1
|
Du X, Feng X, Ming Z, Kiriazis H. Cardiomyopathy characterizing and heart failure risk predicting by echocardiography and pathoanatomy in aged male mice. Physiol Rep 2024; 12:e70061. [PMID: 39411804 PMCID: PMC11480644 DOI: 10.14814/phy2.70061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 10/20/2024] Open
Abstract
Correlation between echocardiographic and pathoanatomic variables and their prognostic value in murine cardiomyopathy models remain unknown. Using echocardiography, morphometrics, and survival monitoring, we characterized transgenic (TG) mice with dilated cardiomyopathy due to cardiac overexpression of β2-adrenoceptors focusing on predicting heart failure (HF) risk and HF mortality. In 12-month-old non-TG and TG mice, echocardiography was performed to determine left ventricular (LV) dimensions (d), wall thickness (h), and fractional shortening (FS). Animals were monitored for 3 months for survival. Organ weights and pathological events indicating left HF were determined. TG mice (n = 76) had reduced FS and enlarged LV, and 79% died of HF or likely arrhythmias during the follow-up period while all non-TG mice (n = 26) survived. These mice with left HF also had pulmonary congestion and hypertrophy/dilatation of the right ventricle (RV). Weights of lungs, RV, and atria were intercorrelated (r = 0.79-0.83) and also negatively correlated with FS × (h/d) index (r = -0.502 to -0.609). By FS × (h/d) tertiles, TG mice of low tertiles were identified with the highest mortality (96%) largely due to HF (76%). In conclusion, in aged cardiomyopathy mice a good correlation existed between echocardiographic and pathoanatomic variables. Echocardiography-derived LV function and remodeling were useful in identifying a subgroup of TG mice with a high risk of HF and HF fatality.
Collapse
Affiliation(s)
- Xiao‐Jun Du
- Department of Physiology and Pathophysiology, School of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'anShaanxiChina
- Experimental CardiologyBaker Heart and Diabetes InstituteMelbourneVictoriaAustralia
| | - Xin‐Heng Feng
- Experimental CardiologyBaker Heart and Diabetes InstituteMelbourneVictoriaAustralia
- Present address:
Department of Cardiology and Institute of Vascular MedicinePeking University Third HospitalBeijingChina
| | - Zi‐Qiu Ming
- Experimental CardiologyBaker Heart and Diabetes InstituteMelbourneVictoriaAustralia
- Present address:
Dandenong NeurologyDandenongVictoriaAustralia
| | - Helen Kiriazis
- Experimental CardiologyBaker Heart and Diabetes InstituteMelbourneVictoriaAustralia
| |
Collapse
|
2
|
Lagunas-Rangel FA. G protein-coupled receptors that influence lifespan of human and animal models. Biogerontology 2021; 23:1-19. [PMID: 34860303 PMCID: PMC8888397 DOI: 10.1007/s10522-021-09945-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/22/2021] [Indexed: 12/16/2022]
Abstract
Humanity has always sought to live longer and for this, multiple strategies have been tried with varying results. In this sense, G protein-coupled receptors (GPCRs) may be a good option to try to prolong our life while maintaining good health since they have a substantial participation in a wide variety of processes of human pathophysiology and are one of the main therapeutic targets. In this way, we present the analysis of a series of GPCRs whose activity has been shown to affect the lifespan of animal and human models, and in which we put a special interest in describing the molecular mechanisms involved. Our compilation of data revealed that the mechanisms most involved in the role of GPCRs in lifespan are those that mimic dietary restriction, those related to insulin signaling and the AMPK and TOR pathways, and those that alter oxidative homeostasis and severe and/or chronic inflammation. We also discuss the possibility of using agonist or antagonist drugs, depending on the beneficial or harmful effects of each GPCR, in order to prolong people's lifespan and healthspan.
Collapse
|
3
|
Wang C, Gaspari TA, Ferens D, Spizzo I, Kemp-Harper BK, Samuel CS. Simultaneous targeting of oxidative stress and fibrosis reverses cardiomyopathy-induced ventricular remodelling and dysfunction. Br J Pharmacol 2021; 178:2424-2442. [PMID: 33660265 DOI: 10.1111/bph.15428] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 02/14/2021] [Accepted: 02/24/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Oxidative stress and fibrosis are hallmarks of cardiomyopathy-induced heart failure yet are not effectively targeted by current frontline therapies. Here, the therapeutic effects of the anti-oxidant, N-acetylcysteine (NAC), were compared and combined with an acute heart failure drug with established anti-fibrotic effects, serelaxin (RLX), in a murine model of cardiomyopathy. EXPERIMENTAL APPROACH Adult male 129sv mice were subjected to repeated isoprenaline (25 mg·kg-1 )-induced cardiac injury for five consecutive days and then left to undergo fibrotic healing until Day 14. Subgroups of isoprenaline-injured mice were treated with RLX (0.5 mg·kg-1 ·day-1 ), NAC (25 mg·kg-1 ·day-1 ) or both combined, given subcutaneously via osmotic minipumps from Day 7 to 14. Control mice received saline instead of isoprenaline. KEY RESULTS Isoprenaline-injured mice showed increased left ventricular (LV) inflammation (~5-fold), oxidative stress (~1-2.5-fold), cardiomyocyte hypertrophy (~25%), cardiac remodelling, fibrosis (~2-2.5-fold) and dysfunction by Day 14 after injury. NAC alone blocked the cardiomyopathy-induced increase in LV superoxide levels, to a greater extent than RLX. Additionally, either treatment alone only partly reduced several measures of LV inflammation, remodelling and fibrosis. In comparison, the combination of RLX and NAC prevented the cardiomyopathy-induced LV macrophage infiltration, remodelling, fibrosis and cardiomyocyte size, to a greater extent than either treatment alone after 7 days. The combination therapy also restored the isoprenaline-induced reduction in LV function, without affecting systolic BP. CONCLUSION AND IMPLICATIONS These findings demonstrated that the simultaneous targeting of oxidative stress and fibrosis is key to treating the pathophysiology and dysfunction induced by cardiomyopathy.
Collapse
Affiliation(s)
- Chao Wang
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Tracey A Gaspari
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Dorota Ferens
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Iresha Spizzo
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Barbara K Kemp-Harper
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia.,Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
4
|
In Experimental Dilated Cardiomyopathy Heart Failure and Survival Are Adversely Affected by a Lack of Sexual Interactions. Int J Mol Sci 2020; 21:ijms21155450. [PMID: 32751757 PMCID: PMC7432836 DOI: 10.3390/ijms21155450] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/28/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
Nearly one in three people in the U.S. will develop heart failure (HF), characterized by fluid retention (edema) in the lungs and elsewhere. This leads to difficult breathing, deterioration of physical capacity, restriction of normal activities and death. There is little data about the safety and effects of sexual interactions in patients with HF. We tested whether a lack of sexual interactions affected pathophysiological outcomes in a pre-clinical mouse model of dilated cardiomyopathy that recapitulates the progressive stages of human HF. Male mice were randomly given access to, or deprived from, sexual interactions with female mice, which were confirmed by videography and generation of offspring. Cohousing with access to sexual interactions markedly prolonged survival, while cohousing without access to sexual activity did not. Sexual interactions improved systolic function, reduced HF-associated edema, altered transcription of heart contractile protein genes and decreased plasma testosterone levels. To determine whether testosterone levels contributed to survival, testosterone levels were experimentally reduced. Reduction of testosterone levels significantly prolonged survival. Taken together, in mice with dilated cardiomyopathy, sexual activity altered cardiac contractile gene transcription, improved systolic function, reduced edema and prolonged survival which may be in part due to lower testosterone levels.
Collapse
|
5
|
Gozalo AS, Zerfas PM, Elkins WR, Gieseck RL. Retrospective Study of Intercalated Disk Defects Associated with Dilated Cardiomyopathy, Atrial Thrombosis, and Heart Failure in BALB/c Mice Deficient in IL4 Receptor α. Comp Med 2020; 70:266-276. [PMID: 32384942 PMCID: PMC7287387 DOI: 10.30802/aalas-cm-19-000059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/01/2019] [Accepted: 10/22/2019] [Indexed: 12/21/2022]
Abstract
An increased incidence of dilated cardiomyopathy and atrial thrombosis was noted in a breeding colony of BALB/c mice deficient in IL4 receptor α. The condition affected mice of both sexes and of various ages, and extensive testing (microbiology, serology, histopathology) failed to ascertain the cause. Transmission electron microscopy of heart samples showed structural defects in the myocardial intercalated disks, characterized by unorganized and heavily convoluted arrangement with lower density and less prominent desmosomes and adherens junctions, widening of the intercellular space, myofibrillar lysis adjacent to intercalated disks, occasional sarcomere lysis with marked myofiber degeneration, vacuolation, accumulation of cell debris, and myelin figures. The intercalated disk contains cell adhesion molecules that form cell junctions, allowing contraction coupling of cardiomyocytes and the electrical and mechanical connection between cardiac fibers. Thus, defects at this level result in poor myocardial contraction, intracardiac blood stagnation, and consequently cardiac dilation with clinical signs of heart failure. The background strain or, potentially, the Cre-loxP-mediated recombination system used to create these mice may have contributed to the elevated incidence of cardiomyopathy and atrial thrombosis in this colony. Due to the backcrossing breeding scheme used, we cannot discount the emergence and colonywide dissemination of a spontaneous mutation that affects the intercalated disk. This report underscores the importance of carefully monitoring genetically modified mice colonies for unexpected phenotypes that may result from spontaneous or unintended mutations or enhanced strain background pathology.
Collapse
Affiliation(s)
- Alfonso S Gozalo
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland;,
| | - Patricia M Zerfas
- Pathology Service, Office of Research Services, National Institutes of Health, Bethesda, Maryland
| | - William R Elkins
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Richard L Gieseck
- Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
6
|
Du X, Zhao W, Nguyen M, Lu Q, Kiriazis H. β-Adrenoceptor activation affects galectin-3 as a biomarker and therapeutic target in heart disease. Br J Pharmacol 2019; 176:2449-2464. [PMID: 30756388 PMCID: PMC6592856 DOI: 10.1111/bph.14620] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/11/2018] [Accepted: 01/29/2019] [Indexed: 12/12/2022] Open
Abstract
Myocardial fibrosis is a key histopathological component that drives the progression of heart disease leading to heart failure and constitutes a therapeutic target. Recent preclinical and clinical studies have implicated galectin-3 (Gal-3) as a pro-fibrotic molecule and a biomarker of heart disease and fibrosis. However, our knowledge is poor on the mechanism(s) that determine the blood level or regulate cardiac expression of Gal-3. Recent studies have demonstrated that enhanced β-adrenoceptor activity is a determinant of both circulating concentration and cardiac expression of Gal-3. Pharmacological or transgenic activation of β-adrenoceptors leads to increased blood levels of Gal-3 and up-regulated cardiac Gal-3 expression, effect that can be reversed with the use of β-adrenoceptor antagonists. Conversely, Gal-3 gene deletion confers protection against isoprenaline-induced cardiotoxicity and fibrogenesis. At the transcription level, β-adrenoceptor stimulation activates cardiac mammalian sterile-20-like kinase 1, a pivotal kinase of the Hippo signalling pathway, which is associated with Gal-3 up-regulation. Recent studies have suggested a role for the β-adrenoceptor-Hippo signalling pathway in the regulation of cardiac Gal-3 expression thereby contributing to the onset and progression of heart disease. This implies a therapeutic potential of the suppression of Gal-3 expression. In this review, we discuss the effects of β-adrenoceptor activity on Gal-3 as a biomarker and causative mediator in the setting of heart disease and point out pivotal knowledge gaps. LINKED ARTICLES: This article is part of a themed section on Adrenoceptors-New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc.
Collapse
Affiliation(s)
- Xiao‐Jun Du
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
- Department of Physiology and Pathophysiology, School of Medical SciencesXi'an Jiaotong University Health Science CenterXi'anChina
| | - Wei‐Bo Zhao
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
| | - My‐Nhan Nguyen
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
| | - Qun Lu
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
- Department of Cardiovascular Medicine, First HospitalXi'an Jiaotong University Health Science CenterXi'anChina
| | - Helen Kiriazis
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
| |
Collapse
|
7
|
Aimo A, Vergaro G, Castiglione V, Barison A, Pasanisi E, Petersen C, Chubuchny V, Giannoni A, Poletti R, Maffei S, Januzzi JL, Passino C, Emdin M. Effect of Sex on Reverse Remodeling in Chronic Systolic Heart Failure. JACC-HEART FAILURE 2018; 5:735-742. [PMID: 28958348 DOI: 10.1016/j.jchf.2017.07.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 06/13/2017] [Accepted: 07/27/2017] [Indexed: 01/15/2023]
Abstract
OBJECTIVES This study sought to investigate sex-related differences in reverse remodeling (RR). BACKGROUND RR, that is, the recovery from left ventricular (LV) dilation and dysfunction in response to treatment for heart failure (HF), is associated with improved prognosis. METHODS Data from patients with stable systolic HF (LV ejection fraction [LVEF] of <50%) undergoing 2 transthoracic echocardiograms within 12 ± 2 months were analyzed. Reverse remodeling was defined as a ≥15% reduction in LV end-systolic volume index. RESULTS A total of 927 patients were evaluated (68 ± 12 years; median LVEF = 35% [interquartile range: 30% to 43%]; 27% women). Ischemic HF was less often encountered in women (33% vs. 60%, respectively; p < 0.001), whereas most characteristics did not differ with regard to sex. Women showed a higher incidence of RR (41% vs. 27%, respectively; p < 0.001), despite similar baseline LV volume and function. RR was more frequent among women in the subgroups with either ischemic or nonischemic HF, as well as in all categories of systolic dysfunction (LVEF ≤35% or >35%, according to current indication for device implantation, and LVEF <40% or 40% to 50% according to the definition of HF with reduced or mid-range EF). In the whole population, female sex was an independent predictor of RR (hazard ratio: 1.54; 95% confidence interval: 1.11 to 2.14; p = 0.011), together with cause of HF, disease duration, and left bundle branch block. Female sex was again an independent predictor of RR in all LVEF categories. CONCLUSIONS Reverse remodeling is more frequent among women, regardless of cause and severity of LV dysfunction. Female sex is an independent predictor of RR in all categories of LV systolic dysfunction.
Collapse
Affiliation(s)
- Alberto Aimo
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Giuseppe Vergaro
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | | | - Andrea Barison
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Emilio Pasanisi
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Christina Petersen
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | | | - Alberto Giannoni
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Roberta Poletti
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Silvia Maffei
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - James L Januzzi
- Cardiology Division, Massachusetts General Hospital and Harvard Clinical Research Institute, Boston, Massachusetts
| | - Claudio Passino
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Michele Emdin
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.
| |
Collapse
|
8
|
Nguyen MN, Su Y, Kiriazis H, Yang Y, Gao XM, McMullen JR, Dart AM, Du XJ. Upregulated galectin-3 is not a critical disease mediator of cardiomyopathy induced by β2-adrenoceptor overexpression. Am J Physiol Heart Circ Physiol 2018; 314:H1169-H1178. [DOI: 10.1152/ajpheart.00337.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Preclinical studies have demonstrated that anti-galectin-3 (Gal-3) interventions are effective in attenuating cardiac remodeling, fibrosis, and dysfunction. We determined, in a transgenic (TG) mouse model of fibrotic cardiomyopathy, whether Gal-3 expression was elevated and whether Gal-3 played a critical role in disease development. We studied mice with fibrotic cardiomyopathy attributable to cardiac overexpression of human β2-adrenoceptors (β2-TG). Cardiac expression levels of Gal-3 and fibrotic or inflammatory genes were determined. The effect of Gal-3 inhibition in β2-TG mice was studied by treatment with Gal-3 inhibitors ( N-acetyllactosamine and modified citrus pectin) or by deletion of Gal-3 through crossing β2-TG and Gal-3 knockout mice. Changes in cardiomyopathy phenotypes were assessed by echocardiography and biochemical assays. In β2-TG mice at 3, 6, and 9 mo of age, upregulation of Gal-3 expression was observed at mRNA (~6- to 15-fold) and protein (~4- to 8-fold) levels. Treatment of β2-TG mice with N-acetyllactosamine (3 wk) or modified citrus pectin (3 mo) did not reverse cardiac fibrosis, inflammation, and cardiomyopathy. Similarly, Gal-3 gene deletion in β2-TG mice aged 3 and 9 mo did not rescue the cardiomyopathy phenotype. In conclusion, the β2-TG model of cardiomyopathy showed a robust upregulation of Gal-3 that correlated with disease severity, but Gal-3 inhibitors or Gal-3 gene deletion had no effect in halting myocardial fibrosis, remodeling, and dysfunction. Gal-3 may not be critical for cardiac fibrogenesis and remodeling in this cardiomyopathy model. NEW & NOTEWORTHY We showed a robust upregulation of cardiac galectin-3 (Gal-3) expression in a mouse model of cardiomyopathy attributable to cardiomyocyte-restricted transgenic activation of β2-adrenoceptors. However, pharmacological and genetic inhibition of Gal-3 did not confer benefit in this model, implying that Gal-3 may not be a critical disease mediator of cardiac remodeling in this model.
Collapse
Affiliation(s)
- My-Nhan Nguyen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Yidan Su
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Yan Yang
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Alfred Heart Centre, The Alfred Hospital, Melbourne, Victoria, Australia
| | - Xiao-Ming Gao
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Julie R. McMullen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Anthony M. Dart
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Alfred Heart Centre, The Alfred Hospital, Melbourne, Victoria, Australia
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
9
|
Prevention of βeta-adrenergic-induced Adverse Cardiac Remodeling by Gonadectomy in Male but Not Female Spontaneously Hypertensive Rats. J Cardiovasc Pharmacol 2018; 70:202-209. [PMID: 28590262 DOI: 10.1097/fjc.0000000000000506] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Chronic β-adrenergic stimulation induces left ventricular (LV) remodeling in male but not in female spontaneously hypertensive rats (SHRs). However, the role of sex steroids in mediating these effects has not been determined. The aim of the present study was to assess the impact of gonadectomy on isoproterenol (ISO)-induced LV remodeling in SHR. Gonadectomy was performed on 9-month-old male and female SHR. LV remodeling was induced by daily ISO injection (0.04 mg/kg) for 6 months. LV dimensions and functions were determined in vivo by echocardiography and ex vivo using isolated perfused heart preparations. In males, ISO increased LV end diastolic (LVED) diameter in sham-operated (in millimeter, ISO: 8.12 ± 0.26 vs. Con: 6.67 ± 0.20, P = 0.0002) but not in castrated SHR (ISO: 6.97 ± 0.31 vs. Con: 6.53 ± 0.15, P = 0.66). Similarly, ISO increased the volume intercept of the LVED pressure-volume relationship in sham-operated (in milliliters, ISO: 0.26 ± 0.02 vs. Con: 0.19 ± 0.01, P = 0.01) but not in castrated SHR (ISO: 0.17 ± 0.02 vs. Con: 0.17 ± 0.01, P = 0.99). In females, ISO only increased LVED diameter (ISO: 6.43 ± 0.13 vs. Con: 6.07 ± 0.09, P = 0.027). However, ovariectomy did not modify any LV dimensions measured in vivo and ex vivo. In conclusion, testosterone may be responsible for the chronic β-adrenergic-induced LV dilation and eccentric remodeling observed in male but not female SHR.
Collapse
|
10
|
Enhanced heart failure, mortality and renin activation in female mice with experimental dilated cardiomyopathy. PLoS One 2017; 12:e0189315. [PMID: 29240788 PMCID: PMC5730114 DOI: 10.1371/journal.pone.0189315] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 10/08/2017] [Indexed: 01/06/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is the major cause of heart failure affecting both women and men. Limited clinical studies show conflicting data in sex-related differences in the progression of dilated cardiomyopathy and heart failure (HF) outcomes. We examined the comparative sex-related progression of cardiomyopathy and the development of HF (at 4, 7, 13 weeks of age) in a well-established, transgenic mouse model of DCM that recapitulates the progressive stages of human HF. By 13 weeks of age, female mice with DCM had more severe left ventricular systolic dysfunction, left ventricular dilation and wall thinning (P<0.001 for all) than age-matched male mice with DCM. Female mice also had greater lung edema (P<0.001), cardiac fibrosis (P<0.01) and pleural effusions, which were not rescued by ovariectomy. By comparison to DCM male mice at 13 weeks, these pathological changes in female mice with DCM, were associated with significant increases in plasma active renin (P<0.01), angiotensin II (P<0.01) and aldosterone levels (P<0.001). In comparison to DCM male mice, DCM female mice also showed differential expression of the natriuretic peptide system with lower corin and higher ANP, BNP and cGMP levels at 13 weeks of age. We conclude, that female mice with experimental DCM have an accelerated progression of cardiomyopathy and HF, which was not corrected by early ovariectomy. These alterations are associated with early renin activation with increased angiotensin II and aldosterone levels, and altered expression of the natriuretic peptide system.
Collapse
|
11
|
Nishiuchi S, Makiyama T, Aiba T, Nakajima K, Hirose S, Kohjitani H, Yamamoto Y, Harita T, Hayano M, Wuriyanghai Y, Chen J, Sasaki K, Yagihara N, Ishikawa T, Onoue K, Murakoshi N, Watanabe I, Ohkubo K, Watanabe H, Ohno S, Doi T, Shizuta S, Minamino T, Saito Y, Oginosawa Y, Nogami A, Aonuma K, Kusano K, Makita N, Shimizu W, Horie M, Kimura T. Gene-Based Risk Stratification for Cardiac Disorders in
LMNA
Mutation Carriers. ACTA ACUST UNITED AC 2017; 10:CIRCGENETICS.116.001603. [DOI: 10.1161/circgenetics.116.001603] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 09/25/2017] [Indexed: 01/01/2023]
Abstract
Background—
Mutations in
LMNA
(
lamin A/C
), which encodes lamin A and C, typically cause age-dependent cardiac phenotypes, including dilated cardiomyopathy, cardiac conduction disturbance, atrial fibrillation, and malignant ventricular arrhythmias. Although the type of
LMNA
mutations have been reported to be associated with susceptibility to malignant ventricular arrhythmias, the gene-based risk stratification for cardiac complications remains unexplored.
Methods and Results—
The multicenter cohort included 77
LMNA
mutation carriers from 45 families; cardiac disorders were retrospectively analyzed. The mean age of patients when they underwent genetic testing was 45±17, and they were followed for a median 49 months. Of the 77 carriers, 71 (92%) were phenotypically affected and showed cardiac conduction disturbance (81%), low left ventricular ejection fraction (<50%; 45%), atrial arrhythmias (58%), and malignant ventricular arrhythmias (26%). During the follow-up period, 9 (12%) died, either from end-stage heart failure (n=7) or suddenly (n=2). Genetic analysis showed truncation mutations in 58 patients from 31 families and missense mutations in 19 patients from 14 families. The onset of cardiac disorders indicated that subjects with truncation mutations had an earlier occurrence of cardiac conduction disturbance and low left ventricular ejection fraction, than those with missense mutations. In addition, the truncation mutation was found to be a risk factor for the early onset of cardiac conduction disturbance and the occurrence of atrial arrhythmias and low left ventricular ejection fraction, as estimated using multivariable analyses.
Conclusions—
The truncation mutations were associated with manifestation of cardiac phenotypes in
LMNA
-related cardiomyopathy, suggesting that genetic analysis might be useful for diagnosis and risk stratification.
Collapse
|
12
|
Abstract
Myocardial injury, mechanical stress, neurohormonal activation, inflammation, and/or aging all lead to cardiac remodeling, which is responsible for cardiac dysfunction and arrhythmogenesis. Of the key histological components of cardiac remodeling, fibrosis either in the form of interstitial, patchy, or dense scars, constitutes a key histological substrate of arrhythmias. Here we discuss current research findings focusing on the role of fibrosis, in arrhythmogenesis. Numerous studies have convincingly shown that patchy or interstitial fibrosis interferes with myocardial electrophysiology by slowing down action potential propagation, initiating reentry, promoting after-depolarizations, and increasing ectopic automaticity. Meanwhile, there has been increasing appreciation of direct involvement of myofibroblasts, the activated form of fibroblasts, in arrhythmogenesis. Myofibroblasts undergo phenotypic changes with expression of gap-junctions and ion channels thereby forming direct electrical coupling with cardiomyocytes, which potentially results in profound disturbances of electrophysiology. There is strong evidence that systemic and regional inflammatory processes contribute to fibrogenesis (i.e., structural remodeling) and dysfunction of ion channels and Ca2+ homeostasis (i.e., electrical remodeling). Recognizing the pivotal role of fibrosis in the arrhythmogenesis has promoted clinical research on characterizing fibrosis by means of cardiac imaging or fibrosis biomarkers for clinical stratification of patients at higher risk of lethal arrhythmia, as well as preclinical research on the development of antifibrotic therapies. At the end of this review, we discuss remaining key questions in this area and propose new research approaches. © 2017 American Physiological Society. Compr Physiol 7:1009-1049, 2017.
Collapse
Affiliation(s)
- My-Nhan Nguyen
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Xiao-Ming Gao
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia
| |
Collapse
|
13
|
Regitz-Zagrosek V, Kararigas G. Mechanistic Pathways of Sex Differences in Cardiovascular Disease. Physiol Rev 2017; 97:1-37. [PMID: 27807199 DOI: 10.1152/physrev.00021.2015] [Citation(s) in RCA: 422] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Major differences between men and women exist in epidemiology, manifestation, pathophysiology, treatment, and outcome of cardiovascular diseases (CVD), such as coronary artery disease, pressure overload, hypertension, cardiomyopathy, and heart failure. Corresponding sex differences have been studied in a number of animal models, and mechanistic investigations have been undertaken to analyze the observed sex differences. We summarize the biological mechanisms of sex differences in CVD focusing on three main areas, i.e., genetic mechanisms, epigenetic mechanisms, as well as sex hormones and their receptors. We discuss relevant subtypes of sex hormone receptors, as well as genomic and nongenomic, activational and organizational effects of sex hormones. We describe the interaction of sex hormones with intracellular signaling relevant for cardiovascular cells and the cardiovascular system. Sex, sex hormones, and their receptors may affect a number of cellular processes by their synergistic action on multiple targets. We discuss in detail sex differences in organelle function and in biological processes. We conclude that there is a need for a more detailed understanding of sex differences and their underlying mechanisms, which holds the potential to design new drugs that target sex-specific cardiovascular mechanisms and affect phenotypes. The comparison of both sexes may lead to the identification of protective or maladaptive mechanisms in one sex that could serve as a novel therapeutic target in one sex or in both.
Collapse
Affiliation(s)
- Vera Regitz-Zagrosek
- Institute of Gender in Medicine & Center for Cardiovascular Research, Charite University Hospital, and DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Georgios Kararigas
- Institute of Gender in Medicine & Center for Cardiovascular Research, Charite University Hospital, and DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| |
Collapse
|
14
|
Michel FS, Magubane M, Mokotedi L, Norton GR, Woodiwiss AJ. Sex-Specific Effects of Adrenergic-Induced Left Ventricular Remodeling in Spontaneously Hypertensive Rats. J Card Fail 2016; 23:161-168. [PMID: 27663100 DOI: 10.1016/j.cardfail.2016.09.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 09/05/2016] [Accepted: 09/15/2016] [Indexed: 01/19/2023]
Abstract
OBJECTIVE The aim of this work was to determine whether adrenergic-induced left ventricular (LV) dilation and eccentric remodeling in pressure-overload hypertrophy is sex specific. METHODS AND RESULTS Chronic β-adrenoreceptor activation was produced in male and female spontaneously hypertensive rats (SHRs) by means of daily administration of isoproterenol (ISO; 0.04 mg/kg daily) from 9 to 15 months of age. LV chamber dimensions were determined in vivo by means of echocardiography and ex vivo in isolated perfused heart preparations. The acute hemodynamic response to ISO, the degree of myocardial necrosis and apoptosis, and collagen distribution were also assessed. Female SHRs demonstrated inotropic and chronotropic responses to ISO similarly to male SHRs. Compared with control subjects (saline solution vehicle), following chronic ISO administration, LV end-diastolic diameter (mm) was increased in male (ISO 7.8 ± 0.3 vs control 6.6 ± 0.2; P < .001) but not in female (ISO 6.3 ± 0.2 vs control 6.2 ± 0.2; P = .23) SHRs. Similarly, compared with control, ISO administration increased the volume intercept of the LV end-diastolic pressure-volume relation (mL) in male (ISO 0.31 ± 0.02 vs control 0.22 ± 0.01; P < .0001) but not in female (ISO 0.17 ± 0.01 vs control 0.17 ± 0.01; P = 1.00) SHRs. Relative wall thickness was also decreased in male SHRs receiving ISO but not in female SHRs receiving ISO. Chronic ISO administration increased the percentage of area covered by interstitial collagen in male but not in female SHRs. Finally, chronic adrenergic stimulation failed to influence LV chamber or myocardial systolic function in either male or female SHRs. CONCLUSIONS Male SHRs are more susceptible to adrenergic-induced LV dilation and eccentric LV remodeling than female SHRs. These effects are associated with increased collagen deposition. In pressure-overload hypertrophy, LV dilation and eccentric LV remodeling occur before LV dysfunction in male rats.
Collapse
Affiliation(s)
- Frederic S Michel
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - Mhlengi Magubane
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Lebogang Mokotedi
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Gavin R Norton
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Angela J Woodiwiss
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
15
|
Nguyen MN, Kiriazis H, Ruggiero D, Gao XM, Su Y, Jian A, Han LP, McMullen JR, Du XJ. Spontaneous ventricular tachyarrhythmias in β2-adrenoceptor transgenic mice in relation to cardiac interstitial fibrosis. Am J Physiol Heart Circ Physiol 2015; 309:H946-57. [PMID: 26116714 DOI: 10.1152/ajpheart.00405.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 06/26/2015] [Indexed: 12/21/2022]
Abstract
Myocardial fibrosis is regarded as a pivotal proarrhythmic substrate, but there have been no comprehensive studies showing a correlation between the severity of fibrosis and ventricular tachyarrhythmias (VTAs). Our purpose was to document this relationship in a transgenic (TG) strain of mice with fibrotic cardiomyopathy. TG mice with cardiac overexpression of β2-adrenoceptors (β2-AR mice) and non-TG (NTG) littermates were studied at 4-12 mo of age. VTA was quantified by ECG telemetry. The effect of pharmacological blockade of β2-ARs on VTA was examined. Myocardial collagen content was determined by hydroxyproline assay. NTG and TG mice displayed circadian variation in heart rate, which was higher in TG mice than in NTG mice (P <0.05). Frequent spontaneous ventricular ectopic beats (VEBs) and ventricular tachycardia (VT) were prominent in TG mice but not present in NTG mice. The frequency of VEB and VT episodes in TG mice increased with age (P < 0.01). Ventricular collagen content was greater in TG mice than in NTG mice (P <0.001) and correlated with age (r = 0.71, P < 0.01). The number of VEBs or VT episodes correlated with age (r = 0.83 and r = 0.73) and the content of total or cross-linked collagen (r = 0.62∼0.66, all P <0.01). While having no effect in younger β2-TG mice, β2-AR blockade reduced the frequency of VTA in old β2-TG mice with more severe fibrosis. In conclusion, β2-TG mice exhibit interstitial fibrosis and spontaneous onset of VTA, becoming more severe with aging. The extent of cardiac fibrosis is a major determinant for both the frequency of VTA and proarrhythmic action of β2-AR activation.
Collapse
Affiliation(s)
- My-Nhan Nguyen
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Helen Kiriazis
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Diego Ruggiero
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; University of Milan, Milan, Italy
| | - Xiao-Ming Gao
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Yidan Su
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Anne Jian
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Li-Ping Han
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; WenZhou Medical University, WenZhou, China; and
| | - Julie R McMullen
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Physiology, Monash University, Melbourne, Australia
| | - Xiao-Jun Du
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; Central Clinical School, Monash University, Melbourne, Victoria, Australia;
| |
Collapse
|
16
|
Xu Q, Jennings NL, Sim K, Chang L, Gao XM, Kiriazis H, Lee YY, Nguyen MN, Woodcock EA, Zhang YY, El-Osta A, Dart AM, Du XJ. Pathological hypertrophy reverses β2-adrenergic receptor-induced angiogenesis in mouse heart. Physiol Rep 2015; 3:3/3/e12340. [PMID: 25780088 PMCID: PMC4393171 DOI: 10.14814/phy2.12340] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
β-adrenergic activation and angiogenesis are pivotal for myocardial function but the link between both events remains unclear. The aim of this study was to explore the cardiac angiogenesis profile in a mouse model with cardiomyocyte-restricted overexpression of β2-adrenoceptors (β2-TG), and the effect of cardiac pressure overload. β2-TG mice had heightened cardiac angiogenesis, which was essential for maintenance of the hypercontractile phenotype seen in this model. Relative to controls, cardiomyocytes of β2-TGs showed upregulated expression of vascular endothelial growth factor (VEGF), heightened phosphorylation of cAMP-responsive-element-binding protein (CREB), and increased recruitment of phospho-CREB, CREB-binding protein (CBP), and p300 to the VEGF promoter. However, when hearts were subjected to pressure overload by transverse aortic constriction (TAC), angiogenic signaling in β2-TGs was inhibited within 1 week after TAC. β2-TG hearts, but not controls, exposed to pressure overload for 1–2 weeks showed significant increases from baseline in phosphorylation of Ca2+/calmodulin-dependent kinase II (CaMKIIδ) and protein expression of p53, reduction in CREB phosphorylation, and reduced abundance of phospho-CREB, p300 and CBP recruited to the CREB-responsive element (CRE) site of VEGF promoter. These changes were associated with reduction in both VEGF expression and capillary density. While non-TG mice with TAC developed compensatory hypertrophy, (2-TGs exhibited exaggerated hypertrophic growth at week-1 post-TAC, followed by LV dilatation and reduced fractional shortening measured by serial echocardiography. In conclusion, angiogenesis was enhanced by the cardiomyocyte (2AR/CREB/VEGF signaling pathway. Pressure overload rapidly inhibited this signaling, likely as a consequence of activated CaMKII and p53, leading to impaired angiogenesis and functional decompensation.
Collapse
Affiliation(s)
- Qi Xu
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Nicole L Jennings
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Kenneth Sim
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Lisa Chang
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Xiao-Ming Gao
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Helen Kiriazis
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Ying Ying Lee
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - My-Nhan Nguyen
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | - You-Yi Zhang
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing, China
| | - Assam El-Osta
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Anthony M Dart
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia Alfred Heart Centre, the Alfred Hospital, Melbourne, Victoria, Australia Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Xiao-Jun Du
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
17
|
Zwadlo C, Schmidtmann E, Szaroszyk M, Kattih B, Froese N, Hinz H, Schmitto JD, Widder J, Batkai S, Bähre H, Kaever V, Thum T, Bauersachs J, Heineke J. Antiandrogenic therapy with finasteride attenuates cardiac hypertrophy and left ventricular dysfunction. Circulation 2015; 131:1071-81. [PMID: 25632043 DOI: 10.1161/circulationaha.114.012066] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND In comparison with men, women have a better prognosis when experiencing aortic valve stenosis, hypertrophic cardiomyopathy, or heart failure. Recent data suggest that androgens like testosterone or the more potent dihydrotestosterone contribute to the development of cardiac hypertrophy and failure. Therefore, we analyzed whether antiandrogenic therapy with finasteride, which inhibits the generation of dihydrotestosterone by the enzyme 5-α-reductase, improves pathological ventricular remodeling and heart failure. METHODS AND RESULTS We found a strongly induced expression of all 3 isoforms of the 5-α-reductase (Srd5a1 to Srd5a3) in human and mouse hearts with pathological hypertrophy, which was associated with increased myocardial accumulation of dihydrotestosterone. Starting 1 week after the induction of pressure overload by transaortic constriction, mice were treated with finasteride for 2 weeks. Cardiac function, hypertrophy, dilation, and fibrosis were markedly improved in response to finasteride treatment in not only male, but also in female mice. In addition, finasteride also very effectively improved cardiac function and mortality after long-term pressure overload and prevented disease progression in cardiomyopathic mice with myocardial Gαq overexpression. Mechanistically, finasteride, by decreasing dihydrotestosterone, potently inhibited hypertrophy and Akt-dependent prohypertrophic signaling in isolated cardiac myocytes, whereas the introduction of constitutively active Akt blunted these effects of finasteride. CONCLUSIONS Finasteride, which is currently used in patients to treat prostate disease, potently reverses pathological cardiac hypertrophy and dysfunction in mice and might be a therapeutic option for heart failure.
Collapse
Affiliation(s)
- Carolin Zwadlo
- From Medizinische Hochschule Hannover, Klinik für Kardiologie und Angiologie, Hanover, Germany (C.Z., E.S., M.S., B.K., N.F., H.H., J.W., J.B., J.H.); Klinik für Herz-, Thorax-, Transplantations- und Gefäßchirurgie, Hanover, Germany (J.D.S.); Institut für Molekulare und Translationale Therapiestrategien (IMTTS), Hannover, Germany (S.B., T.T.); National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.); and Medizinische Hochschule Hannover, Zentrale Forschungseinrichtung Metabolomics, Institut für Pharmakologie, Hannover, Germany (H.B., V.K.)
| | - Elisa Schmidtmann
- From Medizinische Hochschule Hannover, Klinik für Kardiologie und Angiologie, Hanover, Germany (C.Z., E.S., M.S., B.K., N.F., H.H., J.W., J.B., J.H.); Klinik für Herz-, Thorax-, Transplantations- und Gefäßchirurgie, Hanover, Germany (J.D.S.); Institut für Molekulare und Translationale Therapiestrategien (IMTTS), Hannover, Germany (S.B., T.T.); National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.); and Medizinische Hochschule Hannover, Zentrale Forschungseinrichtung Metabolomics, Institut für Pharmakologie, Hannover, Germany (H.B., V.K.)
| | - Malgorzata Szaroszyk
- From Medizinische Hochschule Hannover, Klinik für Kardiologie und Angiologie, Hanover, Germany (C.Z., E.S., M.S., B.K., N.F., H.H., J.W., J.B., J.H.); Klinik für Herz-, Thorax-, Transplantations- und Gefäßchirurgie, Hanover, Germany (J.D.S.); Institut für Molekulare und Translationale Therapiestrategien (IMTTS), Hannover, Germany (S.B., T.T.); National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.); and Medizinische Hochschule Hannover, Zentrale Forschungseinrichtung Metabolomics, Institut für Pharmakologie, Hannover, Germany (H.B., V.K.)
| | - Badder Kattih
- From Medizinische Hochschule Hannover, Klinik für Kardiologie und Angiologie, Hanover, Germany (C.Z., E.S., M.S., B.K., N.F., H.H., J.W., J.B., J.H.); Klinik für Herz-, Thorax-, Transplantations- und Gefäßchirurgie, Hanover, Germany (J.D.S.); Institut für Molekulare und Translationale Therapiestrategien (IMTTS), Hannover, Germany (S.B., T.T.); National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.); and Medizinische Hochschule Hannover, Zentrale Forschungseinrichtung Metabolomics, Institut für Pharmakologie, Hannover, Germany (H.B., V.K.)
| | - Natali Froese
- From Medizinische Hochschule Hannover, Klinik für Kardiologie und Angiologie, Hanover, Germany (C.Z., E.S., M.S., B.K., N.F., H.H., J.W., J.B., J.H.); Klinik für Herz-, Thorax-, Transplantations- und Gefäßchirurgie, Hanover, Germany (J.D.S.); Institut für Molekulare und Translationale Therapiestrategien (IMTTS), Hannover, Germany (S.B., T.T.); National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.); and Medizinische Hochschule Hannover, Zentrale Forschungseinrichtung Metabolomics, Institut für Pharmakologie, Hannover, Germany (H.B., V.K.)
| | - Hebke Hinz
- From Medizinische Hochschule Hannover, Klinik für Kardiologie und Angiologie, Hanover, Germany (C.Z., E.S., M.S., B.K., N.F., H.H., J.W., J.B., J.H.); Klinik für Herz-, Thorax-, Transplantations- und Gefäßchirurgie, Hanover, Germany (J.D.S.); Institut für Molekulare und Translationale Therapiestrategien (IMTTS), Hannover, Germany (S.B., T.T.); National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.); and Medizinische Hochschule Hannover, Zentrale Forschungseinrichtung Metabolomics, Institut für Pharmakologie, Hannover, Germany (H.B., V.K.)
| | - Jan Dieter Schmitto
- From Medizinische Hochschule Hannover, Klinik für Kardiologie und Angiologie, Hanover, Germany (C.Z., E.S., M.S., B.K., N.F., H.H., J.W., J.B., J.H.); Klinik für Herz-, Thorax-, Transplantations- und Gefäßchirurgie, Hanover, Germany (J.D.S.); Institut für Molekulare und Translationale Therapiestrategien (IMTTS), Hannover, Germany (S.B., T.T.); National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.); and Medizinische Hochschule Hannover, Zentrale Forschungseinrichtung Metabolomics, Institut für Pharmakologie, Hannover, Germany (H.B., V.K.)
| | - Julian Widder
- From Medizinische Hochschule Hannover, Klinik für Kardiologie und Angiologie, Hanover, Germany (C.Z., E.S., M.S., B.K., N.F., H.H., J.W., J.B., J.H.); Klinik für Herz-, Thorax-, Transplantations- und Gefäßchirurgie, Hanover, Germany (J.D.S.); Institut für Molekulare und Translationale Therapiestrategien (IMTTS), Hannover, Germany (S.B., T.T.); National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.); and Medizinische Hochschule Hannover, Zentrale Forschungseinrichtung Metabolomics, Institut für Pharmakologie, Hannover, Germany (H.B., V.K.)
| | - Sandor Batkai
- From Medizinische Hochschule Hannover, Klinik für Kardiologie und Angiologie, Hanover, Germany (C.Z., E.S., M.S., B.K., N.F., H.H., J.W., J.B., J.H.); Klinik für Herz-, Thorax-, Transplantations- und Gefäßchirurgie, Hanover, Germany (J.D.S.); Institut für Molekulare und Translationale Therapiestrategien (IMTTS), Hannover, Germany (S.B., T.T.); National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.); and Medizinische Hochschule Hannover, Zentrale Forschungseinrichtung Metabolomics, Institut für Pharmakologie, Hannover, Germany (H.B., V.K.)
| | - Heike Bähre
- From Medizinische Hochschule Hannover, Klinik für Kardiologie und Angiologie, Hanover, Germany (C.Z., E.S., M.S., B.K., N.F., H.H., J.W., J.B., J.H.); Klinik für Herz-, Thorax-, Transplantations- und Gefäßchirurgie, Hanover, Germany (J.D.S.); Institut für Molekulare und Translationale Therapiestrategien (IMTTS), Hannover, Germany (S.B., T.T.); National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.); and Medizinische Hochschule Hannover, Zentrale Forschungseinrichtung Metabolomics, Institut für Pharmakologie, Hannover, Germany (H.B., V.K.)
| | - Volkhard Kaever
- From Medizinische Hochschule Hannover, Klinik für Kardiologie und Angiologie, Hanover, Germany (C.Z., E.S., M.S., B.K., N.F., H.H., J.W., J.B., J.H.); Klinik für Herz-, Thorax-, Transplantations- und Gefäßchirurgie, Hanover, Germany (J.D.S.); Institut für Molekulare und Translationale Therapiestrategien (IMTTS), Hannover, Germany (S.B., T.T.); National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.); and Medizinische Hochschule Hannover, Zentrale Forschungseinrichtung Metabolomics, Institut für Pharmakologie, Hannover, Germany (H.B., V.K.)
| | - Thomas Thum
- From Medizinische Hochschule Hannover, Klinik für Kardiologie und Angiologie, Hanover, Germany (C.Z., E.S., M.S., B.K., N.F., H.H., J.W., J.B., J.H.); Klinik für Herz-, Thorax-, Transplantations- und Gefäßchirurgie, Hanover, Germany (J.D.S.); Institut für Molekulare und Translationale Therapiestrategien (IMTTS), Hannover, Germany (S.B., T.T.); National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.); and Medizinische Hochschule Hannover, Zentrale Forschungseinrichtung Metabolomics, Institut für Pharmakologie, Hannover, Germany (H.B., V.K.)
| | - Johann Bauersachs
- From Medizinische Hochschule Hannover, Klinik für Kardiologie und Angiologie, Hanover, Germany (C.Z., E.S., M.S., B.K., N.F., H.H., J.W., J.B., J.H.); Klinik für Herz-, Thorax-, Transplantations- und Gefäßchirurgie, Hanover, Germany (J.D.S.); Institut für Molekulare und Translationale Therapiestrategien (IMTTS), Hannover, Germany (S.B., T.T.); National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.); and Medizinische Hochschule Hannover, Zentrale Forschungseinrichtung Metabolomics, Institut für Pharmakologie, Hannover, Germany (H.B., V.K.)
| | - Joerg Heineke
- From Medizinische Hochschule Hannover, Klinik für Kardiologie und Angiologie, Hanover, Germany (C.Z., E.S., M.S., B.K., N.F., H.H., J.W., J.B., J.H.); Klinik für Herz-, Thorax-, Transplantations- und Gefäßchirurgie, Hanover, Germany (J.D.S.); Institut für Molekulare und Translationale Therapiestrategien (IMTTS), Hannover, Germany (S.B., T.T.); National Heart and Lung Institute, Imperial College, London, United Kingdom (T.T.); and Medizinische Hochschule Hannover, Zentrale Forschungseinrichtung Metabolomics, Institut für Pharmakologie, Hannover, Germany (H.B., V.K.).
| |
Collapse
|
18
|
Impact of castration on changes in left ventricular diastolic pressure-volume relations induced by chronic adrenergic stimulation in rats. J Cardiovasc Pharmacol 2015; 63:562-6. [PMID: 24477046 DOI: 10.1097/fjc.0000000000000081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
A reduced testosterone concentration characterizes heart failure and independently predicts outcomes. Although testosterone replacement therapy may have non cardiac-related therapeutic benefits in heart failure, whether reduced testosterone concentrations protect against adverse left ventricular remodeling (LV dilatation) is uncertain. We therefore evaluated whether surgical castration modifies LV dilatation after 6 months of daily injections of the β-adrenergic receptor (AR) agonist, isoproterenol (ISO) (0.015 mg·kg(-1)·d(-1)), to rats. The extent of LV dilatation and LV systolic chamber dysfunction were determined using both echocardiography and isolated perfused heart procedures. The extent of LV dilatation was determined from LV diastolic pressure-volume (P-V) relationships. As compared with the saline vehicle-treated group, after 6 months of β-AR activation in sham-castrated rats, a marked right shift in the LV diastolic P-V relationship was noted with an increased LV volume intercept at 0 mm Hg diastolic pressure (LV V(0) in milliliters) (ISO = 0.38 ± 0.02, saline vehicle = 0.30 ± 0.02, P < 0.05). However, chronic β-AR activation did not alter LV systolic chamber function either in vivo (LV endocardial fractional shortening, echocardiography) or ex vivo (LV end systolic elastance). Although castration decreased body weight, castration failed to modify the impact of ISO on the LV diastolic P-V relationships or the LV volume intercept at 0 mm Hg diastolic pressure (LV V(0) in milliliters) (castration ISO = 0.35 ± 0.02, castration saline vehicle = 0.27 ± 0.03, P < 0.05). In conclusion, castration does not influence the extent of LV dilatation induced by chronic adrenergic activation in an animal model, where adverse LV remodeling precedes LV systolic chamber dysfunction.
Collapse
|
19
|
Prabhavathi K, Selvi K, Poornima K, Sarvanan A. Role of biological sex in normal cardiac function and in its disease outcome - a review. J Clin Diagn Res 2014; 8:BE01-4. [PMID: 25302188 PMCID: PMC4190707 DOI: 10.7860/jcdr/2014/9635.4771] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 06/12/2014] [Indexed: 12/21/2022]
Abstract
Biological sex plays an important role in normal cardiac physiology as well as in the heart's response to cardiac disease. Women generally have better cardiac function and survival than do men in the face of cardiac disease; however, this is progressively lost when comparing postmenopausal women with age matched men. Animal model of cardiac disease mirror what is seen in humans. Sex hormones contribute significantly to sex based difference in cardiac functioning and in its disease outcome. Estrogen is considered to be cardioprotective, whereas testosterone is detrimental to heart function.
Collapse
Affiliation(s)
- K. Prabhavathi
- Assistant Professor, Department of Pathology, SRM Medical College and Research Center, Chennai, Tamil Nadu, India
| | - K.Tamarai Selvi
- Professor, Department of Physiology, SRM Medical College and Research Center, Chennai, Tamil Nadu, India
| | - K.N. Poornima
- Tutor, Department of Physiology, SRM Medical College and Research Center, Chennai, Tamil Nadu, India
| | - A. Sarvanan
- Professor, Department of Physiology, SRM Medical College and Research Center, Chennai, Tamil Nadu, India
| |
Collapse
|
20
|
Collette KM, Zhou XD, Amoth HM, Lyons MJ, Papay RS, Sens DA, Perez DM, Doze VA. Long-term α1B-adrenergic receptor activation shortens lifespan, while α1A-adrenergic receptor stimulation prolongs lifespan in association with decreased cancer incidence. AGE (DORDRECHT, NETHERLANDS) 2014; 36:9675. [PMID: 24994537 PMCID: PMC4150908 DOI: 10.1007/s11357-014-9675-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 06/18/2014] [Indexed: 06/03/2023]
Abstract
The α1-adrenergic receptor (α1AR) subtypes, α1AAR and α1BAR, have differential effects in the heart and central nervous system. Long-term stimulation of the α1AAR subtype prolongs lifespan and provides cardio- and neuro-protective effects. We examined the lifespan of constitutively active mutant (CAM)-α1BAR mice and the incidence of cancer in mice expressing the CAM form of either the α1AAR (CAM-α1AAR mice) or α1BAR. CAM-α1BAR mice have a significantly shortened lifespan when compared with wild-type (WT) animals; however, the effect was sex dependent. Female CAM-α1BAR mice lived significantly shorter lives, while the median lifespan of male CAM-α1BAR mice was not different when compared with that of WT animals. There was no difference in the incidence of cancer in either sex of CAM-α1BAR mice. The incidence of cancer was significantly decreased in CAM-α1AAR mice when compared with that in WT, and no sex-dependent effects were observed. Further study is warranted on cancer incidence after activation of each α1AR subtype and the effect of sex on lifespan following activation of the α1BAR. The implications of a decrease in cancer incidence following long-term α1AAR stimulation could lead to improved treatments for cancer.
Collapse
Affiliation(s)
- Katie M. Collette
- />Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, 501 N. Columbia Rd., Grand Forks, ND 58202 USA
| | - Xu Dong Zhou
- />Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202 USA
| | - Haley M. Amoth
- />Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, 501 N. Columbia Rd., Grand Forks, ND 58202 USA
| | - Mariaha J. Lyons
- />Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, 501 N. Columbia Rd., Grand Forks, ND 58202 USA
| | - Robert S. Papay
- />Department of Molecular Cardiology, The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH 44195 USA
| | - Donald A. Sens
- />Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202 USA
| | - Dianne M. Perez
- />Department of Molecular Cardiology, The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH 44195 USA
| | - Van A. Doze
- />Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, 501 N. Columbia Rd., Grand Forks, ND 58202 USA
| |
Collapse
|
21
|
Gao Y, Lin Y, Sun K, Wang Y, Chen J, Wang H, Zhou X, Fan X, Hui R. Orthostatic Blood Pressure Dysregulation and Polymorphisms of β-Adrenergic Receptor Genes in Hypertensive Patients. J Clin Hypertens (Greenwich) 2014; 16:207-13. [PMID: 24552127 DOI: 10.1111/jch.12272] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 12/18/2013] [Accepted: 12/28/2013] [Indexed: 11/28/2022]
Affiliation(s)
- Ying Gao
- State Key Laboratory of Cardiovascular Disease; Department of Cardiology; Fuwai Hospital; National Center for Cardiovascular Diseases; Chinese Academy of Medical Sciences; Peking Union Medical College; Beijing China
| | - Yahui Lin
- State Key Laboratory of Cardiovascular Disease; Sino-German Laboratory for Molecular Medicine; Fuwai Hospital; National Center for Cardiovascular Diseases; Chinese Academy of Medical Sciences; Peking Union Medical College; Beijing China
| | - Kai Sun
- State Key Laboratory of Cardiovascular Disease; Sino-German Laboratory for Molecular Medicine; Fuwai Hospital; National Center for Cardiovascular Diseases; Chinese Academy of Medical Sciences; Peking Union Medical College; Beijing China
| | - Yibo Wang
- State Key Laboratory of Cardiovascular Disease; Sino-German Laboratory for Molecular Medicine; Fuwai Hospital; National Center for Cardiovascular Diseases; Chinese Academy of Medical Sciences; Peking Union Medical College; Beijing China
| | - Jingzhou Chen
- State Key Laboratory of Cardiovascular Disease; Sino-German Laboratory for Molecular Medicine; Fuwai Hospital; National Center for Cardiovascular Diseases; Chinese Academy of Medical Sciences; Peking Union Medical College; Beijing China
| | - Hu Wang
- State Key Laboratory of Cardiovascular Disease; Sino-German Laboratory for Molecular Medicine; Fuwai Hospital; National Center for Cardiovascular Diseases; Chinese Academy of Medical Sciences; Peking Union Medical College; Beijing China
| | - Xianliang Zhou
- State Key Laboratory of Cardiovascular Disease; Department of Cardiology; Fuwai Hospital; National Center for Cardiovascular Diseases; Chinese Academy of Medical Sciences; Peking Union Medical College; Beijing China
| | - Xiaohan Fan
- State Key Laboratory of Cardiovascular Disease; Department of Cardiology; Fuwai Hospital; National Center for Cardiovascular Diseases; Chinese Academy of Medical Sciences; Peking Union Medical College; Beijing China
| | - Rutai Hui
- State Key Laboratory of Cardiovascular Disease; Department of Cardiology; Fuwai Hospital; National Center for Cardiovascular Diseases; Chinese Academy of Medical Sciences; Peking Union Medical College; Beijing China
- State Key Laboratory of Cardiovascular Disease; Sino-German Laboratory for Molecular Medicine; Fuwai Hospital; National Center for Cardiovascular Diseases; Chinese Academy of Medical Sciences; Peking Union Medical College; Beijing China
| |
Collapse
|
22
|
Spangenburg EE, Geiger PC, Leinwand LA, Lowe DA. Regulation of physiological and metabolic function of muscle by female sex steroids. Med Sci Sports Exerc 2013; 44:1653-62. [PMID: 22525764 DOI: 10.1249/mss.0b013e31825871fa] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The ability of female sex steroids to regulate tissue function has long been appreciated; however, their role in the regulation of striated muscle function has received considerably less attention. The purpose of this symposium review was to document recent evidence indicating the role female sex steroids have in defining the functional characteristics of striated muscle. The presentations provide substantial evidence indicating that estrogens are critical to the physiological and metabolic regulations of striated muscle; thus, when considering women's health issues, striated muscle must included as an important target tissue along with other classically thought of estrogen-sensitive tissues.
Collapse
Affiliation(s)
- Espen E Spangenburg
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, MD 21045, USA.
| | | | | | | |
Collapse
|
23
|
Arimura T, Onoue K, Takahashi-Tanaka Y, Ishikawa T, Kuwahara M, Setou M, Shigenobu S, Yamaguchi K, Bertrand AT, Machida N, Takayama K, Fukusato M, Tanaka R, Somekawa S, Nakano T, Yamane Y, Kuba K, Imai Y, Saito Y, Bonne G, Kimura A. Nuclear accumulation of androgen receptor in gender difference of dilated cardiomyopathy due to lamin A/C mutations. Cardiovasc Res 2013; 99:382-94. [DOI: 10.1093/cvr/cvt106] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
24
|
Zhao L, Yang F, Xu K, Cao H, Zheng GY, Zhang Y, Li J, Cui H, Chen X, Zhu Z, He H, Mo X, Kennedy BK, Suh Y, Zeng Y, Tian XL. Common genetic variants of the β2-adrenergic receptor affect its translational efficiency and are associated with human longevity. Aging Cell 2012; 11:1094-101. [PMID: 23020224 DOI: 10.1111/acel.12011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2012] [Indexed: 02/05/2023] Open
Abstract
β-adrenoceptors are the common pharmacological targets for the treatment of cardiovascular diseases and asthma. Genetic modifications of β-adrenergic system in engineered mice affect their lifespan. Here, we tested whether genes encoding for key components of the β-adrenergic signaling pathway are associated with human longevity. We performed a 10-year follow-up study of the Chinese longitudinal healthy longevity survey. The Han Chinese population in this study consisted of 963 long-lived and 1028 geography-matched young individuals. Sixteen SNPs from ADRB1, ADRB2, ADCY5, ADCY6, and MAPK1 were selected and genotyped. Two SNPs, rs1042718 (C/A) and rs1042719 (G/C), of ADRB2 in linkage disequilibrium (D' = 1.0; r2 = 0.67) were found to be associated with enhanced longevity in men in two geographically isolated populations. Bonferroni-corrected P-values in a combined analysis were 0.00053-0.010. Men with haplotype A-C showed an increased probability to become centenarians (the frequency of A-C in long-lived and young individuals are 0.332 and 0.250, respectively, OR = 1.49, CI 95% = 1.17-1.88, P = 0.0007), in contrast to those with haplotype C-G (the frequency of C-G in long-lived and young individuals are 0.523 and 0.635, respectively, OR = 0.63, CI 95% = 0.51-0.78, P = 0.000018). The permuted P-values were 0.00005 and 0.0009, respectively. ADRB2 encodes the β2-adrenergic receptor; the haplotype A-C markedly reduced its translational efficiency compared with C-G (P = 0.002) in transfected HEK293 cells. Thus, our data indicate that enhanced production of β2-adrenergic receptors caused by genetic variants is inversely associated with human lifespan.
Collapse
Affiliation(s)
- Ling Zhao
- Department of Human Population Genetics; Institute of Molecular Medicine; Peking University; 5 Yiheyuan Road; Beijing; 100871; China
| | - Fan Yang
- Department of Human Population Genetics; Institute of Molecular Medicine; Peking University; 5 Yiheyuan Road; Beijing; 100871; China
| | - Ke Xu
- Department of Human Population Genetics; Institute of Molecular Medicine; Peking University; 5 Yiheyuan Road; Beijing; 100871; China
| | - Huiqing Cao
- Department of Human Population Genetics; Institute of Molecular Medicine; Peking University; 5 Yiheyuan Road; Beijing; 100871; China
| | - Gu-Yan Zheng
- Department of Human Population Genetics; Institute of Molecular Medicine; Peking University; 5 Yiheyuan Road; Beijing; 100871; China
| | - Yan Zhang
- Department of Human Population Genetics; Institute of Molecular Medicine; Peking University; 5 Yiheyuan Road; Beijing; 100871; China
| | - Jianxin Li
- Department of Sociology; Peking University; 5 Yiheyuan Road; Beijing; 100871; China
| | - Hanbin Cui
- Key Laboratory of Ningbo First Hospital and Cardiovascular Center of Ningbo First Hospital; Ningbo University; 59 Liuting Street; Ningbo; 315010; China
| | - Xiaomin Chen
- Key Laboratory of Ningbo First Hospital and Cardiovascular Center of Ningbo First Hospital; Ningbo University; 59 Liuting Street; Ningbo; 315010; China
| | - Zhiming Zhu
- Center for Hypertension and Metabolic Diseases; Department of Hypertension and Endocrinology; Daping Hospital; Chongqing Institute of Hypertension; Third Military Medical University; Chongqing; 400042; China
| | - Hongbo He
- Center for Hypertension and Metabolic Diseases; Department of Hypertension and Endocrinology; Daping Hospital; Chongqing Institute of Hypertension; Third Military Medical University; Chongqing; 400042; China
| | - Xianming Mo
- Department of Geriatrics; West China Hospital; West China Medical School; Sichuan University; Chengdu; 610041; China
| | | | - Yousin Suh
- Departments of Medicine and Genetics; Albert Einstein College of Medicine; Bronx; NY; 10461; USA
| | | | - Xiao-Li Tian
- Department of Human Population Genetics; Institute of Molecular Medicine; Peking University; 5 Yiheyuan Road; Beijing; 100871; China
| |
Collapse
|
25
|
Dent MR, Tappia PS, Dhalla NS. Gender related alterations of β-adrenoceptor mechanisms in heart failure due to arteriovenous fistula. J Cell Physiol 2012; 227:3080-7. [PMID: 22015551 DOI: 10.1002/jcp.23058] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
This study was undertaken to determine gender related changes in different components of β-adrenoceptor (β-AR) system in response to arteriovenous fistula (AV-shunt), which is known to produce heart failure due to volume overload. AV-shunt was induced in male and female rats for 16 weeks by the needle technique; ovariectomized (OVX) rats treated with or without estrogen were also used. Although AV-shunt for 16 weeks produced cardiac hypertrophy in both sexes, male animals showed cardiac dysfunction whereas cardiac performance was maintained in females. Both β(1) -AR and β(2) -AR protein content and mRNA levels were decreased in male and increased in female hearts post-AV-shunt. The basal adenylyl cyclase (AC) activity was lower in the female heart; however, AC protein content and the increase in epinephrine (EPi)-stimulated AC activity were greater in the female AV-shunt group as compared to males. While AC V/VI and β-arrestin 2 mRNA levels were decreased in males, mRNA level for GRK2 was increased in females post-AV-shunt. In contrast to intact females, AV-shunt OVX animals showed depressed cardiac function, decreased β(1) -AR, β(2) -AR, and AC protein content, as well as reduced EPi-stimulated AC activity. Treatment of OVX rats with 17-β estradiol attenuated the AV-shunt induced changes in β-AR and AC protein content as well as cardiac dysfunction. These results reveal that β-AR signal transduction system in response to AV-shunt is downregulated in males and upregulated in females. Furthermore, estrogen appears to play an important role in the upregulation of β-AR mechanisms and the maintenance of cardiac function in AV-shunt females.
Collapse
Affiliation(s)
- Melissa R Dent
- Faculty of Medicine, Department of Physiology, University of Manitoba, Institute of Cardiovascular Sciences, St. Boniface Hospital Research, Winnipeg, Canada
| | | | | |
Collapse
|
26
|
Montalvo C, Villar AV, Merino D, García R, Ares M, Llano M, Cobo M, Hurlé MA, Nistal JF. Androgens contribute to sex differences in myocardial remodeling under pressure overload by a mechanism involving TGF-β. PLoS One 2012; 7:e35635. [PMID: 22558184 PMCID: PMC3338422 DOI: 10.1371/journal.pone.0035635] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Accepted: 03/19/2012] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND In clinical studies, myocardial remodeling in aortic valve stenosis appears to be more favorable in women than in men, even after menopause. In the present study, we assessed whether circulating androgens contribute to a less favorable myocardial remodeling under pressure overload in males. We examined sex-related differences in one-year-old male and female mice. Whereas male mice at this age exhibited circulating androgen levels within the normal range for young adults, the circulating estrogens in females were reduced. The contribution of gonadal androgens to cardiac remodeling was analyzed in a group of same-age castrated mice. METHODOLOGY/PRINCIPAL FINDINGS Animals were subjected to transverse aortic constriction (TAC). Echocardiography was performed 2 weeks after TAC and myocardial mRNA levels of TGF-βs, Smads 2 and 3, collagens, fibronectin, β-myosin heavy chain and α-myosin heavy chain were determined by q-PCR. Protein detection of p-SMAD2/3 was performed by Western Blot. Histological staining of fibrosis was performed with picrosirius red and Masson's trichrome. Compared with females, males developed more severe tissue fibrosis, LV dilation and hemodynamic dysfunction. TAC-males showed higher myocardial expression levels of TGF-βs and the treatment with a neutralizing antibody to TGF-β prevented myocardial fibrosis development. Orchiectomy diminished TAC-induced up-regulation of TGF-βs and TGF-β target genes, and it also reduced fibrosis and hemodynamic dysfunction. The capability of androgens to induce TGF-β expression was confirmed in NIH-3T3 fibroblasts and H9C2 cardiomyocytes exposed to dihydrotestosterone. CONCLUSIONS/SIGNIFICANCE Our results indicate that circulating androgens are responsible for the detrimental effects in the myocardium of older male mice subjected to pressure overload through a mechanism involving TGF-βs.
Collapse
Affiliation(s)
- Cecilia Montalvo
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Santander, Spain
- Instituto de Formación e Investigación Marqués de Valdecilla (IFIMAV), Santander, Spain
| | - Ana V. Villar
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Santander, Spain
- Instituto de Formación e Investigación Marqués de Valdecilla (IFIMAV), Santander, Spain
| | - David Merino
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Santander, Spain
- Instituto de Formación e Investigación Marqués de Valdecilla (IFIMAV), Santander, Spain
| | - Raquel García
- Servicio de Cirugía Cardiovascular, Hospital Universitario Marqués de Valdecilla, Santander, Spain
- Instituto de Formación e Investigación Marqués de Valdecilla (IFIMAV), Santander, Spain
| | - Miguel Ares
- Hospital Comarcal de Laredo, Cantabria, Spain
- Instituto de Formación e Investigación Marqués de Valdecilla (IFIMAV), Santander, Spain
| | - Miguel Llano
- Servicio de Cardiología, Hospital Universitario Marqués de Valdecilla, Santander, Spain
- Instituto de Formación e Investigación Marqués de Valdecilla (IFIMAV), Santander, Spain
| | - Manuel Cobo
- Servicio de Cardiología, Hospital Universitario Marqués de Valdecilla, Santander, Spain
- Instituto de Formación e Investigación Marqués de Valdecilla (IFIMAV), Santander, Spain
| | - María A. Hurlé
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Santander, Spain
- Instituto de Formación e Investigación Marqués de Valdecilla (IFIMAV), Santander, Spain
- * E-mail:
| | - J. Francisco Nistal
- Servicio de Cirugía Cardiovascular, Hospital Universitario Marqués de Valdecilla, Santander, Spain
- Instituto de Formación e Investigación Marqués de Valdecilla (IFIMAV), Santander, Spain
| |
Collapse
|
27
|
Vyas PM, Tomamichel WJ, Pride PM, Babbey CM, Wang Q, Mercier J, Martin EM, Payne RM. A TAT-frataxin fusion protein increases lifespan and cardiac function in a conditional Friedreich's ataxia mouse model. Hum Mol Genet 2012; 21:1230-47. [PMID: 22113996 PMCID: PMC3284115 DOI: 10.1093/hmg/ddr554] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 11/21/2011] [Indexed: 11/14/2022] Open
Abstract
Friedreich's ataxia (FRDA) is the most common inherited human ataxia and results from a deficiency of the mitochondrial protein, frataxin (FXN), which is encoded in the nucleus. This deficiency is associated with an iron-sulfur (Fe-S) cluster enzyme deficit leading to progressive ataxia and a frequently fatal cardiomyopathy. There is no cure. To determine whether exogenous replacement of the missing FXN protein in mitochondria would repair the defect, we used the transactivator of transcription (TAT) protein transduction domain to deliver human FXN protein to mitochondria in both cultured patient cells and a severe mouse model of FRDA. A TAT-FXN fusion protein bound iron in vitro, transduced into mitochondria of FRDA deficient fibroblasts and reduced caspase-3 activation in response to an exogenous iron-oxidant stress. Injection of TAT-FXN protein into mice with a conditional loss of FXN increased their growth velocity and mean lifespan by 53% increased their mean heart rate and cardiac output, increased activity of aconitase and reversed abnormal mitochondrial proliferation and ultrastructure in heart. These results show that a cell-penetrant peptide is capable of delivering a functional mitochondrial protein in vivo to rescue a very severe disease phenotype, and present the possibility of TAT-FXN as a protein replacement therapy.
Collapse
Affiliation(s)
- Piyush M. Vyas
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Wendy J. Tomamichel
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - P. Melanie Pride
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Clifford M. Babbey
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Qiujuan Wang
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jennifer Mercier
- Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Elizabeth M. Martin
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - R. Mark Payne
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
28
|
Hewitson TD, Zhao C, Wigg B, Lee SW, Simpson ER, Boon WC, Samuel CS. Relaxin and castration in male mice protect from, but testosterone exacerbates, age-related cardiac and renal fibrosis, whereas estrogens are an independent determinant of organ size. Endocrinology 2012; 153:188-99. [PMID: 22028442 DOI: 10.1210/en.2011-1311] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
This study determined the effects of castration and hormone replacement therapy on the age-related cardiac and renal pathology of male relaxin gene-knockout (RlnKO) and age-matched wild-type (RlnWT) mice and that of aged male aromatase knockout (ArKO) mice, which lack estrogens and have 5-10 times the androgen levels of male wild-type mice. One-month-old RlnWT and RlnKO mice were bilaterally gonadectomized or sham operated and maintained until 12 months. Subgroups of castrated animals received testosterone or 17β-estradiol treatment from 9 to 12 months. Male ArKO mice and aromatase wild-type mice were aged to 12 months. Collected heart and kidney tissues were assessed for changes in organ size and fibrosis. Castration reduced body, heart, left ventricle, and kidney weights in both RlnKO and RlnWT mice, and the cardiac/renal fibrosis that was seen in sham RlnKO animals (all P < 0.05 vs. respective sham). Testosterone normalized organ weights and organ weight to body weight ratio of castrated animals and increased cardiac/renal collagen concentration to levels measured in or beyond that of sham RlnKO mice (all P < 0.05 vs. respective castrated mice). Furthermore, expression of TGF-β1, mothers against decapentaplegic homolog 2 (Smad2), and myofibroblast differentiation paralleled the above changes (all P < 0.05 vs. respective castrated mice), whereas matrix metalloproteinase-13 was decreased in testosterone-treated RlnKO mice. Conversely, 17β-estradiol only restored changes in organ size. Consistent with these findings, intact ArKO mice demonstrated increased cardiac/renal fibrosis in the absence of changes in organ size. These findings suggest that relaxin and castration protect, whereas androgens exacerbate, cardiac and renal fibrosis during ageing, whereas estrogens, in synergy with relaxin, regulates age-related changes in organ size.
Collapse
Affiliation(s)
- Tim D Hewitson
- Howard Florey Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | | | | | | | | | |
Collapse
|
29
|
Sussman MA, Völkers M, Fischer K, Bailey B, Cottage CT, Din S, Gude N, Avitabile D, Alvarez R, Sundararaman B, Quijada P, Mason M, Konstandin MH, Malhowski A, Cheng Z, Khan M, McGregor M. Myocardial AKT: the omnipresent nexus. Physiol Rev 2011; 91:1023-70. [PMID: 21742795 PMCID: PMC3674828 DOI: 10.1152/physrev.00024.2010] [Citation(s) in RCA: 177] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
One of the greatest examples of integrated signal transduction is revealed by examination of effects mediated by AKT kinase in myocardial biology. Positioned at the intersection of multiple afferent and efferent signals, AKT exemplifies a molecular sensing node that coordinates dynamic responses of the cell in literally every aspect of biological responses. The balanced and nuanced nature of homeostatic signaling is particularly essential within the myocardial context, where regulation of survival, energy production, contractility, and response to pathological stress all flow through the nexus of AKT activation or repression. Equally important, the loss of regulated AKT activity is primarily the cause or consequence of pathological conditions leading to remodeling of the heart and eventual decompensation. This review presents an overview compendium of the complex world of myocardial AKT biology gleaned from more than a decade of research. Summarization of the widespread influence that AKT exerts upon myocardial responses leaves no doubt that the participation of AKT in molecular signaling will need to be reckoned with as a seemingly omnipresent regulator of myocardial molecular biological responses.
Collapse
Affiliation(s)
- Mark A Sussman
- Department of Biology, San Diego State University, SDSU Heart Institute, San Diego, California 92182, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Xu Q, Dalic A, Fang L, Kiriazis H, Ritchie RH, Sim K, Gao XM, Drummond G, Sarwar M, Zhang YY, Dart AM, Du XJ. Myocardial oxidative stress contributes to transgenic β₂-adrenoceptor activation-induced cardiomyopathy and heart failure. Br J Pharmacol 2011; 162:1012-28. [PMID: 20955367 DOI: 10.1111/j.1476-5381.2010.01043.x] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE While maintaining cardiac performance, chronic β-adrenoceptor activation eventually exacerbates the progression of cardiac remodelling and failure. We examined the adverse signalling pathways mediated by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and reactive oxygen species (ROS) after chronic β₂-adrenoceptor activation. EXPERIMENTAL APPROACH Mice with transgenic β₂-adrenoceptor overexpression (β₂-TG) and non-transgenic littermates were either untreated or treated with an antioxidant (N-acetylcysteine, NAC) or NADPH oxidase inhibitors (apocynin, diphenyliodonium). Levels of ROS, phosphorylated p38 mitogen-activated protein kinase (MAPK), pro-inflammatory cytokines and collagen content in the left ventricle (LV) and LV function were measured and compared. KEY RESULTS β₂-TG mice showed increased ROS production, phosphorylation of p38 MAPK and heat shock protein 27 (HSP27), expression of pro-inflammatory cytokines and collagen, and progressive ventricular dysfunction. β₂-adrenoceptor stimulation similarly increased ROS production and phosphorylation of p38 MAPK and HSP27 in cultured cardiomyocytes. Treatment with apocynin, diphenyliodonium or NAC reduced phosphorylation of p38 MAPK and HSP27 in both cultured cardiomyocytes and the LV of β₂-TG mice. NAC treatment (500 mg·kg⁻¹ ·day⁻¹) for 2 weeks eliminated the up-regulated expression of pro-inflammatory cytokines and collagen in the LV of β₂-TG mice. Chronic NAC treatment to β₂-TG mice from 7 to 10 months of age largely prevented progression of ventricular dilatation, preserved contractile function (fractional shortening 37 ± 5% vs. 25 ± 3%, ejection fraction 52 ± 5% vs. 32 ± 4%, both P < 0.05), reduced cardiac fibrosis and suppressed matrix metalloproteinase activity. CONCLUSION AND IMPLICATIONS β₂-adrenoceptor stimulation provoked NADPH oxidase-derived ROS production in the heart. Elevated ROS activated p38 MAPK and contributed significantly to cardiac inflammation, remodelling and failure.
Collapse
Affiliation(s)
- Q Xu
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Samuel CS, Cendrawan S, Gao XM, Ming Z, Zhao C, Kiriazis H, Xu Q, Tregear GW, Bathgate RAD, Du XJ. Relaxin remodels fibrotic healing following myocardial infarction. J Transl Med 2011; 91:675-90. [PMID: 21221074 DOI: 10.1038/labinvest.2010.198] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
In the setting of myocardial infarction (MI), implanted stem cell viability is low and scar formation limits stem cell homing, viability, and integration. Thus, interventions that favorably remodel fibrotic healing may benefit stem cell therapies. However, it remains unclear whether it is feasible and safe to remodel fibrotic healing post-MI without compromising ventricular remodeling and dysfunction. This study, therefore, determined the anti-fibrotic and other effects of the hormone, relaxin in a mouse model of MI. Adult male mice underwent left coronary artery ligation-induced MI and were immediately treated with recombinant human relaxin (MI+RLX) or vehicle (MI+VEH) over 7 or 30 days, representing time points of early and mature fibrotic healing. Cardiac function was assessed by echocardiography and catheterization, while comprehensive immunohistochemistry, morphometry, and western blotting were performed to explore the relaxin-induced mechanisms of action post-MI. RLX significantly inhibited the MI-induced progression of cardiac fibrosis over 7 and 30 days, which was associated with a reduction in TGF-β1 expression, myofibroblast differentiation, and cardiomyocyte apoptosis in addition to a promotion of matrix metalloproteinase-13 levels and de novo blood vessel growth (all P<0.05 vs respective measurements from MI+VEH mice). Despite the evident fibrotic healing post-MI, relaxin did not adversely affect the incidence of ventricular free-wall rupture or the extent of LV remodeling and dysfunction. These combined findings demonstrate that RLX favorably remodels the process of fibrotic healing post-infarction by lowering the density of mature scar tissue in the infarcted myocardium, border zone, and non-infarcted myocardium, and may, therefore, facilitate cell-based therapies in the setting of ischemic heart disease.
Collapse
Affiliation(s)
- Chrishan S Samuel
- Howard Florey Institute, University of Melbourne, Parkville, Victoria, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Hossain MA, Chow Suet Man B, Zhao C, Xu Q, Du XJ, Wade JD, Samuel CS. H3 Relaxin Demonstrates Antifibrotic Properties via the RXFP1 Receptor. Biochemistry 2011; 50:1368-75. [DOI: 10.1021/bi1013968] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
| | | | | | - Qi Xu
- Baker IDI Heart and Diabetes Institute, St. Kilda Road Central, Melbourne, Victoria 8008, Australia
| | - Xiao-Jun Du
- Baker IDI Heart and Diabetes Institute, St. Kilda Road Central, Melbourne, Victoria 8008, Australia
| | | | | |
Collapse
|
33
|
Molecular distinction between physiological and pathological cardiac hypertrophy: experimental findings and therapeutic strategies. Pharmacol Ther 2010; 128:191-227. [PMID: 20438756 DOI: 10.1016/j.pharmthera.2010.04.005] [Citation(s) in RCA: 604] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cardiac hypertrophy can be defined as an increase in heart mass. Pathological cardiac hypertrophy (heart growth that occurs in settings of disease, e.g. hypertension) is a key risk factor for heart failure. Pathological hypertrophy is associated with increased interstitial fibrosis, cell death and cardiac dysfunction. In contrast, physiological cardiac hypertrophy (heart growth that occurs in response to chronic exercise training, i.e. the 'athlete's heart') is reversible and is characterized by normal cardiac morphology (i.e. no fibrosis or apoptosis) and normal or enhanced cardiac function. Given that there are clear functional, structural, metabolic and molecular differences between pathological and physiological hypertrophy, a key question in cardiovascular medicine is whether mechanisms responsible for enhancing function of the athlete's heart can be exploited to benefit patients with pathological hypertrophy and heart failure. This review summarizes key experimental findings that have contributed to our understanding of pathological and physiological heart growth. In particular, we focus on signaling pathways that play a causal role in the development of pathological and physiological hypertrophy. We discuss molecular mechanisms associated with features of cardiac hypertrophy, including protein synthesis, sarcomeric organization, fibrosis, cell death and energy metabolism and provide a summary of profiling studies that have examined genes, microRNAs and proteins that are differentially expressed in models of pathological and physiological hypertrophy. How gender and sex hormones affect cardiac hypertrophy is also discussed. Finally, we explore how knowledge of molecular mechanisms underlying pathological and physiological hypertrophy may influence therapeutic strategies for the treatment of cardiovascular disease and heart failure.
Collapse
|
34
|
Thireau J, Aimond F, Poisson D, Zhang B, Bruneval P, Eder V, Richard S, Babuty D. New insights into sexual dimorphism during progression of heart failure and rhythm disorders. Endocrinology 2010; 151:1837-45. [PMID: 20176721 DOI: 10.1210/en.2009-1184] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Neurohormonal imbalance is a key determinant of the progression of heart failure (HF), which results in an elevated risk of mortality. A better understanding of mechanisms involved may influence treatment strategies. The incidence and prevalence of HF are lower in women. We explored sexual dimorphism in the progression of HF using a mice model of neurohormonal-dependent HF. Male and female mice overexpressing the human beta2-adrenergic receptor (TG4 strain) develop HF. We compared TG4 animals with age-matched wild-type controls. Cardiac function was studied in vivo by echocardiography and electrocardiography. Histological studies were performed. Conduction parameters were assessed by intracardiac electrophysiological exploration, as was the occurrence of spontaneous and inducible arrhythmias. The patch-clamp technique was used to determine the cellular electrophysiological profile. The role of hormonal status in HF progression was investigated by surgical gonadectomy. High mortality rate was observed in TG4 mice with a dramatic difference between males and females. Male TG4 mice exhibited intraventricular conduction abnormalities, as measured by infrahisian interval and QRS durations potentially determining reentrant circuits and increasing susceptibility to arrhythmia. The severity of HF was correlated with the degree of fibrosis, which was modulated by the gonadal hormones. Action potentials recorded from male and female left ventricular cardiomyocytes were indistinguishable, although both sexes exhibited delayed repolarization when compared with their wild-type counterparts. In conclusion, female TG4 mice were better protected than males against cardiac remodeling and rhythm disorders. A link between fibrosis, conduction time, and mortality was established in relation with sex hormones.
Collapse
Affiliation(s)
- Jérôme Thireau
- Centre National de la Recherche Scientifique FRE3092, Université François-Rabelais, F-37041 Tours, France
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Adigun AA, Wrench N, Seidler FJ, Slotkin TA. Neonatal organophosphorus pesticide exposure alters the developmental trajectory of cell-signaling cascades controlling metabolism: differential effects of diazinon and parathion. ENVIRONMENTAL HEALTH PERSPECTIVES 2010; 118:210-5. [PMID: 20123610 PMCID: PMC2831919 DOI: 10.1289/ehp.0901237] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Accepted: 09/24/2009] [Indexed: 05/17/2023]
Abstract
BACKGROUND Organophosphorus pesticides (OPs) are developmental neurotoxicants but also produce lasting effects on metabolism. OBJECTIVES/METHODS We administered diazinon (DZN) or parathion (PRT) to rats on postnatal days 14 at doses straddling the threshold for systemic signs of exposure and assessed the effects on hepatic and cardiac cell signaling mediated through the adenylyl cyclase (AC) cascade. RESULTS In the liver, DZN elicited global sensitization, characterized by parallel up-regulation of AC activity itself and of the responses to stimulants acting at beta-adrenergic receptors, glucagon receptors, or G-proteins. The effects intensified over the course from adolescence to adulthood. In contrast, PRT elicited up-regulation in adolescence that waned by adulthood. Superimposed on these general patterns were effects on glucagon receptor coupling to AC and on responses mediated through the Gi inhibitory protein. The effects on the liver were more substantial than those in the heart, which displayed only transient effects of DZN on AC function in adolescence and no significant effects of PRT. Furthermore, the hepatic effects were greater in magnitude than those in a brain region (cerebellum) that shares similar AC cascade elements. CONCLUSIONS These findings indicate that OPs alter the trajectory of hepatic cell signaling in a manner consistent with the observed emergence of prediabetes-like metabolic dysfunction. Notably, the various OPs differ in their net impact on peripheral AC signaling, making it unlikely that the effects on signaling reflect their shared property as cholinesterase inhibitors.
Collapse
Affiliation(s)
| | | | | | - Theodore A. Slotkin
- Address correspondence to T.A. Slotkin, Box 3813 DUMC, Duke University Medical Center, Durham, NC 27710 USA. Telephone: (919) 681-8015. Fax: (919) 684-8922. E-mail:
| |
Collapse
|
36
|
Koshman YE, Piano MR, Russell B, Schwertz DW. Signaling responses after exposure to 5 alpha-dihydrotestosterone or 17 beta-estradiol in norepinephrine-induced hypertrophy of neonatal rat ventricular myocytes. J Appl Physiol (1985) 2009; 108:686-96. [PMID: 20044473 DOI: 10.1152/japplphysiol.00994.2009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Androgens appear to enhance, whereas estrogens mitigate, cardiac hypertrophy. However, signaling pathways in cells for short (3 min) and longer term (48 h) treatment with 17beta-estradiol (E2) or 5 alpha-dihydrotestosterone (DHT) are understudied. We compared the effect of adrenergic stimulation by norepinephrine (NE; 1 microM) alone or in combination with DHT (10 nM) or E2 (10 nM) treatment in neonatal rat ventricular myocytes (NRVMs) by cell area, protein synthesis, sarcomeric structure, gene expression, phosphorylation of extracellular signal-regulated (ERK), and focal adhesion kinases (FAK), and phospho-FAK nuclear localization. NE alone elicited the expected hypertrophy and strong sarcomeric organization, and DHT alone gave a similar but more modest response, whereas E2 did not alter cell size. Effects of NE dominated when used with either E2 or DHT with all combinations. Both sex hormones alone rapidly activated FAK but not ERK. Long-term or brief exposure to E2 attenuated NE-induced FAK phosphorylation, whereas DHT had no effect. Neither hormone altered NE-elicited ERK activation. Longer term exposure to E2 alone reduced FAK phosphorylation and reduced nuclear phospho-FAK, whereas its elevation was seen in the presence of NE with both sex hormones. The mitigating effects of E2 on the NE-elicited increase in cell size and the hypertrophic effect of DHT in NRVMs are in accordance with results observed in whole animal models. This is the first report of rapid, nongenomic sex hormone signaling via FAK activation and altered FAK trafficking to the nucleus in heart cells.
Collapse
Affiliation(s)
- Yevgeniya E Koshman
- Department of Physiology and Biophysics, University of Illinois at Chicago, MC 901, 835 South Wolcott Ave., Chicago, IL 60612-7342, USA
| | | | | | | |
Collapse
|
37
|
Abstract
Biological sex plays an important role in normal cardiac physiology as well as in the heart's response to cardiac disease. Women generally have better cardiac function and survival than do men in the face of cardiac disease; however, this sex difference is lost when comparing postmenopausal women with age-matched men. Animal models of cardiac disease mirror what is seen in humans. Sex steroid hormones contribute significantly to sex-based differences in cardiac disease outcomes. Estrogen is generally considered to be cardioprotective, whereas testosterone is thought to be detrimental to heart function. Environmental estrogen-like molecules, such as phytoestrogens, can also affect cardiac physiology in both a positive and a negative manner.
Collapse
Affiliation(s)
- Elizabeth D Luczak
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado 80309, USA.
| | | |
Collapse
|
38
|
Baumann PQ, Sobel BE, Tarikuz Zaman A, Schneider DJ. Gender-Dependent Differences in Echocardiographic Characteristics of Murine Hearts. Echocardiography 2008; 25:739-48. [DOI: 10.1111/j.1540-8175.2008.00680.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
39
|
Haddad GE, Saunders LJ, Crosby SD, Carles M, del Monte F, King K, Bristow MR, Spinale FG, Macgillivray TE, Semigran MJ, Dec GW, Williams SA, Hajjar RJ, Gwathmey JK. Human cardiac-specific cDNA array for idiopathic dilated cardiomyopathy: sex-related differences. Physiol Genomics 2008; 33:267-77. [DOI: 10.1152/physiolgenomics.00265.2007] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Idiopathic dilated cardiomyopathy (IDCM) constitutes a large portion of patients with heart failure of unknown etiology. Up to 50% of all transplant recipients carry this clinical diagnosis. Female-specific gene expression in IDCM has not been explored. We report sex-related differences in the gene expression profile of ventricular myocardium from patients undergoing cardiac transplantation. We produced and sequenced subtractive cDNA libraries, using human left ventricular myocardium obtained from male transplant recipients with IDCM and nonfailing human heart donors. With the resulting sequence data, we generated a custom human heart failure microarray for IDCM containing 1,145 cardiac-specific oligonucleotide probes. This array was used to characterize RNA samples from female IDCM transplant recipients. We identified a female gene expression pattern that consists of 37 upregulated genes and 18 downregulated genes associated with IDCM. Upon functional analysis of the gene expression pattern, deregulated genes unique to female IDCM were those that are involved in energy metabolism and regulation of transcription and translation. For male patients we found deregulation of genes related to muscular contraction. These data suggest that 1) the gene expression pattern we have detected for IDCM may be specific for this disease and 2) there is a sex-specific profile to IDCM. Our observations further suggest for the first time ever novel targets for treatment of IDCM in women and men.
Collapse
Affiliation(s)
- Georges E. Haddad
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, District of Columbia
| | | | - Seth D. Crosby
- Microarray Core Facility, Washington University Medical School, St. Louis, Missouri
| | - Maria Carles
- Gwathmey, Incorporated, Cambridge, Massachusetts
| | - Federica del Monte
- Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Kindra King
- Gwathmey, Incorporated, Cambridge, Massachusetts
| | - Michael R. Bristow
- Division of Cardiology, School of Medicine, University of Colorado Health Sciences Center, Denver, Colorado
| | - Francis G. Spinale
- Cardiothoracic Surgery, Medical University of South Carolina, Charleston, South Carolina
| | | | - Marc J. Semigran
- Cardiology Division, Gray/Bigelow, Massachusetts General Hospital, Boston
| | - G. William Dec
- Cardiology Division, Gray/Bigelow, Massachusetts General Hospital, Boston
| | - Steven A. Williams
- Department of Biological Sciences, Smith College, Northampton, Massachusetts
| | - Roger J. Hajjar
- Cardiovascular Research Center, Mount Sinai School of Medicine, New York, New York
| | - Judith K. Gwathmey
- Gwathmey, Incorporated, Cambridge, Massachusetts
- Boston University School of Medicine, Cambridge, Massachusetts
| |
Collapse
|
40
|
Yang Y, Ma Y, Han W, Li J, Xiang Y, Liu F, Ma X, Zhang J, Fu Z, Su YD, Du XJ, Gao XM. Age-related differences in postinfarct left ventricular rupture and remodeling. Am J Physiol Heart Circ Physiol 2008; 294:H1815-22. [PMID: 18263717 DOI: 10.1152/ajpheart.00831.2007] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cardiac rupture is more prevalent in elderly patients with first onset of acute myocardial infarct (MI), but the mechanism remains unexplored. We investigated the differences in the incidence of cardiac rupture and early left ventricular (LV) remodeling following coronary artery ligation between old (12-mo) and young (3-mo) C57Bl/6 male mice and explored responsible mechanisms. The incidence of rupture within 1 wk after MI was significantly higher in old than in young mice (40.7 vs. 18.3%, P = 0.013) despite a similar infarct size in both age groups. Old mice dying of rupture had more severe infarct expansion than young counterparts. Echocardiography and catheterization at day 7 revealed more profound LV chamber dilatation and dysfunction as well as higher blood pressures in aged mice. At day 3 after MI immediately before the peak of rupture occurrence, we observed significantly higher content of type I and III collagen, a greater density of macrophage and neutrophil, and markedly enhanced mRNA expression of inflammatory cytokines in the infarcted myocardium in old than in young mice. Furthermore, a more dramatic increment of matrix metalloproteinase (MMP)-9 activity was found in old than in young infarcted hearts, in keeping with enhanced inflammatory response. Collectively, these results revealed that old mice had a higher risk of post-MI cardiac rupture despite a higher level of collagen content and cross-linking. Enhanced inflammatory response and subsequent increase in MMP-9 activity together with higher blood pressure are important factors responsible for the higher risk of cardiac rupture and more severe LV remodeling in the aged heart following acute MI.
Collapse
Affiliation(s)
- Yining Yang
- Cardiovascular Research Institute, Xinjiang Medical University, Xinjinag, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Bathgate RAD, Lekgabe ED, McGuane JT, Su Y, Pham T, Ferraro T, Layfield S, Hannan RD, Thomas WG, Samuel CS, Du XJ. Adenovirus-mediated delivery of relaxin reverses cardiac fibrosis. Mol Cell Endocrinol 2008; 280:30-8. [PMID: 17961912 DOI: 10.1016/j.mce.2007.09.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2007] [Revised: 09/07/2007] [Accepted: 09/12/2007] [Indexed: 11/16/2022]
Abstract
We have evaluated the effectiveness of systemic adenovirally delivered mouse relaxin on reversing fibrosis in a transgenic murine model of fibrotic cardiomyopathy due to beta(2)-adrenergic receptor (beta(2)AR) overexpression. Recombinant adenoviruses expressing green fluorescent protein (Ad-GFP), rat relaxin (Ad-rRLN) and mouse relaxin (Ad-mRLN) were generated and Ad-rRLN and Ad-mRLN were demonstrated to direct the expression of bioactive relaxin peptides in vitro. A single systemic injection of Ad-mRLN resulted in transgene expression in the liver and bioactive relaxin peptide in the plasma. Ad-mRLN, but not Ad-GFP, treatment reversed the increased left ventricular collagen content in beta(2)AR mice to control levels without affecting collagen levels in other heart chambers or in the lung and kidney. Hence a single systemic injection of adenovirus producing mouse relaxin reverses cardiac fibrosis without adversely affecting normal collagen levels in other organs and establishes the potential for the use of relaxin gene therapy for the treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- R A D Bathgate
- Howard Florey Institute, University of Melbourne, Victoria 3010, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Zhou J, Xu X, Liu JJ, Lin YX, Gao GD. Angiotensin II receptors subtypes mediate diverse gene expression profile in adult hypertrophic cardiomyocytes. Clin Exp Pharmacol Physiol 2007; 34:1191-8. [PMID: 17880376 DOI: 10.1111/j.1440-1681.2007.04694.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
1. Although the systemic and cardiac renin-angiotensin systems are known to be activated in the setting of pressure overload, the actions and signaling mechanisms of angiotensin (Ang) II via AT(1) and AT(2) receptors in hypertrophic cardiomyocytes (CM) remain largely unclear. 2. Hypertrophic CM were prepared from rats with aortic banding for 8 weeks, cultured and then treated as follows: (i) 1 micromol/L AngII for 24 h; (ii) 10 micromol/L losartan (an AT(1) receptor antagonist) for 1 h followed by 1 micromol/L AngII for 24 h; and (iii) 10 micromol/L PD123319 (an AT(2) receptor antagonist) for 1 h followed by 1 micromol/L AngII for 24 h. Changes in the expression of genes following stimulation of AT(1) and AT(2) receptors specific to G-protein-coupled receptor (GPCR) signaling pathways were tested using GEArray (Superarray, Bethesda, MD, USA). The effects of AngII, acting via AT(1) and AT(2) receptors, on the expression of tumour necrosis factor (TNF)-alpha, interleukin (IL)-1beta and IL-6 were confirmed by reverse transcription-polymerase chain reaction and radioimmunoassay. 3. The genes regulated via stimulation of AT(1) receptors were mainly restricted to the signaling pathways including cAMP/protein kinase (PK) A, Ca(2+), PKC, protein tyrosine kinase, mitogen-activated protein kinases, phosphatidylinositol 3-kinase and nuclear factor-kappaB. In addition to these pathways related to activation of AT(1) receptors, four additional signaling pathways were found to be associated with stimulation of AT(2) receptors, including phospholipase C, nitric oxide/cGMP, Rho and Janus kinase/signal transducer and activator of transcription. Blockade of AT(2) receptors decreased the mRNA and protein expression of TNF-alpha and IL-1beta, whereas blockade of AT(1) receptors had no such effect. 4. In conclusion, in hypertrophic CM, AngII leads to distinct signaling responses mediated by AT(1) and AT(2) receptors. Stimulation of AT(2) receptors appears to have a greater influence on GPCR-signaling than stimulation of AT(1) receptors. Angiotensin II enhances the synthesis and secretion of TNF-alpha and IL-1beta in hypertrophic CM, which is mediated by AT(2), but not AT(1), receptors.
Collapse
MESH Headings
- Angiotensin II/metabolism
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Angiotensin Receptor Antagonists
- Animals
- Aorta, Abdominal/surgery
- Cardiomegaly/genetics
- Cardiomegaly/metabolism
- Cardiomegaly/pathology
- Cells, Cultured
- Disease Models, Animal
- Gene Expression Profiling
- Gene Expression Regulation/drug effects
- Imidazoles/pharmacology
- Interleukin-1beta/genetics
- Interleukin-1beta/metabolism
- Interleukin-6/genetics
- Interleukin-6/metabolism
- Ligation
- Losartan/pharmacology
- Male
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Oligonucleotide Array Sequence Analysis
- Pyridines/pharmacology
- RNA, Messenger/metabolism
- Radioimmunoassay
- Rats
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1/drug effects
- Receptor, Angiotensin, Type 1/metabolism
- Receptors, Angiotensin/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Time Factors
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Juan Zhou
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | | | | | | | | |
Collapse
|
43
|
Abstract
Clinical observations made over several decades support the existence of gender differences in cardiovascular disease prevalence and severity. For example, women exhibit a delay in the onset of vascular disease compared to men and the temporal link between menopause and the rise in vascular events in women suggests that ovarian hormones may be important in reducing the risk of vascular disease in women. Gender differences have also been observed in the severity and outcome of myocardial diseases such that women with heart failure have a better prognosis than men coupled with gender-specific patterns of ventricular remodeling. These clinical observations have fostered great interest in understanding the mechanisms of gender differences in cardiovascular diseases with the goal being to identify novel therapeutic targets. The purpose of this review is to describe animal models of cardiovascular disease that have demonstrated clear gender differences in the pathophysiologic responses to a given stimulus. Animal models from two broad areas of cardiovascular investigation will be highlighted: vascular disease and heart failure.
Collapse
|
44
|
Kocić I, Gruchała M, Petrusewicz J. Pretreatment of male guinea pigs by 17-beta-estradiol induces hypersensitivity of beta-adrenoceptors in electrically driven left atria. Int J Cardiol 2007; 129:22-5. [PMID: 17707530 DOI: 10.1016/j.ijcard.2007.05.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2006] [Revised: 04/26/2007] [Accepted: 05/11/2007] [Indexed: 11/29/2022]
Abstract
BACKGROUND It is well known that estrogen can modulate distribution and function of adrenergic receptors in the heart of different species. We reported here gender differences in adrenergic responsiveness of electrically driven guinea pig left atria. METHODS Experiments were performed on the guinea pigs divided in four groups: males control (MC), males treated by 17-beta-estradiol (MTE), females control (FC) and females treated by tamoxifen (FTT). After two weeks of treatment, the animals were sacrificed, the left atria were isolated and force of contraction (Fc), velocity of contraction (+dF/dt), velocity of relaxation (-dF/dt) and time to peak contraction (ttp) and relaxation time at 10% of amplitude (tt(10) ) were measured. RESULTS Apart from significantly lower Fc and longer tt10 in FC (0.97+/-0.12 mN, 233+/-7 ms, respectively) vs. MC (1.66+/-0.3, 176.3+/-18 ms, respectively, n=6, P<0.05), isoprenaline (ISO) and noradrenaline (NOR) (in the presence of prazosine) concentration-response curves were strongly shifted leftward in comparison with male group. Additionally, the maximal effects of. NOR was significantly lower in FC (about 40%) than in MC. Application of 17-beta-estradiol to males and tamoxifen to females guinea pigs confirmed crucial role of estrogen in observed phenomenon. CONCLUSION Our results indicate that estrogen not only downregulates beta1-adrenoceptors, but induces its hypersensitivity to catecholamines, at least in guinea pig left atria.
Collapse
Affiliation(s)
- Ivan Kocić
- Department of Toxicology and Clinical Pharmacology, Inter-Faculty Institute of Maritime and Tropical Medicine, Powstania Styczniowego 9b, 81-519 Gdynia, Poland.
| | | | | |
Collapse
|
45
|
Xu Q, Ming Z, Dart AM, Du XJ. OPTIMIZING DOSAGE OF KETAMINE AND XYLAZINE IN MURINE ECHOCARDIOGRAPHY. Clin Exp Pharmacol Physiol 2007; 34:499-507. [PMID: 17439422 DOI: 10.1111/j.1440-1681.2007.04601.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
1. Ketamine and xylazine (KX) mixture is the most commonly used anaesthetic drug during echocardiography in mice to induce sedation and immobility. Nevertheless, the doses of KX reported in the literature vary substantially with associated significant difference in cardiac function. To explore the optimal KX dosage and observation time for murine echocardiography, we compared the effects of various KX combinations on echocardiographic measurement. 2. Mice were anaesthetized with ketamine (50 or 100 mg/kg) and xylazine (0-10 mg/kg). Echocardiography was performed 5, 10, 20 and 40 min after induction of anaesthesia. Also, cardiac function was assessed in mice with and without pressure-overload induced left ventricle (LV) hypertrophy and dysfunction, either under anaesthesia with KX or whilst conscious. 3. Ketamine at 100 mg/kg alone or together with xylazine at 0.1 mg/kg was associated with a high and stable heart rate (HR), a high fractional shortening (FS) and produced the least effect on LV inner dimension at end of diastole (LVIDd). Ketamine and xylazine at 100 and 10 mg/kg, respectively, produced a lower and stable FS, but with a low and unstable HR. All other combinations resulted in depressed and unstable cardiac function during this period. 4. The dose-dependent suppression of FS by xylazine was counteracted partly by ketamine. 5. Although in the chronic pressure-overload model LV hypertrophy can be detected accurately in both the anaesthetized or conscious state, systolic dysfunction was masked partially by higher doses of xylazine (2.5 or 10 mg/kg) combined with ketamine at 100 mg/kg. 6. With KX anaesthesia, both the dose of xylazine and the anaesthetic duration are critical in achieving an ideal condition for murine echocardiography. Ketamine at 100 mg/kg alone produces acceptable anaesthesia, stable cardiac function with a minimal depressant effect and is therefore recommended if single-dose anaesthetic is to be used.
Collapse
Affiliation(s)
- Qi Xu
- Experimental Cardiology Laboratory, Baker Heart Research Institute and Alfred Heart Centre, Alfred Hospital, Melbourne, Victoria, Australia.
| | | | | | | |
Collapse
|
46
|
Fang L, Moore XL, Gao XM, Dart AM, Lim YL, Du XJ. Down-regulation of mitofusin-2 expression in cardiac hypertrophy in vitro and in vivo. Life Sci 2007; 80:2154-2160. [PMID: 17499311 DOI: 10.1016/j.lfs.2007.04.003] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2006] [Revised: 03/22/2007] [Accepted: 04/10/2007] [Indexed: 11/26/2022]
Abstract
Mitofusin-2 (Mfn2) suppresses smooth muscle cell proliferation through inhibition of the Ras-extracellular signal-regulated kinases (ERK1/2) pathway. Since the ERK1/2 pathway is implicated in mediating hypertrophic signaling, we studied the changes in Mfn2 in cardiac hypertrophy using in vitro and in vivo models. Phenylephrine was used to induce hypertrophy in neonatal rat ventricular myocytes (NRVMs). In vivo hypertrophy models included spontaneously hypertensive rats (SHR), pressure-overload hypertrophy by transverse aortic constriction (TAC), hypertrophy of non-infarcted myocardium following myocardial infarction (MI), and cardiomyopathy due to cardiac-restricted overexpression of beta(2)-adrenergic receptors (beta(2)-TG). We determined hypertrophic parameters and analysed expression of atrial natriuretic peptide (ANP) and Mfn2 by real-time PCR. Phosphorylated-ERK1/2 (phospho-ERK) was measured by Western blot. Mfn2 was downregulated in phenylephrine treated NRCMs (by approximately 40%), hypertrophied hearts from SHR (by approximately 80%), mice with TAC (at 1 and 3 weeks, by approximately 50%), and beta(2)-TG mice (by approximately 20%). However, Mfn2 was not downregulated in hypertrophied hearts with 15 weeks of TAC, nor in hypertrophied non-infarcted myocardium following MI. phospho-ERK1/2 was increased in hypertrophied myocardium at 1 week post-TAC, but not in non-infarcted myocardium after MI, indicating that downregulated Mfn2 may be accompanied by an increase of phospho-ERK1/2. This study shows, for the first time, downregulated Mfn2 expression in hypertrophied hearts, which depends on the etiology and time course of hypertrophy. Further study is required to examine the causal relationship between Mfn2 and cardiac hypertrophy.
Collapse
Affiliation(s)
- Lu Fang
- Baker Heart Research Institute and Alfred Heart Centre, Alfred Hospital, Melbourne, Australia; Western Hospital, Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Xiao-Lei Moore
- Baker Heart Research Institute and Alfred Heart Centre, Alfred Hospital, Melbourne, Australia
| | - Xiao-Ming Gao
- Baker Heart Research Institute and Alfred Heart Centre, Alfred Hospital, Melbourne, Australia
| | - Anthony M Dart
- Baker Heart Research Institute and Alfred Heart Centre, Alfred Hospital, Melbourne, Australia
| | - Yean Leng Lim
- Western Hospital, Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Xiao-Jun Du
- Baker Heart Research Institute and Alfred Heart Centre, Alfred Hospital, Melbourne, Australia.
| |
Collapse
|
47
|
Luckey SW, Mansoori J, Fair K, Antos CL, Olson EN, Leinwand LA. Blocking cardiac growth in hypertrophic cardiomyopathy induces cardiac dysfunction and decreased survival only in males. Am J Physiol Heart Circ Physiol 2006; 292:H838-45. [PMID: 17012357 DOI: 10.1152/ajpheart.00615.2006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mutations in myosin heavy chain (MyHC) can cause hypertrophic cardiomyopathy (HCM) that is characterized by hypertrophy, histopathology, contractile dysfunction, and sudden death. The signaling pathways involved in the pathology of HCM have not been elucidated, and an unresolved question is whether blocking hypertrophic growth in HCM may be maladaptive or beneficial. To address these questions, a mouse model of HCM was crossed with an antihypertrophic mouse model of constitutive activated glycogen synthase kinase-3beta (caGSK-3beta). Active GSK-3beta blocked cardiac hypertrophy in both male and female HCM mice. However, doubly transgenic males (HCM/GSK-3beta) demonstrated depressed contractile function, reduced sarcoplasmic (endo) reticulum Ca(2+)-ATPase (SERCA) expression, elevated atrial natriuretic factor (ANF) expression, and premature death. In contrast, female HCM/GSK-3beta double transgenic mice exhibited similar cardiac histology, function, and survival to their female HCM littermates. Remarkably, dietary modification from a soy-based diet to a casein-based diet significantly improved survival in HCM/GSK-3beta males. These findings indicate that activation of GSK-3beta is sufficient to limit cardiac growth in this HCM model and the consequence of caGSK-3beta was sexually dimorphic. Furthermore, these results show that blocking hypertrophy by active GSK-3beta in this HCM model is not therapeutic.
Collapse
Affiliation(s)
- Stephen W Luckey
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Campus Box 347, Boulder, Colorado 80309-0347, USA
| | | | | | | | | | | |
Collapse
|
48
|
Siu CW, Yeung CY, Lau CP, Kung AWC, Tse HF. Incidence, clinical characteristics and outcome of congestive heart failure as the initial presentation in patients with primary hyperthyroidism. Heart 2006; 93:483-7. [PMID: 17005710 PMCID: PMC1861478 DOI: 10.1136/hrt.2006.100628] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND There are limited systematic data on the incidence, clinical characteristics and outcomes of congestive heart failure (CHF) in patients with hyperthyroidism. The aim of this study was to investigate the incidence, clinical characteristics and outcome of CHF as the initial presentation in patients with primary hyperthyroidism. METHODS The prevalence, clinical characteristics and outcome of CHF was studied in 591 consecutive patients (mean (SD) age 45 (1) years, 140 men) who presented with primary hyperthyroidism. RESULTS CHF was the presenting condition in 34 patients (5.8%) with hyperthyroidism. The presence of atrial fibrillation at presentation (OR 37.4, 95% CI 9.72 to 144.0, p<0.001) was an independent predictor for the occurrence of CHF. Of the 34 patients with CHF, 16 (47%) had systolic left ventricular dysfunction with left ventricular ejection fraction (LVEF)<50%. They were predominantly male (OR 26.6, 95% CI 2.6 to 272.5, p = 0.006) and had a lower serum thyroxine level (OR 0.93, 95% CI 0.87 to 0.99, p = 0.044) than patients with preserved left ventricular systolic function. In these patients, LVEF (55 (4)% vs 30 (2)%, p<0.001) and New York Heart Association functional class (1.2 (0.1) vs 2.5 (0.2), p<0.001) improved significantly 3 months after achieving euthyroid status. Systolic left ventricular dysfunction (mean (SD) LVEF 38 (4)%) persisted on long-term follow-up in five PATIENTS no clinical parameter could be identified to predict the occurrence of this persistent cardiomyopathy (p>0.05). CONCLUSION CHF was the initial clinical presentation in approximately 6% of patients with hyperthyroidism, and half of them had left ventricular systolic dysfunction. Symptoms of CHF subsided and LVEF improved after treatment for hyperthyroidism. Nonetheless, one-third of these patients developed persistent dilated cardiomyopathy.
Collapse
Affiliation(s)
- Chung-Wah Siu
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | | | | | | | | |
Collapse
|
49
|
Gao XM, Wong G, Wang B, Kiriazis H, Moore XL, Su YD, Dart A, Du XJ. Inhibition of mTOR reduces chronic pressure-overload cardiac hypertrophy and fibrosis. J Hypertens 2006; 24:1663-70. [PMID: 16877971 DOI: 10.1097/01.hjh.0000239304.01496.83] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND AND OBJECTIVE Inhibition of established left ventricular hypertrophy (LVH) and fibrosis may bring clinical benefits by reducing cardiac morbidity and mortality. The mammalian target of rapamycin, mTOR, is known to play a critical role in determining cell and organ size. We investigated whether mTOR inhibition can inhibit the chronic pressure-overload-induced LVH and fibrosis. METHODS Male FVB/N mice underwent transverse aortic constriction (TAC) for 5 weeks to allow for establishment of LVH, followed by treatment with the mTOR inhibitor, Rapamune (2 mg/kg per day, gavage), for 4 weeks. Echocardiography was used to monitor changes in LVH and function. Haemodynamic, morphometric, histological and molecular analyses were conducted. RESULTS Inhibition of mTOR by Rapamune was confirmed by a suppression of activated phosphorylation of ribosomal S6 protein and eukaryotic translation initiation factor-4E due to pressure overload. Despite a comparable degree of pressure overload between the vehicle- or Rapamune-treated TAC groups, Rapamune treatment for 4 weeks attenuated TAC-induced LVH by 46%, estimated by LV weight or myocyte size, and LV fractional shortening was also preserved versus vehicle-treated control (39 +/- 1 versus 32 +/- 2%, P < 0.05). Inhibition of established LVH by Rapamune was associated with a 38% reduction in collagen content. Moreover, altered gene expression due to pressure overload was largely restored. CONCLUSION Despite sustained pressure overload, inhibition of mTOR by a 4-week period of Rapamune treatment attenuates chronically established LVH and cardiac fibrosis with preserved contractile function.
Collapse
Affiliation(s)
- Xiao-Ming Gao
- Experimental Cardiology Laboratory, Baker Heart Research Institute, Melbourne, Victoria, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Samuel CS, Du XJ, Bathgate RAD, Summers RJ. 'Relaxin' the stiffened heart and arteries: the therapeutic potential for relaxin in the treatment of cardiovascular disease. Pharmacol Ther 2006; 112:529-52. [PMID: 16814863 DOI: 10.1016/j.pharmthera.2005.05.012] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2005] [Accepted: 05/15/2005] [Indexed: 12/20/2022]
Abstract
Although originally characterised as a reproductive hormone, relaxin has emerged as a multi-functional endocrine and paracrine factor that plays a number of important roles in several organs, including the normal and diseased cardiovascular system. The recent discovery of the H3/relaxin-3 gene, and the elusive receptors for relaxin (Relaxin family peptide receptor; RXFP1) and relaxin-3 (RXFP3/RXFP4) have led to the re-classification of a distinct relaxin peptide/receptor family. Additionally, the identification of relaxin and RXFP1 mRNA and/or relaxin binding sites in the heart and blood vessels has confirmed that the cardiovascular system is a target for relaxin peptides. While evidence for the production of relaxins within the cardiovascular system is limited, several studies have established that the relaxin genes are upregulated in the diseased human and rodent heart where they likely act as cardioprotective agents. The ability of relaxin to protect the heart is most likely mediated via its antifibrotic, anti-hypertrophic, anti-inflammatory and vasodilatory actions, but it may also directly stimulate myocardial regeneration and repair. This review describes relaxin and its primary receptor (RXFP1) in relation to the roles and effects of relaxin in the normal and pathological cardiovascular system. It is becoming increasingly clear that relaxin has a number of diverse physiological and pathological roles in the cardiovascular system that may have important therapeutic and clinical implications.
Collapse
Affiliation(s)
- Chrishan S Samuel
- Howard Florey Institute, University of Melbourne, Victoria 3010, Australia; Department of Biochemistry and Molecular Biology, University of Melbourne, Victoria 3010, Australia
| | | | | | | |
Collapse
|