Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Front Immunol ; 12: 753683, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34899705

RESUMO

Human parechovirus type 3 (PeV-A3) infection has been recognized as an emerging etiologic factor causing severe nerve disease or sepsis in infants and young children. But the neuropathogenic mechanisms of PeV-A3 remain unknown. To understand the pathogenesis of PeV-A3 infection in the neuronal system, PeV-A3-mediated cytopathic effects were analyzed in human glioblastoma cells and neuroblastoma cells. PeV-A3 induced interferons and inflammatory cytokine expression in these neuronal cells. The pronounced cytopathic effects accompanied with activation of death signaling pathways of apoptosis, autophagy, and pyroptosis were detected. A new experimental disease model of parechovirus encephalitis was established. In the disease model, intracranial inoculation with PeV-A3 in C57BL/6 neonatal mice showed body weight loss, hindlimb paralysis, and approximately 20% mortality. PeV-A3 infection in the hippocampus and cortex regions of the neonatal mouse brain was revealed. Mechanistic assay supported the in vitro results, indicating detection of PeV-A3 replication, inflammatory cytokine expression, and death signaling transduction in mouse brain tissues. These in vitro and in vivo studies revealed that the activation of death signaling and inflammation responses is involved in PeV-A3-mediated neurological disorders. The present results might account for some of the PeV-A3-associated clinical manifestations.


Assuntos
Efeito Citopatogênico Viral , Modelos Animais de Doenças , Encefalite Viral/metabolismo , Parechovirus/patogenicidade , Infecções por Picornaviridae/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Autofagia , Linhagem Celular Tumoral , Córtex Cerebral/virologia , Chlorocebus aethiops , Citocinas/biossíntese , Citocinas/genética , Encefalite Viral/patologia , Encefalite Viral/virologia , Glioblastoma/patologia , Hipocampo/virologia , Humanos , Inflamação , Interferon Tipo I/biossíntese , Interferon Tipo I/genética , Interferon Tipo I/farmacologia , Interferons/biossíntese , Interferons/genética , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Endogâmicos C57BL , Neuroblastoma/patologia , Parechovirus/efeitos dos fármacos , Parechovirus/fisiologia , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , Piroptose , Células Vero , Replicação Viral/efeitos dos fármacos , Interferon lambda
2.
PLoS One ; 16(12): e0261208, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34890434

RESUMO

PURPOSE: Changes in cerebral cortical regions occur in HIV-infected patients, even in those with mild neurocognitive disorders. Working memory / attention is one of the most affected cognitive domain in these patients, worsening their quality of life. Our objective was to assess whether cortical thickness differs between HIV-infected patients with and without working memory deficit. METHODS: Forty-one adult HIV-infected patients with and without working memory deficit were imaged on a 1.5 T scanner. Working memory deficit was classified by composite Z scores for performance on the Digits and Letter-Number Sequencing subtests of the Wechsler Adult Intelligence Scale (third edition; WAIS-III). Cortical thickness was determined using FreeSurfer software. Differences in mean cortical thickness between groups, corrected for multiple comparisons using Monte-Carlo simulation, were examined using the query design estimate contrast tool of the FreeSurfer software. RESULTS: Greater cortical thickness in left pars opercularis of the inferior frontal gyrus, and rostral and caudal portions of the left middle frontal gyrus (cluster 1; p = .004), and left superior frontal gyrus (cluster 2; p = .004) was observed in HIV-infected patients with working memory deficit compared with those without such deficit. Negative correlations were found between WAIS-III-based Z scores and cortical thickness in the two clusters (cluster 1: ρ = -0.59; cluster 2: ρ = -0.47). CONCLUSION: HIV-infected patients with working memory deficit have regions of greater thickness in the left frontal cortices compared with those without such deficit, which may reflect increased synaptic contacts and/or an inflammatory response related to the damage caused by HIV infection.


Assuntos
Córtex Cerebral/patologia , Córtex Cerebral/virologia , Infecções por HIV/patologia , Transtornos da Memória/virologia , Memória de Curto Prazo/fisiologia , Adulto , Idoso , Brasil/epidemiologia , Feminino , HIV/isolamento & purificação , Infecções por HIV/epidemiologia , Infecções por HIV/psicologia , Infecções por HIV/virologia , Humanos , Masculino , Transtornos da Memória/epidemiologia , Transtornos da Memória/patologia , Transtornos da Memória/psicologia , Pessoa de Meia-Idade , Testes Neuropsicológicos
3.
Cells ; 10(9)2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34571912

RESUMO

COVID-19 presents with a wide range of clinical neurological manifestations. It has been recognized that SARS-CoV-2 infection affects both the central and peripheral nervous system, leading to smell and taste disturbances; acute ischemic and hemorrhagic cerebrovascular disease; encephalopathies and seizures; and causes most surviving patients to have long lasting neurological symptoms. Despite this, typical neuropathological features associated with the infection have still not been identified. Studies of post-mortem examinations of the cerebral cortex are obtained with difficulty due to laboratory safety concerns. In addition, they represent cases with different neurological symptoms, age or comorbidities, thus a larger number of brain autoptic data from multiple institutions would be crucial. Histopathological findings described here are aimed to increase the current knowledge on neuropathology of COVID-19 patients. We report post-mortem neuropathological findings of ten COVID-19 patients. A wide range of neuropathological lesions were seen. The cerebral cortex of all patients showed vascular changes, hyperemia of the meninges and perivascular inflammation in the cerebral parenchyma with hypoxic neuronal injury. Perivascular lymphocytic inflammation of predominantly CD8-positive T cells mixed with CD68-positive macrophages, targeting the disrupted vascular wall in the cerebral cortex, cerebellum and pons were seen. Our findings support recent reports highlighting a role of microvascular injury in COVID-19 neurological manifestations.


Assuntos
COVID-19/patologia , Córtex Cerebral/patologia , Idoso , Idoso de 80 Anos ou mais , Autopsia , Encéfalo/patologia , Encéfalo/virologia , Encefalopatias/patologia , Encefalopatias/virologia , Linfócitos T CD8-Positivos/patologia , Córtex Cerebral/virologia , Feminino , Humanos , Inflamação , Macrófagos/patologia , Masculino , Microvasos/patologia , Microvasos/virologia , Pessoa de Meia-Idade , Doenças do Sistema Nervoso/patologia , Doenças do Sistema Nervoso/virologia , SARS-CoV-2/patogenicidade
4.
J Immunol Res ; 2021: 5317662, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34327243

RESUMO

Zika virus (ZIKV) has attracted the wide global attention due to its causal link to microcephaly. In this study, two amino acid (aa) mutation (E143K and R3394K) were identified at the fourth generation (named ZKC2P4) during the serial passage of ZIKV-Asian lineage ZKC2/2016 strain in the newborn mouse brain, while another seven aa deletions in envelope (E) protein were detected in ZKC2P6. ZKC2P6 is a novel nonglycosylated E protein Asian ZIKV we first identified and provides the first direct supporting evidence that glycosylation motif could be lost during the passage in neonatal mice. To study the impact of E protein glycosylation ablation, we compared the pathogenicity of ZKC2P6 with that of ZKC2P4. The results showed that the loss of E protein glycosylation accelerated the disease progression, as evidenced by an earlier weight loss and death, a thinner cerebral cortex, and more serious tissue lesions and inflammation/necrosis. Furthermore, ZKC2P6 exhibited a greater ability to replicate and caused severer cell apoptosis than that of ZKC2P4. Therefore, the ablation of E glycosylation generally enhances the neurovirulence of ZIKV and cell apoptosis in newborn mice.


Assuntos
Córtex Cerebral/patologia , Proteínas do Envelope Viral/metabolismo , Infecção por Zika virus/imunologia , Zika virus/patogenicidade , Animais , Animais Recém-Nascidos , Apoptose , Linhagem Celular Tumoral , Córtex Cerebral/imunologia , Córtex Cerebral/virologia , Chlorocebus aethiops , Modelos Animais de Doenças , Progressão da Doença , Glicosilação , Humanos , Camundongos , Deleção de Sequência , Células Vero , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Replicação Viral/imunologia , Zika virus/genética , Zika virus/imunologia , Zika virus/metabolismo , Infecção por Zika virus/patologia , Infecção por Zika virus/virologia
5.
J Neurovirol ; 26(6): 846-862, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32910432

RESUMO

Previous studies showed that persons living with HIV (PLWH) demonstrate higher brain prefrontal cortex neuroinflammation and immunoproteasome expression compared to HIV-negative individuals; these associate positively with HIV levels. Lower expression of the antioxidant enzyme heme oxygenase 1 (HO-1) was observed in PLWH with HIV-associated neurocognitive impairment (HIV-NCI) compared to neurocognitively normal PLWH. We hypothesized that similar expression patterns occur throughout cortical, subcortical, and brainstem regions in PLWH, and that neuroinflammation and immunoproteasome expression associate with lower expression of neuronal markers. We analyzed autopsied brains (15 regions) from 9 PLWH without HIV-NCI and 7 matched HIV-negative individuals. Using Western blot and RT-qPCR, we quantified synaptic, inflammatory, immunoproteasome, endothelial, and antioxidant biomarkers, including HO-1 and its isoform heme oxygenase 2 (HO-2). In these PLWH without HIV-NCI, we observed higher expression of neuroinflammatory, endothelial, and immunoproteasome markers in multiple cortical and subcortical regions compared to HIV-negative individuals, suggesting a global brain inflammatory response to HIV. Several regions, including posterior cingulate cortex, globus pallidus, and cerebellum, showed a distinct pattern of higher type I interferon (IFN)-stimulated gene and immunoproteasome expression. PLWH without HIV-NCI also had (i) stable or higher HO-1 expression and positive associations between (ii) HO-1 and HIV levels (CSF, plasma) and (iii) HO-1 expression and neuroinflammation, in multiple cortical, subcortical, and brainstem regions. We observed no differences in synaptic marker expression, suggesting little, if any, associated neuronal injury. We speculate that this may reflect a neuroprotective effect of a concurrent HO-1 antioxidant response despite global neuroinflammation, which will require further investigation.


Assuntos
Córtex Cerebral/metabolismo , Disfunção Cognitiva/genética , Infecções por HIV/genética , HIV-1/patogenicidade , Heme Oxigenase-1/genética , Idoso , Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/virologia , Autopsia , Biomarcadores/metabolismo , Tronco Encefálico/metabolismo , Tronco Encefálico/virologia , Estudos de Casos e Controles , Núcleo Caudado/metabolismo , Núcleo Caudado/virologia , Córtex Cerebral/virologia , Disfunção Cognitiva/complicações , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/virologia , Feminino , Regulação da Expressão Gênica , Infecções por HIV/complicações , Infecções por HIV/metabolismo , Infecções por HIV/virologia , Heme Oxigenase-1/metabolismo , Humanos , Inflamação , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Masculino , Pessoa de Meia-Idade , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo
6.
J Neurovirol ; 26(5): 769-778, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32839948

RESUMO

The blood-brain barrier (BBB) is a major obstacle for the treatment of central nervous system (CNS) disorders. Significant progress has been made in developing adeno-associated virus (AAV) variants with increased ability to cross the BBB in mice. However, these variants are not efficacious in non-human primates. Herein, we employed various bioinformatic techniques to identify lymphocyte antigen-6E (LY6E) as a candidate for mediating transport of AAV across the human BBB based on the previously determined mechanism of transport in mice. Our results provide insight into future discovery and optimization of AAV variants for CNS gene delivery in humans.


Assuntos
Antígenos Ly/metabolismo , Antígenos de Superfície/metabolismo , Barreira Hematoencefálica/metabolismo , Dependovirus/metabolismo , Vetores Genéticos/metabolismo , Proteínas de Membrana/metabolismo , Receptores Virais/metabolismo , Sequência de Aminoácidos , Animais , Antígenos Ly/química , Antígenos Ly/genética , Antígenos de Superfície/química , Antígenos de Superfície/genética , Transporte Biológico , Barreira Hematoencefálica/virologia , Permeabilidade Capilar , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/citologia , Córtex Cerebral/virologia , Biologia Computacional/métodos , Dependovirus/química , Dependovirus/genética , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Endoteliais/virologia , Proteínas Ligadas por GPI/química , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos/química , Humanos , Macaca mulatta , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Modelos Moleculares , Simulação de Acoplamento Molecular , Ligação Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores Virais/química , Receptores Virais/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
7.
J Neurovirol ; 25(4): 605-607, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31140129

RESUMO

Progressive multifocal leukoencephalopathy (PML) is a demyelinating disease caused by JC virus reactivation. Its occurrence is very rare after solid organ transplantation, especially liver transplantation. We report a patient who received liver transplantation due to liver failure resulting from autoimmune hepatitis and advanced PML presenting with aphasia. A 41-year-old female with a history of liver transplantation who received a usual immunosuppression regimen was admitted with a stroke attack resulting in right hemiplegia 2 months after liver transplantation. Surprisingly, she gradually developed dysarthria and left central facial paresis. A brain MRI showed an abnormal multifocal area with a high T2/flair signal in the deep subcortical white matter of the left hemisphere as well as the splenium of the corpus callosum. PCR evaluation of CSF for JCV was positive while other PCR results were negative. A liver transplant recipient receiving immunosuppressive treatment for a long time could develop PML due to JCV reactivation. Only eight cases of JCV infection were reported after liver transplantation by the time of reporting this case. Unfortunately, there is no definite treatment for PML.


Assuntos
Hepatite Autoimune/imunologia , Vírus JC/genética , Leucoencefalopatia Multifocal Progressiva/imunologia , Transplante de Fígado , Adulto , Afasia/diagnóstico por imagem , Afasia/fisiopatologia , Afasia/virologia , Córtex Cerebral/diagnóstico por imagem , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Córtex Cerebral/virologia , Corpo Caloso/diagnóstico por imagem , Corpo Caloso/efeitos dos fármacos , Corpo Caloso/patologia , Corpo Caloso/virologia , Disartria/diagnóstico por imagem , Disartria/fisiopatologia , Disartria/virologia , Feminino , Hemiplegia/diagnóstico por imagem , Hemiplegia/fisiopatologia , Hemiplegia/virologia , Hepatite Autoimune/patologia , Hepatite Autoimune/cirurgia , Hepatite Autoimune/virologia , Humanos , Imunossupressores/administração & dosagem , Vírus JC/imunologia , Vírus JC/isolamento & purificação , Leucoencefalopatia Multifocal Progressiva/diagnóstico por imagem , Leucoencefalopatia Multifocal Progressiva/patologia , Leucoencefalopatia Multifocal Progressiva/cirurgia , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/patologia , Fígado/cirurgia , Imageamento por Ressonância Magnética , Acidente Vascular Cerebral/diagnóstico por imagem , Acidente Vascular Cerebral/fisiopatologia , Acidente Vascular Cerebral/virologia , Ativação Viral/imunologia
8.
Cell Rep ; 21(2): 517-532, 2017 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-29020636

RESUMO

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.


Assuntos
Antirretrovirais/farmacologia , Córtex Cerebral/citologia , Avaliação Pré-Clínica de Medicamentos/métodos , Organoides/virologia , Cultura Primária de Células/métodos , Zika virus/efeitos dos fármacos , Linhagem Celular , Córtex Cerebral/virologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/virologia , Humanos , Neurônios/citologia , Neurônios/metabolismo , Neurônios/virologia , Organoides/citologia , Organoides/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Fator de Transcrição STAT3/metabolismo , c-Mer Tirosina Quinase/metabolismo
9.
Microb Pathog ; 112: 38-49, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28939254

RESUMO

The extracellular signal-regulated kinase (ERK) pathway has been shown to regulate pathogenesis of many viral infections, but its role during rabies virus (RV) infection in vivo is not clear. In the present study, we investigated the potential role of MEK-ERK1/2 signalling pathway in the pathogenesis of rabies in mouse model and its regulatory effects on pro-inflammatory cytokines and other mediators of immunity, and kinetics of immune cells. Mice were infected with 25 LD50 of challenge virus standard (CVS) strain of RV by intracerebral (i.c.) inoculation and were treated i.c. with U0126 (specific inhibitor of MEK1/2) at 10 µM/mouse at 0, 2, 4 and 6 days post-infection. Treatment with U0126 resulted in delayed disease development and clinical signs, increased survival time with lesser mortality than untreated mice. The better survival of inhibitor-treated and RV infected mice was positively correlated with reduced viral load and reduced viral spread in the brain as quantified by real-time PCR, direct fluorescent antibody test and immunohistochemistry. CVS-infected/mock-treated mice developed severe histopathological lesions with increased Fluoro-Jade B positive degenerating neurons in brain, which were associated with higher levels of serum nitric oxide, iNOS, TNF-α, and CXCL10 mRNA. Also CVS-infected/U0126-treated mice revealed significant decrease in caspase 3 but increase in Bcl-2 mRNA levels and less TUNEL positive apoptotic cells. CVS-infected/U0126-treated group also showed significant increase in CD4+, CD8+ T lymphocytes and NK cells in blood and spleen possibly due to less apoptosis of these cells. In conclusion, these data suggest that MEK-ERK1/2 signalling pathway play critical role in the pathogenesis of RV infection in vivo and opens up new avenues of therapeutics.


Assuntos
Butadienos/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Nitrilas/antagonistas & inibidores , Vírus da Raiva/efeitos dos fármacos , Vírus da Raiva/patogenicidade , Raiva/tratamento farmacológico , Animais , Apoptose , Encéfalo/patologia , Encéfalo/virologia , Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos , Caspase 3/metabolismo , Córtex Cerebral/patologia , Córtex Cerebral/virologia , Quimiocina CXCL10/sangue , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Expressão Gênica , Imuno-Histoquímica , Imunofenotipagem , Células Matadoras Naturais , Cinética , Masculino , Camundongos , Degeneração Neural , Óxido Nítrico/sangue , Óxido Nítrico Sintase Tipo II/sangue , RNA Mensageiro/sangue , Raiva/mortalidade , Raiva/virologia , Vírus da Raiva/genética , Transcriptoma , Fator de Necrose Tumoral alfa/sangue , Carga Viral
10.
Artigo em Inglês | MEDLINE | ID: mdl-28516065

RESUMO

Porcine hemagglutinating encephalomyelitis virus (PHEV) invades the central nervous system (CNS) and causes neurodegenerative disease in suckling piglets, but the understanding of its neuropathogenicity for neurological dysfunction remains limited. Here, we report that miR-142-5p is localized to neurons and negatively regulates neuronal morphogenesis in porcine hemagglutinating encephalomyelitis (PHE). This phenotype was mediated by miR-142-5p inhibition of an mRNA encoding unc-51-like-kinase1 (Ulk1), which controls axon outgrowth and dendrite formation. Modulating miR-142-5p activity by microRNA mimics or inhibitors induced neurodegeneration, including stunted axon elongation, unstable dendritic spine formation, and irregular swelling and disconnection in neurites. Relieving Ulk1 mRNA repression in primary cortical neurons by miR-142-5p antagomirs or replication-deficient adenoviruses encoding Ulk1 (Ad5-Ulk1), which improved rescue of nerve injury, restricted viral replication, and increased survival rate in mice underlying PHEV infection. In contrast, disrupting Ulk1 in RNAi-expressing neurons mostly led to significantly shortened axon elongation and/or an abnormally large number of branched dendrites. Taken together, we demonstrated that the abnormal neuronal morphogenesis underlying PHEV infection was mainly caused by functional mRNA repression of the miR-142-5p target Ulk1. Our data revealed that PHEV adapted to use spatiotemporal control of host microRNAs to invade CNS, and provided new insights into the virus-associated neurological dysfunction microenvironment.


Assuntos
Proteína Homóloga à Proteína-1 Relacionada à Autofagia/efeitos dos fármacos , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Betacoronavirus 1/metabolismo , MicroRNAs/antagonistas & inibidores , Morfogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/virologia , Animais , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/genética , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/fisiologia , Axônios/efeitos dos fármacos , Axônios/patologia , Axônios/virologia , Proliferação de Células/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Córtex Cerebral/virologia , Infecções por Coronavirus/virologia , Dendritos/efeitos dos fármacos , Dendritos/patologia , Dendritos/virologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Masculino , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/genética , MicroRNAs/farmacologia , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Replicação Viral/efeitos dos fármacos
11.
Int. j. morphol ; 34(4): 1362-1368, Dec. 2016. ilus
Artigo em Inglês | LILACS | ID: biblio-840894

RESUMO

Rabies is a lethal disease caused by a neurotropic virus that produces inconspicuous morphological changes hardly observable with conventional histopathology. The fatal outcome caused by rabies could be attributed to specific biochemical changes that severely impact neuronal function. The neuronal nuclear protein (NeuN) has become a widely used neuronal marker for the research and the histopathological diagnosis of nervous system diseases. To evaluate the distribution of the protein NeuN in the motor cortex of normal and rabies-infected mice adult ICR mice were inoculated with rabies virus either intramuscularly or intracerebrally. Rabies-infected mice were sacrificed at the terminal stage of the disease. Control mice were also euthanized at the same age. The brains were removed and cut into coronal sections on a vibratome. Immunohistochemistry was used to study the expression of NeuN in the motor area of the cerebral cortex. Neuronal counts, cellular optical densitometry and neuronal diameter measurements were performed to analyze the immunoreactivity of the protein. All parameters revealed decreased immunoreactivity for NeuN in cortical neurons of mice intracerebrally infected with rabies. In contrast, the changes were not statistically significant in mice inoculated intramuscularly. Either the immunoreactivity of NeuN or its expression is affected by the presence of rabies virus in the cerebral cortex depending on the inoculation route. These results contribute to the knowledge of the dynamics of cellular infection on rabies pathogenesis.


La rabia es una enfermedad mortal causada por un virus neurotrópico que produce discretos cambios morfológicos difícilmente observables con la histopatología convencional. El desenlace fatal causado por la rabia puede atribuirse a cambios bioquímicos específicos que afectan gravemente la función neuronal. La proteína nuclear neuronal (NeuN) es un marcador ampliamente utilizado para la investigación y el diagnóstico histopatológico de enfermedades del sistema nervioso. Este trabajo se realizó con el propósito de evaluar la distribución de la proteína NeuN en la corteza motora de ratones normales y ratones infectados con virus de la rabia. Ratones ICR adultos fueron inoculados con virus de la rabia por vía intramuscular o por vía intracerebral. Los animales infectados con rabia fueron sacrificados en la etapa terminal de la enfermedad. Ratones de la misma edad no inoculados con el virus (controles) fueron sacrificados simultáneamente. Se extrajeron los cerebros y se obtuvieron cortes coronales en un vibrátomo. Mediante inmunohistoquímica se estudió la expresión de NeuN en el área motora de la corteza cerebral. Se realizaron conteos neuronales, densitometría óptica celular y mediciones del diámetro de los perfiles neuronales para analizar la inmunorreactividad de la proteína. En los ratones inoculados por vía intracerebral hubo disminución significativa de la inmunorreactividad de NeuN manifestada en los diferentes parámetros evaluados. En contraste, estos cambios no fueron estadísticamente significativos en los cerebros de ratones inoculados por la ruta intramuscular. La inmunorreactividad de NeuN o su expresión es afectada por la presencia del virus de la rabia en la corteza cerebral pero dependiendo de la vía de inoculación. Estos resultados contribuyen al conocimiento de las dinámicas de infección celular en la patogénesis de la rabia.


Assuntos
Animais , Camundongos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Vírus da Raiva/patogenicidade , Raiva/metabolismo , Córtex Cerebral/virologia , Imuno-Histoquímica , Proteínas do Tecido Nervoso/análise , Proteínas Nucleares/análise , Vírus da Raiva/metabolismo
12.
Int. j. morphol ; 33(2): 465-470, jun. 2015. ilus
Artigo em Inglês | LILACS | ID: lil-755496

RESUMO

The microtubule-associated protein MAP-2 is an integral part of the cytoskeleton and plays an important role in neural morphogenesis. This protein is an essential component of the dendritic cytoskeleton, especially in the adult brain, and its expression can be altered under experimental or pathological conditions. The purpose of this study was to evaluate the effect of infection with the rabies virus on MAP-2 immunoreactivity in the cerebral cortex of mice. The mice were inoculated with the rabies virus and the animals were sacrificed when the disease reached its advanced stage, together with uninfected animals of the same age. The brains were extracted after being previously perfusion-fixed with paraformaldehyde; coronal sections were obtained with a vibratome. The coronal sections were processed by immunohistochemistry to reveal the presence of the MAP-2 protein in neurons of the motor area of the cerebral cortex. Rabies-infected mice showed an increase in the immunoreactivity of the somata and apical dendrites in pyramidal neurons of the motor cortex. This is an unexpected result, as dendritic pathology has been previously demonstrated in rabies, and some studies on neurological disorders associate dendritic alterations with loss of expression of the MAP-2 protein. Therefore, whatever the alteration in the expression of this protein, decrease or increase, it could be causing a biochemical imbalance in the integrity and stability of the neuronal cytoskeleton.


La proteína asociada a microtúbulos MAP-2 es una parte integral del citoesqueleto y juega un papel importante en la morfogénesis neuronal. Esta proteína es un componente esencial del citoesqueleto de las dendritas, especialmente en el cerebro adulto, y su expresión puede ser alterada en condiciones experimentales o patológicas. El propósito de este estudio fue evaluar el efecto de la infección con el virus de la rabia sobre la inmunorreactividad de MAP-2 en la corteza cerebral de ratones. Ratones inoculados con el virus de la rabia fueron sacrificados cuando la enfermedad alcanzó su fase avanzada, junto con animales no infectados de la misma edad. Los cerebros se extrajeron después de que los animales fueron tratados con paraformaldehído mediante perfusión intracardiaca. En un vibrátomo se obtuvieron cortes coronales y estos se procesaron mediante inmunohistoquímica para revelar la presencia de la proteína MAP-2 en las neuronas de la zona motora de la corteza cerebral. Los ratones infectados con rabia mostraron un aumento en la inmunorreactividad de los somas y dendritas apicales en las neuronas piramidales de la corteza motora. Este es un resultado inesperado, ya que previamente se ha demostrado patología dendrítica en rabia, y algunos estudios sobre los trastornos neurológicos asocian las alteraciones dendríticas con pérdida de expresión de la proteína MAP-2. Por lo tanto, cualquiera que sea la alteración en la expresión de esta proteína, disminución o aumento, podría ser la causa de un desequilibrio bioquímico en la integridad y estabilidad del citoesqueleto neuronal.


Assuntos
Animais , Feminino , Camundongos , Vírus da Raiva/metabolismo , Córtex Cerebral/patologia , Córtex Cerebral/virologia , Proteínas Associadas aos Microtúbulos/metabolismo , Imuno-Histoquímica , Córtex Cerebral/metabolismo
13.
J Neurovirol ; 21(5): 518-24, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26025330

RESUMO

Bovine herpesvirus type 5 (BoHV-5) is the causative agent of herpetic meningoencephalitis in cattle. The purinergic system is described as a modulator of the immune response and neuroinflammation. These functions are related to the extracellular nucleotides concentration. NTPDase and 5'-nucleotidase are enzymes responsible for controlling the extracellular concentration of adenosine triphosphate (ATP), adenosine diphosphate (ADP), adenosine monophosphate (AMP), and adenosine (ADO). The aim of this study is to determinate the ectonucleotidase activity in cortical synaptosomes and synaptosomes from the hippocampus of rabbits experimentally infected with BoHV-5. Rabbits were divided into four groups, two control groups (non-inoculated animals), and two infected groups (inoculated with BoHV-5). The infected groups received 0.5 ml of BoHV-5 suspension with 10(7.5)TCID50 of viral strain SV-507/99, per paranasal sinuses, and the control groups received 0.5 ml of minimum essential media per paranasal sinuses. Animals were submitted to euthanasia on days 7 and 12 post-inoculation (p.i.); cerebral cortex and hippocampus were collected for the synaptosomes isolation and posterior determination of the ectonucleotidase activities. The results showed a decrease (P < 0.05) in ectonucleotidase activity in synaptosomes from the cerebral cortex of infected rabbits, whereas an increased (P < 0.05) ectonucleotidase activity was observed in synaptosomes from the hippocampus. These differences may be related with the heterogeneous distribution of ectonucleotidases in the different brain regions and also with the viral infectivity. Therefore, it is possible to speculate that BoHV-5 replication results in changes in ectonucleotidase activity in the brain, which may contribute to the neurological signs commonly observed in this disease.


Assuntos
Encefalite Viral/enzimologia , Infecções por Herpesviridae/enzimologia , Meningoencefalite/enzimologia , Nucleotidases/metabolismo , Sinaptossomos/enzimologia , Animais , Córtex Cerebral/enzimologia , Córtex Cerebral/virologia , Herpesvirus Bovino 5 , Hipocampo/enzimologia , Hipocampo/virologia , Coelhos
14.
PLoS One ; 9(9): e107074, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25188269

RESUMO

Regional differences in neurovirulence have been documented among subtype/clade-C HIV-1 isolates in India and Southern Africa. We previously demonstrated that a C31S substitution in Clade-C Tat dicysteine motif reduces monocyte recruitment, cytokine induction and direct neurotoxicity. Therefore, this polymorphism is considered to be a causative factor for these differences in neurovirulence. We previously reported on the genotypic differences in Tat protein between clade-C and rest of the clades showing that approximately 90% of clade-C HIV-1 Tat sequences worldwide contained this C31S polymorphism, while 99% of non-clade C isolates lacked this Tat polymorphism at C31 residue (Ranga et al. (2004) J Virol 78:2586-2590). Subsequently, we documented intra-clade-C differences in the frequency of Tat dicysteine variants between India and Southern Africa, as the basis for differential disease severity and showed the importance of the Tat dicysteine motif for neuropathogenesis using small animal models. We have now examined if determinants of neurovirulence besides Tat are different between the clade-C HIV-1 isolates from Southern Africa and India. Envelope glycoprotein gp120 is a well-documented contributor to neurotoxicity. We found that gp120 sequences of HIV-1 isolates from these two regions are genetically distinct. In order to delineate the contribution of gp120 to neurovirulence, we compared direct in vitro neurotoxicity of HIV-infected supernatants of a representative neurovirulent US clade-B isolate with two isolates each from Southern Africa and India using primary human neurons and SH-SY5Y neuroblastoma cells. Immunodepletion of gp120 of both US clade B and the Southern African clade C isolates revealed robust decreases in neurotoxicity, while that of the Indian isolates showed minimal effect on neurotoxicity. The gp120 as a cause of differential neurotoxicity was further confirmed using purified recombinant gp120 from HIV isolates from these regions. We conclude that gp120 is one of the key factors responsible for the decreased neurovirulence of Indian clade C HIV-1 isolates when compared to South African clade C HIV-1.


Assuntos
Proteína gp120 do Envelope de HIV/genética , HIV-1/genética , HIV-1/patogenicidade , Neurônios/efeitos dos fármacos , Polimorfismo Genético , África Austral , Linhagem Celular Tumoral , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Córtex Cerebral/virologia , Meios de Cultivo Condicionados/toxicidade , Feto , Expressão Gênica , Genótipo , Proteína gp120 do Envelope de HIV/química , Proteína gp120 do Envelope de HIV/metabolismo , HIV-1/classificação , HIV-1/metabolismo , Humanos , Índia , Tipagem Molecular , Neurônios/patologia , Neurônios/virologia , Filogenia , Filogeografia , Cultura Primária de Células , Virulência
15.
PLoS One ; 9(7): e97984, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24988390

RESUMO

Human endogenous retroviruses (HERVs) are differentially expressed depending on the cell type and physiological circumstances. HERV-K has been implicated in the pathogenesis of several diseases although the functional consequences of its expression remain unknown. Human immunodeficiency virus (HIV) infection causes neuroinflammation with neuronal damage and death. Herein, we investigated HERV-K(II)/(HML-2) envelope (Env) expression and its actions in the brain during HIV/AIDS. HERV-K(II) Env expression was assessed in healthy brain tissues, autopsied HIV HIV- infected (HIV+) and uninfected (HIV-) brains and in neural cell cultures by real time RT-PCR, massively parallel (deep) sequencing, immunoblotting and immunohistochemistry. Neuronal and neural stem cells expressing HERV-K(II) Env were analyzed in assays of host responses including cellular viability, immune responses and neurobehavioral outcomes. Deep sequencing of human brain transcriptomes disclosed that RNA sequences encoded by HERV-K were among the most abundant HERV sequences detected in human brain. Comparison of different cell types revealed that HERV-K(II) env RNA abundance was highest in cultured human neurons but was suppressed by epidermal growth factor exposure. HERV-K(II) Env immunoreactivity was increased in the cerebral cortex from persons with HIV/AIDS, principally localized in neurons. Human neuronal cells transfected with HERV-K(II) Env exhibited increased NGF and BDNF expression. Expression of HERV-K(II) Env in neuronal cells increased cellular viability and prevented neurotoxicity mediated by HIV-1 Vpr. Intracerebral delivery of HERV-K(II) Env expressed by neural stem cells suppressed TNF-α expression and microglial activation while also improving neurobehavioral deficits in vpr/RAG1-/- mice. HERV-K(II) Env was highly expressed in human neurons, especially during HIV/AIDS, but in addition exerted neuroprotective effects. These findings imply that HERV gene products might exert adaptive effects in circumstances of pathophysiological stress, perhaps underlying the conservation of HERVs within the human genome.


Assuntos
Retrovirus Endógenos/metabolismo , HIV/metabolismo , Neurônios/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células Cultivadas , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Córtex Cerebral/virologia , Fator de Crescimento Epidérmico/farmacologia , HIV/patogenicidade , Sequenciamento de Nucleotídeos em Larga Escala , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Fator de Crescimento Neural/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/virologia , Neurônios/citologia , Neurônios/virologia , Análise de Sequência de DNA , Regulação para Cima/efeitos dos fármacos , Proteínas do Envelope Viral/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo
16.
Retrovirology ; 11: 35, 2014 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-24886384

RESUMO

BACKGROUND: Human immunodeficiency virus type 1(HIV-1) infects and activates innate immune cells in the brain resulting in inflammation and neuronal death with accompanying neurological deficits. Induction of inflammasomes causes cleavage and release of IL-1ß and IL-18, representing pathogenic processes that underlie inflammatory diseases although their contribution HIV-associated brain disease is unknown. RESULTS: Investigation of inflammasome-associated genes revealed that IL-1ß, IL-18 and caspase-1 were induced in brains of HIV-infected persons and detected in brain microglial cells. HIV-1 infection induced pro-IL-1ß in human microglia at 4 hr post-infection with peak IL-1ß release at 24 hr, which was accompanied by intracellular ASC translocation and caspase-1 activation. HIV-dependent release of IL-1ß from a human macrophage cell line, THP-1, was inhibited by NLRP3 deficiency and high extracellular [K+]. Exposure of microglia to HIV-1 gp120 caused IL-1ß production and similarly, HIV-1 envelope pseudotyped viral particles induced IL-1ß release, unlike VSV-G pseudotyped particles. Infection of cultured feline macrophages by the related lentivirus, feline immunodeficiency virus (FIV), also resulted in the prompt induction of IL-1ß. In vivo FIV infection activated multiple inflammasome-associated genes in microglia, which was accompanied by neuronal loss in cerebral cortex and neurological deficits. Multivariate analyses of data from FIV-infected and uninfected animals disclosed that IL-1ß, NLRP3 and caspase-1 expression in cerebral cortex represented key molecular determinants of neurological deficits. CONCLUSIONS: NLRP3 inflammasome activation was an early and integral aspect of lentivirus infection of microglia, which was associated with lentivirus-induced brain disease. Inflammasome activation in the brain might represent a potential target for therapeutic interventions in HIV/AIDS.


Assuntos
Síndrome da Imunodeficiência Adquirida/metabolismo , Síndrome da Imunodeficiência Adquirida/virologia , Encefalopatias/metabolismo , Encefalopatias/virologia , Infecções por HIV/metabolismo , HIV-1 , Inflamassomos/metabolismo , Microglia/metabolismo , Animais , Caspase 1/metabolismo , Gatos , Linhagem Celular , Córtex Cerebral/metabolismo , Córtex Cerebral/virologia , Feminino , Infecções por HIV/virologia , Humanos , Vírus da Imunodeficiência Felina , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Macrófagos/virologia , Microglia/virologia , Gravidez
17.
Gene Ther ; 21(1): 28-36, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24131981

RESUMO

Canine models have many advantages for evaluating therapy of human central nervous system (CNS) diseases. In contrast to nonhuman primate models, naturally occurring canine CNS diseases are common. In contrast to murine models, the dog's lifespan is long, its brain is large and the diseases affecting it commonly have the same molecular, pathological and clinical phenotype as the human diseases. We compared the ability of four intracerebrally injected adeno-associated virus vector (AAV) serotypes to transduce the dog brain with green fluorescent protein as the first step in using these vectors to evaluate both delivery and efficacy in naturally occurring canine homologs of human diseases. Quantitative measures of transduction, maximum diameter and area, identified both AAV2/9 and AAV2/rh10 as significantly more efficient than either AAV2/1 or AAV2/5 at transducing cerebral cortex, caudate nucleus, thalamus and internal capsule. Fluorescence co-labeling with cell-type-specific antibodies demonstrated that AAV2/9 and AAV2/rh10 were capable of primarily transducing neurons, although glial transduction was also identified and found to be more efficient with the AAV2/9 vector. These data are a prerequisite to evaluating the efficacy of recombinant AAV vectors carrying disease-modifying transgenes to treat naturally occurring canine models in preclinical studies of human CNS disease therapy.


Assuntos
Encéfalo/metabolismo , Dependovirus/genética , Vetores Genéticos , Transdução Genética , Animais , Encéfalo/virologia , Núcleo Caudado/metabolismo , Núcleo Caudado/virologia , Córtex Cerebral/metabolismo , Córtex Cerebral/virologia , Dependovirus/classificação , Dependovirus/fisiologia , Modelos Animais de Doenças , Cães , Proteínas de Fluorescência Verde/genética , Humanos , Cápsula Interna/metabolismo , Cápsula Interna/virologia , Sorotipagem , Tálamo/metabolismo , Tálamo/virologia , Transgenes
18.
J Neurosci Methods ; 220(1): 1-8, 2013 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-23988614

RESUMO

BACKGROUND: Convection-enhanced delivery (CED) is currently under investigation for delivering therapeutic agents to subcortical targets in the brain. Direct delivery of therapies to the cerebral cortex, however, remains a significant challenge. NEW METHOD: We describe a novel method of targeting adeno-associated viral vector (AAV) mediated gene therapies to specific cerebral cortical regions by performing high volume, high flow rate infusions into underlying white matter in a large animal (porcine) model. RESULTS: Infusion volumes of up to 700 µl at flow rates as high as 10 µl/min were successfully performed in white matter without adverse neurological sequelae. Co-infusion of AAV2/5-GFP with 0.2% Gadolinium in artificial CSF confirmed transgene expression in the deep layers of cerebral cortex overlying the infused areas of white matter. COMPARISON WITH EXISTING METHODS: AAV-mediated gene therapies have been previously targeted to the cerebral cortex by performing intrathalamic CED and exploiting axonal transport. The novel method described in this study facilitates delivery of gene therapies to specific regions of the cerebral cortex without targeting deep brain structures. CONCLUSIONS: AAV-mediated gene therapies can be targeted to specific cortical regions by performing CED into underlying white matter. This technique could be applied to the treatment of neurological disorders characterised by cerebral cortical degeneration.


Assuntos
Córtex Cerebral/virologia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Fibras Nervosas Mielinizadas/virologia , Animais , Convecção , Dependovirus , Proteínas de Fluorescência Verde/genética , Imuno-Histoquímica , Infusões Intraventriculares , Imageamento por Ressonância Magnética , Suínos , Transgenes
19.
J Neurovirol ; 19(3): 209-18, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23613008

RESUMO

Emerging evidence suggests that CNS injury and neurocognitive impairment persist in the setting of chronic HIV infection and combination antiretroviral therapy (CART). Yet, whether neurological injury can progress in this setting remains uncertain. Magnetic resonance spectroscopy and neurocognitive and clinical assessments were performed over 2 years in 226 HIV-infected individuals on stable CART, including 138 individuals who were neurocognitively asymptomatic (NA). Concentrations of N-acetylaspartate (NAA), creatine (Cr), choline (Cho), myoinositol, and glutamate/glutamine (Glx) were measured in the midfrontal cortex (MFC), frontal white matter (FWM), and basal ganglia (BG). Longitudinal changes in metabolite levels were determined using linear mixed effect models, as were metabolite changes in relation to global neurocognitive function. HIV-infected subjects showed significant annual decreases in brain metabolite levels in all regions examined, including NAA (2.95 %) and Cho (2.61 %) in the FWM; NAA (1.89 %), Cr (1.84 %), Cho (2.19 %), and Glx (6.05 %) in the MFC; and Glx (2.80 %) in the BG. Similar metabolite decreases were observed in the NA and subclinically impaired subgroups, including subjects with virologic suppression in plasma and CSF. Neurocognitive decline was associated with longitudinal decreases in Glx in the FWM and the BG, and in NAA in the BG. Widespread progressive changes in the brain, including neuronal injury, occur in chronically HIV-infected persons despite stable antiretroviral treatment and virologic suppression and can lead to neurocognitive declines. The basis for these findings is poorly understood and warrants further study.


Assuntos
Complexo AIDS Demência/tratamento farmacológico , Complexo AIDS Demência/patologia , Fármacos Anti-HIV/uso terapêutico , Gânglios da Base/patologia , Córtex Cerebral/patologia , Complexo AIDS Demência/metabolismo , Complexo AIDS Demência/virologia , Adulto , Terapia Antirretroviral de Alta Atividade , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Gânglios da Base/efeitos dos fármacos , Gânglios da Base/metabolismo , Gânglios da Base/virologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/virologia , Colina/metabolismo , Cognição/fisiologia , Progressão da Doença , Feminino , Ácido Glutâmico/metabolismo , HIV/efeitos dos fármacos , HIV/fisiologia , Humanos , Inositol/metabolismo , Masculino , Pessoa de Meia-Idade , Testes Neuropsicológicos
20.
Hum Gene Ther ; 24(5): 526-32, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23517473

RESUMO

The present study builds on previous work showing that infusion of adeno-associated virus type 9 (AAV9) into the cisterna magna (CM) of nonhuman primates resulted in widespread transduction throughout cortex and spinal cord. Transduction efficiency was severely limited, however, by the presence of circulating anti-AAV antibodies. Accordingly, we compared AAV9 to a related serotype, AAV7, which has a high capsid homology. CM infusion of either AAV7 or AAV9 directed high level of cell transduction with similar patterns of distribution throughout brain cortex and along the spinal cord. Dorsal root ganglia and corticospinal tracts were also transduced. Both astrocytes and neurons were transduced. Interestingly, little transduction was observed in peripheral organs. Our results indicate that intrathecal delivery of either AAV7 or AAV9 directs a robust and widespread cellular transduction in the central nervous system and other peripheral neural structures.


Assuntos
Dependovirus/genética , Técnicas de Transferência de Genes/efeitos adversos , Vetores Genéticos/efeitos adversos , Macaca/genética , Transdução Genética , Animais , Astrócitos/patologia , Astrócitos/virologia , Córtex Cerebral/patologia , Córtex Cerebral/virologia , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Gânglios Espinais/virologia , Vetores Genéticos/líquido cefalorraquidiano , Proteínas de Fluorescência Verde/genética , Macaca/virologia , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Neurônios Motores/virologia , Medula Espinal/patologia , Medula Espinal/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA