Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Physiol ; 595(14): 4927-4946, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28383826

RESUMO

KEY POINTS: Afterhyperpolarizations (AHPs) generated by repetitive action potentials in supraoptic magnocellular neurons regulate repetitive firing and spike frequency adaptation but relatively little is known about PIP2 's control of these AHPs. We examined how changes in PIP2 levels affected AHPs, somatic [Ca2+ ]i , and whole cell Ca2+ currents. Manipulations of PIP2 levels affected both medium and slow AHP currents in oxytocin (OT) neurons of the supraoptic nucleus. Manipulations of PIP2 levels did not modulate AHPs by influencing Ca2+ release from IP3 -triggered Ca2+ stores, suggesting more direct modulation of channels by PIP2 . PIP2 depletion reduced spike-evoked Ca2+ entry and voltage-gated Ca2+ currents. PIP2 appears to influence AHPs in OT neurons by reducing Ca2+ influx during spiking. ABSTRACT: Oxytocin (OT)- and vasopressin (VP)-secreting magnocellular neurons of the supraoptic nucleus (SON) display calcium-dependent afterhyperpolarizations (AHPs) following a train of action potentials that are critical to shaping the firing patterns of these cells. Previous work demonstrated that the lipid phosphatidylinositol 4,5-bisphosphate (PIP2 ) enabled the slow AHP component (sAHP) in cortical pyramidal neurons. We investigated whether this phenomenon occurred in OT and VP neurons of the SON. Using whole cell recordings in coronal hypothalamic slices from adult female rats, we demonstrated that inhibition of PIP2 synthesis with wortmannin robustly blocked both the medium and slow AHP currents (ImAHP and IsAHP ) of OT, but not VP neurons with high affinity. We further tested this by introducing a water-soluble PIP2 analogue (diC8 -PIP2 ) into neurons, which in OT neurons not only prevented wortmannin's inhibitory effect, but slowed rundown of the ImAHP and IsAHP . Inhibition of phospholipase C (PLC) with U73122 did not inhibit either ImAHP or IsAHP in OT neurons, consistent with wortmannin's effects not being due to reducing diacylglycerol (DAG) or IP3 availability, i.e. PIP2 modulation of AHPs is not likely to involve downstream Ca2+ release from inositol 1,4,5-trisphosphate (IP3 )-triggered Ca2+ -store release, or channel modulation via DAG and protein kinase C (PKC). We found that wortmannin reduced [Ca2+ ]i increase induced by spike trains in OT neurons, but had no effect on AHPs evoked by uncaging intracellular Ca2+ . Finally, wortmannin selectively reduced whole cell Ca2+ currents in OT neurons while leaving VP neurons unaffected. The results indicate that PIP2 modulates both the ImAHP and IsAHP in OT neurons, most likely by controlling Ca2+ entry through voltage-gated Ca2+ channels opened during spike trains.


Assuntos
Neurônios/fisiologia , Fosfatidilinositol 4,5-Difosfato/fisiologia , Núcleo Supraóptico/fisiologia , Potenciais de Ação/efeitos dos fármacos , Androstadienos/farmacologia , Animais , Feminino , Técnicas In Vitro , Neurônios/efeitos dos fármacos , Ocitocina/fisiologia , Fosfatidilinositol 4,5-Difosfato/antagonistas & inibidores , Ratos Sprague-Dawley , Núcleo Supraóptico/efeitos dos fármacos , Wortmanina
2.
Nat Commun ; 6: 6211, 2015 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-25648615

RESUMO

Most macrophages remain uninfected in HIV-1-infected patients. Nevertheless, the phagocytic capacity of phagocytes from these patients is impaired, favouring the multiplication of opportunistic pathogens. The basis for this phagocytic defect is not known. HIV-1 Tat protein is efficiently secreted by infected cells. Secreted Tat can enter uninfected cells and reach their cytosol. Here we found that extracellular Tat, at the subnanomolar concentration present in the sera of HIV-1-infected patients, inhibits the phagocytosis of Mycobacterium avium or opsonized Toxoplasma gondii by human primary macrophages. This inhibition results from a defect in mannose- and Fcγ-receptor-mediated phagocytosis, respectively. Inhibition relies on the interaction of Tat with phosphatidylinositol (4,5)bisphosphate that interferes with the recruitment of Cdc42 to the phagocytic cup, thereby preventing Cdc42 activation and pseudopod elongation. Tat also inhibits FcγR-mediated phagocytosis in neutrophils and monocytes. This study provides a molecular basis for the phagocytic defects observed in uninfected phagocytes following HIV-1 infection.


Assuntos
HIV-1/fisiologia , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Fagocitose/efeitos dos fármacos , Produtos do Gene tat do Vírus da Imunodeficiência Humana/farmacologia , Efeito Espectador , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Lectinas Tipo C/antagonistas & inibidores , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Macrófagos/metabolismo , Macrófagos/ultraestrutura , Receptor de Manose , Lectinas de Ligação a Manose/antagonistas & inibidores , Lectinas de Ligação a Manose/genética , Lectinas de Ligação a Manose/metabolismo , Monócitos/metabolismo , Monócitos/ultraestrutura , Mycobacterium avium/crescimento & desenvolvimento , Neutrófilos/metabolismo , Neutrófilos/ultraestrutura , Fosfatidilinositol 4,5-Difosfato/antagonistas & inibidores , Fosfatidilinositol 4,5-Difosfato/metabolismo , Cultura Primária de Células , Transporte Proteico/efeitos dos fármacos , Pseudópodes/efeitos dos fármacos , Pseudópodes/metabolismo , Pseudópodes/ultraestrutura , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores de IgG/antagonistas & inibidores , Receptores de IgG/genética , Receptores de IgG/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Toxoplasma/crescimento & desenvolvimento , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/biossíntese
3.
Mol Pharmacol ; 83(4): 793-804, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23313938

RESUMO

Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) is a versatile phospholipid that participates in many membrane-associated signaling processes. PI(4,5)P2 production at the plasma membrane (PM) depends on levels of its precursor, phosphatidylinositol 4-phosphate (PI4P), synthesized principally by two intracellular enzymes, PI4-kinases IIIα and IIIb; the former is preferentially inhibited by phenylarsine oxide (PAO). We found that PAO and quercetin, another lipid kinase inhibitor, rapidly inhibit Ca(2+) responses to antigen in IgE-sensitized rat basophilic leukemia mast cells. Quercetin also rapidly inhibits store-operated Ca(2+) influx stimulated by thapsigargin. In addition, quercetin and PAO effectively inhibit antigen-stimulated ruffling and spreading in these cells, and they inhibit endocytosis of crosslinked IgE receptor complexes, evidently by inhibiting pinching off of endocytic vesicles containing the clustered IgE receptors. A minimal model to account for these diverse effects is inhibition of PI(4,5)P2 synthesis by PAO and quercetin. To characterize the direct effects of these agents on PI(4,5)P2 synthesis, we monitored the reappearance of the PI(4,5)P2-specific PH domain PH-phospholipase C δ-EGFP at the PM after Ca(2+) ionophore (A23187)-induced PI(4,5)P2 hydrolysis, followed by Ca(2+) chelation with excess EGTA. Resynthesized PI(4,5)P2 initially appears as micron-sized patches near the PM. Addition of quercetin subsequent to A23187-induced PI(4,5)P2 hydrolysis reduces PI(4,5)P2 resynthesis in PM-associated patches, and PAO reduces PI(4,5)P2 at the PM while enhancing PI(4,5)P2 accumulation at the Golgi complex. Taken together, these results provide evidence that PI4P generated by PI4-kinase IIIα is dynamically coupled to PI(4,5)P2 pools at the PM that are important for downstream signaling processes activated by IgE receptors.


Assuntos
Mastócitos/metabolismo , Fosfatidilinositol 4,5-Difosfato/antagonistas & inibidores , Fosfatidilinositol 4,5-Difosfato/biossíntese , Receptores de IgE/fisiologia , Transdução de Sinais/fisiologia , Animais , Arsenicais/farmacologia , Linhagem Celular Tumoral , Mastócitos/efeitos dos fármacos , Mastócitos/fisiologia , Fosfatidilinositóis/antagonistas & inibidores , Fosfatidilinositóis/biossíntese , Quercetina/farmacologia , Ratos , Receptores de IgE/metabolismo , Transdução de Sinais/efeitos dos fármacos
4.
Science ; 337(6095): 727-30, 2012 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-22722250

RESUMO

The quantitatively minor phospholipid phosphatidylinositol (4,5)-bisphosphate [PI(4,5)P(2)] fulfills many cellular functions in the plasma membrane (PM), whereas its synthetic precursor, phosphatidylinositol 4-phosphate (PI4P), has no assigned PM roles apart from PI(4,5)P(2) synthesis. We used a combination of pharmacological and chemical genetic approaches to probe the function of PM PI4P, most of which was not required for the synthesis or functions of PI(4,5)P(2). However, depletion of both lipids was required to prevent PM targeting of proteins that interact with acidic lipids or activation of the transient receptor potential vanilloid 1 cation channel. Therefore, PI4P contributes to the pool of polyanionic lipids that define plasma membrane identity and to some functions previously attributed specifically to PI(4,5)P(2), which may be fulfilled by a more general polyanionic lipid requirement.


Assuntos
Membrana Celular/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Animais , Células COS , Chlorocebus aethiops , Endocitose , Células HEK293 , Humanos , Proteínas de Membrana/metabolismo , Fragmentos de Peptídeos/metabolismo , Fosfatidilinositol 4,5-Difosfato/antagonistas & inibidores , Fosfatidilinositol 4,5-Difosfato/biossíntese , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo , Polieletrólitos , Polímeros , Receptor Muscarínico M1/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Transdução de Sinais , Eletricidade Estática , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/metabolismo
5.
Hypertens Res ; 35(9): 909-16, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22592664

RESUMO

Sympathetic nerves regulate vascular tone by releasing neurotransmitters into the vasculature. We previously demonstrated that bradykinin facilitates sympathetic neurotransmission in rat mesenteric arteries. Although little is known about the intracellular mechanism modulating this neurotransmission, recent cell line experiments have shown that the KCNQ channel, which is inhibited by the depletion of membrane phosphatidylinositol-4,5-bisphosphate (PIP2), participates in the control of neurotransmission by bradykinin. In the present study, we examined the mechanism regulating neurotransmitter release from rat perivascular sympathetic nerves. Excitatory junction potentials (EJPs) elicited by repetitive nerve stimulation (1 Hz, 11 pulses, 20 µs, 20-50 V), a measure of sympathetic purinergic neurotransmission, were recorded with a conventional microelectrode technique in rat mesenteric arteries. Bradykinin (10⁻7 mol l⁻¹) significantly enhanced the amplitude of EJPs (n=22, P<0.05). This enhancing effect was abolished by N-type calcium-channel inhibition with ω-conotoxin GVIA (2 × 10⁻9 mol ⁻¹l, n=8). The blockade of phospholipase C with U-73122 (10(-6) mol l⁻¹, n=17) also eliminated the facilitatory effect of bradykinin. In addition, the effects of bradykinin were diminished by the prevention of PIP2 resynthesis with wortmannin (10⁻5 mol l⁻¹ n=7) or KCNQ channel inhibition with XE-991 (10⁻5 mol l⁻¹, n=7). On the other hand, depletion of intracellular calcium stores with cyclopiazonic acid (3 × 10⁻6 mol l⁻¹, n=6) or the inhibition of protein kinase C with bisindolylmaleimide-I (10⁻6 mol l⁻¹, n=9) did not alter the action of bradykinin. These data demonstrate that the hydrolysis of PIP2 by phospholipase C, which is activated by G(q/11)-coupled receptors, and subsequent KCNQ channel inhibition enhance sympathetic purinergic neurotransmission presumably via the activation of N-type calcium channels in rat mesenteric arteries.


Assuntos
Canais de Potássio KCNQ/fisiologia , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/inervação , Fosfatidilinositol 4,5-Difosfato/fisiologia , Sistema Nervoso Simpático/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Androstadienos/farmacologia , Animais , Bradicinina/farmacologia , Cálcio/fisiologia , Bloqueadores dos Canais de Cálcio/farmacologia , Estimulação Elétrica , Estrenos/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Canais de Potássio KCNQ/antagonistas & inibidores , Canais de Potássio KCNQ/efeitos dos fármacos , Masculino , Potenciais da Membrana/efeitos dos fármacos , Microeletrodos , Junção Neuromuscular/efeitos dos fármacos , Fosfatidilinositol 4,5-Difosfato/antagonistas & inibidores , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Pirrolidinonas/farmacologia , Ratos , Ratos Wistar , Receptores Purinérgicos/fisiologia , Fosfolipases Tipo C/antagonistas & inibidores , Wortmanina , ômega-Conotoxina GVIA/farmacologia
6.
J Neurosci ; 31(38): 13596-603, 2011 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-21940450

RESUMO

In the retina, light onset hyperpolarizes photoreceptors and depolarizes ON bipolar cells at the sign inverting photoreceptor-ON bipolar cell synapse. Transmission at this synapse is mediated by a signaling cascade comprised of mGluR6, a G-protein containing G(αo), and the cation channel TRP melastatin 1 (TRPM1). This system is thought to be common to both the rod- and ON-cone-driven pathways, which control vision under scotopic and photopic conditions, respectively. In this study, we present evidence that the rod pathway is uniquely susceptible to modulation by PKCα at the rod-rod bipolar cell synapse. Decreased production of DAG (an activator of PKC) by inhibition of PIP2 (phosphatidylinositol-4,5-bisphosphate) hydrolysis caused depression of the TRPM1 current. Conversely, addition of a DAG analog, 2-acetyl-1-oleoyl-sn-glycerol (OAG), potentiated the current in rod bipolar cells but not in ON-cone bipolar cells. The potentiating effects of OAG were absent both in mutant mice that lack PKCα expression and in wild-type mice in which enzymatic activity of PKCα was pharmacologically inhibited. In addition, we found that, like other members of the TRPM subfamily, TRPM1 current is susceptible to voltage-independent inhibition by intracellular magnesium, and that modulation by PKCα relieves this inhibition, as the potentiating effects of OAG are absent in low intracellular magnesium. We conclude that activation of PKCα initiates a modulatory mechanism at the rod-rod bipolar cell synapse whose function is to reduce inhibition of the TRPM1 current by magnesium, thereby increasing the gain of transmission at this synapse.


Assuntos
Magnésio/fisiologia , Proteína Quinase C-alfa/fisiologia , Células Bipolares da Retina/fisiologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Sinapses/fisiologia , Canais de Cátion TRPM/fisiologia , Animais , Diglicerídeos/farmacologia , Inibidores Enzimáticos/farmacologia , Feminino , Técnicas In Vitro , Magnésio/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Mutantes , Fosfatidilinositol 4,5-Difosfato/antagonistas & inibidores , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfolipase D/antagonistas & inibidores , Proteína Quinase C-alfa/antagonistas & inibidores , Proteína Quinase C-alfa/biossíntese , Proteína Quinase C-alfa/genética , Células Bipolares da Retina/efeitos dos fármacos , Células Bipolares da Retina/metabolismo , Células Fotorreceptoras Retinianas Cones/efeitos dos fármacos , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/efeitos dos fármacos , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Sinapses/metabolismo , Canais de Cátion TRPM/antagonistas & inibidores
7.
Am J Physiol Cell Physiol ; 301(3): C619-29, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21613605

RESUMO

Bovine adrenocortical cells express bTREK-1 K(+) (bovine KCNK2) channels that are inhibited by ANG II through a Gq-coupled receptor by separate Ca(2+) and ATP hydrolysis-dependent signaling pathways. Whole cell and single patch clamp recording from adrenal zona fasciculata (AZF) cells were used to characterize Ca(2+)-dependent inhibition of bTREK-1. In whole cell recordings with pipette solutions containing 0.5 mM EGTA and no ATP, the Ca(2+) ionophore ionomycin (1 µM) produced a transient inhibition of bTREK-1 that reversed spontaneously within minutes. At higher concentrations, ionomycin (5-10 µM) produced a sustained inhibition of bTREK-1 that was reversible upon washing, even in the absence of hydrolyzable [ATP](i). BAPTA was much more effective than EGTA at suppressing bTREK-1 inhibition by ANG II. When intracellular Ca(2+) concentration ([Ca(2+)](i)) was buffered to 20 nM with either 11 mM BAPTA or EGTA, ANG II (10 nM) inhibited bTREK-1 by 12.0 ± 4.5% (n=11) and 59.3 ± 8.4% (n=4), respectively. Inclusion of the water-soluble phosphatidylinositol 4,5-bisphosphate (PIP(2)) analog DiC(8)PI(4,5)P(2) in the pipette failed to increase bTREK-1 expression or reduce its inhibition by ANG II. The open probability (P(o)) of unitary bTREK-1 channels recorded from inside-out patches was reduced by Ca(2+) (10-35 µM) in a concentration-dependent manner. These results are consistent with a model in which ANG II inhibits bTREK-1 K(+) channels by a Ca(2+)-dependent mechanism that does not require the depletion of membrane-associated PIP(2). They further indicate that the Ca(2+) source is located in close proximity within a "Ca(2+) nanodomain" of bTREK-1 channels, where [Ca(2+)](i) may reach concentrations of >10 µM. bTREK-1 is the first two-pore K(+) channel shown to be inhibited by Ca(2+) through activation of a G protein-coupled receptor.


Assuntos
Córtex Suprarrenal/citologia , Angiotensina II/farmacologia , Sinalização do Cálcio/fisiologia , Ativação do Canal Iônico/fisiologia , Ionomicina/farmacologia , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Adenilil Imidodifosfato/farmacologia , Animais , Soluções Tampão , Cálcio/metabolismo , Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Bovinos , Células Cultivadas , Ácido Egtázico/análogos & derivados , Ácido Egtázico/metabolismo , Ácido Egtázico/farmacologia , Estimulação Elétrica , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Fenômenos Eletrofisiológicos/fisiologia , Ativação do Canal Iônico/efeitos dos fármacos , Masculino , Técnicas de Patch-Clamp , Penfluridol/farmacologia , Fosfatidilinositol 4,5-Difosfato/análogos & derivados , Fosfatidilinositol 4,5-Difosfato/antagonistas & inibidores , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canais de Potássio de Domínios Poros em Tandem/antagonistas & inibidores , Uridina Trifosfato/metabolismo , Uridina Trifosfato/farmacologia , Zona Fasciculada/citologia
8.
J Physiol ; 589(Pt 13): 3149-62, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21540350

RESUMO

TASK channels are background K+ channels that contribute to the resting conductance in many neurons. A key feature of TASK channels is the reversible inhibition by Gq-coupled receptors, thereby mediating the dynamic regulation of neuronal activity by modulatory transmitters. The mechanism that mediates channel inhibition is not fully understood. While it is clear that activation of Gαq is required, the immediate signal for channel closure remains controversial. Experimental evidence pointed to either phospholipase C (PLC)-mediated depletion of phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2) as the cause for channel closure or to a direct inhibitory interaction of active Gαq with the channel. Here, we address the role of PI(4,5)P2 for G-protein-coupled receptor (GPCR)-mediated TASK inhibition by using recently developed genetically encoded tools to alter phosphoinositide (PI) concentrations in the living cell.When expressed in CHO cells, TASK-1- and TASK-3-mediated currents were not affected by depletion of plasma membrane PI(4,5)P2 either via the voltage-activated phosphatase Ci-VSP or via chemically triggered recruitment of a PI(4,5)P2-5'-phosphatase. Depletion of both PI(4,5)P2 and PI(4)P via membrane recruitment of a novel engineered dual-specificity phosphatase also did not inhibit TASK currents. In contrast, each of these methods produced robust inhibition of the bona fide PI(4,5)P2-dependent channel KCNQ4. Efficient depletion of PI(4,5)P2 and PI(4)P was further confirmed with a fluorescent phosphoinositide sensor. Moreover, TASK channels recovered normally from inhibition by co-expressed muscarinic M1 receptors when resynthesis of PI(4,5)P2 was prevented by depletion of cellular ATP. These results demonstrate that TASK channel activity is independent of phosphoinositide concentrations within the physiological range. Consequently, Gq-mediated inhibition of TASK channels is not mediated by depletion of PI(4,5)P2.


Assuntos
Proteínas do Tecido Nervoso/fisiologia , Fosfatidilinositol 4,5-Difosfato/fisiologia , Monoéster Fosfórico Hidrolases/fisiologia , Canais de Potássio de Domínios Poros em Tandem/fisiologia , Trifosfato de Adenosina/deficiência , Sequência de Aminoácidos , Animais , Células CHO , Cricetinae , Cricetulus , Genes de Troca , Humanos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/antagonistas & inibidores , Fosfatidilinositol 4,5-Difosfato/antagonistas & inibidores , Monoéster Fosfórico Hidrolases/antagonistas & inibidores , Canais de Potássio de Domínios Poros em Tandem/antagonistas & inibidores , Receptores Acoplados a Proteínas G/fisiologia
9.
J Neurosci ; 30(40): 13338-47, 2010 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-20926660

RESUMO

TRPV2 is a member of the transient receptor potential family of ion channels involved in chemical and thermal pain transduction. Unlike the related TRPV1 channel, TRPV2 does not appear to bind either calmodulin or ATP in its N-terminal ankyrin repeat domain. In addition, it does not contain a calmodulin-binding site in the distal C-terminal region, as has been proposed for TRPV1. We have found that TRPV2 channels transiently expressed in F-11 cells undergo Ca(2+)-dependent desensitization, similar to the other TRPVs, suggesting that the mechanism of desensitization may be conserved in the subfamily of TRPV channels. TRPV2 desensitization was not altered in whole-cell recordings in the presence of calmodulin inhibitors or on coexpression of mutant calmodulin but was sensitive to changes in membrane phosphatidylinositol 4,5-bisphosphate (PIP(2)), suggesting a role of membrane PIP(2) in TRPV2 desensitization. Simultaneous confocal imaging and electrophysiological recording of cells expressing TRPV2 and a fluorescent PIP(2)-binding probe demonstrated that TRPV2 desensitization was concomitant with depletion of PIP(2). We conclude that the decrease in PIP(2) levels on channel activation underlies a major component of Ca(2+)-dependent desensitization of TRPV2 and may play a similar role in other TRP channels.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/fisiologia , Membrana Celular/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Células Receptoras Sensoriais/metabolismo , Canais de Cátion TRPV/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Regulação para Baixo/fisiologia , Humanos , Hidrólise/efeitos dos fármacos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Microscopia Confocal , Técnicas de Patch-Clamp , Fosfatidilinositol 4,5-Difosfato/antagonistas & inibidores , Fosfatidilinositol 4,5-Difosfato/fisiologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Células Receptoras Sensoriais/efeitos dos fármacos , Canais de Cátion TRPV/antagonistas & inibidores
10.
J Cardiovasc Pharmacol ; 55(6): 555-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20179606

RESUMO

The aminosteroid 1-[6-({17beta-3-methoxyestra-1,3,5(10)-trien-17-yl}amino)hexyl]-1H-pyrrole-2,5-dione (U73122) has been extensively used as a pharmacologic inhibitor of phospholipase C (PLC). The inhibitory effect of U73122 on PLC activity is most likely the result of decreased availability of phosphatidylinositol 4,5-bisphosphate (PIP2), the substrate of the PLC signal transduction pathway, rather than direct inhibition of the enzyme. PIP2 is a phospholipid with pleiotropic cellular functions, including a pivotal role in regulating cardiac phospholipase D (PLD) signal transduction. Here, we hypothesized that U73122 acts as an inhibitor of cardiac PLD activity by interference with PIP2. U73122 concentration-dependently inhibited PLD activity in rat myocardial membranes. The inhibitory effect of U73122 was significantly attenuated when assayed on solubilized PLD activity and was completely restored if solubilized PLD activity was assayed in the presence of PIP2. U73122 had no inhibitory effect on purified PLD indicating that the substance does not interact with PLD directly. These data highlight a mechanism of action of U73122 as an inhibitor of myocardial PLD by interaction with PIP2 as a cofactor for optimal PLD activity. Hence, studies using U73122 as a specific inhibitor of PLC have to take into account that PLD may be involved in some of the effects ascribed to PLC.


Assuntos
Fosfatidilinositol 4,5-Difosfato/farmacologia , Fosfolipases Tipo C/antagonistas & inibidores , Animais , Bioensaio , Interações Medicamentosas , Estrenos , Masculino , Membranas/metabolismo , Miocárdio/metabolismo , Fosfatidilinositol 4,5-Difosfato/antagonistas & inibidores , Fosfatos de Fosfatidilinositol/antagonistas & inibidores , Fosfatos de Fosfatidilinositol/metabolismo , Fosfatidilinositóis/antagonistas & inibidores , Fosfolipase D/antagonistas & inibidores , Fosfolipase D/metabolismo , Pirrolidinonas , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Fosfolipases Tipo C/metabolismo
11.
Traffic ; 11(1): 123-37, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19845918

RESUMO

The role of PIP(2) in pancreatic beta cell function was examined here using the beta cell line MIN6B1. Blocking PIP(2) with PH-PLC-GFP or PIP5KIgamma RNAi did not impact on glucose-stimulated secretion although susceptibility to apoptosis was increased. Over-expression of PIP5KIgamma improved cell survival and inhibited secretion with accumulation of endocytic vacuoles containing F-actin, PIP(2), transferrin receptor, caveolin 1, Arf6 and the insulin granule membrane protein phogrin but not insulin. Expression of constitutively active Arf6 Q67L also resulted in vacuole formation and inhibition of secretion, which was reversed by PH-PLC-GFP co-expression. PIP(2) co-localized with gelsolin and F-actin, and gelsolin co-expression partially reversed the secretory defect of PIP5KIgamma-over-expressing cells. RhoA/ROCK inhibition increased actin depolymerization and secretion, which was prevented by over-expressing PIP5KIgamma, while blocking PIP(2) reduced constitutively active RhoA V14-induced F-actin polymerization. In conclusion, although PIP(2) plays a pro-survival role in MIN6B1 cells, excessive PIP(2) production because of PIP5KIgamma over-expression inhibits secretion because of both a defective Arf6/PIP5KIgamma-dependent endocytic recycling of secretory membrane and secretory membrane components such as phogrin and the RhoA/ROCK/PIP5KIgamma-dependent perturbation of F-actin cytoskeleton remodelling.


Assuntos
Apoptose , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Fosfatidilinositol 4,5-Difosfato/fisiologia , Fosfotransferases (Aceptor do Grupo Álcool)/biossíntese , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Técnicas de Cultura de Células , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Eletroforese em Gel de Poliacrilamida , Endocitose/efeitos dos fármacos , Glucose/farmacologia , Marcação In Situ das Extremidades Cortadas , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/enzimologia , Células Secretoras de Insulina/patologia , Camundongos , Microscopia Confocal , Microscopia de Fluorescência , Fosfatidilinositol 4,5-Difosfato/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Transporte Proteico , Vacúolos/efeitos dos fármacos , Vacúolos/enzimologia , Vacúolos/metabolismo
12.
Physiol Plant ; 138(3): 249-55, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19961548

RESUMO

The suggested link between intracellular cytokinin signaling and phospholipase D (PLD, EC 3.1.4.4.) activity (Romanov et al. 2000, 2002) was investigated. The activity of PLD in the early period of cytokinin action was studied in vivo in derooted Amaranthus caudatus seedlings, using the level of phosphatidylbutanol production as a measure of PLD activity. Rapid activation of phosphatidylbutanol synthesis was demonstrated as early as within 5 min of cytokinin administration. Neomycin, a known phosphatidylinositol-4,5-bisphosphate (PIP(2)) antagonist, strongly repressed both physiological cytokinin effect and cytokinin-dependent PLD activation. N-acylethanolamine (NAE 12), an inhibitor of alpha-class PLD, did not influence significantly cytokinin effect on Amaranthus seedlings. Together, results suggest the involvement of PIP(2)-dependent non-class alpha-PLD in the molecular mechanism of cytokinin action.


Assuntos
Amaranthus/enzimologia , Citocininas/farmacologia , Fosfolipase D/metabolismo , Etanolaminas/farmacologia , Glicerofosfolipídeos/biossíntese , Neomicina/farmacologia , Fosfatidilinositol 4,5-Difosfato/antagonistas & inibidores , Lectinas de Plantas/biossíntese , Proteínas Inativadoras de Ribossomos/biossíntese , Proteínas Inativadoras de Ribossomos Tipo 1 , Plântula/enzimologia
13.
J Physiol ; 587(Pt 22): 5361-75, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19770190

RESUMO

We investigate activation mechanisms of native TRPC1/C5/C6 channels (termed TRPC1 channels) by stimulation of endothelin-1 (ET-1) receptor subtypes in freshly dispersed rabbit coronary artery myocytes using single channel recording and immunoprecipitation techniques. ET-1 evoked non-selective cation channel currents with a unitary conductance of 2.6 pS which were not inhibited by either ET(A) or ET(B) receptor antagonists, respectively BQ-123 and BQ788, when administered separately. However, in the presence of both antagonists, ET-1-evoked channel activity was abolished indicating that both ET(A) and ET(B) receptor stimulation activate this conductance. Stimulation of both ET(A) and ET(B) receptors evoked channel activity which was inhibited by the protein kinase C (PKC) inhibitor chelerythrine and by anti-TRPC1 antibodies indicating that activation of both receptor subtypes causes TRPC1 channel activation by a PKC-dependent mechanism. ET(A) receptor-mediated TRPC1 channel activity was selectively inhibited by phosphoinositol-3-kinase (PI-3-kinase) inhibitors wortmannin (50 nM) and PI-828 and by antibodies raised against phosphoinositol-3,4,5-trisphosphate (PIP(3)), the product of PI-3-kinase-mediated phosphorylation of phosphatidylinositol 4,5-bisphosphate (PIP(2)). Moreover, exogenous application of diC8-PIP(3) stimulated PKC-dependent TRPC1 channel activity. These results indicate that stimulation of ET(A) receptors evokes PKC-dependent TRPC1 channel activity through activation of PI-3-kinase and generation of PIP(3). In contrast, ET(B) receptor-mediated TRPC1 channel activity was inhibited by the PI-phospholipase C (PI-PLC) inhibitor U73122. 1-Oleoyl-2-acetyl-sn-glycerol (OAG), an analogue of diacylglycerol (DAG), which is a product of PI-PLC, also activated PKC-dependent TRPC1 channel activity. OAG-induced TRPC1 channel activity was inhibited by anti-phosphoinositol-4,5-bisphosphate (PIP(2)) antibodies and high concentrations of wortmannin (20 microM) which depleted tissue PIP(2) levels. In addition exogenous application of diC8-PIP(2) activated PKC-dependent TRPC1 channel activity. These data indicate that stimulation of ET(B) receptors evokes PKC-dependent TRPC1 activity through PI-PLC-mediated generation of DAG and requires a permissive role of PIP(2). In conclusion, we provide the first evidence that stimulation of ET(A) and ET(B) receptors activate native PKC-dependent TRPC1 channels through two distinct phospholipids pathways involving a novel action of PIP(3), in addition to PIP(2), in rabbit coronary artery myocytes.


Assuntos
Fosfatidilinositol 4,5-Difosfato/fisiologia , Fosfatos de Fosfatidilinositol/fisiologia , Receptor de Endotelina A/fisiologia , Receptor de Endotelina B/fisiologia , Canais de Cátion TRPC/fisiologia , Animais , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/fisiologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Fosfatidilinositol 4,5-Difosfato/antagonistas & inibidores , Fosfatos de Fosfatidilinositol/antagonistas & inibidores , Inibidores de Fosfodiesterase/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Coelhos , Receptor de Endotelina A/agonistas , Receptor de Endotelina B/agonistas , Canais de Cátion TRPC/agonistas , Canal de Cátion TRPC6
14.
Mol Pharmacol ; 76(6): 1349-59, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19770351

RESUMO

The membrane phospholipid phosphatidylinositol (4,5)-bisphosphate (PIP(2)) has been implicated in the regulation of several ion channels and transporters. In this study, we examined the impact of PIP(2) on N-methyl-D-aspartate receptors (NMDARs) in cortical neurons. Blocking PIP(2) synthesis by inhibiting phosphoinositide-4 kinase, or stimulating PIP(2) hydrolysis via activation of phospholipase C (PLC), or blocking PIP(2) function with an antibody caused a significant reduction of NMDAR-mediated currents. On the other hand, inhibition of PLC or application of PIP(2) caused an enhancement of NMDAR currents. These electrophysiological effects were accompanied by changes in NMDAR surface clusters induced by agents that manipulate PIP(2) levels. The PIP(2) regulation of NMDAR currents was abolished by the dynamin inhibitory peptide, which blocks receptor internalization. Agents perturbing actin stability prevented PIP(2) regulation of NMDAR currents, suggesting the actin-dependence of this effect of PIP(2). Cofilin, a major actin depolymerizing factor, which has a common binding sequence for actin and PIP(2), was required for PIP(2) regulation of NMDAR currents. It is noteworthy that the PIP(2) regulation of NMDAR channels was impaired in a transgenic mouse model of Alzheimer's disease, probably because of the amyloid-beta disruption of PIP(2) metabolism. Taken together, our data suggest that continuous synthesis of PIP(2) at the membrane might be important for the maintenance of NMDARs at the cell surface. When PIP(2) is lost, cofilin is released from the PIP(2) complex and is rendered free to depolymerize actin. With the actin cytoskeleton no longer intact, NMDARs are internalized via a dynamin/clathrin-dependent mechanism, leading to reduced NMDAR currents.


Assuntos
Fosfatidilinositol 4,5-Difosfato/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Precursor de Proteína beta-Amiloide/biossíntese , Androstadienos/farmacologia , Animais , Carbacol/farmacologia , Células Cultivadas , Córtex Cerebral/metabolismo , Córtex Cerebral/fisiologia , Relação Dose-Resposta a Droga , Feminino , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Transgênicos , Agonistas Muscarínicos/farmacologia , Neurônios/metabolismo , Neurônios/fisiologia , Fosfatidilinositol 4,5-Difosfato/antagonistas & inibidores , Fosfatidilinositol 4,5-Difosfato/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Wortmanina
15.
Am J Physiol Cell Physiol ; 296(5): C1034-9, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19295171

RESUMO

One of the unanswered questions in muscle hypertrophy is how new contractile units are inserted into a stable existing cytoskeletal meshwork. Regulation of actin capping by CapZ may play a role in remodeling processes, therefore, CapZ dynamics are determined during rapid growth of cardiac cells in vitro. Neonatal rat ventricular myocytes were infected with adenovirus expressing green fluorescent protein-CapZ beta1 and responded normally to hypertrophic stimuli. CapZ dynamics were analyzed by fluorescence recovery after photobleaching in cultured myocytes treated with endothelin-1 (100 nM) or phenylephrine (10 muM). Recovery by 30 s was greater with endothelin treatment. Analysis 30 min postbleach showed CapZ-infected cells treated with endothelin recovered more completely than controls (77 +/- 9% vs. 50 +/- 6%, P < 0.001). Similar results were found with phenylephrine (77 +/- 5%, P < 0.05). A potential mechanism for phosphatidylinositol bisphosphate (PIP2) mediation of increased CapZ exchange in endothelin- and phenylephrine-treated cells was tested. PIP2 sequestration with neomycin (500 muM) blocked both endothelin- (43 +/- 6%, P < 0.001) and phenylephrine (36 +/- 4%, P < 0.001)-mediated recovery. The protein kinase C inhibitor chelerythrine chloride (10 muM) also blocked endothelin- (53 +/- 10%, P < 0.001) and phenylephrine (42 +/- 3%, P < 0.001)-mediated recovery. This study demonstrates for the first time that endothelin and phenylephrine alter CapZ dynamics through PIP2- and PKC-dependent pathways, which might destabilize the existing framework and permit sarcomeric remodelling to proceed.


Assuntos
Proteína de Capeamento de Actina CapZ/metabolismo , Endotelina-1/farmacologia , Miócitos Cardíacos/enzimologia , Fenilefrina/farmacologia , Fosfatidilinositol 4,5-Difosfato/metabolismo , Proteína Quinase C/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Benzofenantridinas/farmacologia , Proteína de Capeamento de Actina CapZ/genética , Cardiomegalia/metabolismo , Cardiotônicos/farmacologia , Células Cultivadas , Recuperação de Fluorescência Após Fotodegradação , Proteínas de Fluorescência Verde/genética , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Neomicina/farmacologia , Fosfatidilinositol 4,5-Difosfato/antagonistas & inibidores , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Proteína Quinase C/antagonistas & inibidores , Inibidores da Síntese de Proteínas/farmacologia , Ratos , Ratos Sprague-Dawley , Sarcômeros/enzimologia
16.
Proc Natl Acad Sci U S A ; 106(8): 2921-6, 2009 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-19193859

RESUMO

D-serine is a physiologic coagonist with glutamate at NMDA-subtype glutamate receptors. As D-serine is localized in glia, synaptically released glutamate presumably stimulates the glia to form and release D-serine, enabling glutamate/D-serine cotransmission. We show that serine racemase (SR), which generates D-serine from L-serine, is physiologically inhibited by phosphatidylinositol (4,5)-bisphosphate (PIP2) presence in membranes where SR is localized. Activation of metabotropic glutamate receptors (mGluR5) on glia leads to phospholipase C-mediated degradation of PIP2, relieving SR inhibition. Thus mutants of SR that cannot bind PIP2 lose their membrane localizations and display a 4-fold enhancement of catalytic activity. Moreover, mGluR5 activation of SR activity is abolished by inhibiting phospholipase C.


Assuntos
Ácido Glutâmico/metabolismo , Fosfatidilinositol 4,5-Difosfato/antagonistas & inibidores , Fosfatidilinositol 4,5-Difosfato/metabolismo , Racemases e Epimerases/metabolismo , Trifosfato de Adenosina/metabolismo , Ligação Competitiva , Linhagem Celular , Polarização de Fluorescência , Humanos , Imuno-Histoquímica , Ligação Proteica , Receptor de Glutamato Metabotrópico 5 , Receptores de Glutamato Metabotrópico/metabolismo
17.
Oncogene ; 28(8): 1076-88, 2009 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-19137008

RESUMO

Sproutys have been shown to negatively regulate growth factor-induced extracellular signal-regulated kinase (ERK) activation, and suggested to be an anti-oncogene. However, molecular mechanism of the suppression has not yet been clarified completely. Sprouty4 inhibits vascular endothelial growth factor (VEGF)-A-induced ERK activation, but not VEGF-C-induced ERK activation. It has been shown that VEGF-A-mediated ERK activation is strongly dependent on protein kinase C (PKC), whereas that by VEGF-C is dependent on Ras. This suggests that Sprouty4 inhibits the PKC pathway more specifically than the Ras pathway. In this study, we confirmed that Sprouty4 suppressed various signals downstream of PKC, such as phosphorylation of MARCKS and protein kinase D (PKD), as well as PKC-dependent nuclear factor (NF)-kappaB activation. Furthermore, Sprouty4 suppressed upstream signals of PKC, such as Ca(2+) mobilization, phosphatidylinositol 4,5-biphosphate (PIP(2)) breakdown and inositol 1,4,5-triphosphate (IP(3)) production in response to VEGF-A. Those effects were dependent on the C-terminal cysteine-rich region, but not on the N-terminal region of Sprouty4, which is critical for the suppression of fibroblast growth factor (FGF)-mediated ERK activation. Sprouty4 overexpression or deletion of the Sprouty4 gene did not affect phospholipase C (PLC) gamma-1 activation, which is an enzyme that catalyzes PIP(2) hydrolysis. Moreover, Sprouty4 inhibited not only VEGF-A-mediated PIP(2) hydrolysis but also inhibited the lysophosphatidic acid (LPA)-induced PIP(2) breakdown that is catalyzed by PLC beta/epsilon activated by G-protein coupled receptor (GPCR). Taken together, Sprouty4 has broader suppression activity for various stimuli than previously thought; it may function as an inhibitor for various types of PLC-dependent signaling as well as for ERK activation.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Fosfatidilinositol 4,5-Difosfato/antagonistas & inibidores , Proteína Quinase C/metabolismo , Animais , Cálcio/metabolismo , Células Cultivadas , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fatores de Crescimento de Fibroblastos/farmacologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Genes ras , Humanos , Hidrólise , Inositol 1,4,5-Trifosfato/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lisofosfolipídeos/farmacologia , Proteínas de Membrana/metabolismo , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Substrato Quinase C Rico em Alanina Miristoilada , NF-kappa B/genética , NF-kappa B/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatidilinositóis/metabolismo , Fosfoinositídeo Fosfolipase C/metabolismo , Fosfolipase C beta/metabolismo , Fosforilação , Proteína Quinase C/genética , Transporte Proteico , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
J Physiol ; 587(3): 531-40, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19047197

RESUMO

In the present study the effect of phosphatidylinositol 4,5-bisphosphate (PIP(2)) was studied on a native TRPC1 store-operated channel (SOC) in freshly dispersed rabbit portal vein myocytes. Application of diC8-PIP(2), a water soluble form of PIP(2), to quiescent inside-out patches evoked single channel currents with a unitary conductance of 1.9 pS. DiC8-PIP(2)-evoked channel currents were inhibited by anti-TRPC1 antibodies and these characteristics are identical to SOCs evoked by cyclopiazonic acid (CPA) and BAPTA-AM. SOCs stimulated by CPA, BAPTA-AM and the phorbol ester phorbol 12,13-dibutyrate (PDBu) were reduced by anti-PIP(2) antibodies and by depletion of tissue PIP(2) levels by pre-treatment of preparations with wortmannin and LY294002. However, these reagents did not alter the ability of PIP(2) to activate SOCs in inside-out patches. Co-immunoprecipitation techniques demonstrated association between TRPC1 and PIP(2) at rest, which was greatly decreased by wortmannin and LY294002. Pre-treatment of cells with PDBu, which activates protein kinase C (PKC), augmented SOC activation by PIP(2) whereas the PKC inhibitor chelerythrine decreased SOC stimulation by PIP(2). Co-immunoprecipitation experiments provide evidence that PKC-dependent phosphorylation of TRPC1 occurs constitutively and was increased by CPA and PDBu but decreased by chelerythrine. These novel results show that PIP(2) can activate TRPC1 SOCs in native vascular myocytes and plays an important role in SOC activation by CPA, BAPTA-AM and PDBu. Moreover, the permissive role of PIP(2) in SOC activation requires PKC-dependent phosphorylation of TRPC1.


Assuntos
Miócitos de Músculo Liso/metabolismo , Fosfatidilinositol 4,5-Difosfato/fisiologia , Canais de Cátion TRPC/metabolismo , 1-Fosfatidilinositol 4-Quinase/antagonistas & inibidores , 1-Fosfatidilinositol 4-Quinase/metabolismo , Androstadienos/farmacologia , Animais , Anticorpos Fosfo-Específicos/farmacologia , Benzofenantridinas/farmacologia , Quelantes/farmacologia , Cromonas/farmacologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Fatores Imunológicos/farmacologia , Imunoprecipitação , Indóis/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Morfolinas/farmacologia , Miócitos de Músculo Liso/efeitos dos fármacos , Fosfatidilinositol 4,5-Difosfato/antagonistas & inibidores , Veia Porta/citologia , Veia Porta/efeitos dos fármacos , Veia Porta/enzimologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Coelhos , Wortmanina
19.
J Physiol ; 587(2): 363-77, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19047199

RESUMO

Caffeine, a prototypic bitter stimulus, produces several physiological actions on taste receptor cells that include inhibition of KIR and KV potassium currents and elevations of intracellular calcium. These responses display adaptation, i.e. their magnitude diminishes in the sustained presence of the stimulus. Levels of the membrane lipid phosphatidylinositol-4,5-bisphosphate (PIP2) are well known to modulate many potassium channels, activating the channel by stabilizing its open state. Here we investigate a putative relationship of KIR and KV with PIP2 levels hypothesizing that inhibition of these currents by caffeine might be allayed by PIP2 resynthesis. Using standard patch-clamp techniques, recordings of either potassium current from rat posterior taste receptor cells produced essentially parallel results when PIP2 levels were manipulated pharmacologically. Increasing PIP2 levels by blocking phosphoinositide-3 kinase with wortmannin or LY294002, or by blocking phospholipase C with U73122 all significantly increased the incidence of adaptation for both KIR and KV. Conversely, lowering PIP2 synthesis by blocking PI4K or using the PIP2 scavengers polylysine or bovine serum albumin reduced the incidence of adaptation. Adaptation could be modulated by activation of protein kinase C but not calcium calmodulin kinase. Collectively, these data support two highly novel conclusions: potassium currents in taste receptor cells are significantly modulated by PIP2 levels and PIP2 resynthesis may play a central role in the gustatory adaptation process at the primary receptor cell level.


Assuntos
Cafeína/farmacologia , Fosfatidilinositol 4,5-Difosfato/biossíntese , Papilas Gustativas/fisiologia , Paladar/fisiologia , Androstadienos/farmacologia , Animais , Benzilaminas/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Cromonas/farmacologia , Canais de Potássio de Retificação Tardia/efeitos dos fármacos , Canais de Potássio de Retificação Tardia/fisiologia , Inibidores Enzimáticos/farmacologia , Estrenos/farmacologia , Masculino , Morfolinas/farmacologia , Técnicas de Patch-Clamp , Fosfatidilinositol 4,5-Difosfato/antagonistas & inibidores , Fosfatidilinositol 4,5-Difosfato/farmacologia , Fosfatos de Fosfatidilinositol/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosfolipase C beta/antagonistas & inibidores , Polilisina/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Proteína Quinase C/metabolismo , Pirrolidinonas/farmacologia , Ratos , Ratos Sprague-Dawley , Soroalbumina Bovina/farmacologia , Sulfonamidas/farmacologia , Paladar/efeitos dos fármacos , Papilas Gustativas/citologia , Papilas Gustativas/efeitos dos fármacos , Acetato de Tetradecanoilforbol/farmacologia , Wortmanina
20.
Pflugers Arch ; 457(1): 77-89, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18461353

RESUMO

The transient receptor potential channel of the ankyrin-binding repeat subfamily, TRPA1, is a Ca(2+)-permeable non-selective cation channel that depolarizes the plasma membrane and causes Ca(2+) influx. A typical feature of TRPA1 is its rapid desensitization following activation by agonists such as mustard oil (MO), cinnamaldehyde, and a high intracellular Ca(2+) concentration. In whole-cell recordings on Chinese hamster ovary (CHO) cells expressing TRPA1, desensitization was delayed when phosphatidylinositol 4,5-biphosphate (PIP(2)) was supplemented via the patch pipette, whereas the PIP(2) scavenger neomycin accelerated desensitization. Preincubation with the PI-4 kinase inhibitor wortmannin reduced both constitutive TRPA1 channels activity and the response to MO. Run down was also accelerated by high intracellular Mg(2+) concentrations, whereas chelating intracellular Mg(2+) with 10 mM ethylenedinitrilotetraacetic acid (EDTA) increased the basal channel activity. In inside-out patches, we observed a rapid run down of TRPA1 activity, which could be prevented by application of diC8-PIP(2) or 2 mM Mg-ATP but not Na(2)-ATP to the cytosolic side of the excised patches. In isolated trigeminal ganglion neurons, preincubation with wortmannin resulted in inhibition of endogenous TRPA1 activation by MO. Taken together, our data indicate that PIP(2) modulates TRPA1, albeit to a lesser extent than other known PIP(2)-dependent TRP channels, and that tools modifying the plasma membrane PIP(2) content often have direct effects on this channel.


Assuntos
Canais de Cálcio/efeitos dos fármacos , Proteínas do Tecido Nervoso/efeitos dos fármacos , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canais de Potencial de Receptor Transitório/efeitos dos fármacos , Animais , Arsenicais/farmacologia , Células CHO , Cricetinae , Cricetulus , Interpretação Estatística de Dados , Eletrofisiologia , Estrenos/farmacologia , Mostardeira , NADPH Oxidases/antagonistas & inibidores , Neomicina/farmacologia , Proteínas do Tecido Nervoso/agonistas , Neurônios/fisiologia , Técnicas de Patch-Clamp , Fosfatidilinositol 4,5-Difosfato/antagonistas & inibidores , Óleos de Plantas/farmacologia , Inibidores da Síntese de Proteínas/farmacologia , Pirrolidinonas/farmacologia , Soluções , Canal de Cátion TRPA1 , Canais de Potencial de Receptor Transitório/agonistas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...