Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Cell Physiol Biochem ; 42(4): 1377-1389, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28704808

RESUMO

BACKGROUND: MicroRNAs (miRNAs) negatively regulate gene expression at a post-transcriptional level. Dicer, a cytoplasmic RNase III enzyme, is required for the maturation of miRNAs from precursor miRNAs. Dicer, therefore, is a critical enzyme involved in the biogenesis and processing of miRNAs. Several biological processes are controlled by miRNAs, including the regulation of T cell development and function. T cells generate reactive oxygen species (ROS) with parallel H+ extrusion accomplished by the Na+/H+-exchanger 1 (NHE1). The present study explored whether ROS production, as well as NHE1 expression and function are sensitive to the lack of Dicer (miRNAs deficient) and could be modified by individual miRNAs. METHODS: CD4+ T cells were isolated from CD4 specific Dicer deficient (DicerΔ/Δ) mice and the respective control mice (Dicerfl/fl). Transcript and protein levels were quantified with RT-PCR and Western blotting, respectively. For determination of intracellular pH (pHi) cells were incubated with the pH sensitive dye bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein (BCECF) and Na+/H+ exchanger (NHE) activity was calculated from re-alkalinization after an ammonium pulse. Changes in cell volume were measured using the forward scatter in flow cytometry, and ROS production utilizing 2',7' -dichlorofluorescin diacetate (DCFDA) fluorescence. Transfection of miRNA-control and mimics in T cells was performed using DharmaFECT3 reagent. RESULTS: ROS production, cytosolic H+ concentration, NHE1 transcript and protein levels, NHE activity, and cell volume were all significantly higher in CD4+ T cells from DicerΔ/Δ mice than in CD4+ T cells from Dicerfl/fl mice. Furthermore, individual miR-200b and miR-15b modify pHi and NHE activity in Dicerfl/fl and DicerΔ/Δ CD4+ T cells, respectively. CONCLUSIONS: Lack of Dicer leads to oxidative stress, cytosolic acidification, upregulated NHE1 expression and activity as well as swelling of CD4+ T cells, functions all reversed by miR-15b or miR-200b.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Proteínas de Transporte de Cátions/genética , RNA Helicases DEAD-box/deficiência , MicroRNAs/genética , Espécies Reativas de Oxigênio/metabolismo , Ribonuclease III/deficiência , Trocadores de Sódio-Hidrogênio/genética , Animais , Linfócitos T CD4-Positivos/imunologia , Proteínas de Transporte de Cátions/imunologia , Citosol/imunologia , Citosol/metabolismo , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/imunologia , Regulação da Expressão Gênica , Concentração de Íons de Hidrogênio , Transporte de Íons/imunologia , Camundongos , Camundongos Knockout , MicroRNAs/imunologia , Espécies Reativas de Oxigênio/imunologia , Ribonuclease III/genética , Ribonuclease III/imunologia , Transdução de Sinais , Trocador 1 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/imunologia
2.
Cell Physiol Biochem ; 38(4): 1652-62, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27119824

RESUMO

BACKGROUND/AIMS: The oxidative stress-responsive kinase 1 (OSR1) and the serine/threonine kinases SPAK (SPS1-related proline/alanine-rich kinase) are under the control of WNK (with-no-K [Lys]) kinases. OSR1 and SPAK participate in diverse functions including cell volume regulation and neuronal excitability. Cell volume and neuronal excitation are further modified by the large conductance Ca2+-activated K+ channels (maxi K+ channel or BK channels). An influence of OSR1 and/or SPAK on BK channel activity has, however, never been shown. The present study thus explored whether OSR1 and/or SPAK modify the activity of BK channels. METHODS: cRNA encoding the Ca2+ insensitive BK channel mutant BKM513I+x0394;899-903 was injected into Xenopus laevis oocytes without or with additional injection of cRNA encoding wild-type OSR1 or wild-type SPAK, constitutively active T185EOSR1, catalytically inactive D164AOSR1, constitutively active T233ESPAK or catalytically inactive D212ASPAK. K+ channel activity was measured utilizing dual electrode voltage clamp. RESULTS: BK channel activity in BKM513I+x0394;899-903 expressing oocytes was significantly decreased by co-expression of OSR1 or SPAK. The effect of wild-type OSR1/SPAK was mimicked by T185EOSR1 and T233ESPAK, but not by D164AOSR1 or D212ASPAK. CONCLUSIONS: OSR1 and SPAK suppress BK channels, an effect possibly contributing to cell volume regulation and neuroexcitability.


Assuntos
Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Citometria de Fluxo , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Alta/genética , Linfócitos/citologia , Linfócitos/metabolismo , Masculino , Camundongos , Mutagênese , Oócitos/metabolismo , Técnicas de Patch-Clamp , Proteínas Serina-Treonina Quinases/genética , Xenopus laevis/crescimento & desenvolvimento , Xenopus laevis/metabolismo
3.
J Membr Biol ; 249(3): 239-49, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26690923

RESUMO

Excitatory amino acid transporters EAAT1 (SLC1A3), EAAT2 (SLC1A2), EAAT3 (SLC1A1), and EAAT4 (SLC1A6) serve to clear L-glutamate from the synaptic cleft and are thus important for the limitation of neuronal excitation. EAAT3 has previously been shown to form complexes with caveolin-1, a major component of caveolae, which participate in the regulation of transport proteins. The present study explored the impact of caveolin-1 on electrogenic transport by excitatory amino acid transporter isoforms EAAT1-4. To this end cRNA encoding EAAT1, EAAT2, EAAT3, or EAAT4 was injected into Xenopus oocytes without or with additional injection of cRNA encoding caveolin-1. The L-glutamate (2 mM)-induced inward current (I Glu) was taken as a measure of glutamate transport. As a result, I Glu was observed in EAAT1-, EAAT2-, EAAT3-, or EAAT4-expressing oocytes but not in water-injected oocytes, and was significantly decreased by coexpression of caveolin-1. Caveolin-1 decreased significantly the maximal transport rate. Treatment of EAATs-expressing oocytes with brefeldin A (5 µM) was followed by a decrease in conductance, which was similar in oocytes expressing EAAT together with caveolin-1 as in oocytes expressing EAAT1-4 alone. Thus, caveolin-1 apparently does not accelerate transporter protein retrieval from the cell membrane. In conclusion, caveolin-1 is a powerful negative regulator of the excitatory glutamate transporters EAAT1, EAAT2, EAAT3, and EAAT4.


Assuntos
Caveolina 1/metabolismo , Transportador 1 de Aminoácido Excitatório/metabolismo , Transportador 2 de Aminoácido Excitatório/metabolismo , Transportador 3 de Aminoácido Excitatório/metabolismo , Transportador 4 de Aminoácido Excitatório/metabolismo , Animais , Transporte Biológico , Caveolina 1/genética , Transportador 1 de Aminoácido Excitatório/genética , Transportador 2 de Aminoácido Excitatório/genética , Transportador 3 de Aminoácido Excitatório/genética , Transportador 4 de Aminoácido Excitatório/genética , Expressão Gênica , Ácido Glutâmico/metabolismo , Humanos , Oócitos/metabolismo , Xenopus laevis
4.
FASEB J ; 29(5): 1701-10, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25609428

RESUMO

Gene variants of the leucine-rich repeat kinase 2 (LRRK2) are associated with susceptibility to Parkinson's disease (PD). Besides brain and periphery, LRRK2 is expressed in various immune cells including dendritic cells (DCs), antigen-presenting cells linking innate and adaptive immunity. However, the function of LRRK2 in the immune system is still incompletely understood. Here, Ca(2+)-signaling was analyzed in DCs isolated from gene-targeted mice lacking lrrk2 (Lrrk2(-/-)) and their wild-type littermates (Lrrk2(+/+)). According to Western blotting, Lrrk2 was expressed in Lrrk2(+/+) DCs but not in Lrrk2(-/-)DCs. Cytosolic Ca(2+) levels ([Ca(2+)]i) were determined utilizing Fura-2 fluorescence and whole cell currents to decipher electrogenic transport. The increase of [Ca(2+)]i following inhibition of sarcoendoplasmatic Ca(2+)-ATPase with thapsigargin (1 µM) in the absence of extracellular Ca(2+) (Ca(2+)-release) and the increase of [Ca(2+)]i following subsequent readdition of extracellular Ca(2+) (SOCE) were both significantly larger in Lrrk2(-/-) than in Lrrk2(+/+) DCs. The augmented increase of [Ca(2+)]i could have been due to impaired Ca(2+) extrusion by K(+)-independent (NCX) and/or K(+)-dependent (NCKX) Na(+)/Ca(2+)-exchanger activity, which was thus determined from the increase of [Ca(2+)]i, (Δ[Ca(2+)]i), and current following abrupt replacement of Na(+) containing (130 mM) and Ca(2+) free (0 mM) extracellular perfusate by Na(+) free (0 mM) and Ca(2+) containing (2 mM) extracellular perfusate. As a result, both slope and peak of Δ[Ca(2+)]i as well as Na(+)/Ca(2+) exchanger-induced current were significantly lower in Lrrk2(-/-) than in Lrrk2(+/+) DCs. A 6 or 24 hour treatment with the LRRK2 inhibitor GSK2578215A (1 µM) significantly decreased NCX1 and NCKX1 transcript levels, significantly blunted Na(+)/Ca(2+)-exchanger activity, and significantly augmented the increase of [Ca(2+)]i following Ca(2+)-release and SOCE. In conclusion, the present observations disclose a completely novel functional significance of LRRK2, i.e., the up-regulation of Na(+)/Ca(2+) exchanger transcription and activity leading to attenuation of Ca(2+)-signals in DCs.


Assuntos
Cálcio/metabolismo , Células Dendríticas/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Trocador de Sódio e Cálcio/metabolismo , Sódio/metabolismo , Animais , Células Apresentadoras de Antígenos , Western Blotting , Células Cultivadas , Células Dendríticas/citologia , Feminino , Citometria de Fluxo , Técnicas Imunoenzimáticas , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Masculino , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Espécies Reativas de Oxigênio
5.
Pflugers Arch ; 467(9): 1871-80, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25319519

RESUMO

Checkpoint kinase 2 (Chk2) is the main effector kinase of ataxia telangiectasia mutated (ATM) and responsible for cell cycle regulation. ATM signaling has been shown to upregulate interferon-regulating factor-1 (IRF-1), a transcription factor also expressed in the kidney. Calcitriol (1,25 (OH)2D3), a major regulator of mineral metabolism, is generated by 25-hydroxyvitamin D 1α-hydroxylase in the kidney. Since 25-hydroxyvitamin D 1α-hydroxylase expression is enhanced by IRF-1, the present study explored the role of Chk2 for calcitriol formation and mineral metabolism. Chk2-deficient mice (chk2 (-/-)) were compared to wild-type mice (chk2 (+/+)). Transcript levels of renal 25-hydroxyvitamin D 1α-hydroxylase, Chk2, and IRF-1 were determined by RT-PCR; Klotho expression by Western blotting; bone density by µCT analysis; serum or plasma 1,25 (OH)2D3, PTH, and C-terminal FGF23 concentrations by immunoassays; and serum, fecal, and urinary calcium and phosphate concentrations by photometry. The renal expression of IRF-1 and 25-hydroxyvitamin D 1α-hydroxylase as well as serum 1,25 (OH)2D3 and FGF23 levels were significantly lower in chk2 (-/-) mice compared to chk2 (+/+) mice. Plasma PTH was not different between the genotypes. Renal calcium and phosphate excretion were significantly higher in chk2 (-/-) mice than in chk2 (+/+) mice despite hypophosphatemia and normocalcemia. Bone density was not different between the genotypes. We conclude that Chk2 regulates renal 25-hydroxyvitamin D 1α-hydroxylase expression thereby impacting on calcium and phosphate metabolism.


Assuntos
25-Hidroxivitamina D3 1-alfa-Hidroxilase/metabolismo , Densidade Óssea/fisiologia , Calcitriol/biossíntese , Quinase do Ponto de Checagem 2/metabolismo , Rim/metabolismo , Animais , Western Blotting , Cálcio/metabolismo , Ensaio de Imunoadsorção Enzimática , Fator de Crescimento de Fibroblastos 23 , Regulação da Expressão Gênica/fisiologia , Glucuronidase/metabolismo , Células HEK293 , Humanos , Proteínas Klotho , Camundongos , Camundongos Knockout , Fosfatos/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Microtomografia por Raio-X
6.
Cell Physiol Biochem ; 37(5): 2032-42, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26584301

RESUMO

BACKGROUND/AIMS: KCNQ1/E1 channels are expressed in diverse tissues and serve a variety of functions including endolymph secretion in the inner ear, cardiac repolarization, epithelial transport and cell volume regulation. Kinases involved in regulation of epithelial transport and cell volume include SPAK (SPS1-related proline/alanine-rich kinase) and OSR1 (oxidative stress-responsive kinase 1), which are under control of WNK (with-no-K[Lys]) kinases. The present study explored whether KCNQ1/E1 channels are regulated by SPAK and/or OSR1. METHODS: cRNA encoding KCNQ1/E1 was injected into Xenopus oocytes with or without additional injection of cRNA encoding wild-type SPAK, constitutively active T233ESPAK, WNK insensitive T233ASPAK, catalytically inactive D212ASPAK, wild-type OSR1, constitutively active T185EOSR1, WNK insensitive T185AOSR1 and catalytically inactive D164AOSR1. Voltage gated K+ channel activity was quantified utilizing dual electrode voltage clamp and KCNQ1/E1 channel protein abundance in the cell membrane utilizing chemiluminescence of KCNQ1/E1 containing an extracellular Flag tag epitope (KCNQ1-Flag/E1). RESULTS: KCNQ1/E1 activity and KCNQ1-Flag/E1 protein abundance were significantly enhanced by wild-type SPAK and T233ESPAK, but not by T233ASPAK and D212ASPAK. Similarly, KCNQ1/E1 activity and KCNQ1-Flag/E1 protein abundance were significantly increased by wild-type OSR1 and T185EOSR1, but not by T185AOSR1 and D164AOSR1. CONCLUSIONS: SPAK and OSR1 participate in the regulation of KCNQ1/E1 protein abundance and activity.


Assuntos
Membrana Celular/metabolismo , Canal de Potássio KCNQ1/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Potenciais de Ação , Animais , Humanos , Íons/química , Íons/metabolismo , Canal de Potássio KCNQ1/genética , Mutagênese Sítio-Dirigida , Oócitos/metabolismo , Técnicas de Patch-Clamp , Potássio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Proteínas Serina-Treonina Quinases/genética , Xenopus laevis/crescimento & desenvolvimento , Xenopus laevis/metabolismo
7.
Cell Physiol Biochem ; 37(6): 2454-63, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26666245

RESUMO

BACKGROUND: The voltage gated K+ channels Kv1.3 and Kv1.5 contribute to the orchestration of cell proliferation. Kinases participating in the regulation of cell proliferation include protein kinase B (PKB/Akt). The present study thus explored whether PKB/Akt modifies the abundance and function of Kv1.3 and Kv1.5. METHODS: Kv1.3 or Kv1.5 was expressed in Xenopus laevis oocytes with or without wild-type PKB/Akt, constitutively active T308D/S473DPKB/Akt or inactive T308A/S473APKB/Akt. The channel activity was quantified utilizing dual electrode voltage clamp. Moreover, HA-tagged Kv1.5 protein was determined utilizing chemiluminescence. RESULTS: Voltage gated K+ currents were observed in Kv1.3 or Kv1.5 expressing oocytes but not in water-injected oocytes or in oocytes expressing PKB/Akt alone. Co-expression of PKB/Akt or T308D/S473DPKB/Akt, but not co-expression of T308A/S473APKB/Akt significantly increased the voltage gated current in both Kv1.3 and Kv1.5 expressing oocytes. As shown for Kv1.5, co-expression of PKB/Akt enhanced the channel protein abundance in the cell membrane. In Kv1.5 expressing oocytes voltage gated current decreased following inhibition of carrier insertion by brefeldin A (5 µM) to similarly low values in the absence and presence of PKB/Akt, suggesting that PKB/Akt stimulated carrier insertion into rather than inhibiting carrier retrieval from the cell membrane. CONCLUSION: PKB/Akt up-regulates both, Kv1.3 and Kv1.5 K+ channels.


Assuntos
Canal de Potássio Kv1.3/metabolismo , Canal de Potássio Kv1.5/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Regulação para Cima , Animais , Xenopus laevis
8.
Cell Physiol Biochem ; 37(6): 2476-85, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26666518

RESUMO

BACKGROUND/AIMS: Janus kinase 3 (JAK3), a kinase mainly expressed in hematopoietic cells, has been shown to down-regulate the Na+/K+ ATPase and participate in the regulation of several ion channels and carriers. Channels expressed in thymus and regulating the abundance of T lymphocytes include the voltage gated K+ channel KCNE1/KCNQ1. The present study explored whether JAK3 contributes to the regulation of KCNE1/KCNQ1. METHODS: cRNA encoding KCNE1/KCNQ1 was injected into Xenopus oocytes with or without additional injection of cRNA encoding wild-type JAK3, constitutively active A568VJAK3, or inactive K851AJAK3. Voltage gated K+ channel activity was measured utilizing two electrode voltage clamp. RESULTS: KCNE1/KCNQ1 activity was significantly increased by wild-type JAK3 and A568VJAK3, but not by K851AJAK3. The difference between oocytes expressing KCNE1/KCNQ1 alone and oocytes expressing KCNE1/KCNQ1 with A568VJAK3 was virtually abrogated by JAK3 inhibitor WHI-P154 (22 µM) but not by inhibition of transcription with actinomycin D (50 nM). Inhibition of KCNE1/KCNQ1 protein insertion into the cell membrane by brefeldin A (5 µM) resulted in a decline of the voltage gated current, which was similar in the absence and presence of A568VJAK3, suggesting that A568VJAK3 did not accelerate KCNE1/KCNQ1 protein retrieval from the cell membrane. CONCLUSION: JAK3 contributes to the regulation of membrane KCNE1/KCNQ1 activity, an effect sensitive to JAK3 inhibitor WHI-P154.


Assuntos
Janus Quinase 3/metabolismo , Canal de Potássio KCNQ1/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Proteínas de Xenopus/fisiologia , Animais , Xenopus laevis
9.
Cell Physiol Biochem ; 37(1): 297-305, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26303250

RESUMO

BACKGROUND/AIMS: Janus kinase 3 (JAK3), a tyrosine kinase contributing to the regulation of cell proliferation and apoptosis of lymphocytes and tumour cells, has been shown to modify the expression and function of several ion channels and transport proteins. Channels involved in the regulation of cell proliferation include the large conductance voltage- and Ca(2+)-activated K(+) channel BK. The present study explored whether JAK3 modifies BK channel protein abundance and current. METHODS: cRNA encoding Ca(2+)-insensitive BK channel (BK(M513I+Δ899-903)) was injected into Xenopus oocytes with or without additional injection of cRNA encoding wild-type JAK3, constitutively active A568VJAK3, or inactive (K851A)JAK3. Voltage gated K(+ )channel activity was measured utilizing dual electrode voltage clamp. Moreover, BK channel protein abundance was determined utilizing flow cytometry in CD19(+) B lymphocyte cell membranes from mice lacking functional JAK3 (jak3(-/-)) and corresponding wild-type mice (jak3(+/+)). RESULTS: BK activity in BK(M513I+Δ899-903) expressing oocytes was slightly but significantly decreased by coexpression of wild-type JAK3 and of (A568V)JAK3, but not by coexpression of (K851A)JAK3. The BK channel protein abundance in the cell membrane was significantly higher in jak3(-/-) than in jak3(+/+) B lymphocytes. The decline of conductance in BK and JAK3 coexpressing oocytes following inhibition of channel protein insertion by brefeldin A (5 µM) was similar in oocytes expressing BK with JAK3 and oocytes expressing BK alone, indicating that JAK3 might slow channel protein insertion into rather than accelerating channel protein retrieval from the cell membrane. CONCLUSION: JAK3 is a weak negative regulator of membrane BK protein abundance and activity.


Assuntos
Janus Quinase 3/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Animais , Antígenos CD19/metabolismo , Apoptose/efeitos dos fármacos , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Brefeldina A/farmacologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Feminino , Camundongos , Oócitos/metabolismo , Xenopus/metabolismo
10.
J Membr Biol ; 248(1): 59-66, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25315612

RESUMO

SPS1-related proline/alanine-rich kinase (SPAK) and oxidative stress-responsive kinase 1 (OSR1) are potent regulators of several transporters and ion channels. The kinases are under regulation of with-no-K(Lys) (WNK) kinases. The present study explored whether SPAK and/or OSR1 modify the expression and/or activity of the voltage-gated K(+) channel Kv1.5, which participates in the regulation of diverse functions including atrial cardiac action potential and tumor cell proliferation. cRNA encoding Kv1.5 was injected into Xenopus oocytes with or without additional injection of cRNA encoding wild-type SPAK, constitutively active (T233E)SPAK, WNK insensitive (T233A)SPAK, catalytically inactive (D212A)SPAK, wild-type OSR1, constitutively active (T185E)OSR1, WNK insensitive (T185A)OSR1, and catalytically inactive (D164A)OSR1. Voltage-gated K(+) channel activity was quantified utilizing dual electrode voltage clamp and Kv1.5 channel protein abundance in the cell membrane utilizing chemiluminescence of Kv1.5 containing an extracellular hemagglutinin epitope (Kv1.5-HA). Kv1.5 activity and Kv1.5-HA protein abundance were significantly decreased by wild-type SPAK and (T233E)SPAK, but not by (T233A)SPAK and (D212A)SPAK. Similarly, Kv1.5 activity and Kv1.5-HA protein abundance were significantly down-regulated by wild-type OSR1 and (T185E)OSR1, but not by (T185A)OSR1 and (D164A)OSR1. Both, SPAK and OSR1 decrease cell membrane Kv1.5 protein abundance and activity.


Assuntos
Canal de Potássio Kv1.5/metabolismo , Oócitos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Canal de Potássio Kv1.5/genética , Proteínas Serina-Treonina Quinases/genética , Xenopus
11.
J Membr Biol ; 248(6): 1061-70, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26100849

RESUMO

The tyrosine kinase Janus kinase 3 (JAK3) participates in the regulation of cell proliferation and apoptosis. The kinase further influences ion channels and transport proteins. The present study explored whether JAK3 contributes to the regulation of the voltage-gated K(+) channel Kv1.5, which participates in the regulation of diverse functions including atrial cardiac action potential and tumor cell proliferation. To this end, cRNA encoding Kv1.5 was injected into Xenopus oocytes with or without additional injection of cRNA encoding wild-type JAK3, constitutively active (A568V)JAK3, or inactive (K851A)JAK3. Voltage-gated K(+) channel activity was measured utilizing dual electrode voltage clamp, and Kv1.5 channel protein abundance in the cell membrane was quantified utilizing chemiluminescence of Kv1.5 containing an extracellular hemagglutinin epitope (Kv1.5-HA). As a result, Kv1.5 activity and Kv1.5-HA protein abundance were significantly decreased by wild-type JAK3 and (A568V)JAK3, but not by (K851A)JAK3. Inhibition of Kv1.5 protein insertion into the cell membrane by brefeldin A (5 µM) resulted in a decline of the voltage-gated current, which was similar in the absence and presence of (A568V)JAK3, suggesting that (A568V)JAK3 did not accelerate Kv1.5 protein retrieval from the cell membrane. A 24 h treatment with ouabain (100 µM) significantly decreased the voltage-gated current in oocytes expressing Kv1.5 without or with (A568V)JAK3 and dissipated the difference between oocytes expressing Kv1.5 alone and oocytes expressing Kv1.5 with (A568V)JAK3. In conclusion, JAK3 contributes to the regulation of membrane Kv1.5 protein abundance and activity, an effect sensitive to ouabain and thus possibly involving Na(+)/K(+) ATPase activity.


Assuntos
Janus Quinase 3/metabolismo , Canal de Potássio Kv1.5/metabolismo , Animais , Brefeldina A/farmacologia , Membrana Celular/metabolismo , Expressão Gênica , Janus Quinase 3/genética , Canal de Potássio Kv1.2/genética , Canal de Potássio Kv1.2/metabolismo , Canal de Potássio Kv1.5/genética , Camundongos , Oócitos/metabolismo , Ouabaína/farmacologia , Ligação Proteica , Ratos , Xenopus
12.
J Membr Biol ; 248(6): 1107-19, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26233565

RESUMO

SPAK (SPS1-related proline/alanine-rich kinase) and OSR1 (oxidative stress-responsive kinase 1) are cell volume-sensitive kinases regulated by WNK (with-no-K[Lys]) kinases. SPAK/OSR1 regulate several channels and carriers. SPAK/OSR1 sensitive functions include neuronal excitability. Orchestration of neuronal excitation involves the excitatory glutamate transporters EAAT1 and EAAT2. Sensitivity of those carriers to SPAK/OSR1 has never been shown. The present study thus explored whether SPAK and/or OSR1 contribute to the regulation of EAAT1 and/or EAAT2. To this end, cRNA encoding EAAT1 or EAAT2 was injected into Xenopus oocytes without or with additional injection of cRNA encoding wild-type SPAK or wild-type OSR1, constitutively active (T233E)SPAK, WNK insensitive (T233A)SPAK, catalytically inactive (D212A)SPAK, constitutively active (T185E)OSR1, WNK insensitive (T185A)OSR1 or catalytically inactive (D164A)OSR1. The glutamate (2 mM)-induced inward current (I Glu) was taken as a measure of glutamate transport. As a result, I Glu was observed in EAAT1- and in EAAT2-expressing oocytes but not in water-injected oocytes, and was significantly decreased by coexpression of SPAK and OSR1. As shown for EAAT2, SPAK, and OSR1 decreased significantly the maximal transport rate but significantly enhanced the affinity of the carrier. The effect of wild-type SPAK/OSR1 on EAAT1 and EAAT2 was mimicked by (T233E)SPAK and (T185E)OSR1, but not by (T233A)SPAK, (D212A)SPAK, (T185A)OSR1, or (D164A)OSR1. Coexpression of either SPAK or OSR1 decreased the EAAT2 protein abundance in the cell membrane of EAAT2-expressing oocytes. In conclusion, SPAK and OSR1 are powerful negative regulators of the excitatory glutamate transporters EAAT1 and EAAT2.


Assuntos
Transportador 1 de Aminoácido Excitatório/metabolismo , Transportador 2 de Aminoácido Excitatório/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Transporte Biológico , Membrana Celular/metabolismo , Regulação para Baixo , Transportador 1 de Aminoácido Excitatório/genética , Transportador 2 de Aminoácido Excitatório/genética , Expressão Gênica , Ácido Glutâmico/metabolismo , Humanos , Mutação , Oócitos/metabolismo , Técnicas de Patch-Clamp , Proteínas Serina-Treonina Quinases/genética , Xenopus laevis
13.
J Membr Biol ; 248(2): 309-17, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25644777

RESUMO

The janus-activated kinase 2 JAK2 participates in the signalling of several hormones including interferon, a powerful regulator of lymphocyte function. Lymphocyte activity and survival depend on the activity of the voltage-gated K(+) channel KCNA3 (Kv1.3). The present study thus explored whether JAK2 modifies the activity of voltage-gated K(+) channel KCNA3. To this end, cRNA encoding KCNA3 was injected in Xenopus oocytes with or without additional injection of cRNA encoding wild-type human JAK2, human inactive (K882E)JAK2 mutant, or human gain-of-function (V617F)JAK2 mutant. KCNA3-dependent depolarization-induced current was determined utilizing dual-electrode voltage clamp, and protein KCNA3 abundance in the cell membrane was quantified by chemiluminescence. Moreover, the effect of interferon-γ on voltage-gated K(+) current was determined by patch clamp in mainly KCNA3-expressing Jurkat T cells with or without prior treatment with JAK2 inhibitor AG490 (40 µM). As a result, KCNA3 channel activity and protein abundance were up-regulated by coexpression of JAK2 or (V617F)JAK2 but not (K882E)JAK2. The effect of JAK2 coexpression was reversed by AG490 treatment. In human Jurkat T lymphoma cells, voltage-gated K(+) current was up-regulated by interferon-γ and down-regulated by AG490 (40 µM). In conclusion, JAK2 participates in the signalling, regulating the voltage-gated K(+) channel KCNA3.


Assuntos
Janus Quinase 2/metabolismo , Canal de Potássio Kv1.3/metabolismo , Animais , Membrana Celular/metabolismo , Células Cultivadas , Expressão Gênica , Humanos , Janus Quinase 2/genética , Canal de Potássio Kv1.3/genética , Potenciais da Membrana , Camundongos , Mutação , Oócitos/metabolismo , Regulação para Cima , Xenopus
14.
J Membr Biol ; 248(2): 223-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25487255

RESUMO

Parvovirus B19 (B19V) has previously been shown to cause endothelial dysfunction. B19V capsid protein VP1 harbors a lysophosphatidylcholine producing phospholipase A2 (PLA2). Lysophosphatidylcholine inhibits Na(+)/K(+) ATPase, which in turn may impact on the activity of inwardly rectifying K(+) channels. The present study explored whether VP1 modifies the activity of Kir2.1 K(+) channels. cRNA encoding Kir2.1 was injected into Xenopus oocytes without or with cRNA encoding VP1 isolated from a patient suffering from fatal B19V-induced inflammatory cardiomyopathy or the VP1 mutant (H153A)VP1 lacking a functional PLA2 activity. K(+) channel activity was determined by dual electrode voltage clamp. In addition, Na(+)/K(+)-ATPase activity was estimated from K(+)-induced pump current (I(pump)) and ouabain-inhibited current (I(ouabain)). Injection of cRNA encoding Kir2.1 into Xenopus oocytes was followed by appearance of inwardly rectifying K(+) channel activity (I(K)), which was significantly decreased by additional injection of cRNA encoding VP1, but not by additional injection of cRNA encoding (H153A)VP1. The effect of VP1 on I K was mimicked by lysophosphatidylcholine (1 µg/ml) and by inhibition of Na(+)/K(+)-ATPase with 0.1 mM ouabain. In the presence of lysophosphatidylcholine, I K was not further decreased by additional treatment with ouabain. The B19V capsid protein VP1 thus inhibits Kir2.1 channels, an effect at least partially due to PLA2-dependent formation of lysophosphatidylcholine with subsequent inhibition of Na(+)/K(+)-ATPase activity.


Assuntos
Proteínas do Capsídeo/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Animais , Proteínas do Capsídeo/genética , Regulação para Baixo , Expressão Gênica , Humanos , Potenciais da Membrana , Oócitos/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Transfecção , Xenopus
15.
Neurosignals ; 23(1): 20-33, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26673921

RESUMO

BACKGROUND/AIMS: Kir2.1 (KCNJ2) channels are expressed in neurons, skeletal muscle and cardiac tissue and maintain the resting membrane potential. The activity of those channels is regulated by diverse signalling molecules. The present study explored whether Kir2.1 channels are sensitive to the transporter and channels regulating kinases SPAK (SPS1-related proline/alanine-rich kinase) and OSR1 (oxidative stress-responsive kinase 1), which are in turn regulated by WNK (with-no-K[Lys]) kinases. METHODS: cRNA encoding Kir2.1 was injected into Xenopus laevis oocytes with or without additional injection of cRNA encoding wild-type SPAK, constitutively active T233E SPAK, WNK insensitive T233A SPAK, catalytically inactive D212A SPAK, wild-type OSR1, constitutively active T185E OSR1, WNK insensitive T185A OSR1 and catalytically inactive D164A OSR1. Inwardly rectifying K+ channel activity was quantified utilizing dual electrode voltage clamp and Kir2.1 channel protein abundance in the cell membrane was measured utilizing chemiluminescence of Kir2.1 containing an extracellular HA-tag epitope. RESULTS: Kir2.1 activity was significantly enhanced by wild-type SPAK and T233E SPAK, but not by T233A SPAK and D212A SPAK, as well as by wild-type OSR1 and T185E OSR1, but not by T185A OSR1 and D164A OSR1. As shown for SPAK, the kinases enhanced Kir2.1 protein abundance in the cell membrane. The difference of current and conductance between oocytes expressing Kir2.1 together with SPAK or OSR1 and oocytes expressing Kir2.1 alone was dissipated following a 24 hours inhibition of channel insertion into the cell membrane by brefeldin A (5 µM). CONCLUSIONS: SPAK and OSR1 are both stimulators of Kir2.1 activity. They are presumably effective by enhancing channel insertion into the cell membrane.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Fenômenos Biofísicos/efeitos dos fármacos , Fenômenos Biofísicos/genética , Brefeldina A/farmacologia , Relação Dose-Resposta a Droga , Estimulação Elétrica , Humanos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Microinjeções , Mutação/genética , Oócitos , Técnicas de Patch-Clamp , Canais de Potássio Corretores do Fluxo de Internalização/genética , Proteínas Serina-Treonina Quinases/genética , Inibidores da Síntese de Proteínas/farmacologia , Xenopus laevis
16.
Kidney Blood Press Res ; 40(6): 555-64, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26506223

RESUMO

BACKGROUND/AIMS: SPAK (SPS1-related proline/alanine-rich kinase) and OSR1 (oxidative stress-responsive kinase 1), kinases controlled by WNK (with-no-K[Lys] kinase), are powerful regulators of cellular ion transport and blood pressure. Observations in gene-targeted mice disclosed an impact of SPAK/OSR1 on phosphate metabolism. The present study thus tested whether SPAK and/or OSR1 contributes to the regulation of the intestinal Na(+)-coupled phosphate co-transporter NaPi-IIb (SLC34A2). METHODS: cRNA encoding NaPi-IIb was injected into Xenopus laevis oocytes without or with additional injection of cRNA encoding wild-type SPAK, constitutively active (T233E)SPAK, WNK insensitive (T233A)SPAK, catalytically inactive (D212A)SPAK, wild-type OSR1, constitutively active (T185E)OSR1, WNK insensitive (T185A)OSR1 or catalytically inactive (D164A)OSR1. The phosphate (1 mM)-induced inward current (I(Pi)) was taken as measure of phosphate transport. RESULTS: I(Pi) was observed in NaPi-IIb expressing oocytes but not in water injected oocytes, and was significantly increased by co-expression of SPAK, (T233E)SPAK, OSR1, (T185E)OSR1 or SPAK+OSR1, but not by co-expression of (T233A)SPAK, (D212A)SPAK, (T185A)OSR1, or (D164A)OSR1. SPAK and OSR1 both increased the maximal transport rate of the carrier. CONCLUSIONS: SPAK and OSR1 are powerful stimulators of the intestinal Na+-coupled phosphate co-transporter NaPi-IIb.


Assuntos
Mucosa Intestinal/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/biossíntese , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/genética , Animais , Humanos , Camundongos , Microinjeções , Oócitos , Técnicas de Patch-Clamp , Fosfatos/metabolismo , RNA/administração & dosagem , RNA/genética , Regulação para Cima , Xenopus laevis
17.
Kidney Blood Press Res ; 40(4): 335-43, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26160150

RESUMO

BACKGROUND/AIMS: The WNK-dependent STE20/SPS1-related proline/alanine-rich kinase SPAK participates in the regulation of NaCl and Na(+),K(+),2Cl(-) cotransport and thus renal salt excretion. The present study explored whether SPAK has similarly the potential to regulate the epithelial Na(+) channel (ENaC). METHODS: ENaC was expressed in Xenopus oocytes with or without additional expression of wild type SPAK, constitutively active (T233E)SPAK, WNK insensitive (T233A)SPAK or catalytically inactive (D212A)SPAK, and ENaC activity estimated from amiloride (50 µM) sensitive current (Iamil) in dual electrode voltage clamp experiments. Moreover, Ussing chamber was employed to determine Iamil in colonic tissue from wild type mice (spak(wt/wt)) and from gene targeted mice carrying WNK insensitive SPAK (spak(tg/tg)). RESULTS: Iamil was observed in ENaC-expressing oocytes, but not in water-injected oocytes. In ENaC expressing oocytes Iamil was significantly increased following coexpression of wild-type SPAK and (T233E)SPAK, but not following coexpression of (T233A)SPAK or (D212A)SPAK. Colonic Iamil was significantly higher in spak(wt/wt) than in spak(tg/tg) mice. CONCLUSION: SPAK has the potential to up-regulate ENaC.


Assuntos
Canais Epiteliais de Sódio/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Aldosterona/sangue , Amilorida/farmacologia , Animais , Colo/metabolismo , Diuréticos/farmacologia , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Canais Epiteliais de Sódio/genética , Feminino , Camundongos , Antígenos de Histocompatibilidade Menor , Ubiquitina-Proteína Ligases Nedd4 , Oócitos/metabolismo , Técnicas de Patch-Clamp , Proteínas Serina-Treonina Quinases/genética , Cloreto de Sódio/metabolismo , Cloreto de Sódio/urina , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Regulação para Cima/fisiologia , Proteína Quinase 1 Deficiente de Lisina WNK , Xenopus laevis
18.
Biochim Biophys Acta ; 1828(11): 2394-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23774524

RESUMO

The Na(+)-coupled glucose transporter SGLT1 (SLC5A1) accomplishes concentrative cellular glucose uptake even at low extracellular glucose concentrations. The carrier is expressed in renal proximal tubules, small intestine and a variety of nonpolarized cells including several tumor cells. The present study explored whether SGLT1 activity is regulated by caveolin-1, which is known to regulate the insertion of several ion channels and carriers in the cell membrane. To this end, SGLT1 was expressed in Xenopus oocytes with or without additional expression of caveolin-1 and electrogenic glucose transport determined by dual electrode voltage clamp experiments. In SGLT1-expressing oocytes, but not in oocytes injected with water or caveolin-1 alone, the addition of glucose to the extracellular bath generated an inward current (Ig), which was increased following coexpression of caveolin-1. Kinetic analysis revealed that caveolin-1 increased maximal Ig without significantly modifying the glucose concentration required to trigger half maximal Ig (KM). According to chemiluminescence and confocal microscopy, caveolin-1 increased SGLT1 protein abundance in the cell membrane. Inhibition of SGLT1 insertion by brefeldin A (5µM) resulted in a decline of Ig, which was similar in the absence and presence of caveolin-1. In conclusion, caveolin-1 up-regulates SGLT1 activity by increasing carrier protein abundance in the cell membrane, an effect presumably due to stimulation of carrier protein insertion into the cell membrane.


Assuntos
Caveolina 1/fisiologia , Transportador 1 de Glucose-Sódio/fisiologia , Regulação para Cima/fisiologia , Animais , Membrana Celular/metabolismo , Cinética , Transportador 1 de Glucose-Sódio/metabolismo , Xenopus
19.
Cell Physiol Biochem ; 33(2): 491-500, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24556932

RESUMO

BACKGROUND/AIMS: The serum & glucocorticoid inducible kinase SGK3, an ubiquitously expressed serine/threonine kinase, regulates a variety of ion channels. It has previously been shown that SGK3 upregulates the outwardly rectifying K(+) channel KV11.1, which is expressed in cardiomyocytes. Cardiomyocytes further express the inward rectifier K(+) channel K(ir)2.1, which contributes to maintenance of resting cell membrane potential. Loss-of-function mutations of KCNJ2 encoding K(ir)2.1 result in Andersen-Tawil syndrome with periodic paralysis, cardiac arrhythmia and dysmorphic features. The present study explored whether SGK3 participates in the regulation of K(ir)2.1. METHODS: cRNA encoding K(ir)2.1 was injected into Xenopus oocytes with and without additional injection of cRNA encoding wild type SGK3, constitutively active (S419D)SGK3 or inactive (K191N)SGK3. Kir2.1 activity was determined by two-electrode voltage-clamp and K(ir)2.1 protein abundance in the cell membrane by immunostaining and subsequent confocal imaging or by chemiluminescence. RESULTS: Injection of 10 ng cRNA encoding wild type SGK3 and (S419D)SGK3, but not (K191N)SGK3 significantly enhanced K(ir)2.1-mediated currents. SGK inhibitor EMD638683 (50 µM) abrogated (S419D)SGK3-induced up-regulation of K(ir)2.1. Moreover, wild type SGK3 enhanced the channel protein abundance in the cell membrane. The decay of K(ir)2.1-mediated currents following inhibition of channel insertion into the cell membrane by brefeldin A (5 µM) was similar in oocytes coexpressing K(ir)2.1 and SGK3 as in oocytes expressing K(ir)2.1 alone, suggesting that SGK3 influences channel insertion into rather than channel retrieval from the cell membrane. CONCLUSIONS: SGK3 is a novel regulator of K(ir)2.1.


Assuntos
Membrana Celular/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/biossíntese , Proteínas Serina-Treonina Quinases/biossíntese , Regulação para Cima/fisiologia , Animais , Antibacterianos/farmacologia , Brefeldina A/farmacologia , Membrana Celular/genética , Humanos , Oócitos , Canais de Potássio Corretores do Fluxo de Internalização/genética , Proteínas Serina-Treonina Quinases/genética , Regulação para Cima/efeitos dos fármacos , Xenopus laevis
20.
Biochem Biophys Res Commun ; 450(4): 1396-401, 2014 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-25010641

RESUMO

Parvovirus B19 (B19V) can cause inflammatory cardiomyopathy and endothelial dysfunction. Pathophysiological mechanisms involved include lysophosphatidylcholine producing phospholipase A2 (PLA2) activity of the B19V capsid protein VP1. Most recently, VP1 and lysophosphatidylcholine have been shown to inhibit Na(+)/K(+) ATPase. The present study explored whether VP1 modifies the activity of Kv1.3 and Kv1.5 K(+) channels. cRNA encoding Kv1.3 or Kv1.5 was injected into Xenopus oocytes without or with cRNA encoding VP1 isolated from a patient suffering from fatal B19V-induced myocarditis. K(+) channel activity was determined by dual electrode voltage clamp. Injection of cRNA encoding Kv1.3 or Kv1.5 into Xenopus oocytes was followed by appearance of Kv K(+) channel activity, which was significantly decreased by additional injection of cRNA encoding VP1, but not by additional injection of cRNA encoding PLA2-negative VP1 mutant (H153A). The effect of VP1 on Kv current was not significantly modified by transcription inhibitor actinomycin (10 µM for 36 h) but was mimicked by lysophosphatidylcholine (1 µg/ml). The B19V capsid protein VP1 inhibits host cell Kv channels, an effect at least partially due to phospholipase A2 (PLA) dependent formation of lysophosphatidylcholine.


Assuntos
Proteínas do Capsídeo/fisiologia , Regulação para Baixo , Parvovirus B19 Humano/metabolismo , Canais de Potássio/fisiologia , Animais , Humanos , Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA