Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Am J Pathol ; 188(12): 2853-2862, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30273599

RESUMO

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that primarily affects motor neurons in the cerebral cortex, brainstem, and spinal cord, leading to progressive paralysis and eventual death. Approximately 95% of all ALS cases are sporadic without known causes. Enteroviruses have been suspected to play a role in ALS because of their ability to target motor neurons and to cause muscle weakness and paralysis. In vitro enteroviral infection results in cytoplasmic aggregation and cleavage of transactive response DNA binding protein-43, a pathologic hallmark of ALS. However, whether enteroviral infection can induce ALS-like pathologies in vivo remains to be characterized. In this study, neonatal BALB/C mice were intracranially inoculated with either a recombinant coxsackievirus B3 expressing enhanced green fluorescent protein or mock-infected for 2, 5, 10, 30, and 90 days. Histologic and immunohistochemical analysis of brain tissues demonstrated sustained inflammation (microglia and astrogliosis) and lesions in multiple regions of the brain (hippocampus, cerebral cortex, striatum, olfactory bulb, and putamen) in parallel with virus detection as early as 2 days for up to 90 days after infection. Most notably, ALS-like pathologies, including cytoplasmic mislocalization of transactive response DNA binding protein-43, p62-, and ubiquitin-positive inclusions, were observed in the areas of infection. These data provide the first pathologic evidence to support a possible link between enteroviral infection and ALS.


Assuntos
Esclerose Lateral Amiotrófica/patologia , Encéfalo/imunologia , Infecções por Coxsackievirus/complicações , Citoplasma/metabolismo , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Enterovirus Humano B/patogenicidade , Esclerose Lateral Amiotrófica/etiologia , Esclerose Lateral Amiotrófica/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/microbiologia , Células Cultivadas , Infecções por Coxsackievirus/virologia , Camundongos , Camundongos Endogâmicos BALB C , Transporte Proteico
2.
Lab Invest ; 94(2): 161-81, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24378643

RESUMO

Coxsackievirus B3 (CVB3) and lymphocytic choriomeningitis virus (LCMV) are both neurotropic RNA viruses, which can establish a persistent infection and cause meningitis and encephalitis in the neonatal host. Utilizing our neonatal mouse model of infection, we evaluated the consequences of early viral infection upon the host central nervous system (CNS) by comparing CVB3 and LCMV infection. Both viruses expressed high levels of viral protein in the choroid plexus and subventricular zone (SVZ), a region of neurogenesis. LCMV infected a greater number of cells in the SVZ and targeted both nestin(+) (neural progenitor cell marker) and olig2(+) (glial progenitor marker) cells at a relatively equal proportion. In contrast, CVB3 preferentially infected nestin(+) cells within the SVZ. Microarray analysis revealed differential kinetics and unique host gene expression changes for each infection. MHC class I gene expression, several developmental-related Hox genes, and transthyretin (TTR), a protein secreted in the cerebrospinal fluid by the choroid plexus, were specifically downregulated following CVB3 infection. Also, we identified severe pathology in the choroid plexus of CVB3-infected animals at 48 h post infection accompanied by a decrease in the level of TTR and carbonic anhydrase II. These results demonstrate broader neural progenitor and stem cell (NPSC) tropism for LCMV in the neonatal CNS, whereas CVB3 targeted a more specific subset of NPSCs, stimulated a distinct early immune response, and induced significant acute damage in the choroid plexus.


Assuntos
Sistema Nervoso Central/virologia , Plexo Corióideo/patologia , Infecções por Coxsackievirus/imunologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/fisiologia , Coriomeningite Linfocítica/imunologia , Células-Tronco Neurais/fisiologia , Animais , Anidrase Carbônica II/metabolismo , Plexo Corióideo/metabolismo , Infecções por Coxsackievirus/patologia , Imunofluorescência , Regulação da Expressão Gênica/genética , Coriomeningite Linfocítica/patologia , Camundongos , Análise em Microsséries , Nestina/metabolismo , Células-Tronco Neurais/virologia
3.
Am J Pathol ; 180(3): 1107-1120, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22214838

RESUMO

Coxsackieviruses are significant human pathogens causing myocarditis, meningitis, and encephalitis. We previously demonstrated the ability of coxsackievirus B3 (CVB3) to persist within the neonatal central nervous system (CNS) and to target neural stem cells. Given that CVB3 is a cytolytic virus and may therefore damage target cells, we characterized the potential reduction in neurogenesis within the developing brain and the subsequent developmental defects that occurred after the loss of these essential neural stem cells. Neonatal mice were inoculated with a recombinant CVB3 expressing eGFP (eGFP-CVB3), and alterations in neurogenesis and brain development were evaluated over time. We observed a reduction in proliferating cells in CNS neurogenic regions simultaneously with the presence of nestin(+) cells undergoing apoptosis. The size of the brain appeared smaller by histology, and a permanent decrease in brain wet weight was observed after eGFP-CVB3 infection. We also observed an inverse relationship between the amount of virus material and brain wet weight up to day 30 postinfection. In addition, signs of astrogliosis and a compaction of the cortical layers were observed at 90 days postinfection. Intriguingly, partial brain wet weight recovery was observed in mice treated with the antiviral drug ribavirin during the persistent stage of infection. Hence, long-term neurological sequelae might be expected after neonatal enteroviral infections, yet antiviral treatment initiated long after the end of acute infection might limit virus-mediated neuropathology.


Assuntos
Sistema Nervoso Central/virologia , Infecções por Coxsackievirus/complicações , Enterovirus Humano B , Células-Tronco Neurais/virologia , Neurogênese/fisiologia , Animais , Animais Recém-Nascidos , Antivirais/farmacologia , Apoptose/fisiologia , Astrócitos/virologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/virologia , Divisão Celular , Proliferação de Células , Sistema Nervoso Central/crescimento & desenvolvimento , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Tamanho do Órgão , Proteínas Recombinantes , Carga Viral
4.
J Virol ; 85(12): 5718-32, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21471247

RESUMO

Enteroviruses, including coxsackieviruses, exhibit significant tropism for the central nervous system, and these viruses are commonly associated with viral meningitis and encephalitis. Previously, we described the ability of coxsackievirus B3 (CVB3) to infect proliferating neuronal progenitor cells located in the neonatal subventricular zone and persist in the adult murine central nervous system (CNS). Here, we demonstrate that cultured murine neurospheres, which comprise neural stem cells and their progeny at different stages of development, were highly susceptible to CVB3 infection. Neurospheres, or neural progenitor and stem cells (NPSCs), isolated from neonatal C57BL/6 mice, supported high levels of infectious virus production and high viral protein expression levels following infection with a recombinant CVB3 expressing enhanced green fluorescent protein (eGFP) protein. Similarly, NPSCs isolated from neonatal actin-promoter-GFP transgenic mice (actin-GFP NPSCs) were highly susceptible to infection with a recombinant CVB3 expressing DsRed (Discosoma sp. red fluorescent protein). Both nestin-positive and NG2(+) progenitor cells within neurospheres were shown to preferentially express high levels of viral protein as soon as 24 h postinfection (p.i.). By day 3 p.i., viral protein expression and viral titers increased dramatically in NPSCs with resultant cytopathic effects (CPE) and eventual cell death. In contrast, reduced viral replication, lower levels of CPE, and diminished viral protein expression levels were observed in NPSCs differentiated for 5 or 16 days in the presence of fetal bovine serum (FBS). Despite the presence of CPE and high levels of cell death following early CVB3 infection, surviving neurospheres were readily observed and continued to express detectable levels of viral protein as long as 37 days after initial infection. Also, CVB3 infection of actin-GFP NPSCs increased the percentage of cells expressing neuronal class III ß-tubulin following their differentiation in the presence of FBS. These results suggest that neural stem cells may be preferentially targeted by CVB3 and that neurogenic regions of the CNS may support persistent viral replication in the surviving host. In addition, normal progenitor cell differentiation may be altered in the host following infection.


Assuntos
Diferenciação Celular , Enterovirus Humano B/fisiologia , Enterovirus Humano B/patogenicidade , Células-Tronco Neurais/virologia , Animais , Células Cultivadas , Efeito Citopatogênico Viral , Enterovirus Humano B/genética , Enterovirus Humano B/ultraestrutura , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Células-Tronco Neurais/citologia , Células-Tronco Neurais/ultraestrutura , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral , Proteína Vermelha Fluorescente
5.
J Neurosci ; 30(25): 8676-91, 2010 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-20573913

RESUMO

Enterovirus infection in newborn infants is a significant cause of aseptic meningitis and encephalitis. Using a neonatal mouse model, we previously determined that coxsackievirus B3 (CVB3) preferentially targets proliferating neural stem cells located in the subventricular zone within 24 h after infection. At later time points, immature neuroblasts, and eventually mature neurons, were infected as determined by expression of high levels of viral protein. Here, we show that blood-derived Mac3(+) mononuclear cells were rapidly recruited to the CNS within 12 h after intracranial infection with CVB3. These cells displayed a myeloid-like morphology, were of a peripheral origin based on green fluorescent protein (GFP)-tagged adoptive cell transplant examination, and were highly susceptible to CVB3 infection during their migration into the CNS. Serial immunofluorescence images suggested that the myeloid cells enter the CNS via the choroid plexus, and that they may be infected during their extravasation and passage through the choroid plexus epithelium; these infected myeloid cells ultimately penetrate into the parenchyma of the brain. Before their migration through the ependymal cell layer, a subset of these infected myeloid cells expressed detectable levels of nestin, a marker for neural stem and progenitor cells. As these nestin(+) myeloid cells infected with CVB3 migrated through the ependymal cell layer, they revealed distinct morphological characteristics typical of type B neural stem cells. The recruitment of these novel myeloid cells may be specifically set in motion by the induction of a unique chemokine profile in the CNS induced very early after CVB3 infection, which includes upregulation of CCL12. We propose that intracranial CVB3 infection may lead to the recruitment of nestin(+) myeloid cells into the CNS which might represent an intrinsic host CNS repair response. In turn, the proliferative and metabolic status of recruited myeloid cells may render them attractive targets for CVB3 infection. Moreover, the migratory ability of these myeloid cells may point to a productive method of virus dissemination within the CNS.


Assuntos
Infecções por Coxsackievirus/virologia , Células Mieloides/virologia , Animais , Animais Recém-Nascidos , Plexo Corióideo/imunologia , Plexo Corióideo/virologia , Infecções por Coxsackievirus/imunologia , Imunofluorescência , Hibridização In Situ , Camundongos , Microscopia Confocal , Células Mieloides/imunologia , Neurônios/imunologia , Neurônios/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/imunologia , Células-Tronco/virologia
6.
J Virol ; 83(18): 9356-69, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19570873

RESUMO

Coxsackieviruses are significant human pathogens, and the neonatal central nervous system (CNS) is a major target for infection. Despite the extreme susceptibility of newborn infants to coxsackievirus infection and viral tropism for the CNS, few studies have been aimed at determining the long-term consequences of infection on the developing CNS. We previously described a neonatal mouse model of coxsackievirus B3 (CVB3) infection and determined that proliferating stem cells in the CNS were preferentially targeted. Here, we describe later stages of infection, the ensuing inflammatory response, and subsequent lesions which remain in the adult CNS of surviving animals. High levels of type I interferons and chemokines (in particular MCP-5, IP10, and RANTES) were upregulated following infection and remained at high levels up to day 10 postinfection (p.i). Chronic inflammation and lesions were observed in the hippocampus and cortex of surviving mice for up to 9 months p.i. CVB3 RNA was detected in the CNS up to 3 months p.i at high abundance ( approximately 10(6) genomes/mouse brain), and viral genomic material remained detectable in culture after two rounds of in vitro passage. These data suggest that CVB3 may persist in the CNS as a low-level, noncytolytic infection, causing ongoing inflammatory lesions. Thus, the effects of a relatively common infection during the neonatal period may be long lasting, and the prognosis for newborn infants recovering from acute infection should be reexplored.


Assuntos
Sistema Nervoso Central/virologia , Infecções por Coxsackievirus/patologia , Enterovirus/patogenicidade , Animais , Animais Recém-Nascidos , Córtex Cerebral/patologia , Córtex Cerebral/virologia , Quimiocinas/análise , Doença Crônica , Infecções por Coxsackievirus/imunologia , Enterovirus/genética , Enterovirus Humano B , Genoma Viral , Hipocampo/patologia , Hipocampo/virologia , Humanos , Inflamação , Interferon Tipo I/análise , Camundongos , RNA Viral/sangue , Fatores de Tempo , Regulação para Cima
7.
J Vis Exp ; (105): e52848, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26555373

RESUMO

The first mouse kidney transplant technique was published in 1973(1) by the Russell laboratory. Although it took some years for other labs to become proficient in and utilize this technique, it is now widely used by many laboratories around the world. A significant refinement to the original technique using the donor aorta to form the arterial anastomosis instead of the renal artery was developed and reported in 1993 by Kalina and Mottram (2) with a further advancement coming from the same laboratory in 1999 (3). While one can become proficient in this model, a search of the literature reveals that many labs still experience a high proportion of graft loss due to arterial thrombosis. We describe here a technique that was devised in our laboratory that vastly reduces the arterial thrombus reported by others (4,5). This is achieved by forming a heel-and-toe cuff of the donor infra-renal aorta that facilitates a larger anastomosis and straighter blood flow into the kidney.


Assuntos
Anastomose Arteriovenosa/cirurgia , Transplante de Rim/veterinária , Rim/cirurgia , Animais , Hemodinâmica , Rim/irrigação sanguínea , Transplante de Rim/métodos , Camundongos , Procedimentos Cirúrgicos Vasculares
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA