Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 52(9): 4784-4798, 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38621757

RESUMO

Antisense oligonucleotide (ASO) therapy is a novel therapeutic approach in which ASO specifically binds target mRNA, resulting in mRNA degradation; however, cellular uptake of ASOs remains critically low, warranting improvement. Transient receptor potential canonical (TRPC) channels regulate Ca2+ influx and are activated upon stimulation by phospholipase C-generated diacylglycerol. Herein, we report that a novel TRPC3/C6/C7 activator, L687, can induce cellular ASO uptake. L687-induced ASO uptake was enhanced in a dose- and incubation-time-dependent manner. L687 enhanced the knockdown activity of various ASOs both in vitro and in vivo. Notably, suppression of TRPC3/C6 by specific siRNAs reduced ASO uptake in A549 cells. Application of BAPTA-AM, a Ca2+ chelator, and SKF96365, a TRPC3/C6 inhibitor, suppressed Ca2+ influx via TRPC3/C6, resulting in reduced ASO uptake, thereby suggesting that Ca2+ influx via TRPC3/C6 is critical for L687-mediated increased ASO uptake. L687 also induced dextran uptake, indicating that L687 increased endocytosis. Adding ASO to L687 resulted in endosome accumulation; however, the endosomal membrane disruptor UNC7938 facilitated endosomal escape and enhanced knockdown activity. We discovered a new function for TRPC activators regarding ASO trafficking in target cells. Our findings provide an opportunity to formulate an innovative drug delivery system for the therapeutic development of ASO.


Assuntos
Cálcio , Oligonucleotídeos Antissenso , Canais de Cátion TRPC , Humanos , Oligonucleotídeos Antissenso/farmacologia , Oligonucleotídeos Antissenso/metabolismo , Canais de Cátion TRPC/metabolismo , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/antagonistas & inibidores , Cálcio/metabolismo , Células A549 , Animais , Camundongos , Imidazóis/farmacologia , Canal de Cátion TRPC6/metabolismo , Canal de Cátion TRPC6/genética , Canal de Cátion TRPC6/antagonistas & inibidores , Ácido Egtázico/farmacologia , Ácido Egtázico/análogos & derivados , Endossomos/metabolismo , Endossomos/efeitos dos fármacos , Linhagem Celular Tumoral
2.
Exp Cell Res ; 421(1): 113374, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36206825

RESUMO

Renal fibrosis is a global health concern with limited curative treatment. Canonical transient receptor potential channel 6 (TRPC6), a nonselective cation channel, has been shown to regulate the renal fibrosis in murine models. However, the molecular mechanism is unclear. Fibroblast-myofibroblast transdifferentiation is one of the critical steps in the progression of renal fibrosis. In the present study, we demonstrate that transforming growth factor (TGF)-ß1 exposure significantly increases the TRPC6 expression in renal interstitial fibroblast NRK-49F cells. Pharmacological inhibition of TRPC6 and knockdown of Trpc6 by siRNA alleviate TGF-ß1-increased expression levels of α-smooth muscle actin (α-SMA) and collagen I, two key markers of myofibroblasts. Although direct activation of TRPC6 by 1-oleoyl-2-acetyl-sn-glycerol (OAG) does not affect the expression of α-SMA and collagen I, OAG potentiates TGF-ß1-induced fibroblast-myofibroblast transdifferentiation. Further study demonstrates that TGF-ß1 exposure increases the phosphorylation level of p38 and Yes-associated protein (YAP) translocation into the nuclei. Inhibition of p38 and YAP decreases TGF-ß1-enhanced TRPC6 and α-SMA expression. In conclusion, we demonstrate that TRPC6 is a key regulator of TGF-ß1-induced fibroblast-myofibroblast transdifferentiation and provides the mechanism of how TGF-ß1 exposure regulates TRPC6 expression in NRK-49F fibroblasts.


Assuntos
Transdiferenciação Celular , Nefropatias , Canal de Cátion TRPC6 , Animais , Camundongos , Actinas/metabolismo , Transdiferenciação Celular/efeitos dos fármacos , Transdiferenciação Celular/fisiologia , Colágeno Tipo I/metabolismo , Fibroblastos/metabolismo , Fibrose , Nefropatias/metabolismo , Miofibroblastos/metabolismo , RNA Interferente Pequeno/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Fator de Crescimento Transformador beta1/metabolismo , Fatores de Crescimento Transformadores/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Canais de Potencial de Receptor Transitório/uso terapêutico , Canal de Cátion TRPC6/antagonistas & inibidores , Canal de Cátion TRPC6/genética , Proteínas de Sinalização YAP , Ratos , Modelos Animais de Doenças
3.
Eur J Pharmacol ; 913: 174616, 2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34780752

RESUMO

High fructose has been reported to drive glomerular podocyte oxidative stress and then induce podocyte foot process effacement in vivo, which could be partly regarded as podocyte hypermotility in vitro. Atractylodin possesses anti-oxidative effect. The aim of this study was to explore whether atractylodin prevented against fructose-induced podocyte hypermotility via anti-oxidative property. In fructose-exposed conditionally immortalized human podocytes, we found that atractylodin inhibited podocyte hypermotility, and up-regulated slit diaphragm proteins podocin and nephrin, and cytoskeleton protein CD2-associated protein (CD2AP), α-Actinin-4 and synaptopodin expression, which were consistent with its anti-oxidative activity evidenced by up-regulation of catalase (CAT) and superoxide dismutase (SOD) 1 expression, and reduction of reactive oxygen species (ROS) production. Atractylodin also significantly suppressed expression of transient receptor potential channels 6 (TRPC6) and phosphorylated Ca2+/calmodulin-dependent protein kinase IV (CaMK4) in cultured podocytes with fructose exposure. Additionally, in fructose-exposed podocytes, CaMK4 siRNA up-regulated synaptopodin and reduced podocyte hypermotility, whereas, silencing of TRPC6 by siRNA decreased p-CaMK4 expression, inhibited podocyte hypermotility, showing TRPC6/p-CaMK4 signaling activation in podocyte hypermotility under fructose condition. Just like atractylodin, antioxidant N-acetyl-L-cysteine (NAC) could inhibit TRPC6/p-CaMK4 signaling activation to reduce fructose-induced podocytes hypermotility. These results first demonstrated that the anti-oxidative property of atractylodin may contribute to the suppression of podocyte hypermotility via inhibiting TRPC6/p-CaMK4 signaling and restoring synaptopodin expression abnormality.


Assuntos
Antioxidantes/farmacologia , Frutose/efeitos adversos , Furanos/farmacologia , Podócitos/efeitos dos fármacos , Edulcorantes/efeitos adversos , Proteína Quinase Tipo 4 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Proteína Quinase Tipo 4 Dependente de Cálcio-Calmodulina/metabolismo , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Humanos , Proteínas dos Microfilamentos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Podócitos/fisiologia , Proteólise/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Canal de Cátion TRPC6/antagonistas & inibidores , Canal de Cátion TRPC6/metabolismo
4.
Am J Physiol Lung Cell Mol Physiol ; 321(6): L1161-L1182, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34704831

RESUMO

Idiopathic pulmonary arterial hypertension (PAH) is a fatal and progressive disease. Sustained vasoconstriction due to pulmonary arterial smooth muscle cell (PASMC) contraction and concentric arterial remodeling due partially to PASMC proliferation are the major causes for increased pulmonary vascular resistance and increased pulmonary arterial pressure in patients with precapillary pulmonary hypertension (PH) including PAH and PH due to respiratory diseases or hypoxemia. We and others observed upregulation of TRPC6 channels in PASMCs from patients with PAH. A rise in cytosolic Ca2+ concentration ([Ca2+]cyt) in PASMC triggers PASMC contraction and vasoconstriction, while Ca2+-dependent activation of PI3K/AKT/mTOR pathway is a pivotal signaling cascade for cell proliferation and gene expression. Despite evidence supporting a pathological role of TRPC6, no selective and orally bioavailable TRPC6 antagonist has yet been developed and tested for treatment of PAH or PH. In this study, we sought to investigate whether block of receptor-operated Ca2+ channels using a nonselective blocker of cation channels, 2-aminoethyl diphenylborinate (2-APB, administered intraperitoneally) and a selective blocker of TRPC6, BI-749327 (administered orally) can reverse established PH in mice. The results from the study show that intrapulmonary application of 2-APB (40 µM) or BI-749327 (3-10 µM) significantly and reversibly inhibited acute alveolar hypoxia-induced pulmonary vasoconstriction. Intraperitoneal injection of 2-APB (1 mg/kg per day) significantly attenuated the development of PH and partially reversed established PH in mice. Oral gavage of BI-749327 (30 mg/kg, every day, for 2 wk) reversed established PH by ∼50% via regression of pulmonary vascular remodeling. Furthermore, 2-APB and BI-749327 both significantly inhibited PDGF- and serum-mediated phosphorylation of AKT and mTOR in PASMC. In summary, the receptor-operated and mechanosensitive TRPC6 channel is a good target for developing novel treatment for PAH/PH. BI-749327, a selective TRPC6 blocker, is potentially a novel and effective drug for treating PAH and PH due to respiratory diseases or hypoxemia.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Hipertensão Pulmonar/patologia , Músculo Liso Vascular/patologia , Artéria Pulmonar/patologia , Canal de Cátion TRPC6/metabolismo , Vasoconstrição , Animais , Compostos de Boro/farmacologia , Sinalização do Cálcio , Células Cultivadas , Humanos , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/metabolismo , Camundongos , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/metabolismo , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Canal de Cátion TRPC6/antagonistas & inibidores , Canal de Cátion TRPC6/genética
5.
Mol Biol Rep ; 48(2): 1453-1464, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33606151

RESUMO

Loss of podocytes is a hallmark of diabetic nephropathy, and a growing body of evidence indicates that podocytes are susceptible to palmitic acid (PA). We have previously shown that AS-IV inhibited PA-induced podocyte apoptosis by activating sarcoendoplasmic reticulum Ca2+ ATPase (SERCA), which indicate calcium regulation may involve in the process. Immunofluorescence staining, Western blot and flow cytometry were used to measure the protective efficacy of AS-IV to ameliorate PA-induced ER stress and podocyte apoptosis. Meanwhile, AS-IV inhibited cytochrome c release, decreased mitochondrial membrane potential, accompany with the depletion of endoplasmic reticulum Ca2+ and elevation of cytosolic and mitochondrial Ca2+. Sequestration of cytosolic calcium with BAPTA-AM limited the response of podocyte apoptosis, while during the process the effect of AS-IV was also restrained. In contrast, elevation of cytosolic calcium with calcium ionophore ionomycin was depressed by AS-IV addition. Furthermore, inhibiting TRPC6 expression with SKF96365 or TRPC6 siRNA counteracted the beneficial effect of AS-IV. Our study provides further evidence to conclude the inhibitory effect of AS-IV to podocyte apoptosis is Ca2+-dependent. And the efficacy correlates with inhibiting TRPC6-mediated Ca2+ influx, and then cellular Ca2+ disturbance was coordinated.


Assuntos
Cálcio/metabolismo , Mitocôndrias/efeitos dos fármacos , Saponinas/farmacologia , Canal de Cátion TRPC6/genética , Triterpenos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Imidazóis/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Mitocôndrias/genética , Ácido Palmítico/farmacologia , Podócitos/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Canal de Cátion TRPC6/antagonistas & inibidores
6.
J Biol Chem ; 296: 100254, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33380424

RESUMO

Melatonin has been reported to induce effective reduction in growth and development in a variety of tumors, including breast cancer. In triple-negative breast cancer (TNBC) cells, melatonin attenuates a variety of cancer features, such as tumor growth and apoptosis resistance, through a number of still poorly characterized mechanisms. One biological process that is important for TNBC cells is store-operated Ca2+ entry (SOCE), which is modulated by TRPC6 expression and function. We wondered whether melatonin might intersect with this pathway as part of its anticancer activity. We show that melatonin, in the nanomolar range, significantly attenuates TNBC MDA-MB-231 cell viability, proliferation, and migration in a time- and concentration-dependent manner, without having any effect on nontumoral breast epithelial MCF10A cells. Pretreatment with different concentrations of melatonin significantly reduced SOCE in MDA-MB-231 cells without altering Ca2+ release from the intracellular stores. By contrast, SOCE in MCF10A cells was unaffected by melatonin. In the TNBC MDA-MB-468 cell line, melatonin not only attenuated viability, migration, and SOCE, but also reduced TRPC6 expression in a time- and concentration-dependent manner, without altering expression or function of the Ca2+ channel Orai1. The expression of exogenous TRPC6 overcame the effect of melatonin on SOCE and cell proliferation, and silencing or inhibition of TRPC6 impaired the inhibitory effect of melatonin on SOCE. These findings indicate that TRPC6 downregulation might be involved in melatonin's inhibitory effects on Ca2+ influx and the maintenance of cancer hallmarks and point toward a novel antitumoral mechanism of melatonin in TNBC cells.


Assuntos
Antioxidantes/farmacologia , Canais de Cálcio/metabolismo , Melatonina/farmacologia , Canal de Cátion TRPC6/antagonistas & inibidores , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Humanos , Canal de Cátion TRPC6/metabolismo , Neoplasias de Mama Triplo Negativas/patologia
7.
Cells ; 9(11)2020 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-33114455

RESUMO

Alzheimer's disease and cerebral ischemia are among the many causative neurodegenerative diseases that lead to disabilities in the middle-aged and elderly population. There are no effective disease-preventing therapies for these pathologies. Recent in vitro and in vivo studies have revealed the TRPC6 channel to be a promising molecular target for the development of neuroprotective agents. TRPC6 channel is a non-selective cation plasma membrane channel that is permeable to Ca2+. Its Ca2+-dependent pharmacological effect is associated with the stabilization and protection of excitatory synapses. Downregulation as well as upregulation of TRPC6 channel functions have been observed in Alzheimer's disease and brain ischemia models. Thus, in order to protect neurons from Alzheimer's disease and cerebral ischemia, proper TRPC6 channels modulators have to be used. TRPC6 channels modulators are an emerging research field. New chemical structures modulating the activity of TRPC6 channels are being currently discovered. The recent publication of the cryo-EM structure of TRPC6 channels should speed up the discovery process even more. This review summarizes the currently available information about potential drug candidates that may be used as basic structures to develop selective, highly potent TRPC6 channel modulators to treat neurodegenerative disorders, such as Alzheimer's disease and cerebral ischemia.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Isquemia Encefálica/etiologia , Isquemia Encefálica/metabolismo , Canal de Cátion TRPC6/deficiência , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Animais , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/patologia , Gerenciamento Clínico , Suscetibilidade a Doenças , Descoberta de Drogas , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Terapia de Alvo Molecular , Fatores de Risco , Transdução de Sinais/efeitos dos fármacos , Canal de Cátion TRPC6/antagonistas & inibidores , Canal de Cátion TRPC6/genética , Canal de Cátion TRPC6/metabolismo
8.
Eur J Pharmacol ; 886: 173357, 2020 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-32758574

RESUMO

Transient receptor potential canonical-6 (TRPC6) channels are non-selective cation channels that can be activated by hyperforin, a constituent of Hypericum perforatum. TRPC6 activation has been linked to a variety of biological functions and pathologies, including focal segmental glomerulosclerosis and the development of various tumor entities. Thus, TRPC6 is an interesting drug target, and a specific pharmacological inhibitor would be very valuable for both basic research and therapy of TRPC6-mediated human pathologies. Here, we assessed the biological activity of various TRP channel inhibitors on hyperforin-stimulated TRPC6 channel signaling. Hyperforin stimulates the activity of the transcription factor AP-1 via TRPC6. Expression experiments involving a TRPC6-specific small hairpin RNA confirmed that hyperforin-induced gene transcription requires TRPC6. Cellular AP-1 activity was measured to assess which compound interrupted the TRPC6-induced intracellular signaling cascade. The results show that the compounds 2-APB, clotrimazole, BCTC, TC-I 2014, SAR 7334, and larixyl acetate blocked TRPC6-mediated activation of AP-1. In contrast, the TRPM8-specific inhibitor RQ-00203078 did not inhibit TRPC6-mediated signaling. 2-APB, clotrimazole, BCTC, and TC-I 2014 are broad-spectrum Ca2+ channel inhibitors, while SAR 7334 and larixyl acetate have been proposed to function as rather TRPC6-specific inhibitors. In this study it is shown that both compounds, in addition to inhibiting TRPC6-induced signaling, completely abolished pregnenolone sulfate-mediated signaling via TRPM3 channels. Thus, SAR 7334 and larixyl acetate are not TRPC6-specific inhibitors.


Assuntos
Canal de Cátion TRPC6/antagonistas & inibidores , Canal de Cátion TRPC6/genética , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética , Bloqueadores dos Canais de Cálcio/farmacologia , Células HEK293 , Humanos , Floroglucinol/análogos & derivados , Floroglucinol/farmacologia , Pregnenolona/farmacologia , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Especificidade por Substrato , Canais de Cátion TRPM/antagonistas & inibidores , Terpenos/farmacologia , Fator de Transcrição AP-1/efeitos dos fármacos
9.
Anesthesiology ; 133(2): 364-376, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32665491

RESUMO

BACKGROUND: Until recently, the mechanism for the malignant hyperthermia crisis has been attributed solely to sustained massive Ca release from the sarcoplasmic reticulum on exposure to triggering agents. This study tested the hypothesis that transient receptor potential cation (TRPC) channels are important contributors to the Ca dyshomeostasis in a mouse model relevant to malignant hyperthermia. METHODS: This study examined the mechanisms responsible for Ca dyshomeostasis in RYR1-p.G2435R mouse muscles and muscle cells using calcium and sodium ion selective microelectrodes, manganese quench of Fura2 fluorescence, and Western blots. RESULTS: RYR1-p.G2435R mouse muscle cells have chronically elevated intracellular resting calcium and sodium and rate of manganese quench (homozygous greater than heterozygous) compared with wild-type muscles. After exposure to 1-oleoyl-2-acetyl-sn-glycerol, a TRPC3/6 activator, increases in intracellular resting calcium/sodium were significantly greater in RYR1-p.G2435R muscles (from 153 ± 11 nM/10 ± 0.5 mM to 304 ± 45 nM/14.2 ± 0.7 mM in heterozygotes P < 0.001] and from 251 ± 25 nM/13.9 ± 0.5 mM to 534 ± 64 nM/20.9 ± 1.5 mM in homozygotes [P < 0.001] compared with 123 ± 3 nM/8 ± 0.1 mM to 196 ± 27 nM/9.4 ± 0.7 mM in wild type). These increases were inhibited both by simply removing extracellular Ca and by exposure to either a nonspecific (gadolinium) or a newly available, more specific pharmacologic agent (SAR7334) to block TRPC6- and TRPC3-mediated cation influx into cells. Furthermore, local pretreatment with SAR7334 partially decreased the elevation of intracellular resting calcium that is seen in RYR1-p.G2435R muscles during exposure to halothane. Western blot analysis showed that expression of TRPC3 and TRPC6 were significantly increased in RYR1-p.G2435R muscles in a gene-dose-dependent manner, supporting their being a primary molecular basis for increased sarcolemmal cation influx. CONCLUSIONS: Muscle cells in knock-in mice expressing the RYR1-p.G2435R mutation are hypersensitive to TRPC3/6 activators. This hypersensitivity can be negated with pharmacologic agents that block TRPC3/6 activity. This reinforces the working hypothesis that transient receptor potential cation channels play a critical role in causing intracellular calcium and sodium overload in malignant hyperthermia-susceptible muscle, both at rest and during the malignant hyperthermia crisis.


Assuntos
Cálcio/metabolismo , Modelos Animais de Doenças , Hipertermia Maligna/metabolismo , Canais de Cátion TRPC/metabolismo , Canal de Cátion TRPC6/metabolismo , Animais , Feminino , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Indanos/farmacologia , Masculino , Hipertermia Maligna/genética , Hipertermia Maligna/patologia , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Canal de Liberação de Cálcio do Receptor de Rianodina/biossíntese , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canais de Cátion TRPC/antagonistas & inibidores , Canais de Cátion TRPC/genética , Canal de Cátion TRPC6/antagonistas & inibidores , Canal de Cátion TRPC6/genética
10.
Elife ; 92020 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-32149605

RESUMO

Transient receptor potential canonical (TRPC) proteins form nonselective cation channels that play physiological roles in a wide variety of cells. Despite growing evidence supporting the therapeutic potential of TRPC6 inhibition in treating pathological cardiac and renal conditions, mechanistic understanding of TRPC6 function and modulation remains obscure. Here we report cryo-EM structures of TRPC6 in both antagonist-bound and agonist-bound states. The structures reveal two novel recognition sites for the small-molecule modulators corroborated by mutagenesis data. The antagonist binds to a cytoplasm-facing pocket formed by S1-S4 and the TRP helix, whereas the agonist wedges at the subunit interface between S6 and the pore helix. Conformational changes upon ligand binding illuminate a mechanistic rationale for understanding TRPC6 modulation. Furthermore, structural and mutagenesis analyses suggest several disease-related mutations enhance channel activity by disrupting interfacial interactions. Our results provide principles of drug action that may facilitate future design of small molecules to ameliorate TRPC6-mediated diseases.


Assuntos
Bloqueadores dos Canais de Cálcio/metabolismo , Diglicerídeos/metabolismo , Canal de Cátion TRPC6/química , Sítios de Ligação , Bloqueadores dos Canais de Cálcio/química , Bloqueadores dos Canais de Cálcio/farmacologia , Microscopia Crioeletrônica , Diglicerídeos/química , Diglicerídeos/farmacologia , Glomerulosclerose Segmentar e Focal/genética , Humanos , Ligantes , Modelos Moleculares , Mutação , Conformação Proteica , Domínios Proteicos , Canal de Cátion TRPC6/agonistas , Canal de Cátion TRPC6/antagonistas & inibidores , Canal de Cátion TRPC6/metabolismo
11.
Drug Des Devel Ther ; 14: 361-370, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32095070

RESUMO

OBJECTIVE: Podocyte damage is common in many renal diseases characterized by proteinuria. Transient receptor potential cation channel protein 6 (TRPC6) plays an important role in renal function through its regulation of intracellular Ca2+ influx and RhoA/ROCK pathways. Chinese herb Stephania tetrandra, with the main active component being tetrandrine, has been used for the treatment of various kidney diseases for several years and has shown a positive effect. This study aimed at investigating the effect and mechanism of tetrandrine in podocyte damage induced by high expression of TRPC6. METHODS: Immortalized, differentiated murine podocytes, MPC5 were treated with valsartan (0-800 µM) and tetrandrine (0-40 µM) for 48 h. The maximum safe concentrations of valsartan and tetrandrine were selected using a cell viability assay. MPC5 podocytes stably expressing TRPC6 were constructed using a lentivirus packaging system, followed by treatment with valsartan, tetrandrine, and Y-27632 for 48 h and U73122 (10 µM) for 10 min. The RhoA/ROCK pathway and podocyte-specific proteins (nephrin and synaptopodin) levels were quantified. Podocyte apoptosis and intracellular Ca2+ concentration were measured. RESULTS: Maximum safe concentrations of 100 µM valsartan and 10 µM tetrandrine showed no observable toxicity in podocytes. MPC5 podocytes stably expressing TRPC6 had higher intracellular Ca2+ influx, apoptotic percentages, and expression of RhoA/ROCK proteins, but lower expression of nephrin and synaptopodin proteins. U73122 treatment for 10 min did not inhibit TRPC6, but suppressed RhoA/ROCK protein. Y-27632 decreased ROCK1 expression, but did not influence the expression of TRPC6 protein. Both 100 µM valsartan and 10 µM tetrandrine for 48 h significantly inhibited intracellular Ca2+ influx, apoptosis, and RhoA/ROCK pathway, and increased nephrin and synaptopodin proteins in podocytes stably expressing TRPC6. CONCLUSION: Elevated TRPC6 expression can lead to podocyte injury by inducing intracellular Ca2+ influx and apoptosis of podocytes, and this effect may be mediated by activation of the RhoA/ROCK1 pathway. Tetrandrine can alleviate podocyte injury induced by TRPC6 expression through inhibition of the RhoA/ROCK pathway, suggesting a protective role in podocyte damage.


Assuntos
Benzilisoquinolinas/farmacologia , Medicamentos de Ervas Chinesas/farmacologia , Podócitos/efeitos dos fármacos , Canal de Cátion TRPC6/antagonistas & inibidores , Quinases Associadas a rho/antagonistas & inibidores , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Medicina Tradicional Chinesa , Camundongos , Podócitos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Stephania tetrandra/química , Relação Estrutura-Atividade , Canal de Cátion TRPC6/genética , Canal de Cátion TRPC6/metabolismo , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
12.
Nephron ; 144(2): 84-95, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31865328

RESUMO

BACKGROUND: The protective effects of taurine supplementation on diabetic kidney disease (DKD) have been defined, but the mechanisms are not quite clear yet. TRPC6 has been shown to function in the homeostasis of podocytes, but whether TRPC6-modulated mitochondrial dysfunctions participating in taurine-induced renal protection during diabetes are unclear. METHODS: A DKD model was constructed using streptozocin (STZ), and an immortalized mouse podocytes cell line MPC-5 was used. Renal histology and western blot were used to analyze the expression levels of certain proteins. Cell proliferation assays, apoptosis assays, calcium influx, and mitochondrial functions were evaluated. RESULTS: In this study, taurine intervention improved STZ-induced DKD injuries, while it decreased both 24-h urinary protein and podocytes apoptosis. In detail, this study showed that taurine treatment decreased mitochondrial ROS productions by suppressing calcium overload and improving mitochondrial respiratory functions. Furthermore, the upregulation of TRPC6 is partially responsible for the calcium overload during high glucose treatment, whereas taurine treatment inhibited TRPC6 expression and partially attenuated high glucose-induced podocytes injuries. In addition, we demonstrated that taurine could upregulate CSE expression and inhibits TRPC6 expression via promoting the synthesis of H2S. CONCLUSION: Our study reveals that taurine intervention could partially attenuate the lesions of DKD by modulating the CSE/TRPC6 axis.


Assuntos
Cistationina gama-Liase/fisiologia , Diabetes Mellitus Experimental/complicações , Nefropatias Diabéticas/prevenção & controle , Mitocôndrias/efeitos dos fármacos , Podócitos/patologia , Canal de Cátion TRPC6/antagonistas & inibidores , Taurina/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Células Cultivadas , Suplementos Nutricionais , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Estreptozocina , Canal de Cátion TRPC6/genética
13.
Trends Pharmacol Sci ; 40(12): 911-918, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31704171

RESUMO

Ion channels are critical to kidney function, and their dysregulation leads to several distinct kidney diseases. Of the diversity of ion channels in kidney cells, the transient receptor potential (TRP) superfamily of proteins plays important and varied roles in both maintaining homeostasis as well as in causing disease. Recent work showed that TRPC5 blockers could successfully protect critical components of the kidney filter both in vitro and in vivo, thus revealing TRPC5 as a tractable therapeutic target for focal and segmental glomerulosclerosis (FSGS), a common cause of kidney failure. Human genetics point to three additional TRP channels as plausible therapeutic targets: TRPC6 in FSGS, PKD2 in polycystic kidney disease, and TRPM6 in familial hypomagnesemia with secondary hypocalcemia (HSH). We conclude that targeting TRP channels could pave the way for much needed therapies for kidney diseases.


Assuntos
Nefropatias/tratamento farmacológico , Canais de Potencial de Receptor Transitório/antagonistas & inibidores , Animais , Humanos , Nefropatias/genética , Nefropatias/metabolismo , Terapia de Alvo Molecular , Canais de Cátion TRPC/antagonistas & inibidores , Canais de Cátion TRPC/metabolismo , Canal de Cátion TRPC6/antagonistas & inibidores , Canal de Cátion TRPC6/genética , Canais de Potencial de Receptor Transitório/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
14.
FASEB J ; 33(8): 9182-9193, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31063704

RESUMO

Soluble klotho (sKlotho), the shed ectodomain of α-klotho, protects the heart by down-regulating transient receptor potential canonical isoform 6 (TRPC6)-mediated calcium signaling. Binding to α2-3-sialyllactose moiety of gangliosides in lipid rafts and inhibition of raft-dependent signaling underlies the mechanism. A recent 3-Å X-ray structure of sKlotho in complex with fibroblast growth factor receptor (FGFR) and fibroblast growth factor 23 (FGF23) indicates that its ß6α6 loop might block access to the proposed binding site for α2-3-sialyllactose. It was concluded that sKlotho only functions in complex with FGFR and FGF23 and that sKlotho's pleiotropic effects all depend on FGF23. Here, we report that sKlotho can inhibit TRPC6 channels expressed in cells lacking endogenous FGFRs. Structural modeling and molecular docking show that a repositioned ß6α6 loop allows sKlotho to bind α2-3-sialyllactose. Molecular dynamic simulations further show the α2-3-sialyllactose-bound sKlotho complex to be stable. Domains mimicking sKlotho's sialic acid-recognizing activity inhibit TRPC6. The results strongly support the hypothesis that sKlotho can exert effects independent of FGF23 and FGFR.-Wright, J. D., An, S.-W., Xie, J., Lim, C., Huang, C.-L. Soluble klotho regulates TRPC6 calcium signaling via lipid rafts, independent of the FGFR-FGF23 pathway.


Assuntos
Sinalização do Cálcio , Glucuronidase/metabolismo , Microdomínios da Membrana/metabolismo , Canal de Cátion TRPC6/metabolismo , Animais , Sítios de Ligação , Ligação Competitiva , Linhagem Celular , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/química , Fatores de Crescimento de Fibroblastos/metabolismo , Glucuronidase/química , Células HEK293 , Humanos , Proteínas Klotho , Lactose/análogos & derivados , Lactose/química , Lactose/metabolismo , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Domínios e Motivos de Interação entre Proteínas , Ratos , Receptores de Fatores de Crescimento de Fibroblastos/química , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Ácidos Siálicos/química , Ácidos Siálicos/metabolismo , Solubilidade , Canal de Cátion TRPC6/antagonistas & inibidores , Canal de Cátion TRPC6/química
15.
Biofactors ; 45(4): 607-615, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31120605

RESUMO

Enhancing soluble receptor for advanced glycation endproducts (sRAGE) is considered as a potent strategy for diabetes therapy. sRAGE secretion is regulated by calcium and transient receptor potential canonical (TRPC) channels. However, the role of TRPC channels in diabetes remains unknown. 18ß-Glycyrrhetinic acid (18ß-GA), produced from liquorice, has shown antidiabetic properties. This study was aimed to investigate the effect of 18ß-GA on sRAGE secretion via TRPC channels in high glucose (HG)-induced THP-1 cells. HG treatment enhanced TRPC3 and TRPC6 expression and consequently caused reactive oxygen species (ROS) accumulation mediated through p47 nicotinamide-adenine dinucleotide phosphate oxidase and inducible nitric oxide synthase (iNOS) associated with uncoupling protein 2 (UCP2) decline and lower sRAGE secretion. Interestingly, 18ß-GA showed the dramatic effects similar to Pyr3 or 2-aminoethyl diphenyl borinate inhibitors and effectively reversed HG-elicited mechanisms including that blocking TRPC3 and TRPC6 protein expressions, suppressing intracellular [Ca2+] concentration, decreasing expressions of ROS, p47s, and iNOS, but increasing UCP2 level and promoting sRAGE secretion. Therefore, 18ß-GA provides a potential implication to diabetes mellitus and its complications.


Assuntos
Glucose/antagonistas & inibidores , Ácido Glicirretínico/análogos & derivados , Glycyrrhiza/química , Hipoglicemiantes/farmacologia , Receptor para Produtos Finais de Glicação Avançada/genética , Canais de Cátion TRPC/genética , Canal de Cátion TRPC6/genética , Compostos de Boro/farmacologia , Cálcio/metabolismo , Regulação da Expressão Gênica , Glucose/toxicidade , Ácido Glicirretínico/isolamento & purificação , Ácido Glicirretínico/farmacologia , Humanos , Hipoglicemiantes/isolamento & purificação , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Extratos Vegetais/química , Pirazóis/farmacologia , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Receptor para Produtos Finais de Glicação Avançada/antagonistas & inibidores , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Transdução de Sinais , Células THP-1 , Canais de Cátion TRPC/antagonistas & inibidores , Canais de Cátion TRPC/metabolismo , Canal de Cátion TRPC6/antagonistas & inibidores , Canal de Cátion TRPC6/metabolismo , Proteína Desacopladora 2/genética , Proteína Desacopladora 2/metabolismo
16.
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
17.
Mol Med Rep ; 19(3): 2421-2430, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30664212

RESUMO

The canonical transient receptor potential channel 6 ion channel is expressed in podocytes and is an important component of the glomerular slit diaphragm. Focal segmental glomerulosclerosis is closely associated with TRPC6 gene mutations, and TRPC6 mediates podocyte injury induced by high glucose. Angiotensin II (AngII) has been revealed to enhance TRPC6 currents in certain types of cells, including podocytes and ventricular myocytes. It has been reported that glucose regulated TRPC6 expression in an AngII­dependent manner in podocytes and that this pathway is critical in diabetic nephropathy. In the present study, the role of TRPC6 detected by western blotting and reverse transcription­quantitative polymerase chain reaction in AngII­mediated podocyte injury was evaluated in rats with type 2 diabetes induced by high­calorie diets and streptozotocin. The results demonstrated that urinary albumin excretion was elevated, and morphological changes, including glomerular basement membrane thickening and podocyte process effacement, were observed. There was increased expression of AngII and TRPC6 in diabetic rats. The angiotensin receptor blocker valsartan significantly reduced TRPC6 and nuclear factor of activated T­cells (NFAT) overexpression in diabetic rats. These results in vivo were confirmed by studies in vitro, which demonstrated that inhibition of TRPC6 ameliorated high glucose­induced podocyte injury by decreasing NFAT mRNA levels. Taken together, the present results suggested that the AngII/TRPC6/NFAT axis may be a crucial signaling pathway in podocytes that is necessary for maintaining the integrity of the glomerular filtration barrier. In addition, TRPC6 may represent a potential therapeutic target for diabetic nephropathy.


Assuntos
Angiotensina II/genética , Diabetes Mellitus Tipo 2/genética , Glomerulosclerose Segmentar e Focal/genética , Fatores de Transcrição NFATC/genética , Canal de Cátion TRPC6/genética , Albuminúria/genética , Animais , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Dieta Hiperlipídica/efeitos adversos , Regulação da Expressão Gênica , Glomerulosclerose Segmentar e Focal/complicações , Glomerulosclerose Segmentar e Focal/metabolismo , Glomerulosclerose Segmentar e Focal/patologia , Glucose/metabolismo , Humanos , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Podócitos/metabolismo , Podócitos/patologia , Ratos , Canal de Cátion TRPC6/antagonistas & inibidores
18.
Cell Physiol Biochem ; 51(1): 393-410, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30453301

RESUMO

BACKGROUND/AIMS: Podocytes are dynamic polarized cells on the surface of glomerular capillaries that are an essential part of the glomerular filtration barrier. AMP-activated protein kinase (AMPK), a key regulator of glucose and fatty acid metabolism, plays a major role in obesity and type 2 diabetes. Accumulating evidence suggests that TRPC6 channels are crucial mediators of calcium transport in podocytes and are involved in regulating glomerular filtration. Here we investigated whether the AMPK-TRPC6 pathway is involved in insulin-dependent cytoskeleton reorganization and glucose uptake in cultured rat podocytes. METHODS: Western blot and immunofluorescence analysis confirmed AMPKα and TRPC6 expression, the phosphorylation of proteins associated with actin cytoskeleton reorganization (PAK, rac1, and cofilin), and the expression of insulin signaling proteins (Akt, Insulin receptor). Coimmunoprecipitation and immunofluorescence results demonstrated AMPKα/TRPC6 interaction. To ask whether TRPC6 is involved in the insulin regulation of glucose transport, we measured insulin-dependent (1, 2-3H)-deoxy-D-glucose uptake into podocytes after reducing TRPC6 activity pharmacologically and biochemically (TRPC6 siRNA). RESULTS: The results suggested a key role for the TRPC6 channel in the mediation of insulin-dependent activation of AMPKα2 and glucose uptake. Moreover, AMPK and TRPC6 activation were required to stimulate the Rac1 signaling pathway. CONCLUSION: These results suggest a potentially important new mechanism that regulates glucose transport in podocytes and that could be injurious during diabetes.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Citoesqueleto/metabolismo , Glucose/metabolismo , Insulina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Canal de Cátion TRPC6/metabolismo , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/química , Actinas/metabolismo , Animais , Cálcio/metabolismo , Citoesqueleto/química , Fosforilação/efeitos dos fármacos , Podócitos/citologia , Podócitos/efeitos dos fármacos , Podócitos/metabolismo , Subunidades Proteicas/antagonistas & inibidores , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Canal de Cátion TRPC6/antagonistas & inibidores , Canal de Cátion TRPC6/genética , Proteínas rac1 de Ligação ao GTP/metabolismo , Quinases Associadas a rho/metabolismo
19.
Eur Spine J ; 27(10): 2621-2630, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29968164

RESUMO

PURPOSE: Prolonged bed rest and microgravity in space cause intervertebral disc (IVD) degeneration. However, the underlying molecular mechanisms are not completely understood. Transient receptor potential canonical (TRPC) channels are implicated in mechanosensing of several tissues, but are poorly explored in IVDs. METHODS: Primary human IVD cells from surgical biopsies composed of both annulus fibrosus and nucleus pulposus (passage 1-2) were exposed to simulated microgravity and to the TRPC channel inhibitor SKF-96365 (SKF) for up to 5 days. Proliferative capacity, cell cycle distribution, senescence and TRPC channel expression were analyzed. RESULTS: Both simulated microgravity and TRPC channel antagonism reduced the proliferative capacity of IVD cells and induced senescence. While significant changes in cell cycle distributions (reduction in G1 and accumulation in G2/M) were observed upon SKF treatment, the effect was small upon 3 days of simulated microgravity. Finally, downregulation of TRPC6 was shown under simulated microgravity. CONCLUSIONS: Simulated microgravity and TRPC channel inhibition both led to reduced proliferation and increased senescence. Furthermore, simulated microgravity reduced TRPC6 expression. IVD cell senescence and mechanotransduction may hence potentially be regulated by TRPC6 expression. This study thus reveals promising targets for future studies. These slides can be retrieved under Electronic Supplementary Material.


Assuntos
Disco Intervertebral , Canal de Cátion TRPC6 , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Humanos , Imidazóis/farmacologia , Disco Intervertebral/citologia , Disco Intervertebral/metabolismo , Mecanotransdução Celular/efeitos dos fármacos , Canal de Cátion TRPC6/antagonistas & inibidores , Canal de Cátion TRPC6/metabolismo , Canal de Cátion TRPC6/fisiologia
20.
Cancer Lett ; 432: 47-55, 2018 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-29859875

RESUMO

Transient receptor potential canonical 6 (TRPC6) proteins form receptor-operated Ca2+-permeable channels, which have been thought to bring benefit to the treatment of diseases, including cancer. However, selective antagonists for TRPC channels are rare and none of them has been tested against gastric cancer. Compound 14a and analogs were synthesized by chemical elaboration of previously reported TRPC3/6/7 agonist 4o. 14a had very weak agonist activity at TRPC6 expressed in HEK293 cells but exhibited strong inhibition on both 4o-mediated and receptor-operated activation of TRPC6 with an IC50 of about 1 µM. When applied to the culture media, 14a suppressed proliferation of AGS and MKN45 cells with IC50 values of 17.1 ±â€¯0.3 and 18.5 ±â€¯1.0 µM, respectively, and inhibited tube formation and migration of cultured human endothelial cells. This anti-tumor effect on gastric cancer was further verified in xenograft models using nude mice. This study has found a new tool compound which shows excellent therapeutic potential against human gastric cancer most likely through targeting TRPC6 channels.


Assuntos
Antineoplásicos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Pirazóis/farmacologia , Pirimidinas/farmacologia , Neoplasias Gástricas/tratamento farmacológico , Canal de Cátion TRPC6/antagonistas & inibidores , Animais , Apoptose , Cálcio/metabolismo , Movimento Celular , Proliferação de Células , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Canal de Cátion TRPC6/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA