1
|
Liu C, Zhang D, Long K, Qi W, Pang L, Li J, Cheng KKY, Cai Y. From exosomes to mitochondria and myocardial infarction: Molecular insight and therapeutic challenge. Pharmacol Res 2024; 209:107468. [PMID: 39426469 DOI: 10.1016/j.phrs.2024.107468] [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: 08/12/2024] [Revised: 09/21/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
Myocardial infarction (MI) remains a leading cause of mortality worldwide. Despite patients with MI benefit from timely reperfusion therapies, the rates of mortality and morbidity remain substantial, suggesting an enduring need for the development of new approaches. Molecular mechanisms underlying myocardial ischemic injury are associated with both cardiomyocytes and non-cardiomyocytes. Exosomes are nano-sized extracellular vesicles released by almost all eukaryotic cells. They facilitate the communication between various cells by transferring information via their cargo and altering different biological activities in recipient cells. Studies have created great prospects for therapeutic applications of exosomes in MI, as demonstrated through their beneficial effect on heart function and reducing ventricular remodeling in association with fibrosis, angiogenesis, apoptosis, and inflammation. Of note, myocardial ischemic injury is primarily due to restricted blood flow, reducing oxygen availability, and causing inefficient utilization of energy substrates. However, the impact of exosomes on cardiac energy metabolism has not been adequately investigated. Although exosomes have been engineered for targeted delivery to enhance clinical efficacy, challenges must be overcome to utilize them reliably in the clinic. In this review, we summarize the research progress of exosomes for MI with a focus on the known and unknown regarding the role of exosomes in energy metabolism in cardiomyocytes and non-cardiomyocytes; as well as potential research avenues of exosome-mitochondrial energy regulation as well as therapeutic challenges. We aim to help identify more efficient molecular targets that may promote the clinical application of exosomes.
Collapse
Affiliation(s)
- Chang Liu
- Department of Anesthesiology, The First Hospital of Jilin University, Jilin, China; Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Dengwen Zhang
- Department of Anesthesiology, Heyuan People's Hospital, Guangdong, China; Department of Anesthesiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangdong, China
| | - Kekao Long
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Wensheng Qi
- Department of Anesthesiology, The First Hospital of Jilin University, Jilin, China; Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Lei Pang
- Department of Anesthesiology, The First Hospital of Jilin University, Jilin, China
| | - Jia Li
- Department of Neurology, Wuhan No.1 Hospital, Hubei, China
| | - Kenneth King-Yip Cheng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China.
| | - Yin Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China; Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong SAR, China; Research Institute for Future Food, The Hong Kong Polytechnic University, Hong Kong SAR, China.
| |
Collapse
|
2
|
Muhammad J, Erlwanger KH, Ibrahim KG, Mokotedi L. Effects of voluntarily consumed sweetened alcohol and naringin on cardiac function in male and female Sprague-Dawley rats. Physiol Rep 2024; 12:e70030. [PMID: 39245811 PMCID: PMC11381194 DOI: 10.14814/phy2.70030] [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: 03/13/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/10/2024] Open
Abstract
This study assessed the impact of sweetened alcohol and naringin on cardiac function in Sprague-Dawley rats. Male (n = 40) and female (n = 40) rats were allocated to control, sweetened alcohol (SOH), naringin (NA), and sweetened alcohol with naringin (SOH + NA) groups. SOH and SOH + NA rats received 10% alcohol + 20% fructose in gelatine; SOH + NA and NA rats received 50 mg/kg naringin in gelatine daily for 10 weeks. Echocardiography was performed to assess left ventricular (LV) function. LV cardiomyocyte diameters and collagen area fraction were determined by H&E and picrosirius-red staining, respectively. In males, sweetened alcohol and naringin did not affect cardiac function. Female SOH rats had increased LV end-diastolic posterior wall (p = 0.04), relative wall thicknesses (p = 0.01), and LV cardiomyocyte diameters (p = 0.005) compared with control. Female SOH and SOH + NA had reduced lateral e' and e'/a' and increased E/e' (p < 0.0001). Female SOH (p = 0.01) and SOH + NA (p = 0.04) rats had increased LV collagen area fraction compared with controls. In males, neither sweetened alcohol nor naringin affected cardiac geometry or diastolic function. In females, sweetened alcohol induced concentric remodelling, impaired LV relaxation, and elevated filling pressures. Naringin may have the potential to improve the sweetened alcohol-induced concentric remodelling; however, it did not ameliorate diastolic dysfunction in females.
Collapse
Affiliation(s)
- Jelani Muhammad
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Federal University Birnin Kebbi, Birnin Kebbi, Kebbi State, Nigeria
| | - Kennedy H Erlwanger
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Kasimu G Ibrahim
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Basic Medical and Dental Sciences, Faculty of Dentistry, Zarqa University, Zarqa, Jordan
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Lebogang Mokotedi
- Department of Physiology, School of Medicine, Sefako Makgatho Health Sciences University, Ga-Rankuwa, South Africa
- Integrated Molecular Physiology Research Initiative (IMPRI), School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
3
|
Zhou L, Nishimura A, Umezawa K, Kato Y, Mi X, Ito T, Urano Y, Akaike T, Nishida M. Supersulfide catabolism participates in maladaptive remodeling of cardiac cells. J Pharmacol Sci 2024; 155:121-130. [PMID: 38880546 DOI: 10.1016/j.jphs.2024.05.002] [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: 03/06/2024] [Revised: 04/29/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
The atrophic myocardium resulting from mechanical unloading and nutritional deprivation is considered crucial as maladaptive remodeling directly associated with heart failure, as well as interstitial fibrosis. Conversely, myocardial hypertrophy resulting from hemodynamic loading is perceived as compensatory stress adaptation. We previously reported the abundant presence of highly redox-active polysulfide molecules, termed supersulfide, with two or more sulfur atoms catenated in normal hearts, and the supersulfide catabolism in pathologic hearts after myocardial infarction correlated with worsened prognosis of heart failure. However, the impact of supersulfide on myocardial remodeling remains unclear. Here, we investigated the involvement of supersulfide metabolism in cardiomyocyte remodeling, using a model of adenosine 5'-triphosphate (ATP) receptor-stimulated atrophy and endothelin-1 receptor-stimulated hypertrophy in neonatal rat cardiomyocytes. Results revealed contrasting changes in intracellular supersulfide and its catabolite, hydrogen sulfide (H2S), between cardiomyocyte atrophy and hypertrophy. Stimulation of cardiomyocytes with ATP decreased supersulfide activity, while H2S accumulation itself did not affect cardiomyocyte atrophy. This supersulfide catabolism was also involved in myofibroblast formation of neonatal rat cardiac fibroblasts. Thus, unraveling supersulfide metabolism during myocardial remodeling may lead to the development of novel therapeutic strategies to improve heart failure.
Collapse
Affiliation(s)
- Liuchenzi Zhou
- National Institute for Physiological Sciences, National Institutes of Natural Sciences (NINS), Okazaki, 444-8787, Japan; Exploratory Research Center on Life and Living Systems, NINS, Okazaki, 444-8787, Japan; SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8787, Japan
| | - Akiyuki Nishimura
- National Institute for Physiological Sciences, National Institutes of Natural Sciences (NINS), Okazaki, 444-8787, Japan; Exploratory Research Center on Life and Living Systems, NINS, Okazaki, 444-8787, Japan; SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8787, Japan
| | - Keitaro Umezawa
- Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Yuri Kato
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Xinya Mi
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Tomoya Ito
- National Institute for Physiological Sciences, National Institutes of Natural Sciences (NINS), Okazaki, 444-8787, Japan; Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yasuteru Urano
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan; Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Takaaki Akaike
- Graduate School of Medicine, Tohoku University, Sendai, 980-8575, Japan
| | - Motohiro Nishida
- National Institute for Physiological Sciences, National Institutes of Natural Sciences (NINS), Okazaki, 444-8787, Japan; Exploratory Research Center on Life and Living Systems, NINS, Okazaki, 444-8787, Japan; SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8787, Japan; Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| |
Collapse
|
4
|
Navarro Cáceres A, Navarro-Matías E, Gómez-Sánchez M, Tamayo-Morales O, Lugones-Sánchez C, González-Sánchez S, Rodríguez-Sánchez E, García-Ortiz L, Gómez-Sánchez L, Gómez-Marcos MA. Increase in Vascular Function Parameters According to Lifestyles in a Spanish Population without Previous Cardiovascular Disease-EVA Follow-Up Study. Nutrients 2023; 15:4614. [PMID: 37960267 PMCID: PMC10648779 DOI: 10.3390/nu15214614] [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: 10/03/2023] [Revised: 10/25/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
The aim of this longitudinal descriptive observational study was to analyze the influence of different lifestyles on arterial stiffness (AS) throughout five years of follow-up and to describe the differences by sex in a Spanish adult population without cardiovascular disease at the start of the study. A random stratified sampling by age and sex was used to obtain 501 subjects included in the initial assessment. No cardiovascular disease was allowed in the subjects. The average age was 55.9 years, and 50.3% were women. A total of 480 subjects were analyzed again five years later. Alcohol and tobacco consumption were collected with standardized questionnaires. Adherence to the Mediterranean diet was assessed with the Mediterranean diet adherence screener (MEDAS) questionnaire. Physical activity was assessed with the short version of the International Physical Activity Questionnaire-Short Form (IPAQ-SF) and sedentary time was assessed with the Marshall Sitting Questionnaire (MSQ). AS was assessed by measuring carotid-femoral pulse wave velocity (cfPWV) and central augmentation index (CAIx) with SphygmoCor System®, and ankle pulse wave velocity (baPWV) and cardio ankle vascular index (CAVI) with Vasera VS-1500®. Increases in vascular function measures per year of follow-up were: cfPWV = 0.228 ± 0.360 m/s, baPWV = 0.186 ± 0.308 m/s, CAVI = 0.041 ± 0.181 m/s, and CAIx = 0.387 ± 2.664 m/s. In multiple regression analysis, positive association was shown between an increase in baPWV and tobacco index (β = 0.007) and alcohol consumption (β = 0.005). Negative association was shown between CAVI and Mediterranean diet score (β = -0.051). In multinomial logistic regression analysis, the OR of tobacco index of subjects with a cfPWV increase >P75 was OR = 1.025 and of subjects classified between P25 and P75 was OR = 1.026 regarding subjects classified with an increase P75 was OR = 1.006 regarding subjects classified with an increase P75, and an OR = 0.841 was found of subjects classified between P25-75 regarding subjects classified with an increase
Collapse
Affiliation(s)
- Alicia Navarro Cáceres
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Institute of Biomedical Research of Salamanca (IBSAL), 37005 Salamanca, Spain; (A.N.C.); (E.N.-M.); (O.T.-M.); (C.L.-S.); (S.G.-S.); (E.R.-S.); (L.G.-O.)
- Castilla and León Health Service-SACYL, Regional Health Management, 37005 Salamanca, Spain
| | - Elena Navarro-Matías
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Institute of Biomedical Research of Salamanca (IBSAL), 37005 Salamanca, Spain; (A.N.C.); (E.N.-M.); (O.T.-M.); (C.L.-S.); (S.G.-S.); (E.R.-S.); (L.G.-O.)
- Castilla and León Health Service-SACYL, Regional Health Management, 37005 Salamanca, Spain
| | - Marta Gómez-Sánchez
- Home Hospitalization Service, Marqués de Valdecilla University Hospital, 39008 Santander, Spain;
| | - Olaya Tamayo-Morales
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Institute of Biomedical Research of Salamanca (IBSAL), 37005 Salamanca, Spain; (A.N.C.); (E.N.-M.); (O.T.-M.); (C.L.-S.); (S.G.-S.); (E.R.-S.); (L.G.-O.)
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), 37005 Salamanca, Spain
| | - Cristina Lugones-Sánchez
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Institute of Biomedical Research of Salamanca (IBSAL), 37005 Salamanca, Spain; (A.N.C.); (E.N.-M.); (O.T.-M.); (C.L.-S.); (S.G.-S.); (E.R.-S.); (L.G.-O.)
- Castilla and León Health Service-SACYL, Regional Health Management, 37005 Salamanca, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), 37005 Salamanca, Spain
| | - Susana González-Sánchez
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Institute of Biomedical Research of Salamanca (IBSAL), 37005 Salamanca, Spain; (A.N.C.); (E.N.-M.); (O.T.-M.); (C.L.-S.); (S.G.-S.); (E.R.-S.); (L.G.-O.)
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), 37005 Salamanca, Spain
| | - Emiliano Rodríguez-Sánchez
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Institute of Biomedical Research of Salamanca (IBSAL), 37005 Salamanca, Spain; (A.N.C.); (E.N.-M.); (O.T.-M.); (C.L.-S.); (S.G.-S.); (E.R.-S.); (L.G.-O.)
- Castilla and León Health Service-SACYL, Regional Health Management, 37005 Salamanca, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), 37005 Salamanca, Spain
- Department of Medicine, University of Salamanca, 28046 Salamanca, Spain
| | - Luis García-Ortiz
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Institute of Biomedical Research of Salamanca (IBSAL), 37005 Salamanca, Spain; (A.N.C.); (E.N.-M.); (O.T.-M.); (C.L.-S.); (S.G.-S.); (E.R.-S.); (L.G.-O.)
- Castilla and León Health Service-SACYL, Regional Health Management, 37005 Salamanca, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), 37005 Salamanca, Spain
- Department of Biomedical and Diagnostic Sciences, University of Salamanca, 37007 Salamanca, Spain
| | - Leticia Gómez-Sánchez
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Institute of Biomedical Research of Salamanca (IBSAL), 37005 Salamanca, Spain; (A.N.C.); (E.N.-M.); (O.T.-M.); (C.L.-S.); (S.G.-S.); (E.R.-S.); (L.G.-O.)
- Emergency Service, University Hospital of La Paz, 37007 Madrid, Spain
| | - Manuel A. Gómez-Marcos
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Institute of Biomedical Research of Salamanca (IBSAL), 37005 Salamanca, Spain; (A.N.C.); (E.N.-M.); (O.T.-M.); (C.L.-S.); (S.G.-S.); (E.R.-S.); (L.G.-O.)
- Castilla and León Health Service-SACYL, Regional Health Management, 37005 Salamanca, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), 37005 Salamanca, Spain
- Department of Medicine, University of Salamanca, 28046 Salamanca, Spain
| | | |
Collapse
|
5
|
Hwang H, Liu R, Eldridge R, Hu X, Forghani P, Jones DP, Xu C. Chronic ethanol exposure induces mitochondrial dysfunction and alters gene expression and metabolism in human cardiac spheroids. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:643-658. [PMID: 36799338 PMCID: PMC10149610 DOI: 10.1111/acer.15026] [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: 12/07/2022] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 02/18/2023]
Abstract
BACKGROUND Chronic alcohol consumption in adults can induce various cardiac toxicities such as arrhythmias, cardiomyopathy, and heart failure. Prenatal alcohol exposure can increase the risk of developing congenital heart defects among offspring. Understanding the molecular mechanisms underlying long-term alcohol exposure-induced cardiotoxicity can help guide the development of therapeutic strategies. METHODS Cardiomyocytes derived from human-induced pluripotent stem cells (hiPSC-CMs) were engineered into cardiac spheroids and treated with clinically relevant concentrations of ethanol (17 and 50 mM) for 5 weeks. The cells were then analyzed for changes in mitochondrial features, transcriptomic and metabolomic profiles, and integrated omics outcomes. RESULTS Following chronic ethanol treatment of hiPSC-CMs, a decrease in mitochondrial membrane potential and respiration and changes in expression of mitochondrial function-related genes were observed. RNA-sequencing analysis revealed changes in various metabolic processes, heart development, response to hypoxia, and extracellular matrix-related activities. Metabolomic analysis revealed dysregulation of energy metabolism and increased metabolites associated with the upregulation of inflammation. Integrated omics analysis further identified functional subclusters and revealed potentially affected pathways associated with cardiac toxicities. CONCLUSION Chronic ethanol treatment of hiPSC-CMs resulted in overall decreased mitochondrial function, increased glycolysis, disrupted fatty acid oxidation, and impaired cardiac structural development.
Collapse
Affiliation(s)
- Hyun Hwang
- Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Rui Liu
- Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Ronald Eldridge
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA 30322, USA
| | - Xin Hu
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Parvin Forghani
- Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Dean P. Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Chunhui Xu
- Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
6
|
Bacova BS, Andelova K, Sykora M, Egan Benova T, Barancik M, Kurahara LH, Tribulova N. Does Myocardial Atrophy Represent Anti-Arrhythmic Phenotype? Biomedicines 2022; 10:2819. [PMID: 36359339 PMCID: PMC9687767 DOI: 10.3390/biomedicines10112819] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/30/2023] Open
Abstract
This review focuses on cardiac atrophy resulting from mechanical or metabolic unloading due to various conditions, describing some mechanisms and discussing possible strategies or interventions to prevent, attenuate or reverse myocardial atrophy. An improved awareness of these conditions and an increased focus on the identification of mechanisms and therapeutic targets may facilitate the development of the effective treatment or reversion for cardiac atrophy. It appears that a decrement in the left ventricular mass itself may be the central component in cardiac deconditioning, which avoids the occurrence of life-threatening arrhythmias. The depressed myocardial contractility of atrophied myocardium along with the upregulation of electrical coupling protein, connexin43, the maintenance of its topology, and enhanced PKCƐ signalling may be involved in the anti-arrhythmic phenotype. Meanwhile, persistent myocardial atrophy accompanied by oxidative stress and inflammation, as well as extracellular matrix fibrosis, may lead to severe cardiac dysfunction, and heart failure. Data in the literature suggest that the prevention of heart failure via the attenuation or reversion of myocardial atrophy is possible, although this requires further research.
Collapse
Affiliation(s)
| | - Katarina Andelova
- Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia
| | - Matus Sykora
- Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia
| | - Tamara Egan Benova
- Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia
| | - Miroslav Barancik
- Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia
| | - Lin Hai Kurahara
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Miki-cho 761-0793, Japan
| | - Narcis Tribulova
- Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovakia
| |
Collapse
|
7
|
Ohlrogge AH, Frost L, Schnabel RB. Harmful Impact of Tobacco Smoking and Alcohol Consumption on the Atrial Myocardium. Cells 2022; 11:2576. [PMID: 36010652 PMCID: PMC9406618 DOI: 10.3390/cells11162576] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/05/2022] [Accepted: 08/11/2022] [Indexed: 11/23/2022] Open
Abstract
Tobacco smoking and alcohol consumption are widespread exposures that are legal and socially accepted in many societies. Both have been widely recognized as important risk factors for diseases in all vital organ systems including cardiovascular diseases, and with clinical manifestations that are associated with atrial dysfunction, so-called atrial cardiomyopathy, especially atrial fibrillation and stroke. The pathogenesis of atrial cardiomyopathy, atrial fibrillation, and stroke in context with smoking and alcohol consumption is complex and multifactorial, involving pathophysiological mechanisms, environmental, and societal aspects. This narrative review summarizes the current literature regarding alterations in the atrial myocardium that is associated with smoking and alcohol.
Collapse
Affiliation(s)
- Amelie H. Ohlrogge
- Department of Cardiology, University Heart and Vascular Centre Hamburg, 20246 Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Lars Frost
- Diagnostic Centre, University Clinic for Development of Innovative Patient Pathways, Silkeborg Regional Hospital, 8600 Silkeborg, Denmark
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Renate B. Schnabel
- Department of Cardiology, University Heart and Vascular Centre Hamburg, 20246 Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| |
Collapse
|
8
|
Association between Alcohol Intake and Arterial Stiffness in Healthy Adults: A Systematic Review. Nutrients 2022; 14:nu14061207. [PMID: 35334865 PMCID: PMC8949071 DOI: 10.3390/nu14061207] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 02/05/2023] Open
Abstract
Background: Arterial stiffness as assessed by Pulse Wave Velocity (PWV) represents an independent predictor of cardiovascular disease. Several dietary compounds and lifestyle factors could influence arterial stiffness. The debate on the significance of the correlation between alcohol consumption and arterial stiffness is still open, given that the relationship is complex and potentially affected by several factors such as alcohol type, consumption levels, gender and age differences. Objective: This systematic literature review aims to examine the evidence supporting an association between alcohol use and PWV, in electronic databases including PubMed/MEDLINE and the Cochrane Library, from January 2010 to November 2020. Screening and full-text reviews were performed by three investigators and data extraction by two. Considering the significant heterogeneity of data only a qualitative analysis (systematic review) was performed. Results: A total of 13 studies met the inclusion criteria. Alcohol consumption was independently associated with arterial stiffness in a J-shaped way in most of the studies included. A benefit of alcohol consumption on arterial stiffness was found in four experimental studies, whilst an unfavorable increasing linear association was found in four others. Associations were confirmed with both oscillometric and tonometric PWV assessment methods. In some studies, a gender and age correlation was found with a more pronounced association in older males. In all studies elevated levels of alcohol consumption were associated with a worsening of arterial stiffness. Conclusions: Despite the variable findings across studies, the current review provides preliminary evidence that light-to-moderate alcohol consumption is associated with arterial stiffness values lower than expected, and evidence that high doses accelerate arterial ageing. These findings could be useful for clinicians who provide recommendations for patients at cardiovascular (CV) risk. Nevertheless, given the heterogeneity of study designs, interventions, measurement methods and statistical evaluations, the protective role of moderate alcohol consumption on arterial stiffness is likely but not certain, warranting additional trials and evidence.
Collapse
|
9
|
Chronic Ethanol Exposure Induces Deleterious Changes in Cardiomyocytes Derived from Human Induced Pluripotent Stem Cells. Stem Cell Rev Rep 2021; 17:2314-2331. [PMID: 34564802 DOI: 10.1007/s12015-021-10267-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2021] [Indexed: 10/20/2022]
Abstract
Chronic alcohol consumption in adults can induce cardiomyopathy, arrhythmias, and heart failure. In newborns, prenatal alcohol exposure can increase the risk of congenital heart diseases. Understanding biological mechanisms involved in the long-term alcohol exposure-induced cardiotoxicity is pivotal to the discovery of therapeutic strategies. In this study, cardiomyocytes derived from human pluripotent stem cells (hiPSC-CMs) were treated with clinically relevant doses of ethanol for various durations up to 5 weeks. The treated cells were characterized for their cellular properties and functions, and global proteomic profiling was conducted to understand the molecular changes associated with long-term ethanol exposure. Increased cell death, oxidative stress, deranged Ca2+ handling, abnormal action potential, altered contractility, and suppressed structure development were observed in ethanol-treated cells. Many dysregulated proteins identified by global proteomic profiling were involved in apoptosis, heart contraction, and extracellular collagen matrix. In addition, several signaling pathways including the Wnt and TGFβ signaling pathways were affected due to long-term ethanol treatment. Therefore, chronic ethanol treatment of hiPSC-CMs induces cardiotoxicity, impairs cardiac functions, and alters protein expression and signaling pathways. This study demonstrates the utility of hiPSC-CMs as a novel model for chronic alcohol exposure study and provides the molecular mechanisms associated with long-term alcohol exposure in human cardiomyocytes.
Collapse
|
10
|
Effects of Boletus Poisoning on Estrogen Receptors and Neurotransmitters in Rats Based on ERk1/2 Pathway. Neural Process Lett 2021. [DOI: 10.1007/s11063-021-10506-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
11
|
Mouton AJ, Flynn ER, Moak SP, Aitken NM, Omoto ACM, Li X, da Silva AA, Wang Z, do Carmo JM, Hall JE. Dimethyl fumarate preserves left ventricular infarct integrity following myocardial infarction via modulation of cardiac macrophage and fibroblast oxidative metabolism. J Mol Cell Cardiol 2021; 158:38-48. [PMID: 34023353 DOI: 10.1016/j.yjmcc.2021.05.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 12/30/2022]
Abstract
Myocardial infarction (MI) is one of the leading causes of mortality and cardiovascular disease worldwide. MI is characterized by a substantial inflammatory response in the infarcted left ventricle (LV), followed by transition of quiescent fibroblasts to active myofibroblasts, which deposit collagen to form the reparative scar. Metabolic shifting between glycolysis and mitochondrial oxidative phosphorylation (OXPHOS) is an important mechanism by which these cell types transition towards reparative phenotypes. Thus, we hypothesized that dimethyl fumarate (DMF), a clinically approved anti-inflammatory agent with metabolic actions, would improve post-MI remodeling via modulation of macrophage and fibroblast metabolism. Adult male C57BL/6J mice were treated with DMF (10 mg/kg) for 3-7 days after MI. DMF attenuated LV infarct and non-infarct wall thinning at 3 and 7 days post-MI, and decreased LV dilation and pulmonary congestion at day 7. DMF improved LV infarct collagen deposition, myofibroblast activation, and angiogenesis at day 7. DMF also decreased pro-inflammatory cytokine expression (Tnf) 3 days after MI, and decreased inflammatory markers in macrophages isolated from the infarcted heart (Hif1a, Il1b). In fibroblasts extracted from the infarcted heart at day 3, RNA-Seq analysis demonstrated that DMF promoted an anti-inflammatory/pro-reparative phenotype. By Seahorse analysis, DMF did not affect glycolysis in either macrophages or fibroblasts at day 3, but enhanced macrophage OXPHOS while impairing fibroblast OXPHOS. Our results indicate that DMF differentially affects macrophage and fibroblast metabolism, and promotes anti-inflammatory/pro-reparative actions. In conclusion, targeting cellular metabolism in the infarcted heart may be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Alan J Mouton
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, United States of America; Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, United States of America.
| | - Elizabeth R Flynn
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, United States of America
| | - Sydney P Moak
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, United States of America
| | - Nikaela M Aitken
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, United States of America
| | - Ana C M Omoto
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, United States of America; Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, United States of America
| | - Xuan Li
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, United States of America; Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, United States of America
| | - Alexandre A da Silva
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, United States of America; Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, United States of America
| | - Zhen Wang
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, United States of America; Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, United States of America
| | - Jussara M do Carmo
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, United States of America; Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, United States of America
| | - John E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, United States of America; Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, United States of America
| |
Collapse
|
12
|
Mouton AJ, Hall JE. Novel roles of immunometabolism and nonmyocyte metabolism in cardiac remodeling and injury. Am J Physiol Regul Integr Comp Physiol 2020; 319:R476-R484. [PMID: 32877243 DOI: 10.1152/ajpregu.00188.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Changes in cardiomyocyte metabolism have been heavily implicated in cardiac injury and heart failure (HF). However, there is emerging evidence that metabolism in nonmyocyte populations, including cardiac fibroblasts, immune cells, and endothelial cells, plays an important role in cardiac remodeling and adaptation to injury. Here, we discuss recent advances and insights into nonmyocyte metabolism in the healthy and injured heart. Metabolic switching from mitochondrial oxidative phosphorylation to glycolysis is critical for immune cell (macrophage and T lymphocyte) and fibroblast phenotypic switching in the inflamed and fibrotic heart. On the other hand, cardiac endothelial cells are heavily reliant on glycolytic metabolism, and thus impairments in glycolytic metabolism underlie endothelial cell dysfunction. Finally, we review current and ongoing metabolic therapies for HF and the potential implications for nonmyocyte metabolism.
Collapse
Affiliation(s)
- Alan J Mouton
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi
| | - John E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|