Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ACS Med Chem Lett ; 9(5): 440-445, 2018 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-29795756

RESUMO

Sodium-phosphate cotransporter 2a, or NaPi2a (SLC34A1), is a solute-carrier (SLC) transporter located in the kidney proximal tubule that reabsorbs glomerular-filtered phosphate. Inhibition of NaPi2a may enhance urinary phosphate excretion and correct maladaptive mineral and hormonal derangements associated with increased cardiovascular risk in chronic kidney disease-mineral and bone disorder (CKD-MBD). To date, only nonselective NaPi inhibitors have been described. Herein, we detail the discovery of the first series of selective NaPi2a inhibitors, resulting from optimization of a high-throughput screening hit. The oral PK profile of inhibitor PF-06869206 (6f) in rodents allows for the exploration of the pharmacology of selective NaPi2a inhibition.

2.
Am J Physiol Renal Physiol ; 301(5): F1035-41, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21835766

RESUMO

Prostaglandins have been implicated as paracrine regulators of renin secretion, but the specific pathways and receptor(s) carrying out these functions have not been fully elucidated. To examine the contributions of prostanoid synthetic pathways and receptors to regulation of renin in the intact animal, we used a panel of mice with targeted disruption of several key genes: cyclooxygenase-2 (COX-2), microsomal PGE synthases 1 and 2 (mPGES1, mPGES2), EP2 and EP4 receptors for PGE(2), and the IP receptor for PGI(2). To activate the macula densa signal for renin stimulation, mice were treated with furosemide over 5 days and renin mRNA levels were determined by real-time RT-PCR. At baseline, there were no differences in renin mRNA levels between wild-type and the various strains of mutant mice. Furosemide caused marked stimulation of renin mRNA expression across all groups of wild-type control mice. This response was completely abrogated in the absence of COX-2, but was unaffected in mice lacking mPGES1 or mPGES2. The absence of G(s)/cAMP-linked EP2 receptors had no effect on stimulation of renin by furosemide and there was only a modest, insignificant reduction in renin responses in mice lacking the IP receptor. By contrast, renin stimulation in EP4(-/-) mice was significantly reduced by ∼70% compared with wild-type controls. These data suggest that stimulation of renin by the macula densa mechanism is mediated by PGE(2) through a pathway requiring COX-2 and the EP4 receptor, but not EP2 or IP receptors. Surprisingly, mPGES1 or mPGES2 are not required, suggesting other alternative mechanisms for generating PGE(2) in response to macula densa stimulation.


Assuntos
Receptores de Prostaglandina E Subtipo EP4/fisiologia , Renina/fisiologia , Animais , Pressão Sanguínea , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , DNA Complementar/biossíntese , DNA Complementar/genética , Dinoprostona/fisiologia , Diuréticos/farmacologia , Furosemida/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Endogâmicos , Camundongos Knockout , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real , Renina/biossíntese , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto , Estimulação Química , Telemetria
4.
Hypertension ; 55(2): 531-8, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20065147

RESUMO

Prostaglandin (PG)E(2) has multiple actions that may affect blood pressure. It is synthesized from arachidonic acid by the sequential actions of phospholipases, cyclooxygenases, and PGE synthases. Although microsomal PGE synthase (mPGES)1 is the only genetically verified PGE synthase, results of previous studies examining the consequences of mPGES1 deficiency on blood pressure (BP) are conflicting. To determine whether genetic background modifies the impact of mPGES1 on BP, we generated mPGES1(-/-) mice on 2 distinct inbred backgrounds, DBA/1lacJ and 129/SvEv. On the DBA/1 background, baseline BP was similar between wild-type (WT) and mPGES1(-/-) mice. By contrast, on the 129 background, baseline BPs were significantly higher in mPGES1(-/-) animals than WT controls. During angiotensin II infusion, the DBA/1 mPGES1(-/-) and WT mice developed mild hypertension of similar magnitude, whereas 129-mPGES1(-/-) mice developed more severe hypertension than WT controls. DBA/1 animals developed only minimal albuminuria in response to angiotensin II infusion. By contrast, WT 129 mice had significantly higher levels of albumin excretion than WT DBA/1 and the extent of albuminuria was further augmented in 129 mPGES1(-/-) animals. In WT mice of both strains, the increase in urinary excretion of PGE(2) with angiotensin II was attenuated in mPGES1(-/-) animals. Urinary excretion of thromboxane was unaffected by angiotensin II in the DBA/1 lines but increased more than 4-fold in 129 mPGES1(-/-) mice. These data indicate that genetic background significantly modifies the BP response to mPGES1 deficiency. Exaggerated production of thromboxane may contribute to the robust hypertension and albuminuria in 129 mPGES1-deficient mice.


Assuntos
Albuminúria/metabolismo , Hipertensão/enzimologia , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/metabolismo , Análise de Variância , Angiotensina II/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Dinoprostona/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Regulação da Expressão Gênica , Hipertensão/induzido quimicamente , Oxirredutases Intramoleculares/deficiência , Rim/efeitos dos fármacos , Rim/metabolismo , Camundongos , Camundongos Endogâmicos DBA , Camundongos Knockout , Microssomos/metabolismo , Probabilidade , Prostaglandina-E Sintases , Prostaglandina-Endoperóxido Sintases/metabolismo , Prostaglandinas/metabolismo , RNA Mensageiro/genética , Distribuição Aleatória , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
J Am Soc Nephrol ; 21(3): 448-59, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20056745

RESUMO

Pharmacologic or genetic deletion of components of the renin-angiotensin system leads to postnatal kidney injury, but the roles of these components in kidney development are unknown. To test the hypothesis that angiotensin II supports angiogenesis during postnatal kidney development, we quantified CD31(+) postglomerular microvessels, performed quantitative PCR analysis of vascular growth factor expression, and measured renal blood flow by magnetic resonance. Treating rats with the angiotensin II type 1 receptor antagonist candesartan for 2 weeks after birth reduced the total length, volume, and surface area of capillaries in both the cortex and the medulla and inhibited the organization of vasa recta bundles. In addition, angiotensin II type 1 antagonism inhibited the transcription of angiogenic growth factors vascular endothelial growth factor, angiopoietin-1, angiopoietin-2, and the angiopoietin receptor Tie-2 in cortex and medulla. Similarly, Agtr1a(-/-);Agtr1b(-/-) mouse kidneys had decreased angiopoietin-1, angiopoietin-2, and Tie-2 mRNAs at postnatal day 14. To test whether increased urinary flow leads to microvascular injury, we induced postnatal polyuria with either lithium or adrenalectomy, but these did not alter vascular endothelial growth factor expression or vasa recta organization. Compared with vehicle-treated rats, renal blood flow was significantly (approximately 20%) lower in candesartan-treated rats even 14 days after candesartan withdrawal. Taken together, these data demonstrate that angiotensin II promotes postnatal expansion of postglomerular capillaries and organization of vasa recta bundles, which are necessary for development of normal renal blood flow.


Assuntos
Angiotensina II/metabolismo , Capilares/crescimento & desenvolvimento , Rim/irrigação sanguínea , Neovascularização Fisiológica/fisiologia , Receptor Tipo 1 de Angiotensina/metabolismo , Circulação Renal/fisiologia , Adrenalectomia , Angiopoietina-1/análogos & derivados , Angiopoietina-1/genética , Angiopoietina-2/genética , Angiotensina II/antagonistas & inibidores , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Animais Recém-Nascidos , Benzimidazóis/farmacologia , Compostos de Bifenilo , Capilares/metabolismo , Feminino , Rim/crescimento & desenvolvimento , Rim/patologia , Camundongos , Camundongos Knockout , Neovascularização Fisiológica/efeitos dos fármacos , Poliúria/patologia , Poliúria/fisiopatologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/genética , Receptor TIE-2/genética , Tetrazóis/farmacologia , Fator A de Crescimento do Endotélio Vascular/genética
6.
Am J Physiol Renal Physiol ; 297(5): F1371-80, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19710239

RESUMO

During nephrogenesis, renin expression shifts from the vessel walls of interlobular arteries to the terminal portions of afferent arterioles in a wavelike pattern. Since the mechanisms responsible for the developmental deactivation of renin expression are as yet unknown, we hypothesized that the developing renin-angiotensin system (RAS) may downregulate itself via negative feedback to prevent overactivity of renin. To test for a possible role of angiotensin II in the developmental deactivation of renin expression, we studied the development of intrarenal renin expression in mice lacking ANG II AT1a, AT1b, or AT2 receptors and in animals with abolished circulating ANG II due to deletion of the gene for angiotensin I-converting enzyme (ACE). The development of intrarenal renin expression was normal in mice lacking ANG II AT1b or AT2 receptors. In animals lacking both ANG II AT1a and AT1b receptors, ACE, or ANG II AT1a receptors, renin expression was normal early and renin disappeared from mature vessels until development of cortical interlobular and afferent arterioles began. The development of cortical vessels in these genotypes was accompanied by a markedly increased number of renin-expressing cells, many of which were ectopically located and attached in a grapelike fashion to the outer vessel perimeter. Although the number of renin-expressing cells declined during final maturation of the kidneys, the atypical distribution pattern of renin cells was maintained. These findings suggest that ANG II does not play a central role in the typical developmental shift in renin expression from the arcuate vessels to the afferent arterioles. During postnatal maturation of mouse kidneys, interruption of the RAS causes severe hyperplasia of renin cells via a mechanism that centrally involves AT(1a) receptors. However, the distribution pattern of renin cells in adult kidneys with an interrupted RAS does not mimic any normal developmental stage since renin expression is frequently found in cells outside the arteriolar vessel walls in RAS mutants.


Assuntos
Sistema Renina-Angiotensina/genética , Sistema Renina-Angiotensina/fisiologia , Renina/biossíntese , Actinas/biossíntese , Actinas/genética , Animais , Retroalimentação Fisiológica/fisiologia , Hiperplasia , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Rim/crescimento & desenvolvimento , Rim/metabolismo , Camundongos , Camundongos Knockout , Músculo Liso/metabolismo , Peptidil Dipeptidase A/biossíntese , Peptidil Dipeptidase A/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/fisiologia , Renina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Hypertension ; 54(3): 652-8, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19652084

RESUMO

Drugs and antibodies that interrupt vascular endothelial growth factor (VEGF) signaling pathways improve outcomes in patients with a variety of cancers by inhibiting tumor angiogenesis. A major adverse effect of these treatments is hypertension, suggesting a critical role for VEGF in blood pressure (BP) regulation. However, the physiological mechanisms underlying the control of BP by VEGF are unclear. To address this question, we administered a specific antibody against the major VEGF receptor, VEGFR2, to normal mice and assessed the consequences on BP. Compared with vehicle-treated controls, administration of the anti-VEGFR2 antibody caused a rapid and sustained increase in BP of approximately 10 mm Hg. This increase in BP was associated with a significant reduction in renin mRNA expression in the kidney (P=0.019) and in urinary excretion of aldosterone (P<0.05). Treatment with the anti-VEGFR2 antibody also caused a marked reduction in the expression of endothelial and neuronal NO synthases in the kidney. To examine the role of NO in the hypertension caused by blocking VEGFR2, mice were treated with N(omega)-nitro-L-arginine methyl ester (L-NAME) (20 mg/kg per day), an inhibitor of NO production. L-NAME administration abolished the difference in BP between the vehicle- and anti-VEGFR2-treated groups. Our data suggest that VEGF, acting via VEGFR2, plays a critical role in BP control by promoting NO synthase expression and NO activity. Interfering with this pathway is likely to be one mechanism underlying hypertension caused by antiangiogenic agents targeting VEGF.


Assuntos
Anticorpos Monoclonais/farmacologia , Pressão Sanguínea/fisiologia , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo I/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Aldosterona/urina , Animais , Anticorpos Monoclonais/efeitos adversos , Pressão Sanguínea/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Expressão Gênica/efeitos dos fármacos , Hibridomas , Hipertensão/etiologia , Hipertensão/fisiopatologia , Rim/efeitos dos fármacos , Rim/enzimologia , Masculino , Manometria , Camundongos , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/metabolismo , Prostaglandinas/urina , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Renina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Cloreto de Sódio na Dieta/administração & dosagem , Cloreto de Sódio na Dieta/efeitos adversos , Fator A de Crescimento do Endotélio Vascular/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia
8.
J Clin Invest ; 119(4): 943-53, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19287096

RESUMO

Studies in humans and animal models indicate a key contribution of angiotensin II to the pathogenesis of glomerular diseases. To examine the role of type 1 angiotensin (AT1) receptors in glomerular inflammation associated with autoimmune disease, we generated MRL-Faslpr/lpr (lpr) mice lacking the major murine type 1 angiotensin receptor (AT1A); lpr mice develop a generalized autoimmune disease with glomerulonephritis that resembles SLE. Surprisingly, AT1A deficiency was not protective against disease but instead substantially accelerated mortality, proteinuria, and kidney pathology. Increased disease severity was not a direct effect of immune cells, since transplantation of AT1A-deficient bone marrow did not affect survival. Moreover, autoimmune injury in extrarenal tissues, including skin, heart, and joints, was unaffected by AT1A deficiency. In murine systems, there is a second type 1 angiotensin receptor isoform, AT1B, and its expression is especially prominent in the renal glomerulus within podocytes. Further, expression of renin was enhanced in kidneys of AT1A-deficient lpr mice, and they showed evidence of exaggerated AT1B receptor activation, including substantially increased podocyte injury and expression of inflammatory mediators. Administration of losartan, which blocks all type 1 angiotensin receptors, reduced markers of kidney disease, including proteinuria, glomerular pathology, and cytokine mRNA expression. Since AT1A-deficient lpr mice had low blood pressure, these findings suggest that activation of type 1 angiotensin receptors in the glomerulus is sufficient to accelerate renal injury and inflammation in the absence of hypertension.


Assuntos
Doenças Autoimunes/etiologia , Nefrite/etiologia , Receptor Tipo 1 de Angiotensina/fisiologia , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Doenças Autoimunes/fisiopatologia , Quimiocinas/genética , Citocinas/genética , Feminino , Rim/lesões , Rim/patologia , Rim/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos MRL lpr , Camundongos Knockout , Nefrite/imunologia , Nefrite/patologia , Nefrite/fisiopatologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor Tipo 1 de Angiotensina/deficiência , Receptor Tipo 1 de Angiotensina/genética , Sistema Renina-Angiotensina/fisiologia
9.
J Biol Chem ; 283(17): 11850-9, 2008 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-18310078

RESUMO

In vivo protein kinases A and G (PKA and PKG) coordinately phosphorylate a broad range of substrates to mediate their various physiological effects. The functions of many of these substrates have yet to be defined genetically. Herein we show a role for smoothelin-like protein 1 (SMTNL1), a novel in vivo target of PKG/PKA, in mediating vascular adaptations to exercise. Aortas from smtnl1(-/-) mice exhibited strikingly enhanced vasorelaxation before exercise, similar in extent to that achieved after endurance training of wild-type littermates. Additionally, contractile responses to alpha-adrenergic agonists were greatly attenuated. Immunological studies showed SMTNL1 is expressed in smooth muscle and type 2a striated muscle fibers. Consistent with a role in adaptations to exercise, smtnl1(-/-) mice also exhibited increased type 2a fibers before training and better performance after forced endurance training compared smtnl1(+/+) mice. Furthermore, exercise was found to reduce expression of SMTNL1, particularly in female mice. In both muscle types, SMTNL1 is phosphorylated at Ser-301 in response to adrenergic signals. In vitro SMTNL1 suppresses myosin phosphatase activity through a substrate-directed effect, which is relieved by Ser-301 phosphorylation. Our findings suggest roles for SMTNL1 in cGMP/cAMP-mediated adaptations to exercise through mechanisms involving direct modulation of contractile activity.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Proteínas Musculares/biossíntese , Proteínas Musculares/fisiologia , Músculo Liso Vascular/metabolismo , Fosfoproteínas/genética , Animais , Feminino , Deleção de Genes , Humanos , Camundongos , Modelos Biológicos , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/genética , Miosinas/metabolismo , Fenótipo , Fosfoproteínas/fisiologia , Fosforilação , Condicionamento Físico Animal
10.
J Am Soc Nephrol ; 18(5): 1466-75, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17442791

RESUMO

Prostaglandin E(2) (PGE(2)) is one of the most ubiquitous prostanoids in the kidney, where it may influence a wide range of physiologic functions. PGE(2) is generated through enzymatic metabolism of prostanoid endoperoxides by specific PGE synthases (PGES). Several putative PGES have been identified and cloned, including the membrane-associated, inducible microsomal PGES1 (mPGES1), which is expressed in the kidney. To evaluate the physiologic role of mPGES1 in the kidney, mice with targeted disruption of mPges1 gene were studied, with a focus on responses where PGE(2) has been implicated, including urinary concentration, regulation of blood pressure, and response to a loop diuretic. The absence of mPGES1 was associated with a 50% decrease in basal excretion of PGE(2) in urine (P < 0.001). In female but not male mPGES1-deficient mice, there was a reciprocal increase in basal excretion of other prostanoids. Nonetheless, urinary osmolalities were similar in mPges1(+/+) and mPges1(-/-) mice at baseline and after 12 h of water deprivation. Likewise, there were no differences in blood pressure between mPGES1-deficient and wild-type mice on control or high- or low-salt diets. The furosemide-induced increase in urinary PGE(2) excretion that was seen in wild-type mice was attenuated in mPGES1-deficient mice. However, furosemide-associated diuresis was reduced only in male, not female, mPGES1-deficient mice. Stimulation of renin by furosemide was not affected by mPGES1 deficiency. These data suggest that mPGES1 contributes to basal synthesis of PGE(2), but there are other pathways that lead to renal PGE(2) synthesis. Moreover, there are significant gender differences in physiologic contributions of mPGES1 to control kidney function.


Assuntos
Oxirredutases Intramoleculares/fisiologia , Rim/fisiologia , Adaptação Fisiológica/efeitos dos fármacos , Animais , Pressão Sanguínea/fisiologia , Diurese/efeitos dos fármacos , Diuréticos/farmacologia , Feminino , Furosemida/farmacologia , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos DBA , Camundongos Knockout , Concentração Osmolar , Prostaglandina-E Sintases , Prostaglandina-Endoperóxido Sintases/genética , Prostaglandina-Endoperóxido Sintases/metabolismo , Prostaglandinas/urina , Renina/genética , Renina/metabolismo , Caracteres Sexuais , Sódio na Dieta/farmacologia
11.
Am J Physiol Renal Physiol ; 289(1): F127-36, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15701815

RESUMO

Although L-type voltage-dependent calcium channels play a major role in mediating vascular smooth muscle cell contraction in the renal vasculature, non-L-type calcium entry mechanisms represent a significant component of vasoactive agonist-induced calcium entry in these cells as well. To investigate the role of these non-voltage-dependent calcium entry pathways in the regulation of renal microvascular reactivity, we have characterized the function of store- and receptor-operated channels (SOCs and ROCs) in renal cortical interlobular arteries (ILAs) of rats. Using fura 2-loaded, microdissected ILAs, we find that the L-type channel antagonist nifedipine blocks less than half the rise in intracellular calcium concentration ([Ca(2+)](i)) elicited by norepinephrine. SOCs were activated in these vessels using the sarco/endoplasmic reticulum Ca(2+) ATPase (SERCA) inhibitors cyclopiazonic acid and thapsigargin and were dose dependently blocked by the SOC antagonists Gd(3+) and 2-aminoethoxydiphenyl borate (2-APB) and the combined SOC/ROC antagonist SKF-96365. Gd(3+) had no effect on the non-L-type Ca(2+) entry activated by 1 microM NE. A low concentration of SKF-96365 that did not affect thapsigargin-induced store-operated Ca(2+) entry blocked 60-70% of the NE-induced Ca(2+) entry. Two different calmodulin inhibitors (W-7 and trifluoperazine) also blocked the NE-induced Ca(2+) entry. These data suggest that in addition to L-type channels, NE primarily activates ROCs rather than SOCs in ILAs and that this receptor-operated Ca(2+) entry mechanism is regulated by calmodulin. Interestingly, 2-APB completely blocked the NE-induced non-L-type Ca(2+) entry, implying that SOCs and ROCs in preglomerular resistance vessels share a common molecular structure.


Assuntos
Canais de Cálcio/fisiologia , Calmodulina/fisiologia , Norepinefrina/fisiologia , Receptores de Detecção de Cálcio/fisiologia , Artéria Renal/fisiologia , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Relação Dose-Resposta a Droga , Técnicas In Vitro , Masculino , Potenciais da Membrana , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
12.
Am J Physiol Renal Physiol ; 286(3): F546-51, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14678949

RESUMO

In the resistance vessels of the renal microcirculation, store- and/or receptor-operated calcium entry contribute to the rise in vascular smooth muscle cell (VSMC) intracellular calcium concentration in response to vasoconstrictor hormones. Short transient receptor potential (TRPC) channels are widely expressed in mammalian tissues and are proposed mediators of voltage-independent cation entry in multiple cell types, including VSMCs. The seven members of the TRPC gene family (TRPC1-7) encode subunit proteins that are thought to form homo- and heterotetrameric channels that are differentially regulated depending on their subunit composition. In the present study, we demonstrate the relative abundance of TRPC mRNA and protein in freshly isolated rat renal resistance vessels, glomeruli, and aorta. TRPC1, 3, 4, 5, and 6 mRNA and protein were detected in both renal resistance vessels and aorta, whereas TRPC2 and TRPC7 mRNA were not expressed. TRPC1, 3, 5, and 6 protein was present in glomeruli. TRPC3 and TRPC6 protein levels were significantly greater in the renal resistance vessels, about six- to eightfold higher than in aorta. These data suggest that TRPC3 and TRPC6 may play a role in mediating voltage-independent calcium entry in renal resistance vessels that is functionally distinct from that in aorta.


Assuntos
Canais de Cálcio/metabolismo , Rim/irrigação sanguínea , Animais , Aorta/metabolismo , Encéfalo/metabolismo , Canais de Cálcio/genética , Rim/metabolismo , Glomérulos Renais/metabolismo , Masculino , Microcirculação , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Circulação Renal
13.
Am J Physiol Heart Circ Physiol ; 282(6): H2364-70, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12003847

RESUMO

Norepinephrine directly induces growth of the vascular wall, which may involve not only proliferation of smooth muscle cells (SMCs) and adventitial fibroblasts (AFBs) but also augmentation of their migration. To test this hypothesis, growth-arrested SMCs and AFBs from rat aorta were exposed to norepinephrine. Norepinephrine caused dose-dependent migration of both cell types that was dependent on chemotaxis. In contrast, platelet-derived growth factor (PDGF)-BB, used as a positive control, stimulated both chemotaxis and chemokinesis. Only alpha(1D)-adrenoceptors (AR) and alpha(2)-AR antagonists inhibited norepinephrine migration of SMCs, whereas norepinephrine migration of AFBs was only inhibited by alpha(1A)-AR and alpha(1B)-AR antagonists; beta-AR blockade was without effect. Norepinephrine and PDGF-BB were additive for AFB, but not SMC, migration. Stimulation of migration was reversed at high norepinephrine concentrations (10 microM); this inhibition was mediated by alpha(2)- and beta-ARs in AFBs but not in SMCs. Thus norepinephrine induces migration of SMCs and AFBs via different alpha-ARs. This action may participate in wall remodeling and norepinephrine potentiation of injury-induced intimal lesion growth.


Assuntos
Movimento Celular , Fibroblastos/citologia , Músculo Liso Vascular/citologia , Receptores Adrenérgicos alfa/fisiologia , Antagonistas Adrenérgicos alfa/farmacologia , Animais , Aorta Torácica , Becaplermina , Movimento Celular/efeitos dos fármacos , Quimiotaxia , Idazoxano/análogos & derivados , Idazoxano/farmacologia , Norepinefrina/administração & dosagem , Norepinefrina/farmacologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Proto-Oncogênicas c-sis , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa 1/fisiologia , Receptores Adrenérgicos alfa 2/fisiologia , Receptores Adrenérgicos beta/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...