1
|
Adasheva DA, Serebryanaya DV. IGF Signaling in the Heart in Health and Disease. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1402-1428. [PMID: 39245453 DOI: 10.1134/s0006297924080042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/11/2024] [Accepted: 06/22/2024] [Indexed: 09/10/2024]
Abstract
One of the most vital processes of the body is the cardiovascular system's proper operation. Physiological processes in the heart are regulated by the balance of cardioprotective and pathological mechanisms. The insulin-like growth factor system (IGF system, IGF signaling pathway) plays a pivotal role in regulating growth and development of various cells and tissues. In myocardium, the IGF system provides cardioprotective effects as well as participates in pathological processes. This review summarizes recent data on the role of IGF signaling in cardioprotection and pathogenesis of various cardiovascular diseases, as well as analyzes severity of these effects in various scenarios.
Collapse
Affiliation(s)
- Daria A Adasheva
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Daria V Serebryanaya
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| |
Collapse
|
2
|
Vlachakis D, Tsilafakis K, Kostavasili I, Kossida S, Mavroidis M. Unraveling Desmin's Head Domain Structure and Function. Cells 2024; 13:603. [PMID: 38607042 PMCID: PMC11012097 DOI: 10.3390/cells13070603] [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: 01/09/2024] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 04/13/2024] Open
Abstract
Understanding the structure and function of intermediate filaments (IFs) is necessary in order to explain why more than 70 related IF genes have evolved in vertebrates while maintaining such dramatically tissue-specific expression. Desmin is a member of the large multigene family of IF proteins and is specifically expressed in myocytes. In an effort to elucidate its muscle-specific behavior, we have used a yeast two-hybrid system in order to identify desmin's head binding partners. We described a mitochondrial and a lysosomal protein, NADH ubiquinone oxidoreductase core subunit S2 (NDUFS2), and saposin D, respectively, as direct desmin binding partners. In silico analysis indicated that both interactions at the atomic level occur in a very similar way, by the formation of a three-helix bundle with hydrophobic interactions in the interdomain space and hydrogen bonds at R16 and S32 of the desmin head domain. The interactions, confirmed also by GST pull-down assays, indicating the necessity of the desmin head domain and, furthermore, point out its role in function of mitochondria and lysosomes, organelles which are disrupted in myopathies due to desmin head domain mutations.
Collapse
Affiliation(s)
- Dimitrios Vlachakis
- Biotechnology Department, Agricultural University of Athens, 11855 Athens, Greece;
| | - Konstantinos Tsilafakis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, 11527 Athens, Greece; (K.T.); (I.K.)
- Biochemistry & Biotechnology Department, University of Thessaly, 41500 Larisa, Greece
| | - Ioanna Kostavasili
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, 11527 Athens, Greece; (K.T.); (I.K.)
| | - Sophia Kossida
- IMGT, The International ImMunoGeneTics Information System, National Center for Scientific Research (CNRS), Institute of Human Genetics (IGH), University of Montpellier (UM), 34090 Montpellier, France;
| | - Manolis Mavroidis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, 11527 Athens, Greece; (K.T.); (I.K.)
| |
Collapse
|
3
|
Hermosilla VE, Gyenis L, Rabalski AJ, Armijo ME, Sepúlveda P, Duprat F, Benítez-Riquelme D, Fuentes-Villalobos F, Quiroz A, Hepp MI, Farkas C, Mastel M, González-Chavarría I, Jackstadt R, Litchfield DW, Castro AF, Pincheira R. Casein kinase 2 phosphorylates and induces the SALL2 tumor suppressor degradation in colon cancer cells. Cell Death Dis 2024; 15:223. [PMID: 38493149 PMCID: PMC10944491 DOI: 10.1038/s41419-024-06591-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 02/29/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
Spalt-like proteins are Zinc finger transcription factors from Caenorhabditis elegans to vertebrates, with critical roles in development. In vertebrates, four paralogues have been identified (SALL1-4), and SALL2 is the family's most dissimilar member. SALL2 is required during brain and eye development. It is downregulated in cancer and acts as a tumor suppressor, promoting cell cycle arrest and cell death. Despite its critical functions, information about SALL2 regulation is scarce. Public data indicate that SALL2 is ubiquitinated and phosphorylated in several residues along the protein, but the mechanisms, biological consequences, and enzymes responsible for these modifications remain unknown. Bioinformatic analyses identified several putative phosphorylation sites for Casein Kinase II (CK2) located within a highly conserved C-terminal PEST degradation motif of SALL2. CK2 is a serine/threonine kinase that promotes cell proliferation and survival and is often hyperactivated in cancer. We demonstrated that CK2 phosphorylates SALL2 residues S763, T778, S802, and S806 and promotes SALL2 degradation by the proteasome. Accordingly, pharmacological inhibition of CK2 with Silmitasertib (CX-4945) restored endogenous SALL2 protein levels in SALL2-deficient breast MDA-MB-231, lung H1299, and colon SW480 cancer cells. Silmitasertib induced a methuosis-like phenotype and cell death in SW480 cells. However, the phenotype was significantly attenuated in CRISPr/Cas9-mediated SALL2 knockout SW480 cells. Similarly, Sall2-deficient tumor organoids were more resistant to Silmitasertib-induced cell death, confirming that SALL2 sensitizes cancer cells to CK2 inhibition. We identified a novel CK2-dependent mechanism for SALL2 regulation and provided new insights into the interplay between these two proteins and their role in cell survival and proliferation.
Collapse
Affiliation(s)
- V E Hermosilla
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Transducción de Señales y Cáncer, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Dept of Orofacial Sciences and Dept of Anatomy, University of California-San Francisco, San Francisco, CA, USA
| | - L Gyenis
- Department of Biochemistry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - A J Rabalski
- Department of Biochemistry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
- Odyssey Therapeutics, Boston, MA, USA
| | - M E Armijo
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Transducción de Señales y Cáncer, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - P Sepúlveda
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Transducción de Señales y Cáncer, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - F Duprat
- Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - D Benítez-Riquelme
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Transducción de Señales y Cáncer, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - F Fuentes-Villalobos
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Transducción de Señales y Cáncer, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Inmunovirología. Departamento de Microbiologia. Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - A Quiroz
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Transducción de Señales y Cáncer, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - M I Hepp
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Transducción de Señales y Cáncer, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Investigación en Ciencias Biomédicas, Departamento de Ciencias Básicas y Morfología, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - C Farkas
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Transducción de Señales y Cáncer, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Investigación en Ciencias Biomédicas, Departamento de Ciencias Básicas y Morfología, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - M Mastel
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg. Cancer Progression and Metastasis Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - I González-Chavarría
- Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - R Jackstadt
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg. Cancer Progression and Metastasis Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - D W Litchfield
- Department of Biochemistry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - A F Castro
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.
- Laboratorio de Transducción de Señales y Cáncer, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.
| | - R Pincheira
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.
- Laboratorio de Transducción de Señales y Cáncer, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.
| |
Collapse
|
4
|
Muluhie M, Castiglioni L, Rzemieniec J, Mercuriali B, Gelosa P, Sironi L. Montelukast, an available and safe anti-asthmatic drug, prevents maladaptive remodelling and maintains cardiac functionality following myocardial infarction. Sci Rep 2024; 14:3371. [PMID: 38337010 PMCID: PMC10858037 DOI: 10.1038/s41598-024-53936-x] [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: 01/02/2023] [Accepted: 02/06/2024] [Indexed: 02/12/2024] Open
Abstract
Preclinical and clinical data indicate that the 5-lipoxygenase pathway becomes activated in cardiovascular diseases suggesting an important role of CysLTs in atherosclerosis and in its ischemic complications. This study aims to investigate the effects of montelukast, a CysLTR-1 antagonist, in a mouse model of myocardial infarction (MI). C57BL/6N female mice were subjected to coronary artery ligation and received montelukast (10 mg/kg/day, intraperitoneal) or vehicle. Montelukast exerted beneficial effects in the infarcted area, decreasing mRNA expression of inflammatory genes, such Il1β and Ccl2 (p < 0.05), at 48 h after MI, and reducing infarct size and preventing ischemic wall thinning (p < 0.05) at 4 weeks. Furthermore, montelukast counteracted maladaptive remodelling of whole heart. Indeed, montelukast reduced LV mass (p < 0.05) and remote wall thickening (p < 0.05), and improved cardiac pumping function, as evidenced by increased global ejection fraction (p < 0.01), and regional contractility in infarcted (p < 0.05) and in remote non-infarcted (p < 0.05) myocardium. Finally, montelukast prevented cardiomyocytes hypertrophy (p < 0.05) in remote myocardium, reducing the phosphorylation of GSK3β, a regulator of hypertrophic pathway (p < 0.05). Our data strongly demonstrate the ability of montelukast to contrast the MI-induced maladaptive conditions, thus sustaining cardiac contractility. The results provide evidences for montelukast "repurposing" in cardiovascular diseases and in particular in myocardial infarction.
Collapse
Affiliation(s)
- Majeda Muluhie
- Department of Pharmaceutical Sciences, University of Milan, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Laura Castiglioni
- Department of Pharmaceutical Sciences, University of Milan, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Joanna Rzemieniec
- Department of Pharmaceutical Sciences, University of Milan, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Benedetta Mercuriali
- Department of Pharmaceutical Sciences, University of Milan, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Paolo Gelosa
- Department of Pharmaceutical Sciences, University of Milan, Via G. Balzaretti 9, 20133, Milan, Italy
| | - Luigi Sironi
- Department of Pharmaceutical Sciences, University of Milan, Via G. Balzaretti 9, 20133, Milan, Italy.
| |
Collapse
|
5
|
Orgil BO, Purevjav E. Molecular Pathways and Animal Models of Cardiomyopathies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:991-1019. [PMID: 38884766 DOI: 10.1007/978-3-031-44087-8_64] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Cardiomyopathies are a heterogeneous group of disorders of the heart muscle that ultimately result in congestive heart failure. Rapid progress in genetics, molecular and cellular biology with breakthrough innovative genetic-engineering techniques, such as next-generation sequencing and multiomics platforms, stem cell reprogramming, as well as novel groundbreaking gene-editing systems over the past 25 years has greatly improved the understanding of pathogenic signaling pathways in inherited cardiomyopathies. This chapter will focus on intracellular and intercellular molecular signaling pathways that are activated by a genetic insult in cardiomyocytes to maintain tissue and organ level regulation and resultant cardiac remodeling in certain forms of cardiomyopathies. In addition, animal models of different clinical forms of human cardiomyopathies with their summaries of triggered key molecules and signaling pathways will be described.
Collapse
Affiliation(s)
- Buyan-Ochir Orgil
- Department of Pediatrics, The Heart Institute, Division of Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Enkhsaikhan Purevjav
- Department of Pediatrics, The Heart Institute, Division of Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
6
|
Zhou YK, Han CS, Zhu ZL, Chen P, Wang YM, Lin S, Chen LJ, Zhuang ZM, Zhou YH, Yang RL. M2 exosomes modified by hydrogen sulfide promoted bone regeneration by moesin mediated endocytosis. Bioact Mater 2024; 31:192-205. [PMID: 37593496 PMCID: PMC10429289 DOI: 10.1016/j.bioactmat.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/31/2023] [Accepted: 08/08/2023] [Indexed: 08/19/2023] Open
Abstract
Bone defects caused by trauma or tumor led to high medical costs and poor life quality for patients. The exosomes, micro vesicles of 30-150 nm in diameter, derived from macrophages manipulated bone regeneration. However, the role of hydrogen sulfide (H2S) in the biogenesis and function of exosomes and its effects on bone regeneration remains elusive. In this study, we used H2S slow releasing donor GYY4137 to stimulate macrophages and found that H2S promoted the polarization of M2 macrophages to increase bone regeneration of MSCs in vitro and in vivo. Moreover, we developed the H2S pre-treated M2 macrophage exosomes and found these exosomes displayed significantly higher capacity to promote bone regeneration in calvarial bone defects by re-establishing the local immune microenvironment. Mechanically, H2S treatment altered the protein profile of exosomes derived from M2 macrophages. One of the significantly enriched exosomal proteins stimulated by H2S, moesin protein, facilitated the exosomes endocytosis into MSCs, leading to activated the β-catenin signaling pathway to promote osteogenic differentiation of MSCs. In summary, H2S pretreated M2 exosomes promoted the bone regeneration of MSCs via facilitating exosomes uptake by MSCs and activate β-catenin signaling pathway. This study not only provides new strategies for promoting bone regeneration, but also provides new insights for the effect and mechanism of exosomes internalization.
Collapse
Affiliation(s)
- Yi-kun Zhou
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Chun-shan Han
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Zi-lu Zhu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Peng Chen
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Yi-ming Wang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Shuai Lin
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Liu-jing Chen
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Zi-meng Zhuang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Yan-heng Zhou
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Rui-li Yang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Haidian District, Beijing, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Haidian District, Beijing, China
- Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| |
Collapse
|
7
|
Sivakumar B, Ali N, Ahmad SF, Nadeem A, Waseem M, Kurian GA. PM 2.5-Induced Cardiac Structural Modifications and Declined Pro-Survival Signalling Pathways Are Responsible for the Inefficiency of GSK-3β Inhibitor in Attenuating Myocardial Ischemia-Reperfusion Injury in Rats. Cells 2023; 12:2064. [PMID: 37626874 PMCID: PMC10453520 DOI: 10.3390/cells12162064] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/27/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Circulatory GSK3β is recognized as a biomarker and therapeutic target for diseases, including myocardial diseases. However, its potential as a target for myocardial ischemia-reperfusion injury (IR) in the presence of PM2.5 exposure is unclear. Wistar rats underwent IR following either a 21-day or single exposure to PM2.5 at a concentration of 250 µg/m3. The effects of GSK3β inhibitor on cardiac physiology, tissue injury, mitochondrial function, and the PI3K/AKT/GSK3β signalling axis were examined. The inhibitor was not effective in improving hemodynamics or reducing IR-induced infarction in the myocardium exposed to PM2.5 for 21 days. However, for a single-day exposure, the inhibitor showed potential in mitigating cardiac injury. In normal hearts undergoing IR, the inhibitor activated the PI3K/AKT signalling pathway, improved mitochondrial function, and reduced oxidative stress. These positive effects were not observed in PM2.5-exposed rats. Furthermore, the inhibitor stimulated autophagy in hearts exposed to PM2.5 for 21 days and subjected to IR, resulting in increased mTOR expression and decreased AMPK expression. In normal hearts and those exposed to a single dose of PM2.5, the inhibitor effectively activated the PI3K/Akt/AMPK axis. These findings suggest that GSK3β may not be a reliable therapeutic target for IR in the presence of chronic PM2.5 exposure.
Collapse
Affiliation(s)
- Bhavana Sivakumar
- Vascular Biology Laboratory, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613401, Tamil Nadu, India;
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (S.F.A.); (A.N.)
| | - Sheikh F. Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (S.F.A.); (A.N.)
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (S.F.A.); (A.N.)
| | - Mohammad Waseem
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Eastern Shore, Princess Anne, MD 21853, USA;
| | - Gino A. Kurian
- Vascular Biology Laboratory, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613401, Tamil Nadu, India;
| |
Collapse
|
8
|
Rami M, Rahdar S, Ahmadi Hekmatikar A, Awang Daud DM. Highlighting the novel effects of high-intensity interval training on some histopathological and molecular indices in the heart of type 2 diabetic rats. Front Endocrinol (Lausanne) 2023; 14:1175585. [PMID: 37274326 PMCID: PMC10235768 DOI: 10.3389/fendo.2023.1175585] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/04/2023] [Indexed: 06/06/2023] Open
Abstract
Background Type 2 diabetes is one of the most common metabolic diseases in recent years and has become an important risk factor for cardiovascular disorders. The first goal is to reduce type 2 diabetes, and in the case of cardiovascular disease, the second goal is to reduce and manage that disorder. Materials and methods The rats were divided into 4 groups: Healthy Control (n=8), Diabetes Control (n=8), Diabetes Training (n=8), and Healthy Training (n=8). The protocol consisted of 8 weeks of High-intensity interval (5 sessions per week), where the training started with 80% of the peak speed in the first week, and 10% was added to this speed every week. To measure the level of B-catenin, c-MYC, GSK3B, and Bcl-2 proteins using the western blot method, cardiac pathological changes were measured using hematoxylin and eosin staining, Masson's trichrome and PAS staining and apoptosis using the TUNEL method. Findings Histological results showed that diabetes causes significant pathological hypertrophy, fibrosis, and severe apoptosis in heart tissue. HIIT training significantly reduced pathological hypertrophy and fibrosis in heart tissue, and the rate of cardiomyocyte apoptosis was greatly reduced. This research showed that diabetes disorder increases the levels of B-catenin and c-Myc proteins and causes a decrease in the expression of GSK3B and Bcl-2 proteins. After eight weeks of HIIT training, the levels of B-catenin and c-Myc proteins decreased significantly, and the levels of GSK3B and Bcl-2 proteins increased. Conclusion This study showed that HIIT could be a suitable strategy to reduce cardiomyopathy in type 2 diabetic rats. However, it is suggested that in future studies, researchers should perform different intensities and exercises to promote exercise goals in type 2 diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Mohammad Rami
- Department of Sport Physiology, Faculty of Sport Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Samane Rahdar
- Department of Basic Sciences, Histology section, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Amirhoseein Ahmadi Hekmatikar
- Department of Physical Education and Sport Sciences, Faculty of Humanities, Tarbiat Modares University, Tehran, Iran
| | - D. Maryama Awang Daud
- Health Through Exercise and Active Living (HEAL) Research Unit, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
| |
Collapse
|
9
|
Alsabeelah N, Kumar V. Protective Effect of Triclosan in Monocrotaline-Induced Pulmonary Arterial Hypertension: FASN Inhibition a Novel Approach. JOURNAL OF PHARMACY AND BIOALLIED SCIENCES 2022; 14:171-177. [PMID: 37051426 PMCID: PMC10084994 DOI: 10.4103/jpbs.jpbs_307_22] [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: 06/05/2022] [Revised: 08/10/2022] [Accepted: 09/27/2022] [Indexed: 02/17/2023] Open
Abstract
Background Novel pharmacological approaches are needed to improve the outcomes of patients with idiopathic pulmonary hypertension. Fatty acid synthase (FASN) inhibitors have shown beneficial effects in preclinical models of pulmonary arterial hypertension (PAH), because of their role in the regulation of pulmonary artery vasoconstrictor tone and remodeling. Objective We compared a Triclosan (FASN inhibitor), for the first time with the dual endothelin receptor antagonist, macitentan, in a monocrotaline-induced rat pulmonary hypertension model. Methods Different methods (hemodynamics, histology of right ventricle and pulmonary vessels, and circulating biomarkers) showed consistently that 30 mg/kg daily of Triclosan (FASN inhibitor) and 10 mg/kg daily of macitentan slowed the progression of PAH both at the functional and structural levels. Results Treatments started on day 14 after monocrotaline injection and lasted 14 days. The findings of all experimental methods show that the FASN inhibitor has more similar effects as compared to macitentan. Conclusion Our study reveals that inhibition of FAS decreases RV hypertrophy and improves cardiac function associated with PAH with the regulation of metabolic functions and governs further studies to establish "FASN inhibitor as a potential therapeutic approach" for the management of PAH.
Collapse
Affiliation(s)
- Nimer Alsabeelah
- Pharmacy Practice Department, Pharmacy College, University of Hafr Al Batin, Saudi Arabia
| | - Vinay Kumar
- Department of Pharmacology, KIET Group of Institutions (KIET School of Pharmacy), Delhi-NCR, Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
10
|
The insulin receptor family in the heart: new light on old insights. Biosci Rep 2022; 42:231495. [PMID: 35766350 PMCID: PMC9297685 DOI: 10.1042/bsr20221212] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/20/2022] [Accepted: 06/29/2022] [Indexed: 11/17/2022] Open
Abstract
Insulin was discovered over 100 years ago. Whilst the first half century defined many of the physiological effects of insulin, the second emphasised the mechanisms by which it elicits these effects, implicating a vast array of G proteins and their regulators, lipid and protein kinases and counteracting phosphatases, and more. Potential growth-promoting and protective effects of insulin on the heart emerged from studies of carbohydrate metabolism in the 1960s, but the insulin receptors (and the related receptor for insulin-like growth factors 1 and 2) were not defined until the 1980s. A related third receptor, the insulin receptor-related receptor remained an orphan receptor for many years until it was identified as an alkali-sensor. The mechanisms by which these receptors and the plethora of downstream signalling molecules confer cardioprotection remain elusive. Here, we review important aspects of the effects of the three insulin receptor family members in the heart. Metabolic studies are set in the context of what is now known of insulin receptor family signalling and the role of protein kinase B (PKB or Akt), and the relationship between this and cardiomyocyte survival versus death is discussed. PKB/Akt phosphorylates numerous substrates with potential for cardioprotection in the contractile cardiomyocytes and cardiac non-myocytes. Our overall conclusion is that the effects of insulin on glucose metabolism that were initially identified remain highly pertinent in managing cardiomyocyte energetics and preservation of function. This alone provides a high level of cardioprotection in the face of pathophysiological stressors such as ischaemia and myocardial infarction.
Collapse
|
11
|
Preserved Left Ventricular Function despite Myocardial Fibrosis and Myopathy in the Dystrophin-Deficient D2.B10-Dmdmdx/J Mouse. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5362115. [PMID: 35340200 PMCID: PMC8942668 DOI: 10.1155/2022/5362115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 02/18/2022] [Accepted: 02/26/2022] [Indexed: 11/18/2022]
Abstract
Duchenne muscular dystrophy involves an absence of dystrophin, a cytoskeletal protein which supports cell structural integrity and scaffolding for signalling molecules in myocytes. Affected individuals experience progressive muscle degeneration that leads to irreversible loss of ambulation and respiratory diaphragm function. Although clinical management has greatly advanced, heart failure due to myocardial cell loss and fibrosis remains the major cause of death. We examined cardiac morphology and function in D2.B10-Dmdmdx/J (D2-mdx) mice, a relatively new mouse model of muscular dystrophy, which we compared to their wild-type background DBA/2J mice (DBA/2). We also tested whether drug treatment with a specific blocker of mitochondrial permeability transition pore opening (Debio-025), or ACE inhibition (Perindopril), had any effect on dystrophy-related cardiomyopathy. D2-mdx mice were treated for six weeks with Vehicle control, Debio-025 (20 mg/kg/day), Perindopril (2 mg/kg/day), or a combination (n = 8/group). At 18 weeks, compared to DBA/2, D2-mdx hearts displayed greater ventricular collagen, lower cell density, greater cell diameter, and greater protein expression levels of IL-6, TLR4, BAX/Bcl2, caspase-3, PGC-1α, and notably monoamine oxidases A and B. Remarkably, these adaptations in D2-mdx mice were associated with preserved resting left ventricular function similar to DBA/2 mice. Compared to vehicle, although Perindopril partly attenuated the increase in heart weight and collagen at 18 weeks, the drug treatments had no marked impact on dystrophic cardiomyopathy.
Collapse
|
12
|
Pathological oligodendrocyte precursor cells revealed in human schizophrenic brains and trigger schizophrenia-like behaviors and synaptic defects in genetic animal model. Mol Psychiatry 2022; 27:5154-5166. [PMID: 36131044 PMCID: PMC9763102 DOI: 10.1038/s41380-022-01777-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 01/19/2023]
Abstract
Although the link of white matter to pathophysiology of schizophrenia is documented, loss of myelin is not detected in patients at the early stages of the disease, suggesting that pathological evolution of schizophrenia may occur before significant myelin loss. Disrupted-in-schizophrenia-1 (DISC1) protein is highly expressed in oligodendrocyte precursor cells (OPCs) and regulates their maturation. Recently, DISC1-Δ3, a major DISC1 variant that lacks exon 3, has been identified in schizophrenia patients, although its pathological significance remains unknown. In this study, we detected in schizophrenia patients a previously unidentified pathological phenotype of OPCs exhibiting excessive branching. We replicated this phenotype by generating a mouse strain expressing DISC1-Δ3 gene in OPCs. We further demonstrated that pathological OPCs, rather than myelin defects, drive the onset of schizophrenic phenotype by hyperactivating OPCs' Wnt/β-catenin pathway, which consequently upregulates Wnt Inhibitory Factor 1 (Wif1), leading to the aberrant synaptic formation and neuronal activity. Suppressing Wif1 in OPCs rescues synaptic loss and behavioral disorders in DISC1-Δ3 mice. Our findings reveal the pathogenetic role of OPC-specific DISC1-Δ3 variant in the onset of schizophrenia and highlight the therapeutic potential of Wif1 as an alternative target for the treatment of this disease.
Collapse
|
13
|
Wogksch MD, Goodenough CG, Finch ER, Partin RE, Ness KK. Physical activity and fitness in childhood cancer survivors: a scoping review. AGING AND CANCER 2021; 2:112-128. [PMID: 35098147 DOI: 10.1002/aac2.12042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Estimates indicate that nearly eight percent of the over 500,000 survivors of childhood cancer living in the United States are frail in their fourth and fifth decades of life, a phenotype typically seen in geriatric populations. Participation in regular physical activity to improve physical fitness in healthy and diseased populations reduces risk for frail health by increasing physiologic reserve. However, physical activity may not have the same effects on fitness in childhood cancer survivors as it does among their peers with no cancer history. AIMS This scoping review seeks to describe associations between physical activity, physical fitness, chronic disease, and mortality in childhood cancer survivors. METHODS Relevant literature was identified through a comprehensive search in the PubMed, Web of Science, CINAHL, and Cochrane databases. A narrative synthesis was performed on observational studies that had physical activity or physical fitness clearly defined and compared with chronic disease outcomes. RESULTS A total of 595 studies were screened, and results from 11 studies are presented. Childhood cancer survivors who participate in regular physical activity have improved markers of cardiovascular health, decreased risk of overt cardiovascular disease, and decreased risk of all-cause mortality compared to survivors who are not physically active. Childhood cancer survivors who are physically fit have increased neurocognition, and decreased risk of all-cause mortality compared to survivor's who are not fit. The differential effects of physical activity on fitness and health among childhood cancer survivors when compared to peers is potentially related to treatment exposures that damage cardiovascular tissue and impact regenerative potential. CONCLUSION Research is needed to determine the optimal timing, frequency, intensity, and duration of physical activity necessary to optimize fitness in childhood cancer survivors.
Collapse
Affiliation(s)
- Matthew D Wogksch
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN
| | - Chelsea G Goodenough
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN
| | - Emily R Finch
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN
| | - Robyn E Partin
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN
| | - Kirsten K Ness
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN
| |
Collapse
|
14
|
Waløen K, Jung-Kc K, Vecchia ED, Pandey S, Gasparik N, Døskeland A, Patil S, Kleppe R, Hritz J, Norton WHJ, Martinez A, Haavik J. Cysteine Modification by Ebselen Reduces the Stability and Cellular Levels of 14-3-3 Proteins. Mol Pharmacol 2021; 100:155-169. [PMID: 34031189 DOI: 10.1124/molpharm.120.000184] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 05/11/2021] [Indexed: 11/22/2022] Open
Abstract
The 14-3-3 proteins constitute a family of adaptor proteins with many binding partners and biological functions, and they are considered promising drug targets in cancer and neuropsychiatry. By screening 1280 small-molecule drugs using differential scanning fluorimetry (DSF), we found 15 compounds that decreased the thermal stability of 14-3-3ζ Among these compounds, ebselen was identified as a covalent, destabilizing ligand of 14-3-3 isoforms ζ, ε, γ, and η Ebselen bonding decreased 14-3-3ζ binding to its partner Ser19-phosphorylated tyrosine hydroxylase. Characterization of site-directed mutants at cysteine residues in 14-3-3ζ (C25, C94, and C189) by DSF and mass spectroscopy revealed covalent modification by ebselen of all cysteines through a selenylsulfide bond. C25 appeared to be the preferential site of ebselen interaction in vitro, whereas modification of C94 was the main determinant for protein destabilization. At therapeutically relevant concentrations, ebselen and ebselen oxide caused decreased 14-3-3 levels in SH-SY5Y cells, accompanied with an increased degradation, most probably by the ubiquitin-dependent proteasome pathway. Moreover, ebselen-treated zebrafish displayed decreased brain 14-3-3 content, a freezing phenotype, and reduced mobility, resembling the effects of lithium, consistent with its proposed action as a safer lithium-mimetic drug. Ebselen has recently emerged as a promising drug candidate in several medical areas, such as cancer, neuropsychiatric disorders, and infectious diseases, including coronavirus disease 2019. Its pleiotropic actions are attributed to antioxidant effects and formation of selenosulfides with critical cysteine residues in proteins. Our work indicates that a destabilization of 14-3-3 may affect the protein interaction networks of this protein family, contributing to the therapeutic potential of ebselen. SIGNIFICANCE STATEMENT: There is currently great interest in the repurposing of established drugs for new indications and therapeutic targets. This study shows that ebselen, which is a promising drug candidate against cancer, bipolar disorder, and the viral infection coronavirus disease 2019, covalently bonds to cysteine residues in 14-3-3 adaptor proteins, triggering destabilization and increased degradation in cells and intact brain tissue when used in therapeutic concentrations, potentially explaining the behavioral, anti-inflammatory, and antineoplastic effects of this drug.
Collapse
Affiliation(s)
- Kai Waløen
- Department of Biomedicine (K.W., K.J.K.C., S.Pan., A.D., S.Pat., A.M., J.Ha.), Proteomics Unit (PROBE), (A.D.), University of Bergen, Bergen, Norway; Department of Neuroscience, Psychology and Behaviour, College of Medicine, Biological Sciences and Psychology, University of Leicester, Leicester, UK (E.D.V., W.H.J.N.); CEITEC-MU, Masaryk University, Brno, Czech Republic (N.G., J.Hr.); Department of Chemistry, Faculty of Science, Masaryk University, Brno, Czech Republic and Norwegian Centre for Maritime and Diving Medicine, Department of Occupational Medicine (R.K.), Division of Psychiatry (J.Ha.), Haukeland University Hospital, Bergen, Norway
| | - Kunwar Jung-Kc
- Department of Biomedicine (K.W., K.J.K.C., S.Pan., A.D., S.Pat., A.M., J.Ha.), Proteomics Unit (PROBE), (A.D.), University of Bergen, Bergen, Norway; Department of Neuroscience, Psychology and Behaviour, College of Medicine, Biological Sciences and Psychology, University of Leicester, Leicester, UK (E.D.V., W.H.J.N.); CEITEC-MU, Masaryk University, Brno, Czech Republic (N.G., J.Hr.); Department of Chemistry, Faculty of Science, Masaryk University, Brno, Czech Republic and Norwegian Centre for Maritime and Diving Medicine, Department of Occupational Medicine (R.K.), Division of Psychiatry (J.Ha.), Haukeland University Hospital, Bergen, Norway
| | - Elisa D Vecchia
- Department of Biomedicine (K.W., K.J.K.C., S.Pan., A.D., S.Pat., A.M., J.Ha.), Proteomics Unit (PROBE), (A.D.), University of Bergen, Bergen, Norway; Department of Neuroscience, Psychology and Behaviour, College of Medicine, Biological Sciences and Psychology, University of Leicester, Leicester, UK (E.D.V., W.H.J.N.); CEITEC-MU, Masaryk University, Brno, Czech Republic (N.G., J.Hr.); Department of Chemistry, Faculty of Science, Masaryk University, Brno, Czech Republic and Norwegian Centre for Maritime and Diving Medicine, Department of Occupational Medicine (R.K.), Division of Psychiatry (J.Ha.), Haukeland University Hospital, Bergen, Norway
| | - Sunil Pandey
- Department of Biomedicine (K.W., K.J.K.C., S.Pan., A.D., S.Pat., A.M., J.Ha.), Proteomics Unit (PROBE), (A.D.), University of Bergen, Bergen, Norway; Department of Neuroscience, Psychology and Behaviour, College of Medicine, Biological Sciences and Psychology, University of Leicester, Leicester, UK (E.D.V., W.H.J.N.); CEITEC-MU, Masaryk University, Brno, Czech Republic (N.G., J.Hr.); Department of Chemistry, Faculty of Science, Masaryk University, Brno, Czech Republic and Norwegian Centre for Maritime and Diving Medicine, Department of Occupational Medicine (R.K.), Division of Psychiatry (J.Ha.), Haukeland University Hospital, Bergen, Norway
| | - Norbert Gasparik
- Department of Biomedicine (K.W., K.J.K.C., S.Pan., A.D., S.Pat., A.M., J.Ha.), Proteomics Unit (PROBE), (A.D.), University of Bergen, Bergen, Norway; Department of Neuroscience, Psychology and Behaviour, College of Medicine, Biological Sciences and Psychology, University of Leicester, Leicester, UK (E.D.V., W.H.J.N.); CEITEC-MU, Masaryk University, Brno, Czech Republic (N.G., J.Hr.); Department of Chemistry, Faculty of Science, Masaryk University, Brno, Czech Republic and Norwegian Centre for Maritime and Diving Medicine, Department of Occupational Medicine (R.K.), Division of Psychiatry (J.Ha.), Haukeland University Hospital, Bergen, Norway
| | - Anne Døskeland
- Department of Biomedicine (K.W., K.J.K.C., S.Pan., A.D., S.Pat., A.M., J.Ha.), Proteomics Unit (PROBE), (A.D.), University of Bergen, Bergen, Norway; Department of Neuroscience, Psychology and Behaviour, College of Medicine, Biological Sciences and Psychology, University of Leicester, Leicester, UK (E.D.V., W.H.J.N.); CEITEC-MU, Masaryk University, Brno, Czech Republic (N.G., J.Hr.); Department of Chemistry, Faculty of Science, Masaryk University, Brno, Czech Republic and Norwegian Centre for Maritime and Diving Medicine, Department of Occupational Medicine (R.K.), Division of Psychiatry (J.Ha.), Haukeland University Hospital, Bergen, Norway
| | - Sudarshan Patil
- Department of Biomedicine (K.W., K.J.K.C., S.Pan., A.D., S.Pat., A.M., J.Ha.), Proteomics Unit (PROBE), (A.D.), University of Bergen, Bergen, Norway; Department of Neuroscience, Psychology and Behaviour, College of Medicine, Biological Sciences and Psychology, University of Leicester, Leicester, UK (E.D.V., W.H.J.N.); CEITEC-MU, Masaryk University, Brno, Czech Republic (N.G., J.Hr.); Department of Chemistry, Faculty of Science, Masaryk University, Brno, Czech Republic and Norwegian Centre for Maritime and Diving Medicine, Department of Occupational Medicine (R.K.), Division of Psychiatry (J.Ha.), Haukeland University Hospital, Bergen, Norway
| | - Rune Kleppe
- Department of Biomedicine (K.W., K.J.K.C., S.Pan., A.D., S.Pat., A.M., J.Ha.), Proteomics Unit (PROBE), (A.D.), University of Bergen, Bergen, Norway; Department of Neuroscience, Psychology and Behaviour, College of Medicine, Biological Sciences and Psychology, University of Leicester, Leicester, UK (E.D.V., W.H.J.N.); CEITEC-MU, Masaryk University, Brno, Czech Republic (N.G., J.Hr.); Department of Chemistry, Faculty of Science, Masaryk University, Brno, Czech Republic and Norwegian Centre for Maritime and Diving Medicine, Department of Occupational Medicine (R.K.), Division of Psychiatry (J.Ha.), Haukeland University Hospital, Bergen, Norway
| | - Jozef Hritz
- Department of Biomedicine (K.W., K.J.K.C., S.Pan., A.D., S.Pat., A.M., J.Ha.), Proteomics Unit (PROBE), (A.D.), University of Bergen, Bergen, Norway; Department of Neuroscience, Psychology and Behaviour, College of Medicine, Biological Sciences and Psychology, University of Leicester, Leicester, UK (E.D.V., W.H.J.N.); CEITEC-MU, Masaryk University, Brno, Czech Republic (N.G., J.Hr.); Department of Chemistry, Faculty of Science, Masaryk University, Brno, Czech Republic and Norwegian Centre for Maritime and Diving Medicine, Department of Occupational Medicine (R.K.), Division of Psychiatry (J.Ha.), Haukeland University Hospital, Bergen, Norway
| | - William H J Norton
- Department of Biomedicine (K.W., K.J.K.C., S.Pan., A.D., S.Pat., A.M., J.Ha.), Proteomics Unit (PROBE), (A.D.), University of Bergen, Bergen, Norway; Department of Neuroscience, Psychology and Behaviour, College of Medicine, Biological Sciences and Psychology, University of Leicester, Leicester, UK (E.D.V., W.H.J.N.); CEITEC-MU, Masaryk University, Brno, Czech Republic (N.G., J.Hr.); Department of Chemistry, Faculty of Science, Masaryk University, Brno, Czech Republic and Norwegian Centre for Maritime and Diving Medicine, Department of Occupational Medicine (R.K.), Division of Psychiatry (J.Ha.), Haukeland University Hospital, Bergen, Norway
| | - Aurora Martinez
- Department of Biomedicine (K.W., K.J.K.C., S.Pan., A.D., S.Pat., A.M., J.Ha.), Proteomics Unit (PROBE), (A.D.), University of Bergen, Bergen, Norway; Department of Neuroscience, Psychology and Behaviour, College of Medicine, Biological Sciences and Psychology, University of Leicester, Leicester, UK (E.D.V., W.H.J.N.); CEITEC-MU, Masaryk University, Brno, Czech Republic (N.G., J.Hr.); Department of Chemistry, Faculty of Science, Masaryk University, Brno, Czech Republic and Norwegian Centre for Maritime and Diving Medicine, Department of Occupational Medicine (R.K.), Division of Psychiatry (J.Ha.), Haukeland University Hospital, Bergen, Norway
| | - Jan Haavik
- Department of Biomedicine (K.W., K.J.K.C., S.Pan., A.D., S.Pat., A.M., J.Ha.), Proteomics Unit (PROBE), (A.D.), University of Bergen, Bergen, Norway; Department of Neuroscience, Psychology and Behaviour, College of Medicine, Biological Sciences and Psychology, University of Leicester, Leicester, UK (E.D.V., W.H.J.N.); CEITEC-MU, Masaryk University, Brno, Czech Republic (N.G., J.Hr.); Department of Chemistry, Faculty of Science, Masaryk University, Brno, Czech Republic and Norwegian Centre for Maritime and Diving Medicine, Department of Occupational Medicine (R.K.), Division of Psychiatry (J.Ha.), Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
15
|
Zhu Y, Zhao P, Sun L, Lu Y, Zhu W, Zhang J, Xiang C, Mao Y, Chen Q, Zhang F. Overexpression of circRNA SNRK targets miR-103-3p to reduce apoptosis and promote cardiac repair through GSK3β/β-catenin pathway in rats with myocardial infarction. Cell Death Discov 2021; 7:84. [PMID: 33875647 PMCID: PMC8055694 DOI: 10.1038/s41420-021-00467-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/25/2021] [Accepted: 03/12/2021] [Indexed: 02/06/2023] Open
Abstract
Ischemic cardiomyopathy seriously endangers human health leading to a poor prognosis. Acute myocardial infarction (AMI) is the primary etiology, and the pathophysiological process concludes with the death of cardiomyocytes caused by acute and persistent ischemia and hypoxia in the coronary arteries. We identified a circRNA (circSNRK) which was downregulated in rats with myocardial infarction (MI), however, the role it plays in the MI environment is still unclear. This study contained experiments to investigate the role of circSNRK in the regulation of cardiac survival and explore the mechanisms underlying circSNRK functions. Quantitative real-time PCR (qRT-PCR) was performed to determine the circSNRK expression patterns in hearts. Gain-of-function assays were also conducted in vitro and in vivo to determine the role of circSNRK in cardiac repair. qRT-PCR, western blot, and luciferase reporter assays were used to study circRNA interactions with micro RNAs (miRNAs). Overexpression of circSNRK in cardiomyocytes reduced apoptosis and increased proliferation. Adeno associated virus 9 (AAV9) mediated myocardium overexpression of circSNRK in post MI hearts reduced cardiomyocyte apoptosis, promoted cardiomyocyte proliferation, enhanced angiogenesis, and improved cardiac functions. Overall, upregulation of circSNRK promotes cardiac survival and functional recovery after MI. Mechanistically, circSNRK regulates cardiomyocyte apoptosis and proliferation by acting as a miR-103-3p sponge and inducing increased expression of SNRK which can bind GSK3β to regulate its phosphorylated activity. And thus circSNRK may be a promising therapeutic target for improving clinical prognosis after MI.
Collapse
Affiliation(s)
- Yeqian Zhu
- Section of Pacing and Electrophysiology, Division of Cardiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Pengcheng Zhao
- Section of Pacing and Electrophysiology, Division of Cardiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Ling Sun
- Section of Pacing and Electrophysiology, Division of Cardiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China.,Department of Cardiology, the Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Yao Lu
- Section of Pacing and Electrophysiology, Division of Cardiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Wenwu Zhu
- Section of Pacing and Electrophysiology, Division of Cardiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Jian Zhang
- Section of Pacing and Electrophysiology, Division of Cardiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Chengyu Xiang
- Section of Pacing and Electrophysiology, Division of Cardiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yangming Mao
- Section of Pacing and Electrophysiology, Division of Cardiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Qiushi Chen
- Section of Pacing and Electrophysiology, Division of Cardiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Fengxiang Zhang
- Section of Pacing and Electrophysiology, Division of Cardiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| |
Collapse
|
16
|
Czeleń P, Szefler B. The Oxindole Derivatives, New Promising GSK-3β Inhibitors as One of the Potential Treatments for Alzheimer's Disease-A Molecular Dynamics Approach. BIOLOGY 2021; 10:biology10040332. [PMID: 33920768 PMCID: PMC8071161 DOI: 10.3390/biology10040332] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/13/2021] [Accepted: 04/13/2021] [Indexed: 12/02/2022]
Abstract
Simple Summary Enzymatic overexpression is a determinant of the development of many diseases. Increased activity of the GSK-3β enzyme is a factor that manifests itself in the development of numerous disease entities such as Alzheimer’s disease, schizophrenia, diabetes and cancers. An important medical procedure in such cases is the inhibition of enzyme activity. Based on the comprehensive use of computational chemistry methods, a group of new compounds derived from 2-oxindole was designed. The conducted research allowed the assessment of the conformational properties of the ligand molecules in the GSK-3β active site, the dynamic stability of the obtained complexes and their exact energetic characteristics. Taking into account the obtained data, a narrow group of derivatives showing an affinity for the active site of the GSK-3β enzyme was selected. The comparison of binding properties of selected 2-oxindole derivatives with an inhibitor with confirmed pharmacological activity indicates the high application potential of the newly developed compounds. Abstract The glycogen synthase kinase 3β (GSK-3β) is a protein kinase involved in regulating numerous physiological processes such as embryonic development, transcription, insulin action, cell division cycle and multiple neuronal functions. The overexpression of this enzyme is related to many diseases such as schizophrenia, Alzheimer’s disease, diabetes and cancer. One of the basic methods of treatment in these cases is the usage of ATP-competitive inhibitors. A significant group of such compounds are indirubin and its analogs, e.g., oxindole derivatives. The compounds considered in this work are 112 newly designed oxindole derivatives. In the first stage, such molecular properties of considered compounds as toxicity and LogP were estimated. The preliminary analysis of the binding capabilities of considered compounds towards the GSK-3β active site was conducted with the use of the docking procedure. Based on obtained molecular properties and docking simulations, a selected group of complexes that were analyzed in the molecular dynamics stage was nominated. The proposed procedure allowed for the identification of compounds such as Oxind_4_9 and Oxind_13_10, which create stable complexes with GSK-3β enzyme and are characterized by the highest values of binding affinity. The key interactions responsible for stabilization of considered ligand–protein complexes were identified, and their dynamic stability was also determined. Comparative analysis including analyzed compounds and reference molecule 3a, which is also an oxindole derivative with a confirmed inhibitory potential towards GSK3B protein, clearly indicates that the proposed compounds exhibit an analogous binding mechanism, and the obtained binding enthalpy values indicate a slightly higher binding potential than the reference molecule.
Collapse
|
17
|
Cardiovascular toxicity of PI3Kα inhibitors. Clin Sci (Lond) 2021; 134:2595-2622. [PMID: 33063821 DOI: 10.1042/cs20200302] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/27/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023]
Abstract
The phosphoinositide 3-kinases (PI3Ks) are a family of intracellular lipid kinases that phosphorylate the 3'-hydroxyl group of inositol membrane lipids, resulting in the production of phosphatidylinositol 3,4,5-trisphosphate from phosphatidylinositol 4,5-bisphosphate. This results in downstream effects, including cell growth, proliferation, and migration. The heart expresses three PI3K class I enzyme isoforms (α, β, and γ), and these enzymes play a role in cardiac cellular survival, myocardial hypertrophy, myocardial contractility, excitation, and mechanotransduction. The PI3K pathway is associated with various disease processes but is particularly important to human cancers since many gain-of-function mutations in this pathway occur in various cancers. Despite the development, testing, and regulatory approval of PI3K inhibitors in recent years, there are still significant challenges when creating and utilizing these drugs, including concerns of adverse effects on the heart. There is a growing body of evidence from preclinical studies revealing that PI3Ks play a crucial cardioprotective role, and thus inhibition of this pathway could lead to cardiac dysfunction, electrical remodeling, vascular damage, and ultimately, cardiovascular disease. This review will focus on PI3Kα, including the mechanisms underlying the adverse cardiovascular effects resulting from PI3Kα inhibition and the potential clinical implications of treating patients with these drugs, such as increased arrhythmia burden, biventricular cardiac dysfunction, and impaired recovery from cardiotoxicity. Recommendations for future directions for preclinical and clinical work are made, highlighting the possible role of PI3Kα inhibition in the progression of cancer-related cachexia and female sex and pre-existing comorbidities as independent risk factors for cardiac abnormalities after cancer treatment.
Collapse
|
18
|
Bcl-2 Is Involved in Cardiac Hypertrophy through PI3K-Akt Pathway. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6615502. [PMID: 33778070 PMCID: PMC7979306 DOI: 10.1155/2021/6615502] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/22/2021] [Accepted: 02/18/2021] [Indexed: 12/25/2022]
Abstract
Cardiac hypertrophy (CH) is a common cause of sudden cardiac death and heart failure, resulting in a significant medical burden. The present study is aimed at exploring potential CH-related pathways and the key downstream effectors. The gene expression profile of GSE129090 was obtained from the Gene Expression Omnibus database (GEO), and 1325 differentially expressed genes (DEGs) were identified, including 785 upregulated genes and 540 downregulated genes. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Reactome pathway enrichment analysis of DEGs were then performed. Although there were no pathways enriched by downregulated genes, many CH-related pathways were identified by upregulated genes, including PI3K-Akt signaling pathway, extracellular matrix- (ECM-) receptor interaction, regulation of actin cytoskeleton, and hypertrophic cardiomyopathy (HCM). In the deeper analysis of PI3K-Akt signaling pathway, we found all the signaling transduction pointed to B cell lymphoma-2- (Bcl-2-) mediated cell survival. We then demonstrated that PI3K-Akt signaling pathway was indeed activated in cardiac hypertrophy. Furthermore, no matter LY294002, an inhibitor of the PI3K/AKT signaling pathway, or Venetoclax, a selective Bcl-2 inhibitor, protected against cardiac hypertrophy. In conclusion, these data indicate that Bcl-2 is involved in cardiac hypertrophy as a key downstream effector of PI3K-Akt signaling pathway, suggesting a potential therapeutic target for the clinical management of cardiac hypertrophy.
Collapse
|
19
|
piRNAs as Modulators of Disease Pathogenesis. Int J Mol Sci 2021; 22:ijms22052373. [PMID: 33673453 PMCID: PMC7956838 DOI: 10.3390/ijms22052373] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/24/2021] [Accepted: 02/24/2021] [Indexed: 12/20/2022] Open
Abstract
Advances in understanding disease pathogenesis correlates to modifications in gene expression within different tissues and organ systems. In depth knowledge about the dysregulation of gene expression profiles is fundamental to fully uncover mechanisms in disease development and changes in host homeostasis. The body of knowledge surrounding mammalian regulatory elements, specifically regulators of chromatin structure, transcriptional and translational activation, has considerably surged within the past decade. A set of key regulators whose function still needs to be fully elucidated are small non-coding RNAs (sncRNAs). Due to their broad range of unfolding functions in the regulation of gene expression during transcription and translation, sncRNAs are becoming vital to many cellular processes. Within the past decade, a novel class of sncRNAs called PIWI-interacting RNAs (piRNAs) have been implicated in various diseases, and understanding their complete function is of vital importance. Historically, piRNAs have been shown to be indispensable in germline integrity and stem cell development. Accumulating research evidence continue to reveal the many arms of piRNA function. Although piRNA function and biogenesis has been extensively studied in Drosophila, it is thought that they play similar roles in vertebrate species, including humans. Compounding evidence suggests that piRNAs encompass a wider functional range than small interfering RNAs (siRNAs) and microRNAs (miRNAs), which have been studied more in terms of cellular homeostasis and disease. This review aims to summarize contemporary knowledge regarding biogenesis, and homeostatic function of piRNAs and their emerging roles in the development of pathologies related to cardiomyopathies, cancer, and infectious diseases.
Collapse
|
20
|
Oh Y, Yang S, Liu X, Jana S, Izaddoustdar F, Gao X, Debi R, Kim DK, Kim KH, Yang P, Kassiri Z, Lakin R, Backx PH. Transcriptomic Bioinformatic Analyses of Atria Uncover Involvement of Pathways Related to Strain and Post-translational Modification of Collagen in Increased Atrial Fibrillation Vulnerability in Intensely Exercised Mice. Front Physiol 2020; 11:605671. [PMID: 33424629 PMCID: PMC7793719 DOI: 10.3389/fphys.2020.605671] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023] Open
Abstract
Atrial Fibrillation (AF) is the most common supraventricular tachyarrhythmia that is typically associated with cardiovascular disease (CVD) and poor cardiovascular health. Paradoxically, endurance athletes are also at risk for AF. While it is well-established that persistent AF is associated with atrial fibrosis, hypertrophy and inflammation, intensely exercised mice showed similar adverse atrial changes and increased AF vulnerability, which required tumor necrosis factor (TNF) signaling, even though ventricular structure and function improved. To identify some of the molecular factors underlying the chamber-specific and TNF-dependent atrial changes induced by exercise, we performed transcriptome analyses of hearts from wild-type and TNF-knockout mice following exercise for 2 days, 2 or 6 weeks of exercise. Consistent with the central role of atrial stretch arising from elevated venous pressure in AF promotion, all 3 time points were associated with differential regulation of genes in atria linked to mechanosensing (focal adhesion kinase, integrins and cell-cell communications), extracellular matrix (ECM) and TNF pathways, with TNF appearing to play a permissive, rather than causal, role in gene changes. Importantly, mechanosensing/ECM genes were only enriched, along with tubulin- and hypertrophy-related genes after 2 days of exercise while being downregulated at 2 and 6 weeks, suggesting that early reactive strain-dependent remodeling with exercise yields to compensatory adjustments. Moreover, at the later time points, there was also downregulation of both collagen genes and genes involved in collagen turnover, a pattern mirroring aging-related fibrosis. By comparison, twofold fewer genes were differentially regulated in ventricles vs. atria, independently of TNF. Our findings reveal that exercise promotes TNF-dependent atrial transcriptome remodeling of ECM/mechanosensing pathways, consistent with increased preload and atrial stretch seen with exercise. We propose that similar preload-dependent mechanisms are responsible for atrial changes and AF in both CVD patients and athletes.
Collapse
Affiliation(s)
- Yena Oh
- Department of Biology, York University, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Sibao Yang
- Department of Biology, York University, Toronto, ON, Canada.,Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xueyan Liu
- Department of Biology, York University, Toronto, ON, Canada.,Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Sayantan Jana
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, AB, Canada
| | | | - Xiaodong Gao
- Department of Biology, York University, Toronto, ON, Canada
| | - Ryan Debi
- Department of Biology, York University, Toronto, ON, Canada
| | - Dae-Kyum Kim
- Donnelly Centre, University of Toronto, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Kyoung-Han Kim
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Ping Yang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zamaneh Kassiri
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, AB, Canada
| | - Robert Lakin
- Department of Biology, York University, Toronto, ON, Canada
| | - Peter H Backx
- Department of Biology, York University, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
21
|
Boileau E, Doroudgar S, Riechert E, Jürgensen L, Ho TC, Katus HA, Völkers M, Dieterich C. A Multi-Network Comparative Analysis of Transcriptome and Translatome Identifies Novel Hub Genes in Cardiac Remodeling. Front Genet 2020; 11:583124. [PMID: 33304386 PMCID: PMC7701244 DOI: 10.3389/fgene.2020.583124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/09/2020] [Indexed: 12/03/2022] Open
Abstract
Our understanding of the transition from physiological to pathological cardiac hypertrophy remains elusive and largely based on reductionist hypotheses. Here, we profiled the translatomes of 15 mouse hearts to provide a molecular blueprint of altered gene networks in early cardiac remodeling. Using co-expression analysis, we showed how sub-networks are orchestrated into functional modules associated with pathological phenotypes. We discovered unappreciated hub genes, many undocumented for their role in cardiac hypertrophy, and genes in the transcriptional network that were rewired in the translational network, and associated with semantically different subsets of enriched functional terms, such as Fam210a, a novel musculoskeletal modulator, or Psmd12, implicated in protein quality control. Using their correlation structure, we found that transcriptome networks are only partially reproducible at the translatome level, providing further evidence of post-transcriptional control at the level of translation. Our results provide novel insights into the complexity of the organization of in vivo cardiac regulatory networks.
Collapse
Affiliation(s)
- Etienne Boileau
- Section of Bioinformatics and Systems Cardiology, Klaus Tschira Institute for Integrative Computational Cardiology, Heidelberg, Germany.,Department of Internal Medicine III (Cardiology, Angiology, and Pneumology), University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
| | - Shirin Doroudgar
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology), University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
| | - Eva Riechert
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology), University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
| | - Lonny Jürgensen
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology), University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
| | - Thanh Cao Ho
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology), University Hospital Heidelberg, Heidelberg, Germany
| | - Hugo A Katus
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology), University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
| | - Mirko Völkers
- Department of Internal Medicine III (Cardiology, Angiology, and Pneumology), University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
| | - Christoph Dieterich
- Section of Bioinformatics and Systems Cardiology, Klaus Tschira Institute for Integrative Computational Cardiology, Heidelberg, Germany.,Department of Internal Medicine III (Cardiology, Angiology, and Pneumology), University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
| |
Collapse
|
22
|
Romic S, Djordjevic A, Tepavcevic S, Culafic T, Stojiljkovic M, Bursac B, Stanisic J, Kostic M, Gligorovska L, Koricanac G. Effects of a fructose-rich diet and chronic stress on insulin signaling and regulation of glycogen synthase kinase-3 beta and the sodium-potassium pump in the hearts of male rats. Food Funct 2020; 11:1455-1466. [PMID: 31974538 DOI: 10.1039/c9fo02306b] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Both a diet rich in fructose and chronic stress exposure induce metabolic and cardiovascular disturbances. The aim of this study was to examine the effects of the fructose-rich diet and chronic stress, separately and in combination, on insulin signaling and molecules regulating glycogen synthesis and ion transport in the heart, and to reveal whether these effects coincide with changes in glucocorticoid receptor (GR) activation. Male Wistar rats were subjected to 10% fructose in drinking water and/or to chronic unpredictable stress for 9 weeks. Protein expression and/or phosphorylation of the insulin receptor (IR), protein tyrosine phosphatase 1B, insulin receptor substrate 1 (IRS1), protein kinase B (Akt), extracellular signal-regulated kinase 1/2 (ERK1/2), glycogen synthase kinase-3β (GSK-3β) and Na+/K+-ATPase α-subunits in cardiac tissue were analyzed by western blot. GR distribution between cytosolic and nuclear fractions was also analyzed. The fructose-rich diet decreased the level of pERK1/2 (Thr202/Tyr204) and pGSK-3β (Ser9) independently of stress, while chronic stress increased the IRS1 content and prevented the fructose diet-induced decrease of the pAkt (Ser473) level. The fructose-rich diet in combination with chronic stress reduced the protein content of cardiac IR and attenuated IRS1 upregulation. Separate treatments increased the protein content of Na+/K+-ATPase α1- and α2-subunits, while after combined treatment the α2 content was at the control level and the α1 content was lower than the control level. The effect of combined treatment on cardiac IR and α2-subunit expression could be mediated by increased GR nuclear accumulation. Our study provides new insights into the effects of chronic stress and a combination of the fructose diet and chronic stress on the studied molecules in the heart.
Collapse
Affiliation(s)
- Snjezana Romic
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
The piRNA CHAPIR regulates cardiac hypertrophy by controlling METTL3-dependent N 6-methyladenosine methylation of Parp10 mRNA. Nat Cell Biol 2020; 22:1319-1331. [PMID: 33020597 DOI: 10.1038/s41556-020-0576-y] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 08/11/2020] [Indexed: 12/21/2022]
Abstract
PIWI-interacting RNAs (piRNAs) are abundantly expressed during cardiac hypertrophy. However, their functions and molecular mechanisms remain unknown. Here, we identified a cardiac-hypertrophy-associated piRNA (CHAPIR) that promotes pathological hypertrophy and cardiac remodelling by targeting METTL3-mediated N6-methyladenosine (m6A) methylation of Parp10 mRNA transcripts. CHAPIR deletion markedly attenuates cardiac hypertrophy and restores heart function, while administration of a CHAPIR mimic enhances the pathological hypertrophic response in pressure-overloaded mice. Mechanistically, CHAPIR-PIWIL4 complexes directly interact with METTL3 and block the m6A methylation of Parp10 mRNA transcripts, which upregulates PARP10 expression. The CHAPIR-dependent increase in PARP10 promotes the mono-ADP-ribosylation of GSK3β and inhibits its kinase activity, which results in the accumulation of nuclear NFATC4 and the progression of pathological hypertrophy. Hence, our findings reveal that a piRNA-mediated RNA epigenetic mechanism is involved in the regulation of cardiac hypertrophy and that the CHAPIR-METTL3-PARP10-NFATC4 signalling axis could be therapeutically targeted for treating pathological hypertrophy and maladaptive cardiac remodelling.
Collapse
|
24
|
Ashraf S, Yilmaz G, Chen X, Harmancey R. Dietary Fat and Sugar Differentially Affect β-Adrenergic Stimulation of Cardiac ERK and AKT Pathways in C57BL/6 Male Mice Subjected to High-Calorie Feeding. J Nutr 2020; 150:1041-1050. [PMID: 31950177 PMCID: PMC7198302 DOI: 10.1093/jn/nxz342] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/16/2019] [Accepted: 12/23/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND High dietary fat and sugar promote cardiac hypertrophy independently from an increase in blood pressure. The respective contribution that each macronutrient exerts on cardiac growth signaling pathways remains unclear. OBJECTIVE The goal of this study was to investigate the mechanisms by which high amounts of dietary fat and sugar affect cardiac growth regulatory pathways. METHODS Male C57BL/6 mice (9 wk old; n = 20/group) were fed a standard rodent diet (STD; kcal% protein-fat-carbohydrate, 29-17-54), a high-fat diet (HFD; 20-60-20), a high-fat and high-sugar Western diet (WD; 20-45-35), a high-sugar diet with mixed carbohydrates (HCD; 20-10-70), or a high-sucrose diet (HSD; 20-10-70). Body composition was assessed weekly by EchoMRI. Whole-body glucose utilization was assessed with an intraperitoneal glucose tolerance test. After 6 wk on diets, mice were treated with saline or 20 mg/kg isoproterenol (ISO), and the activity of cardiac growth regulatory pathways was analyzed by immunoblotting. Data were analyzed by ANOVA with data from the STD group included for references only. RESULTS Compared with HCD and HSD, WD and HFD increased body fat mass 2.7- to 3.8-fold (P < 0.001), induced glucose intolerance (P < 0.001), and increased insulin concentrations >1.5-fold (P < 0.05), thereby enhancing basal and ISO-stimulated AKT phosphorylation at both threonine 308 and serine 473 residues (+25-63%; P < 0.05). Compared with HFD, the high-sugar diets potentiated ISO-mediated stimulation of the glucose-sensitive kinases PYK2 (>47%; P < 0.05 for HCD and HSD) and ERK (>34%; P < 0.05 for WD, HCD, and HSD), thereby leading to increased phosphorylation of protein synthesis regulator S6K1 at threonine 389 residue (>64%; P < 0.05 for WD, HCD, and HSD). CONCLUSIONS Dietary fat and sugar affect cardiac growth signaling pathways in C57BL/6 mice through distinct and additive mechanisms. The findings may provide new insights into the role of overnutrition in pathological cardiac remodeling.
Collapse
Affiliation(s)
- Sadia Ashraf
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS,Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS
| | - Gizem Yilmaz
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS,Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS
| | - Xu Chen
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS,Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS
| | - Romain Harmancey
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS,Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS,Address correspondence to RH (e-mail: )
| |
Collapse
|
25
|
Abstract
The finding of "glycogen synthase kinase-3" (GSK-3) was initially identified as a protein kinase that phosphorylate and inhibited glycogen synthase. However, it was soon discovered that GSK-3 also has significant impact in regulation of truly astonishing number of critical intracellular signaling pathways ranging from regulation of cell growth, neurology, heart failure, diabetes, aging, inflammation, and cancer. Recent studies have validated the feasibility of targeting GSK-3 for its vital therapeutic potential to maintain normal myocardial homeostasis, conversely, its loss is incompatible with life as it can abrupt cell cycle and endorse fatal cardiomyopathy. The current study focuses on its expanding therapeutic action in myocardial tissue, concentrating primarily on its role in diabetes-associated cardiac complication, apoptosis and metabolism, heart failure, cardiac hypertrophy, and myocardial infarction. The current report also includes the finding of our previous investigation that has shown the impact of GSK-3β inhibitor against diabetes-associated myocardial injury and experimentally induced myocardial infarction. We have also discussed some recent identified GSK-3β inhibitors for their cardio-protective potential. The crosstalk of various underlying mechanisms that highlight the significant role of GSK-3β in myocardial pathophysiology have been discussed in the present report. For these literatures, we will rely profoundly on our previous studies and those of others to reconcile some of the deceptive contradictions in the literature.
Collapse
|
26
|
Hamstra SI, Kurgan N, Baranowski RW, Qiu L, Watson CJF, Messner HN, MacPherson REK, MacNeil AJ, Roy BD, Fajardo VA. Low-dose lithium feeding increases the SERCA2a-to-phospholamban ratio, improving SERCA function in murine left ventricles. Exp Physiol 2020; 105:666-675. [PMID: 32087034 DOI: 10.1113/ep088061] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 02/21/2020] [Indexed: 12/24/2022]
Abstract
NEW FINDINGS What is the central question of this study? Inhibition of glycogen synthase kinase-3 (GSK3) has been shown to improve cardiac SERCA2a function. Lithium can inhibit GSK3, but therapeutic doses used in treating bipolar disorder can have toxic effects. It has not been determined whether subtherapeutic doses of lithium can improve cardiac SERCA function. What is the main finding and its importance? Using left ventricles from wild-type mice, we found that subtherapeutic lithium feeding for 6 weeks decreased GSK3 activity and increased cardiac SERCA function compared with control-fed mice. These findings warrant the investigation of low-dose lithium feeding in preclinical models of cardiomyopathy and heart failure to determine the therapeutic benefit of GSK3 inhibition. ABSTRACT The sarco(endo)plasmic reticulum Ca2+ -ATPase (SERCA) pump is responsible for regulating calcium (Ca2+ ) within myocytes, with SERCA2a being the dominant isoform in cardiomyocytes. Its inhibitor, phospholamban (PLN), acts by decreasing the affinity of SERCA for Ca2+ . Changes in the SERCA2a:PLN ratio can cause Ca2+ dysregulation often seen in patients with dilated cardiomyopathy and heart failure. The enzyme glycogen synthase kinase-3 (GSK3) is known to downregulate SERCA function by decreasing the SERCA2a:PLN ratio. In this study, we sought to determine whether feeding mice low-dose lithium, a natural GSK3 inhibitor, would improve left ventricular SERCA function by altering the SERCA2a:PLN ratio. To this end, male wild-type C57BL/6J mice were fed low-dose lithium via drinking water (10 mg kg-1 day-1 LiCl for 6 weeks) and left ventricles were harvested. GSK3 activity was significantly reduced in LiCl-fed versus control-fed mice. The apparent affinity of SERCA for Ca2+ was also increased (pCa50 ; control, 6.09 ± 0.03 versus LiCl, 6.26 ± 0.04, P < 0.0001) along with a 2.0-fold increase in SERCA2a:PLN ratio in LiCl-fed versus control-fed mice. These findings suggest that low-dose lithium supplementation can improve SERCA function by increasing the SERCA2a:PLN ratio. Future studies in murine preclinical models will determine whether GSK3 inhibition via low-dose lithium could be a potential therapeutic strategy for dilated cardiomyopathy and heart failure.
Collapse
Affiliation(s)
- Sophie I Hamstra
- Department of Kinesiology, Brock University, St. Catharines, ON, L2S 3A1, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Nigel Kurgan
- Department of Kinesiology, Brock University, St. Catharines, ON, L2S 3A1, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Ryan W Baranowski
- Department of Kinesiology, Brock University, St. Catharines, ON, L2S 3A1, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Liqun Qiu
- Department of Chemistry, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Colton J F Watson
- Department of Health Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Holt N Messner
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | | | - Adam J MacNeil
- Department of Health Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Brian D Roy
- Department of Kinesiology, Brock University, St. Catharines, ON, L2S 3A1, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Val A Fajardo
- Department of Kinesiology, Brock University, St. Catharines, ON, L2S 3A1, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, L2S 3A1, Canada
| |
Collapse
|
27
|
Gao J, Zhang K, Wang Y, Guo R, Liu H, Jia C, Sun X, Wu C, Wang W, Du J, Chen J. A machine learning-driven study indicates emodin improves cardiac hypertrophy by modulation of mitochondrial SIRT3 signaling. Pharmacol Res 2020; 155:104739. [PMID: 32135248 DOI: 10.1016/j.phrs.2020.104739] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/26/2020] [Accepted: 02/29/2020] [Indexed: 02/06/2023]
Abstract
Cardiac hypertrophy (CH) is an enormous risk factor in the process of heart failure development, however, there is still lack of effective treatment for CH. Mitochondrial protection is an effective way against CH. Rheum palmatum L. (rhubarb) has been used to treat chronic heart diseases such as heart failure, especially to inhibit cardiac compensatory enlargement. The aim of this study was to explore the pharmacodynamic component of rhubarb and reveal its pharmacological effects and targets in the treatment of CH. Based on network pharmacology and machine learning approach, ingredients of rhubarb and targets for CH were extracted and surflex docking was conducted for obtaining the optimal ingredient-target combination(s) and emodin-SIRT3 was identified for further functional analysis. Transverse aortic constriction or isoproterenol induced CH mice and phenylephrine injured cardiomyocytes were used to verify the mitochondria protection effect and CH improvement of emodin in vivo and in vitro by modulation of mitochondrial SIRT3 signaling. The results showed that emodin could block agonist-induced and pressure overload-mediated CH. Emodin prevented mitochondrial dysfunction and its underlying mechanism was attributed to the activation of SIRT3, but the effect was not obvious with the presence of SIRT3 inhibitors (3-TYP)/SIRT3 siRNA. Furthermore, PGC-1ɑ was involved in the process of emodin regulating SIRT3 signaling pathway as an upstream target. Our findings clarified the main material basis and mechanism of rhubarb in the treatment of CH. Emodin, as the major ingredient of rhubarb, has therapeutic potential for CH through mitochondrial protection due to the modulation of SIRT3 signaling.
Collapse
Affiliation(s)
- Jian Gao
- Beijing University of Chinese Medicine, Beijing, 100029, China; The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Kunlin Zhang
- Center for Genetics and BioMedical Informatics Research, CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yi Wang
- Zhejiang University, 866 Yuhangtang Rd, Hangzhou, 310058, China
| | - Rui Guo
- Zhejiang University, 866 Yuhangtang Rd, Hangzhou, 310058, China
| | - Hao Liu
- Zhejiang University, 866 Yuhangtang Rd, Hangzhou, 310058, China
| | - Caixia Jia
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiaoli Sun
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Chaoyong Wu
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wei Wang
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jie Du
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100029, China; Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Jianxin Chen
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
28
|
Role of Akt Activation in PARP Inhibitor Resistance in Cancer. Cancers (Basel) 2020; 12:cancers12030532. [PMID: 32106627 PMCID: PMC7139751 DOI: 10.3390/cancers12030532] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/19/2020] [Accepted: 02/24/2020] [Indexed: 12/12/2022] Open
Abstract
Poly(ADP-ribose) polymerase (PARP) inhibitors have recently been introduced in the therapy of several types of cancers not responding to conventional treatments. However, de novo and acquired PARP inhibitor resistance is a significant limiting factor in the clinical therapy, and the underlying mechanisms are not fully understood. Activity of the cytoprotective phosphatidylinositol-3 kinase (PI3K)-Akt pathway is often increased in human cancer that could result from mutation, expressional change, or amplification of upstream growth-related factor signaling elements or elements of the Akt pathway itself. However, PARP-inhibitor-induced activation of the cytoprotective PI3K-Akt pathway is overlooked, although it likely contributes to the development of PARP inhibitor resistance. Here, we briefly summarize the biological role of the PI3K-Akt pathway. Next, we overview the significance of the PARP-Akt interplay in shock, inflammation, cardiac and cerebral reperfusion, and cancer. We also discuss a recently discovered molecular mechanism that explains how PARP inhibition induces Akt activation and may account for apoptosis resistance and mitochondrial protection in oxidative stress and in cancer.
Collapse
|
29
|
Lima VM, Lino CA, Senger N, de Oliveira Silva T, Fonseca RIB, Bader M, Santos RAS, Júnior JD, Barreto-Chaves MLM, Diniz GP. Angiotensin II type 2 receptor mediates high fat diet-induced cardiomyocyte hypertrophy and hypercholesterolemia. Mol Cell Endocrinol 2019; 498:110576. [PMID: 31520674 DOI: 10.1016/j.mce.2019.110576] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/05/2019] [Accepted: 09/09/2019] [Indexed: 12/30/2022]
Abstract
Obesity is the major risk factor for several cardiovascular and metabolic disorders. Previous studies reported that deletion of Angiotensin II type 2 receptor (AT2R) protects against metabolic dysfunctions induced by high fat (HF) diet. However, the role of AT2R in obesity-induced cardiac hypertrophy remains unclear. Male AT2R knockout (AT2RKO) and wild type (AT2RWT) mice were fed with control or HF diet for 10 weeks. HF diet increased cardiac expression of AT2R in obese mice. Deletion of AT2R did not affect body weight gain, glucose intolerance and fat mass gain induced by HF feeding. However, loss of AT2R prevented HF diet-induced hypercholesterolemia and cardiac remodeling. Mechanistically, we found that pharmacological inhibition or knockdown of AT2R prevented leptin-induced cardiomyocyte hypertrophy in vitro. Collectively, our results suggest that AT2R is involved in obesity-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- Vanessa M Lima
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Caroline A Lino
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Nathalia Senger
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Renata I B Fonseca
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Michael Bader
- Max-Delbruck-Center for Molecular Medicine, Berlin, Germany
| | - Robson A S Santos
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Jose Donato Júnior
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Gabriela P Diniz
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
30
|
Involvement of fatty acid synthase in right ventricle dysfunction in pulmonary hypertension. Exp Cell Res 2019; 383:111569. [DOI: 10.1016/j.yexcr.2019.111569] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 08/09/2019] [Accepted: 08/20/2019] [Indexed: 10/26/2022]
|
31
|
HDAC3 Mediates Cardioprotection of Remifentanil Postconditioning by Targeting GSK-3β in H9c2 Cardiomyocytes in Hypoxia/Reoxygenation Injury. Shock 2019; 50:240-247. [PMID: 28957873 DOI: 10.1097/shk.0000000000001008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Remifentanil postconditioning (RPC) confers robust cardioprotection against ischemia/reperfusion (I/R) injury. We recently determined that HDAC3 was involved in RPC-induced cardioprotection. However, the role of HDAC3 and its possible mechanisms in RPC-induced cardioprotection are unknown, which we aimed to evaluate in an in vitro hypoxia/reoxygenation (HR) model. METHODS Myocardium I/R injury was established after HR with H9c2 cardiomyoblasts. Cell viability and apoptosis were evaluated usingCCK-8 and flow cytometry of HR-injured cardiomyoblasts treated with or without RPC. Furthermore, effects of RPC on HDAC3 protein and mRNA expression were evaluated with Western blot and quantitative real-time PCR analyses, whereas GSK-3β expression was measured with Western blot. RESULTS RPC increased cell viability and reduced cell apoptosis (P < 0.05) in H9c2 cardiomyoblasts subjected to HR injury. In addition, RPC promoted the phosphorylation of GSK-3β at Ser9 site (P < 0.05) and suppressed the protein and mRNA expression of HDAC3 (P < 0.05). Lentiviral-transduced overexpression of HDAC3 had no significant effects on HR injury while attenuating the cardioprotective effects of RPC on cell viability and apoptosis (P < 0.05), GSK-3β phosphorylation (P < 0.05) in H9c2 cardiomyoblasts. CONCLUSIONS RPC attenuates apoptosis in H9c2 cardiomyoblasts after HR injury by downregulating HDAC3-mediated phosphorylation of GSK-3β. Our findings suggest that HDAC3, and its cross talk function with GSK-3β, may be a promising target for myocardium I/R injury.
Collapse
|
32
|
Molecular Mechanisms of Cardiac Remodeling and Regeneration in Physical Exercise. Cells 2019; 8:cells8101128. [PMID: 31547508 PMCID: PMC6829258 DOI: 10.3390/cells8101128] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/19/2019] [Accepted: 09/19/2019] [Indexed: 02/08/2023] Open
Abstract
Regular physical activity with aerobic and muscle-strengthening training protects against the occurrence and progression of cardiovascular disease and can improve cardiac function in heart failure patients. In the past decade significant advances have been made in identifying mechanisms of cardiomyocyte re-programming and renewal including an enhanced exercise-induced proliferational capacity of cardiomyocytes and its progenitor cells. Various intracellular mechanisms mediating these positive effects on cardiac function have been found in animal models of exercise and will be highlighted in this review. 1) activation of extracellular and intracellular signaling pathways including phosphatidylinositol 3 phosphate kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR), EGFR/JNK/SP-1, nitric oxide (NO)-signaling, and extracellular vesicles; 2) gene expression modulation via microRNAs (miR), in particular via miR-17-3p and miR-222; and 3) modulation of cardiac cellular metabolism and mitochondrial adaption. Understanding the cellular mechanisms, which generate an exercise-induced cardioprotective cellular phenotype with physiological hypertrophy and enhanced proliferational capacity may give rise to novel therapeutic targets. These may open up innovative strategies to preserve cardiac function after myocardial injury as well as in aged cardiac tissue.
Collapse
|
33
|
Bonezzi F, Piccoli M, Dei Cas M, Paroni R, Mingione A, Monasky MM, Caretti A, Riganti C, Ghidoni R, Pappone C, Anastasia L, Signorelli P. Sphingolipid Synthesis Inhibition by Myriocin Administration Enhances Lipid Consumption and Ameliorates Lipid Response to Myocardial Ischemia Reperfusion Injury. Front Physiol 2019; 10:986. [PMID: 31447688 PMCID: PMC6696899 DOI: 10.3389/fphys.2019.00986] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/15/2019] [Indexed: 12/19/2022] Open
Abstract
Myocardial infarct requires prompt thrombolytic therapy or primary percutaneous coronary intervention to limit the extent of necrosis, but reperfusion creates additional damage. Along with reperfusion, a maladaptive remodeling phase might occur and it is often associated with inflammation, oxidative stress, as well as a reduced ability to recover metabolism homeostasis. Infarcted individuals can exhibit reduced lipid turnover and their accumulation in cardiomyocytes, which is linked to a deregulation of peroxisome proliferator activated receptors (PPARs), controlling fatty acids metabolism, energy production, and the anti-inflammatory response. We previously demonstrated that Myriocin can be effectively used as post-conditioning therapeutic to limit ischemia/reperfusion-induced inflammation, oxidative stress, and infarct size, in a murine model. In this follow-up study, we demonstrate that Myriocin has a critical regulatory role in cardiac remodeling and energy production, by up-regulating the transcriptional factor EB, PPARs nuclear receptors and genes involved in fatty acids metabolism, such as VLDL receptor, Fatp1, CD36, Fabp3, Cpts, and mitochondrial FA dehydrogenases. The overall effects are represented by an increased β–oxidation, together with an improved electron transport chain and energy production. The potent immunomodulatory and metabolism regulatory effects of Myriocin elicit the molecule as a promising pharmacological tool for post-conditioning therapy of myocardial ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Fabiola Bonezzi
- Stem Cells for Tissue Engineering Laboratory, IRCCS Policlinico San Donato, Milan, Italy
| | - Marco Piccoli
- Stem Cells for Tissue Engineering Laboratory, IRCCS Policlinico San Donato, Milan, Italy
| | - Michele Dei Cas
- Clinical Biochemistry and Mass Spectrometry Laboratory, Health Sciences Department, University of Milan, Milan, Italy
| | - Rita Paroni
- Clinical Biochemistry and Mass Spectrometry Laboratory, Health Sciences Department, University of Milan, Milan, Italy
| | - Alessandra Mingione
- Biochemistry and Molecular Biology Laboratory, Health Sciences Department, University of Milan, Milan, Italy
| | | | - Anna Caretti
- Biochemistry and Molecular Biology Laboratory, Health Sciences Department, University of Milan, Milan, Italy
| | - Chiara Riganti
- Cell Biochemistry Laboratory, Oncology Department, and Interdepartmental Research Center for Molecular Biotechnology, University of Turin, Turin, Italy
| | - Riccardo Ghidoni
- Biochemistry and Molecular Biology Laboratory, Health Sciences Department, University of Milan, Milan, Italy
| | - Carlo Pappone
- Arrhythmology Department, IRCCS Policlinico San Donato, Milan, Italy
| | - Luigi Anastasia
- Stem Cells for Tissue Engineering Laboratory, IRCCS Policlinico San Donato, Milan, Italy.,Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Paola Signorelli
- Biochemistry and Molecular Biology Laboratory, Health Sciences Department, University of Milan, Milan, Italy
| |
Collapse
|
34
|
Agbo E, Liu D, Li M, Saahene RO, Chen L, Zhao L, Wang Y, Tian G. Modulation of PTEN by hexarelin attenuates coronary artery ligation-induced heart failure in rats. Turk J Med Sci 2019; 49:945-958. [PMID: 31091855 PMCID: PMC7018219 DOI: 10.3906/sag-1812-49] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Background/aim Hexarelin is a synthetic growth hormone-releasing peptide that exerts cardioprotective effects. However, its cardioprotective effect against heart failure (HF) is yet to be explained. This study investigated the therapeutic role of hexarelin and the mechanisms underlying its cardioprotective effects against coronary artery ligation (CAL)-induced HF in rats. Materials and methods Rats with four weeks of permanent CAL, induced myocardial infarction, and HF were randomly separated into four groups: the control group (Ctrl), sham group (Sham), hexarelin treatment group (HF + Hx), and heart failure group (HF). The rats were treated with subcutaneous injection of hexarelin (100 µg/kg) in the treatment group or saline in the other groups twice a day for 30 days. Left ventricular (LV) function, oxidative stress, apoptosis, molecular analyses, and cardiac structural and pathological changes in rats were assessed. Results The treatment of HF rats with hexarelin significantly induced the upregulation of phosphatase and tensin homologue (PTEN) expression and inhibited the phosphorylation of protein kinase B (Akt) and mammalian target of rapamycin (mTOR) to significantly improve LV function, ameliorate myocardial remodeling, and reduce oxidative stress. Conclusion These findings indicate that hexarelin attenuates CAL-induced HF in rats by ameliorating myocardial remodeling, LV dysfunction, and oxidative stress via the upmodulation of PTEN signaling and downregulation of the Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Elvis Agbo
- Department of Human Anatomy, Histology, and Embryology, College of Basic Medicine, Jiamusi University, Jiamusi, P.R. China
| | - Donhai Liu
- College of Basic Medicine, Jiamusi University, Jiamusi, P.R. China
| | - Meixiu Li
- Department of Human Anatomy, Histology, and Embryology, College of Basic Medicine, Jiamusi University, Jiamusi, P.R. China
| | - Roland Osei Saahene
- Department of Immunology, College of Basic Medicine, Jiamusi University, Jiamusi, P.R. China
| | - Liqiang Chen
- Department of Human Anatomy, Histology, and Embryology, College of Basic Medicine, Jiamusi University, Jiamusi, P.R. China
| | - Lunpeng Zhao
- Department of Human Anatomy, Histology, and Embryology, College of Basic Medicine, Jiamusi University, Jiamusi, P.R. China
| | - Yiquan Wang
- Department of Human Anatomy, Histology, and Embryology, College of Basic Medicine, Jiamusi University, Jiamusi, P.R. China
| | - Guozhong Tian
- Department of Human Anatomy, Histology, and Embryology, College of Basic Medicine, Jiamusi University, Jiamusi, P.R. China
| |
Collapse
|
35
|
Loonat AA, Curtis MK, Richards MA, Nunez-Alonso G, Michl J, Swietach P. A high-throughput ratiometric method for imaging hypertrophic growth in cultured primary cardiac myocytes. J Mol Cell Cardiol 2019; 130:184-196. [PMID: 30986378 PMCID: PMC6520438 DOI: 10.1016/j.yjmcc.2019.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/14/2019] [Accepted: 04/04/2019] [Indexed: 12/21/2022]
Abstract
Maladaptive hypertrophy of cardiac myocytes increases the risk of heart failure. The underlying signaling can be triggered and interrogated in cultured neonatal ventricular myocytes (NRVMs) using sophisticated pharmacological and genetic techniques. However, the methods for quantifying cell growth are, by comparison, inadequate. The lack of quantitative, calibratable and computationally-inexpensive high-throughput technology has limited the scope for using cultured myocytes in large-scale analyses. We present a ratiometric method for quantifying the hypertrophic growth of cultured myocytes, compatible with high-throughput imaging platforms. Protein biomass was assayed from sulforhodamine B (SRB) fluorescence, and image analysis calculated the quotient of signal from extra-nuclear and nuclear regions. The former readout relates to hypertrophic growth, whereas the latter is a reference for correcting protein-independent (e.g. equipment-related) variables. This ratiometric measure, when normalized to the number of cells, provides a robust quantification of cellular hypertrophy. The method was tested by comparing the efficacy of various chemical agonists to evoke hypertrophy, and verified using independent assays (myocyte area, transcripts of markers). The method's high resolving power and wide dynamic range were confirmed by the ability to generate concentration-response curves, track the time-course of hypertrophic responses with fine temporal resolution, describe drug/agonist interactions, and screen for novel anti-hypertrophic agents. The method can be implemented as an end-point in protocols investigating hypertrophy, and is compatible with automated plate-reader platforms for generating high-throughput data, thereby reducing investigator-bias. Finally, the computationally-minimal workflow required for obtaining measurements makes the method simple to implement in most laboratories. Maladaptive hypertrophy of myocytes can lead to heart failure. Common methods for tracking growth in cultured myocytes are inadequate. We design and test a method for tracking myocyte hypertrophy in vitro. The method provides a ratiometric index of growth for high throughput analyses. Using the method, we characterize further details of (anti)hypertrophic responses.
Collapse
Affiliation(s)
- Aminah A Loonat
- University of Oxford, Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, United Kingdom
| | - M Kate Curtis
- University of Oxford, Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, United Kingdom
| | - Mark A Richards
- University of Oxford, Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, United Kingdom
| | - Graciela Nunez-Alonso
- University of Oxford, Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, United Kingdom
| | - Johanna Michl
- University of Oxford, Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, United Kingdom
| | - Pawel Swietach
- University of Oxford, Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, United Kingdom.
| |
Collapse
|
36
|
Zhao Y, Huang W, Kim TM, Jung Y, Menon LG, Xing H, Li H, Carroll RS, Park PJ, Yang HW, Johnson MD. MicroRNA-29a activates a multi-component growth and invasion program in glioblastoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:36. [PMID: 30683134 PMCID: PMC6347789 DOI: 10.1186/s13046-019-1026-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/06/2019] [Indexed: 12/12/2022]
Abstract
Background Glioblastoma is a malignant brain tumor characterized by rapid growth, diffuse invasion and therapeutic resistance. We recently used microRNA expression profiles to subclassify glioblastoma into five genetically and clinically distinct subclasses, and showed that microRNAs both define and contribute to the phenotypes of these subclasses. Here we show that miR-29a activates a multi-faceted growth and invasion program that promotes glioblastoma aggressiveness. Methods microRNA expression profiles from 197 glioblastomas were analyzed to identify the candidate miRNAs that are correlated to glioblastoma aggressiveness. The candidate miRNA, miR-29a, was further studied in vitro and in vivo. Results Members of the miR-29 subfamily display increased expression in the two glioblastoma subclasses with the worst prognoses (astrocytic and neural). We observed that miR-29a is among the microRNAs that are most positively-correlated with PTEN copy number in glioblastoma, and that miR-29a promotes glioblastoma growth and invasion in part by targeting PTEN. In PTEN-deficient glioblastoma cells, however, miR-29a nevertheless activates AKT by downregulating the metastasis suppressor, EphB3. In addition, miR-29a robustly promotes invasion in PTEN-deficient glioblastoma cells by repressing translation of the Sox4 transcription factor, and this upregulates the invasion-promoting protein, HIC5. Indeed, we identified Sox4 as the most anti-correlated predicted target of miR-29a in glioblastoma. Importantly, inhibition of endogenous miR-29a decreases glioblastoma growth and invasion in vitro and in vivo, and increased miR-29a expression in glioblastoma specimens correlates with decreased patient survival. Conclusions Taken together, these data identify miR-29a as a master regulator of glioblastoma growth and invasion. Electronic supplementary material The online version of this article (10.1186/s13046-019-1026-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yun Zhao
- Department of Neurosurgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Chemotherapy, Tumor Hospital of Guangxi Medical University, No.2, Nanning, Guangxi, China
| | - Wei Huang
- Department of Neurosurgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Tae-Min Kim
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Yuchae Jung
- Department of Neurosurgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Lata G Menon
- Department of Neurosurgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Hongyan Xing
- Department of Neurosurgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Hongwei Li
- Department of Neurosurgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Rona S Carroll
- Department of Neurological Surgery, University of Massachusetts Medical School, Albert Sherman Center AS6-1001, 368 Plantation Street, Worcester, MA, 01605, USA.,Department of Neurosurgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter J Park
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Hong Wei Yang
- Department of Neurological Surgery, University of Massachusetts Medical School, Albert Sherman Center AS6-1001, 368 Plantation Street, Worcester, MA, 01605, USA. .,Department of Neurosurgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Mark D Johnson
- Department of Neurological Surgery, University of Massachusetts Medical School, Albert Sherman Center AS6-1001, 368 Plantation Street, Worcester, MA, 01605, USA. .,Department of Neurosurgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA. .,Program in Neuro-Oncology, Dana Farber Cancer Institute, Boston, MA, USA. .,Department of Neurological Surgery, UMass Memorial Healthcare, University of Massachusetts Medical School, 55 Lake Avenue North, S2-855, Worcester, MA, 01655, USA.
| |
Collapse
|
37
|
Mahdavi Fikejvar E, Rezadoost H, Zakizadeh H, Mozaffarian V. A comparative study on the essential oil composition and antibacterial activities of different organs of wild growingPaeonia dauricasubsp.tomentosafrom Iran. Nat Prod Res 2018; 33:3153-3156. [DOI: 10.1080/14786419.2018.1516665] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Edris Mahdavi Fikejvar
- Department of Horticultural Sciences, University Campus, University of Guilan, Rasht, Iran
| | - Hassan Rezadoost
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, G. C. Evin, Tehran, Iran
| | - Hedayat Zakizadeh
- Department of Horticultural Sciences, University Campus, University of Guilan, Rasht, Iran
| | | |
Collapse
|
38
|
Tuffaha R, Voelkl J, Pieske B, Lang F, Alesutan I. Role of PKB/SGK-dependent phosphorylation of GSK-3α/β in vascular calcification during cholecalciferol overload in mice. Biochem Biophys Res Commun 2018; 503:2068-2074. [PMID: 30119888 DOI: 10.1016/j.bbrc.2018.07.161] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 07/31/2018] [Indexed: 12/22/2022]
Abstract
Medial vascular calcification is a highly regulated process involving osteo-/chondrogenic transdifferentiation of vascular smooth muscle cells. Both, protein kinase B (PKB) and serum- and glucocorticoid-inducible kinase 1 (SGK1) are involved in the intracellular signaling of vascular calcification and both phosphorylate and inactivate glycogen synthase kinase 3 (GSK-3). The present study explored whether PKB/SGK-dependent phosphorylation of GSK-3α/β is involved in vascular calcification. Experiments were performed in Gsk-3α/β double knockin mice lacking functional PKB/SGK phosphorylation sites (gsk-3KI) and corresponding wild-type mice (gsk-3WT) following high-dosed cholecalciferol treatment as well as ex vivo in aortic ring explants from gsk-3KI and gsk-3WT mice treated without and with phosphate. In gsk-3WT mice, high-dosed cholecalciferol induced vascular calcification and aortic osteo-/chondrogenic signaling, shown by increased expression of osteogenic markers Msx2, Cbfa1 and tissue-nonspecific alkaline phosphatase (Alpl). All these effects were suppressed in aortic tissue from gsk-3KI mice. Cholecalciferol decreased aortic Gsk-3α/β phosphorylation (Ser21/9) in gsk-3WT mice, while no phosphorylation was observed in gsk-3KI mice. Moreover, the mRNA expression of type III sodium-dependent phosphate transporter (Pit1) and plasminogen activator inhibitor 1 (Pai1) was increased following cholecalciferol treatment in aortic tissue of gsk-3WT mice, effects again blunted in gsk-3KI mice. In addition, phosphate treatment induced mineral deposition and osteogenic markers expression in aortic ring explants from gsk-3WT mice, effects reduced in aortic ring explants from gsk-3KI mice. In conclusion, vascular PKB/SGK-dependent phosphorylation of GSK-3α/β contributes to the osteoinductive signaling leading to vascular calcification.
Collapse
Affiliation(s)
- Rashad Tuffaha
- Department of Physiology I, Eberhard-Karls University, Wilhelmstr. 56, 72076 Tübingen, Germany
| | - Jakob Voelkl
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, 13353 Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Hessische Str. 3-4, 10115 Berlin, Germany.
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, 13353 Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Hessische Str. 3-4, 10115 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch Str. 2, 10178 Berlin, Germany; Department of Internal Medicine and Cardiology, German Heart Center Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Florian Lang
- Department of Physiology I, Eberhard-Karls University, Wilhelmstr. 56, 72076 Tübingen, Germany
| | - Ioana Alesutan
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, 13353 Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Hessische Str. 3-4, 10115 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch Str. 2, 10178 Berlin, Germany
| |
Collapse
|
39
|
Bulani Y, Sharma SS. Argatroban Attenuates Diabetic Cardiomyopathy in Rats by Reducing Fibrosis, Inflammation, Apoptosis, and Protease-Activated Receptor Expression. Cardiovasc Drugs Ther 2018; 31:255-267. [PMID: 28695302 DOI: 10.1007/s10557-017-6732-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE Chronic diabetes is associated with cardiovascular dysfunctions. Diabetic cardiomyopathy (DCM) is one of the serious cardiovascular complications associated with diabetes. Despite significant efforts in understanding the pathophysiology of DCM, management of DCM is not adequate due to its complex pathophysiology. Recently, involvement of protease-activated receptors (PARs) has been postulated in cardiovascular diseases. These receptors are activated by thrombin, trypsin, or other serine proteases. Expression of PAR has been shown to be increased in cardiac diseases such as myocardial infarction, viral myocarditis, and pulmonary arterial hypertension. However, the role of PAR in DCM has not been elucidated yet. Therefore, in the present study, we have investigated the role of PAR in the condition of DCM using a pharmacological approach. We used argatroban, a direct thrombin inhibitor for targeting PAR. METHODS Type-2 diabetes mellitus (T2DM) was induced by high-fat feeding along with low dose streptozotocin (STZ 35 mg/kg, i.p. single dose) in male Sprague-Dawley rats. After 16 weeks of diabetes induction, animals were treated with argatroban at 0.3 and 1 mg/kg dose daily for 4 weeks. After 20 weeks, ventricular functions were measured using ventricular catheterization. Cardiac histology, TUNEL staining, and immunoblotting were performed to evaluate cardiac fibrosis, DNA fragmentation, and expression level of different proteins, respectively. RESULTS T2DM was associated with cardiac structural and functional disturbances as evidenced from impaired cardiac functional parameters and increased fibrosis. There was a significant increase in PAR expression after 20 weeks of diabetes induction. Four weeks argatroban treatment ameliorated metabolic alterations (reduced plasma glucose and cholesterol), ventricular dysfunctions (improved systolic and diastolic functions), cardiac fibrosis (reduced percentage area of collagen in picro-sirius red staining), and apoptosis (reduced TUNEL positive nuclei). Reduced expression of PAR1 and PAR4 in the argatroban-treated group indicates a response towards inhibition of thrombin. In addition, AKT (Ser-473), GSK-3β (Ser-9), p-65 NFĸB phosphorylation, TGF-β, COX-2, and caspase-3 expression were reduced significantly along with an increase in SERCA expression in argatroban-treated diabetic rats which indicated the anti-fibrotic, anti-inflammatory, and anti-apoptotic potential of argatroban in DCM. CONCLUSION This study suggests the ameliorative effects of argatroban in diabetic cardiomyopathy by improving ventricular functions and reducing fibrosis, inflammation, apoptosis, and PAR expression.
Collapse
Affiliation(s)
- Yogesh Bulani
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector-67, SAS, Nagar (Mohali), Punjab, 160062, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector-67, SAS, Nagar (Mohali), Punjab, 160062, India.
| |
Collapse
|
40
|
Kelaini S, Vilà-González M, Caines R, Campbell D, Eleftheriadou M, Tsifaki M, Magee C, Cochrane A, O'neill K, Yang C, Stitt AW, Zeng L, Grieve DJ, Margariti A. Follistatin-Like 3 Enhances the Function of Endothelial Cells Derived from Pluripotent Stem Cells by Facilitating β-Catenin Nuclear Translocation Through Inhibition of Glycogen Synthase Kinase-3β Activity. Stem Cells 2018; 36:1033-1044. [PMID: 29569797 PMCID: PMC6099345 DOI: 10.1002/stem.2820] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 02/10/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
Abstract
The fight against vascular disease requires functional endothelial cells (ECs) which could be provided by differentiation of induced Pluripotent Stem Cells (iPS Cells) in great numbers for use in the clinic. However, the great promise of the generated ECs (iPS-ECs) in therapy is often restricted due to the challenge in iPS-ECs preserving their phenotype and function. We identified that Follistatin-Like 3 (FSTL3) is highly expressed in iPS-ECs, and, as such, we sought to clarify its possible role in retaining and improving iPS-ECs function and phenotype, which are crucial in increasing the cells' potential as a therapeutic tool. We overexpressed FSTL3 in iPS-ECs and found that FSTL3 could induce and enhance endothelial features by facilitating β-catenin nuclear translocation through inhibition of glycogen synthase kinase-3β activity and induction of Endothelin-1. The angiogenic potential of FSTL3 was also confirmed both in vitro and in vivo. When iPS-ECs overexpressing FSTL3 were subcutaneously injected in in vivo angiogenic model or intramuscularly injected in a hind limb ischemia NOD.CB17-Prkdcscid/NcrCrl SCID mice model, FSTL3 significantly induced angiogenesis and blood flow recovery, respectively. This study, for the first time, demonstrates that FSTL3 can greatly enhance the function and maturity of iPS-ECs. It advances our understanding of iPS-ECs and identifies a novel pathway that can be applied in cell therapy. These findings could therefore help improve efficiency and generation of therapeutically relevant numbers of ECs for use in patient-specific cell-based therapies. In addition, it can be particularly useful toward the treatment of vascular diseases instigated by EC dysfunction. Stem Cells 2018;36:1033-1044.
Collapse
Affiliation(s)
- Sophia Kelaini
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Marta Vilà-González
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Rachel Caines
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - David Campbell
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | | | - Marianna Tsifaki
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Corey Magee
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Amy Cochrane
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Karla O'neill
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Chunbo Yang
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Alan W Stitt
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Lingfang Zeng
- Cardiovascular Division, King's College London, London, United Kingdom
| | - David J Grieve
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Andriana Margariti
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
41
|
Regulation of Aspergillus nidulans CreA-Mediated Catabolite Repression by the F-Box Proteins Fbx23 and Fbx47. mBio 2018; 9:mBio.00840-18. [PMID: 29921666 PMCID: PMC6016232 DOI: 10.1128/mbio.00840-18] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The attachment of one or more ubiquitin molecules by SCF (Skp-Cullin-F-box) complexes to protein substrates targets them for subsequent degradation by the 26S proteasome, allowing the control of numerous cellular processes. Glucose-mediated signaling and subsequent carbon catabolite repression (CCR) are processes relying on the functional regulation of target proteins, ultimately controlling the utilization of this carbon source. In the filamentous fungus Aspergillus nidulans, CCR is mediated by the transcription factor CreA, which modulates the expression of genes encoding biotechnologically relevant enzymes. Although CreA-mediated repression of target genes has been extensively studied, less is known about the regulatory pathways governing CCR and this work aimed at further unravelling these events. The Fbx23 F-box protein was identified as being involved in CCR and the Δfbx23 mutant presented impaired xylanase production under repressing (glucose) and derepressing (xylan) conditions. Mass spectrometry showed that Fbx23 is part of an SCF ubiquitin ligase complex that is bridged via the GskA protein kinase to the CreA-SsnF-RcoA repressor complex, resulting in the degradation of the latter under derepressing conditions. Upon the addition of glucose, CreA dissociates from the ubiquitin ligase complex and is transported into the nucleus. Furthermore, casein kinase is important for CreA function during glucose signaling, although the exact role of phosphorylation in CCR remains to be determined. In summary, this study unraveled novel mechanistic details underlying CreA-mediated CCR and provided a solid basis for studying additional factors involved in carbon source utilization which could prove useful for biotechnological applications.IMPORTANCE The production of biofuels from plant biomass has gained interest in recent years as an environmentally friendly alternative to production from petroleum-based energy sources. Filamentous fungi, which naturally thrive on decaying plant matter, are of particular interest for this process due to their ability to secrete enzymes required for the deconstruction of lignocellulosic material. A major drawback in fungal hydrolytic enzyme production is the repression of the corresponding genes in the presence of glucose, a process known as carbon catabolite repression (CCR). This report provides previously unknown mechanistic insights into CCR through elucidating part of the protein-protein interaction regulatory system that governs the CreA transcriptional regulator in the reference organism Aspergillus nidulans in the presence of glucose and the biotechnologically relevant plant polysaccharide xylan.
Collapse
|
42
|
Angiotensin-(1–7) reduces cardiac effects of thyroid hormone by GSK3Β/NFATc3 signaling pathway. Clin Sci (Lond) 2018; 132:1117-1133. [DOI: 10.1042/cs20171606] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 04/12/2018] [Accepted: 04/23/2018] [Indexed: 12/14/2022]
Abstract
Patients with hyperthyroidism exhibit increased risk of development and progression of cardiac diseases. The activation of the renin–angiotensin system (RAS) has been indirectly implicated in these cardiac effects observed in hyperthyroidism. Angiotensin-(1–7) (Ang-(1–7)) has previously been shown to counterbalance pathological effects of angiotensin II (Ang II). The aim of the present study was to investigate the effects of elevated circulating Ang-(1–7) levels on cardiac effects promoted by hyperthyroidism in a transgenic rat (TG) model that constitutively overexpresses an Ang-(1–7)-producing fusion protein [TGR(A1-7)3292]. TG and wild-type (WT) rats received daily injections (i.p.) of triiodothyronine (T3; 7 µg/100 g of body weight (BW)) or vehicle for 14 days. In contrast with WT rats, the TG rats did not develop cardiac hypertrophy after T3 treatment. Indeed, TG rats displayed reduced systolic blood pressure (SBP) and cardiac hyperdynamic condition induced by hyperthyroidism. Moreover, increased plasma levels of Ang II observed in hyperthyroid WT rats were prevented in TG rats. TG rats were protected from glycogen synthase kinase 3β (GSK3β) inactivation and nuclear factor of activated T cells (NFAT) nuclear accumulation induced by T3. In vitro studies evidenced that Ang-(1–7) prevented cardiomyocyte hypertrophy and GSK3β inactivation induced by T3. Taken together, these data reveal an important cardioprotective action of Ang-(1–7) in experimental model of hyperthyroidism.
Collapse
|
43
|
Chen XX, Guo RR, Cao XP, Tan L, Tan L. The impact of GAB2 genetic variations on cerebrospinal fluid markers in Alzheimer's disease. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:171. [PMID: 29951493 PMCID: PMC5994512 DOI: 10.21037/atm.2018.04.11] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 04/03/2018] [Indexed: 11/06/2022]
Abstract
BACKGROUND Growth factor receptor-bound protein-associated binding protein 2 gene (GAB2) has been regarded as one of the susceptibility gene associated with Alzheimer's disease (AD). However, the role of GAB2 polymorphisms on cerebrospinal fluid (CSF) proteins in AD continuum remains unclear. METHODS We evaluated the connection between four single nucleotide polymorphisms (SNPs) of GAB2 and AD-related CSF biomarkers including amyloid β (Aβ), total tau (T-tau) and phosphorylated tau (P-tau) level in 627 Alzheimer's Disease Neuroimaging Initiative (ADNI) subjects. RESULTS rs1385600 and rs1007837 were significantly associated with all the three biomarkers in CSF (rs1385600: Aβ Pc =0.0112, T-tau Pc =0.0356, P-tau Pc =0.0116; rs1007837: Aβ Pc =0.0058, T-tau Pc =0.0278, P-tau Pc =0.0231). rs2373115 only showed significant association with Aβ and P-tau (Aβ, Pc=0.0398, P-tau, Pc=0.0329). rs10793294 showed no significant association with all the three biomarkers. CONCLUSIONS Our study suggested that GAB2 variants were significantly associated with the level of the three CSF biomarkers, which further supported that GAB2 genetic variation modulates AD risk via the alteration of both Aβ and tau pathology.
Collapse
Affiliation(s)
- Xiao-Xiao Chen
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, China
| | - Run-Rong Guo
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, China
| | - Xi-Peng Cao
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, China
| | - Lin Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, China
| | | |
Collapse
|
44
|
Abstract
Glycogen synthase kinase-3 beta (GSK3β) is principally is a glycogen synthase phosphorylating enzyme that is well known for its role in muscle metabolism. GSK3β is a serine/threonine protein Kinase, which is responsible for several essential roles in mammalian cells. This enzyme is implicated in the pathophysiology of many conditions involved in homeostasis and cellular immigration. GSK3β is involved in several pathways leading to neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. Increasing evidence has shown the potential importance of GSK3β in ischemic heart disease and ischemia-reperfusion pathologies. Reperfusion injury may occur in tissues after prolonged ischemia following reperfusion. Reperfusion injury can be life threatening. Reperfusion injury occurs due to a change in ionic homeostasis, excess free radical production, mitochondrial damage and cell death. There are however clear, cardiac-protective signals; although the molecular pathophysiology is not clearly understood. In normal physiology, GSK3β has a critical role in the cytoprotective pathway. However, it`s controversial role in ischemia and ischemia-reperfusion is a topic of current interest. In this review, we have opted to focus on GSK3β interactions with mitochondria in ischemic heart disease and expand on the therapeutic interventions.
Collapse
|
45
|
Sarikhani M, Mishra S, Maity S, Kotyada C, Wolfgeher D, Gupta MP, Singh M, Sundaresan NR. SIRT2 deacetylase regulates the activity of GSK3 isoforms independent of inhibitory phosphorylation. eLife 2018; 7:32952. [PMID: 29504933 PMCID: PMC5860870 DOI: 10.7554/elife.32952] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 03/02/2018] [Indexed: 12/28/2022] Open
Abstract
Glycogen synthase kinase 3 (GSK3) is a critical regulator of diverse cellular functions involved in the maintenance of structure and function. Enzymatic activity of GSK3 is inhibited by N-terminal serine phosphorylation. However, alternate post-translational mechanism(s) responsible for GSK3 inactivation are not characterized. Here, we report that GSK3α and GSK3β are acetylated at Lys246 and Lys183, respectively. Molecular modeling and/or molecular dynamics simulations indicate that acetylation of GSK3 isoforms would hinder both the adenosine binding and prevent stable interactions of the negatively charged phosphates. We found that SIRT2 deacetylates GSK3β, and thus enhances its binding to ATP. Interestingly, the reduced activity of GSK3β is associated with lysine acetylation, but not with phosphorylation at Ser9 in hearts of SIRT2-deficient mice. Moreover, GSK3 is required for the anti-hypertrophic function of SIRT2 in cardiomyocytes. Overall, our study identified lysine acetylation as a novel post-translational modification regulating GSK3 activity.
Collapse
Affiliation(s)
- Mohsen Sarikhani
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Sneha Mishra
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Sangeeta Maity
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Chaithanya Kotyada
- Molecular Biophysics Unit, Indian Institute of Science, Bengaluru, India
| | - Donald Wolfgeher
- Department of Molecular Genetics and Cell biology, University of Chicago, Chicago, United States
| | - Mahesh P Gupta
- Department of Surgery, University of Chicago, Chicago, United States
| | - Mahavir Singh
- Molecular Biophysics Unit, Indian Institute of Science, Bengaluru, India
| | | |
Collapse
|
46
|
Fisetin Confers Cardioprotection against Myocardial Ischemia Reperfusion Injury by Suppressing Mitochondrial Oxidative Stress and Mitochondrial Dysfunction and Inhibiting Glycogen Synthase Kinase 3 β Activity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9173436. [PMID: 29636855 PMCID: PMC5845518 DOI: 10.1155/2018/9173436] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/01/2018] [Indexed: 01/25/2023]
Abstract
Acute myocardial infarction (AMI) is the leading cause of morbidity and mortality worldwide. Timely reperfusion is considered an optimal treatment for AMI. Paradoxically, the procedure of reperfusion can itself cause myocardial tissue injury. Therefore, a strategy to minimize the reperfusion-induced myocardial tissue injury is vital for salvaging the healthy myocardium. Herein, we investigated the cardioprotective effects of fisetin, a natural flavonoid, against ischemia/reperfusion (I/R) injury (IRI) using a Langendorff isolated heart perfusion system. I/R produced significant myocardial tissue injury, which was characterized by elevated levels of lactate dehydrogenase and creatine kinase in the perfusate and decreased indices of hemodynamic parameters. Furthermore, I/R resulted in elevated oxidative stress, uncoupling of the mitochondrial electron transport chain, increased mitochondrial swelling, a decrease of the mitochondrial membrane potential, and induction of apoptosis. Moreover, IRI was associated with a loss of the mitochondrial structure and decreased mitochondrial biogenesis. However, when the animals were pretreated with fisetin, it significantly attenuated the I/R-induced myocardial tissue injury, blunted the oxidative stress, and restored the structure and function of mitochondria. Mechanistically, the fisetin effects were found to be mediated via inhibition of glycogen synthase kinase 3β (GSK3β), which was confirmed by a biochemical assay and molecular docking studies.
Collapse
|
47
|
Yang Y, Lei T, Du S, Tong R, Wang H, Yang J, Huang J, Sun M, Wang Y, Dong Z. Nuclear GSK3β induces DNA double-strand break repair by phosphorylating 53BP1 in glioblastoma. Int J Oncol 2018; 52:709-720. [PMID: 29328365 PMCID: PMC5807039 DOI: 10.3892/ijo.2018.4237] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 12/20/2017] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma is the most malignant and lethal subtype brain tumors with high risk of recurrence and therapeutic resistance. Emerging evidence has indicated that glycogen synthesis kinase 3 (GSK3)β plays oncogenic roles in multiple tumor types; however, the underlying mechanisms remain largely unknown. It has also been demonstrated that p53 binding protein 1 (53BP1) plays a central role in DNA double-strand break (DSB) repair. This study aimed to reveal the significance of GSK3β translocation from the cytoplasm to the nucleus, and to determine whether GSK3β induces DNA DSB repair in the nuclei of glioblastoma cells via phospho-53BP1. By performing in vitro experiments, we found that GSK3β translocated from the cytoplasm to the nucleus, and it then bound to 53BP1 following exposure to IR (IR). In addition, 53BP1-mediated DNA DSB repair was observed to be abrogated by the inhibition of GSK3β. Further experiments on the phosphorylation site of 53BP1 by GSK3β revealed that the S/T-Q motif may play a critical role. Importantly, our in vivo and in vitro data clearly indicated that GSK3β induced the phosphorylation of 53BP1 at the Ser166 site. Moreover, brain tumor xenograft models revealed that following exposure to IR plus SB216763, a specific GSK3β inhibitor, tumor growth was markedly inhibited and the survival of mice markedly increased. Based on these results, we concluded that the phosphorylation of 53BP1 by GSK3β was indispensable for DNA DSB repair. Our study also suggested that the inhibition of GSK3β by SB216763 significantly inhibited the proliferation and induced the apoptosis of glioblastoma cells. Taken together, our data indicate that GSK3β, a key phosphorylation protein for 53BP1, may be a potential target for enhancing the sensitivity of glioblastoma cells to radiation.
Collapse
Affiliation(s)
- Yong Yang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Tiantian Lei
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, P.R. China
| | - Suya Du
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, P.R. China
| | - Rongsheng Tong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Hailian Wang
- Institute of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Jiao Yang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, P.R. China
| | - Juan Huang
- Department of Pharmacy, Medical Center Hospital of Qionglai, Qionglai, Sichuan 611500, P.R. China
| | - Minghan Sun
- Department of Gynecology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Yi Wang
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Zhi Dong
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
48
|
Rab-GTPase binding effector protein 2 (RABEP2) is a primed substrate for Glycogen Synthase kinase-3 (GSK3). Sci Rep 2017; 7:17682. [PMID: 29247183 PMCID: PMC5732219 DOI: 10.1038/s41598-017-17087-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 11/20/2017] [Indexed: 12/28/2022] Open
Abstract
Glycogen synthase kinase-3 (GSK3) regulates many physiological processes through phosphorylation of a diverse array of substrates. Inhibitors of GSK3 have been generated as potential therapies in several diseases, however the vital role GSK3 plays in cell biology makes the clinical use of GSK3 inhibitors potentially problematic. A clearer understanding of true physiological and pathophysiological substrates of GSK3 should provide opportunities for more selective, disease specific, manipulation of GSK3. To identify kinetically favourable substrates we performed a GSK3 substrate screen in heart tissue. Rab-GTPase binding effector protein 2 (RABEP2) was identified as a novel GSK3 substrate and GSK3 phosphorylation of RABEP2 at Ser200 was enhanced by prior phosphorylation at Ser204, fitting the known consensus sequence for GSK3 substrates. Both residues are phosphorylated in cells while only Ser200 phosphorylation is reduced following inhibition of GSK3. RABEP2 function was originally identified as a Rab5 binding protein. We did not observe co-localisation of RABEP2 and Rab5 in cells, while ectopic expression of RABEP2 had no effect on endosomal recycling. The work presented identifies RABEP2 as a novel primed substrate of GSK3, and thus a potential biomarker for GSK3 activity, but understanding how phosphorylation regulates RABEP2 function requires more information on physiological roles of RABEP2.
Collapse
|
49
|
A partnership with the proteasome; the destructive nature of GSK3. Biochem Pharmacol 2017; 147:77-92. [PMID: 29102676 PMCID: PMC5954166 DOI: 10.1016/j.bcp.2017.10.016] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 10/31/2017] [Indexed: 12/19/2022]
Abstract
Glycogen Synthase Kinase-3 (GSK3) was originally reported as a key enzyme of glucose homeostasis through regulation of the rate of glycogen synthesis. It has subsequently been found to influence most cellular processes, including growth, differentiation and death, as part of its role in modulating response to hormonal, nutritional and cellular stress stimuli. More than 100 protein targets for GSK3 have been proposed although only a small fraction of these have been convincingly validated in physiological cell systems. The effects of GSK3 phosphorylation on substrates include alteration of enzyme activity, protein localisation, protein:protein interaction and protein stability. This latter form of regulation of GSK3 substrates is the focus of this review. There is an ever-growing list of GSK3 substrates that upon phosphorylation are targeted to the beta-transducin repeat containing protein (β-TrCP), thereby allowing ubiquitination of bound protein by cullin-1 and so initiating destruction at the proteasome. We propose the existence of a GSK3-β-TrCP ‘destruction hit-list’ that allows co-ordinated removal (or stabilisation) of a set of proteins with a common physiological purpose, through control of GSK3. We identify 29 proteins where there is relatively strong evidence for regulation by a GSK3-β-TrCP axis and note common features of regulation and pathophysiology. Furthermore, we assess the potential of pre-phosphorylation (priming) of these targets (normally a prerequisite for GSK3 recognition) to provide a second layer of regulation delineated by the priming kinase that allows GSK3 to mark them for destruction. Finally, we discuss whether this knowledge improves options for therapeutic intervention.
Collapse
|
50
|
Mechanisms contributing to cardiac remodelling. Clin Sci (Lond) 2017; 131:2319-2345. [PMID: 28842527 DOI: 10.1042/cs20171167] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 07/26/2017] [Accepted: 07/31/2017] [Indexed: 12/14/2022]
Abstract
Cardiac remodelling is classified as physiological (in response to growth, exercise and pregnancy) or pathological (in response to inflammation, ischaemia, ischaemia/reperfusion (I/R) injury, biomechanical stress, excess neurohormonal activation and excess afterload). Physiological remodelling of the heart is characterized by a fine-tuned and orchestrated process of beneficial adaptations. Pathological cardiac remodelling is the process of structural and functional changes in the left ventricle (LV) in response to internal or external cardiovascular damage or influence by pathogenic risk factors, and is a precursor of clinical heart failure (HF). Pathological remodelling is associated with fibrosis, inflammation and cellular dysfunction (e.g. abnormal cardiomyocyte/non-cardiomyocyte interactions, oxidative stress, endoplasmic reticulum (ER) stress, autophagy alterations, impairment of metabolism and signalling pathways), leading to HF. This review describes the key molecular and cellular responses involved in pathological cardiac remodelling.
Collapse
|