Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Bases de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Immunol ; 186(11): 6398-405, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21525387

RESUMEN

Coxsackievirus B3 (CVB3) infection is the most common cause of viral myocarditis. The pathogenesis of viral myocarditis is strongly controlled by host genetic factors. Although certain indispensable components of immunity have been identified, the genes and pathways underlying natural variation between individuals remain unclear. Previously, we isolated the viral myocarditis susceptibility 1 (Vms1) locus on chromosome 3, which influences pathogenesis. We hypothesized that confirmation and further study of Vms1 controlling CVB3-mediated pathology, combined with pathway analysis and consomic mapping approaches, would elucidate both pathological and protective mechanisms accounting for natural variation in response to CVB3 infection. Vms1 was originally mapped to chromosome 3 using a segregating cross between susceptible A/J and resistant B10.A mice. To validate Vms1, C57BL/6J-Chr 3(A)/NaJ (a chromosome substitution strain that carries a diploid A/J chromosome 3) were used to replicate susceptibility compared with resistant C57BL/6J (B6). A second segregating F2 cross was generated between these, confirming both the localization and effects of Vms1. Microarray analysis of the four strains (A/J, B10.A, C57BL/6J, and C57BL/6J-Chr 3(A)/NaJ) illuminated a core program of response to CVB3 in all strains that is comprised mainly of IFN-stimulated genes. Microarray analysis also revealed strain-specific differential expression programs and genes that may be prognostic or diagnostic of susceptibility to CVB3 infection. A combination of analyses revealed very strong evidence for the existence and location of Vms1. Differentially expressed pathways were identified by microarray, and candidate gene analysis revealed Fpgt, H28, and Tnni3k as likely candidates for Vms1.


Asunto(s)
Infecciones por Coxsackievirus/genética , Predisposición Genética a la Enfermedad/genética , Miocarditis/genética , Sitios de Carácter Cuantitativo/genética , Animales , Mapeo Cromosómico/métodos , Infecciones por Coxsackievirus/inmunología , Enterovirus Humano B/inmunología , Enterovirus Humano B/fisiología , Femenino , Perfilación de la Expresión Génica , Variación Genética , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Masculino , Ratones , Ratones Endogámicos A , Ratones Endogámicos C57BL , Antígenos de Histocompatibilidad Menor/genética , Miocarditis/inmunología , Miocardio/metabolismo , Miocardio/patología , Nucleotidiltransferasas/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Factores de Tiempo
2.
Physiol Genomics ; 44(17): 843-52, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22805347

RESUMEN

Epidemiological studies show that high HDL-cholesterol (HDLc) decreases the risk of cardiovascular disease. To map genes controlling lipid metabolism, particularly HDLc levels, we screened the plasma lipids of 36 AcB/BcA RC mouse strains subjected to either a normal or a high-fat/cholesterol diet. Strains BcA68 and AcB65 showed deviant HDLc plasma levels compared with the parental A/J and C57BL/6J strains; they were thus selected to generate informative F2 crosses. Linkage analyses in the AcB65 strain identified a locus on chromosome 4 (Hdlq78) responsible for high post-high fat diet HDLc levels. This locus has been previously associated at genome-wide significance to two regions in the human genome. A second linkage analysis in strain BcA68 identified linkage in the vicinity of a gene cluster known to control HDLc levels. Sequence analysis of these candidates identified a de novo, loss-of-function mutation in the ApoA1 gene of BcA68 that prematurely truncates the ApoA1 protein. The possibility of dissecting the specific effects of this new ApoA1 deficiency in the context of isogenic controls makes the BcA68 mouse a valuable new tool.


Asunto(s)
Apolipoproteína A-I/genética , HDL-Colesterol/sangre , HDL-Colesterol/genética , Dieta Alta en Grasa , Ratones Congénicos/genética , Animales , Secuencia de Bases , Mapeo Cromosómico , Cruzamientos Genéticos , Sitios Genéticos/genética , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Mutación/genética , Análisis de Secuencia de ADN , Especificidad de la Especie
3.
J Innate Immun ; 7(3): 315-30, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25675947

RESUMEN

Coxsackievirus strain B serotype 3 (CVB3)-induced myocarditis is an important human disease that causes permanent tissue damage and can lead to death from acute infection or long-term morbidity caused by chronic inflammation. The timing and magnitude of immune activation following CVB3 infection can mediate a positive host outcome or increase tissue pathology. To better elucidate the role of endosomal Toll-like receptor (TLR) signaling in acute CVB3 infection, we studied mice with a loss-of-function mutation, known as Letr for 'loss of endosomal TLR response', in Unc93b1, which is a chaperone protein for TLR3, TLR7 and TLR9. Using Unc93b1(Letr/)(Letr) mice, we determined that Unc93b1-dependent TLR activation was essential for the survival of acute CVB3-induced myocarditis. We also determined that a lack of endosomal TLR signaling was associated with a higher viral load in target organs and that it increased inflammation, necrosis and fibrosis in cardiac tissue. Loss of Unc93b1 function was also associated with increased cardiac expression of Ifn-b and markers of tissue injury and fibrosis including Lcn2 and Serpina3n early after CVB3 infection. These observations establish a significant role for Unc93b1 in the regulation of the host inflammatory response to CVB3 infection and also reveal potential mediators of host tissue damage that merit further investigation in acute viral myocarditis.


Asunto(s)
Infecciones por Coxsackievirus/inmunología , Endosomas/inmunología , Enterovirus Humano B/inmunología , Proteínas de Transporte de Membrana/inmunología , Miocarditis/inmunología , Transducción de Señal/inmunología , Receptores Toll-Like/inmunología , Proteínas de Fase Aguda/inmunología , Animales , Infecciones por Coxsackievirus/patología , Endosomas/patología , Células HeLa , Humanos , Interferón beta/inmunología , Lipocalina 2 , Lipocalinas/inmunología , Ratones , Miocarditis/patología , Proteínas Oncogénicas/inmunología , Serpinas/inmunología
4.
PLoS One ; 10(9): e0138222, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26375467

RESUMEN

Coxsackievirus type B3 (CVB3) is a cardiotropic enterovirus. Infection causes cardiomyocyte necrosis and myocardial inflammation. The damaged tissue that results is replaced with fibrotic or calcified tissue, which can lead to permanently altered cardiac function. The extent of pathogenesis among individuals exposed to CVB3 is dictated by a combination of host genetics, viral virulence, and the environment. Here, we aimed to identify genes that modulate cardiopathology following CVB3 infection. 129S1 mice infected with CVB3 developed increased cardiac pathology compared to 129X1 substrain mice despite no difference in viral burden. Linkage analysis identified a major locus on chromosome 7 (LOD: 8.307, P<0.0001) that controlled the severity of cardiac calcification and necrosis following infection. Sub-phenotyping and genetic complementation assays identified Abcc6 as the underlying gene. Microarray expression profiling identified genotype-dependent regulation of genes associated with mitochondria. Electron microscopy examination showed elevated deposition of hydroxyapatite-like material in the mitochondrial matrices of infected Abcc6 knockout (Abcc6-/-) mice but not in wildtype littermates. Cyclosporine A (CsA) inhibits mitochondrial permeability transition pore opening by inhibiting cyclophilin D (CypD). Treatment of Abcc6 -/- mice with CsA reduced cardiac necrosis and calcification by more than half. Furthermore, CsA had no effect on the CVB3-induced phenotype of doubly deficient CypD-/-Abcc6-/- mice. Altogether, our work demonstrates that mutations in Abcc6 render mice more susceptible to cardiac calcification following CVB3 infection. Moreover, we implicate CypD in the control of cardiac necrosis and calcification in Abcc6-deficient mice, whereby CypD inhibition is required for cardioprotection.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Calcinosis/tratamiento farmacológico , Infecciones por Coxsackievirus/tratamiento farmacológico , Ciclosporina/farmacología , Enterovirus Humano B/efectos de los fármacos , Inflamación/tratamiento farmacológico , Miocarditis/tratamiento farmacológico , Animales , Calcinosis/patología , Calcinosis/virología , Infecciones por Coxsackievirus/patología , Infecciones por Coxsackievirus/virología , Peptidil-Prolil Isomerasa F , Ciclofilinas/metabolismo , Femenino , Inmunosupresores/farmacología , Inflamación/patología , Inflamación/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Proteínas de Transporte de Membrana Mitocondrial/efectos de los fármacos , Poro de Transición de la Permeabilidad Mitocondrial , Proteínas Asociadas a Resistencia a Múltiples Medicamentos , Miocarditis/patología , Miocarditis/virología , Necrosis
5.
PLoS One ; 7(2): e31012, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22363534

RESUMEN

Cerebral malaria (CM) is a lethal neurological complication of malaria. We implemented a genome-wide screen in mutagenized mice to identify host proteins involved in CM pathogenesis and whose inhibition may be of therapeutic value. One pedigree (P48) segregated a resistance trait whose CM-protective effect was fully penetrant, mapped to chromosome 8, and identified by genome sequencing as homozygosity for a mis-sense mutation (W81R) in the FERM domain of Janus-associated kinase 3 (Jak3). The causative effect of Jak3(W81R) was verified by complementation testing in Jak3(W81R/-) double heterozygotes that were fully protected against CM. Jak3(W81R) homozygotes showed defects in thymic development with depletion of CD8(+) T cell, B cell, and NK cell compartments, and defective T cell-dependent production of IFN-γ. Adoptive transfer of normal splenocytes abrogates CM resistance in Jak3(W81R) homozygotes, an effect attributed to the CD8(+) T cells. Jak3(W81R) behaves as a dominant negative variant, with significant CM resistance of Jak3(W81R/+) heterozygotes, compared to CM-susceptible Jak3(+/+) and Jak3(+/-) controls. CM resistance in Jak3(W81R/+) heterozygotes occurs in presence of normal T, B and NK cell numbers. These findings highlight the pathological role of CD8(+) T cells and Jak3-dependent IFN-γ-mediated Th1 responses in CM pathogenesis.


Asunto(s)
Genes Dominantes/genética , Janus Quinasa 3/genética , Malaria Cerebral/enzimología , Malaria Cerebral/prevención & control , Mutación/genética , Traslado Adoptivo , Secuencia de Aminoácidos , Animales , Cromosomas de los Mamíferos/genética , Citrobacter/fisiología , Etilnitrosourea , Femenino , Predisposición Genética a la Enfermedad , Heterocigoto , Homocigoto , Inmunofenotipificación , Janus Quinasa 3/química , Malaria Cerebral/genética , Malaria Cerebral/inmunología , Masculino , Ratones , Ratones Mutantes Neurológicos , Datos de Secuencia Molecular , Mycobacterium/fisiología , Linaje , Fenotipo , Plasmodium berghei/fisiología , Estructura Terciaria de Proteína , Bazo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA