Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 215
Filter
1.
Brain Res ; 1769: 147618, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34400123

ABSTRACT

Angiotensin II (AngII) immunoreactive cells, fibers and receptors, were found in the parvocelluar region of paraventricular nucleus (PVNp) and AngII receptors are present on vasopressinergic neurons. However, the mechanism by which vasopressin (AVP) and AngII may interact to regulate arterial pressure is not known. Thus, we tested the cardiovascular effects of blockade of the AngII receptors on AVP neurons and blockade of vasopressin V1a receptors on AngII neurons. We also explored whether the PVNp vasopressin plays a regulatory role during hypotension in anesthetized rat or not. Hypovolemic-hypotension was induced by gradual bleeding from femoral venous catheter. Either AngII or AVP injected into the PVNp produced pressor and tachycardia responses. The responses to AngII were blocked by V1a receptor antagonist. The responses to AVP were partially attenuated by AT1 antagonist and greatly attenuated by AT2 antagonist. Hemorrhage augmented the pressor response to AVP, indicating that during hemorrhage, sensitivity of PVNp to vasopressin was increased. By hemorrhagic-hypotension and bilateral blockade of V1a receptors of the PVNp, we found that vasopressinergic neurons of the PVNp regulate arterial pressure towards normal during hypotension. Taken together these findings and our previous findings about angII (Khanmoradi and Nasimi, 2017a) for the first time, we found that a mutual cooperative system of angiotensinergic and vasopressinergic neurons in the PVNp is a major regulatory controller of the cardiovascular system during hypotension.


Subject(s)
Angiotensin II , Arterial Pressure , Hypotension/physiopathology , Nerve Net/physiopathology , Paraventricular Hypothalamic Nucleus/physiopathology , Vasopressins , Angiotensin I/antagonists & inhibitors , Angiotensin II Type 2 Receptor Blockers/pharmacology , Animals , Hemorrhage/physiopathology , Hypovolemia/physiopathology , Male , Rats , Rats, Sprague-Dawley
2.
Toxicol Appl Pharmacol ; 408: 115255, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33007385

ABSTRACT

Silicosis is a major public health concern with various contributing factors. The renin-angiotensin system (RAS)is a critical regulator in the pathogenesis of this disease. We focused on two key RAS enzymes, angiotensin-converting enzyme (ACE) and angiotensin-converting enzyme 2 (ACE2), to elucidate the activation of the ACE-angiotensin II (Ang II)-angiotensin II receptor 1 (AT1) axis and the inhibition of the ACE2-angiotensin-(1-7) [Ang-(1-7)]-Mas receptor axis in C57BL/6mice following SiO2 treatment. Silica exposure caused nodule formation, pulmonary interstitial fibrosis, epithelial-mesenchymal transition (EMT), abnormal deposition of extracellular matrix, and impaired lung function in mice. These effects were attenuated by the inhibition of ACE (captopril), blockade of the AT1(losartan), or systemic knockdown of the Ace gene. These effects were exacerbated by the inhibition of ACE2 (MLN-4760), blockade of the Mas (A779), or knockdown of the Ace2 gene. N-Acetyl-Seryl-Asparyl-Lysyl-Proline (Ac-SDKP), an anti-fibrotic peptide, ameliorated the silica-exposure-induced pathological changes by targeting the RAS system by activating the protective ACE2-Ang-(1-7)-Mas axis and inhibiting the deleterious ACE-Ang II-AT1 axis, thereby exerting a protective effect. This was confirmed in mouse lung type II epithelial cells (MLE-12) pretreated with Ang II and/or gene silencing separately targeting Ace and Ace2.The effects of Ac-SDKP were similar to those produced by Ace gene silencing and were partly attenuated by Ace2 deficiency. These findings suggested that RAS plays critical roles in the pathomechanism of silicosis fibrosis and that Ac-SDKP regulates lung RAS to inhibit EMT in silicotic mice and MLE-12 cells.


Subject(s)
Epithelial-Mesenchymal Transition , Lung/metabolism , Oligopeptides , Renin-Angiotensin System , Silicosis/metabolism , Angiotensin I/antagonists & inhibitors , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme 2/antagonists & inhibitors , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Captopril/pharmacology , Cell Line , Epithelial-Mesenchymal Transition/drug effects , Fibrosis , Losartan/pharmacology , Lung/drug effects , Lung/pathology , Lung/physiology , Male , Mice, Inbred C57BL , Peptide Fragments/antagonists & inhibitors , Peptidyl-Dipeptidase A , Renin-Angiotensin System/drug effects , Silicosis/pathology , Silicosis/physiopathology
3.
Neuropeptides ; 83: 102076, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32800589

ABSTRACT

The basolateral amygdala (BLA) is critical in the control of the sympathetic output during stress. Studies demonstrated the involvement of the renin-angiotensin system components in the BLA. Angiotensin-(1-7) [Ang-(1-7)], acting through Mas receptors, reduces stress effects. Considering that angiotensin-converting enzyme 2 (ACE2) is the principal enzyme for the production of Ang-(1-7), here we evaluate the cardiovascular reactivity to acute stress after administration of the ACE2 activator, diminazene aceturate (DIZE) into the BLA. We also tested whether systemic treatment with DIZE could modify synaptic activity in the BLA and its effect directly on the expression of the N-methyl-d-aspartate receptors (NMDARs) in NG108 neurons in-vitro. Administration of DIZE into the BLA (200 pmol/100 nL) attenuated the tachycardia to stress (ΔHR, bpm: vehicle = 103 ± 17 vs DIZE = 49 ± 7 p = 0.018); this effect was inhibited by Ang-(1-7) antagonist, A-779 (ΔHR, bpm: DIZE = 49 ± 7 vs A-779 + DIZE = 100 ± 15 p = 0.04). Systemic treatment with DIZE attenuated the excitatory synaptic activity in the BLA (Frequency (Hz): vehicle = 2.9 ± 0.4 vs. DIZE =1.8 ± 0.3 p < 0.04). NG108 cells treated with DIZE demonstrated decreased expression of l subunit NMDAR-NR1 (NR1 expression (a.u): control = 0.534 ± 0.0593 vs. DIZE = 0.254 ± 0.0260) of NMDAR and increases of Mas receptors expression. These data demonstrate that DIZE attenuates the tachycardia evoked by acute stress. This effect results from a central action in the BLA involving activation of Mas receptors. The ACE2 activation via DIZE treatment attenuated the frequency of excitatory synaptic activity in the basolateral amygdala and this effect can be related with the decreases of the NMDAR-NR1 receptor expression.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Basolateral Nuclear Complex/drug effects , Diminazene/analogs & derivatives , Glutamic Acid/metabolism , Heart Rate/drug effects , Neurons/drug effects , Tachycardia/metabolism , Angiotensin I/antagonists & inhibitors , Angiotensin II/analogs & derivatives , Angiotensin II/pharmacology , Animals , Basolateral Nuclear Complex/metabolism , Diminazene/pharmacology , Neurons/metabolism , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/pharmacology , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism
4.
Am J Physiol Renal Physiol ; 317(4): F949-F956, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31411069

ABSTRACT

The role of dopamine D1-like receptors (DR) in the regulation of renal Na+ transporters, natriuresis, and blood pressure is well established. However, the involvement of the angiotensin 1-7 (ANG 1-7)-Mas receptor in the regulation of Na+ balance and blood pressure is not clear. The present study aimed to investigate the hypothesis that ANG 1-7 can regulate Na+ homeostasis by modulating the renal dopamine system. Sprague-Dawley rats were infused with saline alone (vehicle) or saline with ANG 1-7, ANG 1-7 antagonist A-779, DR agonist SKF38393, and antagonist SCH23390. Infusion of ANG 1-7 caused significant natriuresis and diuresis compared with saline alone. Both natriuresis and diuresis were blocked by A-779 and SCH23390. SKF38393 caused a significant, SCH23390-sensitive natriuresis and diuresis, and A-779 had no effect on the SKF38393 response. Concomitant infusion of ANG 1-7 and SKF38393 did not show a cumulative effect compared with either agonist alone. Treatment of renal proximal tubules with ANG 1-7 or SKF38393 caused a significant decrease in Na+-K+-ATPase and Na+/H+ exchanger isoform 3 activity. While SCH23390 blocked both ANG 1-7- and SKF38393-induced inhibition, the DR response was not sensitive to A-779. Additionally, ANG 1-7 activated PKG, enhanced tyrosine hydroxylase activity via Ser40 phosphorylation, and increased renal dopamine production. These data suggest that ANG 1-7, via PKG, enhances tyrosine hydroxylase activity, which increases renal dopamine production and activation of DR and subsequent natriuresis. This study provides evidence for a unidirectional functional interaction between two G protein-coupled receptors to regulate renal Na+ transporters and induce natriuresis.


Subject(s)
Angiotensin I/pharmacology , Kidney/metabolism , Peptide Fragments/pharmacology , Receptors, Dopamine D1/metabolism , Receptors, G-Protein-Coupled/metabolism , Sodium/metabolism , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Angiotensin I/antagonists & inhibitors , Animals , Benzazepines/pharmacology , Cyclic GMP-Dependent Protein Kinases/metabolism , Diuresis/drug effects , Dopamine/biosynthesis , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Natriuresis/drug effects , Peptide Fragments/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/drug effects , Sodium-Hydrogen Exchangers/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism
5.
Heart Fail Rev ; 24(5): 709-723, 2019 09.
Article in English | MEDLINE | ID: mdl-31104255

ABSTRACT

The renin-angiotensin-aldosterone system (RAAS) plays a major role in the regulation of blood pressure and homeostasis. Therefore, it is a commonly used target for pharmacotherapy of cardiovascular diseases in adults. However, the efficacy of this pharmacotherapy can only be limitedly derived into children. Comprehensive knowledge of the humoral parameters acting in the paediatric RAAS (e.g. angiotensin I, angiotensin II, angiotensin 1-7, angiotensin III, and angiotensin IV) might facilitate a more effective and rational pharmacotherapy in children. Therefore, this review aims to provide an overview of the maturing RAAS. Out of 925 identified records, 35 publications were classified as relevant. Physiological and pathophysiological concentrations of angiotensin peptides were compiled and categorised according to European Medicines Agency age groups. Age has a major impact on circulating angiotensin I, angiotensin II, and angiotensin 1-7, which is reflected in an age-dependent decrease during childhood. In contrast to data obtained in adults, no gender-related differences in angiotensin levels were identified. The observed increase in peptide concentrations regarding cardiac- and renal-diseased children is influenced by surgical repair, while evidence for a pharmacological impact is conflicting. A comprehensive set of angiotensin I, angiotensin II, and angiotensin 1-7 values from neonates up to adolescents was compiled. Indicating age as a strong effector. However, evidence about potential promising targets of the RAAS like angiotensin III and angiotensin IV is still lacking in children.


Subject(s)
Angiotensin III/blood , Angiotensin II/analogs & derivatives , Angiotensin I/blood , Hypertension/blood , Kidney Failure, Chronic/blood , Peptide Fragments/blood , Adolescent , Age Factors , Angiotensin I/antagonists & inhibitors , Angiotensin II/blood , Angiotensin III/antagonists & inhibitors , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Antihypertensive Agents/therapeutic use , Child , Child, Preschool , Female , Humans , Hypertension/complications , Hypertension/drug therapy , Infant , Infant, Newborn , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/drug therapy , Male , Peptide Fragments/antagonists & inhibitors , Renin-Angiotensin System/drug effects
6.
J Steroid Biochem Mol Biol ; 185: 137-141, 2019 01.
Article in English | MEDLINE | ID: mdl-30125658

ABSTRACT

Angiotensin 1-7 (Ang 1-7), which is a protein cleaved from angiotensin II (A-II), binds to the MAS receptor. Ang 1-7 has been demonstrated to exert protective effects against A-II-mediated cardiac, atherosclerotic, and renal damages. The aims of our study were to demonstrate the inhibitory role of Ang 1-7 in A-II-mediated aldosterone production by interacting with the MAS receptor in human adrenocortical carcinoma (HAC15) cells, and clarify the intracellular signaling mechanisms underlying the inhibition of aldosterone production by Ang 1-7. Ang 1-7 significantly suppressed A-II-stimulated aldosterone production, and partially abrogated A-II-induced upregulation of CYP11B2 expression. Treatment with a selective Ang 1-7 antagonist abrogated Ang 1-7-mediated inhibition of aldosterone production in HAC15 cells. Incubation of A-II-treated HAC15 cells with conditioned medium containing Ang 1-7 was demonstrated to suppress A-II-mediated aldosterone production and CYP11B2 expression. Proteomic analysis showed that Ang 1-7 predominantly inhibited the phosphorylation of JAK-STAT proteins in A-II stimulated HAC15 cells. Treatment of HAC15 cells with a STAT3 inhibitor partially but significantly repressed A-II-mediated aldosterone production by 63.2%. Similarly, treatment with a STAT5 inhibitor significantly abrogated A-II-stimulated aldosterone production in HAC15 cells by 60.7%. In conclusion, we demonstrated that Ang 1-7 negatively regulates A-II-mediated aldosterone production, and the observed inhibition of aldosterone production was associated with JAK/STAT signaling in human adrenal cells. Therefore, activation of Ang 1-7 or stimulation of the MAS receptor, which inhibits aldosterone production, is a promising therapeutic approach for the prevention of cardiovascular events that can directly affect the target organs.


Subject(s)
Aldosterone/biosynthesis , Angiotensin II/metabolism , Angiotensin I/metabolism , Janus Kinases/antagonists & inhibitors , Peptide Fragments/metabolism , STAT Transcription Factors/antagonists & inhibitors , Adrenocortical Carcinoma/metabolism , Angiotensin I/antagonists & inhibitors , Cardiovascular Diseases/drug therapy , Cell Line, Tumor , Culture Media, Conditioned/pharmacology , Cytochrome P-450 CYP11B2/biosynthesis , Humans , Peptide Fragments/antagonists & inhibitors , Phosphorylation/drug effects , Signal Transduction/drug effects
7.
Neurosci Bull ; 35(1): 47-56, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30328008

ABSTRACT

Angiotensin (Ang)-(1-7) is an important biologically-active peptide of the renin-angiotensin system. This study was designed to determine whether inhibition of Ang-(1-7) in the hypothalamic paraventricular nucleus (PVN) attenuates sympathetic activity and elevates blood pressure by modulating pro-inflammatory cytokines (PICs) and oxidative stress in the PVN in salt-induced hypertension. Rats were fed either a high-salt (8% NaCl) or a normal salt diet (0.3% NaCl) for 10 weeks, followed by bilateral microinjections of the Ang-(1-7) antagonist A-779 or vehicle into the PVN. We found that the mean arterial pressure (MAP), renal sympathetic nerve activity (RSNA), and plasma norepinephrine (NE) were significantly increased in salt-induced hypertensive rats. The high-salt diet also resulted in higher levels of the PICs interleukin-6, interleukin-1beta, tumor necrosis factor alpha, and monocyte chemotactic protein-1, as well as higher gp91phox expression and superoxide production in the PVN. Microinjection of A-779 (3 nmol/50 nL) into the bilateral PVN of hypertensive rats not only attenuated MAP, RSNA, and NE, but also decreased the PICs and oxidative stress in the PVN. These results suggest that the increased MAP and sympathetic activity in salt-induced hypertension can be suppressed by blockade of endogenous Ang-(1-7) in the PVN, through modulation of PICs and oxidative stress.


Subject(s)
Angiotensin I/antagonists & inhibitors , Hypertension/drug therapy , Oxidative Stress/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Peptide Fragments/antagonists & inhibitors , Sodium Chloride, Dietary/pharmacology , Angiotensin I/metabolism , Animals , Antioxidants/pharmacology , Blood Pressure/drug effects , Hypertension/chemically induced , Male , Peptide Fragments/metabolism , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
8.
Biomed Pharmacother ; 109: 639-645, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30404071

ABSTRACT

Local renin-angiotensin system (RAS) in the pancreas is linked to the modulation of glucose-stimulated insulin secretion (GSIS) in beta cells and insulin sensitivity in target tissues, emerging as a promising tool in the prevention and/or treatment of obesity, diabetes, and systemic arterial hypertension. Insulin resistance alters pancreatic islet cell distribution and morphology and hypertrophied islets exhibit upregulated angiotensin II type 1 receptor, which drives oxidative stress, apoptosis, and fibrosis, configuring beta cell dysfunction and diminishing islet lifespan. Pharmacological modulation of RAS has shown beneficial effects in diet-induced obesity model, mainly related to the translational potential that angiotensin receptor blockers and ECA2/ANG (1-7)/MAS receptor axis modulation have when it comes to islet preservation and type 2 diabetes prevention and/or treatment. This review describes the existing evidence for different approaches to blocking RAS elements in the management of insulin resistance and diabetes and focuses on islet remodeling and GSIS in rodents and humans.


Subject(s)
Drug Delivery Systems/trends , Homeostasis/physiology , Islets of Langerhans/metabolism , Renin-Angiotensin System/physiology , Angiotensin I/antagonists & inhibitors , Angiotensin I/metabolism , Angiotensin II/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Drug Delivery Systems/methods , Homeostasis/drug effects , Humans , Insulin Resistance/physiology , Islets of Langerhans/drug effects , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Renin-Angiotensin System/drug effects
9.
Biomed Pharmacother ; 110: 764-774, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30554115

ABSTRACT

Besides assisting the maintenance of blood pressure and sodium homeostasis, the renin-angiotensin system (RAS) plays a pivotal role in pathogenesis of acute kidney injury (AKI). The RAS is equipped with two arms i) the pressor arm composed of Angiotensin II (Ang II)/Angiotensin converting enzyme (ACE)/Angiotensin II type 1 receptor (AT1R) also called conventional RAS, and ii) the depressor arm consisting of Angiotensin (1-7) (Ang 1-7)/Angiotensin converting enzyme 2 (ACE2)/MasR known as non-conventional RAS. Activation of conventional RAS triggers oxidative stress, inflammatory, hypertrophic, apoptotic, and pro-fibrotic signaling cascades which promote AKI. The preclinical and clinical studies have reported beneficial as well as deleterious effects of RAS blockage either by angiotensin receptor blocker or ACE inhibitor in AKI. On the contrary, the depressor arm opposes the conventional RAS, has beneficial effects on the kidney but has been less explored in pathogenesis of AKI. This review focuses on significance of RAS in pathogenesis of AKI and provides better understanding of novel and possible therapeutic approaches to combat AKI.


Subject(s)
Acute Kidney Injury/metabolism , Renin-Angiotensin System/physiology , Acute Kidney Injury/drug therapy , Acute Kidney Injury/pathology , Angiotensin I/antagonists & inhibitors , Angiotensin I/metabolism , Angiotensin Receptor Antagonists/pharmacology , Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme 2 , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Humans , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/metabolism , Proto-Oncogene Mas , Renin-Angiotensin System/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology
10.
Neurosci Lett ; 687: 16-21, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30219484

ABSTRACT

Accumulating evidence suggests that brain angiotensin-converting enzyme (ACE)/angiotensin II/angiotensin II type I receptor axis is activated and thus contributes to the neuronal injury during ischemic stroke. Conversely, inhibition of this axis using centrally active ACE inhibitor captopril was proven neuroprotective in rodents with focal cerebral ischemia. Interestingly, captopril was able to increase angiotensin-(1-7) [Ang-(1-7)] levels in the peripheral organs. As the main component of the alternative renin-angiotensin system axis in the brain, Ang-(1-7) was revealed to protect against focal cerebral ischemia via a MAS1 receptor-dependent manner. Based on this evidence, we hypothesized that Ang-(1-7) might contribute to the neuroprotection of captopril during ischemic stroke. In this study, we evaluated this hypothesis using a rat model of focal cerebral ischemia. We revealed that brain ACE2 activity and Ang-(1-7) levels were significantly elevated following captopril treatment in rats with focal cerebral ischemia. More importantly, we showed that the neuroprotection provided by captopril was partially reversed by A-779, an antagonist for Ang-(1-7) receptor MAS1, indicating that Ang-(1-7) was involved in the neuroprotection of captopril. These findings have uncovered new mechanisms by which captopril protects against focal cerebral ischemia and further suggest that captopril may have practical clinical use for stroke prevention and treatment in addition to its antihypertensive effect.


Subject(s)
Angiotensin I/metabolism , Antihypertensive Agents/therapeutic use , Brain Ischemia/metabolism , Brain Ischemia/prevention & control , Captopril/therapeutic use , Neuroprotection/drug effects , Peptide Fragments/metabolism , Angiotensin I/antagonists & inhibitors , Angiotensin II/analogs & derivatives , Angiotensin II/pharmacology , Animals , Antihypertensive Agents/pharmacology , Brain/drug effects , Brain/metabolism , Brain Ischemia/pathology , Captopril/pharmacology , Male , Neuroprotection/physiology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/pharmacology , Proto-Oncogene Mas , Rats , Rats, Sprague-Dawley , Vasodilator Agents/pharmacology , Vasodilator Agents/therapeutic use
11.
Cell Metab ; 27(6): 1323-1337.e5, 2018 Jun 05.
Article in English | MEDLINE | ID: mdl-29706566

ABSTRACT

Fibroblast growth factor 21 (FGF21) is a metabolic hormone with pleiotropic effects on glucose and lipid metabolism and insulin sensitivity. However, the role of FGF21 in hypertension remains elusive. Here we show that FGF21 deficiency significantly exacerbates angiotensin II-induced hypertension and vascular dysfunction, whereas such negative effects are reversed by replenishment of FGF21. Mechanistically, FGF21 acts on adipocytes and renal cells to promote induction of angiotensin-converting enzyme 2 (ACE2), which in turn converts angiotensin II to angiotensin-(1-7), then inhibits hypertension and reverses vascular damage. In addition, ACE2 deficiency strikingly abrogates these beneficial effects of FGF21 in mice, including alleviation of angiotensin II-associated hypertension and vascular damage. Otherwise, pharmaceutical inhibition of angiotensin-(1-7) attenuates the protective effect of FGF21 on angiotensin II-induced vascular dysfunction, but not on hypertension. Thus, FGF21 protects against angiotensin II-induced hypertension and vascular impairment by activation of the ACE2/angiotensin-(1-7) axis via fine-tuning the multi-organ crosstalk between liver, adipose tissue, kidney, and blood vessels.


Subject(s)
Angiotensin II , Angiotensin I/metabolism , Cardiovascular System/metabolism , Fibroblast Growth Factors , Hypertension/metabolism , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Angiotensin I/antagonists & inhibitors , Angiotensin II/administration & dosage , Angiotensin II/metabolism , Angiotensin-Converting Enzyme 2 , Animals , Blood Pressure/drug effects , Cardiovascular System/drug effects , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/physiology , Kidney/drug effects , Kidney/metabolism , Loss of Function Mutation , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/antagonists & inhibitors , Peptidyl-Dipeptidase A/genetics
12.
Endocrinology ; 159(1): 238-247, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29088382

ABSTRACT

Hyperaldosteronism is associated with hypertension, cardiac hypertrophy, and congestive heart failure. Steroidogenic factors facilitate aldosterone secretion by increasing adrenal blood flow. Angiotensin (Ang) II decreases adrenal vascular tone through release of zona glomerulosa (ZG) cell-derived vasodilatory eicosanoids. However, ZG cell-mediated relaxation of bovine adrenal cortical arteries to Ang II is not altered by angiotensin type 1 or 2 receptor antagonists. Because traditional Ang II receptors do not mediate these vasorelaxations to Ang II, we investigated the role of Ang II metabolites. Ang III was identified by liquid chromatography-mass spectrometry as the primary ZG cell metabolite of Ang II. Ang III stimulated ZG cell-mediated relaxation of adrenal arteries with greater potency than did Ang II. Furthermore, ZG cell-mediated relaxations of adrenal arteries by Ang II were attenuated by aminopeptidase inhibition, and Ang III-stimulated relaxations persisted. Ang IV had little effect compared with Ang II. Moreover, ZG cell-mediated relaxations of adrenal arteries by Ang II were attenuated by an Ang III antagonist but not by an Ang (1-7) antagonist. In contrast, Ang II and Ang III were equipotent in stimulating aldosterone secretion from ZG cells and were unaffected by aminopeptidase inhibition. Additionally, aspartyl and leucyl aminopeptidases, which convert Ang II to Ang III, are the primary peptidase expressed in ZG cells. This was confirmed by enzyme activity. These data indicate that intra-adrenal metabolism of Ang II to Ang III is required for ZG cell-mediated relaxations of adrenal arteries but not aldosterone secretion. These studies have defined an important role of Ang III in the adrenal gland.


Subject(s)
Adrenal Cortex/blood supply , Angiotensin III/metabolism , Angiotensin II/metabolism , Arterioles/metabolism , Endothelium, Vascular/metabolism , Muscle, Smooth, Vascular/metabolism , Zona Glomerulosa/metabolism , Abattoirs , Adrenal Cortex/drug effects , Adrenal Cortex/metabolism , Aldosterone/metabolism , Aminopeptidases/antagonists & inhibitors , Aminopeptidases/genetics , Aminopeptidases/metabolism , Angiotensin I/antagonists & inhibitors , Angiotensin I/metabolism , Angiotensin II/analogs & derivatives , Angiotensin II/chemistry , Angiotensin II/pharmacology , Animals , Arterioles/cytology , Arterioles/drug effects , Cattle , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Gene Expression Regulation, Enzymologic/drug effects , In Vitro Techniques , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protease Inhibitors/pharmacology , Vasodilation/drug effects , Zona Glomerulosa/cytology , Zona Glomerulosa/drug effects
13.
Int J Mol Med ; 41(1): 409-420, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29138810

ABSTRACT

The aim of the present study was to investigate the role of the angiotensin-converting enzyme (ACE)2-angiotensin­(Ang)-(1-7)-Mas axis in the pathogenesis of pancreatitis and the association between this axis and the p38 mitogen-activated protein kinase (p38 MAPK)/nuclear factor (NF-κB) signaling pathway in pancreatic acinar cells. Mouse pancreatic acinar cancer (MPC-83) cells were stimulated with 10 nM caerulein (CAE) to create an in vitro model of acute pancreatitis, and collected for analysis at 2, 6, 12, 24 and 48 h post stimulation. In addition, cells were pretreated with different concentrations of Ang­(1­7), Ang­(1­7) antagonist A779, p38 MAPK inhibitor SB203580 or ACE2 inhibitor DX600 for 30 min, and then stimulated with CAE for 24 h. The ACE2, Mas receptor, p38 MAPK, phosphorylated (p)-p38 MAPK and NF-κB expression levels were evaluated using western blotting and immunofluorescence. p38 MAPK, NF-κB, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), IL-8 and IL-10 mRNA expression levels were assessed using reverse transcription-quantitative polymerase chain reaction. The results of the immunofluorescence assay demonstrated that ACE2 and p38 MAPK were present mainly in the cytoplasm, while the Mas receptor was located mainly in the cell membrane. ACE2, p38 MAPK and p-p38 MAPK protein levels were significantly increased (P<0.05) following stimulation with CAE compared with those in the control group and peaked at 24 h. Mas receptor protein levels were significantly upregulated (P<0.05) between 6 and 24 h, peaking at 12 h. Ang­(1­7) and SB203580 downregulated p-p38 MAPK and NF-κB expression and the mRNA levels of inflammatory factors IL-6, TNF-α and IL-8, but upregulated the mRNA level of inflammatory factor IL-10 compared with those treated with CAE alone. These results were supported by the opposite outcomes observed for cells treated with A779 or DX600. Therefore, it was concluded that the ACE2-Ang­(1­7)-Mas axis significantly inhibits pancreatitis by inhibition of the p38 MAPK/NF-κB signaling pathway.


Subject(s)
Inflammation/drug therapy , Peptidyl-Dipeptidase A/genetics , Proto-Oncogene Proteins/genetics , Receptors, G-Protein-Coupled/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Acinar Cells/drug effects , Acinar Cells/pathology , Angiotensin I/antagonists & inhibitors , Angiotensin I/genetics , Angiotensin II/administration & dosage , Angiotensin II/analogs & derivatives , Angiotensin-Converting Enzyme 2 , Animals , Humans , Imidazoles/administration & dosage , Inflammation/genetics , Inflammation/pathology , Mice , NF-kappa B/genetics , Pancreas/drug effects , Pancreas/pathology , Peptide Fragments/administration & dosage , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/genetics , Peptides/administration & dosage , Peptidyl-Dipeptidase A/drug effects , Proto-Oncogene Mas , Proto-Oncogene Proteins/antagonists & inhibitors , Pyridines/administration & dosage , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction/drug effects
14.
Biochem Pharmacol ; 144: 90-99, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28789938

ABSTRACT

Hyperglycemia up-regulates intracellular angiotensin II (ANG-II) production in cardiac myocytes. This study investigated the hemodynamic and metabolic effects of azilsartan (AZL) treatment in a mouse model of diabetic cardiomyopathy and whether the cardioprotective effects of AZL are mediated by the angiotensin converting enzyme (ACE)-2/ANG 1-7/Mas receptor (R) cascade. Control db/+ and db/db mice (n=5 per group) were treated with vehicle or AZL (1 or 3mg/kg/d oral gavage) from the age of 8 to 16weeks. Echocardiography was then performed and myocardial protein levels of ACE-2, Mas R, AT1R, AT2R, osteopontin, connective tissue growth factor (CTGF), atrial natriuretic peptide (ANP) and nitrotyrosine were measured by Western blotting. Oxidative DNA damage and inflammatory markers were assessed by immunofluorescence of 8-hydroxy-2'-deoxyguanosine (8-OHdG), tumor necrosis factor (TNF)-α and interleukin 6 (IL-6). Compared with db/+ mice, the vehicle-treated db/db mice developed obesity, hyperglycemia, hyperinsulinemia and diastolic dysfunction along with cardiac hypertrophy and fibrosis. AZL treatment lowered blood pressure, fasting blood glucose and reduced peak plasma glucose during an oral glucose tolerance test. AZL-3 treatment resulted in a significant decrease in the expression of cytokines, oxidative DNA damage and cardiac dysfunction. Moreover, AZL-3 treatment significantly abrogated the downregulation of ACE-2 and Mas R protein levels in db/db mice. Furthermore, AZL treatment significantly reduced cardiac fibrosis, hypertrophy and their marker molecules (osteopontin, CTGF, TGF-ß1 and ANP). Short-term treatment with AZL-3 reversed abnormal cardiac structural remodeling and partially improved glucose metabolism in db/db mice by modulating the ACE-2/ANG 1-7/Mas R pathway.


Subject(s)
Angiotensin I/metabolism , Benzimidazoles/therapeutic use , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/metabolism , Oxadiazoles/therapeutic use , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Angiotensin I/antagonists & inhibitors , Angiotensin-Converting Enzyme 2 , Animals , Benzimidazoles/pharmacology , Blood Pressure/drug effects , Blood Pressure/physiology , Diabetic Cardiomyopathies/genetics , Male , Mice , Mice, Transgenic , Oxadiazoles/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Peptide Fragments/antagonists & inhibitors , Proto-Oncogene Mas , Proto-Oncogene Proteins/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology
15.
Dev Comp Immunol ; 74: 200-208, 2017 09.
Article in English | MEDLINE | ID: mdl-28487234

ABSTRACT

We recently demonstrated Ang 1-7 reduced inflammation in the dextran sulfate sodium (DSS) colitis model. In this study we examined the effect of Ang 1-7 on modulation of plasma levels of selected cytokines and chemokines and immune cell effector functions (apoptosis, chemotaxis and superoxide release) in vitro. The degree of neutrophil recruitment to the colon was assessed by immunofluorescence and myeloperoxidase activity. Daily Ang 1-7 treatment at 0.01 mg/kg dose which previously ameliorated colitis severity, showed a significant reduction in circulating levels of several cytokines and chemokines, and neutrophil recruitment to the colonic tissue. It also significantly enhanced immune cell apoptosis, and reduced neutrophil chemotaxis and superoxide release in vitro. In contrast, daily administration of the Ang 1-7R antagonist A779 which previously worsened colitis severity showed significant up-regulation of specific mediators. Our results demonstrate a novel anti-inflammatory action of Ang 1-7 through modulation of plasma levels of cytokines/chemokines and immune cell activity.


Subject(s)
Angiotensin I/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Colitis/therapy , Colon/immunology , Neutrophils/immunology , Peptide Fragments/therapeutic use , Angiotensin I/antagonists & inhibitors , Angiotensin II/administration & dosage , Angiotensin II/analogs & derivatives , Animals , Apoptosis , Cell Movement , Chemokines/blood , Chemotaxis , Colitis/immunology , Cytokines/blood , Dextran Sulfate/immunology , Immunity, Cellular , Immunomodulation , Mice , Mice, Inbred BALB C , Models, Animal , Peptide Fragments/administration & dosage , Peptide Fragments/antagonists & inhibitors , Renin-Angiotensin System , Superoxides/metabolism
16.
Biomed Pharmacother ; 92: 58-68, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28531801

ABSTRACT

The local role of the renin angiotensin system (RAS) was documented recently beside its conventional systemic functions. Studies showed that the effector angiotensin II (AngII) alters bone health, while inhibition of the angiotensin converting enzyme (ACE-1) preserved these effects. The newly identified Ang1-7 exerts numerous beneficial effects opposing the AngII. Thus, the current study examines the role of Ang1-7 in mediating the osteo-preservative effects of ACEI (captopril) through the G-protein coupled Mas receptor using an ovariectomized (OVX) rat model of osteoporosis. 8 weeks after the surgical procedures, captopril was administered orally (40mgkg-1 d-1), while the specific Mas receptor blocker (A-779) was delivered at infusion rate of 400ngkg-1min-1 for 6 weeks. Bone metabolic markers were measured in serum and urine. Minerals concentrations were quantified in serum, urine and femoral bones by inductive coupled plasma mass spectroscopy (ICP-MS). Trabecular and cortical morphometry was analyzed in the right distal femurs using micro-CT. Finally, the expressions of RAS peptides, enzymes and receptors along with the receptor activator of NF-κB ligand (RANKL) and osteoprotegerin (OPG) were determined femurs heads. OVX animals markedly showed altered bone metabolism and mineralization along with disturbed bone micro-structure. Captopril significantly restored the metabolic bone bio-markers and corrected Ca2+ and P values in urine and bones of estrogen deficient rats. Moreover, the trabecular and cortical morphometric features were repaired by captopril in OVX groups. Captopril also improved the expressions of ACE-2, Ang1-7, Mas and OPG, while abolished OVX-induced up-regulation of ACE-1, AngII, Ang type 1 receptor (AT1R) and RANKL. Inhibition of Ang1-7 cascade by A-779 significantly eradicated captopril protective effects on bone metabolism, mineralization and micro-structure. A-779 also restored OVX effects on RANKL expression and ACE-1/AngII/AT1R cascade and down-regulated OPG expression and ACE-2/Ang1-7/Mas pathway. In line with the clinical observations of the bone-preservative properties following ACE-1 inhibition, local activation of ACE-2/Ang1-7/Mas signaling and suppressed osteoclastogenesis seem responsible for the osteo-preservative effect of captopril, which could offers a potential therapeutic value in treatment of disabling bone and skeletal muscular diseases.


Subject(s)
Angiotensin II/analogs & derivatives , Angiotensin I/antagonists & inhibitors , Bone Density Conservation Agents/antagonists & inhibitors , Bone and Bones/drug effects , Captopril/antagonists & inhibitors , Osteoporosis, Postmenopausal/drug therapy , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/toxicity , Signal Transduction/drug effects , Administration, Oral , Angiotensin I/metabolism , Angiotensin II/administration & dosage , Angiotensin II/toxicity , Angiotensin-Converting Enzyme 2 , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/chemistry , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Biomarkers/blood , Biomarkers/urine , Bone Density Conservation Agents/administration & dosage , Bone Density Conservation Agents/therapeutic use , Bone and Bones/chemistry , Bone and Bones/diagnostic imaging , Bone and Bones/metabolism , Captopril/administration & dosage , Captopril/therapeutic use , Delayed-Action Preparations , Female , Femur , Humans , Osteolysis/chemically induced , Osteolysis/prevention & control , Osteoporosis, Postmenopausal/chemically induced , Osteoporosis, Postmenopausal/diagnostic imaging , Osteoporosis, Postmenopausal/metabolism , Ovariectomy , Peptide Fragments/administration & dosage , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/chemistry , Peptidyl-Dipeptidase A/metabolism , Rats, Wistar , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Spectrophotometry, Atomic , X-Ray Microtomography
17.
Can J Physiol Pharmacol ; 93(11): 993-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26488668

ABSTRACT

Female sex hormones are considered to reduce the risk of ischemic stroke. As a part of the renin-angiotensin system, angiotensin-(1-7) [Ang-(1-7)] has recently been reported to play a role in protecting neuronal tissues from ischemic stroke. Thus, we examined the effects of female sex hormones on the levels of Ang-(1-7) and its downstream pathways in the brain. Female rats were ovariectomized and 17ß-estradiol (17ß-EST), progesterone (PGR), or a combination of 17ß-EST plus PGR were administered. Our data demonstrated that lack of female sex hormones significantly decreased the levels of Ang-(1-7) in the cerebral cortex and hippocampal CA1 area. Also, we observed a linear relationship between cortex levels of Ang-(1-7) and plasma brain natriuretic peptide levels (as an indicator for risk of ischemic stroke). We further showed that lack of female sex hormones decreased the expression of Ang-(1-7), Mas-receptor (Mas-R), and neuronal nitric oxide synthase (nNOS). Overall, our findings show for the first time that Ang-(1-7) and Mas-R/nNOS in the cortex are influenced by circulating 17ß-EST and (or) PGR, whereas Ang-(1-7) and its pathways in the hippocampal CA1 area are primarily altered by 17ß-EST. This suggests that female sex hormones play a role in regulating the expression of Ang-(1-7) and its pathways during ischemic brain injuries.


Subject(s)
Angiotensin I/biosynthesis , Brain/metabolism , Gonadal Steroid Hormones/metabolism , Nitric Oxide Synthase Type I/biosynthesis , Peptide Fragments/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Receptors, G-Protein-Coupled/biosynthesis , Signal Transduction/physiology , Angiotensin I/antagonists & inhibitors , Animals , Brain/drug effects , Female , Gene Expression Regulation , Gonadal Steroid Hormones/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Ovariectomy , Peptide Fragments/antagonists & inhibitors , Proto-Oncogene Mas , Proto-Oncogene Proteins/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction/drug effects
18.
J Mol Med (Berl) ; 93(9): 1003-13, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25874965

ABSTRACT

UNLABELLED: Activation of the angiotensin 1-7/Mas receptor (MasR) axis counteracts angiotensin II (Ang II)-mediated cardiovascular disease. Recombinant human angiotensin-converting enzyme 2 (rhACE2) generates Ang 1-7 from Ang II. We hypothesized that the therapeutic effects of rhACE2 are dependent on Ang 1-7 action. Wild type male C57BL/6 mice (10-12 weeks old) were infused with Ang II (1.5 mg/kg/d) and treated with rhACE2 (2 mg/kg/d). The Ang 1-7 antagonist, A779 (200 ng/kg/min), was administered to a parallel group of mice. rhACE2 prevented Ang II-induced hypertrophy and diastolic dysfunction while A779 prevented these beneficial effects and precipitated systolic dysfunction. rhACE2 effectively antagonized Ang II-mediated myocardial fibrosis which was dependent on the action of Ang 1-7. Myocardial oxidative stress and matrix metalloproteinase 2 activity was further increased by Ang 1-7 inhibition even in the presence of rhACE2. Activation of Akt and endothelial nitric oxide synthase (eNOS) by rhACE2 were suppressed by the antagonism of Ang 1-7 while the activation of pathological signaling pathways was maintained. Blocking Ang 1-7 action prevents the therapeutic effects of rhACE2 in the setting of elevated Ang II culminating in systolic dysfunction. These results highlight a key cardioprotective role of Ang 1-7, and increased Ang 1-7 action represents a potential therapeutic strategy for cardiovascular diseases. KEY MESSAGES: Activation of the renin-angiotensin system (RAS) plays a key pathogenic role in cardiovascular disease. ACE2, a monocarboxypeptidase, negatively regulates pathological effects of Ang II. Antagonizing Ang 1-7 prevents the therapeutic effects of recombinant human ACE2. Our results highlight a key protective role of Ang 1-7 in cardiovascular disease.


Subject(s)
Angiotensin II/analogs & derivatives , Angiotensin I/antagonists & inhibitors , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/pharmacology , Peptidyl-Dipeptidase A/therapeutic use , Angiotensin II/metabolism , Angiotensin II/pharmacology , Angiotensin-Converting Enzyme 2 , Animals , Cardiovascular Diseases/blood , Cardiovascular Diseases/drug therapy , Humans , Male , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Nitric Oxide Synthase Type III/metabolism , Peptidyl-Dipeptidase A/blood , Proto-Oncogene Mas , Signal Transduction/drug effects
19.
Toxicon ; 98: 49-53, 2015 May.
Article in English | MEDLINE | ID: mdl-25702959

ABSTRACT

The venom of marine animals is a rich source of compounds with remarkable functional specificity and diversity. Thalassophryne nattereri is a small venomous fish inhabiting the northern and northeastern coast of Brazil, and represents a relatively frequent cause of injuries. Its venom causes severe inflammatory response followed frequently by the necrosis of the affected area. This venom presents characterized components such as proteases (Natterins 1-4) and a lectin (Nattectin) with complex effects on the human organism. A specific inhibitor of tissue kallikrein (TKI) reduces the nociception and the edema caused by the venom in mice. Our study sought to investigate the proteolytic activities against vasopeptides Angiotensin I, Angiotensin II, Angiotensin 1-9 and Bradykinin. The venom indicated angiotensin conversion against angiotensin I, as well as kininase against bradykinin. Captopril conducted the total inhibition of the converting activity, featuring the first report of ACE activity in fish venoms.


Subject(s)
Angiotensins/antagonists & inhibitors , Batrachoidiformes , Fish Venoms/chemistry , Fishes, Poisonous , Angiotensin I/antagonists & inhibitors , Angiotensin I/metabolism , Angiotensin II/metabolism , Angiotensins/metabolism , Animals , Bradykinin/antagonists & inhibitors , Bradykinin/metabolism , Brazil , Chromatography, High Pressure Liquid , Disease Models, Animal , Mice
20.
Am J Pathol ; 185(3): 729-40, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25625676

ABSTRACT

Angiotensin-(1-7) is a ligand for the Mas receptor and may protect against tissue injury associated with renin-angiotensin system activation. We determined the effects of endogenous or exogenous angiotensin-(1-7) in mice with unilateral ureteral obstruction (UUO). Mice with UUO were treated with or without the angiotensin-(1-7) antagonist A779 or with 6, 24, or 62 µg/kg per hour exogenous angiotensin-(1-7). After 10 days, kidneys were harvested for histology, immunoblots, and measurement of NADPH oxidase. Compared with controls, A779 treatment significantly increased fibronectin, transforming growth factor-ß, and α-smooth muscle actin expression in obstructed kidneys and enhanced tubulointerstitial injury, apoptosis, and NADPH oxidase. Unexpectedly, administration of angiotensin-(1-7) to mice with UUO caused injury in obstructed kidneys compared with controls and increased macrophage infiltration. In obstructed kidneys from mice with gene deletion of Mas (Mas(-/-)), apoptosis and macrophage infiltration were increased compared with wild-type mice. Angiotensin-(1-7) (but not A779) further increased apoptosis and macrophage influx in obstructed kidneys from Mas(-/-) mice, compared with untreated Mas(-/-) mice. These data indicate that endogenous angiotensin-(1-7) protects against kidney injury in UUO. In mice with or without the Mas receptor, however, delivery of exogenous angiotensin-(1-7) worsens kidney damage. The results suggest dose-dependent effects of angiotensin-(1-7) in the kidney in UUO, with endogenous angiotensin-(1-7) promoting repair pathways via interaction with Mas and higher amounts exacerbating injury.


Subject(s)
Angiotensin II/analogs & derivatives , Angiotensin I/therapeutic use , Peptide Fragments/therapeutic use , Ureter/drug effects , Ureteral Obstruction/drug therapy , Actins/metabolism , Angiotensin I/antagonists & inhibitors , Angiotensin I/pharmacology , Angiotensin II/pharmacology , Angiotensin II/therapeutic use , Animals , Fibronectins/metabolism , Male , Mice , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/pharmacology , Transforming Growth Factor beta/metabolism , Ureter/metabolism , Ureteral Obstruction/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...