Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
1.
Bioessays ; 43(3): e2000112, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33336824

RESUMEN

This renin-angiotensin system (RAS) interpretation is focused on differences in tissue dependence on RAS and on the topological hierarchy that allows mediators to act only on downstream tissues. Dependence of tissues on RAS: Tested by expectation maximization clustering of the RNA human tissue expression (https://biogps.org/). ACE and vasoconstrictive AT1R clustered with the prorenin receptor. ACE2 and dilatory MAS1 clustered with nine RAS-related genes, highly expressed in: Liver; Cardiac_Myocytes; Skeletal_Muscle; Uterus; Kidney; Lung; Small_Intestine; Smooth_Muscle. RAS and stress accumulation: While prorenin is active after binding to its receptor, binding of soluble renin increases its enzymatic activity several times. Increased renin secretion multiplies the overall capacity for producing Ang I, leading to hypertension and increased vascular resistance. Coronavirus infection and comorbidities: Cardiorespiratory failure during infection is linked to the previously altered RAS role in lungs and myocardium. Reduced vasodilation by ACE2 lead to vasoconstriction and suboptimal tissue perfusion patterns. Also see the video abstract here https://www.youtube.com/watch?v=Jf0Iped-Mws.


Asunto(s)
Enzima Convertidora de Angiotensina 2/genética , COVID-19/genética , Hipertensión/genética , Sistema Renina-Angiotensina/genética , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , Anciano , Angiotensina I/genética , Angiotensina I/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/metabolismo , COVID-19/mortalidad , COVID-19/virología , Regulación de la Expresión Génica , Humanos , Hipertensión/metabolismo , Hipertensión/mortalidad , Hipertensión/virología , Pulmón/metabolismo , Pulmón/patología , Pulmón/virología , Miocardio/metabolismo , Miocardio/patología , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 2/genética , Receptor de Angiotensina Tipo 2/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Renina/genética , Renina/metabolismo , Transducción de Señal , Análisis de Supervivencia
2.
Int J Mol Sci ; 23(24)2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36555207

RESUMEN

Thoracic aortic aneurysm (TAA) involves extracellular matrix (ECM) remodeling of the aortic wall, leading to reduced biomechanical support with risk of aortic dissection and rupture. Activation of the renin-angiotensin system, and resultant angiotensin (Ang) II synthesis, is critically involved in the onset and progression of TAA. The current study investigated the effects of angiotensin (Ang) 1-7 on a murine model of TAA. Male 8-10-week-old ApoEKO mice were infused with Ang II (1.44 mg/kg/day) and treated with Ang 1-7 (0.576 mg/kg/day). ApoEKO mice developed advanced TAA in response to four weeks of Ang II infusion. Echocardiographic and histological analyses demonstrated increased aortic dilatation, excessive structural remodelling, perivascular fibrosis, and inflammation in the thoracic aorta. Ang 1-7 infusion led to attenuation of pathological phenotypic alterations associated with Ang II-induced TAA. Smooth muscle cells (SMCs) isolated from adult murine thoracic aorta exhibited excessive mitochondrial fission, oxidative stress, and hyperproliferation in response to Ang II. Treatment with Ang 1-7 resulted in inhibition of mitochondrial fragmentation, ROS generation, and hyperproliferation. Gene expression profiling used for characterization of the contractile and synthetic phenotypes of thoracic aortic SMCs revealed preservation of the contractile phenotype with Ang 1-7 treatment. In conclusion, Ang 1-7 prevented Ang II-induced vascular remodeling and the development of TAA. Enhancing Ang 1-7 actions may provide a novel therapeutic strategy to prevent or delay the progression of TAA.


Asunto(s)
Aneurisma de la Aorta Torácica , Masculino , Animales , Ratones , Aneurisma de la Aorta Torácica/tratamiento farmacológico , Aneurisma de la Aorta Torácica/prevención & control , Aneurisma de la Aorta Torácica/genética , Angiotensina I/farmacología , Angiotensina I/genética , Fenotipo , Angiotensina II/metabolismo , Miocitos del Músculo Liso/metabolismo , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad
3.
Int J Mol Sci ; 23(14)2022 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-35887034

RESUMEN

The Renin-Angiotensin System (RAS) is expressed in the central nervous system and has important functions that go beyond blood pressure regulation. Clinical and experimental studies have suggested that alterations in the brain RAS contribute to the development and progression of neurodegenerative diseases. However, there is limited information regarding the involvement of RAS components in Huntington's disease (HD). Herein, we used the HD murine model, (BACHD), as well as samples from patients with HD to investigate the role of both the classical and alternative axes of RAS in HD pathophysiology. BACHD mice displayed worse motor performance in different behavioral tests alongside a decrease in the levels and activity of the components of the RAS alternative axis ACE2, Ang-(1-7), and Mas receptors in the striatum, prefrontal cortex, and hippocampus. BACHD mice also displayed a significant increase in mRNA expression of the AT1 receptor, a component of the RAS classical arm, in these key brain regions. Moreover, patients with manifest HD presented higher plasma levels of Ang-(1-7). No significant changes were found in the levels of ACE, ACE2, and Ang II. Our findings provided the first evidence that an imbalance in the RAS classical and counter-regulatory arms may play a role in HD pathophysiology.


Asunto(s)
Angiotensina I , Enzima Convertidora de Angiotensina 2 , Enfermedad de Huntington , Fragmentos de Péptidos , Receptor de Angiotensina Tipo 1 , Sistema Renina-Angiotensina , Angiotensina I/genética , Angiotensina I/metabolismo , Angiotensina II/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Animales , Modelos Animales de Enfermedad , Humanos , Enfermedad de Huntington/genética , Ratones , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Sistema Renina-Angiotensina/genética , Sistema Renina-Angiotensina/fisiología
4.
Clin Sci (Lond) ; 135(18): 2197-2216, 2021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34494083

RESUMEN

Activation of the angiotensin (Ang)-converting enzyme (ACE) 2/Ang-(1-7)/MAS receptor pathway of the renin-angiotensin system (RAS) induces protective mechanisms in different diseases. Herein, we describe the cardiovascular phenotype of a new transgenic rat line (TG7371) that expresses an Ang-(1-7)-producing fusion protein. The transgene-specific mRNA and the corresponding protein were shown to be present in all evaluated tissues of TG7371 with the highest expression in aorta and brain. Plasma Ang-(1-7) levels, measured by radioimmunoassay (RIA) were similar to control Sprague-Dawley (SD) rats, however high Ang-(1-7) levels were found in the hypothalamus. TG7371 showed lower baseline mean arterial pressure (MAP), assessed in conscious or anesthetized rats by telemetry or short-term recordings, associated with increased plasma atrial natriuretic peptide (ANP) and higher urinary sodium concentration. Moreover, evaluation of regional blood flow and hemodynamic parameters with fluorescent microspheres showed a significant increase in blood flow in different tissues (kidneys, mesentery, muscle, spleen, brown fat, heart and skin), with a resulting decrease in total peripheral resistance (TPR). TG7371 rats, on the other hand, also presented increased cardiac and global sympathetic tone, increased plasma vasopressin (AVP) levels and decreased free water clearance. Altogether, our data show that expression of an Ang-(1-7)-producing fusion protein induced a hypotensive phenotype due to widespread vasodilation and consequent fall in peripheral resistance. This phenotype was associated with an increase in ANP together with an increase in AVP and sympathetic drive, which did not fully compensate the lower blood pressure (BP). Here we present the hemodynamic impact of long-term increase in tissue expression of an Ang-(1-7)-fusion protein and provide a new tool to investigate this peptide in different pathophysiological conditions.


Asunto(s)
Angiotensina I/metabolismo , Sistema Cardiovascular/metabolismo , Hemodinámica , Hipertensión/prevención & control , Fragmentos de Péptidos/metabolismo , Sistema Nervioso Simpático/metabolismo , Angiotensina I/genética , Animales , Arginina Vasopresina/metabolismo , Factor Natriurético Atrial/metabolismo , Velocidad del Flujo Sanguíneo , Presión Sanguínea , Sistema Cardiovascular/fisiopatología , Modelos Animales de Enfermedad , Genotipo , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Hemodinámica/genética , Hipertensión/genética , Hipertensión/metabolismo , Hipertensión/fisiopatología , Masculino , Fragmentos de Péptidos/genética , Fenotipo , Ratas Sprague-Dawley , Ratas Transgénicas , Proteínas Recombinantes de Fusión/metabolismo , Flujo Sanguíneo Regional , Sistema Nervioso Simpático/fisiopatología , Factores de Tiempo , Resistencia Vascular
5.
Rev Med Virol ; 30(5): e2119, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32584474

RESUMEN

The novel coronavirus disease 2019 (COVID-19) is rapidly expanding and causing many deaths all over the world with the World Health Organization (WHO) declaring a pandemic in March 2020. Current therapeutic options are limited and there is no registered and/or definite treatment or vaccine for this disease or the causative infection, severe acute respiratory coronavirus 2 syndrome (SARS-CoV-2). Angiotensin-converting enzyme 2 (ACE2), a part of the renin-angiotensin system (RAS), serves as the major entry point into cells for SARS-CoV-2 which attaches to human ACE2, thereby reducing the expression of ACE2 and causing lung injury and pneumonia. Vitamin D, a fat-soluble-vitamin, is a negative endocrine RAS modulator and inhibits renin expression and generation. It can induce ACE2/Ang-(1-7)/MasR axis activity and inhibits renin and the ACE/Ang II/AT1R axis, thereby increasing expression and concentration of ACE2, MasR and Ang-(1-7) and having a potential protective role against acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). Therefore, targeting the unbalanced RAS and ACE2 down-regulation with vitamin D in SARS-CoV-2 infection is a potential therapeutic approach to combat COVID-19 and induced ARDS.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Betacoronavirus/patogenicidad , Infecciones por Coronavirus/tratamiento farmacológico , Peptidil-Dipeptidasa A/genética , Neumonía Viral/tratamiento farmacológico , Receptores Virales/genética , Vitamina D/uso terapéutico , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/virología , Angiotensina I/genética , Angiotensina I/metabolismo , Enzima Convertidora de Angiotensina 2 , Betacoronavirus/genética , Betacoronavirus/metabolismo , COVID-19 , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Pandemias , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/patología , Neumonía Viral/virología , Unión Proteica , Proto-Oncogenes Mas , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Receptores Virales/antagonistas & inhibidores , Receptores Virales/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo
6.
Int J Mol Sci ; 22(11)2021 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-34070749

RESUMEN

Atherosclerosis and nonalcoholic fatty liver disease are leading causes of morbidity and mortality in the Western countries. The renin-angiotensin system (RAS) with its two main opposing effectors, i.e., angiotensin II (Ang II) and Ang-(1-7), is widely recognized as a major regulator of cardiovascular function and body metabolic processes. Angiotensin-converting enzyme 2 (ACE2) by breaking-down Ang II forms Ang-(1-7) and thus favors Ang-(1-7) actions. Therefore, the aim of our study was to comprehensively evaluate the influence of prolonged treatment with ACE2 activator, diminazene aceturate (DIZE) on the development of atherosclerotic lesions and hepatic steatosis in apoE-/- mice fed a high-fat diet (HFD). We have shown that DIZE stabilized atherosclerotic lesions and attenuated hepatic steatosis in apoE-/- mice fed an HFD. Such effects were associated with decreased total macrophages content and increased α-smooth muscle actin levels in atherosclerotic plaques. Moreover, DIZE changed polarization of macrophages towards increased amount of anti-inflammatory M2 macrophages in the atherosclerotic lesions. Interestingly, the anti-steatotic action of DIZE in the liver was related to the elevated levels of HDL in the plasma, decreased levels of triglycerides, and increased biosynthesis and concentration of taurine in the liver of apoE-/- mice. However, exact molecular mechanisms of both anti-atherosclerotic and anti-steatotic actions of DIZE require further investigations.


Asunto(s)
Enzima Convertidora de Angiotensina 2/genética , Aterosclerosis/tratamiento farmacológico , Diminazeno/análogos & derivados , Hígado Graso/tratamiento farmacológico , Placa Aterosclerótica/tratamiento farmacológico , Taurina/biosíntesis , Angiotensina I/genética , Angiotensina I/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/etiología , Aterosclerosis/genética , Aterosclerosis/patología , Dieta Alta en Grasa , Diminazeno/farmacología , Modelos Animales de Enfermedad , Hígado Graso/etiología , Hígado Graso/genética , Hígado Graso/patología , Femenino , Regulación de la Expresión Génica , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Activación de Macrófagos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Placa Aterosclerótica/etiología , Placa Aterosclerótica/genética , Placa Aterosclerótica/patología , Células THP-1 , Taurina/agonistas
7.
Mol Hum Reprod ; 26(6): 367-373, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32365180

RESUMEN

The 2019 novel coronavirus (2019-nCoV) appeared in December 2019 and then spread throughout the world rapidly. The virus invades the target cell by binding to angiotensin-converting enzyme (ACE) 2 and modulates the expression of ACE2 in host cells. ACE2, a pivotal component of the renin-angiotensin system, exerts its physiological functions by modulating the levels of angiotensin II (Ang II) and Ang-(1-7). We reviewed the literature that reported the distribution and function of ACE2 in the female reproductive system, hoping to clarify the potential harm of 2019-nCoV to female fertility. The available evidence suggests that ACE2 is widely expressed in the ovary, uterus, vagina and placenta. Therefore, we believe that apart from droplets and contact transmission, the possibility of mother-to-child and sexual transmission also exists. Ang II, ACE2 and Ang-(1-7) regulate follicle development and ovulation, modulate luteal angiogenesis and degeneration, and also influence the regular changes in endometrial tissue and embryo development. Taking these functions into account, 2019-nCoV may disturb the female reproductive functions through regulating ACE2.


Asunto(s)
Betacoronavirus/patogenicidad , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/patología , Genitales Femeninos/virología , Pandemias , Peptidil-Dipeptidasa A/genética , Neumonía Viral/epidemiología , Neumonía Viral/patología , Glicoproteína de la Espiga del Coronavirus/genética , Adulto , Angiotensina I/genética , Angiotensina I/metabolismo , Angiotensina II/genética , Angiotensina II/metabolismo , Enzima Convertidora de Angiotensina 2 , COVID-19 , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/transmisión , Femenino , Regulación de la Expresión Génica , Genitales Femeninos/patología , Interacciones Huésped-Patógeno/genética , Humanos , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/diagnóstico , Neumonía Viral/transmisión , Embarazo , Unión Proteica , Receptores Virales/genética , Receptores Virales/metabolismo , Sistema Renina-Angiotensina/genética , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/metabolismo
8.
Stress ; 20(2): 189-196, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28288545

RESUMEN

Angiotensin II (Ang II) acts as a pro-stress hormone, while other evidence indicates that angiotensin-(1-7) [Ang-(1-7)] attenuates physiological responses to emotional stress. To further test this hypothesis, in groups of 5-6 rats we evaluated autonomic, cardiovascular and behavioral parameters in male Sprague-Dawley (SD) and transgenic TGR(A1-7)3292 (TG) rats chronically overexpressing Ang-(1-7). Compared to SD rats, TG rats showed reduced baseline heart rate (HR; SD 380 ± 16 versus TG 329 ± 9 beats per minute (bpm), mean ± standard error of mean, p < .05) and renal sympathetic discharge (SD 138 ± 4 versus TG 117 ± 5 spikes/second, p < .05). TG rats had an attenuated tachycardic response to acute air-puff stress (ΔHR: SD 51 ± 20 versus TG 1 ± 3 bpm; p < .05), which was reversed by intracerebroventricular injection of the Mas receptor antagonist, A-779 (ΔHR: SD 51 ± 20 versus TG 63 ± 15 bpm). TG rats showed less anxious behavior on the elevated plus maze, as revealed by more entries into open arms (SD 2 ± 2 versus TG 47 ± 5% relative to total entries; p < .05), and more time spent in the open arms (SD 5 ± 4 versus TG 53 ± 9% relative to total time, p < .05). By contrast with SD rats, diazepam (1.5 mg/kg, intraperitoneally) did not further reduce anxious behavior in TG rats, indicating a ceiling anxiolytic effect of Ang-(1-7) overexpression. Ang-(1-7) concentrations in hypothalamus and plasma, measured by mass spectrometry were two- and three-fold greater, respectively, in TG rats than in SD rats. Hence, increased endogenous Ang-(1-7) levels in TG rats diminishes renal sympathetic outflow and attenuates cardiac reactivity to emotional stress, which may be via central Mas receptors, and reduces anxious behavior. Lay summaryWe used a genetically modified rat model that produces above normal amounts of a peptide hormone called angiotensin-(1-7) to test whether this peptide can reduce some of the effects of stress. We found that angiotensin-(1-7), acting in the brain, can reduce anxiety and reduce the increase in heart rate associated with emotional stress. These findings may provide a lead for design of new drugs to reduce stress.


Asunto(s)
Angiotensina I/genética , Ansiedad/genética , Frecuencia Cardíaca/fisiología , Fragmentos de Péptidos/genética , Estrés Psicológico/fisiopatología , Angiotensina II/análogos & derivados , Angiotensina II/farmacología , Animales , Animales Modificados Genéticamente , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Fragmentos de Péptidos/farmacología , Ratas , Ratas Sprague-Dawley
9.
J Cell Physiol ; 230(3): 691-701, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25200929

RESUMEN

Angiotensin (Ang) II plays an important role in the process of endothelial dysfunction in acute lung injury (ALI) and is degraded by angiotensin-converting enzyme2 (ACE2). However, treatments that target ACE2 to injured endothelium and promote endothelial repair of ALI are lacking. Mesenchymal stem cells (MSCs) are capable of homing to the injured site and delivering a protective gene. Our study aimed to evaluate the effects of genetically modified MSCs, which overexpress the ACE2 protein in a sustained manner via a lentiviral vector, on Ang II production in endothelium and in vitro repair of lipopolysaccharide (LPS)-induced endothelial injury. We found that the efficiency of lentiviral vector transduction of MSCs was as high as 97.8% and was well maintained over 30 passages. MSCs modified with ACE2 showed a sustained high expression of ACE2 mRNA and protein. The modified MSCs secreted soluble ACE2 protein into the culture medium, which reduced the concentration of Ang II and increased the production of Ang 1-7. MSCs modified with ACE2 were more effective at restoring endothelial function than were unmodified MSCs, as shown by the enhanced survival of endothelial cells; the downregulated production of inflammatory mediators, including ICAM-1, VCAM-1, TNF-α, and IL-6; reduced paracellular permeability; and increased expression of VE-cadherin. These data demonstrate that MSCs modified to overexpress the ACE2 gene can produce biologically active ACE2 protein over a sustained period of time and have an enhanced ability to promote endothelial repair after LPS challenge. These results encourage further testing of the beneficial effects of ACE2-modified MSCs in an ALI animal model.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Angiotensina II/metabolismo , Células Madre Mesenquimatosas/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/patología , Angiotensina I/genética , Angiotensina II/genética , Enzima Convertidora de Angiotensina 2 , Animales , Células Endoteliales/metabolismo , Células Endoteliales/patología , Terapia Genética , Células HEK293 , Humanos , Lipopolisacáridos/toxicidad , Células Madre Mesenquimatosas/citología , Ratones , Fragmentos de Péptidos/genética , Peptidil-Dipeptidasa A/genética , Sistema Renina-Angiotensina
10.
Pancreatology ; 15(6): 647-53, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26444748

RESUMEN

BACKGROUND: The objective of this study was to determine if RAS bioactive enzymes and peptides are perturbed in acute pancreatitis and associated lung injury. METHODS: The intervention group of mice were treated with ten hourly intraperitoneal (i.p.) injections of caerulein (50 µg/kg) to induce acute pancreatitis. Animals were euthanized, samples of pancreas, lung and blood were collected, and plasma was prepared and stored for subsequent analysis. ACE and ACE2 activities were determined by spectrofluorometric assay. ACE, ACE2, Ang II and Ang-(1-7) levels were quantified by ELISA. RESULTS: There was a significant decrease in ACE2 enzymatic activity in pancreatic and lung tissues of mice with acute pancreatitis. In contrast, there were no significant changes in measured levels of ACE and ACE2 in the pancreas, and lung or activity of ACE in pancreatic and lung tissue following acute pancreatitis. There were no significant differences in the activities and levels of circulating ACE and ACE2 following acute pancreatitis. The ACE to ACE2 activity ratio was markedly increased in pancreatic and lung tissues of mice with acute pancreatitis. No significant changes were observed in the levels of Ang II except for a decrease in lung tissue. No changes were observed in Ang-(1-7) levels in pancreas, lung and plasma between the groups. The Ang II to Ang-(1-7) ratio was increased in the pancreas but was decreased in the lung following caerulein treatment. CONCLUSION: These data suggest dysregulation of RAS in acute pancreatitis as evidenced by altered Ang II/Ang-(1-7) levels induced by the imbalance of ACE/ACE2 activity.


Asunto(s)
Angiotensina II/metabolismo , Ceruletida/toxicidad , Pancreatitis/inducido químicamente , Pancreatitis/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Angiotensina I/genética , Angiotensina I/metabolismo , Angiotensina II/sangre , Angiotensina II/genética , Enzima Convertidora de Angiotensina 2 , Animales , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Distribución Aleatoria , Sistema Renina-Angiotensina/fisiología
11.
Inflamm Res ; 64(3-4): 253-60, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25721616

RESUMEN

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.


Asunto(s)
Angiotensina II/fisiología , Angiotensina I/fisiología , Aterosclerosis/prevención & control , Endotelio Vascular/fisiología , Inflamación/prevención & control , Fragmentos de Péptidos/fisiología , Peptidil-Dipeptidasa A/fisiología , Transducción de Señal/fisiología , Angiotensina I/genética , Enzima Convertidora de Angiotensina 2 , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/fisiopatología , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Quimiocina CCL2/fisiología , Modelos Animales de Enfermedad , Selectina E/fisiología , Endotelio Vascular/citología , Técnicas de Transferencia de Gen , Humanos , Técnicas In Vitro , Inflamación/fisiopatología , Ratones , Fragmentos de Péptidos/genética , Peptidil-Dipeptidasa A/genética , Molécula 1 de Adhesión Celular Vascular/fisiología
12.
Mol Vis ; 20: 1443-55, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25352750

RESUMEN

PURPOSE: Recent studies have provided evidence that a local renin-angiotensin system (RAS) exists in the retina and plays an important role in retinal neurovascular function. We have recently shown that increased expression of ACE2 and angiotensin (1-7) [Ang (1-7)], two components of the protective axis of the RAS, in the retina via adeno-associated virus (AAV)-mediated gene delivery, conferred protection against diabetes-induced retinopathy. We hypothesized that the protective molecular and cellular mechanisms of Ang (1-7) are mediated by its receptor, Mas, and the expression level and cellular localization dictate the response to Ang (1-7) and activation of subsequent protective signaling pathways. We tested this hypothesis by examining the expression and cellular localization of the Mas receptor in adult and developing mouse retinas. METHODS: The cellular localization of the Mas receptor protein was determined with immunofluorescence of the eyes of adult and postnatal day 1 (P1), P5, P7, P15, and P21 mice using the Mas receptor-specific antibody, and mRNA was detected with in situ hybridization of paraffin-embedded sections. Western blotting and real-time reverse-transcription (RT)-PCR analysis were performed to determine the relative levels of the Mas protein and mRNA in adult and developing retinas, as well as in cultured retinal Müller glial and RPE cells. RESULTS: In the adult eye, the Mas receptor protein was abundantly present in retinal ganglion cells (RGCs) and photoreceptor cells; a lower level of expression was observed in endothelial cells, Müller glial cells, and other neurons in the inner nuclear layer of the retina. In the developing retina, Mas receptor mRNA and protein expression was detected in the inner retina at P1, and the expression levels increased with age to reach the adult level and pattern by P15. In the adult mouse retina, Mas receptor mRNA was expressed at a much higher level when compared to angiotensin II (Ang II) type I (AT1R) and type II (AT2R) receptor mRNA. CONCLUSIONS: The Mas receptor is expressed in developing and adult mouse retinas, and is more abundant in retinal neurons than in endothelial and Müller glial cells. These observations suggest that Mas receptor-mediated signaling may play important roles that extend beyond mediating the vascular effects of Ang (1-7) in developing and adult retinas. In addition, the relatively high expression of the Mas receptor when compared to AT1R suggests that they may play a more important role in maintaining normal retinal physiology than previously considered.


Asunto(s)
Células Fotorreceptoras de Vertebrados/metabolismo , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 2/genética , Receptores Acoplados a Proteínas G/genética , Células Ganglionares de la Retina/metabolismo , Angiotensina I/genética , Angiotensina I/metabolismo , Enzima Convertidora de Angiotensina 2 , Animales , Animales Recién Nacidos , Células Cultivadas , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Neuroglía/metabolismo , Neuroglía/ultraestructura , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Células Fotorreceptoras de Vertebrados/ultraestructura , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Cross-Talk , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 2/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sistema Renina-Angiotensina/genética , Células Ganglionares de la Retina/ultraestructura , Pigmentos Retinianos/metabolismo
13.
J Gastroenterol Hepatol ; 29(6): 1328-37, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24628092

RESUMEN

BACKGROUND AND AIM: Ulinastatin is a drug used effectively to alleviate symptoms and improve the pathophysiology of various types of pancreatitis. However, the molecular mechanism responsible for its action remains unknown. Therefore, we further explore the therapeutic effects of ulinastatin and investigate possible molecular pathways modulated by this drug in the development of severe acute pancreatitis (SAP). METHODS: SAP mouse model was created by administering intraperitoneal injections of cerulein and lipopolysaccharide. Pancreatic injury was assessed by performing biochemical and histological assays and by measuring the inflammatory response of the pancreas. Specifically, we examined changes in the expression of components of the rennin-angiotensin system (RAS), including angiotensin-converting enzyme (ACE)-angiotensin II (Ang II)-angiotensin type 1 receptor (AT-1R), and ACE2-Ang-(1-7)-Mas receptor. RESULTS: When SAP mouse models were treated with ulinastatin at a dosage of 50,000 U/kg body weight, we found, through biochemical and histopathological analyses, that the pancreatic injury was significantly ameliorated. Administration of ulinastatin to SAP mice led to increased expression of ACE2, Ang-(1-7), and Mas receptor, decreased expression of serum Ang II and pancreatic AT-1R, and no alterations in the expression of pancreatic ACE and Ang II when compared to cerulein-treated control group that did not receive ulinastatin. CONCLUSIONS: This study shows that ulinastatin has differential effects on the two axes of the RAS during SAP. Our results further suggest that upregulation of components of the ACE2-Ang-(1-7)-Mas pathway might be an important mechanism contributing to the therapeutic role of ulinastatin in alleviating pancreatitis-associated symptoms.


Asunto(s)
Glicoproteínas/administración & dosificación , Terapia Molecular Dirigida , Pancreatitis/tratamiento farmacológico , Pancreatitis/genética , Sistema Renina-Angiotensina/fisiología , Enfermedad Aguda , Angiotensina I/genética , Angiotensina I/metabolismo , Angiotensina II/genética , Angiotensina II/metabolismo , Enzima Convertidora de Angiotensina 2 , Animales , Ceruletida , Modelos Animales de Enfermedad , Expresión Génica , Lipopolisacáridos , Ratones Endogámicos C57BL , Pancreatitis/inducido químicamente , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Estudios Prospectivos , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Sistema Renina-Angiotensina/genética , Índice de Severidad de la Enfermedad
14.
Clin Exp Pharmacol Physiol ; 41(12): 1023-30, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25225013

RESUMEN

Adenosine triphosphate-binding cassette transporter A1 (ABCA1) and ABCG1 play crucial roles in reverse cholesterol transport, and have anti-atherosclerosis effects, and liver X receptor alpha (LXRα) can stimulate cholesterol efflux through these transporters. Angiotensin (Ang)-(1-7) can protect endothelial cells, inhibit smooth muscle cell growth, ameliorate inflammation and exert anti-atherosclerotic effects. In the present study, we attempted to clarify the effect of Ang-(1-7) on expression of ABCA1 and ABCG1, and explored the role of LXRα in the regulation of ABCA1 and ABCG1 in THP-1 macrophages that had been incubated with angiotensin-II (AngII). Ang-(1-7) increased ABCA1 and ABCG1 expression in a concentration-dependent manner at both the mRNA and protein levels, promoted cholesterol efflux, and decreased cholesterol content in THP-1 macrophages treated with AngII. Furthermore, Ang-(1-7) upregulated the expression of LXRα in a concentration-dependent manner in these cells. LXRα small interfering RNA, as well as the Mas receptor antagonist A-779, completely abolished these effects of Ang-(1-7). In summary, Ang-(1-7) upregulates ABCA1 and ABCG1 expression in THP-1 macrophages treated with AngII through the Mas receptor, via the LXRα pathway. This novel insight into the molecular mechanism underlying Ang-(1-7) and AngII interaction could prove useful for developing new strategies for treatment of cardiovascular diseases.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/genética , Angiotensina I/genética , Receptores Nucleares Huérfanos/genética , Fragmentos de Péptidos/genética , Proteínas Proto-Oncogénicas/genética , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/genética , Regulación hacia Arriba/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Aterosclerosis/genética , Línea Celular Tumoral , Colesterol/metabolismo , Humanos , Receptores X del Hígado , Macrófagos/metabolismo , Proteínas de Transporte de Membrana/genética , Proto-Oncogenes Mas , ARN Mensajero/genética , ARN Interferente Pequeño/genética
15.
Zhongguo Zhong Yao Za Zhi ; 39(8): 1468-72, 2014 Apr.
Artículo en Zh | MEDLINE | ID: mdl-25039184

RESUMEN

OBJECTIVE: To investigate the effect of tanshinone II(A) on the expression of different components in the renin-angiotensin system of left ventricles of renal hypertensive rats. METHOD: The renal hypertension model was established in rats by the two-kidney-one-clip (2K1C) method. In the experiment, all of the rats were randomly divided into four groups (n = 15 per group) before the operation: the sham-operated (Sham) group, the hypertensive model (Model) group, the low-dose tanshinone II(A) group and the high-dose tanshinone II(A) group. At 5 week after the renal artery narrowing, the third and fourth groups were administered with 35 mg kg(-1) x d(-1) and 70 mg x kg(-1) x d(-1) of tanshinone II(A), respectively. The blood pressure in rats was determined by the standard tail-cuff method in each week after the operation. After the drug treatment for 8 weeks, all the rats were put to death, and their left ventricles were separated to determine the ratio of left ventricle weight to body weight (LVW/BW), the myocardial collagen content, and the expressions of different components in myocardial RAS, including angiotensin converting enzyme (ACE), angiotensin converting enzyme 2 (ACE2), angiotensin 1-type receptor (AT1R), Mas receptor mRNA expression and angiotensin II (Ang II) and angiotensin (1-7) [Ang (1-7)] content. RESULT: Compared with the sham group, the hypertensive model group exhibited a markable increase in the content of Ang II and Ang (1-7) and the mRNA expressions of ACE, ACE2, AT1R and Mas (P < 0.01). However, the treatment with tanshinone II(A) showed the does dependence, inhibited left ventricle hypertrophy, decreased myocardial Ang II content and the mRNA expression of ACE and AT, R in renal hypertensive rats (P < 0. 01) , further increased the myocardial Ang (1-7) content and the mRNA expression of ACE2 and Mas (P < 0.01) , but without any change in the blood pressure of hypertensive rats. CONCLUSION: The treatment with tanshinone II(A) could inhibit left ventricle hypertrophy of renal hypertensive rats. Its mechanism may be partially related to the expression of different components in the renin-angiotensin system for regulating myocardial tissues.


Asunto(s)
Abietanos/administración & dosificación , Ventrículos Cardíacos/efectos de los fármacos , Hipertensión/tratamiento farmacológico , Sistema Renina-Angiotensina/efectos de los fármacos , Angiotensina I/genética , Angiotensina I/metabolismo , Angiotensina II/genética , Angiotensina II/metabolismo , Enzima Convertidora de Angiotensina 2 , Animales , Presión Sanguínea/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Humanos , Hipertensión/genética , Hipertensión/metabolismo , Hipertensión/fisiopatología , Masculino , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Ratas , Ratas Sprague-Dawley , Renina/genética , Renina/metabolismo
16.
Pflugers Arch ; 465(1): 79-85, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23463883

RESUMEN

The renin­angiotensin system (RAS) has recently been extended by the addition of a novel axis consisting of the angiotensin-converting enzyme 2 (ACE2), the heptapeptide angiotensin (1­7) (Ang-(1­7)), and the G protein-coupled receptor Mas. ACE2 converts the vasoconstrictive and pro-oxidative peptide angiotensin II (Ang II) into Ang-(1­7) which exerts vasodilatory and antioxidative effects via its receptor Mas. Thereby, ACE2 regulates the local actions of the RAS in cardiovascular tissues and the ACE2/Ang-(1­7)/Mas axis exerts protective actions in hypertension, diabetes, and other cardiovascular disorders. Consequently, this novel RAS axis represents a promising therapeutic target for cardiovascular and metabolic diseases.


Asunto(s)
Angiotensina I/metabolismo , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Angiotensina I/genética , Angiotensina III/genética , Angiotensina III/metabolismo , Enzima Convertidora de Angiotensina 2 , Animales , Encéfalo/metabolismo , Expresión Génica , Humanos , Riñón/metabolismo , Pulmón/metabolismo , Miocardio/metabolismo , Fragmentos de Péptidos/genética , Peptidil-Dipeptidasa A/genética , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/genética , Receptores Acoplados a Proteínas G/genética , Sistema Renina-Angiotensina
17.
Mol Ther ; 20(1): 28-36, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21792177

RESUMEN

Despite evidence that hyperactivity of the vasodeleterious axis (ACE/angiotensin II (Ang II)/AT1 receptor) of the renin-angiotensin system (RAS) is associated with the pathogenesis of diabetic retinopathy (DR) use of the inhibitors of this axis has met with limited success in the control of this pathophysiology. We investigated the hypothesis that enhancing the local activity of the recently established protective axis of the RAS, ACE2/Ang-(1-7), using adeno-associated virus (AAV)-mediated gene delivery of ACE2 or Ang-(1-7) would confer protection against diabetes-induced retinopathy. Genes expressing ACE2 and Ang-(1-7) were cloned in AAV vector. The effects of ocular AAV-ACE2/Ang-(1-7) gene transfer on DR in diabetic eNOS(-/-) mice and Sprague-Dawley (SD) rats were examined. Diabetes was associated with approximately tenfold and greater than threefold increases in the ratios of ACE/ACE2 and AT1R/Mas mRNA levels in the retina respectively. Intraocular administration of AAV-ACE2/Ang-(1-7) resulted in significant reduction in diabetes-induced retinal vascular leakage, acellular capillaries, infiltrating inflammatory cells and oxidative damage in both diabetic mice and rats. Our results demonstrate that DR is associated with impaired balance of retinal RAS. Increased expression of ACE2/Ang-(1-7) overcomes this imbalance and confers protection against DR. Thus, strategies enhancing the protective ACE2/Ang-(1-7) axis of RAS in the eye could serve as a novel therapeutic target for DR.


Asunto(s)
Angiotensina I/genética , Dependovirus/genética , Retinopatía Diabética/terapia , Vectores Genéticos/administración & dosificación , Fragmentos de Péptidos/genética , Peptidil-Dipeptidasa A/genética , Angiotensina I/metabolismo , Enzima Convertidora de Angiotensina 2 , Animales , Dependovirus/metabolismo , Retinopatía Diabética/genética , Retinopatía Diabética/metabolismo , Modelos Animales de Enfermedad , Activación Enzimática/genética , Expresión Génica , Orden Génico , Terapia Genética , Inyecciones Intravítreas , Masculino , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo III/genética , Estrés Oxidativo , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Ratas , Ratas Sprague-Dawley , Sistema Renina-Angiotensina/genética , Retina/metabolismo , Vasos Retinianos/metabolismo , Vasos Retinianos/patología
18.
Poult Sci ; 92(6): 1492-7, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23687144

RESUMEN

This study explored the effect of imidapril on the right ventricular remodeling induced by low ambient temperature in broiler chickens. Twenty-four broiler chickens were randomly divided into 3 groups (n = 8), including the control group, low temperature group, and imidapril group. Chickens in the control group were raised at normal temperature, whereas chickens in the low temperature group and imidapril group were exposed to low ambient temperature (12 to 18°C) from 14 d of age until 45 d of age. At the same time, chickens in the imidapril group were gavaged with imidapril at 3 mg/kg once daily for 30 d. The thickness of the right ventricular wall was observed with echocardiography. The BW and wet lung weight as well as weight of right and left ventricles and ventricular septum were measured. Both wet lung weight index and right ventricular hypertrophy index were calculated. Pulmonary arterial systolic pressure was assessed according to echocardiography. The expression of ACE and ACE2 mRNA in the right ventricular myocardial tissue was quantified by real-time PCR. Proliferating cell nuclear antigen-positive cells were detected by immunohistostaining. The concentration of angiotensin (Ang) II and Ang (1-7) in the right ventricular myocardial tissue was measured with ELISA. The results showed that right ventricular hypertrophy index, wet lung weight index, pulmonary arterial systolic pressure, expression of ACE mRNA in the right ventricular tissue, Ang II concentration, and the thickness of the right ventricular wall in the low temperature group increased significantly compared with those in the control group and imidapril group. The ACE2 mRNA expression increased 36%, whereas Ang (1-7) concentration decreased significantly in the low temperature group compared with that in the control group and imidapril group. In conclusion, imidapril inhibits right ventricular remodeling induced by low ambient temperature in broiler chickens.


Asunto(s)
Pollos , Frío , Ventrículos Cardíacos/patología , Vivienda para Animales , Imidazolidinas/farmacología , Remodelación Ventricular/efectos de los fármacos , Angiotensina I/genética , Angiotensina I/metabolismo , Angiotensina II/genética , Angiotensina II/metabolismo , Enzima Convertidora de Angiotensina 2 , Crianza de Animales Domésticos , Animales , Regulación de la Expresión Génica , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo
19.
Patol Fiziol Eksp Ter ; (4): 46-50, 2013.
Artículo en Ruso | MEDLINE | ID: mdl-24640774

RESUMEN

The aim of this study was to estimate the role of matrix metalloproteinases activity (MMP-9, TIMP-1) and gene polymorphism at the patients with metabolic syndrome in combination with paroxysmal form of atrial fibrillation. The 60 patients with metabolic syndrome in combination with paroxysmal form of atrial fibrillation were investigated with estimation of serum levels of MMP-9, TIMP-1 and mutation of genes of angiotenzinogen 1 Thr 174 Met, angiotenzinogen 2 Met 235 Thr and apolipoprorein C3 C3238 G. In patients with metabolic syndrome in combination with paroxysmal form of atrial fibrillation the significant increase synthesis of MMP-9 level can lead to increase of atrial fibrillation paroxysms because of increase synthesis of collagen tissue in myocard and vessel walls. Also the significant high incidence of angiotenzinogen 2 Met 235 Thr mutation was established.


Asunto(s)
Fibrilación Atrial/sangre , Metaloproteinasa 9 de la Matriz/sangre , Síndrome Metabólico/complicaciones , Inhibidor Tisular de Metaloproteinasa-1/sangre , Adulto , Angiotensina I/genética , Angiotensina II/genética , Apolipoproteínas C/genética , Fibrilación Atrial/etiología , Fibrilación Atrial/genética , Estudios de Casos y Controles , Humanos , Masculino , Persona de Mediana Edad , Mutación , Polimorfismo Genético
20.
Am J Physiol Endocrinol Metab ; 303(5): E669-81, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22811473

RESUMEN

The inhibitory effects of the angiotensin-converting enzyme (ACE)-ANG II-angiotensin type 1 (AT1) receptor axis on jejunal glucose uptake and the reduced expression of this system in type 1 diabetes mellitus (T1DM) have been documented previously. The ACE2-ANG-(1-7)-Mas receptor axis is thought to oppose the actions of the ACE-ANG II-AT1 receptor axis in heart, liver, and kidney. However, the possible involvement of the ACE2-ANG-(1-7)-Mas receptor system on enhanced jejunal glucose transport in T1DM has yet to be determined. Rat everted jejunum and Caco-2 cells were used to determine the effects of ANG-(1-7) on glucose uptake and to study the ACE2-ANG-(1-7)-Mas receptor signaling pathway. Expression of target gene and protein in jejunal enterocytes and human Caco-2 cells were quantified using real-time PCR and Western blotting. T1DM increased jejunal protein and mRNA expression of ACE2 (by 59 and 173%, respectively) and Mas receptor (by 55 and 100%, respectively) in jejunum. One millimolar ANG-(1-7) reduced glucose uptake in jejunum and Caco-2 cells by 30.6 and 30.3%, respectively, effects that were abolished following addition of 1 µM A-779 (a Mas receptor blocker) or 1 µM GF-109203X (protein kinase C inhibitor) to incubation buffer for jejunum or Caco-2 cells, respectively. Finally, intravenous treatment of animals with ANG-(1-7) significantly improved oral glucose tolerance in T1DM but not control animals. In conclusion, enhanced activity of the ACE2-ANG-(1-7)-Mas receptor axis in jejunal enterocytes is likely to moderate the T1DM-induced increase in jejunal glucose uptake resulting from downregulation of the ACE-ANG II-AT1 receptor axis. Therefore, altered activity of both ACE and ACE2 systems during diabetes will determine the overall rate of glucose transport across the jejunal epithelium.


Asunto(s)
Angiotensina I/metabolismo , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Enterocitos/metabolismo , Yeyuno/metabolismo , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Regulación hacia Arriba , Angiotensina I/administración & dosificación , Angiotensina I/genética , Angiotensina I/uso terapéutico , Enzima Convertidora de Angiotensina 2 , Animales , Transporte Biológico/efectos de los fármacos , Células CACO-2 , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Enterocitos/efectos de los fármacos , Enterocitos/patología , Glucosa/metabolismo , Humanos , Hiperglucemia/prevención & control , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/metabolismo , Hipoglucemiantes/uso terapéutico , Técnicas In Vitro , Inyecciones Intravenosas , Yeyuno/efectos de los fármacos , Yeyuno/patología , Masculino , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/uso terapéutico , Peptidil-Dipeptidasa A/genética , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA