Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Acta Physiol (Oxf) ; 214(3): 319-28, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25980432

RESUMO

UNLABELLED: Medullary thick ascending limbs (mTAL) regulate Na balance and therefore blood pressure. We previously showed that cell swelling and luminal flow activates the mechanosensitive channel TRPV4 in mTAL. AIM: We hypothesized that TRPV4 mediates flow-induced increases in intracellular Ca (Cai) in rat mTALs. METHODS: We performed ratiometric measurements of Cai in perfused mTALs. RESULTS: Increasing luminal flow from 0 to 20 nL min(-1) caused Cai to peak 231 ± 29 nmol L(-1) above basal concentrations (n = 18). The general TRPV inhibitor ruthenium red at 15 and 50 µmol L(-1) reduced peak Cai by 41 ± 9 (P < 0.01; n = 5) and 77 ± 10% (P < 0.02; n = 6). The selective TRPV4 inhibitor RN1734 at 10 and 50 µmol L(-1) reduced peak Cai by 46 ± 11 (P < 0.01; n = 7) and 76 ± 5% (P < 0.02; n = 5) respectively. To specifically target TRPV4, mTALs were transduced with adenoviruses expressing TRPV4 small hairpin (sh) RNA. In non-transduced control mTALs, luminal flow generated a peak increase in Cai of 111 ± 21 nmol L(-1) (n = 8). In TRPV4shRNA-transduced mTALs, the Cai peak was reduced to 56 ± 8 nmol L(-1) (P < 0.03, n = 9). Removing extracellular Ca completely abolished flow-induced increases in Cai. Increasing luminal flow in the presence of hexokinase 20 (U mL(-1) ) to scavenge extracellular ATP did not modify significantly the increases in Cai induced by luminal flow. Finally, we studied the effect of the TRPV4 selective agonist GSK1016790A on Cai. In the absence of luminal flow, GSK1016790A (10 nmol L(-1) ) increased Cai from 60 ± 11 nmol L(-1) to 262 ± 71 nmol L(-1) (P < 0.05; n = 7). CONCLUSION: We conclude that flow-induced increases in Cai are mediated primarily by TRPV4 in the rat mTAL.


Assuntos
Cálcio/administração & dosagem , Cálcio/farmacocinética , Ativação do Canal Iônico/fisiologia , Alça do Néfron/metabolismo , Canais de Cátion TRPV/metabolismo , Equilíbrio Hidroeletrolítico/fisiologia , Animais , Relação Dose-Resposta a Droga , Ativação do Canal Iônico/efeitos dos fármacos , Alça do Néfron/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley
2.
Am J Physiol Renal Physiol ; 299(6): F1374-8, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20826574

RESUMO

In vitro experiments showed that the connecting tubule (CNT) sends a signal that dilates the afferent arteriole (Af-Art) when Na(+) reabsorption in the CNT lumen increases. We call this process CNT glomerular feedback (CTGF) to differentiate it from tubuloglomerular feedback (TGF), which is a cross talk between the macula densa (MD) and the Af-Art. In TGF, the MD signals the Af-Art to constrict when NaCl transport by the MD is enhanced by increased luminal NaCl. CTGF is mediated by CNT Na(+) transport via epithelial Na(+) channels (ENaC). However, we do not know whether CTGF occurs in vivo or whether it opposes the increase in Af-Art resistance caused by TGF. We hypothesized that CTGF occurs in vivo and opposes TGF. To test our hypothesis, we conducted in vivo micropuncture of individual rat nephrons, measuring stop-flow pressure (P(SF)) as an index of glomerular filtration pressure. To test whether activation of CTGF opposes TGF, we used benzamil to block CNT Na(+) transport and thus CTGF. CTGF inhibition with the ENaC blocker benzamil (1 µM) potentiated the decrease in P(SF) at 40 and 80 nl/min. Next, we tested whether we could augment CTGF by inhibiting NaCl reabsorption in the distal convoluted tubule with hydrochlorothiazide (HCTZ, 1 mM) to enhance NaCl delivery to the CNT. In the presence of HCTZ, benzamil potentiated the decrease in P(SF) at 20, 40, and 80 nl/min. We concluded that in vivo CTGF occurs and opposes the vasoconstrictor effect of TGF.


Assuntos
Retroalimentação/efeitos dos fármacos , Glomérulos Renais/fisiologia , Túbulos Renais/fisiologia , Amilorida/análogos & derivados , Amilorida/farmacologia , Animais , Arteríolas/efeitos dos fármacos , Glomérulos Renais/efeitos dos fármacos , Túbulos Renais/efeitos dos fármacos , Masculino , Néfrons/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
3.
Kidney Int ; 71(9): 861-6, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17342182

RESUMO

Adenosine triphosphate (ATP) is liberated from macula densa cells in response to increased tubular NaCl delivery. However, it is not known whether ATP from the macula densa is broken down to adenosine, or whether this adenosine mediates efferent arteriole (Ef-Art) tubuloglomerular feedback (TGF). We hypothesized that increased macula densa Ca(2+), release of ATP and degradation of ATP to adenosine are necessary for Ef-Art TGF. Rabbit Ef-Arts and adherent tubular segments (with the macula densa) were simultaneously microperfused in vitro while changing the NaCl concentration at the macula densa. The Ef-Art was perfused orthograde through the end of the afferent arteriole (Af-Art). In Ef-Arts preconstricted with norepinephrine (NE), increasing NaCl concentration from 10 to 80 mM at the macula densa dilated Ef-Arts from 7.5+/-0.7 to 11.1+/-0.3 microm. Buffering increases in macula densa Ca(2+) with the cell-permeant Ca(2+) chelator BAPTA-AM diminished Ef-Art TGF from 3.1+/-0.3 to 0.1+/-0.2 microm. Blocking adenosine formation by adding alpha-beta-methyleneadenosine 5'-diphosphate (MADP) blocked Ef-Art TGF from 2.9+/-0.5 to 0.1+/-0.2 microm. Increasing luminal NaCl at the macula densa from 10 to 45 mM caused a moderate Ef-Art TGF response, 1.3+/-0.1 microm. It was potentiated to 4.0+/-0.3 microm by adding hexokinase, which enhances conversion of ATP into adenosine. Our data show that in vitro changes in macula densa Ca(2+) and ATP release are necessary for Ef-Art TGF. ATP is broken down to form adenosine, which mediates signal transmission of Ef-Art TGF.


Assuntos
Arteríolas/fisiologia , Sistema Justaglomerular/fisiologia , Glomérulos Renais/fisiologia , Túbulos Renais/fisiologia , Adenosina/fisiologia , Animais , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Retroalimentação , Sistema Justaglomerular/efeitos dos fármacos , Coelhos , Fatores de Crescimento Transformadores/fisiologia
4.
Kidney Int ; 71(11): 1116-21, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17361114

RESUMO

The renal afferent arterioles (Af-Arts) account for most of the renal vascular resistance, which is controlled similar to other arterioles and by tubuloglomerular feedback (TGF). The latter signal is generated by sensing sodium chloride concentrations in the macula densa; this in turn results in a signal which acts through the extraglomerular mesangium leading to constriction of the Af-Art. In the outer renal cortex, the connecting tubule (CNT) returns to the glomerular hilus and contacts the Af-Art suggesting that crosstalk may exist here as well. To investigate this, we simultaneously perfused a microdissected Af-Art and adherent CNT. Increasing the sodium chloride concentration perfusing the CNT significantly dilated preconstricted Af-Arts. We called this crosstalk 'connecting tubule glomerular feedback' (CTGF) to differentiate it from TGF. We tested whether entry of Na(+) and/or CI(-) into the CNT is required to induce CTGF by replacing Na(+) with choline(+). Increasing choline chloride concentration did not dilate the Af-Art. To test whether epithelial Na channels (ENaCs) mediate CTGF, we blocked ENaC with amiloride and found that the dilatation induced by CTGF was completely blocked. Inhibiting sodium chloride cotransporters with hydrochlorothiazide failed to prevent Af-Art dilatation. Finally, we tested whether nitric oxide released by the CNT mediates CTGF by the addition of a non-selective nitric oxide synthase inhibitor to the CNT. This potentiated CTGF rather than blocking it. We suggest that crosstalk exists between CNTs and attached Af-Arts, which is initiated by sodium reabsorption through amiloride-sensitive channels and this can contribute to the regulation of renal blood flow and glomerular filtration.


Assuntos
Arteríolas/fisiologia , Túbulos Renais/fisiologia , Rim/irrigação sanguínea , Amilorida/farmacologia , Animais , Arteríolas/cirurgia , Relação Dose-Resposta a Droga , Canais Epiteliais de Sódio/efeitos dos fármacos , Retroalimentação/efeitos dos fármacos , Glomérulos Renais/fisiologia , Túbulos Renais/cirurgia , Microcirculação/fisiologia , Microdissecção , Modelos Biológicos , Óxido Nítrico/fisiologia , Perfusão , Coelhos , Bloqueadores dos Canais de Sódio/farmacologia , Cloreto de Sódio/administração & dosagem , Cloreto de Sódio/farmacologia , Vasodilatação/efeitos dos fármacos
5.
Acta Physiol Scand ; 179(3): 225-32, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14616238

RESUMO

UNLABELLED: The phrase reactive oxygen species covers a number of molecules and atoms, including the quintessential member of the group, O2-; singlet oxygen; H2O2; organic peroxides; and OONO-. While nitric oxide (NO) is also technically a member of the reactive oxygen species family, it is generally considered with a different class of compounds and will not be considered here. To our knowledge, there are currently no published data reporting the effects of reactive oxygen species on net transepithelial flux in the proximal nephron. However, there is evidence that OONO- regulates Na+/K+ adenosine triphosphatase (ATPase) activity as well as paracellular permeability. While it is easy to speculate that such an effect on the pump would decrease net transepithelial solute and water reabsorption, one cannot do so without knowing how other transporters are affected. O2- stimulates NaCl absorption by the thick ascending limb by activating protein kinase C and blunting the effects of NO. The effects of O2- on thick ascending limb NaCl absorption may be important for the initiation of salt-sensitive hypertension. To our knowledge, there are no published data concerning the role of reactive oxygen species in the regulation of solute absorption in either the distal convoluted tubule or the collecting duct. However, OONO- inhibits basolateral K+ channels in the cortical collecting duct, although the net effect of such inhibition is unknown. CONCLUSION: While the regulation of tubular transport by reactive oxygen species is important to overall salt and water balance, we know very little about where and how these regulators act along the nephron.


Assuntos
Túbulos Renais/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transporte Biológico/fisiologia , Humanos , Túbulos Renais Coletores/metabolismo , Néfrons/metabolismo , Oxigênio/metabolismo , Proteína Quinase C/metabolismo , Canais de Sódio/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , ATPase Trocadora de Sódio-Potássio , Superóxidos/metabolismo
6.
Acta Physiol Scand ; 179(2): 107-14, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14510773

RESUMO

In mammalian cells, formation of nitric oxide (NO) is catalysed by a family of enzymes termed NO synthases (NOS). There are three isoforms of this enzyme, NOS I, II and III. NOS III was originally cloned and identified in endothelial cells; thus this isoform is commonly called endothelial NOS (eNOS). The physiological role of NO produced by eNOS has been documented in most organs, including the brain, lung, cardiovascular system, kidney, liver, gastrointestinal tract and reproductive organs. The bioavailability of NO in these tissues is determined by the balance between its rate of production and degradation. The rate of NO production by eNOS is ultimately dependent on the activity of the enzyme. In the past years, co- and post-translational modifications such as myristoylation, palmitoylation, phosphorylation, protein-protein interactions and subcellular localization have been shown to play an important role in determining eNOS activity. In order to maintain specificity, the production of most signalling molecules occurs in an organized spatial and temporal pattern. Spatial localization of eNOS has been shown to be regulated by different mechanisms that control its targeting from the Golgi apparatus to the plasma membrane, correct compartmentalization within the membrane, and internalization from the plasma membrane to the cytoplasm after activation. Thus, regulated localization and trafficking of eNOS may be essential in regulating enzyme activity and maintaining the spatial and temporal organization of NO signalling in different cell types.


Assuntos
Óxido Nítrico Sintase/fisiologia , Membrana Celular/enzimologia , Citoplasma/enzimologia , Células Endoteliais/enzimologia , Endotélio Vascular/enzimologia , Epitélio/enzimologia , Humanos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III , Palmitatos/metabolismo , Proteínas/metabolismo , Frações Subcelulares/enzimologia , Translocação Genética
7.
Kidney Int ; 60(5): 1676-83, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11703584

RESUMO

BACKGROUND: There is evidence that macula densa nitric oxide (NO) inhibits tubuloglomerular feedback (TGF). However, TGF response is not altered in mice deficient in neuronal nitric oxide synthase (nNOS) (-/-). Furthermore, nNOS expression in the macula densa is inversely related to salt intake, yet micropuncture studies have shown that NOS inhibition potentiates TGF in rats on high sodium intake but not in rats on a low-salt diet. These inconsistencies may be due to confounding systemic factors, such as changes in circulating renin. To further clarify the role of macula densa nNOS in TGF response, independent of systemic factors, we tested the hypothesis that (1) TGF response is inversely related to sodium intake, and (2) during low sodium intake, NO produced by macula densa nNOS tonically controls the basal diameter of the afferent arteriole (Af-Art). METHODS: Af-Arts and attached macula densas were simultaneously microperfused in vitro. TGF response was determined by measuring Af-Art diameter before and after increasing NaCl in the macula densa perfusate. TGF response was studied in wild-type (+/+) and nNOS knockout mice (-/-), as well as in juxtaglomerular apparatuses (JGAs) from rabbits fed a low-, normal-, or high-NaCl diet. RESULTS: TGF responses were similar in nNOS +/+ and -/- mice. However, in nNOS +/+ mice, 7-nitroindazole (7-NI) perfused into the macula densa significantly potentiated the TGF response (P = 0.001), while in nNOS -/- mice, this potentiation was absent. In rabbits on three different sodium diets, TGF responses were similar and were potentiated equally by 7-NI. However, in JGAs from rabbits on a low-NaCl diet, adding 7-NI to the macula densa while perfusing it with low-NaCl fluid caused Af-Art vasoconstriction, decreasing the diameter by 14% (from 21.7 +/- 1.3 to 18.6 +/- 1.5 microm; P < 0.001). This effect was not observed in JGAs from rabbits fed a normal- (19.0 +/- 0.5 vs. 19.3 +/- 0.8 microm after 7-NI) or high-NaCl diet (18.6 +/- 0.7 vs. 18.4 +/- 0.7 microm). CONCLUSIONS: First, in this in vitro preparation, chronic changes in macula densa nNOS do not play a major role in the regulation of TGF. Compensatory mechanisms may develop during chronic alteration of nNOS that keep TGF relatively constant. Second, nNOS regulates TGF response acutely. Third, the results obtained in the +/+ and -/- mice also confirm that the effect of 7-NI is due to inhibition of macula densa nNOS. Finally, during low sodium intake (without induction of TGF), the regulation of basal Af-Art resistance by macula densa nNOS suggests that NO in the macula densa helps maintain renal blood flow during the high renin secretion caused by low sodium intake.


Assuntos
Glomérulos Renais/enzimologia , Óxido Nítrico Sintase/fisiologia , Animais , Arteríolas/anatomia & histologia , Arteríolas/efeitos dos fármacos , Retroalimentação , Indazóis/farmacologia , Glomérulos Renais/fisiologia , Túbulos Renais/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo I , Coelhos , Cloreto de Sódio na Dieta/administração & dosagem
8.
Kidney Int ; 60(5): 1851-7, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11703603

RESUMO

BACKGROUND: Recent studies have revealed angiotensin II subtype 1 (AT1) receptors on macula densa cells, raising the possibility that angiotensin II (Ang II) could enhance tubuloglomerular feedback (TGF) by affecting macula densa cell function. We hypothesized that Ang II enhances TGF via activation of AT1 receptors on the luminal membrane of the macula densa. METHODS: Rabbit afferent arterioles and the attached macula densa were simultaneously microperfused in vitro, keeping pressure in the afferent arteriole at 60 mm Hg. RESULTS: The afferent arteriole diameter was measured while the macula densa was perfused with low NaCl (Na+, 5 mmol/L; Cl-, 3 mmol/L) and then with high NaCl (Na+, 79 mmol/L; Cl-, 77 mmol/L) to induce a TGF response. When TGF was induced in the absence of Ang II, the afferent arteriole diameter decreased by 2.4 +/- 0.5 microm (from 17.3 +/- 1.0 to 14.9 +/- 1.2 microm). With Ang II (0.1 nmol/L) present in the lumen of the macula densa, the diameter decreased by 3.8 +/- 0.7 microm (from 17.3 +/- 1.0 to 13.5 +/- 1.2 microm, P < 0.05 vs. TGF with no Ang II, N = 8). To test whether Ang II enhances TGF via activation of AT1 receptors on the luminal membrane of the macula densa, Ang II plus losartan (1 micromol/L) was added to the lumen. Losartan itself did not alter TGF. When TGF was induced in the absence of Ang II and losartan, the afferent arteriole diameter decreased by 2.3 +/- 0.3 microm (from 15.9 +/- 1.0 to 13.6 +/- 1.2 microm). When Ang II and losartan were both present in the macula densa perfusate, the diameter decreased by 2.4 +/- 0.4 microm (from 15.8 +/- 0.9 to 13.4 +/- 0.7 microm, P> 0.8 vs. TGF with no Ang II and losartan, N = 7). We then examined whether AT2 receptors on the macula densa influence the effect of luminal Ang II on TGF. When TGF was induced in the absence of Ang II plus PD 0123319-0121B (1 micromol/L), the afferent arteriole diameter decreased by 2.4 +/- 0.2 microm (from 17.0 +/- 0.9 to 14.6 +/- 0.8 microm). When Ang II and PD 0123319-0121B were both present in the macula densa lumen, the diameter decreased by 3.9 +/- 0.2 microm (from 16.8 +/- 0.9 to 12.9 +/- 0.9 microm, P < 0.001 vs. TGF with no Ang II and PD 0123319-0121B, N = 8). PD 0123319-0121B itself had no effect on TGF. To assure that this effect of Ang II was not due to leakage into the bath, losartan was added to the bath. When TGF was induced in the absence of Ang II with losartan in the bath, the afferent arteriole diameter decreased by 2.8 +/- 0.5 microm (from 19.3 +/- 1.2 to 16.5 +/- 0.8 microm). After Ang II was added to the macula densa perfusate and losartan to the bath, the diameter decreased by 4.0 +/- 0.7 microm (from 18.9 +/- 1.1 to 14.9 +/- 0.5 microm, P < 0.01 vs. TGF with no Ang II in the lumen and losartan in the bath, N = 8). CONCLUSIONS: These results demonstrate that Ang II enhances TGF via activation of AT1 receptors on the luminal membrane of the macula densa.


Assuntos
Angiotensina II/farmacologia , Glomérulos Renais/efeitos dos fármacos , Túbulos Renais/efeitos dos fármacos , Receptores de Angiotensina/efeitos dos fármacos , Animais , Retroalimentação , Glomérulos Renais/fisiologia , Túbulos Renais/fisiologia , Masculino , Coelhos , Receptor Tipo 1 de Angiotensina , Receptores de Angiotensina/fisiologia , Trocadores de Sódio-Hidrogênio/fisiologia
9.
Am J Physiol Renal Physiol ; 281(6): F1102-8, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11704561

RESUMO

The macula densa expresses a luminal Na(+)-K(+)-2Cl(-) cotransporter and a basolateral Cl(-) conductance. Although it is known that cotransport of Na(+), K(+), and Cl(-) is the first step in tubuloglomerular feedback (TGF), subsequent steps are unclear. We hypothesized that Na(+)-K(+)-2Cl(-) entry via the luminal Na(+)-K(+)-2Cl(-) cotransporter elevates intracellular Cl(-), increases electrogenic Cl(-) efflux across the basolateral membrane, and depolarizes the macula densa, initiating TGF. We perfused afferent arterioles with macula densa attached. The macula densa was perfused with solutions containing either 5 mM Na(+) and 3 mM Cl(-) (low NaCl) or 80 mM Na(+) and 77 mM Cl(-) (high NaCl). When the macula densa perfusate was changed from low to high NaCl, afferent arteriole diameter decreased from 15.8 +/- 0.8 to 13.1 +/- 0.7 mm (P < 0.05). Adding 10 microM furosemide to the macula densa lumen blocked TGF. When nystatin, a group I cation ionophore, was added to the macula densa lumen together with furosemide in the presence of low NaCl, it induced TGF (from 18.0 +/- 1.5 to 15.6 +/- 1.6 mm; P = 0.003). When valinomycin, a K(+)-selective ionophore, was added to the macula densa lumen together with furosemide in the presence of low NaCl containing 5 mM K(+), it did not induce TGF. Subsequent addition of 50 mM KCl to the macula densa perfusate induced TGF (from 21.7 +/- 0.8 to 17.5 +/- 1.3 mm; P = 0.0047; n = 6). Adding 50 mM KCl without valinomycin did not induce TGF. When 5-nitro-2-(3-phenylpropylamino)benzoic acid (NPPB; 1 microM), a Cl(-) channel blocker, was added to the bath, it blocked TGF induced by high NaCl, but did not block TGF induced by valinomycin plus 50 mM KCl. NPPB did not alter afferent arteriole constriction induced by norepinephrine. We concluded that increased NaCl in the lumen of the macula densa leads to influx of Cl(-) via the Na(+)-K(+)-2Cl(-) cotransporter. The accelerated transport increases intracellular Cl(-). The subsequent exit of Cl(-) across the basolateral membrane via Cl( -) channels in turn leads to depolarization of the macula densa and thereby induces TGF.


Assuntos
Ionóforos/farmacologia , Glomérulos Renais/fisiologia , Túbulos Renais Distais/fisiologia , Nistatina/farmacologia , Circulação Renal , Valinomicina/farmacologia , Animais , Arteríolas/efeitos dos fármacos , Arteríolas/fisiologia , Canais de Cloreto/antagonistas & inibidores , Cloretos/metabolismo , Técnicas de Cultura , Retroalimentação Fisiológica , Furosemida/farmacologia , Transporte de Íons/efeitos dos fármacos , Glomérulos Renais/irrigação sanguínea , Túbulos Renais Distais/irrigação sanguínea , Nitrobenzoatos/farmacologia , Potássio/metabolismo , Coelhos , Sódio/metabolismo , Inibidores de Simportadores de Cloreto de Sódio e Potássio
10.
Am J Physiol Renal Physiol ; 281(5): F819-25, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11592939

RESUMO

We have reported that nitric oxide (NO) inhibits thick ascending limb (THAL) chloride absorption (J(Cl(-))). NaCl transport in the THAL depends on apical Na(+)-K(+)-2Cl(-) cotransporters, apical K(+) channels, and basolateral Na(+)-K(+)-ATPase. However, the transporter inhibited by NO is unknown. We hypothesized that NO decreases THAL J(Cl(-)) by inhibiting the Na(+)-K(+)-2Cl(-) cotransporter. THALs from Sprague-Dawley rats were isolated and perfused. Intracellular sodium ([Na(+)](i)) and chloride concentrations ([Cl(-)](i)) were measured with sodium green and SPQ, respectively. The NO donor spermine NONOate (SPM) decreased [Na(+)](i) from 13.5 +/- 1.2 to 9.6 +/- 1.6 mM (P < 0.05) and also decreased [Cl(-)](i) (P < 0.01). We next tested whether NO decreases Na(+)-K(+)-2Cl(-) cotransporter activity by measuring the initial rate of Na(+) transport. In the presence of SPM in the bath, initial rates of Na(+) entry were 49.6 +/- 6.0% slower compared with control rates (P < 0.05). To determine whether NO inhibits apical K(+) channel activity, we measured the change in membrane potential caused by an increase in luminal K(+) from 1 to 25 mM using a potential-sensitive fluorescent dye. In the presence of SPM, increasing luminal K(+) concentration depolarized THALs to the same extent as it did in control tubules. We then tested whether a change in apical K(+) permeability could affect NO-induced inhibition of THAL J(Cl(-)). In the presence of luminal valinomycin, which increases K(+) permeability, addition of SPM decreased THAL J(Cl(-)) by 41.2 +/- 10.4%, not significantly different from the inhibition observed in control tubules. We finally tested whether NO alters the affinity or maximal rate of Na(+)-K(+)-ATPase by measuring oxygen consumption rate (QO(2)) in THAL suspensions in the presence of nystatin in varying concentrations of Na(+). In the presence of 10.5 mM Na(+), nystatin increased QO(2) to 119.1 +/- 19.2 and 125.6 +/- 23.4 nmol O(2). mg protein(-1). min(-1) in SPM- and furosemide-treated tubules, respectively. In the presence of 145 mM extracellular Na(+), nystatin increased QO(2) by 104 +/- 7 and 94 +/- 20% in NO- and furosemide-treated tubules, respectively. We concluded that NO decreases THAL J(Cl(-)) by inhibiting Na(+)-K(+)-2Cl(-) cotransport rather than inhibiting apical K(+) channels or the sodium pump.


Assuntos
Cloretos/metabolismo , Alça do Néfron/efeitos dos fármacos , Alça do Néfron/metabolismo , Óxido Nítrico/farmacologia , Inibidores de Simportadores de Cloreto de Sódio e Potássio , Espermina/análogos & derivados , Animais , Condutividade Elétrica , Furosemida/farmacologia , Masculino , Doadores de Óxido Nítrico/farmacologia , Óxidos de Nitrogênio , Nistatina/farmacologia , Potássio/metabolismo , Bloqueadores dos Canais de Potássio , Ratos , Ratos Sprague-Dawley , Sódio/metabolismo , Cloreto de Sódio/administração & dosagem , Simportadores de Cloreto de Sódio-Potássio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Espermina/farmacologia
11.
Hypertension ; 38(3 Pt 2): 621-4, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11566943

RESUMO

Blood pressure is influenced by several vasoactive factors that also regulate nephron transport. An imbalance in regulation of salt reabsorption by the nephron contributes to hypertension. In the spontaneously hypertensive rat (SHR), the responses to dopamine and angiotensin II in the proximal nephron are diminished and enhanced, respectively. This partially explains why the proximal tubule of SHR absorbs more salt and water than that of normotensive controls. In the Dahl salt-sensitive rat, defects in NO signaling and alterations in the arachidonic acid/cytochrome P450 pathways are associated with increased salt reabsorption by the thick ascending limb. In other animal models, such as the deoxycorticosterone acetate (DOCA)-salt rat, hypertension develops as the result of an induced hormonal imbalance. By mimicking the effects of aldosterone, DOCA stimulates sodium reabsorption in the collecting ducts, causing salt and fluid retention. Thus, this model is similar to inherited forms of human hypertension caused by abnormal regulation of transport by mineralocorticoids, such as apparent mineralocorticoid excess and glucocorticoid-remediable aldosteronism. Overall, these findings demonstrate the significance of vasoactive compounds in regulating nephron transport and controlling blood pressure. However, important questions regarding humoral control of nephron transport and its implications in hypertension remain unanswered, and intensive research in these areas is required.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Hipertensão/fisiopatologia , Rim/metabolismo , Angiotensina II/farmacologia , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Arginina/farmacologia , Transporte Biológico/efeitos dos fármacos , Modelos Animais de Doenças , Taxa de Filtração Glomerular/efeitos dos fármacos , Humanos , Ratos , Ratos Endogâmicos Dahl , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY
12.
Am J Physiol Renal Physiol ; 281(4): F679-86, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11553515

RESUMO

Stimulation of alpha(2)-adrenergic receptors inhibits transport in various nephron segments, and the thick ascending limb of the loop of Henle (THAL) expresses alpha(2)-receptors. We hypothesized that selective alpha(2)-receptor activation decreases NaCl absorption by cortical THALs through activation of NOS and increased production of NO. We found that the alpha(2)-receptor agonist clonidine (10 nM) decreased chloride flux (J(Cl)) from 119.5 +/- 15.9 to 67.4 +/- 13.8 pmol. mm(-1). min(-1) (43% reduction; P < 0.02), whereas removal of clonidine from the bath increased J(Cl) by 20%. When NOS activity was inhibited by pretreatment with 5 mM N(G)-nitro-L-arginine methyl ester, the inhibitory effects of clonidine on THAL J(Cl) were prevented (81.7 +/- 10.8 vs. 71.6 +/- 6.9 pmol. mm(-1). min(-1)). Similarly, when the NOS substrate L-arginine was deleted from the bath, addition of clonidine did not decrease THAL J(Cl) from control (106.9 +/- 11.6 vs. 132.2 +/- 21.3 pmol. mm(-1). min(-1)). When we blocked the alpha(2)-receptors with rauwolscine (1 microM), we found that the inhibitory effect of 10 nM clonidine on THAL J(Cl) was abolished, verifying that alpha(2), rather than I(1), receptors mediate the effects of clonidine in the THAL. We investigated the mechanism of NOS activation and found that intracellular calcium concentration did not increase in response to clonidine, whereas pretreatment with 150 nM wortmannin abolished the clonidine-mediated inhibition of THAL J(Cl), indicating activation of phosphatidylinositol 3-kinase and the Akt pathway. We found that pretreatment of THALs with 10 microM LY-83583, an inhibitor of soluble guanylate cyclase, blocked clonidine-mediated inhibition of THAL J(Cl). In conclusion, alpha(2)-receptor stimulation decreases THAL J(Cl) by increasing NO release and stimulating guanylate cyclase. These data suggest that alpha(2)-receptors act as physiological regulators of THAL NO synthesis, thus inhibiting chloride transport and participating in the natriuretic and diuretic effects of clonidine in vivo.


Assuntos
Cloretos/metabolismo , Alça do Néfron/enzimologia , Néfrons/metabolismo , Óxido Nítrico/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Agonistas alfa-Adrenérgicos/farmacologia , Antagonistas Adrenérgicos alfa/farmacologia , Aminoquinolinas/farmacologia , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Clonidina/farmacologia , Inibidores Enzimáticos/farmacologia , Técnicas In Vitro , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Ratos , Ratos Sprague-Dawley , Ioimbina/farmacologia
13.
Am J Physiol Regul Integr Comp Physiol ; 281(3): R861-7, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11507002

RESUMO

ANG II increases fluid absorption in proximal tubules from young rats more than those from adult rats. ANG II increases fluid absorption in the proximal nephron, in part, via activation of protein kinase C (PKC). However, it is unclear how age-related changes in ANG II-induced stimulation of the PKC cascade differ as an animal matures. We hypothesized that the response of the proximal nephron to ANG II decreases as rats mature due to a reduction in the amount and activation of PKC rather than a decrease in the number or affinity of ANG II receptors. Because PKC translocates from the cytosol to the membrane when activated, we first measured PKC activity in the soluble and particulate fractions of proximal tubule homogenates exposed to vehicle or 10(-10) M ANG II from young (26 +/- 1 days old) and adult rats (54 +/- 1 days old). ANG II increased PKC activity to the same extent in homogenates from young rats (from 0.119 +/- 0.017 to 0.146 +/- 0.015 U/mg protein) (P < 0.01) and adult rats (from 0.123 +/- 0.020 to 0.156 +/- 0.023 U/mg protein) (P < 0.01). Total PKC activity did not differ between groups (0.166 +/- 0.018 vs. 0.181 +/- 0.023). We next investigated whether activation of the alpha-, beta-, and gamma-PKC isoforms differed by Western blot. In homogenates from young rats, ANG II significantly increased activated PKC-alpha from 40.2 +/- 6.5 to 60.2 +/- 9.5 arbitrary units (AU) (P < 0.01) but had no effect in adult rats (46.1 +/- 5.1 vs. 48.5 +/- 8.2 AU). Similarly, ANG II increased activated PKC-gamma in proximal tubules from young rats from 47.9 +/- 13.2 to 65.6 +/- 16.7 AU (P < 0.01) but caused no change in adult rats. Activated PKC-beta, however, increased significantly in homogenates from both age groups. Specifically, activated PKC-beta increased from 8.6 +/- 1.4 to 12.2 +/- 2.1 AU (P < 0.01) in homogenates from nine young rats and from 19.0 +/- 5.5 to 25.1 +/- 7.1 AU (P < 0.01) in homogenates from 12 adult rats. ANG II did not alter the amount of soluble PKC-alpha, -beta, and -gamma significantly. The total amount of PKC-alpha and -gamma did not differ between homogenates from young and adult rats, whereas the total amount of PKC-beta was 59.7 +/- 10.7 and 144.9 +/- 41.8 AU taken from young and adult rats, respectively (P < 0.05). Maximum specific binding and affinity of ANG II receptors were not significantly different between young and adult rats. We concluded that the primary PKC isoform activated by ANG II changes during maturation.


Assuntos
Envelhecimento/metabolismo , Angiotensina II/metabolismo , Túbulos Renais Proximais/enzimologia , Proteína Quinase C/metabolismo , Fatores Etários , Angiotensina II/farmacologia , Animais , Western Blotting , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Isoenzimas/metabolismo , Túbulos Renais Proximais/efeitos dos fármacos , Proteína Quinase C beta , Ratos , Ratos Sprague-Dawley
14.
Hypertension ; 37(2 Pt 2): 467-71, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11230320

RESUMO

In the isolated, perfused rat thick ascending limb (THAL), L-arginine (L-Arg) stimulates endogenous nitric oxide (NO) production, which inhibits NaCl absorption. However, the intracellular cascade responsible for the effects of NO has not been studied. We hypothesized that endogenous NO inhibits THAL NaCl transport by increasing cGMP, which activates protein kinase G (PKG) and cGMP-stimulated phosphodiesterase (PDE II), which, in turn, decreases cAMP levels. THALs from rats were isolated and perfused, and net chloride flux (J(Cl-)) was measured. L-Arg was used to stimulate NO production. Adding L-Arg (0.5 mmol/L) to the bath decreased J(Cl-) from 154.4+/-9.9 to 101.9+/-14.1 pmol. mm(-1). min(-1), a 35.2% decrease (n=6; P<0.05). In the presence of the soluble guanylate cyclase inhibitor LY-83583 (10 micromol/L), adding L-Arg to the bath did not affect THAL J(Cl-) (143.7+/-28.1 versus 136.7+/-22.2 pmol. mm(-1). min(-1); n=6). LY-83583 alone had no effect on J(Cl-). In the presence of the PDE II inhibitor erythro-9-(2-hydroxy-3-nonyl) adenine (EHNA) 50 micromol/L, L-Arg reduced J(Cl-) by only 13% (142.1+/-8.9 versus 122.7+/-11.5 pmol. mm(-1). min(-1); P<0.05; n=6). EHNA alone had no effect on THAL J(Cl-). In the presence of 10(-5) mol/L dibutyryl (db)-cAMP, L-Arg did not significantly reduce J(Cl-) (116.3+/-18.2 versus 102.6+/-15.6 pmol. mm(-1). min(-1); n=6). db-cAMP (10(-5) mol/L) had no effect on THAL J(Cl-). In the presence of the PKG inhibitor KT-5823 (2 micromol/L), L-Arg lowered J(Cl-) from 142.6+/-14.1 to 85.9+/-8.3 pmol. mm(-1). min(-1), a decrease of 35.6% (n=8; P<0.05). We conclude that (1) endogenous NO inhibits THAL J(Cl-) by stimulating soluble guanylate cyclase and increasing cGMP; (2) NO inhibits THAL J(Cl-) by stimulation of PDE II, which, in turn, decreases cAMP levels; and (3) PKG does not mediate NO-induced inhibition of THAL J(Cl-).


Assuntos
Adenina/análogos & derivados , Carbazóis , GMP Cíclico/biossíntese , Exonucleases/metabolismo , Indóis , Túbulos Renais Distais/efeitos dos fármacos , Óxido Nítrico/metabolismo , Cloreto de Sódio/metabolismo , Adenina/farmacologia , Alcaloides/farmacologia , Aminoquinolinas/farmacologia , Animais , Arginina/farmacologia , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Exonucleases/antagonistas & inibidores , Guanilato Ciclase/biossíntese , Túbulos Renais Distais/metabolismo , Masculino , Perfusão , Ratos , Ratos Sprague-Dawley
15.
Kidney Int ; 59(1): 222-9, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11135074

RESUMO

BACKGROUND: Afferent and efferent arteriole resistance exerts critical and opposite actions in the regulation of glomerular capillary pressure (PGC) and glomerular filtration rate (GFR). Tubuloglomerular feedback (TGF) plays an important role in the regulation of afferent arteriole resistance; however, the role of TGF in the regulation of efferent arteriole resistance is less well established. We hypothesized that TGF caused by increased NaCl in the tubular fluid stimulates the macula densa to initiate a cascade of events resulting in efferent arteriole vasodilation, mediated by adenosine via its A2 receptor. METHODS: Rabbit efferent arterioles and adherent tubular segments with macula densa were simultaneously microperfused in vitro while changing NaCl concentration at the macula densa. To study whether autacoids produced by the glomerulus participate in the effect of TGF on efferent arterioles, they were perfused orthograde or retrograde. To eliminate the hemodynamic influence of the afferent arteriole during orthograde perfusion, the perfusion pipette was advanced to the distal end of the afferent arteriole, and the tip of the pressure pipette was placed beyond the afferent arteriole; for retrograde perfusion, the efferent arteriole was perfused from its distal end. RESULTS: In efferent arterioles perfused orthograde and preconstricted with norepinephrine (NE), increasing NaCl concentration at the macula densa increased the diameter by 33%. In preconstricted efferent arterioles perfused retrograde, increasing NaCl at the macula densa increased the diameter by 33%. Efferent arteriole vasodilation was completely blocked by a selective adenosine A2 receptor antagonist (3, 7-dimethyl-1-propargylxanthine) but not by an adenosine A1 receptor antagonist (FK838). CONCLUSIONS: Our data show that in vitro, preconstricted efferent arterioles dilate in response to increased macula densa NaCl, and this process is mediated by activation of adenosine A2 receptors. Thus, TGF changes efferent arteriole resistance in the opposite direction from the afferent arteriole, possibly amplifying TGF regulation of PGC and GFR. In vivo efferent arteriole TGF may only buffer the signals that cause efferent arteriole resistance to parallel changes in afferent arteriole resistance. Effects of TGF on efferent arterioles perfused orthograde or retrograde were similar, suggesting that glomerular autacoids do not participate in this process.


Assuntos
Glomérulos Renais/irrigação sanguínea , Túbulos Renais/irrigação sanguínea , Néfrons/irrigação sanguínea , Teobromina/análogos & derivados , Animais , Arteríolas/fisiologia , Retroalimentação/efeitos dos fármacos , Masculino , Perfusão , Antagonistas de Receptores Purinérgicos P1 , Coelhos , Teobromina/farmacologia , Xantinas/farmacologia
16.
Methods Mol Med ; 51: 451-70, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-21331736

RESUMO

Isolation and perfusion of single nephron segments was first described by Burg et al. (1) in 1966. This technique has allowed us to study both transepithelial and transmembrane transport in individual nephron segments, including the proximal tubule, under carefully controlled circumstances. This has obvious advantages for experiments that require the epithelium and its junctional complexes to remain intact, but removed from the influences of endogenous neural or humoral mediators, including angiotensins. In addition, control of bath and luminal perfusate compositions and flow rates allows peptides to be added independently to each side of the epithelium. This has particular relevance in the case of angiotensin, which binds to luminal and basolateral receptors and initiates intracellular signal transduction pathways that may differ between the two sites (2).

17.
Kidney Int ; 58(5): 2053-60, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11044225

RESUMO

BACKGROUND: Previous studies have suggested that nitric oxide (NO) produced within cells of the macula densa (MD) modulates tubuloglomerular feedback (TGF). We tested the hypothesis that NO produced in the MD acts locally as an autacoid to activate soluble guanylate cyclase and cGMP-dependent protein kinase in the MD itself. METHODS: Rabbit afferent arterioles (Af-Arts) and attached MD were simultaneously microperfused in vitro. The TGF response was determined by measuring the Af-Art diameter before and after increasing NaCl in the MD perfusate (from 17 mmol/L of Na and 2 of Cl to 65 mmol/L of Na and 50 of Cl). TGF was studied before (control TGF) and after inhibiting components of the NO-cGMP-dependent cascade in the tubular or vascular compartment. RESULTS: Increasing NaCl concentration in the MD perfusate decreased the Af-Art diameter by 3.2 +/- 0.5 microm (from 18.5 +/- 1.3 to 15.4 +/- 1.3 microm, P < 0.001). Adding a soluble guanylate cyclase inhibitor (LY83583) to the MD increased TGF response to 6.3 +/- 1.1 microm (P < 0.031 vs. control TGF). Similarly, when cGMP-dependent protein kinase was inhibited with KT5823, TGF was augmented from 2.6 +/- 0.3 to 4.0 +/- 0.7 microm (P < 0.023). An analogue of cGMP in the MD reversed the TGF-potentiating effect of both 7-nitroindazole (7NI; an nNOS inhibitor) and LY83583. Inhibition of MD guanylate cyclase did not alter the effect of acetylcholine (a NO-cGMP-dependent vasodilator) on the Af-Art. Perfusing the Af-Art with the guanylate cyclase inhibitor did not potentiate TGF, suggesting that the effect of NO occurred at the MD via a cGMP-dependent mechanism. To determine whether the effect of NO in the MD was entirely mediated by cGMP, TGF was studied after giving (1) LY83583 or (2) LY83583 plus 7NI. Adding the nNOS inhibitor to the MD did not potentiate the TGF response further. CONCLUSIONS: We concluded the following: (1) NO produced by the MD inhibits TGF via stimulation of soluble guanylate cyclase, generating cGMP and activating cGMP-dependent protein kinase; (2) NO acts on the MD itself rather than by diffusing to the Af-Art; and (3) most, if not all, of the effect of NO in the MD is due to a cGMP-dependent mechanism rather than to other NO mediators.


Assuntos
GMP Cíclico/fisiologia , Glomérulos Renais/fisiologia , Túbulos Renais Distais/metabolismo , Túbulos Renais/fisiologia , Óxido Nítrico/fisiologia , Animais , Arteríolas/fisiologia , GMP Cíclico/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Ativação Enzimática/fisiologia , Retroalimentação , Guanilato Ciclase/metabolismo , Túbulos Renais Distais/irrigação sanguínea , Túbulos Renais Distais/citologia , Túbulos Renais Distais/fisiologia , Masculino , Coelhos , Solubilidade
18.
Kidney Int ; 58(5): 2069-74, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11044227

RESUMO

BACKGROUND: In vivo and in vitro studies have shown that nitric oxide (NO) is an important modulator of transport processes along the nephron. The thick ascending limb (TAL) plays a significant role in the urine-concentrating mechanism and in the maintenance of acid/base balance. METHODS: TALs from male Sprague-Dawley rats were isolated and perfused, and net bicarbonate flux (J(HCO3)(-) was determined. RESULTS: In perfused TALs, 0.5 mmol/L L-arginine (L-Arg), the substrate for NO synthase, significantly lowered J(HCO3)(-) from 35.4 +/- 4.6 to 23.2 +/- 2.9 pmol. mm(-1). min(-1), a decrease of 36.9 +/- 11.6% (P < 0.025). D-Arg (0.5 mmol/L) had no effect on J(HCO3)(-) (N = 7). In the presence of 5 mmol/L L-NAME, an NO synthase (NOS) inhibitor, the addition of L-Arg did not affect TAL J(HCO3)(-) (43.4 +/- 4.4 vs. 44.6 +/- 5.0 pmol. mm(-1). min(-1)). L-NAME alone (5 mmol/L) did not affect TAL J(HCO3)(-). After removing L-Arg from the bath, J(HCO3)(-) increased from 26.2 +/- 3.9 to 34.8 +/- 3.2 pmol. mm(-1). min(-1) (P < 0.01), indicating no cytotoxicity of NO. We next investigated the effect of cGMP analogues on TAL J(HCO3)(-). 8-Br-cGMP (50 micromol/L) and db-cGMP (50 micromol/L) significantly decreased J(HCO3)(-) by 26.3 +/- 9.1% and 35.1 +/- 11.6%, respectively. In the presence of cGMP (50 micromol/L), the addition of L-Arg had no effect on J(HCO3)(-). In the presence of KT-5823 (2 mircromol/L), a protein kinase G inhibitor, the addition of L-Arg did not change TAL J(HCO3)(-) (N = 5). CONCLUSIONS: We conclude that (1) endogenously produced NO inhibits TAL J(HCO3)(-) in an autocrine manner, (2) cGMP mediates all the effects of NO, and (3) this effect is mediated by protein kinase G activation.


Assuntos
Comunicação Autócrina , Bicarbonatos/metabolismo , Carbazóis , Indóis , Alça do Néfron/metabolismo , Óxido Nítrico/fisiologia , Alcaloides/farmacologia , Animais , Arginina/farmacologia , Transporte Biológico/fisiologia , GMP Cíclico/análogos & derivados , GMP Cíclico/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Inibidores Enzimáticos/farmacologia , Técnicas In Vitro , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Ratos , Ratos Sprague-Dawley
19.
Am J Physiol Renal Physiol ; 279(2): F326-33, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10919853

RESUMO

Endothelin-1 (ET-1) inhibits transport in various nephron segments, and the thick ascending limb of the loop of Henle (TALH) expresses ET-1 receptors. In many tissues, activation of ET(B) receptors stimulates release of NO, and we recently reported that endogenous NO inhibits TALH chloride flux (J(Cl)). However, the relationship between ET-1 and NO in the control of nephron transport has not been extensively studied. We hypothesized that ET-1 decreases NaCl transport by cortical TALHs through activation of ET(B) receptors and release of NO. Exogenous ET-1 (1 nM) decreased J(Cl) from 118.3 +/- 15.0 to 62.7 +/- 13.6 pmol. mm(-1). min(-1) (48.3 +/- 8.2% reduction), whereas removal of ET-1 increased J(Cl) in a separate group of tubules from 87.6 +/- 10.7 to 115.2 +/- 10.3 pmol. mm(-1). min(-1) (34.5 +/- 6.2% increase). To determine whether NO mediates the inhibitory effects of ET-1 on J(Cl), we examined the effect of inhibiting of NO synthase (NOS) with N(G)-nitro-L-arginine methyl ester (L-NAME) on ET-1-induced changes in J(Cl). L-NAME (5 mM) completely prevented the ET-1-induced reduction in J(Cl), whereas D-NAME did not. L-NAME alone had no effect on J(Cl). These data suggest that the effects of ET-1 are mediated by NO. Blockade of ET(B) receptors with BQ-788 prevented the inhibitory effects of 1 nM ET-1. Activation of ET(B) receptors with sarafotoxin S6c mimicked the inhibitory effect of ET-1 on J(Cl) (from 120.7 +/- 12.6 to 75.4 +/- 13.3 pmol. mm(-1). min(-1)). In contrast, ET(A) receptor antagonism with BQ-610 did not prevent ET-1-mediated inhibition of TALH J(Cl) (from 96.5 +/- 10.4 to 69.5 +/- 8.6 pmol. mm(-1). min(-1)). Endothelin increased intracellular calcium from 96.9 +/- 14.0 to 191.4 +/- 11.9 nM, an increase of 110.8 +/- 26.1%. We conclude that exogenous endothelin indirectly decreases TALH J(Cl) by activating ET(B) receptors, increasing intracellular calcium concentration, and stimulating NO release. These data suggest that endothelin acts as a physiological regulator of TALH NO synthesis, thus inhibiting chloride transport and contributing to the natriuretic effects of ET-1 observed in vivo.


Assuntos
Cloretos/antagonistas & inibidores , Endotelinas/fisiologia , Alça do Néfron/metabolismo , Óxido Nítrico/metabolismo , Receptores de Endotelina/fisiologia , Absorção/efeitos dos fármacos , Animais , Cloretos/metabolismo , Endotelinas/farmacologia , Técnicas In Vitro , Masculino , Ratos , Ratos Sprague-Dawley , Receptor de Endotelina B
20.
Hypertension ; 35(1 Pt 2): 319-23, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10642318

RESUMO

We recently reported that the rat thick ascending limb (THAL) possesses an active isoform of nitric oxide synthase (NOS) that is substrate-limited in vitro. NO produced by THAL NOS inhibits chloride flux. Protein and transcript for each of the primary NOS isoforms-endothelial (eNOS), inducible (iNOS), and neuronal (nNOS)-have been demonstrated in THALs. However, the NOS isoform that mediates NO-induced inhibition of chloride flux is unknown. We hypothesized that NO produced from eNOS in the THAL inhibits NaCl transport. THALs from male eNOS, iNOS, and nNOS knockout mice and C57BL/6J wild-type controls were perfused in vitro and the response of transepithelial chloride flux (J(Cl)) to L-arginine (L-Arg), the substrate for NOS, and spermine NONOate (SPM), an NO donor was measured. We first tested whether isolated mouse THALs could synthesize NO and whether this NO inhibits transport. Addition of 0. 5 mmol/L L-Arg to the bath decreased J(Cl) from 105.8+/-17.5 to 79. 2+/-15.8 pmol/mm per minute (P<0.01) in C57BL/6J wild-type mice, whereas addition of D-Arginine had no effects on J(Cl.) In contrast, addition of 0.5 mmol/L L-Arg to the bath did not alter J(Cl) of THALs from eNOS knockout mice. When 10 micromol/L SPM was added to the bath of eNOS knockout THALs, J(Cl) decreased from 89.1+/-8.6 to 74.8+/-7.5 pmol/mm/min (P<0.05). Thus the lack of responsiveness of eNOS knockout THALs to L-Arg was not due to an inability to respond to NO. We next evaluated the role of iNOS and nNOS in the response to L-Arg. Addition of 0.5 mmol/L L-Arg to the bath decreased J(Cl) in THALs from iNOS and nNOS knockout mice by 37.7+/-6.4% (P<0.05) and 31.8+/-8.3% (P<0.01), respectively. We conclude that eNOS is the active isoform of NOS in the THAL under basal conditions. Mouse THAL eNOS responds to exogenous L-Arg by increasing NO production, which, in turn, inhibits J(Cl).


Assuntos
Arginina/farmacologia , Cloretos/metabolismo , Alça do Néfron/enzimologia , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/genética , Pressão Sanguínea , Regulação Enzimológica da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Sódio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA