Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Am J Physiol Renal Physiol ; 307(3): F287-302, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24920755

RESUMO

In rabbit proximal tubular cells, ANG II type 2-receptor (AT2)-induced arachidonic acid release is PLA2 coupled and dependent of G protein ßγ (Gßγ) subunits. Moreover, ANG II activates ERK1/2 and transactivates EGFR via a c-Src-dependent mechanism. Arachidonic acid has been shown to mimic this effect, at least in part, by an undetermined mechanism. In this study, we determined the effects of ANG II on fibronectin expression in cultured rabbit proximal tubule cells and elucidated the signaling pathways associated with such expression. We found that ANG II and transfection of Gßγ subunits directly increased fibronectin protein expression, and this increase was inhibited by overexpression of ß-adrenergic receptor kinase (ßARK)-ct or DN-Src. Moreover, ANG II-induced fibronectin protein expression was significantly abrogated by the AT2 receptor antagonist PD123319. In addition, inhibition of cystolic PLA2 diminished ANG II-induced fibronectin expression. Endogenous arachidonic acid mimicked ANG II-induced fibronectin expression. We also found that overexpression of Gßγ subunits induced c-Src, ERK1/2, and EGFR tyrosine phosphorylation, which can be inhibited by overexpression of ßARK-ct or DN-Src. Gßγ also induced c-Src SH2 domain association with the EGFR. Supporting these findings, in rabbit proximal tubular epithelium, immunoblot analysis indicated that ßγ expression was significant. Interestingly, arachidonic acid- and eicosatetraenoic acid-induced responses were preserved in the presence of ßARK-ct. This is the first report demonstrating the regulation of EGFR, ERK1/2, c-Src, and fibronectin by Gßγ subunits in renal epithelial cells. Moreover, this work demonstrates a role for Gßγ heterotrimeric proteins in ANG II, but not arachidonic acid, signaling in renal epithelial cells.


Assuntos
Angiotensina II/farmacologia , Ácido Araquidônico/metabolismo , Fibronectinas/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Animais , Proteína Tirosina Quinase CSK , Células Cultivadas , Receptores ErbB/metabolismo , Imidazóis/farmacologia , Túbulos Renais Proximais/citologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Modelos Animais , Piridinas/farmacologia , Coelhos , Receptor Tipo 2 de Angiotensina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Quinases da Família src/metabolismo
2.
J Biol Chem ; 286(3): 1828-35, 2011 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-21084311

RESUMO

The Ca(2+)-sensing receptor (CaR) regulates salt and water transport in the kidney as demonstrated by the association of gain of function CaR mutations with a Bartter syndrome-like, salt-wasting phenotype, but the precise mechanism for this effect is not fully established. We found previously that the CaR interacts with and inactivates an inwardly rectifying K(+) channel, Kir4.1, which is expressed in the distal nephron that contributes to the basolateral K(+) conductance, and in which loss of function mutations are associated with a complex phenotype that includes renal salt wasting. We now find that CaR inactivates Kir4.1 by reducing its cell surface expression. Mutant CaRs reduced Kir4.1 cell surface expression and current density in HEK-293 cells in proportion to their signaling activity. Mutant, activated Gα(q) reduced cell surface expression and current density of Kir4.1, and these effects were blocked by RGS4, a protein that blocks signaling via Gα(i) and Gα(q). Other α subunits had insignificant effects. Knockdown of caveolin-1 blocked the effect of Gα(q) on Kir4.1, whereas knockdown of the clathrin heavy chain had no effect. CaR had no comparable effect on the renal outer medullary K(+) channel, an apical membrane distal nephron K(+) channel that is internalized by clathrin-coated vesicles. Co-immunoprecipitation studies showed that the CaR and Kir4.1 physically associate with caveolin-1 in HEK cells and in kidney extracts. Thus, the CaR decreases cell surface expression of Kir4.1 channels via a mechanism that involves Gα(q) and caveolin. These results provide a novel molecular basis for the inhibition of renal NaCl transport by the CaR.


Assuntos
Regulação da Expressão Gênica , Néfrons/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/biossíntese , Receptores de Detecção de Cálcio/metabolismo , Transdução de Sinais , Cloreto de Sódio/metabolismo , Animais , Transporte Biológico/genética , Caveolina 1/genética , Caveolina 1/metabolismo , Cadeias Pesadas de Clatrina/genética , Cadeias Pesadas de Clatrina/metabolismo , Vesículas Revestidas por Clatrina/genética , Vesículas Revestidas por Clatrina/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Potássio/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Ratos , Receptores de Detecção de Cálcio/genética , Desequilíbrio Hidroeletrolítico/genética , Desequilíbrio Hidroeletrolítico/metabolismo
3.
Am J Physiol Cell Physiol ; 300(3): C397-405, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21123730

RESUMO

The mechanical properties of tissues and cells including renal glomeruli are important determinants of their differentiated state, function, and responses to injury but are not well characterized or understood. Understanding glomerular mechanics is important for understanding renal diseases attributable to abnormal expression or assembly of structural proteins and abnormal hemodynamics. We use atomic force microscopy (AFM) and a new technique, capillary micromechanics, to measure the elastic properties of rat glomeruli. The Young's modulus of glomeruli was 2,500 Pa, and it was reduced to 1,100 Pa by cytochalasin and latunculin, and to 1,400 Pa by blebbistatin. Cytochalasin or latrunculin reduced the F/G actin ratios of glomeruli but did not disrupt their architecture. To assess glomerular biomechanics in disease, we measured the Young's moduli of glomeruli from two mouse models of primary glomerular disease, Col4a3(-/-) mice (Alport model) and Tg26(HIV/nl) mice (HIV-associated nephropathy model), at stages where glomerular injury was minimal by histopathology. Col4a3(-/-) mice express abnormal glomerular basement membrane proteins, and Tg26(HIV/nl) mouse podocytes have multiple abnormalities in morphology, adhesion, and cytoskeletal structure. In both models, the Young's modulus of the glomeruli was reduced by 30%. We find that glomeruli have specific and quantifiable biomechanical properties that are dependent on the state of the actin cytoskeleton and nonmuscle myosins. These properties may be altered early in disease and represent an important early component of disease. This increased deformability of glomeruli could directly contribute to disease by permitting increased distension with hemodynamic force or represent a mechanically inhospitable environment for glomerular cells.


Assuntos
Módulo de Elasticidade/fisiologia , Elasticidade/fisiologia , Nefropatias/fisiopatologia , Glomérulos Renais/fisiologia , Glomérulos Renais/fisiopatologia , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/patologia , Animais , Autoantígenos/genética , Colágeno Tipo IV/deficiência , Colágeno Tipo IV/genética , Módulo de Elasticidade/efeitos dos fármacos , Elasticidade/efeitos dos fármacos , Nefropatias/patologia , Glomérulos Renais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ratos , Ratos Sprague-Dawley
4.
Am J Physiol Renal Physiol ; 297(1): F36-45, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19420109

RESUMO

We previously found that the Ca(2+)-sensing receptor (CaR) interacts with and inactivates the inwardly rectifying K(+) channel Kir4.2 that is expressed in the kidney cortex and that has a COOH-terminal PDZ domain. To identify potential scaffolding proteins that could organize a macromolecular signaling complex involving the CaR and Kir4.2, we used yeast two-hybrid cloning with the COOH-terminal 125 amino acids (AA) of Kir4.2 as bait to screen a human kidney cDNA library. We identified two independent partial cDNAs corresponding to the COOH-terminal 900 AA of MUPP1, a protein containing 13 PDZ binding domains that is expressed in the kidney in tight junctions and lateral borders of epithelial cells. When expressed in human embryonic kidney (HEK)-293 cells, Kir4.2 coimmunoprecipitates reciprocally with MUPP1 but not with a Kir4.2 construct lacking the four COOH-terminal amino acids, Kir5.1, or the CaR. MUPP1 and Kir4.2 coimmunoprecipitate reciprocally from rat kidney cortex extracts. Coexpression of MUPP1 with Kir4.2 in HEK-293 cells leads to reduced cell surface expression of Kir4.2 as assessed by cell surface biotinylation. Coexpression of MUPP1 and Kir4.2 in Xenopus oocytes results in reduced whole cell currents compared with expression of Kir4.2 alone, whereas expression of Kir4.2DeltaPDZ results in minimal currents and is not affected by coexpression with MUPP1. Immunofluorescence studies of oocytes demonstrate that MUPP1 reduces Kir4.2 membrane localization. These results indicate that Kir4.2 interacts selectively with MUPP1 to affect its cell surface expression. Thus MUPP1 and Kir4.2 may participate in a protein complex in the nephron that could regulate transport of K(+) as well as other ions.


Assuntos
Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , Rim/citologia , Rim/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Feminino , Humanos , Proteínas de Membrana , Oócitos/citologia , Oócitos/metabolismo , Domínios PDZ/fisiologia , Técnicas de Patch-Clamp , Plasmídeos , Domínios e Motivos de Interação entre Proteínas/fisiologia , Ratos , Receptores de Detecção de Cálcio/metabolismo , Saccharomyces cerevisiae , Transfecção , Xenopus laevis
5.
Free Radic Biol Med ; 41(2): 213-21, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16814101

RESUMO

Previous studies from this laboratory have demonstrated a critical role of cytosolic phospholipase A2 (cPLA2) and arachidonic acid in angiotensin II (Ang II) AT2 receptor-mediated signal transduction in renal epithelium. In primary proximal tubular epithelial cells exposed to hydrogen peroxide (H2O2), both the selective cPLA2 inhibitors and the cPLA2 antisense oligonucleotides significantly attenuated H2O2-induced arachidonic acid liberation and activation of p38(SAPK), ERK1/2, and Akt1. This H2O2-induced kinase activation was significantly attenuated by a Src kinase inhibitor PP2, or by transient transfection of carboxyl-terminal Src kinase (CSK) that maintained Src in the dormant form. Under basal conditions, Src coimmunoprecipitated with epidermal growth factor receptor (EGFR), while H2O2 increased EGFR phosphorylation in the complex. We observed that inhibition of EGFR kinase activity with AG1478 significantly attenuated H2O2-induced p38(SAPK) and ERK1/2 activation, but did not inhibit Akt1 activation. Furthermore, it seems that p38(SAPK) is upstream of ERK1/2 and Akt1, since a p38(SAPK) inhibitor SB203580 significantly blocked H2O2-induced activation of ERK1/2 and Akt1. Interestingly, overexpression of the dominant-negative p38(SAPK) isoform alpha inhibited ERK1/2 but not Akt1 activation. Our observations demonstrate that in these nontransformed cells, activation of cPLA2 is a converging point for oxidative stress and Ang II, which share common downstream signaling mechanisms including Src and EGFR. In addition, p38(SAPK) provides a positive input to both growth and antiapoptotic signaling pathways induced by acute oxidative stress.


Assuntos
Citosol/enzimologia , Rim/metabolismo , Estresse Oxidativo , Fosfolipases A/metabolismo , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Sequência de Bases , Primers do DNA , DNA Complementar , Epitélio/metabolismo , Fosfolipases A2
6.
J Pharm Pharmacol ; 57(1): 111-6, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15638999

RESUMO

NAD(P)H oxidase contributes to the pathogenesis of cancer and cardiovascular diseases such as hypertension, atherosclerosis, restenosis, cardiac hypertrophy and heart failure. Plumbagin, a plant-derived naphthoquinone, has been shown to exert anticarcinogenic and anti-atherosclerosis effects in animals. However, the molecular mechanisms underlying these effects remain unknown. It is possible that the beneficial effect of plumbagin is due to the inhibition of NAD(P)H oxidase. Human embryonic kidney 293 (HEK293) and brain tumour LN229 cells express mainly Nox-4, a renal NAD(P)H oxidase. We have examined the effect of plumbagin on Nox-4 activity in HEK293 and LN229 cells using lucigenin-dependent chemiluminescence assay. Plumbagin inhibited the activity of Nox-4 in a time- and dose-dependent manner in HEK293 and LN229 cells. Production of superoxide in HEK293 cells was inhibited by diphenyleneiodonium (DPI), a NAD(P)H oxidase inhibitor. The superoxide production in HEK293 cells was NADPH- and NADH-dependent indicating that the superoxide was generated by a NAD(P)H oxidase in HEK293 cells, but not by the redox-cycling of lucigenin. Furthermore, plumbagin inhibited the superoxide production in Nox-4 transfected COS-7 cells. These results indicated that plumbagin directly interacted with Nox-4 and inhibited its activity.


Assuntos
NADPH Oxidases/antagonistas & inibidores , Naftoquinonas/farmacologia , Plumbaginaceae/química , Animais , Neoplasias Encefálicas/metabolismo , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Inibidores Enzimáticos/farmacologia , Humanos , Neoplasias Renais/metabolismo , NAD/farmacologia , NADP/farmacologia , NADPH Oxidase 4 , NADPH Oxidases/biossíntese , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Oniocompostos/farmacologia , Transfecção
7.
J Pharm Pharmacol ; 55(11): 1539-45, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14713365

RESUMO

Oestrogen is known to exert both genomic and non-genomic effects on target tissues. Unlike the genomic effects, the identity of receptors mediating the non-genomic effects of oestrogen remains controversial. 17beta-estradiol has been shown to activate membrane-bound guanylate cyclase GC-A in PC12 cells in a non-genomic manner. To examine whether 17beta-estradiol exerts a similar effect in other cell types, we measured the effect of 17beta-estradiol and tamoxifen, an anti-oestrogen, on guanylate cyclase activity in porcine kidney proximal tubular LLC-PK1 cells. 17beta-estradiol increased cGMP levels in LLC-PK1 cells. Interestingly, addition of tamoxifen also increased cGMP levels in a concentration-dependent manner in LLC-PK1 cells. The effects of both 17beta-estradiol and tamoxifen on guanylate cyclase activity were not additive, suggesting that oestrogen and tamoxifen activate the same enzyme. Similar phenomena were also observed in LLC-PK1 cell membrane preparation. LLC-PK1 cells do not express membrane-bound guanylate cyclase GC-B and express low levels of membrane-bound guanylate cyclase GC-C. Tamoxifen inhibited the activation of GC-A by atrial natriuretic factor (ANF). However, it did not affect membrane-bound guanylate cyclase GC-C stimulated by guanylin or Escherichia coli heat-stable toxin STa. These results indicate that 17beta-estradiol and tamoxifen activate GC-A in LLC-PK1 cells. Thus, tamoxifen functions as an agonist rather than an antagonist for the membrane oestrogen receptor coupled to the activation of GC-A.


Assuntos
Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Guanilato Ciclase/efeitos dos fármacos , Tamoxifeno/farmacologia , Animais , Membrana Celular/efeitos dos fármacos , Células Cultivadas , GMP Cíclico/metabolismo , Ativação Enzimática/efeitos dos fármacos , Hormônios Gastrointestinais/farmacologia , Guanilato Ciclase/metabolismo , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/enzimologia , Peptídeos Natriuréticos , Peptídeos/farmacologia , Suínos
8.
Hypertension ; 46(2): 419-25, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15998700

RESUMO

Internalization of a G-protein-coupled receptor (GPCR) is essential to the desensitization, endocytosis, and signal transduction of the receptor. It has been the general view that conventional homologous internalization of a GPCR requires activation of the G-protein(s) coupled to the receptor. However, whether and how GPCR-mediated G-protein-independent signals trigger receptor internalization remains unknown, although G-protein-independent internalization has been reported. Here we show that an angiotensin II (Ang II) type-1 (AT1) receptor mutant incapable of activating any G-protein still undergoes normal internalization. Substitution of Asp125 with Ala and Arg126 with Leu at the highly conserved DRY motif of the AT1 receptor disabled the ability of the receptor to activate G-proteins, as shown by various Ang II binding studies, GDP-GTP exchange, and inositol phosphate production assays. Surprisingly, the mutant internalized normally in the presence of Ang II and transactivated the epidermal growth factor receptor (EGFR). Similar to the wild-type receptor, overexpression of a dominant-negative K220R mutant GRK2 diminished the internalization of D125A-R126L but not the transactivation of EGFR. These data indicate that G-protein-independent specific signals may also trigger homologous internalizations of the AT1 receptor through beta-arrestin-dependent and -independent pathways, suggesting a possible mechanism for G-protein-independent activation of G-protein-coupled receptor kinases (GRKs). This may represent a general mechanism for triggering GPCR internalization.


Assuntos
Endocitose/fisiologia , Proteínas de Ligação ao GTP/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Transdução de Sinais/fisiologia , Substituição de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Receptores ErbB/metabolismo , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Ligantes , Mutação , Receptor Tipo 1 de Angiotensina/genética , Receptores Acoplados a Proteínas G/metabolismo , Ativação Transcricional
9.
Hypertension ; 44(6): 963-8, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15466660

RESUMO

Cyclophilin A (CypA), a receptor for the immunosuppressive agent cyclosporin A, is a cis-trans-peptidyl-prolyl isomerase (PPIase). It accelerates the cis-trans isomerization of prolyl-peptide bonds. CypA binds and regulates the activity of a variety of proteins. Atrial natriuretic factor (ANF) and its receptor membrane-bound guanylate cyclase-A (GC-A) are involved in the regulation of blood pressure. We examined whether CypA affects the activation of GC-A by ANF. The results showed that CypA associated with GC-A. Interestingly, binding of ANF to GC-A released CypA. Transfection of CypA inhibited ANF-stimulated GC-A activity, indicating that CypA functions as an endogenous inhibitor for GC-A activation. CypA also inhibits the activity of guanylate cyclase-C (GC-c), the catalytic domain of GC-A, indicating that CypA interacts with the catalytic domain of GC-A. In contrast, transfection of CypA R55A, a CypA mutant expressing low PPIase activity, did not significantly attenuate the activity of GC-c and the activation of GC-A. Inhibition of PPIase activity of CypA with cyclosporin A also blocks the inhibitory effect of CypA on GC-c activity. These results demonstrate that PPIase activity is required for CypA to inhibit GC-c activity and GC-A activation by ANF. Furthermore, mutation of Pro 822, 902, or 958 in GC-c abolished its activity. Therefore, it is likely that CypA binds to GC-A and catalyzes the cis-trans isomerization of Pro 822, 902, or 958, which keeps GC-A in the inactive state, and that binding of ANF to GC-A alters the conformation of the catalytic domain that releases CypA from GC-A leading to enzyme activation.


Assuntos
Fator Natriurético Atrial/fisiologia , Ciclofilina A/fisiologia , Guanilato Ciclase/antagonistas & inibidores , Receptores do Fator Natriurético Atrial/antagonistas & inibidores , Animais , Células COS , Domínio Catalítico , Chlorocebus aethiops , Ativação Enzimática , Guanilato Ciclase/fisiologia , Células LLC-PK1 , Peptidilprolil Isomerase/fisiologia , Prolina/metabolismo , Receptores do Fator Natriurético Atrial/fisiologia , Suínos , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA