Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
2.
J Natl Compr Canc Netw ; : 1-10, 2020 Nov 03.
Article in English | MEDLINE | ID: mdl-33142266

ABSTRACT

BACKGROUND: Cancer and cardiovascular disease (CVD) are independently associated with adverse outcomes in patients with COVID-19. However, outcomes in patients with COVID-19 with both cancer and comorbid CVD are unknown. METHODS: This retrospective study included 2,476 patients who tested positive for SARS-CoV-2 at 4 Massachusetts hospitals between March 11 and May 21, 2020. Patients were stratified by a history of either cancer (n=195) or CVD (n=414) and subsequently by the presence of both cancer and CVD (n=82). We compared outcomes between patients with and without cancer and patients with both cancer and CVD compared with patients with either condition alone. The primary endpoint was COVID-19-associated severe disease, defined as a composite of the need for mechanical ventilation, shock, or death. Secondary endpoints included death, shock, need for mechanical ventilation, need for supplemental oxygen, arrhythmia, venous thromboembolism, encephalopathy, abnormal troponin level, and length of stay. RESULTS: Multivariable analysis identified cancer as an independent predictor of COVID-19-associated severe disease among all infected patients. Patients with cancer were more likely to develop COVID-19-associated severe disease than were those without cancer (hazard ratio [HR], 2.02; 95% CI, 1.53-2.68; P<.001). Furthermore, patients with both cancer and CVD had a higher likelihood of COVID-19-associated severe disease compared with those with either cancer (HR, 1.86; 95% CI, 1.11-3.10; P=.02) or CVD (HR, 1.79; 95% CI, 1.21-2.66; P=.004) alone. Patients died more frequently if they had both cancer and CVD compared with either cancer (35% vs 17%; P=.004) or CVD (35% vs 21%; P=.009) alone. Arrhythmias and encephalopathy were also more frequent in patients with both cancer and CVD compared with those with cancer alone. CONCLUSIONS: Patients with a history of both cancer and CVD are at significantly higher risk of experiencing COVID-19-associated adverse outcomes. Aggressive public health measures are needed to mitigate the risks of COVID-19 infection in this vulnerable patient population.

3.
Clin Cardiol ; 43(12): 1494-1500, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32940385

ABSTRACT

BACKGROUND: Left bundle branch block (LBBB) and left ventricular (LV) dyssynchrony likely contribute to progressive systolic dysfunction. The evaluation of newly recognized LBBB includes screening for structural heart abnormalities and coronary artery disease (CAD). In patients whose LV ejection fraction (EF) is preserved during initial testing, the incidence of subsequent cardiomyopathy is not firmly established. HYPOTHESIS: The risk of developing LV systolic dysfunction among LBBB patients with preserved LVEF is high enough to warrant serial imaging. METHODS: We screened records of 1000 consecutive patients with LBBB from our ECG database and identified subjects with an initially preserved LVEF (≥45%) without clinically relevant CAD or other cause for cardiomyopathy. Baseline imaging, clinical data, and follow-up imaging were recorded to determine the risk of subsequent LV systolic dysfunction (LVEF ≤40%). RESULTS: (Data are mean + SD) 784 subjects were excluded, the majority for CAD or depressed LVEF upon initial imaging. Of the remaining 216, 37 (17%) developed a decline in LVEF(≤40%) over a mean follow-up of 55 ± 31 months; 94% of these patients had a baseline LVEF≤60% and LV end systolic diameter (ESD) ≥ 2.9 cm indicating that these measures may be useful to define which patients warrant longitudinal follow-up. The negative predictive value of a LVEF>60% and LVESD <2.9 cm was 98%. CONCLUSIONS: Seventeen percent of patients with LBBB and initial preserved LVEF develop dyssynchrony cardiomyopathy. We believe the risk of developing dyssynchrony cardiomyopathy is high enough to warrant serial assessment of LV systolic function in this high-risk population.


Subject(s)
Bundle-Branch Block/complications , Cardiomyopathies/etiology , Risk Assessment/methods , Stroke Volume/physiology , Ventricular Function, Left/physiology , Aged , Bundle-Branch Block/physiopathology , Cardiomyopathies/epidemiology , Cardiomyopathies/physiopathology , Female , Follow-Up Studies , Humans , Incidence , Male , Retrospective Studies , Risk Factors , United States/epidemiology
4.
Trends Cardiovasc Med ; 30(6): 315-325, 2020 08.
Article in English | MEDLINE | ID: mdl-32474135

ABSTRACT

Patients with pre-existing cardiovascular disease and risk factors are more likely to experience adverse outcomes associated with the novel coronavirus disease-2019 (COVID-19). Additionally, consistent reports of cardiac injury and de novo cardiac complications, including possible myocarditis, arrhythmia, and heart failure in patients without prior cardiovascular disease or significant risk factors, are emerging, possibly due to an accentuated host immune response and cytokine release syndrome. As the spread of the virus increases exponentially, many patients will require medical care either for COVID-19 related or traditional cardiovascular issues. While the COVID-19 pandemic is dominating the attention of the healthcare system, there is an unmet need for a standardized approach to deal with COVID-19 associated and other traditional cardiovascular issues during this period. We provide consensus guidance for the management of various cardiovascular conditions during the ongoing COVID-19 pandemic with the goal of providing the best care to all patients and minimizing the risk of exposure to frontline healthcare workers.


Subject(s)
Betacoronavirus , Cardiovascular Diseases/therapy , Coronavirus Infections/epidemiology , Pneumonia, Viral/epidemiology , COVID-19 , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/etiology , Humans , Pandemics , SARS-CoV-2
5.
Resuscitation ; 147: 53-56, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31891791

ABSTRACT

OBJECTIVES: We sought to determine the outcomes of patients with an Impella CP percutaneous mechanical circulatory support (MCS) device deployed during a cardiac arrest. BACKGROUND: The Impella CP device is indicated for left ventricular support in patients with cardiogenic shock. The utility of percutaneous MCS in the setting of cardiac arrest during cardiopulmonary resuscitation (CPR) remains unclear. METHODS: We retrospectively examined data from patients supported with an Impella CP device for cardiogenic shock complicated by cardiac arrest between April 2015 and April 2017 at a single academic medical center. Patients with cardiac arrest who underwent Impella CP placement during CPR were compared to those who had return of spontaneous circulation (ROSC) prior to Impella CP placement. RESULTS: We identified 22 patients with cardiogenic shock complicated by cardiac arrest (average age 64 years, 23% female) who underwent placement of an Impella CP device. The majority of patients (68%) underwent support for cardiogenic shock secondary to an acute myocardial infarction. Seven of the 22 patients (32%) underwent Impella CP placement during CPR and 15 (68%) underwent Impella CP insertion following ROSC. The in-hospital mortality was 86% in the group of patients who had the Impella CP placed during CPR and 56% in the group with ROSC prior to Impella CP insertion, (p = 0.19). CONCLUSIONS: Based on our single center retrospective analysis, the mortality rate of patients undergoing placement of an Impella CP during CPR is 86%. Further study is necessary to better understand the utility of the Impella CP mechanical circulatory support device during a cardiac arrest.


Subject(s)
Heart Arrest , Heart-Assist Devices , Myocardial Infarction , Female , Heart Arrest/therapy , Humans , Male , Middle Aged , Retrospective Studies , Shock, Cardiogenic/etiology , Shock, Cardiogenic/therapy
6.
Am J Physiol Cell Physiol ; 301(5): C1046-56, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21775709

ABSTRACT

Left ventricular remodeling, including the deposition of excess extracellular matrix, is key to the pathogenesis of heart failure. The stress-inducible transcriptional regulator p8 is increased in failing human hearts and is required both for agonist-stimulated cardiomyocyte hypertrophy and for cardiac fibroblasts matrix metalloprotease-9 (MMP9) induction. In the heart, upregulation of autophagy is an adaptive response to stress and plays a causative role in cardiomyopathies. We have recently shown that p8 ablation in cardiac cells upregulates autophagy and that, in vivo, loss of p8 results in a decrease of cardiac function. Here we investigated the effects of p8 genetic deletion in mediating adverse myocardial remodeling. Unstressed p8-/- mouse hearts manifested complex alterations in the expression of fibrosis markers. In addition, these mice displayed elevated autophagy and apoptosis compared with p8+/+ mice. Transverse aortic constriction (TAC) induced left ventricular p8 expression in p8+/+ mice. Pressure overload caused left ventricular remodeling in both genotypes, however, p8-/- mice showed less cardiac fibrosis induction. Consistent with this, although MMP9 induction was attenuated in the p8-/- mice, induction of MMP2 and MMP3 were strikingly upregulated while TIMP2 was downregulated. Left ventricular autophagy increased after TAC and was significantly higher in the p8-/- mice. Thus p8-deletion results in reduced collagen fibrosis after TAC, but in turn, is associated with a detrimental higher increase in autophagy. These findings suggest a role for p8 in regulating in vivo key signaling pathways involved in the pathogenesis of heart failure.


Subject(s)
Autophagy , DNA-Binding Proteins/metabolism , Matrix Metalloproteinase 9/biosynthesis , Myocardium/pathology , Neoplasm Proteins/metabolism , Ventricular Remodeling , Animals , DNA-Binding Proteins/genetics , Female , Fibrosis , Male , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 3/biosynthesis , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Neoplasm Proteins/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism
7.
Mol Biol Cell ; 21(8): 1335-49, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20181828

ABSTRACT

Autophagy is a cytoprotective pathway used to degrade and recycle cytoplasmic content. Dysfunctional autophagy has been linked to both cancer and cardiomyopathies. Here, we show a role for the transcriptional regulator p8 in autophagy. p8 RNA interference (RNAi) increases basal autophagy markers in primary cardiomyocytes, in H9C2 and U2OS cells, and decreases cellular viability after autophagy induction. This autophagy is associated with caspase activation and is blocked by atg5 silencing and by pharmacological inhibitors. FoxO3 transcription factor was reported to activate autophagy by enhancing the expression of autophagy-related genes. P8 expression represses FoxO3 transcriptional activity, and p8 knockdown affects FoxO3 nuclear localization. Thus, p8 RNAi increases FoxO3 association with bnip3 promoter, a known proautophagic FoxO3 target, resulting in higher bnip3 RNA and protein levels. Accordingly, bnip3 knockdown restores cell viability and blocks apoptosis of p8-deficient cells. In vivo, p8 -/- mice have higher autophagy and express higher cardiac bnip3 levels. These mice develop left ventricular wall thinning and chamber dilation, with consequent impaired cardiac function. Our studies provide evidence of a p8-dependent mechanism regulating autophagy by acting as FoxO3 corepressor, which may be relevant for diseases associated with dysregulated autophagy, as cardiovascular pathologies and cancer.


Subject(s)
Apoptosis , Autophagy , Basic Helix-Loop-Helix Transcription Factors/deficiency , DNA-Binding Proteins/deficiency , Heart Function Tests , Heart/physiopathology , Neoplasm Proteins/deficiency , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carrier Proteins , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Energy Metabolism/genetics , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Deletion , Gene Silencing , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Myocytes, Cardiac/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phenotype , Promoter Regions, Genetic/genetics , Protein Binding , Protein Stability , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Repressor Proteins/metabolism , Stress, Physiological/genetics , Transcriptional Activation/genetics , Ubiquitin-Protein Ligases
10.
J Am Coll Cardiol ; 53(3): 264-71, 2009 Jan 20.
Article in English | MEDLINE | ID: mdl-19147043

ABSTRACT

OBJECTIVES: This study sought to determine the relationship between pre-transplant ventricular assist device (VAD) support and mortality after heart transplantation. BACKGROUND: Increasingly, VADs are being used to bridge patients to heart transplantation. The effect of these devices on post-transplant mortality is unclear. METHODS: Patients 18 years or older who underwent first-time, single-organ heart transplantation in the U.S. between 1995 and 2004 were included in the analyses. This study compared 1,433 patients bridged with intracorporeal and 448 patients bridged with extracorporeal VADs with 9,455 United Network for Organ Sharing status 1 patients not bridged with a VAD with respect to post-transplant mortality. Because the proportional hazards assumption was not met, hazard ratios (HRs) for different time periods were estimated. RESULTS: Intracorporeal VADs were associated with an HR of 1.20 (95% confidence interval [CI]: 1.02 to 1.43; p = 0.03) for mortality in the first 6 months after transplant and an HR of 1.99 (95% CI: 1.44 to 2.75; p < 0.0001) beyond 5 years. Between 6 months and 5 years, the HRs were not significantly different from 1. Extracorporeal VADs were associated with an HR of 1.91 (95% CI: 1.53 to 2.37; p < 0.0001) for mortality in the first 6 months and an HR of 2.93 (95% CI: 1.19 to 7.25; p = 0.02) beyond 5 years. The HRs were not significantly different from 1 between 6 months and 5 years, except for an HR of 0.23 (95% CI: 0.06 to 0.91; p = 0.04) between 24 and 36 months. CONCLUSIONS: Extracorporeal VADs are associated with higher mortality within 6 months and again beyond 5 years after transplantation. Intracorporeal VADs are associated with a small increase in mortality in the first 6 months and a clinically significant increase in mortality beyond 5 years. These data do not provide evidence supporting VAD implantation in stable United Network for Organ Sharing status I patients awaiting heart transplantation.


Subject(s)
Cause of Death , Heart Transplantation/mortality , Heart-Assist Devices/adverse effects , Tissue and Organ Procurement , Adult , Age Factors , Cohort Studies , Female , Follow-Up Studies , Graft Rejection , Graft Survival , Heart Transplantation/methods , Humans , Male , Middle Aged , Preoperative Care , Probability , Reference Values , Registries , Risk Assessment , Sex Factors , Survival Analysis , United States , Waiting Lists
11.
Circ Res ; 104(2): 265-75, 11p following 275, 2009 Jan 30.
Article in English | MEDLINE | ID: mdl-19074476

ABSTRACT

Left ventricular (LV) hypertrophy commonly develops in response to chronic hypertension and is a significant risk factor for heart failure and death. The serine-threonine phosphatase calcineurin (Cn)A plays a critical role in the development of pathological hypertrophy. Previous experimental studies in murine models show that estrogen limits pressure overload-induced hypertrophy; our purpose was to explore further the mechanisms underlying this estrogen effect. Wild-type, ovariectomized female mice were treated with placebo or 17beta-estradiol (E2), followed by transverse aortic constriction (TAC), to induce pressure overload. At 2 weeks, mice underwent physiological evaluation, immediate tissue harvest, or dispersion of cardiomyocytes. E2 replacement limited TAC-induced LV and cardiomyocyte hypertrophy while attenuating deterioration in LV systolic function and contractility. These E2 effects were associated with reduced abundance of CnA. The primary downstream targets of CnA are the nuclear factor of activated T-cell (NFAT) family of transcription factors. In transgenic mice expressing a NFAT-activated promoter/luciferase reporter gene, E2 limited TAC-induced activation of NFAT. Moreover, the inhibitory effects of E2 on LV hypertrophy were absent in CnA knockout mice, supporting the notion that CnA is an important target of E2-mediated inhibition. In cultured rat cardiac myocytes, E2 inhibited agonist-induced hypertrophy while also decreasing CnA abundance and NFAT activation. Agonist stimulation also reduced CnA ubiquitination and degradation that was prevented by E2; all in vitro effects of estrogen were reversed by an estrogen receptor (ER) antagonist. These data support that E2 reduces pressure overload induced hypertrophy by an ER-dependent mechanism that increases CnA degradation, unveiling a novel mechanism by which E2 and ERs regulate pathological LV and cardiomyocyte growth.


Subject(s)
Calcineurin/metabolism , Estradiol/metabolism , Hypertrophy, Left Ventricular/prevention & control , Myocardium/enzymology , Receptors, Estrogen/metabolism , Signal Transduction , Animals , Animals, Newborn , Calcineurin/deficiency , Calcineurin/genetics , Cell Size , Cells, Cultured , Disease Models, Animal , Drug Implants , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Female , Fulvestrant , Hemodynamics , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocardial Contraction , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Ovariectomy , Phenylephrine/pharmacology , Proteasome Endopeptidase Complex/metabolism , Receptors, Estrogen/antagonists & inhibitors , Signal Transduction/drug effects , Time Factors , Ubiquitin/metabolism , Ventricular Function, Left , Ventricular Remodeling
12.
J Heart Lung Transplant ; 27(12): 1350-2, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19059117

ABSTRACT

Heart transplantation from donors with a history of cocaine abuse remains controversial. Therefore, we examined the consequence of donor cocaine-use history on all-cause mortality and the development of coronary artery disease after heart transplantation. Using the United Network for Organ Sharing Thoracic Registry we identified 9,217 first-time heart-only adult transplant recipients between January 1999 and December 2003, and then divided this cohort into sub-groups based on the reported history of donor cocaine use. Multivariate analysis revealed no difference in mortality or development of coronary artery disease at 1 and 5 years between transplant recipients who received an organ from donors with a history of cocaine use when compared with donors having no history of cocaine use.


Subject(s)
Cocaine-Related Disorders/complications , Heart Transplantation/physiology , Tissue Donors , Adult , Follow-Up Studies , Heart Transplantation/mortality , Humans , Proportional Hazards Models , Registries , Retrospective Studies , Risk Assessment , Survival Rate
13.
Circ Res ; 103(5): 493-501, 2008 Aug 29.
Article in English | MEDLINE | ID: mdl-18658259

ABSTRACT

Reduced expression of sarcoplasmic reticulum calcium ATPase (SERCA)2 and other genes in the adult cardiac gene program has raised consideration of an impaired responsiveness to thyroid hormone (T3) that develops in the advanced failing heart. Here, we show that human and murine cardiomyopathy hearts have increased expression of friend of GATA (FOG)-2, a cardiac nuclear hormone receptor corepressor protein. Cardiac-specific overexpression of FOG-2 in transgenic mice led to depressed cardiac function, activation of the fetal gene program, congestive heart failure, and early death. SERCA2 transcript and protein levels were reduced in FOG-2 transgenic hearts, and FOG-2 overexpression impaired T3-mediated SERCA2 expression in cultured cardiomyocytes. FOG-2 physically interacts with thyroid hormone receptor-alpha1 and abrogated even high levels of T3-mediated SERCA2 promoter activity. These results demonstrate that SERCA2 is an important target of FOG-2 and that increased FOG-2 expression may contribute to a decline in cardiac function in end-stage heart failure by impaired T3 signaling.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Heart Failure/physiopathology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Triiodothyronine/metabolism , Animals , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/metabolism , Cardiomyopathies/physiopathology , Cell Line , Echocardiography , Heart Failure/diagnostic imaging , Heart Failure/metabolism , Humans , Kidney/cytology , Mice , Mice, Inbred ICR , Mice, Transgenic , Myocytes, Cardiac/cytology , Oligonucleotide Array Sequence Analysis , Phenotype , Promoter Regions, Genetic/physiology , Rats , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Signal Transduction/physiology , Thyroid Hormone Receptors alpha/metabolism , Transcription, Genetic/physiology , Transfection
14.
J Card Fail ; 14(3): 245-53, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18381189

ABSTRACT

BACKGROUND: We have shown previously that 17beta-estradiol (E2) increases left ventricular (LV) and cardiomyocyte hypertrophy after myocardial infarction (MI). However, E2 decreases hypertrophy in pressure overload models. We hypothesized that the effect of estrogen on cardiac hypertrophy was dependent on the type of hypertrophic stimulus. METHODS AND RESULTS: Ovariectomized wild-type female mice (n = 192) were given vehicle or E2 treatment followed by coronary ligation (MI), transverse aortic constriction (TAC), or sham operation. Signaling pathway activation was studied at 3, 24, and 48 hours, whereas echocardiography and hemodynamic studies were performed at 14 days. MI induced early but transient activation of p38 and p42/44 MAPK pathways, whereas TAC induced sustained activation of both pathways. E2 had no effect on these pathways, but increased Stat3 activation after MI while decreasing Stat3 activation after TAC. MI caused LV dilation and decreased fractional shortening (FS) that were unaltered by E2. TAC caused LV dilation, reduced FS, and increased LV mass, but in this model, E2 improved these parameters. After MI, E2 led to increases in myocyte cross-sectional area, atrial natriuretic peptide (ANP) and beta-myosin heavy chain (MHC) gene expression, but E2 diminished TAC-induced increases ANP and beta-MHC gene expression. CONCLUSIONS: These data demonstrate that the effects of E2 on LV and myocyte remodeling depend on the nature of the hypertrophic stimulus. The opposing influence of E2 on hypertrophy in these models may, in part, result from differential effects of E2 on Stat3 activation. Further work will be necessary to explore this and other potential mechanisms by which estrogen affects hypertrophy in these models.


Subject(s)
Cardiomegaly/drug therapy , Estradiol/pharmacology , Myocardial Infarction/complications , Myocardial Infarction/drug therapy , Myocytes, Cardiac/drug effects , Analysis of Variance , Animals , Blotting, Western , Cardiomegaly/diagnostic imaging , Cardiomegaly/etiology , Cells, Cultured , Disease Models, Animal , Echocardiography, Transesophageal , Female , Hemodynamics/drug effects , Hemodynamics/physiology , Mice , Mice, Inbred C57BL , Myocardial Infarction/pathology , Ovariectomy , Probability , Proportional Hazards Models , Random Allocation , Reference Values , Sensitivity and Specificity , Ventricular Pressure/drug effects , Ventricular Remodeling/drug effects
15.
J Clin Invest ; 118(1): 259-71, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18060044

ABSTRACT

Parasympathetic stimulation of the heart, which provides protection from arrhythmias and sudden death, involves activation of the G protein-coupled inward rectifying K+ channel GIRK1/4 and results in an acetylcholine-sensitive K+ current, I KACh. We describe a unique relationship between lipid homeostasis, the lipid-sensitive transcription factor SREBP-1, regulation of the cardiac parasympathetic response, and the development of ventricular arrhythmia. In embryonic chick atrial myocytes, lipid lowering by culture in lipoprotein-depleted serum increased SREBP-1 levels, GIRK1 expression, and I KACh activation. Regulation of the GIRK1 promoter by SREBP-1 and lipid lowering was dependent on interaction with 2 tandem sterol response elements and an upstream E-box motif. Expression of dominant negative SREBP-1 (DN-SREBP-1) reversed the effect of lipid lowering on I KACh and GIRK1. In SREBP-1 knockout mice, both the response of the heart to parasympathetic stimulation and the expression of GIRK1 were reduced compared with WT. I KACh, attenuated in atrial myocytes from SREBP-1 knockout mice, was stimulated by SREBP-1 expression. Following myocardial infarction, SREBP-1 knockout mice were twice as likely as WT mice to develop ventricular tachycardia in response to programmed ventricular stimulation. These results demonstrate a relationship between lipid metabolism and parasympathetic response that may play a role in arrhythmogenesis.


Subject(s)
Lipid Metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Parasympathetic Nervous System/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Acetylcholine/genetics , Acetylcholine/metabolism , Animals , Cells, Cultured , Chickens , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Heart Atria/innervation , Heart Atria/metabolism , Heart Atria/pathology , Ion Transport/genetics , Lipid Metabolism/genetics , Lipoproteins/metabolism , Mice , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/pathology , Myocytes, Cardiac/pathology , Parasympathetic Nervous System/pathology , Potassium/metabolism , Response Elements/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/pathology , Transcription, Genetic/genetics , Ventricular Fibrillation/genetics , Ventricular Fibrillation/metabolism , Ventricular Fibrillation/pathology
16.
Hypertension ; 49(5): 1000-6, 2007 May.
Article in English | MEDLINE | ID: mdl-17372038

ABSTRACT

Left ventricular (LV) mass and other LV measures have been shown to be heritable. In this study we hypothesized that functional variation in the gene coding for estrogen receptor-alpha (ESR1), known for mediating the effect of estrogens on myocardium, is associated with age-related changes in LV structure. Four genetic markers (ESR1 TA repeat; rs2077647, or +30T>C; rs2234693, or PvuII; and rs9340799, or XbaI) were genotyped in 847 unrelated individuals (488 women) from the Framingham Offspring Study, who attended 2 examination cycles 16 years apart (mean ages at first examination: 43+/-9 years; at follow-up: 59+/-9 years). ANCOVA was used to assess the association of polymorphisms and their haplotypes with cross-sectional measurements and longitudinal changes in LV mass, wall thickness, end-diastolic and end-systolic internal diameter, and fractional shortening after adjustment for factors known to influence these variables. Changes over time were detected for all of the LV measurements (P ranging from <0.0001 to 0.02), except for fractional shortening in men. The SS genotype of the ESR1 TA repeat polymorphism in the promoter region was associated with longitudinal changes in LV mass and LV wall thickness (P ranging from 0.0006 to 0.01). Moreover, the TA[S]-+30[T]-PvuII[T]-XbaI[A] haplotype (frequency: 47.5%) was associated with greater LV changes as compared with the TA[L]-+30[C]-PvuII[C]-XbaI[G] haplotype (frequency: 31.8%). Our results are consistent with the hypothesis that common ESR1 polymorphisms are significantly associated with age-related changes in LV structure. Understanding the mechanisms predisposing to unfavorable LV remodeling of the heart with advancing age may aid in the discovery of new therapeutic targets for the prevention of heart failure.


Subject(s)
Aging/physiology , Echocardiography , Estrogen Receptor alpha/genetics , Genetic Variation/physiology , Ventricular Remodeling/physiology , Adenine , Adult , Cohort Studies , Dinucleotide Repeats , Female , Genotype , Haplotypes , Humans , Male , Middle Aged , Promoter Regions, Genetic , Sex Factors , Thymine
17.
Mol Cell Biol ; 27(3): 993-1006, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17116693

ABSTRACT

Cardiomyocyte hypertrophy and extracellular matrix remodeling, primarily mediated by inflammatory cytokine-stimulated cardiac fibroblasts, are critical cellular events in cardiac pathology. The molecular components governing these processes remain nebulous, and few genes have been linked to both hypertrophy and matrix remodeling. Here we show that p8, a small stress-inducible basic helix-loop-helix protein, is required for endothelin- and alpha-adrenergic agonist-induced cardiomyocyte hypertrophy and for tumor necrosis factor-stimulated induction, in cardiac fibroblasts, of matrix metalloproteases (MMPs) 9 and 13-MMPs linked to general inflammation and to adverse ventricular remodeling in heart failure. In a stimulus-dependent manner, p8 associates with chromatin containing c-Jun and with the cardiomyocyte atrial natriuretic factor (anf) promoter and the cardiac fibroblast mmp9 and mmp13 promoters, established activator protein 1 effectors. p8 is also induced strongly in the failing human heart by a process reversed upon therapeutic intervention. Our results identify an unexpectedly broad involvement for p8 in key cellular events linked to cardiomyocyte hypertrophy and cardiac fibroblast MMP production, both of which occur in heart failure.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Fibroblasts/enzymology , Matrix Metalloproteinase 13/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Myocardium/cytology , Myocytes, Cardiac/pathology , Neoplasm Proteins/metabolism , Animals , Atrial Natriuretic Factor/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Chromatin/drug effects , Endothelin-1/pharmacology , Enzyme Induction/drug effects , Fibroblasts/drug effects , Gene Expression Regulation, Enzymologic/drug effects , HeLa Cells , Heart Failure/therapy , Humans , Hypertrophy , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 9/genetics , Myocytes, Cardiac/drug effects , Neoplasm Proteins/genetics , Phenylephrine/pharmacology , Promoter Regions, Genetic/drug effects , Protein Binding/drug effects , Proto-Oncogene Proteins c-jun/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/pharmacology
18.
Drug Discov Today Dis Models ; 4(4): 227-232, 2007.
Article in English | MEDLINE | ID: mdl-19081826

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.

19.
Mol Cell Biol ; 26(13): 5043-54, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16782890

ABSTRACT

Ischemia in the heart deprives cardiomyocytes of oxygen, triggering cell death (myocardial infarction). Ischemia and its cell culture model, hypoxia, elicit a stress response program that contributes to cardiomyocyte death; however, the molecular components required to promote this process remain nebulous. Gene 33 is a 50-kDa cytosolic adapter protein that suppresses signaling from receptor Tyr kinases of the epidermal growth factor receptor/ErbB family. Here we show that adenoviral expression of Gene 33 swiftly stimulates cardiomyocyte death coincident with reduced Akt and extracellular signal-regulated kinase (ERK) signaling. Subjecting cardiomyocytes to hypoxia and then reoxygenation induces gene 33 mRNA and Gene 33 protein. RNA interference experiments indicate that endogenous Gene 33 reduces Akt and ERK signaling and is required for maximal hypoxia-induced cardiomyocyte death. Gene 33 levels are also strikingly increased in myocardial ischemic injury and infarction. Our results identify a new role for Gene 33 as a component in the molecular pathophysiology of ischemic injury.


Subject(s)
Carrier Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Hypoxia/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Adenoviridae/genetics , Animals , Apoptosis , Carrier Proteins/genetics , Enzyme Activation , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Genetic Vectors/genetics , Hypoxia/genetics , Intracellular Signaling Peptides and Proteins , MAP Kinase Kinase Kinases/metabolism , Mice , Myocardial Infarction/genetics , Myocytes, Cardiac/enzymology , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats
20.
Trends Cardiovasc Med ; 16(3): 69-75, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16546686

ABSTRACT

The effects of estrogen on vascular function have been studied extensively; however, much less is known regarding the effect of estrogen on myocardial function and cardiomyocyte biology. We recently examined the effects of estrogen on infarct size and left ventricular remodeling after induction of myocardial infarction by permanent coronary ligation in ovariectomized female mice. These studies demonstrated that physiologic replacement with 17beta-estradiol (E(2)) reduced infarct size and reduced cardiomyocyte apoptosis at 24 and 72 hours post-myocardial infarction. In vivo, physiologic E(2) replacement rapidly activated the Akt signaling pathway within the myocardium. In vitro, physiologic concentrations of E(2) rapidly activated Akt via estrogen receptor alpha (ERalpha) and phosphoinositide-3 (PI3)-kinase-dependent mechanisms. Moreover, estrogen mitigated anthracycline-induced apoptosis in vitro via ER- and PI3-kinase-Akt-dependent mechanisms. Our data therefore support that estrogen favorably influences cardiomyocyte survival both in a coronary occlusion model in vivo and in cultured cardiomyocytes in vitro. Further work is necessary to more completely understand the complex effects of sex hormones on cardiomyocyte biology.


Subject(s)
Estrogen Replacement Therapy , Myocytes, Cardiac/drug effects , Animals , Apoptosis , Disease Models, Animal , Estradiol/pharmacology , Female , Humans , In Vitro Techniques , Mice , Myocytes, Cardiac/pathology , Rats , Women's Health
SELECTION OF CITATIONS
SEARCH DETAIL
...