Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Pharmaceutics ; 14(5)2022 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-35631690

RESUMEN

BACKGROUND AND PURPOSE: The intranasal administration of oxytocin (OT) reduces migraine headaches through activation of the oxytocin receptor (OTR). Magnesium ion (Mg2+) concentration is critical to the activation of the OTR, and a low serum Mg2+ concentration is predictive of a migraine headache. We, therefore, examined the functional impact of Mg2+ concentration on OT-OTR binding efficacy using two complimentary bioassays. EXPERIMENTAL APPROACH: Current clamp recordings of rat trigeminal ganglia (TG) neurons measured the impact of Mg2+ on an OT-induced reduction in excitability. In addition, we assessed the impact of Mg2+ on intranasal OT-induced craniofacial analgesia in rats. KEY RESULTS: While OT alone dose-dependently hyperpolarized TG neurons, decreasing their excitability, the addition of 1.75 mM Mg2+ significantly enhanced this effect. Similarly, while the intranasal application of OT produced dose-dependent craniofacial analgesia, Mg2+ significantly enhanced these effects. CONCLUSIONS AND IMPLICATIONS: OT efficacy may be limited by low ambient Mg2+ levels. The addition of Mg2+ to OT formulations may improve its efficacy in reducing headache pain as well as for other OT-dependent processes.

2.
Neuron ; 109(22): 3609-3618.e9, 2021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34793707

RESUMEN

Mechanisms governing how immune cells and their derived molecules impact homeostatic brain function are still poorly understood. Here, we elucidate neuronal mechanisms underlying T cell effects on synaptic function and episodic memory. Depletion of CD4 T cells led to memory deficits and impaired long-term potentiation. Severe combined immune-deficient mice exhibited amnesia, which was reversible by repopulation with T cells from wild-type but not from IL-4-knockout mice. Behaviors impacted by T cells were mediated via IL-4 receptors expressed on neurons. Exploration of snRNA-seq of neurons participating in memory processing provided insights into synaptic organization and plasticity-associated pathways regulated by immune cells. IL-4Rα knockout in inhibitory (but not in excitatory) neurons was sufficient to impair contextual fear memory, and snRNA-seq from these mice pointed to IL-4-driven regulation of synaptic function in promoting memory. These findings provide new insights into complex neuroimmune interactions at the transcriptional and functional levels in neurons under physiological conditions.


Asunto(s)
Plasticidad Neuronal , Linfocitos T , Animales , Neuronas GABAérgicas , Hipocampo/fisiología , Potenciación a Largo Plazo/fisiología , Memoria/fisiología , Ratones , Ratones Noqueados , Plasticidad Neuronal/fisiología
3.
Heterocycles ; 101(1): 145-164, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32773946

RESUMEN

Neuropathic pain, epilepsy, insomnia, and tremor disorder may arrive from an increase of intracellular Ca2+ concentration through a dysfunction of T-type Ca2+ channels. Thus, T-type calcium channels could be a target in drug discovery for the treatments of neuropathic pain and epilepsy. From rational drug design approach, a group of 2,5-disubstituted 1,3,4-oxadiazole molecules was synthesized and their selective T-type channel inhibitions were evaluated. The synthetic strategy consists of a short sequence of three reactions: (i) condensation of thiosemicarbazide with acid chlorides; (ii) ring closing by 1,3-dibromo-5,5- dimethylhydantoin; and (iii) coupling with various acid chlorides. 5-Chloro-N-(5- phenyl-1,3,4-oxadiazol-2-yl)thiophene-2-carboxamide (11) was found to selectively inhibit T-type Ca2+ channel over Na+ and K+ channels in mouse dorsal root ganglion neurons and/or human embryonic kidney (HEK)-293 cells and to suppress seizure-induced death in mouse model. Consequently, compound 11 is a useful probe for investigation of physiologic and pathophysiologic roles of the T-channel, and provides a basis to develop a novel therapeutic to treat chronic neuropathic and inflammatory pains.

4.
Sleep ; 42(3)2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30535004

RESUMEN

STUDY OBJECTIVES: A major challenge in treating insomnia is to find effective medicines with fewer side effects. Activation of G-protein-gated inward rectifying K+ channels (GIRKs) by GABAB agonists baclofen or γ-hydroxybutyric acid (GHB) promotes nonrapid eye movement (NREM) sleep and consolidates sleep. However, baclofen has poor brain penetration, GHB possesses abuse liability, and in rodents both drugs cause spike-wave discharges (SWDs), an absence seizure activity. We tested the hypothesis that direct GIRK activation promotes sleep without inducing SWD using ML297, a potent and selective GIRK activator. METHODS: Whole-cell patch-clamp recordings from hypocretin/orexin or hippocampal neurons in mouse brain slices were made to study neuronal excitability and synaptic activity; spontaneous activity, locomotion, contextual and tone-conditioned memory, and novel object recognition were assessed. Electroencephalogram/electromyogram (EEG/EMG) recordings were used to study GIRK modulation of sleep. RESULTS: ML297, like baclofen, caused membrane hyperpolarization, decreased input resistance, and blockade of spontaneous action potentials. Unlike baclofen, ML297 (5-10 µM) did not cause significant depression of postsynaptic excitatory and inhibitory currents (EPSCs-IPSCs), indicating preferential postsynaptic inhibition. ML297 (30 mg/kg, i.p.) inhibited wake activity and locomotion, and preferentially increased NREM sleep without altering EEG delta power, REM sleep, inducing SWDs, or impairing conditioned memory and novel object recognition. CONCLUSIONS: This study finds that direct activation of neuronal GIRK channels modulates postsynaptic membrane excitability and prolongs NREM sleep without changing sleep intensity, inducing SWDs, or impairing memory in rodents. These results suggest that direct GIRK activation with a selective compound may present an innovative approach for the treatment of chronic insomnia.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G/agonistas , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Compuestos de Fenilurea/farmacología , Pirazoles/farmacología , Fases del Sueño/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Electromiografía/efectos de los fármacos , Electromiografía/métodos , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp/métodos , Fases del Sueño/efectos de los fármacos
5.
Mol Psychiatry ; 23(12): 2302-2313, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30254235

RESUMEN

Chronic pain poses a heavy burden for the individual and society, comprising personal suffering, comorbid psychiatric symptoms, cognitive decline, and disability. Treatment options are poor due in large part to pain centralization, where an initial injury can result in lasting CNS maladaptations. Hippocampal cellular plasticity in chronic pain has become a focus of study due to its roles in cognition, memory, and the experience of pain itself. However, the extracellular alterations that parallel and facilitate changes in hippocampal function have not been addressed to date. Here we show structural and biochemical plasticity in the hippocampal extracellular matrix (ECM) that is linked to behavioral, cellular, and synaptic changes in a mouse model of chronic pain. Specifically, we report deficits in working location memory that are associated with decreased hippocampal dendritic complexity, altered ECM microarchitecture, decreased ECM rigidity, and changes in the levels of key ECM components and enzymes, including increased levels of MMP8. We also report aberrations in long-term potentiation (LTP) and a loss of inhibitory interneuron perineuronal ECM nets, potentially accounting for the aberrations in LTP. Finally, we demonstrate that MMP8 is upregulated after injury and that its genetic downregulation normalizes the behavioral, electrophysiological, and extracellular alterations. By linking specific extracellular changes to the chronic pain phenotype, we provide a novel mechanistic understanding of pain centralization that provides new targets for the treatment of chronic pain.


Asunto(s)
Hipocampo/metabolismo , Memoria a Corto Plazo/fisiología , Dolor/metabolismo , Animales , Plasticidad de la Célula/fisiología , Cognición , Disfunción Cognitiva/fisiopatología , Matriz Extracelular/metabolismo , Interneuronas , Potenciación a Largo Plazo/fisiología , Masculino , Metaloproteinasa 8 de la Matriz/metabolismo , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Plasticidad Neuronal/fisiología , Lóbulo Temporal
6.
J Alzheimers Dis ; 58(2): 559-574, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28482635

RESUMEN

There is an urgent unmet need for new therapeutics for Alzheimer's disease (AD), the most common cause of dementia in the elderly. Therapeutic approaches targeting amyloid-ß (Aß) and its downstream toxicities have become major strategies in AD drug development. We have taken a rational design approach and synthesized a class of tricyclic pyrone (TP) compounds that show anti-Aß and other neuroprotective actions. The in vivo efficacy of a lead TP named CP2 to ameliorate AD-like pathologies has been shown in mouse models. Here we report the selection and initial characterization of a new lead TP70, which exhibited an anti-Aß therapeutic index even higher than CP2. Moreover, TP70 was able to reduce oxidative stress, inhibit acyl-coenzyme A:cholesterol acyltransferase (ACAT), and upregulate the expression of ATP-binding cassette subfamily A, member 1 (ABCA1), actions considered neuroprotective in AD. TP70 further showed excellent pharmacokinetic properties, including brain penetration and oral availability. When administered to 5xFAD mice via intraperitoneal or oral route, TP70 enhanced the overall solubility and decreased the level of cerebral Aß, including both fibrillary and soluble Aß species. Interestingly, TP70 enhanced N-methyl-D-aspartate (NMDA) receptor-mediated excitatory post-synaptic potential (EPSP) in the hippocampal CA1 area, increased the magnitude of NMDA-dependent hippocampal long-term potentiation (LTP), a cellular model of learning and memory, and prevented the Aß oligomer-impaired LTP. Significantly, a single dose of TP70 administered to aged 5xFAD mice was effective in mitigating the impaired LTP induction, recorded at 24 h after administration. Our results support a potential of TP70 in clinical development for AD in view of its synergistic neuroprotective actions, ability to positively modulate NMDA receptor-mediated hippocampal plasticity, and favorable pharmacokinetic properties in rodents.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Proteínas Amiloidogénicas/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Pironas/uso terapéutico , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/genética , Proteínas Amiloidogénicas/toxicidad , Animales , Encéfalo/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Conducta de Ingestión de Líquido/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Humanos , Locomoción/efectos de los fármacos , Locomoción/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Mutación/genética , Neuroblastoma/patología , Fármacos Neuroprotectores/química , Presenilina-1/genética , Pironas/síntesis química , Pironas/química
7.
Nature ; 544(7651): 488-492, 2017 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-28424512

RESUMEN

Ageing drives changes in neuronal and cognitive function, the decline of which is a major feature of many neurological disorders. The hippocampus, a brain region subserving roles of spatial and episodic memory and learning, is sensitive to the detrimental effects of ageing at morphological and molecular levels. With advancing age, synapses in various hippocampal subfields exhibit impaired long-term potentiation, an electrophysiological correlate of learning and memory. At the molecular level, immediate early genes are among the synaptic plasticity genes that are both induced by long-term potentiation and downregulated in the aged brain. In addition to revitalizing other aged tissues, exposure to factors in young blood counteracts age-related changes in these central nervous system parameters, although the identities of specific cognition-promoting factors or whether such activity exists in human plasma remains unknown. We hypothesized that plasma of an early developmental stage, namely umbilical cord plasma, provides a reservoir of such plasticity-promoting proteins. Here we show that human cord plasma treatment revitalizes the hippocampus and improves cognitive function in aged mice. Tissue inhibitor of metalloproteinases 2 (TIMP2), a blood-borne factor enriched in human cord plasma, young mouse plasma, and young mouse hippocampi, appears in the brain after systemic administration and increases synaptic plasticity and hippocampal-dependent cognition in aged mice. Depletion experiments in aged mice revealed TIMP2 to be necessary for the cognitive benefits conferred by cord plasma. We find that systemic pools of TIMP2 are necessary for spatial memory in young mice, while treatment of brain slices with TIMP2 antibody prevents long-term potentiation, arguing for previously unknown roles for TIMP2 in normal hippocampal function. Our findings reveal that human cord plasma contains plasticity-enhancing proteins of high translational value for targeting ageing- or disease-associated hippocampal dysfunction.


Asunto(s)
Envejecimiento/metabolismo , Proteínas Sanguíneas/farmacología , Sangre Fetal/química , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Plasticidad Neuronal/efectos de los fármacos , Envejecimiento/efectos de los fármacos , Animales , Proteínas Sanguíneas/administración & dosificación , Proteínas Sanguíneas/metabolismo , Cognición/efectos de los fármacos , Cognición/fisiología , Femenino , Hipocampo/citología , Humanos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Plasticidad Neuronal/fisiología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Análisis por Matrices de Proteínas , Memoria Espacial/efectos de los fármacos , Memoria Espacial/fisiología , Inhibidor Tisular de Metaloproteinasa-2/administración & dosificación , Inhibidor Tisular de Metaloproteinasa-2/antagonistas & inhibidores , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/farmacología
8.
ACS Nano ; 11(1): 395-406, 2017 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-28001346

RESUMEN

Neuropathic pain caused by nerve injury is debilitating and difficult to treat. Current systemic pharmacological therapeutics for neuropathic pain produce limited pain relief and have undesirable side effects, while current local anesthetics tend to nonspecifically block both sensory and motor functions. Calcitonin gene related peptide (CGRP), a neuropeptide released from sensory nerve endings, appears to play a significant role in chronic neuropathic pain. In this study, an analgesic microneedle (AMN) patch was developed using dissolvable microneedles to transdermally deliver selective CGRP antagonist peptide in a painless manner for the treatment of localized neuropathic pain. Local analgesic effects were evaluated in rats by testing behavioral pain sensitivity in response to thermal and mechanical stimuli using neuropathic pain models such as spared-nerve injury and diabetic neuropathy pain, as well as neurogenic inflammatory pain model induced by ultraviolet B (UVB) radiation. Unlike several conventional therapies, the AMN patches produced effective analgesia on neuropathic pain without disturbing the normal nociception and motor function of the rat, resulting from the high specificity of the delivered peptide against CGRP receptors. The AMN patches did not cause skin irritation or systemic side effects. These results demonstrate that dissolvable microneedle patches delivering CGRP antagonist peptide provide an effective, safe, and simple approach to mitigate neuropathic pain with significant advantages over current treatments.


Asunto(s)
Analgésicos/uso terapéutico , Neuropatías Diabéticas/tratamiento farmacológico , Edema/tratamiento farmacológico , Neuralgia/tratamiento farmacológico , Traumatismos de la Médula Espinal/tratamiento farmacológico , Analgésicos/química , Animales , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Agujas , Ratas , Ratas Sprague-Dawley , Piel/efectos de los fármacos , Piel/patología , Rayos Ultravioleta
9.
J Neurosci ; 35(33): 11682-93, 2015 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-26290245

RESUMEN

The nociceptin/orphanin FQ (NOP) receptor, the fourth member of the opioid receptor family, is involved in many processes common to the opioid receptors including pain and drug abuse. To better characterize receptor location and trafficking, knock-in mice were created by inserting the gene encoding enhanced green fluorescent protein (eGFP) into the NOP receptor gene (Oprl1) and producing mice expressing a functional NOP-eGFP C-terminal fusion in place of the native NOP receptor. The NOP-eGFP receptor was present in brain of homozygous knock-in animals in concentrations somewhat higher than in wild-type mice and was functional when tested for stimulation of [(35)S]GTPγS binding in vitro and in patch-clamp electrophysiology in dorsal root ganglia (DRG) neurons and hippocampal slices. Inhibition of morphine analgesia was equivalent when tested in knock-in and wild-type mice. Imaging revealed detailed neuroanatomy in brain, spinal cord, and DRG and was generally consistent with in vitro autoradiographic imaging of receptor location. Multicolor immunohistochemistry identified cells coexpressing various spinal cord and DRG cellular markers, as well as coexpression with µ-opioid receptors in DRG and brain regions. Both in tissue slices and primary cultures, the NOP-eGFP receptors appear throughout the cell body and in processes. These knock-in mice have NOP receptors that function both in vitro and in vivo and appear to be an exceptional tool to study receptor neuroanatomy and correlate with NOP receptor function. SIGNIFICANCE STATEMENT: The NOP receptor, the fourth member of the opioid receptor family, is involved in pain, drug abuse, and a number of other CNS processes. The regional and cellular distribution has been difficult to determine due to lack of validated antibodies for immunohistochemical analysis. To provide a new tool for the investigation of receptor localization, we have produced knock-in mice with a fluorescent-tagged NOP receptor in place of the native NOP receptor. These knock-in mice have NOP receptors that function both in vitro and in vivo and have provided a detailed characterization of NOP receptors in brain, spinal cord, and DRG neurons. They appear to be an exceptional tool to study receptor neuroanatomy and correlate with NOP receptor function.


Asunto(s)
Proteínas Fluorescentes Verdes/metabolismo , Microscopía Fluorescente/métodos , Neuronas/citología , Neuronas/metabolismo , Receptores Opioides/metabolismo , Fracciones Subcelulares/metabolismo , Animales , Células Cultivadas , Técnicas de Sustitución del Gen , Proteínas Fluorescentes Verdes/genética , Masculino , Ratones , Ratones Transgénicos , Imagen Molecular/métodos , Receptores Opioides/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Fracciones Subcelulares/ultraestructura , Distribución Tisular , Receptor de Nociceptina
10.
Bioorg Med Chem ; 23(17): 5985-98, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26190460

RESUMEN

A class of tetracyclic terpenes was synthesized and evaluated for antagonistic activity of endothelin-1 (ET-1) induced vasoconstriction and inhibitory activity of voltage-activated Ca(2+) channels. Three repeated Robinson annulation reactions were utilized to construct the tetracyclic molecules. A stereoselective reductive Robinson annulation was discovered for the formation of optically pure tricyclic terpenes. Stereoselective addition of cyanide to the hindered α-face of tetracyclic enone (-)-18 was found and subsequent transformation into the aldehyde function was affected by the formation of bicyclic hemiiminal (-)-4. Six selected synthetic tetracyclic terpenes show inhibitory activities in ET-1 induced vasoconstriction in the gerbil spiral modiolar artery with putative affinity constants ranging between 93 and 319 nM. Moreover, one compound, (-)-3, was evaluated further and found to inhibit voltage-activated Ca(2+) currents but not to affect Na(+) or K(+) currents in dorsal root ganglion cells under similar concentrations. These observations imply a dual mechanism of action. In conclusion, tetracyclic terpenes represent a new class of hit molecules for the discovery of new drugs for the treatment of pulmonary hypertension and vascular related diseases.


Asunto(s)
Canales de Calcio/química , Hipertensión Pulmonar/terapia , Receptor de Endotelina A/química , Terpenos/química , Terpenos/síntesis química , Estructura Molecular
11.
Exp Neurol ; 263: 190-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25448007

RESUMEN

OBJECTIVE: Charcot-Marie-Tooth (CMT) disease is a group of inherited peripheral neuropathies associated with mutations or copy number variations in over 70 genes encoding proteins with fundamental roles in the development and function of Schwann cells and peripheral axons. Here, we used iPSC-derived cells to identify common pathophysiological mechanisms in axonal CMT. METHODS: iPSC lines from patients with two distinct forms of axonal CMT (CMT2A and CMT2E) were differentiated into spinal cord motor neurons and used to study axonal structure and function and electrophysiological properties in vitro. RESULTS: iPSC-derived motor neurons exhibited gene and protein expression, ultrastructural and electrophysiological features of mature primary spinal cord motor neurons. Cytoskeletal abnormalities were found in neurons from a CMT2E (NEFL) patient and corroborated by a mouse model of the same NEFL point mutation. Abnormalities in mitochondrial trafficking were found in neurons derived from this patient, but were only mildly present in neurons from a CMT2A (MFN2) patient. Novel electrophysiological abnormalities, including reduced action potential threshold and abnormal channel current properties were observed in motor neurons derived from both of these patients. INTERPRETATION: Human iPSC-derived motor neurons from axonal CMT patients replicated key pathophysiological features observed in other models of MFN2 and NEFL mutations, including abnormal cytoskeletal and mitochondrial dynamics. Electrophysiological abnormalities found in axonal CMT iPSC-derived human motor neurons suggest that these cells are hyperexcitable and have altered sodium and calcium channel kinetics. These findings may provide a new therapeutic target for this group of heterogeneous inherited neuropathies.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , Enfermedad de Charcot-Marie-Tooth/fisiopatología , Neuronas Motoras/patología , Adulto , Animales , Separación Celular , Enfermedad de Charcot-Marie-Tooth/patología , Niño , Fenómenos Electrofisiológicos , Femenino , GTP Fosfohidrolasas/genética , Técnicas de Sustitución del Gen , Humanos , Células Madre Pluripotentes Inducidas , Filamentos Intermedios/patología , Masculino , Ratones , Mitocondrias/patología , Proteínas Mitocondriales/genética , Proteínas de Neurofilamentos/genética , Técnicas de Placa-Clamp , Fenotipo , Mutación Puntual , Reacción en Cadena en Tiempo Real de la Polimerasa
12.
Nat Med ; 20(6): 659-63, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24793238

RESUMEN

As human lifespan increases, a greater fraction of the population is suffering from age-related cognitive impairments, making it important to elucidate a means to combat the effects of aging. Here we report that exposure of an aged animal to young blood can counteract and reverse pre-existing effects of brain aging at the molecular, structural, functional and cognitive level. Genome-wide microarray analysis of heterochronic parabionts--in which circulatory systems of young and aged animals are connected--identified synaptic plasticity-related transcriptional changes in the hippocampus of aged mice. Dendritic spine density of mature neurons increased and synaptic plasticity improved in the hippocampus of aged heterochronic parabionts. At the cognitive level, systemic administration of young blood plasma into aged mice improved age-related cognitive impairments in both contextual fear conditioning and spatial learning and memory. Structural and cognitive enhancements elicited by exposure to young blood are mediated, in part, by activation of the cyclic AMP response element binding protein (Creb) in the aged hippocampus. Our data indicate that exposure of aged mice to young blood late in life is capable of rejuvenating synaptic plasticity and improving cognitive function.


Asunto(s)
Envejecimiento/fisiología , Transfusión Sanguínea/métodos , Trastornos del Conocimiento/fisiopatología , Trastornos del Conocimiento/terapia , Plasticidad Neuronal/fisiología , Factores de Edad , Envejecimiento/patología , Animales , Western Blotting , Línea Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Cartilla de ADN/genética , Hipocampo/metabolismo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , Parabiosis/métodos , Reacción en Cadena de la Polimerasa
13.
ACS Chem Neurosci ; 4(9): 1278-86, 2013 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-23730969

RESUMEN

The G-protein activated, inward-rectifying potassium (K(+)) channels, "GIRKs", are a family of ion channels (Kir3.1-Kir3.4) that has been the focus of intense research interest for nearly two decades. GIRKs are comprised of various homo- and heterotetrameric combinations of four different subunits. These subunits are expressed in different combinations in a variety of regions throughout the central nervous system and in the periphery. The body of GIRK research implicates GIRK in processes as diverse as controlling heart rhythm, to effects on reward/addiction, to modulation of response to analgesics. Despite years of GIRK research, very few tools exist to selectively modulate GIRK channels' activity and until now no tools existed that potently and selectively activated GIRKs. Here we report the development and characterization of the first truly potent, effective, and selective GIRK activator, ML297 (VU0456810). We further demonstrate that ML297 is active in two in vivo models of epilepsy, a disease where up to 40% of patients remain with symptoms refractory to present treatments. The development of ML297 represents a truly significant advancement in our ability to selectively probe GIRK's role in physiology as well as providing the first tool for beginning to understand GIRK's potential as a target for a diversity of therapeutic indications.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/agonistas , Compuestos de Fenilurea/uso terapéutico , Pirazoles/uso terapéutico , Convulsiones/tratamiento farmacológico , Animales , Anticonvulsivantes/administración & dosificación , Anticonvulsivantes/química , Anticonvulsivantes/farmacología , Señalización del Calcio/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Electrochoque/efectos adversos , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Inyecciones Intraperitoneales , Ratones , Microsomas Hepáticos/metabolismo , Estructura Molecular , Técnicas de Placa-Clamp , Pentilenotetrazol/toxicidad , Compuestos de Fenilurea/administración & dosificación , Compuestos de Fenilurea/química , Compuestos de Fenilurea/farmacología , Pirazoles/administración & dosificación , Pirazoles/química , Pirazoles/farmacología , Ratas , Receptores de Glutamato Metabotrópico/efectos de los fármacos , Proteínas Recombinantes/efectos de los fármacos , Convulsiones/etiología , Ácido Valproico/uso terapéutico
14.
J Neurosci ; 33(12): 5275-84, 2013 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-23516292

RESUMEN

Hypocretin/orexin (Hcrt)-producing neurons in the lateral hypothalamus project throughout the brain, including to the hippocampus, where Hcrt receptors are widely expressed. Hcrt neurons activate these targets to orchestrate global arousal state, wake-sleep architecture, energy homeostasis, stress adaptation, and reward behaviors. Recently, Hcrt has been implicated in cognitive functions and social interaction. In the present study, we tested the hypothesis that Hcrt neurons are critical to social interaction, particularly social memory, using neurobehavioral assessment and electrophysiological approaches. The validated "two-enclosure homecage test" devices and procedure were used to test sociability, preference for social novelty (social novelty), and recognition memory. A conventional direct contact social test was conducted to corroborate the findings. We found that adult orexin/ataxin-3-transgenic (AT) mice, in which Hcrt neurons degenerate by 3 months of age, displayed normal sociability and social novelty with respect to their wild-type littermates. However, AT mice displayed deficits in long-term social memory. Nasal administration of exogenous Hcrt-1 restored social memory to an extent in AT mice. Hippocampal slices taken from AT mice exhibited decreases in degree of paired-pulse facilitation and magnitude of long-term potentiation, despite displaying normal basal synaptic neurotransmission in the CA1 area compared to wild-type hippocampal slices. AT hippocampi had lower levels of phosphorylated cAMP response element-binding protein (pCREB), an activity-dependent transcription factor important for synaptic plasticity and long-term memory storage. Our studies demonstrate that Hcrt neurons play an important role in the consolidation of social recognition memory, at least in part through enhancements of hippocampal synaptic plasticity and cAMP response element-binding protein phosphorylation.


Asunto(s)
Región CA1 Hipocampal/citología , Región CA1 Hipocampal/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Memoria a Largo Plazo/fisiología , Plasticidad Neuronal/fisiología , Neuropéptidos/fisiología , Conducta Social , Animales , Ataxina-3 , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Femenino , Habituación Psicofisiológica/fisiología , Área Hipotalámica Lateral/citología , Área Hipotalámica Lateral/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/farmacología , Potenciación a Largo Plazo/fisiología , Masculino , Trastornos de la Memoria/genética , Trastornos de la Memoria/patología , Trastornos de la Memoria/fisiopatología , Memoria a Largo Plazo/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Vías Nerviosas/citología , Vías Nerviosas/fisiología , Neuronas/metabolismo , Neuronas/fisiología , Neuropéptidos/genética , Neuropéptidos/farmacología , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Orexinas , Técnicas de Cultivo de Órganos , Filtrado Sensorial/fisiología , Olfato/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología
15.
Stroke ; 44(3): 764-70, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23349191

RESUMEN

BACKGROUND AND PURPOSE: Brain ischemia causes immediate and delayed cell death that is exacerbated by inflammation. Recent studies show that hypocretin-1/orexin-A (Hcrt-1) reduces ischemic brain injury, and Hcrt-positive neurons modulate infection-induced inflammation. Here, we tested the hypothesis that Hcrt plays a protective role against ischemia by modulating inflammation. METHODS: Orexin/ataxin-3 (AT) mice, a transgenic strain in which Hcrt-producing neurons degenerate in early adulthood, and wild-type mice were subjected to transient middle cerebral artery occlusion (MCAO). Infarct volume, neurological score, and spontaneous home cage activity were assessed. Inflammation was measured using immunohistochemistry, ELISA, and assessment of cytokine mRNA levels. RESULTS: Infarct volumes 24 and 48 hours after MCAO were significantly larger, neurological score was worse, and spontaneous activity decreased in AT compared with wild-type mice. Macrophage/microglial infiltration and myeloperoxidase-positive cells were higher in AT compared with wild-type mice. Pre-MCAO intracerebroventricular injection of Hcrt-1 significantly reduced infarct volume and macrophage/microglial infiltration in both genotypes and improved neurological score in AT mice. Post-MCAO treatment decreased infarct size in both wild-type and AT mice, but had no effect on neurological score in either genotype. Microglia express the Hcrt-1 receptor after MCAO. Tumor necrosis factor-α production by lipopolysaccharide-stimulated microglial BV2 cells was significantly reduced by Hcrt-1 pretreatment. Sham AT mice exhibit increased brain tumor necrosis factor-α and interleukin-6 mRNA, suggesting chronic inflammation. CONCLUSIONS: Loss of Hcrt neurons in AT mice resulted in worsened stroke outcomes, which were reversed by administration of exogenous Hcrt-1. The mechanism underlying Hcrt-mediated neuroprotection includes attenuation of inflammatory responses after ischemic insult.


Asunto(s)
Isquemia Encefálica/prevención & control , Isquemia Encefálica/fisiopatología , Encefalitis/prevención & control , Encefalitis/fisiopatología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Péptidos y Proteínas de Señalización Intracelular/uso terapéutico , Neuropéptidos/fisiología , Neuropéptidos/uso terapéutico , Animales , Isquemia Encefálica/patología , Movimiento Celular , Encefalitis/patología , Infarto de la Arteria Cerebral Media/complicaciones , Inyecciones Intraventriculares , Interleucina-6/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Microglía/patología , Modelos Animales , Neuropéptidos/genética , Receptores de Orexina , Orexinas , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/metabolismo , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo
16.
Mol Pain ; 8: 65, 2012 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-22963239

RESUMEN

BACKGROUND: Transforming growth factor beta (TGFß) is upregulated in chronic inflammation, where it plays a key role in wound healing and promoting fibrosis. However, little is known about the peripheral effects of TGFß on nociception. METHODS: We tested the in vitro effects of TGFß1 on the excitability of dorsal root ganglia (DRG) neurons and the function of potassium (K) channels. We also studied the effects of TGFß1 infusion on pain responses to noxious electrical stimulation in healthy rats as well as the effects of neutralization of TGFß1 on evoked pain behaviors in a rat model of chronic pancreatitis. RESULTS: Exposure to TGFß1 in vitro increased sensory neuronal excitability, decreased voltage-gated A-type K(+) currents (IA) and downregulated expression of the Kv1.4 (KCNA4) gene. Further TGFß1 infusion into the naïve rat pancreas in vivo induces hyperalgesia and conversely, neutralization of TGFß1 attenuates hyperalgesia only in rats with experimental chronic pancreatitis. Paradoxically, TGFß1 neutralization in naïve rats results in pancreatic hyperalgesia. CONCLUSIONS: TGFß1 is an important and complex modulator of sensory neuronal function in chronic inflammation, providing a link between fibrosis and nociception and is a potentially novel target for the treatment of persistent pain associated with chronic pancreatitis.


Asunto(s)
Hiperalgesia/inducido químicamente , Canal de Potasio Kv1.4/metabolismo , Pancreatitis Crónica/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Animales , Células Cultivadas , Electrofisiología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Hiperalgesia/metabolismo , Inmunohistoquímica , Canal de Potasio Kv1.4/genética , Masculino , Ratas , Ratas Sprague-Dawley , Células Receptoras Sensoriales
17.
Clin Exp Pharmacol Physiol ; 39(7): 614-22, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22540540

RESUMEN

1. To facilitate investigation of diverse rodent behaviours in rodents' home cages, we have developed an integrated modular platform, the SmartCage(™) system (AfaSci, Inc. Burlingame, CA, USA), which enables automated neurobehavioural phenotypic analysis and in vivo drug screening in a relatively higher-throughput and more objective manner. 2, The individual platform consists of an infrared array, a vibration floor sensor and a variety of modular devices. One computer can simultaneously operate up to 16 platforms via USB cables. 3. The SmartCage(™) detects drug-induced increases and decreases in activity levels, as well as changes in movement patterns. Wake and sleep states of mice can be detected using the vibration floor sensor. The arousal state classification achieved up to 98% accuracy compared with results obtained by electroencephalography and electromyography. More complex behaviours, including motor coordination, anxiety-related behaviours and social approach behaviour, can be assessed using appropriate modular devices and the results obtained are comparable with results obtained using conventional methods. 4. In conclusion, the SmartCage(™) system provides an automated and accurate tool to quantify various rodent behaviours in a 'stress-free' environment. This system, combined with the validated testing protocols, offers powerful a tool kit for transgenic phenotyping and in vivo drug screening.


Asunto(s)
Automatización de Laboratorios/instrumentación , Conducta Animal , Vivienda para Animales , Destreza Motora , Animales , Automatización de Laboratorios/métodos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratas , Ratas Sprague-Dawley , Sueño , Vigilia
18.
Nature ; 477(7362): 90-4, 2011 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-21886162

RESUMEN

In the central nervous system, ageing results in a precipitous decline in adult neural stem/progenitor cells and neurogenesis, with concomitant impairments in cognitive functions. Interestingly, such impairments can be ameliorated through systemic perturbations such as exercise. Here, using heterochronic parabiosis we show that blood-borne factors present in the systemic milieu can inhibit or promote adult neurogenesis in an age-dependent fashion in mice. Accordingly, exposing a young mouse to an old systemic environment or to plasma from old mice decreased synaptic plasticity, and impaired contextual fear conditioning and spatial learning and memory. We identify chemokines--including CCL11 (also known as eotaxin)--the plasma levels of which correlate with reduced neurogenesis in heterochronic parabionts and aged mice, and the levels of which are increased in the plasma and cerebrospinal fluid of healthy ageing humans. Lastly, increasing peripheral CCL11 chemokine levels in vivo in young mice decreased adult neurogenesis and impaired learning and memory. Together our data indicate that the decline in neurogenesis and cognitive impairments observed during ageing can be in part attributed to changes in blood-borne factors.


Asunto(s)
Quimiocinas/sangre , Quimiocinas/metabolismo , Aprendizaje/fisiología , Neurogénesis/fisiología , Envejecimiento , Animales , Quimiocina CCL11/sangre , Quimiocina CCL11/líquido cefalorraquídeo , Quimiocina CCL11/metabolismo , Quimiocina CCL11/farmacología , Quimiocinas/líquido cefalorraquídeo , Femenino , Aprendizaje/efectos de los fármacos , Discapacidades para el Aprendizaje/sangre , Discapacidades para el Aprendizaje/líquido cefalorraquídeo , Discapacidades para el Aprendizaje/fisiopatología , Masculino , Trastornos de la Memoria/sangre , Trastornos de la Memoria/líquido cefalorraquídeo , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Endogámicos C57BL , Neurogénesis/efectos de los fármacos , Parabiosis , Plasma/química , Factores de Tiempo
19.
Am J Physiol Gastrointest Liver Physiol ; 301(4): G644-55, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21817062

RESUMEN

Recent studies have explored the potential of central nervous system-derived neural stem cells (CNS-NSC) to repopulate the enteric nervous system. However, the exact phenotypic fate of gut-transplanted CNS-NSC has not been characterized. The aim of this study was to investigate the effect of the gut microenvironment on phenotypic fate of CNS-NSC in vitro. With the use of Transwell culture, differentiation of mouse embryonic CNS-NSC was studied when cocultured without direct contact with mouse intestinal longitudinal muscle-myenteric plexus preparations (LM-MP) compared with control noncocultured cells, in a differentiating medium. Differentiated cells were analyzed by immunocytochemistry and quantitative RT-PCR to assess the expression of specific markers and by whole cell patch-clamp studies for functional characterization of their phenotype. We found that LM-MP cocultured cells had a significant increase in the numbers of cells that were immune reactive against the panneuronal marker ß-tubulin, neurotransmitters neuronal nitric oxide synthase (nNOS), choline acetyltransferase (ChAT), and neuropeptide vasoactive intestinal peptide (VIP) and showed an increase in expression of these genes, compared with control cells. Whole cell patch-clamp analysis showed that coculture with LM-MP decreases cell excitability and reduces voltage-gated Na(+) currents but significantly enhances A-current and late afterhyperpolarization (AHP) and increases the expression of the four AHP-generating Ca(2+)-dependent K(+) channel genes (KCNN), compared with control cells. In a separate experiment, differentiation of LM-MP cocultured CNS-NSC produced a significant increase in the numbers of cells that were immune reactive against the neurotransmitters nNOS, ChAT, and the neuropeptide VIP compared with CNS-NSC differentiated similarly in the presence of neonatal brain tissue. Our results show that the gut microenvironment induces CNS-NSC to produce neurons that share some of the characteristics of classical enteric neurons, further supporting the therapeutic use of these cells for gastrointestinal disorders.


Asunto(s)
Sistema Nervioso Entérico/crecimiento & desarrollo , Intestinos/fisiología , Células-Madre Neurales/fisiología , Potenciales de Acción/fisiología , Animales , Encéfalo/citología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Sistema Nervioso Entérico/fisiología , Femenino , Masculino , Ratones , Plexo Mientérico/fisiología , Neurogénesis/efectos de los fármacos , Neuronas/citología , Técnicas de Placa-Clamp
20.
ACS Chem Neurosci ; 2(12): 730-742, 2011 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-22368764

RESUMEN

T-type Ca(2+) channel inhibitors hold tremendous therapeutic potential for the treatment of pain, epilepsy, sleep disorders, essential tremor and other neurological disorders; however, a lack of truly selective tools has hindered basic research, and selective tools from the pharmaceutical industry are potentially burdened with intellectual property (IP) constraints. Thus, an MLPCN high-throughput screen (HTS) was conducted to identify novel T-type Ca(2+) channel inhibitors free from IP constraints, and freely available through the MLPCN, for use by the biomedical community to study T-type Ca(2+) channels. While the HTS provided numerous hits, these compounds could not be optimized to the required level of potency to be appropriate tool compounds. Therefore, a scaffold hopping approach, guided by SurflexSim, ultimately afforded ML218 (CID 45115620) a selective T-Type Ca(2+) (Ca(v)3.1, Ca(v)3.2, Ca(v)3.3) inhibitor (Ca(v)3.2, IC(50) = 150 nM in Ca(2+) flux; Ca(v)3.2 IC(50) = 310 nM and Ca(v)3.3 IC(50) = 270 nM, respectively in patch clamp electrophysiology) with good DMPK properties, acceptable in vivo rat PK and excellent brain levels. Electrophysiology studies in subthalamic nucleus (STN) neurons demonstrated robust effects of ML218 on the inhibition of T-Type calcium current, inhibition of low threshold spike and rebound burst activity. Based on the basal ganglia circuitry in Parkinson's disease (PD), the effects of ML218 in STN neurons suggest a therapeutic role for T-type Ca(2+) channel inhibitors, and ML218 was found to be orally efficacious in haloperidol-induced catalepsy, a preclinical PD model, with comparable efficacy to an A(2A) antagonist, a clinically validated PD target. ML218 proves to be a powerful new probe to study T-Type Ca(2+) function in vitro and in vivo, and freely available.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...