Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
2.
Am J Physiol Heart Circ Physiol ; 306(5): H628-40, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24414072

ABSTRACT

The prevalence of left ventricular diastolic dysfunction (LVDD) sharply increases in women after menopause and may lead to heart failure. While evidence suggests that estrogens protect the premenopausal heart from hypertension and ventricular remodeling, the specific mechanisms involved remain elusive. Moreover, whether there is a protective role of estrogens against cardiovascular disease, and specifically LVDD, continues to be controversial. Clinical and basic science have implicated activation of the renin-angiotensin-aldosterone system (RAAS), linked to the loss of ovarian estrogens, in the pathogenesis of postmenopausal diastolic dysfunction. As a consequence of increased tissue ANG II and low estrogen, a maladaptive nitric oxide synthase (NOS) system produces ROS that contribute to female sex-specific hypertensive heart disease. Recent insights from rodent models that mimic the cardiac phenotype of an estrogen-insufficient or -deficient woman (e.g., premature ovarian failure or postmenopausal), including the ovariectomized congenic mRen2.Lewis female rat, provide evidence showing that estrogen modulates the tissue RAAS and NOS system and related intracellular signaling pathways, in part via the membrane G protein-coupled receptor 30 (GPR30; also called G protein-coupled estrogen receptor 1). Complementing the cardiovascular research in this field, the echocardiographic correlates of LVDD as well as inherent limitations to its use in preclinical rodent studies will be briefly presented. Understanding the roles of estrogen and GPR30, their interactions with the local RAAS and NOS system, and the relationship of each of these to LVDD is necessary to identify new therapeutic targets and alternative treatments for diastolic heart failure that achieve the cardiovascular benefits of estrogen replacement without its side effects and contraindications.


Subject(s)
Diastole , Estrogens/metabolism , Myocardium/metabolism , Ventricular Dysfunction, Left/metabolism , Ventricular Function, Left , Age Factors , Animals , Diastole/drug effects , Disease Models, Animal , Estrogen Replacement Therapy , Estrogens/deficiency , Estrogens/therapeutic use , Female , Humans , Nitric Oxide Synthase/metabolism , Postmenopause/metabolism , Reactive Oxygen Species/metabolism , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Renin-Angiotensin System , Risk Factors , Sex Factors , Signal Transduction , Ultrasonography , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left/drug effects
3.
Ther Adv Cardiovasc Dis ; 7(6): 293-305, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24280597

ABSTRACT

BACKGROUND: Knowledge of hemodynamic factors accounting for the development of hypertension should help to tailor therapeutic approaches and improve blood pressure control. Few data exist regarding sex differences of hemodynamic factors contributing to hypertension progression among patients with untreated nondiabetic stage I and II prehypertension (PreHyp) as defined by the Joint National Committee-7 guidelines (JNC-7). METHODS: We utilized noninvasive impedance cardiography, applanation tonometry and plasma measures of angiotensin II, angiotensin (1-7), serum aldosterone, high-sensitivity C-reactive protein (hs-CRP) and cytokine biomarkers of inflammation to characterize the hemodynamic and hormonal profile of 100 patients with untreated hypertension (39 women). RESULTS: Despite there being no differences between women and men in terms of office blood pressure, heart rate and body mass index, men demonstrated lower values of pulse pressure, systemic vascular resistance, brachial artery pulse wave velocity and augmentation index. In each of the three hypertension categories, the increased blood pressure in men was associated with significant augmentations in stroke volume and cardiac output compared with women. Sex-related hemodynamic differences were associated in women with higher plasma levels of leptin, hs-CRP, plasma angiotensin II and serum aldosterone, and no differences in the serum concentrations of cytokinins. In women but not men, hs-CRP correlated with plasma concentrations of transforming growth factor ß1 (TGFß1) and body weight; in addition, plasma TGFß1 correlated with levels of serum vascular cell adhesion molecule 1. CONCLUSION: The impact of sex differences in the hemodynamic factors accounting for the elevation in arterial pressure in subjects with essential hypertension has been poorly characterized or this information is not available. We suggest that this gap in knowledge may adversely influence choices of drug treatment since our study shows for the first time significant differences in the hemodynamic and hormonal mechanisms accounting for the increased blood pressure in women compared to men.


Subject(s)
Hemodynamics , Hormones/blood , Hypertension/blood , Hypertension/physiopathology , Biomarkers/blood , Cardiography, Impedance , Female , Humans , Hypertension/diagnosis , Inflammation Mediators/blood , Male , Manometry , Middle Aged , Prehypertension/blood , Prehypertension/diagnosis , Prehypertension/physiopathology , Prognosis , Renin-Angiotensin System , Sex Factors
4.
PLoS One ; 8(10): e76992, 2013.
Article in English | MEDLINE | ID: mdl-24204720

ABSTRACT

The cardioprotective effects of estrogen are well recognized, but the mechanisms remain poorly understood. Accumulating evidence suggests that the local cardiac renin-angiotensin system (RAS) is involved in the development and progression of cardiac hypertrophy, remodeling, and heart failure. Estrogen attenuates the effects of an activated circulating RAS; however, its role in regulating the cardiac RAS is unclear. Bilateral oophorectomy (OVX; n = 17) or sham-operation (Sham; n = 13) was performed in 4-week-old, female mRen2.Lewis rats. At 11 weeks of age, the rats were randomized and received either 17 ß-estradiol (E2, 36 µg/pellet, 60-day release, n = 8) or vehicle (OVX-V, n = 9) for 4 weeks. The rats were sacrificed, and blood and hearts were used to determine protein and/or gene expression of circulating and tissue RAS components. E2 treatment minimized the rise in circulating angiotensin (Ang) II and aldosterone produced by loss of ovarian estrogens. Chronic E2 also attenuated OVX-associated increases in cardiac Ang II, Ang-(1-7) content, chymase gene expression, and mast cell number. Neither OVX nor OVX+E2 altered cardiac expression or activity of renin, angiotensinogen, angiotensin-converting enzyme (ACE), and Ang II type 1 receptor (AT1R). E2 treatment in OVX rats significantly decreased gene expression of MMP-9, ACE2, and Ang-(1-7) mas receptor, in comparison to sham-operated and OVX littermates. E2 treatment appears to inhibit upsurges in cardiac Ang II expression in the OVX-mRen2 rat, possibly by reducing chymase-dependent Ang II formation. Further studies are warranted to determine whether an E2-mediated reduction in cardiac chymase directly contributes to this response in OVX rats.


Subject(s)
Estrogens/pharmacology , Myocardium/metabolism , Ovariectomy , Renin-Angiotensin System/physiology , Angiotensin I/metabolism , Angiotensinogen/genetics , Angiotensinogen/metabolism , Animals , Blotting, Western , Chymases/genetics , Chymases/metabolism , Estradiol/blood , Estradiol/metabolism , Estradiol/pharmacology , Estrogens/blood , Estrogens/metabolism , Female , Gene Expression/drug effects , Immunohistochemistry , Mast Cells/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Peptide Fragments/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Random Allocation , Rats , Rats, Inbred Lew , Rats, Transgenic , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Renin/genetics , Renin/metabolism , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/genetics , Reverse Transcriptase Polymerase Chain Reaction
5.
Menopause ; 20(8): 860-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23481117

ABSTRACT

OBJECTIVE: mRen2.Lewis rats exhibit exacerbated increases in blood pressure, left ventricular (LV) remodeling, and diastolic impairment after the loss of estrogens. In this same model, depletion of estrogens has marked effects on the cardiac biopterin profile concomitant with suppressed nitric oxide release. With respect to the establishment of overt systolic hypertension after oophorectomy (OVX), we assessed the effects of timing long-term 17ß-estradiol (E2) therapy on myocardial function, myocardial structure, and the cardiac nitric oxide system. METHODS: OVX (n = 24) or sham operation (Sham; n = 13) was performed in 4-week-old female mRen2.Lewis rats. After randomization, OVX rats received E2 immediately (OVX + E2-early; n = 7), E2 at 11 weeks of age (OVX + E2-late; n = 8), or no E2 at all (OVX; n = 9). RESULTS: E2-early was associated with lower body weight, less hypertension-related cardiac remodeling, and decreased LV filling pressure compared with OVX rats without E2 supplementation. E2-late similarly attenuated the adverse effects of ovarian hormone loss on tissue Doppler-derived LV filling pressures and perivascular fibrosis, and significantly improved myocardial relaxation or mitral annular velocity (e'). Early and late exposures to E2 decreased dihydrobiopterin, but only E2-late yielded significant increases in cardiac nitrite concentrations. CONCLUSIONS: Although there are some similarities between E2-early and E2-late treatments in relation to preservation of diastolic function and cardiac structure after OVX, the lusitropic potential of E2 is most consistent with late supplementation. The cardioprotective effects of E2-late are independent of blood pressure and may have occurred through regulation of cardiac biopterins and nitric oxide production.


Subject(s)
Estradiol/administration & dosage , Heart/drug effects , Ovariectomy , Animals , Biopterins/metabolism , Diastole/drug effects , Diastole/physiology , Estradiol/blood , Female , Heart/anatomy & histology , Heart/physiopathology , Hypertension/complications , Hypertension/prevention & control , Myocardium/pathology , Nitric Oxide/biosynthesis , Rats , Rats, Inbred Lew , Time Factors , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/prevention & control
6.
Cardiovasc Res ; 94(1): 96-104, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22328091

ABSTRACT

AIMS: GPR30 is a novel oestrogen receptor expressed in various tissues, including the heart. We determined the role of GPR30 in the maintenance of left ventricular (LV) structure and diastolic function after the surgical loss of ovarian hormones in the female mRen2.Lewis rat, a model emulating the cardiac phenotype of the post-menopausal woman. METHODS AND RESULTS: Bilateral oophorectomy (OVX) or sham surgery was performed in study rats; the selective GPR30 agonist, G-1 (50 µg/kg/day), or vehicle was given subcutaneously to OVX rats from 13-15 weeks of age. Similar to the cardiac phenotype of sham rats, G-1 preserved diastolic function and structure relative to vehicle-treated OVX littermates independent of changes in blood pressure. G-1 limited the OVX-induced increase in LV filling pressure, LV mass, wall thickness, interstitial collagen deposition, atrial natriuretic factor and brain natriuretic peptide mRNA levels, and cardiac NAD(P)H oxidase 4 (NOX4) expression. In vitro studies showed that G-1 inhibited angiotensin II-induced hypertrophy in H9c2 cardiomyocytes, evidenced by reductions in cell size, protein content per cell, and atrial natriuretic factor mRNA levels. The GPR30 antagonist, G15, inhibited the protective effects of both oestradiol and G-1 on this hypertrophy. CONCLUSION: These data show that the GPR30 agonist G-1 mitigates the adverse effects of oestrogen loss on LV remodelling and the development of diastolic dysfunction in the study rats. This expands our knowledge of the sex-specific mechanisms underlying diastolic dysfunction and provides a potential therapeutic target for reducing the progression of this cardiovascular disease process in post-menopausal women.


Subject(s)
Cardiotonic Agents/pharmacology , Cyclopentanes/pharmacology , Myocytes, Cardiac/drug effects , Ovariectomy , Quinolines/pharmacology , Receptors, G-Protein-Coupled/agonists , Renin/genetics , Ventricular Dysfunction, Left/prevention & control , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Angiotensin II/metabolism , Animals , Atrial Natriuretic Factor/metabolism , Blood Pressure/drug effects , Calcium/metabolism , Cardiotonic Agents/administration & dosage , Cell Line , Collagen/metabolism , Cyclopentanes/administration & dosage , Echocardiography, Doppler , Estrogens/metabolism , Female , Gene Expression Regulation/drug effects , Genotype , Hypertrophy, Left Ventricular/diagnostic imaging , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/prevention & control , Injections, Subcutaneous , Mice , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , NADPH Oxidase 4 , NADPH Oxidases/metabolism , Natriuretic Peptide, Brain/metabolism , Phenotype , Quinolines/administration & dosage , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Rats, Transgenic , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Renin/metabolism , Time Factors , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology
7.
J Gerontol A Biol Sci Med Sci ; 66(5): 521-33, 2011 May.
Article in English | MEDLINE | ID: mdl-21393424

ABSTRACT

Fish oil (FO) mediates a number of cardioprotective benefits in patients with cardiovascular disease. In the absence of cardiovascular disease, however, the effects of FO on cardiac structure and function are not clear. In addition, it is not known if an effective dosing strategy for attenuating age-related cardiac dysfunction is also effective at limiting cognitive dysfunction. Therefore, we determined if 4 months of FO supplementation in aged rats would lessen age-related cardiac dysfunction while concomitantly preventing the cognitive decline that is normally observed in this population. The results indicate that FO initiated late in life modifies diastolic function in a small but positive way by attenuating the age-related increases in filling pressure, posterior wall thickness, and interstitial collagen without mitigating age-related deficits in memory or increases in brain inflammation. These data raise the possibility that FO supplementation for purposes of cardiac and brain protection may need to occur earlier in the life span.


Subject(s)
Aging/physiology , Brain/pathology , Diastole/drug effects , Dietary Fats, Unsaturated/pharmacology , Fish Oils/pharmacology , Memory/drug effects , Aging/drug effects , Animals , Body Weight/drug effects , Brain/drug effects , Cell Count , Cognition/drug effects , Cognition/physiology , Diastole/physiology , Encephalitis , Fatty Acids, Omega-3/pharmacology , Hippocampus/drug effects , Hippocampus/pathology , Immunohistochemistry , Male , Motor Activity/drug effects , Motor Activity/physiology , Organ Size/drug effects , Rats , Vision, Ocular/drug effects , Vision, Ocular/physiology
8.
Endocrinology ; 152(6): 2428-36, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21427216

ABSTRACT

After oophorectomy, mRen2.Lewis rats exhibit diastolic dysfunction associated with elevated superoxide, increased cardiac neuronal nitric oxide synthase (nNOS) expression, and diminished myocardial tetrahydrobiopterin (BH4) content, effects that are attenuated with selective nNOS inhibition. BH4 is an essential cofactor of nNOS catalytic activity leading to nitric oxide production. Therefore, we assessed the effect of 4 wk BH4 supplementation on diastolic function and left ventricular (LV) remodeling in oophorectomized mRen2.Lewis rats compared with sham-operated controls. Female mRen2.Lewis rats underwent either bilateral ovariectomy (OVX) (n = 19) or sham operation (n = 13) at 4 wk of age. Beginning at 11 wk of age, OVX rats were randomized to receive either BH4 (10 mg/kg · d) or saline, whereas the sham rats received saline via sc mini-pumps. Loss of ovarian hormones reduced cardiac BH4 when compared with control hearts; this was associated with impaired myocardial relaxation, augmented filling pressures, increased collagen deposition, and thickened LV walls. Additionally, superoxide production increased and nitric oxide decreased in hearts from OVX compared with sham rats. Chronic BH4 supplementation after OVX improved diastolic function and attenuated LV remodeling while restoring myocardial nitric oxide release and preventing reactive oxygen species generation. These data indicate that BH4 supplementation protects against the adverse effects of ovarian hormonal loss on diastolic function and cardiac structure in mRen2.Lewis rats by restoring myocardial NO release and mitigating myocardial O2⁻ generation. Whether BH4 supplementation is a therapeutic option for the management of diastolic dysfunction in postmenopausal women will require direct testing in humans.


Subject(s)
Biopterins/analogs & derivatives , Diastole/drug effects , Heart Diseases/physiopathology , Heart/physiology , Postmenopause/metabolism , Superoxides/metabolism , Animals , Biopterins/administration & dosage , Disease Models, Animal , Female , Heart/drug effects , Heart Diseases/drug therapy , Heart Diseases/metabolism , Humans , Myocardium/metabolism , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Ovariectomy , Postmenopause/drug effects , Rats , Rats, Inbred Lew , Ventricular Remodeling/drug effects
9.
Menopause ; 18(6): 698-708, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21293310

ABSTRACT

OBJECTIVE: The loss of estrogen in mRen2.Lewis rats leads to an exacerbation of diastolic dysfunction. Because specific neuronal nitric oxide synthase (nNOS) inhibition reverses renal damage in the same model, we assessed the effects of inhibiting neuronal nitric oxide on diastolic function, left ventricular remodeling, and the components of the cardiac nitric oxide system in ovariectomized (OVX) and sham-operated mRen2.Lewis rats treated with N5-(1-imino-3-butenyl)-L-ornithine (L-VNIO; 0.5 mg/kg per day for 28 d) or vehicle (saline). METHODS: Female mRen2.Lewis rats underwent either bilateral oophorectomy (OVX; n = 15) or sham operation (or surgical procedure) (sham; n = 19) at 4 weeks of age. Beginning at 11 weeks of age, the rats were randomized to receive either L-VNIO or vehicle. RESULTS: The surgical loss of ovarian hormones, particularly estrogen, led to exacerbated hypertension, impaired myocardial relaxation, diminished diastolic compliance, increased perivascular fibrosis, and increased relative wall thickness. The cardiac tetrahydrobiopterin-to-dihydrobiopterin levels were lower among OVX rats compared with sham-operated rats, and this altered cardiac biopterin profile was associated with enhanced myocardial superoxide production and decreased nitric oxide release. L-VNIO decreased myocardial reactive oxygen species production, increased nitrite concentrations, attenuated cardiac remodeling, and improved diastolic function. CONCLUSIONS: Impaired relaxation, diastolic stiffness, and cardiac remodeling were found among OVX mRen2.Lewis rats. A possible mechanism for this unfavorable cardiac phenotype may have resulted from a deficiency in available tetrahydrobiopterin and subsequent increase in nNOS-derived superoxide and reduction in nitric oxide synthase metabolites within the heart. Selective nNOS inhibition with L-VNIO attenuated cardiac superoxide production and limited remodeling, leading to improved diastolic function in OVX mRen2.Lewis rats.


Subject(s)
Diastole/drug effects , Enzyme Inhibitors/pharmacology , Heart Failure, Diastolic/prevention & control , Nitric Oxide Synthase/biosynthesis , Nitric Oxide/metabolism , Ornithine/analogs & derivatives , Oxidative Stress/drug effects , Animals , Disease Models, Animal , Female , Heart Failure, Diastolic/metabolism , Ornithine/pharmacology , Ovariectomy , Rats , Rats, Inbred Lew
10.
PLoS One ; 5(11): e15433, 2010 Nov 03.
Article in English | MEDLINE | ID: mdl-21082029

ABSTRACT

INTRODUCTION: The G protein-coupled estrogen receptor (GPER) is expressed in various tissues including the heart. Since the mRen2.Lewis strain exhibits salt-dependent hypertension and early diastolic dysfunction, we assessed the effects of the GPER agonist (G-1, 40 nmol/kg/hr for 14 days) or vehicle (VEH, DMSO/EtOH) on cardiac function and structure. METHODS: Intact female mRen2.Lewis rats were fed a normal salt (0.5% sodium; NS) diet or a high salt (4% sodium; HS) diet for 10 weeks beginning at 5 weeks of age. RESULTS: Prolonged intake of HS in mRen2.Lewis females resulted in significantly increased blood pressure, mildly reduced systolic function, and left ventricular (LV) diastolic compliance (as signified by a reduced E deceleration time and E deceleration slope), increased relative wall thickness, myocyte size, and mid-myocardial interstitial and perivascular fibrosis. G-1 administration attenuated wall thickness and myocyte hypertrophy, with nominal effects on blood pressure, LV systolic function, LV compliance and cardiac fibrosis in the HS group. G-1 treatment significantly increased LV lusitropy [early mitral annular descent (e')] independent of prevailing salt, and improved the e'/a' ratio in HS versus NS rats (P<0.05) as determined by tissue Doppler. CONCLUSION: Activation of GPER improved myocardial relaxation in the hypertensive female mRen2.Lewis rat and reduced cardiac myocyte hypertrophy and wall thickness in those rats fed a high salt diet. Moreover, these advantageous effects of the GPER agonist on ventricular lusitropy and remodeling do not appear to be associated with overt changes in blood pressure.


Subject(s)
Blood Pressure/drug effects , Cyclopentanes/pharmacology , Hypertension/prevention & control , Quinolines/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , Cardiomegaly/etiology , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Diastole , Echocardiography , Female , Heart/drug effects , Heart/physiopathology , Hypertension/etiology , Hypertension/physiopathology , Immunohistochemistry , Mice , Myocardium/metabolism , Myocardium/pathology , Rats , Rats, Inbred Lew , Rats, Transgenic , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Renin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sodium Chloride, Dietary/administration & dosage
11.
Am J Physiol Regul Integr Comp Physiol ; 297(2): R446-52, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19515988

ABSTRACT

The effects of chronic mild hypoxemia on the binding of angiotensin receptors in selected brain stem nuclei and reflex responses were studied in fetal sheep. Fetal and maternal catheters were placed at 120 days' gestation, and animals received intratracheal maternal administration of nitrogen (n = 16) or compressed air in controls (n = 19). Nitrogen infusion was adjusted to reduce fetal brachial artery PO(2) by 25% during 5 days. Spontaneous baroreflex sensitivity and spectral analysis of the pulse interval were analyzed during the 5 days hypoxemia period using 90 min of daily recording. Brains of control and hypoxemic animals were collected, and brain stem angiotensin receptor binding was studied by in vitro autoradiography at 130 days of gestation. After 5 days of hypoxemia, some animals in each group were submitted to one complete umbilical cord occlusion during 5 min. [(125)I]sarthran binding showed that chronic mild hypoxemia significantly increases angiotensin type 1 receptor, angiotensin type 2 receptor, and ANG-(1-7) angiotensin receptor binding sites in the nucleus tractus solitarius and dorsal motor nucleus of the vagus (P < 0.05). Hypoxemia induced lower baroreflex sensitivity and a higher low frequency-to-high frequency ratio in the fetus, consistent with a shift from vagal to sympathetic autonomic cardiac regulation. Cord occlusion to elicit a chemoreflex response induced a greater bradycardic response in hypoxemic fetuses (slope of the initial fall in heart rate; 11.3 +/- 1.9 vs. 6.4 +/- 1.2 beats x min(-1) x s(-1), P < 0.05). In summary, chronic mild hypoxemia increased binding of angiotensin receptors in brain stem nuclei, decreased spontaneous baroreflex gain, and increased chemoreflex responses to asphyxia in the fetus. These results suggest hypoxemia-induced alterations in brain stem mechanisms for cardiovascular control.


Subject(s)
Baroreflex/physiology , Brain Stem/physiopathology , Fetus/physiopathology , Gene Expression/physiology , Hypoxia/physiopathology , Receptors, Angiotensin/metabolism , Angiotensin II/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 2 Receptor Blockers , Animals , Area Postrema/metabolism , Asphyxia/physiopathology , Blood Gas Analysis , Blood Pressure/physiology , Brain Stem/metabolism , Female , Fetal Blood/chemistry , Fetal Blood/metabolism , Fetus/metabolism , Heart Rate/physiology , Hemoglobins/metabolism , Hydrogen-Ion Concentration , Hypoxia/blood , Hypoxia/metabolism , Male , Medulla Oblongata/metabolism , Olivary Nucleus/metabolism , Pregnancy , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Sheep , Solitary Nucleus/metabolism
12.
Ther Adv Cardiovasc Dis ; 3(4): 245-57, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19531557

ABSTRACT

BACKGROUND: Hypertension and left ventricular (LV) hypertrophy often precede diastolic dysfunction and are risk factors for diastolic heart failure. Although pharmacologic inhibition of the renin-angiotensin system (RAS) improves diastolic function and functional capacity in hypertensive patients with LV hypertrophy, the effects of combination therapy with an angiotensin converting enzyme inhibitor (ACEi) and an angiotensin receptor blocker (ARB) are unclear. METHOD: We assessed the effects of the combined 10-week administration of lisinopril (10 mg/kg/ day, p.o.) and losartan (10 mg/kg/day, p.o.) (LIS/LOS) on diastolic function and LV structure in seven young (5 weeks), prehypertensive congenic mRen2.Lewis male rat, a model of tissue renin overexpression and angiotensin II (Ang II)-dependent hypertension compared to vehicle (VEH) treated (n = 7), age-matched rats. RESULTS: Systolic blood pressures were 64% lower with the combination therapy (p < 0.001), but there were no differences in heart rate or systolic function between groups. RAS inhibition increased myocardial relaxation, defined by tissue Doppler mitral annular descent (e') by 2.2 fold (p < 0.001). The preserved lusitropy in the LIS/LOS-treated rats was accompanied by a reduction in phospholamban-to-SERCA2 ratio (p < 0.001). Despite lower relative wall thicknesses (VEH: 1.56+/-0.17 versus LIS/LOS: 0.78+/-0.05) and filling pressures, defined by the transmitral Doppler-to-mitral annular descent ratio (E/e', VEH: 28.7+/-1.9 versus LIS/LOS: 17.96+/-1.5), no differences in cardiac collagen were observed. CONCLUSION: We conclude that the lusitropic benefit of early dual RAS blockade may be due to improved vascular hemodynamics and/or cardiac calcium handling rather than effects on extracellular matrix reduction.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Hypertension/drug therapy , Hypertrophy, Left Ventricular/drug therapy , Lisinopril/pharmacology , Losartan/pharmacology , Myocardium/pathology , Renin/genetics , Ventricular Function, Left/drug effects , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Animals , Animals, Congenic , Blood Pressure/drug effects , Calcium-Binding Proteins/metabolism , Collagen/metabolism , Disease Models, Animal , Drug Administration Schedule , Drug Therapy, Combination , Echocardiography, Doppler , Fibrosis , Heart Rate/drug effects , Hypertension/genetics , Hypertension/pathology , Hypertension/physiopathology , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Lisinopril/administration & dosage , Losartan/administration & dosage , Male , Myocardial Contraction/drug effects , Myocardium/metabolism , Rats , Rats, Inbred Lew , Rats, Transgenic , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
13.
Am J Physiol Heart Circ Physiol ; 294(6): H2614-8, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18408132

ABSTRACT

A low expression of angiotensinogen in the heart has been construed as indicating a circulating uptake mechanism to explain the local effects of angiotensin II on tissues. The recent identification of angiotensin-(1-12) in an array of rat organs suggests this propeptide may be an alternate substrate for local angiotensin production. To test this hypothesis, tissues from 11-wk-old spontaneously hypertensive rats (SHR) and Wistar-Kyoto (WKY) rats (n = 14) were stained with purified antibodies directed to the COOH terminus of angiotensin-(1-12). Robust angiotensin-(1-12) staining was predominantly found in ventricular myocytes with less staining found in the medial layer of intracoronary arteries and vascular endothelium. In addition, angiotensin-(1-12) immunoreactivity was present in the proximal, distal, and collecting renal tubules within the deep cortical and outer medullary zones in both strains. Preadsorption of the antibody with angiotensin-(1-12) abolished staining in both tissues. Corresponding tissue measurements by radioimmunoassay showed 47% higher levels of angiotensin-(1-12) in the heart of SHR compared with WKY rats (P < 0.05). In contrast, renal angiotensin-(1-12) levels were 16.5% lower in SHR compared with the WKY rats (P < 0.05). This study shows for first time the localization of angiotensin-(1-12) in both cardiac myocytes and renal tubular components of WKY and SHR. In addition, we show that increased cardiac angiotensin-(1-12) concentrations in SHR is associated with a small, but statistically significant, reduction in renal angiotensin-(1-12) levels.


Subject(s)
Angiotensinogen/analysis , Hypertension/metabolism , Kidney Tubules/chemistry , Myocardium/chemistry , Peptide Fragments/analysis , Angiotensinogen/immunology , Animals , Antibody Specificity , Disease Models, Animal , Immunohistochemistry/methods , Male , Myocytes, Cardiac/chemistry , Peptide Fragments/immunology , Radioimmunoassay , Rats , Rats, Inbred SHR , Rats, Inbred WKY
14.
J Mol Med (Berl) ; 86(6): 663-71, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18437333

ABSTRACT

Accumulation of a large body of evidence during the past two decades testifies to the complexity of the renin-angiotensin system (RAS). The incorporation of novel enzymatic pathways, resulting peptides, and their corresponding receptors into the biochemical cascade of the RAS provides a better understanding of its role in the regulation of cardiovascular and renal function. Hence, in recent years, it became apparent that the balance between the two opposing effector peptides, angiotensin II and angiotensin-(1-7), may have a pivotal role in determining different cardiovascular pathophysiologies. Furthermore, our recent studies provide evidence for the functional relevance of a newly discovered rat peptide, containing two additional amino acid residues compared to angiotensin I, first defined as proangiotensin-12 [angiotensin-(1-12)]. This review focuses on angiotensin-(1-7) and its important contribution to cardiovascular function and growth, while introducing angiotensin-(1-12) as a potential novel angiotensin precursor.


Subject(s)
Angiotensins/metabolism , Animals , Humans , Renin-Angiotensin System
15.
Am J Physiol Heart Circ Physiol ; 294(5): H2242-7, 2008 May.
Article in English | MEDLINE | ID: mdl-18359898

ABSTRACT

Identification of angiotensin-(1-12) as an intermediate precursor derived directly from angiotensinogen led us to explore whether the heart has the capacity to process angiotensin-(1-12) into biologically active angiotensin peptides. The generation of angiotensin I, angiotensin II, and angiotensin-(1-7) from exogenous angiotensin-(1-12) was evaluated in the effluent of isolated perfused hearts mounted on a Langendorff apparatus in three normotensive and two hypertensive strains: Sprague-Dawley, Lewis, congenic mRen2.Lewis, Wistar-Kyoto, and spontaneously hypertensive rats. Hearts were perfused with Krebs solution for 60 min before and after the addition of angiotensin-(1-12) (10 nmol/l). Angiotensin-(1-12) caused the rapid appearance of both angiotensin I and angiotensin II in the perfusate that peaked between 30 and 60 min of recirculation. Production of angiotensin-(1-7) from exogenous angiotensin-(1-12) rose steadily over the course of the 60-min experiment. These data directly demonstrate that angiotensin-(1-12) is a substrate for the formation of angiotensin peptides in cardiac tissue. This finding further suggests that this angiotensinogen-derived product is a previously unrecognized important precursor peptide to the renin-angiotensin system cascade.


Subject(s)
Angiotensinogen/metabolism , Angiotensins/metabolism , Hypertension/metabolism , Myocardium/metabolism , Peptide Fragments/metabolism , Renin-Angiotensin System , Angiotensin I/metabolism , Angiotensin II/metabolism , Animals , Animals, Genetically Modified , Disease Models, Animal , Male , Perfusion , Rats , Rats, Inbred Lew , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Sprague-Dawley , Renin/genetics , Renin/metabolism , Time Factors
16.
J Am Soc Hypertens ; 2(2): 106-15, 2008.
Article in English | MEDLINE | ID: mdl-19343087

ABSTRACT

Fifty years since their introduction, thiazide diuretics are established as first-line therapy in the treatment of hypertension. Because the dosing profile for thiazide agents lessened, the mechanism responsible for the blood pressure lowering effect may lie outside their diuretic properties. We evaluated the mechanism driving blood pressure reductions in spontaneously hypertensive rats (SHR) and normotensive WKY by examining the effects of low-dose hydrochlorothiazide (HCTZ) administration on renin-angiotensin system (RAS) components. The 7-day, 1.5 mg/kg/day HCTZ did not change systolic pressure in WKY, but decreased SBP by 41 +/- 2 mm Hg (p < 0.0001) in SHR. This reduction was independent of increases in water intake, urine output, or alterations in electrolyte excretion. HCTZ significantly increased the plasma concentrations of angiotensin I (Ang I) and angiotensin II (Ang II) in both WKY and SHR while reducing angiotensin converting enzyme (ACE) activity and the Ang II/Ang I ratio (17.1 +/- 2.9 before versus 10.3 +/- 2.9 after, p < 0.05) only in SHR. HCTZ increased cardiac ACE2 mRNA and activity, and neprilysin mRNA in WKY, but not SHR. Conversely in SHR, ACE2 activity was decreased and aside from a 75% increase in AT(1) mRNA in the HCTZ-treated SHR, the expression of the other variables remained unaltered. Measures of cardiac mas receptor mRNA showed no changes in response to treatment in both strains, although cardiac mas mRNA was significantly lower in untreated SHR. These data, which document for the first time the effect of low-dose thiazide on the activity of the ACE2/Ang-(1-7)/mas-receptor axis, suggest that the opposing arm of the system does not substantially contribute to the antihypertensive effect of low-dose thiazides in SHR.

17.
Article in English | MEDLINE | ID: mdl-17703433

ABSTRACT

INTRODUCTION: The presence of Angiotensin (1-7) (Ang 1-7) and ACE 2 in the ventricle of cardiomyopathic hamsters as well as the influence of Ang (1-7) on membrane potential, impulse propagation and cardiac excitability were investigated. METHODS: Histology and immunochemistry were used to demonstrate the presence of Ang (1-7) and ACE 2 in the ventricle of cardiomyopathic hamsters. Measurements of transmembrane potentials, conduction velocity and refractoriness were made using conventional intracellular microelectrodes. The influence of Ang (1-7) on sodium pump current was investigated in voltageclamped myocytes isolated from the ventricle. RESULTS: The results indicated the presence of Ang (1-7) and ACE 2 in myocytes of cardiomyopathic hamsters. Moreover,Ang (1-7) (10(-8) M) hyperpolarised the heart cell, increased the conduction velocity, and reduced transiently the action potential duration. The cardiac refractoriness was also increased by the heptapeptide, an effect in part reduced by an inhibitor of mas receptor. These findings indicate that Ang (1-7) has important antiarrhythmic properties. However, the beneficial effects of Ang (1-7) are dose-dependent because at higher concentration (10(-7) M) the heptapeptide elicited an appreciable increase of action potential duration and early-after depolarisations. Since losartan (10(-7) M) did not counteract this effect of the high dose of the heptapeptide, it is possible to conclude that activation of AT(1)-receptors is not involved in this effect of Ang (1-7). To investigate the mechanism of the hyperpolarising action of Ang (1-7) the influence of the heptapeptide on the sodium potassium pump current was studied in myocytes isolated from the ventricle of cardiomyopathic hamsters. The peak pump current density was measured under voltage clamp using the whole cell configuration. The results indicated that Ang (1-7) (10(-8) M) enhanced the electrogenic sodium pump, an effect suppressed by ouabain (10(-7) M). CONCLUSIONS: Ang (1-7) has beneficial effects on the failing heart by activating the sodium pump, hyperpolarising the cell membrane and increasing the conduction velocity. These effects as well as the increment of refractoriness indicate that Ang (1-7) has antiarrhythmic properties. At higher concentrations (10(-7) M), however, the heptapeptide induced early-after depolarisations which leads to the conclusion that an optimal generation of Ang (1-7) must be achieved to permit a protective role of Ang (1-7) on cardiac arrhythmias.


Subject(s)
Angiotensin I/therapeutic use , Cardiomyopathies/physiopathology , Heart Rate/drug effects , Heart/physiopathology , Membrane Potentials/physiology , Peptide Fragments/therapeutic use , Animals , Antihypertensive Agents/therapeutic use , Cardiomyopathies/drug therapy , Cardiomyopathies/pathology , Cricetinae , Disease Models, Animal , Heart/drug effects , Immunohistochemistry , Membrane Potentials/drug effects , Myocardium/pathology , Sodium Channels/drug effects
18.
Am J Physiol Heart Circ Physiol ; 291(5): H2166-72, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16766648

ABSTRACT

The generation of the Lew.Tg(mRen2) congenic hypertensive rat strain, developed through a backcross of the hypertensive (mRen2)27 transgenic rat with normotensive Lewis rats, provides a new model by which primary hypertension can be studied without the genetic variability found in the original strain. The purpose of this study was to characterize the Lew.Tg(mRen2) rats by dually investigating the effects of type 1 angiotensin II (ANG II) receptor (AT(1)) blockade and angiotensin-converting enzyme (ACE) activity inhibition on the ANG-(1-7)/ACE2 axis of the renin-angiotensin system in this new hypertensive model. The control of blood pressure elicited by 12-day administration of either lisinopril (mean difference change = 92 +/- 2, P < 0.05) or losartan (mean difference change = 69 +/- 2, P < 0.05) was associated with 54% and 33% increases in cardiac ACE2 mRNA and 54% and 43% increases in cardiac ACE mRNA, respectively. Lisinopril induced a 3.1-fold (P < 0.05) increase in renal cortical expression of ACE2, whereas losartan increased ACE2 mRNA 3.5-fold (P < 0.05). Both treatment regimens increased renal ACE mRNA 2.6-fold (P < 0.05). The two therapies augmented ACE2 protein activity, as well as increased cardiac and renal AT(1) receptor mRNAs. ACE inhibition reduced plasma ANG II levels (81%, P < 0.05) and increased plasma ANG-(1-7) (265%, P < 0.05), whereas losartan had no effect on the peptides. In contrast with what had been shown in normotensive rats, ACE inhibition decreased renal ANG II excretion and transiently decreased ANG-(1-7) excretion, whereas losartan treatment was associated with a consistent decrease in ANG-(1-7) urinary excretion rates. In response to the treatments, the expression of both renal cortical renin and angiotensinogen mRNAs was significantly augmented. The paradoxical effects of blockade of ANG II synthesis and activity on urinary excretion rates of the peptides and plasma angiotensins levels suggest that, in Lew.Tg(mRen2) congenic rats, a failure of compensatory ACE2 and ANG-(1-7)-dependent vasodepressor mechanisms may contribute both to the development and progression of hypertension driven by increased formation of endogenous ANG II.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin Receptor Antagonists , Hypertension, Renal/genetics , Models, Genetic , Peptidyl-Dipeptidase A/drug effects , Renin/genetics , Angiotensin I/blood , Angiotensin I/urine , Angiotensin II/antagonists & inhibitors , Angiotensin II/blood , Angiotensin II/metabolism , Angiotensin-Converting Enzyme 2 , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Animals, Congenic , Animals, Genetically Modified , Crosses, Genetic , Hypertension, Renal/metabolism , Kinetics , Lisinopril/pharmacology , Losartan/pharmacology , Male , Peptide Fragments/blood , Peptide Fragments/urine , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/physiology , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Time Factors
19.
Kidney Int ; 68(5): 2189-96, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16221218

ABSTRACT

BACKGROUND: Angiotensin-converting enzyme (ACE)2, a homologue of ACE, which is insensitive to ACE inhibitors and forms angiotensin-(1-7) [Ang-(1-7)] from angiotensin II (Ang II) with high efficiency was investigated in response to chronic blockade with lisinopril, losartan, and both drugs combined. METHODS: Thirty-six adult Lewis rats were assigned to receive these medications in their drinking water for 2 weeks while their arterial pressure, water intake, and urine volume were recorded throughout the study. Measures of renal excretory variables included assessing excretion rates of angiotensin I (Ang I), Ang II and Ang-(1-7) while blood collected at the completion of the study was used for measures of plasma angiotensin concentrations. Samples from renal cortex were assayed for renin, angiotensinogen (Aogen), neprilysin, angiotensin types 1 and 2 (AT(1) and AT(2)) and mas receptor mRNAs by semiquantitative reverse transcriptase (RT) real-time polymerase chain reaction (PCR). ACE2 activity was determined as the rate of Ang II conversion into Ang-(1-7). RESULTS: Comparable blood pressure reductions were obtained in rats medicated with either lisinopril or losartan, whereas both drugs produced a greater decrease in arterial pressure. Polyuria was recorded in all three forms of treatment associated with reduced osmolality but no changes in creatinine excretion. Lisinopril augmented plasma levels and urinary excretion rates of Ang I and Ang-(1-7), while plasma Ang II was reduced with no effect on urinary Ang II. Losartan produced similar changes in plasma and urinary Ang-(1-7) but increased plasma Ang II without changing urinary Ang II excretion. Combination therapy mimicked the effects obtained with lisinopril on plasma and urinary Ang I and Ang-(1-7) levels. Renal cortex Aogen mRNA increased in rats medicated with either lisinopril or the combination, whereas all three treatments produced a robust increase in renal renin mRNA. In contrast, ACE, ACE2, neprilysin, AT(1), and mas receptor mRNAs remained unchanged with all three treatments. Renal cortex ACE2 activity was significantly augmented in rats medicated with lisinopril or losartan but not changed in those given the combination. CONCLUSION: Our data revealed a role for ACE2 in Ang-(1-7) formation from Ang II in the kidney of normotensive rats as primarily reflected by the increased ACE2 activity measured in renal membranes from the kidney of rats given either lisinopril or losartan. The data further indicate that increased levels of Ang-(1-7) in the urine of animals after ACE inhibition or AT(1) receptor blockade reflect an intrarenal formation of the heptapeptide.


Subject(s)
Angiotensin I/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Hypertension, Renal/drug therapy , Lisinopril/pharmacology , Peptide Fragments/metabolism , Renin-Angiotensin System/drug effects , Angiotensin I/blood , Angiotensin II/blood , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme 2 , Animals , Carboxypeptidases/genetics , Carboxypeptidases/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Hypertension, Renal/metabolism , Kidney/metabolism , Losartan/pharmacology , Male , Peptidyl-Dipeptidase A , Rats , Rats, Inbred Lew , Receptors, Angiotensin/metabolism , Renin-Angiotensin System/physiology
20.
Am J Physiol Heart Circ Physiol ; 289(6): H2281-90, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16055515

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2) is the first human homologue of ACE to be described. ACE2 is a type I integral membrane protein that functions as a carboxypeptidase, cleaving a single hydrophobic/basic residue from the COOH-terminus of its substrates. Because ACE2 efficiently hydrolyzes the potent vasoconstrictor angiotensin II to angiotensin (1-7), this has changed our overall perspective about the classical view of the renin angiotensin system in the regulation of hypertension and heart and renal function, because it represents the first example of a feedforward mechanism directed toward mitigation of the actions of angiotensin II. This paper reviews the new data regarding the biochemistry of angiotensin-(1-7)-forming enzymes and discusses key findings such as the elucidation of the regulatory mechanisms participating in the expression of ACE2 and angiotensin-(1-7) in the control of the circulation.


Subject(s)
Angiotensin I/metabolism , Biochemistry/trends , Blood Pressure , Carboxypeptidases/metabolism , Cardiovascular System/physiopathology , Hypertension/physiopathology , Peptide Fragments/metabolism , Renin-Angiotensin System , Angiotensin-Converting Enzyme 2 , Animals , Feedback , Humans , Myocardial Infarction/physiopathology , Peptidyl-Dipeptidase A
SELECTION OF CITATIONS
SEARCH DETAIL
...