Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Am J Pathol ; 194(5): 810-827, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38325553

RESUMEN

Corneal nerve impairment contributes significantly to dry eye disease (DED) symptoms and is thought to be secondary to corneal epithelial damage. Transient receptor potential vanilloid-1 (TRPV1) channels abound in corneal nerve fibers and respond to inflammation-derived ligands, which increase in DED. TRPV1 overactivation promotes axonal degeneration in vitro, but whether it participates in DED-associated corneal nerve dysfunction is unknown. To explore this, DED was surgically induced in wild-type and TRPV1-knockout mice, which developed comparable corneal epithelial damage and reduced tear secretion. However, corneal mechanosensitivity decreased progressively only in wild-type DED mice. Sensitivity to capsaicin (TRPV1 agonist) increased in wild-type DED mice, and consistently, only this strain displayed DED-induced pain signs. Wild-type DED mice exhibited nerve degeneration throughout the corneal epithelium, whereas TRPV1-knockout DED mice only developed a reduction in the most superficial nerve endings that failed to propagate to the deeper subbasal corneal nerves. Pharmacologic TRPV1 blockade reproduced these findings in wild-type DED mice, whereas CD4+ T cells from both strains were equally pathogenic when transferred, ruling out a T-cell-mediated effect of TRPV1 deficiency. These data show that ocular desiccation triggers superficial corneal nerve damage in DED, but proximal propagation of axonal degeneration requires TRPV1 expression. Local inflammation sensitized TRPV1 channels, which increased ocular pain. Thus, ocular TRPV1 overactivation drives DED-associated corneal nerve impairment.


Asunto(s)
Lesiones de la Cornea , Síndromes de Ojo Seco , Canales de Potencial de Receptor Transitorio , Animales , Ratones , Córnea/patología , Lesiones de la Cornea/patología , Síndromes de Ojo Seco/metabolismo , Inflamación/patología , Dolor , Canales de Potencial de Receptor Transitorio/farmacología
2.
Neurogastroenterol Motil ; 36(2): e14718, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38009899

RESUMEN

BACKGROUND: Psychological stress is a major trigger for visceral hypersensitivity (VH) in irritable bowel syndrome. The zinc finger protein ZBTB20 (ZBTB20) is implicated in somatic nociception via modulating transient receptor potential (TRP) channels, but its role in the development of VH is unclear. This study aimed to investigate the role of ZBTB20/TRP channel axis in stress-induced VH. METHODS: Rats were subjected to water avoidance stress (WAS) for 10 consecutive days. Small interfering RNA (siRNA) targeting ZBTB20 was intrathecally administered. Inhibitors of TRP channels, stress hormone receptors, and nuclear factor kappa-B (NF-κB) were administered. Visceromotor response to colorectal distension was recorded. Dorsal root ganglia (DRGs) were dissected for Western blot, coimmunoprecipitation, and chromatin immunoprecipitation. The DRG-derived neuron cell line was applied for specific research. KEY RESULTS: WAS-induced VH was suppressed by the inhibitor of TRPV1, TRPA1, or TRPM8, with enhanced expression of these channels in L6-S2 DRGs. The inhibitor of glucocorticoid receptor or ß2-adrenergic receptor counteracted WAS-induced VH and TRP channel expression. Concurrently, WAS-induced stress hormone-dependent ZBTB20 expression and NF-κB activation in DRGs. Intrathecally injected ZBTB20 siRNA or an NF-κB inhibitor repressed WAS-caused effect. In cultured DRG-derived neurons, stress hormones promoted nuclear translocation of ZBTB20, which preceded p65 nuclear translocation. And, ZBTB20 siRNA suppressed stress hormone-caused NF-κB activation. Finally, WAS enhanced p65 binding to the promoter of TRPV1, TRPA1, or TRPM8 in rat DRGs. CONCLUSIONS AND INFERENCES: ZBTB20 mediates stress-induced VH via activating NF-κB/TRP channel pathway in nociceptive sensory neurons.


Asunto(s)
Canales de Potencial de Receptor Transitorio , Ratas , Animales , Canales de Potencial de Receptor Transitorio/metabolismo , Canales de Potencial de Receptor Transitorio/farmacología , FN-kappa B/metabolismo , Canales Catiónicos TRPV/metabolismo , Células Receptoras Sensoriales/metabolismo , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , Hormonas , Ganglios Espinales/metabolismo
3.
Antimicrob Agents Chemother ; 67(10): e0041923, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37728916

RESUMEN

Filarial nematode infections are a major health concern in several countries. Lymphatic filariasis is caused by Wuchereria bancrofti and Brugia spp. affecting over 120 million people. Heavy infections can lead to elephantiasis, which has serious effects on individuals' lives. Although current anthelmintics are effective at killing microfilariae in the bloodstream, they have little to no effect against adult parasites found in the lymphatic system. The anthelmintic diethylcarbamazine is one of the central pillars of lymphatic filariasis control. Recent studies have reported that diethylcarbamazine can open transient receptor potential (TRP) channels in the muscles of adult female Brugia malayi, leading to contraction and paralysis. Diethylcarbamazine has synergistic effects in combination with emodepside on Brugia, inhibiting motility: emodepside is an anthelmintic that has effects on filarial nematodes and is under trial for the treatment of river blindness. Here, we have studied the effects of diethylcarbamazine on single Brugia muscle cells by measuring the change in Ca2+ fluorescence in the muscle using Ca2+-imaging techniques. Diethylcarbamazine interacts with the transient receptor potential channel, C classification (TRPC) ortholog receptor TRP-2 to promote Ca2+ entry into the Brugia muscle cells, which can activate Slopoke (SLO-1) Ca2+-activated K+ channels, the putative target of emodepside. A combination of diethylcarbamazine and emodepside leads to a bigger Ca2+ signal than when either compound is applied alone. Our study shows that diethylcarbamazine targets TRP channels to promote Ca2+ entry that is increased by emodepside activation of SLO-1 K+ channels.


Asunto(s)
Antihelmínticos , Brugia Malayi , Filariasis Linfática , Canales de Potencial de Receptor Transitorio , Animales , Adulto , Femenino , Humanos , Dietilcarbamazina/farmacología , Dietilcarbamazina/uso terapéutico , Brugia Malayi/fisiología , Filariasis Linfática/tratamiento farmacológico , Filariasis Linfática/parasitología , Canales de Potencial de Receptor Transitorio/farmacología , Canales de Potencial de Receptor Transitorio/uso terapéutico , Antihelmínticos/farmacología , Músculos
4.
Hypertens Res ; 46(10): 2356-2367, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37532951

RESUMEN

Recent studies have focused on the contribution of vascular endothelial transient receptor potential vanilloid 4 (TRPV4) channels to hypertension. However, in hypertension, TRPV4 channels in vascular smooth muscle remain unexplored. In the present study, we performed wire myograph experiments in isolated aortas from endothelial cell specific TRPV4 channel knockout (TRPV4EC-/-) mice to demonstrate that GSK1016790A (a specific TRPV4 channel agonist) triggered aortic smooth muscle-dependent contractions from mice on a normal-salt diet, and the contractions were enhanced in high-salt diet (HSD) mice. Intracellular Ca2+ concentration ([Ca2+]i) and Ca2+ imaging assays showed that TRPV4-induced [Ca2+]i was significantly higher in aortic smooth muscle cells (ASMCs) from HSD-induced hypertensive mice, and application of an inositol trisphosphate receptor (IP3R) inhibitor markedly attenuated TRPV4-induced [Ca2+]i. IP3R2 expression was enhanced in ASMCs from HSD-induced hypertensive mice and the contractile response induced by TRPV4 was inhibited by the IP3R inhibitor. Whole-transcriptome analysis by RNA-seq and western blot assays revealed the involvement of interferon regulatory factor 7 (IRF7) in TRPV4-IRF7-IP3R2 signaling in HSD-induced hypertension. These results suggested that TRPV4 channels regulate smooth muscle-dependent contractions in high salt-induced hypertension, and this contraction involves increased [Ca2+]i, IP3R2, and IRF7 activity. Our study revealed a considerable effect of TRPV4 channels in smooth muscle-dependent contraction in mice during high-salt induced hypertension.


Asunto(s)
Aorta , Hipertensión , Canales Catiónicos TRPV , Canales de Potencial de Receptor Transitorio , Animales , Ratones , Aorta/citología , Aorta/metabolismo , Hipertensión/inducido químicamente , Hipertensión/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Cloruro de Sodio , Cloruro de Sodio Dietético/efectos adversos , Cloruro de Sodio Dietético/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Canales de Potencial de Receptor Transitorio/farmacología , Vasoconstricción
5.
J Sex Med ; 20(9): 1153-1160, 2023 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-37490314

RESUMEN

BACKGROUND: Hypoandrogenism is a cause of erectile dysfunction (ED). Vascular smooth muscle cell contraction and relaxation are regulated by TRPV1-4 channels. However, the influence of hypoandrogenism on TRPV1-4 and its relationship with erectile function remain unclear. AIM: To reveal whether hypoandrogenism affects erectile function by influencing TRPV1-4 expression in the corpus cavernosum of rats. METHODS: Male Sprague-Dawley rats (N = 36) aged 8 weeks were assigned to 6 groups at random (n = 6): sham operation, castrated, castrated + testosterone replacement, sham operation + transfection, castrated + transfection, and castrated + empty transfection. Four weeks after castration, 20 µL of lentiviral vector (1 × 108 TU/mL) carrying the TRPV4 gene was injected into the penile cavernous tissue of the transfection groups. One week after transfection, the maximum intracavernous pressure (ICPmax)/mean arterial pressure (MAP) and the content of TRPV1-4, phosphorylated eNOS (p-eNOS)/eNOS, and nitric oxide (NO) in penile cavernous tissue of each group were measured. OUTCOMES: Under low androgen conditions, TRPV4 expression in endothelial cells in the rat penile cavernosum was sharply reduced, resulting in a decrease in p-eNOS/eNOS and NO content, which could inhibit erectile function. RESULTS: In rat penile cavernous tissue, TRPV1-4 was expressed in the cell membranes of endothelial cells and smooth muscle cells. The ICPmax/MAP and the content of TRPV4, p-eNOS/eNOS, and NO end product nitrite level in rat penile cavernous tissue was markedly reduced in the castrated group as compared with the sham group (P < .05). The ICPmax/MAP and the content of TRPV4, p-eNOS/eNOS, and NO end product nitrite level in rat penile cavernous tissue were markedly improved in the castrated + transfection group vs the castrated group (P < .01). CLINICAL IMPLICATIONS: Upregulation of TRPV4 expression in penile cavernosum tissue might be a viable therapeutic for ED caused by hypoandrogenism. STRENGTHS AND LIMITATIONS: The specific mechanism of TRPV4 in ED needs to be further verified by androgen receptor or TRPV4 gene knockout experiments. CONCLUSION: Hypoandrogenism may cause ED by reducing the expression of TRPV4 in rat penile cavernous tissue. Upregulation of TRPV4 expression in penile cavernous tissue can increase the ratio of p-eNOS/eNOS and NO levels and ameliorate the erectile function of castrated rats.


Asunto(s)
Disfunción Eréctil , Canales de Potencial de Receptor Transitorio , Humanos , Ratas , Masculino , Animales , Disfunción Eréctil/etiología , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Canales Catiónicos TRPV/farmacología , Ratas Sprague-Dawley , Canales de Potencial de Receptor Transitorio/metabolismo , Canales de Potencial de Receptor Transitorio/farmacología , Canales de Potencial de Receptor Transitorio/uso terapéutico , Células Endoteliales/metabolismo , Nitritos/metabolismo , Nitritos/farmacología , Nitritos/uso terapéutico , Erección Peniana/fisiología , Pene , Óxido Nítrico Sintasa de Tipo III/metabolismo
6.
J Vet Med Sci ; 85(7): 705-714, 2023 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-37225446

RESUMEN

Transient receptor potential melastatin 4 (TRPM4) cation channels are expressed in prostate glands. However, the precise role of these channels in prostate contractility remains unclear. In this study, we examined whether TRPM4 channels were involved in adrenergic contractions in the mouse prostate gland. Adrenergic contractile responses elicited by noradrenaline or electrical field stimulation of the sympathetic nerve were isometrically recorded, and the effects of 9-phenanthrol, a specific TRPM4 channel inhibitor, on those contractile responses were investigated in mouse ventral prostate preparations. 9-phenanthrol (10 or 30 µM) inhibited noradrenaline- and sympathetic nerve-evoked contractions in a concentration-dependent manner. A similar inhibitory effect was observed with another TRPM4 channel inhibitor, 4-chloro-2-(2-(naphthalene-1-yloxy) acetamido) benzoic acid (NBA; 10 µM). Inhibition by 9-phenanthrol and NBA were much greater at lower noradrenaline concentrations and lower stimulus frequencies than those of higher concentrations or frequencies. However, 9-phenanthrol did not inhibit the noradrenaline-induced contractile response when the membrane potential was decreased to approximately 0 mV in the 140 mM K+ medium. Moreover, 9-phenanthrol does not affect noradrenaline-induced increases in spontaneous contractions of cardiac atrial preparation. This agent inhibited noradrenaline-induced contractions in the posterior aorta preparation. However, the inhibitory effect was significantly weaker than that observed in the prostate gland. These results suggest that TRPM4 channels are involved in adrenergic contractions in the mouse prostate gland, possibly through membrane depolarization by their opening; therefore, they might be potential candidates for treating benign prostatic hyperplasia.


Asunto(s)
Canales Catiónicos TRPM , Canales de Potencial de Receptor Transitorio , Masculino , Ratones , Animales , Próstata , Músculo Liso , Canales de Potencial de Receptor Transitorio/farmacología , Adrenérgicos/farmacología , Contracción Muscular , Norepinefrina/farmacología
7.
Exp Eye Res ; 228: 109405, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36773739

RESUMEN

This study aimed to determine the role of transient receptor potential vanilloid 4 (TRPV4), a calcium (Ca2+)-permeable cation channel, in the pathophysiology of retinal vascular disease. The retinal vein occlusion (RVO) murine model was created by irradiating retinal veins using lasers. TRPV4 expression and localization were evaluated in RVO mice retinas. In addition, we examined the effects of TRPV4 antagonists (RQ-00317310, HC-067047, GSK2193874, and GSK2798745) on retinal edema, blood flow, and ischemic areas in RVO mice. Furthermore, changes in the retinal expression of tumor necrosis factor (TNF)-α and aquaporin4 (AQP4) by RQ-00317310 were analyzed using Western blot. We also assessed the barrier integrity of epithelial cell monolayers using trans-endothelial electrical resistance (TEER) in Human Retinal Microvascular Endothelial Cells (HRMECs). The expression of TRPV4 was significantly increased and co-localized with glutamine synthetase (GS), a Müller glial marker, in the ganglion cell layer (GCL) of the RVO mice. Moreover, RQ-00317310 administration ameliorated the development of retinal edema and ischemia in RVO mice. In addition, the up regulation of TNF-α and down-regulation of AQP4 were lessened by the treatment with RQ-00317310. Treatment with GSK1016790A, a TRPV4 agonist, increased vascular permeability, while RQ-00317310 treatment decreased vascular endothelial growth factor (VEGF)- or TRPV4-induced retinal vascular hyperpermeability in HRMECs. These findings suggest that TRPV4 plays a role in the development of retinal edema and ischemia. Thus, TRPV4 could be a new therapeutic target against the pathological symptoms of retinal vascular diseases.


Asunto(s)
Papiledema , Enfermedades de la Retina , Canales de Potencial de Receptor Transitorio , Enfermedades Vasculares , Ratones , Humanos , Animales , Permeabilidad Capilar , Canales Catiónicos TRPV/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Canales de Potencial de Receptor Transitorio/farmacología , Células Endoteliales/metabolismo , Papiledema/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Enfermedades de la Retina/metabolismo , Enfermedades Vasculares/metabolismo
8.
Microvasc Res ; 145: 104443, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36208670

RESUMEN

OBJECTIVE: To investigate the nitric oxide synthase (NOS) and reactive oxygen species (ROS) contributions of the cutaneous vasodilator response to transient receptor potential ankyrin-1 channel (TRPA1) activation in young and older adults. MATERIALS AND METHODS: In sixteen young (20 ± 2 years, 8 females) and sixteen older adults (61 ± 5 years, 8 females), cutaneous vascular conductance normalized to maximum vasodilation (%CVCmax) was assessed at four dorsal forearm skin sites continuously perfused via microdialysis with: 1) vehicle solution (Control, 2 % dimethyl sulfoxide, 2 % Ringer, 96 % propylene glycol), 2) 10 mM Ascorbate (non-specific ROS inhibitor), 3) 10 mM L-NAME (non-specific NOS inhibitor), or 4) Ascorbate+L-NAME. The TRPA1 agonist cinnamaldehyde was co-administered at all sites [0 % (baseline), 2.9 %, 8.8 %, 26.4 %; ≥ 30 min per dose]. RESULTS: %CVCmax was not different between groups for Control, L-NAME, and Ascorbate (all p > 0.05). However, there were significant main dose effects for each site wherein %CVCmax was greater than baseline from 2.9 % to 26.4 % cinnamaldehyde for Control and Ascorbate, and at 26.4 % cinnamaldehyde for L-NAME and Ascorbate+L-NAME (all p < 0.05). For Ascorbate+L-NAME, there was a significant main group effect, wherein perfusion was 6 %CVCmax [95% CI: 2, 11, p < 0.05] greater in the older compared to the young group across all cinnamaldehyde doses. There was a significant main site effect for area under the curve wherein L-NAME and Ascorbate+L-NAME were lower than Control and Ascorbate across groups (all p < 0.05). CONCLUSION: The NOS-dependent cutaneous vasodilator response to TRPA1 activation is maintained in older adults, with no detectable contribution of ascorbate-sensitive ROS in either age group.


Asunto(s)
Canales de Potencial de Receptor Transitorio , Vasodilatación , Anciano , Femenino , Humanos , Ácido Ascórbico/farmacología , Microdiálisis , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa , Especies Reactivas de Oxígeno , Flujo Sanguíneo Regional , Piel/irrigación sanguínea , Canales de Potencial de Receptor Transitorio/farmacología , Vasodilatadores/farmacología , Masculino , Adulto Joven , Persona de Mediana Edad
9.
Hypertens Res ; 45(12): 1954-1963, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36056206

RESUMEN

Silybin is a flavonolignan extracted from the seeds of Silybum marianum that has been used as a dietary supplement for treating hepatic diseases and components of metabolic syndrome such as diabetes, obesity and hypertension. Transient receptor potential vanilloid 4 (TRPV4) channels are Ca2+-permeable, nonselective cation channels that regulate vascular endothelial function and blood flow. However, the relationship between silybin and TRPV4 channels in small mesenteric arteries remains unknown. In our study, we carried out a molecular docking experiment by using Discovery Studio v3.5 to predict the binding of silybin to TRPV4. Activation of TRPV4 with silybin was detected via intracellular Ca2+ concentration ([Ca2+]i) measurement and patch clamp experiments. The molecular docking results showed that silybin was likely to bind to the ankyrin repeat domain of TPRV4. [Ca2+]i measurements in mesenteric arterial endothelial cells (MAECs) and TRPV4-overexpressing HEK293 (TRPV4-HEK293) cells demonstrated that silybin induced Ca2+ influx by activating TRPV4 channels. The patch clamp experiments indicated that in TRPV4-HEK293 cells, silybin induced TRPV4-mediated cation currents. In addition, in high-salt-induced hypertensive mice, oral administration of silybin decreased systolic blood pressure (SBP) and significantly improved the arterial dilatory response to acetylcholine. Our findings provide the first evidence that silybin could induce mesenteric endothelium-dependent vasodilation and reduce blood pressure in high-salt-induced hypertensive mice via TRPV4 channels, thereby revealing the potential effect of silybin on preventing endothelial dysfunction-related cardiovascular diseases.


Asunto(s)
Hipertensión , Canales de Potencial de Receptor Transitorio , Ratones , Humanos , Animales , Vasodilatación/fisiología , Canales Catiónicos TRPV , Silibina/farmacología , Silibina/metabolismo , Células HEK293 , Células Endoteliales/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Canales de Potencial de Receptor Transitorio/farmacología , Simulación del Acoplamiento Molecular , Endotelio Vascular , Arterias Mesentéricas
10.
Cephalalgia ; 42(13): 1375-1386, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35861239

RESUMEN

BACKGROUND: Calcitonin gene-related peptide release in trigeminovascular system is a pivotal component of neurogenic inflammation underlying migraine pathophysiology. Transient receptor potential channels and voltage-gated KCNQ/Kv7 potassium channels expressed throughout trigeminovascular system are important targets for modulation of calcitonin gene-related peptide release. We investigated the effects of certain transient receptor potential (TRP) channels the vanilloid 1 and 4 (TRPV1 and TRPV4), the ankyrin 1 (TRPA1), and metastatin type 8 (TRPM8), and voltage-gated potassium channel (Kv7) opener retigabine on calcitonin gene-related peptide release from peripheral (dura mater and trigeminal ganglion) and central (trigeminal nucleus caudalis) trigeminal components of rats. METHODS: The experiments were carried out using well-established in-vitro preparations (hemiskull, trigeminal ganglion and trigeminal nucleus caudalis) from male Wistar rats. Agonists and antagonists of TRPV1, TRPV4, TRPA1 and TRPM8 channels, and also retigabine were tested on the in-vitro release of calcitonin gene-related peptide. Calcitonin gene-related peptide concentrations were measured using enzyme-linked immunosorbent assay. RESULTS: Agonists of these transient receptor potential channels induced calcitonin gene-related peptide release from hemiskull, trigeminal ganglion and trigeminal nucleus caudalis, respectively. The transient receptor potential channels-induced calcitonin gene-related peptide releases were blocked by their specific antagonists and reduced by retigabine. Retigabine also decreased basal calcitonin gene-related peptide releases in all preparations. CONCLUSION: Our findings suggest that favorable antagonists of these transient receptor potential channels, or Kv7 channel opener retigabine may be effective in migraine therapy by inhibiting neurogenic inflammation that requires calcitonin gene-related peptide release.


Asunto(s)
Trastornos Migrañosos , Canales de Potencial de Receptor Transitorio , Ratas , Masculino , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Canales de Potencial de Receptor Transitorio/farmacología , Canales Catiónicos TRPV , Inflamación Neurogénica , Ratas Wistar , Ganglio del Trigémino/metabolismo , Trastornos Migrañosos/tratamiento farmacológico
11.
Andrologia ; 54(8): e14477, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35596534

RESUMEN

The exact mechanism by which testosterone deficiency causes ED has not yet been elucidated. TRPC is involved in the process of smooth muscle cell contraction and relaxation. The effect of androgens on TRPCs and their relationship with erectile function are currently unclear. Thirty male SD rats were randomly divided into six groups: control group, castration group, castration + testosterone (T) group (cast + T), control + transfection group (control + trans), control + empty transfection group and castration + transfection group (cast + trans). The transfection group rats were given with lentivirus (1 × 108 TU/mL, 15 µl) carrying the siRNA targeting TRPC4 gene in the rat penile cavernous tissue at 4 weeks after castration. The tests were performed at 5 weeks after castration. Comparing the cast group with the control, the ICPmax/MAP, p-eNOS/eNOS and NO levels in the rat penile tissue were significantly lower (p < 0.01) and the level of TRPC3, TRPC4 and TRPC6 in the rat penile tissue was significantly increased (p < 0.01). When the cast + trans group was compared to the cast group, ICPmax/MAP was markedly higher (p < 0.05), and the level of the TRPC4 was remarkably lower (p < 0.05). Low androgen levels might inhibit an erectile function through up-regulation of the expression of TRPC3, TRPC4 and TRPC6 in rat penile cavernous tissue. Inhibition the level of TRPC4 in rat penile tissue may improve the erectile function in low androgen levels.


Asunto(s)
Disfunción Eréctil , Canales de Potencial de Receptor Transitorio , Andrógenos/farmacología , Animales , Humanos , Masculino , Erección Peniana , Pene , Ratas , Ratas Sprague-Dawley , Canal Catiónico TRPC6/metabolismo , Testosterona/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Canales de Potencial de Receptor Transitorio/farmacología
12.
Phytother Res ; 36(7): 2952-2963, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35537691

RESUMEN

This study investigated the vasorelaxant effects of schwarzinicine A, an alkaloid recently reported from Ficus schwarzii Koord. Regulation of calcium homeostasis in vascular smooth muscle cells (VSMC) is viewed as one of the main mechanisms for controlling blood pressure. L-type voltage-gated calcium channel (VGCC) blockers are commonly used for controlling hypertension. Recently, the transient receptor potential canonical (TRPC) channels were found in blood vessels of different animal species with evidence of their roles in the regulation of vascular contractility. In this study, we studied the mechanism of actions of schwarzinicine A focusing on its regulation of L-type VGCC and TRPC channels. Schwarzinicine A exhibited the highest vasorelaxant effect (123.1%) compared to other calcium channel blockers. It also overtly attenuated calcium-induced contractions of the rat isolated aortae in a calcium-free environment showing its mechanism to inhibit calcium influx. Fluorometric intracellular calcium recordings confirmed its inhibition of hTRPC3-, hTRPC4-, hTRPC5- and hTRPC6-mediated calcium influx into HEK cells with IC50 values of 3, 17, 19 and 7 µM, respectively. The evidence gathered in this study suggests that schwarzinicine A blocks multiple TRPC channels and L-type VGCC to exert a significant vascular relaxation response.


Asunto(s)
Canales de Potencial de Receptor Transitorio , Vasodilatación , Animales , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/farmacología , Ratas , Canales de Potencial de Receptor Transitorio/farmacología , Vasodilatadores/farmacología
13.
J Cardiovasc Pharmacol ; 79(3): 375-382, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34983913

RESUMEN

ABSTRACT: Transient receptor potential ankyrin 1 (TRPA1) channel activation induces cutaneous vasodilation in humans in vivo. However, the mechanisms underlying this response remains equivocal. We hypothesized that nitric oxide synthase (NOS) and Ca2+ activated K+ (KCa) channels contribute to the TRPA1 channel-induced cutaneous vasodilation with no involvement of cyclooxygenase (COX). Cutaneous vascular conductance (CVC) in 9 healthy young adults was assessed at 4 dorsal forearm skin sites treated by intradermal microdialysis with (1) 1.985% dimethyl sulfoxide + 0.015% lactated Ringer solution with propylene glycol (vehicle control), (2) 10 mM l-NAME, a nonselective NOS inhibitor, (3) 10 mM ketorolac, a nonselective COX inhibitor, or (4) 50 mM tetraethylammonium, a nonselective KCa channel blocker. Cinnamaldehyde, a TRPA1 channel activator, was administered to each skin site in a dose-dependent manner (2.9%, 8.8%, 26%, and 80%, each lasting ≥30 minutes). Administration of ≥8.8% cinnamaldehyde increased CVC from baseline at the vehicle control site by as much as 27.4% (95% confidence interval of 5.3; P < 0.001). NOS inhibitor attenuated the cinnamaldehyde-induced increases in CVC at the 8.8%, 26%, and 80% concentrations relative to the vehicle control site (all P ≤ 0.05). In contrast, both the COX inhibitor and KCa channel blockers did not attenuate the cinnamaldehyde induced-increases in CVC relative to the vehicle control site for all concentrations (all P ≥ 0.130). We conclude that in human skin in vivo, NOS plays a role in modulating the regulation of cutaneous vasodilation in response to TRPA1 channel activation with no detectable contributions of COX and KCa channels.


Asunto(s)
Prostaglandina-Endoperóxido Sintasas , Canales de Potencial de Receptor Transitorio , Acroleína/análogos & derivados , Inhibidores de la Ciclooxigenasa/farmacología , Humanos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Piel , Sudoración , Canal Catiónico TRPA1 , Canales de Potencial de Receptor Transitorio/farmacología , Vasodilatación , Adulto Joven
14.
Int J Obes (Lond) ; 46(1): 153-161, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34564707

RESUMEN

BACKGROUND: Bioactive dietary constituents activating Transient receptor potential (TRP) channels have emerged as promising candidates for the prevention of metabolic disorders. OBJECTIVE: The present study is an attempt to evaluate anti-obesity potential of a dietary TRP-based tri-agonist, combination of sub-effective doses of capsaicin (TRPV1 agonist), menthol (TRPM8 agonist), and cinnamaldehyde (TRPA1 agonist) in high-fat diet (HFD)-fed mice. DESIGN: Male C57BL/6 J mice divided into three groups (n = 8), were fed on normal pellet diet (NPD), or high-fat diet (HFD) (60% energy by fat) and HFD + CB (combination of capsaicin 0.4 mg/Kg, menthol 20 mg/Kg, and cinnamaldehyde 2 mg/Kg; p.o) for 12 weeks. Effects on HFD-induced weight gain, biochemical, histological and genomic changes in the WAT, BAT, liver and hypothalamus tissues were studied. RESULTS: Administration of tri-agonist prevented HFD-induced increase in weight gain, improved altered morphometric parameters, glucose homeostasis, and adipose tissue hypertrophy. Tri-agonist supplementation was found to induce browning of white adipose tissue and promote brown adipose tissue activation. Enhanced glucose utilization and prevention of lipid accumulation and insulin resistance in the liver was observed in mice supplemented with a tri-agonist. CONCLUSION: The present work provides evidence that the new approach based on combination of sub-effective doses of TRP channel agonists (TRI-AGONIST) can be employed to develop concept-based functional food for therapeutic and preventive strategies against HFD-associated pathological complications.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Canales de Potencial de Receptor Transitorio/agonistas , Acroleína/administración & dosificación , Acroleína/análogos & derivados , Acroleína/uso terapéutico , Animales , Capsaicina/administración & dosificación , Capsaicina/uso terapéutico , Dieta Alta en Grasa/efectos adversos , Dieta Alta en Grasa/métodos , Modelos Animales de Enfermedad , Mentol/administración & dosificación , Mentol/uso terapéutico , Ratones , Ratones Endogámicos C57BL/crecimiento & desarrollo , Ratones Endogámicos C57BL/metabolismo , Fenotipo , Canales de Potencial de Receptor Transitorio/farmacología
15.
Neurol Res ; 44(1): 24-37, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34256685

RESUMEN

Transient Receptor Potential Melastatin-like 7 (TRPM7), Transient Receptor Potential Melastatin-like 8 (TRPM8) and Transient Receptor Potential Vanilloid-like 1 (TRPV1) channels are expressed in neurological tissues such as brain cortex, dorsal root ganglion and hippocampal neurons and involved in several neurological diseases. The SH-SY5Y neuronal cell line is frequently used as a cellular model of neurodegenerative diseases including Parkinson's disease. The differentiated SH-SY5Y cells have much neuronal structure, function and exaggerated neuronal marker expression. However, we have less data about how differentiation induces TRP channel expression and how TRP channels have a role in cellular functions in Parkinson's disease model in SH-SY5Y cells. Hence, we aimed to investigate the effects of differentiation phenomena on TRPM7, TRPM8 and TRPV1 cation channel expression and related Ca2+ signaling. We also made some other analysis to elucidate TRP channels' function in MPP induced apoptosis, mitochondrial membrane potential levels, intracellular reactive oxygen species production, caspase 3 and caspase 9 enzyme activities in differentiated or undifferentiated SH-SY5Y neuronal cells. Herein we concluded that TRPM7, TRPM8 and TRPV1 cation channels have pivotal effects on differentiation and MPP induced Parkinson's disease model in SH-SY5Y cells.


Asunto(s)
Enfermedad de Parkinson , Canales Catiónicos TRPM , Canales de Potencial de Receptor Transitorio , Diferenciación Celular , Ganglios Espinales , Humanos , Proteínas de la Membrana/metabolismo , Enfermedad de Parkinson/metabolismo , Proteínas Serina-Treonina Quinasas , Canales Catiónicos TRPV/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Canales de Potencial de Receptor Transitorio/farmacología
16.
Stem Cell Res Ther ; 12(1): 584, 2021 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-34809715

RESUMEN

BACKGROUND: Ischemic heart diseases is one of the leading causes of death worldwide. Although revascularization timely is an effective therapeutic intervention to salvage the ischemic myocardium, reperfusion itself causes additional myocardial injury called ischemia/reperfusion (I/R) injury. Bone marrow-derived mesenchymal stem cells (MSCs) is one of the promising cells to alleviate ischemic myocardial injury. However, this cell therapy is limited by poor MSCs survival after transplantation. Here, we investigated whether sevoflurane preconditioning could promote MSCs to attenuate myocardial I/R injury via transient receptor potential canonical channel 6 (TRPC6)-induced angiogenesis. METHODS: The anti-apoptotic effect of sevoflurane preconditioning on MSCs was determined by Annexin V-FITC/propidium iodide staining. TRPC6, hypoxia-inducible factor-1α (HIF-1α), Chemokine receptor 4 (CXCR4) and vascular endothelial growth factor (VEGF) protein expressions and VEGF release from MSCs were determined after hypoxia and reoxygenation (H/R). Small interfering RNA (siRNA) was used to knock down TRPC6 gene expression in MSCs. The angiogenesis of human umbilical vein endothelial cells (HUVECs) co-cultured with MSCs was determined by Matrigel tube formation. Myocardial I/R mouse model was induced by occluding left anterior descending coronary artery for 30 min and then reperfusion. MSCs or sevoflurane preconditioned MSCs were injected around the ligature border zone 5 min before reperfusion. Left ventricle systolic function, infarction size, serum LDH, cTnI and inflammatory cytokines were determined after reperfusion. RESULTS: Sevoflurane preconditioning up-regulated TRPC6, HIF-1α, CXCR4 and VEGF expressions in MSCs and VEGF release from MSCs under H/R, which were reversed by knockdown of TRPC6 gene using siRNA in MSCs. Furthermore, sevoflurane preconditioning promoted the angiogenic and anti-inflammatory effect of HUVECs co-cultured with MSCs. Sevoflurane preconditioned MSCs improved left ventricle systolic function and alleviated myocardial infarction and inflammation in mice subjected to I/R insult. CONCLUSION: The current findings reveal that sevoflurane preconditioned MSCs boost angiogenesis in HUVECs subjected to H/R insult and attenuate myocardial I/R injury, which may be mediated by TRPC6 up-regulated HIF-1α, CXCR4 and VEGF.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Daño por Reperfusión Miocárdica , Canales de Potencial de Receptor Transitorio , Animales , Células Endoteliales/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/terapia , Sevoflurano/metabolismo , Sevoflurano/farmacología , Canal Catiónico TRPC6/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Canales de Potencial de Receptor Transitorio/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
Medicina (Kaunas) ; 55(7)2019 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-31315301

RESUMEN

Objectives: Many studies indicate the involvement of transient receptor potential (TRP) channels in the development of heart hypertrophy. However, the data is often conflicted and has originated in animal models. Here, we provide systematic analysis of TRP channels expression in human failing myocardium. Methods and results: Left-ventricular tissue samples were isolated from explanted hearts of NYHA III-IV patients undergoing heart transplants (n = 43). Quantitative real-time PCR was performed to assess the mRNA levels of TRPC, TRPM and TRPV channels. Analysis of functional, clinical and biochemical data was used to confirm an end-stage heart failure diagnosis. Compared to myocardium samples from healthy donor hearts (n = 5), we detected a distinct increase in the expression of TRPC1, TRPC5, TRPM4 and TRPM7, and decreased expression of TRPC4 and TRPV2. These changes were not dependent on gender, clinical or biochemical parameters, nor functional parameters of the heart. We detected, however, a significant correlation of TRPC1 and MEF2c expression. Conclusions: The end-stage heart failure displays distinct expressional changes of TRP channels. Our findings provide a systematic description of TRP channel expression in human heart failure. The results highlight the complex interplay between TRP channels and the need for deeper analysis of early stages of hypertrophy and heart failure development.


Asunto(s)
Insuficiencia Cardíaca/fisiopatología , Trasplante de Corazón/efectos adversos , Canales de Potencial de Receptor Transitorio/análisis , Análisis de Varianza , Femenino , Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/complicaciones , Trasplante de Corazón/métodos , Humanos , Masculino , Persona de Mediana Edad , Proteínas Serina-Treonina Quinasas/análisis , Proteínas Serina-Treonina Quinasas/sangre , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Estadísticas no Paramétricas , Canales Catiónicos TRPC/análisis , Canales Catiónicos TRPC/sangre , Canales Catiónicos TRPM/análisis , Canales Catiónicos TRPM/sangre , Canales de Potencial de Receptor Transitorio/sangre , Canales de Potencial de Receptor Transitorio/farmacología
18.
Acta Physiol (Oxf) ; 203(1): 87-98, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20950342

RESUMEN

Transient receptor potential anykrin 1 (TRPA1) is a member of the TRP superfamily, representing the sole member of the TRPA subfamily. It has many identified endogenous and exogenous agonists, comprising largely of chemical irritants and products of oxidative stress. Classically located on sensory neurone endings, TRPA1 has developed a strong presence in pain and inflammatory studies, where it is now becoming an intriguing clinical drug target. TRPA1 is increasingly recognized in a growing number of neuronal and non-neuronal locations with expanding expression and activity profiles providing evidence of a role for TRPA1 in other systems. Interest in discovering the pharmacological and functional roles of TRPA1 is increasing and diversifying into many areas. Historically, compounds now known as TRPA1 agonists have demonstrated cardiovascular activity, modulating activities in both the heart and the vasculature. Now TRPA1 has been identified as the receptor via which these compounds can act, these studies are being revisited and expanded on using current techniques. It is therefore timely to review the current knowledge of TRPA1 receptor presence and activities of relevance to the cardiovascular system, summarizing findings to date and identifying potential areas for future investigation.


Asunto(s)
Canales de Calcio/metabolismo , Canales de Calcio/farmacología , Fenómenos Fisiológicos Cardiovasculares , Sistema Cardiovascular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/farmacología , Canales de Potencial de Receptor Transitorio/metabolismo , Canales de Potencial de Receptor Transitorio/farmacología , Animales , Fenómenos Fisiológicos Cardiovasculares/efectos de los fármacos , Sistema Cardiovascular/efectos de los fármacos , Humanos , Canal Catiónico TRPA1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA