Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Biol ; 21(8): e3002281, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37643163

RESUMEN

The central circadian clock of the suprachiasmatic nucleus (SCN) is a network consisting of various types of neurons and glial cells. Individual cells have the autonomous molecular machinery of a cellular clock, but their intrinsic periods vary considerably. Here, we show that arginine vasopressin (AVP) neurons set the ensemble period of the SCN network in vivo to control the circadian behavior rhythm. Artificial lengthening of cellular periods by deleting casein kinase 1 delta (CK1δ) in the whole SCN lengthened the free-running period of behavior rhythm to an extent similar to CK1δ deletion specific to AVP neurons. However, in SCN slices, PER2::LUC reporter rhythms of these mice only partially and transiently recapitulated the period lengthening, showing a dissociation between the SCN shell and core with a period instability in the shell. In contrast, in vivo calcium rhythms of both AVP and vasoactive intestinal peptide (VIP) neurons in the SCN of freely moving mice demonstrated stably lengthened periods similar to the behavioral rhythm upon AVP neuron-specific CK1δ deletion, without changing the phase relationships between each other. Furthermore, optogenetic activation of AVP neurons acutely induced calcium increase in VIP neurons in vivo. These results indicate that AVP neurons regulate other SCN neurons, such as VIP neurons, in vivo and thus act as a primary determinant of the SCN ensemble period.


Asunto(s)
Arginina Vasopresina , Calcio , Animales , Ratones , Neuronas , Núcleo Supraquiasmático , Neuroglía , Péptido Intestinal Vasoactivo
2.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33526663

RESUMEN

The suprachiasmatic nucleus (SCN), the central circadian pacemaker in mammals, is a network structure composed of multiple types of γ-aminobutyric acid (GABA)-ergic neurons and glial cells. However, the roles of GABA-mediated signaling in the SCN network remain controversial. Here, we report noticeable impairment of the circadian rhythm in mice with a specific deletion of the vesicular GABA transporter in arginine vasopressin (AVP)-producing neurons. These mice showed disturbed diurnal rhythms of GABAA receptor-mediated synaptic transmission in SCN neurons and marked lengthening of the activity time in circadian behavioral rhythms due to the extended interval between morning and evening locomotor activities. Synchrony of molecular circadian oscillations among SCN neurons did not significantly change, whereas the phase relationships between SCN molecular clocks and circadian morning/evening locomotor activities were altered significantly, as revealed by PER2::LUC imaging of SCN explants and in vivo recording of intracellular Ca2+ in SCN AVP neurons. In contrast, daily neuronal activity in SCN neurons in vivo clearly showed a bimodal pattern that correlated with dissociated morning/evening locomotor activities. Therefore, GABAergic transmission from AVP neurons regulates the timing of SCN neuronal firing to temporally restrict circadian behavior to appropriate time windows in SCN molecular clocks.


Asunto(s)
Relojes Circadianos , Ritmo Circadiano , Neuronas/metabolismo , Núcleo Supraquiasmático/metabolismo , Vasopresinas/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Conducta Animal , Calcio/metabolismo , Relojes Circadianos/genética , Ritmo Circadiano/genética , Regulación de la Expresión Génica , Locomoción , Ratones , Sinapsis/fisiología , Transmisión Sináptica/fisiología , Factores de Tiempo , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/deficiencia , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo
3.
PLoS Pathog ; 15(5): e1007756, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31095648

RESUMEN

ADP-ribosylation is a ubiquitous post-translational addition of either monomers or polymers of ADP-ribose to target proteins by ADP-ribosyltransferases, usually by interferon-inducible diphtheria toxin-like enzymes known as PARPs. While several PARPs have known antiviral activities, these activities are mostly independent of ADP-ribosylation. Consequently, less is known about the antiviral effects of ADP-ribosylation. Several viral families, including Coronaviridae, Togaviridae, and Hepeviridae, encode for macrodomain proteins that bind to and hydrolyze ADP-ribose from proteins and are critical for optimal replication and virulence. These results suggest that macrodomains counter cellular ADP-ribosylation, but whether PARPs or, alternatively, other ADP-ribosyltransferases cause this modification is not clear. Here we show that pan-PARP inhibition enhanced replication and inhibited interferon production in primary macrophages infected with macrodomain-mutant but not wild-type coronavirus. Specifically, knockdown of two abundantly expressed PARPs, PARP12 and PARP14, led to increased replication of mutant but did not significantly affect wild-type virus. PARP14 was also important for the induction of interferon in mouse and human cells, indicating a critical role for this PARP in the regulation of innate immunity. In summary, these data demonstrate that the macrodomain is required to prevent PARP-mediated inhibition of coronavirus replication and enhancement of interferon production.


Asunto(s)
Infecciones por Coronavirus/virología , Coronavirus/inmunología , Inmunidad Innata/inmunología , Interferones/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Replicación Viral , ADP-Ribosilación , Animales , Coronavirus/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/metabolismo , Humanos , Inmunidad Innata/efectos de los fármacos , Ratones , Poli(ADP-Ribosa) Polimerasas/química , Poli(ADP-Ribosa) Polimerasas/genética , Dominios Proteicos , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Virulencia
4.
Circulation ; 139(1): 78-96, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30586693

RESUMEN

BACKGROUND: Chronic kidney disease (CKD) increases cardiovascular risk. Underlying mechanisms, however, remain obscure. The uremic toxin indoxyl sulfate is an independent cardiovascular risk factor in CKD. We explored the potential impact of indoxyl sulfate on proinflammatory activation of macrophages and its underlying mechanisms. METHODS: We examined in vitro the effects of clinically relevant concentrations of indoxyl sulfate on proinflammatory responses of macrophages and the roles of organic anion transporters and organic anion transporting polypeptides (OATPs). A systems approach, involving unbiased global proteomics, bioinformatics, and network analysis, then explored potential key pathways. To address the role of Delta-like 4 (Dll4) in indoxyl sulfate-induced macrophage activation and atherogenesis in CKD in vivo, we used 5/6 nephrectomy and Dll4 antibody in low-density lipoprotein receptor-deficient (Ldlr-/-) mice. To further determine the relative contribution of OATP2B1 or Dll4 to proinflammatory activation of macrophages and atherogenesis in vivo, we used siRNA delivered by macrophage-targeted lipid nanoparticles in mice. RESULTS: We found that indoxyl sulfate-induced proinflammatory macrophage activation is mediated by its uptake through transporters, including OATP2B1, encoded by the SLCO2B1 gene. The global proteomics identified potential mechanisms, including Notch signaling and the ubiquitin-proteasome pathway, that mediate indoxyl sulfate-triggered proinflammatory macrophage activation. We chose the Notch pathway as an example of key candidates for validation of our target discovery platform and for further mechanistic studies. As predicted computationally, indoxyl sulfate triggered Notch signaling, which was preceded by the rapid induction of Dll4 protein. Dll4 induction may result from inhibition of the ubiquitin-proteasome pathway, via the deubiquitinating enzyme USP5. In mice, macrophage-targeted OATP2B1/Slco2b1 silencing and Dll4 antibody inhibited proinflammatory activation of peritoneal macrophages induced by indoxyl sulfate. In low-density lipoprotein receptor-deficient mice, Dll4 antibody abolished atherosclerotic lesion development accelerated in Ldlr-/- mice. Moreover, coadministration of indoxyl sulfate and OATP2B1/Slco2b1 or Dll4 siRNA encapsulated in macrophage-targeted lipid nanoparticles in Ldlr-/- mice suppressed lesion development. CONCLUSIONS: These results suggest that novel crosstalk between OATP2B1 and Dll4-Notch signaling in macrophages mediates indoxyl sulfate-induced vascular inflammation in CKD.


Asunto(s)
Aterosclerosis/metabolismo , Indicán/toxicidad , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Transportadores de Anión Orgánico/metabolismo , Receptores Notch/metabolismo , Insuficiencia Renal Crónica/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/prevención & control , Proteínas de Unión al Calcio , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Transportadores de Anión Orgánico/genética , Fenotipo , Placa Aterosclerótica , Células RAW 264.7 , Receptores de LDL/deficiencia , Receptores de LDL/genética , Receptores Notch/genética , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/patología , Transducción de Señal/efectos de los fármacos , Calcificación Vascular/metabolismo , Calcificación Vascular/patología
5.
Addict Biol ; 25(1): e12723, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-30734456

RESUMEN

In drug addiction, environmental stimuli previously associated with cocaine use readily elicit cocaine-associated memories, which persist long after abstinence and trigger cocaine craving and consumption. Although previous studies suggest that the medial prefrontal cortex (mPFC) is involved in the expression of cocaine-addictive behaviors, it remains unclear whether excitatory and inhibitory neurons in the mPFC are causally related to the formation and retrieval of cocaine-associated memories. To address this issue, we used the designer receptors exclusively activated by designer drugs (DREADD) technology combined with a cocaine-induced conditioned place preference (CPP) paradigm. We suppressed mPFC neuronal activity in a cell-type- and timing-dependent manner. C57BL/6J wild-type mice received bilateral intra-mPFC infusion of an adeno-associated virus (AAV) expressing inhibitory DREADD (hM4Di) under the control of CaMKII promotor to selectively suppress mPFC pyramidal neurons. GAD67-Cre mice received bilateral intra-mPFC infusion of a Cre-dependent AAV expressing hM4Di to specifically silence GABAergic neurons. Chemogenetic suppression of mPFC pyramidal neurons significantly attenuated both the acquisition and expression of cocaine CPP, while suppression of mPFC GABAergic neurons affected neither the acquisition nor expression of cocaine CPP. Moreover, chemogenetic inhibition of mPFC glutamatergic neurons did not affect the acquisition and expression of lithium chloride-induced conditioned place aversion. These results suggest that the activation of glutamatergic, but not GABAergic, neurons in the mPFC mediates both the formation and retrieval of cocaine-associated memories.


Asunto(s)
Trastornos Relacionados con Cocaína/fisiopatología , Cocaína/farmacología , Neuronas GABAérgicas/efectos de los fármacos , Memoria/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiopatología , Animales , Modelos Animales de Enfermedad , Inhibidores de Captación de Dopamina/farmacología , Electrofisiología , Técnica del Anticuerpo Fluorescente , Masculino , Ratones , Ratones Endogámicos C57BL
6.
Proc Natl Acad Sci U S A ; 114(17): E3526-E3535, 2017 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-28396432

RESUMEN

Narcolepsy is a sleep disorder caused by the loss of orexin (hypocretin)-producing neurons and marked by excessive daytime sleepiness and a sudden weakening of muscle tone, or cataplexy, often triggered by strong emotions. In a mouse model for narcolepsy, we previously demonstrated that serotonin neurons of the dorsal raphe nucleus (DRN) mediate the suppression of cataplexy-like episodes (CLEs) by orexin neurons. Using an optogenetic tool, in this paper we show that the acute activation of DRN serotonin neuron terminals in the amygdala, but not in nuclei involved in regulating rapid eye-movement sleep and atonia, suppressed CLEs. Not only did stimulating serotonin nerve terminals reduce amygdala activity, but the chemogenetic inhibition of the amygdala using designer receptors exclusively activated by designer drugs also drastically decreased CLEs, whereas chemogenetic activation increased them. Moreover, the optogenetic inhibition of serotonin nerve terminals in the amygdala blocked the anticataplectic effects of orexin signaling in DRN serotonin neurons. Taken together, the results suggest that DRN serotonin neurons, as a downstream target of orexin neurons, inhibit cataplexy by reducing the activity of amygdala as a center for emotional processing.


Asunto(s)
Amígdala del Cerebelo , Catalepsia , Núcleo Dorsal del Rafe , Neuronas Serotoninérgicas/metabolismo , Transducción de Señal , Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/patología , Amígdala del Cerebelo/fisiopatología , Animales , Catalepsia/genética , Catalepsia/metabolismo , Catalepsia/patología , Catalepsia/fisiopatología , Núcleo Dorsal del Rafe/metabolismo , Núcleo Dorsal del Rafe/patología , Núcleo Dorsal del Rafe/fisiopatología , Movimientos Oculares , Masculino , Ratones , Ratones Noqueados , Neuronas Serotoninérgicas/patología , Serotonina/metabolismo
7.
J Neurosci ; 38(28): 6366-6378, 2018 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-29915137

RESUMEN

The hypothalamus plays an important role in the regulation of sleep/wakefulness states. While the ventrolateral preoptic nucleus (VLPO) plays a critical role in the initiation and maintenance of sleep, the lateral posterior part of the hypothalamus contains neuronal populations implicated in maintenance of arousal, including orexin-producing neurons (orexin neurons) in the lateral hypothalamic area (LHA) and histaminergic neurons in the tuberomammillary nucleus (TMN). During a search for neurons that make direct synaptic contact with histidine decarboxylase-positive (HDC+), histaminergic neurons (HDC neurons) in the TMN and orexin neurons in the LHA of male mice, we found that these arousal-related neurons are heavily innervated by GABAergic neurons in the preoptic area including the VLPO. We further characterized GABAergic neurons electrophysiologically in the VLPO (GABAVLPO neurons) that make direct synaptic contact with these hypothalamic arousal-related neurons. These neurons (GABAVLPO→HDC or GABAVLPO→orexin neurons) were both potently inhibited by noradrenaline and serotonin, showing typical electrophysiological characteristics of sleep-promoting neurons in the VLPO. This work provides direct evidence of monosynaptic connectivity between GABAVLPO neurons and hypothalamic arousal neurons and identifies the effects of monoamines on these neuronal pathways.SIGNIFICANCE STATEMENT Rabies-virus-mediated tracing of input neurons of two hypothalamic arousal-related neuron populations, histaminergic and orexinergic neurons, showed that they receive similar distributions of input neurons in a variety of brain areas, with rich innervation by GABAergic neurons in the preoptic area, including the ventrolateral preoptic area (VLPO), a region known to play an important role in the initiation and maintenance of sleep. Electrophysiological experiments found that GABAergic neurons in the VLPO (GABAVLPO neurons) that make direct input to orexin or histaminergic neurons are potently inhibited by noradrenaline and serotonin, suggesting that these monoamines disinhibit histamine and orexin neurons. This work demonstrated functional and structural interactions between GABAVLPO neurons and hypothalamic arousal-related neurons.


Asunto(s)
Nivel de Alerta/fisiología , Neuronas GABAérgicas/fisiología , Área Hipotalámica Lateral/fisiología , Área Preóptica/fisiología , Sueño/fisiología , Animales , Neuronas GABAérgicas/citología , Área Hipotalámica Lateral/citología , Masculino , Ratones , Ratones Transgénicos , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Norepinefrina/metabolismo , Área Preóptica/citología , Serotonina/metabolismo
8.
J Proteome Res ; 18(4): 1607-1622, 2019 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-30848916

RESUMEN

ADP-ribosylation is a post-translational modification that, until recently, has remained elusive to study at the cellular level. Previously dependent on radioactive tracers to identify ADP-ribosylation targets, several advances in mass spectrometric workflows now permit global identification of ADP-ribosylated substrates. In this study, we capitalized on two ADP-ribosylation enrichment strategies, and multiple activation methods performed on the Orbitrap Fusion Lumos, to identify IFN-γ-induced ADP-ribosylation substrates in macrophages. The ADP-ribosyl binding protein, Af1521, was used to enrich ADP-ribosylated peptides, and the antipoly-ADP-ribosyl antibody, 10H, was used to enrich ADP-ribosylated proteins. ADP-ribosyl-specific mass spectra were further enriched by an ADP-ribose product ion triggered EThcD and HCD activation strategy, in combination with multiple acquisitions that segmented the survey scan into smaller ranges. HCD and EThcD resulted in overlapping and unique ADP-ribosyl peptide identifications, with HCD providing more peptide identifications but EThcD providing more reliable ADP-ribosyl acceptor sites. Our acquisition strategies also resulted in the first ever characterization of ADP-ribosyl on three poly-ADP-ribose polymerases, ARTD9/PARP9, ARTD10/PARP10, and ARTD8/PARP14. IFN-γ increased the ADP-ribosylation status of ARTD9/PARP9, ARTD8/PARP14, and proteins involved in RNA processes. This study therefore summarizes specific molecular pathways at the intersection of IFN-γ and ADP-ribosylation signaling pathways.


Asunto(s)
ADP-Ribosilación/fisiología , Interferón gamma/metabolismo , Proteínas de Neoplasias/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Humanos , Macrófagos/metabolismo , Proteínas de Neoplasias/química , Poli(ADP-Ribosa) Polimerasas/química , Mapas de Interacción de Proteínas/fisiología , Proteómica , Células THP-1
9.
J Neurosci ; 33(12): 5162-74, 2013 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-23516282

RESUMEN

Ataxia, episodic dyskinesia, and thalamocortical seizures are associated with an inherited loss of P/Q-type voltage-gated Ca(2+) channel function. P/Q-type channels are widely expressed throughout the neuraxis, obscuring identification of the critical networks underlying these complex neurological disorders. We showed recently that the conditional postnatal loss of P/Q-type channels in cerebellar Purkinje cells (PCs) in mice (purky) leads to these aberrant phenotypes, suggesting that intrinsic alteration in PC output is a sufficient pathogenic factor for disease initiation. The question arises whether P/Q-type channel deletion confined to a single upstream cerebellar synapse might induce the pathophysiological abnormality of genomically inherited P/Q-type channel disorders. PCs integrate two excitatory inputs, climbing fibers from inferior olive and parallel fibers (PFs) from granule cells (GCs) that receive mossy fiber (MF) input derived from precerebellar nuclei. In this study, we introduce a new mouse model with a selective knock-out of P/Q-type channels in rhombic-lip-derived neurons including the PF and MF pathways (quirky). We found that in quirky mice, PF-PC synaptic transmission is reduced during low-frequency stimulation. Using focal light stimulation of GCs that express optogenetic light-sensitive channels, channelrhodopsin-2, we found that modulation of PC firing via GC input is reduced in quirky mice. Phenotypic analysis revealed that quirky mice display ataxia, dyskinesia, and absence epilepsy. These results suggest that developmental alteration of patterned input confined to only one of the main afferent cerebellar excitatory synaptic pathways has a significant role in generating the neurological phenotype associated with the global genomic loss of P/Q-type channel function.


Asunto(s)
Ataxia/fisiopatología , Canales de Calcio Tipo N/fisiología , Canales de Calcio Tipo P/fisiología , Canales de Calcio Tipo Q/fisiología , Cerebelo/anomalías , Epilepsia Tipo Ausencia/fisiopatología , Células de Purkinje/fisiología , Animales , Ataxia/genética , Ataxia/patología , Canales de Calcio Tipo N/genética , Canales de Calcio Tipo P/genética , Canales de Calcio Tipo Q/genética , Cerebelo/fisiopatología , Electroencefalografía , Epilepsia Tipo Ausencia/genética , Epilepsia Tipo Ausencia/patología , Potenciales Postsinápticos Excitadores/genética , Potenciales Postsinápticos Excitadores/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes Neurológicos , Fenotipo , Transmisión Sináptica/fisiología , Grabación de Cinta de Video
10.
Sci Rep ; 13(1): 16974, 2023 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-37813987

RESUMEN

Prokineticin 2 (Prok2) is a small protein expressed in a subpopulation of neurons in the suprachiasmatic nucleus (SCN), the primary circadian pacemaker in mammals. Prok2 has been implicated as a candidate output molecule from the SCN to control multiple circadian rhythms. Genetic manipulation specific to Prok2-producing neurons would be a powerful approach to understanding their function. Here, we report the generation of Prok2-tTA knock-in mice expressing the tetracycline transactivator (tTA) specifically in Prok2 neurons and an application of these mice to in vivo recording of Ca2+ rhythms in these neurons. First, the specific and efficient expression of tTA in Prok2 neurons was verified by crossing the mice with EGFP reporter mice. Prok2-tTA mice were then used to express a fluorescent Ca2+ sensor protein to record the circadian Ca2+ rhythm in SCN Prok2 neurons in vivo. Ca2+ in these cells showed clear circadian rhythms in both light-dark and constant dark conditions, with their peaks around midday. Notably, the hours of high Ca2+ nearly coincided with the rest period of the behavioral rhythm. These observations fit well with the predicted function of Prok2 neurons as a candidate output pathway of the SCN by suppressing locomotor activity during both daytime and subjective daytime.


Asunto(s)
Calcio , Neuronas del Núcleo Supraquiasmático , Ratones , Animales , Calcio/metabolismo , Núcleo Supraquiasmático/fisiología , Ritmo Circadiano/fisiología , Neuronas del Núcleo Supraquiasmático/metabolismo , Neuronas/metabolismo , Transactivadores/metabolismo , Calcio de la Dieta/metabolismo , Mamíferos/metabolismo
11.
Neuroscience ; 532: 87-102, 2023 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-37778689

RESUMEN

The thalamic reticular nucleus (TRN) is a thin sheet of GABAergic neurons surrounding the thalamus, and it regulates the activity of thalamic relay neurons. The TRN has been reported to be involved in sensory gating, attentional regulation, and some other functions. However, little is known about the contribution of the TRN to sequence learning. In the present study, we examined whether the TRN is involved in reward-based learning of action sequence with no eliciting stimuli (operant conditioning), by analyzing the performance of male and female Avp-Vgat-/- mice (Vgatflox/flox mice crossed to an Avp-Cre driver line) on tasks conducted in an operant box having three levers. Our histological and electrophysiological data demonstrated that in adult Avp-Vgat-/- mice, vesicular GABA transporter (VGAT) was absent in most TRN neurons and the GABAergic transmission from the TRN to the thalamus was largely suppressed. The performance on a task in which mice needed to press an active lever for food reward showed that simple operant learning of lever pressing and learning of win-stay and lose-shift strategies are not affected in Avp-Vgat-/- mice. In contrast, the performance on a task in which mice needed to press three levers in a correct order for food reward showed that learning of the order of lever pressing (action sequence learning) was impaired in Avp-Vgat-/- mice. These results suggest that the TRN plays an important role in action sequence learning.


Asunto(s)
Núcleos Talámicos , Tálamo , Ratones , Masculino , Femenino , Animales , Núcleos Talámicos/fisiología , Neuronas GABAérgicas/fisiología , Aprendizaje/fisiología , Condicionamiento Operante
12.
J Neurosci ; 31(11): 4311-26, 2011 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-21411672

RESUMEN

Inherited loss of P/Q-type calcium channel function causes human absence epilepsy, episodic dyskinesia, and ataxia, but the molecular "birthdate" of the neurological syndrome and its dependence on prenatal pathophysiology is unknown. Since these channels mediate transmitter release at synapses throughout the brain and are expressed early in embryonic development, delineating the critical circuitry and onset underlying each of the emergent phenotypes requires targeted control of gene expression. To visualize P/Q-type Ca(2+) channels and dissect their role in neuronal networks at distinct developmental stages, we created a novel conditional Cacna1a knock-in mouse by inserting the floxed green fluorescent protein derivative Citrine into the first exon of Cacna1a and then crossed it with a postnatally expressing PCP2-Cre line for delayed Purkinje cell (PC) gene deletion within the cerebellum and sparsely in forebrain (purky). PCs in purky mice lacked P/Q-type calcium channel protein and currents within the first month after birth, displayed altered spontaneous firing, and showed impaired neurotransmission. Unexpectedly, adult purky mice exhibited the full spectrum of neurological deficits seen in mice with genomic Cacna1a ablation. Our results show that the ataxia, dyskinesia, and absence epilepsy caused by inherited disorders of the P/Q-type channel arise from signaling defects beginning in late infancy, revealing an early window of opportunity for therapeutic intervention.


Asunto(s)
Ataxia/genética , Canales de Calcio Tipo P/genética , Canales de Calcio Tipo Q/genética , Discinesias/genética , Epilepsia Tipo Ausencia/genética , Células de Purkinje/metabolismo , Análisis de Varianza , Animales , Ataxia/metabolismo , Canales de Calcio Tipo P/metabolismo , Canales de Calcio Tipo Q/metabolismo , Cerebelo/metabolismo , Discinesias/metabolismo , Electroencefalografía , Electrofisiología , Epilepsia Tipo Ausencia/metabolismo , Inmunohistoquímica , Potenciales Postsinápticos Inhibidores/fisiología , Ratones , Ratones Transgénicos , Mutación , Fenotipo
13.
J Biol Chem ; 286(29): 25848-58, 2011 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-21628464

RESUMEN

G protein-coupled receptors are involved in the modulation of complex neuronal networks in the brain. To investigate the impact of a cell-specific G(i/o) protein-mediated signaling pathway on brain function, we created a new optogenetic mouse model in which the G(i/o) protein-coupled receptor vertebrate rhodopsin can be cell-specifically expressed with the aid of Cre recombinase. Here we use this mouse model to study the functional impact of G(i/o) modulation in cerebellar Purkinje cells (PCs). We show that in vivo light activation of vertebrate rhodopsin specifically expressed in PCs reduces simple spike firing that is comparable with the reduction in firing observed for the activation of cerebellar G(i/o)-coupled GABA(B) receptors. Notably, the light exposure of the cerebellar vermis in freely moving mice changes the motor behavior. Thus, our studies directly demonstrate that spike modulation via G(i/o)-mediated signaling in cerebellar PCs affects motor coordination and show a new promising approach for studying the physiological function of G protein-coupled receptor-mediated signaling in a cell type-specific manner.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Actividad Motora/genética , Actividad Motora/efectos de la radiación , Fenómenos Ópticos , Células de Purkinje/metabolismo , Células de Purkinje/efectos de la radiación , Rodopsina/metabolismo , Animales , Conducta Animal/efectos de la radiación , Regulación de la Expresión Génica/efectos de la radiación , Luz , Masculino , Ratones , Ratones Transgénicos , Rodopsina/genética , Transducción de Señal/efectos de la radiación
14.
Mol Brain ; 15(1): 47, 2022 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-35606816

RESUMEN

Self-grooming plays an essential role in hygiene maintenance, thermoregulation, and stress response. However, the neural populations involved in self-grooming remain largely unknown. The paraventricular hypothalamic nucleus (PVH) has been implicated in the regulation of self-grooming. Arginine vasopressin-producing neurons are among the major neuronal populations in the PVH (PVHAVP), which play important roles in water homeostasis, blood pressure regulation, feeding, and stress response. Here, we report the critical role of PVHAVP neurons in the induction of self-grooming. Optogenetic activation of PVHAVP neurons immediately induced self-grooming in freely moving mice. Chemogenetic activation of these neurons also increased time spent self-grooming. In contrast, their chemogenetic inhibition significantly reduced naturally occurring self-grooming, suggesting that PVHAVP-induced grooming has physiological relevance. Notably, optogenetic activation of PVHAVP neurons triggered self-grooming over other adaptive behaviors, such as voracious feeding induced by fasting and social interaction with female mice. Thus, our study proposes the novel role of PVHAVP neurons in regulating self-grooming behavior and, consequently, hygiene maintenance and stress response. Furthermore, uncontrolled activation of these neurons may be potentially relevant to diseases characterized by compulsive behaviors and impaired social interaction, such as autism, obsessive-compulsive disorder, and anorexia nervosa.


Asunto(s)
Arginina Vasopresina , Núcleo Hipotalámico Paraventricular , Animales , Arginina Vasopresina/genética , Arginina Vasopresina/metabolismo , Femenino , Aseo Animal , Ratones , Neuronas/metabolismo , Optogenética , Núcleo Hipotalámico Paraventricular/metabolismo
15.
Curr Biol ; 32(18): 3871-3885.e4, 2022 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-35907397

RESUMEN

The sleep-wakefulness cycle is regulated by complicated neural networks that include many different populations of neurons throughout the brain. Arginine vasopressin neurons in the paraventricular nucleus of the hypothalamus (PVHAVP) regulate various physiological events and behaviors, such as body-fluid homeostasis, blood pressure, stress response, social interaction, and feeding. Changes in arousal level often accompany these PVHAVP-mediated adaptive responses. However, the contribution of PVHAVP neurons to sleep-wakefulness regulation has remained unknown. Here, we report the involvement of PVHAVP neurons in arousal promotion. Optogenetic stimulation of PVHAVP neurons rapidly induced transitions to wakefulness from both NREM and REM sleep. This arousal effect was dependent on AVP expression in these neurons. Similarly, chemogenetic activation of PVHAVP neurons increased wakefulness and reduced NREM and REM sleep, whereas chemogenetic inhibition of these neurons significantly reduced wakefulness and increased NREM sleep. We observed dense projections of PVHAVP neurons in the lateral hypothalamus with potential connections to orexin/hypocretin (LHOrx) neurons. Optogenetic stimulation of PVHAVP neuronal fibers in the LH immediately induced wakefulness, whereas blocking orexin receptors attenuated the arousal effect of PVHAVP neuronal activation drastically. Monosynaptic rabies-virus tracing revealed that PVHAVP neurons receive inputs from multiple brain regions involved in sleep-wakefulness regulation, as well as those involved in stress response and energy metabolism. Moreover, PVHAVP neurons mediated the arousal induced by novelty stress and a melanocortin receptor agonist melanotan-II. Thus, our data suggested that PVHAVP neurons promote wakefulness via LHOrx neurons in the basal sleep-wakefulness and some stressful conditions.


Asunto(s)
Área Hipotalámica Lateral , Vigilia , Arginina Vasopresina/metabolismo , Área Hipotalámica Lateral/fisiología , Hipotálamo/metabolismo , Neuronas/fisiología , Receptores de Orexina/metabolismo , Orexinas/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Receptores de Melanocortina/metabolismo , Sueño/fisiología , Vasopresinas/metabolismo , Vasopresinas/farmacología , Vigilia/fisiología
16.
Front Cell Neurosci ; 16: 877131, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36274991

RESUMEN

Astrocytes play key roles in supporting the central nervous system structure, regulating synaptic functions, and maintaining brain homeostasis. The number of astrocytes in the cerebrum has markedly increased through evolution. However, the manner by which astrocytes change their features during evolution remains unknown. Compared with the rodent brain, the brain of the ferret, a carnivorous animal, has a folded cerebral cortex and higher white to gray matter ratio, which are common features of the human brain. To further clarify the features of ferret astrocytes, we isolated astrocytes from ferret neonatal brains, cultured these cells, and compared their morphology, gene expression, calcium response, and proliferating ability with those of mouse astrocytes. The morphology of cultured ferret astrocytes differed from that of mouse astrocytes. Ferret astrocytes had longer and more branched processes, smaller cell bodies, and different calcium responses to glutamate, as well as had a greater ability to proliferate, compared to mouse astrocytes. RNA sequencing analysis revealed novel ferret astrocyte-specific genes, including several genes that were the same as those in humans. Astrocytes in the ferret brains had larger cell size, longer primary processes in larger numbers, and a higher proliferation rate compared to mouse astrocytes. Our study shows that cultured ferret astrocytes have different features from rodent astrocytes and similar features to human astrocytes, suggesting that they are useful in studying the roles of astrocytes in brain evolution and cognitive functions in higher animals.

17.
J Biol Chem ; 285(40): 30825-36, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20643652

RESUMEN

Understanding serotonergic (5-HT) signaling is critical for understanding human physiology, behavior, and neuropsychiatric disease. 5-HT mediates its actions via ionotropic and metabotropic 5-HT receptors. The 5-HT(1A) receptor is a metabotropic G protein-coupled receptor linked to the G(i/o) signaling pathway and has been specifically implicated in the pathogenesis of depression and anxiety. To understand and precisely control 5-HT(1A) signaling, we created a light-activated G protein-coupled receptor that targets into 5-HT(1A) receptor domains and substitutes for endogenous 5-HT(1A) receptors. To induce 5-HT(1A)-like targeting, vertebrate rhodopsin was tagged with the C-terminal domain (CT) of 5-HT(1A) (Rh-CT(5-HT1A)). Rh-CT(5-HT1A) activates G protein-coupled inward rectifying K(+) channels in response to light and causes membrane hyperpolarization in hippocampal neurons, similar to the agonist-induced responses of the 5-HT(1A) receptor. The intracellular distribution of Rh-CT(5-HT1A) resembles that of the 5-HT(1A) receptor; Rh-CT(5-HT1A) localizes to somatodendritic sites and is efficiently trafficked to distal dendritic processes. Additionally, neuronal expression of Rh-CT(5-HT1A), but not Rh, decreases 5-HT(1A) agonist sensitivity, suggesting that Rh-CT(5-HT1A) and 5-HT(1A) receptors compete to interact with the same trafficking machinery. Finally, Rh-CT(5-HT1A) is able to rescue 5-HT(1A) signaling of 5-HT(1A) KO mice in cultured neurons and in slices of the dorsal raphe showing that Rh-CT(5-HT1A) is able to functionally compensate for native 5-HT(1A). Thus, as an optogenetic tool, Rh-CT(5-HT1A) has the potential to directly correlate in vivo 5-HT(1A) signaling with 5-HT neuron activity and behavior in both normal animals and animal models of neuropsychiatric disease.


Asunto(s)
Dendritas/metabolismo , Hipocampo/metabolismo , Receptor de Serotonina 5-HT1A/metabolismo , Rodopsina/metabolismo , Animales , Línea Celular , Humanos , Trastornos Mentales/genética , Trastornos Mentales/metabolismo , Ratones , Ratones Noqueados , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/metabolismo , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo , Transporte de Proteínas , Ratas , Receptor de Serotonina 5-HT1A/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Rodopsina/genética
18.
J Pharmacol Sci ; 116(1): 107-15, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21521932

RESUMEN

Hepatic ATP-binding cassette transporter A1 (ABCA1) plays a key role in high-density lipoprotein (HDL) production by apolipoprotein A-I (ApoA-I) lipidation. 3-Hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors, statins, increase ABCA1 mRNA levels in hepatoma cell lines, but their mechanism of action is not yet clear. We investigated how statins increase ABCA1 in rat hepatoma McARH7777 cells. Pitavastatin, atorvastatin, and simvastatin increased total ABCA1 mRNA levels, whereas pravastatin had no effect. Pitavastatin also increased ABCA1 protein. Hepatic ABCA1 expression in rats is regulated by both liver X receptor (LXR) and sterol regulatory element-binding protein (SREBP2) pathways. Pitavastatin repressed peripheral type ABCA1 mRNA levels and its LXR-driven promoter, but activated the liver-type SREBP-driven promoter, and eventually increased total ABCA1 mRNA expression. Furthermore, pitavastatin increased peroxisome proliferator-activated receptor α (PPARα) and its downstream gene expression. Knockdown of PPARα attenuated the increase in ABCA1 protein, indicating that pitavastatin increased ABCA1 protein via PPARα activation, although it repressed LXR activation. Furthermore, the degradation of ABCA1 protein was retarded in pitavastatin-treated cells. These data suggest that pitavastatin increases ABCA1 protein expression by dual mechanisms: SREBP2-mediated mRNA transcription and PPARα-mediated ABCA1 protein stabilization, but not by the PPAR-LXR-ABCA1 pathway. [Supplementary Figures: available only at http://dx.doi.org/10.1254/jphs.10241FP].


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Hígado/efectos de los fármacos , PPAR alfa/metabolismo , Quinolinas/farmacología , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Animales , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Genes Reporteros/efectos de los fármacos , Cinética , Hígado/metabolismo , Neoplasias Hepáticas/metabolismo , Receptores X del Hígado , Receptores Nucleares Huérfanos/metabolismo , PPAR alfa/antagonistas & inhibidores , PPAR alfa/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Ratas , Transducción de Señal/efectos de los fármacos , Proteínas de Unión a los Elementos Reguladores de Esteroles/genética , Activación Transcripcional/efectos de los fármacos
19.
Mol Cell Biol ; 27(12): 4248-60, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17403900

RESUMEN

Cholesterol homeostasis is maintained by coordinate regulation of cholesterol synthesis and its conversion to bile acids in the liver. The excretion of cholesterol from liver and intestine is regulated by ATP-binding cassette half-transporters ABCG5 and ABCG8. The genes for these two proteins are closely linked and divergently transcribed from a common intergenic promoter region. Here, we identified a binding site for hepatocyte nuclear factor 4alpha (HNF4alpha) in the ABCG5/ABCG8 intergenic promoter, through which HNF4alpha strongly activated the expression of a reporter gene in both directions. The HNF4alpha-responsive element is flanked by two conserved GATA boxes that were also required for stimulation by HNF4alpha. GATA4 and GATA6 bind to the GATA boxes, coexpression of GATA4 and HNF4alpha leads to a striking synergistic activation of both the ABCG5 and the ABCG8 promoters, and binding sites for HNF4alpha and GATA were essential for maximal synergism. We also show that HNF4alpha, GATA4, and GATA6 colocalize in the nuclei of HepG2 cells and that a physical interaction between HNF4alpha and GATA4 is critical for the synergistic response. This is the first demonstration that HNF4alpha acts synergistically with GATA factors to activate gene expression in a bidirectional fashion.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Factor de Transcripción GATA4/metabolismo , Factor de Transcripción GATA6/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Lipoproteínas/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 5 , Transportador de Casete de Unión a ATP, Subfamilia G, Miembro 8 , Transportadoras de Casetes de Unión a ATP/genética , Adenoviridae/genética , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular , Línea Celular Tumoral , Secuencia de Consenso , Secuencia Conservada , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA6/genética , Eliminación de Gen , Genes Reporteros , Factor Nuclear 4 del Hepatocito/química , Humanos , Lipoproteínas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Luciferasas/metabolismo , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Unión Proteica , Estructura Terciaria de Proteína , Interferencia de ARN , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico
20.
Neuron ; 45(2): 257-68, 2005 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-15664177

RESUMEN

Endocannabinoids mediate retrograde signal and modulate transmission efficacy at various central synapses. Although endocannabinoid release is induced by either depolarization or activation of G(q/11)-coupled receptors, it is markedly enhanced by the coincidence of depolarization and receptor activation. Here we report that this coincidence is detected by phospholipase Cbeta1 (PLCbeta1) in hippocampal neurons. By measuring cannabinoid-sensitive synaptic currents, we found that the receptor-driven endocannabinoid release was dependent on physiological levels of intracellular Ca(2+) concentration ([Ca(2+)](i)), and markedly enhanced by depolarization-induced [Ca(2+)](i) elevation. Furthermore, we measured PLC activity in intact neurons by using exogenous TRPC6 channel as a biosensor for the PLC product diacylglycerol and found that the receptor-driven PLC activation exhibited similar [Ca(2+)](i) dependence to that of endocannabinoid release. Neither endocannabinoid release nor PLC activation was induced by receptor activation in PLCbeta1 knockout mice. We therefore conclude that PLCbeta1 serves as a coincidence detector through its Ca(2+) dependency for endocannabinoid release in hippocampal neurons.


Asunto(s)
Moduladores de Receptores de Cannabinoides/metabolismo , Endocannabinoides , Hipocampo/metabolismo , Isoenzimas/metabolismo , Neuronas/metabolismo , Transducción de Señal/fisiología , Transmisión Sináptica/fisiología , Fosfolipasas de Tipo C/metabolismo , Animales , Técnicas Biosensibles , Calcio/metabolismo , Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , Células Cultivadas , Diglicéridos/metabolismo , Líquido Intracelular/metabolismo , Isoenzimas/genética , Ratones , Ratones Noqueados , Fosfolipasa C beta , Ratas , Receptores Muscarínicos/metabolismo , Canales Catiónicos TRPC , Canal Catiónico TRPC6 , Fosfolipasas de Tipo C/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA