Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Cell Physiol ; 234(8): 13917-13930, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30633335

RESUMO

A high renal oxygen (O2 ) need is primarily associated with the renal tubular O2 consumption (VO2 ) necessary for a high rate of sodium (Na+ ) transport. Limited O2 availability leads to increased levels of adenosine, which regulates the kidney via activation of both A1 and A2A adenosine receptors (A1R and A2AR, respectively). The relative contributions of A1R and A2AR to the regulation of renal Na+ transport and VO2 have not been determined. We demonstrated that A1R activation has a dose-dependent biphasic effect on both renal Na+ /H+ exchanger-3 (NHE3), a major player in Na+ transport, and VO2 . Here, we report concentration-dependent effects of adenosine: less than 5 × 10-7 M adenosine-stimulated NHE3 activity; between 5 × 10-7 M and 10-5 M adenosine-inhibited NHE3 activity; and greater than 10-5 M adenosine reversed the change in NHE3 activity (returned to baseline). A1R activation mediated the activation and inhibition of NHE3 activity, whereas 10-4 M adenosine had no effect on the NHE3 activity due to A2AR activation. The following occurred when A1R and A2AR were activated: (a) Blockade of the A2AR receptor restored the NHE3 inhibition mediated by A1R activation, (b) the NHE-dependent effect on VO2 mediated by A1R activation became NHE independent, and (c) A2AR bound to A1R. In summary, A1R affects VO2 via NHE-dependent mechanisms, whereas A2AR acts via NHE-independent mechanisms. When both A1R and A2AR are activated, the A2AR effect on NHE3 and VO2 predominates, possibly via an A1R-A2AR protein interaction. A2AR-A1R heterodimerization is proposed as the molecular mechanism enabling the NHE-independent control of renal VO2 .


Assuntos
Rim/metabolismo , Consumo de Oxigênio , Receptor A1 de Adenosina/metabolismo , Receptor A2A de Adenosina/metabolismo , Sódio/metabolismo , Adenosina/farmacologia , Animais , Transporte Biológico/efeitos dos fármacos , Linhagem Celular , Humanos , Masculino , Modelos Biológicos , Gambás , Consumo de Oxigênio/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Ratos Sprague-Dawley , Trocador 3 de Sódio-Hidrogênio/metabolismo
2.
J Cell Sci ; 129(6): 1128-40, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26823603

RESUMO

The most common mutation of the cystic fibrosis transmembrane regulator (CFTR) gene, F508del, produces a misfolded protein resulting in its defective trafficking to the cell surface and an impaired chloride secretion. Pharmacological treatments partially rescue F508del CFTR activity either directly by interacting with the mutant protein and/or indirectly by altering the cellular protein homeostasis. Here, we show that the phosphorylation of ezrin together with its binding to phosphatidylinositol-4,5-bisphosphate (PIP2) tethers the F508del CFTR to the actin cytoskeleton, stabilizing it on the apical membrane and rescuing the sub-membrane compartmentalization of cAMP and activated PKA. Both the small molecules trimethylangelicin (TMA) and VX-809, which act as 'correctors' for F508del CFTR by rescuing F508del-CFTR-dependent chloride secretion, also restore the apical expression of phosphorylated ezrin and actin organization and increase cAMP and activated PKA submembrane compartmentalization in both primary and secondary cystic fibrosis airway cells. Latrunculin B treatment or expression of the inactive ezrin mutant T567A reverse the TMA and VX-809-induced effects highlighting the role of corrector-dependent ezrin activation and actin re-organization in creating the conditions to generate a sub-cortical cAMP pool of adequate amplitude to activate the F508del-CFTR-dependent chloride secretion.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/metabolismo , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Actinas/metabolismo , Animais , Cloretos/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/genética , Fibrose Cística/enzimologia , Fibrose Cística/genética , Proteínas do Citoesqueleto/genética , Citoesqueleto/genética , Humanos , Fosforilação , Ratos , Deleção de Sequência , Transdução de Sinais
3.
J Cell Physiol ; 230(12): 3093-104, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26010290

RESUMO

The high requirement of O2 in the renal proximal tubule stems from a high rate of Na(+) transport. Adenosine A1 receptor (A1R) activation regulates Na(+) transport in this nephron segment. Thus, the effect of the acute activation and the mechanisms of A1R on the rate of O2 consumption were evaluated. The A1R-antagonist, 8-cyclopentyl-1,3-dipropylxanthine (CPX) and adenosine deaminase (ADA), which metabolize endogenous adenosine, reduced O2 consumption (40-50%). Replacing Na(+) in the buffer reversed the ADA- or CPX-mediated reduction of O2 consumption. Blocking the Na/H-exchanger activity, which decreases O2 usage per se, did not enhance the ADA- or CPX-induced inhibition of O2 consumption. These data indicate that endogenous adenosine increases O2 usage via the activation of Na(+) transport. In the presence of endogenous adenosine, A1R was further activated by the A1R-agonist N(6)-cyclopentyladenosine (CPA); CPA inhibited O2 usage (30%) and this effect also depended on Na(+) transport. Moreover, a low concentration of CPA activated O2 usage in tissue pretreated with ADA, whereas a high concentration of CPA inhibited O2 usage; both effects depended on Na(+). Protein kinase C signaling mediated the inhibitory effect of A1R, while adenylyl cyclase mediated its stimulatory effect on O2 consumption. In summary, increasing the local concentrations of adenosine can either activate or inhibit O2 consumption via A1R, and this mechanism depends on Na(+) transport. The inhibition of O2 usage by A1R activation might restore the compromised balance between energy supply and demand under pathophysiological conditions, such as renal ischemia, which results in high adenosine production.


Assuntos
Adenosina/metabolismo , Córtex Renal/metabolismo , Túbulos Renais Proximais/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Receptor A1 de Adenosina/metabolismo , Agonistas do Receptor A1 de Adenosina/farmacologia , Adenilil Ciclases/metabolismo , Animais , Técnicas In Vitro , Córtex Renal/efeitos dos fármacos , Túbulos Renais Proximais/efeitos dos fármacos , Cinética , Masculino , Proteína Quinase C/metabolismo , Antagonistas de Receptores Purinérgicos P1/farmacologia , Ratos Sprague-Dawley , Receptor A1 de Adenosina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Trocadores de Sódio-Hidrogênio/metabolismo
4.
FASEB J ; 27(11): 4646-58, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23934281

RESUMO

Epithelial Na(+)/H(+) exchanger-3 (NHE3) transport is fundamental for renal and intestinal sodium reabsorption. Cytoplasmic protons are thought to serve as allosteric modifiers of the exchanger and to trigger its transport through protein conformational change. This effect presupposes an intracellular pH (pHi) dependence of NHE3 activity, although the biophysical and molecular basis of NHE3 pHi sensitivity have not been defined. NHE3, when complexed with the calcineurin homologous protein-1 (CHP1), had a shift in pHi sensitivity (0.4 units) toward the acidic side in comparison with NHE3 alone, as measured by oscillating pH electrodes combined with whole-cell patch clamping. Indeed, CHP1 interaction with NHE3 inhibited NHE3 transport in a pHi -dependent manner. CHP1 binding to NHE3 also affected its acute regulation. Intracellular perfusion of peptide from the CHP1 binding region (or pHi modification to reduce the CHP1 amount bound to NHE3) was permissive and cooperative for dopamine inhibition of NHE3 but reversed that of adenosine. Thus, CHP1 interaction with NHE3 apparently establishes the exchanger set point for pHi, and modification in this set point is effective in the hormonal stimuli-mediated regulation of NHE3. CHP1 may serve as a regulatory cofactor for NHE3 conformational change, dependent on intracellular protonation.


Assuntos
Espaço Intracelular/metabolismo , Prótons , Trocadores de Sódio-Hidrogênio/metabolismo , Potenciais de Ação , Adenosina/farmacologia , Animais , Sítios de Ligação , Células CHO , Proteínas de Ligação ao Cálcio/metabolismo , Cricetinae , Cricetulus , Dopamina/farmacologia , Concentração de Íons de Hidrogênio , Ligação Proteica , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Trocadores de Sódio-Hidrogênio/química
5.
Am J Physiol Renal Physiol ; 304(9): F1169-80, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23427139

RESUMO

The intrarenal autocrine/paracrine dopamine (DA) system contributes to natriuresis in response to both acute and chronic Na(+) loads. While the acute DA effect is well described, how DA induces natriuresis chronically is not known. We used an animal and a cell culture model to study the chronic effect of DA on a principal renal Na(+) transporter, Na(+)/H(+) exchanger-3 (NHE3). Intraperitoneal injection of Gludopa in rats for 2 days elevated DA excretion and decreased total renal cortical and apical brush-border NHE3 antigen. Chronic treatment of an opossum renal proximal cell line with DA decreased NHE3 activity, cell surface and total cellular NHE3 antigen, but not NHE3 transcript. The decrease in NHE3 antigen was dose and time dependent with maximal inhibition at 16-24 h and half maximal effect at 3 × 10(-7) M. This is in contradistinction to the acute effect of DA on NHE3 (half maximal at 2 × 10(-6) M), which was not associated with changes in total cellular NHE3 protein. The DA-induced decrease in total NHE3 protein was associated with decrease in NHE3 translation and mediated by cis-sequences in the NHE3 5'-untranslated region. DA also decreased cell surface and total cellular NHE3 protein half-life. The DA-induced decrease in total cellular NHE3 was partially blocked by proteasome inhibition but not by lysosome inhibition, and DA increased ubiquitylation of total and surface NHE3. In summary, chronic DA inhibits NHE3 with mechanisms distinct from its acute action and involves decreased NHE3 translation and increased NHE3 degradation, which are novel mechanisms for NHE3 regulation.


Assuntos
Dopamina/farmacologia , Rim/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Trocadores de Sódio-Hidrogênio/genética , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Di-Hidroxifenilalanina/análogos & derivados , Di-Hidroxifenilalanina/farmacologia , Dopaminérgicos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Meia-Vida , Técnicas In Vitro , Rim/citologia , Rim/efeitos dos fármacos , Modelos Animais , Gambás , Ratos , Ratos Sprague-Dawley , Trocador 3 de Sódio-Hidrogênio , Ubiquitinação/efeitos dos fármacos
6.
Am J Physiol Cell Physiol ; 302(11): C1569-87, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22460714

RESUMO

The Na(+)/H(+) exchanger-3 (NHE3) belongs to the mammalian NHE protein family and catalyzes the electro-neutral exchange of extracellular sodium for intracellular proton across cellular membranes. Its transport function is of essential importance for the maintenance of the body's salt and water homeostasis as well as acid-base balance. Indeed, NHE3 activity is finely regulated by a variety of stimuli, both acutely and chronically, and its transport function is fundamental for a multiplicity of severe and world-wide infection-pathological conditions. This review aims to provide a concise overview of NHE3 physiology and discusses the role of NHE3 in clinical conditions of prominent importance, specifically in hypertension, diabetic nephropathy, heart failure, acute kidney injury, and diarrhea. Study of NHE3 function in models of these diseases has contributed to the deciphering of mechanisms that control the delicate ion balance disrupted in these disorders. The majority of the findings indicate that NHE3 transport function is activated before the onset of hypertension and inhibited thereafter; NHE3 transport function is also upregulated in diabetic nephropathy and heart failure, while it is reported to be downregulated in acute kidney injury and in diarrhea. The molecular mechanisms activated during these pathological conditions to regulate NHE3 transport function are examined with the aim of linking NHE3 dysfunction to the analyzed clinical disorders.


Assuntos
Diarreia/metabolismo , Insuficiência Cardíaca/metabolismo , Hipertensão/metabolismo , Nefropatias/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Sódio/metabolismo , Equilíbrio Ácido-Base , Injúria Renal Aguda/patologia , Animais , Transporte Biológico Ativo/fisiologia , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Diarreia/patologia , Insuficiência Cardíaca/patologia , Humanos , Hipertensão/patologia , Transporte de Íons/fisiologia , Nefropatias/patologia , Trocador 3 de Sódio-Hidrogênio
7.
Am J Physiol Renal Physiol ; 303(2): F165-79, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22189947

RESUMO

The calcineurin homologous protein (CHP) belongs to an evolutionarily conserved Ca(2+)-binding protein subfamily. The CHP subfamily is composed of CHP1, CHP2, and CHP3, which in vertebrates share significant homology at the protein level with each other and between other Ca(2+)-binding proteins. The CHP structure consists of two globular domains containing from one to four EF-hand structural motifs (calcium-binding regions composed of two helixes, E and F, joined by a loop), the myristoylation, and nuclear export signals. These structural features are essential for the function of the three members of the CHP subfamily. Indeed, CHP1-CHP3 have multiple and diverse essential functions, ranging from the regulation of the plasma membrane Na(+)/H(+) exchanger protein function, to carrier vesicle trafficking and gene transcription. The diverse functions attributed to the CHP subfamily rendered an understanding of its action highly complex and often controversial. This review provides a comprehensive and organized examination of the properties and physiological roles of the CHP subfamily with a view to revealing a link between CHP diverse functions.


Assuntos
Proteínas de Ligação ao Cálcio/fisiologia , Sequência de Aminoácidos , Animais , Proteínas de Ligação ao Cálcio/análise , Proteínas de Ligação ao Cálcio/química , Humanos , Dados de Sequência Molecular , Transporte Proteico/fisiologia , Trocadores de Sódio-Hidrogênio/fisiologia , Transcrição Gênica/fisiologia
8.
Biochem Pharmacol ; 190: 114617, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34023293

RESUMO

Clinical reports indicate a bidirectional relationship between mental illness and chronic systemic diseases. However, brain mechanisms linking chronic stress and development of mood disorders to accompanying peripheral organ dysfunction are still not well characterized in animal models. In the current study, we investigated whether activation of hippocampal mitogen-activated protein kinase phosphatase-1 (MKP-1), a key factor in depression pathophysiology, also acts as a mediator of systemic effects of stress. First, we demonstrated that treatment with the glucocorticoid receptor (GR) agonist dexamethasone or acute restraint stress (ARS) significantly increased Mkp-1 mRNA levels within the rat hippocampus. Conversely, administration of the GR antagonist mifepristone 30 min before ARS produced a partial blockade of Mkp-1 upregulation, suggesting that stress activates MKP-1, at least in part, through upstream GR signaling. Chronic corticosterone (CORT) administration evoked comparable increases in hippocampal MKP-1 protein levels and produced a robust increase in behavioral emotionality. In addition to behavioral deficits, chronic CORT treatment also produced systemic pathophysiological effects. Elevated levels of renal inflammation protein markers (NGAL and IL18) were observed suggesting tissue damage and early kidney impairment. In a rescue experiment, the effects of CORT on development of depressive-like behaviors and increased NGAL and IL18 protein levels in the kidney were blocked by CRISPR-mediated knockdown of hippocampal Mkp-1 prior to CORT exposure. In sum, these findings further demonstrate that MKP-1 is necessary for development of enhanced behavioral emotionality, while also suggesting a role in stress mechanisms linking brain dysfunction and systemic illness such as kidney disease.


Assuntos
Corticosterona/administração & dosagem , Corticosterona/efeitos adversos , Fosfatase 1 de Especificidade Dupla/biossíntese , Hipocampo/metabolismo , Estresse Psicológico/induzido quimicamente , Estresse Psicológico/metabolismo , Animais , Linhagem Celular Tumoral , Dexametasona/administração & dosagem , Dexametasona/efeitos adversos , Esquema de Medicação , Glucocorticoides/administração & dosagem , Glucocorticoides/efeitos adversos , Hipocampo/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley
9.
Am J Physiol Renal Physiol ; 299(6): F1496-506, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20926631

RESUMO

Serum and glucocorticoid-regulated kinase 2 (sgk2) is 80% identical to the kinase domain of sgk1, an important mediator of mineralocorticoid-regulated sodium (Na(+)) transport in the distal nephron of the kidney. The expression pattern and role in renal function of sgk2 are virtually uncharacterized. In situ hybridization and immunohistochemistry of rodent kidney coupled with real-time RT-PCR of microdissected rat kidney tubules showed robust sgk2 expression in the proximal straight tubule and thick ascending limb of the loop of Henle. Sgk2 expression was minimal in distal tubule cells with aquaporin-2 immunostaining but significant in proximal tubule cells with Na(+)/H(+) exchanger 3 (NHE3) immunostaining. To ascertain whether mineralocorticoids regulate expression of sgk2 in a manner similar to sgk1, we examined sgk2 mRNA expression in the kidneys of adrenalectomized rats treated with physiological doses of aldosterone together with the glucocorticoid receptor antagonist RU486. Northern blot analysis and in situ hybridization showed that, unlike sgk1, sgk2 expression in the kidney was not altered by aldosterone treatment. Based on the observation that sgk2 is expressed in proximal tubule cells that also express NHE3, we asked whether sgk2 regulates NHE3 activity. We heterologously expressed sgk2 in opossum kidney (OKP) cells and measured Na(+)/H(+) exchange activity by Na(+)-dependent cell pH recovery. Constitutively active sgk2, but not sgk1, stimulated Na(+)/H(+) exchange activity by >30%. Moreover, the sgk2-mediated increase in Na(+)/H(+) exchange activity correlated with an increase in cell surface expression of NHE3. Together, these results suggest that the pattern of expression, regulation, and role of sgk2 within the mammalian kidney are distinct from sgk1 and that sgk2 may play a previously unrecognized role in the control of transtubular Na(+) transport through NHE3 in the proximal tubule.


Assuntos
Proteínas Imediatamente Precoces/biossíntese , Proteínas Serina-Treonina Quinases/biossíntese , Trocadores de Sódio-Hidrogênio/metabolismo , Aldosterona/farmacologia , Animais , Linhagem Celular , Regulação da Expressão Gênica , Proteínas Imediatamente Precoces/fisiologia , Hibridização In Situ , Rim/efeitos dos fármacos , Rim/metabolismo , Túbulos Renais/metabolismo , Masculino , Camundongos , Gambás , Proteínas Serina-Treonina Quinases/fisiologia , Ratos , Ratos Sprague-Dawley , Trocador 3 de Sódio-Hidrogênio
10.
Am J Physiol Renal Physiol ; 298(5): F1205-13, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20181665

RESUMO

Nephrogenic dopamine is a potent natriuretic paracrine/autocrine hormone that is central for mammalian sodium homeostasis. In the renal proximal tubule, dopamine induces natriuresis partly via inhibition of the sodium/proton exchanger NHE3. The signal transduction pathways and mechanisms by which dopamine inhibits NHE3 are complex and incompletely understood. This manuscript describes the role of the serine/threonine protein phosphatase 2A (PP2A) in the regulation of NHE3 by dopamine. The PP2A regulatory subunit B56δ (coded by the Ppp2r5d gene) directly associates with more than one region of the carboxy-terminal hydrophilic putative cytoplasmic domain of NHE3 (NHE3-cyto), as demonstrated by yeast-two-hybrid, coimmunoprecipitation, blot overlay, and in vitro pull-down assays. Phosphorylated NHE3-cyto is a substrate for purified PP2A in an in vitro dephosphorylation reaction. In cultured renal cells, inhibition of PP2A by either okadaic acid or by overexpression of the simian virus 40 (SV40) small T antigen blocks the ability of dopamine to inhibit NHE3 activity and to reduce surface NHE3 protein. Dopamine-induced NHE3 redistribution is also blocked by okadaic acid ex vivo in rat kidney cortical slices. These studies demonstrate that PP2A is an integral and critical participant in the signal transduction pathway between dopamine receptor activation and NHE3 inhibition.


Assuntos
Dopamina/farmacologia , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Proteína Fosfatase 2/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Antígenos Transformantes de Poliomavirus/farmacologia , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Técnicas In Vitro , Túbulos Renais Proximais/citologia , Modelos Animais , Ácido Okadáico/farmacologia , Gambás , Proteína Fosfatase 2/antagonistas & inibidores , Proteína Fosfatase 2/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Trocador 3 de Sódio-Hidrogênio
11.
J Am Soc Nephrol ; 20(8): 1776-86, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19556366

RESUMO

The Na(+)/H(+)-exchanger 3 (NHE3) is essential for regulation of Na(+) transport in the renal and intestinal epithelium. Although changes in cell surface abundance control NHE3 function, the molecular signals that regulate NHE3 surface expression are not well defined. We found that overexpression of the calcineurin homologous protein-1 (CHP1) in opossum kidney cells increased NHE3 transport activity, surface protein abundance, and ezrin phosphorylation. CHP1 knockdown by small interfering RNA had the opposite effects. Overexpression of wild-type ezrin increased both NHE3 transport activity and surface protein abundance, confirming that NHE3 is downstream of ezrin. Expression of a pseudophosphorylated ezrin enhanced these effects, whereas expression of an ezrin variant that could not be phosphorylated prevented the downstream effects on NHE3. Furthermore, CHP1 knockdown reversed the activation of NHE3 by wild-type ezrin but not by the pseudophosphorylated ezrin. Taken together, these results demonstrate that CHP1 increases NHE3 abundance and constitutive function in a manner dependent on ezrin phosphorylation.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Proteínas do Citoesqueleto/metabolismo , Fosfotransferases/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Linhagem Celular , Humanos , Gambás , Fosforilação , Transdução de Sinais , Trocador 3 de Sódio-Hidrogênio , Treonina/metabolismo , Regulação para Cima
13.
J Cell Physiol ; 216(1): 221-33, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18286509

RESUMO

The renal function of the A(3) adenosine receptor (A3AR) is poorly characterized. In this study, we report that the A3AR-selective agonist, 1-[2-chloro-6-[[(3-iodophenyl)methyl]amino]-9H-purine-9-yl]-1-deoxy-N-methyl-b-D-ribofuranuronamide (2-Cl-IBMECA) regulates the Na+/H+ exchanger-3 (NHE3) in a dose- and time-dependent fashion. In opossum kidney (OK) cells, 2-Cl-IBMECA at high (10(-6) M) and low (10(-8) M) dose inhibits NHE3 by a multiphasic time course with an acute phase of NHE3 inhibition from 15 min to 1 h, followed by a chronic phase of NHE3 inhibition from 24 to 48 h. Pre-incubation with either the selective A3AR-antagonist MRS1523 (10(-7) M) or the protein kinase C inhibitor, Calphostin C (10(-8) M) completely blocked 10(-6) M 2-Cl-IBMECA-induced acute (15 min) and chronic (24 h) phases of NHE3 inhibition. In contrast, the acute inhibitory phase (15 min) of 10(-8) M 2-Cl-IBMECA was completely prevented only when Calphostin C (10(-8) M) was added in conjunction with the protein kinase A inhibitor, H89 (10(-7) M). Acute (15 or 30 min depending on the A3AR-agonist concentration) A3AR-dependent inhibition of NHE3 activity was accompanied by decrease in cell surface NHE3 protein with no change in total NHE3 antigen. Chronic (24 h) A3AR-mediated down-regulation of NHE3 was associated with reduction of surface NHE3, decreased total NHE3 protein (70%) and a paradoxical rise of NHE3 RNA (40%). In summary, these results indicate that A3AR directly regulates NHE3 at multiple levels in a complex pattern. A3AR-dependent short- and long-term inhibition of NHE3 may be a fundamental mechanism of net sodium and fluid balance.


Assuntos
Células Epiteliais/metabolismo , Rim/citologia , Receptor A3 de Adenosina/metabolismo , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Trocadores de Sódio-Hidrogênio/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/farmacologia , Agonistas do Receptor A3 de Adenosina , Animais , Anti-Inflamatórios/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/metabolismo , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Humanos , Hidrocortisona/metabolismo , Hidrogênio/metabolismo , Concentração de Íons de Hidrogênio , Rim/metabolismo , Naftalenos/metabolismo , Gambás , Proteína Quinase C/metabolismo , Receptor A3 de Adenosina/genética , Transdução de Sinais/fisiologia , Sódio/metabolismo , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/genética
14.
J Vis Exp ; (132)2018 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-29443070

RESUMO

The transport of ions through cell membranes ensures the fine control of ion content within and outside the cell that is indispensable for cell survival. These transport mechanisms are mediated by the activities of specialized transporter proteins. Specifically,pH dynamics are finely controlled by plasma membrane proton (H+) extrusion systems, such as the Na+/H+ exchanger (NHE) protein family. Despite extensive efforts to study the mechanisms underlying NHE regulation, our current understanding of the biophysical and molecular properties of the NHE family is inadequate because of the limited availability of methods to effectively measure NHE activity. In this manuscript, we used H+-selective electrodes during whole-cell patch clamping recording to measure NHE-induced H+ flux. We proposed this approach to overcome some limitations of typically used methods to measure NHE activity, such as radioactive uptake and fluorescent membrane permeants. Measurement of NHE activity using the described method enables high sensitivity and time resolution and more efficient control of intracellular H+ concentrations. H+-selective electrodes are based on the fact that transporter activity creates an ion gradient in close proximity to the cell membrane. An H+-selective electrode moving up to and away from the cell membrane in a repetitive, oscillatory fashion records a voltage difference that is dependent on H+ flux. While H+-selective electrodes are used to detect H+ flux moving out of the cell, the patch clamp method in the whole-cell configuration is used to control the intracellular ion composition. Moreover, application of the giant patch clamp technique allows modification of the intracellular composition of not only ions but also lipids. The transporter activity of NHE isoform 3 (NHE3) was measured using this technical approach to study the molecular basis of NHE3 regulation by phosphoinositides.


Assuntos
Eletrofisiologia/métodos , Membrana Celular/metabolismo , Proteínas de Membrana Transportadoras/metabolismo
15.
Nutrients ; 10(11)2018 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-30380720

RESUMO

Copper, which can potentially be a highly toxic agent, is an essential nutrient due to its role as a cofactor for cuproenzymes and its participation in signaling pathways. In mammals, the liver is a central organ that controls copper turnover throughout the body, including copper absorption, distribution, and excretion. In ontogenesis, there are two types of copper metabolism, embryonic and adult, which maintain the balance of copper in each of these periods of life, respectively. In the liver cells, these types of metabolism are characterized by the specific expression patterns and activity levels of the genes encoding ceruloplasmin, which is the main extracellular ferroxidase and copper transporter, and the proteins mediating ceruloplasmin metalation. In newborns, the molecular genetic mechanisms responsible for copper homeostasis and the ontogenetic switch from embryonic to adult copper metabolism are highly adapted to milk ceruloplasmin as a dietary source of copper. In the mammary gland cells, the level of ceruloplasmin gene expression and the alternative splicing of its pre-mRNA govern the amount of ceruloplasmin in the milk, and thus, the amount of copper absorbed by a newborn is controlled. In newborns, the absorption, distribution, and accumulation of copper are adapted to milk ceruloplasmin. If newborns are not breast-fed in the early stages of postnatal development, they do not have this natural control ensuring alimentary copper balance in the body. Although there is still much to be learned about the neonatal consequences of having an imbalance of copper in the mother/newborn system, the time to pay attention to this problem has arrived because the neonatal misbalance of copper may provoke the development of copper-related disorders.


Assuntos
Ceruloplasmina/metabolismo , Cobre/metabolismo , Leite Humano/química , Adulto , Feminino , Humanos , Recém-Nascido , Lactação/metabolismo , Fígado/metabolismo , Masculino
18.
PLoS One ; 10(6): e0129306, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26042733

RESUMO

Na+/H+ exchanger-3 (NHE3) plays an essential role in maintaining sodium and fluid homeostasis in the intestine and kidney epithelium. Thus, NHE3 is highly regulated and its function depends on binding to multiple regulatory proteins. Ezrin complexed with NHE3 affects its activity via not well-defined mechanisms. This study investigates mechanisms by which ezrin regulates NHE3 activity in epithelial Opossum Kidney cells. Ezrin is activated sequentially by phosphatidylinositol-4,5-bisphosphate (PIP2) binding and phosphorylation of threonine 567. Expression of ezrin lacking PIP2 binding sites inhibited NHE3 activity (-40%) indicating that ezrin binding to PIP2 is required for preserving NHE3 activity. Expression of a phosphomimetic ezrin mutated at the PIP2 binding region was sufficient not only to reverse NHE3 activity to control levels but also to increase its activity (+80%) similar to that of the expression of ezrin carrying the phosphomimetic mutation alone. Calcineurin Homologous Protein-1 (CHP1) is part, with ezrin, of the NHE3 regulatory complex. CHP1-mediated activation of NHE3 activity was blocked by expression of an ezrin variant that could not be phosphorylated but not by an ezrin variant unable to bind PIP2. Thus, for NHE3 activity under baseline conditions not only ezrin phosphorylation, but also ezrin spatial-temporal targeting on the plasma membrane via PIP2 binding is required; however, phosphorylation of ezrin appears to overcome the control of NHE3 transport. CHP1 action on NHE3 activity is not contingent on ezrin binding to PIP2 but rather on ezrin phosphorylation. These findings are important in understanding the interrelation and dynamics of a CHP1-ezrin-NHE3 regulatory complex.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Linhagem Celular , Humanos , Proteínas Mutantes/metabolismo , Gambás , Fosforilação , Fosfotreonina/metabolismo , Ligação Proteica , Transporte Proteico , Frações Subcelulares/metabolismo
19.
J Nephrol ; 15 Suppl 5: S3-21, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12027219

RESUMO

Na+/H+ exchangers (NHEs) extrude protons from, and take up sodium ions into cells. Six isoforms, NHE-1 - NHE-6, have been cloned. NHE proteins are composed of an N-terminal domain, which most likely crosses the cell membrane 12 times and constitutes the cation exchange machinery, and a C-terminal tail, which modulates the exchanger by interacting with protein kinases and regulatory factors. The "house-keeping" NHE-1 is located at the basolateral membrane of most renal tubule cells; NHE-2 is located apically in selected nephron segments. As suggested from data with NHE-1 and NHE-2 deficient mice, both isoforms play a minor role in renal salt and water handling. NHE-3 is located at the apical membrane of proximal tubule and thick ascending limb cells, is involved in Na+ absorption, and is responsible for the majority of bicarbonate absorption. NHE-3 is modulated by the NHE regulating factor, which interacts with further proteins, protein kinases, and the cytoskeleton. Downregulation of NHE-3 by parathyroid hormone, dopamine, and by an increase in blood pressure leads to saluresis/diuresis. The failure of dopamine to downregulate NHE-3 may cause hypertension through renal salt and water retention. NHE-3 knockouts are hypotonic and can not survive on low salt diet. In chronic acidosis, NHE-3 is upregulated possibly through increased local endothelin production. NHE4 has been found mostly in renal medulla. The precise function of this isoform, which is activated by hypertonicity and can perform K+/H+ exchange, is not clear. The segmental location and function of NHE-5 and NHE-6 in the kidney are unknown at present.


Assuntos
Rim/fisiologia , Família Multigênica/fisiologia , Trocadores de Sódio-Hidrogênio/genética , Absorção , Animais , Bicarbonatos/metabolismo , Humanos , Distribuição Tecidual
20.
PLoS One ; 8(9): e75113, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24086451

RESUMO

Extracellular matrix (ECM) degradation is a critical process in tumor cell invasion and requires matrix degrading protrusions called invadopodia. The Na(+)/H(+) exchanger (NHE1) has recently been shown to be fundamental in the regulation of invadopodia actin cytoskeleton dynamics and activity. However, the structural link between the invadopodia cytoskeleton and NHE1 is still unknown. A candidate could be ezrin, a linker between the NHE1 and the actin cytoskeleton known to play a pivotal role in invasion and metastasis. However, the mechanistic basis for its role remains unknown. Here, we demonstrate that ezrin phosphorylated at T567 is highly overexpressed in the membrane of human breast tumors and positively associated with invasive growth and HER2 overexpression. Further, in the metastatic cell line, MDA-MB-231, p-ezrin was almost exclusively expressed in invadopodia lipid rafts where it co-localized in a functional complex with NHE1, EGFR, ß1-integrin and phosphorylated-NHERF1. Manipulation by mutation of ezrins T567 phosphorylation state and/or PIP2 binding capacity or of NHE1s binding to ezrin or PIP2 demonstrated that p-ezrin expression and binding to PIP2 are required for invadopodia-mediated ECM degradation and invasion and identified NHE1 as the membrane protein that p-ezrin regulates to induce invadopodia formation and activity.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas do Citoesqueleto/metabolismo , Integrina beta1/metabolismo , Microdomínios da Membrana/metabolismo , Invasividade Neoplásica/fisiopatologia , Pseudópodes/fisiologia , Transdução de Sinais/fisiologia , Análise de Variância , Primers do DNA/genética , Matriz Extracelular/metabolismo , Feminino , Imunofluorescência , Regulação da Expressão Gênica/genética , Humanos , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Imunoprecipitação , Itália , Fosforilação , Receptor ErbB-2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA