Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 211
Filtrar
1.
Cells ; 10(5)2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-34069822

RESUMEN

Sodium salicylate (SA), a cyclooxygenase inhibitor, has been shown to increase insulin sensitivity and to suppress inflammation in obese patients and animal models. Transient receptor potential vanilloid 1 (TRPV1) is a nonselective cation channel expressed in afferent nerve fibers. Cyclooxygenase-derived prostaglandins are involved in the activation and sensitization of TRPV1. This study tested whether the metabolic and renal effects of SA were mediated by the TRPV1 channel. Wild-type (WT) and TRPV1-/- mice were fed a Western diet (WD) for 4 months and received SA infusion (120mg/kg/day) or vehicle for the last 4 weeks of WD feeding. SA treatment significantly increased blood pressure in WD-fed TRPV1-/- mice (p < 0.05) but not in WD-fed WT mice. Similarly, SA impaired renal blood flow in TRPV1-/- mice (p < 0.05) but not in WT mice. SA improved insulin and glucose tolerance in both WT and TRPV1-/- mice on WD (all p < 0.05). In addition, SA reduced renal p65 and urinary prostaglandin E2, prostaglandin F1α, and interleukin-6 in both WT and TRPV1-/- mice (all p < 0.05). SA decreased urine noradrenaline levels, increased afferent renal nerve activity, and improved baroreflex sensitivity in WT mice (all p < 0.05) but not in TRPV1-/- mice. Importantly, SA increased serum creatinine and urine kidney injury molecule-1 levels and decreased the glomerular filtration rate in obese WT mice (all p < 0.05), and these detrimental effects were significantly exacerbated in obese TRPV1-/- mice (all p < 0.05). Lastly, SA treatment increased urine albumin levels in TRPV1-/- mice (p < 0.05) but not in WT mice. Taken together, SA-elicited metabolic benefits and anti-inflammatory effects are independent of TRPV1, while SA-induced sympathetic suppression is dependent on TRPV1 channels. SA-induced renal dysfunction is dependent on intact TRPV1 channels. These findings suggest that SA needs to be cautiously used in patients with obesity or diabetes, as SA-induced renal dysfunction may be exacerbated due to impaired TRPV1 in obese and diabetic patients.


Asunto(s)
Inhibidores de la Ciclooxigenasa/toxicidad , Dieta Alta en Grasa , Enfermedades Renales/inducido químicamente , Riñón/efectos de los fármacos , Obesidad/tratamiento farmacológico , Salicilato de Sodio/toxicidad , Sistema Nervioso Simpático/efectos de los fármacos , Canales Catiónicos TRPV/deficiencia , Animales , Barorreflejo/efectos de los fármacos , Modelos Animales de Enfermedad , Metabolismo Energético/efectos de los fármacos , Eliminación de Gen , Tasa de Filtración Glomerular/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Resistencia a la Insulina , Riñón/inervación , Riñón/metabolismo , Enfermedades Renales/metabolismo , Enfermedades Renales/fisiopatología , Enfermedades Renales/prevención & control , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/complicaciones , Obesidad/metabolismo , Obesidad/fisiopatología , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/fisiopatología , Canales Catiónicos TRPV/genética
2.
Am J Physiol Heart Circ Physiol ; 320(3): H1156-H1169, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33449852

RESUMEN

The TRPV4 channel is a calcium-permeable channel (PCa/PNa ∼ 10). Its expression has been reported in ventricular myocytes, where it is involved in several cardiac pathological mechanisms. In this study, we investigated the implication of TRPV4 in ventricular electrical activity. Left ventricular myocytes were isolated from trpv4+/+ and trpv4-/- mice. TRPV4 membrane expression and its colocalization with L-type calcium channels (Cav1.2) was confirmed using Western blot biotinylation, immunoprecipitation, and immunostaining experiments. Then, electrocardiograms (ECGs) and patch-clamp recordings showed shortened QTc and action potential (AP) duration in trpv4-/- compared with trpv4+/+ mice. Thus, TRPV4 activator GSK1016790A produced a transient and dose-dependent increase in AP duration at 90% of repolarization (APD90) in trpv4+/+ but not in trpv4-/- myocytes or when combined with TRPV4 inhibitor GSK2193874 (100 nM). Hence, GSK1016790A increased calcium transient (CaT) amplitude in trpv4+/+ but not in trpv4-/- myocytes, suggesting that TRPV4 carries an inward Ca2+ current in myocytes. Conversely, TRPV4 inhibitor GSK2193874 (100 nM) alone reduced APD90 in trpv4+/+ but not in trpv4-/- myocytes, suggesting that TRPV4 prolongs AP duration in basal condition. Finally, introducing TRPV4 parameters in a mathematical model predicted the development of an inward TRPV4 current during repolarization that increases AP duration and CaT amplitude, in accord with what was found experimentally. This study shows for the first time that TRPV4 modulates AP and QTc durations. It would be interesting to evaluate whether TRPV4 could be involved in long QT-mediated ventricular arrhythmias.NEW & NOTEWORTHY Transient receptor potential vanilloid 4 (TRPV4) is expressed at the membrane of mouse ventricular myocytes and colocalizes with non-T-tubular L-type calcium channels. Deletion of trpv4 gene in mice results in shortened QT interval on electrocardiogram and reduced action potential duration of ventricular myocytes. Pharmacological activation of TRPV4 channel leads to increased action potential duration and increased calcium transient amplitude in trpv4-/- but not in trpv4-/- ventricular myocytes. To the contrary, TRPV4 channel pharmacological inhibition reduces action potential duration in trpv4+/+ but not in trpv4-/- myocytes. Integration of TRPV4 channel in a computational model of mouse action potential shows that the channel carries an inward current contributing to slowing down action potential repolarization and to increase calcium transient amplitude, similarly to what is observed experimentally. This study highlights for the first time the involvement of TRPV4 channel in ventricular electrical activity.


Asunto(s)
Potenciales de Acción , Señalización del Calcio , Frecuencia Cardíaca , Miocitos Cardíacos/metabolismo , Canales Catiónicos TRPV/metabolismo , Función Ventricular Izquierda , Potenciales de Acción/efectos de los fármacos , Animales , Canales de Calcio Tipo L/metabolismo , Señalización del Calcio/efectos de los fármacos , Simulación por Computador , Células HEK293 , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Leucina/análogos & derivados , Leucina/farmacología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Cardiovasculares , Miocitos Cardíacos/efectos de los fármacos , Piperidinas/farmacología , Quinolinas/farmacología , Sulfonamidas/farmacología , Canales Catiónicos TRPV/deficiencia , Canales Catiónicos TRPV/genética , Factores de Tiempo , Función Ventricular Izquierda/efectos de los fármacos
3.
Cell Prolif ; 54(1): e12955, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33159483

RESUMEN

OBJECTIVES: Calcium ion signals are important for osteoclast differentiation. Transient receptor potential vanilloid 6 (TRPV6) is a regulator of bone homeostasis. However, it was unclear whether TRPV6 was involved in osteoclast formation. Therefore, the aim of this study was to evaluate the role of TPRV6 in bone metabolism and to clarify its regulatory role in osteoclasts at the cellular level. MATERIALS AND METHODS: Bone structure and histological changes in Trpv6 knockout mice were examined using micro-computed tomography and histological analyses. To investigate the effects of Trpv6 on osteoclast function, we silenced or overexpressed Trpv6 in osteoclasts via lentivirus transfection, respectively. Osteoclast differentiation and bone resorption viability were measured by tartrate-resistant acid phosphatase (TRAP) staining and pit formation assays. The expression of osteoclast marker genes, including cathepsin k, DC-STAMP, Atp6v0d2 and TRAP, was measured by qRT-PCR. Cell immunofluorescence and Western blotting were applied to explore the mechanisms by which the IGF-PI3K-AKT pathway was involved in the regulation of osteoclast formation and bone resorption by Trpv6. RESULTS: We found that knockout of Trpv6 induced osteoporosis and enhanced bone resorption in mice, but did not affect bone formation. Further studies showed that Trpv6, which was distributed on the cell membrane of osteoclasts, acted as a negative regulator for osteoclast differentiation and function. Mechanistically, Trpv6 suppressed osteoclastogenesis by decreasing the ratios of phosphoprotein/total protein in the IGF-PI3K-AKT signalling pathway. Blocking of the IGF-PI3K-AKT pathway significantly alleviated the inhibitory effect of Trpv6 on osteoclasts formation. CONCLUSIONS: Our study confirmed the important role of Trpv6 in bone metabolism and clarified its regulatory role in osteoclasts at the cellular level. Taken together, this study may inspire a new strategy for the treatment of osteoporosis.


Asunto(s)
Resorción Ósea/metabolismo , Canales de Calcio/metabolismo , Diferenciación Celular , Osteoclastos/metabolismo , Ligando RANK/metabolismo , Transducción de Señal , Canales Catiónicos TRPV/metabolismo , Animales , Canales de Calcio/deficiencia , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Somatomedinas/metabolismo , Canales Catiónicos TRPV/deficiencia
4.
Eur J Pharmacol ; 888: 173567, 2020 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-32946867

RESUMEN

This study focuses on exploring the role of sensory cation channel Transient Receptor Potential channel subfamily Vanilloid 1 (TRPV1) in gut health, specifically mucus production and microflora profile in gut. We employed resiniferatoxin (ultrapotent TRPV1 agonist) induced chemo-denervation model in rats and studied the effects of TRPV1 ablation on colonic mucus secretion patterns. Histological and transcriptional analysis showed substantial decrease in mucus production as well as in expression of genes involved in goblet cell differentiation, mucin production and glycosylation. 16S metagenome analysis revealed changes in abundance of various gut bacteria, including decrease in beneficial bacteria like Lactobacillus spp and Clostridia spp. Also, TRPV1 ablation significantly decreased the levels of short chain fatty acids, i.e. acetate and butyrate. The present study provides first evidence that systemic TRPV1 ablation leads to impairment in mucus production and causes dysbiosis in gut. Further, it suggests to address mucin production and gut microbiota related adverse effects during the development of TRPV1 antagonism/ablation-based therapeutic and preventive strategies.


Asunto(s)
Colon/metabolismo , Microbioma Gastrointestinal/fisiología , Mucinas/antagonistas & inhibidores , Mucinas/biosíntesis , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/deficiencia , Técnicas de Ablación/métodos , Animales , Disbiosis/genética , Disbiosis/metabolismo , Masculino , Ratas , Ratas Wistar , Canales Catiónicos TRPV/genética
5.
Chem Senses ; 45(7): 573-579, 2020 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-32572463

RESUMEN

Exposure of the oral cavity to acidic solutions evokes not only a sensation of sour, but also of sharp or tangy. Acidic substances potentially stimulate both taste buds and acid-sensitive mucosal free nerve endings. Mice lacking taste function (P2X2/P2X3 double-KO mice) refuse acidic solutions similar to wildtype (WT) mice and intraoral infusion of acidic solutions in these KO animals evokes substantial c-Fos activity within orosensory trigeminal nuclei as well as of the nucleus of the solitary tract (nTS) (Stratford, Thompson, et al. 2017). This residual acid-evoked, non-taste activity includes areas that receive inputs from trigeminal and glossopharyngeal peptidergic (CGRP-containing) nerve fibers that express TrpA1 and TrpV1 both of which are activated by low pH. We compared avoidance responses in WT and TrpA1/V1 double-KO (TRPA1/V1Dbl-/-) mice in brief-access behavioral assay (lickometer) to 1, 3, 10, and 30 mM citric acid, along with 100 µM SC45647 and H2O. Both WT and TRPA1/V1Dbl-/- show similar avoidance, including to higher concentrations of citric acid (10 and 30 mM; pH 2.62 and pH 2.36, respectively), indicating that neither TrpA1 nor TrpV1 is necessary for the acid-avoidance behavior in animals with an intact taste system. Similarly, induction of c-Fos in the nTS and dorsomedial spinal trigeminal nucleus was similar in the WT and TRPA1/V1Dbl-/- animals. Taken together these results suggest non-TrpV1 and non-TrpA1 receptors underlie the residual responses to acids in mice lacking taste function.


Asunto(s)
Reacción de Prevención/efectos de los fármacos , Ácido Cítrico/farmacología , Canal Catiónico TRPA1/genética , Canales Catiónicos TRPV/genética , Animales , Reacción de Prevención/fisiología , Ácido Cítrico/química , Femenino , Guanidinas/química , Guanidinas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-fos/metabolismo , Núcleo Solitario/metabolismo , Canal Catiónico TRPA1/deficiencia , Canales Catiónicos TRPV/deficiencia , Núcleos del Trigémino/metabolismo
6.
Ann Neurol ; 88(2): 297-308, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32418267

RESUMEN

OBJECTIVE: Myotonia is caused by involuntary firing of skeletal muscle action potentials and causes debilitating stiffness. Current treatments are insufficiently efficacious and associated with side effects. Myotonia can be triggered by voluntary movement (electrically induced myotonia) or percussion (mechanically induced myotonia). Whether distinct molecular mechanisms underlie these triggers is unknown. Our goal was to identify ion channels involved in mechanically induced myotonia and to evaluate block of the channels involved as a novel approach to therapy. METHODS: We developed a novel system to enable study of mechanically induced myotonia using both genetic and pharmacologic mouse models of myotonia congenita. We extended ex vivo studies of excitability to in vivo studies of muscle stiffness. RESULTS: As previous work suggests activation of transient receptor potential vanilloid 4 (TRPV4) channels by mechanical stimuli in muscle, we examined the role of this cation channel. Mechanically induced myotonia was markedly suppressed in TRPV4-null muscles and in muscles treated with TRPV4 small molecule antagonists. The suppression of mechanically induced myotonia occurred without altering intrinsic muscle excitability, such that myotonia triggered by firing of action potentials (electrically induced myotonia) was unaffected. When injected intraperitoneally, TRPV4 antagonists lessened the severity of myotonia in vivo by approximately 80%. INTERPRETATION: These data demonstrate that there are distinct molecular mechanisms triggering electrically induced and mechanically induced myotonia. Our data indicates that activation of TRPV4 during muscle contraction plays an important role in triggering myotonia in vivo. Elimination of mechanically induced myotonia by TRPV4 inhibition offers a new approach to treating myotonia. ANN NEUROL 2020;88:297-308.


Asunto(s)
Contracción Isométrica/fisiología , Morfolinas/farmacología , Miotonía Congénita/genética , Miotonía Congénita/metabolismo , Pirroles/farmacología , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/deficiencia , Animales , Antracenos/farmacología , Contracción Isométrica/efectos de los fármacos , Ratones , Ratones Noqueados , Morfolinas/uso terapéutico , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiología , Miotonía Congénita/prevención & control , Pirroles/uso terapéutico
7.
J Clin Invest ; 130(5): 2527-2541, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31999644

RESUMEN

Elevated pressure in the pancreatic gland is the central cause of pancreatitis following abdominal trauma, surgery, endoscopic retrograde cholangiopancreatography, and gallstones. In the pancreas, excessive intracellular calcium causes mitochondrial dysfunction, premature zymogen activation, and necrosis, ultimately leading to pancreatitis. Although stimulation of the mechanically activated, calcium-permeable ion channel Piezo1 in the pancreatic acinar cell is the initial step in pressure-induced pancreatitis, activation of Piezo1 produces only transient elevation in intracellular calcium that is insufficient to cause pancreatitis. Therefore, how pressure produces a prolonged calcium elevation necessary to induce pancreatitis is unknown. We demonstrate that Piezo1 activation in pancreatic acinar cells caused a prolonged elevation in intracellular calcium levels, mitochondrial depolarization, intracellular trypsin activation, and cell death. Notably, these effects were dependent on the degree and duration of force applied to the cell. Low or transient force was insufficient to activate these pathological changes, whereas higher and prolonged application of force triggered sustained elevation in intracellular calcium, leading to enzyme activation and cell death. All of these pathological events were rescued in acinar cells treated with a Piezo1 antagonist and in acinar cells from mice with genetic deletion of Piezo1. We discovered that Piezo1 stimulation triggered transient receptor potential vanilloid subfamily 4 (TRPV4) channel opening, which was responsible for the sustained elevation in intracellular calcium that caused intracellular organelle dysfunction. Moreover, TRPV4 gene-KO mice were protected from Piezo1 agonist- and pressure-induced pancreatitis. These studies unveil a calcium signaling pathway in which a Piezo1-induced TRPV4 channel opening causes pancreatitis.


Asunto(s)
Canales Iónicos/agonistas , Pancreatitis/etiología , Pancreatitis/fisiopatología , Canales Catiónicos TRPV/fisiología , Células Acinares/efectos de los fármacos , Células Acinares/patología , Células Acinares/fisiología , Animales , Calcio/metabolismo , Señalización del Calcio , Muerte Celular , Modelos Animales de Enfermedad , Femenino , Canales Iónicos/genética , Canales Iónicos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Páncreas/efectos de los fármacos , Páncreas/patología , Páncreas/fisiopatología , Pancreatitis/patología , Presión , Pirazinas/farmacología , Canales Catiónicos TRPV/deficiencia , Canales Catiónicos TRPV/genética , Tiadiazoles/farmacología
8.
Basic Res Cardiol ; 115(2): 14, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31925567

RESUMEN

Cardiac fibrosis caused by adverse cardiac remodeling following myocardial infarction can eventually lead to heart failure. Although the role of soluble factors such as TGF-ß is well studied in cardiac fibrosis following myocardial injury, the physiological role of mechanotransduction is not fully understood. Here, we investigated the molecular mechanism and functional role of TRPV4 mechanotransduction in cardiac fibrosis. TRPV4KO mice, 8 weeks following myocardial infarction (MI), exhibited preserved cardiac function compared to WT mice. Histological analysis demonstrated reduced cardiac fibrosis in TRPV4KO mice. We found that WT CF exhibited hypotonicity-induced calcium influx and extracellular matrix (ECM)-stiffness-dependent differentiation in response to TGF-ß1. In contrast, TRPV4KO CF did not display hypotonicity-induced calcium influx and failed to differentiate on high-stiffness ECM gels even in the presence of saturating amounts of TGF-ß1. Mechanistically, TRPV4 mediated cardiac fibrotic gene promoter activity and fibroblast differentiation through the activation of the Rho/Rho kinase pathway and the mechanosensitive transcription factor MRTF-A. Our findings suggest that genetic deletion of TRPV4 channels protects heart from adverse cardiac remodeling following MI by modulating Rho/MRTF-A pathway-mediated cardiac fibroblast differentiation and cardiac fibrosis.


Asunto(s)
Diferenciación Celular , Fibroblastos/metabolismo , Eliminación de Gen , Infarto del Miocardio/prevención & control , Miocardio/metabolismo , Canales Catiónicos TRPV/deficiencia , Remodelación Ventricular , Animales , Señalización del Calcio , Células Cultivadas , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Fibroblastos/patología , Fibrosis , Mecanotransducción Celular , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Canales Catiónicos TRPV/genética , Transactivadores/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/metabolismo
9.
Prostate ; 80(1): 28-37, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31573117

RESUMEN

BACKGROUND: Chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) is a disorder that is characterized by persistent pelvic pain in men of any age. Although several studies suggest that the transient receptor potential vanilloid 1 (TRPV1) channel is involved in various pathways of chronic pain, the TRPV1 channel has not been implicated in chronic pelvic pain associated with CP/CPPS. METHODS: Male C57BL/6J (B6) and TRPV1 knockout (TRPV1 KO) mice (5-7 weeks old) were used to study the development of pelvic allodynia in a murine model of CP/CPPS called experimental autoimmune prostatitis (EAP). The prostate lobes, dorsal root ganglia (DRG), and spinal cord were excised at day 20. The prostate lobes were assessed for inflammation, TRPV1 expression, and mast cell activity. DRG and spinal cord, between the L6-S4 regions, were analyzed to determine the levels of phosphorylated ERK1/2 (p-ERK 1/2). To examine the therapeutic potential of TRPV1, B6 mice with EAP received intraurethral infusion of a TRPV1 antagonist at day 20 (repeated every 2 days) and pelvic pain was evaluated at days 20, 25, 30, and 35. RESULTS: Our data showed that B6 mice with EAP developed pelvic tactile allodynia at days 7, 14, and 20. In contrast, TRPV1 KO mice with EAP do not develop pelvic tactile allodynia at any time point. Although we observed no change in the levels of TRPV1 protein expression in the prostate from B6 mice with EAP, there was evidence of significant inflammation and elevated mast cell activation. Interestingly, the prostate from TRPV1 KO mice with EAP showed a lack of mast cell activation despite evidence of prostate inflammation. Next, we observed a significant increase of p-ERK1/2 in the DRG and spinal cord from B6 mice with EAP; however, p-ERK1/2 expression was unaltered in TRPV1 KO mice with EAP. Finally, we confirmed that intraurethral administration of a TRPV1 antagonist peptide reduced pelvic tactile allodynia in B6 mice with EAP after day 20. CONCLUSIONS: We demonstrated that in a murine model of CP/CPPS, the TRPV1 channel is key to persistent pelvic tactile allodynia and blocking TRPV1 in the prostate may be a promising strategy to quell chronic pelvic pain.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Prostatitis/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Arginina/análogos & derivados , Arginina/farmacología , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ganglios Espinales/metabolismo , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/inmunología , Hiperalgesia/metabolismo , Hiperalgesia/patología , Masculino , Mastocitos/inmunología , Mastocitos/metabolismo , Mastocitos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oligopéptidos/farmacología , Dolor Pélvico/tratamiento farmacológico , Dolor Pélvico/inmunología , Dolor Pélvico/metabolismo , Dolor Pélvico/patología , Fosforilación , Prostatitis/tratamiento farmacológico , Prostatitis/inmunología , Prostatitis/patología , Médula Espinal/metabolismo , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/deficiencia
10.
Neuromolecular Med ; 22(1): 68-72, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31468327

RESUMEN

Mutations in TRPV4 are linked to a group of clinically distinct, but also overlapping axonal neuropathies, including Charcot-Marie-Tooth disease type 2C (CMT2C), scapuloperoneal spinal muscular atrophy, and congenital distal spinal muscular atrophy. The incidence of TRPV4-linked cases ranges from 0 to 7% in overall axonal neuropathy cohorts from European countries and Australia. However, the data from other areas remain largely unknown. In this study, we screened for TRPV4 mutations in a well-characterized USA cohort of 62 unrelated CMT2 patients without mutations in MFN2, GARS, NEFL, and GDAP1. All 15 coding exons of TRPV4 were analyzed by Sanger-sequencing. Clinical features of TRPV4-linked patients were compared with those lacking TRPV4 mutations. We identified two TRPV4 mutations in two patients. A TRPV4-R316C was identified in a patient with family history, while a TRPV4-R269C in an apparently sporadic case. Marked clinical variations were observed in the patients with TRPV4 mutations. Our data suggest that TRPV4-linked CMT2C accounts for a sizable fraction in this USA cohort of CMT2; it has a wide phenotypic spectrum, and vocal cord paralysis, scapular weakness and wasting, skeletal dysplasia, and hearing loss are suggestive signs for TRPV4-linked CMT2C.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/epidemiología , Atrofia Muscular Espinal/epidemiología , Mutación , Canales Catiónicos TRPV/genética , Adulto , Axones/patología , Enfermedades del Desarrollo Óseo/etiología , Enfermedad de Charcot-Marie-Tooth/genética , Enfermedad de Charcot-Marie-Tooth/fisiopatología , Exones/genética , Femenino , Asesoramiento Genético , Pérdida Auditiva/etiología , Humanos , Incidencia , Masculino , Debilidad Muscular/etiología , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/fisiopatología , Mutación Missense , Linaje , Fenotipo , Mutación Puntual , Canales Catiónicos TRPV/deficiencia , Estados Unidos/epidemiología , Parálisis de los Pliegues Vocales/etiología
12.
Am J Physiol Renal Physiol ; 318(2): F298-F314, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31790304

RESUMEN

Interstitial cystitis/bladder pain syndrome (IC/BPS) is a common chronic pelvic disorder with sensory symptoms of urinary urgency, frequency, and pain, indicating a key role for hypersensitivity of bladder-innervating sensory neurons. The inflammatory mast cell mediator histamine has long been implicated in IC/BPS, yet the direct interactions between histamine and bladder afferents remain unclear. In the present study, we show, using a mouse ex vivo bladder afferent preparation, that intravesical histamine enhanced the mechanosensitivity of subpopulations of afferents to bladder distension. Histamine also recruited "silent afferents" that were previously unresponsive to bladder distension. Furthermore, in vivo intravesical histamine enhanced activation of dorsal horn neurons within the lumbosacral spinal cord, indicating increased afferent signaling in the central nervous system. Quantitative RT-PCR revealed significant expression of histamine receptor subtypes (Hrh1-Hrh3) in mouse lumbosacral dorsal root ganglia (DRG), bladder detrusor smooth muscle, mucosa, and isolated urothelial cells. In DRG, Hrh1 was the most abundantly expressed. Acute histamine exposure evoked Ca2+ influx in select populations of DRG neurons but did not elicit calcium transients in isolated primary urothelial cells. Histamine-induced mechanical hypersensitivity ex vivo was abolished in the presence of the histamine H1 receptor antagonist pyrilamine and was not present in preparations from mice lacking transient receptor potential vanilloid 1 (TRPV1). Together, these results indicate that histamine enhances the sensitivity of bladder afferents to distension via interactions with histamine H1 receptor and TRPV1. This hypersensitivity translates to increased sensory input and activation in the spinal cord, which may underlie the symptoms of bladder hypersensitivity and pain experienced in IC/BPS.


Asunto(s)
Cistitis Intersticial/metabolismo , Histamina/administración & dosificación , Hiperalgesia/metabolismo , Mecanorreceptores/efectos de los fármacos , Mecanotransducción Celular/efectos de los fármacos , Receptores Histamínicos H1/efectos de los fármacos , Canales Catiónicos TRPV/metabolismo , Vejiga Urinaria/inervación , Administración Intravesical , Animales , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Cistitis Intersticial/fisiopatología , Femenino , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Ganglios Espinales/fisiopatología , Hiperalgesia/fisiopatología , Masculino , Mecanorreceptores/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Umbral del Dolor/efectos de los fármacos , Presión , Receptores Histamínicos H1/metabolismo , Canales Catiónicos TRPV/deficiencia , Canales Catiónicos TRPV/genética , Urotelio/efectos de los fármacos , Urotelio/metabolismo
13.
Elife ; 82019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31526479

RESUMEN

Epithelial homeostasis and regeneration require a pool of quiescent cells. How the quiescent cells are established and maintained is poorly understood. Here, we report that Trpv6, a cation channel responsible for epithelial Ca2+ absorption, functions as a key regulator of cellular quiescence. Genetic deletion and pharmacological blockade of Trpv6 promoted zebrafish epithelial cells to exit from quiescence and re-enter the cell cycle. Reintroducing Trpv6, but not its channel dead mutant, restored the quiescent state. Ca2+ imaging showed that Trpv6 is constitutively open in vivo. Mechanistically, Trpv6-mediated Ca2+ influx maintained the quiescent state by suppressing insulin-like growth factor (IGF)-mediated Akt-Tor and Erk signaling. In zebrafish epithelia and human colon carcinoma cells, Trpv6/TRPV6 elevated intracellular Ca2+ levels and activated PP2A, which down-regulated IGF signaling and promoted the quiescent state. Our findings suggest that Trpv6 mediates constitutive Ca2+ influx into epithelial cells to continuously suppress growth factor signaling and maintain the quiescent state.


Asunto(s)
Canales de Calcio/metabolismo , Proliferación Celular , Células Epiteliales/fisiología , Canales Catiónicos TRPV/metabolismo , Animales , Calcio/metabolismo , Eliminación de Gen , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Transducción de Señal , Canales Catiónicos TRPV/deficiencia , Pez Cebra , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/metabolismo
14.
J Neuroinflammation ; 16(1): 114, 2019 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-31142341

RESUMEN

BACKGROUND: Hypoxic-ischemic encephalopathy (HIE) is a serious birth complication with high incidence in both advanced and developing countries. Children surviving from HIE often have severe long-term sequela including cerebral palsy, epilepsy, and cognitive disabilities. The severity of HIE in infants is tightly associated with increased IL-1ß expression and astrocyte activation which was regulated by transient receptor potential vanilloid 1 (TRPV1), a non-selective cation channel in the TRP family. METHODS: Neonatal hypoxic ischemia (HI) and oxygen-glucose deprivation (OGD) were used to simulate HIE in vivo and in vitro. Primarily cultured astrocytes were used for investigating the expression of glial fibrillary acidic protein (GFAP), IL-1ß, Janus kinase 2 (JAK2), signal transducer and activator of transcription 3 (STAT3), and activation of the nucleotide-binding, oligomerization domain (NOD)-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome by using Western blot, q-PCR, and immunofluorescence. Brain atrophy, infarct size, and neurobehavioral disorders were evaluated by Nissl staining, 2,3,5-triphenyltetrazolium chloride monohydrate (TTC) staining and neurobehavioral tests (geotaxis reflex, cliff aversion reaction, and grip test) individually. RESULTS: Astrocytes were overactivated after neonatal HI and OGD challenge. The number of activated astrocytes, the expression level of IL-1ß, brain atrophy, and shrinking infarct size were all downregulated in TRPV1 KO mice. TRPV1 deficiency in astrocytes attenuated the expression of GFAP and IL-1ß by reducing phosphorylation of JAK2 and STAT3. Meanwhile, IL-1ß release was significantly reduced in TRPV1 deficiency astrocytes by inhibiting activation of NLRP3 inflammasome. Additionally, neonatal HI-induced neurobehavioral disorders were significantly improved in the TRPV1 KO mice. CONCLUSIONS: TRPV1 promotes activation of astrocytes and release of astrocyte-derived IL-1ß mainly via JAK2-STAT3 signaling and activation of the NLRP3 inflammasome. Our findings provide mechanistic insights into TRPV1-mediated brain damage and neurobehavioral disorders caused by neonatal HI and potentially identify astrocytic TRPV1 as a novel therapeutic target for treating HIE in the subacute stages (24 h).


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Interleucina-1beta/metabolismo , Canales Catiónicos TRPV/deficiencia , Animales , Astrocitos/patología , Encéfalo/patología , Células Cultivadas , Femenino , Hipoxia-Isquemia Encefálica/genética , Hipoxia-Isquemia Encefálica/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Canales Catiónicos TRPV/genética
15.
Sci Rep ; 9(1): 6686, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-31040368

RESUMEN

Taste buds are comprised of taste cells, which are classified into types I to IV. Transient receptor potential (TRP) channels play a significant role in taste perception. TRP vanilloid 4 (TRPV4) is a non-selective cation channel that responds to mechanical, thermal, and chemical stimuli. The present study aimed to define the function and expression of TRPV4 in taste buds using Trpv4-deficient mice. In circumvallate papillae, TRPV4 colocalized with a type IV cell and epithelial cell marker but not type I, II, or III markers. Behavioural studies showed that Trpv4 deficiency reduced sensitivity to sourness but not to sweet, umami, salty, and bitter tastes. Trpv4 deficiency significantly reduced the expression of type III cells compared with that in wild type (WT) mice in vivo and in taste bud organoid experiments. Trpv4 deficiency also significantly reduced Ki67-positive cells and ß-catenin expression compared with those in WT circumvallate papillae. Together, the present results suggest that TRPV4 contributes to sour taste sensing by regulating type III taste cell differentiation in mice.


Asunto(s)
Diferenciación Celular/genética , Canales Catiónicos TRPV/genética , Papilas Gustativas/citología , Papilas Gustativas/metabolismo , Percepción del Gusto/genética , Animales , Biomarcadores , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Ratones , Canales Catiónicos TRPV/deficiencia , Canales Catiónicos TRPV/metabolismo , beta Catenina/metabolismo
16.
Am J Physiol Renal Physiol ; 316(5): F948-F956, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30838874

RESUMEN

Tight regulation of K+ balance is fundamental for normal physiology. Reduced dietary K+ intake, which is common in Western diets, often leads to hypokalemia and associated cardiovascular- and kidney-related pathologies. The distal nephron, and, specifically, the collecting duct (CD), is the major site of controlled K+ reabsorption via H+-K+-ATPase in the state of dietary K+ deficiency. We (Mamenko MV, Boukelmoune N, Tomilin VN, Zaika OL, Jensen VB, O'Neil RG, Pochynyuk OM. Kidney Int 91: 1398-1409, 2017) have previously demonstrated that the transient receptor potential vanilloid type 4 (TRPV4) Ca2+ channel, abundantly expressed in the CD, contributes to renal K+ handling by promoting flow-induced K+ secretion. Here, we investigated a potential role of TRPV4 in controlling H+-K+-ATPase-dependent K+ reabsorption in the CD. Treatment with a K+-deficient diet (<0.01% K+) for 7 days reduced serum K+ levels in wild-type (WT) mice from 4.3 ± 0.2 to 3.3 ± 0.2 mM but not in TRPV4-/- mice (4.3 ± 0.1 and 4.2 ± 0.3 mM, respectively). Furthermore, we detected a significant reduction in 24-h urinary K+ levels in TRPV4-/- compared with WT mice upon switching to K+-deficient diet. TRPV4-/- animals also had significantly more acidic urine on a low-K+ diet, but not on a regular (0.9% K+) or high-K+ (5% K+) diet, which is consistent with increased H+-K+-ATPase activity. Moreover, we detected a greatly accelerated H+-K+-ATPase-dependent intracellular pH extrusion in freshly isolated CDs from TRPV4-/- compared with WT mice fed a K+-deficient diet. Overall, our results demonstrate a novel kaliuretic role of TRPV4 by inhibiting H+-K+-ATPase-dependent K+ reabsorption in the CD. We propose that TRPV4 inhibition could be a novel strategy to manage certain hypokalemic states in clinical settings.


Asunto(s)
Hipopotasemia/prevención & control , Túbulos Renales Colectores/metabolismo , Deficiencia de Potasio/metabolismo , Potasio en la Dieta/metabolismo , Reabsorción Renal , Canales Catiónicos TRPV/deficiencia , Animales , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Concentración de Iones de Hidrógeno , Hipopotasemia/genética , Hipopotasemia/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Deficiencia de Potasio/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Canales Catiónicos TRPV/genética
17.
Eur J Neurosci ; 50(1): 1685-1699, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30633415

RESUMEN

The proper function of the nervous system is dependent on the balance of ions and water between the intracellular and extracellular space (ECS). It has been suggested that the interaction of aquaporin-4 (AQP4) and the transient receptor potential vaniloid isoform 4 (TRPV4) channels play a role in water balance and cell volume regulation, and indirectly, of the ECS volume. Using the real-time iontophoretic method, we studied the changes of the ECS diffusion parameters: ECS volume fraction α (α = ECS volume fraction/total tissue volume) and tortuosity λ (λ2  = free/apparent diffusion coefficient) in mice with a genetic deficiency of AQP4 or TRPV4 channels, and in control animals. The used models of cytotoxic edema included: mild and severe hypotonic stress or oxygen-glucose deprivation (OGD) in situ and terminal ischemia/anoxia in vivo. This study shows that an AQP4 or TRPV4 deficit slows down the ECS volume shrinkage during severe ischemia in vivo. We further demonstrate that a TRPV4 deficit slows down the velocity and attenuates an extent of the ECS volume decrease during OGD treatment in situ. However, in any of the cytotoxic edema models in situ (OGD, mild or severe hypotonic stress), we did not detect any alterations in the cell swelling or volume regulation caused by AQP4 deficiency. Overall, our results indicate that the AQP4 and TRPV4 channels may play a crucial role in severe pathological states associated with their overexpression and enhanced cell swelling. However, detailed interplay between AQP4 and TRPV4 channels requires further studies and additional research.


Asunto(s)
Acuaporina 4/metabolismo , Edema Encefálico/metabolismo , Espacio Extracelular/metabolismo , Corteza Somatosensorial/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Acuaporina 4/deficiencia , Modelos Animales de Enfermedad , Electrocardiografía , Femenino , Paro Cardíaco/metabolismo , Hipoglucemia/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Potasio/metabolismo , Canales Catiónicos TRPV/deficiencia
18.
Glia ; 67(5): 999-1012, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30637823

RESUMEN

Pruritus is a common and disabling symptom in patients with hepatobiliary disorders, particularly in those with cholestatic features. Serum levels of lysophosphatidic acid (LPA) and its forming enzyme autotaxin were increased in patients suffering from hepatic pruritus, correlated with itch severity and response to treatment. Here we show that in a culture of dorsal root ganglia LPA 18:1 surprisingly activated a large fraction of satellite glia cells, and responses to LPA 18:1 correlated inversely with responses to neuronal expressed transient receptor potential channels. LPA 18:1 caused only a marginal activation of heterologously expressed TRPV1, and responses in dorsal root ganglion cultures from TRPV1-deficient mice were similar to controls. LPA 18:1 desensitized subsequent responsiveness to chloroquine and TGR5 agonist INT-777. The LPA 18:1-induced increase in cytoplasmatic calcium stems from the endoplasmatic reticulum. LPA receptor expression in dorsal root ganglia and Schwann cells, LPAR1 immunohistochemistry, and pharmacological results indicate a signaling pathway through LPA receptor 1. Peripheral rat Schwann cells, which are of glial lineage as the satellite glia cells, were also responsive to LPA 18:1. Summarizing, LPA 18:1 primarily activates rather glial cells than neurons, which may subsequently modulate neuronal responsiveness and sensory sensations such as itch and pain.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Lisofosfolípidos/farmacología , Neuroglía/efectos de los fármacos , Células Satélites Perineuronales/efectos de los fármacos , Células de Schwann/efectos de los fármacos , Animales , Calcio/metabolismo , Células Cultivadas , Femenino , Ganglios Espinales/citología , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/metabolismo , Receptores del Ácido Lisofosfatídico/genética , Receptores del Ácido Lisofosfatídico/metabolismo , Nervio Ciático/citología , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Canal Catiónico TRPA1/deficiencia , Canal Catiónico TRPA1/genética , Canales Catiónicos TRPV/deficiencia , Canales Catiónicos TRPV/genética
19.
J Cell Mol Med ; 23(2): 761-774, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30450767

RESUMEN

Substrate stiffness (or rigidity) of the extracellular matrix has important functions in numerous pathophysiological processes including fibrosis. Emerging data support a role for both a mechanical signal, for example, matrix stiffness, and a biochemical signal, for example, transforming growth factor ß1 (TGFß1), in epithelial-mesenchymal transition (EMT), a process critically involved in fibrosis. Here, we report evidence showing that transient receptor potential vanilloid 4 (TRPV4), a mechanosensitive channel, is the likely mediator of EMT in response to both TGFß1 and matrix stiffness. Specifically, we found that: (a) genetic ablation or pharmacological inhibition of TRPV4 blocked matrix stiffness and TGFß1-induced EMT in normal mouse primary epidermal keratinocytes (NMEKs) as determined by changes in morphology, adhesion, migration and alterations of expression of EMT markers including E-cadherin, N-cadherin (NCAD) and α-smooth muscle actin (α-SMA), and (b) TRPV4 deficiency prevented matrix stiffness-induced EMT in NMEKs over a pathophysiological range. Intriguingly, TRPV4 deletion in mice suppressed expression of mesenchymal markers, NCAD and α-SMA, in a bleomycin-induced murine skin fibrosis model. Mechanistically, we found that: (a) TRPV4 was essential for the nuclear translocation of YAP/TAZ (yes-associated protein/transcriptional coactivator with PDZ-binding motif) in response to matrix stiffness and TGFß1, (b) TRPV4 deletion inhibited both matrix stiffness- and TGFß1-induced expression of YAP/TAZ proteins and (c) TRPV4 deletion abrogated both matrix stiffness- and TGFß1-induced activation of AKT, but not Smad2/3, suggesting a mechanism by which TRPV4 activity regulates EMT in NMEKs. Altogether, these data identify a novel role for TRPV4 in regulating EMT.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas de Ciclo Celular/genética , Epidermis/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Canales Catiónicos TRPV/genética , Transactivadores/genética , Actinas/genética , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Bleomicina/administración & dosificación , Cadherinas/genética , Cadherinas/metabolismo , Adhesión Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Epidermis/metabolismo , Epidermis/patología , Transición Epitelial-Mesenquimal/genética , Matriz Extracelular/metabolismo , Fibrosis/inducido químicamente , Regulación de la Expresión Génica , Humanos , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Mecanotransducción Celular , Ratones , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/deficiencia , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Proteínas Señalizadoras YAP
20.
Cancer Lett ; 442: 15-20, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30401632

RESUMEN

The transient receptor potential vanilloid 4 (TRPV4) channel is a mechanosensor in endothelial cells (EC) that regulates cyclic strain-induced reorientation and flow-mediated nitric oxide production. We have recently demonstrated that TRPV4 expression is reduced in tumor EC and tumors grown in TRPV4KO mice exhibited enhanced growth and immature leaky vessels. However, the mechanism by which TRPV4 regulates tumor vascular integrity and metastasis is not known. Here, we demonstrate that VE-cadherin expression at the cell-cell contacts is significantly reduced in TRPV4-deficient tumor EC and TRPV4KO EC. In vivo angiogenesis assays with Matrigel of varying stiffness (700-900 Pa) revealed a significant stiffness-dependent reduction in VE-cadherin-positive vessels in Matrigel plugs from TRPV4KO mice compared with WT mice, despite an increase in vessel growth. Further, syngeneic Lewis Lung Carcinomatumor experiments demonstrated a significant decrease in VE-cadherin positive vessels in TRPV4KO tumors compared with WT. Functionally, enhanced tumor cell metastasis to the lung was observed in TRPV4KO mice. Our findings demonstrate that TRPV4 channels regulate tumor vessel integrity by maintaining VE-cadherin expression at cell-cell contacts and identifies TRPV4 as a novel target for metastasis.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Movimiento Celular , Células Endoteliales/metabolismo , Uniones Intercelulares/metabolismo , Neoplasias Pulmonares/irrigación sanguínea , Mecanotransducción Celular , Neovascularización Patológica , Canales Catiónicos TRPV/metabolismo , Animales , Antígenos CD/genética , Cadherinas/genética , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/secundario , Células Endoteliales/patología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Uniones Intercelulares/genética , Uniones Intercelulares/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Invasividad Neoplásica , Canales Catiónicos TRPV/deficiencia , Canales Catiónicos TRPV/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...