Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Curr Neurovasc Res ; 20(1): 112-123, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37013433

RESUMEN

BACKGROUND: Diabetic cardiac autonomic neuropathy (DCAN) is a serious diabetic complication with no approved pharmacological agents for its treatment. Parasympathetic system dysfunction characterized by vagal nerve damage is one of the major drivers of DCAN. The TRPC5 or transient receptor potential canonical 5 channel is a promising target in autonomic dysfunction; however, its role in vagal nerve damage and subsequent DCAN has not yet been elucidated. The present study investigated the role of the TRPC5 channel in DCAN using [N-{3-(adamantan-2-yloxy)-propyl}-3-(6-methyl-1,1-dioxo-2H-1λ6,2,4-benzothiadiazin-3-yl) propanamide)] or BTD, which is a potent TRPC5 activator. OBJECTIVES: The role of the TRPC5 channel and its activator, BTD, was investigated in the treatment of parasympathetic dysfunction associated with DCAN. METHODS: Type 1 diabetes was induced in male Sprague-Dawley rats using streptozotocin. The alterations in cardiac autonomic parameters in diabetic animals were assessed by heart rate variability, hemodynamic parameters, and baroreflex sensitivity. TRPC5's role in DCAN was investigated by treating diseased rats with BTD (1 and 3 mg/kg, i.p. for 14 days). BTD's beneficial effects in parasympathetic dysfunction were assessed by western blotting, estimating oxidative stress and inflammatory markers in the vagus nerve. RESULTS: BTD treatment (3 mg/kg, i.p.) once daily for 14 days ameliorated heart rate variability, hemodynamic dysfunction, and baroreflex sensitivity in diseased rats. BTD treatment down regulated TRPC5 expression by increasing the activity of protein kinase C in the vagus nerve. It also down-regulated the apoptotic marker CASPASE-3 and also exerted potent anti-inflammatory action on pro-inflammatory cytokines levels in the vagus. CONCLUSION: BTD ameliorated parasympathetic dysfunction associated with DCAN by virtue of its TRPC5 modulatory, anti-inflammatory, and anti-apoptotic properties.


Asunto(s)
Diabetes Mellitus Experimental , Neuropatías Diabéticas , Canales Catiónicos TRPC , Animales , Masculino , Ratas , Diabetes Mellitus Experimental/complicaciones , Neuropatías Diabéticas/tratamiento farmacológico , Corazón/inervación , Frecuencia Cardíaca , Ratas Sprague-Dawley , Canales Catiónicos TRPC/agonistas
2.
Heart Vessels ; 35(12): 1755-1765, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32844288

RESUMEN

Increased blood pressure variability (BPV) has been proved to be associated with cardiovascular morbidity and mortality. It is of great significance to elucidate the mechanism of BPV increase. The cation channel transient receptor potential canonical 6 (TRPC6) is involved in a series of cardiovascular disease. Our experiment aimed to explore the role of TRPC6 in the development of BPV increase. Sino-aortic denervation (SAD) operation was applied to establish the model of BPV increase in rats. The BPV was presented as the standard deviation to the mean of systolic or diastolic blood pressure every 1 h during 12 h of the light period. SAD was performed in male Sprague Dawley (SD) rats at the age of 10 weeks. At 8 weeks after SAD operation, the hemodynamic parameters were determined non-invasively via a Rodent Blood Pressure Analysis System. The TRPC6 expressions in myocardial and thoracic aortic tissue was determined utilizing Western Blot, immunofluorescence and quantitative RT-PCR. The expression of TRPC3 was detected as well. To investigate whether TRPC6 was a causative factor of BPV increase in SAD rats, TRPC6 activator and inhibitor with three progressively increasing doses were intraperitoneally injected to the SAD rats. We found that SAD rats presented significant augmentation of systolic and diastolic BPV with no change of BP level and heart rate. The mRNA and protein expression levels of TRPC6 in myocardial and thoracic aortic tissue in SAD rats were substantially increased, but there was no obvious change in TRPC3 expression. The systolic and diastolic BPV increase were dose-dependently exacerbated after TRPC6 activation with GSK1702934A but were dose-dependently attenuated after TRPC6 inhibition with SAR7334. In Conclusion, the TRPC6 (but not TRPC3) expressions in myocardial and thoracic aortic tissue were substantially increased in SAD rats, and TRPC6 probably played an important role in the development of BPV elevation.


Asunto(s)
Aorta Torácica/metabolismo , Presión Arterial , Barorreflejo , Frecuencia Cardíaca , Miocardio/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/inervación , Presión Arterial/efectos de los fármacos , Desnervación Autonómica , Barorreflejo/efectos de los fármacos , Seno Carotídeo/inervación , Frecuencia Cardíaca/efectos de los fármacos , Indanos/farmacología , Masculino , Ratas Sprague-Dawley , Transducción de Señal , Canales Catiónicos TRPC/agonistas , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales Catiónicos TRPC/genética , Regulación hacia Arriba
3.
Br J Pharmacol ; 176(7): 832-846, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30656647

RESUMEN

Canonical or classical transient receptor potential 4 and 5 proteins (TRPC4 and TRPC5) assemble as homomers or heteromerize with TRPC1 protein to form functional nonselective cationic channels with high calcium permeability. These channel complexes, TRPC1/4/5, are widely expressed in nervous and cardiovascular systems, also in other human tissues and cell types. It is debatable that TRPC1 protein is able to form a functional ion channel on its own. A recent explosion of molecular information about TRPC1/4/5 has emerged including knowledge of their distribution, function, and regulation suggesting these three members of the TRPC subfamily of TRP channels play crucial roles in human physiology and pathology. Therefore, these ion channels represent potential drug targets for cancer, epilepsy, anxiety, pain, and cardiac remodelling. In recent years, a number of highly selective small-molecule modulators of TRPC1/4/5 channels have been identified as being potent with improved pharmacological properties. This review will focus on recent remarkable small-molecule agonists: (-)-englerin A and tonantzitlolone and antagonists: Pico145 and HC7090, of TPRC1/4/5 channels. In addition, this work highlights other recently identified modulators of these channels such as the benzothiadiazine derivative, riluzole, ML204, clemizole, and AC1903. Together, these treasure troves of agonists and antagonists of TRPC1/4/5 channels provide valuable hints to comprehend the functional importance of these ion channels in native cells and in vivo animal models. Importantly, human diseases and disorders mediated by these proteins can be studied using these compounds to perhaps initiate drug discovery efforts to develop novel therapeutic agents.


Asunto(s)
Canales Catiónicos TRPC , Animales , Humanos , Canales Catiónicos TRPC/agonistas , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPC/fisiología
4.
Br J Pharmacol ; 175(16): 3361-3368, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29859013

RESUMEN

BACKGROUND AND PURPOSE: The diterpene ester tonantzitlolone (TZL) is a natural product, which displays cytotoxicity towards certain types of cancer cell such as renal cell carcinoma cells. The effect is similar to that of (-)-englerin A, and so, although it is chemically distinct, we investigated whether TZL also targets transient receptor potential canonical (TRPC) channels of the 1, 4 and 5 type (TRPC1/4/5 channels). EXPERIMENTAL APPROACH: The effects of TZL on renal cell carcinoma A498 cells natively expressing TRPC1 and TRPC4, modified HEK293 cells overexpressing TRPC4, TRPC5, TRPC4-TRPC1 or TRPC5-TRPC1 concatemer, TRPC3 or TRPM2, or CHO cells overexpressing TRPV4 were studied by determining changes in intracellular Ca2+ , or whole-cell or excised membrane patch-clamp electrophysiology. KEY RESULTS: TZL induced an elevation of intracellular Ca2+ in A498 cells, similar to that evoked by englerin A. TZL activated overexpressed channels with EC50 values of 123 nM (TRPC4), 83 nM (TRPC5), 140 nM (TRPC4-TRPC1) and 61 nM (TRPC5-TRPC1). These effects of TZL were reversible on wash-out and potently inhibited by the TRPC1/4/5 inhibitor Pico145. TZL activated TRPC5 channels when bath-applied to excised outside-out but not inside-out patches. TZL failed to activate endogenous store-operated Ca2+ entry or overexpressed TRPC3, TRPV4 or TRPM2 channels in HEK 293 cells. CONCLUSIONS AND IMPLICATIONS: TZL is a novel potent agonist for TRPC1/4/5 channels, which should be useful for testing the functionality of this type of ion channel and understanding how TRPC1/4/5 agonists achieve selective cytotoxicity against certain types of cancer cell.


Asunto(s)
Antineoplásicos/farmacología , Diterpenos/farmacología , Compuestos Macrocíclicos/farmacología , Canales Catiónicos TRPC/agonistas , Animales , Células CHO , Calcio/fisiología , Línea Celular Tumoral , Cricetulus , Células HEK293 , Humanos , Canales Catiónicos TRPC/fisiología
5.
J Am Soc Nephrol ; 29(2): 409-415, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29061651

RESUMEN

Transient receptor potential channel 5 (TRPC5) is highly expressed in brain and kidney and mediates calcium influx and promotes cell migration. In the kidney, loss of TRPC5 function has been reported to benefit kidney filter dynamics by balancing podocyte cytoskeletal remodeling. However, in vivo gain-in-function studies of TRPC5 with respect to kidney function have not been reported. To address this gap, we developed two transgenic mouse models on the C57BL/6 background by overexpressing either wild-type TRPC5 or a TRPC5 ion-pore mutant. Compared with nontransgenic controls, neither transgenic model exhibited an increase in proteinuria at 8 months of age or a difference in LPS-induced albuminuria. Moreover, activation of TRPC5 by Englerin A did not stimulate proteinuria, and inhibition of TRPC5 by ML204 did not significantly lower the level of LPS-induced proteinuria in any group. Collectively, these data suggest that the overexpression or activation of the TRPC5 ion channel does not cause kidney barrier injury or aggravate such injury under pathologic conditions.


Asunto(s)
Albuminuria/genética , Enfermedades Renales/genética , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo , Albuminuria/inducido químicamente , Animales , Encéfalo/metabolismo , Femenino , Indoles/farmacología , Enfermedades Renales/inducido químicamente , Enfermedades Renales/mortalidad , Lipopolisacáridos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Piperidinas/farmacología , Podocitos/ultraestructura , Sesquiterpenos de Guayano/farmacología , Canales Catiónicos TRPC/agonistas , Canales Catiónicos TRPC/antagonistas & inhibidores
6.
Cell Calcium ; 66: 10-18, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28807145

RESUMEN

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


Asunto(s)
Benzotiadiazinas/farmacología , Potenciales de la Membrana/efectos de los fármacos , Metilprednisolona/farmacología , Canales Catiónicos TRPC/metabolismo , Animales , Benzotiadiazinas/química , Señalización del Calcio/efectos de los fármacos , Células HEK293 , Humanos , Metilprednisolona/química , Ratones , Microscopía Confocal , Técnicas de Placa-Clamp , Fosfoinositido Fosfolipasa C/metabolismo , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/metabolismo , Subunidades de Proteína/agonistas , Subunidades de Proteína/metabolismo , Canales Catiónicos TRPC/agonistas , Canales Catiónicos TRPC/genética
7.
J Pharmacol Exp Ther ; 362(3): 368-377, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28620120

RESUMEN

The nonselective cation channel transient receptor potential ankryn subtype family 1 (TRPA1) is expressed in neurons of dorsal root ganglia and trigeminal ganglia and also in vagal afferent neurons that innervate the lungs and gastrointestinal tract. Many TRPA1 agonists are reactive electrophilic compounds that form covalent adducts with TRPA1. Allyl isothiocyanate (AITC), the common agonist used to identify TRPA1, contains an electrophilic group that covalently binds with cysteine residues of TRPA1 and confers a structural change on the channel. There is scientific motivation to identify additional compounds that can activate TRPA1 with different mechanisms of channel gating. We provide evidence that ethyl vanillin (EVA) is a TRPA1 agonist. Using fluorescent calcium imaging and whole-cell patch-clamp electrophysiology on dissociated rat vagal afferent neurons and TRPA1-transfected COS-7 cells, we discovered that EVA activates cells also activated by AITC. Both agonists display similar current profiles and conductances. Pretreatment with A967079, a selective TRPA1 antagonist, blocks the EVA response as well as the AITC response. Furthermore, EVA does not activate vagal afferent neurons from TRPA1 knockout mice, showing selectivity for TRPA1 in this tissue. Interestingly, EVA appears to be pharmacologically different from AITC as a TRPA1 agonist. When AITC is applied before EVA, the EVA response is occluded. However, they both require intracellular oxidation to activate TRPA1. These findings suggest that EVA activates TRPA1 but via a distinct mechanism that may provide greater ease for study in native systems compared with AITC and may shed light on differential modes of TRPA1 gating by ligand types.


Asunto(s)
Benzaldehídos/farmacología , Canales Catiónicos TRPC/agonistas , Animales , Células COS , Chlorocebus aethiops , Células HEK293 , Humanos , Isotiocianatos/farmacología , Masculino , Ratones , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/metabolismo , Ganglio Nudoso/citología , Ganglio Nudoso/efectos de los fármacos , Oximas/farmacología , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Canal Catiónico TRPA1 , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales Catiónicos TRPC/genética
8.
J Med Chem ; 60(11): 4680-4692, 2017 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-28395140

RESUMEN

Transient receptor potential canonical 3/6/7 (TRPC3/6/7) are highly homologous receptor-operated nonselective cation channels. Despite their physiological significance, very few selective and potent agonists are available for functional examination of these channels. Using a cell-based high throughput screening approach, a lead compound with the pyrazolopyrimidine skeleton was identified as a TRPC6 agonist. Synthetic schemes for the lead and its analogues were established, and structural-activity relationship studies were carried out. A series of potent and direct agonists of TRPC3/6/7 channels were identified, and among them, 4m-4p have a potency order of TRPC3 > C7 > C6, with 4n being the most potent with an EC50 of <20 nM on TRPC3. Importantly, these compounds exhibited no stimulatory activity on related TRP channels. The potent and selective compounds described here should be suitable for evaluation of the roles of TRPC channels in the physiology and pathogenesis of diseases, including glomerulosclerosis and cancer.


Asunto(s)
Pirazoles/química , Pirimidinas/farmacología , Canales Catiónicos TRPC/agonistas , Células HEK293 , Humanos , Pirimidinas/química , Relación Estructura-Actividad , Canal Catiónico TRPC6
10.
Inflamm Res ; 66(4): 311-322, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27904941

RESUMEN

BACKGROUND: Although TRPA1, SP, histamine and 5-hydroxytryptamine (5-HT) have recognized contribution to nociceptive mechanisms, little is known about how they interact with each other to mediate inflammatory pain in vivo. In this study we evaluated whether TRPA1, SP, histamine and 5-HT interact, in an interdependent way, to induce nociception in vivo. METHODS AND RESULTS: The subcutaneous injection of the TRPA1 agonist allyl isothiocyanate (AITC) into the rat's hind paw induced a dose-dependent and short lasting behavioral nociceptive response that was blocked by the co-administration of the TRPA1 antagonist, HC030031, or by the pretreatment with antisense ODN against TRPA1. AITC-induced nociception was significantly decreased by the co-administration of selective antagonists for the NK1 receptor for substance P, the H1 receptor for histamine and the 5-HT1A or 3 receptors for 5-HT. Histamine- or 5-HT-induced nociception was decreased by the pretreatment with antisense ODN against TRPA1. These findings suggest that AITC-induced nociception depends on substance P, histamine and 5-HT, while histamine- or 5-HT-induced nociception depends on TRPA1. Most important, AITC interact in a synergistic way with histamine, 5-HT or substance P, since their combination at non-nociceptive doses induced a nociceptive response much higher than that expected by the sum of the effect of each one alone. This synergistic effect is dependent on the H1, 5-HT1A or 3 receptors. CONCLUSION: Together, these findings suggest a self-sustainable cycle around TRPA1, no matter where the cycle is initiated each step is achieved and even subeffective activation of more than one step results in a synergistic activation of the overall cycle.


Asunto(s)
Histamina/metabolismo , Dolor/metabolismo , Serotonina/metabolismo , Sustancia P/metabolismo , Canales Catiónicos TRPC/metabolismo , Acetanilidas/farmacología , Animales , Antagonistas de los Receptores Histamínicos H1/farmacología , Isotiocianatos , Masculino , Oligonucleótidos Antisentido/farmacología , Dolor/inducido químicamente , Piperazinas/farmacología , Purinas/farmacología , Pirilamina/farmacología , Quinuclidinas/farmacología , Ratas Wistar , Receptor de Serotonina 5-HT1A/metabolismo , Receptores Histamínicos H1/metabolismo , Receptores de Neuroquinina-1/metabolismo , Receptores de Serotonina 5-HT3/metabolismo , Antagonistas de la Serotonina/farmacología , Canal Catiónico TRPA1 , Canales Catiónicos TRPC/agonistas , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales Catiónicos TRPC/genética , p-Metoxi-N-metilfenetilamina/farmacología
11.
J Neurosci ; 36(47): 11837-11850, 2016 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-27881772

RESUMEN

Mushroom dendritic spine structures are essential for memory storage and the loss of mushroom spines may explain memory defects in aging and Alzheimer's disease (AD). The stability of mushroom spines depends on stromal interaction molecule 2 (STIM2)-mediated neuronal-store-operated Ca2+ influx (nSOC) pathway, which is compromised in AD mouse models, in aging neurons, and in sporadic AD patients. Here, we demonstrate that the Transient Receptor Potential Canonical 6 (TRPC6) and Orai2 channels form a STIM2-regulated nSOC Ca2+ channel complex in hippocampal mushroom spines. We further demonstrate that a known TRPC6 activator, hyperforin, and a novel nSOC positive modulator, NSN21778 (NSN), can stimulate activity of nSOC pathway in the spines and rescue mushroom spine loss in both presenilin and APP knock-in mouse models of AD. We further show that NSN rescues hippocampal long-term potentiation impairment in APP knock-in mouse model. We conclude that the STIM2-regulated TRPC6/Orai2 nSOC channel complex in dendritic mushroom spines is a new therapeutic target for the treatment of memory loss in aging and AD and that NSN is a potential candidate molecule for therapeutic intervention in brain aging and AD. SIGNIFICANCE STATEMENT: Mushroom dendritic spine structures are essential for memory storage and the loss of mushroom spines may explain memory defects in Alzheimer's disease (AD). This study demonstrated that Transient Receptor Potential Canonical 6 (TRPC6) and Orai2 form stromal interaction molecule 2 (STIM2)-regulated neuronal-store-operated Ca2+ influx (nSOC) channel complex in hippocampal synapse and the resulting Ca2+ influx is critical for long-term maintenance of mushroom spines in hippocampal neurons. A novel nSOC-positive modulator, NSN21778 (NSN), rescues mushroom spine loss and synaptic plasticity impairment in AD mice models. The TRPC6/Orai2 nSOC channel complex is a new therapeutic target and NSN is a potential candidate molecule for therapeutic intervention in brain aging and AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Agonistas de los Canales de Calcio/administración & dosificación , Señalización del Calcio/fisiología , Espinas Dendríticas/metabolismo , Proteína ORAI2/metabolismo , Canales Catiónicos TRPC/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Espinas Dendríticas/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína ORAI2/agonistas , Sinapsis/efectos de los fármacos , Potenciales Sinápticos/efectos de los fármacos , Potenciales Sinápticos/fisiología , Canales Catiónicos TRPC/agonistas , Canal Catiónico TRPC6
12.
Neuroscience ; 339: 1-11, 2016 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-27687800

RESUMEN

We previously showed that magnesium sulfate (MS) has systemic antinociceptive and local peripheral pronociceptive effects. The role of transient receptor potential (TRP) channels and acid-sensing ion channels (ASICs) in the mechanism of action of MS has not been investigated in detail. The aim of this study was to explore the participation of TRP channels in the pronociceptive action of MS in rats after its intraplantar injection. The paw withdrawal threshold (PWT) to mechanical stimuli was measured by the electronic von Frey test. Drugs that were tested were either co-administered with an isotonic pH-unadjusted or pH-adjusted solution of MS intraplantarily, or to the contralateral paw to exclude systemic effects. We found that the subcutaneous administration of both pH-adjusted (7.4) and pH-unadjusted (about 6.0) isotonic (6.2% w/v in water) solutions of MS induce the pain at the injection site. The pH-unadjusted MS solution-induced mechanical hyperalgesia decreased in a dose-dependent manner as a consequence of co-injection of capsazepine, a selective TRPV1 antagonist (20, 100 and 500pmol/paw), RN-1734, a selective TRPV4 antagonist (1.55, 3.1 and 6.2µmol/paw), HC-030031, a selective TRPA1 antagonist (5.6, 28.1 and 140nmol/paw), and amiloride hydrochloride, a non-selective ASIC inhibitor (0.83, 2.5 and 7.55µmol/paw). In pH-adjusted MS-induced hyperalgesia, the highest doses of TRPV1, TRPV4 and TRPA1 antagonists displayed effects that were, respectively, either similar, less pronounced or delayed in comparison to the effect induced by administration of the pH-unadjusted MS solution; the ASIC antagonist did not have any effect. These results suggest that the MS-induced local peripheral mechanical hyperalgesia is mediated via modulation of the activity of peripheral TRPV1, TRPV4, TRPA1 and ASICs. Specific local inhibition of TRP channels represents a novel approach to treating local injection-related pain.


Asunto(s)
Canales Iónicos Sensibles al Ácido/metabolismo , Dolor/metabolismo , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPV/metabolismo , Analgésicos/farmacología , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Concentración de Iones de Hidrógeno , Sulfato de Magnesio , Masculino , Moduladores del Transporte de Membrana/farmacología , Dolor/tratamiento farmacológico , Distribución Aleatoria , Ratas Wistar , Canal Catiónico TRPA1 , Canales Catiónicos TRPC/agonistas , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/antagonistas & inhibidores , Tacto
13.
J Physiol Pharmacol ; 67(4): 587-594, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27779479

RESUMEN

Methylglyoxal (MG) is a reactive carbonyl compound generated in diabetes mellitus. MG is an established transient receptor potential ankyrin 1 (TRPA1) channel agonist that contributes to TRPA1-mediated diabetic pain hypersensitivity. Here we studied whether exposure to diabetes and thereby to elevated endogenous MG modulates hypersensitivity induced by intradermal MG. Moreover, since diabetes induces endoplasmic reticulum (ER) stress, we compared the role of TRPA1 in diabetes and ER stress by assessing whether tunicamycin-induced ER stress, without diabetes, produces TRPA1-mediated pain hypersensitivity and by assessing whether ER stress and diabetes have similar modulatory effects on MG-induced hypersensitivity. In vitro patch clamp recording was performed to assess whether tunicamycin is a TRPA1 agonist. Behavioral tests showed that mechanical hypersensitivity induced by MG is reduced in diabetes and ER stress. In healthy controls, hypersensitivity induced by MG was reduced when MG was administered for the second time in the same but not adjacent plantar sites. Hypersensitivity induced by ER stress was reversed by pharmacological blocking of TRPA1. In vitro patch clamp recording indicated that tunicamycin itself (30 µM) is not a TRPA1 agonist. The results indicate that pain hypersensitivity induced by non-diabetic ER stress as well as that induced by diabetes is mediated TRPA1. Reduction of MG-induced hypersensitivity in diabetes or ER stress may, at least partly, be explained by peripheral mechanisms.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Hiperalgesia/inducido químicamente , Piruvaldehído/farmacología , Canales Catiónicos TRPC/agonistas , Tunicamicina/farmacología , Administración Cutánea , Animales , Conducta Animal/efectos de los fármacos , Diabetes Mellitus Experimental , Células HEK293 , Humanos , Masculino , Dimensión del Dolor , Estimulación Física , Ratas , Piel/efectos de los fármacos , Canal Catiónico TRPA1 , Canales Catiónicos TRPC/fisiología
14.
Mol Pain ; 122016.
Artículo en Inglés | MEDLINE | ID: mdl-27411353

RESUMEN

The primary complaint of burn victims is an intense, often devastating spontaneous pain, with persistence of mechanical and thermal allodynia. The transient receptor potential channels, TRPV1 and TRPA1, are expressed by a subset of nociceptive sensory neurons and contribute to inflammatory hypersensitivity. Although their function in the periphery is well known, a role for these TRP channels in central pain mechanisms is less well defined. Lipid agonists of TRPV1 are released from peripheral tissues via enzymatic oxidation after burn injury; however, it is not known if burn injury triggers the release of oxidized lipids in the spinal cord. Accordingly, we evaluated whether burn injury evoked the central release of oxidized lipids . Analysis of lipid extracts of spinal cord tissue with HPLC-MS revealed a significant increase in levels of the epoxide and diol metabolites of linoleic acid: 9,10-DiHOME, 12,13-DiHOME, 9(10)-EpOME, and 12(13)-EpOME, that was reduced after intrathecal (i.t.) injection of the oxidative enzyme inhibitor ketoconazole. Moreover, we found that these four lipid metabolites were capable of specifically activating both TRPV1 and TRPA1. Intrathecal injection of specific antagonists to TRPV1 (AMG-517) or TRPA1 (HC-030031) significantly reduced post-burn mechanical and thermal allodynia. Finally, i.t. injection of ketoconazole significantly reversed post-burn mechanical and thermal allodynia. Our data indicate that spinal cord TRPV1 and TRPA1 contributes to pain after burn and identifies a novel class of oxidized lipids elevated in the spinal cord after burn injury. Since the management of burn pain is problematic, these findings point to a novel approach for treating post-burn pain.


Asunto(s)
Quemaduras/complicaciones , Hiperalgesia/etiología , Hiperalgesia/metabolismo , Activación del Canal Iónico , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Quemaduras/patología , Células CHO , Cricetinae , Cricetulus , Hiperalgesia/patología , Activación del Canal Iónico/efectos de los fármacos , Ácido Linoleico/metabolismo , Ácido Linoleico/farmacología , Masculino , Oxidación-Reducción/efectos de los fármacos , Ratas Sprague-Dawley , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/patología , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Médula Espinal/patología , Canal Catiónico TRPA1 , Canales Catiónicos TRPC/agonistas , Canales Catiónicos TRPV/agonistas , Factores de Tiempo
15.
Naunyn Schmiedebergs Arch Pharmacol ; 389(9): 1009-20, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27342418

RESUMEN

Besides their deleterious action on cardiac muscle, anthracycline-type cytostatic agents exert significant neurotoxic effects on primary sensory neurons. Since cardiac sensory nerves confer protective effects on heart muscle and share common traits with cutaneous chemosensitive nerves, this study examined the effects of cardiotoxic doses of adriamycin on the function and morphology of epidermal nerves. Sensory neurogenic vasodilatation, plasma extravasation, and the neural CGRP release evoked by TRPV1 and TRPA1 agonists in vitro were examined by using laser Doppler flowmetry, the Evans blue technique, and ELISA, respectively. Carrageenan-induced hyperalgesia was assessed with the Hargreaves method. Immunohistochemistry was utilized to study cutaneous innervation. Adriamycin treatment resulted in profound reductions in the cutaneous neurogenic sensory vasodilatation and plasma extravasation evoked by the TRPV1 and TRPA1 agonists capsaicin and mustard oil, respectively. The in vitro capsaicin-, but not high potassium-evoked neural release of the major sensory neuropeptide, CGRP, was markedly attenuated after adriamycin treatment. Carrageenan-induced inflammatory hyperalgesia was largely abolished following the administration of adriamycin. Immunohistochemistry revealed a substantial loss of epidermal TRPV1-expressing nociceptive nerves and a marked thinning of the epidermis. These findings indicate impairments in the functions of TRPV1 and TRPA1 receptors expressed on cutaneous chemosensitive nociceptive nerves and the loss of epidermal axons following the administration of cardiotoxic doses of adriamycin. Monitoring of the cutaneous nociceptor function in the course of adriamycin therapy may well be of predictive value for early detection of the deterioration of cardiac nerves which confer protection against the deleterious effects of the drug.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Doxorrubicina/toxicidad , Hiperalgesia/prevención & control , Nocicepción/efectos de los fármacos , Células Receptoras Sensoriales/efectos de los fármacos , Piel/inervación , Animales , Biomarcadores/metabolismo , Péptido Relacionado con Gen de Calcitonina/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Capsaicina/farmacología , Cardiotoxicidad , Carragenina , Modelos Animales de Enfermedad , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Masculino , Actividad Motora/efectos de los fármacos , Planta de la Mostaza , Aceites de Plantas/farmacología , Ratas Wistar , Flujo Sanguíneo Regional/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Piel/irrigación sanguínea , Canal Catiónico TRPA1 , Canales Catiónicos TRPC/agonistas , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/metabolismo , Factores de Tiempo , Vasodilatación/efectos de los fármacos
16.
Am J Physiol Cell Physiol ; 310(11): C1001-9, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27147559

RESUMEN

Anoctamin-1 [ANO1, also known as transmembrane protein 16A (TMEM16A)] is a Ca(2+)-activated Cl(-) channel expressed in arterial myocytes that regulates membrane potential and contractility. Signaling mechanisms that control ANO1 activity in arterial myocytes are poorly understood. In cerebral artery myocytes, ANO1 channels are activated by local Ca(2+) signals generated by plasma membrane nonselective cation channels, but the molecular identity of these proteins is unclear. Arterial myocytes express several different nonselective cation channels, including multiple members of the transient receptor potential receptor (TRP) family. The goal of this study was to identify localized ion channels that control ANO1 currents in cerebral artery myocytes. Coimmunoprecipitation and immunofluorescence resonance energy transfer microscopy experiments indicate that ANO1 and canonical TRP 6 (TRPC6) channels are present in the same macromolecular complex and localize in close spatial proximity in the myocyte plasma membrane. In contrast, ANO1 is not near TRPC3, TRP melastatin 4, or inositol trisphosphate receptor 1 channels. Hyp9, a selective TRPC6 channel activator, stimulated Cl(-) currents in myocytes that were blocked by T16Ainh-A01, an ANO1 inhibitor, ANO1 knockdown using siRNA, and equimolar replacement of intracellular EGTA with BAPTA, a fast Ca(2+) chelator that abolishes local Ca(2+) signaling. Hyp9 constricted pressurized cerebral arteries, and this response was attenuated by T16Ainh-A01. In contrast, T16Ainh-A01 did not alter depolarization-induced (60 mM K(+)) vasoconstriction. These data indicate that TRPC6 channels generate a local intracellular Ca(2+) signal that activates nearby ANO1 channels in myocytes to stimulate vasoconstriction.


Asunto(s)
Canales de Cloruro/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Canales Catiónicos TRPC/metabolismo , Vasoconstricción , Animales , Anoctamina-1 , Quelantes del Calcio/farmacología , Señalización del Calcio , Arterias Cerebrales/metabolismo , Canales de Cloruro/antagonistas & inhibidores , Canales de Cloruro/genética , Masculino , Potenciales de la Membrana , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Interferencia de ARN , Ratas Sprague-Dawley , Canales Catiónicos TRPC/agonistas , Técnicas de Cultivo de Tejidos , Transfección , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología
17.
Cephalalgia ; 36(9): 875-86, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26984967

RESUMEN

BACKGROUND: Administration of onabotulinumtoxinA (BoNT-A) to peripheral tissues outside the calvaria reduces the number of days chronic migraine patients experience headache. Because the headache phase of a migraine attack, especially those preceded by aura, is thought to involve activation of meningeal nociceptors by endogenous stimuli such as changes in intracranial pressure (i.e. mechanical) or chemical irritants that appear in the meninges as a result of a yet-to-be-discovered sequence of molecular/cellular events triggered by the aura, we sought to determine whether extracranial injections of BoNT-A alter the chemosensitivity of meningeal nociceptors to stimulation of their intracranial receptive fields. MATERIAL AND METHODS: Using electrophysiological techniques, we identified 161 C- and 135 Aδ-meningeal nociceptors in rats and determined their mechanical response threshold and responsiveness to chemical stimulation of their dural receptive fields with TRPV1 and TRPA1 agonists seven days after BoNT-A administration to different extracranial sites. Two paradigms were compared: distribution of 5 U BoNT-A to the lambdoid and sagittal sutures alone, and 1.25 U to the sutures and 3.75 U to the temporalis and trapezius muscles. RESULTS: Seven days after it was administered to tissues outside the calvaria, BoNT-A inhibited responses of C-type meningeal nociceptors to stimulation of their intracranial dural receptive fields with the TRPV1 agonist capsaicin and the TRPA1 agonist mustard oil. BoNT-A inhibition of responses to capsaicin was more effective when the entire dose was injected along the suture lines than when it was injected into muscles and sutures. As in our previous study, BoNT-A had no effect on non-noxious mechanosensitivity of C-fibers or on responsiveness of Aδ-fibers to mechanical and chemical stimulation. DISCUSSION: This study demonstrates that extracranial administration of BoNT-A suppresses meningeal nociceptors' responses to stimulation of their intracranial dural receptive fields with capsaicin and mustard oil. The findings suggest that surface expression of TRPV1 and TRPA1 channels in dural nerve endings of meningeal nociceptors is reduced seven days after extracranial administration of BoNT-A. In the context of chronic migraine, reduced sensitivity to molecules that activate meningeal nociceptors through the TRPV1 and TRPA1 channels can be important for BoNT-A's ability to act as a prophylactic.


Asunto(s)
Toxinas Botulínicas Tipo A/farmacología , Meninges/efectos de los fármacos , Trastornos Migrañosos/fisiopatología , Fármacos Neuromusculares/farmacología , Nociceptores/efectos de los fármacos , Animales , Capsaicina/farmacología , Suturas Craneales/efectos de los fármacos , Masculino , Músculo Esquelético/efectos de los fármacos , Planta de la Mostaza , Aceites de Plantas/farmacología , Ratas , Ratas Sprague-Dawley , Fármacos del Sistema Sensorial/farmacología , Canal Catiónico TRPA1 , Canales Catiónicos TRPC/agonistas , Canales Catiónicos TRPV/agonistas
18.
Mol Pharmacol ; 89(3): 348-63, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26733543

RESUMEN

Transient receptor potential canonical (TRPC) proteins form Ca(2+)-permeable cation channels activated upon stimulation of metabotropic receptors coupled to phospholipase C. Among the TRPC subfamily, TRPC3 and TRPC6 channels activated directly by diacylglycerol (DAG) play important roles in brain-derived neurotrophic factor (BDNF) signaling, promoting neuronal development and survival. In various disease models, BDNF restores neurologic deficits, but its therapeutic potential is limited by its poor pharmacokinetic profile. Elucidation of a framework for designing small molecules, which elicit BDNF-like activity via TRPC3 and TRPC6, establishes a solid basis to overcome this limitation. We discovered, through library screening, a group of piperazine-derived compounds that activate DAG-activated TRPC3/TRPC6/TRPC7 channels. The compounds [4-(5-chloro-2-methylphenyl)piperazin-1-yl](3-fluorophenyl)methanone (PPZ1) and 2-[4-(2,3-dimethylphenyl)piperazin-1-yl]-N-(2-ethoxyphenyl)acetamide (PPZ2) activated, in a dose-dependent manner, recombinant TRPC3/TRPC6/TRPC7 channels, but not other TRPCs, in human embryonic kidney cells. PPZ2 activated native TRPC6-like channels in smooth muscle cells isolated from rabbit portal vein. Also, PPZ2 evoked cation currents and Ca(2+) influx in rat cultured central neurons. Strikingly, both compounds induced BDNF-like neurite growth and neuroprotection, which were abolished by a knockdown or inhibition of TRPC3/TRPC6/TRPC7 in cultured neurons. Inhibitors of Ca(2+) signaling pathways, except calcineurin, impaired neurite outgrowth promotion induced by PPZ compounds. PPZ2 increased activation of the Ca(2+)-dependent transcription factor, cAMP response element-binding protein. These findings suggest that Ca(2+) signaling mediated by activation of DAG-activated TRPC channels underlies neurotrophic effects of PPZ compounds. Thus, piperazine-derived activators of DAG-activated TRPC channels provide important insights for future development of a new class of synthetic neurotrophic drugs.


Asunto(s)
Factores de Crecimiento Nervioso/metabolismo , Piperazinas/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Evaluación Preclínica de Medicamentos/métodos , Femenino , Células HEK293 , Humanos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Factores de Crecimiento Nervioso/química , Factores de Crecimiento Nervioso/farmacología , Piperazinas/química , Piperazinas/farmacología , Conejos , Ratas , Ratas Wistar , Canales Catiónicos TRPC/agonistas
19.
Behav Pharmacol ; 27(1): 29-36, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26274042

RESUMEN

A sensitive response of the nervous system to changes in temperature is of predominant importance for homeotherms to maintain a stable body temperature. A number of temperature-sensitive transient receptor potential (TRP) ion channels have been studied as nociceptors that respond to extreme temperatures and harmful chemicals. Recent findings in the field of pain have established a family of six thermo-TRP channels (TRPA1, TRPM8, TRPV1, TRPV2, TRPV3, and TRPV4) that exhibit sensitivity to increases or decreases in temperature, as well as to chemical substances eliciting the respective hot or cold sensations. In this study, we used behavioral methods to investigate whether mustard oil (allyl isothiocyanate) and capsaicin affect the sensitivity to heat, innocuous and noxious cold, and mechanical stimuli in male rats. The results obtained indicate that TRPA1 and TRPV1 channels are clearly involved in pain reactions, and the TRPA1 agonist allyl isothiocyanate enhances the heat pain sensitivity, possibly by indirectly modulating TRPV1 channels coexpressed in nociceptors with TRPA1. Overall, our data support the role of thermosensitive TRPA1 and TRPV1 channels in pain modulation and show that these two thermoreceptor channels are in a synergistic and/or conditional relationship with noxious heat and cold cutaneous stimulation.


Asunto(s)
Nocicepción/fisiología , Dolor Nociceptivo/metabolismo , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Capsaicina/farmacología , Frío , Relación Dosis-Respuesta a Droga , Calor , Masculino , Planta de la Mostaza , Aceites de Plantas/farmacología , Ratas Wistar , Fármacos del Sistema Sensorial/farmacología , Canal Catiónico TRPA1 , Canales Catiónicos TRPC/agonistas , Canales Catiónicos TRPV/agonistas , Tacto
20.
Sci Rep ; 5: 17637, 2015 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-26656101

RESUMEN

Injury to podocytes is considered a major contributor to diabetic kidney disease: their loss causes proteinuria and progressive glomerulosclerosis. Podocyte depletion may result from improper calcium handling due to abnormal activation of the calcium permeant TRPC (Transient Receptor Potential Canonical) channels. Angiotensin II (Ang II) levels are found to be elevated in diabetes; furthermore, it was reported that Ang II causes activation of TRPC6 in podocytes. We hypothesized here that Ang II-mediated calcium influx is aggravated in the podocytes under the conditions of type 1 diabetic nephropathy (DN). Diabetes was induced in the Dahl Salt-Sensitive rats by an injection of streptozotocin (STZ-SS). Eleven weeks post treatment was sufficient for the animals to develop hyperglycemia, excessive urination, weight loss, microalbuminuria, nephrinuria and display renal histological lesions typical for patients with DN. Patch-clamp electrophysiology performed on podocytes of the freshly isolated glomeruli showed enhanced basal TRPC channel activity in the STZ-SS rats, and increased response to Ang II; total calcium influx triggered by Ang II application was also augmented in podocytes of these rats. Our studies have a strong potential for advancing the understanding of TRPC-mediated effects on podocytopenia in DN initiation.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Podocitos/metabolismo , Podocitos/patología , Albuminuria/etiología , Angiotensina II/metabolismo , Angiotensina II/farmacología , Animales , Calcio/metabolismo , Nefropatías Diabéticas/genética , Modelos Animales de Enfermedad , Expresión Génica , Podocitos/efectos de los fármacos , Ratas , Ratas Endogámicas Dahl , Canales Catiónicos TRPC/agonistas , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA