Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Kidney Int ; 72(9): 1130-7, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17728707

RESUMO

Elevated serum phosphorus and calcium are associated with arterial calcification and mortality in dialysis patients. Unlike calcium-based binders, sevelamer attenuates arterial calcification but it is unknown whether sevelamer affects mortality or morbidity. In a multicenter, randomized, open-label, parallel design trial we compared sevelamer and calcium-based binders on all-cause and cause-specific mortality (cardiovascular, infection, and other) in prevalent hemodialysis patients. A total of 2103 patients were initially randomized to treatment and 1068 patients completed the study. All-cause mortality rates and cause-specific mortality rates were not significantly different. There was a significant age interaction on the treatment effect. Only in patients over 65 years of age was there a significant effect of sevelamer in lowering the mortality rate. There was a suggestion that sevelamer was associated with lower overall, but not cardiovascular-linked, mortality in older patients. We suggest that further research is needed to confirm these findings.


Assuntos
Acetatos/uso terapêutico , Carbonato de Cálcio/uso terapêutico , Doenças Cardiovasculares/prevenção & controle , Quelantes/uso terapêutico , Nefropatias/terapia , Poliaminas/uso terapêutico , Diálise Renal/mortalidade , Acetatos/efeitos adversos , Adulto , Fatores Etários , Idoso , Cálcio/sangue , Carbonato de Cálcio/efeitos adversos , Compostos de Cálcio/efeitos adversos , Compostos de Cálcio/uso terapêutico , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/mortalidade , Quelantes/efeitos adversos , Doença Crônica , Feminino , Humanos , Nefropatias/complicações , Masculino , Pessoa de Meia-Idade , Fósforo/sangue , Poliaminas/efeitos adversos , Sevelamer , Resultado do Tratamento
2.
Kidney Int ; 53(5): 1259-68, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9573541

RESUMO

Erythropoietin (EPO) increases Ca2+ influx in vascular smooth muscle cells and acts both as a direct vasoconstrictor and vascular growth factor (that is, angiogenesis). However, the mechanism by which EPO promotes extracellular Ca2+ entry in contractile cells has not been elucidated. In hematopoietic cells, EPO induces tyrosine kinase (TK)-dependent activation of phospholipase C (PLC)-gamma 1 and Ca2+ influx via a voltage-independent Ca2+ conductance. In contractile mesangial cells, we have recently characterized a voltage-independent, 1 pS Ca2+ channel that is dependent on both TK and PLC-gamma 1 activity. Therefore, we examined cultured rat glomerular mesangial cells after timed exposure to recombinant human EPO (20 U/ml). Erythropoietin increased the tyrosine phosphorylation of PLC-gamma 1, promoted membrane complex formation between PLC-gamma 1 and the EPO receptor itself, and raised the levels of intracellular inositol 1,4,5-trisphosphate and intracellular Ca2+. Consistent with our previous studies, 1 pS Ca2+ channel activity was extremely low under basal, unstimulated conditions in cell-attached patches, but was dramatically increased when EPO was present in the patch pipette. Tyrosine kinase inhibition with 100 micron genistein or 1 micron PP1 (Src; selective tyrosine kinase inhibitor) prevented all of these EPO-induced responses. We conclude that: (1) EPO-induced stimulation of 1 pS Ca2+ channels is mediated via a cytosolic Src TK in glomerular mesangial cells. (2) Stimulation of this Ca2(+)-activated, Ca2(+)-permeable channel is dependent on the tyrosine phosphorylation/activation of PLC-gamma 1. (3) This cascade provides a possible mechanism for the vasoconstriction and hypertension observed with clinical EPO use for the treatment of chronic anemias.


Assuntos
Canais de Cálcio/metabolismo , Isoenzimas/metabolismo , Receptores da Eritropoetina/metabolismo , Fosfolipases Tipo C/metabolismo , Animais , Cálcio/metabolismo , Canais de Cálcio/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Eritropoetina/farmacologia , Eritropoetina/toxicidade , Mesângio Glomerular/efeitos dos fármacos , Mesângio Glomerular/metabolismo , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Líquido Intracelular/metabolismo , Modelos Biológicos , Fosfolipase C gama , Fosforilação , Ratos , Proteínas Recombinantes , Tirosina/metabolismo , Quinases da Família src/metabolismo
3.
J Biol Chem ; 273(13): 7703-8, 1998 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-9516477

RESUMO

An early event in signaling by the G-protein-coupled angiotensin II (Ang II) AT1 receptor in vascular smooth muscle cells is the tyrosine phosphorylation and activation of phospholipase Cgamma1 (PLCgamma1). In the present study, we show that stimulation of this event by Ang II in vascular smooth muscle cells is accompanied by binding of PLCgamma1 to the AT1 receptor in an Ang II- and tyrosine phophorylation-dependent manner. The PLCgamma1-AT1 receptor interaction appears to depend on phosphorylation of tyrosine 319 in a YIPP motif in the C-terminal intracellular domain of the AT1 receptor and binding of the phosphorylated receptor by the most C-terminal of two Src homology 2 domains in PLCgamma1. PLCgamma1 thus binds to the same site in the receptor previously identified for binding by the SHP-2 phosphotyrosine phosphatase.JAK2 tyrosine kinase complex. A single site in the C-terminal tail of the AT1 receptor can, therefore, be bound in a ligand-dependent manner by two different downstream effector proteins. These data demonstrate that G-protein-coupled receptors can physically associate with intracellular proteins other than G proteins, creating membrane-delimited signal transduction complexes similar to those observed for classic growth factor receptors.


Assuntos
Angiotensina II/farmacologia , Isoenzimas/metabolismo , Proteínas Proto-Oncogênicas , Receptores de Angiotensina/metabolismo , Fosfolipases Tipo C/metabolismo , Animais , Linhagem Celular , Ativação Enzimática , Janus Quinase 2 , Masculino , Músculo Liso Vascular/metabolismo , Fosfolipase C gama , Ligação Proteica/efeitos dos fármacos , Proteínas Tirosina Quinases/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina , Receptor Tipo 2 de Angiotensina , Proteínas Recombinantes de Fusão/metabolismo , Domínios de Homologia de src
4.
J Biol Chem ; 272(39): 24684-90, 1997 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-9305939

RESUMO

In vascular smooth muscle cells, the induction of early growth response genes involves the Janus kinase (JAK)/signal transducer and activators of transcription (STAT) and the Ras/Raf-1/mitogen-activated protein kinase cascades. In the present study, we found that electroporation of antibodies against MEK1 or ERK1 abolished vascular smooth muscle cell proliferation in response to either platelet-derived growth factor or angiotensin II. However, anti-STAT1 or -STAT3 antibody electroporation abolished proliferative responses only to angiotensin II and not to platelet-derived growth factor. AG-490, a specific inhibitor of the JAK2 tyrosine kinase, prevented proliferation of vascular smooth muscle cells, complex formation between JAK2 and Raf-1, the tyrosine phosphorylation of Raf-1, and the activation of ERK1 in response to either angiotensin II or platelet-derived growth factor. However, AG-490 had no effect on angiotensin II- or platelet-derived growth factor-induced Ras/Raf-1 complex formation. Our results indicate that: 1) STAT proteins play an essential role in angiotensin II-induced vascular smooth muscle cell proliferation, 2) JAK2 plays an essential role in the tyrosine phosphorylation of Raf-1, and 3) convergent mitogenic signaling cascades involving the cytosolic kinases JAK2, MEK1, and ERK1 mediate vascular smooth muscle cell proliferation in response to both growth factor and G protein-coupled receptors.


Assuntos
Angiotensina II/farmacologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Divisão Celular/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas , Transativadores/metabolismo , Animais , Proteínas de Ligação ao GTP/metabolismo , Janus Quinase 2 , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Ratos , Transdução de Sinais
5.
Am J Physiol ; 273(2 Pt 1): C548-57, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9277352

RESUMO

In A6 distal nephron cells, short-circuit current (Isc) was increased by basolateral exposure to prostaglandin E2 (PGE2; peak response at 1 microM). The effect was only partially abolished by either apical amiloride, an Na+ channel blocker, or 5-nitro-2-(3-phenylpropylamino)benzoic acid (NPPB), a Cl- channel blocker. In apical cell-attached patches, we observed a 7-pS Cl- channel with a linear current-voltage relationship, a reversal potential near resting membrane potential, and open probability > 0.5. The channel was blocked by diphenylamine-2-carboxylate, glibenclamide, and NPPB but not by 4,4'-diisothiocyanostilbene-2,2'-disulfonic acid. The frequency of observed Cl- channel activity increased 7-fold with 10-min exposure to PGE2 and 3.7-fold with longer (10-50 min) exposure to PGE2. The PGE2-induced increase in Cl- channel activity was due primarily to an increase in the number of functional channels. The following conclusions were made: 1) activation of apical, 7-pS Cl- channels in A6 cells accounts for the PGE2-induced increase in the amiloride-insensitive Isc, and 2) 7-pS Cl- channel activation was mediated via an increase in channel density without substantial effects on channel kinetics.


Assuntos
Canais de Cloreto/metabolismo , Dinoprostona/farmacologia , Néfrons/efeitos dos fármacos , Néfrons/metabolismo , Amilorida/farmacologia , Linhagem Celular , Membrana Celular/metabolismo , Canais de Cloreto/fisiologia , Dinoprostona/metabolismo , Condutividade Elétrica , Eletrofisiologia , Membranas Intracelulares/metabolismo , Néfrons/citologia , Distribuição Tecidual
6.
Am J Physiol ; 272(6 Pt 1): C2019-30, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9227431

RESUMO

The binding of vasoactive peptides to their respective G protein-coupled receptors has been implicated in the pathogenesis of vascular smooth muscle cell proliferation, leading to the development of hypertension, arteriosclerosis, and restenosis after vascular injury. We previously showed that the cytosolic tyrosine kinase pp60c-src is crucial for angiotensin II (ANG II)-induced activation of the protooncogene p21ras. Therefore, we investigated the role of pp60c-src and p21ras in rat aortic smooth muscle cell proliferation induced by several G protein-coupled receptors. ANG II, endothelin-1, or thrombin increased cell proliferation and DNA synthesis. Electroporation of anti-pp60c-src antibodies into cells abolished proliferation in response to these G protein-coupled receptor ligands but not in response to platelet-derived growth factor-BB (PDGF-BB). In contrast, electroporation of anti-p21ras antibody completely blocked DNA synthesis and cell proliferation in response to ANG II, endothelin-1, thrombin, and PDGF-BB. Our data indicate that the pp60c-src tyrosine kinase is necessary and specific for vascular smooth muscle cell proliferation and DNA synthesis in response to G protein-coupled receptors but not classic growth factor receptors.


Assuntos
Proteínas de Ligação ao GTP/fisiologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Receptores de Superfície Celular/fisiologia , Angiotensina II/farmacologia , Animais , Anticorpos/farmacologia , Aorta , Becaplermina , Compostos de Bifenilo/farmacologia , Divisão Celular/efeitos dos fármacos , Células Cultivadas , DNA/biossíntese , Eletroporação , Endotelina-1/farmacologia , Imidazóis/farmacologia , Cinética , Losartan , Músculo Liso Vascular/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Proto-Oncogênicas c-sis , Ratos , Receptores de Angiotensina/efeitos dos fármacos , Receptores de Angiotensina/fisiologia , Receptores de Superfície Celular/efeitos dos fármacos , Receptores de Endotelina/efeitos dos fármacos , Receptores de Endotelina/fisiologia , Receptores do Fator de Crescimento Derivado de Plaquetas/efeitos dos fármacos , Receptores do Fator de Crescimento Derivado de Plaquetas/fisiologia , Receptores de Trombina/efeitos dos fármacos , Receptores de Trombina/fisiologia , Tetrazóis/farmacologia , Trombina/farmacologia
7.
J Biol Chem ; 272(19): 12373-9, 1997 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-9139682

RESUMO

Angiotensin II (Ang II) and insulin-like growth factor I (IGF I) stimulate intracellular signaling events through binding to their respective G-protein-coupled and growth factor receptors. In rat aortic vascular smooth muscle cells, IGF I (20 ng/ml) induced a sustained (>30 min) increase in the tyrosine phosphorylation of both Src-homology 2 domain-docking insulin receptor substrate 1 (IRS-1) and Src-homology 2-binding tyrosine phosphatase 1D (PTP-1D). In addition, IGF I stimulated PTP-1D phosphatase activity. Ang II (10(-7) M) also increased the tyrosine phosphorylation of IRS-1 (4-fold), PTP-1D (5-fold), and PTP-1D activity (3-4-fold), but with a more transient time course. Ang II also induced PTP-1D.IRS-1 complex formation. These Ang II-induced events were not affected by preincubation with an anti-IGF I antibody, suggesting that Ang II's actions were not mediated via the autocrine secretion of IGF I. Anti-PTP-1D antibody electroporation attenuated Ang II-induced PTP-1D.IRS-1 complex formation and PTP-1D tyrosine phosphorylation and activation. Our findings show that the tyrosine phosphorylation of IRS-1 and PTP-1D represents a convergent intracellular signaling cascade stimulated by both growth factor (i.e. IGF I) and G-protein-coupled (i.e. AT1) receptors.


Assuntos
Angiotensina II/farmacologia , Músculo Liso Vascular/enzimologia , Fosfoproteínas/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Tirosina/metabolismo , Animais , Anticorpos , Eletroporação , Ativação Enzimática , Proteínas Substratos do Receptor de Insulina , Fator de Crescimento Insulin-Like I/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular , Substâncias Macromoleculares , Masculino , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Ratos , Ratos Sprague-Dawley
8.
J Biol Chem ; 272(1): 594-600, 1997 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-8995302

RESUMO

Mutations in a Cl- channel (cystic fibrosis transmembrane conductance regulator or CFTR) are responsible for the cystic fibrosis (CF) phenotype. Increased Na+ transport rates are observed in CF airway epithelium, and recent studies suggest that this is due to an increase in Na+ channel open probability (Po). The Xenopus renal epithelial cell line, A6, expresses both cAMP-activated 8-picosiemen (pS) Cl- channels and amiloride-sensitive 4-pS Na+ channels, and provides a model system for examining the interactions of CFTR and epithelial Na+ channels. A6 cells express CFTR mRNA, as demonstrated by reverse transcriptase-polymerase chain reaction and partial sequence analysis. A phosphorothioate antisense oligonucleotide, complementary to the 5' end of the open reading frame of Xenopus CFTR, was used to inhibit functional expression of CFTR in A6 cells. Parallel studies utilized the corresponding sense oligonucleotide as a control. CFTR protein expression was markedly reduced in cells incubated with the antisense oligonucleotide. Incubation of A6 cells with the antisense oligonucleotide led to inhibition of forskolin-activated amiloride-insensitive short circuit current (Isc). After a 30-min exposure to 10 microM forskolin, 8-pS Cl- channel activity was detected in only 1 of 31 (3%) cell-attached patches on cells treated with antisense oligonucleotide, compared to 5 of 19 (26%) patches from control cells. A shift in the single-channel current-voltage relationship derived from antisense-treated cells was also consistent with a reduction in Cl- reabsorption. Both amiloride-sensitive Isc and Na+ channel Po were significantly increased in antisense-treated, forskolin-stimulated A6 cells, when compared with forskolin-stimulated controls. These data suggest that the regulation of Na+ channels by CFTR is not limited to respiratory epithelia and to epithelial cells in culture overexpressing CFTR and epithelial Na+ channels.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Fibrose Cística/fisiopatologia , Canais de Sódio/fisiologia , Amilorida/farmacologia , Animais , Células Cultivadas , Técnica Indireta de Fluorescência para Anticorpo , Expressão Gênica/efeitos dos fármacos , Modelos Biológicos , Oligonucleotídeos Antissenso/farmacologia , Técnicas de Patch-Clamp , Fenótipo , Sódio/fisiologia , Bloqueadores dos Canais de Sódio , Xenopus laevis
9.
Am J Physiol ; 271(5 Pt 2): F994-1003, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8945993

RESUMO

Platelet-derived growth factor (PDGF)-induced Ca2+ signaling mechanisms were examined in cultured rat glomerular mesangial cells. PDGF-BB stimulated the tyrosine phosphorylation of phospholipase C (PLC)-gamma 1, the formation of a PLC-gamma 1/PDGF-beta receptor membrane complex, and the generation of intracellular inositol 1,4,5-trisphosphate (IP3). Preincubation with a tyrosine kinase inhibitor (genistein) abolished these PDGF-induced responses. Activation of 1-pS Ca2+ channels in cell-attached patches by intrapipette PDGF-BB was also abolished by tyrosine kinase inhibition. In the absence of PDGF-BB, channels were activated in cell-attached patches exposed to intrapipette thapsigargin (IP3-independent releaser of intracellular Ca2+ stores) and in excised inside-out patches exposed to increasing "cytoplasmic" Ca2+ (10(-8) to 10(-6) M). In cell-attached patches, channel activation by PDGF-BB was abolished when extracellular Ca2+ was < 1 mM. In glomerular mesangial cells 1) PDGF-BB stimulates tyrosine phosphorylation of PLC-gamma 1, PDGF-beta receptor/PLC-gamma 1 membrane complex formation, IP3 production, and 1-pS Ca2+ channel activity; 2) all four PDGF-induced responses are abolished by tyrosine kinase inhibition; 3) PDGF receptor-operated Ca2+ channels are sensitive to both intra- and extracellular Ca2+.


Assuntos
Canais de Cálcio/metabolismo , Mesângio Glomerular/metabolismo , Isoenzimas/fisiologia , Receptores do Fator de Crescimento Derivado de Plaquetas/fisiologia , Fosfolipases Tipo C/fisiologia , Animais , Becaplermina , Cálcio/metabolismo , Células Cultivadas , Espaço Extracelular/metabolismo , Mesângio Glomerular/citologia , Mesângio Glomerular/efeitos dos fármacos , Inositol 1,4,5-Trifosfato/metabolismo , Membranas Intracelulares/metabolismo , Masculino , Técnicas de Patch-Clamp , Fosfolipase C gama , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteína Quinase C/fisiologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-sis , Ratos , Ratos Sprague-Dawley , Receptor beta de Fator de Crescimento Derivado de Plaquetas , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo
10.
Am J Physiol ; 271(2 Pt 2): F451-60, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8770179

RESUMO

We used patch-clamp methods to investigate the effects of basolateral endothelin-1 (ET-1) on the amiloride-sensitive Na+ channel in A6 distal nephron cells. One hundred picomolar ET-1 decreased channel activity via an increase in mean time closed (P < 0.01, n = 10). Channel inhibition by pM ET-1 was mimicked by an ET-B receptor agonist (P < 0.05, n = 7) and was prevented by ET-B antagonists (P = 0.14, n = 10) but not by an ET-A antagonist (P < 0.05, n = 4). With the inhibitory ET-B receptor blocked, higher doses of ET-1 (10 nM) actually increased channel activity through an increase in mean time open (P < 0.001, n = 12). The current-voltage relationship and the number of channels were not changed by basolateral ET-1 exposure. We conclude that 1) basolateral ET-1 regulates amiloride-sensitive Na+ channels; 2) binding of picomolar ET-1 to ET-B receptors inhibits, whereas the binding of nanomolar ET-1 to a different ET receptor (likely ET-A) stimulates, channel activity; and 3) these dose-dependent, distal nephron responses provide a potential mechanism for the in vivo natriuresis and antinatriuresis observed in response to "subpressor" and "pressor" concentrations of ET-1, respectively.


Assuntos
Amilorida/farmacologia , Endotelina-1/farmacologia , Néfrons/metabolismo , Canais de Sódio/efeitos dos fármacos , Animais , Linhagem Celular , Membrana Celular/metabolismo , Relação Dose-Resposta a Droga , Néfrons/citologia , Concentração Osmolar , Técnicas de Patch-Clamp , Receptor de Endotelina B , Receptores de Endotelina/fisiologia , Bloqueadores dos Canais de Sódio , Xenopus laevis
11.
Am J Physiol ; 270(6 Pt 1): C1834-42, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8764169

RESUMO

Angiotensin II (ANG II)-induced, activation of phospholipase C (PLC) and Ca(2+)-dependent Cl-channels is an important signal transduction pathway for mesangial cell contraction and growth. Although ANG II receptors are traditionally though to be G protein coupled, recent evidence suggests that they may also mediate protein tyrosine phosphorylation. In cultured rat mesangial cells, 10(-7) MANG II stimulated the tyrosine phosphorylation of PLC-gamma 1 and elevation of intracellular inositol 1,4,5-trisphosphate (IP3) and Ca2+ levels; peak response occurred within 0.5 min. In cell-attached patches, ANG II stimulated the activity of Ca(2+)-dependent, 3- to 4-pS Cl-channels (number of channels x open probability) from 0.063 +/- 0.022 to 0.77 +/- 0.20. Tyrosine kinase inhibition with genistein or herbimycin A blocked all four ANG II-induced responses. We conclude the following. 1) Stimulation of inositol phosphate hydrolysis by PLC, release of IP3-dependent intracellular Ca2+ stores, and activation of Ca(2+)-dependent C1-channels by ANG II are dependent on the tyrosine phosphorylation of PLC-gamma 1.2) This ANG II-induced signal transduction cascade provides a possible mechanism for both the contractile and growth-stimulating effects of ANG II on glomerular mesangial cells.


Assuntos
Angiotensina II/farmacologia , Canais de Cloreto/efeitos dos fármacos , Canais de Cloreto/metabolismo , Mesângio Glomerular/metabolismo , Isoenzimas/metabolismo , Fosfolipases Tipo C/metabolismo , Tirosina/metabolismo , Animais , Cálcio/metabolismo , Mesângio Glomerular/citologia , Inositol 1,4,5-Trifosfato/biossíntese , Membranas Intracelulares/metabolismo , Masculino , Fosfolipase C gama , Fosforilação , Ratos , Ratos Sprague-Dawley
12.
Am J Physiol ; 270(5 Pt 2): F798-805, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-8928841

RESUMO

To investigate the effects of luminal adenosine on amiloride-sensitive Na+ channels, we applied the cell-attached patch-clamp technique to A6 distal nephron cells. Exposure to luminal 30 nM adenosine increased number of channels x open probability (NP0) from 0.38 +/- 0.08 to 0.77 +/- 0.09 (means +/- SE; P < 0.01, n = 17). Luminal exposure to an A1-receptor antagonist (30 nM 8-cyclopentyl-1,3-dipropylxanthine) abolished (P = 0.17, n = 11), whereas an A1 agonist (30 nM N6-cyclohexyladenosine) reproduced (P < 0.02, n = 6) the stimulatory effect of 30 nM adenosine. In contrast, higher concentrations of luminal adenosine (1 or 10 microM) decreased NP0 from 0.65 +/- 0.09 to 0.24 +/- 0.10 (P < 0.02, n = 11) and from 0.80 +/- 0.11 to 0.19 +/- 0.03 (P < 0.01, n = 8), respectively. Channel inhibition by high-dose luminal adenosine was abolished by an A2 antagonist (30 microM 3,7-dimethyl-1-propargylxanthine; P = 0.2, n = 10) and mimicked by an A2 agonist (100 nM CGS-21680 hydrochloride; P < 0.0005, n = 8). We conclude that 1) purinergic regulation of distal nephron Na+ channels is mediated by stimulatory apical A1 receptors and inhibitory apical A2 receptors; 2) basal urinary adenosine concentrations (in nM) would stimulate Na+ reabsorption, whereas higher urinary concentrations (in microM), e.g., renal ischemia and elevations in filtered NaCl load, would increase Na+ excretion; and 3) urinary adenosine may be involved in feedback regulation of distal nephron Na+ transport.


Assuntos
Amilorida/farmacologia , Néfrons/metabolismo , Receptores Purinérgicos P1/fisiologia , Canais de Sódio/efeitos dos fármacos , Adenosina/análogos & derivados , Adenosina/farmacologia , Linhagem Celular , Relação Dose-Resposta a Droga , Néfrons/citologia , Técnicas de Patch-Clamp , Fenetilaminas/farmacologia , Agonistas do Receptor Purinérgico P1 , Antagonistas de Receptores Purinérgicos P1 , Teobromina/análogos & derivados , Teobromina/farmacologia , Xantinas/farmacologia
13.
J Clin Invest ; 97(10): 2332-41, 1996 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-8636414

RESUMO

We investigated the signaling pathways mediating 1-pS Ca2+ channel activation by PDGF in cultured rat mesangial cells. In cell-attached patches, intrapipette PDGF-BB (PDGF B chain homodimer isoform) (50 ng/ml) dramatically stimulates channel activity (P < 0.003, n = 6). Tyrosine kinase inhibition (100 microM genistein or 10 microM tryphostin 9) abolished PDGF-induced channel activation (P < 0.02, n = 6). In excised patches, the effect of tyrosine kinase inhibition could be reversed by 200 microM GTPgammaS (P < 0.02, n = 4). In contrast, 200 microM GDPbetaS inhibited PDGF-induced channel activity (P < 0.04, n = 6). Pertussis toxin (250 ng/ml) had no effect on PDGF-induced channel activity (P = 0.45, n = 6). When excised patches were exposed to anti-Ras antibody (5 microg/ml), PDGF-induced channel activity was abolished (P < 0.002, n = 11). Western immunoblots revealed that PDGF-BB binding stimulates the formation of a membrane-bound complex consisting of growth factor receptor-binding protein 2, son of sevenless, and the PDGF-beta receptor. Complex formation was abolished by genistein. In mesangial cells, the intrinsic tyrosine kinase activity of the PDGF-beta receptor stimulates the formation of a membrane-bound growth factor receptor-binding protein 2/son of sevenless/PDGF-beta receptor complex and activation of the pertussis toxin-insensitive GTP-binding protein, p21-Ras, which leads to the opening of 1-pS Ca2+ channels.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Canais de Cálcio/efeitos dos fármacos , Mesângio Glomerular/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Tirosina/metabolismo , Animais , Proteína Adaptadora GRB2 , Proteínas de Ligação ao GTP/fisiologia , Masculino , Proteínas de Membrana/fisiologia , Fosforilação , Proteínas Tirosina Quinases/fisiologia , Proteínas/fisiologia , Ratos , Ratos Sprague-Dawley , Proteínas Son Of Sevenless , Proteínas ras/fisiologia
14.
Clin Exp Pharmacol Physiol ; 23(1): 83-8, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8713501

RESUMO

1. Angiotensin II (AngII)-induced, activation of phospholipase C (PLC) and Ca2+-dependent Cl- channels is an important signal transduction pathway for the regulation of vascular smooth muscle cell (VSMC) and glomerular mesangial cell contraction and growth. While AT receptors are traditionally thought to be G-protein coupled to the beta isoform of PLC, recent evidence suggests that in some tissues AT receptors may also activate the PLC-gamma isoform via tyrosine phosphorylation. 2. By western analysis, we identified PLC-gamma1 in the above cell types. We found that within 3 min of exposure to 10(-7) mol/L AngII, tyrosine phosphorylation of PLC-gamma1 was observed; however, peak response (>3-fold increase) occurred within 0.5 min. In addition, pre-incubation of these cells with the tyrosine kinase inhibitor genistein blocked the tyrosine phosphorylation of PLC-gamma1 by AngII. In contrast, preincubation with the tyrosine phosphatase inhibitor sodium vanadate increased the levels of tyrosine phosphorylation of PLC-gamma1. Similar results were found when intracellular levels of 1,4,5-IP3 were measured after AngII exposure. 3. By using patch clamp techniques on cultured rat mesangial cells exposed to AngII, we found that the release of 1,4,5-IP3-sensitive intracellular Ca2+ stores stimulated low conductance Cl- channels. Preincubation with genistein, abolished the usual 10-fold increase in Cl- channel activity observed with AngII. 4. Therefore, we conclude that in VSMC and glomerular mesangial cells (i) AngII transiently stimulates PLC activity via tyrosine phosphorylation of the gamma1 isoenzyme, (ii) tyrosine phosphorylation of PLC-gamma1 and production of 1,4,5-IP3 in response to AngII is dramatically inhibited by tyrosine kinase inhibition and stimulated by tyrosine phosphatase inhibition, (iii) activation of Ca2+-dependent Cl- channels by AngII-induced release of 1,4,5-IP3-dependent intracellular Ca2+ stores is also abolished by tyrosine kinase inhibition. In summary, this AngII-induced signal transduction cascade provides a possible mechanism for both the contractile and growth-stimulating effects of AngII on VSMC and glomerular mesangial cells.


Assuntos
Angiotensina II/farmacologia , Mesângio Glomerular/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Tirosina/metabolismo , Animais , Aorta/efeitos dos fármacos , Canais de Cloreto/metabolismo , Fosforilação , Ratos
15.
Clin Exp Pharmacol Physiol ; 23(1): 89-94, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8713502

RESUMO

1. In response to vasoactive peptides (e.g. angiotensin II (AngII), vasopressin, endothelin-1, platelet-activating factor), glomerular mesangial cell contraction is mediated through activation of a Ca2+-dependent Cl- conductance that, in turn, promotes membrane depolarization and voltage-activated Ca2+ entry. 2. Using patch clamp technology, our laboratory was the first to characterize a candidate Ca2+-dependent, 4 pS Cl- channel that is stimulated by vasoactive peptides in cultured rat mesangial cells. In the absence of extracellular insulin, the activation of Cl- channels by AngII is abolished. We find that Cl- channel sensitivity to intracellular Ca2+ and the membrane density of AngII receptors is also dependent on the presence of insulin. 3. Our studies also show that high extracellular glucose interferes with mesangial cell IP3 generation and Cl- channel stimulation. Importantly, we find that the insulin-dependency of Cl- channels occurs within the range of plasma insulin concentrations observed in normal, obese, hypertensive and diabetic humans (i.e. 1-100 mu U/mL). Similarly, normal regulation of Cl- channel activity is also modulated by glucose concentrations commonly observed in the plasma of diabetic humans (5-30 mmol/L). 4. There is substantial evidence, both in diabetic humans and animal models, that the provision of insulin and improved glycaemic control corrects or prevents glomerular hyperfiltration. The requirement for normal insulin and glucose levels, for the proper regulation of the 4 pS Cl- channel, provides a mechanism for impaired Ca2+ uptake and contraction observed in glomerular mesangial cells in association with insulin deficiency and hyperglocaemia.


Assuntos
Canais de Cloreto/efeitos dos fármacos , Nefropatias Diabéticas/metabolismo , Glucose/farmacologia , Insulina/farmacologia , Animais , Modelos Biológicos
16.
Cell Signal ; 8(1): 21-6, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8777137

RESUMO

Angiotensin II is a potent vasoconstrictor that is important in the control of systemic blood pressure. All the hemodynamic effects of angiotensin II result from the AT1 receptor which has the structural features of a seven transmembrane receptor. Both in cultured rat aortic smooth muscle cells and rat glomerular mesangial cells, angiotensin II stimulates the rapid tyrosine phosphorylation of phospholipase C-gamma 1 (PLC-gamma 1). Tyrosine kinase inhibitors that block this phosphorylation also block the angiotensin II-mediated production of 1,4,5 inositol trisphosphate (1,4,5-IP3) and the intracellular release of Ca2+. The cellular tyrosine kinase c-src appears to play a critical role in the angiotensin II-stimulated tyrosine phosphorylation of PLC-gamma 1 and the generation of 1,4,5-IP3. We have also found that angiotensin II stimulates the tyrosine phosphorylation and activation of the JAK family of intracellular kinases. This in turn activates the STAT family of transcription factors. Angiotensin II, working through the AT1 receptor, uses tyrosine phosphorylation as a mechanism to convey signals from the cell surface to the cell nucleus.


Assuntos
Angiotensina II/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Proteínas Proto-Oncogênicas , Receptores Proteína Tirosina Quinases/fisiologia , Transdução de Sinais/fisiologia , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Proteínas de Ligação a DNA/fisiologia , Humanos , Isoenzimas/fisiologia , Janus Quinase 1 , Janus Quinase 2 , Modelos Moleculares , Fosfolipase C gama , Fosforilação , Proteínas Tirosina Quinases/fisiologia , Ratos , Receptores de Angiotensina/química , Receptores de Angiotensina/fisiologia , Sistema Renina-Angiotensina/fisiologia , Fator de Transcrição STAT1 , Fator de Transcrição STAT2 , Transativadores/fisiologia , Fosfolipases Tipo C/fisiologia
17.
Kidney Int ; 49(1): 82-7, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8770952

RESUMO

This study was designed to test the hypothesis that the antikaliuresis caused by trimethoprim could be diminished by alkalinizing the luminal fluid in the CCD, thereby converting trimethoprim from its cationic, active form to an electroneutral, inactive, form. Trimethoprim-induced inhibition of transepithelial Na+ transport was examined in A6 distal nephron cells by analysis of short circuit current. The voltage-dependence of the trimethoprim-induced block of Na+ channels was examined with patch clamp recordings of A6 cells. The antikaliuretic effect of trimethoprim was examined in vivo in rats pretreated with deoxycorticosterone and with NH4Cl to lower urine pH, and in rats also receiving acetazolamide to raise urine pH. We found that the concentration of trimethoprim required to inhibit the amiloride sensitive component of short circuit current by 50% (IC50) was 340 microM (at pH 8.2) and 50 microM (at pH 6.3). The IC50S of protonated trimethoprim were similar (34 microM at pH 8.2 and 45 microM at pH 6.3). The mean time open for the high selectivity, Na+ channel was reduced from 1679 +/- 387 msec to 502 +/- 98 msec with addition of 10-5 M trimethoprim to patch pipette solution at the resting membrane potential (-Vpipette = 0 mV). further decreases in mean time open were observed as -Vpipette was reduced (that is, apical membrane hyperpolarization) to -40 mV (mean time open = 217 +/- 85 msec) and to -80 mV (mean time open = 69 +/- 13 msec). In vivo, trimethoprim caused a > 50% reduction in potassium (K+) excretion due primarily to a fall in the [K+] in the lumen of the terminal CCD. This effect of trimethoprim was markedly attenuated in an alkaline urine induced by acetazolamide. We conclude that it is the charged, protonated species of trimethoprim which blocks epithelial Na+ channels. Increasing urinary pH decreases the concentration of the charged species of trimethoprim and minimizes its antikaliuretic effect.


Assuntos
Anti-Infecciosos Urinários/administração & dosagem , Túbulos Renais/efeitos dos fármacos , Canais de Potássio/metabolismo , Potássio/urina , Canais de Sódio/metabolismo , Trimetoprima/administração & dosagem , Animais , Concentração de Íons de Hidrogênio , Injeções Intraperitoneais , Túbulos Renais/metabolismo , Masculino , Potenciais da Membrana , Técnicas de Patch-Clamp , Canais de Potássio/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Bloqueadores dos Canais de Sódio
18.
Kidney Int ; 48(4): 1158-66, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8569077

RESUMO

Our single channel work has characterized two ion channels capable of depolarizing mesangial cells and activating classic, voltage-activated Ca2+ channels in response to growth-stimulatory peptides (such as Ang II, ET and insulin): (1) Ca(2+)-dependent, 4 pS Cl- channel promoting Cl- efflux; and (2) Ca(2+)-dependent, 27 pS nonselective cation channels promoting cation influx. We have also characterized a third channel which provides an alternative, receptor-operated pathway for Ca2+ entry in response to the growth factor, PDGF: (3) Ca(2+)-permeable, 1 pS cation channel. Consistent with our model of mesangial cell signal transduction (Fig. 1), these three mesangial cell ion channels are activated by binding of growth factors to membrane receptors (Fig. 8). Defective channel regulation, such as occurs in early diabetes mellitus, would promote mesangial cell relaxation and pathogenic glomerular hyperfiltration. Glomerular hyperfiltration and hypertension have been proposed to be major pathogenic factors in renal disease progression [4, 29, 38, 39]. Compensatory renal growth factor responses initially provide adaptive changes in glomerular hemodynamics after loss of functional renal mass. However, chronic stimulation of these mesangial cell ion channels by renal growth factors would promote sustained extracellular Ca2+ entry, resulting in mesangial cell contraction and growth, and progressive decreases in Kf and GFR. Eventually, this process leads to irreversible renal damage due to the development of glomerulosclerosis and interstitial fibrosis.


Assuntos
Mesângio Glomerular/metabolismo , Substâncias de Crescimento/metabolismo , Canais Iônicos/metabolismo , Angiotensina II/farmacologia , Animais , Cálcio/metabolismo , Canais de Cloreto/efeitos dos fármacos , Canais de Cloreto/metabolismo , Ciclosporina/farmacologia , Nefropatias Diabéticas/etiologia , Endotelinas/farmacologia , Furosemida/farmacologia , Mesângio Glomerular/citologia , Mesângio Glomerular/efeitos dos fármacos , Humanos , Insulina/farmacologia , Canais Iônicos/efeitos dos fármacos , Transporte de Íons/efeitos dos fármacos , Modelos Biológicos , Receptores de Angiotensina/metabolismo
20.
Am J Physiol ; 269(3 Pt 2): F389-97, 1995 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-7573488

RESUMO

Abnormal cellular ion homeostasis is a well-recognized component of diabetic glomerular disease. In cultured rat glomerular mesangial cells, we have previously shown that insulin regulates Ca(2+)-dependent activation of 4-pS Cl- channels and 27-pS nonselective cation channels (NSCC) by angiotensin II (ANG II). To assess whether extracellular glucose also affects mesangial ion channels, we applied patch-clamp techniques to cells incubated in constant insulin (100 mU/ml) and either "normal" (5 mM) or "high" (30 mM) glucose for 1 wk. In normal glucose, 100 nM ANG II increased Cl- and NSCC activity by > 16-fold and > 60-fold, respectivley. Direct release of intracellular Ca2+ ([Ca2+]i) stores (0.25 microM thapsigargin) mimicked ANG II-induced channel stimulation. In high glucose, Cl- and NSCC stimulation by ANG II was attenuated (< 7-fold), whereas channel activation by thapsigargin was unaffected. Protein kinase C (PKC) inhibition (30-min exposure to 0.5 microM calphostin) or downregulation (24-h exposure to 0.1 microM 4 beta-phorbol 12-myristate 13-acetate), but not aldose reductase inhibition (0.5 mM sorbinil), restored channel responsiveness to ANG II despite high glucose. Channel responsiveness was also restored if mesangial cells were coincubated in both high glucose and 500 microM myo-inositol. Acute exposure to a synthetic diacylglycerol (100 microM 1-oleoyl-2-acetyl glycerol) reestablished channel unresponsiveness to ANG II. We conclude the following in rat mesangial cell cultures: 1) Activation of Ca(2+)-dependent Cl- and NSCCs by ANG II is reduced by high extracellular glucose.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Angiotensina II/farmacologia , Espaço Extracelular/metabolismo , Mesângio Glomerular/metabolismo , Glucose/metabolismo , Canais Iônicos/efeitos dos fármacos , Aldeído Redutase/fisiologia , Animais , Células Cultivadas , Diglicerídeos/fisiologia , Mesângio Glomerular/citologia , Inositol/fisiologia , Masculino , Técnicas de Patch-Clamp , Proteína Quinase C/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de Angiotensina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA