Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Drugs Today (Barc) ; 46(6): 371-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20571606

ABSTRACT

Nicotinic acid (niacin) is one of the oldest drugs used to treat dyslipidemia. In addition to modestly lowering low-density lipoprotein cholesterol (LDL-C) and lipoprotein(a), niacin is currently the most effective available agent for raising high-density lipoprotein cholesterol (HDL-C). Despite its well-documented beneficial effects on lipids, the clinical use of niacin has been limited by its side effect profile, notably flushing. This sensation of cutaneous vasodilatation and burning has limited patient compliance and is a frequent cause of discontinuation of the drug. While pretreatment with nonsteroidal anti-inflammatory agents, such as aspirin, may reduce the incidence of flushing, present-day niacin still results in flushing in many patients. Recent studies have elucidated what we currently understand as the molecular mechanism that mediates niacin-induced flushing, specifically that niacin acting through its receptor stimulates the production of several prostaglandins, including prostaglandin (PG) I(2), PGE(2) and PGD(2). Laropiprant is a potent, highly selective prostaoid DP(1) receptor antagonist that decreases the incidence and intensity of niacin-induced flushing without affecting its beneficial lipid effects. Thus, laropiprant, when used in conjunction with niacin, can improve the tolerability of niacin and aid in medication compliance. This paper reviews the data suggesting the importance of raising HDL with niacin, describes the pharmacology of the drug, and examines the potential beneficial effects of combining niacin with laropiprant.


Subject(s)
Indoles/therapeutic use , Niacin/therapeutic use , Cholesterol, HDL/drug effects , Cholesterol, HDL/metabolism , Drug Tolerance , Exercise , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Indoles/pharmacokinetics , Lipoproteins, HDL/drug effects , Lipoproteins, HDL/metabolism , Receptors, Immunologic/antagonists & inhibitors , Receptors, Prostaglandin/antagonists & inhibitors , Safety , Weight Loss
2.
J Hum Hypertens ; 23(9): 590-6, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19225528

ABSTRACT

Low circulating levels of high-density lipoprotein cholesterol (HDL-C) are associated with increased risk for cardiovascular events. HDL-C has a variety of poorly understood atheroprotective effects, including altering lipid metabolism and reducing inflammation. Increased arterial stiffness is an important predictor of subsequent cardiovascular risk. Therefore, in this study, we sought to determine whether HDL-C levels are associated with carotid arterial stiffness. In addition, we examined potential correlates of this association, such as inflammatory factors, cardiorespiratory fitness and body fat percentage. Carotid artery beta-stiffness was measured by ultrasound in 47 (23 years old) healthy pre-hypertensive men. Low HDL-C was defined as <1.0 mmol l(-1). Body fat was measured by air displacement plethysmography. Cardiorespiratory fitness was measured using a maximal exercise test, with metabolic gas analysis and inflammatory markers consisting of C-reactive protein (CRP), white blood cell (WBC) count and absolute neutrophil count. Men with a low HDL-C had significantly higher carotid artery stiffness, CRP, WBC count, neutrophil count, body fat, fasting glucose and lower cardiorespiratory fitness (P<0.05). Co-varying for cardiorespiratory fitness, % body fat and glucose had no effect on group differences in carotid artery stiffness. Co-varying for inflammatory markers resulted in groups having similar carotid artery stiffness. Pre-hypertensive men with low HDL-C have a higher carotid artery stiffness when compared with those with higher HDL-C. The detrimental effects of low HDL-C on large artery stiffness in pre-hypertensive men may be mediated by inflammation and not by cardiorespiratory fitness or body fat levels.


Subject(s)
Carotid Arteries/physiopathology , Cholesterol, HDL/blood , Hypertension/physiopathology , Body Composition , Elasticity , Humans , Hypertension/blood , Inflammation/physiopathology , Male , Physical Fitness/physiology , Young Adult
3.
Transplant Proc ; 38(9): 3016-20, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17112888

ABSTRACT

BACKGROUND: Early atherosclerosis may be associated with compensatory vessel enlargement, termed positive remodeling. Enlarged brachial artery diameter has been reported in patients with risk factors for atherosclerosis and in individuals with coronary atherosclerosis, indicating that brachial artery enlargement is a marker for the presence of atherosclerotic changes. Cardiac transplant recipients often have abnormal lipid levels, but the effect of specific lipid abnormalities on vascular remodeling in this population has not been evaluated. This study examined the relationship between lipid levels and brachial artery diameter in cardiac transplant recipients. METHODS: Thirty-five stable cardiac transplant recipients underwent high-resolution brachial artery ultrasound to evaluate resting brachial artery diameter. Levels of high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), and triglycerides were determined and the presence of other cardiac risk factors was assessed. RESULTS: Brachial artery diameter was larger (4.3 +/- 0.1 mm) in subjects with low levels of HDL-C (< 40 mg/dL, n = 11) compared to subjects with high HDL-C (> or = 40 mg/dL, n = 24), who had a mean brachial artery diameter of 3.7 +/- 0.1 mm (P = .006). Neither high LDL-C (> or = 100 mg/dL) nor high triglycerides (> or = 200 mg/dL) were associated with differences in brachial artery diameter. Multivariate analysis demonstrated that the relationship between low HDL-C and increased brachial artery diameter was independent of body surface area or statin use. CONCLUSIONS: Low levels of HDL-C are an independent predictor of brachial artery enlargement in stable cardiac transplant recipients. These findings suggest that suboptimal HDL-C levels may be associated with the development of vascular remodeling and atherosclerosis in this population.


Subject(s)
Brachial Artery/diagnostic imaging , Cholesterol, HDL/blood , Heart Transplantation/physiology , Adult , Aged , Brachial Artery/anatomy & histology , Brachial Artery/physiopathology , Cholesterol, LDL/blood , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Male , Middle Aged , Triglycerides/blood , Ultrasonography
5.
J Am Coll Cardiol ; 38(7): 1843-9, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11738283

ABSTRACT

OBJECTIVES: We studied whether assessment of endothelium-dependent vasomotion (EDV) with brachial artery ultrasound (BAUS) imaging predicts the presence or absence of coronary artery disease (CAD) as defined by exercise myocardial perfusion imaging (ExMPI). BACKGROUND: Abnormalities in EDV can be detected in arteries before the development of overt atherosclerosis, and its presence may predict poor long-term prognosis. Brachial artery ultrasound during reactive hyperemia is a noninvasive method of assessing peripheral EDV. METHODS: Clinically-indicated ExMPI along with BAUS were performed in 94 subjects (43 women, 51 men). Coronary artery disease was defined by myocardial ischemia or infarction on single photon emission computed tomography images. Flow-mediated dilation (FMD) after upper arm occlusion was defined as the percent change in arterial diameter during reactive hyperemia relative to the baseline. RESULTS: Subjects with CAD by ExMPI (n = 23) had a lower FMD (6.3 +/- 0.7%) than those without CAD by ExMPI (n = 71) (10.5 +/- 0.6%; p = 0.0004). Flow-mediated dilation was highly predictive for CAD with an odds ratio of 1.32 for each percent decrease in FMD (p = 0.001). Based on a receiver-operator analysis, an FMD of 10% was used as a cut-point for further analysis. Twenty-one of 23 subjects who were positive for ExMPI had an FMD < 10% (sensitivity 91%), whereas only two of 40 subjects with an FMD > or =10% were ExMPI-positive (negative predictive value: 95%). There was a correlation between the number of cardiac risk factors and FMD. Individuals with an FMD < 10% exercised for a shorter duration than those with an FMD > or =10% (456 +/- 24 vs. 544 +/- 31 s, respectively; p = 0.02). CONCLUSIONS: Assessment of EDV with BAUS has a high sensitivity and an excellent negative predictive value for CAD and, thus, has the potential for use as a screening tool to exclude CAD in low-risk subjects. Further standardization of BAUS is required, however, before specific cut-points for excluding CAD can be established.


Subject(s)
Coronary Artery Disease/diagnostic imaging , Endothelium, Vascular/diagnostic imaging , Vascular Resistance/physiology , Brachial Artery/diagnostic imaging , Brachial Artery/physiopathology , Coronary Artery Disease/physiopathology , Coronary Circulation/physiology , Endothelium, Vascular/physiopathology , Exercise Test , Female , Humans , Male , Middle Aged , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/physiopathology , Predictive Value of Tests , Prognosis , Risk Factors , Tomography, Emission-Computed, Single-Photon , Ultrasonography
7.
FEBS Lett ; 502(3): 103-8, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11583108

ABSTRACT

Physiological effects of estrogen on myocardium are mediated by two intracellular estrogen receptors, ERalpha and ERbeta, that regulate transcription of target genes through binding to specific DNA target sequences. To define the role of ERbeta in the transcriptional activation of both endothelial (eNOS) and inducible nitric oxide synthase (iNOS) in cardiac myocytes, we used the complete ER-specific antagonist R,R-tetrahydrochrysene (R,R-THC). R,R-THC inhibited activation of iNOS/eNOS promoter-luciferase reporter constructs (iNOS/eNOS-Luc) in a dose-dependent fashion in COS7 cells selectively transfected with ERbeta, but failed to influence ERalpha-mediated increase of iNOS/ eNOS-Luc. In neonatal rat cardiomyocytes transfected with eNOS-Luc or iNOS-Luc, incubation with 17betaestradiol (E2, 10(-8) M) for 24 h stimulated expression of eNOS and iNOS. R,R-THC (10(-5) M) completely inhibited this effect. Furthermore, eNOS and iNOS protein expression in cardiac myocytes induced by E2 was completely blocked by R,R-THC as shown by immunoblot analysis. Taken together, these results show that ERbeta mediates transcriptional activation of eNOS and iNOS by E2.


Subject(s)
Estradiol/pharmacology , Myocardium/enzymology , Nitric Oxide Synthase/metabolism , Receptors, Estrogen/metabolism , Animals , Blotting, Western , COS Cells , Cells, Cultured , Chrysenes/pharmacology , Estrogen Receptor beta , Heart/drug effects , Immunoblotting , Luciferases/metabolism , Male , Myocardium/chemistry , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Promoter Regions, Genetic , Rats , Rats, Wistar , Transfection , Up-Regulation
8.
Circ Res ; 89(6): 534-9, 2001 Sep 14.
Article in English | MEDLINE | ID: mdl-11557741

ABSTRACT

The two known estrogen receptors, ERalpha and ERbeta, mediate the effects of estrogen in all target tissues, including blood vessels. We have shown previously that estrogen inhibits vascular injury response to the same extent in female wild-type (WT), ERalpha knockout (ERalphaKO(CH)), and ERbeta knockout (ERbetaKO(CH)) mice. We generated mice harboring disruptions of both ERalpha and ERbeta genes (ERalpha,betaKO(CH)) by breeding and studied the effect of 17beta-estradiol (E2) on vascular injury responses in ovariectomized female ERalpha,betaKO(CH) mice and WT littermates. E2 inhibited increases in vascular medial area following injury in the WT mice but not in the ERalpha,betaKO(CH) mice, demonstrating for the first time that the two known estrogen receptors are necessary and sufficient to mediate estrogen inhibition of a component of the vascular injury response. Surprisingly, as in WT littermates, E2 still significantly increased uterine weight and inhibited vascular smooth muscle cell (VSMC) proliferation following injury in the ERalpha,betaKO(CH) mice. These data support that the role of estrogen receptors differs for specific components of the vascular injury response in the ERalpha,betaKO(CH) mice. The results leave unresolved whether E2 inhibition of VSMC proliferation in ERalpha,betaKO(CH) mice is caused by a receptor-independent mechanism, an unidentified receptor responsive to estrogen, or residual activity of the ERalpha splice variant reported previously in the parental ERalphaKO(CH) mice. These possibilities may be resolved by studies of mice in which ERalpha has been fully disrupted (ERalphaKO(St)), which are in progress.


Subject(s)
Carotid Artery Injuries/drug therapy , Estrogens/therapeutic use , Receptors, Estrogen/genetics , Animals , Bromodeoxyuridine/metabolism , Carotid Arteries/drug effects , Carotid Arteries/pathology , Carotid Artery Injuries/genetics , Cell Division/drug effects , Estradiol/therapeutic use , Estrogen Receptor alpha , Estrogen Receptor beta , Female , Genotype , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Ovariectomy
9.
J Clin Invest ; 108(4): 611-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11518735

ABSTRACT

Clinical studies of hormone replacement therapy to prevent cardiovascular diseases have heightened interest in the cardiovascular effects of progestins. However, the role of the progesterone receptor (PR) in vascular biology has not been studied in vivo. We studied ovariectomized female PR knockout (PRKO) mice and their wild-type (WT) littermates using the mouse carotid artery injury model. Placebo-treated PRKO mice showed significantly greater vascular medial hypertrophy and vascular smooth muscle cell (VSMC) proliferation in response to vascular injury than did WT mice. Progesterone had no significant effect in the PRKO mice, but worsened the response to injury in WT mice. VSMCs cultured from PRKO mouse aortae were markedly hyperproliferative, and their growth was not affected by progesterone. In contrast to the in vivo findings, progesterone inhibited proliferation of WT-derived VSMCs. Furthermore, reintroduction of PR into PRKO-derived VSMCs using adenoviral methods restored progesterone-mediated inhibition of proliferation to these cells. This effect was reversed by the PR antagonist, RU 486. Thus, the effects of PR and progesterone differ markedly between cultured VSMCs and intact blood vessels. These data demonstrate a direct role for the PR in regulating the response to vascular injury and VSMC proliferation.


Subject(s)
Carotid Artery Injuries , Receptors, Progesterone/physiology , Animals , Carotid Artery, Common/pathology , Cell Division/drug effects , Cells, Cultured/drug effects , DNA Replication/drug effects , Female , Hormone Antagonists/pharmacology , Hyperplasia , Mice , Mice, Knockout , Mifepristone/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Ovariectomy , Progesterone/antagonists & inhibitors , Progesterone/pharmacology , Receptors, Progesterone/deficiency , Receptors, Progesterone/drug effects , Receptors, Progesterone/genetics , Recombinant Fusion Proteins/physiology , Transfection
11.
J Am Coll Cardiol ; 37(1): 195-200, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11153738

ABSTRACT

OBJECTIVES: The goal of this study was to examine the effect of heart failure etiology on peripheral vascular endothelial function in cardiac transplant recipients. BACKGROUND: Peripheral vascular endothelial dysfunction occurs in patients with heart failure of either ischemic or nonischemic etiology. The effect of heart failure etiology on peripheral endothelial function after cardiac transplantation is unknown. METHODS: Using brachial artery ultrasound, endothelium-dependent, flow-mediated dilation (FMD) was assessed in patients with heart failure with either nonischemic cardiomyopathy (n = 10) or ischemic cardiomyopathy (n = 7), cardiac transplant recipients with prior nonischemic cardiomyopathy (n = 10) or prior ischemic cardiomyopathy (n = 10) and normal controls (n = 10). RESULTS: Patients with heart failure with either ischemic cardiomyopathy or nonischemic cardiomyopathy had impaired FMD (3.6 +/- 1.0% and 5.1 +/- 1.2%, respectively, p = NS) compared with normal subjects (13.9 +/- 1.3%, p < 0.01 compared with either heart failure group). In transplant recipients with antecedent nonischemic cardiomyopathy, FMD was markedly higher than that of heart failure patients with nonischemic cardiomyopathy (13.0 +/- 2.4%, p < 0.001) and similar to that of normal subjects (p = NS). However, FMD remained impaired in transplant recipients with prior ischemic cardiomyopathy (5.5 +/- 1.5%, p = 0.001 compared with normal, p = 0.002 vs. transplant recipients with previous nonischemic cardiomyopathy). CONCLUSIONS: Peripheral vascular endothelial function is normal in cardiac transplant recipients with antecedent nonischemic cardiomyopathy, but remains impaired in those with prior ischemic cardiomyopathy. In contrast, endothelial function is uniformly abnormal for patients with heart failure, regardless of etiology. These findings indicate that cardiac transplantation corrects peripheral endothelial function for patients without ischemic heart disease, but not in those with prior atherosclerotic coronary disease.


Subject(s)
Endothelium, Vascular/physiopathology , Heart Failure/etiology , Heart Transplantation/physiology , Postoperative Complications/etiology , Vasodilation/physiology , Adult , Brachial Artery/physiopathology , Female , Heart Failure/physiopathology , Humans , Male , Middle Aged , Myocardial Ischemia/complications , Myocardial Ischemia/physiopathology , Myocardial Ischemia/surgery , Postoperative Complications/physiopathology , Risk Factors
12.
Proc Natl Acad Sci U S A ; 96(26): 15133-6, 1999 Dec 21.
Article in English | MEDLINE | ID: mdl-10611350

ABSTRACT

The protective effects of estrogen in the cardiovascular system result from both systemic effects and direct actions of the hormone on the vasculature. Two estrogen receptors have been identified, ERalpha and ERbeta. We demonstrated previously that estrogen inhibits the response to vascular injury in both wild-type and ERalpha-deficient mice, and that ERbeta is expressed in the blood vessels of each, suggesting a role for ERbeta in the vascular protective effects of estrogen. In the present study, we examined the effect of estrogen administration on mouse carotid arterial injury in ERbeta-deficient mice. Surprisingly, in ovariectomized female wild-type and ERbeta knockout mice, 17beta-estradiol markedly and equally inhibited the increase in vascular medial area and the proliferation of vascular smooth muscle cells after vascular injury. These data demonstrate that ERbeta is not required for estrogen-mediated inhibition of the response to vascular injury, and suggest that either of the two known estrogen receptors is sufficient to protect against vascular injury, or that another unidentified estrogen receptor mediates the vascular protective effects of estrogen.


Subject(s)
Carotid Artery Injuries/drug therapy , Estradiol/therapeutic use , Muscle, Smooth, Vascular/drug effects , Receptors, Estrogen/genetics , Animals , Cell Division/drug effects , Estrogen Receptor beta , Female , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Ovariectomy
14.
J Clin Invest ; 103(3): 401-6, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9927501

ABSTRACT

Estrogen is an important vasoprotective molecule that causes the rapid dilation of blood vessels by activating endothelial nitric oxide synthase (eNOS) through an unknown mechanism. In studies of intact ovine endothelial cells, 17beta-estradiol (E2) caused acute (five-minute) activation of eNOS that was unaffected by actinomycin D but was fully inhibited by concomitant acute treatment with specific estrogen receptor (ER) antagonists. Overexpression of the known transcription factor ERalpha led to marked enhancement of the acute response to E2, and this was blocked by ER antagonists, was specific to E2, and required the ERalpha hormone-binding domain. In addition, the acute response of eNOS to E2 was reconstituted in COS-7 cells cotransfected with wild-type ERalpha and eNOS, but not by transfection with eNOS alone. Furthermore, the inhibition of tyrosine kinases or mitogen-activated protein (MAP) kinase kinase prevented the activation of eNOS by E2, and E2 caused rapid ER-dependent activation of MAP kinase. These findings demonstrate that the short-term effects of estrogen central to cardiovascular physiology are mediated by ERalpha functioning in a novel, nongenomic manner to activate eNOS via MAP kinase-dependent mechanisms.


Subject(s)
Endothelium, Vascular/metabolism , Estrogens/pharmacology , Nitric Oxide Synthase/metabolism , Receptors, Estrogen/metabolism , Animals , Enzyme Activation , Estrogen Receptor alpha , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type III , Receptors, Estrogen/agonists , Sheep , Signal Transduction/drug effects
15.
Circ Res ; 83(2): 224-9, 1998 Jul 27.
Article in English | MEDLINE | ID: mdl-9686763

ABSTRACT

Estrogen exerts direct effects on vascular endothelial and smooth muscle cells that are important for vascular protection. Estrogen receptor-alpha (ERalpha) is expressed in vascular cells from males and females and may mediate some of the effects of estrogen on vascular tissue. However, we recently found that estrogen is able to protect against vascular injury in ovariectomized female ERalpha knockout mice. These mice express the newly described estrogen receptor-beta (ERbeta) in their aortas, suggesting that ERbeta may also mediate some of the direct effects of estrogen on the vasculature. In this study, the level of expression of ERalpha and ERbeta mRNA in male rat aortas was examined before and after vascular injury using en face (Häutchen) preparations and in situ hybridization. Little or no change in ERalpha expression was observed after vascular injury in either vascular endothelial or smooth muscle cells at any time point. In contrast, ERbeta mRNA was found to be expressed markedly after balloon injury. In endothelial cells, ERbeta was increased by 2 days after injury, and high levels of expression were maintained at 8 and 14 days. Furthermore, ERbeta expression was high in luminal smooth muscle cells at 8 and 14 days after injury and had decreased to low levels by 28 days after injury. These data demonstrate the presence of ERbeta in male vascular tissues and the induction of ERbeta mRNA expression after vascular injury, supporting a role for ERbeta in the direct vascular effects of estrogen.


Subject(s)
Aorta/injuries , RNA, Messenger/biosynthesis , Receptors, Estrogen/biosynthesis , Tunica Intima/metabolism , Animals , Aorta/pathology , Catheterization/adverse effects , Estrogen Receptor alpha , Estrogen Receptor beta , Gene Expression Regulation , In Situ Hybridization , Male , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/genetics , Sex Characteristics
16.
J Clin Invest ; 101(12): 2851-61, 1998 Jun 15.
Article in English | MEDLINE | ID: mdl-9637719

ABSTRACT

The classical estrogen receptor ERalpha mediates many of the known cardiovascular effects of estrogen and is expressed in male and female vascular cells. Estrogen-independent activation of ERalpha is known to occur in cells from reproductive tissues, but has not been investigated previously in vascular cells. In this study, transient transfection assays in human saphenous vein smooth muscle cells (HSVSMC) and pulmonary vein endothelial cells (PVEC) demonstrated ERalpha-dependent activation of estrogen response element-based, and vascular endothelial growth factor-based reporter plasmids by both estrogen-deficient FBS (ED-FBS) and EGF. In nonvascular cells, ERalpha-mediated gene expression can be activated via mitogen-activated protein (MAP) kinase- induced phosphorylation of serine 118 of ERalpha. However, in vascular cells, we found that pharmacologic inhibition of MAP kinase did not alter EGF-mediated ERalpha activation. In addition, a mutant ER containing an alanine-for-serine substitution at position 118 was activated to the same degree as the wild-type receptor by ED-FBS and EGF in both HSVSMC and PVEC. Furthermore, constitutively active MAP kinase kinase (MAPKK) activated ERalpha in Cos1 cells as expected, but MAPKK inhibited ER activation in PVEC. We conclude that growth factors also stimulate ERalpha-mediated gene expression in vascular cells, but find that this occurs via a MAP kinase-independent pathway distinct from that reported previously in nonvascular cells.


Subject(s)
Growth Substances/pharmacology , Muscle, Smooth, Vascular/physiology , Receptors, Estrogen/physiology , Signal Transduction/physiology , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cells, Cultured , Endothelium, Vascular/physiology , Female , Humans , Male , Mitogen-Activated Protein Kinase Kinases , Protein Kinases/physiology , Signal Transduction/drug effects
18.
FEBS Lett ; 416(1): 107-12, 1997 Oct 13.
Article in English | MEDLINE | ID: mdl-9369244

ABSTRACT

Gender-based differences found in cardiovascular diseases raise the possibility that estrogen may have direct effects on cardiac tissue. Therefore we investigated whether cardiac myocytes and fibroblasts express functional estrogen receptors. Immunofluorescence demonstrated estrogen receptor protein expression in both female and male rat cardiac myocytes and fibroblasts. Nuclear translocation of the estrogen receptor protein was observed after stimulation of cardiomyocytes with 17beta-estradiol (E2). Cells transfected with an estrogen-responsive reporter plasmid showed that treatment with E2 induced a significant increase in reporter activity. Furthermore, E2 induced a significant increase in expression of the estrogen receptors alpha and beta, progesterone receptor and connexin 43 in cardiac myocytes. Cardiac myocytes and fibroblasts contain functional estrogen receptors and estrogen regulates expression of specific cardiac genes. These data suggest that gender-based differences in cardiac diseases may in part be due to direct effects of estrogen on the heart.


Subject(s)
Myocardium/metabolism , Receptors, Estrogen/metabolism , Animals , Female , Fibroblasts/metabolism , Fluorescent Antibody Technique , Male , Myocardium/cytology , Rats , Rats, Inbred WKY , Receptors, Progesterone/metabolism
19.
Nat Med ; 3(5): 545-8, 1997 May.
Article in English | MEDLINE | ID: mdl-9142124

ABSTRACT

The atheroprotective effects of estrogen in women are well recognized, but the underlying mechanisms responsible are not well understood. Blood vessel cells express the classic estrogen receptor, ER alpha (ref. 2-6), and are directly affected by estrogen, which inhibits the development of atherosclerotic and injury-induced vascular lesions. We have generated mice in which the ER alpha gene is disrupted and have used a mouse model of carotid arterial injury to compare the effects of estrogen on wild-type and estrogen receptor-deficient mice. Increases in vascular medial area and smooth muscle cell proliferation were quantified following vascular injury in ovariectomized mice treated with vehicle or with physiologic levels of 17 beta-estradiol. Surprisingly, in both wild-type and estrogen receptor-deficient mice, 17 beta-estradiol markedly inhibited to the same degree all measures of vascular injury. These data demonstrate that estrogen inhibits vascular by a novel mechanism that is independent of the classic estrogen receptor, ER alpha.


Subject(s)
Endothelium, Vascular/drug effects , Estradiol/pharmacology , Muscle, Smooth, Vascular/drug effects , Receptors, Estrogen/physiology , Animals , Carotid Arteries , Cell Division , Endothelium, Vascular/pathology , Female , Gene Expression , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/pathology , Receptors, Estrogen/genetics
20.
JAMA ; 277(14): 1140-7, 1997 Apr 09.
Article in English | MEDLINE | ID: mdl-9087469

ABSTRACT

OBJECTIVE: To examine the effect of hormone replacement therapy on life expectancy in postmenopausal women with different risk profiles for heart disease, breast cancer, and hip fracture. DESIGN: Decision analysis using a Markov model. Published regression models were used to link risk factors to disease incidence and to estimate the lifetime risks of developing coronary heart disease (CHD), breast cancer, hip fracture, and endometrial cancer. The impact of hormone therapy on disease incidence was estimated from published epidemiologic studies. SETTING: Mathematical model applicable to primary care. INTERVENTIONS: Treatment with hormone replacement therapy or no hormone replacement therapy. MAIN OUTCOME MEASURE: Life expectancy. RESULTS: Hormone replacement therapy should increase life expectancy for nearly all postmenopausal women, with some gains exceeding 3 years, depending mainly on an individual's risk factors for CHD and breast cancer. For women with at least 1 risk factor for CHD, hormone therapy should extend life expectancy, even for women having first-degree relatives with breast cancer. Women without any risk factors for CHD or hip fracture, but who have 2 first-degree relatives with breast cancer, however, should not receive hormone therapy. CONCLUSIONS: The benefit of hormone replacement therapy in reducing the likelihood of developing CHD appears to outweigh the risk of breast cancer for nearly all women in whom this treatment might be considered. Our analysis supports the broader use of hormone replacement therapy.


Subject(s)
Decision Support Techniques , Estrogen Replacement Therapy , Aged , Black People , Breast Neoplasms/epidemiology , Coronary Disease/epidemiology , Endometrial Neoplasms/epidemiology , Estrogen Replacement Therapy/adverse effects , Estrogen Replacement Therapy/statistics & numerical data , Female , Hip Fractures/epidemiology , Humans , Life Expectancy , Middle Aged , Models, Theoretical , Postmenopause , Risk , Risk Factors , Sensitivity and Specificity , White People
SELECTION OF CITATIONS
SEARCH DETAIL
...