Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Cell ; 184(2): 534-544.e11, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33373586

RESUMO

Determination of what is the specificity of subunits composing a protein complex is essential when studying gene variants on human pathophysiology. The pore-forming α-subunit KCNQ1, which belongs to the voltage-gated ion channel superfamily, associates to its ß-auxiliary subunit KCNE1 to generate the slow cardiac potassium IKs current, whose dysfunction leads to cardiac arrhythmia. Using pharmacology, gene invalidation, and single-molecule fluorescence assays, we found that KCNE1 fulfils all criteria of a bona fide auxiliary subunit of the TMEM16A chloride channel, which belongs to the anoctamin superfamily. Strikingly, assembly with KCNE1 switches TMEM16A from a calcium-dependent to a voltage-dependent ion channel. Importantly, clinically relevant inherited mutations within the TMEM16A-regulating domain of KCNE1 abolish the TMEM16A modulation, suggesting that the TMEM16A-KCNE1 current may contribute to inherited pathologies. Altogether, these findings challenge the dogma of the specificity of auxiliary subunits regarding protein complexes and questions ion channel classification.


Assuntos
Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Subunidades Proteicas/metabolismo , Animais , Anoctamina-1/metabolismo , Cálcio/metabolismo , Canais de Cloreto/metabolismo , Células HEK293 , Humanos , Túbulos Renais Proximais/metabolismo , Camundongos , Proteínas Mutantes/metabolismo , Peptídeos/metabolismo , Polimorfismo Genético , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Ligação Proteica , Domínios Proteicos , Sistema Renina-Angiotensina
2.
Respir Physiol Neurobiol ; 245: 13-28, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-27838333

RESUMO

TASK-1 potassium channels have been implicated in central and peripheral chemoreception; however, the precise contribution of TASK-1 for the control of respiration is still under debate. Here, we investigated the respiration of unrestrained adult and neonatal TASK-1 knockout mice (TASK-1-/-) using a plethysmographic device. Respiration in adult female TASK-1-/- mice under control (21% O2), hypoxia and hypercapnia was unaffected. Under acute hypoxia male TASK-1-/- mice exhibited a reduced increase of the respiratory frequency (fR) compared to wildtypes. However, the tidal volume (VT) of male TASK-1-/- mice was strongly enhanced. The volatile anesthetic isoflurane induced in male TASK-1-/- and male wild type mice (TASK-1+/+) a similar respiratory depression. Neonatal TASK-1-/- mice demonstrated a 30-40% decrease of the minute volume, caused by a reduction of the fR under control condition (21% O2). Under hypoxia, neonatal TASK-1-/- mice more frequently stopped breathing (apnea>3s) suggesting an increased hypoxia-sensitivity. As reported before, this increased hypoxia sensitivity had no influence on the survival rate of neonatal TASK-1-/- mice. In adult and neonatal mice, TASK-1 gene deletion induced a significant prolongation of the relaxation time (RT), which is a parameter for expiration kinetics. Additionally, screening for mutations in the human TASK-1 gene in 155 cases of sudden infant death syndrome (SIDS) was inconclusive. In conclusion, these data are suggestive for an increased hypoxia-sensitivity of neonatal TASK-1-/- mice, however, without causing an increase in neonatal lethality. In adult female TASK-1-/- mice respiration was unaffected, whereas adult male TASK-1-/- mice showed a modified breathing pattern. These results are suggestive for sex-specific mechanisms for compensating the inactivation of TASK-1 in mice.


Assuntos
Proteínas do Tecido Nervoso/deficiência , Canais de Potássio de Domínios Poros em Tandem/deficiência , Respiração , Caracteres Sexuais , Envelhecimento/metabolismo , Anestésicos Inalatórios/farmacologia , Animais , Animais Recém-Nascidos , Estudos de Coortes , Feminino , Humanos , Hipercapnia/fisiopatologia , Lactente , Isoflurano/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Pletismografia Total , Canais de Potássio de Domínios Poros em Tandem/genética , Respiração/efeitos dos fármacos , Morte Súbita do Lactente/genética , Volume de Ventilação Pulmonar/fisiologia
3.
Trends Endocrinol Metab ; 27(3): 123-131, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26803728

RESUMO

Primary aldosteronism (PA) is caused by excessive production of aldosterone by the adrenal cortex and is determined by a benign aldosterone-producing adenoma (APA) in a significant proportion of cases. Local mechanisms, as opposed to circulatory ones, that control aldosterone production in the adrenal cortex are particularly relevant in the physiopathological setting and in the pathogenesis of PA. A breakthrough in our understanding of the pathogenetic mechanisms in APA has been the identification of somatic mutations in genes controlling membrane potential and intracellular calcium concentrations. However, recent data show that the processes of nodule formation and aldosterone hypersecretion can be dissociated in pathological adrenals and suggest a model envisaging different molecular events for the pathogenesis of APA.


Assuntos
Córtex Suprarrenal/metabolismo , Aldosterona/metabolismo , Regulação da Expressão Gênica , Hiperaldosteronismo/fisiopatologia , Modelos Biológicos , Córtex Suprarrenal/fisiologia , Córtex Suprarrenal/fisiopatologia , Neoplasias do Córtex Suprarrenal/metabolismo , Neoplasias do Córtex Suprarrenal/fisiopatologia , Adenoma Adrenocortical/metabolismo , Adenoma Adrenocortical/fisiopatologia , Animais , Regulação Neoplásica da Expressão Gênica , Humanos , Hiperaldosteronismo/etiologia , Hiperaldosteronismo/metabolismo
4.
FASEB J ; 30(2): 909-22, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26527067

RESUMO

Brown adipose tissue (BAT) is essential for adaptive thermogenesis and dissipation of caloric excess through the activity of uncoupling protein (UCP)-1. BAT in humans is of great interest for the treatment of obesity and related diseases. In this study, the expression of Twik-related acid-sensitive K(+) channel (TASK)-1 [a pH-sensitive potassium channel encoded by the potassium channel, 2-pore domain, subfamily K, member 3 (Kcnk3) gene] correlated highly with Ucp1 expression in obese and cold-exposed mice. In addition, Task1-null mice, compared with their controls, became overweight, mainly because of an increase in white adipose tissue mass and BAT whitening. Task1(-/-)-mouse-derived brown adipocytes, compared with wild-type mouse-derived brown adipocytes, displayed an impaired ß3-adrenergic receptor response that was characterized by a decrease in oxygen consumption, Ucp1 expression, and lipolysis. This phenotype was thought to be caused by an exacerbation of mineralocorticoid receptor (MR) signaling, given that it was mimicked by corticoids and reversed by an MR inhibitor. We concluded that the K(+) channel TASK1 controls the thermogenic activity in brown adipocytes through modulation of ß-adrenergic receptor signaling.


Assuntos
Adipócitos Marrons/metabolismo , Tecido Adiposo Marrom/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Receptores de Mineralocorticoides/metabolismo , Transdução de Sinais/fisiologia , Adipócitos Marrons/citologia , Tecido Adiposo Marrom/citologia , Animais , Feminino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Consumo de Oxigênio/fisiologia , Canais de Potássio de Domínios Poros em Tandem/genética , Receptores de Mineralocorticoides/genética , Termogênese/fisiologia
5.
Nat Commun ; 6: 8780, 2015 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-26549439

RESUMO

In the cochlea, K(+) is essential for mechano-electrical transduction. Here, we explore cochlear structure and function in mice lacking K(+) channels of the two-pore domain family. A profound deafness associated with a decrease in endocochlear potential is found in adult Kcnk5(-/-) mice. Hearing occurs around postnatal day 19 (P19), and completely disappears 2 days later. At P19, Kcnk5(-/-) mice have a normal endolymphatic [K(+)] but a partly lowered endocochlear potential. Using Lac-Z as a gene reporter, KCNK5 is mainly found in outer sulcus Claudius', Boettcher's and root cells. Low levels of expression are also seen in the spiral ganglion, Reissner's membrane and stria vascularis. Essential channels (KCNJ10 and KCNQ1) contributing to K(+) secretion in stria vascularis have normal expression in Kcnk5(-/-) mice. Thus, KCNK5 channels are indispensable for the maintenance of hearing. Among several plausible mechanisms, we emphasize their role in K(+) recycling along the outer sulcus lateral route.


Assuntos
Cóclea/metabolismo , Surdez/genética , Audição/genética , Canais de Potássio de Domínios Poros em Tandem/genética , Animais , Cóclea/patologia , Cóclea/fisiologia , Surdez/fisiopatologia , Endolinfa/química , Potenciais Evocados Auditivos do Tronco Encefálico , Audição/fisiologia , Imuno-Histoquímica , Canal de Potássio KCNQ1/metabolismo , Potenciais da Membrana/genética , Camundongos , Camundongos Knockout , Mutação , Proteínas do Tecido Nervoso/genética , Potássio , Canais de Potássio/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Janela da Cóclea/fisiopatologia , Gânglio Espiral da Cóclea/citologia , Gânglio Espiral da Cóclea/patologia , Estria Vascular/metabolismo , Testes de Função Vestibular
6.
Hum Mol Genet ; 24(2): 471-9, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25205110

RESUMO

Andersen's syndrome (AS) is a rare and dominantly inherited pathology, linked to the inwardly rectifying potassium channel Kir2.1. AS patients exhibit a triad of symptoms that include periodic paralysis, cardiac dysrhythmia and bone malformations. Some progress has been made in understanding the contribution of the Kir2.1 channel to skeletal and cardiac muscle dysfunctions, but its role in bone morphogenesis remains unclear. We isolated myoblast precursors from muscle biopsies of healthy individuals and typical AS patients with dysmorphic features. Myoblast cultures underwent osteogenic differentiation that led to extracellular matrix mineralization. Osteoblastogenesis was monitored through the activity of alkaline phosphatase, and through the hydroxyapatite formation using Alizarin Red and Von Kossa staining techniques. Patch-clamp recordings revealed the presence of an inwardly rectifying current in healthy cells that was absent in AS osteoblasts, showing the dominant-negative effect of the Kir2.1 mutant allele in osteoblasts. We also found that while control cells actively synthesize hydroxyapatite, AS osteoblasts are unable to efficiently form any extracellular matrix. To further demonstrate the role of the Kir2.1 channels during the osteogenesis, we inhibited Kir2.1 channel activity in healthy patient cells by applying extracellular Ba(2+) or using adenoviruses carrying mutant Kir2.1 channels. In both cases, cells were no longer able to produce extracellular matrixes. Moreover, osteogenic activity of AS osteoblasts was restored by rescue experiments, via wild-type Kir2.1 channel overexpression. These observations provide a proof that normal Kir2.1 channel function is essential during osteoblastogenesis.


Assuntos
Doença de Depósito de Glicogênio Tipo IV/metabolismo , Mioblastos/metabolismo , Osteogênese , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Células Cultivadas , Matriz Extracelular/metabolismo , Doença de Depósito de Glicogênio Tipo IV/genética , Doença de Depósito de Glicogênio Tipo IV/fisiopatologia , Humanos , Mioblastos/citologia , Osteoblastos/citologia , Osteoblastos/metabolismo , Técnicas de Patch-Clamp , Canais de Potássio Corretores do Fluxo de Internalização/genética
7.
Pflugers Arch ; 467(5): 917-29, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25346157

RESUMO

A number of the subunits within the family of K2P background K(+) channels are sensitive to changes in extracellular pH in the physiological range, making them likely candidates to mediate various pH-dependent processes. Based on expression patterns within several brainstem neuronal cell groups that are believed to function in CO2/H(+) regulation of breathing, three TASK subunits-TASK-1, TASK-2, and TASK-3-were specifically hypothesized to contribute to this central respiratory chemoreflex. For the acid-sensitive TASK-1 and TASK-3 channels, despite widespread expression at multiple levels within the brainstem respiratory control system (including presumptive chemoreceptor populations), experiments in knockout mice provided no evidence for their involvement in CO2 regulation of breathing. By contrast, the alkaline-activated TASK-2 channel has a more restricted brainstem distribution and was localized to the Phox2b-expressing chemoreceptor neurons of the retrotrapezoid nucleus (RTN). Remarkably, in a Phox2b(27Ala/+) mouse genetic model of congenital central hypoventilation syndrome (CCHS) that is characterized by reduced central respiratory chemosensitivity, selective ablation of Phox2b-expressing RTN neurons was accompanied by a corresponding loss of TASK-2 expression. Furthermore, genetic deletion of TASK-2 blunted RTN neuronal pH sensitivity in vitro, reduced alkaline-induced respiratory network inhibition in situ and diminished the ventilatory response to CO2/H(+) in vivo. Notably, a subpopulation of RTN neurons from TASK-2(-/-) mice retained their pH sensitivity, at least in part due to a residual pH-sensitive background K(+) current, suggesting that other mechanisms (and perhaps other K2P channels) for RTN neuronal pH sensitivity are yet to be identified.


Assuntos
Células Quimiorreceptoras/metabolismo , Concentração de Íons de Hidrogênio , Neurônios/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Potássio/metabolismo , Animais , Humanos , Respiração
8.
Pflugers Arch ; 467(5): 1027-42, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25339223

RESUMO

The physiological control of steroid hormone secretion from the adrenal cortex depends on the function of potassium channels. The "two-pore domain K(+) channels" (K2P) TWIK-related acid sensitive K(+) channel 1 (TASK1), TASK3, and TWIK-related K(+) channel 1 (TREK1) are strongly expressed in adrenocortical cells. They confer a background K(+) conductance to these cells which is important for the K(+) sensitivity as well as for angiotensin II and adrenocorticotropic hormone-dependent stimulation of aldosterone and cortisol synthesis. Mice with single deletions of the Task1 or Task3 gene as well as Task1/Task3 double knockout mice display partially autonomous aldosterone synthesis. It appears that TASK1 and TASK3 serve different functions: TASK1 affects cell differentiation and prevents expression of aldosterone synthase in the zona fasciculata, while TASK3 controls aldosterone secretion in glomerulosa cells. TREK1 is involved in the regulation of cortisol secretion in fasciculata cells. These data suggest that a disturbed function of K2P channels could contribute to adrenocortical pathologies in humans.


Assuntos
Córtex Suprarrenal/metabolismo , Aldosterona/metabolismo , Angiotensina II/metabolismo , Membrana Celular/metabolismo , Canais de Potássio/metabolismo , Animais , Humanos , Proteínas do Tecido Nervoso/metabolismo
9.
Front Cell Neurosci ; 7: 246, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24368895

RESUMO

TWIK-1 two-pore domain K(+) channels are expressed abundantly in astrocytes. In the present study, we examined the extent to which TWIK-1 contributes to the linear current-voltage (I-V) relationship (passive) K(+) membrane conductance, a dominant electrophysiological feature of mature hippocampal astrocytes. Astrocytes from TWIK-1 knockout mice have a more negative resting potential than those from wild type animals and a reduction in both inward rectification and Cs(+) permeability. Nevertheless, the overall whole-cell passive conductance is not altered significantly in TWIK-1 knockout astrocytes. The expression of Kir4.1 and TREK-1, two other major astrocytic K(+) channels, or of other two-pore K(+) channels is not altered in TWIK-1 knockout mice, suggesting that the mild effect of TWIK-1 knockout does not result from compensation by these channels. Fractionation experiments showed that TWIK-1 is primarily localized in intracellular cytoplasmic fractions (55%) and mildly hydrophobic internal compartment fractions (41%), with only 5% in fractions containing plasma membranes. Our study revealed that TWIK-1 proteins are mainly located in the intracellular compartments of hippocampal astrocyte under physiological condition, therefore a minimal contribution of TWIK-1 channels to whole-cell currents is likely attributable to a relatively low level presence of channels in the plasma membrane.

10.
J Neurosci ; 33(41): 16033-44, 2013 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-24107938

RESUMO

Phox2b-expressing glutamatergic neurons of the retrotrapezoid nucleus (RTN) display properties expected of central respiratory chemoreceptors; they are directly activated by CO2/H(+) via an unidentified pH-sensitive background K(+) channel and, in turn, facilitate brainstem networks that control breathing. Here, we used a knock-out mouse model to examine whether TASK-2 (K2P5), an alkaline-activated background K(+) channel, contributes to RTN neuronal pH sensitivity. We made patch-clamp recordings in brainstem slices from RTN neurons that were identified by expression of GFP (directed by the Phox2b promoter) or ß-galactosidase (from the gene trap used for TASK-2 knock-out). Whereas nearly all RTN cells from control mice were pH sensitive (95%, n = 58 of 61), only 56% of GFP-expressing RTN neurons from TASK-2(-/-) mice (n = 49 of 88) could be classified as pH sensitive (>30% reduction in firing rate from pH 7.0 to pH 7.8); the remaining cells were pH insensitive (44%). Moreover, none of the recorded RTN neurons from TASK-2(-/-) mice selected based on ß-galactosidase activity (a subpopulation of GFP-expressing neurons) were pH sensitive. The alkaline-activated background K(+) currents were reduced in amplitude in RTN neurons from TASK-2(-/-) mice that retained some pH sensitivity but were absent from pH-insensitive cells. Finally, using a working heart-brainstem preparation, we found diminished inhibition of phrenic burst amplitude by alkalization in TASK-2(-/-) mice, with apneic threshold shifted to higher pH levels. In conclusion, alkaline-activated TASK-2 channels contribute to pH sensitivity in RTN neurons, with effects on respiration in situ that are particularly prominent near apneic threshold.


Assuntos
Células Quimiorreceptoras/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Centro Respiratório/metabolismo , Animais , Feminino , Concentração de Íons de Hidrogênio , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Endocrinology ; 154(8): 2712-22, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23698720

RESUMO

Task3 K(+) channels are highly expressed in the adrenal cortex and contribute to the angiotensin II and K(+) sensitivity of aldosterone-producing glomerulosa cells. Adult Task3(-/-) mice display a partially autonomous aldosterone secretion, subclinical hyperaldosteronism, and salt-sensitive hypertension. Here, we investigated the age dependence of the adrenal phenotype of Task3(-/-) mice. Compared with adults, newborn Task3(-/-) mice displayed a severe adrenal phenotype with strongly increased plasma levels of aldosterone, corticosterone, and progesterone. This adrenocortical dysfunction was accompanied by a modified gene expression profile. The most strongly up-regulated gene was the protease renin. Real-time PCR corroborated the strong increase in adrenal renin expression, and immunofluorescence revealed renin-expressing cells in the zona fasciculata. Together with additional factors, activation of the local adrenal renin system is probably causative for the severely disturbed steroid hormone secretion of neonatal Task3(-/-) mice. The changes in gene expression patterns of neonatal Task3(-/-) mice could also be relevant for other forms of hyperaldosteronism.


Assuntos
Glândulas Suprarrenais/metabolismo , Hiperaldosteronismo/genética , Canais de Potássio/genética , Sistema Renina-Angiotensina/genética , Aldosterona/sangue , Aldosterona/metabolismo , Animais , Animais Recém-Nascidos , Corticosterona/sangue , Corticosterona/metabolismo , Citocromo P-450 CYP11B2/genética , Citocromo P-450 CYP11B2/metabolismo , Feminino , Imunofluorescência , Perfilação da Expressão Gênica , Hiperaldosteronismo/sangue , Hiperaldosteronismo/metabolismo , Rim/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Canais de Potássio/deficiência , Progesterona/sangue , Progesterona/metabolismo , Renina/sangue , Renina/genética , Renina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Zona Fasciculada/metabolismo
12.
Endocrinology ; 153(10): 4740-8, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22878402

RESUMO

Task1 and Task3 potassium channels (Task: tandem of P domains in a weak inward rectifying K(+) channel-related acid-sensitive K(+) channel) are believed to control the membrane voltage of aldosterone-producing adrenal glomerulosa cells. This study aimed at understanding the role of Task3 for the control of aldosterone secretion. The adrenal phenotype of Task3(-/-) mice was investigated using electrophysiology, adrenal slices, and blood pressure measurements. Primary adrenocortical cells of Task3(-/-) mice were strongly depolarized compared with wild-type (-52 vs. -79 mV), and in fresh adrenal slices Ca(2+) signaling of Task3(-/-) glomerulosa cells was abnormal. In living Task3(-/-) mice, the regulation of aldosterone secretion showed specific deficits: Under low Na(+) and high K(+) diets, protocols known to increase aldosterone, and under standard diet, Task3 inactivation was compensated and aldosterone was normal. However, high Na(+) and low K(+) diets, two protocols known to lower aldosterone, failed to lower aldosterone in Task3(-/-) mice. The physiological regulation of aldosterone was disturbed: aldosterone-renin ratio, an indicator of autonomous aldosterone secretion, was 3-fold elevated at standard and high Na(+) diets. Isolated adrenal glands of Task3(-/-) produced 2-fold more aldosterone. As a consequence, Task3(-/-) mice showed salt-sensitive arterial hypertension (plus 10 mm Hg). In conclusion, Task3 plays an important role in the adaptation of aldosterone secretion to dietary salt intake.


Assuntos
Glândulas Suprarrenais/metabolismo , Pressão Sanguínea/genética , Hipertensão/genética , Canais de Potássio/genética , Renina/sangue , Cloreto de Sódio na Dieta , Adaptação Fisiológica/genética , Glândulas Suprarrenais/citologia , Aldosterona/sangue , Animais , Cálcio/metabolismo , Células Cultivadas , Hipertensão/metabolismo , Camundongos , Camundongos Knockout , Fenótipo , Canais de Potássio/metabolismo , Zona Glomerulosa/citologia , Zona Glomerulosa/metabolismo
13.
Hum Mol Genet ; 21(22): 4922-9, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22918120

RESUMO

Primary aldosteronism (PA, autonomous aldosterone production from the adrenal cortex) causes the most common form of secondary arterial hypertension (HT), which is also the most common curable form of HT. Recent studies have highlighted an important role of mutations in genes encoding potassium channels in the pathogenesis of PA, both in human disease and in animal models. Here, we have exploited the unique features of the hyperaldosteronemic phenotype of Kcnk3 null mice, which is dependent on sexual hormones, to identify genes whose expression is modulated in the adrenal gland according to the dynamic hyperaldosteronemic phenotype of those animals. Genetic inactivation of one of the genes identified by our strategy, dickkopf-3 (Dkk3), whose expression is increased by calcium influx into adrenocortical cells, in the Kcnk3 null background results in the extension of the low-renin, potassium-rich diet insensitive hyperaldosteronemic phenotype to the male sex. Compound Kcnk3/Dkk3 animals display an increased expression of Cyp11b2, the rate-limiting enzyme for aldosterone biosynthesis in the adrenal zona glomerulosa (ZG). Our data show that Dkk3 can act as a modifier gene in a mouse model for altered potassium channel function and suggest its potential involvement in human PA syndromes.


Assuntos
Córtex Suprarrenal/metabolismo , Aldosterona/biossíntese , Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Córtex Suprarrenal/patologia , Animais , Cálcio/metabolismo , Análise por Conglomerados , Feminino , Perfilação da Expressão Gênica , Inativação Gênica , Hiperaldosteronismo/genética , Hiperaldosteronismo/metabolismo , Masculino , Camundongos , Camundongos Knockout , Fenótipo
14.
Proc Natl Acad Sci U S A ; 109(14): 5499-504, 2012 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-22431633

RESUMO

TWIK1 belongs to the family of background K(+) channels with two pore domains. In native and transfected cells, TWIK1 is detected mainly in recycling endosomes. In principal cells in the kidney, TWIK1 gene inactivation leads to the loss of a nonselective cationic conductance, an unexpected effect that was attributed to adaptive regulation of other channels. Here, we show that TWIK1 ion selectivity is modulated by extracellular pH. Although TWIK1 is K(+) selective at neutral pH, it becomes permeable to Na(+) at the acidic pH found in endosomes. Selectivity recovery is slow after restoration of a neutral pH. Such hysteresis makes plausible a role of TWIK1 as a background channel in which selectivity and resulting inhibitory or excitatory influences on cell excitability rely on its recycling rate between internal acidic stores and the plasma membrane. TWIK1(-/-) pancreatic ß cells are more polarized than control cells, confirming a depolarizing role of TWIK1 in kidney and pancreatic cells.


Assuntos
Canais de Potássio/metabolismo , Sequência de Aminoácidos , Animais , Concentração de Íons de Hidrogênio , Modelos Moleculares , Dados de Sequência Molecular , Canais de Potássio/química , Homologia de Sequência de Aminoácidos , Xenopus
15.
Physiology (Bethesda) ; 26(6): 424-37, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22170960

RESUMO

Background K(2P) channels are tightly regulated by different stimuli including variations of external and internal pH. pH sensitivity relies on proton-sensing residues that influence channel gating and activity. Gene inactivation in the mouse is a revealing implication of K(2P) channels in many physiological functions ranging from hormone secretion to central respiratory adaptation. Surprisingly, only a few phenotypic traits of these mice have yet been directly related to the pH sensitivity of K(2P) channels.


Assuntos
Neurônios/fisiologia , Canais de Potássio de Domínios Poros em Tandem/química , Canais de Potássio de Domínios Poros em Tandem/fisiologia , Sequência de Aminoácidos , Animais , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Canais de Potássio de Domínios Poros em Tandem/metabolismo
16.
J Neurosci ; 31(46): 16541-9, 2011 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-22090480

RESUMO

The mechanotransduction of vestibular sensory cells depends on the high endolymphatic potassium concentration ([K+]) maintained by a fine balance between K+ secretion and absorption by epithelial cells. Despite the crucial role of endolymph as an electrochemical motor for mechanotransduction, little is known about the processes that govern endolymph formation. To address these, we took advantage of an organotypic rodent model, which regenerates a genuine neonatal vestibular endolymphatic compartment, facilitating the determination of endolymphatic [K+] and transepithelial potential (Vt) during endolymph formation. While mature Vt levels are almost immediately achieved, K+ accumulates to reach a steady [K+] by day 5 in culture. Inhibition of sensory cell K+ efflux enhances [K+] regardless of the blocker used (FM1.43, amikacin, gentamicin, or gadolinium). Targeting K+ secretion with bumetanide partially and transiently reduces [K+], while ouabain application and Kcne1 deletion almost abolishes it. Immunofluorescence studies demonstrate that dark cells do not express Na-K-2Cl cotransporter 1 (the target of bumetanide) in cultured and young mouse utricles, while Na/K-ATPase (the target of ouabain) is found in dark cells and transitional cells. This global analysis of the involvement of endolymphatic homeostasis actors in the immature organ (1) confirms that KCNE1 channels are necessary for K+ secretion, (2) highlights Na/K-ATPase as the key endolymphatic K+ provider and shows that Na-K-2Cl cotransporter 1 has a limited impact on K+ influx, and (3) demonstrates that transitional cells are involved in K+ secretion in the early endolymphatic compartment.


Assuntos
Endolinfa/metabolismo , Células Epiteliais/fisiologia , Sáculo e Utrículo/crescimento & desenvolvimento , Sáculo e Utrículo/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Aminoglicosídeos/farmacologia , Animais , Animais Recém-Nascidos , Bumetanida/farmacologia , Endocitose/genética , Inibidores Enzimáticos/farmacologia , Células Epiteliais/efeitos dos fármacos , Feminino , Gadolínio/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Cultura de Órgãos , Ouabaína/farmacologia , Potássio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Compostos de Piridínio/farmacocinética , Compostos de Amônio Quaternário/farmacocinética , Ratos , Ratos Wistar , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Fatores de Tempo
17.
Cell Physiol Biochem ; 25(4-5): 367-78, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20332617

RESUMO

Most bacteria initiate host inflammatory responses through interactions with epithelial cells. Lipopolysaccharide (LPS), a component of the bacterial cell wall is a major cause of septic shock in emergency care units and in the pathogenesis of acute renal failure. Kidney cells exposed to LPS undergo apoptotic changes, including cell volume decrease, phosphatidylserine exposure, caspase-3- and membrane K+ conductance -activation. Whole-cell configuration was used to identify K+ channels in primary and immortalized culture of mice distal convoluted tubules. LPS exposure induced a 3 fold increase in intracellular cAMP concentration and the activation of an outwardly rectifying K+ conductance in both immortalized and primary culture of distal cells. This LPS-induced current exhibited KCNQ1 K+ channel characteristics, i.e. inhibition by quinidine, chromanol293B and low dose of HMR1556 (IC50<1 microM) and insensitive to TEA and charybdotoxin. The background-like biophysical properties of the current suggest that the KCNQ1 pore-forming subunit is associated with a KCNE2 or KCNE3 ancillary subunit. RT-PCR experiments confirmed the presence of KCNQ1 and KCNE3 mRNA transcripts in primary culture of distal segments. Activation of the KCNQ1/KCNE3 K+ current appeared to be an essential step in the LPS-induced apoptosis process since HMR1556 blocked the LPS-induced- cell volume decrease, -caspase-3 activation and -phosphatidylserine exposure.


Assuntos
Apoptose , Canal de Potássio KCNQ1/metabolismo , Túbulos Renais Distais/metabolismo , Animais , Caspase 3/metabolismo , Células Cultivadas , Cromanos/farmacologia , AMP Cíclico/metabolismo , Canal de Potássio KCNQ1/antagonistas & inibidores , Túbulos Renais Distais/citologia , Lipopolissacarídeos/toxicidade , Camundongos , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Subunidades Proteicas/metabolismo , Quinidina/farmacologia , Sulfonamidas/farmacologia
18.
Proc Natl Acad Sci U S A ; 107(5): 2325-30, 2010 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-20133877

RESUMO

Task2 K(+) channel expression in the central nervous system is surprisingly restricted to a few brainstem nuclei, including the retrotrapezoid (RTN) region. All Task2-positive RTN neurons were lost in mice bearing a Phox2b mutation that causes the human congenital central hypoventilation syndrome. In plethysmography, Task2(-/-) mice showed disturbed chemosensory function with hypersensitivity to low CO(2) concentrations, leading to hyperventilation. Task2 probably is needed to stabilize the membrane potential of chemoreceptive cells. In addition, Task2(-/-) mice lost the long-term hypoxia-induced respiratory decrease whereas the acute carotid-body-mediated increase was maintained. The lack of anoxia-induced respiratory depression in the isolated brainstem-spinal cord preparation suggested a central origin of the phenotype. Task2 activation by reactive oxygen species generated during hypoxia could silence RTN neurons, thus contributing to respiratory depression. These data identify Task2 as a determinant of central O(2) chemoreception and demonstrate that this phenomenon is due to the activity of a small number of neurons located at the ventral medullary surface.


Assuntos
Dióxido de Carbono/fisiologia , Oxigênio/fisiologia , Canais de Potássio de Domínios Poros em Tandem/fisiologia , Centro Respiratório/fisiologia , Animais , Animais Recém-Nascidos , Tronco Encefálico/patologia , Tronco Encefálico/fisiologia , Tronco Encefálico/fisiopatologia , Células Quimiorreceptoras/patologia , Células Quimiorreceptoras/fisiologia , Modelos Animais de Doenças , Feminino , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Humanos , Hipercapnia/fisiopatologia , Hipóxia/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Pletismografia Total , Canais de Potássio de Domínios Poros em Tandem/deficiência , Canais de Potássio de Domínios Poros em Tandem/genética , Gravidez , Fenômenos Fisiológicos Respiratórios , Apneia do Sono Tipo Central/etiologia , Apneia do Sono Tipo Central/genética , Apneia do Sono Tipo Central/fisiopatologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia
19.
J Biol Chem ; 285(10): 7165-75, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20051516

RESUMO

The KCNE3 beta-subunit constitutively opens outwardly rectifying KCNQ1 (Kv7.1) K(+) channels by abolishing their voltage-dependent gating. The resulting KCNQ1/KCNE3 heteromers display enhanced sensitivity to K(+) channel inhibitors like chromanol 293B. KCNE3 was also suggested to modify biophysical properties of several other K(+) channels, and a mutation in KCNE3 was proposed to underlie forms of human periodic paralysis. To investigate physiological roles of KCNE3, we now disrupted its gene in mice. kcne3(-/-) mice were viable and fertile and displayed neither periodic paralysis nor other obvious skeletal muscle abnormalities. KCNQ1/KCNE3 heteromers are present in basolateral membranes of intestinal and tracheal epithelial cells where they might facilitate transepithelial Cl(-) secretion through basolateral recycling of K(+) ions and by increasing the electrochemical driving force for apical Cl(-) exit. Indeed, cAMP-stimulated electrogenic Cl(-) secretion across tracheal and intestinal epithelia was drastically reduced in kcne3(-/-) mice. Because the abundance and subcellular localization of KCNQ1 was unchanged in kcne3(-/-) mice, the modification of biophysical properties of KCNQ1 by KCNE3 is essential for its role in intestinal and tracheal transport. Further, these results suggest KCNE3 as a potential modifier gene in cystic fibrosis.


Assuntos
Cloretos/metabolismo , Mucosa Intestinal/metabolismo , Canal de Potássio KCNQ1/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Subunidades Proteicas/metabolismo , Traqueia/metabolismo , Animais , Linhagem Celular , Mucosa Gástrica/metabolismo , Humanos , Intestinos/citologia , Canal de Potássio KCNQ1/química , Canal de Potássio KCNQ1/genética , Camundongos , Camundongos Knockout , Músculo Esquelético/metabolismo , Técnicas de Patch-Clamp , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Estrutura Quaternária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/genética , Teste de Desempenho do Rota-Rod , Estômago/citologia , Distribuição Tecidual , Traqueia/citologia
20.
J Biol Chem ; 285(7): 4798-805, 2010 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-19959478

RESUMO

Tandem of P domains in a weak inwardly rectifying K(+) channel 1 (TWIK1) is a K(+) channel that produces unusually low levels of current. Replacement of lysine 274 by a glutamic acid (K274E) is associated with stronger currents. This mutation would prevent conjugation of a small ubiquitin modifier peptide to Lys-274, a mechanism proposed to be responsible for channel silencing. However, we found no biochemical evidence of TWIK1 sumoylation, and we showed that the conservative change K274R did not increase current, suggesting that K274E modifies TWIK1 gating through a charge effect. Now we rule out an eventual effect of K274E on TWIK1 trafficking, and we provide convincing evidence that TWIK1 silencing results from its rapid retrieval from the cell surface. TWIK1 is internalized via a dynamin-dependent mechanism and addressed to the recycling endosomal compartment. Mutation of a diisoleucine repeat located in its cytoplasmic C terminus (I293A,I294A) stabilizes TWIK1 at the plasma membrane, resulting in robust currents. The effects of I293A,I294A on channel trafficking and of K274E on channel activity are cumulative, promoting even more currents. Activation of serotoninergic receptor 5-HT(1)R or adrenoreceptor alpha2A-AR stimulates TWIK1 but has no effect on TWIK1I293A,I294A, suggesting that G(i) protein activation is a physiological signal for increasing the number of active channels at the plasma membrane.


Assuntos
Endocitose/fisiologia , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Transporte Proteico/fisiologia , Animais , Linhagem Celular , Membrana Celular/metabolismo , Cães , Eletrofisiologia , Endocitose/genética , Humanos , Imuno-Histoquímica , Microscopia Eletrônica , Mutação , Fosforilação/efeitos dos fármacos , Canais de Potássio de Domínios Poros em Tandem/genética , Transporte Proteico/genética , Receptores 5-HT1 de Serotonina/metabolismo , Serotonina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA