Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 253
Filter
1.
J Eur Acad Dermatol Venereol ; 34(3): 558-564, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31746507

ABSTRACT

BACKGROUND: Systemic sclerosis (SSc) is a multisystemic disease with an extensive microvasculopathy. Previously, disturbances in plasma levels of angiotensin II (Ang II) and its antagonistic angiotensin-(1-7) (Ang-(1-7)) were found in patients with SSc. Their significance in a pathogenesis of SSc stays unclear due to discrepancies of earlier studies. OBJECTIVES: To evaluate a significance of disturbances in production pathway of angiotensins in a development of SSc. METHODS: There were enrolled 27 patients with established SSc, 23 subjects with very early SSc and 23 healthy controls. The diagnosis of SSc was established in patients who met EULAR/ACR 2013 classification criteria. Very early SSc described patients with Raynaud's phenomenon having SSc-specific antinuclear antibodies and SSc-like abnormalities in nailfold videocapillaroscopy. Patients were submitted to evaluation of internal organ involvement and blood sampling to assay plasma levels of angiotensin I, angiotensin II and angiotensin-(1-7) with ELISA technique. RESULTS: Plasma level of angiotensin-(1-7) was significantly reduced in both SSc group (median = 47.2 pg/mL; P < 0.001) and ones with very early SSc (median = 102.7 pg/mL; P = 0.002) when compared to healthy controls (median = 176.1 pg/mL). A tendency to higher than in control group (median = 214 pg/mL) plasma level of angiotensin I was seen in SSc group (median = 392 pg/mL; P = 0.059). Differences in plasma level of angiotensin II were insignificant between all study groups. Those disturbances produced unfavourable angiotensin-(1-7)/angiotensin II (%) ratio in both groups of patients, which achieved statistical significance in subjects with established SSc (P < 0.001). Production pathway of angiotensins showed a dependence on a subtype of SSc, immune profile and a presence of interstitial lung disease. CONCLUSIONS: Production of angiotensin-(1-7) was significantly reduced in both SSc patients and those ones with very early SSc, although a significant imbalance between angiotensin II and angiotensin-(1-7) occurred only in subjects with established disease.


Subject(s)
Angiotensin II/physiology , Angiotensin I/physiology , Peptide Fragments/physiology , Scleroderma, Systemic/etiology , Adult , Aged , Angiotensin I/blood , Angiotensin II/blood , Female , Humans , Male , Middle Aged , Peptide Fragments/blood , Scleroderma, Systemic/blood , Young Adult
2.
Clin Sci (Lond) ; 132(10): 1021-1038, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29802208

ABSTRACT

All the components of the classic renin-angiotensin system (RAS) have been identified in the brain. Today, the RAS is considered to be composed mainly of two axes: the pressor axis, represented by angiotensin (Ang) II/angiotensin-converting enzyme/AT1 receptors, and the depressor and protective one, represented by Ang-(1-7)/ angiotensin-converting enzyme 2/Mas receptors. Although the RAS exerts a pivotal role on electrolyte homeostasis and blood pressure regulation, their components are also implicated in higher brain functions, including cognition, memory, anxiety and depression, and several neurological disorders. Overactivity of the pressor axis of the RAS has been implicated in stroke and several brain disorders, such as cognitive impairment, dementia, and Alzheimer or Parkinson's disease. The present review is focused on the role of the protective axis of the RAS in brain disorders beyond its effects on blood pressure regulation. Furthermore, the use of drugs targeting centrally RAS and its beneficial effects on brain disorders are also discussed.


Subject(s)
Brain Diseases/physiopathology , Renin-Angiotensin System/physiology , Alzheimer Disease/physiopathology , Angiotensin I/physiology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Anxiety/physiopathology , Brain Diseases/prevention & control , Cognition/physiology , Humans , Peptide Fragments/physiology , Renin-Angiotensin System/drug effects , Stroke/physiopathology , Translational Research, Biomedical/methods
3.
Clin Sci (Lond) ; 132(11): 1117-1133, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29685981

ABSTRACT

Patients with hyperthyroidism exhibit increased risk of development and progression of cardiac diseases. The activation of the renin-angiotensin system (RAS) has been indirectly implicated in these cardiac effects observed in hyperthyroidism. Angiotensin-(1-7) (Ang-(1-7)) has previously been shown to counterbalance pathological effects of angiotensin II (Ang II). The aim of the present study was to investigate the effects of elevated circulating Ang-(1-7) levels on cardiac effects promoted by hyperthyroidism in a transgenic rat (TG) model that constitutively overexpresses an Ang-(1-7)-producing fusion protein [TGR(A1-7)3292]. TG and wild-type (WT) rats received daily injections (i.p.) of triiodothyronine (T3; 7 µg/100 g of body weight (BW)) or vehicle for 14 days. In contrast with WT rats, the TG rats did not develop cardiac hypertrophy after T3 treatment. Indeed, TG rats displayed reduced systolic blood pressure (SBP) and cardiac hyperdynamic condition induced by hyperthyroidism. Moreover, increased plasma levels of Ang II observed in hyperthyroid WT rats were prevented in TG rats. TG rats were protected from glycogen synthase kinase 3ß (GSK3ß) inactivation and nuclear factor of activated T cells (NFAT) nuclear accumulation induced by T3. In vitro studies evidenced that Ang-(1-7) prevented cardiomyocyte hypertrophy and GSK3ß inactivation induced by T3. Taken together, these data reveal an important cardioprotective action of Ang-(1-7) in experimental model of hyperthyroidism.


Subject(s)
Angiotensin I/physiology , Cardiomegaly/etiology , Glycogen Synthase Kinase 3 beta/physiology , Hyperthyroidism/complications , NFATC Transcription Factors/physiology , Peptide Fragments/physiology , Angiotensin I/blood , Angiotensin I/pharmacology , Animals , Cardiomegaly/blood , Cardiomegaly/diagnostic imaging , Cells, Cultured , Echocardiography , Hyperthyroidism/blood , Hyperthyroidism/chemically induced , Male , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Peptide Fragments/blood , Peptide Fragments/pharmacology , Rats, Sprague-Dawley , Rats, Transgenic , Rats, Wistar , Renin-Angiotensin System/physiology , Signal Transduction/physiology , Triiodothyronine/pharmacology
4.
Circ Res ; 118(8): 1313-26, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-27081112

ABSTRACT

Heart failure (HF) remains the most common cause of death and disability, and a major economic burden, in industrialized nations. Physiological, pharmacological, and clinical studies have demonstrated that activation of the renin-angiotensin system is a key mediator of HF progression. Angiotensin-converting enzyme 2 (ACE2), a homolog of ACE, is a monocarboxypeptidase that converts angiotensin II into angiotensin 1-7 (Ang 1-7) which, by virtue of its actions on the Mas receptor, opposes the molecular and cellular effects of angiotensin II. ACE2 is widely expressed in cardiomyocytes, cardiofibroblasts, and coronary endothelial cells. Recent preclinical translational studies confirmed a critical counter-regulatory role of ACE2/Ang 1-7 axis on the activated renin-angiotensin system that results in HF with preserved ejection fraction. Although loss of ACE2 enhances susceptibility to HF, increasing ACE2 level prevents and reverses the HF phenotype. ACE2 and Ang 1-7 have emerged as a key protective pathway against HF with reduced and preserved ejection fraction. Recombinant human ACE2 has been tested in phase I and II clinical trials without adverse effects while lowering and increasing plasma angiotensin II and Ang 1-7 levels, respectively. This review discusses the transcriptional and post-transcriptional regulation of ACE2 and the role of the ACE2/Ang 1-7 axis in cardiac physiology and in the pathophysiology of HF. The pharmacological and therapeutic potential of enhancing ACE2/Ang 1-7 action as a novel therapy for HF is highlighted.


Subject(s)
Angiotensin I/physiology , Heart Failure/metabolism , Peptide Fragments/physiology , Peptidyl-Dipeptidase A/physiology , Renin-Angiotensin System/physiology , Angiotensin-Converting Enzyme 2 , Animals , Heart Failure/diagnosis , Heart Failure/physiopathology , Humans , Signal Transduction/physiology
5.
J Am Soc Nephrol ; 28(5): 1350-1361, 2017 May.
Article in English | MEDLINE | ID: mdl-28151411

ABSTRACT

Inappropriate activation of the renin-angiotensin system (RAS) exacerbates renal and vascular injury. Accordingly, treatment with global RAS antagonists attenuates cardiovascular risk and slows the progression of proteinuric kidney disease. By reducing BP, RAS inhibitors limit secondary immune activation responding to hemodynamic injury in the target organ. However, RAS activation in hematopoietic cells has immunologic effects that diverge from those of RAS stimulation in the kidney and vasculature. In preclinical studies, activating type 1 angiotensin (AT1) receptors in T lymphocytes and myeloid cells blunts the polarization of these cells toward proinflammatory phenotypes, protecting the kidney from hypertensive injury and fibrosis. These endogenous functions of immune AT1 receptors temper the pathogenic actions of renal and vascular AT1 receptors during hypertension. By counteracting the effects of AT1 receptor stimulation in the target organ, exogenous administration of AT2 receptor agonists or angiotensin 1-7 analogs may similarly limit inflammatory injury to the heart and kidney. Moreover, although angiotensin II is the classic effector molecule of the RAS, several RAS enzymes affect immune homeostasis independently of canonic angiotensin II generation. Thus, as reviewed here, multiple components of the RAS signaling cascade influence inflammatory cell phenotype and function with unpredictable and context-specific effects on innate and adaptive immunity.


Subject(s)
Renin-Angiotensin System/immunology , Angiotensin I/physiology , Angiotensin-Converting Enzyme 2 , Animals , Humans , Peptide Fragments/physiology , Peptidyl-Dipeptidase A/physiology , Receptor, Angiotensin, Type 1/physiology , Receptor, Angiotensin, Type 2/physiology
6.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 43(7): 738-746, 2018 Jul 28.
Article in Zh | MEDLINE | ID: mdl-30124209

ABSTRACT

OBJECTIVE: To investigate changes in the angiotensin converting enzyme 2 (ACE2) and angiotensin (1-7) [Ang (1-7)] and to explore the role of ACE2-Ang (1-7)-Mas receptor axis in hypertension with heart failure with preserved ejection fraction (HFPEF).
 Methods: A total of 70 patients with primary hypertension and preserved left ventricular ejection fraction (LVEF>50%) were recruited and patients were divided into a hypertension group (HBP) and a heart failure with preserved ejection fraction group (HFpEF) according to the diagnostic criteria of HFpEF. Thirty-five healthy participants were selected randomly as a control group. Enzyme linked immunosorbent assays (ELISA) method was used to detect concentration of Ang (1-7), ACE2, angiotensin II (Ang II), brain natriuretic peptide (BNP) in plasma. Male Sprague- Dawley (SD) rats was randomly divided into 2 groups: An HFpEF group (n=16) and a sham group (n=8). Rats (n=8) in the AAC group were given Ang (1-7) [0.5 mg/(kg.d), intraperitoneally] for 6 weeks, and the rest were given equal dose normal saline. Then all the rats were killed, and the hearts were taken out for hematoxylineosin (HE) staining. The protein expressions of angiotensin converting enzyme (ACE), ACE2, and Mas receptor were detected by Western blot.
 Results: The BNP and Ang II were significantly increased in the HBP group and the HFpEF group compared with the control group (P<0.01). There were not significantly different in levels of ACE2 and Ang (1-7) between the HBP group and control group (P>0.05), whereas those levels were significantly increased in the HFpEF group compared with the HBP group and control group (P<0.01). HE staining showed obvious hypertrophy of myocardial cell in the AAC group compared with the sham group. Hypertrophy of myocardial cell in the AAC+Ang (1-7) group was significantly higher than that in the AAC group. Expressions of ACE, ACE2, and Mas receptor proteins were significantly higher in the AAC group than those in the sham group (P<0.05), while the expressions of ACE2 and Mas receptor proteins in the AAC+Ang (1-7) group were significantly higher than those in the AAC group (P<0.05). There was no significant difference in the ACE protein expression between groups (P>0.05).
 Conclusion: ACE2 and Ang (1-7) are important predictive factors for the severity of heart failure and myocardial remodeling of HFpEF with hypertension; ACE2-Ang (1-7)-Mas receptor axis may play a protective role in preventing myocardial remodeling in HFpEF with hypertension.


Subject(s)
Angiotensin I/physiology , Heart Failure/metabolism , Hypertension/metabolism , Peptide Fragments/physiology , Peptidyl-Dipeptidase A/physiology , Angiotensin II , Angiotensin-Converting Enzyme 2 , Animals , Atrial Remodeling/physiology , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Heart Failure/physiopathology , Humans , Hypertension/physiopathology , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/physiology , Stroke Volume , Ventricular Function, Left/physiology , Ventricular Remodeling/physiology
8.
Neurobiol Learn Mem ; 127: 27-33, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26642920

ABSTRACT

Inappropriate defense-alerting reaction to fear is a common feature of neuropsychiatric diseases. Therefore, impairments in brain circuits, as well as in molecular pathways underlying the neurovegetative adjustments to fear may play an essential role on developing neuropsychiatric disorders. Here we tested the hypothesis that interfering with angiotensin-(1-7) [Ang-(1-7)]/Mas receptor axis homeostasis, which appears to be essential to arterial pressure control, would affect fear memory and extinction. Mas knockout (MasKO) mice, in FVB/N background, showed normal cued fear memory and extinction, but increased freezing in response to context. Next, as FVB/N has poor performance in contextual fear memory, we tested MasKO in mixed 129xC57BL/6 background. MasKO mice behaved similarly to wild-type (WT), but memory extinction was slower in contextual fear conditioning to a weak protocol (1CS/US). In addition, delayed extinction in MasKO mice was even more pronounced after a stronger protocol (3CS/US). We showed previously that Angiotensin II receptor AT1 antagonist, losantan, rescued object recognition memory deficit in MasKO mice. Here, losartan was also effective. Memory extinction was accelerated in MasKO mice after treatment with losartan. In conclusion, we showed for the first time that Ang-(1-7)/Mas axis may modulate fear memory extinction. Furthermore, we suggest MasKO mice as an animal model to study post-traumatic stress disorder (PTSD).


Subject(s)
Angiotensin I/physiology , Conditioning, Classical/physiology , Extinction, Psychological/physiology , Fear/physiology , Peptide Fragments/physiology , Proto-Oncogene Proteins/physiology , Receptor, Angiotensin, Type 1/physiology , Receptors, G-Protein-Coupled/physiology , Acoustic Stimulation , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Electroshock , Losartan/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Receptors, G-Protein-Coupled/genetics
10.
Circ Res ; 112(8): 1104-11, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23446738

ABSTRACT

RATIONALE: The renin-angiotensin system (RAS) is a key regulator of the cardiovascular system, electrolyte, and water balance. Here, we report identification and characterization of alamandine, a new heptapeptide generated by catalytic action of angiotensin-converting enzyme-2 angiotensin A or directly from angiotensin-(1-7). OBJECTIVE: To characterize a novel component of the RAS, alamandine. METHODS AND RESULTS: Using mass spectrometry we observed that alamandine circulates in human blood and can be formed from angiotensin-(1-7) in the heart. Alamandine produces several physiological actions that resemble those produced by angiotensin-(1-7), including vasodilation, antifibrosis, antihypertensive, and central effects. Interestingly, our data reveal that its actions are independent of the known vasodilator receptors of the RAS, Mas, and angiotensin II type 2 receptor. Rather, we demonstrate that alamandine acts through the Mas-related G-protein-coupled receptor, member D. Binding of alamandine to Mas-related G-protein-coupled receptor, member D is blocked by D-Pro(7)-angiotensin-(1-7), the Mas-related G-protein-coupled receptor, member D ligand ß-alanine and PD123319, but not by the Mas antagonist A-779. In addition, oral administration of an inclusion compound of alamandine/ß-hydroxypropyl cyclodextrin produced a long-term antihypertensive effect in spontaneously hypertensive rats and antifibrotic effects in isoproterenol-treated rats. Alamandine had no noticeable proliferative or antiproliferative effect in human tumoral cell lines. CONCLUSIONS: The identification of these 2 novel components of the RAS, alamandine and its receptor, provides new insights for the understanding of the physiological and pathophysiological role of the RAS and may help to develop new therapeutic strategies for treating human cardiovascular diseases and other related disorders.


Subject(s)
Angiotensin I/chemistry , Antihypertensive Agents/chemistry , Antihypertensive Agents/pharmacology , Drug Discovery , Oligopeptides/chemistry , Peptide Fragments/chemistry , Renin-Angiotensin System/physiology , Angiotensin I/physiology , Angiotensin II/analogs & derivatives , Angiotensin II/chemistry , Angiotensin II/physiology , Angiotensin-Converting Enzyme 2 , Animals , Antihypertensive Agents/isolation & purification , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Drug Discovery/methods , Humans , Male , Oligopeptides/physiology , Peptide Fragments/physiology , Peptidyl-Dipeptidase A/physiology , Proto-Oncogene Mas , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/physiology , Rats , Rats, Inbred F344 , Rats, Inbred SHR , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/physiology
11.
Pharmacol Res ; 101: 41-55, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26238180

ABSTRACT

Insulin-like growth factor-1, angiotensin-(1-7) and angiotensin-(1-9) have been proposed to be important mediators in cardioprotection. A large body of evidence indicates that insulin like growth factor-1 has pleotropic actions in the heart (i.e., contractility, metabolism, hypertrophy, autophagy, senescence and cell death) and, conversely, its deficiency is associated with impaired cardiac function. Recently, we reported that insulin like growth factor-1 receptor is also located in plasma membrane invaginations with perinuclear localization, highlighting the role of nuclear Ca(2+) signaling in the heart. In parallel, angiotensin-(1-7) and angiotensin (1-9) acting through Mas receptor and angiotensin type 2 receptor have emerged as a novel anti-hypertensive molecules promoting vasodilatation and preventing heart hypertrophy. In this review we discuss the scientific evidence available regarding insulin-like growth factor-1, angiotensin-(1-7) and angiotensin-(1-9) in cardioprotection and its potential application as novel therapeutic targets for treating cardiac diseases.


Subject(s)
Angiotensin I/physiology , Cardiovascular Agents/pharmacology , Insulin-Like Growth Factor I/physiology , Peptide Fragments/physiology , Animals , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/physiopathology , Cardiovascular Physiological Phenomena , Cardiovascular System/drug effects , Humans , Models, Cardiovascular , Proto-Oncogene Mas , Proto-Oncogene Proteins/physiology , Receptor, Angiotensin, Type 2/physiology , Receptor, IGF Type 1/physiology , Receptors, G-Protein-Coupled/physiology , Renin-Angiotensin System/physiology , Signal Transduction
12.
Inflamm Res ; 64(3-4): 253-60, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25721616

ABSTRACT

BACKGROUND: Angiotensin-converting enzyme 2 (ACE2) is a counter-regulator against ACE by converting angiotensin II (Ang-II) to Ang-(1-7), but the effect of ACE2 and Ang-(1-7) on endothelial cell function and atherosclerotic evolution is unknown. We hypothesized that ACE2 overexpression and Ang-(1-7) may protect endothelial cell function by counterregulation of angiotensin II signaling and inhibition of inflammatory response. METHODS: We used a recombinant adenovirus vector to locally overexpress ACE2 gene (Ad-ACE2) in human endothelial cells in vitro and in apoE-deficient mice in vivo. The Ang II-induced MCP-1, VCAM-1 and E-selectin expression, endothelial cell migration and adhesion of human monocytic cells (U-937) to HUVECs by ACE2 gene transfer were evaluated in vitro. Accelerated atherosclerosis was studied in vivo, and atherosclerosis was induced in apoE-deficient mice which were divided randomly into four groups that received respectively a ACE2 gene transfer, Ad-ACE2, Ad-EGFP, Ad-ACE2 + A779, an Ang-(1-7) receptor antagonist, control group. After a gene transfer for 4 weeks, atherosclerotic pathology was evaluated. RESULTS: ACE2 gene transfer not only promoted HUVECs migration, inhibited adhesion of monocyte to HUVECs and decreased Ang II-induced MCP-1, VCAM-1 and E-selectin protein production in vitro, but also decreased the level of MCP-1, VCAM-1 and interleukin 6 and inhibit atherosclerotic plaque evolution in vivo. Further, administration of A779 increased the level of MCP-1, VCAM-1 and interleukin 6 in vivo and led to further advancements in atherosclerotic extent. CONCLUSIONS: ACE2 and Ang-(1-7) significantly inhibit early atherosclerotic lesion formation via protection of endothelial function and inhibition of inflammatory response.


Subject(s)
Angiotensin II/physiology , Angiotensin I/physiology , Atherosclerosis/prevention & control , Endothelium, Vascular/physiology , Inflammation/prevention & control , Peptide Fragments/physiology , Peptidyl-Dipeptidase A/physiology , Signal Transduction/physiology , Angiotensin I/genetics , Angiotensin-Converting Enzyme 2 , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/physiopathology , Cell Adhesion/physiology , Cell Movement/physiology , Chemokine CCL2/physiology , Disease Models, Animal , E-Selectin/physiology , Endothelium, Vascular/cytology , Gene Transfer Techniques , Humans , In Vitro Techniques , Inflammation/physiopathology , Mice , Peptide Fragments/genetics , Peptidyl-Dipeptidase A/genetics , Vascular Cell Adhesion Molecule-1/physiology
13.
Am J Physiol Renal Physiol ; 306(10): F1136-42, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24647710

ABSTRACT

ANG (1-7) contributes to the blood pressure (BP)-lowering effect of angiotensin receptor blockers (ARBs) in male experimental animals. Females have greater ANG (1-7) concentrations than males; however, the contribution of ANG (1-7) to ARB-mediated decreases in BP in females is unknown. The current study tested the hypothesis that female spontaneously hypertensive rats (SHR) have a larger ANG (1-7) contribution to the BP-lowering effects of the ARB candesartan than male SHR. Twelve-week-old male and female SHR were randomized to receive candesartan (0.5 mg·kg(-1)·day(-1); 7 days), candesartan plus ANG II (200 ng·kg(-1)·min(-1); 7 days), the ANG (1-7) antagonist A-779 (48 µg·kg(-1)·h(-1)) plus candesartan and ANG II. Candesartan decreased basal BP in males and females (baseline vs. candesartan: 142 ± 2 vs. 122 ± 3 and 129 ± 1 vs. 115 ± 1 mmHg, respectively; P < 0.05); however, the decrease was greater in males. ANG II increased BP in males in the presence of candesartan (149 ± 2 mmHg; P < 0.05); candesartan blocked ANG II-induced increases in BP in females (116 ± 1 mmHg). Pretreatment with A-779 abolished candesartan-mediated decreases in BP in females, but not males. A-779 also exacerbated ANG II-induced proteinuria (26 ± 6 vs. 77 ± 11 µg·kg(-1)·day(-1), respectively; P < 0.05) and nephrinuria (20 ± 5 vs. 202 ± 58 µg·kg(-1)·day(-1), respectively; P < 0.05) in candesartan-treated female SHR, with no effect in males. In conclusion, females are more sensitive to the BP-lowering effect of ARBs during ANG II infusion, whereas males are more sensitive under basal conditions. In addition, ANG (1-7) has a greater contribution to ARB-mediated decreases in BP, protein, and nephrin excretion in females relative to males.


Subject(s)
Angiotensin I/physiology , Angiotensin Receptor Antagonists/pharmacology , Blood Pressure/physiology , Hypertension/physiopathology , Peptide Fragments/physiology , Receptor, Angiotensin, Type 1/drug effects , Sex Factors , Angiotensin II/analogs & derivatives , Angiotensin II/pharmacology , Animals , Benzimidazoles/pharmacology , Biphenyl Compounds , Blood Pressure/drug effects , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Hypertension/metabolism , Male , Membrane Proteins/metabolism , Peptide Fragments/pharmacology , Rats , Rats, Inbred SHR , Receptor, Angiotensin, Type 1/physiology , Tetrazoles/pharmacology
14.
Am J Physiol Regul Integr Comp Physiol ; 307(5): R487-9, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24944244

ABSTRACT

The kidney is a key target organ for bioactive components of the renin-angiotensin system (RAS); however, various renal cells such as the tubular epithelium contain an intrinsic RAS. The renal RAS can be functionally divided into ANG II-AT1 receptor and ANG-(1-7)-AT7/Mas receptor arms that functionally oppose one another. The current review considers both extracellular and intracellular pathways that potentially govern the formation and metabolism of angiotensin peptides within the renal proximal tubules.


Subject(s)
Angiotensins/metabolism , Kidney Tubules, Proximal/metabolism , Angiotensin I/physiology , Animals , Humans , Peptide Fragments/physiology , Receptor, Angiotensin, Type 1/physiology , Renin-Angiotensin System/physiology
15.
Clin Sci (Lond) ; 126(12): 815-27, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24593683

ABSTRACT

The RAS (renin-angiotensin system) is integral to cardiovascular physiology; however, dysregulation of this system largely contributes to the pathophysiology of CVD (cardiovascular disease). It is well established that AngII (angiotensin II), the main effector of the RAS, engages the AT1R (angiotensin type 1 receptor) and promotes cell growth, proliferation, migration and oxidative stress, all processes which contribute to remodelling of the heart and vasculature, ultimately leading to the development and progression of various CVDs, including heart failure and atherosclerosis. The counter-regulatory axis of the RAS, which is centred on the actions of ACE2 (angiotensin-converting enzyme 2) and the resultant production of Ang-(1-7) [angiotensin-(1-7)] from AngII, antagonizes the actions of AngII via the receptor Mas, thereby providing a protective role in CVD. More recently, another ACE2 metabolite, Ang-(1-9) [angiotensin-(1-9)], has been reported to be a biologically active peptide within the counter-regulatory axis of the RAS. The present review will discuss the role of the counter-regulatory RAS peptides Ang-(1-7) and Ang-(1-9) in the cardiovascular system, with a focus on their effects in remodelling of the heart and vasculature.


Subject(s)
Angiotensin I/physiology , Heart/physiology , Peptide Fragments/physiology , Humans , Renin-Angiotensin System
16.
Clin Sci (Lond) ; 126(3): 195-205, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24102099

ABSTRACT

The goal of the present review is to examine the evidence for beneficial actions of manipulation of the RAS (renin-angiotensin system) in stroke, with particular focus on Ang-(1-7) [angiotensin-(1-7)] and its receptor Mas. The RAS appears to be highly involved in the multifactorial pathophysiology of stroke. Blocking the effects of AngII (angiotensin II) at AT1R (AngII type 1 receptor), through the use of commonly prescribed ACE (angiotensin-converting enzyme) inhibitors or AT1R blockers, has been shown to have therapeutic effects in both ischaemic and haemorrhagic stroke. In contrast with the deleterious actions of over activation of AT1R by AngII, stimulation of AT2Rs (AngII type 2 receptors) in the brain has been demonstrated to elicit beneficial effects in stroke. Likewise, the ACE2/Ang-(1-7)/Mas axis of the RAS has been shown to have therapeutic effects in stroke when activated, countering the effects of the ACE/AngII/AT1R axis. Studies have demonstrated that activating this axis in the brain elicits beneficial cerebral effects in rat models of ischaemic stroke, and we have also demonstrated the cerebroprotective potential of this axis in haemorrhagic stroke using stroke-prone spontaneously hypertensive rats and collagenase-induced striatal haemorrhage. The mechanism of cerebroprotection elicited by ACE2/Ang-(1-7)/Mas activation includes anti-inflammatory effects within the brain parenchyma. The major hurdle to overcome in translating these results to humans is devising strategies to activate the ACE2/Ang-(1-7)/Mas cerebroprotective axis using post-stroke treatments that can be administered non-invasively.


Subject(s)
Angiotensin I/therapeutic use , Peptide Fragments/therapeutic use , Peptides/physiology , Peptides/therapeutic use , Proto-Oncogene Proteins/agonists , Receptors, G-Protein-Coupled/agonists , Renin-Angiotensin System/physiology , Stroke/drug therapy , Vasodilator Agents/therapeutic use , Angiotensin I/physiology , Animals , Humans , Peptide Fragments/physiology , Proto-Oncogene Mas , Translational Research, Biomedical , Vasodilator Agents/pharmacology
17.
Article in English | MEDLINE | ID: mdl-25149042

ABSTRACT

Using embryonic chickens (Gallus gallus domesticus), we examined the role of the renin-angiotensin system (RAS) in cardiovascular and osmotic homeostasis through chronic captopril, an angiotensin-converting enzyme (ACE) inhibitor. Captopril (5 mg kg⁻¹ embryo wet mass) or saline (control) was delivered via the egg air cell daily from embryonic day 5-18. Mean arterial pressure (MAP), heart rate (ƒ(H)), fluid osmolality and ion concentration, and embryonic and organ masses were measured on day 19. Exogenous angiotensin I (ANG I) injection did not change MAP or ƒ(H) in captopril-treated embryos, confirming ACE inhibition. Captopril-treated embryos were significantly hypotensive, with MAP 15% lower than controls, which we attributed to the loss of vasoconstrictive ANG II action. Exogenous ANG II induced a relatively greater hypertensive response in captopril-treated embryos compared to controls. Changes in response to ANG II following pre-treatment with phentolamine (α-adrenergic antagonist) indicated a portion of the ANG II response was due to circulating catecholamines in captopril-treated embryos. An increase in MAP and ƒ(H) in response to hexamethonium indicated vagal tone was also increased in the absence of ACE activity. Captopril-treated embryos had lower osmolality, lower Na⁺ and higher K⁺ concentration in the blood, indicating osmoregulatory changes. Larger kidney mass in captopril-treated embryos suggests disrupting the RAS may stimulate kidney growth by decreasing resistance at the efferent arteriole and increasing the fraction of cardiac output to the kidneys. This study suggests that the RAS, most likely through ANG II action, influences the development of the cardiovascular and osmoregulatory systems.


Subject(s)
Avian Proteins/physiology , Cardiovascular System/embryology , Chick Embryo/physiology , Heart Rate , Osmoregulation , Renin-Angiotensin System , Vascular Resistance , Adrenergic alpha-Antagonists/pharmacology , Angiotensin I/antagonists & inhibitors , Angiotensin I/physiology , Angiotensin II/physiology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Avian Proteins/antagonists & inhibitors , Cardiovascular System/drug effects , Catecholamines/antagonists & inhibitors , Catecholamines/physiology , Chick Embryo/drug effects , Heart Rate/drug effects , Kidney/drug effects , Kidney/embryology , Nicotinic Antagonists/pharmacology , Organogenesis/drug effects , Osmoregulation/drug effects , Renin-Angiotensin System/drug effects , Vascular Resistance/drug effects , Vasoconstriction/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology
18.
J Physiol Pharmacol ; 75(2): 185-194, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38736265

ABSTRACT

We have previously described local aldosterone synthesis in mouse colon. In the renin-angiotensin-aldosterone system (RAAS), angiotensin II (Ang II) peptide is the physiological factor which stimulates aldosterone synthesis in the adrenal glands. We have recently demonstrated that Ang II stimulates aldosterone synthesis also in mouse colon. Here, we conducted a 75-min ex vivo incubation of murine colonic tissue and evaluated the effects of three other Ang peptides, Ang I (1 µM), Ang III (0.1 µM) and Ang (1-7) (0.1 µM) on aldosterone synthesis. As a possible mechanism, their effects on tissue levels of the rate-limiting enzyme, aldosterone synthase (CYP11B2) were measured by ELISA and Western blot. Ang III significantly elevated the amount of tissue CYP11B2 protein in colon. The values of released aldosterone in colon tissue incubation were increased over the control in the presence of Ang I, II or III, however, being statistically non-significant. In Western blot analysis, the values of tissue CYP11B2 protein content were elevated by Ang I and II. Ang (1-7) alone in colon did not influence CYP11B2 protein levels in the incubation experiment but showed higher aldosterone release without statistical significance. Ang (1-7) showed an antagonistic effect towards Ang II in release of aldosterone in adrenal gland. An overall estimation of a single peptide (three measured variables), the results were always in an increasing direction. The responses of aldosterone synthesis to high levels of glucose (44 mM) and potassium (18.8 mM) as physiological stimulators in vivo were investigated in the colon incubation. Glucose, equal to four times the concentration of the control buffer in the incubation, showed higher values of aldosterone release in colon than control without statistical significance similarly to the effect seen in adrenal glands. Increasing the concentration of potassium in the incubation buffer exerted no effect on colonic aldosterone production. Intriguingly, no correlation was found between aldosterone release and the tissue CYP11B2 protein content in colon. In summary, the response of colonic aldosterone synthesis to different Ang peptides resembles, but is not identical to, the situation in the adrenal glands.


Subject(s)
Aldosterone , Colon , Cytochrome P-450 CYP11B2 , Glucose , Potassium , Animals , Male , Mice , Aldosterone/metabolism , Angiotensin I/physiology , Angiotensin II/physiology , Angiotensin III/physiology , Colon/metabolism , Colon/drug effects , Cytochrome P-450 CYP11B2/metabolism , Glucose/metabolism , Peptide Fragments/physiology , Potassium/metabolism
19.
Eur J Clin Invest ; 43(9): 978-85, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23909886

ABSTRACT

BACKGROUND: The renin-angiotensin system (RAS) has been shown to play an active role within the erectile tissues. The aim of this narrative review is to summarize the literature addressing the pathophysiological role of RAS on erectile function. Additionally, we update evidence on recent findings on the role of the Ang-(1-7) and Mas receptor on the erectile function and its therapeutic potential for treating erectile dysfunction (ED). MATERIALS AND METHODS: This narrative review is based on the material searched and obtained via MEDLINE and PubMed up to November 2012. The search terms we used are 'angiotensin, erectile dysfunction, renin, Mas receptor' in combination with 'pathophysiology, fibrosis, pathways'. RESULTS: The levels of angiotensin (Ang) II, the main component of this system, are increased in the corpus cavernosum as compared to those found in the systemic circulation. Moreover, emerging evidence indicates that an increased activity of Ang II via AT1 receptor might contribute to the development of ED, whereas the pharmacological blockage of Ang II/AT1 actions has beneficial effects on the erection. On the other hand, the heptapeptide Ang-(1-7), known as a major endogenous counter-regulator of Ang II actions, favours penile erection via the activation of Mas receptor. CONCLUSIONS: Ang-(1-7) and Mas receptor pathway might be considered as a promising therapeutic target for the treatment of ED.


Subject(s)
Erectile Dysfunction/etiology , Renin-Angiotensin System/physiology , Angiotensin I/physiology , Angiotensin II/physiology , Humans , Male , Penile Erection/physiology , Peptide Fragments/physiology , Proto-Oncogene Mas , Proto-Oncogene Proteins/physiology , Receptor, Angiotensin, Type 1/physiology , Receptors, G-Protein-Coupled/physiology
20.
Clin Sci (Lond) ; 124(7): 443-56, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23249272

ABSTRACT

It is well known that the RAS (renin-angiotensin system) plays a key role in the modulation of many functions in the body. AngII (angiotensin II) acting on AT1R (type 1 AngII receptor) has a central role in mediating most of the actions of the RAS. However, over the past 10 years, several studies have presented evidence for the existence of a new arm of the RAS, namely the ACE (angiotensin-converting enzyme) 2/Ang-(1-7) [angiotensin-(1-7)]/Mas axis. Ang-(1-7) can be produced from AngI or AngII via endo- or carboxy-peptidases respectively. ACE2 appears to play a central role in Ang-(1-7) formation. As described for AngII, Ang-(1-7) also has a broad range of effects in different organs and tissues which goes beyond its initially described cardiovascular and renal actions. Those effects are mediated by Mas and can counter-regulate most of the deleterious effects of AngII. The interaction Ang-(1-7)/Mas regulates different signalling pathways, such as PI3K (phosphoinositide 3-kinase)/AKT and ERK (extracellularsignal-regulated kinase) pathways and involves downstream effectors such as NO, FOXO1 (forkhead box O1) and COX-2 (cyclo-oxygenase-2). Through these mechanisms, Ang-(1-7) is able to improve pathological conditions including fibrosis and inflammation in organs such as lungs, liver and kidney. In addition, this heptapeptide has positive effects on metabolism, increasing the glucose uptake and lipolysis while decreasing insulin resistance and dyslipidaemia. Ang-(1-7) is also able to improve cerebroprotection against ischaemic stroke, besides its effects on learning and memory. The reproductive system can also be affected by Ang-(1-7) treatment, with enhanced ovulation, spermatogenesis and sexual steroids synthesis. Finally, Ang-(1-7) is considered a potential anti-cancer treatment since it is able to inhibit cell proliferation and angiogenesis. Thus the ACE2/Ang-(1-7)/Mas pathway seems to be involved in many physiological and pathophysiological processes in several systems and organs especially by opposing the detrimental effects of inappropriate overactivation of the ACE/AngII/AT1R axis.


Subject(s)
Angiotensin I/physiology , Angiotensin I/therapeutic use , Peptide Fragments/physiology , Peptide Fragments/therapeutic use , Peptidyl-Dipeptidase A/metabolism , Proto-Oncogene Proteins/physiology , Receptors, G-Protein-Coupled/physiology , Angiogenesis Inhibitors/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Brain Ischemia/prevention & control , Cell Proliferation/drug effects , Female , Fibrosis/prevention & control , Glucose/metabolism , Humans , Insulin/metabolism , Kidney/metabolism , Lipid Metabolism/drug effects , Male , Metabolic Syndrome/prevention & control , Proto-Oncogene Mas , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Reproduction/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL