Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Brain Struct Funct ; 225(5): 1459-1482, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32394093

RESUMEN

Human obesity epidemic is increasing worldwide with major adverse consequences on health. Among other possible causes, the hypothesis of an infectious contribution is worth it to be considered. Here, we report on an animal model of virus-induced obesity which might help to better understand underlying processes in human obesity. Eighty Wistar rats, between 30 and 60 days of age, were intracerebrally inoculated with Borna disease virus (BDV-1), a neurotropic negative-strand RNA virus infecting an unusually broad host spectrum including humans. Half of the rats developed fatal encephalitis, while the other half, after 3-4 months, continuously gained weight. At tripled weights, rats were sacrificed by trans-cardial fixative perfusion. Neuropathology revealed prevailing inflammatory infiltrates in the median eminence (ME), progressive degeneration of neurons of the paraventricular nucleus, the entorhinal cortex and the amygdala, and a strikingly high-grade involution of the hippocampus with hydrocephalus. Immune histology revealed that major BDV-1 antigens were preferentially present at glutamatergic receptor sites, while GABAergic areas remained free from BDV-1. Virus-induced suppression of the glutamatergic system caused GABAergic predominance. In the hypothalamus, this shifted the energy balance to the anabolic appetite-stimulating side governed by GABA, allowing for excessive fat accumulation in obese rats. Furthermore, inflammatory infiltrates in the ME and ventro-medial arcuate nucleus hindered free access of appetite-suppressing hormones leptin and insulin. The hormone transport system in hypothalamic areas outside the ME became blocked by excessively produced leptin, leading to leptin resistance. The resulting hyperleptinemic milieu combined with suppressed glutamatergic mechanisms was a characteristic feature of the found metabolic pathology. In conclusion, the study provided clear evidence that BDV-1 induced obesity in the rat model is the result of interdependent structural and functional metabolic changes. They can be explained by an immunologically induced hypothalamic microcirculation-defect, combined with a disturbance of neurotransmitter regulatory systems. The proposed mechanism may also have implications for human health. BDV-1 infection has been frequently found in depressive patients. Independently, comorbidity between depression and obesity has been reported, either. Future studies should address the exciting question of whether BDV-1 infection could be a link, whatsoever, between these two conditions.


Asunto(s)
Enfermedad de Borna/complicaciones , Virus de la Enfermedad de Borna/fisiología , Encefalitis Viral/patología , Hipotálamo/patología , Hipotálamo/virología , Neuropéptidos/metabolismo , Obesidad/virología , Animales , Enfermedad de Borna/metabolismo , Enfermedad de Borna/patología , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/virología , Hipotálamo/metabolismo , Neuronas/metabolismo , Neuronas/patología , Neuronas/virología , Obesidad/metabolismo , Obesidad/patología , Ratas Wistar
2.
J Neuropathol Exp Neurol ; 78(12): 1160-1170, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31675093

RESUMEN

Japanese encephalitis (JE) is a known CNS viral infection that often involves the thalamus early. To investigate the possible role of sensory peripheral nervous system (PNS) in early neuroinvasion, we developed a left hindlimb footpad-inoculation mouse model to recapitulate human infection by a mosquito bite. A 1-5 days postinfection (dpi) study, demonstrated focal viral antigens/RNA in contralateral thalamic neurons at 3 dpi in 50% of the animals. From 4 to 5 dpi, gradual increase in viral antigens/RNA was observed in bilateral thalami, somatosensory, and piriform cortices, and then the entire CNS. Infection of neuronal bodies and adjacent nerves in dorsal root ganglia (DRGs), trigeminal ganglia, and autonomic ganglia (intestine, etc.) was also observed from 5 dpi. Infection of explant organotypic whole brain slice cultures demonstrated no viral predilection for the thalamus, while DRG and intestinal ganglia organotypic cultures confirmed sensory and autonomic ganglia susceptibility to infection, respectively. Early thalamus and sensory-associated cortex involvement suggest an important role for sensory pathways in neuroinvasion. Our results suggest that JE virus neuronotropism is much more extensive than previously known, and that the sensory PNS and autonomic system are susceptible to infection.


Asunto(s)
Encéfalo/virología , Virus de la Encefalitis Japonesa (Especie)/fisiología , Neuronas/virología , Sistema Nervioso Periférico/virología , Tálamo/virología , Animales , Encéfalo/patología , Células Cultivadas , Infecciones del Sistema Nervioso Central/patología , Infecciones del Sistema Nervioso Central/virología , Modelos Animales de Enfermedad , Virus de la Encefalitis Japonesa (Especie)/aislamiento & purificación , Ratones Endogámicos ICR , Neuronas/patología , Sistema Nervioso Periférico/patología , Corteza Piriforme/patología , Corteza Piriforme/virología , Corteza Somatosensorial/patología , Corteza Somatosensorial/virología , Tálamo/patología
3.
Cell Rep ; 21(2): 517-532, 2017 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-29020636

RESUMEN

The human cerebral cortex possesses distinct structural and functional features that are not found in the lower species traditionally used to model brain development and disease. Accordingly, considerable attention has been placed on the development of methods to direct pluripotent stem cells to form human brain-like structures termed organoids. However, many organoid differentiation protocols are inefficient and display marked variability in their ability to recapitulate the three-dimensional architecture and course of neurogenesis in the developing human brain. Here, we describe optimized organoid culture methods that efficiently and reliably produce cortical and basal ganglia structures similar to those in the human fetal brain in vivo. Neurons within the organoids are functional and exhibit network-like activities. We further demonstrate the utility of this organoid system for modeling the teratogenic effects of Zika virus on the developing brain and identifying more susceptibility receptors and therapeutic compounds that can mitigate its destructive actions.


Asunto(s)
Antirretrovirales/farmacología , Corteza Cerebral/citología , Evaluación Preclínica de Medicamentos/métodos , Organoides/virología , Cultivo Primario de Células/métodos , Virus Zika/efectos de los fármacos , Línea Celular , Corteza Cerebral/virología , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/virología , Humanos , Neuronas/citología , Neuronas/metabolismo , Neuronas/virología , Organoides/citología , Organoides/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Factor de Transcripción STAT3/metabolismo , Tirosina Quinasa c-Mer/metabolismo
4.
J Neurovirol ; 23(2): 205-215, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27739033

RESUMEN

Semliki Forest virus (SFV), a neurotropic virus, has been used to deliver heterologous genes into cells in vitro and in vivo. In this study, we constructed a reporter SFV4-FL-EGFP and found that it can deliver EGFP into neurons located at the injection site without disseminating throughout the brain. Lacking of the capsid gene of SFV4-FL-EGFP does not block its life cycle, while forming replication-competent virus-like particles (VLPs). These VLPs hold subviral genome by using the packaging sequence (PS) located within the nsP2 gene, and can transfer their genome into cells. In addition, we found that the G protein of vesicular stomatitis virus (VSVG) can package SFV subviral genome, which is consistent with the previous reports. The G protein of rabies virus (RVG) could also package SFV subviral genome. These pseudo-typed SFV can deliver EGFP gene into neurons. Taken together, these findings may be used to construct various SFV-based delivery systems for virological studies, gene therapy, and neural circuit labeling.


Asunto(s)
Ingeniería Genética , Terapia Genética/métodos , Vectores Genéticos/metabolismo , Hipotálamo/virología , Neuronas/virología , Virus de los Bosques Semliki/genética , Animales , Línea Celular , Cricetulus , Células Epiteliales/ultraestructura , Células Epiteliales/virología , Expresión Génica , Genes Reporteros , Vectores Genéticos/química , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipotálamo/ultraestructura , Inyecciones Intraventriculares , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas/ultraestructura , Cultivo Primario de Células , Virus de la Rabia/genética , Virus de la Rabia/metabolismo , Virus de los Bosques Semliki/metabolismo , Vesiculovirus/genética , Vesiculovirus/metabolismo , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Virión/genética , Virión/metabolismo , Ensamble de Virus/genética
5.
Cell Cycle ; 15(24): 3482-3489, 2016 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-27830988

RESUMEN

The cell cycle-associated neuronal death hypothesis, which has been proposed as a common mechanism for most neurodegenerative diseases, is notably supported by evidencing cell cycle effectors in neurons. However, in naturally occurring nervous system diseases, these markers are not expressed in neuron nuclei but in cytoplasmic compartments. In other respects, the Feline Panleukopenia Virus (FPV) is able to complete its cycle in mature brain neurons in the feline species. As a parvovirus, the FPV is strictly dependent on its host cell reaching the cell cycle S phase to start its multiplication. In this retrospective study on the whole brain of 12 cats with naturally-occurring, FPV-associated cerebellar atrophy, VP2 capsid protein expression was detected by immunostaining not only in some brain neuronal nuclei but also in neuronal cytoplasm in 2 cats, suggesting that viral mRNA translation was still occurring. In these cats, double immunostainings demonstrated the expression of cell cycle S phase markers cyclin A, cdk2 and PCNA in neuronal nuclei. Parvoviruses are able to maintain their host cells in S phase by triggering the DNA damage response. S139 phospho H2A1, a key player in the cell cycle arrest, was detected in some neuronal nuclei, supporting that infected neurons were also blocked into the S phase. PCR studies did not support a co-infection with an adeno or herpes virus. ERK1/2 nuclear accumulation was observed in some neurons suggesting that the ERK signaling pathway might be involved as a mechanism driving these neurons far into the cell cycle.


Asunto(s)
Biomarcadores/metabolismo , Núcleo Celular/metabolismo , Cerebro/patología , Virus de la Panleucopenia Felina/fisiología , Panleucopenia Felina/patología , Panleucopenia Felina/virología , Neuronas/patología , Fase S , Animales , Anticuerpos Antivirales/metabolismo , Especificidad de Anticuerpos , Emparejamiento Base , Proteínas de la Cápside/metabolismo , Gatos , Núcleo Celular/enzimología , ADN Viral/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Virus de la Panleucopenia Felina/genética , Femenino , Genes Virales , Células HEK293 , Humanos , Inmunohistoquímica , Masculino , Neuronas/virología , Reacción en Cadena de la Polimerasa , Reproducibilidad de los Resultados , Tálamo/metabolismo
6.
Virus Res ; 205: 63-72, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26031763

RESUMEN

Herpes simplex virus type-1 (HSV-1) is ubiquitous and is able to establish a lifelong persistent latent infection in neurons of infected individuals. It has been estimated that in approximately 70% of the population over 50 years old, the virus enters the brain and infects neurons, and possibly undergoes recurrent reactivation episodes during lifetime, especially in immunodepressed individuals. We previously showed that the sensors AMP-dependent kinase (AMPK) and Sirtuin 1 (Sirt1), involved in survival pathways and neuroprotection, were affected during the course of HSV-1 infection. To evaluate if natural activators of the AMPK/Sirt1 axis, such as Resveratrol and Quercetin could reduce viral propagation and/or counteract the effects of neuronal infection, we analyzed progeny virion production, neuronal viability and neurodegenerative events during HSV-1 infection. We found that the activators of AMPK/Sirt1 axis, increased the viability of infected neurons, significantly reduced the viral titer in the supernatant and the expression of viral genes. More importantly, pretreatment of neurons with Resveratrol or Quercetin significantly reduced the levels of caspase-3 cleaved- and hyperphosphorylated tau associated with HSV-1 infection. These results suggest that activators of the AMPK/Sirt1 axis could be potentially useful in reducing the risk of HSV-1 productive infection in neurons and the cellular damage associated with reactivation episodes.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Suplementos Dietéticos/análisis , Activadores de Enzimas/farmacología , Herpes Simple/enzimología , Herpesvirus Humano 1/fisiología , Neuronas/virología , Sirtuina 1/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Herpes Simple/genética , Herpes Simple/virología , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/genética , Humanos , Neuronas/efectos de los fármacos , Quercetina/farmacología , Resveratrol , Sirtuina 1/genética , Estilbenos/farmacología
7.
PLoS One ; 9(11): e112818, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25415340

RESUMEN

Alzheimer's disease (AD) is characterized by progressive dysfunction of memory and higher cognitive functions with abnormal accumulation of extracellular amyloid plaques and intracellular neurofibrillary tangles throughout cortical and limbic brain regions. Withania somnifera (WS) also known as 'ashwagandha' (ASH) is used widely in Ayurvedic medicine as a nerve tonic and memory enhancer. However, there is paucity of data on potential neuroprotective effects of ASH against ß-Amyloid (1-42) (Aß) induced neuropathogenesis. In the present study, we have tested the neuroprotective effects of Methanol: Chloroform (3:1) extract of ASH and its constituent Withanolide A (WA) against Aß induced toxicity, HIV-1(Ba-L) (clade B) infection and the effects of drugs of abuse using a human neuronal SK-N-MC cell line. Aß when tested individually, induced cytotoxic effects in SK-N-MC cells as shown by increased trypan blue stained cells. However, when ASH was added to Aß treated cells the toxic effects were neutralized. This observation was supported by cellular localization of Aß, MTT formazan exocytosis, and the levels of acetylcholinesterase activity, confirming the chemopreventive or protective effects of ASH against Aß induced toxicity. Further, the levels of MAP2 were significantly increased in cells infected with HIV-1(Ba-L) (clade B) as well as in cells treated with Cocaine (COC) and Methamphetamine (METH) compared with control cells. In ASH treated cells the MAP2 levels were significantly less compared to controls. Similar results were observed in combination experiments. Also, WA, a purified constituent of ASH, showed same pattern using MTT assay as a parameter. These results suggests that neuroprotective properties of ASH observed in the present study may provide some explanation for the ethnopharmacological uses of ASH in traditional medicine for cognitive and other HIV associated neurodegenerative disorders and further ASH could be a potential novel drug to reduce the brain amyloid burden and/or improve the HIV-1 associated neurocognitive impairments.


Asunto(s)
Péptidos beta-Amiloides/fisiología , VIH-1/fisiología , Drogas Ilícitas/toxicidad , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/fisiología , Witanólidos/farmacología , Línea Celular , Humanos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/virología , Withania/química
8.
J Neurosci ; 33(8): 3624-32, 2013 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-23426689

RESUMEN

POMC-derived melanocortins inhibit food intake. In the adult rodent brain, POMC-expressing neurons are located in the arcuate nucleus (ARC) and the nucleus tractus solitarius (NTS), but it remains unclear how POMC neurons in these two brain nuclei regulate feeding behavior and metabolism differentially. Using pharmacogenetic methods to activate or deplete neuron groups in separate brain areas, in the present study, we show that POMC neurons in the ARC and NTS suppress feeding behavior at different time scales. Neurons were activated using the DREADD (designer receptors exclusively activated by designer drugs) method. The evolved human M3-muscarinic receptor was expressed in a selective population of POMC neurons by stereotaxic infusion of Cre-recombinase-dependent, adeno-associated virus vectors into the ARC or NTS of POMC-Cre mice. After injection of the human M3-muscarinic receptor ligand clozapine-N-oxide (1 mg/kg, i.p.), acute activation of NTS POMC neurons produced an immediate inhibition of feeding behavior. In contrast, chronic stimulation was required for ARC POMC neurons to suppress food intake. Using adeno-associated virus delivery of the diphtheria toxin receptor gene, we found that diphtheria toxin-induced ablation of POMC neurons in the ARC but not the NTS, increased food intake, reduced energy expenditure, and ultimately resulted in obesity and metabolic and endocrine disorders. Our results reveal different behavioral functions of POMC neurons in the ARC and NTS, suggesting that POMC neurons regulate feeding and energy homeostasis by integrating long-term adiposity signals from the hypothalamus and short-term satiety signals from the brainstem.


Asunto(s)
Tronco Encefálico/fisiología , Conducta Alimentaria/fisiología , Hipotálamo/fisiología , Inhibición Neural/fisiología , Neuronas/fisiología , Proopiomelanocortina/fisiología , Adiposidad/genética , Animales , Tronco Encefálico/virología , Dependovirus/genética , Femenino , Vectores Genéticos/administración & dosificación , Células HEK293 , Homeostasis/genética , Humanos , Masculino , Ratones , Ratones Transgénicos , Inhibición Neural/genética , Vías Nerviosas/fisiopatología , Neuronas/virología , Proopiomelanocortina/antagonistas & inhibidores
9.
PLoS One ; 7(5): e37540, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22629415

RESUMEN

BACKGROUND: HIV-1 viral protein Tat partially mediates the neural dysfunction and neuronal cell death associated with HIV-1 induced neurodegeneration and neurocognitive disorders. Soy isoflavones provide protection against various neurotoxic insults to maintain neuronal function and thus help preserve neurocognitive capacity. METHODOLOGY/PRINCIPAL FINDINGS: We demonstrate in primary cortical cell cultures that 17ß-estradiol or isoflavones (genistein or daidzein) attenuate Tat(1-86)-induced expression of apoptotic proteins and subsequent cell death. Exposure of cultured neurons to the estrogen receptor antagonist ICI 182,780 abolished the anti-apoptotic actions of isoflavones. Use of ERα or ERß specific antagonists determined the involvement of both ER isoforms in genistein and daidzein inhibition of caspase activity; ERß selectively mediated downregulation of mitochondrial pro-apoptotic protein Bax. The findings suggest soy isoflavones effectively diminished HIV-1 Tat-induced apoptotic signaling. CONCLUSIONS/SIGNIFICANCE: Collectively, our results suggest that soy isoflavones represent an adjunctive therapeutic option with combination anti-retroviral therapy (cART) to preserve neuronal functioning and sustain neurocognitive abilities of HIV-1 infected persons.


Asunto(s)
Apoptosis/efectos de los fármacos , Genisteína/farmacología , VIH-1 , Isoflavonas/farmacología , Neuronas/efectos de los fármacos , Fitoestrógenos/farmacología , Receptores de Estrógenos/metabolismo , Animales , Regulación hacia Abajo/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Neuronas/metabolismo , Neuronas/virología , Fragmentos de Péptidos/farmacología , Ratas , Ratas Sprague-Dawley , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/farmacología
10.
Am J Pathol ; 176(6): 2776-84, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20382699

RESUMEN

The molecular mechanisms underlying learning and memory impairment in patients with HIV-associated neurological disease have remained unclear. Calcium/calmodulin-dependent kinase II (CaMKII) has key roles in synaptic potentiation and memory storage in neurons and also may have immunomodulatory functions. To determine whether HIV and simian immunodeficiency virus (SIV) induce alterations in CaMKII expression and/or activation (autophosphorylation) in the brain, we measured CaMKII alterations by quantitative immunoblotting in both an in vitro HIV/neuronal culture model and in vivo in an SIV-infected macaque model of HIV-associated neurological damage. Using primary rat hippocampal neuronal cultures treated with culture supernatants harvested from HIV-1-infected human monocyte-derived macrophages (HIV/MDM), we found that CaMKII activation declined after exposure of neurons to HIV/MDM. Consistent with our in vitro measurements, a significant decrease in CaMKII activation was present in both the hippocampus and frontal cortex of SIV-infected macaques compared with uninfected animals. In SIV-infected animals, total CaMKII expression in the hippocampus correlated well with levels of synaptophysin. Furthermore, CaMKII expression in both the hippocampus and frontal cortex was inversely correlated with viral load in the brain. These findings suggest that alterations in CaMKII may compromise synaptic function in the early phases of chronic neurodegenerative processes induced by HIV.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Lóbulo Frontal/enzimología , Lóbulo Frontal/virología , VIH/metabolismo , Hipocampo/enzimología , Hipocampo/virología , Virus de la Inmunodeficiencia de los Simios/metabolismo , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Células Cultivadas , Activación Enzimática , Lóbulo Frontal/citología , Lóbulo Frontal/patología , Infecciones por VIH/metabolismo , Hipocampo/citología , Hipocampo/patología , Humanos , Macaca mulatta/metabolismo , Macaca mulatta/virología , Neuronas/metabolismo , Neuronas/virología , Ratas , Síndrome de Inmunodeficiencia Adquirida del Simio/metabolismo , Sinapsis/metabolismo , Sinaptofisina/metabolismo , Carga Viral
11.
Brain Res ; 1268: 135-141, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19269279

RESUMEN

The clinical picture of viral encephalitis is determined by the affinity and persistence of the virus to different brain regions. Therefore, the present study was aimed to investigate the neuropathological changes following Japanese encephalitis virus (JEV) infection in rat at different time points. Twelve days old Wistar rats were infected by intracerebral inoculation of JEV. Presence of JEV antigen was detected in thalamus, striatum, cortex and mid brain on 3, 6, 10 and 20 days post inoculation (d.p.i.). Histopathological changes were also studied in different brain regions at different time points. The highest expression of JEV antigen was found on 6 dpi in all the brain regions studied. JEV antigen was maximum in thalamus on 6 d.p.i. and mid brain on 10 d.p.i. JEV antigen, however, was almost undetectable on 20 d.p.i. in all the regions. The classical pathological changes such as cellular infiltration, perivascular cuffing, meningeal disruption, neuronal damage, neuronal shrinkage, and plaque formation were observed up to 10 d.p.i. The present study reveals high affinity of JEV to thalamus, brainstem and striatum. Rat model of JEV infection may serve as a useful model for studying mechanism of cell injury and recovery in JE.


Asunto(s)
Encéfalo/patología , Encéfalo/virología , Virus de la Encefalitis Japonesa (Especie)/aislamiento & purificación , Encefalitis Japonesa/patología , Encefalitis Japonesa/virología , Análisis de Varianza , Animales , Animales Lactantes , Corteza Cerebral/patología , Corteza Cerebral/virología , Cuerpo Estriado/patología , Cuerpo Estriado/virología , Femenino , Inmunohistoquímica , Masculino , Mesencéfalo/patología , Mesencéfalo/virología , Neuronas/patología , Neuronas/virología , Ratas , Ratas Wistar , Tálamo/patología , Tálamo/virología
12.
Brain Struct Funct ; 213(1-2): 239-45, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18247051

RESUMEN

Although it has been reported by several laboratories that vestibular stress activates the hypothalamo-pituitary-adrenocortical axis (HPA), the existence of neuronal connections between vestibular and hypothalamic paraventricular neurons has not yet been demonstrated. By the use of a virus-based retrograde trans-synaptic tracing technique in the rat, here we demonstrate vestibular projections to the paraventricular nucleus (PVN). Pseudorabies virus (Bartha strain, type BDR62) was injected into the PVN, and the progression of the infection along synaptically connected neurons was followed in the pons and the medulla, 3 and 4 days post-inoculation. Virus-infected neurons were revealed mainly in the medial vestibular nucleus. Labeled cells were scattered in the spinal, and very rarely in the superior nuclei, but none of them in the lateral vestibular nucleus. Injections of cholera toxin B subunit, a monosynaptic retrograde tracer into the PVN failed to label any cells in the vestibular nuclei. These results provide anatomical evidence for the existence of a vestibulo-paraventricular polysynaptic pathway and support the view that the HPA axis is modulated by vestibular stress.


Asunto(s)
Hipotálamo/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Estrés Fisiológico/fisiología , Núcleos Vestibulares/metabolismo , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/metabolismo , Animales , Transporte Biológico/fisiología , Toxina del Cólera/administración & dosificación , Toxina del Cólera/metabolismo , Herpesvirus Suido 1/fisiología , Hipotálamo/patología , Inmunohistoquímica , Masculino , Bulbo Raquídeo/metabolismo , Bulbo Raquídeo/patología , Bulbo Raquídeo/virología , Microinyecciones , Vías Nerviosas/metabolismo , Vías Nerviosas/patología , Vías Nerviosas/virología , Neuronas/metabolismo , Neuronas/patología , Neuronas/virología , Neuronas Eferentes/metabolismo , Neuronas Eferentes/patología , Neuronas Eferentes/virología , Núcleo Hipotalámico Paraventricular/patología , Núcleo Hipotalámico Paraventricular/virología , Puente/metabolismo , Puente/patología , Puente/virología , Seudorrabia/fisiopatología , Seudorrabia/virología , Ratas , Ratas Sprague-Dawley , Médula Espinal/metabolismo , Médula Espinal/patología , Médula Espinal/virología , Núcleos Vestibulares/patología , Núcleos Vestibulares/virología
13.
J Mol Biol ; 375(5): 1222-33, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18082765

RESUMEN

Prion infections induce severe disruption of the central nervous system with neuronal vacuolation and extensive glial reactions, and invariably lead to death of affected individuals. The molecular underpinnings of these events are not well understood. To better define the molecular consequences of prion infections, we analyzed the transcriptional response to persistent prion infection in a panel of three murine neural cell lines in vitro. Colony spot immunochemistry assays indicated that 65-100% of cells were infected in each line. Only the Nav1 gene was marginally modulated in one cell line, whereas transcripts previously reported to be derailed in prion-infected cells were not confirmed in the present study. We attribute these discrepancies to the experimental stringency of the current study, which was performed under conditions designed to minimize potential genetic drifts. These findings are at striking variance with gene expression studies performed on whole brains upon prion infections in vivo, suggesting that many of the latter changes represent secondary reactions to infection. We conclude that, surprisingly, there are no universal transcriptional changes induced by prion infection of neural cells in vitro.


Asunto(s)
Enfermedades por Prión/virología , Priones/genética , Transcripción Genética , Animales , Western Blotting , Técnicas de Cultivo de Célula , Línea Celular , Células Cultivadas , Perfilación de la Expresión Génica , Hipotálamo/citología , Inmunohistoquímica , Ratones , Neuroblastoma/patología , Neuronas/metabolismo , Neuronas/virología , Hibridación de Ácido Nucleico , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas PrPSc/genética , Proteínas PrPSc/metabolismo , Priones/patogenicidad , ARN Complementario/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Neuron ; 50(1): 75-88, 2006 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-16600857

RESUMEN

Activity-dependent synaptic delivery of GluR1-, GluR2L-, and GluR4-containing AMPA receptors (-Rs) and removal of GluR2-containing AMPA-Rs mediate synaptic potentiation and depression, respectively. The obvious puzzle is how synapses maintain the capacity for bidirectional plasticity if different AMPA-Rs are utilized for potentiation and depression. Here, we show that synaptic AMPA-R exchange is essential for maintaining the capacity for bidirectional plasticity. The exchange process consists of activity-independent synaptic removal of GluR1-, GluR2L-, or GluR4-containing AMPA-Rs and refilling with GluR2-containing AMPA-Rs at hippocampal and cortical synapses in vitro and in intact brains. In GluR1 and GluR2 knockout mice, initiation or completion of synaptic AMPA-R exchange is compromised, respectively. The complementary AMPA-R removal and refilling events in the exchange process ultimately maintain synaptic strength unchanged, but their long rate time constants ( approximately 15-18 hr) render transmission temporarily depressed in the middle of the exchange. These results suggest that the previously hypothesized "slot" proteins, rather than AMPA-Rs, code and maintain transmission efficacy at central synapses.


Asunto(s)
Encéfalo/citología , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Receptores AMPA/fisiología , Sinapsis/metabolismo , Anestésicos Locales/farmacología , Animales , Animales Recién Nacidos , Estimulación Eléctrica/métodos , Proteínas Fluorescentes Verdes/metabolismo , Técnicas In Vitro , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Potenciación a Largo Plazo/efectos de la radiación , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Depresión Sináptica a Largo Plazo/fisiología , Depresión Sináptica a Largo Plazo/efectos de la radiación , Magnesio/farmacología , Ratones , Ratones Noqueados , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/efectos de la radiación , Neuronas/efectos de los fármacos , Neuronas/efectos de la radiación , Neuronas/virología , Técnicas de Placa-Clamp/métodos , Ratas , Receptores AMPA/clasificación , Receptores AMPA/deficiencia , Receptores AMPA/genética , Sinapsis/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Transmisión Sináptica/efectos de la radiación , Tetrodotoxina/farmacología , Factores de Tiempo , Transfección/métodos , Vibrisas/inervación , Vibrisas/fisiología
15.
Cell ; 123(4): 669-82, 2005 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-16290037

RESUMEN

In order to gain insight into sensory processing modulating reproductive behavioral and endocrine changes, we have aimed at identifying afferent pathways to neurons synthesizing luteinizing hormone-releasing hormone (LHRH, also known as gonadotropin-releasing hormone [GnRH]), a key neurohormone of reproduction. Injection of conditional pseudorabies virus into the brain of an LHRH::CRE mouse line led to the identification of neuronal networks connected to LHRH neurons. Remarkably, and in contrast to established notions on the nature of LHRH neuronal inputs, our data identify major olfactory projection pathways originating from a discrete population of olfactory sensory neurons but fail to document any synaptic connectivity with the vomeronasal system. Accordingly, chemosensory modulation of LHRH neuronal activity and mating behavior are dramatically impaired in absence of olfactory function, while they appear unaffected in mouse mutants lacking vomeronasal signaling. Further visualization of afferents to LHRH neurons across the brain offers a unique opportunity to uncover complex polysynaptic circuits modulating reproduction and fertility.


Asunto(s)
Fertilidad/fisiología , Hipotálamo/fisiología , Vías Olfatorias/fisiología , Reproducción/fisiología , Animales , Encéfalo/citología , Encéfalo/fisiología , Encéfalo/virología , Canales Catiónicos Regulados por Nucleótidos Cíclicos , Femenino , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Liberadora de Gonadotropina/fisiología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Herpesvirus Suido 1/genética , Hipotálamo/citología , Hipotálamo/virología , Integrasas/genética , Integrasas/metabolismo , Canales Iónicos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Vías Nerviosas/citología , Vías Nerviosas/fisiología , Vías Nerviosas/virología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/virología , Nitrilos/farmacología , Bulbo Olfatorio/citología , Bulbo Olfatorio/fisiología , Bulbo Olfatorio/virología , Mucosa Olfatoria/efectos de los fármacos , Mucosa Olfatoria/inervación , Mucosa Olfatoria/fisiología , Vías Olfatorias/citología , Vías Olfatorias/virología , Fosforilación , Área Preóptica/fisiología , Área Preóptica/virología , Tabique del Cerebro/fisiología , Tabique del Cerebro/virología , Atractivos Sexuales/farmacología , Conducta Sexual Animal/efectos de los fármacos , Conducta Sexual Animal/fisiología , Canales Catiónicos TRPC/genética , Timidina Quinasa/genética , Órgano Vomeronasal/inervación , Órgano Vomeronasal/fisiología
16.
Science ; 308(5718): 83-8, 2005 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15746389

RESUMEN

To elucidate molecular, cellular, and circuit changes that occur in the brain during learning, we investigated the role of a glutamate receptor subtype in fear conditioning. In this form of learning, animals associate two stimuli, such as a tone and a shock. Here we report that fear conditioning drives AMPA-type glutamate receptors into the synapse of a large fraction of postsynaptic neurons in the lateral amygdala, a brain structure essential for this learning process. Furthermore, memory was reduced if AMPA receptor synaptic incorporation was blocked in as few as 10 to 20% of lateral amygdala neurons. Thus, the encoding of memories in the lateral amygdala is mediated by AMPA receptor trafficking, is widely distributed, and displays little redundancy.


Asunto(s)
Amígdala del Cerebelo/fisiología , Aprendizaje por Asociación/fisiología , Memoria/fisiología , Plasticidad Neuronal , Neuronas/fisiología , Receptores AMPA/metabolismo , Sinapsis/fisiología , Amígdala del Cerebelo/citología , Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/virología , Animales , Condicionamiento Psicológico , Electrofisiología , Miedo , Femenino , Vectores Genéticos , Proteínas Fluorescentes Verdes/metabolismo , Potenciación a Largo Plazo , Masculino , Vías Nerviosas/fisiología , Neuronas/metabolismo , Neuronas/virología , Técnicas de Placa-Clamp , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/metabolismo , Simplexvirus/genética , Sinapsis/metabolismo , Transmisión Sináptica , Tálamo/fisiología
17.
Neuroreport ; 15(10): 1663-7, 2004 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-15232303

RESUMEN

In scrapie-infected cells, the abnormal isoform of the prion protein, PrP(Sc), accumulates in endosomes/lysosomes. In this study, the involvement of two lysosomal proteases, cathepsin B and L, in cellular processing of PrP(Sc) was analyzed in immortalized neuronal gonadotropin-releasing hormone cells (GT1-1) infected with scrapie. Treatment with inhibitors of either cathepsin B or L resulted in accumulation of PrP(Sc). Such an increased accumulation also occurred when the activities of both cathepsins were inhibited using RNA interference. We conclude that cathepsin B and L are involved in the degradation of PrP(Sc) in scrapie-infected GT1-1 cells and that they can compensate for each other's functions. This study shows that specific proteases, abundantly present in neurons, have the capacity to degrade PrP(Sc).


Asunto(s)
Catepsina B/metabolismo , Catepsinas/metabolismo , Neuronas/metabolismo , Priones/metabolismo , Animales , Western Blotting/métodos , Catepsina B/antagonistas & inhibidores , Catepsina B/genética , Catepsina L , Catepsinas/antagonistas & inhibidores , Catepsinas/genética , Línea Celular , Cisteína Endopeptidasas , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo , Ratones , Neuronas/virología , Proteínas PrPSc/metabolismo , ARN Mensajero/biosíntesis , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Transfección/métodos
18.
J Neurosci ; 23(23): 8432-44, 2003 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-12968006

RESUMEN

We used transneuronal transport of neurotropic viruses to examine the topographic organization of circuits linking the cerebellar cortex with the arm area of the primary motor cortex (M1) and with area 46 in dorsolateral prefrontal cortex of monkeys. Retrograde transneuronal transport of the CVS-11 (challenge virus strain 11) strain of rabies virus in cerebello-thalamocortical pathways revealed that the arm area of M1 receives input from Purkinje cells located primarily in lobules IV-VI of the cerebellar cortex. In contrast, transneuronal transport of rabies from area 46 revealed that it receives input from Purkinje cells located primarily in Crus II of the ansiform lobule. Thus, both M1 and area 46 are the targets of output from the cerebellar cortex. However, the output to each area of the cerebral cortex originates from Purkinje cells in different regions of the cerebellar cortex. Anterograde transneuronal transport of the H129 strain of herpes simplex virus type 1 (HSV1) revealed that neurons in the arm area of M1 project via the pons to granule cells primarily in lobules IV-VI, whereas neurons in area 46 project to granule cells primarily in Crus II. Together, the findings from rabies and HSV1 experiments indicate that the regions of the cerebellar cortex that receive input from M1 are the same as those that project to M1. Similarly, the regions of the cerebellar cortex that receive input from area 46 are the same as those that project to area 46. Thus, our observations suggest that multiple closed-loop circuits represent a fundamental architectural feature of cerebrocerebellar interactions.


Asunto(s)
Cebus , Cerebelo/citología , Corteza Motora/citología , Corteza Prefrontal/citología , Animales , Transporte Biológico , Mapeo Encefálico/métodos , Núcleos Cerebelosos/citología , Núcleos Cerebelosos/virología , Cerebelo/virología , Herpesvirus Humano 1/fisiología , Corteza Motora/virología , Vías Nerviosas/citología , Vías Nerviosas/virología , Neuronas/citología , Neuronas/virología , Corteza Prefrontal/virología , Células de Purkinje/citología , Células de Purkinje/virología , Virus de la Rabia/fisiología , Tálamo/citología , Tálamo/virología , Factores de Tiempo
19.
J Neurosci Methods ; 127(2): 179-92, 2003 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12906947

RESUMEN

Analysis of gene regulatory sequences in primary cultures of neurons has been hampered by inefficient transfection of post-mitotic neurons with reporter plasmids. We describe detailed conditions that allowed a significant improvement of transfection efficiency in primary cultures of serum-supplemented rat fetal hypothalamic cells. Transfected cells expressed the green fluorescent protein (GFP) under the control of the strong but non-cell-specific cytomegalo virus (CMV) promoter or under the thyrotropin-releasing hormone (TRH) promoter, to direct expression only in TRH neurons. Using the CMV promoter-GFP plasmid, we tested several commercially available transfection reagents; the best results were obtained with polyethylenimine (PEI) and Lipofectamine 2000. We optimized the transfection procedure with PEI because it rendered more reproducible results. Transfection with PEI was optimal when cells were transfected at a cellular density of 2.9 x 10(6) cells in 35-mm dishes, with 10 microg of DNA, a PEI/DNA ratio of 8.8 and PEI pH of 6.9. Using these conditions, we were able to detect GFP positive neurons after transfecting the TRH promoter-GFP plasmid. GFP positive cells were successfully purified by FACS. This opens the possibility to use transfection of mammalian CNS post-mitotic neurons for new applications including the purification of specific neuronal subtypes.


Asunto(s)
Hipotálamo/citología , Neuronas/efectos de los fármacos , Polietileneimina/farmacología , Transfección/métodos , Células 3T3 , Animales , Recuento de Células , Tamaño de la Célula , Supervivencia Celular , Células Cultivadas , Citomegalovirus/metabolismo , Embrión de Mamíferos , Femenino , Citometría de Flujo/instrumentación , Citometría de Flujo/métodos , Proteínas Fluorescentes Verdes , Hipotálamo/virología , Hibridación in Situ/métodos , Luciferasas/metabolismo , Proteínas Luminiscentes/metabolismo , Ratones , Neuronas/fisiología , Neuronas/virología , Embarazo , Regiones Promotoras Genéticas , ARN Mensajero/biosíntesis , Ratas , Ratas Wistar , Hormona Liberadora de Tirotropina/metabolismo , Factores de Tiempo
20.
J Neuropathol Exp Neurol ; 62(6): 617-26, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12834106

RESUMEN

Recent in vitro studies suggest that the alpha chemokine stromal-derived factor-1alpha (SDF-1alpha) and its receptor CXCR-4 may contribute to neuronal apoptosis in HIV infection of the brain. The cellular and regional expression of this chemokine and its relationship to the AIDS dementia complex (ADC), however, have remained undetermined. Using immunohistochemistry and semiquantitative RT-PCR, we examined the expression of SDF-1alpha in the frontal cortex (FC), the adjacent deep white matter (DWM). and the basal ganglia (BG) of 17 patients with ADC and 5 normal controls, and the FC and temporal cortex of 6 patients with Alzheimer disease (AD). Additionally, SDF-1alpha expression was studied in 3 different neuronal cultures: differentiated SK-N-MC cells, primary human fetal neuronal, and mouse hippocampal cultures. SDF-1alpha staining was predominantly localized to astrocytes in all 3 groups in the gray matter of the FC and the BG, often in the vicinity of cortical and basal ganglia neurons, but was generally absent in the DWM. Further, the number of positive neurons was significantly greater in the BG of AIDS subjects with advanced brain disease compared to subjects with lesser disease (p = 0.029). All cultures showed prominent SDF-1alpha staining of neurons within the cytoplasm and in neurites, whereas preferential expression in GABA-ergic neurons was found in hippocampal cultures. This is the first study to show that SDF-1alpha is constitutively expressed in astrocytes of the deep and cortical gray matter as well as in neurons of the human brain. Its increased expression in basal ganglia neurons of patients with advanced HIV CNS disease suggests it may also contribute to pathogenesis.


Asunto(s)
Complejo SIDA Demencia/metabolismo , Astrocitos/metabolismo , Quimiocinas CXC/metabolismo , Neuronas/metabolismo , Complejo SIDA Demencia/genética , Complejo SIDA Demencia/patología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Astrocitos/patología , Astrocitos/virología , Ganglios Basales/metabolismo , Ganglios Basales/patología , Ganglios Basales/virología , Northern Blotting/métodos , Southern Blotting/métodos , Recuento de Células/métodos , Células Cultivadas/virología , Quimiocina CXCL12 , Quimiocinas CXC/genética , Feto , Lóbulo Frontal/metabolismo , Lóbulo Frontal/patología , Lóbulo Frontal/virología , Regulación de la Expresión Génica , Productos del Gen tat/metabolismo , VIH/patogenicidad , Proteína gp120 de Envoltorio del VIH/metabolismo , Compuestos Heterocíclicos con 1 Anillo/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/virología , Humanos , Inmunohistoquímica/métodos , Técnicas In Vitro , Ratones , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Neuronas/patología , Neuronas/virología , ARN Mensajero/biosíntesis , Proteínas Recombinantes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA