Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
China Journal of Chinese Materia Medica ; (24): 461-468, 2022.
Article in Chinese | WPRIM | ID: wpr-927990

ABSTRACT

To investigate the effects of leonurine(Leo) on abdominal aortic constriction(AAC)-induced cardiac hypertrophy in rats and its mechanism. A rat model of pressure overload-induced cardiac hypertrophy was established by AAC method. After 27-d intervention with high-dose(30 mg·kg~(-1)) and low-dose(15 mg·kg~(-1)) Leo or positive control drug losartan(5 mg·kg~(-1)), the cardiac function was evaluated by hemodynamic method, followed by the recording of left ventricular systolic pressure(LVSP), left ventricular end-diastolic pressure(LVESP), as well as the maximum rate of increase and decrease in left ventricular pressure(±dp/dt_(max)). The degree of left ventricular hypertrophy was assessed based on heart weight index(HWI) and left ventricular mass index(LVWI). Myocardial tissue changes and the myocardial cell diameter(MD) were measured after hematoxylin-eosin(HE) staining. The contents of angiotensin Ⅱ(AngⅡ) and angiotensin Ⅱ type 1 receptor(AT1 R) in myocardial tissue were detected by ELISA. The level of Ca~(2+) in myocardial tissue was determined by colorimetry. The protein expression levels of phospholipase C(PLC), inositol triphosphate(IP3), AngⅡ, and AT1 R were assayed by Western blot. Real-time quantitative PCR(qRT-PCR) was employed to determine the mRNA expression levels of β-myosin heavy chain(β-MHC), atrial natriuretic factor(ANF), AngⅡ, and AT1 R. Compared with the model group, Leo decreased the LVSP, LVEDP, HWI, LVWI and MD values, but increased ±dp/dt_(max) of the left ventricle. Meanwhile, it improved the pathological morphology of myocardial tissue, reduced cardiac hypertrophy, edema, and inflammatory cell infiltration, decreased the protein expression levels of PLC, IP3, AngⅡ, AT1 R, as well as the mRNA expression levels of β-MHC, ANF, AngⅡ, AT1 R, c-fos, and c-Myc in myocardial tissue. Leo inhibited AAC-induced cardiac hypertrophy possibly by influencing the RAS system.


Subject(s)
Animals , Rats , Angiotensin II/metabolism , Cardiomegaly/genetics , Gallic Acid/analogs & derivatives , Hypertrophy, Left Ventricular/pathology , Myocardium/pathology
2.
Clinics ; 73: e246, 2018. tab, graf
Article in English | LILACS | ID: biblio-952795

ABSTRACT

OBJECTIVES: The present study aimed to investigate cardiovascular autonomic modulation and angiotensin II (Ang II) activity in diabetic mice that were genetically engineered to harbor two or three copies of the angiotensin-converting enzyme gene. METHODS: Diabetic and non-diabetic mice harboring 2 or 3 copies of the angiotensin-converting enzyme gene were used in the present study. Animals were divided into 4 groups: diabetic groups with two and three copies of the angiotensin-converting enzyme gene (2CD and 3CD) and the respective age-matched non-diabetic groups (2C and 3C). Hemodynamic, cardiovascular, and autonomic parameters as well as renal Ang II expression were evaluated. RESULTS: Heart rate was lower in diabetic animals than in non-diabetic animals. Autonomic modulation analysis indicated that the 3CD group showed increased sympathetic modulation and decreased vagal modulation of heart rate variability, eliciting increased cardiac sympathovagal balance, compared with all the other groups. Concurrent diabetes and either angiotensin-converting enzyme polymorphism resulted in a significant increase in Ang II expression in the renal cortex. CONCLUSION: Data indicates that a small increase in angiotensin-converting enzyme activity in diabetic animals leads to greater impairment of autonomic function, as demonstrated by increased sympathetic modulation and reduced cardiac vagal modulation along with increased renal expression of Ang II.


Subject(s)
Animals , Male , Mice , Autonomic Nervous System/physiopathology , Angiotensin II/analysis , Cardiovascular System/physiopathology , Peptidyl-Dipeptidase A/genetics , Gene Dosage/physiology , Diabetes Mellitus, Experimental/physiopathology , Kidney/enzymology , Vagus Nerve/physiopathology , Blood Glucose/analysis , Angiotensin II/metabolism , Immunohistochemistry , Random Allocation , Polymerase Chain Reaction , Heart Rate/physiology
3.
Indian J Biochem Biophys ; 2015 Apr; 52 (2): 119-124
Article in English | IMSEAR | ID: sea-158207

ABSTRACT

The role of angiotensin II in regulating Na+/K+-ATPase activity has been investigated in bovine pulmonary artery smooth muscle cells (BPASMCs). Our study reveals that angiotensin II inhibits the Na+/K+ATPase activity via glutathionylation of the pump with the involvement of an increase in NADPH oxidase-derived O2.-. Additionally, angiotensin II treatment to the cells increases the inhibitory potency of the 15.6 kDa inhibitor towards the Na+/K+ATPase activity.


Subject(s)
Angiotensin II/metabolism , Enzyme Inhibitors/chemistry , Glutathione/pharmacology , /enzymology , Oxidation-Reduction , Pulmonary Artery/enzymology , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/chemistry
4.
Rev. méd. Chile ; 143(1): 109-111, ene. 2015.
Article in Spanish | LILACS | ID: lil-742558

ABSTRACT

Professor Alessandri died in 1980. We started our residency in Internal Medicine about 30 years later. Considering the profound changes our society has witnessed, including medical practice, I would like to approach the meaning of his work for our generation. It is not the Father’s figure nor his Aura what inspires us today. Neither is his personality nor his shape. His universality comes from his transcendent image as a teacher. Today’s teachers live rough times, their social status has changed, their professional requirements have grown exponentially, they have to adapt to social phenomena like the Internet and multiculturalism. Being a teacher nowadays demands to be a multifaceted expert. Things have changed since Professor Alessandri made rounds with his patients. But a deeper look allows us to understand that everything returns to where it started: professional deontology of the teacher, never fading but transcendent. We know that Doctor Alessandri had the natural gift to keep faithful to that code with consistency and perseverance. He excelled with integrity in every aspect including professional betterment, constructive work for his institution, collegiality, a warm relationship with students and a model of social values. Beyond virtues and personal defects he will keep on being the mould in which present teachers should be formed, engraved in their souls and in the subconscious of students that seek to learn.


Subject(s)
Animals , Dogs , Humans , Mice , /metabolism , Oxygen/chemistry , Sick Sinus Syndrome/genetics , Sinoatrial Node/pathology , Angiotensin II/metabolism , Apoptosis , Biomarkers/metabolism , Electrocardiography/methods , Mice, Transgenic , NADPH Oxidases/genetics , Reactive Oxygen Species , Sick Sinus Syndrome/metabolism
5.
Medicina (B.Aires) ; 74(3): 254-258, jun. 2014. ilus
Article in Spanish | LILACS | ID: lil-734375

ABSTRACT

La muerte celular programada y la fibrosis renal son procesos inherentes a la enfermedad renal crónica y, en tal sentido, ha sido recientemente descripta una clara desregulación de la maquinaria respiratoria mitocondrial en pacientes con enfermedad renal crónica asociada con un aumento del estrés oxidativo. Las células tubulares lesionadas vinculadas a los macrófagos intersticiales y miofibroblastos producen citoquinas y factores de crecimiento que promueven un estado inflamatorio, inducen la apoptosis de las células tubulares y facilitan la acumulación de matriz extracelular. La angiotensina II desempeña un papel central en la fibrogénesis renal y conduce a una rápida progresión de la enfermedad renal crónica. Los niveles crecientes de la angiotensina II inducen citoquinas pro-inflamatorias, la activación de NF-kB, moléculas de adhesión, quimiocinas, factores de crecimiento y estrés oxidativo. Toda la evidencia actual sugiere que la angiotensina II aumenta el estrés oxidativo mitocondrial, regula la inducción de apoptosis y condiciona al estado inflamatorio. Por lo tanto, existiría un papel determinante de las mitocondrias y el estrés oxidativo en el proceso inflamatorio renal. Finalmente, esta revisión resume nuestro actual conocimiento acerca de los posibles mecanismos que contribuirían con la apoptosis modulada por la inflamación y/o el estrés oxidativo durante la enfermedad renal crónica. Además, se propone un nuevo concepto de herramientas anti-inflamatorias que regulan el estrés oxidativo mitocondrial lo cual afectaría directamente al proceso inflamatorio y la apoptosis. Esta idea podría tener consecuencias atractivas sobre el tratamiento de patologías inflamatorias renales y de otras afines.


The apoptosis and renal fibrosis are processes inherent to the chronic kidney disease, and consequently a clear deregulation of the mitochondrial respiratory mechanism has been described in patients with chronic renal disease associated to an increase of the oxidative stress. The injured tubular cells linked to the interstitial macrophages and myofibroblasts produce cytokines and growth factors that encourage an inflammatory condition, inducing the apoptosis of the tubular cells and enabling the accumulation of the extracellular matrix. The angiotensin II has a central role in the renal fibrogenesis leading to a rapid progression of the chronic kidney disease. The growing levels of the angiotensin II induce pro-inflammatory cytokines, the activation of NF-kB, adhesion molecules,chemokines, growth factors, and oxidative stress. The current evidence suggests that the angiotensin II increases the mitochondrial oxidative stress, regulates the induction of the apoptosis and conditions the inflammatory process. Therefore the mitochondria and the oxidative stress would play a determinant role in the renal inflammatory process. Finally, this review summarizes our present knowledge regarding the possible mechanisms that would contribute to the apoptosis conditioned by inflammation and/or oxidative stress during the chronic renal disease. Additionally, a new concept of the anti-inflammatory tools is proposed to regulate the mitochondrial oxidative stress that would directly affect the inflammatory process and apoptosis. This concept could have positive consequences on the treatment of renal inflammatory pathologies and related diseases.


Subject(s)
Animals , Humans , Apoptosis/physiology , Mitochondria/metabolism , Mitochondria/pathology , Nephritis/etiology , Oxidative Stress/physiology , Renal Insufficiency, Chronic/etiology , Angiotensin II/metabolism , Cytoprotection , Ergocalciferols/pharmacology , Kidney Cortex/drug effects , Kidney Cortex/pathology , Kidney Tubules/drug effects , Kidney Tubules/pathology , NF-kappa B/metabolism , Nephritis/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Vitamins/pharmacology
6.
Braz. j. med. biol. res ; 46(9): 746-751, 19/set. 2013. graf
Article in English | LILACS | ID: lil-686569

ABSTRACT

Macrophage migration inhibitory factor (MIF), a pleiotropic cytokine, plays an important role in the pathogenesis of atrial fibrillation; however, the upstream regulation of MIF in atrial myocytes remains unclear. In the present study, we investigated whether and how MIF is regulated in response to the renin-angiotensin system and oxidative stress in atrium myocytes (HL-1 cells). MIF protein and mRNA levels in HL-1 cells were assayed using immunofluorescence, real-time PCR, and Western blot. The result indicated that MIF was expressed in the cytoplasm of HL-1 cells. Hydrogen peroxide (H2O2), but not angiotensin II, stimulated MIF expression in HL-1 cells. H2O2-induced MIF protein and gene levels increased in a dose-dependent manner and were completely abolished in the presence of catalase. H2O2-induced MIF production was completely inhibited by tyrosine kinase inhibitors genistein and PP1, as well as by protein kinase C (PKC) inhibitor GF109203X, suggesting that redox-sensitive MIF production is mediated through tyrosine kinase and PKC-dependent mechanisms in HL-1 cells. These results suggest that MIF is upregulated by HL-1 cells in response to redox stress, probably by the activation of Src and PKC.


Subject(s)
Animals , Mice , Hydrogen Peroxide/pharmacology , Intramolecular Oxidoreductases/drug effects , Macrophage Migration-Inhibitory Factors/drug effects , Myocytes, Cardiac/metabolism , Oxidants/pharmacology , Protein Kinase C/metabolism , src-Family Kinases/metabolism , Angiotensin II/metabolism , Blotting, Western , Cell Line , Immunohistochemistry , Intramolecular Oxidoreductases/genetics , Microscopy, Confocal , Macrophage Migration-Inhibitory Factors/genetics , Oxidative Stress/physiology , Protein Kinase Inhibitors/pharmacology , Real-Time Polymerase Chain Reaction , Renin-Angiotensin System/physiology
7.
Indian J Exp Biol ; 2013 Jan; 51(1): 48-55
Article in English | IMSEAR | ID: sea-147567

ABSTRACT

Atrial natriuretic peptide (ANP) exerts anti-hypertrophic effects in the heart via natriuretic peptide receptor-A (NPR-A). However, ANP mediated anti-hypertrophic activity is decreased in the cardiomyopathic conditions. In the present investigation the in vivo effects of angiotensin II (Ang II), a hypertrophic agonist have been studied on the ventricular expression level of NPR-A in Wistar rat hearts. NPR-A expression at the protein and mRNA levels were found to be markedly reduced by 5-fold respectively in Ang II infused rats heart as compared with sham rat hearts. Moreover, cGMP production in response to ANP was reduced by 77% in the isolated cardiac membrane preparation from the Ang II infused rat hearts. Losartan treatment reversed NPR-A expression and responsiveness to ANP. This study suggests that Ang II down regulates cardiac NPR-A activity by suppressing Npr1 gene transcription.


Subject(s)
Angiotensin II/metabolism , Animals , Atrial Natriuretic Factor/chemistry , Down-Regulation , Gene Expression Regulation , Guanylate Cyclase/metabolism , Heart/physiology , Heart Ventricles/pathology , Hypertrophy, Left Ventricular/pathology , Male , Models, Biological , Myocardium/metabolism , Rats , Rats, Wistar , Receptors, Atrial Natriuretic Factor/metabolism , Signal Transduction
8.
Yonsei Medical Journal ; : 258-261, 2012.
Article in English | WPRIM | ID: wpr-154820

ABSTRACT

Increased arterial stiffness is an independent predictor of cardiovascular disease independent from blood pressure. Recent studies have shed new light on the importance of inflammation on the pathogenesis of arterial stiffness. Arterial stiffness is associated with the increased activity of angiotensin II, which results in increased NADPH oxidase activity, reduced NO bioavailability and increased production of reactive oxygen species. Angiotensin II signaling activates matrix metalloproteinases (MMPs) which degrade TGFbeta precursors to produce active TGFbeta, which then results in increased arterial fibrosis. Angiotensin II signaling also activates cytokines, including monocyte chemoattractant protein-1, TNF-alpha, interleukin-1, interleukin-17 and interleukin-6. There is also ample clinical evidence that demonstrates the association of inflammation with increased arterial stiffness. Recent studies have shown that reductions in inflammation can reduce arterial stiffness. In patients with rheumatoid arthritis, increased aortic pulse wave velocity in patients was significantly reduced by anti tumor necrosis factor-alpha therapy. Among the major classes of anti hypertensive drugs, drugs that block the activation of the RAS system may be more effective in reducing the progression of arterial stiffness. Thus, there is rationale for targeting specific inflammatory pathways involved in arterial stiffness in the development of future drugs. Understanding the role of inflammation in the pathogenesis of arterial stiffness is important to understanding the complex puzzle that is the pathophysiology of arterial stiffening and may be important for future development of novel treatments.


Subject(s)
Humans , Angiotensin II/metabolism , Inflammation/drug therapy , Matrix Metalloproteinases/metabolism , Vascular Stiffness/drug effects
9.
The Korean Journal of Hepatology ; : 251-257, 2011.
Article in English | WPRIM | ID: wpr-58543

ABSTRACT

NADPH oxidase (NOX) is a multicomponent enzyme complex that generates reactive oxygen species (ROS) in response to a wide range of stimuli. ROS is involved as key secondary messengers in numerous signaling pathways, and NADPH oxidases complex has been increasingly recognized as key elements of intracellular signaling of hepatic fibrogenesis. In the liver, NADPH oxidase is functionally expressed both in the phagocytic form and in the non-phagocytic form. The non-phagocytic NADPH oxidase complex is structurally and functionally similar to the phagocytic NADPH, resulting in reduction of molecular oxygen to generate superoxide. There are six homologous NOX proteins in the non-phagocytic forms of NADPH oxidase. An emerging concept is that both phagocytic and nonphagocytic NADPH oxidase components in hepatic stellate cells (HSCs) mediate hepatic fibrosis, suggesting its potential role as a pharmacological target for anti-fibrotic therapy. The molecular components and signaling pathways of various NADPH oxidase homologues that are critical for the fibrotic activity in HSCs need to be more clearly identified.


Subject(s)
Humans , Angiotensin II/metabolism , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/enzymology , NADPH Oxidases/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism
10.
Journal of Veterinary Science ; : 291-293, 2011.
Article in English | WPRIM | ID: wpr-108423

ABSTRACT

Blood-borne angiotensin-II (Ang-II) has profound effects in the brain. We tested the hypothesis that Ang-II-dependent hypertension involves differential Ang-II type I (AT1) receptors expression in the subfornical organ (SFO) and the rostral ventrolateral medulla (RVLM). Male Wistar rats were implanted with 14-day osmotic minipump filled with Ang-II (150 ng/kg/min) or saline. AT1 receptor mRNA levels were detected in the SFO and RVLM by reverse transcription-polymerase chain reaction (RT-PCR). Ang-II caused hypertension (134 +/- 10 mmHg vs. 98 +/- 9 mmHg, n = 9, p < 0.05). RT-PCR revealed that Ang-II infusion induced increased AT1 receptor mRNA levels in RVLM and decreased in SFO. Our data suggest that Ang-II-induced hypertension involves differential expression of brain AT1 receptors.


Subject(s)
Animals , Male , Rats , Angiotensin II/metabolism , Hypertension/chemically induced , Medulla Oblongata/metabolism , RNA, Messenger/genetics , Rats, Wistar , Receptor, Angiotensin, Type 1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Subfornical Organ/metabolism
11.
Experimental & Molecular Medicine ; : 569-576, 2009.
Article in English | WPRIM | ID: wpr-34743

ABSTRACT

Angiotensin II (AngII) is a crucial hormone that affects vasoconstriction and exerts hypertrophic effects on vascular smooth muscle cells. Here, we showed that phosphatidylinositol 3-kinase-dependent calcium mobilization plays pivotal roles in AngII-induced vascular constriction. Stimulation of rat aortic vascular smooth muscle cell (RASMC)-embedded collagen gel with AngII rapidly induced contraction. AngII-induced collagen gel contraction was blocked by pretreatment with a phosphatidylinositol 3-kinase (PI3K) inhibitor (LY294002) whereas ERK inhibitor (PD98059) was not effective. AngII-induced collagen gel contraction was significantly blocked by extracellular calcium depletion by EGTA or by nifedipine which is an L-type calcium channel blocker. In addition, AngII-induced calcium mobilization was also blocked by nifedipine and EGTA, whereas intracellular calcium store-depletion by thapsigargin was not effective. Finally, pretreatment of rat aortic ring with LY294002 and nifedipine significantly reduced AngII-induced constriction. Given these results, we suggest that PI3K-dependent activation of L-type calcium channels might be involved in AngII-induced vascular constriction.


Subject(s)
Animals , Rats , Phosphatidylinositol 3-Kinase/metabolism , Angiotensin II/metabolism , Aorta, Thoracic/drug effects , Calcium Channels, L-Type/drug effects , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Rats, Sprague-Dawley , Signal Transduction/drug effects , Specific Pathogen-Free Organisms , Vasoconstriction/drug effects
12.
Experimental & Molecular Medicine ; : 387-396, 2009.
Article in English | WPRIM | ID: wpr-196698

ABSTRACT

Angiotensin II (Ang II) stimulates migration of vascular smooth muscle cell (VSMC) in addition to its contribution to contraction and hypertrophy. It is well established that Rho GTPases regulate cellular contractility and migration by reorganizing the actin cytoskeleton. Ang II activates Rac1 GTPase, but its upstream guanine nucleotide exchange factor (GEF) remains elusive. Here, we show that Ang II-induced VSMC migration occurs in a betaPIX GEF-dependent manner. betaPIX-specific siRNA treatment significantly inhibited Ang II-induced VSMC migration. Ang II activated the catalytic activity of betaPIX towards Rac1 in dose- and time-dependent manners. Activity reached a peak at 10 min and declined close to a basal level by 30 min following stimulation. Pharmacological inhibition with specific kinase inhibitors revealed the participation of protein kinase C, Src family kinase, and phosphatidylinositol 3-kinase (PI3-K) upstream of betaPIX. Both p21-activated kinase and reactive oxygen species played key roles in cytoskeletal reorganization downstream of betaPIX-Rac1. Taken together, our results suggest that betaPIX is involved in Ang II-induced VSMC migration.


Subject(s)
Animals , Rats , Phosphatidylinositol 3-Kinase/metabolism , Angiotensin II/metabolism , Cell Movement , Cells, Cultured , Guanine Nucleotide Exchange Factors/genetics , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , NADPH Oxidases/metabolism , Protein Kinase C/metabolism , RNA, Small Interfering/genetics , Rats, Sprague-Dawley , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , src-Family Kinases/metabolism
13.
Indian J Exp Biol ; 2008 Nov; 46(11): 749-54
Article in English | IMSEAR | ID: sea-61111

ABSTRACT

The use of oral contraceptive (OC) steroids is associated with high blood pressure, although mechanisms responsible are still unclear. This study sought to investigate the possible roles that renin-angiotensin system (RAS) and sympathetic nervous system (SNS) may play in the development of OC-induced hypertension. Administration of OC led to significant increases in blood pressure, heart weight and significant decrease in urinary output in OC-treated and OC+clonidine-treated groups but not in OC+captopril-treated group. The pressor response to angiostensin II was significantly greater in the OC-treated rats than in the control rats. However, the pressor responses induced by norepinephrine were not significantly affected by OC administration. The results of the present study demonstrate that OC-induced high blood pressure is associated with cardiac hypertrophy, enhanced pressor response to angiotensin II and preserved pressor response to sympathetic activation. The study also suggests that the development of the OC-induced hypertension and cardiac hypertrophy is mediated by RAS, but not by SNS.


Subject(s)
Angiotensin II/metabolism , Animals , Blood Pressure , Clonidine/therapeutic use , Contraceptives, Oral/pharmacology , Dose-Response Relationship, Drug , Female , Hypertension/etiology , Models, Biological , Norepinephrine/pharmacology , Rats , Rats, Sprague-Dawley , Renin-Angiotensin System , Sympathetic Nervous System/drug effects , Treatment Outcome
14.
Arq. bras. endocrinol. metab ; 51(6): 901-912, ago. 2007. ilus
Article in Portuguese | LILACS | ID: lil-464281

ABSTRACT

O principal determinante da nefropatia diabética é a hiperglicemia, mas hipertensão e fatores genéticos também estão envolvidos. O glomérulo é o foco de lesão, onde proliferação celular mesangial e produção excessiva de matriz extracelular decorrem do aumento da glicose intracelular, por excesso de glicose extracelular e hiperexpressão de GLUT1. Seguem-se aumento do fluxo pela via dos polióis, estresse oxidativo intracelular, produção intracelular aumentada de produtos avançados da glicação não enzimática (AGEs), ativação da via da PKC, aumento da atividade da via das hexosaminas e ativação de TGF-beta1. Altas concentrações de glicose também aumentam angiotensina II (AII) nas células mesangiais por aumento intracelular da atividade da renina (ações intrácrinas, mediando efeitos proliferativos e inflamatórios diretamente). Portanto, glicose e AII exercem efeitos proliferativos celulares e de matriz extracelular nas células mesangiais, utilizando vias de transdução de sinais semelhantes, que levam a aumento de TGF-beta1. Nesse estudo são revisadas as vias que sinalizam os efeitos da glicose e AII nas células mesangiais em causar os eventos-chaves relacionados à gênese da glomerulopatia diabética. As alterações das vias de sinalização implicadas na glomerulopatia, aqui revisadas, suportam dados de estudos observacionais/ensaios clínicos, onde controle metabólico e anti-hipertensivo, especificamente com inibidores do sistema renina-angiotensina, têm-se mostrado importantes - e aditivos - na prevenção do início e progressão da nefropatia. Novas estratégias terapêuticas dirigidas aos eventos intracelulares descritos deverão futuramente promover benefício adicional.


The determinant of the diabetic nephropathy is hyperglycemia, but hypertension and other genetic factors are also involved. Glomerulus is the focus of the injury, where mesangial cell proliferation and extracellular matrix occur because of the increase of the intra- and extracellular glucose concentration and overexpression of GLUT1. Sequentially, there are increases in the flow by the poliol pathway, oxidative stress, increased intracellular production of advanced glycation end products (AGEs), activation of the PKC pathway, increase of the activity of the hexosamine pathway, and activation of TGF-beta1. High glucose concentrations also increase angiotensin II (AII) levels. Therefore, glucose and AII exert similar effects in inducing extracellular matrix formation in the mesangial cells, using similar transductional signal, which increases TGF-beta1 levels. In this review we focus in the effect of glucose and AII in the mesangial cells in causing the events related to the genesis of diabetic nephropathy. The alterations in the signal pathways discussed in this review give support to the observational studies and clinical assays, where metabolic and antihypertensive controls obtained with angiotensin-converting inhibitors have shown important and additive effect in the prevention of the beginning and progression of diabetic nephropathy. New therapeutic strategies directed to the described intracellular events may give future additional benefits.


Subject(s)
Humans , Diabetic Nephropathies/etiology , Glomerular Mesangium , Hyperglycemia/complications , Angiotensin II/metabolism , Cell Proliferation/drug effects , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/physiopathology , Endothelium-Dependent Relaxing Factors/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Glomerular Mesangium/metabolism , Glomerular Mesangium/pathology , Glomerular Mesangium/physiopathology , Glucose Transporter Type 1/metabolism , /metabolism , Hyperglycemia/metabolism , Hyperglycemia/physiopathology , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Renin-Angiotensin System/drug effects , Sclerosis/metabolism , Sclerosis/physiopathology , Transforming Growth Factor beta1/metabolism , Vasoconstrictor Agents/metabolism
15.
Biocell ; 31(1): 1-12, abr. 2007. ilus, graf
Article in English | LILACS | ID: lil-491532

ABSTRACT

Congenital obstructive nephropathy is the primary cause of end-stage renal disease in children. Rapid diagnosis and initiation of the treatment are vital to preserve function and/or to slow down renal injury. Obstructive uropathy effects -decline in the plasmatic renal flow and glomerular filtration rate, interstitial infiltrate of leukocytes, significant decrease of the urine concentration, loss of the capacity to concentrate urine as well as fibrosis and apoptosis- are a consequence of a variety of factors that work in complex ways and are still not fully understood. Mediators as angiotensin II, transforming growth factor-beta(TGF-beta) and nitric oxide (NO) have been implicated in congenital obstructive nephropathy. The renin-angiotensin system is regulated in different ways, affecting both renal structure and function, and that it in turn depends upon the duration of the obstruction. On the other hand, the role of nitric oxide in renal injury remains somewhat controversial due to the fact that it can exert opposite effects such as cytoprotective and prooxidant / proapoptotic efects as well as proinflammatory and anti-inflammatory effects. In addition, reactive oxidative species (ROS) might contribute to the progression of renal disease. During unilateral ureteral obstruction induced uncoordinated and aberrant growth may lead to the loss of cellular phenotype and apoptosis. Promoting inflammatory responses, the oxidizers can regulate the adherence of certain molecules and proinflammatory mediators, transcription factors and fibrogenic cytokines, that are clearly involved in the progression of renal disease. The congenital obstructive nephropathy is characterized by tubular atrophy, cellular proliferation, apoptosis and fibrosis; immature kidney is more susceptible than adult kidney to showing the above mentioned alterations.


Subject(s)
Humans , Animals , Child , Adult , Angiotensin II/metabolism , Angiotensin II/urine , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/pathology , Intercellular Adhesion Molecule-1/metabolism , Intercellular Adhesion Molecule-1/urine , Nitric Oxide/metabolism , Nitric Oxide/urine , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/urine , Apoptosis , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/urine , Biomarkers/metabolism , Oxidative Stress , Ureteral Obstruction/physiopathology , Ureteral Obstruction/metabolism , Ureteral Obstruction/urine
16.
Rev. chil. cardiol ; 26(1): 63-76, 2007. tab, ilus
Article in Spanish | LILACS | ID: lil-499058

ABSTRACT

Recientemente se ha descubierto el gen de una nueva enzima convertidora de angiotensina I- homóloga a ECA y denominada ECA-2 que finalmente lleva a producir angiotensina 1-7. Esta segunda vía del sistema reninaangiotensina (SRA), con la ECA-2 tendría acciones opuestas a la ECA. Objetivo y Métodos: Determinar la actividad y expresión de la ECA2 en el tratamiento de la hipertrofia y fibrosis ventricular izquierda tardía post IAM en la rata. Se usaron ratas Sprague Dawley 200 +/- 10 g sometidas a ligadura de la coronaria izquierda. Como controles se usaron ratas sham (S). 48 hrs. post cirugía, un grupo de ratas IAM recibió enalapril (IAM-ENA; 10 mg/kg, gavage). A las 8 semanas post-operatorias se determinó la presión arterial sistólica (PAS), masa corporal(MC), masa cardíaca relativa (MCR, mg VI/100 g MC, expresión proteica de la cadena pesada de la miosina beta(betaMHC) por Western blot, mRNA por RT-PCR, las actividades enzimáticas de ECA y de ECA-2 por fluorimetría tanto circulante como en ventrículo izquierdo (VI), el contenido de colágeno total por rojo picrosirio y la actividad de metaloproteasa 2 (MMP-2) por zimografía. Conclusión: El aumento de la actividad y expresión de la ECA2 (a nivel circulante y tisular cardíaco) se asocia a menor fibrosis e hipertrofia ventricular izquierda, lo que podría aumentar – en ese periodo - el efecto cardioprotector de Ang-(1-7).


Background. Recently the gene of a new angiotensin-1 converting homologous enzyme (ACE-2) which leads to the production of angiotensin 1-7 (Ang (1-7) has been reported. This new pathway of the renin-angiotensin system(RAS) is supposed to have opposite effects to those of ACE. Aim: To determine the activity and expression of ACE-2 in the development of left ventricular hypertrophy and fibrosis late after induced myocardial infarction (AMI) in rats. Methods: 200 +/- 10g Sprague-Dawley rats were submitted to left coronary artery ligation. Sham operated rats were used as controls. 48 hr after surgery, one group of AMI rats received enalapril (AMI-En), 10mg/Kg. 8 weeks after surgery the systolic blood pressure (SBP), body mass (BM) and relative cardiac mass (RCM, mg/100g BM) were measured. The protein expression of heavy weight chain beta myosin (beta HCM) was determined by Western Blot, mRNA through RT-PCR, circulating and left ventricular ACE and ACE-2 activities through fluorometry, total collagen content by the pycrosirius red method and metaloproteinase-2 (MMP-2) through zymography were determined. Conclusion: The increased activity and expression of ACE-2 both in plasma and the LV is associated to less fibrosis and left ventricular hypertrophy after AMI. This could temporarily boost the cardioproctive effect of Ang (1-7).


Subject(s)
Animals , Rats , Myocardial Infarction/enzymology , Peptidyl-Dipeptidase A , Peptidyl-Dipeptidase A/metabolism , Angiotensin II/metabolism , Blotting, Western , Myosin Heavy Chains/analysis , Cardiomyopathies/enzymology , Enalapril/pharmacology , Fibrosis , Fluorometry , Hypertrophy, Left Ventricular/enzymology , Metalloproteases/metabolism , /metabolism , Rats, Sprague-Dawley , Renin-Angiotensin System/physiology
17.
Rev. chil. cardiol ; 25(3): 291-300, oct.-dic. 2006. tab, graf
Article in Spanish | LILACS | ID: lil-451693

ABSTRACT

Antecedentes: El polimorfismo de la enzima convertidora de angiotensina I (ECA) determina mayor actividad de ECA yniveles de angiotensina (Ang) II en ratas Brown Norway (BN) y menor actividad de ECA y niveles de Ang II en ratas Lewis (L). La relación entre niveles de Ang II y la vía transduccional Rho A/Rho kinasa no ha sido explorada.Objetivo: Determinar la participación de la vía Rho A/ Rho kinasa mediante la fosforilación de la proteína blanco 1 de la fosfatasa de la miosina (MYPT1) vascular en ratas con niveles genéticamente bajos (L) y altos de Ang II (BN) y su relación con la expresión de algunos genes que determinan remodelado de la pared arterial: el gen del activador de plasminógeno 1 (PAI-1), el gen de la proteína quimioatractante de monocitos (MCP-1) y el gen del factor de transformación beta 1 (TGF- b1) en la pared arterial. Métodos: Se usaron ratas machos homocigotos de 150 grs BN y L. Para inhibir la vía Rho A / Rho kinasa, se administró fasudil (100 mg/Kg/día por gavage) a ratas BN, durante 7 días. Se determinó la presión arterial sistólica (PAS), la expresión vascular (en la aorta) de MYPT1 total (MYPT1-T) y MYPT1 fosforilada (MYPT1-P) y la relación entre ambas por Western blot además de la expresión génica de PAI-1, MCP-1 y de TGF-b1 por RT-PCR (en unidades de densidad óptica). Resultados como promedio(ES): El antagonista del receptor tipo I de angiotensina II Candesartán por 7 días (10 mg/Kg/d, n = 8) redujo en un 50 por ciento los niveles elevados de MYPT1-P/T en las ratas BN (p <0.05) sin modificar la mayor expresión de los genes evaluados. Conclusión: La vía Rho A/ Rho kinasa se encuentra activada en la pared arterial de ratas con niveles elevados de Ang II (BN) y causa mayor expresión génica de PAI-1 y de MCP-1 ya que la sobre expresión de ambos tiende a normalizarse con fasudil. La mayor expresión génica de TGF-b1 en la aorta en ratas BN parece no estar relacionada con la activación de de Rho A/ Rho kinasa vía Angiotensina II ya que no...


Subject(s)
Rats , Animals , /pharmacology , Angiotensin I/metabolism , Angiotensin II/metabolism , Aorta , Aorta/metabolism , Blood Pressure/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Ventricular Remodeling , Gene Amplification , Genotype , Enzyme Inhibitors/pharmacology , Phosphorylation , Polymerase Chain Reaction , Polymorphism, Genetic
19.
Rev. argent. cardiol ; 73(5): 346-351, sept.-oct. 2005. tab, graf
Article in Spanish | LILACS | ID: lil-434918

ABSTRACT

El objetivo del presente trabajo fue evaluar el efecto de la administración intrahipotalámica de la angiotensina -[1-7](Ang-[1-7]) sobre la presión arterial y la frecuencia cardíaca y sobre la actividad presora de la angiotensina II (Ang-II) en ratas con operación simulada (OS) o con coartación aórtica (CoAo) en un estadio temprano y en uno crónico de la hipertensión. Se utilizaron ratas Wistar. A los 7 y 42 días de la operación correspondiente se canuló una arteria carótida para la medición de la presión arterial media (PAM) en ratas anestesiadas y se insertó una aguja inyectora en el hipotálamo anterior para la administración de Ang-II (50 ng), Ang-[1-7] (50ng) y Ang-II + Ang-[1-7] (50 + 50 ng). La administración hipotálamica de Ang-II produjo un aumento de la PAM en las ratas CoAo con respecto al grupo control en los estadios temprano y crónico de hipertensión, mientras que la Ang-[1-7] redujo la actividad presora de la Ang-II en las ratas CoAo, mientras que careció de efecto sobre la actividad presora de la Ang-II en las ratas normotensas. En conclusión, la actividad presora de la Ang-II está aumentada en ratas en los estadios temprano y crónico de hipertensión arterial, mientras que el efecto antagónico de la Ang-[1-7] indicaría que ésta modularía la actividad presora de la angiotensina II cuando la Ang-II estuviera exacerbada, ya que se ha observado únicamente en el grupo de ratas hipertensas.


Subject(s)
Animals , Female , Rats , Aortic Coarctation , Angiotensin I/metabolism , Angiotensin II/metabolism , Blood Pressure , Hypothalamus, Anterior , Hypertension , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL