1
|
Sommerfeld LC, Holmes AP, Yu TY, O'Shea C, Kavanagh DM, Pike JM, Wright T, Syeda F, Aljehani A, Kew T, Cardoso VR, Kabir SN, Hepburn C, Menon PR, Broadway-Stringer S, O'Reilly M, Witten A, Fortmueller L, Lutz S, Kulle A, Gkoutos GV, Pavlovic D, Arlt W, Lavery GG, Steeds R, Gehmlich K, Stoll M, Kirchhof P, Fabritz L. Reduced plakoglobin increases the risk of sodium current defects and atrial conduction abnormalities in response to androgenic anabolic steroid abuse. J Physiol 2024; 602:4409-4436. [PMID: 38345865 DOI: 10.1113/jp284597] [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: 02/28/2023] [Accepted: 01/16/2024] [Indexed: 03/07/2024] Open
Abstract
Androgenic anabolic steroids (AAS) are commonly abused by young men. Male sex and increased AAS levels are associated with earlier and more severe manifestation of common cardiac conditions, such as atrial fibrillation, and rare ones, such as arrhythmogenic right ventricular cardiomyopathy (ARVC). Clinical observations suggest a potential atrial involvement in ARVC. Arrhythmogenic right ventricular cardiomyopathy is caused by desmosomal gene defects, including reduced plakoglobin expression. Here, we analysed clinical records from 146 ARVC patients to identify that ARVC is more common in males than females. Patients with ARVC also had an increased incidence of atrial arrhythmias and P wave changes. To study desmosomal vulnerability and the effects of AAS on the atria, young adult male mice, heterozygously deficient for plakoglobin (Plako+/-), and wild type (WT) littermates were chronically exposed to 5α-dihydrotestosterone (DHT) or placebo. The DHT increased atrial expression of pro-hypertrophic, fibrotic and inflammatory transcripts. In mice with reduced plakoglobin, DHT exaggerated P wave abnormalities, atrial conduction slowing, sodium current depletion, action potential amplitude reduction and the fall in action potential depolarization rate. Super-resolution microscopy revealed a decrease in NaV1.5 membrane clustering in Plako+/- atrial cardiomyocytes after DHT exposure. In summary, AAS combined with plakoglobin deficiency cause pathological atrial electrical remodelling in young male hearts. Male sex is likely to increase the risk of atrial arrhythmia, particularly in those with desmosomal gene variants. This risk is likely to be exaggerated further by AAS use. KEY POINTS: Androgenic male sex hormones, such as testosterone, might increase the risk of atrial fibrillation in patients with arrhythmogenic right ventricular cardiomyopathy (ARVC), which is often caused by desmosomal gene defects (e.g. reduced plakoglobin expression). In this study, we observed a significantly higher proportion of males who had ARVC compared with females, and atrial arrhythmias and P wave changes represented a common observation in advanced ARVC stages. In mice with reduced plakoglobin expression, chronic administration of 5α-dihydrotestosterone led to P wave abnormalities, atrial conduction slowing, sodium current depletion and a decrease in membrane-localized NaV1.5 clusters. 5α-Dihydrotestosterone, therefore, represents a stimulus aggravating the pro-arrhythmic phenotype in carriers of desmosomal mutations and can affect atrial electrical function.
Collapse
Affiliation(s)
- Laura C Sommerfeld
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- University Center of Cardiovascular Science, University Heart and Vascular Center, UKE Hamburg, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Standort Hamburg/Kiel/Lübeck, Germany
| | - Andrew P Holmes
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- School of Biomedical Sciences, Institute of Clinical Sciences, University of Birmingham, Birmingham, UK
| | - Ting Y Yu
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Research and Training Centre in Physical Sciences for Health, Birmingham, UK
| | - Christopher O'Shea
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Research and Training Centre in Physical Sciences for Health, Birmingham, UK
| | - Deirdre M Kavanagh
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK
| | - Jeremy M Pike
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK
| | - Thomas Wright
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Fahima Syeda
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Areej Aljehani
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Tania Kew
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Victor R Cardoso
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - S Nashitha Kabir
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Claire Hepburn
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Priyanka R Menon
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | | | - Molly O'Reilly
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Anika Witten
- Genetic Epidemiology, Institute for Human Genetics, University of Münster, Münster, Germany
- Core Facility Genomics of the Medical Faculty, University of Münster, Münster, Germany
| | - Lisa Fortmueller
- University Center of Cardiovascular Science, University Heart and Vascular Center, UKE Hamburg, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Standort Hamburg/Kiel/Lübeck, Germany
- Genetic Epidemiology, Institute for Human Genetics, University of Münster, Münster, Germany
| | - Susanne Lutz
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
| | - Alexandra Kulle
- Division of Paediatric Endocrinology and Diabetes, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Georgios V Gkoutos
- University Center of Cardiovascular Science, University Heart and Vascular Center, UKE Hamburg, Hamburg, Germany
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Institute of Translational Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- MRC Health Data Research UK (HDR), Midlands Site, UK
| | - Davor Pavlovic
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism (CEDAM), Birmingham Health Partners, Birmingham, UK
- Medical Research Council London Institute of Medical Sciences, London UK & Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London, UK
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism (CEDAM), Birmingham Health Partners, Birmingham, UK
| | - Richard Steeds
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Department of Cardiology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Katja Gehmlich
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Monika Stoll
- Genetic Epidemiology, Institute for Human Genetics, University of Münster, Münster, Germany
- Core Facility Genomics of the Medical Faculty, University of Münster, Münster, Germany
- Cardiovascular Research Institute Maastricht, Department of Biochemistry, Maastricht University, Maastricht, The Netherlands
| | - Paulus Kirchhof
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- German Center for Cardiovascular Research (DZHK), Standort Hamburg/Kiel/Lübeck, Germany
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Larissa Fabritz
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- University Center of Cardiovascular Science, University Heart and Vascular Center, UKE Hamburg, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Standort Hamburg/Kiel/Lübeck, Germany
- Department of Cardiology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
2
|
Lv Z, Chen X, Chen P, Li Q, Guo Z, Lu Q, Ding S. Colchicine prevents ventricular arrhythmias vulnerability in diet-induced obesity rats. Biochem Biophys Res Commun 2022; 610:127-132. [PMID: 35462093 DOI: 10.1016/j.bbrc.2022.03.114] [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: 03/16/2022] [Accepted: 03/22/2022] [Indexed: 11/02/2022]
Abstract
AIMS This study aimed to assess the role of colchicine in ventricular arrhythmias (VAs) induced by high fat diet (HFD)-fed rats. METHODS AND RESULTS Male rats were divided into four groups: CTL, normal diet plus saline; CTC, normal diet plus colchicine; HFD, HFD plus saline; HFC: HFD plus colchicine. Metabolic parameters, ECG parameters, ventricular electrophysiological parameters, ventricular histology, Western blot and RT-qPCR were measured. Compared with the HFD group, colchicine treatment significantly improved metabolic parameters, reduced ventricular fibrosis, increased the expression of Cav1.2, Kv4.2, Nav1.5, and Cx43, reduced CaMKII, p-CaMKII, p-RyR2 (S2808), and p-RyR2 (S2814) expression in LV. Furthermore, colchicine inhibited the inflammatory responses, prolonged ventricular effective refractory period (ERP), reduced corrected QT interval (QTc) and Tpeak-Tend interval, so as to reduce the susceptibility to VAs in obesity rats. CONCLUSIONS Colchicine could mitigate ventricular fibrosis, ventricular electrical remodeling, as well as the expression of ion channels, and inhibit obesity-induced inflammatory responses, which provides a new idea for colchicine to prevent VAs in obese individuals.
Collapse
Affiliation(s)
- Zhiyang Lv
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, PR China; Institute of Cardiovascular Diseases, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, PR China
| | - Xiaodi Chen
- Department of Ultrasound, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, PR China
| | - Ping Chen
- National Standardized Training Base for Resident, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, PR China
| | - Qianyuan Li
- Clinical Laboratory, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, PR China
| | - Zhuli Guo
- Institute of Cardiovascular Diseases, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, PR China
| | - Qing Lu
- Department of Cardiology, General Hospital of Central Theater Command, Wuhan Clinical Medicine College of Southern Medical University, Wuhan, 430070, PR China
| | - Shifang Ding
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, PR China; Department of Cardiology, General Hospital of Central Theater Command, Wuhan Clinical Medicine College of Southern Medical University, Wuhan, 430070, PR China.
| |
Collapse
|
3
|
Gagnon LR, Sadasivan C, Perera K, Oudit GY. Cardiac Complications of Common Drugs of Abuse: Pharmacology, Toxicology and Management. Can J Cardiol 2021; 38:1331-1341. [PMID: 34737034 DOI: 10.1016/j.cjca.2021.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 12/23/2022] Open
Abstract
Cardiovascular complications from drugs of abuse are becoming more apparent due to increased usage worldwide. Substance abuse can cause both acute and chronic cardiovascular complications and is increasing in prevalence especially in young adults. These substances contribute to the development of acute coronary syndrome, type II myocardial injury, arrhythmias, cardiomyopathies and have numerous other cardiovascular complications. Although no screening guidelines exist, clinical awareness of these potential complications and their prevention, clinical presentation, diagnosis, and treatment are critically important. Management of cardiovascular disease should be coupled with appropriate social and mental health interventions to provide sustained clinical benefit. The higher the number of substances used recreationally, the greater the risk of premature heart disease. Epidemiological studies showed that 1 in 5 young adults misuse several substances and often start using at younger ages with a greater risk for adverse health outcomes over the long-term. The aim of this review is to highlight the basic epidemiology, cardiac complications, and disease-specific treatment options of commonly abused substances including methamphetamine, cocaine, alcohol, anabolic-androgenic steroids, cannabis, and tobacco.
Collapse
Affiliation(s)
- Luke R Gagnon
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Chandu Sadasivan
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Kevin Perera
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Gavin Y Oudit
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
4
|
Severe Cardiac and Metabolic Pathology Induced by Steroid Abuse in a Young Individual. Diagnostics (Basel) 2021; 11:diagnostics11081313. [PMID: 34441248 PMCID: PMC8394374 DOI: 10.3390/diagnostics11081313] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 11/17/2022] Open
Abstract
Androgenic-Anabolic Steroids (AAS) abuse is known to play an important role in causing the systemic inflammatory response and multiple-organ dysfunction in healthy individuals. Although many of the undesirable effects of steroid abuse have been reported, at present, little is known about the effect of anabolic supplements and the correlation between cardiac and metabolic pathology. This paper presents a case of a 25 year old patient with a complex medical history after 6 months of steroid administration. Myocardial infraction, dyslipidemia, obesity, hyperuricemia, secondary diabetes, and chronic renal disease were identified after clinical and para-clinical examinations. The particularities of this case were interpreted in the context of a literature review, highlighting the effect of multi-organ damage as a result of the uncontrolled use of anabolic steroid supplements.
Collapse
|
5
|
Ronchi SN, Mass EMSW, Bernardina NRD, de Melo Júnior AF, Dos Santos WC, de Andrade TU, Brasil GA, Bissoli NS. Low and high doses of oxandrolone promote pathological cardiac remodeling in young male rats. Steroids 2021; 170:108814. [PMID: 33727119 DOI: 10.1016/j.steroids.2021.108814] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 02/18/2021] [Accepted: 02/24/2021] [Indexed: 01/09/2023]
Abstract
Oxandrolone (OXA) used in clinical practice, however, its misuse is frequent, including by adolescents pursuing an aesthetic goal. However, the impacts of noxious doses on the cardiovascular system remain unknown. AIM To investigate cardiac effects of OXA in low (LD) and high (HD) doses. METHODS Male Wistar prepubescent rats were separated into 3 experimental groups: control (CON), LD, and HD. Only the CON group received the carrier (carboxymethylcellulose, 0.5%), while the LD and HD groups received, respectively, 2.5 and 37.5 mg/kg/day of OXA via gavage for 4 weeks. The hemodynamic parameters (+dP/dtmax, -dP/dtmin, and Tau) and cardiac autonomic tonus were assessed. Hearts were retrieved for histological analyses and oxidative stress evaluation. Expression levels of calcium-handling proteins were measured by western blot. RESULTS The OXA treatment changed neither the cardiac contractility nor the cardiac autonomic tonus. However, cardiac hypertrophy, collagen deposition, and increased angiotensin-converting enzyme (ACE) expression were observed in a dose-dependent way. Also, the p-phospholamban (p-PLB)/PLB ratio was observed to decrease and increase, respectively, in the LD and HD groups; the sarcoplasmic/endoplasmic reticulum calcium ATPase 2a (SERCA2a)/PLB ratio being higher in both groups. OXA increased SOD1 expression and decreased catalase expression only in the LD group, and protein oxidation was increased in HD. CONCLUSION Both doses of OXA could promote pathological cardiac remodeling, probably via increased ACE, and these effects were exacerbated in the HD treatment, but cardiac contractility was not affected regardless of the dose.
Collapse
Affiliation(s)
- Silas Nascimento Ronchi
- Department of Physiological Sciences, Federal University of Espírito Santo, Vitória, ES, Brazil
| | | | | | | | | | | | | | - Nazaré Souza Bissoli
- Department of Physiological Sciences, Federal University of Espírito Santo, Vitória, ES, Brazil
| |
Collapse
|
6
|
Adverse Effects of Anabolic-Androgenic Steroids: A Literature Review. Healthcare (Basel) 2021; 9:healthcare9010097. [PMID: 33477800 PMCID: PMC7832337 DOI: 10.3390/healthcare9010097] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/03/2021] [Accepted: 01/11/2021] [Indexed: 12/19/2022] Open
Abstract
Anabolic-androgenic steroids (AASs) are a large group of molecules including endogenously produced androgens, such as testosterone, as well as synthetically manufactured derivatives. AAS use is widespread due to their ability to improve muscle growth for aesthetic purposes and athletes’ performance, minimizing androgenic effects. AAS use is very popular and 1–3% of US inhabitants have been estimated to be AAS users. However, AASs have side effects, involving all organs, tissues and body functions, especially long-term toxicity involving the cardiovascular system and the reproductive system, thereby, their abuse is considered a public health issue. The aim of the proposed review is to highlight the most recent evidence regarding the mechanisms of action of AASs and their unwanted effects on organs and lifestyle, as well as suggesting that AAS misuse and abuse lead to adverse effects in all body tissues and organs. Oxidative stress, apoptosis, and protein synthesis alteration are common mechanisms involved in AAS-related damage in the whole body. The cardiovascular system and the reproductive system are the most frequently involved apparatuses. Epidemiology as well as the molecular and pathological mechanisms involved in the neuropsychiatric side-effects of AAS abuse are still unclear, further research is needed in this field. In addition, diagnostically reliable tests for AAS abuse should be standardized. In this regard, to prevent the use of AASs, public health measures in all settings are crucial. These measures consist of improved knowledge among healthcare workers, proper doping screening tests, educational interventions, and updated legislation.
Collapse
|
7
|
Nandrolone Decanoate: Use, Abuse and Side Effects. ACTA ACUST UNITED AC 2020; 56:medicina56110606. [PMID: 33187340 PMCID: PMC7696474 DOI: 10.3390/medicina56110606] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/28/2020] [Accepted: 11/09/2020] [Indexed: 12/13/2022]
Abstract
Background and Objectives: Androgens play a significant role in the development of male reproductive organs. The clinical use of synthetic testosterone derivatives, such as nandrolone, is focused on maximizing the anabolic effects and minimizing the androgenic ones. Class II anabolic androgenic steroids (AAS), including nandrolone, are rapidly becoming a widespread group of drugs used both clinically and illicitly. The illicit use of AAS is diffused among adolescent and bodybuilders because of their anabolic proprieties and their capacity to increase tolerance to exercise. This systematic review aims to focus on side effects related to illicit AAS abuse, evaluating the scientific literature in order to underline the most frequent side effects on AAS abusers’ bodies. Materials and Methods: A systematic review of the scientific literature was performed using the PubMed database and the keywords “nandrolone decanoate”. The inclusion criteria for articles or abstracts were English language and the presence of the following words: “abuse” or “adverse effects”. After applying the exclusion and inclusion criteria, from a total of 766 articles, only 148 were considered eligible for the study. Results: The most reported adverse effects (found in more than 5% of the studies) were endocrine effects (18 studies, 42%), such as virilization, gynecomastia, hormonal disorders, dyslipidemia, genital alterations, and infertility; cardiovascular dysfunctions (six studies, 14%) such as vascular damage, coagulation disorders, and arteriosus hypertension; skin disorders (five studies, 12%) such as pricking, acne, and skin spots; psychiatric and mood disorders (four studies, 9%) such as aggressiveness, sleep disorders and anxiety; musculoskeletal disorders (two studies, 5%), excretory disorders (two studies, 5%), and gastrointestinal disorders (two studies, 5%). Conclusions: Based on the result of our study, the most common adverse effects secondary to the abuse of nandrolone decanoate (ND) involve the endocrine, cardiovascular, skin, and psychiatric systems. These data could prove useful to healthcare professionals in both sports and clinical settings.
Collapse
|
8
|
Torrisi M, Pennisi G, Russo I, Amico F, Esposito M, Liberto A, Cocimano G, Salerno M, Li Rosi G, Di Nunno N, Montana A. Sudden Cardiac Death in Anabolic-Androgenic Steroid Users: A Literature Review. ACTA ACUST UNITED AC 2020; 56:medicina56110587. [PMID: 33158202 PMCID: PMC7694262 DOI: 10.3390/medicina56110587] [Citation(s) in RCA: 40] [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: 09/30/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023]
Abstract
Background and objectives: Anabolic-androgenic steroids (AASs) are a group of synthetic molecules derived from testosterone and its related precursors. AASs are widely used illicitly by adolescents and athletes, especially by bodybuilders, both for aesthetic uses and as performance enhancers to increase muscle growth and lean body mass. When used illicitly they can damage health and cause disorders affecting several functions. Sudden cardiac death (SCD) is the most common medical cause of death in athletes. SCD in athletes has also been associated with the use of performance-enhancing drugs. This review aimed to focus on deaths related to AAS abuse to investigate the cardiac pathophysiological mechanism that underlies this type of death, which still needs to be fully investigated. Materials and Methods: This review was conducted using PubMed Central and Google Scholar databases, until 21 July 2020, using the following key terms: “((Sudden cardiac death) OR (Sudden death)) AND ((androgenic anabolic steroid) OR (androgenic anabolic steroids) OR (anabolic-androgenic steroids) OR (anabolic-androgenic steroid))”. Thirteen articles met the inclusion and exclusion criteria, for a total of 33 reported cases. Results: Of the 33 cases, 31 (93.9%) were males while only 2 (61%) were females. Mean age was 29.79 and, among sportsmen, the most represented sports activity was bodybuilding. In all cases there was a history of AAS abuse or a physical phenotype suggesting AAS use; the total usage period was unspecified in most cases. In 24 cases the results of the toxicological analysis were reported. The most detected AASs were nandrolone, testosterone, and stanozolol. The most frequently reported macroscopic alterations were cardiomegaly and left ventricular hypertrophy, while the histological alterations were foci of fibrosis and necrosis of the myocardial tissue. Conclusions: Four principal mechanisms responsible for SCD have been proposed in AAS abusers: the atherogenic model, the thrombosis model, the model of vasospasm induced by the release of nitric oxide, and the direct myocardial injury model. Hypertrophy, fibrosis, and necrosis represent a substrate for arrhythmias, especially when combined with exercise. Indeed, AAS use has been shown to change physiological cardiac remodeling of athletes to pathophysiological cardiac hypertrophy with an increased risk of life-threatening arrhythmias.
Collapse
Affiliation(s)
- Marco Torrisi
- Legal Medicine, Department of Medical, Surgical and Advanced Technologies, “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (M.T.); (G.P.); (I.R.); (F.A.); (M.E.); (A.L.); (G.C.); (M.S.)
| | - Giuliana Pennisi
- Legal Medicine, Department of Medical, Surgical and Advanced Technologies, “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (M.T.); (G.P.); (I.R.); (F.A.); (M.E.); (A.L.); (G.C.); (M.S.)
| | - Ilenia Russo
- Legal Medicine, Department of Medical, Surgical and Advanced Technologies, “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (M.T.); (G.P.); (I.R.); (F.A.); (M.E.); (A.L.); (G.C.); (M.S.)
| | - Francesco Amico
- Legal Medicine, Department of Medical, Surgical and Advanced Technologies, “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (M.T.); (G.P.); (I.R.); (F.A.); (M.E.); (A.L.); (G.C.); (M.S.)
| | - Massimiliano Esposito
- Legal Medicine, Department of Medical, Surgical and Advanced Technologies, “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (M.T.); (G.P.); (I.R.); (F.A.); (M.E.); (A.L.); (G.C.); (M.S.)
| | - Aldo Liberto
- Legal Medicine, Department of Medical, Surgical and Advanced Technologies, “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (M.T.); (G.P.); (I.R.); (F.A.); (M.E.); (A.L.); (G.C.); (M.S.)
| | - Giuseppe Cocimano
- Legal Medicine, Department of Medical, Surgical and Advanced Technologies, “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (M.T.); (G.P.); (I.R.); (F.A.); (M.E.); (A.L.); (G.C.); (M.S.)
| | - Monica Salerno
- Legal Medicine, Department of Medical, Surgical and Advanced Technologies, “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (M.T.); (G.P.); (I.R.); (F.A.); (M.E.); (A.L.); (G.C.); (M.S.)
| | - Giuseppe Li Rosi
- Department of Law, Criminology, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy;
| | - Nunzio Di Nunno
- Department of History, Society and Studies on Humanity, University of Salento, 73100 Lecce, Italy;
| | - Angelo Montana
- Legal Medicine, Department of Medical, Surgical and Advanced Technologies, “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (M.T.); (G.P.); (I.R.); (F.A.); (M.E.); (A.L.); (G.C.); (M.S.)
- Correspondence: ; Tel.: +39-3287655428
| |
Collapse
|
9
|
Seara FAC, Pereira-Junior PP, Silva-Almeida C, Dos-Santos RC, Souza RN, Costa CRM, Domingos AE, Barbosa RAQ, Ferraz AP, Machado AAN, Ceccato VM, Takiya CM, Ponte CG, Reis LC, Olivares EL, Nascimento JHM. Anabolic steroid excess promotes hydroelectrolytic and autonomic imbalance in adult male rats: Is it enough to alter blood pressure? Steroids 2020; 163:108711. [PMID: 32739291 DOI: 10.1016/j.steroids.2020.108711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/20/2020] [Accepted: 07/27/2020] [Indexed: 11/29/2022]
Abstract
AIM The present study investigated the effects of anabolic steroid (AS) excess on blood pressure regulation. METHODS Male Wistar rats were treated with nandrolone decanoate (AS) or vehicle (CTL) for 8 or 10 weeks. Saline (1.8%) and water intake were measured in metabolic cages. Urinary volume, osmolarity, Na+ and K+ concentrations, and plasma osmolarity were measured. The autonomic balance was estimated by heart rate variability at baseline or after icv injection of losartan. Cardiac function was assessed by echocardiography and ex vivo recordings. Myocardial collagen deposition was evaluated by Picrosirius-Red staining. Vascular reactivity and wall thickness were investigated in aortic sections. Blood pressure (BP) was assessed by tail-cuff plethysmography. Angiotensin II type I receptor (AT1R), renin, and mineralocorticoid receptor (MR) mRNA expression was measured in the kidneys and whole hypothalamus. RESULTS AS group exhibited decreased urinary volume and Na+ concentration, while urinary K+ concentration, plasma osmolarity, and renal AT1R and renin mRNA levels were increased compared to CTL (p < 0.05). Water intake was increased, and saline intake was decreased in the AS group (p < 0.01). AS group exhibited increased low-frequency/high-frequency-ratio, while it was decreased by icv injection of losartan (p < 0.05) compared to baseline. Neither cardiac function nor vascular reactivity/morphology was affected by AS excess (p > 0.05). Ultimately, BP levels were not altered by AS excess (p > 0.05). CONCLUSION AS excess promoted hydroelectrolytic and autonomic imbalance but did not alter vascular or cardiac function/morphology.
Collapse
Affiliation(s)
- Fernando A C Seara
- Department of Physiological Sciences, Institute of Biological and Health Sciences, Federal Rural University of Rio de Janeiro, Brazil; Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Brazil.
| | - Pedro P Pereira-Junior
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Brazil
| | - Claudio Silva-Almeida
- Department of Physiological Sciences, Institute of Biological and Health Sciences, Federal Rural University of Rio de Janeiro, Brazil; Estácio de Sá University, Brazil
| | - Raoni C Dos-Santos
- Department of Physiological Sciences, Institute of Biological and Health Sciences, Federal Rural University of Rio de Janeiro, Brazil
| | - Raquel N Souza
- Department of Physiological Sciences, Institute of Biological and Health Sciences, Federal Rural University of Rio de Janeiro, Brazil
| | - César R M Costa
- Department of Physiological Sciences, Institute of Biological and Health Sciences, Federal Rural University of Rio de Janeiro, Brazil; Multicenter Postgraduate Program of Physiological Sciences, Brazilian Society of Physiology, Brazil
| | - Ainá E Domingos
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Brazil
| | - Raiana A Q Barbosa
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Brazil
| | - Ana P Ferraz
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Brazil
| | - André A N Machado
- Laboratory of Biochemistry and Gene Expression, University of Ceará, Brazil
| | - Vânia M Ceccato
- Laboratory of Biochemistry and Gene Expression, University of Ceará, Brazil
| | - Christina M Takiya
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Brazil
| | - Cristiano G Ponte
- Federal Institute of Education, Science, and Technology of Rio de Janeiro, Brazil
| | - Luis C Reis
- Department of Physiological Sciences, Institute of Biological and Health Sciences, Federal Rural University of Rio de Janeiro, Brazil; Multicenter Postgraduate Program of Physiological Sciences, Brazilian Society of Physiology, Brazil
| | - Emerson L Olivares
- Department of Physiological Sciences, Institute of Biological and Health Sciences, Federal Rural University of Rio de Janeiro, Brazil; Multicenter Postgraduate Program of Physiological Sciences, Brazilian Society of Physiology, Brazil
| | - Jose H M Nascimento
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Brazil
| |
Collapse
|
10
|
Arteyeva NV. Dispersion of ventricular repolarization: Temporal and spatial. World J Cardiol 2020; 12:437-449. [PMID: 33014291 PMCID: PMC7509993 DOI: 10.4330/wjc.v12.i9.437] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/11/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023] Open
Abstract
Repolarization heterogeneity (RH) is an intrinsic property of ventricular myocardium and the reason for T-wave formation on electrocardiogram (ECG). Exceeding the physiologically based RH level is associated with appearance of life-threatening ventricular arrhythmias and sudden cardiac death. In this regard, an accurate and comprehensive evaluation of the degree of RH parameters is of importance for assessment of heart state and arrhythmic risk. This review is devoted to comprehensive consideration of RH phenomena in terms of electrophysiological processes underlying RH, cardiac electric field formation during ventricular repolarization, as well as clinical significance of RH and its reflection on ECG parameters. The formation of transmural, apicobasal, left-to-right and anterior-posterior gradients of action potential durations and end of repolarization times resulting from the heterogenous distribution of repolarizing ion currents and action potential morphology throughout the heart ventricles, and the different sensitivity of myocardial cells in different ventricular regions to the action of pharmacological agents, temperature, frequency of stimulation, etc., are being discussed. The review is focused on the fact that RH has different aspects – temporal and spatial, global and local; ECG reflection of various RH aspects and their clinical significance are being discussed. Strategies for comprehensive assessment of ventricular RH using different ECG indices reflecting various RH aspects are presented.
Collapse
Affiliation(s)
- Natalia V Arteyeva
- Laboratory of Cardiac Physiology, Institute of Physiology of Komi Science Centre of the Ural Branch of the Russian Academy of Sciences, Syktyvkar 167982, Russia
| |
Collapse
|
11
|
Seara FAC, Olivares EL, Nascimento JHM. Anabolic steroid excess and myocardial infarction: From ischemia to reperfusion injury. Steroids 2020; 161:108660. [PMID: 32492466 DOI: 10.1016/j.steroids.2020.108660] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/05/2020] [Accepted: 05/17/2020] [Indexed: 01/06/2023]
Abstract
Anabolic steroids (AS) are synthetic testosterone-derivatives developed by the pharmaceutical industry to mimic testosterone biological effects. So far, AS have been implicated in the treatment of pathological conditions, such as hypogonadism, anemia, and cachexia. Since their discovery, though, AS have been illicitly used by elite and recreational athletes, bodybuilders and weightlifters in order to enhance athletic and aesthetic performance. This practice is characterized by cycles of administration and withdrawal, the combination of different AS compounds, and administration of doses 50 - 1000 times higher than those recommended for therapeutic purposes. AS excess has been correlated to cardiovascular detrimental effects, including cardiac hypertrophy, arrhythmias, and hypertension. Particularly, acute myocardial infarction (AMI) has been extensively reported by clinical and post-mortem studies. Atherosclerosis, hypercoagulability state, increased thrombogenesis and vasospasm have arisen as potential causes of myocardial ischemia in AS users. Additionally, several experimental reports have demonstrated that AS can increase the susceptibility to cardiac ischemia/reperfusion injury, whereas the cardioprotection elicited by physical exercise and ischemic postconditioning is blunted. Altogether, these factors can contribute to increased AMI morbidity and mortality during AS excess, particularly when AS are combined with other compounds, such as thyroid hormones, growth hormones, insulin, and diuretics.
Collapse
Affiliation(s)
- Fernando A C Seara
- Laboratory of Cardiovascular Physiology and Pharmacology, Department of Physiological Sciences, Federal Rural University of Rio de Janeiro, Seropédica, RJ, Brazil; Multicenter Graduate Program in Physiological Sciences, Department of Physiological Sciences, Institute of Biological and Health Sciences, Federal Rural University of Rio de Janeiro, Seropédica, Brazil; Laboratory of Cardiac Electrophysiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Emerson L Olivares
- Laboratory of Cardiovascular Physiology and Pharmacology, Department of Physiological Sciences, Federal Rural University of Rio de Janeiro, Seropédica, RJ, Brazil; Multicenter Graduate Program in Physiological Sciences, Department of Physiological Sciences, Institute of Biological and Health Sciences, Federal Rural University of Rio de Janeiro, Seropédica, Brazil
| | - Jose H M Nascimento
- Laboratory of Cardiac Electrophysiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
12
|
Yang HJ, Kong B, Shuai W, Zhang JJ, Huang H. Shensong Yangxin Protects Against Metabolic Syndrome-Induced Ventricular Arrhythmias by Inhibiting Electrical Remodeling. Front Pharmacol 2020; 11:993. [PMID: 32733242 PMCID: PMC7363804 DOI: 10.3389/fphar.2020.00993] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 06/19/2020] [Indexed: 12/19/2022] Open
Abstract
Shensong Yangxin (SSYX) is a traditional Chinese medicine, which has been proven to improve the clinical symptoms of arrhythmia. However, the role of SSYX in metabolic syndrome (MetS)-induced electrical remodeling remains to be fully elucidated. Here, we sought to clarify whether SSYX can alter the electrophysiological remodeling of cardiac myocytes from MetS rats by regulating transient outward potassium current (Ito) and calcium current (ICa-L). Male Wistar rats were subjected to 16 weeks of high-carbohydrate, high-fat to produce a MetS model group. SSYX (0.4 g/kg) was administrated by daily gavage 8 weeks following high-carbohydrate, high-fat for 8 weeks. In vivo electrophysiological study was performed to evaluated ventricular arrhythmias (VA) vulnerability and electrophysiological properties. The potential electrical mechanisms were estimated by whole-cell patch-clamp and molecular analysis. The H9C2 cells were used to verify the protective role of SSYX in vitro. After 16-week high-carbohydrate, high-fat feeding, MetS model rats showed increased body weight (BW), blood pressure (BP), blood sugar (BS), heart rate (HR) and heart weights to tibia length (HW/TL) ratio. Furthermore, MetS rats depicted increased VA inducibility, shortened effective refractory period (ERP) and prolonged action potential duration (APD). Lower ICa-L and Ito current densities were observed in MetS rats than CTL rats. Additionally, MetS rats exhibited significantly increased cardiac fibrosis, decreased Cx43 expression and protein levels of Cav1.2, Kv4.2, Kv4.3 than CTL group. As expected, these MetS-induced effects above were reversed when SSYX was administrated. Mechanistically, SSYX administrated significantly down-regulated the TLR4/MyD88/CaMKII signaling pathway both in vivo and in vitro. Collectively, our data indicated that the electrical remodeling induced by MetS contributed to the increased VA susceptibility. SSYX protects against MetS-induced VA by inhibiting electrical remodeling through TLR4/MyD88/CaMKII signaling pathway.
Collapse
Affiliation(s)
- Hong-Jie Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Bin Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wei Shuai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jing-Jing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
13
|
Barber M, Nguyen LS, Wassermann J, Spano JP, Funck-Brentano C, Salem JE. Cardiac arrhythmia considerations of hormone cancer therapies. Cardiovasc Res 2020; 115:878-894. [PMID: 30698686 DOI: 10.1093/cvr/cvz020] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/14/2018] [Accepted: 01/24/2019] [Indexed: 12/28/2022] Open
Abstract
Breast and prostate cancers are among the most prevalent cancers worldwide. Oestradiol and progesterone are major drivers for breast cancer proliferation, and androgens for prostate cancer. Endocrine therapies are drugs that interfere with hormone-activated pathways to slow cancer progression. Multiple new breakthrough drugs improving overall survival have recently been developed within this class. As the use of these latter drugs grows, incidence of cardiac arrhythmias has emerged as an unappreciated complication. These changes are not surprising given that sex hormones alter ventricular repolarization. Testosterone shortens action potential duration and QT interval duration, while oestradiol has an opposite effect. In patients with breast cancer, selective oestrogen receptor modulators are associated with more reports for long QT and torsade de pointes (TdP) than aromatase inhibitors, likely through an oestradiol-like effect on the heart. Cyclin-dependent kinase 4/6 inhibitors, a new class of anticancer drugs used in combination with endocrine therapies in hormone receptor positive breast cancer, are also variably associated with drug-induced long QT, particularly with ribociclib. In prostate cancer, androgen deprivation therapy is associated with long QT and TdP, and possibly atrial fibrillation for abiraterone. In this review, we have summarized the clinical and preclinical data focusing on cardiac arrhythmia considerations of hormone cancer therapies.
Collapse
Affiliation(s)
- Mary Barber
- Department of Medicine and Clinical Pharmacology, Cardio-Oncology Program, Vanderbilt University Medical Center, 1211 Medical Center Dr, Nashville, TN, USA
| | - Lee S Nguyen
- Department of Pharmacology, Sorbonne Université, INSERM CIC Paris-Est, AP-HP, ICAN, Pitié-Salpêtrière Hospital, Paris F-75013, France
| | - Johanna Wassermann
- Department of Oncology, Sorbonne Université, AP-HP, Pitié-Salpêtrière Hospital, Paris F-75013, France
| | - Jean-Philippe Spano
- Department of Oncology, Sorbonne Université, AP-HP, Pitié-Salpêtrière Hospital, Paris F-75013, France
| | - Christian Funck-Brentano
- Department of Pharmacology, Sorbonne Université, INSERM CIC Paris-Est, AP-HP, ICAN, Pitié-Salpêtrière Hospital, Paris F-75013, France
| | - Joe-Elie Salem
- Department of Medicine and Clinical Pharmacology, Cardio-Oncology Program, Vanderbilt University Medical Center, 1211 Medical Center Dr, Nashville, TN, USA.,Department of Pharmacology, Sorbonne Université, INSERM CIC Paris-Est, AP-HP, ICAN, Pitié-Salpêtrière Hospital, Paris F-75013, France
| |
Collapse
|
14
|
Shuai W, Kong B, Fu H, Jiang X, Huang H. The effect of MD1 on potassium and L-type calcium current of cardiomyocytes from high-fat diet mice. Channels (Austin) 2020; 14:181-189. [PMID: 32491968 PMCID: PMC7515570 DOI: 10.1080/19336950.2020.1772628] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Myeloid differentiation protein 1 (MD1) is exerted an anti-arrhythmic effect in obese mice. Therefore, we sought to clarify whether MD1 can alter the electrophysiological remodeling of cardiac myocytes from obese mice by regulating voltage-gated potassium current and calcium current. MD1 knock-out (KO) and wild type (WT) mice were given a high-fat diet (HFD) for 20 weeks, starting at the age of 6 weeks. The potential electrophysiological mechanisms were estimated by whole-cell patch-clamp and molecular analysis. After 20-week HFD feeding, action potential duration (APD) from left ventricular myocytes of MD1-KO mice revealed APD20, APD50, and APD90 were profoundly enlarged. Furthermore, HFD mice showed a decrease in the fast transient outward potassium currents (Ito,f), slowly inactivating potassium current (IK, slow), and inward rectifier potassium current (IK1). Besides, HFD-fed mice showed that the current density of ICaL was significantly lower, and the haft inactivation voltage was markedly shifted right. These HFD induced above adverse effects were further exacerbated in KO mice. The mRNA expression of potassium ion channels (Kv4.2, Kv4.3, Kv2.1, Kv1.5, and Kir2.1) and calcium ion channel (Cav1.2) was markedly decreased in MD1-KO HFD-fed mice. MD1 deletion led to down-regulated potassium currents and slowed inactivation of L-type calcium channel in an obese mice model.
Collapse
Affiliation(s)
- Wei Shuai
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan, Hubei, China.,Cardiovascular Research Institute of Wuhan University , Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology , Wuhan, Hubei, China
| | - Bin Kong
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan, Hubei, China.,Cardiovascular Research Institute of Wuhan University , Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology , Wuhan, Hubei, China
| | - Hui Fu
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan, Hubei, China.,Cardiovascular Research Institute of Wuhan University , Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology , Wuhan, Hubei, China
| | - Xiaobo Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan, Hubei, China.,Cardiovascular Research Institute of Wuhan University , Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology , Wuhan, Hubei, China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan, Hubei, China.,Cardiovascular Research Institute of Wuhan University , Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology , Wuhan, Hubei, China
| |
Collapse
|
15
|
Fattoretti P, Malatesta M, Mariotti R, Zancanaro C. Testosterone administration increases synaptic density in the gyrus dentatus of old mice independently of physical exercise. Exp Gerontol 2019; 125:110664. [DOI: 10.1016/j.exger.2019.110664] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 05/17/2019] [Accepted: 07/15/2019] [Indexed: 10/26/2022]
|
16
|
Abstract
OBJECTIVE Anabolic-androgenic steroids (AAS) represents a group of synthetic testosterone derivatives that play an important role in clinical treatment. These drugs are widely abused among the general public to increase lean weight and improve athletic performance. It has been reported that AAS use can produce many adverse effects, especially the occurrence of cardiovascular risk. Although there are many related studies, there has been no consensus on AAS use and cardiovascular risk. The present study was to review the effect of AAS on the cardiovascular system. DATA SOURCES The data in this review were obtained from articles included in PubMed and the National Center for Biotechnology Information database. STUDY SELECTION Original articles, case reports, and systematic reviews about AAS were selected for the article. RESULTS The use/abuse of AAS is correlated with higher cardiovascular risks, and many AAS users/abusers had cardiovascular diseases. However, there are many confounding factors in the studies that explored the causality between AAS intake and disease development, and additional studies are required to determine AAS toxicity. CONCLUSION AAS produces toxic effects on the cardiovascular system, and it is necessary to ensure that more people know this about AAS, including medical personnel.
Collapse
Affiliation(s)
- Jian-Di Liu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | | |
Collapse
|
17
|
Marocolo M, Katayama PL, Meireles A, Barbosa Neto O. Combined effects of exercise training and high doses of anabolic steroids on cardiac autonomic modulation and ventricular repolarization properties in rats. Can J Physiol Pharmacol 2019; 97:1185-1192. [PMID: 31505126 DOI: 10.1139/cjpp-2019-0286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Several studies have reported that high doses of synthetic anabolic androgenic steroids (AAS) can have serious negative effects on health, including the cardiovascular system. The aim of this study was to evaluate the combined effects of AAS and exercise training on ventricular repolarization and cardiac autonomic modulation in rats. Male Wistar rats were allocated into 4 groups: sedentary rats treated with vehicle, sedentary rats treated with nandrolone decanoate, swimming-trained rats treated with vehicle, and swimming-trained rats treated with nandrolone decanoate. Ventricular repolarization was evaluated by electrocardiographic analysis of QT interval and QT dispersion. Cardiac autonomic modulation was assessed by heart rate variability. Our results show that AAS increased QT interval and QT dispersion in sedentary rats treated with nandrolone decanoate as compared to sedentary rats treated with vehicle, indicating AAS-induced ventricular repolarization abnormalities. When rats treated with nandrolone decanoate were subjected to concomitant exercise training, ventricular repolarization was normalized. On the other hand, AAS-induced reduction in cardiac parasympathetic modulation was not prevented by exercise training. In conclusion, AAS produced cardiac autonomic dysfunction and ventricular repolarization disturbances in rats. Combining an exercise training protocol during the AAS treatment attenuated the ventricular repolarization abnormalities and did not prevent cardiac autonomic dysfunction.
Collapse
Affiliation(s)
- Moacir Marocolo
- Physiology and Human Performance Research Group, Department of Physiology, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil
| | - Pedro L Katayama
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University, Araraquara, SP, Brazil
| | - Anderson Meireles
- Physiology and Human Performance Research Group, Department of Physiology, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil
| | - Octávio Barbosa Neto
- Sport Sciences Department, Federal University of Triangulo Mineiro, Uberaba, MG, Brazil
| |
Collapse
|
18
|
Shuai W, Kong B, Fu H, Shen C, Huang H. Loss of MD1 increases vulnerability to ventricular arrhythmia in diet-induced obesity mice via enhanced activation of the TLR4/MyD88/CaMKII signaling pathway. Nutr Metab Cardiovasc Dis 2019; 29:991-998. [PMID: 31353205 DOI: 10.1016/j.numecd.2019.06.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 05/09/2019] [Accepted: 06/10/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIM Obesity is an important risk factor for ventricular arrhythmia (VA), and myeloid differentiation protein 1 (MD1) has been reported to decrease in obese hearts. Nevertheless, underlying mechanisms linking MD1 and VA have not been fully studied. This study aims to investigate the regulatory role of MD1 in VA caused by diet-induced obesity. METHODS AND RESULTS MD1 knock-out (KO) and wild type (WT) mice from experimental groups were fed with a high-fat diet (HFD) since the age of six weeks for 20 weeks. The body weight gain, fast glucose and serum lipid levels were measured and recorded. In addition, pathological analysis, echocardiography, electrocardiography, langendorff-perfused heart and molecular analysis were performed to detect HFD-induced vulnerability to VA and its underlying mechanisms. After a 20-week HFD feeding, the mice showed an increase in body weight, glycemic, lipid levels, QTc interval, LVEDd, LVEDs and LVFS. HFD feeding also increased vulnerability to VA, as shown by the prolonged action potential duration (APD), enhanced APD alternans threshold and greater incidence of VA. Moreover, HFD feeding caused LV hypertrophy and fibrosis, and decreased the protein expressions of Kv4.2, Kv4.3, Kv1.5, Kv2.1 and Cav1.2 channels. At last, the above-mentioned HFD-induced adverse effects were further exacerbated in KO mice compared with WT mice. Mechanistically, MD1 deletion markedly enhanced the activation of TLR4/MyD88/CaMKII signaling pathway in HFD-fed mice. CONCLUSION MD1 deficiency increased HFD-induced vulnerability to VA. This is mainly caused by the aggravated maladaptive LV hypertrophy, fibrosis and decreased protein expressions of ion channels, which are induced by the enhanced activation of the TLR4/MyD88/CaMKII signaling pathway.
Collapse
Affiliation(s)
- Wei Shuai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Bin Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Hui Fu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Caijie Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China.
| |
Collapse
|
19
|
Seara FAC, Arantes PC, Domingos AE, Barbosa RAQ, Olivares EL, Sudo RT, Campos de Carvalho AC, Nascimento JHM. Cardiac electrical and contractile disorders promoted by anabolic steroid overdose are associated with late autonomic imbalance and impaired Ca 2+ handling. Steroids 2019; 148:1-10. [PMID: 31028764 DOI: 10.1016/j.steroids.2019.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/02/2019] [Accepted: 04/08/2019] [Indexed: 02/05/2023]
Abstract
AIM Investigate cardiac electrical and mechanical dysfunctions elicited by chronic anabolic steroid (AS) overdose. METHODS Male Wistar rats were treated with nandrolone decanoate (DECA) or vehicle (CTL) for 8 weeks. Electrocardiography and heart rate variability were assessed at weeks 2, 4, and 8. Cardiac reactivity to isoproterenol was investigated in isolated rat hearts. Action potential duration (APD) was measured from left ventricular (LV) muscle strips. L-type Ca2+ current (ICaL), and transient outward potassium current (Ito) were recorded by whole-cell patch-clamp in LV cardiomyocytes. Sarcoplasmic reticulum (SR) Ca2+ mobilization and Ca2+-induced contractile response sensitivity were evaluated in skinned cardiac fibers. Muscarinic type 2 receptor (M2R), β1-adrenergic receptor (β1AR), sarcoplasmic Ca2+ ATPase (SERCA-2a), type 2 ryanodine receptor (RyR2), L-type Ca2+ channel (CACNA1), Kv4.2 (KCND2), and Kv4.3 (KCND3) mRNA expression levels were measured by quantitative RT-PCR. RESULTS Compared with CTL group, DECA group exhibited decreased high frequency band power density (HF) and increased low frequency power density (LF), Cardiac M2R mRNA level was decreased. QTc interval at 2nd, 4th, and 8th week as well as APD30 and APD90 were increased by DECA. Ito density was decreased, while ICaL density was increased by DECA. SR Ca2+ loading and release were decreased by DECA, while contractile sensitivity to Ca2+ was increased versus CTL group. CONCLUSION DECA overdose induced cardiac rhythmic and mechanical abnormalities that can be associated with autonomic imbalance, up-regulated ICaL and down-regulated Ito, abnormal SR Ca2+ mobilization, and increased contractile sensitivity to Ca2+.
Collapse
Affiliation(s)
- Fernando A C Seara
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil.
| | - Paulo C Arantes
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Ainá E Domingos
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Raiana A Q Barbosa
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Emerson L Olivares
- Department of Physiological Sciences, Institute of Biology and Health Sciences, Federal Rural University of Rio de Janeiro, Seropedica 23890-000, RJ, Brazil
| | - Roberto T Sudo
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Antonio C Campos de Carvalho
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Jose H M Nascimento
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| |
Collapse
|
20
|
Nandrolone alter left ventricular contractility and promotes remodelling involving calcium-handling proteins and renin-angiotensin system in male SHR. Life Sci 2018; 208:239-245. [PMID: 30040952 DOI: 10.1016/j.lfs.2018.07.041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/12/2018] [Accepted: 07/20/2018] [Indexed: 11/24/2022]
Abstract
AIMS Hypertension is a highly prevalent disease that has been correlated to severe organ damage and mortality. However, the role of androgens in hypertension is controversial. The aim of this study was to evaluate the cardiac effects of the nandrolone decanoate (NDL) in male SHR. MAIN METHODS At 12 weeks of age, male SHR rats were separated into three groups: Control (CON), Nandrolone 10 mg/kg twice weekly (NDL), and NDL plus Enalapril 10 mg/kg/day (NDL-E) groups. The animals were treated for 4 weeks. Haemodynamic parameters were acquired through ventricular catheter implantation. The left ventricle was stained with haematoxylin/eosin or picrosirius red. Western blot analysis of TNF-α, ACE, AT1R, β1-AR, PLB, p-PLBser16 and SERCA2a was performed. KEY FINDINGS Nandrolone increased hypertension in SHR rats and enalapril reduced blood pressure to values below those of the control. NDL increased +dP/dtmax, -dP/dtmax and cardiac hypertrophy, which were prevented in the NDL-E group. Cardiac collagen deposition was increased in the NDL group, with this effect being attenuated by enalapril in NDL-E animals. TNF-α, ACE, AT1R and β1-AR proteins were increased in the NDL, and enalapril decreased them, except for TNF-α. The ratio p-PLBser16/PLB revealed an increase after nandrolone, which was prevented in the NDL-E group. The SERCA2a expression protein and SERCA2a/PLB were increased in NDL animals, which did not occur in the NDL-E group. SIGNIFICANCE Nandrolone has distinct effects on cardiac function and remodelling in male SHR, altering the hypertension development process in the heart through modulation of calcium handling proteins and the renin-angiotensin system.
Collapse
|
21
|
Marocolo M, Silva-Neto JA, Barbosa Neto O. Acute interruption of treatment with nandrolone decanoate is not sufficient to reverse cardiac autonomic dysfunction and ventricular repolarization disturbances in rats. Steroids 2018; 132:12-17. [PMID: 29366731 DOI: 10.1016/j.steroids.2018.01.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/10/2018] [Accepted: 01/16/2018] [Indexed: 11/26/2022]
Abstract
Anabolic androgenic steroids are a class of synthetic compounds derived from testosterone, eventually used by athletes, to improve physical performance. However, anabolic steroids can also modify normal cardiovascular function. Thus, we investigated cardiac electrophysiological and autonomic abnormalities in rats, through a electrocardiographic variability protocol during and after interruption of administration of nandrolone decanoate (DECA) anabolic steroid. Twenty male Wistar rats (60-70 days old) received DECA (10 mg. kg-1i.m) once a week or vehicle, during eight weeks. Electrocardiogram was recorded in conscious rats by a noninvasive method, and time and domain analysis of heart rate variability as well as electrocardiogram intervals (QTc / QTd) were performed. Body mass was lower in treated rats compared to control after 4th and 8th weeks, but not at the end of 14th week. QTc and QTd were longer in DECA group compared to control on 4th, 8th, 11th, but equal on 14th week. Cardiac autonomic dysfunction (vagal attenuation) was present on DECA group after 4th week and did not normalize after interruption of treatment. The animals of DECA group showed a correlation between attenuated parasympathetic modulation and increased correct QT interval. Our data allow us to conclude that long-term treatment with DECA impairs autonomic cardiac physiology, predisposing to cardiovascular risk and sudden death, and interruption of administration does not recovery the normality immediately.
Collapse
Affiliation(s)
- Moacir Marocolo
- Physiology and Human Performance Research Group, Department of Physiology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil.
| | - Julio A Silva-Neto
- "Paes de Carvalho" Laboratory of Biophysics of Heart, Department of Physiology, Federal University of Sergipe, Aracaju, Brazil
| | - Octávio Barbosa Neto
- Department of Sport Sciences, Federal University of Triângulo Mineiro, Uberaba, Brazil
| |
Collapse
|
22
|
Goldman A, Basaria S. Adverse health effects of androgen use. Mol Cell Endocrinol 2018; 464:46-55. [PMID: 28606866 DOI: 10.1016/j.mce.2017.06.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 06/08/2017] [Indexed: 01/21/2023]
Abstract
Anabolic androgenic steroids (AAS) are performance enhancing drugs commonly used by athletes and bodybuilders to improve appearance and athletic capability. Unfortunately, these testosterone derivatives can be associated with serious and potentially irreversible side effects, and can impact multiple organ systems. It is important that physicians be familiar with these adverse consequences so that they can appropriately counsel patients whom they suspect of AAS-abuse. In this chapter, we will review the negative effects of these compounds on various organ systems in men using AAS.
Collapse
Affiliation(s)
- Anna Goldman
- Research Program in Men's Health: Aging and Metabolism, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shehzad Basaria
- Research Program in Men's Health: Aging and Metabolism, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
23
|
Morentin B, Callado LF, García-Hernández S, Bodegas A, Lucena J. The role of toxic substances in sudden cardiac death. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.remle.2017.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
24
|
Barbosa Neto O, da Mota GR, De Sordi CC, Resende EAMR, Resende LAPR, Vieira da Silva MA, Marocolo M, Côrtes RS, de Oliveira LF, Dias da Silva VJ. Long-term anabolic steroids in male bodybuilders induce cardiovascular structural and autonomic abnormalities. Clin Auton Res 2017; 28:231-244. [DOI: 10.1007/s10286-017-0470-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 09/12/2017] [Indexed: 10/18/2022]
|
25
|
Baggish AL, Weiner RB, Kanayama G, Hudson JI, Lu MT, Hoffmann U, Pope HG. Cardiovascular Toxicity of Illicit Anabolic-Androgenic Steroid Use. Circulation 2017; 135:1991-2002. [PMID: 28533317 DOI: 10.1161/circulationaha.116.026945] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 03/13/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Millions of individuals have used illicit anabolic-androgenic steroids (AAS), but the long-term cardiovascular associations of these drugs remain incompletely understood. METHODS Using a cross-sectional cohort design, we recruited 140 experienced male weightlifters 34 to 54 years of age, comprising 86 men reporting ≥2 years of cumulative lifetime AAS use and 54 nonusing men. Using transthoracic echocardiography and coronary computed tomography angiography, we assessed 3 primary outcome measures: left ventricular (LV) systolic function (left ventricular ejection fraction), LV diastolic function (early relaxation velocity), and coronary atherosclerosis (coronary artery plaque volume). RESULTS Compared with nonusers, AAS users demonstrated relatively reduced LV systolic function (mean±SD left ventricular ejection fraction = 52±11% versus 63±8%; P<0.001) and diastolic function (early relaxation velocity = 9.3±2.4 cm/second versus 11.1±2.0 cm/second; P<0.001). Users currently taking AAS at the time of evaluation (N=58) showed significantly reduced LV systolic (left ventricular ejection fraction = 49±10% versus 58±10%; P<0.001) and diastolic function (early relaxation velocity = 8.9±2.4 cm/second versus 10.1±2.4 cm/second; P=0.035) compared with users currently off-drug (N=28). In addition, AAS users demonstrated higher coronary artery plaque volume than nonusers (median [interquartile range] 3 [0, 174] mL3 versus 0 [0, 69] mL3; P=0.012). Lifetime AAS dose was strongly associated with coronary atherosclerotic burden (increase [95% confidence interval] in rank of plaque volume for each 10-year increase in cumulative duration of AAS use: 0.60 SD units [0.16-1.03 SD units]; P=0.008). CONCLUSIONS Long-term AAS use appears to be associated with myocardial dysfunction and accelerated coronary atherosclerosis. These forms of AAS-associated adverse cardiovascular phenotypes may represent a previously underrecognized public-health problem.
Collapse
Affiliation(s)
- Aaron L Baggish
- From Cardiovascular Performance Program, Division of Cardiology, Massachusetts General Hospital, Boston (A.L.B., R.B.W.); Department of Medicine, Harvard Medical School, Boston, MA (A.L.B., R.B.W.); Biological Psychiatry Laboratory and Psychiatric Epidemiology Research Program, McLean Hospital, Belmont, MA (G.K., J.I.H., H.G.P.); Department of Psychiatry, Harvard Medical School, Boston, MA (G.K., J.I.H., H.G.P.); Cardiac MR PET CT Program, Massachusetts General Hospital and Department of Radiology, Harvard Medical School, Boston (M.T.L., U.H.).
| | - Rory B Weiner
- From Cardiovascular Performance Program, Division of Cardiology, Massachusetts General Hospital, Boston (A.L.B., R.B.W.); Department of Medicine, Harvard Medical School, Boston, MA (A.L.B., R.B.W.); Biological Psychiatry Laboratory and Psychiatric Epidemiology Research Program, McLean Hospital, Belmont, MA (G.K., J.I.H., H.G.P.); Department of Psychiatry, Harvard Medical School, Boston, MA (G.K., J.I.H., H.G.P.); Cardiac MR PET CT Program, Massachusetts General Hospital and Department of Radiology, Harvard Medical School, Boston (M.T.L., U.H.)
| | - Gen Kanayama
- From Cardiovascular Performance Program, Division of Cardiology, Massachusetts General Hospital, Boston (A.L.B., R.B.W.); Department of Medicine, Harvard Medical School, Boston, MA (A.L.B., R.B.W.); Biological Psychiatry Laboratory and Psychiatric Epidemiology Research Program, McLean Hospital, Belmont, MA (G.K., J.I.H., H.G.P.); Department of Psychiatry, Harvard Medical School, Boston, MA (G.K., J.I.H., H.G.P.); Cardiac MR PET CT Program, Massachusetts General Hospital and Department of Radiology, Harvard Medical School, Boston (M.T.L., U.H.)
| | - James I Hudson
- From Cardiovascular Performance Program, Division of Cardiology, Massachusetts General Hospital, Boston (A.L.B., R.B.W.); Department of Medicine, Harvard Medical School, Boston, MA (A.L.B., R.B.W.); Biological Psychiatry Laboratory and Psychiatric Epidemiology Research Program, McLean Hospital, Belmont, MA (G.K., J.I.H., H.G.P.); Department of Psychiatry, Harvard Medical School, Boston, MA (G.K., J.I.H., H.G.P.); Cardiac MR PET CT Program, Massachusetts General Hospital and Department of Radiology, Harvard Medical School, Boston (M.T.L., U.H.)
| | - Michael T Lu
- From Cardiovascular Performance Program, Division of Cardiology, Massachusetts General Hospital, Boston (A.L.B., R.B.W.); Department of Medicine, Harvard Medical School, Boston, MA (A.L.B., R.B.W.); Biological Psychiatry Laboratory and Psychiatric Epidemiology Research Program, McLean Hospital, Belmont, MA (G.K., J.I.H., H.G.P.); Department of Psychiatry, Harvard Medical School, Boston, MA (G.K., J.I.H., H.G.P.); Cardiac MR PET CT Program, Massachusetts General Hospital and Department of Radiology, Harvard Medical School, Boston (M.T.L., U.H.)
| | - Udo Hoffmann
- From Cardiovascular Performance Program, Division of Cardiology, Massachusetts General Hospital, Boston (A.L.B., R.B.W.); Department of Medicine, Harvard Medical School, Boston, MA (A.L.B., R.B.W.); Biological Psychiatry Laboratory and Psychiatric Epidemiology Research Program, McLean Hospital, Belmont, MA (G.K., J.I.H., H.G.P.); Department of Psychiatry, Harvard Medical School, Boston, MA (G.K., J.I.H., H.G.P.); Cardiac MR PET CT Program, Massachusetts General Hospital and Department of Radiology, Harvard Medical School, Boston (M.T.L., U.H.)
| | - Harrison G Pope
- From Cardiovascular Performance Program, Division of Cardiology, Massachusetts General Hospital, Boston (A.L.B., R.B.W.); Department of Medicine, Harvard Medical School, Boston, MA (A.L.B., R.B.W.); Biological Psychiatry Laboratory and Psychiatric Epidemiology Research Program, McLean Hospital, Belmont, MA (G.K., J.I.H., H.G.P.); Department of Psychiatry, Harvard Medical School, Boston, MA (G.K., J.I.H., H.G.P.); Cardiac MR PET CT Program, Massachusetts General Hospital and Department of Radiology, Harvard Medical School, Boston (M.T.L., U.H.).
| |
Collapse
|
26
|
de Andrade EC, de Castro Paiva KC, da Silva Guedes S, Souza MLC, Pereira MN, Miana LP, de Figueiredo AA, de Bessa J, Netto JMB. Echocardiographic evaluation of left ventricular mass index in children with hypospadias after hormonal stimulation with topical testosterone: A randomized controlled trial. J Pediatr Urol 2017; 13:352.e1-352.e7. [PMID: 28434633 DOI: 10.1016/j.jpurol.2017.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 03/06/2017] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Testosterone is often used in the preoperative period of hypospadias surgery. Previous studies have demonstrated the presence of androgen receptors in cardiac myocytes that can modulate the phenotype. The use of supraphysiological doses of androgens can lead to toxicity on the heart muscle and, in some cases, to left ventricular hypertrophy. This randomized double blind controlled clinical trial aims to evaluate the effect of topical testosterone on left ventricular mass index in boys with hypospadias. MATERIALS AND METHODS Boys with hypospadias aged 6 months to 9 years were included. Children were divided into two groups: G1 - boys who received testosterone propionate 1% ointment twice a day for 30 days, and G2 - boys receiving placebo ointment in the same regimen. All children were submitted to bi-dimensional echocardiographic evaluation to compare the left ventricular mass index, blood pressure, and body mass index before and after treatment (30 and 90 days). Levels of serum testosterone, LH, and FSH were measured. RESULTS Thirty-five children were analyzed: 17 in G1 and 18 in G2. No differences were found in left ventricular mass index (left ventricular mass indexed by body surface area) prior to treatment. Left ventricular mass index was 59.21 ± 11.91 g/m2 in G1 and 55.12 ± 8.29 g/m2 in G2 (p = 0.244) after 30 days of treatment, and 61.13 ± 11.69 g/m2 in G1 and 62.84 ± 35.99 g/m2 in G2 (p = 0.852) after 90 days. Serum testosterone levels were 12 (7-80) ng/dL in G1 and 5 (5-7) ng/dL in G2 (p = 0.018) after 30 days of treatment, and 10 (5-11) ng/dL in G1 and 5 (4-5) ng/dL in G2 (p = 0.155), after 90 days (Figure). There was a small increase in systolic blood pressure (SBP) after 30 days (83.82 ± 7.18 mmHg) in the group who receive testosterone (G1) compared with controls (77.5 ± 6.69 mmHg) (p = 0.010). After 90 days, SBP levels returned to basal levels in G1 (82.35 ± 5.62 mmHg) and in G2 (81.38 ± 4.79 mmHg) (p = 0.588). CONCLUSION Topical testosterone can be considered safe in the preoperative period of children with hypospadias with no risk of left ventricular hypertrophy. An increase in systolic blood pressure occurs while using testosterone but it is transitory, returning to normal levels after 90 days.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - José de Bessa
- Department of Surgery, Division of Urology, State University of Feira de Santana, UEFS, Brazil
| | - José Murillo B Netto
- Department of Surgery, Division of Urology, Federal University of Juiz de Fora, UFJF, Brazil; Department of Surgery, Division of Urology, Hospital e Maternidade Therezinha de Jesus da Faculdade de Ciências Médicas e da Saúde de Juiz de Fora, Suprema, Brazil.
| |
Collapse
|
27
|
Pergolizzi B, Carriero V, Abbadessa G, Penna C, Berchialla P, De Francia S, Bracco E, Racca S. Subchronic nandrolone administration reduces cardiac oxidative markers during restraint stress by modulating protein expression patterns. Mol Cell Biochem 2017; 434:51-60. [DOI: 10.1007/s11010-017-3036-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 04/12/2017] [Indexed: 01/20/2023]
|
28
|
Argenziano M, Tiscornia G, Moretta R, Casal L, Potilinski C, Amorena C, Gras EG. Arrhythmogenic effect of androgens on the rat heart. J Physiol Sci 2017; 67:217-225. [PMID: 27241707 PMCID: PMC10717165 DOI: 10.1007/s12576-016-0459-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 05/06/2016] [Indexed: 01/05/2023]
Abstract
In most species androgens shorten the cardiac action potential and reduce the risk of afterdepolarizations. Despite the central role of the rat model in physiological studies, the effects of androgens on the rat heart are still inconclusive. We therefore performed electrophysiological studies on the perfused rat right ventricular free wall. We found a correlation between androgenic activity and a propensity to generate ventricular ectopic action potentials. We also found that the testosterone treatment increased action potential duration at 90 % of repolarization (APD90), while androgenic inhibition increased the time to peak and decreased APD90. We observed that the voltage-gated potassium channel Kv4.3 and the bi-directional membrane ion transporter NCX in the rat myocardium were regulated by androgenic hormones. One possible explanation for these findings is that due to the expression of specific ion channels in the rat myocardium, the action potential response to its hormonal background is different from those described in other experimental models. Our results indicate that androgenic control of NCX expression plays a key role in determining arrhythmogenicity in the rat heart.
Collapse
Affiliation(s)
- Mariana Argenziano
- Centro de Estudios en Salud y Medio Ambiente (CESyMA), Escuela de Ciencia y Tecnología (ECyT), Universidad Nacional de General San Martín (UNSAM), Av. Gral. Paz 5445, INTI, Edificio 23, 1650, San Martin, Buenos Aires, Argentina
| | - Gisela Tiscornia
- Centro de Estudios en Salud y Medio Ambiente (CESyMA), Escuela de Ciencia y Tecnología (ECyT), Universidad Nacional de General San Martín (UNSAM), Av. Gral. Paz 5445, INTI, Edificio 23, 1650, San Martin, Buenos Aires, Argentina
| | - Rosalia Moretta
- Centro de Estudios en Salud y Medio Ambiente (CESyMA), Escuela de Ciencia y Tecnología (ECyT), Universidad Nacional de General San Martín (UNSAM), Av. Gral. Paz 5445, INTI, Edificio 23, 1650, San Martin, Buenos Aires, Argentina
| | - Leonardo Casal
- Centro de Estudios en Salud y Medio Ambiente (CESyMA), Escuela de Ciencia y Tecnología (ECyT), Universidad Nacional de General San Martín (UNSAM), Av. Gral. Paz 5445, INTI, Edificio 23, 1650, San Martin, Buenos Aires, Argentina
| | - Constanza Potilinski
- Centro de Estudios en Salud y Medio Ambiente (CESyMA), Escuela de Ciencia y Tecnología (ECyT), Universidad Nacional de General San Martín (UNSAM), Av. Gral. Paz 5445, INTI, Edificio 23, 1650, San Martin, Buenos Aires, Argentina
- The National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Carlos Amorena
- Centro de Estudios en Salud y Medio Ambiente (CESyMA), Escuela de Ciencia y Tecnología (ECyT), Universidad Nacional de General San Martín (UNSAM), Av. Gral. Paz 5445, INTI, Edificio 23, 1650, San Martin, Buenos Aires, Argentina
- The National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Eduardo Garcia Gras
- Centro de Estudios en Salud y Medio Ambiente (CESyMA), Escuela de Ciencia y Tecnología (ECyT), Universidad Nacional de General San Martín (UNSAM), Av. Gral. Paz 5445, INTI, Edificio 23, 1650, San Martin, Buenos Aires, Argentina.
- The National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
29
|
Kassa RM, Bonafede R, Boschi F, Bentivoglio M, Mariotti R. Effect of physical exercise and anabolic steroid treatment on spinal motoneurons and surrounding glia of wild-type and ALS mice. Brain Res 2016; 1657:269-278. [PMID: 28048973 DOI: 10.1016/j.brainres.2016.12.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/07/2016] [Accepted: 12/29/2016] [Indexed: 12/12/2022]
Abstract
Motoneuron degeneration is the hallmark of amyotrophic lateral sclerosis (ALS). The cause and predisposing factors for sporadic ALS are still unknown. Exposure to a specific environmental risk factors in subjects with a susceptibility genotype may increase the risk of the disease. The role of physical activity and the use of anabolic steroids are still debated in epidemiological studies on patients and murine models of ALS. To assess at the cellular level the role (beneficial or detrimental) of physical exercise and the use of anabolic steroid, we here investigated, in SOD1(G93A) (mSOD1) mice and wild-type littermates, changes in the ventral horn after regular exercise, treatment with the anabolic androgenic steroid 19-nortestosterone (nandrolone), and their combination, compared with matched control sedentary mice. The experiments were pursued for several weeks until symptom onset in mSOD1 mice. Lumbar motoneurons, astrocytes and microglia were analyzed. In wild-type mice, cytological alterations of motoneurons were observed especially after nandrolone treatment. The following main findings were observed in treated mSOD1 mice versus untreated ones: i) nandrolone treatment markedly enhanced motoneuron loss; this detrimental effect was reverted by the combination with exercise, resulting in increased motoneuron survival; ii) astrocytic activation was most marked after nandrolone treatment when motoneuron damage was most severe; iii) microglia activation was most marked after physical exercise when motoneuron damage was less severe. The results indicate a vulnerability of mSOD1 motoneurons to nandrolone treatment, a potential neuroprotective effect of physical exercise, and a modulation by glial cells in the ALS murine model in the examined paradigms.
Collapse
Affiliation(s)
- Roman M Kassa
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Roberta Bonafede
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Federico Boschi
- Department of Computer Sciences, University of Verona, Verona, Italy
| | - Marina Bentivoglio
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Raffaella Mariotti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.
| |
Collapse
|
30
|
Rodrigues Junior LF, de Azevedo Carvalho AC, Pimentel EB, Mill JG, Nascimento JHM. Chronic enalapril treatment increases transient outward potassium current in cardiomyocytes isolated from right ventricle of spontaneously hypertensive rats. Naunyn Schmiedebergs Arch Pharmacol 2016; 390:225-234. [PMID: 27915452 DOI: 10.1007/s00210-016-1322-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 11/21/2016] [Indexed: 01/19/2023]
Abstract
It has been well established that chronic pressure overload resulting from hypertension leads to ventricular hypertrophy and electrophysiological remodeling. The transient outward potassium current (I to) reduction described in hypertensive animals delays ventricular repolarization, leading to complex ventricular arrhythmias and sudden death. Antihypertensive drugs, as angiotensin-converting enzyme inhibitors (ACEi), can restore I to and reduce the incidence of arrhythmic events. The purpose of this study was to evaluate the differential effects of long-term treatment with ACEi or direct-acting smooth muscle relaxant on the I to of left and right ventricle myocytes of spontaneously hypertensive rats (SHR). Animals were divided into four groups: normotensive Wistar-Kyoto rats (WKY), hypertensive (SHR), SHR treated for 6 weeks with enalapril 10 mg/kg/day (SHRE), or hydralazine 20 mg/kg/day (SHRH). Systolic blood pressure (SBP) and hypertrophy index (heart weight/body weight (HW/BW)) were determined at the end of treatment period. Cell membrane capacitance (C m) and I to were assessed in cardiomyocytes isolated from left and right ventricles. The SHR exhibited significantly increased SBP and HW/BW when compared to the WKY. The treated groups, SHRE and SHRH, restored normal SBP but not HW/BW. The SHR group exhibited a diminished I to in the left but not the right ventricle. Both the treated groups restored I to in the left ventricle. However, in the right ventricle, only enalapril treatment modified I to. The SHRE group exhibited a significant increase in I to compared to all the other groups. These findings suggest that enalapril may increase I to by a pressure overload independent mechanism.
Collapse
Affiliation(s)
- Luiz Fernando Rodrigues Junior
- Institute of Biophysics Carlos Chagas Filho, Laboratory of Cardiac Electrophysiology Antonio Paes de Carvalho, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, 373 - CCS Bloco G - Ilha do Fundao, 21, Rio de Janeiro, RJ, 941-902, Brazil.,Department of Physiological Sciences, Laboratory of Cardiovascular Biophysics, Federal University of State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Carolina de Azevedo Carvalho
- Institute of Biophysics Carlos Chagas Filho, Laboratory of Cardiac Electrophysiology Antonio Paes de Carvalho, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, 373 - CCS Bloco G - Ilha do Fundao, 21, Rio de Janeiro, RJ, 941-902, Brazil
| | | | - José Geraldo Mill
- Department of Physiological Sciences, Federal University of Espírito Santo, Vitória, Brazil
| | - José Hamilton Matheus Nascimento
- Institute of Biophysics Carlos Chagas Filho, Laboratory of Cardiac Electrophysiology Antonio Paes de Carvalho, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, 373 - CCS Bloco G - Ilha do Fundao, 21, Rio de Janeiro, RJ, 941-902, Brazil.
| |
Collapse
|
31
|
The Effects of Nandrolone Decanoate Along with Prolonged Low-Intensity Exercise on Susceptibility to Ventricular Arrhythmias. Cardiovasc Toxicol 2016; 16:23-33. [PMID: 25636207 DOI: 10.1007/s12012-015-9313-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We examined the influence of chronic administration of nandrolone decanoate with low-intensity endurance swimming exercise on susceptibility to lethal ventricular arrhythmias in rat. The animal groups included the control group, exercise group (EX), nandrolone group (Nan), vehicle group (Arach), trained vehicle group (Arach + Ex) and trained nandrolone group (Nan + Ex) that treated for 8 weeks. Then, arrhythmia induction was performed by intravenous infusion of aconitine and electrocardiogram recorded. Then, malondialdehyde (MDA), hydroxyproline (HYP) and glutathione peroxidase of heart tissue were measured. Chronic administration of nandrolone with low-intensity endurance swimming exercise had no significant effect on blood pressure, heart rate and basal ECG parameters except RR interval that showed increase (P < 0.05). Low-intensity exercise could prevent the incremental effect of nandrolone on MDA and HYP significantly. It also increased the heart hypertrophy index (P < 0.05) and reduced the abating effect of nandrolone on animal weighting. Nandrolone along with exercise significantly increased the duration of VF (P < 0.05) and reduced the VF latency (P < 0.05). The findings suggest that chronic co-administration of nandrolone with low-intensity endurance swimming exercise to some extent facilitates the occurrence of ventricular fibrillation in rat. Complementary studies are needed to elucidate the involved mechanisms of this abnormality.
Collapse
|
32
|
Abstract
Professional and amateur athletic training can cause tremendous overload of the cardiovascular system and thus become a trigger of serious and often fatal cardiac events in athletes with a previously undetected underlying cardiovascular disease. Therefore, every athlete should undergo a specialized diagnostic and qualification screening before a training program is prescribed or continued. However, it is still an unresolved issue which of the accessible diagnostic tools should be routinely applied in order to increase the safety of extreme physical training and reduce the risk of sudden cardiac death. Pre-participation athlete evaluation including a standard electrocardiogram (ECG), physical examination, and familial history of cardiovascular diseases is important, but does not always guarantee high diagnostic accuracy. Hence, the aim of this review article is to discuss the controversy over the usefulness of ECG for the detection of cardiovascular diseases in athletes as well as the views on athlete screening methods in Europe and the USA. Differential diagnostic options and screening schemes are also described in particular groups of athletes in reference to their age, cardiovascular risk factors, as well as intensity and type of sport discipline.
Collapse
Affiliation(s)
- R Skalik
- Department of Physiology, Medical University of Wroclaw, T. Chalubinskiego ST 10, 50-368, Wroclaw, Poland,
| |
Collapse
|
33
|
Trentin-Sonoda M, da Silva RC, Kmit FV, Abrahão MV, Monnerat Cahli G, Brasil GV, Muzi-Filho H, Silva PA, Tovar-Moll FF, Vieyra A, Medei E, Carneiro-Ramos MS. Knockout of Toll-Like Receptors 2 and 4 Prevents Renal Ischemia-Reperfusion-Induced Cardiac Hypertrophy in Mice. PLoS One 2015; 10:e0139350. [PMID: 26448184 PMCID: PMC4598103 DOI: 10.1371/journal.pone.0139350] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 08/06/2015] [Indexed: 11/30/2022] Open
Abstract
We investigated whether the pathways linked to Toll-like receptors 2 and 4 (TLRs) are involved in renal ischemia-reperfusion (I/R)-induced cardiac hypertrophy. Wild type (WT) C57BL/6J, TLR2-/- and TLR4-/- mice were subjected to left kidney ischemia for 60 min followed by reperfusion for 5, 8, 12 and 15 days. Proton density magnetic resonance showed alterations in the injured kidney from WT mice, together with signs of parenchymal edema and higher levels of vimentin mRNA, accompanied by: (i) small, but significant, increase in serum urea after 24 h, (ii) 100% increase in serum creatinine at 24 h. A serum peak of inflammatory cytokines occurred after 5 days of reperfusion. Heart weight/body weight and heart weight/tibia length ratios increased after 12 and 15 days of reperfusion, respectively. Cardiac hypertrophy markers, B-type natriuretic peptide (BNP) and α-actin, left ventricle mass, cardiac wall thickness and myocyte width increased after 15 days of reperfusion, together with longer QTc and action potential duration. Cardiac TLRs, MyD88, HSP60 and HSP70 mRNA levels also increased. After 15 days of reperfusion, absence of TLRs prevented cardiac hypertrophy, as reflected by similar values of left ventricular cardiac mass and heart weight/body weight ratio compared to the transgenic Sham. Renal tissular injury also ameliorated in both knockout mice, as revealed by the comparison of their vimentin mRNA levels with those found in the WT on the same day after I/R. The I/R TLR2-/- group had TNF-α, IFN-γ and IL-1β levels similar to the non-I/R group, whereas the TLR4-/- group conserved the p-NF-κB/NF- κB ratio contrasting with that found in TLR2-/-. We conclude: (i) TLRs are involved in renal I/R-induced cardiac hypertrophy; (ii) absence of TLRs prevents I/R-induced cardiac hypertrophy, despite renal lesions seeming to evolve towards those of chronic disease; (iii) TLR2 and TLR4 selectively regulate the systemic inflammatory profile and NF- κB activation.
Collapse
Affiliation(s)
- Mayra Trentin-Sonoda
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, Brazil
| | | | - Fernanda Vieira Kmit
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, Brazil
| | | | - Gustavo Monnerat Cahli
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Rio de Janeiro, Brazil
| | - Guilherme Visconde Brasil
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Rio de Janeiro, Brazil
| | - Humberto Muzi-Filho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Rio de Janeiro, Brazil
| | - Paulo André Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Rio de Janeiro, Brazil
| | - Fernanda Freire Tovar-Moll
- Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Rio de Janeiro, Brazil
- Instituto D’Or de Pesquisa e Ensino, Rio de Janeiro, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Adalberto Vieyra
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Rio de Janeiro, Brazil
| | - Emiliano Medei
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Rio de Janeiro, Brazil
| | | |
Collapse
|
34
|
Bayat G, Javan M, Safari F, Khalili A, Shokri S, Goudarzvand M, Salimi M, Hajizadeh S. Nandrolone decanoate negatively reverses the beneficial effects of exercise on cardiac muscle via sarcolemmal, but not mitochondrial K(ATP) channel. Can J Physiol Pharmacol 2015; 94:324-31. [PMID: 26909616 DOI: 10.1139/cjpp-2015-0040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
ATP-sensitive potassium channels are supposed to have a substantial role in improvement of cardiac performance. This study was performed to evaluate whether nandrolone decanoate (ND) and (or) exercise training could affect the expression of cardiac K(ATP) channel subunits. Thirty-five male albino Wistar rats were randomly divided into 5 groups, including sedentary control (SC), sedentary vehicle (SV), sedentary ND (SND), exercise control (EC), and exercise and ND (E+ND). Exercise training was performed on a treadmill 5 times per week. ND was injected (10 mg/kg/week, i.m.) to the rats in the SND and E+ND groups. Following cardiac isolation, the expression of both sarcolemmal and mitochondrial subunits of K(ATP) channel was measured using Western blot method. The expression of sarcolemmal, but not mitochondrial, subunits of K(ATP) channel (Kir6.2 and SUR2) of EC group was significantly higher compared with SC group while ND administration (SND group) did not show any change in their expression. In the E+ND group, ND administration led to decrease of the over-expression of sarcolemmal Kir6.2 and SUR2 which was previously induced by exercise. There was no significant association between the mitochondrial expression of either Kir6.2 or SUR2 proteins and administration of ND or exercise. Supra-physiological dosage of ND negatively reverses the effects of exercise on the cardiac muscle expression of sarcolemmal, but not mitochondrial, K(ATP) channel subunits.
Collapse
Affiliation(s)
- Gholamreza Bayat
- a Department of Physiology and Pharmacology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Javan
- b Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Safari
- c Deptartment of Physiology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Azadeh Khalili
- d Department of Pharmacology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Shokri
- e Department of Anatomy, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mahdi Goudarzvand
- a Department of Physiology and Pharmacology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Mehdi Salimi
- b Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sohrab Hajizadeh
- b Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
35
|
Ghorbani Baravati H, Joukar S, Fathpour H, Kordestani Z. Nandrolone Plus Moderate Exercise Increases the Susceptibility to Lethal Arrhythmias. Res Cardiovasc Med 2015; 4:e26233. [PMID: 26396972 PMCID: PMC4576214 DOI: 10.5812/cardiovascmed.26233v2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Revised: 01/14/2015] [Accepted: 01/26/2015] [Indexed: 11/30/2022] Open
Abstract
Background: Until now, no experimental study has directly assessed the arrhythmogenesis of chronic consumption of anabolic androgenic steroids along with moderate-intensity endurance exercise. Objectives: We evaluated the influence of integration of anabolic androgenic steroids along with moderate-intensity endurance exercise on susceptibility to lethal ventricular arrhythmias in rat. Materials and Methods: The animal groups were as follows: control group (CTL); exercise group (EX) which were under 6 weeks of treadmill exercise; nandrolone group (Nan) which received 5 mg/kg of nandrolone decanoate twice a week; vehicle group (Arach) which received Arachis oil (solvent of nandrolone); trained vehicle group (Arach + Ex); and trained nandrolone group (Nan + Ex). One day after ending of the intervention period, arrhythmia was inducted by intravenous infusion of aconitine and ventricular arrhythmias were recorded. Then malondialdehyde (MDA) and glutathione peroxidase (GPX) of heart tissue were measured. Results: Nandrolone, exercise, and their combination were associated with heart hypertrophy. Exercise could prevent the incremental effect of nandrolone on MDA/GPX ratio. Chronic administration of nandrolone with moderate-intensity endurance exercise had no significant effect on blood pressure, heart rate, and basal electrocardiographic parameters. Combination of nandrolone and exercise significantly increased the incidence of ventricular fibrillation (VF) and reduced the VF latency (P < 0.05). Conclusions: The findings suggest that chronic coadministration of nandrolone with moderate-intensity endurance exercise facilitates the VF occurrence in rat. Complementary studies are needed to elucidate the involved mechanisms of this abnormality.
Collapse
Affiliation(s)
- Hamideh Ghorbani Baravati
- Deptarment of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, IR Iran
| | - Siyavash Joukar
- Physiology Research Center, Kerman University of Medical Sciences, Kerman, IR Iran
- Department of Physiology and Pharmacology, Kerman University of Medical Sciences, Kerman, IR Iran
- Corresponding author: Siyavash Joukar, Department of Physiology and Pharmacology, Kerman University of Medical Sciences, P. O. Box: 76169-14115, Kerman, IR Iran. Tel/Fax: +98-3433220081, E-mail: ,
| | - Hossein Fathpour
- Deptarment of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, IR Iran
| | - Zeinab Kordestani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, IR Iran
| |
Collapse
|
36
|
Frati P, Busardò FP, Cipolloni L, Dominicis ED, Fineschi V. Anabolic Androgenic Steroid (AAS) related deaths: autoptic, histopathological and toxicological findings. Curr Neuropharmacol 2015; 13:146-59. [PMID: 26074749 PMCID: PMC4462039 DOI: 10.2174/1570159x13666141210225414] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Revised: 10/01/2014] [Accepted: 10/25/2014] [Indexed: 12/22/2022] Open
Abstract
Anabolic androgenic steroids (AASs) represent a large group of synthetic derivatives of testosterone, produced to maximize anabolic effects and minimize the androgenic ones. AAS can be administered orally, parenterally by intramuscular injection and transdermally. Androgens act by binding to the nuclear androgen receptor (AR) in the cytoplasm and then translocate into the nucleus. This binding results in sequential conformational changes of the receptor affecting the interaction between receptor and protein, and receptor and DNA. Skeletal muscle can be considered as the main target tissue for the anabolic effects of AAS, which are mediated by ARs which after exposure to AASs are up-regulated and their number increases with body building. Therefore, AASs determine an increase in muscle size as a consequence of a dose-dependent hypertrophy resulting in an increase of the cross-sectional areas of both type I and type II muscle fibers and myonuclear domains. Moreover, it has been reported that AASs can increase tolerance to exercise by making the muscles more capable to overload therefore shielding them from muscle fiber damage and improving the level of protein synthesis during recovery. Despite some therapeutic use of AASs, there is also wide abuse among athletes especially bodybuilders in order to improve their performances and to increase muscle growth and lean body mass, taking into account the significant anabolic effects of these drugs. The prolonged misuse and abuse of AASs can determine several adverse effects, some of which may be even fatal especially on the cardiovascular system because they may increase the risk of sudden cardiac death (SCD), myocardial infarction, altered serum lipoproteins, and cardiac hypertrophy. The aim of this review is to focus on deaths related to AAS abuse, trying to evaluate the autoptic, histopathological and toxicological findings in order to investigate the pathophysiological mechanism that underlines this type of death, which is still obscure in several aspects. The review of the literature allowed us to identify 19 fatal cases between 1990 and 2012, in which the autopsy excluded in all cases, extracardiac causes of death.
Collapse
Affiliation(s)
- Paola Frati
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161, Rome, Italy
- Neuromed, Istituto Mediterraneo Neurologico (IRCCS), Via Atinense 18, Pozzilli, 86077 Isernia, Italy
| | - Francesco P. Busardò
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161, Rome, Italy
| | - Luigi Cipolloni
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161, Rome, Italy
| | | | - Vittorio Fineschi
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161, Rome, Italy
| |
Collapse
|
37
|
Toll-like receptor 4 activation promotes cardiac arrhythmias by decreasing the transient outward potassium current (Ito) through an IRF3-dependent and MyD88-independent pathway. J Mol Cell Cardiol 2014; 76:116-25. [PMID: 25169970 DOI: 10.1016/j.yjmcc.2014.08.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/31/2014] [Accepted: 08/15/2014] [Indexed: 11/21/2022]
Abstract
Cardiac arrhythmias are one of the main causes of death worldwide. Several studies have shown that inflammation plays a key role in different cardiac diseases and Toll-like receptors (TLRs) seem to be involved in cardiac complications. In the present study, we investigated whether the activation of TLR4 induces cardiac electrical remodeling and arrhythmias, and the signaling pathway involved in these effects. Membrane potential was recorded in Wistar rat ventricle. Ca(2+) transients, as well as the L-type Ca(2+) current (ICaL) and the transient outward K(+) current (Ito), were recorded in isolated myocytes after 24 h exposure to the TLR4 agonist, lipopolysaccharide (LPS, 1 μg/ml). TLR4 stimulation in vitro promoted a cardiac electrical remodeling that leads to action potential prolongation associated with arrhythmic events, such as delayed afterdepolarization and triggered activity. After 24 h LPS incubation, Ito amplitude, as well as Kv4.3 and KChIP2 mRNA levels were reduced. The Ito decrease by LPS was prevented by inhibition of interferon regulatory factor 3 (IRF3), but not by inhibition of interleukin-1 receptor-associated kinase 4 (IRAK4) or nuclear factor kappa B (NF-κB). Extrasystolic activity was present in 25% of the cells, but apart from that, Ca(2+) transients and ICaL were not affected by LPS; however, Na(+)/Ca(2+) exchanger (NCX) activity was apparently increased. We conclude that TLR4 activation decreased Ito, which increased AP duration via a MyD88-independent, IRF3-dependent pathway. The longer action potential, associated with enhanced Ca(2+) efflux via NCX, could explain the presence of arrhythmias in the LPS group.
Collapse
|
38
|
Silva PA, Monnerat-Cahli G, Pereira-Acácio A, Luzardo R, Sampaio LS, Luna-Leite MA, Lara LS, Einicker-Lamas M, Panizzutti R, Madeira C, Vieira-Filho LD, Castro-Chaves C, Ribeiro VS, Paixão ADO, Medei E, Vieyra A. Mechanisms involving Ang II and MAPK/ERK1/2 signaling pathways underlie cardiac and renal alterations during chronic undernutrition. PLoS One 2014; 9:e100410. [PMID: 24983243 PMCID: PMC4077653 DOI: 10.1371/journal.pone.0100410] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 05/27/2014] [Indexed: 02/07/2023] Open
Abstract
Background Several studies have correlated protein restriction associated with other nutritional deficiencies with the development of cardiovascular and renal diseases. The driving hypothesis for this study was that Ang II signaling pathways in the heart and kidney are affected by chronic protein, mineral and vitamin restriction. Methodology/Principal Findings Wistar rats aged 90 days were fed from weaning with either a control or a deficient diet that mimics those used in impoverished regions worldwide. Such restriction simultaneously increased ouabain-insensitive Na+-ATPase and decreased (Na++K+)ATPase activity in the same proportion in cardiomyocytes and proximal tubule cells. Type 1 angiotensin II receptor (AT1R) was downregulated by that restriction in both organs, whereas AT2R decreased only in the kidney. The PKC/PKA ratio increased in both tissues and returned to normal values in rats receiving Losartan daily from weaning. Inhibition of the MAPK pathway restored Na+-ATPase activity in both organs. The undernourished rats presented expanded plasma volume, increased heart rate, cardiac hypertrophy, and elevated systolic pressure, which also returned to control levels with Losartan. Such restriction led to electrical cardiac remodeling represented by prolonged ventricular repolarization parameters, induced triggered activity, early after-depolarization and delayed after-depolarization, which were also prevented by Losartan. Conclusion/Significance The mechanisms responsible for these alterations are underpinned by an imbalance in the PKC- and PKA-mediated pathways, with participation of angiotensin receptors and by activation of the MAPK/ERK1/2 pathway. These cellular and molecular alterations culminate in cardiac electric remodeling and in the onset of hypertension in adulthood.
Collapse
Affiliation(s)
- Paulo A. Silva
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
| | - Gustavo Monnerat-Cahli
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
| | - Amaury Pereira-Acácio
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
| | - Ricardo Luzardo
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
| | - Luzia S. Sampaio
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
| | - Marcia A. Luna-Leite
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lucienne S. Lara
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Einicker-Lamas
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
| | - Rogério Panizzutti
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Caroline Madeira
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leucio D. Vieira-Filho
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife, Brazil
| | - Carmen Castro-Chaves
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife, Brazil
| | - Valdilene S. Ribeiro
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife, Brazil
| | - Ana D. O. Paixão
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife, Brazil
| | - Emiliano Medei
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
| | - Adalberto Vieyra
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
39
|
Pope HG, Wood RI, Rogol A, Nyberg F, Bowers L, Bhasin S. Adverse health consequences of performance-enhancing drugs: an Endocrine Society scientific statement. Endocr Rev 2014; 35:341-75. [PMID: 24423981 PMCID: PMC4026349 DOI: 10.1210/er.2013-1058] [Citation(s) in RCA: 351] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite the high prevalence of performance-enhancing drug (PED) use, media attention has focused almost entirely on PED use by elite athletes to illicitly gain a competitive advantage in sports, and not on the health risks of PEDs. There is a widespread misperception that PED use is safe or that adverse effects are manageable. In reality, the vast majority of PED users are not athletes but rather nonathlete weightlifters, and the adverse health effects of PED use are greatly underappreciated. This scientific statement synthesizes available information on the medical consequences of PED use, identifies gaps in knowledge, and aims to focus the attention of the medical community and policymakers on PED use as an important public health problem. PED users frequently consume highly supraphysiologic doses of PEDs, combine them with other PEDs and/or other classical drugs of abuse, and display additional associated risk factors. PED use has been linked to an increased risk of death and a wide variety of cardiovascular, psychiatric, metabolic, endocrine, neurologic, infectious, hepatic, renal, and musculoskeletal disorders. Because randomized trials cannot ethically duplicate the large doses of PEDs and the many factors associated with PED use, we need observational studies to collect valid outcome data on the health risks associated with PEDs. In addition, we need studies regarding the prevalence of PED use, the mechanisms by which PEDs exert their adverse health effects, and the interactive effects of PEDs with sports injuries and other high-risk behaviors. We also need randomized trials to assess therapeutic interventions for treating the adverse effects of PEDs, such as the anabolic-androgen steroid withdrawal syndrome. Finally, we need to raise public awareness of the serious health consequences of PEDs.
Collapse
Affiliation(s)
- Harrison G Pope
- McLean Hospital (H.G.P.), Harvard Medical School, Belmont, Massachusetts 02478; University of Southern California (R.I.W.), Los Angeles, California 90089; University of Virginia (A.R.), Charlottesville, Virginia 22904; Department of Pharmaceutical Biosciences, (F.N.), Upsala University, SE-751 24, Upsala, Sweden; United States Anti-Doping Agency (L.B.), Colorado Springs, Colorado 80919; and Brigham and Women's Hospital (S.B.), Harvard Medical School, Boston, Massachusetts 02115
| | | | | | | | | | | |
Collapse
|
40
|
Coutinho DCO, Monnerat-Cahli G, Ferreira AJ, Medei E. Activation of angiotensin-converting enzyme 2 improves cardiac electrical changes in ventricular repolarization in streptozotocin-induced hyperglycaemic rats. Europace 2014; 16:1689-96. [PMID: 24741027 DOI: 10.1093/europace/euu070] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
AIMS Diabetic patients present a high level of cardiac arrhythmias and risk of cardiac sudden death. The renin-angiotensin system (RAS) plays a key role in diabetes and cardiac diseases. The present study aimed to evaluate whether an angiotensin-converting enzyme 2 (ACE2) activator, diminazene aceturate (DIZE), could improve the streptozotocin (STZ)-induced electrical changes in ventricular repolarization in hyperglycaemic rats. METHODS AND RESULTS Hyperglycaemia was induced in Wistar male rats with STZ (60 mg/kg/iv). After 4 weeks of STZ injection, rats were daily treated with saline (control) or DIZE (1 mg/kg/gavage) for four consecutive weeks. The cardiac electrical function was evaluated in vivo by electrocardiogram and in vitro by cardiac action potential records in different pacing frequencies. Treatment with DIZE was not able to reverse hyperglycaemia nor body weight loss. However, DIZE reversed hyperglycaemia-induced cardiac electrical changes in ventricular repolarization. Specifically, animals treated with DIZE showed shorter QT and QTc intervals. In addition, ACE2 activation was capable to shorten the cardiac action potential and also reverse the arrhythmic markers. Diminazene aceturate treatment did not induce arrhythmic events in normal, as well as in hyperglycaemic animals. CONCLUSION Our data indicate that activation of ACE2 has a beneficial effect in hyperglycaemic rats, improving the cardiac electrical function. Thus, DIZE represents a promising new therapeutic agent to treat hyperglycaemia-induced cardiac electrical changes in ventricular repolarization.
Collapse
Affiliation(s)
- Danielle C O Coutinho
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Antônio Carlos, 6627-31.270-901, Belo Horizonte, MG, Brazil
| | - Gustavo Monnerat-Cahli
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Ilha do Fundão, 21.949-000, Rio de Janeiro, Brazil
| | - Anderson J Ferreira
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Antônio Carlos, 6627-31.270-901, Belo Horizonte, MG, Brazil
| | - Emiliano Medei
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Ilha do Fundão, 21.949-000, Rio de Janeiro, Brazil
| |
Collapse
|
41
|
Androgens affect muscle, motor neuron, and survival in a mouse model of SOD1-related amyotrophic lateral sclerosis. Neurobiol Aging 2014; 35:1929-38. [PMID: 24630363 DOI: 10.1016/j.neurobiolaging.2014.02.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 01/28/2014] [Accepted: 02/02/2014] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by selective loss of upper and lower motor neurons and skeletal muscle atrophy. Epidemiologic and experimental evidence suggest the involvement of androgens in ALS pathogenesis, but the mechanism through which androgens modify the ALS phenotype is unknown. Here, we show that androgen ablation by surgical castration extends survival and disease duration of a transgenic mouse model of ALS expressing mutant human SOD1 (hSOD1-G93A). Furthermore, long-term treatment of orchiectomized hSOD1-G93A mice with nandrolone decanoate (ND), an anabolic androgenic steroid, worsened disease manifestations. ND treatment induced muscle fiber hypertrophy but caused motor neuron death. ND negatively affected survival, thereby dissociating skeletal muscle pathology from life span in this ALS mouse model. Interestingly, orchiectomy decreased androgen receptor levels in the spinal cord and muscle, whereas ND treatment had the opposite effect. Notably, stimulation with ND promoted the recruitment of endogenous androgen receptor into biochemical complexes that were insoluble in sodium dodecyl sulfate, a finding consistent with protein aggregation. Overall, our results shed light on the role of androgens as modifiers of ALS pathogenesis via dysregulation of androgen receptor homeostasis.
Collapse
|
42
|
Chaves EA, Fortunato RS, Carvalho DP, Nascimento JHM, Oliveira MF. Exercise-induced cardioprotection is impaired by anabolic steroid treatment through a redox-dependent mechanism. J Steroid Biochem Mol Biol 2013; 138:267-72. [PMID: 23831356 DOI: 10.1016/j.jsbmb.2013.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 06/13/2013] [Accepted: 06/15/2013] [Indexed: 10/26/2022]
Abstract
High doses of anabolic androgenic steroids (AAS) impair the cardioprotective effects of exercise against ischemia/reperfusion (I/R) insult, possibly through cellular redox imbalance. Here, the effect of nandrolone decanoate (DECA) treatment on heart redox metabolism was investigated during I/R in sedentary and exercised rats. DECA treatment significantly reduced superoxide dismutase and glutathione reductase activities in exercised rats after heart reperfusion. Catalase and glutathione peroxidase activities were not affected by DECA in both sedentary and trained rats, regardless the I/R period. DECA also induced myocardial oxidative stress, as evidenced by the reduced levels of total reduced thiols after heart reperfusion in exercised rats treated with the anabolic steroid. These results indicate that cardiotoxic effects of supraphysiological doses of AAS involve reduced heart antioxidant capacity.
Collapse
Affiliation(s)
- Elen A Chaves
- Laboratório de Eletrofisiologia Cardíaca Antonio Paes de Carvalho, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro, Brazil; Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro, Brazil; Laboratório de Bioquímica de Resposta ao Estresse, Instituto de Bioquímica Médica, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro, Brazil; Laboratório de Inflamação e Metabolismo, Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem (INBEB), Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | | | | |
Collapse
|
43
|
Hassan AF, Kamal MM. Effect of exercise training and anabolic androgenic steroids on hemodynamics, glycogen content, angiogenesis and apoptosis of cardiac muscle in adult male rats. Int J Health Sci (Qassim) 2013; 7:47-60. [PMID: 23559905 DOI: 10.12816/0006020] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVES To investigate the effects of exercise training and anabolic androgenic steroids (AAS) on hemodynamics, glycogen content, angiogenesis, apoptosis and histology of cardiac muscle. METHODS Forty rats were divided into 4 groups; control, steroid, exercise-trained and exercise-trained plus steroid groups. The exercise-trained and trained plus steroid groups, after one week of water adaptation, were exercised by jumping into water for 5 weeks. The steroid and trained plus steroid groups received nandrolone decanoate, for 5 weeks. Systolic blood pressure and heart rate (HR) were monitored weekly. Heart weight/body weight ratio (HW/BW ratio) were determined. Serum testosterone, vascular endothelial growth factor (VEGF), cardiac caspase-3 activity and glycogen content were measured. RESULTS Compared with control, the steroid group had significantly higher blood pressure, HR, sympathetic nerve activity, testosterone level, HW/BW and cardiac caspase-3 activity. Histological examination revealed apoptotic changes and hypertrophy of cardiomyocytes. In exercise-trained group, cardiac glycogen, VEGF and testosterone levels were significantly higher while HR was significantly lower than control. HW/BW was more than control confirmed by hypertrophy of cardiomyocytes with angiogenesis on histological examination. Trained plus steroid group, had no change in HR, with higher blood pressure and HW/BW than control, cardiac glycogen and serum VEGF were higher than control but lower than exercise-trained group. Histological examination showed hypertrophy of cardiomyoctes with mild angiogenesis rather than apoptosis. CONCLUSION When exercise is augmented with AAS, exercise-associated cardiac benefits may not be fully gained with potential cardiac risk from AAS if used alone or combined with exercise.
Collapse
Affiliation(s)
- Asmaa F Hassan
- Department of Medical Physiology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | | |
Collapse
|
44
|
Marques Neto SR, Silva ADH, Santos MCPD, Ferraz EF, Nascimento JHM. The blockade of angiotensin AT1 and aldosterone receptors protects rats from synthetic androgen-induced cardiac autonomic dysfunction. Acta Physiol (Oxf) 2013; 208:166-71. [PMID: 23279762 DOI: 10.1111/apha.12056] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 10/15/2012] [Accepted: 12/15/2012] [Indexed: 11/29/2022]
Abstract
AIM This study aimed to evaluate the combined effects of exercise and antagonists of the angiotensin II and aldosterone receptors on cardiac autonomic regulation and ventricular repolarization in rats chronically treated with nandrolone decanoate (ND), a synthetic androgen. METHODS Thirty male Wistar rats were divided into six groups: sedentary, trained, ND-treated, trained and ND-treated, trained and treated with both ND and spironolactone, and trained and treated with both ND and losartan. ND (10 mg kg(-1) weekly) and the antagonists (20 mg kg(-1) daily) of the angiotensin II AT1 (losartan) and aldosterone (spironolactone) receptors were administered for 8 weeks. Exercise training was performed using a treadmill five times each week for 8 weeks. Following this 8-week training and treatment period, electrocardiogram recordings were obtained to determine the time and frequency domains of heart rate variability (HRV) and corrected QT interval (QTc). RESULTS Nandrolone decanoate treatment increased the QTc interval and reduced the parasympathetic indexes of HRV (RMSSD, pNN5 and high-frequency power) in sedentary and trained rats. The ratio between low- and high-frequency power (LF/HF) was higher in ND-treated groups. Both losartan and spironolactone treatments prevented the effects of ND on the QTc interval and the HRV parameters (RMSSD, pNN5, high-frequency power, and the LF/HF ratio). CONCLUSION Our results show that chronic treatment with a high dose of ND induces cardiac parasympathetic dysfunction and disturbances in ventricular repolarization in both sedentary and exercised rats. Furthermore, inhibiting the renin-angiotensin-aldosterone system using losartan, or spironolactone, prevented these deleterious effects.
Collapse
Affiliation(s)
| | - A. da H. Silva
- Instituto de Biofisica Carlos Chagas Filho; Universidade Federal do Rio de Janeiro; Rio de Janeiro; Brazil
| | - M. C. P. dos Santos
- Instituto de Biofisica Carlos Chagas Filho; Universidade Federal do Rio de Janeiro; Rio de Janeiro; Brazil
| | - E. F. Ferraz
- Instituto de Biofisica Carlos Chagas Filho; Universidade Federal do Rio de Janeiro; Rio de Janeiro; Brazil
| | - J. H. M. Nascimento
- Instituto de Biofisica Carlos Chagas Filho; Universidade Federal do Rio de Janeiro; Rio de Janeiro; Brazil
| |
Collapse
|
45
|
Kaabia Z, Dervilly-Pinel G, Hanganu F, Cesbron N, Bichon E, Popot M, Bonnaire Y, Le Bizec B. Ultra high performance liquid chromatography/tandem mass spectrometry based identification of steroid esters in serum and plasma: An efficient strategy to detect natural steroids abuse in breeding and racing animals. J Chromatogr A 2013; 1284:126-40. [DOI: 10.1016/j.chroma.2013.02.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 01/31/2013] [Accepted: 02/05/2013] [Indexed: 11/16/2022]
|
46
|
Cappello V, Vezzoli E, Righi M, Fossati M, Mariotti R, Crespi A, Patruno M, Bentivoglio M, Pietrini G, Francolini M. Analysis of neuromuscular junctions and effects of anabolic steroid administration in the SOD1G93A mouse model of ALS. Mol Cell Neurosci 2012; 51:12-21. [PMID: 22800606 DOI: 10.1016/j.mcn.2012.07.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 07/06/2012] [Accepted: 07/08/2012] [Indexed: 12/11/2022] Open
Abstract
Several lines of evidence indicate that neuromuscular junction (NMJ) destruction and disassembly is an early phenomenon in amyotrophic lateral sclerosis (ALS). Here we analyzed by confocal and electron microscopy the NMJ structure in the diaphragm of SOD1G93A mice at symptom onset. In these mice, which provide a model for familial ALS, diaphragm denervation (~50%) as well as gastrocnemius denervation (~40%) was found. In addition, the size of the synaptic vesicle pool was reduced and alterations of mitochondria were observed in approximately 40% of the remaining presynaptic terminals. Chronic treatment of SOD1G93A mice with the anabolic steroid nandrolone during the presymptomatic stage preserved the diaphragm muscle mass and features indicative of synaptic activity. These features were represented by the number of vesicles docked within 200 nm from the presynaptic membrane and area of acetylcholine receptor clusters. Structural preservation of mitochondria was documented in presynaptic terminals. However, innervation of diaphragm muscle fibers was only slightly increased in nandrolone-treated SOD1-mutant mice. Altogether the results point out and define fine structural alterations of diaphragm NMJs in the murine model of familial ALS at symptom onset, and indicate that nandrolone may prevent or delay structural alterations in NMJ mitochondria and stimulate presynaptic activity but does not prevent muscle denervation during the disease.
Collapse
Affiliation(s)
- Valentina Cappello
- Dept. of Medical Biotechnology and Translational Medicine, University of Milan, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Golestani R, Slart RHJA, Dullaart RPF, Glaudemans AWJM, Zeebregts CJ, Boersma HH, Tio RA, Dierckx RAJO. Adverse cardiovascular effects of anabolic steroids: pathophysiology imaging. Eur J Clin Invest 2012; 42:795-803. [PMID: 22299602 DOI: 10.1111/j.1365-2362.2011.02642.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Anabolic-androgenic steroids (AAS) are widely abused for enhancing muscle mass, strength, growth and improving athletic performance. MATERIALS AND METHODS In recent years, many observational and interventional studies have shown important adverse cardiovascular effects of AAS abuse. CONCLUSIONS This review discusses established and future perspectives of novel molecular imaging techniques that may serve as potential tools for early detection of AAS-associated cardiovascular disorders.
Collapse
Affiliation(s)
- Reza Golestani
- Departments of Nuclear Medicine and Molecular Imaging Endocrinology, Division of Vascular Surgery, Department of University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Oberlander JG, Penatti CAA, Porter DM, Henderson LP. The Buzz about anabolic androgenic steroids: electrophysiological effects in excitable tissues. Neuroendocrinology 2012; 96:141-51. [PMID: 22576754 PMCID: PMC3488447 DOI: 10.1159/000339123] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 04/23/2012] [Indexed: 01/04/2023]
Abstract
Anabolic androgenic steroids (AAS) comprise a large and growing class of synthetic androgens used clinically to promote tissue-building in individuals suffering from genetic disorders, injuries, and diseases. Despite these beneficial therapeutic applications, the predominant use of AAS is illicit: these steroids are self-administered to promote athletic performance and body image. Hand in hand with the desired anabolic actions of the AAS are untoward effects on the brain and behavior. While the signaling routes by which the AAS impose both beneficial and harmful actions may be quite diverse, key endpoints are likely to include ligand-gated and voltage-dependent ion channels that govern the activity of electrically excitable tissues. Here, we review the known effects of AAS on molecular targets that play critical roles in controlling electrical activity, with a specific focus on the effects of AAS on neurotransmission mediated by GABA(A) receptors in the central nervous system.
Collapse
Affiliation(s)
- Joseph G. Oberlander
- Department of Physiology and Neurobiology, Dartmouth Medical School, Hanover, NH 03755 USA
| | - Carlos A. A. Penatti
- Departamento de Ciências Médicas, Universidade Nove de Julho - UNINOVE, São Paulo, SP 01504-000 Brasil
| | - Donna M. Porter
- Department of Physiology and Neurobiology, Dartmouth Medical School, Hanover, NH 03755 USA
| | - Leslie P. Henderson
- Department of Physiology and Neurobiology, Dartmouth Medical School, Hanover, NH 03755 USA
- To Whom Correspondence Should be Addressed:
| |
Collapse
|
49
|
Maior AS, Menezes P, Pedrosa RC, Carvalho DP, Soares PP, Nascimento JHM. Abnormal cardiac repolarization in anabolic androgenic steroid users carrying out submaximal exercise testing. Clin Exp Pharmacol Physiol 2011; 37:1129-33. [PMID: 20880189 DOI: 10.1111/j.1440-1681.2010.05452.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
1. The aim of the present study was to investigate the cardiovascular effects of anabolic androgenic steroid (AAS) abuse by comparing the electrocardiographic parameters before and after submaximal exercise between AAS users and non-AAS users. 2. A total of 22 men who regularly engaged in both resistance and aerobic exercise at fitness academies volunteered for the study (control group: n = 11, age 25 ± 4 years; AAS group: n = 11, age 27 ± 5 years). All subjects were submitted to submaximal exercise testing using an Astrand-Rhyming protocol. Heart rate and electrocardiography parameters were measured at rest and at the third minute of the post-exercise recovery time. 3. AAS users presented higher QTc and QTd at rest (10% and 55%, respectively) and at the post-exercise period (17% and 43%, respectively), compared with control subjects. The maximal and minimum QTc interval of the AAS group was significantly prolonged at the post-exercise period (12% and 15%, respectively). The haemodynamic parameters were similar in both groups (P > 0.05). The AAS group showed a lower heart rate recovery at the first minute after the test (P = 0.0001), and a higher exertion score (P < 0.0001) at a lower workload, compared with the control group. 4. Our results show that the QTc interval and dispersion are increased in individuals who abuse AAS, suggesting the presence of ventricular repolarization abnormalities that could potentially increase the risk of cardiac arrhythmias and sudden cardiac death.
Collapse
Affiliation(s)
- Alex S Maior
- Cardiac Electrophysiology Laboratory, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
During the past few years the results from molecular biological, biochemical, chemical, physical and theoretical approaches expanded the knowledge about metallo-beta-lactamases considerably. The main reason for the attracted interest is a persisting medical problem. Bacteria expressing metallo-beta-lactamases can be resistant to treatment with all the known beta-lactam antibiotics, and they are additionally invulnerable to combined treatment with inhibitors for the wider-spread serine-beta-lactamases. However, clinically useful inhibitors for metallo-beta-lactamases are not yet available. In spite of the rapidly expanding knowledge base a central question is still controversially discussed: is it the mononuclear, the binuclear or the metal-free state which might serve as the physiologically relevant target for inhibitor design? A summary of the present views of the roles and coordination geometries of metal ion(s) in metallo-beta-lactamases is combined with a discussion of the possibly variable metal ion content under physiological conditions.
Collapse
Affiliation(s)
- U Heinz
- Department of Natural Sciences, The Royal Veterinary and Agricultural University, 1871, Frederiksberg C, Denmark.
| | | |
Collapse
|