1
|
Brandão SR, Oliveira PF, Guerra-Carvalho B, Reis-Mendes A, Neuparth MJ, Carvalho F, Ferreira R, Costa VM. Enduring metabolic modulation in the cardiac tissue of elderly CD-1 mice two months post mitoxantrone treatment. Free Radic Biol Med 2024; 223:199-211. [PMID: 39059512 DOI: 10.1016/j.freeradbiomed.2024.07.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/21/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Mitoxantrone (MTX) is a therapeutic agent used in the treatment of solid tumors and multiple sclerosis, recognized for its cardiotoxicity, with underlying molecular mechanisms not fully disclosed. The cardiotoxicity is influenced by risk factors, including age. Our study intended to assess the molecular effect of MTX on the cardiac muscle of old male CD-1 mice. Mice aged 19 months received a total cumulative dose of 4.5 mg/kg of MTX (MTX group) or saline solution (CTRL group). Two months post treatment, blood was collected, animals sacrificed, and the heart removed. MTX caused structural cardiac changes, which were accompanied by extracellular matrix remodeling, as indicated by the increased ratio between matrix metallopeptidase 2 and metalloproteinase inhibitor 2. At the metabolic level, decreased glycerol levels were found, together with a trend towards increased content of the electron transfer flavoprotein dehydrogenase. In contrast, lower glycolysis, given by the decreased content of glucose transporter GLUT4 and phosphofructokinase, seemed to occur. The findings suggest higher reliance on fatty acids oxidation, despite no major remodeling occurring at the mitochondrial level. Furthermore, the levels of glutamine and other amino acids (although to a lesser extent) were decreased, which aligns with decreased content of the E3 ubiquitin-protein ligase Atrogin-1, suggesting a decrease in proteolysis. As far as we know, this was the first study made in old mice with a clinically relevant dose of MTX, evaluating its long-term cardiac effects. Even two months after MTX exposure, changes in metabolic fingerprint occurred, highlighting enduring cardiac effects that may require clinical vigilance.
Collapse
Affiliation(s)
- Sofia Reis Brandão
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; LAQV - REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Pedro Fontes Oliveira
- LAQV - REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Bárbara Guerra-Carvalho
- LAQV - REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal.
| | - Ana Reis-Mendes
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Maria João Neuparth
- Laboratory for Integrative and Translational Research in Population Health (ITR), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sports, University of Porto, 4200-450 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal.
| | - Félix Carvalho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Rita Ferreira
- LAQV - REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Vera Marisa Costa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
2
|
Ajoolabady A, Pratico D, Ren J. Endothelial dysfunction: mechanisms and contribution to diseases. Acta Pharmacol Sin 2024; 45:2023-2031. [PMID: 38773228 PMCID: PMC11420364 DOI: 10.1038/s41401-024-01295-8] [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: 02/18/2024] [Accepted: 04/16/2024] [Indexed: 05/23/2024] Open
Abstract
The endothelium, lining the inner surface of blood vessels and spanning approximately 3 m2, serves as the largest organ in the body. Comprised of endothelial cells, the endothelium interacts with other bodily components including the bloodstream, circulating cells, and the lymphatic system. Functionally, the endothelium primarily synchronizes vascular tone (by balancing vasodilation and vasoconstriction) and prevents vascular inflammation and pathologies. Consequently, endothelial dysfunction disrupts vascular homeostasis, leading to vascular injuries and diseases such as cardiovascular, cerebral, and metabolic diseases. In this opinion/perspective piece, we explore the recently identified mechanisms of endothelial dysfunction across various disease subsets and critically evaluate the strengths and limitations of current therapeutic interventions at the pre-clinical level.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| |
Collapse
|
3
|
Røsand Ø, Wang J, Scrimgeour N, Marwarha G, Høydal MA. Exosomal Preconditioning of Human iPSC-Derived Cardiomyocytes Beneficially Alters Cardiac Electrophysiology and Micro RNA Expression. Int J Mol Sci 2024; 25:8460. [PMID: 39126028 PMCID: PMC11313350 DOI: 10.3390/ijms25158460] [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: 06/13/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Experimental evidence, both in vitro and in vivo, has indicated cardioprotective effects of extracellular vesicles (EVs) derived from various cell types, including induced pluripotent stem cell-derived cardiomyocytes. The biological effects of EV secretion, particularly in the context of ischemia and cardiac electrophysiology, remain to be fully explored. Therefore, the goal of this study was to unveil the effects of exosome (EXO)-mediated cell-cell signaling during hypoxia by employing a simulated preconditioning approach on human-induced pluripotent stem cell-derived cardiomyocytes (hIPSC-CMs). Electrophysiological activity of hIPSC-CMs was measured using a multielectrode array (MEA) system. A total of 16 h of hypoxic stress drastically increased the beat period. Moreover, hIPSC-CMs preconditioned with EXOs displayed significantly longer beat periods compared with non-treated cells after 16 h of hypoxia (+15.7%, p < 0.05). Furthermore, preconditioning with hypoxic EXOs resulted in faster excitation-contraction (EC) coupling compared with non-treated hIPSC-CMs after 16 h of hypoxia (-25.3%, p < 0.05). Additionally, microRNA (miR) sequencing and gene target prediction analysis of the non-treated and pre-conditioned hIPSC-CMs identified 10 differentially regulated miRs and 44 gene targets. These results shed light on the intricate involvement of miRs, emphasizing gene targets associated with cell survival, contraction, apoptosis, reactive oxygen species (ROS) regulation, and ion channel modulation. Overall, this study demonstrates that EXOs secreted by hIPSC-CM during hypoxia beneficially alter electrophysiological properties in recipient cells exposed to hypoxic stress, which could play a crucial role in the development of targeted interventions to improve outcomes in ischemic heart conditions.
Collapse
Affiliation(s)
| | | | | | | | - Morten Andre Høydal
- Group of Molecular and Cellular Cardiology, Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology (NTNU), 7030 Trondheim, Norway; (Ø.R.); (J.W.); (N.S.); (G.M.)
| |
Collapse
|
4
|
Hu Y, Yi L, Yang Y, Wu Z, Kong M, Kang Z, Yang Z. Acetylation of FOXO1 activates Bim expression involved in CVB3 induced cardiomyocyte apoptosis. Apoptosis 2024; 29:1271-1287. [PMID: 38127284 PMCID: PMC11263423 DOI: 10.1007/s10495-023-01924-3] [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] [Accepted: 11/25/2023] [Indexed: 12/23/2023]
Abstract
Viral myocarditis (VMC) is the major reason for sudden cardiac death among both children and young adults. Of these, coxsackievirus B3 (CVB3) is the most common causative agent of myocarditis. Recently, the role of signaling pathways in the pathogenesis of VMC has been evaluated in several studies, which has provided a new perspective on identifying potential therapeutic targets for this hitherto incurable disease. In the present study, in vivo and in vitro experiments showed that CVB3 infection leads to increased Bim expression and triggers apoptosis. In addition, by knocking down Bim using RNAi, we further confirmed the biological function of Bim in apoptosis induced by CVB3 infection. We additionally found that Bim and forkhead box O1 class (FOXO1) inhibition significantly increased the viability of CVB3-infected cells while blocking viral replication and viral release. Moreover, CVB3-induced Bim expression was directly dependent on FOXO1 acetylation, which is catalyzed by the co-regulation of CBP and SirTs. Furthermore, the acetylation of FOXO1 was an important step in Bim activation and apoptosis induced by CVB3 infection. The findings of this study suggest that CVB3 infection induces apoptosis through the FOXO1 acetylation-Bim pathway, thus providing new insights for developing potential therapeutic targets for enteroviral myocarditis.
Collapse
Affiliation(s)
- Yanan Hu
- Department of Pediatrics, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Lu Yi
- Department of Pediatrics, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Yeyi Yang
- Department of Medicine, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Zhixiang Wu
- Department of Pediatrics, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Min Kong
- Department of Pediatrics, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Zhijuan Kang
- Department of Pediatrics, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Zuocheng Yang
- Department of Pediatrics, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China.
| |
Collapse
|
5
|
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
|
6
|
Rebboah E, Rezaie N, Williams BA, Weimer AK, Shi M, Yang X, Liang HY, Dionne LA, Reese F, Trout D, Jou J, Youngworth I, Reinholdt L, Morabito S, Snyder MP, Wold BJ, Mortazavi A. The ENCODE mouse postnatal developmental time course identifies regulatory programs of cell types and cell states. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598567. [PMID: 38915583 PMCID: PMC11195270 DOI: 10.1101/2024.06.12.598567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Postnatal genomic regulation significantly influences tissue and organ maturation but is under-studied relative to existing genomic catalogs of adult tissues or prenatal development in mouse. The ENCODE4 consortium generated the first comprehensive single-nucleus resource of postnatal regulatory events across a diverse set of mouse tissues. The collection spans seven postnatal time points, mirroring human development from childhood to adulthood, and encompasses five core tissues. We identified 30 cell types, further subdivided into 69 subtypes and cell states across adrenal gland, left cerebral cortex, hippocampus, heart, and gastrocnemius muscle. Our annotations cover both known and novel cell differentiation dynamics ranging from early hippocampal neurogenesis to a new sex-specific adrenal gland population during puberty. We used an ensemble Latent Dirichlet Allocation strategy with a curated vocabulary of 2,701 regulatory genes to identify regulatory "topics," each of which is a gene vector, linked to cell type differentiation, subtype specialization, and transitions between cell states. We find recurrent regulatory topics in tissue-resident macrophages, neural cell types, endothelial cells across multiple tissues, and cycling cells of the adrenal gland and heart. Cell-type-specific topics are enriched in transcription factors and microRNA host genes, while chromatin regulators dominate mitosis topics. Corresponding chromatin accessibility data reveal dynamic and sex-specific regulatory elements, with enriched motifs matching transcription factors in regulatory topics. Together, these analyses identify both tissue-specific and common regulatory programs in postnatal development across multiple tissues through the lens of the factors regulating transcription.
Collapse
Affiliation(s)
- Elisabeth Rebboah
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, USA
| | - Narges Rezaie
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, USA
| | - Brian A. Williams
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, USA
| | - Annika K. Weimer
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, USA
| | - Minyi Shi
- Department of Next Generation Sequencing and Microchemistry, Proteomics and Lipidomics, Genentech, San Francisco, USA
| | - Xinqiong Yang
- Department of Genetics, Stanford University School of Medicine, Palo Alto, USA
| | - Heidi Yahan Liang
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
| | | | - Fairlie Reese
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
| | - Diane Trout
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, USA
| | - Jennifer Jou
- Department of Genetics, Stanford University School of Medicine, Palo Alto, USA
| | - Ingrid Youngworth
- Department of Genetics, Stanford University School of Medicine, Palo Alto, USA
| | | | - Samuel Morabito
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, USA
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Palo Alto, USA
| | - Barbara J. Wold
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, USA
| | - Ali Mortazavi
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, USA
| |
Collapse
|
7
|
Song M, Choi DB, Im JS, Song YN, Kim JH, Lee H, An J, Kim A, Choi H, Kim JC, Han C, Jeon YK, Kim SJ, Woo DH. Modeling acute myocardial infarction and cardiac fibrosis using human induced pluripotent stem cell-derived multi-cellular heart organoids. Cell Death Dis 2024; 15:308. [PMID: 38693114 PMCID: PMC11063052 DOI: 10.1038/s41419-024-06703-9] [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: 10/15/2023] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/03/2024]
Abstract
Heart disease involves irreversible myocardial injury that leads to high morbidity and mortality rates. Numerous cell-based cardiac in vitro models have been proposed as complementary approaches to non-clinical animal research. However, most of these approaches struggle to accurately replicate adult human heart conditions, such as myocardial infarction and ventricular remodeling pathology. The intricate interplay between various cell types within the adult heart, including cardiomyocytes, fibroblasts, and endothelial cells, contributes to the complexity of most heart diseases. Consequently, the mechanisms behind heart disease induction cannot be attributed to a single-cell type. Thus, the use of multi-cellular models becomes essential for creating clinically relevant in vitro cell models. This study focuses on generating self-organizing heart organoids (HOs) using human-induced pluripotent stem cells (hiPSCs). These organoids consist of cardiomyocytes, fibroblasts, and endothelial cells, mimicking the cellular composition of the human heart. The multi-cellular composition of HOs was confirmed through various techniques, including immunohistochemistry, flow cytometry, q-PCR, and single-cell RNA sequencing. Subsequently, HOs were subjected to hypoxia-induced ischemia and ischemia-reperfusion (IR) injuries within controlled culture conditions. The resulting phenotypes resembled those of acute myocardial infarction (AMI), characterized by cardiac cell death, biomarker secretion, functional deficits, alterations in calcium ion handling, and changes in beating properties. Additionally, the HOs subjected to IR efficiently exhibited cardiac fibrosis, displaying collagen deposition, disrupted calcium ion handling, and electrophysiological anomalies that emulate heart disease. These findings hold significant implications for the advancement of in vivo-like 3D heart and disease modeling. These disease models present a promising alternative to animal experimentation for studying cardiac diseases, and they also serve as a platform for drug screening to identify potential therapeutic targets.
Collapse
Affiliation(s)
- Myeongjin Song
- Department of Commercializing Organoid Technology, NEXEL Co., Ltd., Seoul, 07802, Korea
| | - Da Bin Choi
- Department of Commercializing Organoid Technology, NEXEL Co., Ltd., Seoul, 07802, Korea
| | - Jeong Suk Im
- Department of Commercializing Organoid Technology, NEXEL Co., Ltd., Seoul, 07802, Korea
| | - Ye Na Song
- Department of Commercializing Organoid Technology, NEXEL Co., Ltd., Seoul, 07802, Korea
| | - Ji Hyun Kim
- Department of Commercializing Organoid Technology, NEXEL Co., Ltd., Seoul, 07802, Korea
| | - Hanbyeol Lee
- Centre for Research, Hudson Institute of Medical Research, Monash University, Clayton, VIC, 3168, Australia
| | - Jieun An
- Department of Commercializing iPSC Technology, NEXEL Co., Ltd., Seoul, 07802, Korea
| | - Ami Kim
- Department of Commercializing iPSC Technology, NEXEL Co., Ltd., Seoul, 07802, Korea
| | - Hwan Choi
- Department of Commercializing iPSC Technology, NEXEL Co., Ltd., Seoul, 07802, Korea
| | - Joon-Chul Kim
- Department of Commercializing Organoid Technology, NEXEL Co., Ltd., Seoul, 07802, Korea
| | - Choongseong Han
- Department of Commercializing Organoid Technology, NEXEL Co., Ltd., Seoul, 07802, Korea
- Department of Commercializing iPSC Technology, NEXEL Co., Ltd., Seoul, 07802, Korea
| | - Young Keul Jeon
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University, College of Medicine, Seoul, 03080, Korea
| | - Sung Joon Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University, College of Medicine, Seoul, 03080, Korea
| | - Dong-Hun Woo
- Department of Commercializing Organoid Technology, NEXEL Co., Ltd., Seoul, 07802, Korea.
- Department of Commercializing iPSC Technology, NEXEL Co., Ltd., Seoul, 07802, Korea.
| |
Collapse
|
8
|
Fan X, Cao J, Li M, Zhang D, El‐Battrawy I, Chen G, Zhou X, Yang G, Akin I. Stroke Related Brain-Heart Crosstalk: Pathophysiology, Clinical Implications, and Underlying Mechanisms. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307698. [PMID: 38308187 PMCID: PMC11005719 DOI: 10.1002/advs.202307698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/09/2024] [Indexed: 02/04/2024]
Abstract
The emergence of acute ischemic stroke (AIS) induced cardiovascular dysfunctions as a bidirectional interaction has gained paramount importance in understanding the intricate relationship between the brain and heart. Post AIS, the ensuing cardiovascular dysfunctions encompass a spectrum of complications, including heart attack, congestive heart failure, systolic or diastolic dysfunction, arrhythmias, electrocardiographic anomalies, hemodynamic instability, cardiac arrest, among others, all of which are correlated with adverse outcomes and mortality. Mounting evidence underscores the intimate crosstalk between the heart and the brain, facilitated by intricate physiological and neurohumoral complex networks. The primary pathophysiological mechanisms contributing to these severe cardiac complications involve the hypothalamic-pituitary-adrenal (HPA) axis, sympathetic and parasympathetic hyperactivity, immune and inflammatory responses, and gut dysbiosis, collectively shaping the stroke-related brain-heart axis. Ongoing research endeavors are concentrated on devising strategies to prevent AIS-induced cardiovascular dysfunctions. Notably, labetalol, nicardipine, and nitroprusside are recommended for hypertension control, while β-blockers are employed to avert chronic remodeling and address arrhythmias. However, despite these therapeutic interventions, therapeutic targets remain elusive, necessitating further investigations into this complex challenge. This review aims to delineate the state-of-the-art pathophysiological mechanisms in AIS through preclinical and clinical research, unraveling their intricate interplay within the brain-heart axis, and offering pragmatic suggestions for managing AIS-induced cardiovascular dysfunctions.
Collapse
Affiliation(s)
- Xuehui Fan
- Key Laboratory of Medical ElectrophysiologyMinistry of Education and Medical Electrophysiological Key Laboratory of Sichuan ProvinceCollaborative Innovation Center for Prevention of Cardiovascular DiseasesInstitute of Cardiovascular ResearchSouthwest Medical UniversityLuzhou646000China
- CardiologyAngiologyHaemostaseologyand Medical Intensive CareMedical Centre MannheimMedical Faculty MannheimHeidelberg University68167HeidelbergGermany
- European Center for AngioScience (ECAS)German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheimand Centre for Cardiovascular Acute Medicine Mannheim (ZKAM)Medical Centre MannheimHeidelberg University68167HeidelbergGermany
| | - Jianyang Cao
- School of Physical EducationSouthwest Medical UniversityLuzhouSichuan Province646000China
- Acupuncture and Rehabilitation DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhou646000China
| | - Mingxia Li
- School of Physical EducationSouthwest Medical UniversityLuzhouSichuan Province646000China
- Acupuncture and Rehabilitation DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhou646000China
| | - Dechou Zhang
- Department of NeurologyThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhou646000China
| | - Ibrahim El‐Battrawy
- Department of Cardiology and AngiologyRuhr University44780BochumGermany
- Institut für Forschung und Lehre (IFL)Department of Molecular and Experimental CardiologyRuhr‐University Bochum44780BochumGermany
| | - Guiquan Chen
- Acupuncture and Rehabilitation DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhou646000China
| | - Xiaobo Zhou
- Key Laboratory of Medical ElectrophysiologyMinistry of Education and Medical Electrophysiological Key Laboratory of Sichuan ProvinceCollaborative Innovation Center for Prevention of Cardiovascular DiseasesInstitute of Cardiovascular ResearchSouthwest Medical UniversityLuzhou646000China
- CardiologyAngiologyHaemostaseologyand Medical Intensive CareMedical Centre MannheimMedical Faculty MannheimHeidelberg University68167HeidelbergGermany
- European Center for AngioScience (ECAS)German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheimand Centre for Cardiovascular Acute Medicine Mannheim (ZKAM)Medical Centre MannheimHeidelberg University68167HeidelbergGermany
| | - Guoqiang Yang
- CardiologyAngiologyHaemostaseologyand Medical Intensive CareMedical Centre MannheimMedical Faculty MannheimHeidelberg University68167HeidelbergGermany
- European Center for AngioScience (ECAS)German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheimand Centre for Cardiovascular Acute Medicine Mannheim (ZKAM)Medical Centre MannheimHeidelberg University68167HeidelbergGermany
- Acupuncture and Rehabilitation DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhou646000China
| | - Ibrahim Akin
- CardiologyAngiologyHaemostaseologyand Medical Intensive CareMedical Centre MannheimMedical Faculty MannheimHeidelberg University68167HeidelbergGermany
- European Center for AngioScience (ECAS)German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheimand Centre for Cardiovascular Acute Medicine Mannheim (ZKAM)Medical Centre MannheimHeidelberg University68167HeidelbergGermany
| |
Collapse
|
9
|
Isop LM, Neculau AE, Necula RD, Kakucs C, Moga MA, Dima L. Metformin: The Winding Path from Understanding Its Molecular Mechanisms to Proving Therapeutic Benefits in Neurodegenerative Disorders. Pharmaceuticals (Basel) 2023; 16:1714. [PMID: 38139841 PMCID: PMC10748332 DOI: 10.3390/ph16121714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/25/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Metformin, a widely prescribed medication for type 2 diabetes, has garnered increasing attention for its potential neuroprotective properties due to the growing demand for treatments for Alzheimer's, Parkinson's, and motor neuron diseases. This review synthesizes experimental and clinical studies on metformin's mechanisms of action and potential therapeutic benefits for neurodegenerative disorders. A comprehensive search of electronic databases, including PubMed, MEDLINE, Embase, and Cochrane library, focused on key phrases such as "metformin", "neuroprotection", and "neurodegenerative diseases", with data up to September 2023. Recent research on metformin's glucoregulatory mechanisms reveals new molecular targets, including the activation of the LKB1-AMPK signaling pathway, which is crucial for chronic administration of metformin. The pleiotropic impact may involve other stress kinases that are acutely activated. The precise role of respiratory chain complexes (I and IV), of the mitochondrial targets, or of the lysosomes in metformin effects remains to be established by further research. Research on extrahepatic targets like the gut and microbiota, as well as its antioxidant and immunomodulatory properties, is crucial for understanding neurodegenerative disorders. Experimental data on animal models shows promising results, but clinical studies are inconclusive. Understanding the molecular targets and mechanisms of its effects could help design clinical trials to explore and, hopefully, prove its therapeutic effects in neurodegenerative conditions.
Collapse
Affiliation(s)
- Laura Mihaela Isop
- Department of Fundamental, Prophylactic and Clinical Sciences, Faculty of Medicine, Transilvania University of Brasov, 500036 Brașov, Romania; (L.M.I.)
| | - Andrea Elena Neculau
- Department of Fundamental, Prophylactic and Clinical Sciences, Faculty of Medicine, Transilvania University of Brasov, 500036 Brașov, Romania; (L.M.I.)
| | - Radu Dan Necula
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov, 500036 Brașov, Romania
| | - Cristian Kakucs
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov, 500036 Brașov, Romania
| | - Marius Alexandru Moga
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov, 500036 Brașov, Romania
| | - Lorena Dima
- Department of Fundamental, Prophylactic and Clinical Sciences, Faculty of Medicine, Transilvania University of Brasov, 500036 Brașov, Romania; (L.M.I.)
| |
Collapse
|
10
|
Olejnik A, Radajewska A, Krzywonos-Zawadzka A, Bil-Lula I. Klotho inhibits IGF1R/PI3K/AKT signalling pathway and protects the heart from oxidative stress during ischemia/reperfusion injury. Sci Rep 2023; 13:20312. [PMID: 37985893 PMCID: PMC10662387 DOI: 10.1038/s41598-023-47686-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023] Open
Abstract
Ischemia/reperfusion injury (IRI) of the heart involves the activation of oxidative and proapoptotic pathways. Simultaneously Klotho protein presents anti-aging, antiapoptotic and antioxidative properties. Therefore, this study aimed to evaluate the effect of Klotho protein on oxidative stress in hearts subjected to IRI. Isolated rat hearts perfused with the Langendorff method were subjected to ischemia, followed by reperfusion, in the presence or absence of recombinant rat Klotho protein. The factors involved in the activation of insulin-like growth factor receptor (IGF1R)/phosphoinositide-3-kinase (PI3K)/protein kinase B (AKT) signalling pathway were evaluated. IRI caused activation of the IGF1R (p = 0.0122)/PI3K (p = 0.0022) signalling, as compared to the aerobic control group. Infusion supply of Klotho protein during IRI significantly reduced the level of phospho-IGF1R (p = 0.0436), PI3K (p = 0.0218) and phospho-AKT (p = 0.0020). Transcriptional activity of forkhead box protein O3 (FOXO3) was reduced (p = 0.0207) in hearts subjected to IRI, compared to aerobic control. Administration of Klotho decreased phosphorylation of FOXO3 (p = 0.0355), and enhanced activity of glutathione peroxidase (p = 0.0452) and superoxide dismutase (p = 0.0060) in IRI + Klotho group. The levels of reactive oxygen/nitrogen species (ROS/RNS) (p = 0.0480) and hydrogen peroxide (H2O2) (p = 0.0460), and heart injury (p = 0.0005) were significantly increased in hearts from the IRI group in comparison to the aerobic group. Klotho reduced NADPH oxidase 2 (NOX2) (p = 0.0390), ROS/RNS (p = 0.0435) and H2O2 (p = 0.0392) levels, and heart damage (p = 0.0286) in the hearts subjected to IRI. In conclusion, Klotho contributed to the protection of the heart against IRI and oxidative stress via inhibition of the IGF1R/PI3K/AKT pathway, thus can be recognized as a novel cardiopreventive/cardioprotective agent.
Collapse
Affiliation(s)
- Agnieszka Olejnik
- Division of Clinical Chemistry and Laboratory Haematology, Department of Medical Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A St., 50-556, Wrocław, Poland
| | - Anna Radajewska
- Division of Clinical Chemistry and Laboratory Haematology, Department of Medical Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A St., 50-556, Wrocław, Poland
| | - Anna Krzywonos-Zawadzka
- Division of Clinical Chemistry and Laboratory Haematology, Department of Medical Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A St., 50-556, Wrocław, Poland
| | - Iwona Bil-Lula
- Division of Clinical Chemistry and Laboratory Haematology, Department of Medical Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A St., 50-556, Wrocław, Poland.
| |
Collapse
|
11
|
Morris BJ, Donlon TA. Genes That Extend Lifespan May Do So by Mitigating the Increased Risk of Death Posed by Having Hypertension. Am J Hypertens 2023; 36:631-640. [PMID: 37561089 PMCID: PMC10647014 DOI: 10.1093/ajh/hpad070] [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: 05/12/2023] [Revised: 07/22/2023] [Accepted: 08/09/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Genetic factors influence lifespan. In humans, there appears to be a particularly strong genetic effect in those aged ≥ 90 years. An important contribution is nutrient sensing genes which confer cell resilience. METHODS Our research has been investigating the genetic factors by longitudinal studies of American men of Japanese descent living on the island of Oahu in Hawaii. This cohort began as the Honolulu Heart Program in the mid-1960s and most subjects are now deceased. RESULTS We previously discovered various genes containing polymorphisms associated with longevity. In recent investigations of the mechanism involved we found that the longevity genotypes ameliorated the risk of mortality posed by having a cardiometabolic disease (CMD)-most prominently hypertension. For the gene FOXO3 the protective alleles mitigated the risk of hypertension, coronary heart disease (CHD) and diabetes. For the kinase MAP3K5 it was hypertension, CHD and diabetes, for the kinase receptor PIK3R1 hypertension, CHD and stroke, and for the growth hormone receptor gene (GHR) and vascular endothelial growth factor receptor 1 gene (FLT1), it was nullifying the higher mortality risk posed by hypertension. Subjects with a CMD who had a longevity genotype had similar survival as men without CMD. No variant protected against risk of death from cancer. We have postulated that the longevity-associated genotypes reduced mortality risk by effects on intracellular resilience mechanisms. In a proteomics study, 43 "stress" proteins and associated biological pathways were found to influence the association of FOXO3 genotype with reduced mortality. CONCLUSIONS Our landmark findings indicate how heritable genetic components affect longevity.
Collapse
Affiliation(s)
- Brian J Morris
- Department of Research, NIH Center of Biomedical Research Excellence on Aging, Kuakini Medical Center, Honolulu, Hawaii 96817, USA
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii 96813, USA
- School of Medical Sciences, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Timothy A Donlon
- Department of Research, NIH Center of Biomedical Research Excellence on Aging, Kuakini Medical Center, Honolulu, Hawaii 96817, USA
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii 96813, USA
| |
Collapse
|
12
|
Zhang B, Shi L, Tan Y, Zhou Y, Cui J, Song Y, Liu Y, Zhang M, Duan W, Jin Z, Liu J, Yi D, Sun Y, Yi W. Forkhead box O6 (FoxO6) promotes cardiac pathological remodeling and dysfunction by activating Kif15-TGF-β1 under aggravated afterload. MedComm (Beijing) 2023; 4:e383. [PMID: 37799807 PMCID: PMC10547936 DOI: 10.1002/mco2.383] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 08/08/2023] [Accepted: 08/24/2023] [Indexed: 10/07/2023] Open
Abstract
Pathological cardiac hypertrophy exhibits complex and abnormal gene expression patterns and progresses to heart failure. Forkhead box protein O6 (FoxO6) is a key transcription factor involved in many biological processes. This study aimed to explore the role of FoxO6 in cardiac hypertrophy. Three groups of mice were established: wild-type, FoxO6 knockout, and FoxO6-overexpressing. The mice received daily administration of angiotensin-II (Ang-II) or saline for 4 weeks, after which they were examined for cardiac hypertrophy, fibrosis, and function. Elevated cardiac expression of FoxO6 was observed in Ang-II-treated mice. FoxO6 deficiency attenuated contractile dysfunction and cardiac remodeling, including cardiomyocyte hypertrophy and fibroblast proliferation and differentiation. Conversely, FoxO6 overexpression aggravated the cardiomyopathy and heart dysfunction. Further studies identified kinesin family member 15 (Kif15) as downstream molecule of FoxO6. Kif15 inhibition attenuated the aggravating effect of FoxO6 overexpression. In vitro, FoxO6 overexpression increased Kif15 expression in cardiomyocytes and elevated the concentration of transforming growth factor-β1 (TGF-β1) in the medium where fibroblasts were grown, exhibiting increased proliferation and differentiation, while FoxO6 knockdown attenuated this effect. Cardiac-derived FoxO6 promoted pathological cardiac remodeling induced by aggravated afterload largely by activating the Kif15/TGF-β1 axis. This result further complements the mechanisms of communication among different cells in the heart, providing novel therapeutic targets for heart failure.
Collapse
Affiliation(s)
- Bing Zhang
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Lei Shi
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Yanzhen Tan
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Yenong Zhou
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Jun Cui
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Yujie Song
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Yingying Liu
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Miao Zhang
- Department of GeriatricsXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Weixun Duan
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Zhenxiao Jin
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Jincheng Liu
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Dinghua Yi
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Yang Sun
- Department of GeriatricsXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Wei Yi
- Department of Cardiovascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| |
Collapse
|
13
|
Cheng C, Wang S, Dong J, Zhang S, Yu D, Wang Z. Effects of targeted lung cancer drugs on cardiomyocytes studied by atomic force microscopy. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:4077-4084. [PMID: 37565311 DOI: 10.1039/d3ay00784g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
The epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKIs) has become one of the important targeted drugs for the treatment of non-small cell lung cancer (NSCLC). But the cardiac adverse events (AEs) related to the EGFR-TKI treatment occur frequently. And the cases of TKI-associated cardiac AEs remain poorly understood. In order to study the effects of EGFR-TKIs on cardiomyocytes, atomic force microscopy (AFM) was used to measure and analyze the physical properties of cardiomyocytes under the actions of three drugs (gefitinib, afatinib and osimertinib) with different concentrations. By comparing the height, adhesion, Young's modulus, the amplitude and the time of the contraction and relaxation process, it was found that the changes of the mechanical properties of cells were well correlated with the symptoms of AEs, such as cardiomyocyte hypertrophy, QT prolongation, atrial fibrillation, ejection fraction reductions, and cardiac failure. In addition, osimertinib has the most obvious effect on cardiomyocytes at a low concentration, and gefitinib has the greatest effect with the increase of concentration, while afatinib has the least effect on cardiomyocytes. This provides a new method for screening drugs and exploring the principle of action in the process of cancer treatment at the cellular level.
Collapse
Affiliation(s)
- Can Cheng
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China.
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China
| | - Shuwei Wang
- Affiliated Hospital of Jilin Medical University, Jilin City, Jilin, China
| | - Jianjun Dong
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China.
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China
| | - Shengli Zhang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China.
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China
| | - Dongliang Yu
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China.
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, China
| | - Zuobin Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China.
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China
- JR3CN & IRAC, University of Bedfordshire, Luton LU1 3JU, UK
| |
Collapse
|
14
|
Donlon TA, Morris BJ, Chen R, Lim E, Morgen EK, Fortney K, Shah N, Masaki KH, Willcox BJ. Proteomic basis of mortality resilience mediated by FOXO3 longevity genotype. GeroScience 2023; 45:2303-2324. [PMID: 36881352 PMCID: PMC10651822 DOI: 10.1007/s11357-023-00740-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/23/2023] [Indexed: 03/08/2023] Open
Abstract
FOXO3 is a ubiquitous transcription factor expressed in response to cellular stress caused by nutrient deprivation, inflammatory cytokines, reactive oxygen species, radiation, hypoxia, and other factors. We showed previously that the association of inherited FOXO3 variants with longevity was the result of partial protection against mortality risk posed by aging-related life-long stressors, particularly cardiometabolic disease. We then referred to the longevity-associated genotypes as conferring "mortality resilience." Serum proteins whose levels change with aging and are associated with mortality risk may be considered as "stress proteins." They may serve as indirect measures of life-long stress. Our aims were to (1) identify stress proteins that increase with aging and are associated with an increased risk of mortality, and (2) to determine if FOXO3 longevity/resilience genotype dampens the expected increase in mortality risk they pose. A total of 4500 serum protein aptamers were quantified using the Somalogic SomaScan proteomics platform in the current study of 975 men aged 71-83 years. Stress proteins associated with mortality were identified. We then used age-adjusted multivariable Cox models to investigate the interaction of stress protein with FOXO3 longevity-associated rs12212067 genotypes. For all the analyses, the p values were corrected for multiple comparisons by false discovery rate. This led to the identification of 44 stress proteins influencing the association of FOXO3 genotype with reduced mortality. Biological pathways were identified for these proteins. Our results suggest that the FOXO3 resilience genotype functions by reducing mortality in pathways related to innate immunity, bone morphogenetic protein signaling, leukocyte migration, and growth factor response.
Collapse
Affiliation(s)
- Timothy A Donlon
- Department of Research, NIH Center of Biomedical Research Excellence for Clinical and Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, 96817, USA
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Brian J Morris
- Department of Research, NIH Center of Biomedical Research Excellence for Clinical and Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, 96817, USA.
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA.
- School of Medical Sciences, University of Sydney, Sydney, New South Wales, Australia.
| | - Randi Chen
- Department of Research, NIH Center of Biomedical Research Excellence for Clinical and Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, 96817, USA
| | - Eunjung Lim
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Eric K Morgen
- BioAge Labs Inc., 1445A S 50th St, Richmond, California, USA
| | - Kristen Fortney
- BioAge Labs Inc., 1445A S 50th St, Richmond, California, USA
| | - Naisha Shah
- BioAge Labs Inc., 1445A S 50th St, Richmond, California, USA
| | - Kamal H Masaki
- Department of Research, NIH Center of Biomedical Research Excellence for Clinical and Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, 96817, USA
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Bradley J Willcox
- Department of Research, NIH Center of Biomedical Research Excellence for Clinical and Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, 96817, USA
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| |
Collapse
|
15
|
Lei YQ, Ye ZJ, Wei YL, Zhu LP, Zhuang XD, Wang XR, Cao H. Nono deficiency impedes the proliferation and adhesion of H9c2 cardiomyocytes through Pi3k/Akt signaling pathway. Sci Rep 2023; 13:7134. [PMID: 37130848 PMCID: PMC10154399 DOI: 10.1038/s41598-023-32572-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/29/2023] [Indexed: 05/04/2023] Open
Abstract
Congenital heart disease (CHD) is the most common type of birth defect and the main noninfectious cause of death during the neonatal stage. The non-POU domain containing, octamer-binding gene, NONO, performs a variety of roles involved in DNA repair, RNA synthesis, transcriptional and post-transcriptional regulation. Currently, hemizygous loss-of-function mutation of NONO have been described as the genetic origin of CHD. However, essential effects of NONO during cardiac development have not been fully elucidated. In this study, we aim to understand role of Nono in cardiomyocytes during development by utilizing the CRISPR/Cas9 gene editing system to deplete Nono in the rat cardiomyocytes H9c2. Functional comparison of H9c2 control and knockout cells showed that Nono deficiency suppressed cell proliferation and adhesion. Furthermore, Nono depletion significantly affected the mitochondrial oxidative phosphorylation (OXPHOS) and glycolysis, resulting in H9c2 overall metabolic deficits. Mechanistically we demonstrated that the Nono knockout impeded the cardiomyocyte function by attenuating phosphatidyl inositol 3 kinase-serine/threonine kinase (Pi3k/Akt) signaling via the assay for transposase-accessible chromatin using sequencing in combination with RNA sequencing. From these results we propose a novel molecular mechanism of Nono to influence cardiomyocytes differentiation and proliferation during the development of embryonic heart. We conclude that NONO may represent an emerging possible biomarkers and targets for the diagnosis and treatment of human cardiac development defects.
Collapse
Affiliation(s)
- Yu-Qing Lei
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350000, China
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, 350000, China
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350011, China
| | - Zhou-Jie Ye
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350000, China
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, 350000, China
| | - Ya-Lan Wei
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350000, China
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, 350000, China
| | - Li-Ping Zhu
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350000, China
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, 350000, China
| | - Xu-Dong Zhuang
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350000, China
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, 350000, China
| | - Xin-Rui Wang
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350000, China.
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, 350000, China.
| | - Hua Cao
- Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350000, China.
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, 350000, China.
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350011, China.
| |
Collapse
|
16
|
Danesh Yazdi M, Nassan FL, Kosheleva A, Wang C, Xu Z, Di Q, Requia WJ, Comfort NT, Wu H, Laurent LC, DeHoff P, Vokonas P, Baccarelli AA, Schwartz JD. Short-term air pollution and temperature exposure and changes in the extracellular microRNA profile of Normative Aging Study (NAS) participants. ENVIRONMENT INTERNATIONAL 2023; 171:107735. [PMID: 36640488 PMCID: PMC10159015 DOI: 10.1016/j.envint.2023.107735] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 05/06/2023]
Abstract
BACKGROUND While the health effects of air pollution and temperature are widely studied, the molecular effects are poorly understood. Extracellular microRNAs (ex-miRNAs) have the potential to serve as diagnostic or prognostic biomarkers and/or to act as intercellular signaling molecules that mediate the effects of environmental exposures on health outcomes. METHODS We examined the relationship between short-term exposure to air pollution and ambient temperature and the ex-miRNA profiles of participants in the Normative Aging Study (NAS) from 1999 to 2015. Our exposures were defined as same-day, two-day, three-day, one-week, two-week, and three-week moving averages of PM2.5, NO2, O3, and temperature which were derived from high-resolution spatio-temporal models. The ex-miRNA profiles of the subjects were obtained during follow-up visits. We analyzed the data using a longitudinal quantile regression model adjusted for individual covariates, batch effects, and time trends. We adjusted for multiple comparisons using a false discovery rate (FDR) correction. Ex-miRNAs that were significantly associated with exposures were further investigated using pathway analyses. RESULTS We found that all the examined exposures were associated with changes in ex-miRNA profiles in our study, particularly PM2.5 which was responsible for most of the statistically significant results. We found 110 statistically significant exposure-outcome relationships that revealed associations with the levels of 52 unique ex-miRNAs. Pathway analyses showed these ex-miRNAs have been linked to target mRNAs, genes, and biological mechanisms that could affect virtually every organ system, and as such may be linked to multiple clinical disease presentations such as cardiovascular disease, respiratory disease, and neurological disease. CONCLUSIONS Air pollution and temperature exposures were significantly associated with alterations in the ex-miRNA profiles of NAS subjects with possible biological consequences.
Collapse
Affiliation(s)
- Mahdieh Danesh Yazdi
- Program in Public Health, Department of Family, Population, and Preventive Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA; Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA.
| | - Feiby L Nassan
- Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA; Biogen Inc, Cambridge, MA, USA
| | - Anna Kosheleva
- Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Cuicui Wang
- Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Zongli Xu
- Laboratory of Molecular Carcinogenesis and Biostatistics Branch, National Institute of Environmental Health Sciences, Durham, NC, USA
| | - Qian Di
- Vanke School of Public Health, Tsinghua University, Beijing, China
| | - Weeberb J Requia
- School of Public Policy and Government, Fundação Getúlio Vargas, Brasília, Distrito Federal, Brazil
| | - Nicole T Comfort
- Department of Environmental Health Sciences, Columbia Mailman School of Public Health, New York, NY, USA
| | - Haotian Wu
- Department of Environmental Health Sciences, Columbia Mailman School of Public Health, New York, NY, USA
| | - Louise C Laurent
- Department of Obstetrics, Gynecology, & Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Peter DeHoff
- Department of Obstetrics, Gynecology, & Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Pantel Vokonas
- Department of Veterans Affairs, Boston, MA, USA; Department of Medicine, Boston University Chobanian and Avidisian School of Medicine, Boston, MA, USA
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Columbia Mailman School of Public Health, New York, NY, USA
| | - Joel D Schwartz
- Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
17
|
Yang X, Wang C, Zheng Q, Liu Q, Wawryk NJP, Li XF. Emerging Disinfection Byproduct 2,6-Dichlorobenzoquinone-Induced Cardiovascular Developmental Toxicity of Embryonic Zebrafish and Larvae: Imaging and Transcriptome Analysis. ACS OMEGA 2022; 7:45642-45653. [PMID: 36530307 PMCID: PMC9753109 DOI: 10.1021/acsomega.2c06296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/23/2022] [Indexed: 06/17/2023]
Abstract
Epidemiological studies have observed the potential association of water disinfection byproduct (DBP) exposure with cardiac defects. Aromatic DBPs represent a significant portion of total DBPs, but their effects on cardiovascular development are unclear. In this study, we examined the effects of an aromatic DBP, 2,6-dichlorobenzoquinone (DCBQ), on the cardiovascular development of zebrafish embryos. After exposure to 2, 4, and 8 μM DCBQ, morphological images of growing zebrafish embryos clearly showed cardiovascular malformation. Fluorescent images of transgenic zebrafish strains with fluorescently labeled heart and blood vessels show that DCBQ exposure resulted in deformed atrium-ventricle looping, degenerated abdomen and trunk vessels, pericardial edema, and decreased blood flow. Furthermore, the expression of the marker gene myl7 (essential for the differentiation and motility of cardiomyocytes) was inhibited in a dose-dependent manner by DCBQ exposure. Finally, transcriptome analysis found that in the 4 μM DCBQ exposure group, the numbers of differentially expressed genes (DEGs) were 113 (50 upregulated and 63 downregulated) at 24 hpf, 2123 (762 upregulated and 1361 downregulated) at 48 hpf, and 61 (11 upregulated and 50 downregulated) at 120 hpf; in the 8 μM DCBQ exposure group, the number of DEGs was 1407 (647 upregulated and 760 downregulated) at 120 hpf. The FoxO signaling pathway was significantly altered. The in vivo results demonstrate the effects of 2,6-DCBQ (0-8 μM) on cardiovascular development, contributing to the understanding of the developmental toxicity of aromatic DBP halobenzoquinones (HBQs).
Collapse
Affiliation(s)
- Xue Yang
- Hubei
Key Laboratory of Environmental and Health Effects of Persistent Toxic
Substances, School of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Chang Wang
- Hubei
Key Laboratory of Environmental and Health Effects of Persistent Toxic
Substances, School of Environment and Health, Jianghan University, Wuhan 430056, China
- Division
of Analytical and Environmental Toxicology, Department of Laboratory
Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2G3, Canada
| | - Qi Zheng
- Hubei
Key Laboratory of Environmental and Health Effects of Persistent Toxic
Substances, School of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Qiongyu Liu
- Hubei
Key Laboratory of Environmental and Health Effects of Persistent Toxic
Substances, School of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Nicholas J. P. Wawryk
- Division
of Analytical and Environmental Toxicology, Department of Laboratory
Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2G3, Canada
| | - Xing-Fang Li
- Division
of Analytical and Environmental Toxicology, Department of Laboratory
Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2G3, Canada
| |
Collapse
|
18
|
Chen K, Gao P, Li Z, Dai A, Yang M, Chen S, Su J, Deng Z, Li L. Forkhead Box O Signaling Pathway in Skeletal Muscle Atrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1648-1657. [PMID: 36174679 DOI: 10.1016/j.ajpath.2022.09.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/01/2022] [Accepted: 09/13/2022] [Indexed: 06/16/2023]
Abstract
Skeletal muscle atrophy is the consequence of protein degradation exceeding protein synthesis because of disease, aging, and physical inactivity. Patients with skeletal muscle atrophy have decreased muscle mass and fiber cross-sectional area, and experience reduced survival quality and motor function. The forkhead box O (FOXO) signaling pathway plays an important role in the pathogenesis of skeletal muscle atrophy by regulating E3 ubiquitin ligases and some autophagy factors. However, the mechanism of FOXO signaling pathway leading to skeletal muscle atrophy is still unclear. The development of treatment strategies for skeletal muscle atrophy has been a thorny clinical problem. FOXO-targeted therapy to treat skeletal muscle atrophy is a promising approach, and an increasing number of relevant studies have been reported. This article reviews the mechanism and therapeutic targets of the FOXO signaling pathway mediating skeletal muscle atrophy, and provides ideas for the clinical treatment of this condition.
Collapse
Affiliation(s)
- Kun Chen
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Peng Gao
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Zongchao Li
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Aonan Dai
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Ming Yang
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Siyu Chen
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China; School of Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Jingyue Su
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China; School of Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Zhenhan Deng
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China; School of Medicine, Guangxi University of Chinese Medicine, Nanning, China.
| | - Liangjun Li
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China.
| |
Collapse
|
19
|
Zhang L, Men X, Yu S, Guo H, Luo Y, Chen H, Mi S. Ozone protects cardiomyocytes from myocardial ischemia-reperfusion injury through miR-200c/FOXO3 axis. J Recept Signal Transduct Res 2022; 42:531-539. [PMID: 35579073 DOI: 10.1080/10799893.2022.2060259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE Myocardial ischemia-reperfusion injury (I/R) is a detrimental process contributing to the pathological progression of coronary artery diseases. Studies indicate that miRNAs are implicated in ischemic heart disease, and ozone therapy could protect the heart from ischemic heart disease. In this study, we investigated the effect of ozone on miR-200c expression and the potential role of miR-200c in an I/R myocardial injury model. METHODS A myocardial cellular model of I/R was established to detect the expression of miR-200c. Cardiomyocytes with I/R induction were treated with ozone as a cellular model to detect miR-200 expression and investigate its functional roles. The downstream target of miR-200c was predicted with Starbase online tools and validated by dual luciferase reporter assay. The function of miR-200c/FOXO3 axis in I/R was examined by CCK-8 proliferation and apoptotic assays. RESULTS miR-200c was upregulated in primary cardiomyocytes of the I/R model. In cardiomyocyte cells, cell proliferation in the I/R group was significantly impaired, which could be partially rescued by miR-200c inhibitor or ozone treatment. Cell death detected by LDH release and apoptosis assay in the I/R model could also be inhibited by miR-200c inhibitor or ozone treatment. FOXO3 was identified as a downstream target of miR-200c, which was induced by ozone treatment and suppressed by miR-200c. Silencing FOXO3 abrogated the protective effect of ozone treatment on the I/R cell model. CONCLUSION Overall, our results suggest that ozone plays a cardio-protective role in I/R through regulating miR-200/FOXO3 axis, and indicate that targeting miR-200/FOXO3 axis could potentially alleviate I/R.
Collapse
Affiliation(s)
- Lian Zhang
- The Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun, China.,Department of Pathology, The Second Hospital of Jilin University, Changchun, China
| | - Xingping Men
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Shenglong Yu
- Jinan University, Guangzhou, China.,Department of Cardiovascular, Panyu Central Hospital (Cardiovascular Institute of Panyu District), Guangzhou, China
| | - Huizhuang Guo
- Department of Radiology, Panyu Central Hospital (Medical Imaging Institute of Panyu District), Guangzhou, China
| | - Yi Luo
- Jinan University, Guangzhou, China.,Department of Cardiovascular, Panyu Central Hospital (Cardiovascular Institute of Panyu District), Guangzhou, China
| | - Hanwei Chen
- Jinan University, Guangzhou, China.,Department of Radiology, Panyu Central Hospital (Medical Imaging Institute of Panyu District), Guangzhou, China
| | - Shaohua Mi
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| |
Collapse
|
20
|
Myostatin/AKT/FOXO Signaling Is Altered in Human Non-Ischemic Dilated Cardiomyopathy. Life (Basel) 2022; 12:life12091418. [PMID: 36143454 PMCID: PMC9506454 DOI: 10.3390/life12091418] [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: 08/08/2022] [Revised: 09/04/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Disturbances in the ubiquitin proteasome system, and especially changes of the E3 ligases, are subjects of interest when searching for causes and therapies for cardiomyopathies. The aim of this study was to clarify whether the myostatin/AKT/forkhead box O (FOXO) pathway, which regulates the expression of the E3 ligases muscle atrophy F-box gene (MAFbx) and muscle ring-finger protein-1 (MuRF1), is changed in dilated cardiomyopathy of ischemic origin (IDCM) and dilated cardiomyopathy of non-ischemic origin (NIDCM). The mRNA and protein expression of myostatin, AKT, FOXO1, FOXO3, MAFbx and MuRF1 were quantified by real-time polymerase chain reaction and ELISA, respectively, in myocardial tissue from 26 IDCM and 23 NIDCM patients. Septal tissue from 17 patients undergoing Morrow resection served as a control. MAFbx and FOXO1 mRNA and protein expression (all p < 0.05), AKT mRNA (p < 0.01) and myostatin protein expression (p = 0.02) were decreased in NIDCM patients compared to the control group. Apart from decreases of AKT and MAFbx mRNA expression (both p < 0.01), no significant differences were detected in IDCM patients compared to the control group. Our results demonstrate that the myostatin/AKT/FOXO pathway is altered in NIDCM but not in IDCM patients. FOXO1 seems to be an important drug target for regulating the expression of MAFbx in NIDCM patients.
Collapse
|
21
|
Zhang X, You LY, Zhang ZY, Jiang DX, Qiu Y, Ruan YP, Mao ZJ. Integrating pharmacological evaluation and computational identification for deciphering the action mechanism of Yunpi-Huoxue-Sanjie formula alleviates diabetic cardiomyopathy. Front Pharmacol 2022; 13:957829. [PMID: 36147338 PMCID: PMC9487204 DOI: 10.3389/fphar.2022.957829] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Yunpi-Huoxue-Sanjie (YP-SJ) formula is a Chinese herbal formula with unique advantages for the treatment of diabetic cardiovascular complications, such as Diabetic cardiomyopathy (DCM). However, potential targets and molecular mechanisms remain unclear. Therefore, our research was designed to evaluate rat myocardial morphology, fat metabolism and oxidative stress to verify myocardial protective effect of YP-SJ formula in vivo. And then to explore and validate its probable mechanism through network pharmacology and experiments in vitro and in vivo. Methods: In this study, DCM rats were randomly divided into five groups: control group, model group, and three YP-SJ formula groups (low-dose, middle-dose, and high-dose groups). Experimental rats were treated with 6 g/kg/d, 12 g/kg/d and 24 g/kg/d YP-SJ formula by gavage for 10 weeks, respectively. Cardiac function of rats was measured by high-resolution small-animal imaging system. The cells were divided into control group, high glucose group, high glucose + control serum group, high glucose + dosed serum group, high glucose + NC-siRNA group, high glucose + siRNA-FoxO1 group. The extent of autophagy was measured by flow cytometry, immunofluorescence, and western blotting. Results: It was found that YP-SJ formula could effectively improve cardiac systolic function in DCM rats. We identified 46 major candidate YP-SJ formula targets that are closely related to the progression of DCM. Enrichment analysis revealed key targets of YP-SJ formula related to environmental information processing, organic systems, and the metabolic occurrence of reactive oxygen species. Meanwhile, we verified that YP-SJ formula can increase the expression of forkhead box protein O1 (FoxO1), autophagy-related protein 7 (Atg7), Beclin 1, and light chain 3 (LC3), and decrease the expression of phosphorylated FoxO1 in vitro and in vivo. The results showed that YP-SJ formula could activate the FoxO1 signaling pathway associated with DCM rats. Further experiments showed that YP-SJ formula could improve cardiac function by regulating autophagy. Conclusion: YP-SJ formula treats DCM by modulating targets that play a key role in autophagy, improving myocardial function through a multi-component, multi-level, multi-target, multi-pathway, and multi-mechanism approach.
Collapse
Affiliation(s)
- Xin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Chinese Medicine Plant Essential Oil Zhejiang Engineering Research Center, Zhejiang, China
| | - Li-Yan You
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Chinese Medicine Plant Essential Oil Zhejiang Engineering Research Center, Zhejiang, China
| | - Ze-Yu Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Dong-Xiao Jiang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Chinese Medicine Plant Essential Oil Zhejiang Engineering Research Center, Zhejiang, China
| | - Yu Qiu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ye-Ping Ruan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Chinese Medicine Plant Essential Oil Zhejiang Engineering Research Center, Zhejiang, China
- *Correspondence: Zhu-Jun Mao, ; Ye-Ping Ruan,
| | - Zhu-Jun Mao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Chinese Medicine Plant Essential Oil Zhejiang Engineering Research Center, Zhejiang, China
- *Correspondence: Zhu-Jun Mao, ; Ye-Ping Ruan,
| |
Collapse
|
22
|
Micro RNA-411 Expression Improves Cardiac Phenotype Following Myocardial Infarction in Mice. JACC Basic Transl Sci 2022; 7:859-875. [PMID: 36317138 PMCID: PMC9617134 DOI: 10.1016/j.jacbts.2022.05.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 01/23/2023]
Abstract
Induction of endogenous regenerative capacity has emerged as one promising approach to repair damaged hearts following myocardial infarction (MI). Re-expression of factors that are exclusively expressed during embryonic development may reactivate the ability of adult cardiomyocytes to regenerate. Here, we identified miR-411 as a potent inducer of cardiomyocyte proliferation. Overexpression of miR-411 in the heart significantly increased cardiomyocyte proliferation and survival in a model MI. We found that miR-411 enhances the activity of YAP, the main downstream effector of the Hippo pathway, in cardiomyocytes. In conclusion, miR-411 induces cardiomyocyte regeneration and improves cardiac function post-MI likely by modulating the Hippo/YAP pathway.
Collapse
Key Words
- CVEC, cardiac vascular endothelial cells
- EdU, 5-ethynyl-2'-deoxyuridine
- Hippo pathway
- LAD, left anterior descending coronary artery
- MI, myocardial infarction
- MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
- NFAT, nuclear factor of activated T cells
- NRCF, neonatal rat cardiac fibroblast
- NRCM, neonatal rat cardiomyocytes
- PCR, polymerase chain reaction
- PEI, polyethylenimine
- cTnI, cardiac troponin I
- cardiac remodeling
- heart failure
- miRNA, microRNA
- microRNA-411
- myocardial infarction
- pHH3, phosphohistone H3
- qPCR, quantitative PCR
Collapse
|
23
|
Acharya A, Nemade H, Papadopoulos S, Hescheler J, Neumaier F, Schneider T, Rajendra Prasad K, Khan K, Hemmersbach R, Gusmao EG, Mizi A, Papantonis A, Sachinidis A. Microgravity-induced stress mechanisms in human stem cell-derived cardiomyocytes. iScience 2022; 25:104577. [PMID: 35789849 PMCID: PMC9249673 DOI: 10.1016/j.isci.2022.104577] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/25/2022] [Accepted: 06/07/2022] [Indexed: 11/17/2022] Open
Abstract
Exposure to outer space microgravity poses a risk for the development of various pathologies including cardiovascular disease. To study this, we derived cardiomyocytes (CMs) from human-induced pluripotent stem cells and exposed them to simulated microgravity (SMG). We combined different “omics” and chromosome conformation capture technologies with live-cell imaging of various transgenic lines to discover that SMG impacts on the contractile velocity and function of CMs via the induction of senescence processes. This is linked to SMG-induced changes of reactive oxygen species (ROS) generation and energy metabolism by mitochondria. Taken together, we uncover a microgravity-controlled axis causing contractile dysfunctions to CMs. Our findings can contribute to the design of preventive and therapeutic strategies against senescence-associated disease. Simulated microgravity (SMG) causes ROS production in human cardiomyocytes (CMs) SMG inhibits mitochondria function and energy metabolism and induces senescence of CMs SMG attenuates contractile velocity, beating frequency and Ca2+ influx in CMs SMG induces chromosomal changes and modifies the chromosomal architecture in CMs
Collapse
|
24
|
Anti-diabetic potential, crystal structure, molecular docking, DFT, and optical-electrochemical studies of new dimethyl and diethyl carbamoyl-N, N′-disubstituted based thioureas. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.132207] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
25
|
Guo S, Mangal R, Dandu C, Geng X, Ding Y. Role of Forkhead Box Protein O1 (FoxO1) in Stroke: A Literature Review. Aging Dis 2022; 13:521-533. [PMID: 35371601 PMCID: PMC8947839 DOI: 10.14336/ad.2021.0826] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/26/2021] [Indexed: 12/11/2022] Open
Abstract
Stroke is one of the most prevalent causes of death around the world. When a stroke occurs, many cellular signaling cascades and regulators are activated, which results in severe cellular dysfunction and debilitating long-term disability. One crucial regulator of cell fate and function is mammalian Forkhead box protein O1 (FoxO1). Many studies have found FoxO1 to be implicated in many cellular processes, including regulating gluconeogenesis and glycogenolysis. During a stroke, modifications of FoxO1 have been linked to a variety of functions, such as inducing cell death and inflammation, inhibiting oxidative injury, affecting the blood brain barrier (BBB), and regulating hepatic gluconeogenesis. For these functions of FoxO1, different measures and treatments were applied to FoxO1 after ischemia. However, the subtle mechanisms of post-transcriptional modification and the role of FoxO1 are still elusive and even contradictory in the development of stroke. The determination of these mechanisms will lead to further enlightenment for FoxO1 signal transduction and the identification of targeted drugs. The regulation and function of FoxO1 may provide an important way for the prevention and treatment of diseases. Overall, the functions of FoxO1 are multifactorial, and this paper will summarize all of the significant pathways in which FoxO1 plays an important role during stroke damage and recovery.
Collapse
Affiliation(s)
- Sichao Guo
- 1Luhe Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,3Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Ruchi Mangal
- 3Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Chaitu Dandu
- 3Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Xiaokun Geng
- 1Luhe Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,2Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,3Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yuchuan Ding
- 3Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
26
|
Kansakar U, Varzideh F, Mone P, Jankauskas SS, Santulli G. Functional Role of microRNAs in Regulating Cardiomyocyte Death. Cells 2022; 11:983. [PMID: 35326433 PMCID: PMC8946783 DOI: 10.3390/cells11060983] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 02/08/2023] Open
Abstract
microRNAs (miRNA, miRs) play crucial roles in cardiovascular disease regulating numerous processes, including inflammation, cell proliferation, angiogenesis, and cell death. Herein, we present an updated and comprehensive overview of the functional involvement of miRs in the regulation of cardiomyocyte death, a central event in acute myocardial infarction, ischemia/reperfusion, and heart failure. Specifically, in this systematic review we are focusing on necrosis, apoptosis, and autophagy.
Collapse
Affiliation(s)
- Urna Kansakar
- Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (F.V.); (P.M.); (S.S.J.)
| | - Fahimeh Varzideh
- Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (F.V.); (P.M.); (S.S.J.)
| | - Pasquale Mone
- Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (F.V.); (P.M.); (S.S.J.)
| | - Stanislovas S. Jankauskas
- Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (F.V.); (P.M.); (S.S.J.)
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Gaetano Santulli
- Department of Medicine (Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA; (U.K.); (F.V.); (P.M.); (S.S.J.)
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
27
|
Liu Y, Li S, Gao Z, Li S, Tan Q, Li Y, Wang D, Wang Q. Indoleamine 2,3-Dioxygenase 1 (IDO1) Promotes Cardiac Hypertrophy via a PI3K-AKT-mTOR-Dependent Mechanism. Cardiovasc Toxicol 2021; 21:655-668. [PMID: 34021461 PMCID: PMC8211584 DOI: 10.1007/s12012-021-09657-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/04/2021] [Indexed: 01/25/2023]
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1) is an enzyme for tryptophan metabolism, involved in immune cell differentiation/maturation and cancer biology. IDO1 is also expressed in cardiomyocytes, but its roles in the cardiovascular system are not fully understood. Here, we reported the functions of IDO1 during cardiac hypertrophy. Quantitative real-time PCR and Western blot experiments demonstrated the upregulation of IDO1 mRNA and protein levels in human and hypertrophic mouse hearts, as well as in angiotensin II (Ang II)-induced hypertrophic rat cardiomyocytes. IDO1 activity and metabolite product kynurenine were upregulated in rodent hypertrophic hearts and cardiomyocytes. Inhibition of IDO1 activity with PF-06840003 reduced Ang II-induced cardiac hypertrophy and rescued cardiac function in mice. siRNA-mediated knockdown of Ido1 repressed Ang II-induced growth in cardiomyocyte size and overexpression of hypertrophy-associated genes atrial natriuretic peptide (Anp or Nppa), brain natriuretic peptide (Bnp or Nppb), β-myosin heavy chain (β-Mhc or Myh7). By contrast, adenovirus-mediated rat Ido1 overexpression in cardiomyocytes promoted hypertrophic growth induced by Ang II. Mechanism analysis showed that IDO1 overexpression was associated with PI3K-AKT-mTOR signaling to activate the ribosomal protein S6 kinase 1 (S6K1), which promoted protein synthesis in Ang II-induced hypertrophy of rat cardiomyocytes. Finally, we provided evidence that inhibition of PI3K with pictilisib, AKT with perifosine, or mTOR with rapamycin, blocked the effects of IDO1 on protein synthesis and cardiomyocyte hypertrophy in Ang II-treated cells. Collectively, our findings identify that IDO1 promotes cardiomyocyte hypertrophy partially via PI3K-AKT-mTOR-S6K1 signaling.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Cardiomegaly/drug therapy
- Cardiomegaly/enzymology
- Cardiomegaly/pathology
- Case-Control Studies
- Cells, Cultured
- Disease Models, Animal
- Female
- Humans
- Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Kynurenine/metabolism
- Male
- Mice
- Middle Aged
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Phosphatidylinositol 3-Kinase/metabolism
- Protein Kinase Inhibitors/pharmacology
- Proto-Oncogene Proteins c-akt/antagonists & inhibitors
- Proto-Oncogene Proteins c-akt/metabolism
- Rats, Sprague-Dawley
- Signal Transduction
- TOR Serine-Threonine Kinases/antagonists & inhibitors
- TOR Serine-Threonine Kinases/metabolism
- Rats
Collapse
Affiliation(s)
- Yang Liu
- Emergency Department, First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Shuang Li
- Emergency Department, First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Zhanqun Gao
- Emergency Department, First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Shuangjia Li
- Emergency Department, First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Qingyun Tan
- Department of Anesthesiology, First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Yanmei Li
- Emergency Department, First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Dongwei Wang
- Department of Anesthesiology, First Affiliated Hospital of Jiamusi University, Jiamusi, China.
| | - Qingdong Wang
- Department of Anesthesiology, First Affiliated Hospital of Jiamusi University, Jiamusi, China.
| |
Collapse
|
28
|
Zhou F, Wang YK, Zhang CG, Wu BY. miR-19a/b-3p promotes inflammation during cerebral ischemia/reperfusion injury via SIRT1/FoxO3/SPHK1 pathway. J Neuroinflammation 2021; 18:122. [PMID: 34051800 PMCID: PMC8164774 DOI: 10.1186/s12974-021-02172-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023] Open
Abstract
Background Stroke affects 3–4% of adults and kills numerous people each year. Recovering blood flow with minimal reperfusion-induced injury is crucial. However, the mechanisms underlying reperfusion-induced injury, particularly inflammation, are not well understood. Here, we investigated the function of miR-19a/b-3p/SIRT1/FoxO3/SPHK1 axis in ischemia/reperfusion (I/R). Methods MCAO (middle cerebral artery occlusion) reperfusion rat model was used as the in vivo model of I/R. Cultured neuronal cells subjected to OGD/R (oxygen glucose deprivation/reperfusion) were used as the in vitro model of I/R. MTT assay was used to assess cell viability and TUNEL staining was used to measure cell apoptosis. H&E staining was employed to examine cell morphology. qRT-PCR and western blot were performed to determine levels of miR-19a/b-3p, SIRT1, FoxO3, SPHK1, NF-κB p65, and cytokines like TNF-α, IL-6, and IL-1β. EMSA and ChIP were performed to validate the interaction of FoxO3 with SPHK1 promoter. Dual luciferase assay and RIP were used to verify the binding of miR-19a/b-3p with SIRT1 mRNA. Results miR-19a/b-3p, FoxO3, SPHK1, NF-κB p65, and cytokines were elevated while SIRT1 was reduced in brain tissues following MCAO/reperfusion or in cells upon OGD/R. Knockdown of SPHK1 or FoxO3 suppressed I/R-induced inflammation and cell death. Furthermore, knockdown of FoxO3 reversed the effects of SIRT1 knockdown. Inhibition of the miR-19a/b-3p suppressed inflammation and this suppression was blocked by SIRT1 knockdown. FoxO3 bound SPHK1 promoter and activated its transcription. miR-19a/b-3p directly targeted SIRT1 mRNA. Conclusion miR-19a/b-3p promotes inflammatory responses during I/R via targeting SIRT1/FoxO3/SPHK1 axis.
Collapse
Affiliation(s)
- Feng Zhou
- Research Center of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China.,Department of Neurology, First People's Hospital of Foshan, Foshan, 528000, Guangdong Province, People's Republic of China
| | - Yu-Kai Wang
- Department of Neurology, First People's Hospital of Foshan, Foshan, 528000, Guangdong Province, People's Republic of China
| | - Cheng-Guo Zhang
- Department of Neurology, First People's Hospital of Foshan, Foshan, 528000, Guangdong Province, People's Republic of China.
| | - Bing-Yi Wu
- Research Center of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China.
| |
Collapse
|
29
|
PXDN reduces autophagic flux in insulin-resistant cardiomyocytes via modulating FoxO1. Cell Death Dis 2021; 12:418. [PMID: 33903591 PMCID: PMC8076187 DOI: 10.1038/s41419-021-03699-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/11/2022]
Abstract
Autophagy, a well-observed intracellular lysosomal degradation process, is particularly important to the cell viability in diabetic cardiomyopathy (DCM). Peroxidasin (PXDN) is a heme-containing peroxidase that augments oxidative stress and plays an essential role in cardiovascular diseases, while whether PXDN contributes to the pathogenesis of DCM remains unknown. Here we reported the suppression of cell viability and autophagic flux, as shown by autophagosomes accumulation and increased expression level of LC3-II and p62 in cultured H9C2 and human AC16 cells that treated with 400 μM palmitate acid (PA) for 24 h. Simultaneously, PXDN protein level increased. Moreover, cell death, autophagosomes accumulation as well as increased p62 expression were suppressed by PXDN silence. In addition, knockdown of PXDN reversed PA-induced downregulated forkhead box-1 (FoxO1) and reduced FoxO1 phosphorylation, whereas did not affect AKT phosphorylation. Not consistent with the effects of si-PXDN, double-silence of PXDN and FoxO1 significantly increased cell death, suppressed autophagic flux and declined the level of FoxO1 and PXDN, while the expression of LC3-II was unchanged under PA stimulation. Furthermore, inhibition of FoxO1 in PA-untreated cells induced cell death, inhibited autophagic flux, and inhibited FoxO1 and PXDN expression. Thus, we come to conclusion that PXDN plays a key role in PA-induced cell death by impairing autophagic flux through inhibiting FoxO1, and FoxO1 may also affect the expression of PXDN. These findings may develop better understanding of potential mechanisms regarding autophagy in insulin-resistant cardiomyocytes.
Collapse
|
30
|
Hartwig J, Loebel M, Steiner S, Bauer S, Karadeniz Z, Roeger C, Skurk C, Scheibenbogen C, Sotzny F. Metformin Attenuates ROS via FOXO3 Activation in Immune Cells. Front Immunol 2021; 12:581799. [PMID: 33953705 PMCID: PMC8089390 DOI: 10.3389/fimmu.2021.581799] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 03/31/2021] [Indexed: 12/16/2022] Open
Abstract
Forkhead box O 3 (FOXO3) is a transcription factor involved in cell metabolism, inflammation and longevity. Here, we investigated if metformin can activate FOXO3 in human immune cells and affects the subsequent level of reactive oxygen/nitrogen species (ROS/RNS) in immune cells. AMP-activated protein kinase (AMPK) and FOXO3 activation were investigated by immunoblot or flow cytometry (FC) analysis, respectively. FOXO3 target gene expression was quantified by real-time PCR. ROS/RNS measurement using dichlorodihydrofluorescein diacetate (DCFH-DA) dye was investigated by FC. The role of the FOXO3 single nucleotide polymorphisms (SNPs) rs12212067, rs2802292 and rs12206094 on ROS/RNS production was studied using allelic discrimination PCR. Metformin induced activation of AMPK (pT172) and FOXO3 (pS413). ROS/RNS level was reduced in immune cells after metformin stimulation accompanied by induction of the FOXO3 targets mitochondrial superoxide dismutase and cytochrome c. Studies in Foxo3 deficient (Foxo3-/- ) mouse splenocytes confirmed that metformin mediates its effects via Foxo3 as it attenuates ROS/RNS in myeloid cells of wildtype (WT) but not of Foxo3-/- mice. Our results suggest that FOXO3 can be activated by metformin leading to reduced ROS/RNS level in immune cells. This may add to the beneficial clinical effects of metformin observed in large cohort studies on longevity, cardiovascular and cancer risk.
Collapse
Affiliation(s)
- Jelka Hartwig
- Institute of Medical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität (FU) Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Madlen Loebel
- Science Center, Carl-Thiem-Klinikum Cottbus, Cottbus, Germany
| | - Sophie Steiner
- Institute of Medical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität (FU) Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Sandra Bauer
- Institute of Medical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität (FU) Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Zehra Karadeniz
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Carsten Roeger
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Carsten Skurk
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research) Partner Site Berlin, Berlin, Germany
| | - Carmen Scheibenbogen
- Institute of Medical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität (FU) Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health (BIH), Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Franziska Sotzny
- Institute of Medical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität (FU) Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
31
|
Weeks KL, Tham YK, Yildiz SG, Alexander Y, Donner DG, Kiriazis H, Harmawan CA, Hsu A, Bernardo BC, Matsumoto A, DePinho RA, Abel ED, Woodcock EA, McMullen JR. FoxO1 is required for physiological cardiac hypertrophy induced by exercise but not by constitutively active PI3K. Am J Physiol Heart Circ Physiol 2021; 320:H1470-H1485. [PMID: 33577435 DOI: 10.1152/ajpheart.00838.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/09/2021] [Indexed: 02/06/2023]
Abstract
The insulin-like growth factor 1 receptor (IGF1R) and phosphoinositide 3-kinase p110α (PI3K) are critical regulators of exercise-induced physiological cardiac hypertrophy and provide protection in experimental models of pathological remodeling and heart failure. Forkhead box class O1 (FoxO1) is a transcription factor that regulates cardiomyocyte hypertrophy downstream of IGF1R/PI3K activation in vitro, but its role in physiological hypertrophy in vivo was unknown. We generated cardiomyocyte-specific FoxO1 knockout (cKO) mice and assessed the phenotype under basal conditions and settings of physiological hypertrophy induced by 1) swim training or 2) cardiac-specific transgenic expression of constitutively active PI3K (caPI3KTg+). Under basal conditions, male and female cKO mice displayed mild interstitial fibrosis compared with control (CON) littermates, but no other signs of cardiac pathology were present. In response to exercise training, female CON mice displayed an increase (∼21%) in heart weight normalized to tibia length vs. untrained mice. Exercise-induced hypertrophy was blunted in cKO mice. Exercise increased cardiac Akt phosphorylation and IGF1R expression but was comparable between genotypes. However, differences in Foxo3a, Hsp70, and autophagy markers were identified in hearts of exercised cKO mice. Deletion of FoxO1 did not reduce cardiac hypertrophy in male or female caPI3KTg+ mice. Cardiac Akt and FoxO1 protein expressions were significantly reduced in hearts of caPI3KTg+ mice, which may represent a negative feedback mechanism from chronic caPI3K, and negate any further effect of reducing FoxO1 in the cKO. In summary, FoxO1 contributes to exercise-induced hypertrophy. This has important implications when one is considering FoxO1 as a target for treating the diseased heart.NEW & NOTEWORTHY Regulators of exercise-induced physiological cardiac hypertrophy and protection are considered promising targets for the treatment of heart failure. Unlike pathological hypertrophy, the transcriptional regulation of physiological hypertrophy has remained largely elusive. To our knowledge, this is the first study to show that the transcription factor FoxO1 is a critical mediator of exercise-induced cardiac hypertrophy. Given that exercise-induced hypertrophy is protective, this finding has important implications when one is considering FoxO1 as a target for treating the diseased heart.
Collapse
Affiliation(s)
- Kate L Weeks
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Diabetes Central Clinical School, Monash University, Clayton, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Yow Keat Tham
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Diabetes Central Clinical School, Monash University, Clayton, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Suzan G Yildiz
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Yonali Alexander
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Daniel G Donner
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, Victoria, Australia
| | | | - Amy Hsu
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Bianca C Bernardo
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Diabetes Central Clinical School, Monash University, Clayton, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Aya Matsumoto
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Ronald A DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - E Dale Abel
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Carver College of Medicine University of Iowa, Iowa City, Iowa
| | | | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Diabetes Central Clinical School, Monash University, Clayton, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, Victoria, Australia
- Department of Physiology and Department of Medicine Alfred Hospital, Monash University, Victoria, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| |
Collapse
|
32
|
Vivar R, Anfossi R, Humeres C, Catalán M, Reyes C, Cárdenas S, Contreras A, Aránguiz P, González F, Diaz-Araya G. FoxO1 is required for high glucose-dependent cardiac fibroblasts into myofibroblast phenoconversion. Cell Signal 2021; 83:109978. [PMID: 33722671 DOI: 10.1016/j.cellsig.2021.109978] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 03/05/2021] [Accepted: 03/09/2021] [Indexed: 11/16/2022]
Abstract
In the normal heart, cardiac fibroblasts (CFs) maintain extracellular matrix (ECM) homeostasis, whereas in pathological conditions, such as diabetes mellitus (DM), CFs converse into cardiac myofibroblasts (CMFs) and this CFs phenoconversion increase the synthesis and secretion of ECM proteins, promoting cardiac fibrosis and heart dysfunction. High glucose (HG) conditions increase TGF-β1 expression and FoxO1 activity, whereas FoxO1 is crucial to CFs phenoconversion induced by TGF-β1. In addition, FoxO1 increases CTGF expression, whereas CTGF plays an active role in the fibrotic process induced by hyperglycemia. However, the role of FoxO1 and CTGF in CFs phenoconversion induced by HG is not clear. In this study, we investigated the effects of FoxO1 pharmacological inhibition on CFs phenoconversion in both in vitro and ex vivo models of DM. Our results demonstrate that HG induces CFs phenoconversion and FoxO1 activation. Moreover, AS1842856, a pharmacological inhibitor of FoxO1 activity, prevents CFs phenoconversion and CTGF expression increase induced by HG, whereas these results were corroborated by FoxO1 silencing. Additionally, K252a, a pharmacological blocker of CTGF receptor, prevents HG-induced CFs phenoconversion, which was corroborated with CTGF expression knockdown. Furthermore, through CFs isolation from heart of diabetic rats, we showed that hyperglycemia induces FoxO1 activation, the increase of CTGF expression and CFs phenoconversion, whereas the FoxO1 activity inhibition reverses the effects induced by hyperglycemia on CFs. Altogether, our results demonstrate that FoxO1 and CTGF are necessary for CFs phenoconversion induced by HG and suggest that both proteins are likely to become a potential targeted drug for fibrotic response induced by hyperglycemic conditions.
Collapse
Affiliation(s)
- Raúl Vivar
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Renatto Anfossi
- Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile
| | - Claudio Humeres
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Mabel Catalán
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Christopher Reyes
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Simone Cárdenas
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Alejandra Contreras
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Pablo Aránguiz
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andrés Bello, 2520000 Viña del Mar, Chile
| | - Fabiola González
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Guillermo Diaz-Araya
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile.; Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile.
| |
Collapse
|
33
|
Zhu N, Huang B, Jiang W. Targets of Vitamin C With Therapeutic Potential for Cardiovascular Disease and Underlying Mechanisms: A Study of Network Pharmacology. Front Pharmacol 2021; 11:591337. [PMID: 33603661 PMCID: PMC7884818 DOI: 10.3389/fphar.2020.591337] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/09/2020] [Indexed: 12/14/2022] Open
Abstract
Vitamin C (ascorbic acid) is a nutrient used to treat cardiovascular disease (CVD). However, the pharmacological targets of vitamin C and the mechanisms underlying the therapeutic effects of vitamin C on CVD remain to be elucidated. In this study, we used network pharmacology approach to investigate the pharmacological mechanisms of vitamin C for the treatment of CVD. The core targets, major hubs, enriched biological processes, and key signaling pathways were identified. A protein-protein interaction network and an interaction diagram of core target-related pathways were constructed. Three core targets were identified, including phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha isoform, signal transducer and activator of transcription-3 (STAT3), and prothrombin. The GO and KEGG analyses identified top 20 enriched biological processes and signaling pathways involved in the therapeutic effects of vitamin C on CVD. The JAK-STAT, STAT, PD1, EGFR, FoxO, and chemokines signaling pathways may be highly involved in the protective effects of vitamin C against CVD. In conclusion, our bioinformatics analyses provided evidence on the possible therapeutic mechanisms of vitamin C in CVD treatment, which may contribute to the development of novel drugs for CVD.
Collapse
Affiliation(s)
- Ning Zhu
- Department of Cardiology, The Third Affiliated Hospital of Shanghai University, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People’s Hospital, Wenzhou, China
| | - Bingwu Huang
- Department of Anesthesiology, The Third Affiliated Hospital of Shanghai University, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People’s Hospital, Wenzhou, China
| | - Wenbing Jiang
- Department of Cardiology, The Third Affiliated Hospital of Shanghai University, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People’s Hospital, Wenzhou, China
| |
Collapse
|
34
|
Yu W, Chen C, Cheng J. The role and molecular mechanism of FoxO1 in mediating cardiac hypertrophy. ESC Heart Fail 2020; 7:3497-3504. [PMID: 33089967 PMCID: PMC7755013 DOI: 10.1002/ehf2.13065] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/18/2020] [Accepted: 09/25/2020] [Indexed: 02/05/2023] Open
Abstract
Cardiac hypertrophy can lead to heart failure and cardiovascular events and has become a research hotspot in the field of cardiovascular disease. Despite extensive and in-depth research, the pathogenesis of cardiac hypertrophy is far from being fully understood. Increasing evidence has shown that the transcription factor forkhead box protein O 1 (FoxO1) is closely related to the occurrence and development of cardiac hypertrophy. This review summarizes the current literature on the role and molecular mechanism of FoxO1 in cardiac hypertrophy. We searched the database MEDLINE via PubMed for available evidence on the effect of FoxO1 on cardiac hypertrophy. FoxO1 has many effects on multiple diseases, including cardiovascular diseases, diabetes, cancer, aging, and stem cell activity. Recent studies have shown that FoxO1 plays a critical role in the development of cardiac hypertrophy. Evidence for this relationship includes the following. (i) FoxO1 can regulate cardiac growth/protein synthesis, calcium homeostasis, cell apoptosis, and autophagy and (ii) is controlled by several upstream signalling molecules (e.g. phosphatidylinositol 3-kinase/Akt, AMP-activated protein kinase, and sirtuins) and regulates many downstream transcription proteins (e.g. ubiquitin ligases muscle RING finger 1/muscle atrophy F-box, calcineurin/nuclear factor of activated T cells, and microRNAs). In response to stress or external stimulation (e.g. low energy, oxidative stress, or growth factor signalling), FoxO1 undergoes post-translational modification and transfers from the cytoplasm to nucleus, thus regulating the expression of a series of target genes in myocardium that are involved in cardiac growth/protein synthesis, calcium homeostasis, cell apoptosis, and autophagy. (iii) Finally, targeted regulation of FoxO1 is an effective method of intervening in myocardial hypertrophy. The information reviewed here should be significant for understanding the roles of FoxO1 in cardiac hypertrophy and should contribute to the design of further studies related to FoxO1 and the hypertrophic response. It should also shed light on a potential treatment for cardiac hypertrophy.
Collapse
Affiliation(s)
- Wei Yu
- Department of Internal MedicineXiang'an Hospital of Xiamen UniversityXiamen361102China
| | - Chunjuan Chen
- Department of CardiologySecond Affiliated Hospital of Shantou University Medical College69 Dong Xia North RoadShantou515041China
| | - Jidong Cheng
- Department of Internal MedicineXiang'an Hospital of Xiamen UniversityXiamen361102China
| |
Collapse
|
35
|
Abstract
FOXO proteins are transcription factors that are involved in numerous physiological processes and in various pathological conditions, including cardiovascular disease, cancer, diabetes and chronic neurological diseases. For example, FOXO proteins are context-dependent tumour suppressors that are frequently inactivated in human cancers, and FOXO3 is the second most replicated gene associated with extreme human longevity. Therefore, pharmacological manipulation of FOXO proteins is a promising approach to developing therapeutics for cancer and for healthy ageing. In this Review, we overview the role of FOXO proteins in health and disease and discuss the pharmacological approaches to modulate FOXO function.
Collapse
|
36
|
Duddu S, Chakrabarti R, Ghosh A, Shukla PC. Hematopoietic Stem Cell Transcription Factors in Cardiovascular Pathology. Front Genet 2020; 11:588602. [PMID: 33193725 PMCID: PMC7596349 DOI: 10.3389/fgene.2020.588602] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022] Open
Abstract
Transcription factors as multifaceted modulators of gene expression that play a central role in cell proliferation, differentiation, lineage commitment, and disease progression. They interact among themselves and create complex spatiotemporal gene regulatory networks that modulate hematopoiesis, cardiogenesis, and conditional differentiation of hematopoietic stem cells into cells of cardiovascular lineage. Additionally, bone marrow-derived stem cells potentially contribute to the cardiovascular cell population and have shown potential as a therapeutic approach to treat cardiovascular diseases. However, the underlying regulatory mechanisms are currently debatable. This review focuses on some key transcription factors and associated epigenetic modifications that modulate the maintenance and differentiation of hematopoietic stem cells and cardiac progenitor cells. In addition to this, we aim to summarize different potential clinical therapeutic approaches in cardiac regeneration therapy and recent discoveries in stem cell-based transplantation.
Collapse
Affiliation(s)
| | | | | | - Praphulla Chandra Shukla
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
37
|
Kaur N, Raja R, Ruiz-Velasco A, Liu W. Cellular Protein Quality Control in Diabetic Cardiomyopathy: From Bench to Bedside. Front Cardiovasc Med 2020; 7:585309. [PMID: 33195472 PMCID: PMC7593653 DOI: 10.3389/fcvm.2020.585309] [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: 07/20/2020] [Accepted: 09/09/2020] [Indexed: 12/14/2022] Open
Abstract
Heart failure is a serious comorbidity and the most common cause of mortality in diabetes patients. Diabetic cardiomyopathy (DCM) features impaired cellular structure and function, culminating in heart failure; however, there is a dearth of specific clinical therapy for treating DCM. Protein homeostasis is pivotal for the maintenance of cellular viability under physiological and pathological conditions, particularly in the irreplaceable cardiomyocytes; therefore, it is tightly regulated by a protein quality control (PQC) system. Three evolutionarily conserved molecular processes, the unfolded protein response (UPR), the ubiquitin-proteasome system (UPS), and autophagy, enhance protein turnover and preserve protein homeostasis by suppressing protein translation, degrading misfolded or unfolded proteins in cytosol or organelles, disposing of damaged and toxic proteins, recycling essential amino acids, and eliminating insoluble protein aggregates. In response to increased cellular protein demand under pathological insults, including the diabetic condition, a coordinated PQC system retains cardiac protein homeostasis and heart performance, on the contrary, inappropriate PQC function exaggerates cardiac proteotoxicity with subsequent heart dysfunction. Further investigation of the PQC mechanisms in diabetes propels a more comprehensive understanding of the molecular pathogenesis of DCM and opens new prospective treatment strategies for heart disease and heart failure in diabetes patients. In this review, the function and regulation of cardiac PQC machinery in diabetes mellitus, and the therapeutic potential for the diabetic heart are discussed.
Collapse
Affiliation(s)
- Namrita Kaur
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| | - Rida Raja
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| | - Andrea Ruiz-Velasco
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| | - Wei Liu
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
38
|
Shimizu T, Taguchi A, Higashijima Y, Takubo N, Kanki Y, Urade Y, Wada Y. PERK-Mediated Suppression of microRNAs by Sildenafil Improves Mitochondrial Dysfunction in Heart Failure. iScience 2020; 23:101410. [PMID: 32768667 PMCID: PMC7378464 DOI: 10.1016/j.isci.2020.101410] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/03/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
Oxidative/nitrosative stress is a major trigger of cardiac dysfunction, involving the unfolded protein response and mitochondrial dysfunction. Activation of nitric oxide-cyclic guanosine monophosphate-protein kinase G signaling by sildenafil improves cardiac mal-remodeling during pressure-overload-induced heart failure. Transcriptome analysis was conducted in failing hearts with or without sildenafil treatment. Protein kinase R-like endoplasmic reticulum (ER) kinase (PERK) downstream signaling pathways, EIF2 and NRF2, were significantly altered. Although EIF2 signaling was suppressed, NRF2 signaling was upregulated, inhibiting the maturation of miR 24-3p through EGFR-mediated Ago2 phosphorylation. To study the effect of sildenafil on these pathways, we generated cardiac-specific PERK knockout mice. In these mice, sildenafil could not inhibit the maturations, the nuclear translocation of NRF2 was suppressed, and mitochondrial dysfunction advanced. Altogether, these results show that PERK-mediated suppression of miRNAs by sildenafil is vital for maintaining mitochondrial homeostasis through NRF2-mediated oxidative stress response.
Collapse
Affiliation(s)
- Takashi Shimizu
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan; Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.
| | - Akashi Taguchi
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan
| | - Yoshiki Higashijima
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan; Department of Bioinformational Pharmacology, Tokyo Medical and Dental University, Tokyo 113-8510, Japan; Department of Proteomics, The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark
| | - Naoko Takubo
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan
| | - Yasuharu Kanki
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan
| | - Yoshihiro Urade
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan
| | - Youichiro Wada
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan
| |
Collapse
|
39
|
Zhang Q, Feng R, Chaudhary O, Mahmood E, Baribeau Y, Rashid R, Khabbaz KR, Chu LM, Liu DC, Senthilnathan V, Cassavaugh J, Mahmood F, Robson SC, Matyal R. Cardiopulmonary Bypass Suppresses Forkhead Box O3 and Downstream Autophagy in the Diabetic Human Heart. Ann Thorac Surg 2020; 111:937-944. [PMID: 32712101 DOI: 10.1016/j.athoracsur.2020.05.142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 05/07/2020] [Accepted: 05/18/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Autophagy is an integral component of cellular homeostasis and metabolism. The exact mechanism of impaired autophagy in diabetes mellitus is unknown. Forkhead Box O3 (FOXO3α) is a key regulator of oxidative stress-related responses. We hypothesize FOXO3α is a direct upstream regulator of the autophagy pathway, and its upregulation is compromised in diabetic patients during stress of cardiopulmonary bypass (CPB). METHODS The study enrolled 32 diabetic and 33 nondiabetic patients undergoing a cardiac surgical procedure on CPB. Right atrial tissue and serum samples were collected before and after CPB per protocol. A set of key components were quantitatively assessed and compared by microarray, immunoblotting, and immunohistochemistry studies. Data were analyzed using paired or unpaired student test. A P of <.05 or less was considered significant. RESULTS Serum microarray showed FOXO3α was upregulated in the diabetic vs nondiabetic group after CPB (P = .033), autophagy-related 4B gene and Beclin 1 gene were greatly upregulated in the nondiabetic group (P = .028 and P = .002, respectively). On immunoblotting, there was upregulation of FOXO3α in the nondiabetic patients after CPB (P = .003). There were increased levels of Beclin-1, Bcl-2, and light chain 3B after CPB in the nondiabetic group only (P = .016, P = .005, P = .002, respectively). Sirtuin 1, Unc-51-like autophagy activating kinase 1 (ULK1), peroxisome proliferator-activated receptor gamma coactivator 1α (PGC1α), and mammalian target of rapamycin (mTOR) were not significantly changed in the nondiabetic group after CPB. CONCLUSIONS Compared with nondiabetic patients, there was no significant upregulation of FOXO3α in diabetic patients, which could possibly explain the lack of upregulation of the autophagy process after CPB. FOXO3α could potentially serve as a therapeutic target to improve cellular homeostasis.
Collapse
Affiliation(s)
- Qianqian Zhang
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Ruby Feng
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Omar Chaudhary
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Eitezaz Mahmood
- Department of Cardiothoracic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Yanick Baribeau
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Rayan Rashid
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Kamal R Khabbaz
- Department of Cardiothoracic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Louis M Chu
- Department of Cardiothoracic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - David C Liu
- Department of Cardiothoracic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Venkatachalam Senthilnathan
- Department of Cardiothoracic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Jessica Cassavaugh
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Feroze Mahmood
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Simon C Robson
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Robina Matyal
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
40
|
Gao W, Guo N, Zhao S, Chen Z, Zhang W, Yan F, Liao H, Chi K. Carboxypeptidase A4 promotes cardiomyocyte hypertrophy through activating PI3K-AKT-mTOR signaling. Biosci Rep 2020; 40:BSR20200669. [PMID: 32347291 PMCID: PMC7214395 DOI: 10.1042/bsr20200669] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 11/28/2022] Open
Abstract
Carboxypeptidase A4 (CPA4) is a member of the metallocarboxypeptidase family. Current studies have identified the roles of CPA4 in cancer biology and insulin sensitivity. However, the roles of CPA4 in other diseases are not known. In the present study, we investigated the roles of CPA4 in cardiac hypertrophy. The expression of CPA4 was significantly increased in the hypertrophic heart tissues of human patients and isoproterenol (ISO)-induced hypertrophic heart tissues of mice. We next knocked down Cpa4 with shRNA or overexpressed Cpa4 using adenovirus in neonatal rat cardiomyocytes and induced cardiomyocyte hypertrophy with ISO. We observed that Cpa4 overexpression promoted whereas Cpa4 knockdown reduced ISO-induced growth of cardiomyocyte size and overexpression of hypertrophy marker genes, such as myosin heavy chain β (β-Mhc), atrial natriuretic peptide (Anp), and brain natriuretic peptide (Bnp). Our further mechanism study revealed that the mammalian target of rapamycin (mTOR) signaling was activated by Cpa4 in cardiomyocytes, which depended on the phosphoinositide 3-kinase (PI3K)-AKT signaling. Besides, we showed that the PI3K-AKT-mTOR signaling was critically involved in the roles of Cpa4 during cardiomyocyte hypertrophy. Collectively, these results demonstrated that CPA4 is a regulator of cardiac hypertrophy by activating the PI3K-AKT-mTOR signaling, and CPA4 may serve as a promising target for the treatment of hypertrophic cardiac diseases.
Collapse
Affiliation(s)
- Weinian Gao
- Department of Cardiac Macrovascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Na Guo
- Department of Cardiology, Shijiazhuang Translational Chinese Medicine Hospital, Shijiazhuang 050000, China
| | - Shuguang Zhao
- Department of Cardiac Macrovascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Ziying Chen
- Department of Cardiac Macrovascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Wenli Zhang
- Department of Cardiac Macrovascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Fang Yan
- Department of Cardiac Macrovascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Hongjuan Liao
- Department of Cardiac Macrovascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Kui Chi
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| |
Collapse
|
41
|
Su X, Tian J, Li B, Zhou L, Kang H, Pei Z, Zhang M, Li C, Wu M, Wang Q, Han B, Chu C, Pang Y, Ning J, Zhang B, Niu Y, Zhang R. Ambient PM2.5 caused cardiac dysfunction through FoxO1-targeted cardiac hypertrophy and macrophage-activated fibrosis in mice. CHEMOSPHERE 2020; 247:125881. [PMID: 31978653 DOI: 10.1016/j.chemosphere.2020.125881] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/31/2019] [Accepted: 01/08/2020] [Indexed: 06/10/2023]
Abstract
Plenty of epidemiological evidences have shown that ambient particulate matter (PM2.5) exposure increased the prevalence of cardiovascular disease, but the potential mechanism has not been known clearly. We established mice models by ambient PM2.5 exposure system to explore the adverse effects of PM2.5 on cardiac function in mice. Forty-eight C57BL/6 mice were randomly divided into 3 groups and exposed to filtered air (FA), unfiltered air (UA) and concentrated PM2.5 air (CA) for 8 or 16 weeks, 6 hours per day, 7 days per week, respectively. The changes of cardiac structure and function, histological analysis and related mechanism were investigated. The main manifestations of cardiac structure were cardiac hypertrophy and fibrosis in a dose- and time-dependent manner after PM2.5 exposure, which led to the decrease of cardiac systolic function. Cardiac hypertrophy in mice might be regulated by PI3K/Akt/FoxO1 signal. Cardiac fibrosis might be attributed to inflammatory infiltration caused by macrophage activation. Consequently, our data indicated that cardiac hypertrophy and fibrosis might be important factors of PM2.5-induced cardiac dysfunction in mice.
Collapse
Affiliation(s)
- Xuan Su
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Junzhi Tian
- Department of Physical Examination, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, PR China
| | - Binghua Li
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Lixiao Zhou
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Hui Kang
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Zijie Pei
- Department of Pathology, Medical School, China Three Gorge University, Yichang, 443002, PR China
| | - Mengyue Zhang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Chen Li
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Mengqi Wu
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Qian Wang
- Department of Experimental Center, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Bin Han
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Chen Chu
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Yaxian Pang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Jie Ning
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Boyuan Zhang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Yujie Niu
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, PR China
| | - Rong Zhang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, 050017, PR China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, PR China.
| |
Collapse
|
42
|
Battaglini D, Robba C, Lopes da Silva A, Dos Santos Samary C, Leme Silva P, Dal Pizzol F, Pelosi P, Rocco PRM. Brain-heart interaction after acute ischemic stroke. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:163. [PMID: 32317013 PMCID: PMC7175494 DOI: 10.1186/s13054-020-02885-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/13/2020] [Indexed: 02/07/2023]
Abstract
Early detection of cardiovascular dysfunctions directly caused by acute ischemic stroke (AIS) has become paramount. Researchers now generally agree on the existence of a bidirectional interaction between the brain and the heart. In support of this theory, AIS patients are extremely vulnerable to severe cardiac complications. Sympathetic hyperactivity, hypothalamic-pituitary-adrenal axis, the immune and inflammatory responses, and gut dysbiosis have been identified as the main pathological mechanisms involved in brain-heart axis dysregulation after AIS. Moreover, evidence has confirmed that the main causes of mortality after AIS include heart attack, congestive heart failure, hemodynamic instability, left ventricular systolic dysfunction, diastolic dysfunction, arrhythmias, electrocardiographic anomalies, and cardiac arrest, all of which are more or less associated with poor outcomes and death. Therefore, intensive care unit admission with continuous hemodynamic monitoring has been proposed as the standard of care for AIS patients at high risk for developing cardiovascular complications. Recent trials have also investigated possible therapies to prevent secondary cardiovascular accidents after AIS. Labetalol, nicardipine, and nitroprusside have been recommended for the control of hypertension during AIS, while beta blockers have been suggested both for preventing chronic remodeling and for treating arrhythmias. Additionally, electrolytic imbalances should be considered, and abnormal rhythms must be treated. Nevertheless, therapeutic targets remain challenging, and further investigations might be essential to complete this complex multi-disciplinary puzzle. This review aims to highlight the pathophysiological mechanisms implicated in the interaction between the brain and the heart and their clinical consequences in AIS patients, as well as to provide specific recommendations for cardiovascular management after AIS.
Collapse
Affiliation(s)
- Denise Battaglini
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, Genoa, Italy.,Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genoa, Italy
| | - Chiara Robba
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, Genoa, Italy
| | - Adriana Lopes da Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cynthia Dos Santos Samary
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Physiotherapy, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro Leme Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Felipe Dal Pizzol
- Unidade Acadêmica de Ciências da Saude, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, Santa Catarina, Brazil
| | - Paolo Pelosi
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, Genoa, Italy.,Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genoa, Italy
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil. .,Rio de Janeiro Network on Neuroinflammation, Carlos Chagas Filho Foundation for Supporting Research in the State of Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil.
| |
Collapse
|
43
|
Hao Q, Zhang F, Wang Y, Li Y, Qi X. Cardiac Contractility Modulation Attenuates Chronic Heart Failure in a Rabbit Model via the PI3K/AKT Pathway. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1625362. [PMID: 31998779 PMCID: PMC6973194 DOI: 10.1155/2020/1625362] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 11/27/2019] [Indexed: 01/01/2023]
Abstract
The Akt plays an important role in regulating cardiac growth, myocardial angiogenesis, and cell death in cardiac myocytes. However, there are few studies to focus on the responses of the Akt pathway to cardiac contractility modulation (CCM) in a chronic heart failure (HF) model. In this study, the effects of CCM on the treatment of HF in a rabbit model were investigated. Thirty six-month-old rabbits were randomly separated into control, HF, and CCM groups. The rabbits in HF and CCM groups were pressure uploaded, which can cause an aortic constriction. Then, CCM was gradually injected to the myocardium of rabbits in the CCM group, and this process lasted for four weeks with six hours per day. Rabbit body weight, heart weight, and heart beating rates were recorded during the experiment. To assess the CCM impacts, rabbit myocardial histology was examined as well. Additionally, western blot analysis was employed to measure the protein levels of Akt, FOXO3, Beclin, Pi3k, mTOR, GSK-3β, and TORC2 in the myocardial histology of rabbits. Results showed that the body and heart weight of rabbits decreased significantly after suffering HF when compared with those in the control group. However, they gradually recovered after CCM application. The CCM significantly decreased collagen volume fraction in myocardial histology of HF rabbits, indicating that CCM therapy attenuated myocardial fibrosis and collagen deposition. The levels of Akt, FOXO3, Beclin, mTOR, GSK-3β, and TORC2 were significantly downregulated, but Pi3k concentration was greatly upregulated after CCM utilization. Based on these findings, it was concluded that CCM could elicit positive effects on HF therapy, which was potentially due to the variation in the Pi3k/Akt signaling pathway.
Collapse
Affiliation(s)
- Qingqing Hao
- School of Graduate, Hebei Medical University, Shijiazhuang, China
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, China
| | - Feifei Zhang
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, China
| | - Yudan Wang
- School of Graduate, Hebei Medical University, Shijiazhuang, China
| | - Yingxiao Li
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, China
| | - Xiaoyong Qi
- School of Graduate, Hebei Medical University, Shijiazhuang, China
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
44
|
Li L, Li X, Zhang Z, Liu L, Liu T, Li S, Liu S, Zhou Y, Liu F. Effects of Hydrogen-rich Water on the PI3K/AKT Signaling Pathway in Rats with Myocardial Ischemia-reperfusion Injury. Curr Mol Med 2019; 20:396-406. [PMID: 31702499 DOI: 10.2174/1566524019666191105150709] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/15/2019] [Accepted: 10/21/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND The effects of hydrogen-rich water on PI3K/AKT-mediated apoptosis were studied in rats subjected to myocardial ischemia-reperfusion injury (MIRI). Methdos: Sixty rats were divided randomly into a hydrogen-rich water group and a control group. The hearts were removed and fixed in a Langendorff device. Hearts from the control group were perfused with K-R solution, and hearts from the hydrogen-rich water group was perfused with K-R solution + hydrogen-rich water. The two treatment groups were then divided randomly into pre-ischemic period, ischemic period and reperfusion period groups(10 rats per group), which were subjected to reverse perfusion for 10 min, normal treatment for 20 min, and reperfusion for 20 min, respectively. The mRNA and protein expression levels of PI3K, AKT, p-AKT, FoxO1, Bim and Caspase-3 in each group were detected by RT-qPCR, immunohistochemistry (IHC) and Western blotting. Caspase-3 activity was detected by spectrophotometry. RESULTS Among the hydrogen-rich water group, the PI3K/AKT signaling pathway was significantly activated, and FoxO1, Bim, and Caspase-3 mRNA and protein levels were significantly decreased in ischemia-reperfusion subgroup compared with the preischemic and ischemic subgroups. In the ischemia-reperfusion hydrogen-rich water group, PI3K, AKT and p-AKT mRNA and protein expression levels were increased while the FoxO1, Bim and Caspase-3 expression levels were significantly decreased compared with those in the corresponding control group (P<0.05). CONCLUSION Hydrogen-rich water can activate the PI3K/AKT signaling pathway, alleviate ischemia-reperfusion injury in isolated rat hearts, and inhibit cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Liangtong Li
- Medical College, Hebei University, Baoding, 071000, China
| | - Xiangzi Li
- Medical College, Hebei University, Baoding, 071000, China
| | - Zhe Zhang
- Medical College, Hebei University, Baoding, 071000, China
| | - Li Liu
- Medical College, Hebei University, Baoding, 071000, China
| | - Tongtong Liu
- Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Shaochun Li
- Medical College, Hebei University, Baoding, 071000, China
| | - Sen Liu
- Medical College, Hebei University, Baoding, 071000, China
| | - Yujuan Zhou
- Medical College, Hebei University, Baoding, 071000, China
| | - Fulin Liu
- Affiliated Hospital of Hebei University, Baoding, 071000, China
| |
Collapse
|
45
|
Kwak HJ, Um JY, Lee SS. Mild NO preconditioning protects H9c2 cells against NO-induced apoptosis through activation of PI3K/Akt and PKA-dependent pathways. Mol Cell Toxicol 2019. [DOI: 10.1007/s13273-019-0033-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
46
|
Aubert G, Barefield DY, Demonbreun AR, Ramratnam M, Fallon KS, Warner JL, Rossi AE, Hadhazy M, Makielski JC, McNally EM. Deletion of Sulfonylurea Receptor 2 in the Adult Myocardium Enhances Cardiac Glucose Uptake and Is Cardioprotective. JACC Basic Transl Sci 2019; 4:251-268. [PMID: 31061927 PMCID: PMC6488756 DOI: 10.1016/j.jacbts.2018.11.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/24/2018] [Accepted: 11/26/2018] [Indexed: 11/05/2022]
Abstract
In the heart, SUR2 couples with a potassium channel to form an adenosine triphosphate–sensitive complex that responds to the energy state of the cell. The authors deleted SUR2 in adult cardiomyocytes and found a shift of the heart toward glycolytic metabolism, which is protective under cardiac stress. SUR2 was found to complex with glucose transporter type 4, the major glucose transporter. Drugs that antagonize the SUR2 receptor may be cardioprotective and useful for managing heart failure.
The adult myocardium relies on oxidative metabolism. In ischemic myocardium, such as the embryonic heart, glycolysis contributes more prominently as a fuel source. The sulfonylurea receptor 2 (SUR2) was previously implicated in the normal myocardial transition from glycolytic to oxidative metabolism that occurs during adaptation to postnatal life. This receptor was now selectively deleted in adult mouse myocardium resulting in protection from ischemia reperfusion injury. SUR2-deleted cardiomyocytes had enhanced glucose uptake, and SUR2 forms a complex with the major glucose transporter. These data identify the SUR2 receptor as a target to shift cardiac metabolism to protect against myocardial injury.
Collapse
Key Words
- 2DG, 2-deoxy-D-glucose
- ABCC9
- EDTA, ethylenediaminetetraacetic acid
- FL Ex5, LoxP sites flanking exon 5
- GFP, green fluorescent protein
- GLUT, glucose transporter
- HEK293T, human embryonic kidney 293T
- KATP, adenosine triphosphate–sensitive potassium
- Kir, inward rectifying potassium channel
- LVDP, left ventricular developed pressure
- MCM, αMHC-MerCreMer
- PCR, polymerase chain reaction
- SUR, sulfonylurea receptor
- ischemia
- potassium ATP channels
- sulfonylurea
Collapse
Affiliation(s)
- Gregory Aubert
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago Illinois
| | - David Y Barefield
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago Illinois
| | - Alexis R Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago Illinois
| | - Mohun Ramratnam
- Division of Cardiology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Katherine S Fallon
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago Illinois
| | - James L Warner
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago Illinois
| | - Ann E Rossi
- Section of Cardiology, University of Chicago, Chicago Illinois
| | - Michele Hadhazy
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago Illinois
| | - Jonathan C Makielski
- Division of Cardiology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | | |
Collapse
|
47
|
Bakuchiol: A newly discovered warrior against organ damage. Pharmacol Res 2019; 141:208-213. [DOI: 10.1016/j.phrs.2019.01.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/24/2018] [Accepted: 01/01/2019] [Indexed: 12/17/2022]
|
48
|
Sárközy M, Gáspár R, Zvara Á, Siska A, Kővári B, Szűcs G, Márványkövi F, Kovács MG, Diószegi P, Bodai L, Zsindely N, Pipicz M, Gömöri K, Kiss K, Bencsik P, Cserni G, Puskás LG, Földesi I, Thum T, Bátkai S, Csont T. Chronic kidney disease induces left ventricular overexpression of the pro-hypertrophic microRNA-212. Sci Rep 2019; 9:1302. [PMID: 30718600 PMCID: PMC6362219 DOI: 10.1038/s41598-018-37690-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 12/12/2018] [Indexed: 12/22/2022] Open
Abstract
Chronic kidney disease (CKD) is a public health problem that increases the risk of cardiovascular morbidity and mortality. Heart failure with preserved ejection fraction (HFpEF) characterized by left ventricular hypertrophy (LVH) and diastolic dysfunction is a common cardiovascular complication of CKD. MicroRNA-212 (miR-212) has been demonstrated previously to be a crucial regulator of pathologic LVH in pressure-overload-induced heart failure via regulating the forkhead box O3 (FOXO3)/calcineurin/nuclear factor of activated T-cells (NFAT) pathway. Here we aimed to investigate whether miR-212 and its hypertrophy-associated targets including FOXO3, extracellular signal-regulated kinase 2 (ERK2), and AMP-activated protein kinase (AMPK) play a role in the development of HFpEF in CKD. CKD was induced by 5/6 nephrectomy in male Wistar rats. Echocardiography and histology revealed LVH, fibrosis, preserved systolic function, and diastolic dysfunction in the CKD group as compared to sham-operated animals eight and/or nine weeks later. Left ventricular miR-212 was significantly overexpressed in CKD. However, expressions of FOXO3, AMPK, and ERK2 failed to change significantly at the mRNA or protein level. The protein kinase B (AKT)/FOXO3 and AKT/mammalian target of rapamycin (mTOR) pathways are also proposed regulators of LVH induced by pressure-overload. Interestingly, phospho-AKT/total-AKT ratio was increased in CKD without significantly affecting phosphorylation of FOXO3 or mTOR. In summary, cardiac overexpression of miR-212 in CKD failed to affect its previously implicated hypertrophy-associated downstream targets. Thus, the molecular mechanism of the development of LVH in CKD seems to be independent of the FOXO3, ERK1/2, AMPK, and AKT/mTOR-mediated pathways indicating unique features in this form of LVH.
Collapse
Affiliation(s)
- Márta Sárközy
- Metabolic Diseases and Cell Signaling Group, Department of Biochemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 9, Szeged, H-6720, Hungary.
| | - Renáta Gáspár
- Metabolic Diseases and Cell Signaling Group, Department of Biochemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 9, Szeged, H-6720, Hungary
| | - Ágnes Zvara
- Laboratory for Functional Genomics, Institute of Genetics, Biological Research Center of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6701, Szeged, Hungary
| | - Andrea Siska
- Department of Laboratory Medicine, Faculty of Medicine, University of Szeged, Semmelweis utca 6, Szeged, H-6725, Hungary
| | - Bence Kővári
- Department of Pathology, University of Szeged, Állomás utca 1, Szeged, H-6725, Hungary
| | - Gergő Szűcs
- Metabolic Diseases and Cell Signaling Group, Department of Biochemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 9, Szeged, H-6720, Hungary
| | - Fanni Márványkövi
- Metabolic Diseases and Cell Signaling Group, Department of Biochemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 9, Szeged, H-6720, Hungary
| | - Mónika G Kovács
- Metabolic Diseases and Cell Signaling Group, Department of Biochemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 9, Szeged, H-6720, Hungary
| | - Petra Diószegi
- Metabolic Diseases and Cell Signaling Group, Department of Biochemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 9, Szeged, H-6720, Hungary
| | - László Bodai
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, Szeged, H-6726, Hungary
| | - Nóra Zsindely
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, Szeged, H-6726, Hungary
| | - Márton Pipicz
- Metabolic Diseases and Cell Signaling Group, Department of Biochemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 9, Szeged, H-6720, Hungary
| | - Kamilla Gömöri
- Cardiovascular Research Group, Department of Biochemistry, Faculty of Medicine, University of Szeged, Dóm tér 9, Szeged, H-6720, Hungary
| | - Krisztina Kiss
- Cardiovascular Research Group, Department of Biochemistry, Faculty of Medicine, University of Szeged, Dóm tér 9, Szeged, H-6720, Hungary
| | - Péter Bencsik
- Cardiovascular Research Group, Department of Biochemistry, Faculty of Medicine, University of Szeged, Dóm tér 9, Szeged, H-6720, Hungary
| | - Gábor Cserni
- Department of Pathology, University of Szeged, Állomás utca 1, Szeged, H-6725, Hungary
| | - László G Puskás
- Laboratory for Functional Genomics, Institute of Genetics, Biological Research Center of the Hungarian Academy of Sciences, Temesvári krt. 62, H-6701, Szeged, Hungary
| | - Imre Földesi
- Department of Laboratory Medicine, Faculty of Medicine, University of Szeged, Semmelweis utca 6, Szeged, H-6725, Hungary
| | - Thomas Thum
- IMTTS, Hannover Medical School, Carl-Neuberg Strasse 1, Hannover, 30625, Germany
| | - Sándor Bátkai
- IMTTS, Hannover Medical School, Carl-Neuberg Strasse 1, Hannover, 30625, Germany
| | - Tamás Csont
- Metabolic Diseases and Cell Signaling Group, Department of Biochemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 9, Szeged, H-6720, Hungary
| |
Collapse
|
49
|
Blice-Baum AC, Vogler G, Viswanathan MC, Trinh B, Limpitikul WB, Cammarato A. Quantifying Tissue-Specific Overexpression of FOXO in Drosophila via mRNA Fluorescence In Situ Hybridization Using Branched DNA Probe Technology. Methods Mol Biol 2019; 1890:171-190. [PMID: 30414154 PMCID: PMC7906431 DOI: 10.1007/978-1-4939-8900-3_15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
While the highly conserved FOXO transcription factors have been studied in Drosophila melanogaster for decades, the ability to accurately control and measure their tissue-specific expression is often cumbersome due to a lack of reagents and to limited, nonhomogeneous samples. The need for quantitation within a distinct cell type is particularly important because transcription factors must be expressed in specific amounts to perform their functions properly. However, the inherent heterogeneity of many samples can make evaluating cell-specific FOXO and/or FOXO load difficult. Here, we describe an extremely sensitive fluorescence in situ hybridization (FISH) approach for visualizing and quantifying multiple mRNAs with single-cell resolution in adult Drosophila cardiomyocytes. The procedure relies upon branched DNA technology, which allows several fluorescent molecules to label an individual transcript, drastically increasing the signal-to-noise ratio compared to other FISH assays. This protocol can be modified for use in various small animal models, tissue types, and for assorted nucleic acids.
Collapse
Affiliation(s)
- Anna C Blice-Baum
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Science Department, Iadarola Center for Science, Education and Technology, Cabrini University, Radnor, PA, USA.
| | - Georg Vogler
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| | - Meera C Viswanathan
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bosco Trinh
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Worawan B Limpitikul
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anthony Cammarato
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
50
|
Duan P, Wang J, Li Y, Wei S, Su F, Zhang S, Duan Y, Wang L, Zhu Q. Opening of mitoKATP improves cardiac function and inhibits apoptosis via the AKT-Foxo1 signaling pathway in diabetic cardiomyopathy. Int J Mol Med 2018; 42:2709-2719. [PMID: 30132505 PMCID: PMC6192784 DOI: 10.3892/ijmm.2018.3832] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 08/16/2018] [Indexed: 12/21/2022] Open
Abstract
Decreasing phosphorylation of AKT-Foxo1 is closely associated with the onset of insulin resistance and apoptosis during diabetic cardiomyopathy (DCM). Opening of mitochondrial ATP-sensitive potassium channels (mitoKATP) increases the expression of p-AKT in the process of reperfusion injury. It was therefore hypothesized that opening of mitoKATP may regulate the AKT-Foxo1 signaling pathway and improve cardiac function in DCM. In the present study, opening of mitoKATP by diazoxide (DZX) was found to improve cardiac function and attenuate cardiomyocyte apoptosis in db/db mice. DZX also significantly increased the expression of p-AKT and p-Foxo1. Similarly, DZX decreased the expression of the heart failure marker NT-proBNP, increased mitochondrial membrane potential, inhibited apoptosis, and increased the expression of p-AKT and p-Foxo1 when mimicking insulin resistance in cultured cardiomyocytes. Moreover, the protective effects of DZX were completely blocked by the specific AKT inhibitor MK-2206. These data suggest that the regulation of the AKT-Foxo1 signaling pathway by mitoKATP plays an important role in improving cardiac function and inhibiting apoptosis in DCM, and may therefore be a new potential therapeutic target for DCM.
Collapse
Affiliation(s)
- Peng Duan
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Jinxin Wang
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Yang Li
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Shiqiang Wei
- Department of Cardiology, Chinese PLA No. 371 Hospital, Xinxiang, Henan 453000, P.R. China
| | - Feng Su
- Department of Medical Administration, Chinese PLA No. 371 Hospital, Xinxiang, Henan 453000, P.R. China
| | - Sanlin Zhang
- Department of Cardiology, Chinese PLA No. 371 Hospital, Xinxiang, Henan 453000, P.R. China
| | - Yuhui Duan
- Department of Cardiology, Chinese PLA No. 371 Hospital, Xinxiang, Henan 453000, P.R. China
| | - Lin Wang
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Qinglei Zhu
- Department of Cardiology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| |
Collapse
|