Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Neurosci ; 41(20): 4367-4377, 2021 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-33827934

RESUMEN

Early-life inflammatory stress increases seizure susceptibility later in life. However, possible sex- and age-specific differences and the associated mechanisms are largely unknown. C57BL/6 mice were bred in house, and female and male pups were injected with lipopolysaccharide (LPS; 100 µg/kg, i.p.) or vehicle control (saline solution) at postnatal day 14 (P14). Seizure threshold was assessed in response to pentylenetetrazol (1% solution, i.v.) in adolescence (∼P40) and adulthood (∼P60). We found that adult, but not adolescent, mice treated with LPS displayed ∼34% lower seizure threshold compared with controls. Females and males showed similar increased seizure susceptibility, suggesting that altered brain excitability was age dependent, but not sex dependent. Whole-cell recordings revealed no differences in excitatory synaptic activity onto CA1 pyramidal neurons from control or neonatally inflamed adolescent mice of either sex. However, adult mice of both sexes previously exposed to LPS displayed spontaneous EPSC frequency approximately twice that of controls, but amplitude was unchanged. Although these changes were not associated with alterations in dendritic spines or in the NMDA/AMPA receptor ratio, they were linked to an increased glutamate release probability from Schaffer collateral, but not temporoammonic pathway. This glutamate increase was associated with reduced activity of presynaptic GABAB receptors and was independent of the endocannabinoid-mediated suppression of excitation. Our new findings demonstrate that early-life inflammation leads to long-term increased hippocampal excitability in adult female and male mice associated with changes in glutamatergic synaptic transmission. These alterations may contribute to enhanced vulnerability of the brain to subsequent pathologic challenges such as epileptic seizures.SIGNIFICANCE STATEMENT Adult physiology has been shown to be affected by early-life inflammation. Our data reveal that early-life inflammation increases excitatory synaptic transmission onto hippocampal CA1 pyramidal neurons in an age-dependent manner through disrupted presynaptic GABAB receptor activity on Schaffer collaterals. This hyperexcitability was seen only in adult, and not in adolescent, animals of either sex. The data suggest a maturation process, independent of sex, in the priming action of early-life inflammation and highlight the importance of studying mature brains to reveal cellular changes associated with early-life interventions.


Asunto(s)
Inflamación/fisiopatología , Células Piramidales/fisiología , Convulsiones/fisiopatología , Transmisión Sináptica/fisiología , Animales , Convulsivantes/toxicidad , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/fisiopatología , Inflamación/inducido químicamente , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Pentilenotetrazol/toxicidad , Convulsiones/inducido químicamente
2.
J Neurosci ; 39(37): 7244-7259, 2019 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-31308096

RESUMEN

Early life, systemic inflammation causes long-lasting changes in behavior. To unmask possible mechanisms associated with this phenomenon, we asked whether the intrinsic membrane properties in hippocampal neurons were altered as a consequence of early life inflammation. C57BL/6 mice were bred in-house and both male and female pups from multiple litters were injected with lipopolysaccharide (LPS; 100 µg/kg, i.p.) or vehicle at postnatal day (P)14, and kept until adolescence (P35-P45) or adulthood (P60-P70), when brain slices were prepared for whole-cell and perforated-patch recordings from CA1 hippocampal pyramidal neurons. In neurons of adult male mice pretreated with LPS, the number of action potentials elicited by depolarizing current pulses was significantly increased compared with control neurons, concomitant with increased input resistance, and a lower action potential threshold. Although these changes were not associated with changes in relevant sodium channel expression or differences in capacitance or dendritic architecture, they were linked to a mechanism involving intracellular chloride overload, revealed through a depolarized GABA reversal potential and increased expression of the chloride transporter, NKCC1. In contrast, no significant changes were observed in neurons of adult female mice pretreated with LPS, nor in adolescent mice of either sex. These data uncover a potential mechanism involving neonatal inflammation-induced plasticity in chloride homeostasis, which may contribute to early life inflammation-induced behavioral alterations.SIGNIFICANCE STATEMENT Early life inflammation results in long-lasting changes in many aspects of adult physiology. In this paper we reveal that a brief exposure to early life peripheral inflammation with LPS increases excitability in hippocampal neurons in a sex- and age-dependent manner through a chloride homeostasis disruption. As this hyperexcitability was only seen in adult males, and not in adult females or adolescent animals of either sex, it raises the possibility of a hormonal interaction with early life inflammation. Furthermore, as neonatal inflammation is a normal feature of early life in most animals, as well as humans, these findings may be very important for the development of animal models of disease that more appropriately resemble the human condition.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Homeostasis/fisiología , Inflamación/metabolismo , Células Piramidales/metabolismo , Caracteres Sexuales , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Factores de Edad , Animales , Región CA1 Hipocampal/efectos de los fármacos , Femenino , Homeostasis/efectos de los fármacos , Inflamación/inducido químicamente , Lipopolisacáridos/toxicidad , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Células Piramidales/efectos de los fármacos , Factores Sexuales
3.
J Neurosci ; 38(42): 9019-9033, 2018 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-30185466

RESUMEN

Emotional dysfunction is common in multiple sclerosis (MS) patients and in mouse models of MS, including experimental autoimmune encephalomyelitis (EAE); however, the etiology of these behaviors is poorly understood. To identify CNS changes associated with these behaviors, we focused on the basolateral amygdala (BLA) because of its central role in the regulation of emotional behavior. Whole-cell recordings were performed in the principal neurons of the BLA in early EAE, before demyelination, T-cell invasion, and motor dysfunction. EAE female mice displayed increased frequency of mEPSCs, with no alteration in amplitude or evoked EPSC paired-pulse ratio compared with controls. We found an increase in the AMPA-NMDA ratio and dendritic spine density, indicating increased numbers of glutamatergic synapses. We saw similar electrophysiological changes in BLA principal neurons after microglia were either inactivated (minocycline) or depleted (Mac1-Saporin) in the BLA. Microglia regulate synapses through pruning, directed by complement protein 3 (C3) expression. C3 was downregulated in the BLA in EAE. Ultrastructural analysis of microglia revealed more complex ramifications and reduced extracellular digestion of cellular elements. We also observed reduced IBA-1 and CD68 staining and lack of proinflammatory cytokine expression in the amygdala. Thus, early EAE is a state of microglial "deactivation" associated with reduced synaptic pruning. This contrasts with the prototypic microglial activation commonly associated with inflammatory CNS disease. Additionally, these data support a role for the acquired immune system to influence both neuronal and microglial function in early CNS autoimmunity.SIGNIFICANCE STATEMENT Microglia help regulate synaptic homeostasis, but there has been little evidence for how this might be important in neuroinflammatory diseases. The data from this study reveal increased synaptic activity and spine density in early stages of experimental autoimmune encephalomyelitis (an animal model of multiple sclerosis) in the basolateral amygdala, a nucleus important in the types of behavioral changes we have previously described. These electrophysiological and morphological effects occurred without significant elevation of local inflammatory cytokines or local demyelination. Unexpectedly, in the context of inflammatory state, we found that microglia were "deactivated." This study provides strong evidence for a link between microglial activity and synaptic function; the conclusions contrast with the generally accepted view that microglia are activated in inflammatory disease.


Asunto(s)
Complejo Nuclear Basolateral/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Ácido Glutámico/inmunología , Microglía/inmunología , Esclerosis Múltiple/inmunología , Neuronas/inmunología , Transmisión Sináptica/inmunología , Animales , Proteínas del Sistema Complemento/inmunología , Citocinas/inmunología , Espinas Dendríticas/inmunología , Potenciales Postsinápticos Excitadores , Femenino , Ratones Endogámicos C57BL , Potenciales Postsinápticos Miniatura , Receptores AMPA/inmunología
4.
Brain Behav Immun ; 40: 74-84, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24607605

RESUMEN

BACKGROUND: Neuropsychiatric disorders during HIV/AIDS are common although the contribution of HIV-1 infection within the brain, and in particular individual HIV-1 proteins, to the development of these brain disorders is unknown. Herein, an in vivo transgenic mouse model was generated in which the HIV-1 Nef protein was expressed in microglia cells, permitting investigation of neurobehavioral phenotypes and associated cellular and molecular properties. METHODS: Transgenic (Tg) mice that expressed full length HIV-1 nef under the control of the c-fms promoter and wildtype (Wt) littermates were investigated using different measures of neurobehavioral performance including locomotory, forced swim (FST), elevated plus maze (EPM) and T-maze tests. Host gene and transgene expression were assessed by RT-PCR, immunoblotting, enzymatic activity and immunohistochemistry. Biogenic amine levels were measured by HPLC with electrochemical detection. RESULTS: Tg animals exhibited Nef expression in brain microglia and cultured macrophages. Tg males displayed hyperactive behaviors including augmented locomotor activity, decreased immobility in the FST and increased open-arm EPM exploration compared to Wt littermates (p<0.05). Tg animals showed increased CCL2 expression with concurrent IFN-α suppression in striatum compared with Wt littermates (p<0.05). Dopamine levels, MAO activity and the dopamine transporter (DAT) expression were reduced in the striatum of Tg animals (p<0.05). CONCLUSIONS: HIV-1 Nef expression in microglia induced CCL2 expression together with disrupting striatal dopaminergic transmission, resulting in hyperactive behaviors which are observed in mania and other psychiatric comorbidities among HIV-infected persons. These findings emphasize the selective effects of individual viral proteins in the brain and their participation in neuropathogenesis.


Asunto(s)
Dopamina/metabolismo , Microglía/virología , Actividad Motora , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Animales , Trastorno Bipolar/inmunología , Trastorno Bipolar/virología , Encéfalo/inmunología , Encéfalo/metabolismo , Cuerpo Estriado/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Femenino , Macrófagos/inmunología , Masculino , Ratones , Ratones Transgénicos , Microglía/inmunología , Serotonina/metabolismo , Aprendizaje Espacial/fisiología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética
5.
Brain Behav Immun ; 33: 164-72, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23886782

RESUMEN

Multiple sclerosis (MS) is often associated with co-morbid behavioural and cognitive impairments; however the presence of these symptoms does not necessarily correlate with neurological damage. This suggests that an alternate mechanism may subserve these impairments relative to motor deficits. We investigated whether these abnormalities could be studied in experimental autoimmune encephalomyelitis (EAE), an animal model of MS. In myelin oligodendrocyte glycoprotein peptide (MOG35-55)-induced EAE mice, no motor deficits were observed until d9 after immunization. This enabled us to carry out a series of neurobehavioral tests during the presymptomatic stage, between d6 and d8 post-immunization. EAE mice spent more time in the outer zone in an open field test and in the closed arms of an elevated plus maze and, showed decreased latency for immobility in the tail suspension and forced swim tests and reduced social interaction compared with controls. These results are indicative of anxiety- and depression- like behavior. In addition, EAE mice appeared to exhibit memory impairment compared to controls based on their reduced time spent in the target quadrant in the Morris water maze and their faster memory extinction in the fear conditioning test. No demyelination, microglial activation or astrogliosis was observed in the brain at this early stage. Transcript analysis by RT-PCR from d6 to d8 brain revealed elevated interleukin (IL)-1ß and TNF-α in the hypothalamus but not in the amygdala or hippocampus of EAE mice. Lastly, plasma corticosterone levels increased in EAE mice compared to controls. In conclusion, emotional and cognitive deficits are observed in EAE prior to demyelination and are associated with elevated IL-1ß and TNF-α in the hypothalamus and changes in the hypothalamic-pituitary-adrenal axis.


Asunto(s)
Conducta Animal/fisiología , Trastornos del Conocimiento/diagnóstico , Trastornos del Conocimiento/fisiopatología , Corticosterona/sangre , Citocinas/fisiología , Emociones/fisiología , Encefalomielitis Autoinmune Experimental/diagnóstico , Encefalomielitis Autoinmune Experimental/fisiopatología , Glicoproteína Mielina-Oligodendrócito/administración & dosificación , Animales , Trastornos del Conocimiento/psicología , Diagnóstico Precoz , Encefalomielitis Autoinmune Experimental/psicología , Femenino , Relaciones Interpersonales , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/inmunología , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Estrés Psicológico/inmunología , Natación
6.
J Immunol ; 187(9): 4788-99, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21964030

RESUMEN

Endoplasmic reticulum (ER) stress is a homeostatic mechanism, which is used by cells to adapt to intercellular and intracellular changes. Moreover, ER stress is closely linked to inflammatory pathways. We hypothesized that ER stress is an integral component of neuroinflammation and contributes to the development of neurological diseases. In autopsied brain specimens from multiple sclerosis (MS) and non-MS patients, XBP-1 spliced variant (XBP-1/s) was increased in MS brains (p < 0.05) and was correlated with the expression of the human endogenous retrovirus-W envelope transcript, which encodes the glycoprotein, Syncytin-1 (p < 0.05). In primary human fetal astrocytes transfected with a Syncytin-1-expressing plasmid, XBP-1/s, BiP, and NOS2 were induced, which was suppressed by crocin treatment (p < 0.05). Crocin also protected oligodendrocytes exposed to cytotoxic supernatants derived from Syncytin-1-expressing astrocytes (p < 0.05) and NO-mediated oligodendrocytotoxicity (p < 0.05). During experimental autoimmune encephalomyelitis (EAE), the transcript levels of the ER stress genes XBP-1/s, BiP, PERK, and CHOP were increased in diseased spinal cords compared with healthy littermates (p < 0.05), although CHOP expression was not involved in the EAE disease phenotype. Daily treatment with crocin starting on day 7 post-EAE induction suppressed ER stress and inflammatory gene expression in spinal cords (p < 0.05), which was accompanied by preserved myelination and axonal density, together with reduced T cell infiltration and macrophage activation. EAE-associated neurobehavioral deficits were also ameliorated by crocin treatment (p < 0.05). These findings underscored the convergent roles of pathogenic ER stress and immune pathways in neuroinflammatory disease and point to potential therapeutic applications for crocin.


Asunto(s)
Carotenoides/uso terapéutico , Enfermedades Desmielinizantes/prevención & control , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Mediadores de Inflamación/uso terapéutico , Enfermedades Neurodegenerativas/prevención & control , Animales , Carotenoides/administración & dosificación , Células Cultivadas , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/patología , Retículo Endoplásmico/efectos de los fármacos , Femenino , Depuradores de Radicales Libres/uso terapéutico , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/metabolismo , Lóbulo Frontal/patología , Humanos , Mediadores de Inflamación/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Enfermedades Neurodegenerativas/patología , Ratas , Ratas Sprague-Dawley
7.
FASEB J ; 25(7): 2211-20, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21427211

RESUMEN

Immunosuppressive lentivirus infections, including human, simian, and feline immunodeficiency viruses (HIV, SIV, and FIV, respectively), cause the acquired immunodeficiency syndrome (AIDS), frequently associated with AIDS enteropathy. Herein, we investigated the extent to which lentivirus infections affected mucosal integrity and intestinal permeability in conjunction with immune responses and activation of endoplasmic reticulum (ER) stress pathways. Duodenal biopsies from individuals with HIV/AIDS exhibited induction of IL-1ß, CD3ε, HLA-DRA, spliced XBP-1(Xbp-1s), and CHOP expression compared to uninfected persons (P<0.05). Gut epithelial cells exposed to HIV-1 Vpr demonstrated elevated TNF-α, IL-1ß, spliced Xbp-1s, and CHOP expression (P<0.05) together with calcium activation and disruption of epithelial cell monolayer permeability. In addition to reduced blood CD4(+) T lymphocyte levels, viral loads in the gut and plasma were high in FIV-infected animals (P<0.05). FIV-infected animals also exhibited a failure to gain weight and increased lactulose/mannitol ratios compared with uninfected animals (P<0.05). Proinflammatory and ER stress gene expression were activated in the ileum of FIV-infected animals (P<0.05), accompanied by intestinal epithelial damage with loss of epithelial cells and leukocyte infiltration of the lamina propria. Lentivirus infections cause gut inflammation and ensuing damage to intestinal epithelial cells, likely through induction of ER stress pathways, resulting in disruption of gut functional integrity.


Asunto(s)
Retículo Endoplásmico/metabolismo , Enteritis/genética , Células Epiteliales/metabolismo , Enteropatía por VIH/genética , Animales , Complejo CD3/genética , Complejo CD3/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Gatos , Línea Celular Tumoral , Duodeno/inmunología , Duodeno/metabolismo , Duodeno/virología , Retículo Endoplásmico/inmunología , Enteritis/inmunología , Enteritis/virología , Células Epiteliales/patología , Células Epiteliales/virología , Femenino , Expresión Génica , Enteropatía por VIH/inmunología , Enteropatía por VIH/virología , Infecciones por VIH/genética , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/inmunología , VIH-1/fisiología , Antígenos HLA-DR/genética , Antígenos HLA-DR/metabolismo , Cadenas alfa de HLA-DR , Interacciones Huésped-Patógeno , Humanos , Virus de la Inmunodeficiencia Felina/inmunología , Virus de la Inmunodeficiencia Felina/fisiología , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Infecciones por Lentivirus/genética , Infecciones por Lentivirus/inmunología , Infecciones por Lentivirus/virología , Embarazo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Carga Viral/inmunología , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/inmunología
8.
Brain ; 134(Pt 11): 3209-21, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22021895

RESUMEN

Distal sensory polyneuropathy is a frequent complication of lentivirus infections of the peripheral nervous system including both human immunodeficiency virus and feline immunodeficiency virus. Proteinase-activated receptors are G protein-coupled receptors implicated in the pathogenesis of neuroinflammation and neurodegeneration. Proteinase-activated receptor-1 is expressed on different cell types within the nervous system including neurons and glia, but little is known about its role in the pathogenesis of inflammatory peripheral nerve diseases, particularly lentivirus-related distal sensory polyneuropathy. Herein, the expression and functions of proteinase-activated receptor-1 in the peripheral nervous system during human immunodeficiency virus and feline immunodeficiency virus infections were investigated. Proteinase-activated receptor-1 expression was most evident in autopsied dorsal root ganglion neurons from subjects infected with human immunodeficiency virus, compared with the dorsal root ganglia of uninfected subjects. Human immunodeficiency virus or feline immunodeficiency virus infection of cultured human or feline dorsal root ganglia caused upregulation of interleukin-1ß and proteinase-activated receptor-1 expression. In the human immunodeficiency virus- or feline immunodeficiency virus-infected dorsal root ganglia, interleukin-1ß activation was principally detected in macrophages, while neurons showed induction of proteinase-activated receptor-1. Binding of proteinase-activated receptor-1 by the selective proteinase-activated receptor-1-activating peptide resulted in neurite retraction and soma atrophy in conjunction with cytosolic calcium activation in human dorsal root ganglion neurons. Interleukin-1ß exposure to feline or human dorsal root ganglia caused upregulation of proteinase-activated receptor-1 in neurons. Exposure of feline immunodeficiency virus-infected dorsal root ganglia to the interleukin-1 receptor antagonist prevented proteinase-activated receptor-1 induction and neurite retraction. In vivo feline immunodeficiency virus infection was associated with increased proteinase-activated receptor-1 expression on neurons and interleukin-1ß induction in macrophages. Moreover, feline immunodeficiency virus infection caused hyposensitivity to mechanical stimulation. These data indicated that activation and upregulation of proteinase-activated receptor-1 by interleukin-1ß contributed to dorsal root ganglion neuronal damage during lentivirus infections leading to the development of distal sensory polyneuropathy and might also provide new targets for future therapeutic interventions.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/patología , Ganglios Espinales/patología , Infecciones por VIH/patología , Degeneración Nerviosa/patología , Receptor PAR-1/metabolismo , Síndrome de Inmunodeficiencia Adquirida/metabolismo , Animales , Gatos , Ganglios Espinales/metabolismo , Infecciones por VIH/metabolismo , Seropositividad para VIH/metabolismo , Seropositividad para VIH/patología , Humanos , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Degeneración Nerviosa/metabolismo , Neuronas/metabolismo , Neuronas/patología , Receptores de Interleucina-1/metabolismo
9.
Retrovirology ; 8: 44, 2011 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-21645334

RESUMEN

BACKGROUND: Viral diversity and abundance are defining properties of human immunodeficiency virus (HIV)-1's biology and pathogenicity. Despite the increasing availability of antiretroviral therapy, HIV-associated dementia (HAD) continues to be a devastating consequence of HIV-1 infection of the brain although the underlying disease mechanisms remain uncertain. Herein, molecular diversity within the HIV-1 non-structural gene, Vpr, was examined in RNA sequences derived from brain and blood of HIV/AIDS patients with or without HIV-associated dementia (HAD) together with the ensuing pathobiological effects. RESULTS: Cloned brain- and blood-derived full length vpr alleles revealed that amino acid residue 77 within the brain-derived alleles distinguished HAD (77Q) from non-demented (ND) HIV/AIDS patients (77R) (p < 0.05) although vpr transcripts were more frequently detected in HAD brains (p < 0.05). Full length HIV-1 clones encoding the 77R-ND residue induced higher IFN-α, MX1 and BST-2 transcript levels in human glia relative to the 77Q-HAD encoding virus (p < 0.05) but both viruses exhibited similar levels of gene expression and replication. Myeloid cells transfected with 77Q-(pVpr77Q-HAD), 77R (pVpr77R-ND) or Vpr null (pVpr(-))-containing vectors showed that the pVpr77R-ND vector induced higher levels of immune gene expression (p < 0.05) and increased neurotoxicity (p < 0.05). Vpr peptides (amino acids 70-96) containing the 77Q-HAD or 77R-ND motifs induced similar levels of cytosolic calcium activation when exposed to human neurons. Human glia exposed to the 77R-ND peptide activated higher transcript levels of IFN-α, MX1, PRKRA and BST-2 relative to 77Q-HAD peptide (p < 0.05). The Vpr 77R-ND peptide was also more neurotoxic in a concentration-dependent manner when exposed to human neurons (p < 0.05). Stereotaxic implantation of full length Vpr, 77Q-HAD or 77R-ND peptides into the basal ganglia of mice revealed that full length Vpr and the 77R-ND peptide caused greater neurobehavioral deficits and neuronal injury compared with 77Q-HAD peptide-implanted animals (p < 0.05). CONCLUSIONS: These observations underscored the potent neuropathogenic properties of Vpr but also indicated viral diversity modulates innate neuroimmunity and neurodegeneration.


Asunto(s)
Encéfalo/virología , VIH-1/inmunología , VIH-1/patogenicidad , Polimorfismo Genético , Tropismo Viral , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/inmunología , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo , Secuencia de Aminoácidos , Animales , Antígenos CD/biosíntesis , Sangre/virología , Proteínas Ligadas a GPI/biosíntesis , Proteínas de Unión al GTP/biosíntesis , Perfilación de la Expresión Génica , VIH-1/genética , Humanos , Inmunidad Innata , Interferón-alfa/biosíntesis , Masculino , Ratones , Datos de Secuencia Molecular , Proteínas de Resistencia a Mixovirus , Neuroglía/inmunología , Neuroglía/virología , ARN Viral/genética , Análisis de Secuencia de ADN , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/genética
10.
FASEB J ; 24(11): 4343-53, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20628092

RESUMEN

Painful peripheral neuropathy has become the principal neurological disorder in HIV/AIDS patients. Herein, we investigated the effects of a cytotoxic HIV-1 accessory protein, viral protein R (Vpr), on the peripheral nervous system (PNS). Host and viral gene expression was investigated in peripheral nerves from HIV-infected individuals and in HIV-infected human dorsal root ganglion (DRG) cultures by RT-PCR and immunocytochemistry. Cytosolic calcium ([Ca(2+)]) fluxes and neuronal membrane responses were analyzed in cultured DRGs. Neurobehavioral responses and cytokine levels were assessed in a transgenic mouse model in which the vpr transgene was expressed in an immunodeficient background (vpr/RAG1(-/-)). Vpr transcripts and proteins were detected in peripheral nerves and DRGs from HIV-infected patients. Exposure of rat or human cultured DRG neurons to Vpr rapidly increased [Ca(2+)] and action potential frequency while increasing input resistance. HIV infection of human DRG cultures caused neurite retraction (P<0.05), accompanied by induction of interferon-α (IFN-α) transcripts (P<0.05). vpr/RAG1(-/-) mice expressed Vpr together with increased IFN-α (P<0.05) in the PNS and also exhibited mechanical allodynia, unlike their vpr/RAG1(-/-) littermates (P<0.05). Herein, Vpr caused DRG neuronal damage, likely through cytosolic calcium activation and cytokine perturbation, highlighting Vpr's contribution to HIV-associated peripheral neuropathy and ensuing neuropathic pain.


Asunto(s)
Productos del Gen vpr/metabolismo , VIH-1 , Neuralgia/complicaciones , Enfermedades del Sistema Nervioso Periférico/complicaciones , Traumatismos del Sistema Nervioso/complicaciones , Animales , Células Cultivadas , Ganglios Espinales/fisiopatología , Ganglios Espinales/virología , Regulación de la Expresión Génica , Productos del Gen vpr/genética , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Sci Rep ; 11(1): 14319, 2021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-34253764

RESUMEN

Microglia play an important role in the pathogenesis of multiple sclerosis and the mouse model of MS, experimental autoimmune encephalomyelitis (EAE). To more fully understand the role of microglia in EAE we characterized microglial transcriptomes before the onset of motor symptoms (pre-onset) and during symptomatic EAE. We compared the transcriptome in brain, where behavioral changes are initiated, and spinal cord, where damage is revealed as motor and sensory deficits. We used a RiboTag strategy to characterize ribosome-bound mRNA only in microglia without incurring possible transcriptional changes after cell isolation. Brain and spinal cord samples clustered separately at both stages of EAE, indicating regional heterogeneity. Differences in gene expression were observed in the brain and spinal cord of pre-onset and symptomatic animals with most profound effects in the spinal cord of symptomatic animals. Canonical pathway analysis revealed changes in neuroinflammatory pathways, immune functions and enhanced cell division in both pre-onset and symptomatic brain and spinal cord. We also observed a continuum of many pathways at pre-onset stage that continue into the symptomatic stage of EAE. Our results provide additional evidence of regional and temporal heterogeneity in microglial gene expression patterns that may help in understanding mechanisms underlying various symptomology in MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Esclerosis Múltiple/metabolismo , Animales , Secuencia de Bases , Femenino , Técnica del Anticuerpo Fluorescente , Ratones , Microglía , ARN Mensajero/metabolismo , Transmisión Sináptica/fisiología , Transcriptoma/genética
12.
J Exp Neurosci ; 13: 1179069519825882, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30733631

RESUMEN

Multiple sclerosis, and its animal model-experimental autoimmune encephalomyelitis (EAE), is a demyelinating disease causing motor and sensory dysfunction, as well as behavioral comorbidities. In exploring possible functional changes underlying behavioral comorbidities in EAE, we observed increased excitatory drive onto the major cells of the basolateral amygdala. This was associated with increased numbers of dendritic spines. An unexpected finding was that microglial cells at this time were in a "deactivated" state, and further studies suggested that the microglial deactivation was responsible for the increased excitatory drive. This is the first report of microglial deactivation in an inflammatory disease and raises many questions as to the underlying mechanisms and functional relevance.

13.
AIDS ; 30(4): 543-52, 2016 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-26558720

RESUMEN

OBJECTIVE: The neurotoxic actions of the HIV protease inhibitors, amprenavir (APV) and lopinavir (LPV) were investigated. DESIGN: With combination antiretroviral therapy (cART), HIV-infected persons exhibit neurocognitive impairments, raising the possibility that cART might exert adverse central nervous system (CNS) effects. We examined the effects of LPV and APV using in-vitro and in-vivo assays of CNS function. METHODS: Gene expression, cell viability and amino-acid levels were measured in human astrocytes, following exposure to APV or LPV. Neurobehavioral performance, amino-acid levels and neuropathology were examined in HIV-1 Vpr transgenic mice after treatment with APV or LPV. RESULTS: Excitatory amino-acid transporter-2 (EAAT2) expression was reduced in astrocytes treated with LPV or APV, especially LPV (P < 0.05), which was accompanied by reduced intracellular L-glutamate levels in LPV-treated cells (P < 0.05). Treatment of astrocytes with APV or LPV reduced the expression of proliferating cell nuclear antigen (PCNA) and Ki-67 (P < 0.05) although cell survival was unaffected. Exposure of LPV to astrocytes augmented glutamate-evoked transient rises in [Cai] (P < 0.05). Vpr mice treated with LPV showed lower concentrations of L-glutamate, L-aspartate and L-serine in cortex compared with vehicle-treated mice (P < 0.05). Total errors in T-maze assessment were increased in LPV and APV-treated animals (P < 0.05). EAAT2 expression was reduced in the brains of protease inhibitor-treated animals, which was associated with gliosis (P < 0.05). CONCLUSION: These results indicated that contemporary protease inhibitors disrupt astrocyte functions at therapeutic concentrations with enhanced sensitivity to glutamate, which can lead to neurobehavioral impairments. ART neurotoxicity should be considered in future therapeutic regimens for HIV/AIDS.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG/antagonistas & inhibidores , Astrocitos/efectos de los fármacos , Astrocitos/enzimología , Carbamatos/efectos adversos , Inhibidores de la Proteasa del VIH/efectos adversos , Lopinavir/efectos adversos , Enfermedades del Sistema Nervioso/inducido químicamente , Sulfonamidas/efectos adversos , Animales , Química Encefálica , Carbamatos/administración & dosificación , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Femenino , Furanos , Perfilación de la Expresión Génica , Inhibidores de la Proteasa del VIH/administración & dosificación , VIH-1 , Humanos , Lopinavir/administración & dosificación , Masculino , Ratones Transgénicos , Enfermedades del Sistema Nervioso/patología , Examen Neurológico , Sulfonamidas/administración & dosificación
14.
J Neuroimmune Pharmacol ; 7(2): 319-31, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21918813

RESUMEN

HIV-associated neurocognitive disorders (HAND) represent a constellation of neurological disabilities defined by neuropsychological impairments, neurobehavioral abnormalities and motor deficits. To gain insights into the mechanisms underlying the development of these disabilities, several transgenic models have been developed over the past two decades, which have provided important information regarding the cellular and molecular factors contributing to the neuropathogenesis of HAND. Herein, we concentrate on the neuropathogenic effects of HIV-1 Vpr expressed under the control of c-fms, resulting transgene expression in myeloid cells in both the central and peripheral nervous systems. Vpr's actions, possibly through its impact on cell cycle machinery, in brain culminate in neuronal and astrocyte injury and death through apoptosis involving activation of caspases-3, -6 and -9 depending on the individual target cell type. Indeed, these outcomes are also induced by soluble Vpr implying Vpr's effects stem from direct interaction with target cells. Remarkably, in vivo transgenic Vpr expression induces a neurodegenerative phenotype defined by neurobehavioral deficits and neuronal loss in the absence of frank inflammation. Implantation of another viral protein, hepatitis C virus (HCV) core, into Vpr transgenic animals' brains stimulated neuroinflammation and amplified the neurodegenerative disease phenotype, thereby recapitulating HCV's putative neuropathogenic actions. The availability of different transgenic models to study HIV neuropathogenesis represents exciting and innovative approaches to understanding disease mechanisms and perhaps developing new therapeutic strategies in the future.


Asunto(s)
Complejo SIDA Demencia/metabolismo , Complejo SIDA Demencia/virología , Modelos Animales de Enfermedad , VIH-1/patogenicidad , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo , Complejo SIDA Demencia/genética , Animales , Animales Modificados Genéticamente , VIH-1/genética , VIH-1/metabolismo , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/genética
15.
G3 (Bethesda) ; 2(1): 59-69, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22384382

RESUMEN

Mitochondria located within neuronal presynaptic terminals have been shown to play important roles in the release of chemical neurotransmitters. In the present study, a genetic screen for synaptic transmission mutants of Drosophila has identified the first mutation in a Drosophila homolog of the mitochondrial protein P32. Although P32 is highly conserved and has been studied extensively, its physiological role in mitochondria remains unknown and it has not previously been implicated in neural function. The Drosophila P32 mutant, referred to as dp32(EC1), exhibited a temperature-sensitive (TS) paralytic behavioral phenotype. Moreover, electrophysiological analysis at adult neuromuscular synapses revealed a TS reduction in the amplitude of excitatory postsynaptic currents (EPSC) and indicated that dP32 functions in neurotransmitter release. These studies are the first to address P32 function in Drosophila and expand our knowledge of mitochondrial proteins contributing to synaptic transmission.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA