Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 116(20): 10156-10161, 2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-31028142

RESUMO

Transient receptor potential canonical type 6 (TRPC6) is a nonselective receptor-operated cation channel that regulates reactive fibrosis and growth signaling. Increased TRPC6 activity from enhanced gene expression or gain-of-function mutations contribute to cardiac and/or renal disease. Despite evidence supporting a pathophysiological role, no orally bioavailable selective TRPC6 inhibitor has yet been developed and tested in vivo in disease models. Here, we report an orally bioavailable TRPC6 antagonist (BI 749327; IC50 13 nM against mouse TRPC6, t1/2 8.5-13.5 hours) with 85- and 42-fold selectivity over the most closely related channels, TRPC3 and TRPC7. TRPC6 calcium conductance results in the stimulation of nuclear factor of activated T cells (NFAT) that triggers pathological cardiac and renal fibrosis and disease. BI 749327 suppresses NFAT activation in HEK293T cells expressing wild-type or gain-of-function TRPC6 mutants (P112Q, M132T, R175Q, R895C, and R895L) and blocks associated signaling and expression of prohypertrophic genes in isolated myocytes. In vivo, BI 749327 (30 mg/kg/day, yielding unbound trough plasma concentration ∼180 nM) improves left heart function, reduces volume/mass ratio, and blunts expression of profibrotic genes and interstitial fibrosis in mice subjected to sustained pressure overload. Additionally, BI 749327 dose dependently reduces renal fibrosis and associated gene expression in mice with unilateral ureteral obstruction. These results provide in vivo evidence of therapeutic efficacy for a selective pharmacological TRPC6 inhibitor with oral bioavailability and suitable pharmacokinetics to ameliorate cardiac and renal stress-induced disease with fibrosis.


Assuntos
Cardiomegalia/tratamento farmacológico , Nefroesclerose/tratamento farmacológico , Canal de Cátion TRPC6/antagonistas & inibidores , Animais , Avaliação Pré-Clínica de Medicamentos , Fibrose , Células HEK293 , Coração/efeitos dos fármacos , Humanos , Rim/efeitos dos fármacos , Camundongos
2.
PLoS One ; 13(1): e0191225, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29385160

RESUMO

BACKGROUND: Forty million adults in the US suffer from anxiety disorders, making these the most common forms of mental illness. Transient receptor potential channel canonical subfamily (TRPC) members 4 and 5 are non-selective cation channels highly expressed in regions of the cortex and amygdala, areas thought to be important in regulating anxiety. Previous work with null mice suggests that inhibition of TRPC4 and TRPC5 may have anxiolytic effects. HC-070 IN VITRO: To assess the potential of TRPC4/5 inhibitors as an avenue for treatment, we invented a highly potent, small molecule antagonist of TRPC4 and TRPC5 which we call HC-070. HC-070 inhibits recombinant TRPC4 and TRPC5 homomultimers in heterologous expression systems with nanomolar potency. It also inhibits TRPC1/5 and TRPC1/4 heteromultimers with similar potency and reduces responses evoked by cholecystokinin tetrapeptide (CCK-4) in the amygdala. The compound is >400-fold selective over a wide range of molecular targets including ion channels, receptors, and kinases. HC-070 IN VIVO: Upon oral dosing in mice, HC-070 achieves exposure levels in the brain and plasma deemed sufficient to test behavioral activity. Treatment with HC-070 attenuates the anxiogenic effect of CCK-4 in the elevated plus maze (EPM). The compound recapitulates the phenotype observed in both null TRPC4 and TRPC5 mice in a standard EPM. Anxiolytic and anti-depressant effects of HC-070 are also observed in pharmacological in vivo tests including marble burying, tail suspension and forced swim. Furthermore, HC-070 ameliorates the increased fear memory induced by chronic social stress. A careful evaluation of the pharmacokinetic-pharmacodynamic relationship reveals that substantial efficacy is observed at unbound brain levels similar to, or even lower than, the 50% inhibitory concentration (IC50) recorded in vitro, increasing confidence that the observed effects are indeed mediated by TRPC4 and/or TRPC5 inhibition. Together, this experimental data set introduces a novel, high quality, small molecule antagonist of TRPC4 and TRPC5 containing channels and supports the targeting of TRPC4 and TRPC5 channels as a new mechanism of action for the treatment of psychiatric symptoms.


Assuntos
Ansiolíticos/farmacologia , Antidepressivos/farmacologia , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Canais de Cátion TRPC/antagonistas & inibidores , Animais , Ansiolíticos/química , Ansiolíticos/farmacocinética , Antidepressivos/química , Antidepressivos/farmacocinética , Ansiedade/tratamento farmacológico , Ansiedade/metabolismo , Ansiedade/psicologia , Complexo Nuclear Basolateral da Amígdala/efeitos dos fármacos , Complexo Nuclear Basolateral da Amígdala/metabolismo , Comportamento Animal/efeitos dos fármacos , Depressão/tratamento farmacológico , Depressão/metabolismo , Depressão/psicologia , Modelos Animais de Doenças , Medo/efeitos dos fármacos , Medo/fisiologia , Medo/psicologia , Compostos Heterocíclicos de 4 ou mais Anéis/química , Compostos Heterocíclicos de 4 ou mais Anéis/farmacocinética , Ensaios de Triagem em Larga Escala , Humanos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL
3.
Br J Pharmacol ; 175(12): 2185-2203, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-28924972

RESUMO

Control of chronic pain is frequently inadequate and/or associated with intolerable adverse effects, prompting a frantic search for new therapeutics and new therapeutic targets. Nearly two decades of preclinical and clinical research supports the involvement of transient receptor potential (TRP) channels in temperature perception, nociception and sensitization. Although there has been considerable excitement around the therapeutic potential of this channel family since the cloning and identification of TRPV1 cation channels as the capsaicin receptor more than 20 years ago, only modulators of a few channels have been tested clinically. TRPV1 channel antagonists have suffered from side effects related to the channel's role in temperature sensation; however, high dose formulations of capsaicin have reached the market and shown therapeutic utility. A number of potent, small molecule antagonists of TRPA1 channels have recently advanced into clinical trials for the treatment of inflammatory and neuropathic pain, and TRPM8 antagonists are following closely behind for cold allodynia. TRPV3, TRPV4, TRPM2 and TRPM3 channels have also been of significant interest. This review discusses the preclinical promise and status of novel analgesic agents that target TRP channels and the challenges that these compounds may face in development and clinical practice. LINKED ARTICLES: This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.


Assuntos
Analgésicos/farmacologia , Analgésicos/uso terapêutico , Dor Crônica/tratamento farmacológico , Canais de Potencial de Receptor Transitório/antagonistas & inibidores , Animais , Dor Crônica/metabolismo , Humanos , Canais de Potencial de Receptor Transitório/metabolismo
4.
Annu Rev Pharmacol Toxicol ; 58: 309-330, 2018 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-28945977

RESUMO

The transient receptor potential (TRP) superfamily of channels comprises a diverse group of cation channels. Four TRP channel subunits coassemble to form functional homo- or heterotetramers that pass sodium, calcium, or both in the inward direction. Modulating TRP channel activity provides an important way to impact cellular function by regulating both membrane excitability and intracellular calcium levels. The import of these channels is underscored by the number of genetic diseases caused when they are mutated: Skeletal, skin, sensory, ocular, cardiac, and neuronal disturbances all arise from aberrant TRP function. Not surprisingly, there has been significant pharmaceutical interest in targeting these fascinating channels. Compounds that modulate TRP vanilloid 1 (TRPV1), TRPV3, TRPV4, TRP ankyrin 1 (TRPA1), and TRP melastatin 8 (TRPM8) have all entered clinical trials. The goal of this review is to familiarize the readers with the rationale behind the pursuit of these channels in drug discovery and the status of those efforts.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Ensaios Clínicos como Assunto , Descoberta de Drogas/métodos , Humanos
5.
J Clin Invest ; 124(12): 5225-38, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25365224

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a fatal fibrotic lung disorder with no effective medical treatments available. The generation of myofibroblasts, which are critical for fibrogenesis, requires both a mechanical signal and activated TGF-ß; however, it is not clear how fibroblasts sense and transmit the mechanical signal(s) that promote differentiation into myofibroblasts. As transient receptor potential vanilloid 4 (TRPV4) channels are activated in response to changes in plasma membrane stretch/matrix stiffness, we investigated whether TRPV4 contributes to generation of myofibroblasts and/or experimental lung fibrosis. We determined that TRPV4 activity is upregulated in lung fibroblasts derived from patients with IPF. Moreover, TRPV4-deficient mice were protected from fibrosis. Furthermore, genetic ablation or pharmacological inhibition of TRPV4 function abrogated myofibroblast differentiation, which was restored by TRPV4 reintroduction. TRPV4 channel activity was elevated when cells were plated on matrices of increasing stiffness or on fibrotic lung tissue, and matrix stiffness-dependent myofibroblast differentiation was reduced in response to TRVP4 inhibition. TRPV4 activity modulated TGF-ß1-dependent actions in a SMAD-independent manner, enhanced actomyosin remodeling, and increased nuclear translocation of the α-SMA transcription coactivator (MRTF-A). Together, these data indicate that TRPV4 activity mediates pulmonary fibrogenesis and suggest that manipulation of TRPV4 channel activity has potential as a therapeutic approach for fibrotic diseases.


Assuntos
Diferenciação Celular , Pulmão/metabolismo , Miofibroblastos/metabolismo , Fibrose Pulmonar/metabolismo , Canais de Cátion TRPV/biossíntese , Regulação para Cima , Animais , Antibióticos Antineoplásicos/efeitos adversos , Antibióticos Antineoplásicos/farmacologia , Bleomicina/efeitos adversos , Bleomicina/farmacologia , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Feminino , Pulmão/patologia , Camundongos , Camundongos Mutantes , Miofibroblastos/patologia , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , Fibrose Pulmonar/patologia , Canais de Cátion TRPV/genética , Transativadores/genética , Transativadores/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
7.
Nat Rev Drug Discov ; 10(8): 601-20, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21804597

RESUMO

Transient receptor potential (TRP) cation channels have been among the most aggressively pursued drug targets over the past few years. Although the initial focus of research was on TRP channels that are expressed by nociceptors, there has been an upsurge in the amount of research that implicates TRP channels in other areas of physiology and pathophysiology, including the skin, bladder and pulmonary systems. In addition, mutations in genes encoding TRP channels are the cause of several inherited diseases that affect a variety of systems including the renal, skeletal and nervous system. This Review focuses on recent developments in the TRP channel-related field, and highlights potential opportunities for therapeutic intervention.


Assuntos
Descoberta de Drogas , Canais de Potencial de Receptor Transitório/antagonistas & inibidores , Animais , Regulação da Temperatura Corporal , Canais de Cálcio/fisiologia , Humanos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/fisiologia , Canal de Cátion TRPA1 , Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/fisiologia , Canais de Cátion TRPV/agonistas , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/fisiologia , Canais de Potencial de Receptor Transitório/química , Canais de Potencial de Receptor Transitório/genética , Canais de Potencial de Receptor Transitório/fisiologia , Doenças da Bexiga Urinária/tratamento farmacológico
8.
J Neurosci ; 30(45): 15165-74, 2010 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-21068322

RESUMO

TRPA1 is a nonselective cation channel expressed by nociceptors. Although it is widely accepted that TRPA1 serves as a broad irritancy receptor for a variety of reactive chemicals, its role in cold sensation remains controversial. Here, we demonstrate that mild cooling markedly increases agonist-evoked rat TRPA1 currents. In the absence of an agonist, even noxious cold only increases current amplitude slightly. These results suggest that TRPA1 is a key mediator of cold hypersensitivity in pathological conditions in which reactive oxygen species and proinflammatory activators of the channel are present, but likely plays a comparatively minor role in acute cold sensation. Supporting this, cold hypersensitivity can be induced in wild-type but not Trpa1(-/-) mice by subcutaneous administration of a TRPA1 agonist. Furthermore, the selective TRPA1 antagonist HC-030031 [2-(1,3-dimethyl-2,6-dioxo-1,2,3,6-tetrahydro-7H-purin-7-yl)-N-(4-isopropylphenyl)acetamide] reduces cold hypersensitivity in rodent models of inflammatory and neuropathic pain.


Assuntos
Temperatura Baixa , Hiperalgesia/metabolismo , Nociceptores/fisiologia , Sensação Térmica/fisiologia , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Eletrofisiologia , Gânglios Espinais/fisiologia , Hiperalgesia/fisiopatologia , Camundongos , Camundongos Knockout , Ratos , Canal de Cátion TRPA1 , Canais de Potencial de Receptor Transitório/agonistas , Canais de Potencial de Receptor Transitório/antagonistas & inibidores
9.
Proc Natl Acad Sci U S A ; 107(44): 19084-9, 2010 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-20956320

RESUMO

Reduced functional bladder capacity and concomitant increased micturition frequency (pollakisuria) are common lower urinary tract symptoms associated with conditions such as cystitis, prostatic hyperplasia, neurological disease, and overactive bladder syndrome. These symptoms can profoundly affect the quality of life of afflicted individuals, but available pharmacological treatments are often unsatisfactory. Recent work has demonstrated that the cation channel TRPV4 is highly expressed in urothelial cells and plays a role in sensing the normal filling state of the bladder. In this article, we show that the development of cystitis-induced bladder dysfunction is strongly impaired in Trpv4(-/-) mice. Moreover, we describe HC-067047, a previously uncharacterized, potent, and selective TRPV4 antagonist that increases functional bladder capacity and reduces micturition frequency in WT mice and rats with cystitis. HC-067047 did not affect bladder function in Trpv4(-/-) mice, demonstrating that its in vivo effects are on target. These results indicate that TRPV4 antagonists may provide a promising means of treating bladder dysfunction.


Assuntos
Antineoplásicos Alquilantes/efeitos adversos , Ciclofosfamida/efeitos adversos , Cistite , Moduladores de Transporte de Membrana/farmacologia , Morfolinas/farmacologia , Pirróis/farmacologia , Canais de Cátion TRPV/antagonistas & inibidores , Bexiga Urinária/fisiopatologia , Urotélio/fisiopatologia , Animais , Antineoplásicos Alquilantes/farmacologia , Ciclofosfamida/farmacologia , Cistite/induzido quimicamente , Cistite/tratamento farmacológico , Cistite/metabolismo , Cistite/fisiopatologia , Humanos , Camundongos , Camundongos Knockout , Ratos , Ratos Wistar , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo , Bexiga Urinária/metabolismo , Micção/efeitos dos fármacos , Urotélio/metabolismo
10.
PLoS One ; 4(8): e6844, 2009 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-19718436

RESUMO

The four sperm-specific CatSper ion channel proteins are required for hyperactivated motility and male fertility, and for Ca(2+) entry evoked by alkaline depolarization. In the absence of external Ca(2+), Na(+) carries current through CatSper channels in voltage-clamped sperm. Here we show that CatSper channel activity can be monitored optically with the [Na(+)](i)-reporting probe SBFI in populations of intact sperm. Removal of external Ca(2+) increases SBFI signals in wild-type but not CatSper2-null sperm. The rate of the indicated rise of [Na(+)](i) is greater for sperm alkalinized with NH(4)Cl than for sperm acidified with propionic acid, reflecting the alkaline-promoted signature property of CatSper currents. In contrast, the [Na(+)](i) rise is slowed by candidate CatSper blocker HC-056456 (IC(50) approximately 3 microM). HC-056456 similarly slows the rise of [Ca(2+)](i) that is evoked by alkaline depolarization and reported by fura-2. HC-056456 also selectively and reversibly decreased CatSper currents recorded from patch-clamped sperm. HC-056456 does not prevent activation of motility by HCO(3) (-) but does prevent the development of hyperactivated motility by capacitating incubations, thus producing a phenocopy of the CatSper-null sperm. When applied to hyperactivated sperm, HC-056456 causes a rapid, reversible loss of flagellar waveform asymmetry, similar to the loss that occurs when Ca(2+) entry through the CatSper channel is terminated by removal of external Ca(2+). Thus, open CatSper channels and entry of external Ca(2+) through them sustains hyperactivated motility. These results indicate that pharmacological targeting of the CatSper channel may impose a selective late-stage block to fertility, and that high-throughput screening with an optical reporter of CatSper channel activity may identify additional selective blockers with potential for male-directed contraception.


Assuntos
Canais de Cálcio/efeitos dos fármacos , Espermatozoides/efeitos dos fármacos , Animais , Cálcio/metabolismo , Transporte de Íons , Masculino , Camundongos , Sódio/metabolismo
11.
Proc Natl Acad Sci U S A ; 106(22): 9099-104, 2009 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-19458046

RESUMO

Asthma is an inflammatory disorder caused by airway exposures to allergens and chemical irritants. Studies focusing on immune, smooth muscle, and airway epithelial function revealed many aspects of the disease mechanism of asthma. However, the limited efficacies of immune-directed therapies suggest the involvement of additional mechanisms in asthmatic airway inflammation. TRPA1 is an irritant-sensing ion channel expressed in airway chemosensory nerves. TRPA1-activating stimuli such as cigarette smoke, chlorine, aldehydes, and scents are among the most prevalent triggers of asthma. Endogenous TRPA1 agonists, including reactive oxygen species and lipid peroxidation products, are potent drivers of allergen-induced airway inflammation in asthma. Here, we examined the role of TRPA1 in allergic asthma in the murine ovalbumin model. Strikingly, genetic ablation of TRPA1 inhibited allergen-induced leukocyte infiltration in the airways, reduced cytokine and mucus production, and almost completely abolished airway hyperreactivity to contractile stimuli. This phenotype is recapitulated by treatment of wild-type mice with HC-030031, a TRPA1 antagonist. HC-030031, when administered during airway allergen challenge, inhibited eosinophil infiltration and prevented the development of airway hyperreactivity. Trpa1(-/-) mice displayed deficiencies in chemically and allergen-induced neuropeptide release in the airways, providing a potential explanation for the impaired inflammatory response. Our data suggest that TRPA1 is a key integrator of interactions between the immune and nervous systems in the airways, driving asthmatic airway inflammation following inhaled allergen challenge. TRPA1 may represent a promising pharmacological target for the treatment of asthma and other allergic inflammatory conditions.


Assuntos
Asma/fisiopatologia , Hiper-Reatividade Brônquica/fisiopatologia , Células Receptoras Sensoriais/fisiologia , Canais de Potencial de Receptor Transitório/fisiologia , Animais , Asma/imunologia , Hiper-Reatividade Brônquica/imunologia , Sistema Imunitário/imunologia , Sistema Imunitário/fisiopatologia , Inflamação/imunologia , Inflamação/fisiopatologia , Camundongos , Camundongos Knockout , Células Receptoras Sensoriais/imunologia , Canal de Cátion TRPA1 , Canais de Potencial de Receptor Transitório/genética
12.
Mol Pain ; 5: 19, 2009 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-19383149

RESUMO

BACKGROUND: TRPA1 has been implicated in both chemo- and mechanosensation. Recent work demonstrates that inhibiting TRPA1 function reduces mechanical hypersensitivity produced by inflammation. Furthermore, a broad range of chemical irritants require functional TRPA1 to exert their effects. In this study we use the ex-vivo skin-nerve preparation to directly determine the contribution of TRPA1 to mechanical- and chemical-evoked responses at the level of the primary afferent terminal. RESULTS: Acute application of HC-030031, a selective TRPA1 antagonist, inhibited all formalin responses in rat C fibers but had no effect on TRPV1 function, assessed by capsaicin responsiveness. Genetic ablation experiments corroborated the pharmacological findings as C fibers from wild type mice responded to both formalin and capsaicin, but fibers from their TRPA1-deficient littermates responded only to capsaicin. HC-030031 markedly reduced the mechanically-evoked action potential firing in rat and wild type mouse C fibers, particularly at high-intensity forces, but had no effect on the mechanical responsiveness of Adelta fiber nociceptors. Furthermore, HC-030031 had no effect on mechanically-evoked firing in C fibers from TRPA1-deficient mice, indicating that HC-030031 inhibits mechanically-evoked firing via a TRPA1-dependent mechanism. CONCLUSION: Our data show that acute pharmacological blockade of TRPA1 at the cutaneous receptive field inhibits formalin-evoked activation and markedly reduces mechanically-evoked action potential firing in C fibers. Thus, functional TRPA1 at sensory afferent terminals in skin is required for their responsiveness to both noxious chemical and mechanical stimuli.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Moduladores de Transporte de Membrana/farmacologia , Nociceptores/efeitos dos fármacos , Nociceptores/metabolismo , Canais de Potencial de Receptor Transitório/antagonistas & inibidores , Animais , Capsaicina/farmacologia , Feminino , Formaldeído/farmacologia , Masculino , Camundongos , Camundongos Knockout , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Fibras Nervosas Mielinizadas/fisiologia , Fibras Nervosas Amielínicas/efeitos dos fármacos , Fibras Nervosas Amielínicas/fisiologia , Ratos , Ratos Sprague-Dawley , Canal de Cátion TRPA1 , Canais de Potencial de Receptor Transitório/deficiência
13.
Proc Natl Acad Sci U S A ; 104(33): 13525-30, 2007 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-17686976

RESUMO

The formalin model is widely used for evaluating the effects of analgesic compounds in laboratory animals. Injection of formalin into the hind paw induces a biphasic pain response; the first phase is thought to result from direct activation of primary afferent sensory neurons, whereas the second phase has been proposed to reflect the combined effects of afferent input and central sensitization in the dorsal horn. Here we show that formalin excites sensory neurons by directly activating TRPA1, a cation channel that plays an important role in inflammatory pain. Formalin induced robust calcium influx in cells expressing cloned or native TRPA1 channels, and these responses were attenuated by a previously undescribed TRPA1-selective antagonist. Moreover, sensory neurons from TRPA1-deficient mice lacked formalin sensitivity. At the behavioral level, pharmacologic blockade or genetic ablation of TRPA1 produced marked attenuation of the characteristic flinching, licking, and lifting responses resulting from intraplantar injection of formalin. Our results show that TRPA1 is the principal site of formalin's pain-producing action in vivo, and that activation of this excitatory channel underlies the physiological and behavioral responses associated with this model of pain hypersensitivity.


Assuntos
Canais de Cálcio/fisiologia , Formaldeído/toxicidade , Proteínas do Tecido Nervoso/fisiologia , Dor/induzido quimicamente , Canais de Potencial de Receptor Transitório/fisiologia , Animais , Anquirinas , Gânglios Espinais/efeitos dos fármacos , Humanos , Neurônios/efeitos dos fármacos , Ratos , Proteínas Recombinantes/metabolismo , Canal de Cátion TRPA1 , Canais de Cátion TRPC
14.
Proc Natl Acad Sci U S A ; 104(4): 1219-23, 2007 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-17227845

RESUMO

Mammalian spermatozoa become motile at ejaculation, but before they can fertilize the egg, they must acquire more thrust to penetrate the cumulus and zona pellucida. The forceful asymmetric motion of hyperactivated spermatozoa requires Ca2+ entry into the sperm tail by an alkalinization-activated voltage-sensitive Ca2+-selective current (ICatSper). Hyperactivation requires CatSper1 and CatSper2 putative ion channel genes, but the function of two other related genes (CatSper3 and CatSper4) is not known. Here we show that targeted disruption of murine CatSper3 or CatSper4 also abrogated ICatSper, sperm cell hyperactivated motility and male fertility but did not affect spermatogenesis or initial motility. Direct protein interactions among CatSpers, the sperm specificity of these proteins, and loss of ICatSper in each of the four CatSper-/- mice indicate that CatSpers are highly specialized flagellar proteins.


Assuntos
Canais de Cálcio/fisiologia , Fertilidade/fisiologia , Isoformas de Proteínas/fisiologia , Motilidade dos Espermatozoides/fisiologia , Animais , Masculino , Camundongos , Dados de Sequência Molecular
15.
Nature ; 440(7088): 1213-6, 2006 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-16554753

RESUMO

Voltage changes across the cell membrane control the gating of many cation-selective ion channels. Conserved from bacteria to humans, the voltage-gated-ligand superfamily of ion channels are encoded as polypeptide chains of six transmembrane-spanning segments (S1-S6). S1-S4 functions as a self-contained voltage-sensing domain (VSD), in essence a positively charged lever that moves in response to voltage changes. The VSD 'ligand' transmits force via a linker to the S5-S6 pore domain 'receptor', thereby opening or closing the channel. The ascidian VSD protein Ci-VSP gates a phosphatase activity rather than a channel pore, indicating that VSDs function independently of ion channels. Here we describe a mammalian VSD protein (H(V)1) that lacks a discernible pore domain but is sufficient for expression of a voltage-sensitive proton-selective ion channel activity. H(v)1 currents are activated at depolarizing voltages, sensitive to the transmembrane pH gradient, H+-selective, and Zn2+-sensitive. Mutagenesis of H(v)1 identified three arginine residues in S4 that regulate channel gating and two histidine residues that are required for extracellular inhibition of H(v)1 by Zn2+. H(v)1 is expressed in immune tissues and manifests the characteristic properties of native proton conductances (G(vH+)). In phagocytic leukocytes, G(vH+) are required to support the oxidative burst that underlies microbial killing by the innate immune system. The data presented here identify H(v)1 as a long-sought voltage-gated H+ channel and establish H(v)1 as the founding member of a family of mammalian VSD proteins.


Assuntos
Ativação do Canal Iônico , Canais Iônicos/química , Canais Iônicos/metabolismo , Prótons , Linhagem Celular , Condutividade Elétrica , Humanos , Concentração de Íons de Hidrogênio , Sistema Imunitário/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Canais Iônicos/antagonistas & inibidores , Canais Iônicos/genética , Ligantes , Mutação/genética , Estrutura Terciária de Proteína , Zinco/farmacologia
16.
Mol Cell Biol ; 25(7): 2632-43, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15767669

RESUMO

The lack of direct targets for TATA-binding protein (TBP)-like factors (TLFs) confounds the understanding of their role in gene expression. Here we report that human TLF (also called TBP-related factor 2 [TRF2]) activates a number of different genes, including the neurofibromatosis type 1 (NF1) gene. The overexpression of TLF increases the amount of NF1 mRNA in cells. In vivo, TLF binds to and upregulates transcription from a fragment of the NF1 promoter. In vitro, purified TLF-TFIIA binds directly to the same NF1 promoter fragment that is required for TLF responsiveness in cells. Furthermore, targeted deletion of TLF in mice reduces NF1 levels. In contrast, TLF inhibits transcription driven by a fragment from the TATA-containing c-fos promoter by sequestering TFIIA. TBP affects the NF1 and c-fos promoters in a manner reciprocal to that of TLF, stimulating the c-fos promoter and inhibiting NF1 transcription. We conclude that TLF is a functional regulator of transcription with targets distinct from those of TBP.


Assuntos
Regulação da Expressão Gênica/genética , Genes da Neurofibromatose 1 , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Semelhantes à Proteína de Ligação a TATA-Box/metabolismo , Proteína de Ligação a TATA-Box/metabolismo , Transcrição Gênica/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops , Humanos , Camundongos , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Semelhantes à Proteína de Ligação a TATA-Box/química , Proteínas Semelhantes à Proteína de Ligação a TATA-Box/genética
17.
Curr Opin Neurobiol ; 14(3): 362-9, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15194117

RESUMO

The transient receptor potential (TRP) superfamily comprises a group of non-selective cation channels that sense and respond to changes in their local environments. TRP channels are found in many eukaryotes, from yeast to mammals. They are a diverse group of proteins organized into six families: classical (TRPC), vanilloid (TRPV), melastatin (TRPM), muclopins (TRPML), polycystin (TRPP), and ANKTM1 (TRPA). In the peripheral nervous system, stimuli including temperature, pressure, inflammatory agents, and receptor activation effect TRP-mediated responses. In the central nervous system, TRPs participate in neurite outgrowth, receptor signalling and excitotoxic cell death resulting from anoxia. TRP channels are emerging as essential cellular switches that allow animals to respond to their environments.


Assuntos
Canais de Cálcio/metabolismo , Membrana Celular/metabolismo , Sistema Nervoso/metabolismo , Animais , Comunicação Celular/fisiologia , Diferenciação Celular/fisiologia , Membrana Celular/ultraestrutura , Humanos , Sistema Nervoso/crescimento & desenvolvimento , Sistema Nervoso/ultraestrutura , Filogenia , Sensação/fisiologia , Transdução de Sinais/fisiologia , Canais de Cátion TRPC
18.
Nature ; 418(6894): 181-6, 2002 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-12077604

RESUMO

Transient receptor potential (TRP) proteins are cation-selective channels that function in processes as diverse as sensation and vasoregulation. Mammalian TRP channels that are gated by heat and capsaicin (>43 degrees C; TRPV1 (ref. 1)), noxious heat (>52 degrees C; TRPV2 (ref. 2)), and cooling (< 22 degrees C; TRPM8 (refs 3, 4)) have been cloned; however, little is known about the molecular determinants of temperature sensing in the range between approximately 22 degrees C and 40 degrees C. Here we have identified a member of the vanilloid channel family, human TRPV3 (hTRPV3) that is expressed in skin, tongue, dorsal root ganglion, trigeminal ganglion, spinal cord and brain. Increasing temperature from 22 degrees C to 40 degrees C in mammalian cells transfected with hTRPV3 elevated intracellular calcium by activating a nonselective cationic conductance. As in published recordings from sensory neurons, the current was steeply dependent on temperature, sensitized with repeated heating, and displayed a marked hysteresis on heating and cooling. On the basis of these properties, we propose that hTRPV3 is thermosensitive in the physiological range of temperatures between TRPM8 and TRPV1.


Assuntos
Cálcio/metabolismo , Proteínas de Transporte de Cátions , Permeabilidade da Membrana Celular , Ativação do Canal Iônico , Canais Iônicos/metabolismo , Temperatura , Sequência de Aminoácidos , Animais , Células CHO , Sinalização do Cálcio , Cátions Bivalentes/metabolismo , Clonagem Molecular , Cricetinae , Condutividade Elétrica , Eletrofisiologia , Evolução Molecular , Perfilação da Expressão Gênica , Humanos , Canais Iônicos/química , Canais Iônicos/genética , Dados de Sequência Molecular , Estrutura Terciária de Proteína , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Alinhamento de Sequência , Canais de Cátion TRPV
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...