Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 157(3): 664-75, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24746027

RESUMEN

Sensory systems for detecting tactile stimuli have evolved from touch-sensing nerves in invertebrates to complicated tactile end organs in mammals. Merkel discs are tactile end organs consisting of Merkel cells and Aß-afferent nerve endings and are localized in fingertips, whisker hair follicles, and other touch-sensitive spots. Merkel discs transduce touch into slowly adapting impulses to enable tactile discrimination, but their transduction and encoding mechanisms remain unknown. Using rat whisker hair follicles, we show that Merkel cells rather than Aß-afferent nerve endings are primary sites of tactile transduction and identify the Piezo2 ion channel as the Merkel cell mechanical transducer. Piezo2 transduces tactile stimuli into Ca(2+)-action potentials in Merkel cells, which drive Aß-afferent nerve endings to fire slowly adapting impulses. We further demonstrate that Piezo2 and Ca(2+)-action potentials in Merkel cells are required for behavioral tactile responses. Our findings provide insights into how tactile end-organs function and have clinical implications for tactile dysfunctions.


Asunto(s)
Canales Iónicos/metabolismo , Células de Merkel/metabolismo , Tacto , Vibrisas/citología , Vibrisas/fisiología , Potenciales de Acción , Animales , Calcio/metabolismo , Técnicas de Silenciamiento del Gen , Canales Iónicos/genética , Mecanorreceptores/metabolismo , Mecanotransducción Celular , Ratas
2.
Pharmacol Res ; 164: 105391, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33352230

RESUMEN

Baroreflex plays a crucial role in regulation of arterial blood pressure (BP). Recently, Piezo1 and Piezo2, the mechanically-activated (MA) ion channels, have been identified as baroreceptors. However, the underlying molecular mechanism for regulating these baroreceptors in hypertension remains unknown. In this study, we used spontaneously hypertensive rats (SHR) and NG-Nitro-l-Arginine (L-NNA)- and Angiotensin II (Ang II)-induced hypertensive model rats to determine the role and mechanism of Piezo1 and Piezo2 in hypertension. We found that Piezo2 was dominantly expressed in baroreceptor nodose ganglia (NG) neurons and aortic nerve endings in Wistar-Kyoto (WKY) rats. The expression of Piezo2 not Piezo1 was significantly downregulated in these regions in SHR and hypertensive model rats. Electrophysiological results showed that the rapidly adapting mechanically-activated (RA-MA) currents and the responsive neuron numbers were significantly reduced in baroreceptor NG neurons in SHR. In WKY rats, the arterial BP was elevated by knocking down the expression of Piezo2 or inhibiting MA channel activity by GsMTx4 in NG. Knockdown of Piezo2 in NG also attenuated the baroreflex and increased serum norepinephrine (NE) concentration in WKY rats. Co-immunoprecipitation experiment suggested that Piezo2 interacted with Neural precursor cell-expressed developmentally downregulated gene 4 type 2 (Nedd4-2, also known as Nedd4L); Electrophysiological results showed that Nedd4-2 inhibited Piezo2 MA currents in co-expressed HEK293T cells. Additionally, Nedd4-2 was upregulated in NG baroreceptor neurons in SHR. Collectively, our results demonstrate that Piezo2 not Piezo1 may act as baroreceptor to regulate arterial BP in rats. Nedd4-2 induced downregulation of Piezo2 in baroreceptor NG neurons leads to hypertension in rats. Our findings provide a novel insight into the molecular mechanism for the regulation of baroreceptor Piezo2 and its critical role in the pathogenesis of hypertension.


Asunto(s)
Hipertensión/fisiopatología , Canales Iónicos/fisiología , Ubiquitina-Proteína Ligasas Nedd4/fisiología , Neuronas/fisiología , Ganglio Nudoso/fisiología , Presorreceptores/fisiología , Animales , Aorta Torácica/inervación , Barorreflejo , Células Cultivadas , Humanos , Masculino , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Transducción de Señal
3.
Pharmacol Res ; 161: 105128, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32805354

RESUMEN

Cannabidiol (CBD) is a major phytocannabinoid in Cannabis sativa. CBD is being increasingly reported as a clinical treatment for neurological diseases. Febrile seizure is one of the most common diseases in children with limited therapeutic options. We investigated possible therapeutic effects of CBD on febrile seizures and the underlying mechanism. Use of a hyperthermia-induced seizures model revealed that CBD significantly prolonged seizure latency and reduced the severity of thermally-induced seizures. Hippocampal neuronal excitability was significantly decreased by CBD. Further, CBD significantly reduced the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) mediated evoked excitatory postsynaptic currents (eEPSCs) and the amplitude and frequency of miniature EPSCs (mEPSCs). Furthermore, CBD significantly accelerated deactivation in GluA1 and GluA2 subunits. Interestingly, CBD slowed receptor recovery from desensitization of GluA1, but not GluA2. These effects on kinetics were even more prominent when AMPAR was co-expressed with γ-8, the high expression isoform 8 of transmembrane AMPAR regulated protein (TARPγ8) in the hippocampus. The inhibitory effects of CBD on AMPAR depended on its interaction with the distal N-terminal domain of GluA1/GluA2. CBD inhibited AMPAR activity and reduced hippocampal neuronal excitability, thereby improving the symptoms of febrile seizure in mice. The putative binding site of CBD in the N-terminal domain of GluA1/GluA2 may be a drug target for allosteric gating modulation of AMPAR.


Asunto(s)
Anticonvulsivantes/farmacología , Ondas Encefálicas/efectos de los fármacos , Región CA1 Hipocampal/efectos de los fármacos , Cannabidiol/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Hipertermia/complicaciones , Receptores AMPA/antagonistas & inhibidores , Convulsiones Febriles/prevención & control , Animales , Anticonvulsivantes/metabolismo , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/fisiopatología , Cannabidiol/metabolismo , Modelos Animales de Enfermedad , Antagonistas de Aminoácidos Excitadores/metabolismo , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Células HEK293 , Humanos , Activación del Canal Iónico/efectos de los fármacos , Cinética , Ratones , Ratones Endogámicos C57BL , Potenciales Postsinápticos Miniatura/efectos de los fármacos , Modelos Moleculares , Unión Proteica , Tiempo de Reacción/efectos de los fármacos , Receptores AMPA/genética , Receptores AMPA/metabolismo , Convulsiones Febriles/etiología , Convulsiones Febriles/metabolismo , Convulsiones Febriles/fisiopatología
4.
Exp Cell Res ; 371(1): 20-30, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29842877

RESUMEN

Although resident cardiac stem cells have been reported, regeneration of functional cardiomyocytes (CMs) remains a challenge. The present study identifies an alternative progenitor source for CM regeneration without the need for genetic manipulation or invasive heart biopsy procedures. Unlike limb skeletal muscles, masseter muscles (MM) in the mouse head are developed from Nkx2-5 mesodermal progenitors. Adult masseter muscle satellite cells (MMSCs) display heterogeneity in developmental origin and cell phenotypes. The heterogeneous MMSCs that can be characterized by cell sorting based on stem cell antigen-1 (Sca1) show different lineage potential. While cardiogenic potential is preserved in Sca1+ MMSCs as shown by expression of cardiac progenitor genes (including Nkx2-5), skeletal myogenic capacity is maintained in Sca1- MMSCs with Pax7 expression. Sca1+ MMSC-derived beating cells express cardiac genes and exhibit CM-like morphology. Electrophysiological properties of MMSC-derived CMs are demonstrated by calcium transients and action potentials. These findings show that MMSCs could serve as a novel cell source for cardiomyocyte replacement.


Asunto(s)
Diferenciación Celular , Músculo Masetero/citología , Desarrollo de Músculos/genética , Miocitos Cardíacos/citología , Células Satélite del Músculo Esquelético/citología , Potenciales de Acción/fisiología , Animales , Ataxina-1/genética , Ataxina-1/metabolismo , Biomarcadores/metabolismo , Calcio/metabolismo , Linaje de la Célula/genética , Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteína Homeótica Nkx-2.5/genética , Proteína Homeótica Nkx-2.5/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Músculo Masetero/metabolismo , Ratones , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Factor de Transcripción PAX7/genética , Factor de Transcripción PAX7/metabolismo , Fenotipo , Regeneración , Células Satélite del Músculo Esquelético/metabolismo , Proteína Fluorescente Roja
5.
Cell Physiol Biochem ; 50(5): 1891-1902, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30396162

RESUMEN

BACKGROUND/AIMS: Myocardial ischemia/reperfusion (I/R) or hypoxia/reoxygenation (H/R) injury is always characterized by Ca2+ overload, energy metabolism disorder and necrocytosis of cardiomyocytes. We showed previously that chronic intermittent hypobaric hypoxia (CIHH) improves cardiac function during I/R through improving cardiac glucose metabolism. However, the underlying cellular and molecular mechanisms of CIHH treatment improving energy metabolism in cardiomyocytes are still unclear. In this study, we determined whether and how CIHH protects cardiomyocytes from Ca2+ overload and necrocytosis through energy regulating pathway. METHODS: Adult male Sprague-Dawley rats were randomly divided into two groups: control (CON) and CIHH group. CIHH rats received a hypobaric hypoxia simulating 5,000-m altitude for 28 days, 6 hours each day, in hypobaric chamber. Rat ventricular myocytes were obtained by enzymatic dissociation. The intracellular calcium concentration ([Ca2+]i) and cTnI protein expression were used to evaluate the degree of cardiomyocytes injury during and after H/R. The mRNA and protein expressions involved in cardiac energy metabolism were determined using quantitative PCR and Western blot techniques. PGC-1α siRNA adenovirus transfection was used to knock down PGC-1α gene expression of cardiomyocytes to determine the effect of PGC-1α in the energy regulating pathway. RESULTS: H/R increased [Ca2+]i and cTnI protein expression in cardiomyocytes. CIHH treatment decreased [Ca2+]i (p< 0.01) and cTnI protein expression (p< 0.01) in cardiomyocytes after H/R. Both mRNA and protein expression of PGC-1α increased after CIHH treatment, which was reversed by PGC-1α siRNA adenovirus transfection. Furthermore, CIHH treatment increased the expression of HIF-1α, AMPK and p-AMPK in cardiomyocytes, and pretreatment with AMPK inhibitor dorsomorphin abolished the enhancement of PGC-1α protein expression in cardiomyocytes by CIHH (p< 0.01). In addition, PGC-1α knock down also abolished the increased protein level of GLUT4 (p< 0.01) and decreased the protein level of CPT-1b (p< 0.05) in cardiomyocytes by CIHH treatment. CONCLUSION: CIHH treatment could reduce the calcium overload and H/R injury in cardiomyocytes by up-regulating the expression of PGC-1α and regulating the energy metabolism of glucose and lipid. The HIF-1α-AMPK signaling pathway might be involved in the process.


Asunto(s)
Miocitos Cardíacos/fisiología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Calcio/metabolismo , Carnitina O-Palmitoiltransferasa/metabolismo , Hipoxia de la Célula/genética , Células Cultivadas , Transportador de Glucosa de Tipo 4/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/antagonistas & inhibidores , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Pirazoles/farmacología , Pirimidinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Troponina I/metabolismo , Regulación hacia Arriba/efectos de los fármacos
6.
J Biol Chem ; 291(17): 9087-104, 2016 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-26929410

RESUMEN

The Piezo2 channel is a newly identified mammalian mechanical transducer that confers rapidly adapting mechanically activated (RA-MA) currents in primary afferent neurons. The Piezo2 channels sense rapid membrane displacement, but it is not clear whether they are sensitive to osmotic swelling, which slowly increases static plasma membrane tension (SPMT). Here, we show that SPMT exerts a profound impact on the mechanical sensitivity of RA-MA channels in primary afferent neurons. RA-MA currents are greatly enhanced, and the mechanical threshold was reduced in both primary afferent neurons of rat dorsal root ganglia (DRG) and HEK293 cells heterologously expressing Piezo2 when these cells undergo osmotic swelling to increase SPMT. Osmotic swelling switches the kinetics of RA-MA currents to the slowly adapting type in both cultured DRG neurons and HEK293 cells heterologously expressing Piezo2. The potentiation of RA-MA currents is abolished when cultured DRG neurons are treated with cytochalasin D, an actin filament disruptor that prevents SPMT of cultured DRG neurons from an increase by osmotic swelling. Osmotic swelling significantly increases DRG neuron mechano-excitability such that a subthreshold mechanical stimulus can result in action potential firing. Behaviorally, the mechanical hind paw withdrawal threshold in rats is reduced following the injection of a hypotonic solution, but this osmotic effect is abolished when cytochalasin D or Gd(3+) is co-administered with the hypo-osmotic solution. Taken together, our findings suggest that Piezo2-mediated mechanotransduction is regulated by SPMT in primary afferent neurons. Because SPMT can be changed by multiple biological factors, our findings may have broad implications in mechanical sensitivity under physiological and pathological conditions.


Asunto(s)
Membrana Celular/metabolismo , Ganglios Espinales/metabolismo , Canales Iónicos/metabolismo , Mecanotransducción Celular/fisiología , Neuronas Aferentes/metabolismo , Presión Osmótica/fisiología , Animales , Membrana Celular/genética , Células Cultivadas , Femenino , Ganglios Espinales/citología , Células HEK293 , Humanos , Canales Iónicos/genética , Masculino , Neuronas Aferentes/citología , Ratas , Ratas Sprague-Dawley , Tensión Superficial
8.
Mol Pain ; 11: 45, 2015 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-26227020

RESUMEN

BACKGROUND: Hyperexcitability of nociceptive afferent fibers is an underlying mechanism of neuropathic pain and ion channels involved in neuronal excitability are potentially therapeutic targets. KCNQ channels, a subfamily of voltage-gated K(+) channels mediating M-currents, play a key role in neuronal excitability. It is unknown whether KCNQ channels are involved in the excitability of nociceptive cold-sensing trigeminal afferent fibers and if so, whether they are therapeutic targets for orofacial cold hyperalgesia, an intractable trigeminal neuropathic pain. METHODS: Patch-clamp recording technique was used to study M-currents and neuronal excitability of cold-sensing trigeminal ganglion neurons. Orofacial operant behavioral assessment was performed in animals with trigeminal neuropathic pain induced by oxaliplatin or by infraorbital nerve chronic constrictive injury. RESULTS: We showed that KCNQ channels were expressed on and mediated M-currents in rat nociceptive cold-sensing trigeminal ganglion (TG) neurons. The channels were involved in setting both resting membrane potentials and rheobase for firing action potentials in these cold-sensing TG neurons. Inhibition of KCNQ channels by linopirdine significantly decreased resting membrane potentials and the rheobase of these TG neurons. Linopirdine directly induced orofacial cold hyperalgesia when the KCNQ inhibitor was subcutaneously injected into rat orofacial regions. On the other hand, retigabine, a KCNQ channel potentiator, suppressed the excitability of nociceptive cold-sensing TG neurons. We further determined whether KCNQ channel could be a therapeutic target for orofacial cold hyperalgesia. Orofacial cold hyperalgesia was induced in rats either by the administration of oxaliplatin or by infraorbital nerve chronic constrictive injury. Using the orofacial operant test, we showed that retigabine dose-dependently alleviated orofacial cold hyperalgesia in both animal models. CONCLUSION: Taken together, these findings indicate that KCNQ channel plays a significant role in controlling cold sensitivity and is a therapeutic target for alleviating trigeminal neuropathic pain that manifests orofacial cold hyperalgesia.


Asunto(s)
Frío , Hiperalgesia/metabolismo , Hiperalgesia/terapia , Canales de Potasio KCNQ/metabolismo , Neuronas/metabolismo , Nocicepción , Sensación Térmica , Potenciales de Acción/efectos de los fármacos , Animales , Carbamatos/farmacología , Enfermedad Crónica , Constricción , Modelos Animales de Enfermedad , Cara , Hiperalgesia/fisiopatología , Masculino , Terapia Molecular Dirigida , Neuronas/efectos de los fármacos , Compuestos Organoplatinos , Oxaliplatino , Fenilendiaminas/farmacología , Ratas Sprague-Dawley , Ganglio del Trigémino/efectos de los fármacos , Ganglio del Trigémino/patología , Ganglio del Trigémino/fisiopatología
9.
Cell Rep ; 43(4): 114014, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38568807

RESUMEN

The transmembrane channel-like (TMC) protein family comprises eight members, with TMC1 and TMC2 being extensively studied. This study demonstrates substantial co-expression of TMC7 with the mechanosensitive channel Piezo2 in somatosensory neurons. Genetic deletion of TMC7 in primary sensory ganglia neurons in vivo enhances sensitivity in both physiological and pathological mechanosensory transduction. This deletion leads to an increase in proportion of rapidly adapting (RA) currents conducted by Piezo2 in dorsal root ganglion (DRG) neurons and accelerates RA deactivation kinetics. In HEK293 cells expressing both proteins, TMC7 significantly suppresses the current amplitudes of co-expressed Piezo2. Our findings reveal that TMC7 and Piezo2 exhibit physical interactions, and both proteins also physically interact with cytoskeletal ß-actin. We hypothesize that TMC7 functions as an inhibitory modulator of Piezo2 in DRG neurons, either through direct inhibition or by disrupting the transmission of mechanical forces from the cytoskeleton to the channel.


Asunto(s)
Ganglios Espinales , Canales Iónicos , Mecanotransducción Celular , Células Receptoras Sensoriales , Humanos , Células Receptoras Sensoriales/metabolismo , Animales , Canales Iónicos/metabolismo , Canales Iónicos/genética , Ganglios Espinales/metabolismo , Células HEK293 , Ratones , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Actinas/metabolismo
10.
Acta Pharm Sin B ; 13(8): 3365-3381, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37655331

RESUMEN

Vincristine, a widely used chemotherapeutic agent for treating different cancer, often induces severe peripheral neuropathic pain. A common symptom of vincristine-induced peripheral neuropathic pain is mechanical allodynia and hyperalgesia. However, mechanisms underlying vincristine-induced mechanical allodynia and hyperalgesia are not well understood. In the present study, we show with behavioral assessment in rats that vincristine induces mechanical allodynia and hyperalgesia in a PIEZO2 channel-dependent manner since gene knockdown or pharmacological inhibition of PIEZO2 channels alleviates vincristine-induced mechanical hypersensitivity. Electrophysiological results show that vincristine potentiates PIEZO2 rapidly adapting (RA) mechanically-activated (MA) currents in rat dorsal root ganglion (DRG) neurons. We have found that vincristine-induced potentiation of PIEZO2 MA currents is due to the enhancement of static plasma membrane tension (SPMT) of these cells following vincristine treatment. Reducing SPMT of DRG neurons by cytochalasin D (CD), a disruptor of the actin filament, abolishes vincristine-induced potentiation of PIEZO2 MA currents, and suppresses vincristine-induced mechanical hypersensitivity in rats. Collectively, enhancing SPMT and subsequently potentiating PIEZO2 MA currents in primary afferent neurons may be an underlying mechanism responsible for vincristine-induced mechanical allodynia and hyperalgesia in rats. Targeting to inhibit PIEZO2 channels may be an effective analgesic method to attenuate vincristine-induced mechanical hypersensitivity.

11.
Mol Pharmacol ; 81(5): 689-700, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22328719

RESUMEN

Transient receptor potential vanilloid type 1 (TRPV1) is a nonselective cation channel activated by capsaicin, low pH, and noxious heat and plays a key role in nociception. Understanding mechanisms for functional modulation of TRPV1 has important implications. One characteristic of TRPV1 is that channel activity induced by either capsaicin or other activators can be sensitized or modulated by factors involving different cell signaling mechanisms. In this study, we describe a novel mechanism for the modulation of TRPV1 function: TRPV1 function is modulated by 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid (DIDS) and its analogs. We found that, in rat dorsal root ganglion neurons, although DIDS did not induce the activation of TRPV1 per se but drastically increased the TRPV1 currents induced by either capsaicin or low pH. DIDS also blocked the tachyphylaxis of the low pH-induced TRPV1 currents. 4-Acetamido-4'-isothiocyanatostilbene-2,2'-disulfonic acid (SITS), a DIDS analog, failed to enhance the capsaicin-evoked TRPV1 current but increased the low pH-evoked TRPV1 currents, with an effect comparable with that of DIDS. SITS also blocked the low pH-induced tachyphylaxis. DIDS also potentiated the currents of TRPV1 channels expressed in human embryonic kidney 293 cells, with an effect of left-shifting the concentration-response curve of the capsaicin-induced TRPV1 currents. This study demonstrates that DIDS and SITS, traditionally used chloride channel blockers, can modify TRPV1 channel function in an agonist-dependent manner. The results provide new input for understanding TRPV1 modulation and developing new modulators of TRPV1 function.


Asunto(s)
Estilbenos/farmacología , Canales Catiónicos TRPV/efectos de los fármacos , Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/farmacología , Ácido 4-Acetamido-4'-isotiocianatostilbeno-2,2'-disulfónico/farmacología , Animales , Ácidos Araquidónicos/farmacología , Calcio/metabolismo , Capsaicina/farmacología , Células Cultivadas , Endocannabinoides , Humanos , Concentración de Iones de Hidrógeno , Alcamidas Poliinsaturadas/farmacología , Ratas , Ratas Sprague-Dawley , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/fisiología
12.
Brain Res Bull ; 191: 93-106, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36330921

RESUMEN

Ferric Chelate Reductase 1 Like (FRRS1L) protein has been identified as an auxiliary regulatory protein for the α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR). FRRS1L is highly expressed in the cerebellum and other brain regions associated with the control of motor function. Loss of FRRS1L has been shown to lead to impaired synaptic transmission via AMPARs and to movement disorders. We found that deletion of the FRRS1L gene causes hyperactivity, reduced muscle strength, impaired coordination, and ataxia in mice. Deletion also impairs Purkinje cell dendritic spine formation and AMPAR expression in the cerebellum and damages the electrophysiological discharge rhythm of Purkinje cells. Cerebrospinal fluid examination and oleic acid (OA)-induced lipid accumulation monitoring in FRRS1L-knockdown SH-SY5Y cells indicated that FRRS1L deficiency could lead to aberrant metabolism of amino acids, glucose, and lipids. In summary, we found that the deletion of FRRS1L leads to impaired motor coordination and cerebellar ataxia in mice, which might be related to the reduced expression of AMPARs, metabolic deviations, and dysplastic functional defects in Purkinje cells.


Asunto(s)
Trastornos del Movimiento , Neuroblastoma , Humanos , Animales , Ratones , Células de Purkinje/fisiología , Cerebelo/metabolismo , Trastornos del Movimiento/genética , Ataxia , Ratones Noqueados , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo
13.
Exp Gerontol ; 151: 111429, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34052348

RESUMEN

Cav3 channels play a critical role in maintaining calcium homeostasis, and its dysregulation is related to age-related diseases, such as age-related hearing loss (AHL). However, the underlying mechanism of the Cav3 channels involved in AHL remains unknown. Previous studies have shown that the degeneration of spiral ganglion neurons (SGNs) plays a critical role in AHL. Here, we explored the involvement of Cav3 channels in the dysregulation of SGNs in AHL. We used C57BL/6 mice as the AHL mouse model and found that the expression of Cav3 channels was increased in SGNs associated with age. The three subtypes of Cav3 channels were present in the apical, middle, and basal SGNs from young and older (AHL) mice. The immunostaining data suggest that Cav3.1 and Cav3.2 may contribute to Cav3 upregulation in SGNs of AHL mice. Additionally, we found that calpain-2 and apoptosis-inducing factor (AIF) were activated in SGNs from AHL mice. The inhibition of Cav3 channels or calpain-2 reduced AIF-activation in SGNs may affect neuronal survival. In conclusion, the findings suggest that Cav3 channels are upregulated in SGNs from AHL mice that may contribute to the degeneration of SGNs through the calpain-2-AIF apoptosis pathway in AHL mice.


Asunto(s)
Pérdida Auditiva , Ganglio Espiral de la Cóclea , Animales , Ratones , Ratones Endogámicos C57BL , Neuronas , Regulación hacia Arriba
14.
Front Mol Neurosci ; 13: 105, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32694980

RESUMEN

M/Kv7 potassium channels play a key role in regulation of neuronal excitability. Modulation of neuronal excitability of primary sensory neurons determines the itch sensation induced by a variety of itch-causing substances including chloroquine (CQ). In the present study, we demonstrate that suppression of M/Kv7 channel activity contributes to generation of itch in mice. CQ enhances excitability of the primary sensory neurons through inhibiting M/Kv7 potassium currents in a Ca2+ influx-dependent manner. Specific M/Kv7 channel opener retigabine (RTG) or tannic acid (TA) not only reverses the CQ-induced enhancement of neuronal excitability but also suppresses the CQ-induced itch behavior. Systemic application of RTG or TA also significantly inhibits the itch behavior induced by a variety of pruritogens. Taken together, our findings provide novel insight into the molecular basis of CQ-induced itch sensation in mammals that can be applied to the development of strategies to mitigate itch behavior.

15.
Aging (Albany NY) ; 12(10): 8923-8938, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32463790

RESUMEN

The two most common aging-related diseases, Alzheimer's disease and type 2 diabetes mellitus, are associated with accumulation of amyloid proteins (ß-amyloid and amylin, respectively). This amylin aggregation is reportedly cytotoxic to neurons. We found that aggregation of human amylin (hAmylin) induced neuronal apoptosis without obvious microglial infiltration in vivo. High concentrations of hAmylin irreversibly aggregated on the surface of the neuronal plasma membrane. Long-term incubation with hAmylin induced morphological changes in neurons. Moreover, hAmylin permeabilized the neuronal membrane within 1 min in a manner similar to Triton X-100, allowing impermeable fluorescent antibodies to enter the neurons and stain intracellular antigens. hAmylin also permeabilized the cell membrane of astrocytes, though more slowly. Under scanning electron microscopy, we observed that hAmylin destroyed the integrity of the cell membranes of both neurons and astrocytes. Additionally, it increased intracellular reactive oxygen species generation and reduced the mitochondrial membrane potential. Thus, by aggregating on the surface of neurons, hAmylin impaired the cell membrane integrity, induced reactive oxygen species production, reduced the mitochondrial membrane potential, and ultimately induced neuronal apoptosis.


Asunto(s)
Membrana Celular/efectos de los fármacos , Hipocampo/citología , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Neuronas/efectos de los fármacos , Animales , Células Cultivadas , Humanos , Neuronas/citología , Ratas , Especies Reactivas de Oxígeno/metabolismo
16.
Front Mol Neurosci ; 12: 296, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31866822

RESUMEN

The transient receptor potential cation channel, vanilloid type (TRPV) 3, is a member of the TRPV subfamily that is expressed predominantly in the skin, hair follicles, and gastrointestinal tract. It is also distributed in the organ of Corti of the inner ear and colocalizes with TRPV1 or TRPV4, but its role in auditory function is unknown. In the present study, we demonstrate that TRPV3 is expressed in inner hair cells (HCs) but mainly in cochlear outer HCs in mice, with expression limited to the cytoplasm and not detected in stereocilia. We compared the number of HCs as well as distortion product otoacoustic emissions (DPOAE) and auditory brainstem response (ABR) thresholds between TRPV3 knockout (V3KO) and wild-type (V3WT) mice and found that although most mutants (72.3%) had normal hearing, a significant proportion (27.7%) showed impaired hearing associated with loss of cochlear HCs. Compensatory upregulation of TRPV4 in HCs prevented HC damage and kanamycin-induced hearing loss and preserved normal auditory function in most of these mice. Thus, TRPV4 and TRPV3 in cochlear HCs protect hearing in mice; moreover, the results suggest some functional redundancy in the functions of TRPV family members. Our findings provide novel insight into the molecular basis of auditory function in mammals that can be applied to the development of strategies to mitigate hearing loss.

17.
Int J Oncol ; 55(3): 629-644, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31322184

RESUMEN

Prostate cancer is one of the most common types of cancer affecting men worldwide; however, its etiology and pathological mechanisms remain poorly understood. Mechanical stimulation plays a key role in prostate cancer development. Piezo type mechanosensitive ion channel component 1 (Piezo1), which functions as a cell sensor and transducer of mechanical stimuli, may have a crucial role in the development of prostate cancer. In the present study, the expression of the Piezo1 channel was demonstrated to be significantly elevated in prostate cancer cell lines and in human prostate malignant tumor tissues. Downregulation of Piezo1 significantly suppressed the viability, proliferation and migration of prostate cancer cells in vitro, and inhibited prostate tumor growth in vivo. The activation of the Akt/mTOR pathway or acceleration of cell cycle progression from G0/G1 to S phase may downstream consequences of Piezo 1 signal pathway activation. Downregulation of Piezo1 considerably suppressed Ca2+ signal increments, inhibited the phosphorylation of Akt and mTOR and arrested the cell cycle of prostate cancer cells at G0/G1 phase in while inhibiting the activation of CDK4 and cyclin D1. Taken together, these findings suggest that Piezo1 channels have a crucial role in prostate cancer development and may, therefore, be a novel therapeutic target in the treatment of prostate cancer.


Asunto(s)
Canales Iónicos/genética , Canales Iónicos/metabolismo , Neoplasias de la Próstata/patología , Transducción de Señal , Regulación hacia Arriba , Animales , Señalización del Calcio , Ciclo Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Trasplante de Neoplasias , Fosforilación , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
18.
J Neurosci ; 27(10): 2503-12, 2007 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-17344388

RESUMEN

KCNQ2/3 currents are the molecular basis of the neuronal M currents that play a critical role in neuron excitability. Many neurotransmitters modulate M/KCNQ currents through their G-protein-coupled receptors. Membrane PtdIns(4,5)P2 hydrolysis and channel phosphorylation are two mechanisms that have been proposed for modulation of KCNQ2/3 currents. In this study, we studied regulation of KCNQ2/3 currents by the epidermal growth factor (EGF) receptor, a member of another family of membrane receptors, receptor tyrosine kinases. We demonstrate here that EGF induces biphasic inhibition of KCNQ2/3 currents in human embryonic kidney 293 cells and in rat superior cervical ganglia neurons, an initial fast inhibition and a later slow inhibition. Additional studies indicate that the early and late inhibitions resulted from PtdIns(4,5)P2 hydrolysis and tyrosine phosphorylation, respectively. We further demonstrate that these two processes are mutually dependent. This study indicates that EGF is a potent modulator of M/KCNQ currents and provides a new dimension to the understanding of the modulation of these channels.


Asunto(s)
Receptores ErbB/metabolismo , Canal de Potasio KCNQ2/antagonistas & inhibidores , Canal de Potasio KCNQ2/fisiología , Canal de Potasio KCNQ3/antagonistas & inhibidores , Canal de Potasio KCNQ3/fisiología , Fosfatos de Fosfatidilinositol/metabolismo , Animales , Células Cultivadas , Conductividad Eléctrica , Factor de Crecimiento Epidérmico/farmacología , Humanos , Hidrólisis , Canal de Potasio KCNQ2/metabolismo , Canal de Potasio KCNQ3/metabolismo , Riñón/citología , Riñón/embriología , Riñón/metabolismo , Membranas/metabolismo , Neuronas/metabolismo , Fosfatidilinositol 4,5-Difosfato , Fosforilación , Ratas , Ratas Sprague-Dawley , Ganglio Cervical Superior/citología , Ganglio Cervical Superior/metabolismo , Tirosina/metabolismo
19.
Mol Pharmacol ; 73(4): 1185-94, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18202303

RESUMEN

Kir2.0 channels play a significant role in setting the resting membrane potential, modulating action potential wave form, and buffering extracellular potassium. One member of this family, Kir2.3, is highly expressed in the heart and brain and is modulated by a variety of factors, including arachidonic acid (AA). Using two-electrode voltage clamp and inside-out patch clamp recordings from Xenopus laevis oocytes expressing Kir2.3 channels, we found that AA selectively activated Kir2.3 channels with an EC(50) of 0.59 muM and that this activation required phosphatidyl inositol 4,5-bisphosphate (PIP(2)). We found that AA activated Kir2.3 by enhancing channel-PIP(2) interactions as demonstrated by a shift in PIP(2) activation curve. EC(50) for channel activation by PIP(2) were 36 and 12 muM in the absence and presence of AA, respectively. A single point mutation on the channel C terminus that enhanced basal channel-PIP(2) interactions reduced the sensitivity of the channel to AA. Effects of AA are mediated through cytoplasmic sites on the channel by increasing the open probability, mainly due to more frequent bursts of opening in the presence of PIP(2). Therefore, enhanced interaction with PIP(2) is the molecular mechanism for Kir2.3 channel activation by AA.


Asunto(s)
Ácido Araquidónico/farmacología , Activación del Canal Iónico/efectos de los fármacos , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Animales , Bovinos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Citosol/efectos de los fármacos , Citosol/metabolismo , Diglicéridos/farmacología , Humanos , Cinética , Ratones , Oocitos/citología , Oocitos/efectos de los fármacos , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Receptor Muscarínico M1/metabolismo , Albúmina Sérica Bovina , Factores de Tiempo , Xenopus laevis
20.
Eur J Pharmacol ; 558(1-3): 21-6, 2007 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-17214982

RESUMEN

In the present study, the effects of antihistamines on inwardly rectifying potassium (Kir) channels expressed in Xenopus oocyte were investigated using two-electrode voltage clamp technique. Firstly, effects of antihistamines on two members of Kir2.0 sub-family, Kir2.1 and Kir2.3 were compared. For antihistamines that selectively block histamine H(1) receptor, the first-generation antihistamines mepyramine and diphenhydramine inhibited Kir2.3 current by 25.0+/-2.9% and 17.3+/-0.7% at concentrations of 100 microM, respectively. In contrast, the second- and third-generation antihistamines astemizole and desloratadine were completely devoid of any inhibitory effect on Kir2.3 current. Histamine H(2) receptor antagonist cimetidine, at 100 microM, failed to inhibit Kir2.3 current. On the other hand, Kir2.1 current was not sensitive to any of these drugs. The mepyramine-induced inhibition of Kir2.3 current was significantly reduced by a single point mutation in Kir2.3 (Kir2.3(I213L)), which enhances Kir2.3-PIP(2) interaction. Secondly, the effect of mepyramine was also tested on Kir3.4*, another member of Kir family. 100 microM mepyramine produced a 30.3+/-4.6% inhibition on Kir3.4* current. These results suggest that the first-generation histamine H(1) receptor antagonists selectively inhibit Kir currents. The inhibitory effect of antihistamines on Kir currents may be involved in their neuronal and cardiac toxic effects caused by drug overdosing.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G/antagonistas & inhibidores , Antagonistas de los Receptores Histamínicos H1/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Animales , Femenino , Fosfatidilinositol 4,5-Difosfato/metabolismo , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA