Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Br J Pharmacol ; 180(21): 2736-2749, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37254803

RESUMO

BACKGROUND AND PURPOSE: In macrophages, transient receptor potential vanilloid 2 (TRPV2) channel contributes to various cellular processes such as cytokine production, differentiation, phagocytosis and migration. Due to a lack of selective pharmacological tools, its function in immunological processes is not well understood and the identification of novel and selective TRPV2 modulators is highly desirable. EXPERIMENTAL APPROACH: Novel and selective TRPV2 modulators were identified by screening a compound library using Ca2+ influx assays with human embryonic kidney 293 (HEK293) cells heterologously expressing rat TRPV2. Hits were further characterized and validated with Ca2+ influx and electrophysiological assays. Phagocytosis and migration of macrophages were analysed and the contribution of TRPV2 to the generation of Ca2+ microdomains was studied by total internal reflection fluorescence microscopy (TIRFM). KEY RESULTS: The compound IV2-1, a dithiolane derivative (1,3-dithiolan-2-ylidene)-4-methyl-5-phenylpentan-2-one), is a potent inhibitor of heterologously expressed TRPV2 channels (IC50 = 6.3 ± 0.7 µM) but does not modify TRPV1, TRPV3 or TRPV4 channels. IV2-1 also inhibits TRPV2-mediated Ca2+ influx in macrophages. IV2-1 inhibits macrophage phagocytosis along with valdecoxib and after siRNA-mediated knockdown. Moreover, TRPV2 inhibition inhibits lipopolysaccharide-induced migration of macrophages whereas TRPV2 activation promotes migration. After activation, TRPV2 shapes Ca2+ microdomains predominantly at the margin of macrophages, which are important cellular regions to promote phagocytosis and migration. CONCLUSIONS AND IMPLICATIONS: IV2-1 is a novel TRPV2-selective blocker and underline the role of TRPV2 in macrophage-mediated phagocytosis and migration. Furthermore, we provide evidence that TRPV2 activation generates Ca2+ microdomains, which may be involved in phagocytosis and migration of macrophages.


Assuntos
Lipopolissacarídeos , Macrófagos , Humanos , Ratos , Animais , Lipopolissacarídeos/farmacologia , Células HEK293 , Fagocitose , Expressão Gênica , Canais de Cátion TRPV/genética
2.
Cell Calcium ; 106: 102640, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36030694

RESUMO

The transient receptor potential cation channel, subfamily M, members 6 and 7 (TRPM6 and TRPM7) are homologous membrane proteins encompassing cation channel units fused to cytosolic serine/threonine-protein kinase domains. Clinical studies and experiments with animal disease models suggested that selective inhibition of TRPM6 and TRPM7 currents might be beneficial for subjects with immune and cardiovascular disorders, tumours and other pathologies, but the suitable pharmacological toolkit remains underdeveloped. The present study identified small synthetic molecules acting specifically on the channel moieties of TRPM6 and TRPM7. Using electrophysiological analysis in conjunction with Ca2+ imaging, we show that iloperidone and ifenprodil inhibit the channel activity of recombinant TRPM6 with IC50 values of 0.73 and 3.33 µM, respectively, without an impact on the TRPM7 channel. We also found that VER155008 suppresses the TRPM7 channel with an IC50 value of 0.11 µM but does not affect TRPM6. Finally, the effects of iloperidone and VER155008 were found to be suitable for blocking native endogenous TRPM6 and TRPM7 in a collection of mouse and human cell models. Hence, the identification of iloperidone, ifenprodil, and VER155008 allows for the first time to selectively manipulate TRPM6 and TRPM7 currents.


Assuntos
Canais de Cátion TRPM , Animais , Humanos , Isoxazóis/farmacologia , Magnésio/metabolismo , Camundongos , Piperidinas/farmacologia , Proteínas Serina-Treonina Quinases , Nucleosídeos de Purina/farmacologia , Canais de Cátion TRPM/efeitos dos fármacos , Canais de Cátion TRPM/metabolismo , Canais de Potencial de Receptor Transitório/efeitos dos fármacos , Canais de Potencial de Receptor Transitório/metabolismo
3.
Br J Pharmacol ; 179(24): 5290-5304, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35916168

RESUMO

BACKGROUND AND PURPOSE: Ca2+ signalling mediated by the thermosensitive, non-selective, Ca2+ -permeable transient receptor potential channel TRPV3 is assumed to play a critical role in regulating several aspects of skin functions, such as keratinocyte proliferation, differentiation, skin barrier formation and wound healing. Studying the function of TRPV3 in skin homeostasis, however, is still constrained by a lack of potent and selective pharmacological modulators of TRPV3. EXPERIMENTAL APPROACH: By screening an in-house compound library using fluorometric intracellular Ca2+ assays, we identified two chemically related hits. The more potent and efficient TRPV3 activator 2-(2-chloro-3-isopropylcyclopent-2-en-1-yl)-4-methylphenol (KS0365) was further evaluated in fluo-4-assisted Ca2+ assays, different Ca2+ imaging approaches, electrophysiological studies, cytotoxicity and migration assays. KEY RESULTS: KS0365 activated recombinant and native mouse TRPV3 more potently and with a higher efficacy compared with 2-APB and did not activate TRPV2 or TRPV4 channels. The activation of TRPV3 by KS0365 super-additively accelerated the EGF-induced keratinocyte migration, which was inhibited by the TRP channel blocker ruthenium red or by siRNA-mediated TRPV3 knockdown. Moreover, KS0365 induced strong Ca2+ responses in migrating front cells and in leading edges of keratinocytes. CONCLUSIONS AND IMPLICATIONS: The selective TRPV3 activator KS0365 triggers increases in [Ca2+ ]i with most prominent signals in the leading edge and accelerates migration of keratinocytes. TRPV3 activators may promote re-epithelialization upon skin wounding.


Assuntos
Queratinócitos , Canais de Cátion TRPV , Animais , Camundongos , Diferenciação Celular , Movimento Celular , Proliferação de Células , Canais de Cátion TRPV/agonistas , Canais de Cátion TRPV/fisiologia , Cicatrização/fisiologia
4.
Eur J Pharmacol ; 915: 174702, 2022 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-34919887

RESUMO

The transient receptor potential vanilloid 2 (TRPV2) channel is broadly expressed in a multitude of different tissues and is implicated in the pathology of several diseases, such as the progression of different cancer types. However, a lack of specific, potent and non-toxic TRPV2 activators and inhibitors complicate further studies to clarify the role of TRPV2. We here present valdecoxib as a novel inhibitor of heterologously expressed rat TRPV2 channels in HEK293 cells and native TRPV2 channels, endogenously expressed in the rat basophilic leukemia (RBL-2H3) cell line. Fluorometric assays reveal an IC50 of 9 µM and 11 µM for TRPV2 in HEK293 and RBL-2H3 cells, respectively. Closely related TRPV1, TRPV3 or TRPV4 channels are not blocked by valdecoxib. The inhibition is reversible and direct as confirmed by whole-cell and excised inside-out electrophysiological recordings. Other cyclooxygenase-2 inhibitors do not affect TRPV2 activity. Furthermore, we demonstrate that the combined application of 2-aminoethoxydiphenyl borate (2-APB) and probenecid at concentrations, which, on their own, elicit only small TRPV2 currents, act in a highly synergistic manner when applied simultaneously. Taken together, we here provide novel tools and chemical lead structures for further studying TRPV2 channel function in native tissues.


Assuntos
Isoxazóis , Sulfonamidas
5.
Cell Calcium ; 92: 102310, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33161279

RESUMO

TRPV3 is a Ca2+-permeable cation channel, prominently expressed by keratinocytes where it contributes to maintaining the skin barrier, skin regeneration, and keratinocyte differentiation. However, much less is known about its physiological function in other tissues and there is still a need for identifying novel and efficient TRPV3 channel blockers. By screening a compound library, we identified 26E01 as a novel TRPV3 blocker. 26E01 blocks heterologously expressed TRPV3 channels overexpressed in HEK293 cells as assessed by fluorometric intracellular free Ca2+ assays (IC50 = 8.6 µM) but does not affect TRPV1, TRPV2 or TRPV4 channels. Electrophysiological whole-cell recordings confirmed the reversible block of TRPV3 currents by 26E01, which was also effective in excised inside-out patches, hinting to a rather direct mode of action. 26E01 suppresses endogenous TRPV3 currents in the mouse 308 keratinocyte cell line and in the human DLD-1 colon carcinoma cell line (IC50 = 12 µM). In sections of the gastrointestinal epithelium of mice, the expression of TRPV3 mRNA follows a gradient along the gastrointestinal tract, with the highest expression in the distal colon. 26E01 efficiently attenuates 2-aminoethoxydiphenyl borate-induced calcium influx in primary colonic epithelial cells isolated from the distal colon. As 26E01 neither shows toxic effects on DLD-1 cells at concentrations of up to 100 µM in MTT assays nor on mouse primary colonic crypts as assessed by calcein-AM/propidium iodide co-staining, it may serve as a useful tool to further study the physiological function of TRPV3 in various tissues.


Assuntos
Colo/citologia , Células Epiteliais/metabolismo , Canais de Cátion TRPV/antagonistas & inibidores , Animais , Compostos de Boro/farmacologia , Células Epiteliais/efeitos dos fármacos , Células HEK293 , Temperatura Alta , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Camundongos , Canais de Cátion TRPV/metabolismo
6.
Cell Calcium ; 66: 10-18, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28807145

RESUMO

The transient receptor potential canonical channel 5 (TRPC5) is a Ca2+-permeable ion channel, which is predominantly expressed in the brain. TRPC5-deficient mice exhibit a reduced innate fear response and impaired motor control. In addition, outgrowth of hippocampal and cerebellar neurons is retarded by TRPC5. However, pharmacological evidence of TRPC5 function on cellular or organismic levels is sparse. Thus, there is still a need for identifying novel and efficient TRPC5 channel modulators. We, therefore, screened compound libraries and identified the glucocorticoid methylprednisolone and N-[3-(adamantan-2-yloxy)propyl]-3-(6-methyl-1,1-dioxo-2H-1λ6,2,4-benzothiadiazin-3-yl)propanamide (BTD) as novel TRPC5 activators. Comparisons with closely related chemical structures from the same libraries indicate important substructures for compound efficacy. Methylprednisolone activates TRPC5 heterologously expressed in HEK293 cells with an EC50 of 12µM, while BTD-induced half-maximal activation is achieved with 5-fold lower concentrations, both in Ca2+ assays (EC50=1.4µM) and in electrophysiological whole cell patch clamp recordings (EC50=1.3 µM). The activation resulting from both compounds is long lasting, reversible and sensitive to clemizole, a recently established TRPC5 inhibitor. No influence of BTD on homotetrameric members of the remaining TRPC family was observed. On the main sensory TRP channels (TRPA1, TRPV1, TRPM3, TRPM8) BTD exerts only minor activity. Furthermore, BTD can activate heteromeric channel complexes consisting of TRPC5 and its closest relatives TRPC1 or TRPC4, suggesting a high selectivity of BTD for channel complexes bearing at least one TRPC5 subunit.


Assuntos
Benzotiadiazinas/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Metilprednisolona/farmacologia , Canais de Cátion TRPC/metabolismo , Animais , Benzotiadiazinas/química , Sinalização do Cálcio/efeitos dos fármacos , Células HEK293 , Humanos , Metilprednisolona/química , Camundongos , Microscopia Confocal , Técnicas de Patch-Clamp , Fosfoinositídeo Fosfolipase C/metabolismo , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/metabolismo , Subunidades Proteicas/agonistas , Subunidades Proteicas/metabolismo , Canais de Cátion TRPC/agonistas , Canais de Cátion TRPC/genética
7.
Sci Rep ; 7(1): 5447, 2017 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-28710476

RESUMO

Phospholipids occurring in cell membranes and lipoproteins are converted into oxidized phospholipids (OxPL) by oxidative stress promoting atherosclerotic plaque formation. Here, OxPL were characterized as novel targets in acute and chronic inflammatory pain. Oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine (OxPAPC) and its derivatives were identified in inflamed tissue by mass spectrometry and binding assays. They elicited calcium influx, hyperalgesia and induced pro-nociceptive peptide release. Genetic, pharmacological and mass spectrometric evidence in vivo as well as in vitro confirmed the role of transient receptor potential channels (TRPA1 and TRPV1) as OxPAPC targets. Treatment with the monoclonal antibody E06 or with apolipoprotein A-I mimetic peptide D-4F, capturing OxPAPC in atherosclerosis, prevented inflammatory hyperalgesia, and in vitro TRPA1 activation. Administration of D-4F or E06 to rats profoundly ameliorated mechanical hyperalgesia and inflammation in collagen-induced arthritis. These data reveal a clinically relevant role for OxPAPC in inflammation offering therapy for acute and chronic inflammatory pain treatment by scavenging OxPAPC.


Assuntos
Anticorpos Monoclonais/farmacologia , Apolipoproteína A-I/farmacologia , Artrite Experimental/tratamento farmacológico , Hiperalgesia/tratamento farmacológico , Dor/tratamento farmacológico , Fosfatidilcolinas/antagonistas & inibidores , Canal de Cátion TRPA1/genética , Canais de Cátion TRPV/genética , Animais , Artrite Experimental/induzido quimicamente , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Colágeno Tipo II/administração & dosagem , Feminino , Expressão Gênica , Células HEK293 , Membro Posterior , Humanos , Hiperalgesia/induzido quimicamente , Hiperalgesia/metabolismo , Hiperalgesia/patologia , Masculino , Nociceptividade/efeitos dos fármacos , Nociceptividade/fisiologia , Dor/induzido quimicamente , Dor/metabolismo , Dor/patologia , Técnicas de Patch-Clamp , Fosfatidilcolinas/metabolismo , Fosfatidilcolinas/farmacologia , Ratos , Ratos Endogâmicos Lew , Ratos Wistar , Canal de Cátion TRPA1/antagonistas & inibidores , Canal de Cátion TRPA1/metabolismo , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/metabolismo
8.
Br J Pharmacol ; 174(16): 2696-2705, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28567799

RESUMO

BACKGROUND AND PURPOSE: The transient receptor potential vanilloid 3 (TRPV3) channel is a heat-sensitive ion channel, which is predominantly expressed in keratinocytes. TRPV3 channels are involved in numerous physiological and pathophysiological processes within the skin, including cutaneous nociception, temperature sensation and development of itch. The role of TRPV3 channels in such processes is poorly understood; therefore, the establishment of selective modulators of TRPV3 channels is highly desirable. EXPERIMENTAL APPROACH: Novel TRPV3-modulating compounds were identified using fluorometric intracellular Ca2+ assays and further evaluated with electrophysiological techniques. KEY RESULTS: TRPV3 activity, elicited by 2-aminoethoxydiphenyl borate (2-APB), was efficaciously enhanced by deracoxib and celecoxib, two COX-2-selective inhibitors. They exerted their potentiating effect via a direct interaction with TRPV3 as evident from excised inside-out recordings. Structurally-related COX-2 inhibitors affected TRPV3 channel gating to a much lesser degree. Similar results were obtained in HEK293 cells stably expressing cyan fluorescent protein-tagged mouse TRPV3 channels and in a mouse keratinocyte cell line, endogenously expressing TRPV3. The effects of celecoxib and deracoxib on TRPV3 were dependent on the stimulus used to activate TRPV3. While 2-APB and heat-activated TRPV3 channels were potentiated by celecoxib, carvacrol-activated channels were inhibited by celecoxib. CONCLUSIONS AND IMPLICATIONS: We identified a new class of drugs that modulate TRPV3 channels. The most potent compound celecoxib is an approved analgesic and anti-inflammatory drug, which is currently being investigated for its topical application in the treatment of skin cancer. As TRPV3 is highly expressed in skin, celecoxib might affect TRPV3 activity in vivo when used at high local concentrations.


Assuntos
Celecoxib/farmacologia , Inibidores de Ciclo-Oxigenase 2/farmacologia , Sulfonamidas/farmacologia , Canais de Cátion TRPV/fisiologia , Animais , Compostos de Boro , Linhagem Celular , Células HEK293 , Temperatura Alta , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/fisiologia , Camundongos
9.
Anal Bioanal Chem ; 408(29): 8529-8538, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27722942

RESUMO

The analysis of receptor activity, especially in its native cellular environment, has always been of great interest to evaluate its intrinsic but also downstream biological activity. An important group of cellular receptors are ion channels. Since they are involved in a broad range of crucial cell functions, they represent important therapeutic targets. Thus, novel analytical techniques for the quantitative monitoring and screening of biological receptor activity are of great interest. In this context, we developed an impedance spectroscopy-based label-free and non-invasive monitoring system that enabled us to analyze the activation of the transient receptor potential channel Vanilloid 1 (TRPV1) in detail. TRPV1 channel activation by capsaicin resulted in a reproducible impedance decrease. Moreover, concentration response curves with an EC50 value of 0.9 µM could be determined. Control experiments with non TRPV1 channel expressing HEK cells as well as experiments with the TRPV1 channel blocker ruthenium red validated the specificity of the observed impedance decrease. More strikingly, through correlative studies with a cytoskeleton restructuring inhibitor mixture and equivalent circuit analysis of the acquired impedance spectra, we could quantitatively discriminate between the direct TRPV1 channel activation and downstream-induced biological effects. In summary, we developed a quantitative impedimetric monitoring system for the analysis of TRPV1 channel activity as well as downstream-induced biological activity in living cells. It has the capabilities to identify novel ion channel activators as well as inhibitors for the TRPV1 channel but could also easily be applied to other ion channel-based receptors.


Assuntos
Capsaicina/farmacologia , Espectroscopia Dielétrica/métodos , Canais de Cátion TRPV/metabolismo , Eletrodos , Células HEK293 , Humanos , Reprodutibilidade dos Testes , Rutênio Vermelho/farmacologia , Sensibilidade e Especificidade , Canais de Cátion TRPV/antagonistas & inibidores
10.
J Nat Prod ; 79(4): 697-703, 2016 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-26905390

RESUMO

A series of seven oxyprenylated phenylpropanoids and naphthoquinones were tested regarding their ability to activate transient receptor potential ankyrin subtype 1 channel (TRPA1). Three of the assayed compounds, namely, boropinal (3), juglone (5), and plumbagin (7), acted as strong modulators of TRPA1 channels with EC50 values of 9.8, 1.7, and 0.5 µM, respectively, as assessed by Ca(2+) assays. Moreover, the compounds elicited TRPA1 currents in electrophysiological whole cell recordings. We additionally provide evidence that plumbagin activated TRPA1-positive neurons isolated from mouse dorsal root ganglion neurons but did not affect sensory neurons from TRPA1-deficient mice. The high potencies of plumbagin and juglone to activate TRPA1 channels may explain the molecular basis of the mucosal irritant properties of these compounds as well as of related naphthoquinones and phytopreparations, as widely reported in the literature.


Assuntos
Naftoquinonas/farmacologia , Fenilpropionatos/farmacologia , Canais de Potencial de Receptor Transitório/antagonistas & inibidores , Animais , Anquirinas , Cálcio/análise , Cálcio/metabolismo , Cumarínicos/química , Cumarínicos/farmacologia , Feminino , Gânglios Espinais/efeitos dos fármacos , Células HEK293 , Humanos , Masculino , Camundongos , Estrutura Molecular , Naftoquinonas/química , Fenilpropionatos/química , Células Receptoras Sensoriais/efeitos dos fármacos , Canal de Cátion TRPA1
11.
Mol Pharmacol ; 86(5): 514-21, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25140002

RESUMO

Canonical transient receptor potential channel 5 (TRPC5) is a nonselective, Ca(2+)-permeable cation channel that belongs to the large family of transient receptor potential channels. It is predominantly found in the central nervous system with a high expression density in the hippocampus, the amygdala, and the frontal cortex. Several studies confirm that TRPC5 channels are implicated in the regulation of neurite length and growth cone morphology. We identified clemizole as a novel inhibitor of TRPC5 channels. Clemizole efficiently blocks TRPC5 currents and Ca(2+) entry in the low micromolar range (IC50 = 1.0-1.3 µM), as determined by fluorometric intracellular free Ca(2+) concentration ([Ca(2+)]i) measurements and patch-clamp recordings. Clemizole blocks TRPC5 currents irrespectively of the mode of activation, for example, stimulation of G protein-coupled receptors, hypo-osmotic buffer conditions, or by the direct activator riluzole. Electrophysiological whole-cell recordings revealed that the block was mostly reversible. Moreover, clemizole was still effective in blocking TRPC5 single channels in excised inside-out membrane patches, hinting to a direct block of TRPC5 by clemizole. Based on fluorometric [Ca(2+)]i measurements, clemizole exhibits a sixfold selectivity for TRPC5 over TRPC4ß (IC50 = 6.4 µM), the closest structural relative of TRPC5, and an almost 10-fold selectivity over TRPC3 (IC50 = 9.1 µM) and TRPC6 (IC50 = 11.3 µM). TRPM3 and M8 as well as TRPV1, V2, V3, and V4 channels were only weakly affected by markedly higher clemizole concentrations. Clemizole was not only effective in blocking heterologously expressed TRPC5 homomers but also TRPC1:TRPC5 heteromers as well as native TRPC5-like currents in the U-87 glioblastoma cell line.


Assuntos
Benzimidazóis/farmacologia , Canais de Cátion TRPC/antagonistas & inibidores , Canais de Potencial de Receptor Transitório/antagonistas & inibidores , Cálcio/metabolismo , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Glioblastoma/metabolismo , Células HEK293 , Humanos , Técnicas de Patch-Clamp/métodos , Receptores Acoplados a Proteínas G/metabolismo , Canais de Cátion TRPC/metabolismo
12.
Cell Calcium ; 55(4): 200-7, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24636274

RESUMO

Transient receptor potential A1 (TRPA1) is widely expressed throughout the human and animal organism, including the dorsal root ganglia as well as the bladder, stomach and small intestine. Here, we examined the effect of three platelet aggregation inhibitors on TRPA1: ticlopidine, clopidogrel and prasugrel. Utilising fluorometric Ca(2+) influx analysis and electrophysiological whole cell measurements in TRPA1-expressing HEK293 and in human enterochromaffin-like QGP-1 cells, we found that ticlopidine, clopidogrel and prasugrel are direct activators of TRPA1. Although this polymodal channel commonly contributes to the perception of pain, temperature and chemical irritants, recent studies provide evidence for its involvement in the release of serotonin (5-HT) from enterochromaffin cells. Therefore, we further investigated the ability of ticlopidine, clopidogrel and prasugrel to stimulate 5-HT release from QGP-1 cells. We could determine 5-HT in supernatants from cultured QGP-1 cells upon treatment with ticlopidine and clopidogrel but not with prasugrel. These findings indicate that a robust TRPA1 activation by ticlopidine and clopidogrel correlates with the stimulatory effect on the secretion of 5-HT. As recipients of ticlopidine and clopidogrel frequently complain about gastrointestinal adverse events such as nausea, vomiting and diarrhoea, an activation of TRPA1 may contribute to adverse effects of such drugs in the digestive system.


Assuntos
Canais de Cálcio/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Inibidores da Agregação Plaquetária/farmacologia , Piridinas/farmacologia , Canais de Potencial de Receptor Transitório/metabolismo , Cálcio/metabolismo , Canais de Cálcio/genética , Linhagem Celular , Clopidogrel , Celulas Tipo Enterocromafim/efeitos dos fármacos , Celulas Tipo Enterocromafim/metabolismo , Células HEK293 , Humanos , Mutação , Proteínas do Tecido Nervoso/genética , Piperazinas/química , Piperazinas/farmacologia , Inibidores da Agregação Plaquetária/química , Cloridrato de Prasugrel , Piridinas/química , Serotonina/metabolismo , Canal de Cátion TRPA1 , Tiofenos/química , Tiofenos/farmacologia , Ticlopidina/análogos & derivados , Ticlopidina/química , Ticlopidina/farmacologia , Imagem com Lapso de Tempo , Canais de Potencial de Receptor Transitório/genética
13.
Br J Pharmacol ; 171(1): 158-70, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24117252

RESUMO

BACKGROUND AND PURPOSE: The transient receptor potential channel C5 (TRPC5) is a Ca(2+)-permeable cation channel, which is predominantly expressed in the brain. TRPC5 is activated in a PLC-dependent manner by, as yet, unidentified endogenous messengers. Recently, modulators of TRPC5, like Ca(2+), pH and phospholipids, have been identified. However, the role of TRPC5 in vivo is only poorly understood. Novel specific modulators of TRPC5 might help to elucidate its function. EXPERIMENTAL APPROACH: Novel modulators of TRPC5 were identified in a compound screening of approved drugs and natural compounds. The potency and selectivity of TRPC5-activating compounds were determined by fluorometric calcium imaging. The biophysical properties of channel activation by these compounds were analysed using electrophysiological measurements. KEY RESULTS: Riluzole was identified as a novel activator of TRPC5 (EC50 9.2 ± 0.5 µM) and its mechanism of action was shown to be independent of G protein signalling and PLC activity. Riluzole-induced TRPC5 currents were potentiated by La(3+) and, utilizing TRPC5 mutants that lack La(3+) binding sites, it was confirmed that riluzole and La(3+) activate TRPC5 by different mechanisms. Recordings of excised inside-out patches revealed a relatively direct effect of riluzole on TRPC5. CONCLUSIONS AND IMPLICATIONS: Riluzole can activate TRPC5 heterologously expressed in HEK293 cells as well as those endogenously expressed in the U-87 glioblastoma cell line. Riluzole does not activate any other member of the TRPC family and could, therefore, despite its action on other ion channels, be a useful pharmacological tool for identifying TRPC5-specific currents in immortalized cell lines or in acutely isolated primary cells.


Assuntos
Ativação do Canal Iônico/efeitos dos fármacos , Riluzol/farmacologia , Canais de Cátion TRPC/agonistas , Fosfolipases Tipo C/metabolismo , Animais , Sítios de Ligação , Sinalização do Cálcio/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Lantânio/metabolismo , Potenciais da Membrana , Camundongos , Microscopia de Fluorescência , Microscopia de Vídeo , Dados de Sequência Molecular , Mutação , Interferência de RNA , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/metabolismo , Fatores de Tempo , Transfecção
14.
Mol Pharmacol ; 83(2): 542-51, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23220749

RESUMO

Apomorphine is a non-narcotic derivative of morphine, which acts as a dopamine agonist and is clinically used to treat "off-states" in patients suffering from Parkinson's disease. Adverse effects of apomorphine treatment include severe emesis and nausea, and ulceration and pain at the injection site. We wanted to test whether sensory transient receptor potential (TRP) channels are a molecular target for apomorphine. Here, we show that rTRPV1, rTRPV2, rTRPV3, and mTRPV4, as well as hTRPM8, and rTRPM3, which are expressed in dorsal root ganglion neurons, are insensitive toward apomorphine treatment. This also applied to the cellular redox sensor hTRPM2. On the contrary, human TRPA1 could be concentration-dependently modulated by apomorphine. Whereas the addition of apomorphine in the low micromolar range produced an irreversible activation of the channel, application of higher concentrations caused a reversible voltage-dependent inhibition of heterologously expressed TRPA1 channels, resulting from a reduction of single-channel open times. In addition, we provide evidence that apomorphine also acts on endogenous TRPA1 in cultured dorsal root ganglion neurons from rats and in the enterochromaffin model cell line QGP-1, from which serotonin is released upon activation of TRPA1. Our study shows that human TRPA1 is a target for apomorphine, suggesting that an activation of TRPA1 might contribute to adverse side effects such as nausea and painful injections, which can occur during treatment with apomorphine.


Assuntos
Apomorfina/farmacologia , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Cálcio/metabolismo , Linhagem Celular , Feminino , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Células HEK293 , Humanos , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Wistar , Serotonina/metabolismo
15.
Naunyn Schmiedebergs Arch Pharmacol ; 385(6): 555-63, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22426997

RESUMO

Melanoma is the most dangerous form of skin cancer occurring in Caucasians with rising incidence. They are remarkably resistant to conventional anti-tumour therapies like chemotherapy and radiotherapy. Therefore, new treatment strategies are urgently needed. Anti-tumour effects of phytochemicals such as allyl isothiocyanate or cinnamaldehyde have been demonstrated in various melanoma models in vitro and in vivo. Considering their high potency as transient receptor potential A1 (TRPA1)-activating compounds, we examined the functional expression of TRPA1 channels in different melanoma cell lines as well as in non-malignantly transformed primary melanocytes. The presence of TRPA1 transcripts could be detected in most of the melanoma cell lines. Furthermore, single-cell calcium imaging and patch clamp electrophysiology confirmed the presence of functional TRPA1 channels in those cell lines. Proliferation assays revealed that allyl isothiocyanate and cinnamaldehyde clearly reduce the proliferation of melanoma cells, but this effect is independent of an activation of TRPA1 channels, making it unlikely that ionic currents through TRPA1 are responsible for the anti-tumour effects of mustard oil and cinnamaldehyde.


Assuntos
Canais de Cálcio/fisiologia , Melanoma/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Canais de Potencial de Receptor Transitório/fisiologia , Acetanilidas/farmacologia , Acroleína/análogos & derivados , Acroleína/farmacologia , Antineoplásicos/farmacologia , Cálcio/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Células HEK293 , Humanos , Isotiocianatos/farmacologia , Melanócitos/fisiologia , Melanoma/fisiopatologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Técnicas de Patch-Clamp , Purinas/farmacologia , RNA Mensageiro/metabolismo , Rutênio Vermelho/farmacologia , Canal de Cátion TRPA1 , Canais de Potencial de Receptor Transitório/antagonistas & inibidores
16.
Cell Calcium ; 51(2): 194-206, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22280812

RESUMO

The Ca(2+)-permeable, nonselective cation channel TRPC6 is gated via phospholipase C-activating receptors and has recently been implicated in hypoxia-induced pulmonary vasoconstriction (HPV), idiopathic pulmonary hypertension and focal segmental glomerulosclerosis (FSGS). Therefore, TRPC6 is a promising target for pharmacological interference. To identify and develop TRPC6-blocking compounds, we screened the Chembionet library, a collection of 16,671 chemically diverse drug-like compounds, for biological activity to prevent the 1-oleoyl-2-acetyl-sn-glycerol-triggered Ca(2+) influx in a stably transfected HEK(TRPC6-YFP) cell line. Hits were validated and characterised by fluorometric and electrophysiological methods. Six compounds displayed inhibitory potency at low micromolar concentrations, lack of cytotoxicity and blocked the receptor-dependent mode of TRPC6 activation. The specificity was tested towards closely (TRPC3 and TRPC7) and more distantly related TRP channels. One of the compounds, 8009-5364, displayed a 2.5-fold TRPC6-selectivity compared to TRPC3, and almost no inhibition of TRPC7 or the other TRP channels tested. Block of native TRPC3/6-like responses was confirmed in dissociated pulmonary artery smooth muscle cells. Two non-polar blockers effectively suppressed the HPV responses in the perfused mouse lung model. We conclude that pharmacological targeting of TRPC6 is feasible and provide a promising concept to treat pulmonary diseases that are characterised by excessive hypoxic vasoconstriction.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Hipertensão Pulmonar/tratamento farmacológico , Hipóxia/fisiopatologia , Canais de Cátion TRPC/antagonistas & inibidores , Vasoconstrição/efeitos dos fármacos , Animais , Bloqueadores dos Canais de Cálcio/química , Células HEK293 , Humanos , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/metabolismo , Hipóxia/complicações , Hipóxia/genética , Masculino , Camundongos , Camundongos Knockout , Ratos , Bibliotecas de Moléculas Pequenas , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/metabolismo , Canal de Cátion TRPC6
17.
Neuropharmacology ; 62(4): 1797-807, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22186081

RESUMO

Endocannabinoids exert numerous effects in the CNS under physiological and pathological conditions. The aim of the present study was to examine whether the endocannabinoid N-arachidonoyldopamine (NADA) may protect neurons in excitotoxically lesioned organotypic hippocampal slice cultures (OHSC). OHSC were excitotoxically lesioned by application of N-methyl-d-aspartate (NMDA, 50 µM) for 4 h and subsequently treated with different NADA concentrations (0.1 pM-50 µM) alone or in combination with cannabinoid receptor antagonists. NADA protected dentate gyrus granule cells and caused a slight reduction in the number of microglial cells. The number of degenerated neurons significantly decreased between 100 pM and 10 µM NADA (p < 0.05). To identify the responsive receptor type of NADA mediated neuroprotection, we applied the cannabinoid (CB) receptor 1 (CB(1)) inverse agonist/antagonist AM251, CB(2) inverse agonist/antagonist AM630, abnormal-cannabidiol (abn-CBD)-sensitive receptor antagonist O-1918, transient receptor potential channel V1 (TRPV1) antagonist 6-iodonordihydrocapsaicin and A1 (TRPA1) antagonist HC-030031. Neuroprotective properties of low (1 nM) but not high (10 µM) NADA concentrations were solely blocked by AM251 and were absent in CB(1)(-/-) mice. AM630, O-1918, 6-iodonordihydrocapsaicin and HC-030031 showed no effects at all NADA concentrations applied. Our findings demonstrate that NADA protects dentate gyrus granule cells by acting via CB(1). NADA reduced the number of microglial cells at distinct concentrations. TRPV1 and TRPA1 were not involved in NADA mediated neuroprotection. Thus, our data implicate that NADA mediated activation of neuronal CB(1) may serve as a novel pharmacological target to mitigate symptoms of neuronal damage.


Assuntos
Ácidos Araquidônicos/farmacologia , Dopamina/análogos & derivados , Hipocampo/efeitos dos fármacos , Degeneração Neural/metabolismo , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Receptor CB1 de Canabinoide/metabolismo , Animais , Células Cultivadas , Dopamina/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Hipocampo/citologia , Hipocampo/metabolismo , Camundongos , Neurônios/citologia , Neurônios/metabolismo , Piperidinas/farmacologia , Pirazóis/farmacologia , Ratos , Ratos Wistar
18.
Cell Calcium ; 45(2): 155-64, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18814910

RESUMO

TRPA1, a Ca(2+)-permeable cation channel that is expressed in sensory neurones, is involved in the perception of chemical irritants and mechanical hyperalgesia. TRPA1 is activated by either covalent or reversible binding of various chemical compounds, including allylisothiocyanate or acrolein, and is further sensitised by increases in the intracellular Ca(2+) concentration. We here demonstrate that TRPA1 confers a sensitivity towards near ultraviolet (UVA) light, which rapidly causes Ca(2+) entry. In electrophysiological recordings in whole cell and inside out modes, exposure to UVA light activated typical TRPA1 currents in a wavelength-dependent and membrane-delimited manner. In the presence of the photosensitising agents acridine orange (100 nM) or hypericin (10 nM), the sensitivity of light-induced TRPA1 activation was increased and extended towards the visible spectrum. Since extracellular application of hydrogen peroxide mimicked the effect of UVA irradiation and since dithiothreitol partly reversed the activation by UVA exposure, we conclude that reactive oxygen species may mediate the light-induced activation of TRPA1. Accordingly, hydrogen peroxide induced a TRPA1 activation with a membrane-delimited mode of action that was attenuated by dithiothreitol. Intracellular but not extracellular application of FeSO(4), which catalyses the formation of highly reactive hydroxyl radicals potentiated the hydrogen peroxide-stimulated TRPA1 activation. We conclude that, via generation of reactive oxygen species, light-induced TRPA1 activation provides an additional mode of activation, which renders TRPA1 a likely molecular candidate in processes leading to painful or burning sensations during photodynamic therapy or upon local application of hydrogen peroxide.


Assuntos
Canais de Cálcio/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/efeitos da radiação , Proteínas do Tecido Nervoso/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/efeitos da radiação , Fármacos Fotossensibilizantes/farmacologia , Canais de Potencial de Receptor Transitório/metabolismo , Raios Ultravioleta , Laranja de Acridina/farmacologia , Animais , Antracenos , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/efeitos da radiação , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Ditiotreitol/farmacologia , Compostos Ferrosos/farmacologia , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/efeitos da radiação , Humanos , Peróxido de Hidrogênio/farmacologia , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Espaço Intracelular/efeitos da radiação , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/efeitos da radiação , Perileno/análogos & derivados , Perileno/farmacologia , Ratos , Ratos Wistar , Canal de Cátion TRPA1
19.
Proc Natl Acad Sci U S A ; 105(49): 19538-43, 2008 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-19033464

RESUMO

SNARE-mediated exocytosis is a multistage process central to synaptic transmission and hormone release. Complexins (CPXs) are small proteins that bind very rapidly and with a high affinity to the SNARE core complex, where they have been proposed recently to inhibit exocytosis by clamping the complex and inhibiting membrane fusion. However, several other studies also suggest that CPXs are positive regulators of neurotransmitter release. Thus, whether CPXs are positive or negative regulators of exocytosis is not known, much less the stage in the vesicle life cycle at which they function. Here, we systematically dissect the vesicle stages leading up to exocytosis using a knockout-rescue strategy in a mammalian model system. We show that adrenal chromaffin cells from CPX II knockout mice exhibit markedly diminished releasable vesicle pools (comprising the readily and slowly releasable pools), while showing no change in the kinetics of fusion pore dilation or morphological vesicle docking. Overexpression of WT CPX II-but not of SNARE-binding-deficient mutants-restores the size of the the releasable pools in knockout cells, and in WT cells it markedly enlarges them. Our results show that CPXs regulate the size of the primed vesicle pools and have a positive role in Ca(2+)-triggered exocytosis.


Assuntos
Cálcio/metabolismo , Células Cromafins/fisiologia , Exocitose/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Vesículas Secretórias/metabolismo , Proteínas Adaptadoras de Transporte Vesicular , Animais , Catecolaminas/metabolismo , Células Cromafins/metabolismo , Células Cromafins/ultraestrutura , Feminino , Masculino , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Mutantes , Microscopia Eletrônica , Proteínas do Tecido Nervoso/genética , Técnicas de Patch-Clamp , Proteínas SNARE/metabolismo , Vesículas Secretórias/ultraestrutura
20.
J Biol Chem ; 283(49): 33942-54, 2008 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-18818211

RESUMO

The protective epithelial barrier in our skin undergoes constant regulation, whereby the balance between differentiation and proliferation of keratinocytes plays a major role. Impaired keratinocyte differentiation and proliferation are key elements in the pathophysiology of several important dermatological diseases, including atopic dermatitis and psoriasis. Ca(2+) influx plays an essential role in this process presumably mediated by different transient receptor potential (TRP) channels. However, investigating their individual role was hampered by the lack of specific stimulators or inhibitors. Because we have recently identified hyperforin as a specific TRPC6 activator, we investigated the contribution of TRPC6 to keratinocyte differentiation and proliferation. Like the endogenous differentiation stimulus high extracellular Ca(2+) concentration ([Ca(2+)](o)), hyperforin triggers differentiation in HaCaT cells and in primary cultures of human keratinocytes by inducing Ca(2+) influx via TRPC6 channels and additional inhibition of proliferation. Knocking down TRPC6 channels prevents the induction of Ca(2+)- and hyperforin-induced differentiation. Importantly, TRPC6 activation is sufficient to induce keratinocyte differentiation similar to the physiological stimulus [Ca(2+)](o). Therefore, TRPC6 activation by hyperforin may represent a new innovative therapeutic strategy in skin disorders characterized by altered keratinocyte differentiation.


Assuntos
Queratinócitos/citologia , Canais de Cátion TRPC/fisiologia , Compostos Bicíclicos com Pontes/farmacologia , Cálcio/química , Cátions , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Queratinócitos/metabolismo , Modelos Biológicos , Técnicas de Cultura de Órgãos/métodos , Floroglucinol/análogos & derivados , Floroglucinol/farmacologia , Pele/metabolismo , Dermatopatias/metabolismo , Canais de Cátion TRPC/química , Canal de Cátion TRPC6 , Terpenos/farmacologia , Fatores de Tempo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA