Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-36378366

RESUMEN

Transient receptor potential vanilloid type 4 (TRPV4) channels are Ca2+-permeable non-selective cation channels which mediate a wide range of physiological functions and are activated and modulated by a diverse array of stimuli. One of this ion channel's least discussed functions is in relation to the generation and maintenance of certain pain sensations. However, in the two decades which have elapsed since the identification of this ion channel, considerable data has emerged concerning its function in mediating pain sensations. TRPV4 is a mediator of mechanical hyperalgesia in the various contexts in which a mechanical stimulus, comprising trauma (at the macro-level) or discrete extracellular pressure or stress (at the micro-level), results in pain. TRPV4 is also recognised as constituting an essential component in mediating inflammatory pain. It also plays a role in relation to many forms of neuropathic-type pain, where it functions in mediating mechanical allodynia and hyperalgesia.Here, we review the role of TRPV4 in mediating pain sensations.


Asunto(s)
Antineoplásicos , Neuralgia , Humanos , Canales Catiónicos TRPV/uso terapéutico , Hiperalgesia/tratamiento farmacológico
2.
Sci Rep ; 12(1): 19562, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36380063

RESUMEN

Mouse monoclonal antibody M4M was recently designed to block human TRPM4 channel. The polypeptide for generating M4M is composed of peptide A1 between the transmembrane segment 5 (S5) and the pore, and a second peptide A2 between the pore and the transmembrane segment 6 (S6). Using peptide microarray, a 4-amino acid sequence EPGF within the A2 was identified to be the binding epitope for M4M. Substitution of EPGF with other amino acids greatly reduced binding affinity. Structural analysis of human TRPM4 structure indicates that EPGF is located externally to the channel pore. A1 is close to the EPGF binding epitope in space, albeit separated by a 37-amino acid peptide. Electrophysiological study reveals that M4M could block human TRPM4, but with no effect on rodent TRPM4 which shares a different amino acid sequence ERGS for the binding motif. Our results demonstrate that M4M is a specific inhibitor for human TRPM4.


Asunto(s)
Anticuerpos Monoclonales , Canales Catiónicos TRPM , Ratones , Animales , Humanos , Epítopos , Anticuerpos Monoclonales/metabolismo , Secuencia de Aminoácidos , Péptidos/metabolismo , Mutación Missense , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPM/metabolismo
3.
Sci Rep ; 11(1): 10411, 2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-34002002

RESUMEN

TRPM4 is a calcium-activated non-selective monovalent cation channel implicated in diseases such as stroke. Lack of potent and selective inhibitors remains a major challenge for studying TRPM4. Using a polypeptide from rat TRPM4, we have generated a polyclonal antibody M4P which could alleviate reperfusion injury in a rat model of stroke. Here, we aim to develop a monoclonal antibody that could block human TRPM4 channel. Two mouse monoclonal antibodies M4M and M4M1 were developed to target an extracellular epitope of human TRPM4. Immunohistochemistry and western blot were used to characterize the binding of these antibodies to human TRPM4. Potency of inhibition was compared using electrophysiological methods. We further evaluated the therapeutic potential on a rat model of middle cerebral artery occlusion. Both M4M and M4M1 could bind to human TRPM4 channel on the surface of live cells. Prolonged incubation with TRPM4 blocking antibody internalized surface TRPM4. Comparing to M4M1, M4M is more effective in blocking human TRPM4 channel. In human brain microvascular endothelial cells, M4M successfully inhibited TRPM4 current and ameliorated hypoxia-induced cell swelling. Using wild type rats, neither antibody demonstrated therapeutic potential on stroke. Human TRPM4 channel can be blocked by a monoclonal antibody M4M targeting a key antigenic sequence. For future clinical translation, the antibody needs to be humanized and a transgenic animal carrying human TRPM4 sequence is required for in vivo characterizing its therapeutic potential.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Daño por Reperfusión/prevención & control , Canales Catiónicos TRPM/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/uso terapéutico , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Células HEK293 , Humanos , Infarto de la Arteria Cerebral Media/complicaciones , Masculino , Técnicas de Placa-Clamp , Ratas , Daño por Reperfusión/etiología , Daño por Reperfusión/patología , Canales Catiónicos TRPM/metabolismo
4.
Am J Respir Cell Mol Biol ; 59(1): 87-95, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29393654

RESUMEN

We previously described several ionic conductances in human pulmonary fibroblasts, including one activated by two structurally distinct TRPV4 (transient receptor potential, vanilloid-type, subtype 4)-channel agonists: 4αPDD (4α-phorbol-12,13-didecanoate) and GSK1016790A. However, the TRPV4-activated current exhibited peculiar properties: it developed slowly over many minutes, exhibited reversal potentials that could vary by tens of millivolts even within a given cell, and was not easily reversed by subsequent addition of two distinct TRPV4-selective blockers (RN-1734 and HC-067047). In this study, we characterized that conductance more carefully. We found that 4αPDD stimulated a delayed release of ATP into the extracellular space, which was reduced by genetic silencing of pannexin expression, and that the 4αPDD-evoked current could be blocked by apyrase (which rapidly degrades ATP) or by the P2Y purinergic receptor/channel blocker pyridoxalphosphate-6-azophenyl-2',4'-disulphonic acid (PPADS), and could be mimicked by exogenous addition of ATP. In addition, we found that the 4αPDD-evoked current was blocked by pretreatment with RN-1734 or HC-067047, by Gd3+ or La3+, or by two distinct blockers of pannexin channels (carbenoxolone and probenecid), but not by a blocker of connexin hemichannels (flufenamic acid). We also found expression of TRPV4- and pannexin-channel proteins. 4αPDD markedly increased calcium flashing in our cells. The latter was abrogated by the P2Y channel blocker PPADS, and the 4αPDD-evoked current was eliminated by loading the cytosol with 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid or by inhibiting Ca2+/calmodulin-sensitive kinase II using KN93. Altogether, we interpret these findings as suggesting that 4αPDD triggers the release of ATP via pannexin channels, which in turn acts in an autocrine and/or paracrine fashion to stimulate PPADS-sensitive purinergic receptors on human pulmonary fibroblasts.


Asunto(s)
Adenosina Trifosfato/metabolismo , Conexinas/metabolismo , Fibroblastos/metabolismo , Pulmón/citología , Proteínas del Tejido Nervioso/metabolismo , Canales Catiónicos TRPV/metabolismo , Anciano , Anciano de 80 o más Años , Calcio/metabolismo , Femenino , Humanos , Espacio Intracelular/metabolismo , Activación del Canal Iónico/efectos de los fármacos , Masculino , Persona de Mediana Edad , Modelos Biológicos , Ésteres del Forbol/farmacología , Fosfato de Piridoxal/análogos & derivados , Fosfato de Piridoxal/farmacología , Factores de Tiempo
5.
Semin Cancer Biol ; 47: 154-167, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28627410

RESUMEN

Mitochondria are essential intracellular organelles that regulate energy metabolism, cell death, and signaling pathways that are important for cell proliferation and differentiation. Therefore, mitochondria are fundamentally implicated in cancer biology, including initiation, growth, metastasis, relapse, and acquired drug resistance. Based on these implications, mitochondria have been proposed as a major therapeutic target for cancer treatment. In addition to classical view of mitochondria in cancer biology, recent studies found novel pathophysiological roles of mitochondria in cancer. In this review, we introduce recent concepts of mitochondrial roles in cancer biology including mitochondrial DNA mutation and epigenetic modulation, energy metabolism reprogramming, mitochondrial channels, involvement in metastasis and drug resistance, and cancer stem cells. We also discuss the role of mitochondria in emerging cancer therapeutic strategies, especially cancer immunotherapy and CRISPR-Cas9 system gene therapy.


Asunto(s)
Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Animales , Sistemas CRISPR-Cas , ADN Mitocondrial , Resistencia a Antineoplásicos/genética , Metabolismo Energético/efectos de los fármacos , Humanos , Inmunoterapia , Mitocondrias/genética , Mutación , Metástasis de la Neoplasia , Neoplasias/etiología , Neoplasias/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo
6.
J Physiol ; 594(24): 7327-7340, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27779758

RESUMEN

KEY POINTS: Increase in blood pressure in the renal afferent arteriole is known to induce an increase in cytosolic calcium concentration ([Ca2+ ]i ) of juxtaglomerular (JG) cells and to result in a decreased secretion of renin. Mechanical stimulation of As4.1 JG cells induces an increase in [Ca2+ ]i that is inhibited by HC067047 and RN1734, two inhibitors of TRPV4, or by siRNA-mediated repression of TRPV4. Inhibition of TRPV4 impairs pressure-induced decrease in renin secretion. Compared to wild-type mice, Trpv4-/- mice present increased resting plasma levels of renin and aldosterone and present a significantly altered pressure-renin relationship. We suggest that TRPV4 channel participates in mechanosensation at the juxtaglomerular apparatus. ABSTRACT: The renin-angiotensin system is a crucial blood pressure regulation system. It consists of a hormonal cascade where the rate-limiting enzyme is renin, which is secreted into the blood flow by renal juxtaglomerular (JG) cells in response to low pressure in the renal afferent arteriole. In contrast, an increase in blood pressure results in a decreased renin secretion. This is accompanied by a transitory increase in [Ca2+ ]i of JG cells. The inverse relationship between [Ca2+ ]i and renin secretion has been called the 'calcium paradox' of renin release. How increased pressure induces a [Ca2+ ]i transient in JG cells, is however, unknown. We observed that [Ca2+ ]i transients induced by mechanical stimuli in JG As4.1 cells were completely abolished by HC067047 and RN1734, two inhibitors of TRPV4. They were also reduced by half by siRNA-mediated repression of TRPV4 but not after repression or inhibition of TRPV2 or Piezo1 ion channels. Interestingly, the stimulation of renin secretion by the adenylate cyclase activator forskolin was totally inhibited by cyclic stretching of the cells. This effect was mimicked by stimulation with GSK1016790A and 4αPDD, two activators of TRPV4 and inhibited in the presence of HC067047. Moreover, in isolated perfused kidneys from Trpv4-/- mice, the pressure-renin relationship was significantly altered. In vivo, Trpv4-/- mice presented increased plasma levels of renin and aldosterone compared to wild-type mice. Altogether, our results suggest that TRPV4 is involved in the pressure-induced entry of Ca2+ in JG cells, which inhibits renin release and allows the negative feedback regulation on blood pressure.


Asunto(s)
Aparato Yuxtaglomerular/metabolismo , Mecanotransducción Celular/fisiología , Renina/antagonistas & inhibidores , Canales Catiónicos TRPV/fisiología , Aldosterona/sangre , Animales , Calcio/fisiología , Línea Celular Tumoral , Masculino , Ratones Noqueados , Presión , Renina/sangre , Renina/metabolismo , Canales Catiónicos TRPV/genética
7.
Pflugers Arch ; 468(8): 1299-309, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27343012

RESUMEN

Cereblon (CRBN) is a substrate receptor of the E3 ubiquitin ligase complex that has been linked to autosomal recessive non-syndromic mental retardation. Several key findings suggest diverse roles of CRBN, including its regulation of the large-conductance calcium- and voltage-activated potassium (BKCa) channels, regulation of thalidomide-binding proteins, and mediation of lenalidomide treatment in multiple myeloma. Recent studies also indicate that CRBN is involved in energy metabolism and negatively regulates AMP-activated protein kinase signaling. Mice with genetic depletion of CRBN are resistant to various stress conditions including a high-fat diet, endoplasmic reticulum stress, ischemia/reperfusion injury, and alcohol-related liver damage. In this review, we discuss the various roles of CRBN in human health and disease and suggest avenues for further research to enhance our basic knowledge and clinical application of CRBN.


Asunto(s)
Péptido Hidrolasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Proteínas Portadoras/metabolismo , Humanos , Unión Proteica/fisiología , Transducción de Señal/fisiología
8.
Pflugers Arch ; 468(3): 371-83, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26739710

RESUMEN

The volume-regulated anion channel (VRAC), also known as the volume-sensitive outwardly rectifying (VSOR) anion channel or the volume-sensitive organic osmolyte/anion channel (VSOAC), is essential for cell volume regulation after swelling in most vertebrate cell types studied to date. In addition to its role in cell volume homeostasis, VRAC has been implicated in numerous other physiological and pathophysiological processes, including cancer, ischemic brain edema, cell motility, proliferation, angiogenesis, programmed cell death, and excitotoxic glutamate release. Although VRAC has been extensively biophysically, pharmacologically, and functionally characterized, its molecular identity was highly controversial until the recent identification of the leucine-rich repeats containing 8A (LRRC8A) protein as essential for the VRAC current in multiple cell types and a likely pore-forming subunit of VRAC. Members of this distantly pannexin-1-related protein family form heteromers, and in addition to LRRC8A, at least another LRRC8 family member is required for the formation of a functional VRAC. This review summarizes the biophysical and pharmacological properties of VRAC, highlights its main physiological functions and pathophysiological implications, and outlines the search for its molecular identity.


Asunto(s)
Aniones/metabolismo , Tamaño de la Célula , Canales Iónicos/metabolismo , Potenciales de Acción , Animales , Apoptosis , Humanos , Transporte Iónico , Neuronas/citología , Neuronas/metabolismo , Neuronas/fisiología
10.
Pflugers Arch ; 466(4): 623-6, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24563221

RESUMEN

T-type Ca(2+) channels have gained, 15 years after cloning, an immense interest as novel players in very unexpected cell functions, and its many relations to diseases have been discovered. This special issue provides a state-of-the-art overview on novel functional properties of T-type Ca(2+) channels, unexpected cellular functions, and most importantly will also summarizes and review the involvement of this "tiny, transient" type of Ca(2+) channels in several diseases. It is tried to bridge the gap between molecular biophysical properties of T-type Ca(2+) channels and diseases providing finally a translational view on this amazing ion channel.


Asunto(s)
Canales de Calcio Tipo T/fisiología , Investigación Biomédica Traslacional/tendencias , Animales , Epilepsia/genética , Epilepsia/metabolismo , Humanos , Activación del Canal Iónico/fisiología , Neoplasias/genética , Neoplasias/metabolismo , Dolor/genética , Dolor/metabolismo
11.
Chem Senses ; 39(4): 295-311, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24452633

RESUMEN

Transient receptor potential cation channel subfamily M member 5 (TRPM5) is a Ca(2+)-activated nonselective cation channel involved in the transduction of sweet, bitter, and umami tastes. We previously showed that TRPM5 is a locus for the modulation of taste perception by temperature changes, and by quinine and quinidine, 2 bitter compounds that suppress gustatory responses. Here, we determined whether other bitter compounds known to modulate taste perception also affect TRPM5. We found that nicotine inhibits TRPM5 currents with an effective inhibitory concentration of ~1.3mM at -50 mV. This effect may contribute to the inhibitory effect of nicotine on gustatory responses in therapeutic and experimental settings, where nicotine is often employed at millimolar concentrations. In addition, it implies the existence of a TRPM5-independent pathway for the detection of nicotine bitterness. Nicotine seems to act from the extracellular side of the channel, reducing the maximal whole-cell conductance and inducing an acceleration of channel closure that leads to a negative shift of the activation curve. TRPM5 currents were unaffected by nicotine's metabolite cotinine, the intensive sweetener saccharin or by the bitter xanthines caffeine, theobromine, and theophylline. We also tested the effects of bitter compounds on another essential element of the sweet taste transduction pathway, the type 3 IP3 receptor (IP3R3). We found that IP3R3-mediated Ca(2+) flux is slightly enhanced by nicotine, not affected by saccharin, modestly inhibited by caffeine, theobromine, and theophylline, and strongly inhibited by quinine. Our results demonstrate that bitter compounds have differential effects on key elements of the sweet taste transduction pathway, suggesting for heterogeneous mechanisms of bitter-sweet taste interactions.


Asunto(s)
Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Nicotina/farmacología , Transducción de Señal/efectos de los fármacos , Canales Catiónicos TRPM/metabolismo , Animales , Calcio/metabolismo , Células HEK293 , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , Ratones , Técnicas de Placa-Clamp , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Quinidina/farmacología , Quinina/farmacología , Edulcorantes/farmacología , Canales Catiónicos TRPM/antagonistas & inhibidores
12.
Neurourol Urodyn ; 33(4): 461-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23616342

RESUMEN

AIMS: The KIT receptor is considered as a reliable marker for a subpopulation of interstitial cells (IC), and by persistent neonatal inhibition of KIT we have investigated the role of this receptor in the development of IC-networks in bladder and we have observed the functional consequences of this inhibition. METHODS: Newborn rat pups were treated daily with the KIT inhibitor imatinib mesylate (IM). After 7 days animals were sacrificed and bladder samples were dissected for morphological and functional studies. Morphological research consisted of immunohistochemistry with IC specific antigens (KIT and vimentin) and electron microscopy. The functional studies were based on isolated bladder strips in organ baths, in which spontaneous bladder contractility and the response to a non-subtype selective muscarinic agonist was evaluated. RESULTS: Suburothelial and intramuscular IC were found and characterized in neonatal rat bladder. IM-treatment induced a significant decrease in numbers of IC based on specific immunohistochemical markers, and electron microscopy revealed evidence of IC cell injury. These morphological alterations were observed on intramuscular IC only and not on IC in the suburothelium. Isolated muscle strips from IM-treated animals had a lower contractile frequency and an altered response to muscarinic agonists. CONCLUSIONS: The present study shows the presence of regional subpopulations of IC in neonatal rat bladder, provides evidence for a dependence on KIT of the development of intramuscular IC and supports the hypothesis that a poor development of networks of intramuscular IC might have repercussions on spontaneous and muscarinic-induced bladder contractility.


Asunto(s)
Antineoplásicos/toxicidad , Benzamidas/toxicidad , Células Intersticiales de Cajal/efectos de los fármacos , Músculo Liso/citología , Músculo Liso/efectos de los fármacos , Piperazinas/toxicidad , Pirimidinas/toxicidad , Vejiga Urinaria/citología , Vejiga Urinaria/efectos de los fármacos , Animales , Animales Recién Nacidos , Femenino , Crecimiento/efectos de los fármacos , Mesilato de Imatinib , Inmunohistoquímica , Técnicas In Vitro , Células Intersticiales de Cajal/ultraestructura , Contracción Muscular/efectos de los fármacos , Músculo Liso/ultraestructura , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , Ratas , Ratas Wistar
13.
Cell Calcium ; 55(1): 38-47, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24342753

RESUMEN

The importance of the TRPV4 channel for human physiology has been highlighted in recent years with the identification of an increasing number of hereditary diseases associated with mutations of this channel. However, the functional understanding of TRPV4 associated pathologies remains a puzzle due to incomplete understanding of the polymodal regulation of TRPV4 channels and lack of insight into the structure-function relationship of the channel. In this work, we identified a series of highly conserved aromatic residues in transmembrane (TM) helices 5-6 with profound importance for TRPV4 activity. Substituting F617, Y621 or F624 in TM5 with leucine reduced channel sensitivity to the agonist 4α-PDD and heat, yet two of these mutants - F617L and Y621L - showed increased activation in response to cell swelling. In TM6, a Y702L mutation significantly reduced sensitivity to all of the above stimuli. In conclusion, we have identified residues in TM5-6 which differentially affect heat and agonist activation, and we have demonstrated distinct activation pathways for 4α-PDD and osmolarity.


Asunto(s)
Soluciones Hipotónicas/farmacología , Ésteres del Forbol/farmacología , Mutación Puntual/genética , Porinas/fisiología , Canales Catiónicos TRPV/efectos de los fármacos , Canales Catiónicos TRPV/genética , Secuencia de Aminoácidos , Calcio/fisiología , Carcinógenos/farmacología , Fenómenos Electrofisiológicos , Células HEK293 , Humanos , Datos de Secuencia Molecular , Concentración Osmolar , Estructura Secundaria de Proteína/efectos de los fármacos , Estructura Secundaria de Proteína/genética , Canales Catiónicos TRPV/química
14.
PLoS One ; 8(8): e73424, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23977387

RESUMEN

OBJECTIVE: Regulation of apical calcium entry is important for the function of principal cells of the collecting duct. However, the molecular identity and the regulators of the transporter/channel, which is responsible for apical calcium entry and what factors regulate the calcium conduction remain unclear. METHODS AND RESULTS: We report that endogenous TRPP2 and TRPV4 assemble to form a 23-pS divalent cation-permeable non-selective ion channel at the apical membrane of renal principal cells of the collecting duct. TRPP2\TRPV4 channel complex was identified by patch-clamp, immunofluorescence and co-immunprecipitation studies in both principal cells that either possess normal cilia (cilia (+)) or in which cilia are absent (cilia (-)). This channel has distinct biophysical and pharmacological and regulatory profiles compared to either TRPP2 or TRPV4 channels. The rate of occurrence detected by patch clamp was higher in cilia (-) compared to cilia (+) cells. In addition, shRNA knockdown of TRPP2 increased the prevalence of TRPV4 channel activity while knockdown of TRPV4 resulted in TRPP2 activity and knockdown of both proteins vastly decreased the 23-pS channel activity. Epidermal growth factor (EGF) stimulated TRPP2\TRPV4 channel through the EGF receptor (EGFR) tyrosine kinase-dependent signaling. With loss of cilia, apical EGF treatment resulted in 64-fold increase in channel activity in cilia (-) but not cilia (+) cells. In addition EGF increased cell proliferation in cilia (-) cell that was dependent upon TRPP2\TRPV4 channel mediated increase in intracellular calcium. CONCLUSION: We conclude that in the absence of cilia, an EGF activated TRPP2\TRPV4 channel may play an important role in increased cell proliferation and cystogenesis.


Asunto(s)
Agonistas de los Canales de Calcio/farmacología , Calcio/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Túbulos Renales Colectores/metabolismo , Canales Catiónicos TRPP/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Cationes Bivalentes/metabolismo , Proliferación Celular/efectos de los fármacos , Cilios/metabolismo , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Técnica del Anticuerpo Fluorescente , Silenciador del Gen/efectos de los fármacos , Inmunoprecipitación , Activación del Canal Iónico/efectos de los fármacos , Túbulos Renales Colectores/citología , Túbulos Renales Colectores/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Fosforilación/efectos de los fármacos
15.
Mol Pharmacol ; 84(3): 325-34, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23757176

RESUMEN

Allyl isothiocyanate (AITC; aka, mustard oil) is a powerful irritant produced by Brassica plants as a defensive trait against herbivores and confers pungency to mustard and wasabi. AITC is widely used experimentally as an inducer of acute pain and neurogenic inflammation, which are largely mediated by the activation of nociceptive cation channels transient receptor potential ankyrin 1 and transient receptor potential vanilloid 1 (TRPV1). Although it is generally accepted that electrophilic agents activate these channels through covalent modification of cytosolic cysteine residues, the mechanism underlying TRPV1 activation by AITC remains unknown. Here we show that, surprisingly, AITC-induced activation of TRPV1 does not require interaction with cysteine residues, but is largely dependent on S513, a residue that is involved in capsaicin binding. Furthermore, AITC acts in a membrane-delimited manner and induces a shift of the voltage dependence of activation toward negative voltages, which is reminiscent of capsaicin effects. These data indicate that AITC acts through reversible interactions with the capsaicin binding site. In addition, we show that TRPV1 is a locus for cross-sensitization between AITC and acidosis in nociceptive neurons. Furthermore, we show that residue F660, which is known to determine the stimulation by low pH in human TRPV1, is also essential for the cross-sensitization of the effects of AITC and low pH. Taken together, these findings demonstrate that not all reactive electrophiles stimulate TRPV1 via cysteine modification and help understanding the molecular bases underlying the surprisingly large role of this channel as mediator of the algesic properties of AITC.


Asunto(s)
Isotiocianatos/farmacología , Canales Catiónicos TRPV/metabolismo , Animales , Sitios de Unión , Capsaicina/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Cultivadas , Ganglios Espinales/citología , Humanos , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos C57BL , Mutación , Técnicas de Placa-Clamp , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/genética
16.
Pflugers Arch ; 465(9): 1303-16, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23605066

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is characterized by hepatic lipid deposition and coincides often with cardiometabolic diseases. Several dietary factors attenuate NAFLD. Here, we report beneficial effects of chronic dietary capsaicin intake on NAFLD which is mediated by the transient receptor potential vanilloid 1 (TRPV1) activation. The results showed that TRPV1 activation by capsaicin reduced free fatty acids (FFAs) induced the intracellular lipid droplets in HepG2 cells and prevented fatty liver in vivo. Chronic dietary capsaicin promoted lipolysis by increasing hepatic phosphorylated hormone-sensitive lipase (phospho-HSL), carnitine palmitoyltransferase 1 (CPT1), and peroxisome proliferator-activated receptor δ (PPARδ) in wild-type (WT) mice. This effect was absent in TRPV1(-/-) mice. Dietary capsaicin did not affect lipogenesis, as indicated by the detection of hepatic fatty acid synthase (FAS), sterol regulatory element-binding protein-1 (SREBP-1), PPARα, and liver X receptor (LXR) in mice. Importantly, TRPV1 causes PPARδ activation which significantly increased the expression of autophagy-related proteins, such as light chain 3 (LC3)II, Beclin1, Atg5, and Atg7 in HepG2 cells. In the in vivo study, TRPV1 activation by dietary capsaicin enhanced hepatic PPARδ and autophagy-related proteins and reduced hepatic enzymes and inflammatory factor in WT but not TRPV1(-/-) mice. TRPV1 activation by dietary capsaicin prevents NAFLD through PPARδ-dependent autophagy enhancement in mice. Dietary capsaicin may represent a beneficial intervention in populations at high risk for NAFLD.


Asunto(s)
Capsaicina/farmacología , Hígado Graso/metabolismo , PPAR delta/metabolismo , Canales Catiónicos TRPV/genética , Administración Oral , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia , Proteína 7 Relacionada con la Autofagia , Beclina-1 , Capsaicina/administración & dosificación , Capsaicina/uso terapéutico , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Ácido Graso Sintasas/genética , Ácido Graso Sintasas/metabolismo , Ácidos Grasos/metabolismo , Hígado Graso/dietoterapia , Hígado Graso/prevención & control , Células Hep G2 , Humanos , Lipólisis/efectos de los fármacos , Receptores X del Hígado , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Enfermedad del Hígado Graso no Alcohólico , Receptores Nucleares Huérfanos/genética , Receptores Nucleares Huérfanos/metabolismo , PPAR delta/genética , Esterol Esterasa/genética , Esterol Esterasa/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Canales Catiónicos TRPV/metabolismo , Enzimas Activadoras de Ubiquitina/genética , Enzimas Activadoras de Ubiquitina/metabolismo
17.
Artículo en Inglés | MEDLINE | ID: mdl-23605179

RESUMEN

Spicy food does not only provide an important hedonic input in daily life, but has also been anedoctically associated to beneficial effects on our health. In this context, the discovery of chemesthetic trigeminal receptors and their spicy ligands has provided the mechanistic basis and the pharmacological means to investigate this enticing possibility. This review discusses in molecular terms the connection between the neurophysiology of pungent spices and the "systemic" effects associated to their trigeminality. It commences with a cultural and historical overview on the Western fascination for spices, and, after analysing in detail the mechanisms underlying the trigeminality of food, the main dietary players from the transient receptor potential (TRP) family of cation channels are introduced, also discussing the "alien" distribution of taste receptors outside the oro-pharingeal cavity. The modulation of TRPV1 and TRPA1 by spices is next described, discussing how spicy sensations can be turned into hedonic pungency, and analyzing the mechanistic bases for the health benefits that have been associated to the consumption of spices. These include, in addition to a beneficial modulation of gastro-intestinal and cardio-vascular function, slimming, the optimization of skeletal muscle performance, the reduction of chronic inflammation, and the prevention of metabolic syndrome and diabetes. We conclude by reviewing the role of electrophilic spice constituents on cancer prevention in the light of their action on pro-inflammatory and pro-cancerogenic nuclear factors like NFκB, and on their interaction with the electrophile sensor protein Keap1 and the ensuing Nrf2-mediated transcriptional activity. Spicy compounds have a complex polypharmacology, and just like any other bioactive agent, show a balance of beneficial and bad actions. However, at least for moderate consumption, the balance seems definitely in favour of the positive side, suggesting that a spicy diet, a caveman-era technology, could be seriously considered in addition to caloric control and exercise as a measurement to prevent and control many chronic diseases associate to malnutrition from a Western diet.


Asunto(s)
Especias , Animales , Humanos , Neoplasias/prevención & control , Dolor/tratamiento farmacológico , Plantas , Células Receptoras Sensoriales/fisiología , Canales Catiónicos TRPM/fisiología , Canales Catiónicos TRPV/fisiología , Gusto
18.
EMBO Rep ; 14(2): 152-63, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23306656

RESUMEN

Hereditary channelopathies, that is, mutations in channel genes that alter channel function and are causal for the pathogenesis of the disease, have been described for several members of the transient receptor potential channel family. Mutations in the TRPV4 gene, encoding a polymodal Ca(2+) permeable channel, are causative for several human diseases, which affect the skeletal system and the peripheral nervous system, with highly variable phenotypes. In this review, we describe the phenotypes of TRPV4 channelopathies and overlapping symptoms. Putative mechanisms to explain the puzzle, and how mutations in the same region of the channel cause different diseases, are discussed and experimental approaches to tackle this surprising problem are suggested.


Asunto(s)
Canalopatías/genética , Canales Catiónicos TRPV/genética , Animales , Enfermedades del Desarrollo Óseo/genética , Enfermedades del Desarrollo Óseo/metabolismo , Canalopatías/metabolismo , Neuropatía Hereditaria Motora y Sensorial/genética , Neuropatía Hereditaria Motora y Sensorial/metabolismo , Humanos , Enfermedades Musculoesqueléticas/genética , Enfermedades Musculoesqueléticas/metabolismo , Mutación Missense , Fenotipo
19.
Pflugers Arch ; 464(5): 425-58, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23001121

RESUMEN

The Transient Receptor Potential Ankyrin 1 channel (TRPA1), is a member of the large TRP family of ion channels, and functions as a Ca(2+) permeable non-selective cation channel in many different cell processes, ranging from sensory to homeostatic tasks. TRPA1 is highly conserved across the animal kingdom. The only mammalian TRPA subfamily member, TRPA1, is widely expressed in neuronal (e.g. sensory dorsal root and trigeminal ganglia neurons)- and in non-neuronal cells (e.g. epithelial cells, hair cells). It exhibits 14-19 amino-(N-)terminal ankyrin repeats, an unusual structural feature. The TRPA1 channel is activated by noxious cold (<17 °C) as well as by a plethora of chemical compounds that includes not only electrophilic compounds and oxidants that can modify, in an alkylative or oxidative fashion, nucleophilic cysteine residues in the channel's N-terminus, but also compounds that do not covalently bind to the channel proteins (e.g. menthol, nifedipin). Based on localization and functional properties, TRPA1 is considered a key player in acute and chronic (neuropathic) pain and inflammation. Moreover, its role in the (patho)physiology of nearly all organ systems is anticipated, and will be discussed along with the potential of TRPA1 as a drug target for the management of various pathological conditions.


Asunto(s)
Canales de Potencial de Receptor Transitorio/genética , Canales de Potencial de Receptor Transitorio/fisiología , Animales , Agonistas de los Canales de Calcio/metabolismo , Canalopatías/genética , Humanos , Inflamación/metabolismo , Activación del Canal Iónico , Nocicepción , Dolor/metabolismo , Filogenia , Estructura Terciaria de Proteína , Canal Catiónico TRPA1 , Canales de Potencial de Receptor Transitorio/química , Canales de Potencial de Receptor Transitorio/metabolismo
20.
J Vis Exp ; (66): e3869, 2012 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-22929055

RESUMEN

The lower urinary tract (LUT) functions as a dynamic reservoir that is able to store urine and to efficiently expel it at a convenient time. While storing urine, however, the bladder is exposed for prolonged periods to waste products. By acting as a tight barrier, the epithelial lining of the LUT, the urothelium, avoids re-absorption of harmful substances. Moreover, noxious chemicals stimulate the bladder's nociceptive innervation and initiate voiding contractions that expel the bladder's contents. Interestingly, the bladder's sensitivity to noxious chemicals has been used successfully in clinical practice, by intravesically infusing the TRPV1 agonist capsaicin to treat neurogenic bladder overactivity. This underscores the advantage of viewing the bladder as a chemosensory organ and prompts for further clinical research. However, ethical issues severely limit the possibilities to perform, in human subjects, the invasive measurements that are necessary to unravel the molecular bases of LUT clinical pharmacology. A way to overcome this limitation is the use of several animal models. Here we describe the implementation of cystometry in mice and rats, a technique that allows measuring the intravesical pressure in conditions of controlled bladder perfusion. After laparotomy, a catheter is implanted in the bladder dome and tunneled subcutaneously to the interscapular region. Then the bladder can be filled at a controlled rate, while the urethra is left free for micturition. During the repetitive cycles of filling and voiding, intravesical pressure can be measured via the implanted catheter. As such, the pressure changes can be quantified and analyzed. Moreover, simultaneous measurement of the voided volume allows distinguishing voiding contractions from non-voiding contractions. Importantly, due to the differences in micturition control between rodents and humans, cystometric measurements in these animals have only limited translational value. Nevertheless, they are quite instrumental in the study of bladder pathophysiology and pharmacology in experimental pre-clinical settings. Recent research using this technique has revealed the key role of novel molecular players in the mechano- and chemo-sensory properties of the bladder.


Asunto(s)
Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/fisiología , Animales , Femenino , Ratones , Ratas , Estimulación Química , Cateterismo Urinario/métodos , Urodinámica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA