-Qing Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-Sen University, Guangzhou, PR China.
| |
Collapse
|
50
|
Lu J, Xiang G, Liu M, Mei W, Xiang L, Dong J. Irisin protects against endothelial injury and ameliorates atherosclerosis in apolipoprotein E-Null diabetic mice. Atherosclerosis 2015; 243:438-48. [PMID: 26520898 DOI: 10.1016/j.atherosclerosis.2015.10.020] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 10/01/2015] [Accepted: 10/14/2015] [Indexed: 12/24/2022]
Abstract
OBJECTIVE The circulating irisin increases energy expenditure and improves insulin resistance in mice and humans. The improvement of insulin resistance ameliorates atherosclerosis. Therefore, we hypothesized that irisin alleviates atherosclerosis in diabetes. METHODS Endothelial function was measured by acetylcholine-induced endothelium-dependent vasodilation using aortic rings in apolipoprotein E-Null (apoE(-/-)) streptozotocin-induced diabetic mice. Atherosclerotic lesion was evaluated by plaque area and inflammatory response in aortas. In addition, the endothelium-protective effects of irisin were also further investigated in primary human umbilical vein endothelial cells (HUVECs) in vitro. RESULTS The in vivo experiments showed that irisin treatment significantly improved endothelial dysfunction, decreased endothelial apoptosis, and predominantly decreased atherosclerotic plaque area of both en face and cross sections when compared with normal saline-treated diabetic mice. Moreover, the infiltrating macrophages and T lymphocytes within plaque and the mRNA expression levels of inflammatory cytokines in aortas were also significantly reduced by irisin treatment in mice. The in vitro experiments revealed that irisin inhibited high glucose-induced apoptosis, oxidative stress and increased antioxidant enzymes expression in HUVECs, and pretreatment with LY294002, l-NAME, AMPK-siRNA or eNOS-siRNA, attenuated the protection of irisin on HUVECs apoptosis induced by high glucose. In addition, the in vivo and in vitro experiments showed that irisin increased the phosphorylation of AMPK, Akt and eNOS in aortas and cultured HUVECs. CONCLUSIONS The present study indicates that systemic administration of irisin may be protected against endothelial injury and ameliorated atherosclerosis in apoE(-/-) diabetic mice. The endothelium-protective action of irisin was through activation of AMPK-PI3K-Akt-eNOS signaling pathway. Irisin could be therapeutic for atherosclerotic vascular diseases in diabetes.
Collapse
Affiliation(s)
- Junyan Lu
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China.
| | - Guangda Xiang
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China.
| | - Min Liu
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China.
| | - Wen Mei
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China.
| | - Lin Xiang
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China.
| | - Jing Dong
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China.
| |
Collapse
|