Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Am J Physiol Renal Physiol ; 316(5): F970-F973, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30838871

RESUMO

In individuals on a regular "Western" diet, furosemide induces a kaliuresis and reduction in plasma K concentration by inhibiting Na reabsorption in the thick ascending limb of Henle's loop, enhancing delivery of Na to the aldosterone-sensitive distal nephron. In the aldosterone-sensitive distal nephron, the increased Na delivery stimulates K wasting due to an exaggerated exchange of epithelial Na channel-mediated Na reabsorption of secreted K. The effects of furosemide are different in mice fed a high-K, alkaline (HK) diet: the large-conductance Ca-activated K (BK) channel, in conjunction with the BK ß4-subunit (BK-α/ß4), mediates K secretion from intercalated cells (IC) of the connecting tubule and collecting ducts. The urinary alkaline load is necessary for BK-α/ß4-mediated K secretion in HK diet-fed mice. However, furosemide acidifies the urine by increasing vacuolar ATPase expression and acid secretion from IC, thereby inhibiting BK-α/ß4-mediated K secretion and sparing K. In mice fed a low-Na, high-K (LNaHK) diet, furosemide causes a greater increase in plasma K concentration and reduction in K excretion than in HK diet-fed mice. Micropuncture of the early distal tubule of mice fed a LNaHK diet, but not a regular or a HK diet, reveals K secretion in the thick ascending limb of Henle's loop. The sites of action of K secretion in individuals consuming a high-K diet should be taken into account when diuretic agents known to waste K with low or moderate K intakes are prescribed.


Assuntos
Diurético Poupador de Potássio/farmacologia , Furosemida/farmacologia , Rim/efeitos dos fármacos , Potássio na Dieta/urina , Eliminação Renal/efeitos dos fármacos , Animais , Diurético Poupador de Potássio/toxicidade , Furosemida/toxicidade , Rim/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Camundongos , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo
2.
Am J Physiol Renal Physiol ; 316(2): F341-F350, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30484346

RESUMO

Special high-K diets have cardioprotective effects and are often warranted in conjunction with diuretics such as furosemide for treating hypertension. However, it is not understood how a high-K diet (HK) influences the actions of diuretics on renal K+ handling. Furosemide acidifies the urine by increasing acid secretion via the Na+-H+ exchanger 3 (NHE3) in TAL and vacuolar H+-ATPase (V-ATPase) in the distal nephron. We previously found that an alkaline urine is required for large conductance Ca2+-activated K+ (BK)-αß4-mediated K+ secretion in mice on HK. We therefore hypothesized that furosemide could reduce BK-αß4-mediated K+ secretion by acidifying the urine. Treating with furosemide (drinking water) for 11 days led to decreased urine pH in both wild-type (WT) and BK-ß4-knockout mice (BK-ß4-KO) with increased V-ATPase expression and elevated plasma aldosterone levels. However, furosemide decreased renal K+ clearance and elevated plasma [K+] in WT but not BK-ß4-KO. Western blotting and immunofluorescence staining showed that furosemide treatment decreased cortical expression of BK-ß4 and reduced apical localization of BK-α in connecting tubules. Addition of the carbonic anhydrase inhibitor, acetazolamide, to furosemide water restored urine pH along with renal K+ clearance and plasma [K+] to control levels. Acetazolamide plus furosemide also restored the cortical expression of BK-ß4 and BK-α in connecting tubules. These results indicate that in mice adapted to HK, furosemide reduces BK-αß4-mediated K+ secretion by acidifying the urine.


Assuntos
Dieta , Furosemida/farmacologia , Rim/efeitos dos fármacos , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Potássio/metabolismo , Eliminação Renal/efeitos dos fármacos , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacologia , Equilíbrio Ácido-Base , Animais , Feminino , Concentração de Íons de Hidrogênio , Rim/metabolismo , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/deficiência , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Potássio/sangue , Potássio/urina , ATPases Vacuolares Próton-Translocadoras/metabolismo
3.
Kidney Int ; 92(4): 864-875, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28688582

RESUMO

Because of its cardio-protective effects, a low-Na, high-K diet (LNaHK) is often warranted in conjunction with diuretics to treat hypertensive patients. However, it is necessary to understand the renal handling of such diets in order to choose the best diuretic. Wild-type (WT) or Renal Outer Medullary K channel (ROMK) knockout mice (KO) were given a regular (CTRL), LNaHK, or high-K diet (HK) for 4-7 days. On LNaHK, mice treated with either IP furosemide for 12 hrs, or given furosemide in drinking water for 7 days, exhibited decreased K clearance. We used free-flow micropuncture to measure the [K+] in the early distal tubule (EDT [K+]) before and after furosemide treatment. Furosemide increased the EDT [K+] in WT on CTRL but decreased that in WT on LNaHK. Furosemide did not affect the EDT [K+] of KO on LNaHK or WT on HK. Furosemide-sensitive Na+ excretion was significantly greater in mice on LNaHK than those on CTRL or HK. Patch clamp analysis of split-open TALs revealed that 70-pS ROMK exhibited a higher open probability (Po) but similar density in mice on LNaHK, compared with CTRL. No difference was found in the density or Po of the 30 pS K channels between the two groups. These results indicate mice on LNaHK exhibited furosemide-sensitive net K+ secretion in the TAL that is dependent on increased NKCC2 activity and mediated by ROMK. We conclude that furosemide is a K-sparing diuretic by decreasing the TAL net K+ secretion in subjects on LNaHK.


Assuntos
Diuréticos/efeitos adversos , Hipertensão/terapia , Túbulos Renais Distais/metabolismo , Potássio na Dieta/metabolismo , Sódio na Dieta/metabolismo , Animais , Dieta Mediterrânea , Dieta Paleolítica , Furosemida/efeitos adversos , Humanos , Hipertensão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Eliminação Renal/efeitos dos fármacos , Membro 1 da Família 12 de Carreador de Soluto/metabolismo
4.
Am J Physiol Renal Physiol ; 310(7): F581-F595, 2016 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-26739887

RESUMO

A low-Na+, high-K+ diet (LNaHK) is considered a healthier alternative to the "Western" high-Na+ diet. Because the mechanism for K+ secretion involves Na+ reabsorptive exchange for secreted K+ in the distal nephron, it is not understood how K+ is eliminated with such low Na+ intake. Animals on a LNaHK diet produce an alkaline load, high urinary flows, and markedly elevated plasma ANG II and aldosterone levels to maintain their K+ balance. Recent studies have revealed a potential mechanism involving the actions of alkalosis, urinary flow, elevated ANG II, and aldosterone on two types of K+ channels, renal outer medullary K+ and large-conductance K+ channels, located in principal and intercalated cells. Here, we review these recent advances.


Assuntos
Dieta , Potássio na Dieta/metabolismo , Sódio na Dieta/metabolismo , Animais , Humanos
5.
Am J Physiol Renal Physiol ; 309(6): F489-91, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26136555

RESUMO

The electrogenic Na(+)-HCO3 (-) cotransporter 2 (NBCe2) is a newly discovered protein in the distal nephron. Our understanding is minimal regarding its physiological role in renal electrolyte transport. In this mini-review, we summarize the potential function of NBCe2 in the regulation of blood pressure, acid-base, and K(+) and Ca(2+) transport in the distal nephron.


Assuntos
Eletrólitos/metabolismo , Néfrons/metabolismo , Simportadores de Sódio-Bicarbonato/metabolismo , Animais , Transporte Biológico , Humanos , Hipertensão Renal/metabolismo , Rim/metabolismo , Equilíbrio Hidroeletrolítico/fisiologia
6.
Am J Physiol Renal Physiol ; 309(6): F523-30, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26109087

RESUMO

In many circumstances, the pathogenesis of distal renal tubular acidosis (dRTA) is not understood. In the present study, we report that a mouse model lacking the electrogenic Na(+)-HCO3 (-) cotransporter [NBCe2/Slc4a5; NBCe2 knockout (KO) mice] developed dRTA after an oral acid challenge. NBCe2 expression was identified in the connecting tubule (CNT) of wild-type mice, and its expression was significantly increased after acid loading. NBCe2 KO mice did not have dRTA when on a standard mouse diet. However, after acid loading, NBCe2 KO mice exhibited complete features of dRTA, characterized by insufficient urinary acidification, hyperchloremic hypokalemic metabolic acidosis, and hypercalciuria. Additional experiments showed that NBCe2 KO mice had decreased luminal transepithelial potential in the CNT, as revealed by micropuncture. Further immunofluorescence and Western blot experiments found that NBCe2 KO mice had increased expression of H(+)-ATPase B1 in the plasma membrane. These results showed that NBCe2 KO mice with acid loading developed increased urinary K(+) and Ca(2+) wasting due to decreased luminal transepithelial potential in the CNT. NBCe2 KO mice compensated to maintain systemic pH by increasing H(+)-ATPase in the plasma membrane. Therefore, defects in NBCe2 can cause dRTA, and NBCe2 has an important role to regulate urinary acidification and the transport of K(+) and Ca(2+) in the distal nephron.


Assuntos
Acidose Tubular Renal/metabolismo , Túbulos Renais Distais/metabolismo , Simportadores de Sódio-Bicarbonato/genética , Simportadores de Sódio-Bicarbonato/fisiologia , Animais , Membrana Celular/metabolismo , Cloro/metabolismo , Hipercalciúria/metabolismo , Hipopotassemia/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , ATPases Translocadoras de Prótons/metabolismo , Simportadores de Sódio-Bicarbonato/metabolismo
7.
Hypertension ; 66(1): 68-74, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25941340

RESUMO

The gene SLC4A5 encodes the Na(+)-HCO3 (-) cotransporter electrogenic 2, which is located in the distal nephron. Genetically deleting Na(+)-HCO3 (-) cotransporter electrogenic 2 (knockout) causes Na(+)-retention and hypertension, a phenotype that is diminished with alkali loading. We performed experiments with acid-loaded mice and determined whether overactive epithelial Na(+) channels (ENaC) or the Na(+)-Cl(-) cotransporter causes the Na(+) retention and hypertension in knockout. In untreated mice, the mean arterial pressure was higher in knockout, compared with wild-type (WT); however, treatment with amiloride, a blocker of ENaC, abolished this difference. In contrast, hydrochlorothiazide, an inhibitor of Na(+)-Cl(-) cotransporter, decreased mean arterial pressure in WT, but not knockout. Western blots showed that quantity of plasmalemmal full-length ENaC-α was significantly higher in knockout than in WT. Amiloride treatment caused a 2-fold greater increase in Na(+) excretion in knockout, compared with WT. In knockout, but not WT, amiloride treatment decreased plasma [Na(+)] and urinary K(+) excretion, but increased hematocrit and plasma [K(+)] significantly. Micropuncture with microelectrodes showed that the [K(+)] was significantly higher and the transepithelial potential (Vte) was significantly lower in the late distal tubule of the knockout compared with WT. The reduced Vte in knockout was amiloride sensitive and therefore revealed an upregulation of electrogenic ENaC-mediated Na(+) reabsorption in this segment. These results show that, in the absence of Na(+)-HCO3 (-) cotransporter electrogenic 2 in the late distal tubule, acid-loaded mice exhibit disinhibition of ENaC-mediated Na(+) reabsorption, which results in Na(+) retention, K(+) wasting, and hypertension.


Assuntos
Canais Epiteliais de Sódio/fisiologia , Hipertensão Renal/metabolismo , Simportadores de Sódio-Bicarbonato/deficiência , Amilorida/farmacologia , Amilorida/uso terapêutico , Animais , Anti-Hipertensivos/uso terapêutico , Modelos Animais de Doenças , Diuréticos/uso terapêutico , Canais Epiteliais de Sódio/efeitos dos fármacos , Hematócrito , Hidroclorotiazida/uso terapêutico , Concentração de Íons de Hidrogênio , Hipertensão Renal/tratamento farmacológico , Hipertensão Renal/genética , Hipopotassemia/etiologia , Túbulos Renais Distais/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Natriurese/efeitos dos fármacos , Natriurese/genética , Polimorfismo de Nucleotídeo Único , Potássio/metabolismo , Sódio/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Bloqueadores dos Canais de Sódio/uso terapêutico , Simportadores de Sódio-Bicarbonato/genética , Simportadores de Sódio-Bicarbonato/fisiologia
8.
PLoS One ; 10(1): e0115515, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25607984

RESUMO

A low Na, high K diet (LNaHK) is associated with a low rate of cardiovascular (CV) disease in many societies. Part of the benefit of LNaHK relies on its diuretic effects; however, the role of aldosterone (aldo) in the diuresis is not understood. LNaHK mice exhibit an increase in renal K secretion that is dependent on the large, Ca-activated K channel, (BK-α with accessory BK-ß4; BK-α/ß4). We hypothesized that aldo causes an osmotic diuresis by increasing BK-α/ß4-mediated K secretion in LNaHK mice. We found that the plasma aldo concentration (P[aldo]) was elevated by 10-fold in LNaHK mice compared with control diet (Con) mice. We subjected LNaHK mice to either sham surgery (sham), adrenalectomy (ADX) with low aldo replacement (ADX-LA), or ADX with high aldo replacement (ADX-HA). Compared to sham, the urinary flow, K excretion rate, transtubular K gradient (TTKG), and BK-α and BK-ß4 expressions, were decreased in ADX-LA, but not different in ADX-HA. BK-ß4 knockout (ß4KO) and WT mice exhibited similar K clearance and TTKG in the ADX-LA groups; however, in sham and ADX-HA, the K clearance and TTKG of ß4KO were less than WT. In response to amiloride treatment, the osmolar clearance was increased in WT Con, decreased in WT LNaHK, and unchanged in ß4KO LNaHK. These data show that the high P[aldo] of LNaHK mice is necessary to generate a high rate of BK-α/ß4-mediated K secretion, which creates an osmotic diuresis that may contribute to a reduction in CV disease.


Assuntos
Aldosterona/metabolismo , Dieta , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Potássio/farmacologia , Potássio/farmacocinética , Sódio/farmacologia , Animais , Doenças Cardiovasculares/dietoterapia , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Camundongos
9.
Curr Opin Pharmacol ; 15: 28-32, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24721651

RESUMO

Large conductance, Ca-activated K channels (BK) are abundantly located in cells of vasculature, glomerulus, and distal nephron, where they are involved in maintaining blood volume, blood pressure, and K homeostasis. In mesangial cells and smooth muscle cells of vessels, the BK-α pore associates with BK-ß1 subunits and regulates contraction in a Ca-mediated feedback manner. The BK-ß1 also resides in connecting tubule cells of the nephron. BK-ß1 knockout mice (ß1KO) exhibit fluid retention, hypertension, and compromised K handling. The BK-α/ß4 resides in acid/base transporting intercalated cells (IC) of the distal nephron, where they mediate K secretion in mammals on a high K, alkaline diet. BK-α expression in IC is increased by a high K diet via aldosterone. The BK-ß4 subunit and alkaline urine are necessary for the luminal expression and function of BK-α in mouse IC. In distal nephron cells, membrane BK-α expression is inhibited by WNK4 in in vitro expression systems, indicating a role in the hyperkalemic phenotype in patients with familial hyperkalemic hypertension type 2 (FHHt2). ß1KO and BK-ß4 knockout mice (ß4KO) are hypertensive because of exaggerated epithelial Na channels (ENaC) mediated Na retention in an effort to secrete K via only renal outer medullary K channels (ROMK). BK hypertension is resistant to thiazides and furosemide, and would be more amenable to ENaC and aldosterone inhibiting drugs. Activators of BK-α/ß1 or BK-α/ß4 might be effective blood pressure lowering agents for a subset of hypertensive patients. Inhibitors of renal BK would effectively spare K in patients with Bartter Syndrome, a renal K wasting disease.


Assuntos
Dieta , Diuréticos/farmacologia , Interações Alimento-Droga , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Potássio/metabolismo , Animais , Homeostase/efeitos dos fármacos , Camundongos , Camundongos Knockout
10.
Kidney Int ; 86(1): 139-45, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24573316

RESUMO

The large-conductance, calcium-activated BK-α/ß4 potassium channel, localized to the intercalated cells of the distal nephron, mediates potassium secretion during high-potassium, alkaline diets. Here we determine whether BK-α/ß4-mediated potassium transport is dependent on epithelial sodium channel (ENaC)-mediated sodium reabsorption. We maximized sodium-potassium exchange in the distal nephron by feeding mice a low-sodium, high-potassium diet. Wild-type and BK-ß4 knockout mice were maintained on a low-sodium, high-potassium, alkaline diet or a low-sodium, high-potassium, acidic diet for 7-10 days. Wild-type mice maintained potassium homeostasis on the alkaline, but not acid, diet. BK-ß4 knockout mice could not maintain potassium homeostasis on either diet. During the last 12 h of diet, wild-type mice on either a regular, alkaline, or an acid diet, or knockout mice on an alkaline diet, were administered amiloride (an ENaC inhibitor). Amiloride enhanced sodium excretion in all wild-type and knockout groups to similar values; however, amiloride diminished potassium excretion by 59% in wild-type but only by 33% in knockout mice on an alkaline diet. Similarly, amiloride decreased the trans-tubular potassium gradient by 68% in wild-type but only by 42% in knockout mice on an alkaline diet. Amiloride treatment equally enhanced sodium excretion and diminished potassium secretion in knockout mice on an alkaline diet and wild-type mice on an acid diet. Thus, the enhanced effect of amiloride on potassium secretion in wild-type compared to knockout mice on the alkaline diet clarify a BK- α/ß4-mediated potassium secretory pathway in intercalated cells driven by ENaC-mediated sodium reabsorption linked to bicarbonate secretion.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Néfrons/metabolismo , Sódio/metabolismo , Amilorida/farmacologia , Animais , Bloqueadores do Canal de Sódio Epitelial/farmacologia , Hidroclorotiazida/farmacologia , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/deficiência , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/deficiência , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Néfrons/efeitos dos fármacos , Potássio/metabolismo , Potássio na Dieta/administração & dosagem , Inibidores de Simportadores de Cloreto de Sódio/farmacologia , Sódio na Dieta/administração & dosagem
11.
Am J Physiol Renal Physiol ; 305(4): F463-76, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23761673

RESUMO

In the distal nephron, the large-conductance Ca-activated K (BK) channel, comprised of a pore-forming-α (BK-α) and the BK-ß4 subunit, promotes K excretion when mice are maintained on a high-K alkaline diet (HK-alk). We examined whether BK-ß4 and the acid-base status regulate apical membrane expression of BK-α in the cortical (CCD) and medullary collecting ducts (MCD) using immunohistochemical analysis (IHC) and Western blot. With the use of IHC, BK-α of mice on acontrol diet localized mostly cytoplasmically in intercalated cells (IC) of the CCD and in the perinuclear region of both principle cells (PC) and IC of the MCD. HK-alk wild-type mice (WT), but not BK-ß4 knockout mice (ß4KO), exhibited increased apical BK-α in both the CCD and MCD. When given a high-K acidic diet (HK-Cl), BK-α expression increased but remained cytoplasmic in the CCD and perinuclear in the MCD of both WT and ß4KO. Western blot confirmed that total BK-α expression was enhanced by either HK-alk or HK-Cl but only increased in the plasma membrane with HK-alk. Compared with controls, mice drinking NaHCO3 water exhibited more apical BK-α and total cellular BK-ß4. Spironolactone given to mice on HK-alk significantly reduced K secretion and decreased total cellular BK-α but did not affect cellular BK-ß4 and apical BK-α. Experiments with MDCK-C11 cells indicated that BK-ß4 stabilizes surface BK-α by inhibiting degradation through a lysosomal pathway. These data suggest that aldosterone mediates a high-K-induced increase in BK-α and urinary alkalinization increases BK-ß4 expression, which promotes the apical localization of BK-α.


Assuntos
Aldosterona/metabolismo , Túbulos Renais Coletores/metabolismo , Rim/metabolismo , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Néfrons/metabolismo , Potássio/farmacologia , Animais , Western Blotting , Técnicas de Cultura de Células , Concentração de Íons de Hidrogênio , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Potássio/metabolismo
12.
J Biol Chem ; 287(46): 38552-8, 2012 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-23002235

RESUMO

The epithelial Na(+) channel (ENaC) in the aldosterone-sensitive distal nephron (ASDN) is under tonic inhibition by a local purinergic signaling system responding to changes in dietary sodium intake. Normal BK(Ca) channel function is required for flow-sensitive ATP secretion in the ASDN. We tested here whether ATP secreted through connexin channels in a coupled manner with K(+) efflux through BK(Ca) channels is required for inhibitory purinergic regulation of ENaC in response to increases in sodium intake. Inhibition of connexin channels relieves purinergic inhibition of ENaC. Deletion of the BK-ß4 regulatory subunit, which is required for normal BK(Ca) channel function and flow-sensitive ATP secretion in the ASDN, suppresses increases in urinary ATP in response to increases in sodium intake. As a consequence, ENaC activity, particularly in the presence of high sodium intake, is inappropriately elevated in BK-ß4 null mice. ENaC in BK-ß4 null mice, however, responds normally to exogenous ATP, indicating that increases in activity do not result from end-organ resistance but rather from lowered urinary ATP. Consistent with this, disruption of purinergic regulation increases ENaC activity in wild type but not BK-ß4 null mice. Consequently, sodium excretion is impaired in BK-ß4 null mice. These results demonstrate that the ATP secreted in the ASDN in a BK(Ca) channel-dependent manner is physiologically available for purinergic inhibition of ENaC in response to changes in sodium homeostasis. Impaired sodium excretion resulting form loss of normal purinergic regulation of ENaC in BK-ß4 null mice likely contributes to their elevated blood pressure.


Assuntos
Trifosfato de Adenosina/química , Canais Epiteliais de Sódio/metabolismo , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Néfrons/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Pressão Sanguínea , Conexinas/química , Homeostase , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Técnicas de Patch-Clamp , Sódio/química , Sódio/metabolismo , Sódio na Dieta/metabolismo
13.
Am J Physiol Renal Physiol ; 303(11): F1563-71, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22993067

RESUMO

Ca-activated K channels (BK), which are stimulated by high distal nephron flow, are utilized during high-K conditions to remove excess K. Because BK predominantly reside with BK-ß4 in acid/base-transporting intercalated cells (IC), we determined whether BK-ß4 knockout mice (ß4KO) exhibit deficient K excretion when consuming a high-K alkaline diet (HK-alk) vs. high-K chloride diet (HK-Cl). When wild type (WT) were placed on HK-alk, but not HK-Cl, renal BK-ß4 expression increased (Western blot). When WT and ß4KO were placed on HK-Cl, plasma K concentration ([K]) was elevated compared with control K diets; however, K excretion was not different between WT and ß4KO. When HK-alk was consumed, the plasma [K] was lower and K clearance was greater in WT compared with ß4KO. The urine was alkaline in mice on HK-alk; however, urinary pH was not different between WT and ß4KO. Immunohistochemical analysis of pendrin and V-ATPase revealed the same increases in ß-IC, comparing WT and ß4KO on HK-alk. We found an amiloride-sensitive reduction in Na excretion in ß4KO, compared with WT, on HK-alk, indicating enhanced Na reabsorption as a compensatory mechanism to secrete K. Treating mice with an alkaline, Na-deficient, high-K diet (LNaHK) to minimize Na reabsorption exaggerated the defective K handling of ß4KO. When WT on LNaHK were given NH(4)Cl in the drinking water, K excretion was reduced to the magnitude of ß4KO on LNaHK. These results show that WT, but not ß4KO, efficiently excretes K on HK-alk but not on HK-Cl and suggest that BK-α/ß4-mediated K secretion is promoted by bicarbonaturia.


Assuntos
Bicarbonatos/farmacologia , Túbulos Renais Distais/metabolismo , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Néfrons/metabolismo , Potássio na Dieta/farmacologia , Potássio/metabolismo , Animais , Canais Epiteliais de Sódio/metabolismo , Túbulos Renais Distais/citologia , Túbulos Renais Distais/efeitos dos fármacos , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/deficiência , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Néfrons/citologia , Néfrons/efeitos dos fármacos , Cloreto de Potássio/farmacologia , Sódio/metabolismo
14.
Curr Opin Nephrol Hypertens ; 20(5): 512-7, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21670674

RESUMO

PURPOSE OF REVIEW: This review summarizes recent studies of hypertension associated with a defect in renal K excretion due to genetic deletions of various components of the large, Ca-activated K channel (BK), and describes new evidence and theories regarding K secretory roles of BK in intercalated cells. RECENT FINDINGS: Isolated perfused tubule methods have revealed the importance of BK in flow-induced K secretion. Subsequently, mice with genetically deleted BK subunits revealed the complexities of BK-mediated K secretion. Deletion of BKα results in extreme aldosteronism, hypertension, and an absence of flow-induced K secretion. Deletion of the BKß1 ancillary subunit results in decreased handling of a K load, increased plasma K, mild aldosteronism and hypertension that is exacerbated by a high K diet. Deletion of BKß4 (ß4KO) leads to insufficient K handling, high plasma K, fluid retention, but with milder hypertension. Fluid retention in ß4KO may be the result of insufficient flow-induced secretion of adenosine triphosphate (ATP), which normally inhibits epithelial Na channels (ENaCs). SUMMARY: Classical physiological analysis of electrolyte handling in knockout mice has enlightened our understanding of the mechanism of handling K loads by renal K channels. Studies have focused on the different roles of BK-α/ß1 and BK-α/ß4 in the kidney. BKß1 hypertension may be a 'three-hit' hypertension, involving a K secretory defect, elevated production of aldosterone, and increased vascular tone. The disorders observed in BK knockout mice have shed new insights on the importance of proper renal K handling for maintaining volume balance and blood pressure.


Assuntos
Hipertensão/metabolismo , Túbulos Renais/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Potássio/metabolismo , Aldosterona/metabolismo , Animais , Pressão Sanguínea , Humanos , Hipertensão/fisiopatologia , Transporte de Íons , Túbulos Renais/fisiopatologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/deficiência , Canais de Potássio Ativados por Cálcio de Condutância Alta/genética , Camundongos , Camundongos Knockout , Potássio/sangue , Equilíbrio Hidroeletrolítico
15.
Am J Physiol Renal Physiol ; 300(6): F1319-26, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21454249

RESUMO

Increased flow in the distal nephron induces K secretion through the large-conductance, calcium-activated K channel (BK), which is primarily expressed in intercalated cells (IC). Since flow also increases ATP release from IC, we hypothesized that purinergic signaling has a role in shear stress (τ; 10 dynes/cm(2)) -induced, BK-dependent, K efflux. We found that 10 µM ATP led to increased IC Ca concentration, which was significantly reduced in the presence of the P(2) receptor blocker suramin or calcium-free buffer. ATP also produced BK-dependent K efflux, and IC volume decrease. Suramin inhibited τ-induced K efflux, suggesting that K efflux is at least partially dependent on purinergic signaling. BK-ß4 small interfering (si) RNA, but not nontarget siRNA, decreased ATP secretion and both ATP-dependent and τ-induced K efflux. Similarly, carbenoxolone (25 µM), which blocks connexins, putative ATP pathways, blocked τ-induced K efflux and ATP secretion. Compared with BK-ß4(-/-) mice, wild-type mice with high distal flows exhibited significantly more urinary ATP excretion. These data demonstrate coupled electrochemical efflux between K and ATP as part of the mechanism for τ-induced ATP release in IC.


Assuntos
Trifosfato de Adenosina/metabolismo , Cálcio/metabolismo , Néfrons/metabolismo , Canais de Potássio Cálcio-Ativados/metabolismo , Potássio/metabolismo , Animais , Western Blotting , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Canais de Potássio Cálcio-Ativados/genética , RNA Interferente Pequeno , Estatísticas não Paramétricas
16.
Kidney Int ; 78(10): 956-62, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20720523

RESUMO

Large, Ca-activated K channels (BK) are comprised of an α pore (BKα) and one of four ß subunits (BKß1-4). When the gene for BKß1 is knocked out (BKß1-KO), the result is increased myogenic tone of vascular smooth muscle and hypertension. We reexamined whether the hypertension is entirely due to increased vascular tone, because most monogenic forms of hypertension have renal origins and BKß1 resides in renal connecting tubule (CNT) cells. Moreover, BKß1 is localized in the adrenal glands, where it may control production of aldosterone. This review will summarize our report that a majority of the hypertension of BKß1-KO is the result of insufficient handling of dietary K, resulting in increased plasma K and hyperaldosteronism, the latter promoting Na and fluid retention. The fluid retention and hypertension are exacerbated by a high-K diet and reduced by eplerenone, an aldosterone receptor inhibitor. Genetic knockout of BKß4 (BKß4-KO), which resides in intercalated cells, also exhibits deficient K excretion, fluid retention, and mild hypertension that is not exacerbated when animals are treated with a high-K diet. These results show that the hypertension associated with BKß1-KO occurs because of enhanced fluid retention, as well as because of the previously described vascular dysfunction.


Assuntos
Hiperaldosteronismo/fisiopatologia , Hipertensão/fisiopatologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Medula Suprarrenal/fisiopatologia , Animais , Modelos Animais de Doenças , Humanos , Hipertensão/genética , Rim/fisiopatologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/genética , Camundongos , Camundongos Knockout , Músculo Liso Vascular/fisiopatologia , Potássio/metabolismo
17.
Am J Physiol Renal Physiol ; 299(3): F507-16, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20576683

RESUMO

Large-conductance, calcium-activated potassium channels (BK) are expressed in principal cells (PC) and intercalated cells (IC) in mammalian nephrons as BK-alpha/beta1 and BK-alpha/beta4, respectively. IC, which protrude into the lumens of tubules, express substantially more BK than PC despite lacking sufficient Na-K-ATPase to support K secretion. We previously showed in mice that IC exhibit size reduction when experiencing high distal flows induced by a high-K diet. We therefore tested the hypothesis that BK-alpha/beta4 are regulators of IC volume via a shear stress (tau)-induced, calcium-dependent mechanism, resulting in a reduction in intracellular K content. We determined by Western blot and immunocytochemical analysis that C11-Madin-Darby canine kidney cells contained a predominance of BK-alpha/beta4. To determine the role of BK-alpha/beta4 in tau-induced volume reduction, we exposed C11 cells to tau and measured K efflux by flame photometry and cell volume by calcein staining, which changes inversely to cell volume. With 10 dynes/cm(2), calcein intensity significantly increased 39% and monovalent cationic content decreased significantly by 37% compared with static conditions. Furthermore, the shear-induced K loss from C11 was abolished by the reduction of extracellular calcium, addition of 5 mM TEA, or BK-beta4 small interfering (si) RNA, but not by addition of nontarget siRNA. These results show that BK-alpha/beta4 plays a role in shear-induced K loss from IC, suggesting that BK-alpha/beta4 regulate IC volume during high-flow conditions. Furthermore, these results support the use of C11 cells as in vitro models for studying BK-related functions in IC of the kidney.


Assuntos
Tamanho Celular , Rim/citologia , Rim/metabolismo , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Estresse Mecânico , Animais , Cálcio/metabolismo , Linhagem Celular , Cães , Modelos Animais , Potássio/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , RNA Interferente Pequeno/farmacologia , Tetraetilamônio/farmacologia
18.
J Am Soc Nephrol ; 21(4): 634-45, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20299355

RESUMO

The large-conductance, calcium-activated potassium (BK) channels help eliminate potassium in mammals consuming potassium-rich diets. In the distal nephron, principal cells contain BK-alpha/beta1 channels and intercalated cells contain BK-alpha/beta4 channels. We studied whether BK-beta4-deficient mice (Kcnmb4(-/-)) have altered renal sodium and potassium clearances compared with wild-type mice when fed a regular or potassium-rich diet for ten days. We did not detect differences in urinary flow or fractional excretions of potassium (FE(K)) or sodium (FE(Na)) between Kcnmb4-deficient and wild-type mice fed a regular diet. However, a potassium-rich diet led to >4-fold increases in urinary flows for both groups of mice, although Kcnmb4-deficient mice exhibited less urinary flow, higher plasma potassium concentration, more fluid retention, and significantly lower FE(K) and FE(Na) than wild-type mice despite similar plasma aldosterone levels. Immunohistochemical analysis revealed increased basolateral Na-K-ATPase in principal cells of all potassium-adapted mice, but expression of Na-K-ATPase in intercalated cells was >10-fold lower. The size of intercalated cells reduced and luminal volume increased among potassium-adapted wild-type but not Kcnmb4-deficient mice. Paradoxically, this led to increased urinary fluid velocity in potassium-adapted Kcnmb4-deficient mice compared with wild-type mice. Taken together, these data suggest that BK-alpha/beta4 channels in intercalated cells reduce cell size, increasing luminal volume to accommodate higher distal flow rates during potassium adaptation. These changes streamline flow across the principal cells, producing gradients more favorable for potassium secretion and less favorable for sodium reabsorption.


Assuntos
Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/fisiologia , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/fisiologia , Potássio/metabolismo , Sódio/metabolismo , Adaptação Fisiológica , Animais , Camundongos
19.
Kidney Int ; 76(9): 968-76, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19710627

RESUMO

Patients with Alport's syndrome develop a number of pro-inflammatory cytokine and matrix metalloproteinase (MMP) abnormalities that contribute to progressive renal failure. Changes in the composition and structure of the glomerular basement membranes likely alter the biomechanics of cell adhesion and signaling in these patients. To test if enhanced strain on the capillary tuft due to these structural changes contributes to altered gene regulation, we subjected cultured podocytes to cyclic biomechanical strain. There was robust induction of interleukin (IL)-6, along with MMP-3, -9, -10, and -14, but not MMP-2 or -12 by increased strain. Neutralizing antibodies against IL-6 attenuated the strain-mediated induction of MMP-3 and -10. Alport mice treated with a general inhibitor of nitric oxide synthase (L-NAME) developed significant hypertension and increased IL-6 and MMP-3 and -10 in their glomeruli relative to those of normotensive Alport mice. These hypertensive Alport mice also had elevated proteinuria along with more advanced histological and ultrastructural glomerular basement membrane damage. We suggest that MMP and cytokine dysregulation may constitute a maladaptive response to biomechanical strain in the podocytes of Alport patients, thus contributing to glomerular disease initiation and progression.


Assuntos
Membrana Basal Glomerular/metabolismo , Interleucina-6/genética , Glomérulos Renais/metabolismo , Metaloproteinases da Matriz/genética , Nefrite Hereditária/genética , Podócitos/metabolismo , Adaptação Fisiológica/genética , Animais , Pressão Sanguínea , Células Cultivadas , Citoesqueleto/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Hipertensão/induzido quimicamente , Hipertensão/genética , Hipertensão/fisiopatologia , Interleucina-6/metabolismo , Glomérulos Renais/fisiopatologia , Metaloproteinase 10 da Matriz/genética , Metaloproteinase 14 da Matriz/genética , Metaloproteinase 3 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NG-Nitroarginina Metil Éster , Nefrite Hereditária/metabolismo , Nefrite Hereditária/fisiopatologia , Proteinúria/induzido quimicamente , Proteinúria/genética , Proteinúria/fisiopatologia , RNA Mensageiro/metabolismo , Cloreto de Sódio na Dieta , Estresse Mecânico , Fatores de Tempo
20.
Proc Natl Acad Sci U S A ; 106(28): 11800-5, 2009 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-19556540

RESUMO

Mice lacking the beta1-subunit (gene, Kcnmb1; protein, BK-beta1) of the large Ca-activated K channel (BK) are hypertensive. This phenotype is thought to result from diminished BK currents in vascular smooth muscle where BK-beta1 is an ancillary subunit. However, the beta1-subunit is also expressed in the renal connecting tubule (CNT), a segment of the aldosterone-sensitive distal nephron, where it associates with BK and facilitates K secretion. Because of the correlation between certain forms of hypertension and renal defects, particularly in the distal nephron, it was determined whether the hypertension of Kcnmb1(-/-) has a renal origin. We found that Kcnmb1(-/-) are hypertensive, volume expanded, and have reduced urinary K and Na clearances. These conditions are exacerbated when the animals are fed a high K diet (5% K; HK). Supplementing HK-fed Kcnmb1(-/-) with eplerenone (mineralocorticoid receptor antagonist) corrected the fluid imbalance and more than 70% of the hypertension. Finally, plasma [aldo] was elevated in Kcnmb1(-/-) under basal conditions (control diet, 0.6% K) and increased significantly more than wild type when fed the HK diet. We conclude that the majority of the hypertension of Kcnmb1(-/-) is due to aldosteronism, resulting from renal potassium retention and hyperkalemia.


Assuntos
Hiperaldosteronismo/complicações , Hiperpotassemia/complicações , Hipertensão/genética , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/deficiência , Potássio/metabolismo , Análise de Variância , Animais , Eplerenona , Hiperaldosteronismo/etiologia , Hipertensão/etiologia , Hipertensão/metabolismo , Túbulos Renais Coletores/metabolismo , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Camundongos , Camundongos Knockout , Espironolactona/análogos & derivados
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...