Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Pathol ; 188(12): 2853-2862, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30273599

RESUMEN

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.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Encéfalo/inmunología , Infecciones por Coxsackievirus/complicaciones , Citoplasma/metabolismo , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Enterovirus Humano B/patogenicidad , Esclerosis Amiotrófica Lateral/etiología , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/microbiología , Células Cultivadas , Infecciones por Coxsackievirus/virología , Ratones , Ratones Endogámicos BALB C , Transporte de Proteínas
2.
J Vis Exp ; (105): e52848, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26555373

RESUMEN

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.


Asunto(s)
Anastomosis Arteriovenosa/cirugía , Trasplante de Riñón/veterinaria , Riñón/cirugía , Animales , Hemodinámica , Riñón/irrigación sanguínea , Trasplante de Riñón/métodos , Ratones , Procedimientos Quirúrgicos Vasculares
3.
Cancer Cell ; 27(3): 354-69, 2015 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-25759021

RESUMEN

Many tumor cells are fueled by altered metabolism and increased glutamine (Gln) dependence. We identify regulation of the L-glutamine carrier proteins SLC1A5 and SLC38A2 (SLC1A5/38A2) by the ubiquitin ligase RNF5. Paclitaxel-induced ER stress to breast cancer (BCa) cells promotes RNF5 association, ubiquitination, and degradation of SLC1A5/38A2. This decreases Gln uptake, levels of TCA cycle components, mTOR signaling, and proliferation while increasing autophagy and cell death. Rnf5-deficient MMTV-PyMT mammary tumors were less differentiated and showed elevated SLC1A5 expression. Whereas RNF5 depletion in MDA-MB-231 cells promoted tumorigenesis and abolished paclitaxel responsiveness, SLC1A5/38A2 knockdown elicited opposing effects. Inverse RNF5(hi)/SLC1A5/38A2(lo) expression was associated with positive prognosis in BCa. Thus, RNF5 control of Gln uptake underlies BCa response to chemotherapies.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/metabolismo , Sistema de Transporte de Aminoácidos A/metabolismo , Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Proteínas de Unión al ADN/fisiología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Paclitaxel/farmacología , Ubiquitina-Proteína Ligasas/fisiología , Sistema de Transporte de Aminoácidos A/genética , Sistema de Transporte de Aminoácidos ASC/genética , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Ciclo del Ácido Cítrico/efectos de los fármacos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Femenino , Humanos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Antígenos de Histocompatibilidad Menor , Paclitaxel/uso terapéutico , Proteolisis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
4.
Lab Invest ; 94(2): 161-81, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24378643

RESUMEN

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.


Asunto(s)
Sistema Nervioso Central/virología , Plexo Coroideo/patología , Infecciones por Coxsackievirus/inmunología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/fisiología , Coriomeningitis Linfocítica/inmunología , Células-Madre Neurales/fisiología , Animales , Anhidrasa Carbónica II/metabolismo , Plexo Coroideo/metabolismo , Infecciones por Coxsackievirus/patología , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica/genética , Coriomeningitis Linfocítica/patología , Ratones , Análisis por Micromatrices , Nestina/metabolismo , Células-Madre Neurales/virología
5.
Pigment Cell Melanoma Res ; 26(1): 136-42, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23035722

RESUMEN

To date, there are no effective therapies for tumors bearing NRAS mutations, which are present in 15-20% of human melanomas. Here we extend our earlier studies where we demonstrated that the small molecule BI-69A11 inhibits the growth of melanoma cell lines. Gene expression analysis revealed the induction of interferon- and cell death-related genes that were associated with responsiveness of melanoma cell lines to BI-69A11. Strikingly, the administration of BI-69A11 inhibited melanoma development in genetically modified mice bearing an inducible form of activated Nras and a deletion of the Ink4a gene (Nras((Q61K)) ::Ink4a(-/-) ). Biweekly administration of BI-69A11 starting at 10 weeks or as late as 24 weeks after the induction of mutant Nras expression inhibited melanoma development (100 and 36%, respectively). BI-69A11 treatment did not inhibit the development of histiocytic sarcomas, which constitute about 50% of the tumors in this model. BI-69A11-resistant Nras((Q61K)) ::Ink4a(-/-) tumors exhibited increased CD45 expression, reflective of immune cell infiltration and upregulation of gene networks associated with the cytoskeleton, DNA damage response, and small molecule transport. The ability to attenuate the development of NRAS mutant melanomas supports further development of BI-69A11 for clinical assessment.


Asunto(s)
Sustitución de Aminoácidos/genética , Bencimidazoles/uso terapéutico , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Melanoma/tratamiento farmacológico , Lesiones Precancerosas/tratamiento farmacológico , Quinolonas/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Proteínas ras/genética , Animales , Bencimidazoles/administración & dosificación , Bencimidazoles/farmacología , Proliferación Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inyecciones Intraperitoneales , Antígeno Ki-67/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Melanoma/genética , Melanoma/patología , Ratones , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas B-raf/genética , Quinolonas/administración & dosificación , Quinolonas/farmacología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Am J Pathol ; 180(3): 1107-1120, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22214838

RESUMEN

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.


Asunto(s)
Sistema Nervioso Central/virología , Infecciones por Coxsackievirus/complicaciones , Enterovirus Humano B , Células-Madre Neurales/virología , Neurogénesis/fisiología , Animales , Animales Recién Nacidos , Antivirales/farmacología , Apoptosis/fisiología , Astrocitos/virología , Encéfalo/crecimiento & desarrollo , Encéfalo/virología , División Celular , Proliferación Celular , Sistema Nervioso Central/crecimiento & desarrollo , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Endogámicos BALB C , Tamaño de los Órganos , Proteínas Recombinantes , Carga Viral
7.
J Virol ; 85(12): 5718-32, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21471247

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Enterovirus Humano B/fisiología , Enterovirus Humano B/patogenicidad , Células-Madre Neurales/virología , Animales , Células Cultivadas , Efecto Citopatogénico Viral , Enterovirus Humano B/genética , Enterovirus Humano B/ultraestructura , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Células-Madre Neurales/citología , Células-Madre Neurales/ultraestructura , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral , Proteína Fluorescente Roja
8.
J Neurosci ; 30(25): 8676-91, 2010 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-20573913

RESUMEN

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.


Asunto(s)
Infecciones por Coxsackievirus/virología , Células Mieloides/virología , Animales , Animales Recién Nacidos , Plexo Coroideo/inmunología , Plexo Coroideo/virología , Infecciones por Coxsackievirus/inmunología , Técnica del Anticuerpo Fluorescente , Hibridación in Situ , Ratones , Microscopía Confocal , Células Mieloides/inmunología , Neuronas/inmunología , Neuronas/virología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/inmunología , Células Madre/virología
9.
J Virol ; 83(18): 9356-69, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19570873

RESUMEN

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.


Asunto(s)
Sistema Nervioso Central/virología , Infecciones por Coxsackievirus/patología , Enterovirus/patogenicidad , Animales , Animales Recién Nacidos , Corteza Cerebral/patología , Corteza Cerebral/virología , Quimiocinas/análisis , Enfermedad Crónica , Infecciones por Coxsackievirus/inmunología , Enterovirus/genética , Enterovirus Humano B , Genoma Viral , Hipocampo/patología , Hipocampo/virología , Humanos , Inflamación , Interferón Tipo I/análisis , Ratones , ARN Viral/sangre , Factores de Tiempo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA