Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 6: 35395, 2016 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-27734949

RESUMO

Cystinosis is a rare autosomal recessive disorder characterized by lysosomal cystine accumulation due to loss of function of the lysosomal cystine transporter (CTNS). The most common mutation in cystinosis patients of Northern Europe consists of a 57-kb deletion. This deletion not only inactivates the CTNS gene but also extends into the non-coding region upstream of the start codon of the TRPV1 gene, encoding the capsaicin- and heat-sensitive ion channel TRPV1. To evaluate the consequences of the 57-kb deletion on functional TRPV1 expression, we compared thermal, mechanical and chemical sensitivity of cystinosis patients with matched healthy controls. Whereas patients heterozygous for the 57-kb deletion showed normal sensory responses, homozygous subjects exhibited a 60% reduction in vasodilation and pain evoked by capsaicin, as well as an increase in heat detection threshold. Responses to cold, mechanical stimuli or cinnamaldehyde, an agonist of the related nociceptor channel TRPA1, were unaltered. We conclude that cystinosis patients homozygous for the 57-kb deletion exhibit a strong reduction of TRPV1 function, leading to sensory deficiencies akin to the phenotype of TRPV1-deficient mice. These deficits may account for the reported sensory alterations and thermoregulatory deficits in these patients, and provide a paradigm for life-long TRPV1 deficiency in humans.


Assuntos
Cistinose/metabolismo , Deleção de Genes , Homozigoto , Canais de Cátion TRPV/metabolismo , Acroleína/análogos & derivados , Acroleína/química , Adolescente , Adulto , Alelos , Capsaicina/química , Códon , Cistinose/genética , Europa (Continente) , Feminino , Temperatura Alta , Humanos , Lisossomos/metabolismo , Masculino , Mutação , Deleção de Sequência , Canal de Cátion TRPA1/metabolismo , Canais de Cátion TRPV/genética , Adulto Jovem
2.
Neurourol Urodyn ; 35(4): 450-6, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25727376

RESUMO

AIMS: Neurogenic bladder dysfunction is a major issue in Multiple Sclerosis (MS). High intravesical pressure should be treated early. Available therapies are insufficient and there is need for drug development and investigation of pathogenesis. Experimental Autoimmune Encephalomyelitis (EAE) in rodents is a well validated model to study MS. Previous research has shown that these animals develop urinary symptoms. However, from clinical studies, we know that symptoms do not necessarily reflect changes in bladder pressure. This paper aims to provide a complete overview of urodynamic changes in a model for detrusor overactivity in MS. METHODS: Female C57Bl/6J mice, injected with MOG35-55 and control mice, injected with vehicle (Complete Freund's adjuvant), were monitored daily for neurologic symptoms. Within 1 month after symptom development, mice were used for cystometry or histology of the bladder. RESULTS: Increasing disease score correlated with increased micturition frequency, basal pressure, and average pressure, and with a decrease in functional bladder capacity, voiding amplitude, and maximum pressure. CONCLUSIONS: This paper provides a detailed description of bladder function in C57Bl/6J mice with Myelin Oligodendrocyte Glycoprotein peptide (MOG35-55 ) induced EAE. This EAE model induces detrusor overactivity in close relationship to neurological impairment. EAE in mice is a suitable model to study detrusor overactivity in MS. Neurourol. Urodynam. 35:450-456, 2016. © 2015 Wiley Periodicals, Inc.


Assuntos
Encefalomielite Autoimune Experimental/fisiopatologia , Bexiga Urinaria Neurogênica/fisiopatologia , Bexiga Urinária Hiperativa/fisiopatologia , Bexiga Urinária/fisiopatologia , Urodinâmica/fisiologia , Animais , Feminino , Camundongos , Micção
3.
Br J Pharmacol ; 171(10): 2537-51, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24895732

RESUMO

Lower urinary tract dysfunction (LUTd) represents a major healthcare problem. Although it is mostly not lethal, associated social disturbance, medical costs, loss of productivity and especially diminished quality of life should not be underestimated. Although more than 15% of people suffer from a form of LUTd to some extent, pathophysiology often remains obscure. In the past 20 years, transient receptor potential (TRP) channels have become increasingly important in this field of research. These intriguing ion channels are believed to be the main molecular sensors that generate bladder sensation. Therefore, they are intensely pursued as new drug targets for both curative and symptomatic treatment of different forms of LUTd. TRPV1 was the first of its class to be investigated. Actually, even before this channel was cloned, it had already been targeted in the bladder, with clinical trials of intravesical capsaicin instillations. Several other polymodally gated TRP channels, particularly TRPM8, TRPA1 and TRPV4, also appear to play a prominent role in bladder (patho)physiology. With this review, we provide a brief overview of current knowledge on the role of these TRP channels in LUTd and their potential as molecular targets for treatment.


Assuntos
Sintomas do Trato Urinário Inferior/metabolismo , Células Receptoras Sensoriais/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Doenças da Bexiga Urinária/metabolismo , Bexiga Urinária/inervação , Animais , Desenho de Fármacos , Humanos , Sintomas do Trato Urinário Inferior/tratamento farmacológico , Sintomas do Trato Urinário Inferior/fisiopatologia , Moduladores de Transporte de Membrana/farmacologia , Terapia de Alvo Molecular , Células Receptoras Sensoriais/efeitos dos fármacos , Transdução de Sinais , Canais de Potencial de Receptor Transitório/efeitos dos fármacos , Doenças da Bexiga Urinária/tratamento farmacológico , Doenças da Bexiga Urinária/fisiopatologia , Agentes Urológicos/farmacologia
4.
Neurobiol Aging ; 29(5): 639-52, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-17222948

RESUMO

The function of presenilin1 (PS1) in intra-membrane proteolysis is undisputed, as is its role in neurodegeneration in FAD, in contrast to its exact function in normal conditions. In this study, we analyzed synaptic plasticity and its underlying mechanisms biochemically in brain of mice with a neuron-specific deficiency in PS1 (PS1(n-/-)) and compared them to mice that expressed human mutant PS1[A246E] or wild-type PS1. PS1(n-/-) mice displayed a subtle impairment in Schaffer collateral hippocampal long-term potentiation (LTP) as opposed to normal LTP in wild-type PS1 mice, and a facilitated LTP in mutant PS1[A246E] mice. This finding correlated with, respectively, increased and reduced NMDA receptor responses in PS1[A246E] mice and PS1(n-/-) mice in hippocampal slices. Postsynaptically, levels of NR1/NR2B NMDA-receptor subunits and activated alpha-CaMKII were reduced in PS1(n-/-) mice, while increased in PS1[A246E] mice. In addition, PS1(n-/-) mice, displayed reduced paired pulse facilitation, increased synaptic fatigue and lower number of total and docked synaptic vesicles, implying a presynaptic function for wild-type presenilin1, unaffected by the mutation in PS1[A246E] mice. In contrast to the deficiency in PS1, mutant PS1 activated GSK-3beta by decreasing phosphorylation on Ser-9, which correlated with increased phosphorylation of protein tau at Ser-396-Ser-404 (PHF1/AD2 epitope). The synaptic functions of PS1, exerted on presynaptic vesicles and on postsynaptic NMDA-receptor activity, were concluded to be independent of alterations in GSK-3beta activity and phosphorylation of protein tau.


Assuntos
Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Presenilina-1/metabolismo , Sinapses/fisiologia , Sinapses/ultraestrutura , Transmissão Sináptica/fisiologia , Proteínas tau/metabolismo , Animais , Células Cultivadas , Hipocampo/citologia , Hipocampo/fisiologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mutação , Neurônios/citologia , Fosforilação , Presenilina-1/genética
5.
Handb Exp Pharmacol ; (179): 329-44, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17217067

RESUMO

Originally cloned as a prostate-specific protein, TRPM8 is now best known as a cold- and menthol-activated channel implicated in thermosensation. In this chapter we provide a brief review of current knowledge concerning the biophysical properties, gating mechanisms, pharmacology and (patho)physiology of this TRP channel.


Assuntos
Canais de Cátion TRPM/genética , Canais de Cátion TRPM/fisiologia , Animais , Cálcio/metabolismo , Regulação da Expressão Gênica , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Neoplasias/genética , Neoplasias/patologia , Canais de Cátion TRPM/agonistas , Canais de Cátion TRPM/antagonistas & inibidores , Sensação Térmica/genética , Sensação Térmica/fisiologia
6.
Biochem Soc Trans ; 35(Pt 1): 105-8, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17233613

RESUMO

TRP (transient receptor potential) channels respond to a plethora of stimuli in a fine-tuned manner. We show here that both membrane potential and the level of PI (phosphatidylinositol) phosphates are efficient regulators of TRP channel gating. Recent work has shown that this regulation applies to several members of the TRPV (TRP vanilloid) subfamily (TRPV1 and TRPV5) and the TRPM (TRP melastatin) subfamily (TRPM4/TRPM5/TRPM7/TRPM8), whereas regulation of members of the TRPC subfamily is still disputed. The mechanism whereby PIP(2) (PI 4,5-bisphosphate) acts on TRPM4, a Ca(2+)- and voltage-activated channel, is shown in detail in this paper: (i) PIP(2) may bind directly to the channel, (ii) PIP(2) induces sensitization to activation by Ca(2+), and (iii) PIP(2) shifts the voltage dependence towards negative and physiologically more meaningful potentials. A PIP(2)-binding pocket seems to comprise a part of the TRP domain and especially pleckstrin homology domains in the C-terminus.


Assuntos
Lipídeos/química , Canais de Cátion TRPC/fisiologia , Canais de Cátion TRPM/fisiologia , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Membrana Celular/metabolismo , Eletrofisiologia/métodos , Humanos , Potenciais da Membrana , Modelos Biológicos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Canais de Cátion TRPC/química , Canais de Cátion TRPM/química
7.
Cell Mol Life Sci ; 64(4): 377-81, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17180301

RESUMO

Daily experience tells us that temperature has a strong influence on how we taste. Despite the longstanding interest of many specialists in this aspect of taste, we are only starting to understand the molecular mechanisms underlying the temperature dependence of different taste modalities. Recent research has led to the identification of some strong thermosensitive molecules in the taste transduction pathway. The cold activation of the epithelial Na(+) channel and the heat activation of the taste variant of the vanilloid receptor (TRPV1t) may underlie the temperature dependence of salt responses. Heat activation of the transient receptor potential channel TRPM5 explains the enhancement of sweet taste perception by warm temperatures. Current development of methods to study taste cell physiology will help to determine the contribution of other temperature-sensitive events in the taste transduction pathways. Vice versa, the analysis of the thermodynamic properties of these events may assist to unveil the nature of several taste processes.


Assuntos
Paladar/fisiologia , Temperatura , Animais , Humanos , Sorvetes , Ativação do Canal Iônico/fisiologia , Ratos , Canais de Sódio/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo
8.
Artigo em Inglês | MEDLINE | ID: mdl-16634147

RESUMO

Transient receptor potential (TRP) channels are involved in the perception of a wide range of physical and chemical stimuli, including temperature and osmolarity changes, light, pain, touch, taste and pheromones, and in the initiation of cellular responses thereupon. Since the last decade, rapid progress has been made in the identification and characterization of new members of the TRP superfamily. They constitute a large superfamily of cation channels that are expressed in almost all cell types in both invertebrates and vertebrates. This review summarizes and discusses the current knowledge on the TRP protein structure and its impact on the regulation of the channel function.


Assuntos
Canais de Cálcio/fisiologia , Canais de Potencial de Receptor Transitório/fisiologia , Animais , Cálcio/metabolismo , Canais de Cálcio/genética , Sinalização do Cálcio/fisiologia , Humanos , Modelos Moleculares , Família Multigênica/genética , Filogenia , Canais de Potencial de Receptor Transitório/química , Canais de Potencial de Receptor Transitório/classificação , Canais de Potencial de Receptor Transitório/genética
9.
Circ Res ; 97(9): 908-15, 2005 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-16179585

RESUMO

TRPV4 is a broadly expressed Ca2+-permeable cation channel in the vanilloid subfamily of transient receptor potential channels. TRPV4 gates in response to a large variety of stimuli, including cell swelling, warm temperatures, the synthetic phorbol ester 4alpha-phorbol 12,13-didecanoate (4alpha-PDD), and the endogenous lipid arachidonic acid (AA). Activation by cell swelling and AA requires cytochrome P450 (CYP) epoxygenase activity to convert AA to epoxyeicosatrienoic acids (EETs) such as 5,6-EET, 8,9-EET, which both act as direct TRPV4 agonists. To evaluate the role of TRPV4 and its modulation by the CYP pathway in vascular endothelial cells, we performed Ca2+ imaging and patch-clamp measurements on mouse aortic endothelial cells (MAECs) isolated from wild-type and TRPV4(-/-) mice. All TRPV4-activating stimuli induced robust Ca2+ responses in wild-type MAECs but not in MAECs isolated from TRPV4(-/-) mice. Upregulation of CYP2C expression by preincubation with nifedipine enhanced the responses to AA and cell swelling in wild-type MAECs, whereas responses to other stimuli remained unaffected. Conversely, inhibition of CYP2C9 activity with sulfaphenazole abolished the responses to AA and hypotonic solution (HTS). Moreover, suppression of EET hydrolysis using 1-adamantyl-3-cyclo-hexylurea or indomethacin, inhibitors of soluble epoxide hydrolases (sEHs), and cyclooxygenases, respectively, enhanced the TRPV4-dependent responses to AA, HTS, and EETs but not those to 4alpha-PDD or heat. Together, our data establish that CYP-derived EETs modulate the activity of TRPV4 channels in endothelial cells and shows the unraveling of novel modulatory pathways via CYP2C modulation and sEH inhibition.


Assuntos
Cálcio/metabolismo , Sistema Enzimático do Citocromo P-450/fisiologia , Células Endoteliais/metabolismo , Epóxido Hidrolases/fisiologia , Canais de Cátion TRPV/fisiologia , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Ácido 8,11,14-Eicosatrienoico/metabolismo , Animais , Células Cultivadas , Epóxido Hidrolases/antagonistas & inibidores , Camundongos , Nifedipino/farmacologia
10.
Proc Natl Acad Sci U S A ; 101(1): 396-401, 2004 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-14691263

RESUMO

TRPV4 is a Ca(2+)- and Mg(2+)-permeable cation channel within the vanilloid receptor subgroup of the transient receptor potential (TRP) family, and it has been implicated in Ca(2+)-dependent signal transduction in several tissues, including brain and vascular endothelium. TRPV4-activating stimuli include osmotic cell swelling, heat, phorbol ester compounds, and 5',6'-epoxyeicosatrienoic acid, a cytochrome p450 epoxygenase metabolite of arachidonic acid (AA). It is presently unknown how these distinct activators converge on opening of the channel. Here, we demonstrate that blockers of phospholipase A(2) (PLA(2)) and cytochrome p450 epoxygenase inhibit activation of TRPV4 by osmotic cell swelling but not by heat and 4alpha-phorbol 12,13-didecanoate. Mutating a tyrosine residue (Tyr-555) in the N-terminal part of the third transmembrane domain to an alanine strongly impairs activation of TRPV4 by 4alpha-phorbol 12,13-didecanoate and heat but has no effect on activation by cell swelling or AA. We conclude that TRPV4-activating stimuli promote channel opening by means of distinct pathways. Cell swelling activates TRPV4 by means of the PLA(2)-dependent formation of AA, and its subsequent metabolization to 5',6'-epoxyeicosatrienoic acid by means of a cytochrome p450 epoxygenase-dependent pathway. Phorbol esters and heat operate by means of a distinct, PLA(2)- and cytochrome p450 epoxygenase-independent pathway, which critically depends on an aromatic residue at the N terminus of the third transmembrane domain.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Canais Iônicos/metabolismo , Animais , Proteínas de Transporte de Cátions/agonistas , Proteínas de Transporte de Cátions/química , Proteínas de Transporte de Cátions/genética , Cátions/metabolismo , Linhagem Celular , Citocromo P-450 CYP2J2 , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/metabolismo , Inibidores Enzimáticos/farmacologia , Temperatura Alta , Humanos , Canais Iônicos/agonistas , Canais Iônicos/química , Canais Iônicos/genética , Camundongos , Mutagênese Sítio-Dirigida , Pressão Osmótica , Oxigenases/antagonistas & inibidores , Oxigenases/metabolismo , Ésteres de Forbol/farmacologia , Fosfolipases A/antagonistas & inibidores , Fosfolipases A/metabolismo , Fosforilação , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Canais de Cátion TRPV , Tirosina/química
11.
J Membr Biol ; 192(1): 1-8, 2003 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-12647029

RESUMO

Drosophila flies with the trp mutation exhibit impaired vision due to the lack of a specific Ca2+ influx pathway in the photoreceptors. The identification of the trp gene product as a Ca2+-permeable ion channel and the search for TRP homologues in flies, worms and mammals has opened the way to the discovery of a whole superfamily of cation channels, baptized TRP channels. In contrast to voltage-gated K+, Na+, or Ca2+ channels, with whom they share their transmembrane architecture, TRP channels are not activated by voltage but by a variety of signals including intra- and extracellular ligands, Ca2+-store depletion and mechanical or thermal stress. Due to the promiscuity of these gating mechanisms, TRP channels are privileged candidates as primary sensing molecules for the recognition and integration of physical and chemical signals from the environment. In this review we discuss recent evidence that implicates members of the TRP superfamily in sensory signal transduction.


Assuntos
Canais de Cálcio/classificação , Canais de Cálcio/fisiologia , Proteínas de Drosophila , Sensação/fisiologia , Transdução de Sinais/fisiologia , Animais , Drosophila , Humanos , Proteínas de Insetos/classificação , Proteínas de Insetos/fisiologia , Ativação do Canal Iônico/fisiologia , Estimulação Física , Estimulação Química , Canais de Cátion TRPC , Canais de Potencial de Receptor Transitório
12.
EMBO J ; 22(4): 776-85, 2003 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-12574114

RESUMO

The molecular assembly of the epithelial Ca(2+) channels (TRPV5 and TRPV6) was investigated to determine the subunit stoichiometry and composition. Immunoblot analysis of Xenopus laevis oocytes expressing TRPV5 and TRPV6 revealed two specific bands of 75 and 85-100 kDa, corresponding to the core and glycosylated proteins, respectively, for each channel. Subsequently, membranes of these oocytes were sedimented on sucrose gradients. Immuno blotting revealed that TRPV5 and TRPV6 complexes migrate with a mol. wt of 400 kDa, in line with a tetrameric structure. The tetrameric stoichiometry was confirmed in an electrophysiological analysis of HEK293 cells co-expressing concatemeric channels together with a TRPV5 pore mutant that reduced Cd(2+) sensitivity and voltage-dependent gating. Immuno precipitations using membrane fractions from oocytes co-expressing TRPV5 and TRPV6 demonstrated that both channels can form heteromeric complexes. Expression of all possible heterotetrameric TRPV5/6 complexes in HEK293 cells resulted in Ca(2+) channels that varied with respect to Ca(2+)-dependent inactivation, Ba(2+) selectivity and pharmacological block. Thus, Ca(2+)-transporting epithelia co-expressing TRPV5 and TRPV6 can generate a pleiotropic set of functional heterotetrameric channels with different Ca(2+) transport kinetics.


Assuntos
Canais de Cálcio/química , Cálcio/metabolismo , Epitélio/metabolismo , Animais , Canais de Cálcio/metabolismo , Epitélio/química , Rim/química , Rim/metabolismo , Camundongos , Testes de Precipitina , Estrutura Terciária de Proteína , Canais de Cátion TRPV
13.
Cell Calcium ; 31(6): 253-64, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12098215

RESUMO

Calcium influx into the cell from the extracellular medium is crucial for important processes including muscle contraction, secretion and gene expression. This calcium influx is mainly mediated through calcium influx channels, which on the basis of their activation mechanism can be subdivided in voltage-gated calcium channels, which have already been thoroughly characterized and non-voltage-gated calcium permeable channels. This latter group includes ion channels activated by binding of extra and intracellular messengers, mechanical stress or depletion of intracellular calcium stores. Currently little molecular data is available concerning this class of calcium influx channels. However, recent studies have indicated that members of the transient receptor potential (TRP) family of ion channels can function as calcium influx channels both in excitable and non-excitable tissues. On the basis of structural information the TRP family is subdivided in three main subfamilies: the TRPC (canonical) group, the TRPV (vanilloid) group and the TRPM (melastatin) group. The cloning and characterization of members of this cation channel family has exploded during recent years, leading to a plethora of data concerning TRPs in a variety of tissues and species, including mammals, insects and yeast. This review summarizes the currently available information concerning members of the TRP family expressed in mammalian tissues.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Canais Iônicos/metabolismo , Animais , Humanos , Canais Iônicos/classificação , Canais Iônicos/genética , Família Multigênica , Filogenia , Estrutura Secundária de Proteína , Distribuição Tecidual
14.
J Biol Chem ; 276(51): 47767-70, 2001 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-11687570

RESUMO

The calcium release-activated calcium channel (CRAC) is a highly Ca(2+)-selective ion channel that is activated on depletion of inositol triphosphate (IP(3))-sensitive intracellular Ca(2+) stores. It was recently reported that CaT1, a member of the TRP family of cation channels, exhibits the unique biophysical properties of CRAC, which led to the conclusion that CaT1 comprises all or part of the CRAC pore (Yue, L., Peng, J. B., Hediger, M. A., and Clapham, D. E. (2001) Nature 410, 705-709). Here, we directly compare endogenous CRAC with heterologously expressed CaT1 and show that they manifest several clearly distinct properties. CaT1 can be distinguished from CRAC in the following features: sensitivity to store-depleting agents; inward rectification in the absence of divalent cations; relative permeability to Na(+) and Cs(+); effect of 2-aminoethoxydiphenyl borate (2-APB). Moreover, CaT1 displays a mode of voltage-dependent gating that is fully absent in CRAC and originates from the voltage-dependent binding/unbinding of Mg(2+) inside the channel pore. Our results imply that the pores of CaT1 and CRAC are not identical and indicate that CaT1 is a Mg(2+)-gated channel not directly related to CRAC.


Assuntos
Canais de Cálcio/fisiologia , Linhagem Celular , Humanos , Técnicas de Patch-Clamp , Canais de Cátion TRPV
15.
Neuron ; 31(4): 581-91, 2001 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-11545717

RESUMO

Secretory vesicles dock at the plasma membrane before Ca(2+) triggers their exocytosis. Exocytosis requires the assembly of SNARE complexes formed by the vesicle protein Synaptobrevin and the membrane proteins Syntaxin-1 and SNAP-25. We analyzed the role of Munc18-1, a cytosolic binding partner of Syntaxin-1, in large dense-core vesicle (LDCV) secretion. Calcium-dependent LDCV exocytosis was reduced 10-fold in mouse chromaffin cells lacking Munc18-1, but the kinetic properties of the remaining release, including single fusion events, were not different from controls. Concomitantly, mutant cells displayed a 10-fold reduction in morphologically docked LDCVs. Moreover, acute overexpression of Munc18-1 in bovine chromaffin cells increased the amount of releasable vesicles and accelerated vesicle supply. We conclude that Munc18-1 functions upstream of SNARE complex formation and promotes LDCV docking.


Assuntos
Células Cromafins/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Vesículas Sinápticas/metabolismo , Proteínas de Transporte Vesicular , Animais , Antígenos de Superfície/metabolismo , Bovinos , Membrana Celular/metabolismo , Células Cromafins/ultraestrutura , Exocitose/fisiologia , Feminino , Feto/citologia , Deleção de Genes , Expressão Gênica/fisiologia , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Mutantes , Microscopia Eletrônica , Proteínas Munc18 , Proteínas do Tecido Nervoso/genética , Técnicas de Patch-Clamp , Gravidez , Sintaxina 1
16.
Proc Natl Acad Sci U S A ; 98(20): 11680-5, 2001 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-11562488

RESUMO

Synaptotagmin I is a synaptic vesicle-associated protein essential for synchronous neurotransmission. We investigated its impact on the intracellular Ca(2+)-dependence of large dense-core vesicle (LDCV) exocytosis by combining Ca(2+)-uncaging and membrane capacitance measurements in adrenal slices from mouse synaptotagmin I null mutants. Synaptotagmin I-deficient chromaffin cells displayed prolonged exocytic delays and slow, yet Ca(2+)-dependent fusion rates, resulting in strongly reduced LDCV release in response to short depolarizations. Vesicle recruitment, the shape of individual amperometric events, and endocytosis appeared unaffected. These findings demonstrate that synaptotagmin I is required for rapid, highly Ca(2+)-sensitive LDCV exocytosis and indicate that it regulates the equilibrium between a slowly releasable and a readily releasable state of the fusion machinery. Alternatively, synaptotagmin I could function as calcium sensor for the readily releasable pool, leading to the destabilization of the pool in its absence.


Assuntos
Glândulas Suprarrenais/fisiologia , Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/metabolismo , Células Cromafins/fisiologia , Exocitose/fisiologia , Líquido Intracelular/fisiologia , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Animais Recém-Nascidos , Proteínas de Ligação ao Cálcio/deficiência , Técnicas In Vitro , Glicoproteínas de Membrana/deficiência , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Sinaptotagmina I , Sinaptotagminas
17.
Neuron ; 29(3): 681-90, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11301027

RESUMO

Release of neurotransmitter at the inner hair cell (IHC) afferent synapse is a fundamental step in translating sound into auditory nerve excitation. To study the Ca2+ dependence of the underlying vesicle fusion and subsequent endocytosis, we combined Ca2+ uncaging with membrane capacitance measurements in mouse IHCs. Rapid elevations in [Ca2+]i above 8 microM caused a biphasic capacitance increase corresponding to the fusion of approximately 40,000 vesicles. The kinetics of exocytosis displayed a fifth-order Ca2+ dependence reaching maximal rates of >3 x 10(7) vesicle/s. Exocytosis was always followed by slow, compensatory endocytosis (tau congruent with 15 s). Higher [Ca2+]i increased the contribution of a faster mode of endocytosis with a Ca2+ independent time constant of approximately 300 ms. These properties provide for rapid and sustained transmitter release from this large presynaptic terminal.


Assuntos
Cálcio/fisiologia , Endocitose/fisiologia , Exocitose/fisiologia , Células Ciliadas Auditivas Internas/fisiologia , Sinapses/fisiologia , Animais , Cálcio/metabolismo , Cálcio/farmacologia , Canais de Cálcio/fisiologia , Membrana Celular/fisiologia , Condutividade Elétrica , Ativação do Canal Iônico/fisiologia , Cinética , Camundongos , Neurotransmissores/metabolismo , Fotólise
18.
J Physiol ; 529 Pt 2: 385-94, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11101648

RESUMO

This study investigated the volume-regulated anion channel (VRAC) of human cervical cancer SiHa cells under various culture conditions, testing the hypothesis that the progression of the cell cycle is accompanied by differential expression of VRAC activity. Exponentially growing SiHa cells expressed VRACs, as indicated by the presence of large outwardly rectifying currents activated by hypotonic stress with the anion permeability sequence I- > Br- > Cl-. VRACs were potently inhibited by tamoxifen with an IC50 of 4.6 [mu]M. Fluorescence-activated cell sorting (FACS) experiments showed that 59 +/- 0.5, 5 +/- 0.5 and 36 +/- 1.1% of unsynchronized, exponentially growing cervical cancer SiHa cells were in G0/G1, S and G2/M stage, respectively. Treatment with aphidicolin (5 [mu]M) arrested 88 +/- 1.4% of cells at the G0/G1 stage. Arrest of cell growth in the G0/G1 phase was accompanied by a significant decrease of VRAC activity. The normalized hypotonicity-induced current decreased from 48 +/- 5.2 pA pF-1 at +100 mV in unsynchronized cells to 15 +/- 2.6 pA pF-1 at +100 mV in aphidicolin-treated cells. After removal of aphidicolin, culturing in medium containing 10% fetal calf serum triggered a rapid re-entry into the cell cycle and a concomitant recovery of VRAC density. Pharmacological blockade of VRACs by tamoxifen or NPPB caused proliferating cervical cancer cells to arrest in the G0/G1 stage, suggesting that activity of this channel is critical for G1/S checkpoint progression. This study provides new information on the functional significance of VRACs in the cell cycle clock of human cervical cancer cells.


Assuntos
Ciclo Celular , Tamanho Celular , Canais Iônicos/metabolismo , Neoplasias do Colo do Útero/patologia , Cloretos/metabolismo , Meios de Cultura/farmacologia , Condutividade Elétrica , Feminino , Humanos , Soluções Hipotônicas/farmacologia , Canais Iônicos/efeitos dos fármacos , Nitrobenzoatos/farmacologia , Tamoxifeno/farmacologia , Células Tumorais Cultivadas , Neoplasias do Colo do Útero/metabolismo
19.
J Neurosci ; 20(22): 8377-83, 2000 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11069944

RESUMO

Maturation of exocytic vesicles to the release-ready state is regulated by several factors, including intracellular calcium concentration ([Ca(2+)](int)) and the state of protein phosphorylation. Here we investigated the effects of temperature on the recovery from depletion of the readily releasable pool (RRP) of vesicles in adrenal chromaffin cells. Exocytosis and [Ca(2+)](int) were monitored by combined membrane capacitance and fura-2 measurements. At higher temperatures, a faster pool refilling and a larger RRP size were observed. The time constants of the recovery from depletion ranged from 3.6 to 1.1 sec (22 and 37 degrees C, respectively) yielding a Q(10) of 2.3. The changes of the Ca(2+) signal between the different temperatures could not account for the differences in recovery kinetics. At 32 and 37 degrees C, we observed a transient overfilling of the RRP after pool depletion, which stands in clear contrast to the sustained secretory depression seen at lower temperatures. The overshoot in RRP size was very prominent in cells with lower basal [Ca(2+)](int), hence with a large difference between prestimulus and poststimulus [Ca(2+)](int). In cells with higher basal [Ca(2+)](int), the pool was larger under steady-state conditions but showed less overfilling on stimulation. We conclude that vesicle maturation is markedly accelerated at physiological temperature, thus allowing for a rapid adaptation of the pool size to the relatively short-lived Ca(2+) transient.


Assuntos
Células Cromafins/metabolismo , Fura-2/análogos & derivados , Vesículas Secretórias/metabolismo , Animais , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Bovinos , Membrana Celular/metabolismo , Células Cultivadas , Células Cromafins/citologia , Condutividade Elétrica , Exocitose/fisiologia , Corantes Fluorescentes , Líquido Intracelular/metabolismo , Técnicas de Patch-Clamp , Tempo de Reação/fisiologia , Temperatura
20.
Gen Pharmacol ; 34(2): 107-16, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10974418

RESUMO

Osmotic cell swelling activates an outwardly rectifying Cl(-) current in endothelial cells that is mediated by volume-regulated anion channels (VRACs). In the past, we have shown that serum-induced proliferation of endothelial cells is arrested in the presence of compounds that potently block the endothelial VRACs. Here we report on the effects of four chemically distinct VRAC blockers [5-nitro-2-(3-phenylpropylamino)benzoic acid] (NPPB), mibefradil, tamoxifen, and clomiphene-on several models of experimental angiogenesis. Mibefradil (20 microM), NPPB (100 microM), tamoxifen (20 microM), and clomiphene (20 microM) inhibited tube formation by rat microvascular endothelial cells plated on matrigel by 42.9 +/- 8.8%, 25.3 +/- 10.4%, 32.2 +/- 4.5%, and 20 +/- 5.8%, respectively (p < 0.05). Additionally, NPPB (50-100 microM) and mibefradil (10-30 microM) significantly inhibited bFGF (10 ng/ml) + TNFalpha (2.5 ng/ml)-stimulated microvessel formation by human microvascular endothelial cells plated on fibrin by 30-70%. Furthermore, NPPB, mibefradil, and clomiphene concentration dependently inhibited spontaneous microvessel formation in the rat aorta-ring assay and vessel development in the chick chorioallantoic membrane assay. These results suggest that VRAC blockers are potent inhibitors of angiogenesis and thus might serve as therapeutic tools in tumor growth and other angiogenesis-dependent diseases.


Assuntos
Inibidores da Angiogênese/farmacologia , Canais Iônicos/antagonistas & inibidores , Neovascularização Fisiológica/efeitos dos fármacos , Alantoide/irrigação sanguínea , Alantoide/efeitos dos fármacos , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/crescimento & desenvolvimento , Bovinos , Células Cultivadas , Embrião de Galinha , Córion/irrigação sanguínea , Córion/efeitos dos fármacos , Clomifeno/farmacologia , Colágeno , Combinação de Medicamentos , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Fibrina , Humanos , Técnicas In Vitro , Laminina , Masculino , Mibefradil/farmacologia , Nitrobenzoatos/farmacologia , Proteoglicanas , Ratos , Ratos Wistar , Tamoxifeno/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...