1
|
Favre J, Vessieres E, Guihot AL, Proux C, Grimaud L, Rivron J, Garcia MC, Réthoré L, Zahreddine R, Davezac M, Fébrissy C, Adlanmerini M, Loufrani L, Procaccio V, Foidart JM, Flouriot G, Lenfant F, Fontaine C, Arnal JF, Henrion D. Membrane estrogen receptor alpha (ERα) participates in flow-mediated dilation in a ligand-independent manner. eLife 2021; 10:68695. [PMID: 34842136 PMCID: PMC8676342 DOI: 10.7554/elife.68695] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 11/26/2021] [Indexed: 12/13/2022] Open
Abstract
Estrogen receptor alpha (ERα) activation by estrogens prevents atheroma through its nuclear action, whereas plasma membrane-located ERα accelerates endothelial healing. The genetic deficiency of ERα was associated with a reduction in flow-mediated dilation (FMD) in one man. Here, we evaluated ex vivo the role of ERα on FMD of resistance arteries. FMD, but not agonist (acetylcholine, insulin)-mediated dilation, was reduced in male and female mice lacking ERα (Esr1-/- mice) compared to wild-type mice and was not dependent on the presence of estrogens. In C451A-ERα mice lacking membrane ERα, not in mice lacking AF2-dependent nuclear ERα actions, FMD was reduced, and restored by antioxidant treatments. Compared to wild-type mice, isolated perfused kidneys of C451A-ERα mice revealed a decreased flow-mediated nitrate production and an increased H2O2 production. Thus, endothelial membrane ERα promotes NO bioavailability through inhibition of oxidative stress and thereby participates in FMD in a ligand-independent manner.
Collapse
Affiliation(s)
- Julie Favre
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France
| | - Emilie Vessieres
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Anne-Laure Guihot
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Coralyne Proux
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Linda Grimaud
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France
| | - Jordan Rivron
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Manuela Cl Garcia
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Léa Réthoré
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France
| | - Rana Zahreddine
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Morgane Davezac
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Chanaelle Fébrissy
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Marine Adlanmerini
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Laurent Loufrani
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,University Hospital (CHU) of Angers, Angers, France
| | - Vincent Procaccio
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,University Hospital (CHU) of Angers, Angers, France
| | - Jean-Michel Foidart
- Groupe Interdisciplinaire de Génoprotéomique Appliquée, Université de Liège, Liège, Belgium
| | - Gilles Flouriot
- INSERM U1085, IRSET (Institut de Recherche en Santé, Environnement et Travail), University of Rennes, Rennes, France
| | - Françoise Lenfant
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Coralie Fontaine
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Jean-François Arnal
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Daniel Henrion
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France.,University Hospital (CHU) of Angers, Angers, France
| |
Collapse
|
2
|
Adekunle AO, Adzika GK, Mprah R, Ndzie Noah ML, Adu-Amankwaah J, Rizvi R, Akhter N, Sun H. Predominance of Heart Failure With Preserved Ejection Fraction in Postmenopausal Women: Intra- and Extra-Cardiomyocyte Maladaptive Alterations Scaffolded by Estrogen Deficiency. Front Cell Dev Biol 2021; 9:685996. [PMID: 34660569 PMCID: PMC8511782 DOI: 10.3389/fcell.2021.685996] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 09/09/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) remains a public health concern as it is associated with high morbidity and death rates. In particular, heart failure with preserved ejection fraction (HFpEF) represents the dominant (>50%) form of HF and mostly occurring among postmenopausal women. Hence, the initiation and progression of the left ventricular diastolic dysfunctions (LVDD) (a typically clinical manifestation of HFpEF) in postmenopausal women have been attributed to estrogen deficiency and the loss of its residue cardioprotective effects. In this review, from a pathophysiological and immunological standpoint, we discuss the probable multiple pathomechanisms resulting in HFpEF, which are facilitated by estrogen deficiency. The initial discussions recap estrogen and estrogen receptors (ERs) and β-adrenergic receptors (βARs) signaling under physiological/pathological states to facilitate cardiac function/dysfunction, respectively. By reconciling these prior discussions, attempts were made to explain how the loss of estrogen facilitates the disruptions both ERs and βARs-mediated signaling responsible for; the modulation of intra-cardiomyocyte calcium homeostasis, maintenance of cardiomyocyte cytoskeletal and extracellular matrix, the adaptive regulation of coronary microvascular endothelial functions and myocardial inflammatory responses. By scaffolding the disruption of these crucial intra- and extra-cardiomyocyte physiological functions, estrogen deficiency has been demonstrated to cause LVDD and increase the incidence of HFpEF in postmenopausal women. Finally, updates on the advancements in treatment interventions for the prevention of HFpEF were highlighted.
Collapse
Affiliation(s)
| | | | - Richard Mprah
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | | | | | | | - Nazma Akhter
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Hong Sun
- Department of Physiology, Xuzhou Medical University, Xuzhou, China.,Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
3
|
Dechichi JGC, Mariano IM, Giolo JS, Batista JP, Amaral AL, Ribeiro PAB, de Oliveira EP, Puga GM. Isoflavone Supplementation Does Not Potentiate the Effect of Combined Exercise Training on Resting and Ambulatory Blood Pressure in Non-Obese Postmenopausal Women: A Randomized Double-Blind Controlled Trial-A Pilot Study. Nutrients 2020; 12:nu12113495. [PMID: 33203003 PMCID: PMC7697944 DOI: 10.3390/nu12113495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 11/16/2022] Open
Abstract
Physical exercise and isoflavone supplementation are potential strategies to prevent and treat cardiovascular diseases in postmenopausal women. The aim of this study was to investigate whether there are additive effects of isoflavone supplementation when associated with combined aerobic and resistance exercise on resting and ambulatory blood pressure monitoring (ABPM) and in blood pressure variability (BPV). Thirty-one non-obese postmenopausal women were randomly allocated into two groups: placebo and exercise (Placebo n = 19); and isoflavone supplementation (100 mg/day) and exercise (isoflavone n = 19). ABPM and BPV were evaluated before and after 10 weeks of moderate combined (aerobic and resistance) exercise training. Generalized Estimating Equation (GEE) with Bonferroni correction and intention-to-treat analysis was used to compare the effects of interventions on resting BP, ABPM and BPV. Combined exercise training decreased resting systolic (SBP) and diastolic blood pressure (DBP) and reduced 24 h and awake ambulatory SBP, DBP and mean blood pressure over time, with no additional effects of isoflavone supplementation. No changes were observed in sleep period, or in BPV indexes (Standard Deviation of 24 h (SD), daytime and nighttime interval (SDdn) and average real variability (ARV) in both groups. We conclude that isoflavone supplementation does not potentiate the effects of combined training on resting and ambulatorial systolic and diastolic blood pressure in non-obese postmenopausal women.
Collapse
Affiliation(s)
- Juliene G. C. Dechichi
- Laboratory of Cardiorespiratory and Metabolic Physiology, Physical Education and Physical Therapy Department, Federal University of Uberlândia, 38400-678 Uberlândia, MG, Brazil; (J.G.C.D.); (I.M.M.); (J.S.G.); (J.P.B.); (A.L.A.)
| | - Igor M. Mariano
- Laboratory of Cardiorespiratory and Metabolic Physiology, Physical Education and Physical Therapy Department, Federal University of Uberlândia, 38400-678 Uberlândia, MG, Brazil; (J.G.C.D.); (I.M.M.); (J.S.G.); (J.P.B.); (A.L.A.)
| | - Jéssica S. Giolo
- Laboratory of Cardiorespiratory and Metabolic Physiology, Physical Education and Physical Therapy Department, Federal University of Uberlândia, 38400-678 Uberlândia, MG, Brazil; (J.G.C.D.); (I.M.M.); (J.S.G.); (J.P.B.); (A.L.A.)
| | - Jaqueline P. Batista
- Laboratory of Cardiorespiratory and Metabolic Physiology, Physical Education and Physical Therapy Department, Federal University of Uberlândia, 38400-678 Uberlândia, MG, Brazil; (J.G.C.D.); (I.M.M.); (J.S.G.); (J.P.B.); (A.L.A.)
| | - Ana Luiza Amaral
- Laboratory of Cardiorespiratory and Metabolic Physiology, Physical Education and Physical Therapy Department, Federal University of Uberlândia, 38400-678 Uberlândia, MG, Brazil; (J.G.C.D.); (I.M.M.); (J.S.G.); (J.P.B.); (A.L.A.)
| | - Paula A. B. Ribeiro
- Research Center of University of Montreal Hospital Centre, Montreal, QC H2X 0A9, Canada;
| | - Erick P. de Oliveira
- Laboratory of Nutrition, Exercise and Health (LaNES), School of Medicine, Federal University of Uberlandia (UFU), 38402-022 Uberlândia, MG, Brazil;
| | - Guilherme M. Puga
- Laboratory of Cardiorespiratory and Metabolic Physiology, Physical Education and Physical Therapy Department, Federal University of Uberlândia, 38400-678 Uberlândia, MG, Brazil; (J.G.C.D.); (I.M.M.); (J.S.G.); (J.P.B.); (A.L.A.)
- Correspondence: ; Tel.: +55-343218-2965
| |
Collapse
|
4
|
The Effect of Hypertension Duration and the Age of Onset on CV Risk Factors Expression in Perimenopausal Women. Int J Hypertens 2019; 2019:9848125. [PMID: 31885899 PMCID: PMC6925926 DOI: 10.1155/2019/9848125] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/29/2019] [Accepted: 11/04/2019] [Indexed: 12/31/2022] Open
Abstract
Background The impact of hypertension duration and the time of onset on the expression of metabolic syndrome (MS) and other CV risk factors, in perimenopausal women, have not been studied so far. Methods. A total of 202 women, old 47–59 years, and diagnosed with hypertension, were recruited from primary care practices in eastern Croatia. The categories of hypertension duration were defined as <5, 5–10, and >10 years. Data were analyzed by standard statistical procedures. Results The proportion of women with MS increases in parallel with hypertension duration (p = 0.025). Among the examined CV risk factors, significant increase in parallel with hypertension duration was found for body mass index (p = 0.007) and triglycerides (p = 0.07). The highest proportion of women with diabetes duration of less than 5 years, indicating recent diabetes onset, was found in the category of hypertension duration of less than 5 years, corresponding with the onset of hypertension in the time around menopause (p = 0.003). The strongest linear correlations with BMI and waist circumference were found for total serum cholesterol (r = 0.355 and 0.499, respectively). Conclusion Hypertension onset at the time around menopause appears together with abdominal obesity and may be a driving force for CV risk factor accumulation in postmenopausal women.
Collapse
|
5
|
Tingskov SJ, Mutsaers HAM, Nørregaard R. Estrogen regulates aquaporin-2 expression in the kidney. VITAMINS AND HORMONES 2019; 112:243-264. [PMID: 32061343 DOI: 10.1016/bs.vh.2019.08.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Estrogens are primarily identified as sex hormones that, for a long time, have been known as important regulators of female reproductive physiology. However, our understanding of the role of estrogens has changed over the past years. It is now well accepted that estrogens are also involved in other physiological and pathological processes in both genders. This is due to the fact that estrogen can act both local as well as on a systemic level. Next to its role in reproductive physiology, there is accumulating evidence that estrogen influences multiple systems involved in water homeostasis. This chapter will delineate the regulatory effects of estrogen on the water channel aquaporin-2 (AQP2) found in the renal collecting duct. We will first provide an introduction to estrogen, the estrogen receptors and their role in renal physiology as well as describe the effect of selective estrogen receptor modulators (SERMs) on the kidney. Subsequently, we will focus on how estrogen and SERMs influence water balance and regulate AQP2 expression in principal cells of the collecting duct. Finally, we will describe how estrogen regulates AQP2 functionality in other organ systems.
Collapse
Affiliation(s)
| | | | - Rikke Nørregaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
6
|
Deng W, Duan M, Qian B, Zhu Y, Lin J, Zheng L, Zhang C, Qi X, Luo M. NADPH oxidase 1/4 inhibition attenuates the portal hypertensive syndrome via modulation of mesenteric angiogenesis and arterial hyporeactivity in rats. Clin Res Hepatol Gastroenterol 2019; 43:255-265. [PMID: 30413372 DOI: 10.1016/j.clinre.2018.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 09/25/2018] [Accepted: 10/11/2018] [Indexed: 02/08/2023]
Abstract
AIM NADPH oxidase (NOX)-derived reactive oxygen species (ROS) plays key roles in the development of portal hypertension (PHT) and represents a potential therapeutic method. The objective of this study was to investigate whether pharmacological inhibition of NADPH oxidase activity could ameliorate PHT in rats. METHOD PHT model was established by partial portal vein ligation (PPVL). Rats were treated with 30 mg/kg GKT137831 (the most specific Nox1/4 inhibitor) or vehicle daily by gavage for 14 days beginning at the day of PPVL or sham operation (SO). Hemodynamics, severity of portal-systemic shunting, vascular contractility, vascular endothelial growth factor (VEGF), VEGFR-2, CD31, AKT, phospho-AKT (p-AKT, at Ser473), endothelial nitric oxide synthase (eNOS), and phospho-eNOS (p-eNOS, at Ser1177) expressions were evaluated. Nitric oxide (NO) production and oxidative stress in mesenteric arteries, and hydrogen peroxide (H2O2) in both mesenteric tissues and arteries were measured. RESULT Inhibition of NOX1/4 with GKT137831 significantly decreased cardiac index, increased portal flow resistance, reduced portal pressure (PP), portal blood flow, mesenteric angiogenesis and portal-systemic shunting (PSS) in PPVL rats. GKT137831 reduced the production of H2O2, down regulated mesenteric angiogenesis markers (CD31, vascular endothelial growth factor (VEGF) and VEGFR-2 expression. Compared with controls), the mesenteric artery contraction to norepinephrine (NE) was impaired in PPVL rats, which was reversed by exposure to GKT137831. In addition, GKT137831 markedly decrease NADPH oxidase activity and ROS production in mesenteric arteries, and reduced NO production by decreasing the level of phosphor-AKT and eNOS. CONCLUSION Inhibition of NOX1/4 decreased PP, ameliorated hyperdynamic circulation, mesenteric angiogenesis and arterial hyporesonse in portal hypertensive rats. Pharmacological inhibition of NOX1/4 activity may be a potential treatment for PHT-related complications.
Collapse
Affiliation(s)
- Wensheng Deng
- Department of Liver surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China
| | - Ming Duan
- Department of General Surgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, PR China
| | - Binbin Qian
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, PR China
| | - Yiming Zhu
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, PR China
| | - Jiayun Lin
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, PR China
| | - Lei Zheng
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, PR China
| | - Chihao Zhang
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, PR China
| | - Xiaoliang Qi
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, PR China.
| | - Meng Luo
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, PR China.
| |
Collapse
|
7
|
Morrison HW, Filosa JA. Stroke and the neurovascular unit: glial cells, sex differences, and hypertension. Am J Physiol Cell Physiol 2019; 316:C325-C339. [PMID: 30601672 DOI: 10.1152/ajpcell.00333.2018] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A functional neurovascular unit (NVU) is central to meeting the brain's dynamic metabolic needs. Poststroke damage to the NVU within the ipsilateral hemisphere ranges from cell dysfunction to complete cell loss. Thus, understanding poststroke cell-cell communication within the NVU is of critical importance. Loss of coordinated NVU function exacerbates ischemic injury. However, particular cells of the NVU (e.g., astrocytes) and those with ancillary roles (e.g., microglia) also contribute to repair mechanisms. Epidemiological studies support the notion that infarct size and recovery outcomes are heterogeneous and greatly influenced by modifiable and nonmodifiable factors such as sex and the co-morbid condition common to stroke: hypertension. The mechanisms whereby sex and hypertension modulate NVU function are explored, to some extent, in preclinical laboratory studies. We present a review of the NVU in the context of ischemic stroke with a focus on glial contributions to NVU function and dysfunction. We explore the impact of sex and hypertension as modifiable and nonmodifiable risk factors and the underlying cellular mechanisms that may underlie heterogeneous stroke outcomes. Most of the preclinical investigative studies of poststroke NVU dysfunction are carried out primarily in male stroke models lacking underlying co-morbid conditions, which is very different from the human condition. As such, the evolution of translational medicine to target the NVU for improved stroke outcomes remains elusive; however, it is attainable with further research.
Collapse
|
8
|
de Oliveira TS, de Oliveira LM, de Oliveira LP, Costa RMD, Tostes RDC, Georg RDC, Costa EA, Lobato NDS, Filgueira FP, Ghedini PC. Activation of PI3K/Akt pathway mediated by estrogen receptors accounts for estrone-induced vascular activation of cGMP signaling. Vascul Pharmacol 2018; 110:42-48. [PMID: 30075228 DOI: 10.1016/j.vph.2018.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 05/08/2018] [Accepted: 07/29/2018] [Indexed: 01/13/2023]
Abstract
Estrone (E1) produces remarkable vascular effects, including relaxation, modulation of proliferation, apoptosis and cell adhesion. This study investigated the role of estrogen receptors and endothelial signaling pathways in the vascular relaxation promoted by E1. Aortic rings from male Wistar rats (250-300 g) were contracted with phenylephrine and stimulated with graded concentrations of E1. The concentration-dependent relaxation induced by E1 was abolished after removal of the endothelium or incubation with the estrogen receptor antagonist ICI 182,780. G protein-coupled estrogen receptor antagonism did not alter the E1 effect. Pretreatment of endothelium-intact arteries with inhibitors of nitric oxide synthase, guanylyl cyclase, calmodulin (CaM) and PI3K reduced the E1-induced vasorelaxation. Incubation with inhibitors of the MEK/ERK1/2 or p38MAPK pathways did not alter the E1 vasorelaxation. Similarly, inhibition of cyclooxygenase or blockade of potassium channels did not change the E1 effect. Western blot analysis evidenced that E1 induces phosphorylation of eNOS, PI3K and Akt in rat aorta. Our data demonstrate that E1 induces aortic vascular relaxation through classic estrogen receptors activation on the endothelium. We also identify CaM and PI3K/Akt pathways as critical mediators of the NO-cGMP signaling activation by E1. These findings contribute to the notion that this estrogen regulates arterial function and represents another link, besides 17β-estradiol (E2), between postmenopause and vascular dysfunction.
Collapse
Affiliation(s)
| | - Lais Moraes de Oliveira
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Goias, Goiania, Brazil
| | | | - Rafael Menezes da Costa
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Faculty of Medicine, Federal University of Jatai, Jatai, Brazil
| | - Rita de Cássia Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Raphaela de Castro Georg
- Department of Biochemistry and Molecular Biology, Institute of Biological Sciences, Federal University of Goias, Goiania, Brazil
| | - Elson Alves Costa
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Goias, Goiania, Brazil
| | | | | | - Paulo César Ghedini
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Goias, Goiania, Brazil
| |
Collapse
|
9
|
Naiberg MR, Hatch JK, Selkirk B, Fiksenbaum L, Yang V, Black S, Kertes PJ, Goldstein BI. Retinal photography: A window into the cardiovascular-brain link in adolescent bipolar disorder. J Affect Disord 2017; 218:227-237. [PMID: 28477501 DOI: 10.1016/j.jad.2017.04.066] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/21/2017] [Accepted: 04/28/2017] [Indexed: 01/03/2023]
Abstract
OBJECTIVE The burden of cardiovascular disease in bipolar disorder (BD) exceeds what can be explained by traditional cardiovascular risk factors (CVRFs), lifestyle, and/or medications. Moreover, neurocognitive deficits are a core feature of BD, and are also related to CVRFs. We examined retinal vascular photography, a proxy for cerebral microvasculature, in relation to CVRFs, peripheral microvascular function, and neurocognition among BD adolescents. METHODS Subjects were 30 adolescents with BD and 32 healthy controls (HC). Retinal photography was conducted using a Topcon TRC 50 DX, Type IA camera, following pupil dilation. Retinal arteriolar and venular caliber was measured, from which the arterio-venular ratio (AVR) was computed. All measures were conducted masked to participant diagnosis. Peripheral arterial tonometry measured endothelial function. Neurocognition was assessed using the Cambridge Neuropsychological Tests Automated Battery. RESULTS AVR was not significantly different between groups (Cohen's d=0.18, p=0.103). Higher diastolic blood pressure (BP) was associated with lower (worse) AVR in BD (r=-0.441, p=0.015) but not HC (r=-0.192, p=0.293). Similarly, in the BD group only, higher (better) endothelial function was associated with higher AVR (r=0.375, p=0.041). Hierarchical regression models confirmed that, independent of covariates, retinal vascular caliber was significantly associated with diastolic BP and endothelial function in BD. Within the BD group, mood scores were significantly negatively correlated with AVR (β=-0.451, p=0.044). LIMITATIONS This study's limitations include a small sample size, a cross-sectional study design, and a heterogeneous sample. CONCLUSION Retinal photography may offer unique insights regarding the cardiovascular and neurocognitive burden of BD. Larger longitudinal studies are warranted.
Collapse
Affiliation(s)
- Melanie R Naiberg
- Sunnybrook Health Sciences Centre, Centre for Youth Bipolar Disorder, Toronto, Canada; University of Toronto, Department of Pharmacology and Toxicology, Toronto, Canada; Univeristy of Toronto, Department of Neurology, Toronto, Canada
| | - Jessica K Hatch
- Sunnybrook Health Sciences Centre, Centre for Youth Bipolar Disorder, Toronto, Canada; University of Toronto, Department of Pharmacology and Toxicology, Toronto, Canada; Univeristy of Toronto, Department of Neurology, Toronto, Canada
| | - Beth Selkirk
- The John and Liz Tory Eye Centre, Sunnybrook Health Sciences Centre, Department of Ophthalmology and Vision Sciences, Toronto, Canada; Univeristy of Toronto, Department of Neurology, Toronto, Canada
| | - Lisa Fiksenbaum
- Sunnybrook Health Sciences Centre, Centre for Youth Bipolar Disorder, Toronto, Canada; Univeristy of Toronto, Department of Neurology, Toronto, Canada
| | - Victor Yang
- Sunnybrook Research Institute, Hurvitz Brain Sciences Research Program, Toronto, Canada; University of Toronto, Department of Neurosurgery, Toronto, Canada; Univeristy of Toronto, Department of Neurology, Toronto, Canada
| | - Sandra Black
- Sunnybrook Research Institute, Hurvitz Brain Sciences Research Program, Toronto, Canada; University of Toronto, Department of Neurosurgery, Toronto, Canada; Sunnybrook Health Sciences Centre, Department of Medicine (Neurology), Toronto, Canada; Univeristy of Toronto, Department of Neurology, Toronto, Canada
| | - Peter J Kertes
- The John and Liz Tory Eye Centre, Sunnybrook Health Sciences Centre, Department of Ophthalmology and Vision Sciences, Toronto, Canada; University of Toronto, Ophthalmology and Vision Sciences, Toronto, Canada; Univeristy of Toronto, Department of Neurology, Toronto, Canada
| | - Benjamin I Goldstein
- Sunnybrook Health Sciences Centre, Centre for Youth Bipolar Disorder, Toronto, Canada; University of Toronto, Department of Pharmacology and Toxicology, Toronto, Canada; Univeristy of Toronto, Department of Neurology, Toronto, Canada.
| |
Collapse
|
10
|
Arnal JF, Lenfant F, Metivier R, Flouriot G, Henrion D, Adlanmerini M, Fontaine C, Gourdy P, Chambon P, Katzenellenbogen B, Katzenellenbogen J. Membrane and Nuclear Estrogen Receptor Alpha Actions: From Tissue Specificity to Medical Implications. Physiol Rev 2017; 97:1045-1087. [DOI: 10.1152/physrev.00024.2016] [Citation(s) in RCA: 213] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 12/19/2016] [Accepted: 01/18/2017] [Indexed: 12/22/2022] Open
Abstract
Estrogen receptor alpha (ERα) has been recognized now for several decades as playing a key role in reproduction and exerting functions in numerous nonreproductive tissues. In this review, we attempt to summarize the in vitro studies that are the basis of our current understanding of the mechanisms of action of ERα as a nuclear receptor and the key roles played by its two activation functions (AFs) in its transcriptional activities. We then depict the consequences of the selective inactivation of these AFs in mouse models, focusing on the prominent roles played by ERα in the reproductive tract and in the vascular system. Evidence has accumulated over the two last decades that ERα is also associated with the plasma membrane and activates non-nuclear signaling from this site. These rapid/nongenomic/membrane-initiated steroid signals (MISS) have been characterized in a variety of cell lines, and in particular in endothelial cells. The development of selective pharmacological tools that specifically activate MISS and the generation of mice expressing an ERα protein impeded for membrane localization have begun to unravel the physiological role of MISS in vivo. Finally, we discuss novel perspectives for the design of tissue-selective ER modulators based on the integration of the physiological and pathophysiological roles of MISS actions of estrogens.
Collapse
Affiliation(s)
- Jean-Francois Arnal
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Françoise Lenfant
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Raphaël Metivier
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Gilles Flouriot
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Daniel Henrion
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Marine Adlanmerini
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Coralie Fontaine
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Pierre Gourdy
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Pierre Chambon
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Benita Katzenellenbogen
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - John Katzenellenbogen
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| |
Collapse
|
11
|
Abstract
Regular exercise is promoted as a therapeutic strategy for age-associated endothelial dysfunction. Improvements in endothelial function are observed with endurance exercise in older men, but are diminished or absent in older women. This article examines the hypothesis that sex hormones modulate vascular adaptations to exercise training by influencing antioxidant defense systems, mitochondrial function, oxidative stress, and intracellular signaling.
Collapse
Affiliation(s)
- Kerrie L. Moreau
- Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
- Denver Veterans Administration Medical Center, Geriatric Research, Education and Clinical Center, University of Illinois at Chicago, Chicago, IL
| | - Cemal Ozemek
- Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
12
|
Kypreos KE, Zafirovic S, Petropoulou PI, Bjelogrlic P, Resanovic I, Traish A, Isenovic ER. Regulation of endothelial nitric oxide synthase and high-density lipoprotein quality by estradiol in cardiovascular pathology. J Cardiovasc Pharmacol Ther 2014; 19:256-68. [PMID: 24414281 DOI: 10.1177/1074248413513499] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Estrogens have been recognized, in the last 3 decades, as important hormones in direct and indirect modulation of vascular health. In addition to their direct benefit on cardiovascular health, the presence of esterified estrogen in the lipid core of high-density lipoprotein (HDL) particles indirectly contributes to atheroprotection by significantly improving HDL quality and functionality. Estrogens modulate their physiological activity via genomic and nongenomic mechanisms. Genomic mechanisms are thought to be mediated directly by interaction of the hormone receptor complex with the hormone response elements that regulate gene expression. Nongenomic mechanisms are thought to occur via interaction of the estrogen with membrane-bound receptors, which rapidly activate intracellular signaling without binding of the hormone receptor complex to its hormone response elements. Estradiol in particular mediates early and late endothelial nitric oxide synthase (eNOS) activation via interaction with estrogen receptors through both nongenomic and genomic mechanisms. In the vascular system, the primary endogenous source of nitric oxide (NO) generation is eNOS. Nitric oxide primarily influences blood vessel relaxation, the heart rate, and myocyte contractility. The abnormalities in expression and/or functions of eNOS lead to the development of cardiovascular diseases, both in animals and in humans. Although considerable research efforts have been dedicated to understanding the mechanisms of action of estradiol in regulating cardiac eNOS, more research is needed to fully understand the details of such mechanisms. This review focuses on recent findings from animal and human studies on the regulation of eNOS and HDL quality by estradiol in cardiovascular pathology.
Collapse
Affiliation(s)
- Kyriakos E Kypreos
- 1Department of Medicine, University of Patras Medical School, Pharmacology Laboratory, Panepistimioupolis, Rio, Greece
| | | | | | | | | | | | | |
Collapse
|
13
|
Affiliation(s)
- Akos Koller
- Department of Pathophysiology and Gerontology , Medical School, University of Pecs , Szigeti Str. 12, Pécs H-7624 , Hungary ; Department of Physiology , New York Medical College , 15 Dana Rd Valhalla NY 10595 , USA
| |
Collapse
|
14
|
Moreau KL, Stauffer BL, Kohrt WM, Seals DR. Essential role of estrogen for improvements in vascular endothelial function with endurance exercise in postmenopausal women. J Clin Endocrinol Metab 2013; 98:4507-15. [PMID: 24092827 PMCID: PMC3816259 DOI: 10.1210/jc.2013-2183] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVE In contrast to age-matched men, endurance exercise training is not consistently associated with enhanced endothelial function in estrogen-deficient postmenopausal women. We determined whether endurance exercise training improves endothelial function in postmenopausal women treated with estrogen. In a substudy, we determined if oxidative stress is mechanistically linked to endothelial function adaptations to endurance exercise training. PARTICIPANTS AND DESIGN Brachial artery flow-mediated dilation (FMD) was measured in 36 sedentary, estrogen-deficient postmenopausal women (45-65 y) at study entry (baseline), after 12 weeks of either placebo, oral (1 mg/d) estradiol, or transdermal estradiol (0.05 mg/d) (randomized), and after an additional 12 weeks of continued estradiol or placebo treatment with concurrent endurance exercise training. In subgroups of women, FMD also was measured during the infusion of ascorbic acid at baseline and following estradiol/placebo plus endurance exercise training, and in seven habitually endurance-trained estrogen-deficient controls. RESULTS FMD increased in the estrogen-treated groups (both P < .01) after 12 weeks and remained unchanged in placebo. FMD further increased following 12 weeks of endurance exercise training in estrogen-treated (both P < .025), but not placebo-treated women (P = .55). In the substudy, baseline FMD was similar between sedentary and endurance-trained controls. Ascorbic acid increased FMD at baseline in sedentary women and endurance-trained controls, and following endurance exercise training in placebo-treated, but not in estrogen-treated women. CONCLUSIONS Estrogen status appears to play an important modulatory role in improvements in endothelial function with endurance exercise training in postmenopausal women. The restored endurance exercise training adaptation in estrogen-treated postmenopausal women may be related to mitigation of oxidative stress.
Collapse
Affiliation(s)
- Kerrie L Moreau
- PhD, AssociateDivision of Geriatric Medicine, University of Colorado School of Medicine, Building L15, Room 8111, 12631 East 17th Avenue, PO Box 6511, Aurora, CO 80045.
| | | | | | | |
Collapse
|
15
|
Subramanian M, Balasubramanian P, Garver H, Northcott C, Zhao H, Haywood JR, Fink GD, MohanKumar SMJ, MohanKumar PS. Chronic estradiol-17β exposure increases superoxide production in the rostral ventrolateral medulla and causes hypertension: reversal by resveratrol. Am J Physiol Regul Integr Comp Physiol 2011; 300:R1560-8. [PMID: 21411770 DOI: 10.1152/ajpregu.00020.2011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Women are exposed to estrogen in several forms, such as oral contraceptive pills and hormone replacement therapy. Although estrogen was believed to be cardioprotective, lately, its beneficial effects are being questioned. Recent studies indicate that oxidative stress in the rostral ventrolateral medulla (RVLM) may play a role in the development of hypertension. Therefore, we hypothesized that chronic exposure to low levels of estradiol-17β (E(2)) leads to hypertension in adult-cycling female Sprague Dawley (SD) rats potentially through generation of superoxide in the RVLM. To test this hypothesis, young adult (3 or 4 mo old) female SD rats were either sham-implanted or implanted (subcutaneously) with slow-release E(2) pellets (20 ng/day) for 90 days. A group of control and E(2)-treated animals were fed lab chow or chow containing resveratrol (0.84 g/kg of chow), an antioxidant. Rats were implanted with telemeters to continuously monitor blood pressure (BP) and heart rate (HR). At the end of treatment, the RVLM was isolated for measurements of superoxide. E(2) treatment significantly increased mean arterial pressure (mmHg) and HR (beats/min) compared with sham rats (119.6 ± 0.8 vs. 105.1 ± 0.7 mmHg and 371.7 ± 1.5 vs. 354.4 ± 1.3 beats/min, respectively; P < 0.0001). Diastolic and systolic BP were significantly increased in E(2)-treated rats compared with control animals. Superoxide levels in the RVLM increased significantly in the E(2)-treated group (0.833 ± 0.11 nmol/min·mg) compared with control (0.532 ± 0.04 nmol/min·mg; P < 0.05). Treatment with resveratrol reversed the E(2)-induced increases in BP and superoxide levels in the RVLM. In conclusion, these findings support the hypothesis that chronic exposure to low levels of E(2) induces hypertension and increases superoxide levels in the RVLM and that this effect can be reversed by resveratrol treatment.
Collapse
Affiliation(s)
- Madhan Subramanian
- Department of Pathobiology and Diagnostic Investigation, Colleges of Veterinary, Osteopathic and Human Medicine, Michigan State University, East Lansing, 48824, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Labinskyy N, Hicks S, Grijalva J, Edwards J. The Contrary Impact Of Diabetes And Exercise On Endothelial Nitric Oxide Synthase Function. WEBMEDCENTRAL 2010; 1. [PMID: 27683619 DOI: 10.9754/journal.wmc.2010.00137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
17
|
Gopinath B, Wang JJ, Flood VM, Burlutsky G, Wong TY, Mitchell P. The associations between blood levels of homocysteine, folate, vitamin B12, and retinal vascular caliber. Am J Ophthalmol 2009; 148:902-9. [PMID: 19796756 DOI: 10.1016/j.ajo.2009.07.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Revised: 06/30/2009] [Accepted: 07/16/2009] [Indexed: 11/18/2022]
Abstract
PURPOSE To assess the association of total serum levels of homocysteine (tHcy), folate, and vitamin B12 with retinal vascular caliber in older adults. DESIGN Cross-sectional, population-based study. METHODS One thousand seven hundred and seventy-two of 2334 Blue Mountains Eye Study participants examined had fundus photographs taken and retinal arteriolar and venular caliber measured using computer-assisted techniques and summarized. Serum folate and vitamin B12 levels and tHcy were determined from venous blood samples. Linear regression models adjusting for age, gender, mean arterial blood pressure, smoking, and diabetes were used to assess whether serum levels of tHcy, folate, and vitamin B12 were associated with retinal arteriolar and venular caliber. RESULTS Arteriolar and venular caliber was not associated with tHcy in the total population. Further analysis demonstrated a significant serum homocysteine-gender interaction (P = .04). A significant inverse association between tHcy and arteriolar caliber was found in men only (P = .03), with a threshold detected at a level of 17 micromol/l. Above this threshold, increasing tHcy was linearly associated with narrower arteriolar caliber (0.86 microm reduction per 1.0 micromol/l increase in tHcy) in men, but no significant association was found below this threshold. No significant associations were observed between serum folate or vitamin B12 levels and either retinal vessel caliber. CONCLUSIONS Retinal vascular caliber is associated with tHcy in men but not women. This finding may reflect the stronger association between blood pressure and tHcy in men than in women.
Collapse
Affiliation(s)
- Bamini Gopinath
- Centre for Vision Research, Department of Ophthalmology, Westmead Millennium Institute, University of Sydney, Sydney, New South Wales, Australia
| | | | | | | | | | | |
Collapse
|
18
|
Gardner JD, Murray DB, Voloshenyuk TG, Brower GL, Bradley JM, Janicki JS. Estrogen attenuates chronic volume overload induced structural and functional remodeling in male rat hearts. Am J Physiol Heart Circ Physiol 2009; 298:H497-504. [PMID: 19933421 DOI: 10.1152/ajpheart.00336.2009] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have previously reported gender differences in ventricular remodeling and development of heart failure using the aortocaval fistula model of chronic volume overload in rats. In contrast to males, female rats exhibited no adverse ventricular remodeling and less mortality in response to volume overload. This gender-specific cardioprotection was lost following ovariectomy and was partially restored using estrogen replacement. However, it is not known if estrogen treatment would be as effective in males. The purpose of this study was to evaluate the structural and functional effects of estrogen in male rats subjected to chronic volume overload. Four groups of male rats were studied at 3 days and 8 wk postsurgery as follows: fistula and sham-operated controls, with and without estrogen treatment. Biochemical and histological studies were performed at 3 days postsurgery, with chronic structural and functional effects studied at 8 wk. Measurement of systolic and diastolic pressure-volume relationships was obtained using a blood-perfused isolated heart preparation. Both fistula groups developed significant ventricular hypertrophy after 8 wk of volume overload. Untreated rats with fistula exhibited extensive ventricular dilatation, which was coupled with a loss of systolic function. Estrogen attenuated left ventricular dilatation and maintained function in treated rats. Estrogen treatment was also associated with a reduction in oxidative stress and circulating endothelin-1 levels, as well as prevention of matrix metalloproteinase-2 and -9 activation and breakdown of ventricular collagen in the early stage of remodeling. These data demonstrate that estrogen attenuates ventricular remodeling and disease progression in male rats subjected to chronic volume overload.
Collapse
Affiliation(s)
- Jason D Gardner
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.
| | | | | | | | | | | |
Collapse
|
19
|
Al Malty AM, Petrofsky J, Akhavan S. Aging in Women: The Effect of Menopause on Skin Blood Flow and the Response to Electrical Stimulation. PHYSICAL & OCCUPATIONAL THERAPY IN GERIATRICS 2009. [DOI: 10.1080/02703180802335659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
20
|
Whitley GSJ, Cartwright JE. Trophoblast-mediated spiral artery remodelling: a role for apoptosis. J Anat 2009; 215:21-6. [PMID: 19215319 DOI: 10.1111/j.1469-7580.2008.01039.x] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
In the first 20 weeks of pregnancy a number of important changes take place in the maternal uterine vasculature. Vascular endothelial and smooth muscle cells are lost from the spiral arteries and are replaced by fetal trophoblast cells. The resulting increase in blood flow to the intervillous space ensures that the fetus receives the nutrients and respiratory gases required for growth. Failure of the vessels to remodel sufficiently is a common feature of pregnancy pathologies such as early pregnancy loss, intrauterine growth restriction and pre-eclampsia. Although there is evidence to suggest that some vascular changes occur prior to trophoblast invasion, it is clear that in the absence of trophoblast invasion the remodelling of the spiral arteries is reduced. The cellular and molecular mechanisms by which trophoblasts influence vessel structure have been little studied. Trophoblasts synthesize and release a plethora of cytokines and growth factors including members of the tumour necrosis factor family such as tumour necrosis factor alpha, Fas-ligand and tumour necrosis factor-related apoptosis-inducing ligand. Recent studies suggest that these factors may be important in regulating the remodelling process by inducing both endothelial cell and vascular smooth muscle cell apoptosis.
Collapse
Affiliation(s)
- Guy St J Whitley
- Centre for Developmental and Endocrine Signalling, Division of Basic Medical Sciences, St George's University of London, Cranmer Terrace, London, UK.
| | | |
Collapse
|
21
|
LeBlanc AJ, Cumpston JL, Chen BT, Frazer D, Castranova V, Nurkiewicz TR. Nanoparticle inhalation impairs endothelium-dependent vasodilation in subepicardial arterioles. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2009; 72:1576-84. [PMID: 20077232 PMCID: PMC2808198 DOI: 10.1080/15287390903232467] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Exposure to fine particulate matter (PM, mean aerodynamic diameter <or=2.5 microm) has been shown to be a risk factor for cardiovascular disease mortality and may contribute to acute coronary events such as myocardial infarction (MI). There is sufficient reason to believe that smaller particles, such as nanoparticles, might be even more detrimental than larger sized particles due to their increased surface area and higher pulmonary deposition. Our laboratory showed that nanoparticle inhalation impairs endothelium-dependent arteriolar vasodilation in skeletal muscle. However, it is not known whether coronary microvascular endothelial function is affected in a similar manner. Rats were exposed to filtered air (control) or TiO(2) nanoparticles (primary particle diameter, approximately 21 nm) via inhalation at concentrations that produced measured depositions (10 microg) relevant to ambient air pollution. Subepicardial arterioles ( approximately 150 mum in diameter) were isolated and responses to transmural pressure, flow-induced dilation (FID), acetylcholine (ACh), the Ca(2+) ionophore A23187, and sodium nitroprusside (SNP) were assessed. Myogenic responsiveness was preserved between groups. In addition, there was no difference in the vasodilation to SNP, signifying that smooth muscle sensitivity to nitric oxide (NO) is unaffected by nano-TiO(2) exposure. However, inhalation of nano-TiO(2) produced an increase in spontaneous tone in coronary arterioles and also impaired endothelium-dependent FID. In addition, ACh-induced and A23187-induced vasodilation was also blunted in arterioles after inhalation of nano-TiO(2). Data showed that nanoparticle exposure significantly impairs endothelium-dependent vasodilation in subepicardial arterioles. Such disturbances in coronary microvascular function are consistent with the cardiac events associated with particle pollution exposure.
Collapse
Affiliation(s)
- AJ LeBlanc
- Center for Cardiovascular and Respiratory Sciences, West Virginia University School of Medicine, Morgantown, WV, 26506 USA
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, 26506 USA
| | - JL Cumpston
- Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, 26506 USA
| | - BT Chen
- Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, 26506 USA
| | - D Frazer
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, 26506 USA
- Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, 26506 USA
| | - V Castranova
- Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, 26506 USA
| | - TR Nurkiewicz
- Center for Cardiovascular and Respiratory Sciences, West Virginia University School of Medicine, Morgantown, WV, 26506 USA
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, 26506 USA
- Department of Neurobiology and Anatomy, West Virginia University School of Medicine, Morgantown, WV, 26506 USA
| |
Collapse
|
22
|
Ricchiuti V, Lian CG, Oestreicher EM, Tran L, Stone JR, Yao T, Seely EW, Williams GH, Adler GK. Estradiol increases angiotensin II type 1 receptor in hearts of ovariectomized rats. J Endocrinol 2009; 200:75-84. [PMID: 18931023 PMCID: PMC2927364 DOI: 10.1677/joe-08-0199] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
We tested the hypothesis that 17beta-estradiol (E(2)) has dual effects on the heart, increasing levels of proteins thought to have beneficial cardiovascular effects (e.g. endothelial nitric oxide (NO) synthase (eNOS)) as well as those thought to have detrimental cardiovascular effects (e.g. type 1 angiotensin II (AngII) receptor (AT(1)R)). Ovariectomized Wistar rats consuming a high-sodium diet received one of four treatments (n=7 per group): group 1, placebo pellets; group 2, E(2) (0 x 5 mg/pellet, 21-day release); group 3, NOS inhibitor, N(omega)-nitro-L-arginine-methyl-ester (L-NAME; 40 mg/kg per day for 14 days) plus Ang II (0 x 225 mg/kg per day on days 11-14); group 4, E(2) plus L-NAME/Ang II. E(2) increased cardiac levels of estrogen receptors ESR1 and ESR2, an ESR-associated membrane protein caveolin-3, eNOS, and phosphorylated (p)eNOS, thus, exerting potentially beneficial cardiovascular effects on NO. However, E(2) also increased cardiac levels of proteins associated with cardiovascular injury and inflammation including, AT(1)R, protein kinase C delta (PRKCD), phosphorylated PRKC, and phosphorylated extracellular signal regulated kinase (pMAPK)3/1, plasminogen activator inhibitor-1 (PAI-1), osteopontin and ED-1, a monocyte/macrophage-specific protein. E(2) treatment led to similar protein changes in the hearts of L-NAME/Ang II-treated rats except that the increase in peNOS was prevented, and L-NAME/Ang II and E(2) had additive effects in increasing cardiac PRKCD and PAI-1. Thus, the highest levels of cardiac PAI-1 and PRKCD occurred in L-NAME/Ang II-treated rats receiving E(2). In summary, E(2) treatment increased cardiac expression of AT(1)R as well as the expression of pro-inflammatory and prothrombotic factors.
Collapse
Affiliation(s)
- Vincent Ricchiuti
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Lane PH. Estrogen receptors in the kidney: lessons from genetically altered mice. ACTA ACUST UNITED AC 2008; 5 Suppl A:S11-8. [PMID: 18395676 DOI: 10.1016/j.genm.2008.03.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2007] [Indexed: 10/22/2022]
Abstract
BACKGROUND Sex differences in human and animal models of kidney disease suggest that estrogen receptor (ER)-mediated events may modulate these processes. Genetically altered mice lacking one or both ERs provide a powerful tool to study these phenomena. OBJECTIVE This article examines sex differences in the kidney, particularly the role of ERs. METHODS To identify pertinent studies in genetically altered mice, a literature search was conducted on the MEDLINE database from January 1966 to July 2007, using the search terms estrogen receptor, kidney, and mice. Our group examined the effect of the ER-alpha knockout genotype on the kidney in streptozotocin-induced diabetes mellitus and compensatory kidney growth after uninephrectomy. RESULTS Female mice lacking ERa had reduced renal growth, including glomerular enlargement after 2 weeks of streptozotocin-induced diabetes mellitus and compensatory kidney growth 48 hours after uninephrectomy. CONCLUSION ER-mediated events influence kidney growth and disease in female mice.
Collapse
Affiliation(s)
- Pascale H Lane
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| |
Collapse
|
24
|
Nurkiewicz TR, Porter DW, Hubbs AF, Cumpston JL, Chen BT, Frazer DG, Castranova V. Nanoparticle inhalation augments particle-dependent systemic microvascular dysfunction. Part Fibre Toxicol 2008; 5:1. [PMID: 18269765 PMCID: PMC2276228 DOI: 10.1186/1743-8977-5-1] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2007] [Accepted: 02/12/2008] [Indexed: 11/16/2022] Open
Abstract
Background We have shown that pulmonary exposure to fine particulate matter (PM) impairs endothelium dependent dilation in systemic arterioles. Ultrafine PM has been suggested to be inherently more toxic by virtue of its increased surface area. The purpose of this study was to determine if ultrafine PM (or nanoparticle) inhalation produces greater microvascular dysfunction than fine PM. Rats were exposed to fine or ultrafine TiO2 aerosols (primary particle diameters of ~1 μm and ~21 nm, respectively) at concentrations which do not alter bronchoalveolar lavage markers of pulmonary inflammation or lung damage. Results By histopathologic evaluation, no significant inflammatory changes were seen in the lung. However, particle-containing macrophages were frequently seen in intimate contact with the alveolar wall. The spinotrapezius muscle was prepared for in vivo microscopy 24 hours after inhalation exposures. Intraluminal infusion of the Ca2+ ionophore A23187 was used to evaluate endothelium-dependent arteriolar dilation. In control rats, A23187 infusion produced dose-dependent arteriolar dilations. In rats exposed to fine TiO2, A23187 infusion elicited vasodilations that were blunted in proportion to pulmonary particle deposition. In rats exposed to ultrafine TiO2, A23187 infusion produced arteriolar constrictions or significantly impaired vasodilator responses as compared to the responses observed in control rats or those exposed to a similar pulmonary load of fine particles. Conclusion These observations suggest that at equivalent pulmonary loads, as compared to fine TiO2, ultrafine TiO2 inhalation produces greater remote microvascular dysfunction.
Collapse
Affiliation(s)
- Timothy R Nurkiewicz
- Center for Interdisciplinary Research in Cardiovascular Sciences, West Virginia University School of Medicine, Morgantown, WV, USA.
| | | | | | | | | | | | | |
Collapse
|
25
|
Douglas G, Natalia Cruz M, Poston L, Gustafsson JÅ, Kublickiene K. Functional characterization and sex differences in small mesenteric arteries of the estrogen receptor-β knockout mouse. Am J Physiol Regul Integr Comp Physiol 2008; 294:R112-20. [DOI: 10.1152/ajpregu.00421.2007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The role of the estrogen receptor (ER) subtypes in the modulation of vascular function is poorly understood. The aim of this study was to characterize ex vivo the functional properties of small arteries and their response to estrogens in the mesenteric circulation of female and male ER-β knockout mice (β-ERKO) and their wild-type (WT) littermates. Responses to changes in intraluminal flow and pressure were obtained before and after incubation with 17β-estradiol or ER-α agonist propyl-pyrazole-triol (3 h; 10 nM). Cumulative concentration-response curves to acetylcholine, norepinephrine, and passive distensibility were compared with respect to sex and genotype. The collagen and elastin content within the vascular wall and ER expression were also determined. Endothelial morphology was visualized by scanning electron microscopy. 17β-Estradiol and propyl-pyrazole-triol-treated arteries from female β-ERKO and WT mice showed enhanced flow-mediated dilation, but this was not evident in males. Distensibility was decreased in arteries from β-ERKO females. Sex differences in myogenic tone were observed in 17β-estradiol-treated arteries, but were similar between β-ERKO and WT mice. Acetylcholine- and norepinephrine-induced responses were similar between groups and sexes. ER-α was similarly expressed in the endothelium and media of arteries from all groups studied, as well as ER-β in WT animals. Endothelial morphology was similar in arteries from animals of both sexes and genotype; however, arterial elastin content was decreased, and collagen content was increased in β-ERKO male compared with WT male and with β-ERKO female. We suggest that ERs play a sex-specific role in estrogen-mediated flow responses and distensibility, and that deletion of ER-β affects artery structure but only in male animals. Further studies in β-ERKO mice with established hypertension and in α-ERKO mice are warranted.
Collapse
|
26
|
Toblli JE, Cao G, Casabé AR, Bechara AJ. Effects of ACE inhibition and beta-blockade on female genital structures in spontaneously hypertensive rats. J Sex Med 2007; 4:1593-603. [PMID: 17888071 DOI: 10.1111/j.1743-6109.2007.00597.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
INTRODUCTION AND AIM This study evaluated the possible differences between an angiotensin converting enzyme (ACE) inhibitor and a beta-blocker concerning their potential protective role on female external genitalia in spontaneously hypertensive rats (SHR). MAIN OUTCOME MEASURES Morphological changes in the clitoris after antihypertensive treatments. METHODS For 6 months, SHR received no treatment; SHR + ramipril (RAM), SHR + atenolol (AT), and control Wistar Kyoto (WKY) rats received no treatment. Clitorises were processed for immunohistochemistry using anti-alpha-smooth muscle actin (alpha-SMA), anti-collagen I and III, anti-transforming growth factor beta(1) (TGFbeta(1)), and anti-endothelial nitric oxide synthase (eNOS) antibodies. RESULTS SHR + RAM and SHR + AT presented significantly lower blood pressure in both groups vs. untreated SHR. Compared with WKY, alpha-SMA was increased in the arteries and in the cavernous spaces of the clitoris together with a marked increase in wall/lumen ratio in clitoral vessels in untreated SHR. All these alterations were diminished in SHR + AT (P < 0.01). SHR + RAM presented differences with respect to SHR + AT in the reduction of these variables. TGFbeta(1) expression in the vessel wall from the clitoris and collagen I and III deposition in the interstitium from the clitoris in untreated SHR were significantly more (P < 0.01) than in WKY. While SHR + AT showed a mild decrease in these variables, SHR + RAM presented a significant reduction (P < 0.01) in TGFbeta(1) expression interstitial fibrosis and in both types of collagens. Positive immunostaining of eNOS in the sinusoidal endothelium from the clitoris was less (P < 0.01) in untreated SHR (3.4 +/- 1.3%) and SHR + AT (5.1 +/- 1.2%) than in SHR + RAM (17.2 +/- 1.6%) and WKY (15.9 +/- 1.7%). Untreated SHR and SHR + AT presented more surrounding connective tissue at the perineurium in the clitoris (P < 0.01) than SHR + RAM. CONCLUSION ACE inhibition provided a considerable protective role on the female external genitalia structures in SHR by a mechanism that may be, at least in part, independent of the degree of blood pressure lowering.
Collapse
Affiliation(s)
- Jorge E Toblli
- Laboratory of Experimental Medicine, Hospital Alemán, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| | | | | | | |
Collapse
|
27
|
Nichols WW, Pierce GL, Braith RW. Does hormone treatment alter arterial properties in postmenopausal women? Expert Rev Endocrinol Metab 2007; 2:653-665. [PMID: 30736128 DOI: 10.1586/17446651.2.5.653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Arterial elastic properties and wave reflections constitute left ventricular pulsatile afterload and are directly related to cardiovascular morbidity and mortality. During the aging process, elastic arteries stiffen and increase pulse wave velocity, causing the reflected pressure wave to arrive at the heart during systole, and augment systolic and pulse blood pressure, resulting in left ventricular hypertrophy. The incidence of cardiovascular disease is much greater in men aged 30-50 years compared with women of a similar age. Among women, the incidence of cardiovascular disease increases dramatically after menopause, which occurs at an average age of 52 years. This observation has lead to the belief that sex hormones produced premenopausally impede or delay the progression of cardiovascular disease. Therefore, it appears logical that the administration of female sex hormones, hormone treatment (HT), estrogen alone or with progesterone should provide some degree of cardiovascular protection. This idea was supported by experiments in animals and isolated arterial segments demonstrating that HT increases plasma nitric oxide, decreases endothelin-1 levels and causes smooth muscle relaxation. Also, 17β-estradiol administration decreases collagen and increases elastin in the aortic wall of rats. These experimental studies suggest an improvement in both elastic and muscular artery properties and favorabe modifications of arterial wave reflection characteristics. Furthermore, HT improves lipoprotein metabolism and reduces coronary artery plaque formation in animal models. Unfortunately, observational and interventional studies in postmenopausal women that have evaluated the impact of HT on cardiovascular changes have produced inconsistent and inconclusive results. Most studies agree that arterial stiffness increases after menopause, partly due to advancing age and reduced estrogen production. Results from most studies that were designed to investigate the effects of HT on arterial properties have shown a selective decrease in elastic artery stiffness with little effect on muscular arteries. This beneficial effect was observed only if estrogen alone was administered. The main objective of this review is to discuss the ill effects of arterial stiffness in general and attempt to translate information from previous experimental studies to those in postmenopausal women and explain the beneficial effects of HT on arterial stiffness and improvement in left ventricular afterload.
Collapse
Affiliation(s)
- Wilmer W Nichols
- a University of Florida College of Medicine, Division of Cardiovascular Medicine, PO Box 100277, Gainesville, FL 32610-0277, USA.
| | - Gary L Pierce
- b University of Colorado, Department of Integrative Physiology, Boulder, CO, USA.
| | - Randy W Braith
- c University of Florida, Department of Applied Physiology, Gainesville, FL 32611C, USA.
| |
Collapse
|
28
|
van den Meiracker AH, de Ronde W. Gender differences in vascular disease: not a simple explanation. J Hypertens 2007; 25:1193-4. [PMID: 17563529 DOI: 10.1097/hjh.0b013e32812908d3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
29
|
Sun D, Yan C, Jacobson A, Jiang H, Carroll MA, Huang A. Contribution of epoxyeicosatrienoic acids to flow-induced dilation in arteries of male ERalpha knockout mice: role of aromatase. Am J Physiol Regul Integr Comp Physiol 2007; 293:R1239-46. [PMID: 17634204 PMCID: PMC4536954 DOI: 10.1152/ajpregu.00185.2007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We studied the roles of estrogen receptors (ER) and aromatase in the mediation of flow-induced dilation (FID) in isolated arteries of male ERalpha-knockout (ERalpha-KO) and wild-type (WT) mice. FID was comparable between gracilis arteries of WT and ERalpha-KO mice. In WT arteries, inhibition of NO and prostaglandins eliminated FID. In ERalpha-KO arteries, N(omega)-nitro-L-arginine methyl ester (L-NAME) inhibited FID by approximately 26%, whereas indomethacin inhibited dilations by approximately 50%. The remaining portion of the dilation was abolished by additional administration of 6-(2-proparglyoxyphenyl)hexanoic acid (PPOH) or iberiotoxin, inhibitors of epoxyeicosatrienoic acid (EET) synthesis and large-conductance potassium channels, respectively. By using an electrophysiological technique, we found that, in the presence of 10 dyne/cm(2) shear stress, perfusate passing through donor vessels isolated from gracilis muscle of ERalpha-KO mice subjected to L-NAME and indomethacin elicited smooth muscle hyperpolarization and a dilator response of endothelium-denuded detector vessels. These responses were prevented by the presence of iberiotoxin in detector or PPOH in donor vessels. Gas chromatography-mass spectrometry (GC-MS) analysis indicated a significant increase in arterial production of EETs in ERalpha-KO compared with WT mice. Western blot analysis showed a significantly reduced endothelial nitric oxide synthase expression but enhanced expressions of aromatase and ERbeta in ERalpha-KO arteries. Treatment of ERalpha-KO arteries with specific aromatase short-interfering RNA for 72 h, knocked down the aromatase mRNA and protein associated with elimination of EET-mediation of FID. Thus, FID in male ERalpha-KO arteries is maintained via an endothelium-derived hyperpolarizing factor/EET-mediated mechanism compensating for reduced NO mediation due, at least in part, to estrogen aromatized from testosterone.
Collapse
MESH Headings
- 8,11,14-Eicosatrienoic Acid/antagonists & inhibitors
- 8,11,14-Eicosatrienoic Acid/pharmacology
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Aromatase/physiology
- Arteries/physiology
- Blotting, Western
- Estrogen Receptor alpha/drug effects
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/physiology
- Gas Chromatography-Mass Spectrometry
- In Vitro Techniques
- Indomethacin/pharmacology
- Male
- Membrane Potentials/drug effects
- Mice
- Mice, Knockout
- Muscle, Skeletal/enzymology
- Muscle, Skeletal/physiology
- NG-Nitroarginine Methyl Ester/pharmacology
- Nitric Oxide Synthase Type III/antagonists & inhibitors
- Organophosphorus Compounds/pharmacology
- Peptides/pharmacology
- Perfusion
- RNA, Small Interfering/pharmacology
- Reverse Transcriptase Polymerase Chain Reaction
- Stress, Mechanical
- Vasodilation/genetics
- Vasodilation/physiology
Collapse
Affiliation(s)
- Dong Sun
- Department of Physiology, New York Medical College, Valhalla, NY 10595, USA
| | | | | | | | | | | |
Collapse
|
30
|
Ling S, Zhou L, Li H, Dai A, Liu JP, Komesaroff PA, Sudhir K. Effects of 17beta-estradiol on growth and apoptosis in human vascular endothelial cells: influence of mechanical strain and tumor necrosis factor-alpha. Steroids 2006; 71:799-808. [PMID: 16806337 DOI: 10.1016/j.steroids.2006.05.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2005] [Revised: 05/11/2006] [Accepted: 05/16/2006] [Indexed: 11/22/2022]
Abstract
Vascular endothelial cell (EC) integrity is key to arterial health; endothelial dysfunction is linked to atherogenesis. Atherosclerosis shows a male preponderance, possibly related to the protective effect of estrogens in women. This study examined the effect of estrogens on growth, apoptosis and adhesion molecule expression in cultured human EC. The effects of 17beta-estradiol (E2) were studied in human umbilical vein endothelial cells (HUVEC) under normal culture conditions, and following exposure to cyclic mechanical strain or tumor necrosis factor alpha (TNFalpha). E2 enhanced HUVEC growth in serum-enriched media, in a concentration-dependent manner. This up-regulation of EC growth by E2 was associated with an increase in telomerase activity, assessed by PCR-based TRAP analysis. Cyclic strain enhanced [(3)H]-thymidine incorporation into DNA, and increased activation of mitogen-activated protein (MAP) kinase ERK1/2 and expression of early growth genes (Egr-1 and Sp-1); E2 attenuated the strain-induced ERK1/2 activation but not the early growth gene expression or DNA synthesis. TNFalpha (20 ng/mL) induced apoptosis in HUVEC, causing a decrease in DNA synthesis, increase in floating and Annexin-V-stained cell numbers, and morphological changes. TNFalpha also upregulated ERK1/2 activity and expression of adhesion molecules (ICAM-1, VCAM-1 and E-selectin). E2 significantly attenuated the effects of TNFalpha on ERK1/2 activity, apoptosis, and E-selectin expression in the cells. Thus, estradiol enhances growth and reduces TNFalpha-induced apoptosis in EC; enhanced EC growth may be mediated via upregulation of telomerase activity. These effects are possible cellular mechanisms underlying female gender-associated cardiovascular protection.
Collapse
Affiliation(s)
- Shanhong Ling
- Monash University Central and Eastern Clinical School, Alfred Hospital, Commercial Road, Melbourne, Vic. 3181, Australia.
| | | | | | | | | | | | | |
Collapse
|
31
|
Hwang J, Rouhanizadeh M, Hamilton RT, Lin TC, Eiserich JP, Hodis HN, Hsiai TK. 17beta-Estradiol reverses shear-stress-mediated low density lipoprotein modifications. Free Radic Biol Med 2006; 41:568-78. [PMID: 16863990 PMCID: PMC3320656 DOI: 10.1016/j.freeradbiomed.2006.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2005] [Revised: 03/23/2006] [Accepted: 04/10/2006] [Indexed: 12/21/2022]
Abstract
Within arterial bifurcations or branching points, oscillatory shear stress (OSS) induces oxidative stress mainly via the reduced nicotinamide adenine dinucleodtide phosphate (NADPH) oxidase system. It is unknown whether 17beta-estradiol (E(2)) can regulate OSS-mediated low-density lipoprotein (LDL) modifications. Bovine aortic endothelial cells were pretreated with E(2) at 5 nmol/L, followed by exposure to OSS (0 +/- 3.0 dynes/cm(2) s and 60 cycles/min) in a flow system. E(2) decreased OSS-mediated NADPH oxidase mRNA expression, and E(2)-mediated (.-)NO production was mitigated by the NO synthase inhibitor N(G)-nitro-l-argenine methyl ester. The rates of O(2)(-.) production in response to OSS increased steadily as determined by superoxide-dismutase-inhibited ferricytochrome c reduction; whereas, pretreatment with E(2) decreased OSS-mediated O(2)(-.) production (n = 4, p < 0.05). In the presence of native LDL (50 microg/mL), E(2) also significantly reversed OSS-mediated LDL oxidation as determined by high-performance liquid chromatography. In the presence of O(2)(-.) donor, xanthine oxidase (XO), E(2) further reversed XO-induced LDL lipid peroxidation (n = 3, p < 0.001). Mass spectra acquired in the m/z 400-1800 range, revealed XO-mediated LDL protein nitration involving tyrosine 2535 in the alpha-2 domains, whereas pretreatment with E(2) reversed nitration, as supported by the changes in nitrotyrosine intensities. Thus, E(2) plays an indirect antioxidative role. In addition to upregulation of endothelial (.-)NO synthase and downregulation of Nox4 expression, E(2) influences LDL modifications via lipid peroxidation and protein nitration.
Collapse
Affiliation(s)
- Juliana Hwang
- Department of Biomedical Engineering and Cardiovascular Medicine, Atherosclerosis Research Unit, University of Southern California, Los Angeles, CA 90081, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Giménez J, García MP, Serna M, Bonacasa B, Carbonell LF, Quesada T, Hernández I. 17β-Oestradiol enhances the acute hypotensive effect of captopril in female ovariectomized spontaneously hypertensive rats. Exp Physiol 2006; 91:715-22. [PMID: 16627572 DOI: 10.1113/expphysiol.2006.033449] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The objective of this study was to investigate whether the acute haemodynamic effects of angiotensin-converting enzyme inhibition with captopril could be enhanced by oestrogen administration, and then to evaluate the mechanisms involved in this enhancement. All experiments were performed in 18-week-old female spontaneously hypertensive rats arranged in three experimental groups: intact; ovariectomized (OVX); and ovariectomized plus treatment with 17beta-oestradiol (OVX + E2). These groups were used to evaluate the effects of captopril administration alone, or following bradykinin B2 receptor blockade or nitric oxide synthase inhibition, on a number of haemodynamic parameters (mean arterial pressure, cardiac index, vascular resistance and heart rate). The drop in mean arterial pressure and vascular resistance index in response to captopril was more pronounced in intact and ovariectomized rats treated with 17beta-oestradiol than in ovariectomized animals. Blockade of bradykinin B2 receptors or inhibition of nitric oxide synthesis attenuated the synergy between 17beta-oestradiol and captopril. It is concluded that ovariectomy blunted the blood pressure and vascular resistance index drop observed in intact rats in response to captopril. Treatment with 17beta-oestradiol prevented the blunted response to captopril in ovariectomized rats. Kinins and nitric oxide may be involved in the mechanisms of 17beta-oestradiol potentiation of the haemodynamic effects of captopril.
Collapse
Affiliation(s)
- José Giménez
- Department of Physiology, Faculty of Medicine, University of Murcia, Murcia, Spain
| | | | | | | | | | | | | |
Collapse
|
33
|
Sun D, Liu H, Yan C, Jacobson A, Ojaimi C, Huang A, Kaley G. COX-2 contributes to the maintenance of flow-induced dilation in arterioles of eNOS-knockout mice. Am J Physiol Heart Circ Physiol 2006; 291:H1429-35. [PMID: 16632543 PMCID: PMC4536927 DOI: 10.1152/ajpheart.01130.2005] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Our previous studies demonstrated that, in gracilis muscle arterioles of male mice deficient in the gene for endothelial nitric oxide synthase (eNOS), flow-induced dilation (FID) is mediated by endothelial PGs. Thus the present study aimed to identify the specific isoform of cyclooxygenase (COX) responsible for the compensatory mediation of FID in arterioles of eNOS-knockout (KO) mice. Experiments were conducted on gracilis muscle arterioles of male eNOS-KO and wild-type (WT) mice. Basal tone and magnitude of FID of arterioles were comparable in the two strains of mice. A role for COX isoforms in the mediation of the responses was assessed by use of valeryl salicylate (3 mM) and NS-398 (10 microM), inhibitors of COX-1 and COX-2, respectively. In eNOS-KO arterioles, valeryl salicylate or NS-398 alone inhibited FID (at maximal flow rate) by approximately 51% and approximately 58%, respectively. Administration of both inhibitors eliminated the dilation. In WT arterioles, inhibition of COX-2 did not significantly affect FID, whereas inhibition of COX-1 decreased the dilation by approximately 57%. The residual portion of the response was abolished by additional administration of Nomega-nitro-L-arginine methyl ester. Western blot analysis indicated a comparable content of COX-1 protein in arterioles of WT and eNOS-KO mice. COX-2 protein, which was not detectable in arterioles of WT mice, was strongly expressed in arterioles of eNOS-KO mice, together with an upregulation of COX-2 gene expression. Immunohistochemical staining confirmed the presence of COX-2 in the endothelium of eNOS-KO arterioles. In conclusion, COX-2-derived PGs are the mediators responsible for maintenance of FID in arterioles of eNOS-deficient mice.
Collapse
Affiliation(s)
- Dong Sun
- Department of Physiology, New York Medical College, Valhalla, NY 10595, USA.
| | | | | | | | | | | | | |
Collapse
|
34
|
Chancey AL, Gardner JD, Murray DB, Brower GL, Janicki JS. Modulation of cardiac mast cell-mediated extracellular matrix degradation by estrogen. Am J Physiol Heart Circ Physiol 2005; 289:H316-21. [PMID: 15722408 DOI: 10.1152/ajpheart.00765.2004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
There are fundamental differences between males and females with regard to susceptibility to heart disease. Although numerous animal models of heart failure have demonstrated that premenopausal females are afforded cardioprotection and, therefore, fare better in the face of cardiac disease than their male counterparts, many questions as to how this occurs still exist. Recently, we showed that 1) increased mast cell density is associated with adverse ventricular remodeling and 2) chemically induced mast cell degranulation using compound 48/80 resulted in remarkable changes in matrix metalloproteinase (MMP) activity, cardiac collagen structure, and cardiac diastolic function in normal male rats. With the known gender differences in cardiac disease in mind, we sought to examine the effects of chemically induced cardiac mast cell degranulation in isolated, blood-perfused hearts of intact female rats, ovariectomized female rats, and ovariectomized female rats treated with 17β-estradiol. In response to mast cell degranulation, no significant differences in cardiac function, MMP-2 activity, or collagen volume fraction were observed between intact female rats and ovariectomized female rats treated with estrogen. In the ovariectomized female group, a significant rightward shift in the left ventricular pressure-volume relation, accompanied by a marked 133% increase in active MMP-2 values over that in the intact female group, was noted after treatment with compound 48/80 ( P ≤ 0.05), along with a significant reduction in collagen volume fraction below control (0.46 ± 0.23 vs. 0.73 ± 0.13%, P ≤ 0.05). These findings indicate that estrogen's cardioprotective role can be partially mediated by its effects on cardiac mast cells, MMPs, and the extracellular matrix.
Collapse
Affiliation(s)
- Amanda L Chancey
- Dept. of Cell and Developmental Biology, School of Medicine, University of South Carolina, Columbia, SC 29208, USA
| | | | | | | | | |
Collapse
|
35
|
Ajayi AA, Ogungbade GO, Okorodudu AO. Sex hormone regulation of systemic endothelial and renal microvascular reactivity in type-2 diabetes: studies in gonadectomized and sham-operated Zucker diabetic rats. Eur J Clin Invest 2004; 34:349-57. [PMID: 15147332 DOI: 10.1111/j.1365-2362.2004.01339.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Male Zucker diabetic rats exhibit a more severe endotheliopathy in comparison with their female diabetic litter mates. The plasma concentrations of both thromboxanes and endothelins are elevated in diabetes, and the receptor cross-talk between TXA(2) and ET-1 receptors may be enhanced in type-2 diabetic Zucker rats. AIMS To determine the role of the endogenous sex steroid hormones, testosterone and estradiol on the systemic and renal microvascular reactivity to ET-1, thromboxane-mimetic U46619, ET-TXA(2) receptor interaction, and the nitric oxide vasodilator system in Zucker hypertensive-diabetic rats. METHODS Male and female Zucker rats aged 8-10 weeks were each divided into two groups. The male rats were castrated or underwent a sham operation. The female rats were spayed (bilateral ovariectomy and hysterectomy) or had a sham operation. All rats were studied 4-6 weeks after the gonadectomy or sham operations. Blood glucose and insulin as well as plasma concentrations of testosterone and estradiol were determined. Haemodynamic studies were undertaken with determination of the dose-response curve for mean arterial pressure (MAP), renal cortical flow (RCF) and renal medullary blood flow (MBF) in response to ET-1 and U46619, and the effect of interdiction of the ET-TXA(2) interaction with ET-antagonists BQ610 and BQ788. The role of endogenous NO was assessed by its response to graded acetylcholine doses and to a L-NG-nitro-arginine methyl ester (L-NAME) infusion. RESULTS Castrated male rats had a significantly lower blood glucose concentration (295 +/- 33 mg dL(-1)) compared with their sham-controls (481 +/- 40 mg dL(-1)), P = 0.008. Mean arterial pressure tended to be lower in the castrated rats. Gonadectomy reduced the plasma testosterone and estradiol concentrations. Castration abolished the hypotensive action of U46619 compared with sham-operated male rats (P < 0.0001, anova). Conversely, the pressor action of U46619 seen in the sham-operated female rats was reversed to a profound hypotensive action in the spayed rats (P < 0.001, anova). The change in MAP after U46619 was inversely correlated to the plasma testosterone concentration (r = -0.73, P = 0.027). The paradoxical hypotensive response elicited by ET-1 in the Zucker diabetic rats of both sexes was abolished by castration only (P < 0.005, anova). Castration caused a significant (P = 0.011) augmentation of the vasodilator response to acetylcholine, while spaying caused a slight attenuation. Castration, but not spaying, resulted in significant increases in MBF after U46619 (P = 0.003, anova), ET-1 (P = 0.005, anova) and acetylcholine (P = 0.053, anova). The ET-(B) antagonist BQ788 augmented the U46619-induced rise in MAP in castrated male rats, and also abolished the U46619-induced increase in MBF (P < 0.01 anova). L-NAME (25 mg kg(-1)) increased MAP and decreased MBF in the gonadectomized and sham-operated rats, except for the castrated male Zucker rats, where it significantly increased MBF (+90 +/- 31 PU) (P = 0.0004, anova) despite the increase in MAP. CONCLUSIONS Testosterone and estradiol regulate systemic and microvascular reactivity to TXA(2) receptor stimulation in type-2 diabetic Zucker rats. The impact of testosterone on blood glucose concentration, blood pressure, and the systemic and renal microcirculatory response to ET-1 and NO, as well as the endothelin-thromboxane receptor cross talk, is greater, and opposite to that of estradiol. The effects of testosterone withdrawal may at least in part be mediated by the ET-B receptor subtype and NO generation. Androgen blockade should be investigated further for the reversal or delay of hypertensive-diabetic endotheliopathy.
Collapse
Affiliation(s)
- A A Ajayi
- Center for Cardiovascular Diseases, Texas Southern University, Houston, Texas 77004, USA.
| | | | | |
Collapse
|
36
|
Graham D, Hamilton C, Beattie E, Spiers A, Dominiczak AF. Comparison of the effects of omapatrilat and irbesartan/hydrochlorothiazide on endothelial function and cardiac hypertrophy in the stroke-prone spontaneously hypertensive rat. J Hypertens 2004; 22:329-37. [PMID: 15076191 DOI: 10.1097/00004872-200402000-00017] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The novel antihypertensive agent, omapatrilat, is both an inhibitor of neutral endopeptidase and angiotensin-converting enzyme. This study investigated the effects of omapatrilat in comparison with an angiotensin I-receptor antagonist/diuretic combination on blood pressure, endothelial function and cardiac hypertrophy in stroke-prone spontaneously hypertensive rats (SHRSP). METHODS Male and female SHRSP were treated orally with omapatrilat or irbesartan plus hydrochlorothiazide (I + H) or vehicle for 8 weeks. Systolic blood pressure was measured weekly by tail-cuff. Cardiac hypertrophy was monitored by echocardiography at 8, 12 and 16 weeks of age. Endothelial function [basal nitric oxide (NO) bioavailability and stimulated NO release] was examined in carotid arteries using organ bath pharmacology and in mesenteric resistance arteries using wire myography. RESULTS Compared with untreated controls, omapatrilat and I + H significantly attenuated hypertension [male control, 198.3 +/- 6.9 mmHg versus omapatrilat, 149.6 +/- 3.8 mmHg (F = 8.63 P < 0.0001), versus I + H, 145.6 +/- 5.1 mmHg (F = 7.38 P < 0.0001); female control, 170.3 +/-8.3 mmHg versus omapatrilat, 120.0 +/- 4.6 mmHg (F = 8.36, P < 0.0001), versus I + H, 112.2 +/- 2.9 mmHg (F = 9.08, P < 0.0001)] and left ventricular hypertrophy [male + female controls, 3.02 +/- 0.38 mg/g versus omapatrilat, 2.47 +/- 0.26 mg/g (P < 0.0001; 95% confidence interval, 0.27, 0.83), versus I + H, 2.49 +/- 0.21 mg/g (P < 0.0001; 95% confidence interval, 0.25, 0.83)]. Both treatments also significantly increased male carotid artery basal NO bioavailability relative to control [control, 0.62 +/- 0.17 g/g versus omapatrilat, 1.95 +/- 0.17 g/g (P < 0.0001; 95% confidence interval, -1.83, -0.36), versus I + H, 1.57 +/- 0.21 g/g (P < 0.026; 95% confidence interval, -1.31, -0.12)]. However, stimulated NO (EC50) was only improved in omapatrilat-treated males [controls, 0.19 +/- 0.06 micromol/l versus omapatrilat, 0.05 +/- 0.01 micromol/l (P = 0.05; 95% confidence interval, -1.16, -0.03)]. CONCLUSIONS Omapatrilat treatment significantly reduced left ventricular hypertrophy and improved endothelial function in carotid arteries from male SHRSP by NO-dependent mechanisms. Despite equivalent antihypertensive and antihypertrophic actions, a similar improvement in endothelial function, specifically stimulated NO release, was not observed after treatment with I + H.
Collapse
Affiliation(s)
- Delyth Graham
- BHF Glasgow Cardiovascular Research Centre, Division of Cardiovascular and Medical Sciences, Western Infirmary, Glasgow, UK.
| | | | | | | | | |
Collapse
|
37
|
Rosano GMC, Vitale C, Silvestri A, Fini M. Hormone replacement therapy and cardioprotection: the end of the tale? Ann N Y Acad Sci 2004; 997:351-7. [PMID: 14644842 DOI: 10.1196/annals.1290.038] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of morbidity and mortality in women after the age of 50 years in most developed countries. Estrogen deficiency plays a key role in causing CVD in women. Apart from the direct effect of ovarian hormones on the vessel wall, cessation of ovarian function and the consequent reduction of sex steroid hormone levels have important metabolic and pathological implications that negatively influence the cardiovascular system. Therefore, the increased incidence of CVD observed in women after menopause should be considered on a multifactorial basis. Data available for the effects of ERT and HRT in the primary prevention of CVD are mainly observational. However, despite limitations related to this kind of study, it must be noted that their results consistently show a reduction in cardiovascular events in hormone users. Meta-analysis of epidemiological studies found that women who had ever used estrogens had a 34% overall reduction in the relative risk of cardiovascular events compared to those who had never used hormones. Most of the early epidemiological studies were conducted using unopposed estrogen replacement therapy. The number of studies evaluating the effects of estrogen-progestin replacement therapy is limited. Recently, the estrogen-progestin arm of the Women's Health Initiative (WHI) study has been stopped because of an increased incidence of breast cancer, and too early on to give any insight into possible cardiovascular effects. Comments on the cardiovascular effects of HRT from the results of the WHI study are therefore not warranted, as the study did not continue for a duration long enough to enable a calculation of cardiovascular end points. The WHI study included only one single type of estrogen-progestin association; whether different estrogen-progestin combinations, more commonly used outside the United States, may have a different effect is still a matter of speculation. A major difference between observational and randomized studies on the effect of ovarian hormones on cardiovascular function is the time of HRT initiation since menopause, which is significantly shorter in observational studies. In conclusion, the average 35-50% risk reduction in CVD with HRT in primary prevention in postmenopausal women is based on nonrandomized observational data.
Collapse
Affiliation(s)
- Giuseppe M C Rosano
- Department of Medical Sciences, Cardiovascular Research Unit, San Raffaele-Roma, Via della Pisana 234, 00163 Rome, Italy.
| | | | | | | |
Collapse
|
38
|
Huang A, Sun D, Wu Z, Yan C, Carroll MA, Jiang H, Falck JR, Kaley G. Estrogen elicits cytochrome P450--mediated flow-induced dilation of arterioles in NO deficiency: role of PI3K-Akt phosphorylation in genomic regulation. Circ Res 2003; 94:245-52. [PMID: 14670845 PMCID: PMC4536912 DOI: 10.1161/01.res.0000111525.96232.46] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This study investigated the mechanisms responsible for the estrogen-dependent, cytochrome P450 (CYP)-mediated dilator responses to shear stress in arterioles of NO-deficient female rats and mice. Flow-induced dilation (FID) was assessed in isolated arterioles from N(G)-nitro-L-arginine methyl ester (L-NAME)-treated male and ovariectomized female rats before and after overnight incubation with 17beta-estradiol (17beta-E2, 10(-9) mol/L). In control conditions, prostaglandins (PGs) mediated FID, because indomethacin (INDO) abolished the responses. After incubation of the vessels with 17beta-E2, the basal tone of arterioles was significantly reduced and FID was augmented. INDO did not affect the dilation of the vessels incubated with 17beta-E2. Dilations of these vessels, however, were eliminated by PPOH and miconazole, inhibitors of CYP/epoxygenase. Simultaneous incubation of the vessels with 17beta-E2 plus ICI, 182,780, an estrogen receptor antagonist, or wortmannin, an inhibitor of phosphatidylinositol 3-kinase (PI3K) phosphorylation or the transcriptional inhibitor DRB, prevented the reduced arteriolar tone and the enhanced CYP-mediated FID caused by incubation of vessels with 17beta-E2. Western blot analysis indicated a significantly increased phospho-Akt level in arterioles incubated with 17beta-E2 compared with those without 17beta-E2. The enhanced phospho-Akt in response to 17beta-E2 was localized, by immunohistochemistry, to arteriolar endothelial cells. Moreover, GC-MS analysis indicated a significantly increased production of epoxyeicosatrienoic acids, vasodilator metabolites of CYP/epoxygenase, in arterioles incubated with 17beta-E2, a response that was prevented by ICI 182780 and wortmannin, respectively. Thus, estrogen, via a receptor-dependent, PI3K/Akt-mediated pathway, transcriptionally upregulates CYP activity, leading to an enhanced arteriolar response to shear stress.
Collapse
Affiliation(s)
- An Huang
- Department of Physiology, New York Medical College, Valhalla, NY 10595, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
The cardiovascular protective actions of estrogen are partially mediated by a direct effect on the vessel wall. Estrogen is active both on vascular smooth muscle and endothelial cells where functionally competent estrogen receptors have been identified. Estrogen administration promotes vasodilation in humans and in experimental animals, in part by stimulating prostacyclin and nitric oxide synthesis, as well as by decreasing the production of vasoconstrictor agents such as cyclooxygenase-derived products, reactive oxygen species, angiotensin II, and endothelin-1. In vitro, estrogen exerts a direct inhibitory effect on smooth muscle by activating potassium efflux and by inhibiting calcium influx. In addition, estrogen inhibits vascular smooth muscle cell proliferation. In vivo, 17beta-estradiol prevents neointimal thickening after balloon injury and also ameliorates the lesions occurring in atherosclerotic conditions. As is the case for other steroids, the effect of estrogen on the vessel wall has a rapid non-genomic component involving membrane phenomena, such as alteration of membrane ionic permeability and activation of membrane-bound enzymes, as well as the classical genomic effect involving estrogen receptor activation and gene expression.
Collapse
Affiliation(s)
- R C Tostes
- Grupo de Pesquisa sobre Hipertensão Arterial, Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil.
| | | | | | | |
Collapse
|
40
|
Ajayi AA, Hercule H, Cory J, Hayes BE, Oyekan AO. Gender difference in vascular and platelet reactivity to thromboxane A(2)-mimetic U46619 and to endothelial dependent vasodilation in Zucker fatty (hypertensive, hyperinsulinemic) diabetic rats. Diabetes Res Clin Pract 2003; 59:11-24. [PMID: 12482637 DOI: 10.1016/s0168-8227(02)00180-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We examined the hypothesis that gender differences exist in platelet and vascular reactivity in type-2 diabetes mellitus, using Zucker fatty diabetic rats of both sexes and their lean littermates. Type-2 diabetes is characterized by excessive platelet production of TXA(2), which is thrombogenic. Testosterone up-regulates platelet TXA(2) receptors and the aggregation response to thromboxane mimetics. Conversely, estrogen increases vascular nitric oxide (NO) production and inhibits platelet aggregation. Hemodynamic studies were undertaken with the determination of dose-response curve for MAP and renal cortical blood flow (RCF) in response to U46619, angiotensin-II, phenylephrine and endothelin-1, as well as the systemic hemodynamic response to acetylcholine and L-NG nitro-arginine methylester (L-NAME). Platelet aggregation response was evaluated using whole blood impedance aggregometry. There were significant gender differences in the systemic blood pressure and RCF response to TXA(2)-mimetic U46619 and angiotensin-II (P<0.02, ANOVA) but not to phenylephrine or endothelin-1. Male rats exhibited a paradoxical hypotensive response to U46619 (-18+/-11 mmHg) compared with a peak pressor response of +6+/-1 mmHg in female rats (P<0.01, ANOVA). The male rats exhibited an attenuated systemic vasodilator response (P<0.001, ANOVA) to acetylcholine (fall in MAP in male diabetic rats being -24+/-8 mmHg, compared with a fall of -50+/-8 mmHg in females), but a greater rise in the renal cortical resistance in response to NO inhibition by L-NAME (P<0.03) compared with the female rats. Both the slope (46+/-2) and the peak magnitude of the U46619-induced whole blood platelet aggregation (13+/-1) ohms were significantly higher (P<0.01, ANOVA) in male (n=10) compared with female diabetic rats (n=8) (29+/-0.8 slope, 10.0+/-0.8 ohms, respectively). Thus, the male diabetic Zucker rats exhibited an impaired response to vasoconstrictors (U46619 and angiotensin-II) and to endothelial (NO)-mediated vasodilation. The male gender may therefore be associated with the greater prothrombotic activity and a worse impairment of endothelial reactivity in the type-2 diabetic state.
Collapse
Affiliation(s)
- A A Ajayi
- Center for Cardiovascular Diseases, Texas Southern University, 3100 Cleburne Avenue, Houston, TX 77004, USA.
| | | | | | | | | |
Collapse
|
41
|
Adamopoulos S, Leftheriotis D, Sbarouni E, Karavolias G, Kremastinos DT. Acute haemodynamic effects of oestrogen administration in male patients with chronic heart failure. Eur J Heart Fail 2002; 4:719-26. [PMID: 12453542 DOI: 10.1016/s1388-9842(02)00170-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Although there are many studies concerning the effects of long-term oestrogen administration on systemic haemodynamics in postmenopausal women, the effects of oestrogen in patients with chronic heart failure are not defined. AIM The goal of this study was to evaluate the acute haemodynamic effects of oestrogen in male patients with chronic heart failure. METHODS AND RESULTS We studied 15 men with advanced heart failure (NYHA II-IV, EF < 35%). A Swan-Ganz thermodilution catheter was advanced in their pulmonary artery and central haemodynamics were assessed at baseline, after placebo administration, and following 0.625 and 1.25 mg of oestrogen infusion. Simultaneously, all patients underwent limb plethysmography. Analysis of variance with repeated measures was used to compare the sequential measurements. Following oestrogen administration, right atrial, pulmonary artery and pulmonary capillary wedge pressures, as well as systemic, pulmonary and forearm vascular resistance were decreased; cardiac output, cardiac index, stroke volume, stroke volume index, stroke work index and forearm blood flow were increased. CONCLUSIONS In male patients with chronic heart failure, acute oestrogen administration improves the indices of cardiac systolic performance and decreases pulmonary and systemic vascular resistance. These findings imply a beneficial effect of oestrogen in selected patients with chronic heart failure.
Collapse
Affiliation(s)
- Stamatis Adamopoulos
- Second Department of Cardiology, Onassis Cardiac Surgery Centre, 356 Syngrou Avenue, 176 74 Athens, Greece
| | | | | | | | | |
Collapse
|
42
|
Svedas E, Nisell H, Vanwijk MJ, Nikas Y, Kublickiene KR. Endothelial dysfunction in uterine circulation in preeclampsia: can estrogens improve it? Am J Obstet Gynecol 2002; 187:1608-16. [PMID: 12501072 DOI: 10.1067/mob.2002.127378] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The purpose of this study was to evaluate whether a 3-hour incubation with 17beta-estradiol will enhance blood flow- and bradykinin-mediated dilatation and alter pressure-induced basal tone in myometrial resistance arteries from women with preeclampsia and to evaluate the role of nitric oxide in the responses that were observed. STUDY DESIGN Blood flow- and bradykinin-mediated dilatation and responses to intraluminal pressure of 60 and 80 mm Hg were compared before and after 3 hours of incubation with 17beta-estradiol (10(-8) mol/L) in isolated myometrial arteries with the pressure myography technique. In separate experiments, the role of nitric oxide on 17beta-estradiol-induced responses was evaluated in the presence of the nitric oxide synthase inhibitor (10(-4) mol/L). Endothelial morphologic condition was evaluated by scanning electron microscopy. RESULTS Incubation with 17beta-estradiol significantly improved blood flow-mediated dilatation compared with initial blood flow-mediated response in arteries from women with preeclampsia. This effect was nitric oxide mediated, because the nitric oxide synthase inhibitor abolished the response. Arteries from women with preeclampsia demonstrated impaired bradykinin-mediated dilatation compared with that obtained in arteries from normal pregnant women. The 17beta-estradiol had no effect on bradykinin-mediated dilatation in arteries from women with preeclampsia. The enhanced pressure-induced tone at 80 mm Hg compared with the tone that developed at 60 mm Hg in arteries from women with preeclampsia was reduced after incubation with 17beta-estradiol. This reduction was also nitric oxide mediated. Morphologic signs of endothelial dysfunction were evident in arteries from women with preeclampsia. CONCLUSION The 17beta-estradiol improved impaired blood flow-mediated dilatation and reduced basal tone through a nitric oxide-mediated pathway in isolated myometrial arteries from women with preeclampsia.
Collapse
Affiliation(s)
- Eimantas Svedas
- Section for Obstetrics and Gynecology, Department of Clinical Science, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
43
|
Abstract
Puberty accelerates microvascular complications of diabetes mellitus, including nephropathy. Animal studies confirm a different renal hypertrophic response to diabetes before and after puberty, probably due to differences in the production of transforming growth factor-beta (TGF-beta). Many of the complex physiological changes during puberty could affect potentially pathogenic mechanisms of diabetic kidney disease. Increased blood pressure, activation of the growth hormone-insulin-like growth factor I axis, and production of sex steroids could all play a role in pubertal susceptibility to diabetic renal hypertrophy and nephropathy. These factors may influence the effects of hyperglycemia and several systems that ultimately control TGF-beta production, including the renin-angiotensin system, cellular redox systems, the polyol pathway, and protein kinase C. These phenomena may also explain gender differences in kidney function and incidence of end-stage renal disease. Normal changes during puberty, when coupled with diabetes and superimposed on a genetically susceptible milieu, are capable of accelerating diabetic hypertrophy and microvascular lesions. A better understanding of these processes may lead to new treatments to prevent renal failure in diabetes mellitus.
Collapse
Affiliation(s)
- Pascale H Lane
- Department of Pediatrics, University of Nebraska Medical Center, Omaha 68198-2169, USA.
| |
Collapse
|
44
|
Nevala R, Lassila M, Finckenberg P, Paukku K, Korpela R, Vapaatalo H. Genistein treatment reduces arterial contractions by inhibiting tyrosine kinases in ovariectomized hypertensive rats. Eur J Pharmacol 2002; 452:87-96. [PMID: 12323389 DOI: 10.1016/s0014-2999(02)02270-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The aim of the present study was to evaluate the vascular effects of genistein in a short-term study. The ovariectomized spontaneously hypertensive rats (SHR) were divided into four groups (n = 8 in each), which received the following subcutaneous treatments either for 2 days or for 2 weeks: (1) solvent control (96% dimethylsulphoxide (DMSO) 1 ml/kg), (2) estradiol-17beta (25 microg/kg), (3) genistein (2.5 mg/kg; low-dose), and (4) genistein (25 mg/kg; high-dose). The renal arterial rings were studied using organ bath system. The renal artery contractions were attenuated by the 2-day low-dose genistein treatment as follows: angiotensin II (46%), noradrenaline (42%) KCl (36%), and endothelin-1 (34%). Only the angiotensin II-induced contractions were reduced by the 2-week treatment with estradiol-17beta (38%) and with the low-dose of genistein (31%). The 2-day genistein treatment reduced tyrosine phosphorylation, while the other treatments or treatment times had no effect. The 2-day low-dose genistein treatment had no estrogenic effect on the uterine morphology. The mechanism for attenuated contractility in the renal arteries after the 2-day low-dose genistein treatment is independent of the estrogenic effect of genistein, but is due to the tyrosine kinase inhibitory property of genistein.
Collapse
Affiliation(s)
- Riikka Nevala
- Biomedicum, Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Helsinki, PO Box 63, FIN-00014 Helsinki, Finland.
| | | | | | | | | | | |
Collapse
|
45
|
Li H, Wallerath T, Förstermann U. Physiological mechanisms regulating the expression of endothelial-type NO synthase. Nitric Oxide 2002; 7:132-47. [PMID: 12223183 DOI: 10.1016/s1089-8603(02)00127-1] [Citation(s) in RCA: 163] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Although endothelial nitric oxide synthase (eNOS) is a constitutively expressed enzyme, its expression is regulated by a number of biophysical, biochemical, and hormonal stimuli, both under physiological conditions and in pathology. This review summarizes the recent findings in this field. Shear stress, growth factors (such as transforming growth factor-beta, fibroblast growth factor, vascular endothelial growth factor, and platelet-derived growth factor), hormones (such as estrogens, insulin, angiotensin II, and endothelin 1), and other compounds (such as lysophosphatidylcholine) upregulate eNOS expression. On the other hand, the cytokine tumor necrosis factor-alpha and bacterial lipopolysaccharide downregulate the expression of this enzyme. The growth status of cells, the actin cytoskeleton, and NO itself are also important regulators of eNOS expression. Both transcriptional and posttranscriptional mechanisms are involved in the expressional regulation of eNOS. Different signaling pathways are involved in the regulation of eNOS promoter activity and eNOS mRNA stability. Changes in eNOS expression and activity under pathophysiological conditions and the pharmacological modulation of eNOS expression are subject of a subsequent brief review (part 2) to be published in the next issue of this journal.
Collapse
Affiliation(s)
- Huige Li
- Department of Pharmacology, Johannes Gutenberg University, Obere Zahlbacher Strasse 67, Mainz D-55101, Germany
| | | | | |
Collapse
|
46
|
Christopher TA, Lopez BL, Stillwagon JC, Gao F, Gao E, Ma XL, Ohlstein EH, Yue TL. Idoxifene causes endothelium-dependent, nitric oxide-mediated vasorelaxation in male rats. Eur J Pharmacol 2002; 446:139-43. [PMID: 12098595 DOI: 10.1016/s0014-2999(02)01821-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Selective estrogen receptor modulators are estrogen-like compounds that lack the deleterious effects of estrogen. The present study was designed to determine whether idoxifene, a new selective estrogen receptor modulator, may have a vasodilatory effect on aortic vessels from male animals, and if so, to investigate the mechanism (i.e., endothelium-independent, direct vasorelaxation vs. endothelium-dependent, nitric oxide mediated vasorelaxation) by which idoxifene may exert its vasodilatory effect. Superior mesenteric arterial rings from adult male Sprague-Dawley rats were suspended in Krebs-Henseleit ring baths. Rings were contracted with 50 nM U-46619 (9,11-epoxymethano-PGH(2)), a thromboxane A(2) mimetic. Cumulative dose-response vasorelaxation to idoxifene (0.01 to 3 microM) was studied in the presence and absence of 200 microM N(omega)-nitro-L-arginine methyl ester (L-NAME, a nitric oxide synthase (NOS) inhibitor). The results obtained from idoxifene were compared with those from 17beta-estradiol. Our experimental results demonstrated that addition of idoxifene to superior mesenteric arterial rings isolated from male rats resulted in a dose-dependent vasorelaxation in the range of 0.1 microM (minimal vasodilatory concentration) to 3 microM (maximal vasodilatory concentration). Pre-treatment with L-NAME to block nitric oxide (NO) production virtually abolished idoxifene-induced vasodilatation, indicating that idoxifene caused an NO-mediated vasorelaxation in vessels from male animals. Addition of 17beta-estradiol also resulted in an endothelium-dependent vasorelaxation in aortic rings from male rats. However, these vessels were 30-fold less sensitive to 17beta-estradiol than to idoxifene in their vasorelaxation responses. Taken together, these results demonstrate that selective estrogen receptor modulators are superior to traditional estrogen in their vascular protection and may thus have potential therapeutic use in protection against cardiovascular disease, especially in male patients.
Collapse
Affiliation(s)
- Theodore A Christopher
- Department of Emergency Medicine, Jefferson Medical College, 1020 Sansom Street, Philadelphia, PA 19107-5004, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Bucci M, Roviezzo F, Cicala C, Pinto A, Cirino G. 17-beta-oestradiol-induced vasorelaxation in vitro is mediated by eNOS through hsp90 and akt/pkb dependent mechanism. Br J Pharmacol 2002; 135:1695-700. [PMID: 11934809 PMCID: PMC1573306 DOI: 10.1038/sj.bjp.0704641] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
1. The L-arginine-NO pathway has been implicated in the vasorelaxant effect of 17-beta-oestradiol. Here we have addressed the involvement of two distinct activation steps of endothelial nitric oxide synthase (eNOS) in the 17-beta-oestradiol-induced vasorelaxant effect on rat aortic rings. 2. Rat aortic rings contracted with phenylephrine (PE) 1 microM relaxed in a concentration related fashion to 17-beta-oestradiol water soluble cyclodextrin-encapsulated (E2) only when endothelium was present. The pure anti-oestrogen of E2 receptor ICI 182,780 (20 microM) significantly inhibited E2-induced vasorelaxation. 3. Geldanamycin (10 microM), a specific inhibitor of heat shock protein 90 (hsp90) and N(omega)-nitro-L-arginine-methyl ester (L-NAME, 100 microM), a nitric oxide synthase inhibitor, significantly inhibited E2-induced vasorelaxation. 4. Incubation of rat aortic rings up to 6 h with LY 294002 (25 microM), a specific inhibitor of PI(3)K akt/pkb pathway reduced E2-induced vasorelaxation. 5. Incubation of rat isolated aorta with E2, induced prostacyclin (PGI(2)) release. PGI(2) levels, measured as 6-keto PGF(1alpha), were abolished by ibuprofen (10 microM), both L-NAME and GA did not influence basal or E2-stimulated PGI(2) confirming the specificity of these two compounds on eNOS pathway. 6. In conclusion, we demonstrate that E2 interaction with its receptor is followed by a vasorelaxant effect in rat aortic rings mediated by eNOS activation through both hsp90 and akt/pkb dependent mechanisms.
Collapse
Affiliation(s)
- Mariarosaria Bucci
- Department of Pharmaceutical Science, Faculty of Pharmacy, University of Salerno, Fisciano (SA) Italy
| | - Fiorentina Roviezzo
- Department of Experimental Pharmacology, Faculty of Pharmacy, University of Naples – Federico II, Naples Italy
| | - Carla Cicala
- Department of Experimental Pharmacology, Faculty of Pharmacy, University of Naples – Federico II, Naples Italy
| | - Aldo Pinto
- Department of Pharmaceutical Science, Faculty of Pharmacy, University of Salerno, Fisciano (SA) Italy
| | - Giuseppe Cirino
- Department of Experimental Pharmacology, Faculty of Pharmacy, University of Naples – Federico II, Naples Italy
- Author for correspondence:
| |
Collapse
|
48
|
Gardner JD, Brower GL, Janicki JS. Gender differences in cardiac remodeling secondary to chronic volume overload. J Card Fail 2002; 8:101-7. [PMID: 12016634 DOI: 10.1054/jcaf.2002.32195] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND Gender differences in the prevalence of cardiovascular disease, both clinical and experimental, led us to evaluate the influence of gender on ventricular remodeling induced by chronic volume overload. METHODS AND RESULTS Chronic volume overload was induced in male and female rats via infrarenal aortocaval fistula. Ventricular function was assessed 8 weeks after fistula surgery in surviving rats. Left ventricular, right ventricular, and lung weights were measured. Mortality in female rats was 10-fold less than in male rats after 8 weeks of volume overload. Both sexes had significant increases in left ventricular weights relative to controls (77% increase for female v 114% for male rats; P <.05). Corresponding increases were also observed in right ventricular weight (134% for female v 161% for male rats; P <.05). However, lung weight was significantly increased only in males. In contrast to males, female rats had no indications of congestive heart failure. Male rats had marked dilatation and increased compliance, whereas female rats had no significant change in left ventricular dilatation or compliance. CONCLUSIONS There are clear gender-specific differences in ventricular function, structural remodeling, and mortality induced by chronic volume overload in this model of heart failure.
Collapse
Affiliation(s)
- Jason D Gardner
- Department of Anatomy, Physiology, and Pharmacology, Auburn University, Auburn, Alabama 36849-5517, USA
| | | | | |
Collapse
|
49
|
Mayhan WG, Sun H, Irvine SD. Influence of gender on dilatation of the basilar artery during diabetes mellitus. Brain Res 2002; 930:182-90. [PMID: 11879808 DOI: 10.1016/s0006-8993(02)02245-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The first goal of the present study was to examine the influence of gender on reactivity of the basilar artery. The second goal of this study was to examine the effect of diabetes mellitus on reactivity of the basilar artery in male and female rats. We examined in vivo responses of the basilar artery in male and female nondiabetic and diabetic rats in response to a nitric oxide synthase (NOS)-dependent (acetylcholine) and -independent (nitroglycerin) agonist. In nondiabetic male and female rats, acetylcholine and nitroglycerin produced dose-related dilatation of the basilar artery. However, the magnitude of vasodilatation in response to acetylcholine and a high concentration of nitroglycerin was significantly greater in female than in male rats. Acetylcholine (1.0 microM) dilated the basilar artery by 11 +/- 2% in nondiabetic males versus 25 +/- 4% in nondiabetic females (P<0.05). Nitroglycerin (1.0 microM) dilated the basilar artery by 37 +/- 8% in nondiabetic males versus 62 +/- 5% in nondiabetic females (P<0.05). Thus, there is a significant effect of gender on reactivity of the basilar artery during physiologic conditions. Dilatation of the basilar artery in response to acetylcholine, but not nitroglycerin, was impaired in diabetic male and female rats compared to their nondiabetic counterparts. Acetylcholine (1.0 microM) dilated the basilar artery by only 5 +/- 1% in diabetic males and by only 4 +/- 1% in diabetic females. In summary, dilatation of the basilar artery in response to NOS-dependent agonist was significantly greater in nondiabetic female than in nondiabetic male rats. In addition, diabetes mellitus impaired NOS-dependent dilatation of the basilar artery not only in male rats, but also in female rats. We suggest that the results of these studies provide insight into the pathogenesis of cerebrovascular abnormalities observed in postmenopausal women.
Collapse
Affiliation(s)
- William G Mayhan
- Department of Physiology and Biophysics, University of Nebraska Medical Center, 984575 Nebraska Medical Center, Omaha, NE 68198-4575, USA.
| | | | | |
Collapse
|
50
|
Joyce JM, Phernetton TM, Shaw CE, Modrick ML, Magness RR. Endothelial vasodilator production by uterine and systemic arteries. IX. eNOS gradients in cycling and pregnant ewes. Am J Physiol Heart Circ Physiol 2002; 282:H342-8. [PMID: 11748080 DOI: 10.1152/ajpheart.00438.2001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The follicular phase (FOL) and pregnancy exhibit increases in uterine blood flow (UBF), estrogen levels, and uterine artery (UA) endothelial nitric oxide synthase (eNOS) expression. UA branching within the mesometrium increases the total vascular cross-sectional area, which reduces the vascular perfusion pressure gradient, thus locally decreasing the blood flow velocity. Shear stress (SS) activates eNOS and may be associated with UBF elevations during FOL and pregnancy. We hypothesized that regional differences in eNOS responses are observed with both decreases in vessel diameter and during the ovarian cycle and pregnancy. Endothelial isolated proteins were collected from renal (RA) and internal iliac arteries (II) as well as from primary (UA 1 degrees ), secondary (UA 2 degrees), and tertiary (UA 3 degrees) UA branches of nonpregnant luteal phase (LUT; n = 6) and FOL (n = 6) as well as midpregnant (MP; 82 +/- 1 days gestation, n = 6) and late pregnant (LP; 127 +/- 3 days gestation, n = 6) ewes (term = 145 +/- 3 days gestation) for Western blot analysis. LUT RA, II, and UA 1 degrees eNOS levels were similar. There was a 60.7 +/- 9.8% reduction in eNOS expression in UA 2 degrees and UA 3 degrees. A similar decreasing eNOS regional expression gradient was observed in LP ewes. No eNOS regional expression gradient was observed in FOL or MP ewes because eNOS increased in UA 2 degrees and UA 3 degrees. In UA 2 degrees and UA 3 degrees, MP > LP = FOL > LUT. Thus, with increasing UBF, FOL and pregnancy rises in SS may regulate eNOS protein expression in smaller diameter UAs. A decrease in LUT and LP UA 2 degrees and UA 3 degrees endothelial eNOS suggest a possible negative feedback mechanism due to downregulation of eNOS if SS is normalized.
Collapse
Affiliation(s)
- Jeffrey M Joyce
- Perinatal Research Laboratories, Department of Obstetrics and Gynecology, University of Wisconsin, 202 S Park St., Madison, WI 53715, USA
| | | | | | | | | |
Collapse
|