Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 123(10): 4219-31, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24051376

RESUMO

Inactivation of the B1 proton pump subunit (ATP6V1B1) in intercalated cells (ICs) leads to type I distal renal tubular acidosis (dRTA), a disease associated with salt- and potassium-losing nephropathy. Here we show that mice deficient in ATP6V1B1 (Atp6v1b1-/- mice) displayed renal loss of NaCl, K+, and water, causing hypovolemia, hypokalemia, and polyuria. We demonstrated that NaCl loss originated from the cortical collecting duct, where activity of both the epithelial sodium channel (ENaC) and the pendrin/Na(+)-driven chloride/bicarbonate exchanger (pendrin/NDCBE) transport system was impaired. ENaC was appropriately increased in the medullary collecting duct, suggesting a localized inhibition in the cortex. We detected high urinary prostaglandin E2 (PGE2) and ATP levels in Atp6v1b1-/- mice. Inhibition of PGE2 synthesis in vivo restored ENaC protein levels specifically in the cortex. It also normalized protein levels of the large conductance calcium-activated potassium channel and the water channel aquaporin 2, and improved polyuria and hypokalemia in mutant mice. Furthermore, pharmacological inactivation of the proton pump in ß-ICs induced release of PGE2 through activation of calcium-coupled purinergic receptors. In the present study, we identified ATP-triggered PGE2 paracrine signaling originating from ß-ICs as a mechanism in the development of the hydroelectrolytic imbalance associated with dRTA. Our data indicate that in addition to principal cells, ICs are also critical in maintaining sodium balance and, hence, normal vascular volume and blood pressure.


Assuntos
Túbulos Renais Coletores/metabolismo , Potássio na Dieta/sangue , Sódio na Dieta/sangue , Equilíbrio Hidroeletrolítico , Trifosfato de Adenosina/metabolismo , Animais , Aquaporina 2/metabolismo , Dinoprostona/metabolismo , Canais Epiteliais de Sódio/metabolismo , Técnicas In Vitro , Medula Renal/citologia , Medula Renal/metabolismo , Túbulos Renais Coletores/citologia , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Camundongos , Camundongos Knockout , Comunicação Parácrina , ATPases Vacuolares Próton-Translocadoras/deficiência , ATPases Vacuolares Próton-Translocadoras/genética
2.
Proc Natl Acad Sci U S A ; 110(19): 7928-33, 2013 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-23610411

RESUMO

The Na(+) concentration of the intracellular milieu is very low compared with the extracellular medium. Transport of Na(+) along this gradient is used to fuel secondary transport of many solutes, and thus plays a major role for most cell functions including the control of cell volume and resting membrane potential. Because of a continuous leak, Na(+) has to be permanently removed from the intracellular milieu, a process that is thought to be exclusively mediated by the Na(+)/K(+)-ATPase in animal cells. Here, we show that intercalated cells of the mouse kidney are an exception to this general rule. By an approach combining two-photon imaging of isolated renal tubules, physiological studies, and genetically engineered animals, we demonstrate that inhibition of the H(+) vacuolar-type ATPase (V-ATPase) caused drastic cell swelling and depolarization, and also inhibited the NaCl absorption pathway that we recently discovered in intercalated cells. In contrast, pharmacological blockade of the Na(+)/K(+)-ATPase had no effects. Basolateral NaCl exit from ß-intercalated cells was independent of the Na(+)/K(+)-ATPase but critically relied on the presence of the basolateral ion transporter anion exchanger 4. We conclude that not all animal cells critically rely on the sodium pump as the unique bioenergizer, but can be replaced by the H(+) V-ATPase in renal intercalated cells. This concept is likely to apply to other animal cell types characterized by plasma membrane expression of the H(+) V-ATPase.


Assuntos
Rim/metabolismo , ATPase Trocadora de Sódio-Potássio/fisiologia , Sódio/metabolismo , Absorção , Animais , Membrana Celular/metabolismo , Células Cultivadas , Antiportadores de Cloreto-Bicarbonato/genética , Imuno-Histoquímica , Íons , Potenciais da Membrana , Camundongos , Camundongos Knockout , Perfusão , Bombas de Próton/fisiologia , Cloreto de Sódio/farmacologia , ATPase Trocadora de Sódio-Potássio/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo
3.
Proc Natl Acad Sci U S A ; 107(50): 21872-7, 2010 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-21098262

RESUMO

Acid-base transport in the renal collecting tubule is mediated by two canonical cell types: the ß-intercalated cell secretes HCO(3) by an apical Cl:HCO(3) named pendrin and a basolateral vacuolar (V)-ATPase. Acid secretion is mediated by the α-intercalated cell, which has an apical V-ATPase and a basolateral Cl:HCO(3) exchanger (kAE1). We previously suggested that the ß-cell converts to the α-cell in response to acid feeding, a process that depended on the secretion and deposition of an extracellular matrix protein termed hensin (DMBT1). Here, we show that deletion of hensin from intercalated cells results in the absence of typical α-intercalated cells and the consequent development of complete distal renal tubular acidosis (dRTA). Essentially all of the intercalated cells in the cortex of the mutant mice are canonical ß-type cells, with apical pendrin and basolateral or diffuse/bipolar V-ATPase. In the medulla, however, a previously undescribed cell type has been uncovered, which resembles the cortical ß-intercalated cell in ultrastructure, but does not express pendrin. Polymerization and deposition of hensin (in response to acidosis) requires the activation of ß1 integrin, and deletion of this gene from the intercalated cell caused a phenotype that was identical to the deletion of hensin itself, supporting its critical role in hensin function. Because previous studies suggested that the conversion of ß- to α-intercalated cells is a manifestation of terminal differentiation, the present results demonstrate that this differentiation proceeds from HCO(3) secreting to acid secreting phenotypes, a process that requires deposition of hensin in the ECM.


Assuntos
Acidose Tubular Renal/metabolismo , Túbulos Renais Coletores/citologia , Mucinas/metabolismo , Animais , Proteínas de Transporte de Ânions/genética , Proteínas de Transporte de Ânions/metabolismo , Bicarbonatos/metabolismo , Proteínas de Ligação ao Cálcio , Proteínas de Ligação a DNA , Deleção de Genes , Concentração de Íons de Hidrogênio , Integrina beta1/metabolismo , Túbulos Renais Coletores/metabolismo , Túbulos Renais Coletores/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mucinas/genética , Transportadores de Sulfato , Proteínas Supressoras de Tumor
4.
Proc Natl Acad Sci U S A ; 107(30): 13526-31, 2010 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-20624970

RESUMO

Tissue kallikrein (TK) is a serine protease synthetized in renal tubular cells located upstream from the collecting duct where renal potassium balance is regulated. Because secretion of TK is promoted by K+ intake, we hypothesized that this enzyme might regulate plasma K+ concentration ([K+]). We showed in wild-type mice that renal K+ and TK excretion increase in parallel after a single meal, representing an acute K+ load, whereas aldosterone secretion is not modified. Using aldosterone synthase-deficient mice, we confirmed that the control of TK secretion is aldosterone-independent. Mice with TK gene disruption (TK-/-) were used to assess the impact of the enzyme on plasma [K+]. A single large feeding did not lead to any significant change in plasma [K+] in TK+/+, whereas TK-/- mice became hyperkalemic. We next examined the impact of TK disruption on K+ transport in isolated cortical collecting ducts (CCDs) microperfused in vitro. We found that CCDs isolated from TK-/- mice exhibit net transepithelial K+ absorption because of abnormal activation of the colonic H+,K+-ATPase in the intercalated cells. Finally, in CCDs isolated from TK-/- mice and microperfused in vitro, the addition of TK to the perfusate but not to the peritubular bath caused a 70% inhibition of H+,K+-ATPase activity. In conclusion, we have identified the serine protease TK as a unique kalliuretic factor that protects against hyperkalemia after a dietary K+ load.


Assuntos
Adaptação Fisiológica/fisiologia , Rim/fisiologia , Potássio/metabolismo , Calicreínas Teciduais/metabolismo , Adaptação Fisiológica/efeitos dos fármacos , Aldosterona/metabolismo , Aldosterona/urina , Animais , Transporte Biológico , Citocromo P-450 CYP11B2/deficiência , Citocromo P-450 CYP11B2/genética , ATPase Trocadora de Hidrogênio-Potássio/genética , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Rim/metabolismo , Túbulos Renais Coletores/metabolismo , Túbulos Renais Coletores/fisiologia , Camundongos , Camundongos Knockout , Potássio/sangue , Potássio/urina , Potássio na Dieta/administração & dosagem , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sódio/metabolismo , Calicreínas Teciduais/genética
6.
J Clin Invest ; 120(5): 1627-35, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20389022

RESUMO

Regulation of sodium balance is a critical factor in the maintenance of euvolemia, and dysregulation of renal sodium excretion results in disorders of altered intravascular volume, such as hypertension. The amiloride-sensitive epithelial sodium channel (ENaC) is thought to be the only mechanism for sodium transport in the cortical collecting duct (CCD) of the kidney. However, it has been found that much of the sodium absorption in the CCD is actually amiloride insensitive and sensitive to thiazide diuretics, which also block the Na-Cl cotransporter (NCC) located in the distal convoluted tubule. In this study, we have demonstrated the presence of electroneutral, amiloride-resistant, thiazide-sensitive, transepithelial NaCl absorption in mouse CCDs, which persists even with genetic disruption of ENaC. Furthermore, hydrochlorothiazide (HCTZ) increased excretion of Na+ and Cl- in mice devoid of the thiazide target NCC, suggesting that an additional mechanism might account for this effect. Studies on isolated CCDs suggested that the parallel action of the Na+-driven Cl-/HCO3- exchanger (NDCBE/SLC4A8) and the Na+-independent Cl-/HCO3- exchanger (pendrin/SLC26A4) accounted for the electroneutral thiazide-sensitive sodium transport. Furthermore, genetic ablation of SLC4A8 abolished thiazide-sensitive NaCl transport in the CCD. These studies establish what we believe to be a novel role for NDCBE in mediating substantial Na+ reabsorption in the CCD and suggest a role for this transporter in the regulation of fluid homeostasis in mice.


Assuntos
Antiportadores de Cloreto-Bicarbonato/metabolismo , Túbulos Renais Coletores/metabolismo , Simportadores de Sódio-Bicarbonato/metabolismo , Sódio/química , Amilorida/farmacologia , Animais , Eletrofisiologia/métodos , Hidroclorotiazida/farmacologia , Rim/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Biológicos , Oócitos/metabolismo , Inibidores de Simportadores de Cloreto de Sódio/farmacologia , Xenopus
7.
Crit Care ; 13(4): R120, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19622169

RESUMO

INTRODUCTION: In septic patients, an unpredictable response to epinephrine may be due to pharmacodynamic factors or to non-linear pharmacokinetics. The purpose of this study was to investigate the pharmacokinetics of epinephrine and its determinants in patients with septic shock. METHODS: Thirty-eight consecutive adult patients with septic shock were prospectively recruited immediately before epinephrine infusion. A baseline blood sample (C0) was taken to assess endogenous epinephrine, norepinephrine, renin, aldosterone, and plasma cortisol levels before epinephrine infusion. At a fixed cumulative epinephrine dose adjusted to body weight and under steady-state infusion, a second blood sample (C1) was taken to assess epinephrine and norepinephrine concentrations. Data were analyzed using the nonlinear mixed effect modeling software program NONMEM. RESULTS: Plasma epinephrine concentrations ranged from 4.4 to 540 nmol/L at steady-state infusion (range 0.1 to 7 mg/hr; 0.026 to 1.67 microg/kg/min). A one-compartment model adequately described the data. Only body weight (BW) and New Simplified Acute Physiologic Score (SAPSII) at intensive care unit admission significantly influenced epinephrine clearance: CL (L/hr) = 127 x (BW/70)0.60 x (SAPS II/50)-0.67. The corresponding half-life was 3.5 minutes. Endogenous norepinephrine plasma concentration significantly decreased during epinephrine infusion (median (range) 8.8 (1 - 56.7) at C0 vs. 4.5 (0.3 - 38.9) nmol/L at C1, P < 0.001). CONCLUSIONS: Epinephrine pharmacokinetics is linear in septic shock patients, without any saturation at high doses. Basal neurohormonal status does not influence epinephrine pharmacokinetics. Exogenous epinephrine may alter the endogenous norepinephrine metabolism in septic patients.


Assuntos
Epinefrina/farmacocinética , Neurotransmissores/sangue , Choque Séptico/tratamento farmacológico , Idoso , Cromatografia Líquida de Alta Pressão , Eletroquímica , Epinefrina/sangue , Epinefrina/uso terapêutico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Choque Séptico/sangue
8.
Am J Physiol Renal Physiol ; 289(6): F1281-90, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16077082

RESUMO

NH(4)(+) transport by the distal nephron and NH(4)(+) detoxification by the liver are critical for achieving regulation of acid-base balance and to avoid hyperammonemic hepatic encephalopathy, respectively. Therefore, it has been proposed that rhesus type B glycoprotein (Rhbg), a member of the Mep/Amt/Rh NH(3) channel superfamily, may be involved in some forms of distal tubular acidosis and congenital hyperammonemia. We have tested this hypothesis by inactivating the RHbg gene in the mouse by insertional mutagenesis. Histochemical studies analyses confirmed that RHbg knockout (KO) mice did not express Rhbg protein. Under basal conditions, the KO mice did not exhibit encephalopathy and survived well. They did not exhibit hallmarks of distal tubular acidosis because neither acid-base status, serum potassium concentration, nor bone mineral density was altered by RHbg disruption. They did not have hyperammonemia or disturbed hepatic NH(3) metabolism. Moreover, the KO mice adapted to a chronic acid-loading challenge by increasing urinary NH(4)(+) excretion as well as their wild-type controls. Finally, transepithelial NH(3) diffusive permeability, or NH(3) and NH(4)(+) entry across the basolateral membrane of cortical collecting duct cells, measured by in vitro microperfusion of collecting duct from KO and wild-type mice, was identical with no apparent effect of the absence of Rhbg protein. We conclude that Rhbg is not a critical determinant of NH(4)(+) excretion by the kidney and of NH(4)(+) detoxification by the liver in vivo.


Assuntos
Amônia/metabolismo , Glicoproteínas/genética , Rim/fisiologia , Proteínas de Membrana Transportadoras/genética , Acidose Tubular Renal/fisiopatologia , Amônia/urina , Animais , Proteína 1 de Troca de Ânion do Eritrócito/metabolismo , Densidade Óssea , Camundongos , Camundongos Knockout , Mutagênese Insercional , ATPases Translocadoras de Prótons/biossíntese
9.
Am J Physiol Renal Physiol ; 287(3): F404-10, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15100097

RESUMO

NaCl reabsorption in the medullary thick ascending limb of Henle (MTALH) contributes to NaCl balance and is also responsible for the creation of medullary interstitial hypertonicity. Despite the presence of angiotensin II subtype 1 (AT(1)) receptors in both the luminal and the basolateral plasma membranes of MTALH cells, no information is available on the effect of angiotensin II on NaCl reabsorption in MTALH and, furthermore, on angiotensin II-dependent medullary interstitial osmolality. MTALHs from male Sprague-Dawley rats were isolated and microperfused in vitro; transepithelial net chloride absorption (J(Cl)) as well as transepithelial voltage (V(te)) were measured. Luminal or peritubular 10(-11) and 10(-10) M angiotensin II had no effect on J(Cl) or V(te). However, 10(-8) M luminal or peritubular angiotensin II reversibly decreased both J(Cl) and V(te). The effect of both luminal and peritubular angiotensin II was prevented by the presence of losartan (10(-6) M). By contrast, PD-23319, an AT(2)-receptor antagonist, did not alter the inhibitory effect of 10(-8) M angiotensin II. Finally, no additive effect of luminal and peritubular angiotensin II was observed. We conclude that both luminal and peritubular angiotensin II inhibit NaCl absorption in the MTALH via AT(1) receptors. Because of intrarenal angiotensin II synthesis, angiotensin II concentration in medullary tubular and interstitial fluids may be similar in vivo to the concentration that displays an inhibitory effect on NaCl reabsorption under the present experimental conditions.


Assuntos
Angiotensina II/farmacologia , Alça do Néfron/metabolismo , Cloreto de Sódio/farmacocinética , Vasoconstritores/farmacologia , Absorção/efeitos dos fármacos , Animais , Técnicas In Vitro , Medula Renal/metabolismo , Alça do Néfron/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/metabolismo , Organismos Livres de Patógenos Específicos , Equilíbrio Hidroeletrolítico/fisiologia
10.
Kidney Int ; 62(6): 2144-51, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12427139

RESUMO

BACKGROUND: To balance medullary interstitium hypertonicity generated by transepithelial NaCl absorption, medullary thick ascending limb (MTAL) cells accumulate myo-inositol (MI). Expression of Na+-MI cotransporter (SMIT) mRNA in TAL is correlated with the NaCl absorption rate. Our present study aimed to determine the plasma membrane location and functional properties of the Na+-MI cotransporter in MTAL cells. METHODS: Preparation of basolateral (BLMV) and luminal (LMV) membrane vesicles were simultaneously isolated from purified rat MTAL suspension, and uptake of [3H]myo-inositol ([3H]MI) was used to assess Na+-MI cotransport activity. RESULTS: In the presence of an inside-negative membrane potential, imposing an inwardly-directed Na+-gradient versus tetramethylammonium (TMA) stimulated the initial [3H]MI uptake in BLMV and LMV. Phlorizin inhibited Na+ gradient-dependent initial [3H]MI uptake in both preparations, with IC50 values of 565 and 29 micromol/L in BLMV and LMV, respectively. 2-0,C-methylene myo-inositol (MMI), a competitive inhibitor of MI transport, only inhibited the BLMV Na+-MI cotransporter. Phlorizin-sensitive Na+ gradient-dependent initial [3H]MI uptake showed Michaelis-Menten kinetics in both preparations, with similar Vmax but different Km values of 51 and 107 micromol/L in BLMV and LMV, respectively. Finally, BLMV but not LMV Na+-MI cotransporter exhibited a marked pH dependence with sigmoidal patterns of activation, as intravesicular pH (pHi) was decreased from 8.0 to 6.0 at extravesicular pH (pHe) 8.0, and as pHe was increased from 6.0 to 8.0 at pHi 6.0. Maximal activation was observed at pHi 6.5 and pHe 7.5. CONCLUSIONS: In rat MTAL cells, Na+-MI cotransporter activity is present in both BLM and LM, and has markedly different functional properties, indicating the presence of distinct transporters. Basolateral Na+-MI cotransporter activity is maximal at physiological pH values of MTAL cells and interstitium, and a powerful modulation of the transporter activity may be exerted by pHe and pHi.


Assuntos
Equilíbrio Ácido-Base/fisiologia , Proteínas de Choque Térmico/metabolismo , Concentração de Íons de Hidrogênio , Alça do Néfron/metabolismo , Proteínas de Membrana , Simportadores/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Inositol/farmacocinética , Medula Renal/metabolismo , Masculino , Florizina/farmacologia , Ratos , Ratos Sprague-Dawley , Sódio/farmacocinética , Trítio
11.
Am J Physiol Renal Physiol ; 282(5): F835-43, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11934693

RESUMO

In the proximal tubule, the apical Na(+)/H(+) exchanger identified as NHE3 mediates most NaCl and NaHCO(3) absorption. The purpose of this study was to analyze the long-term regulation of NHE3 during alkalosis induced by dietary NaHCO(3) loading and changes in NaCl intake. Sprague-Dawley rats exposed to a low-NaCl, high-NaCl, or NaHCO(3) diet for 6 days were studied. Renal cortical apical membrane vesicles (AMV) were prepared from treated and normal rats. Na(+)/H(+) exchange was assayed as the initial rate of (22)Na(+) uptake in the presence of an outward H(+) gradient. (22)Na(+) uptake measured in the presence of high-dose 5-(N-ethyl-N-isopropyl) amiloride was not different among models. Changes in NaCl intake did not affect NHE3 activity, whereas NaHCO(3) loading inhibited (22)Na(+) uptake by 30%. AMV NHE3 protein abundance assessed by Western blot analysis was unaffected during changes in NaCl intake. During NaHCO(3) loading, NHE3 protein abundance was decreased by 65%. We conclude that proximal NHE3 adapts to chronic metabolic acid-base disorders but not to changes in dietary NaCl intake.


Assuntos
Desequilíbrio Ácido-Base/metabolismo , Cloreto de Sódio na Dieta/administração & dosagem , Trocadores de Sódio-Hidrogênio/metabolismo , Adaptação Fisiológica , Alcalose/induzido quimicamente , Alcalose/metabolismo , Animais , Transporte Biológico , Western Blotting , Membrana Celular/metabolismo , Concentração de Íons de Hidrogênio , Córtex Renal/metabolismo , Cinética , Masculino , Natriurese , Ratos , Ratos Sprague-Dawley , Sódio/administração & dosagem , Bicarbonato de Sódio/administração & dosagem , Radioisótopos de Sódio/metabolismo , Trocador 3 de Sódio-Hidrogênio , alfa-Glucosidases/análise , gama-Glutamiltransferase/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...