Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Drug Des Devel Ther ; 14: 361-370, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32095070

RESUMEN

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.


Asunto(s)
Bencilisoquinolinas/farmacología , Medicamentos Herbarios Chinos/farmacología , Podocitos/efectos de los fármacos , Canal Catiónico TRPC6/antagonistas & inhibidores , Quinasas Asociadas a rho/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Medicina Tradicional China , Ratones , Podocitos/metabolismo , Transducción de Señal/efectos de los fármacos , Stephania tetrandra/química , Relación Estructura-Actividad , Canal Catiónico TRPC6/genética , Canal Catiónico TRPC6/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
2.
Nephron ; 144(2): 84-95, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31865328

RESUMEN

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.


Asunto(s)
Cistationina gamma-Liasa/fisiología , Diabetes Mellitus Experimental/complicaciones , Nefropatías Diabéticas/prevención & control , Mitocondrias/efectos de los fármacos , Podocitos/patología , Canal Catiónico TRPC6/antagonistas & inhibidores , Taurina/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Células Cultivadas , Suplementos Dietéticos , Ratones , Ratones Endogámicos C57BL , Mitocondrias/fisiología , Especies Reactivas de Oxígeno/metabolismo , Estreptozocina , Canal Catiónico TRPC6/genética
3.
Biofactors ; 45(4): 607-615, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31120605

RESUMEN

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.


Asunto(s)
Glucosa/antagonistas & inhibidores , Ácido Glicirretínico/análogos & derivados , Glycyrrhiza/química , Hipoglucemiantes/farmacología , Receptor para Productos Finales de Glicación Avanzada/genética , Canales Catiónicos TRPC/genética , Canal Catiónico TRPC6/genética , Compuestos de Boro/farmacología , Calcio/metabolismo , Regulación de la Expresión Génica , Glucosa/toxicidad , Ácido Glicirretínico/aislamiento & purificación , Ácido Glicirretínico/farmacología , Humanos , Hipoglucemiantes/aislamiento & purificación , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Extractos Vegetales/química , Pirazoles/farmacología , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Transducción de Señal , Células THP-1 , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales Catiónicos TRPC/metabolismo , Canal Catiónico TRPC6/antagonistas & inhibidores , Canal Catiónico TRPC6/metabolismo , Proteína Desacopladora 2/genética , Proteína Desacopladora 2/metabolismo
4.
Proc Natl Acad Sci U S A ; 116(20): 10156-10161, 2019 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-31028142

RESUMEN

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.


Asunto(s)
Cardiomegalia/tratamiento farmacológico , Nefroesclerosis/tratamiento farmacológico , Canal Catiónico TRPC6/antagonistas & inhibidores , Animales , Evaluación Preclínica de Medicamentos , Fibrosis , Células HEK293 , Corazón/efectos de los fármacos , Humanos , Riñón/efectos de los fármacos , Ratones
5.
J Nat Prod ; 81(4): 913-917, 2018 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-29469570

RESUMEN

Two new acylated ß-hydroxynitrile glycosides, ribemansides A (1) and B (2), were isolated from the aerial parts of Ribes manshuricum. Their structures were elucidated by comprehensive spectroscopic analysis. Ribemansides A and B inhibited transforming growth factor ß1 (TGF-ß1)-induced expression of α-smooth muscle actin, fibronectin release, and changes in cell morphology in the human proximal tubular epithelial cell line (human kidney-2, HK-2). Further biological evaluation demonstrated that both 1 and 2 inhibit the activity of canonical transient receptor potential cation channel 6 (TRPC6), with IC50 values of 24.5 and 25.6 µM, respectively. The antifibrogenic effect of these compounds appears to be mediated through TRPC6 inhibition, since the TRPC6 inhibitor, SAR7334, also suppressed TGF-ß1-induced fibrogenesis in HK-2 cells.


Asunto(s)
Glicósidos/farmacología , Extractos Vegetales/farmacología , Ribes/química , Canal Catiónico TRPC6/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/metabolismo , Actinas/metabolismo , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Fibronectinas/metabolismo , Glicósidos/química , Humanos , Extractos Vegetales/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA