Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 40(8): 111249, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-36001963

RESUMEN

The microtubule-associated protein tau is an abundant component of neurons of the central nervous system. In Alzheimer's disease and other neurodegenerative tauopathies, tau is found hyperphosphorylated and aggregated in neurofibrillary tangles. To obtain a better understanding of the cellular perturbations that initiate tau pathogenesis, we performed a CRISPR-Cas9 screen for genetic modifiers that enhance tau aggregation. This initial screen yielded three genes, BANF1, ANKLE2, and PPP2CA, whose inactivation promotes the accumulation of tau in a phosphorylated and insoluble form. In a complementary screen, we identified three additional genes, LEMD2, LEMD3, and CHMP7, that, when overexpressed, provide protection against tau aggregation. The proteins encoded by the identified genes are mechanistically linked and recognized for their roles in the maintenance and repair of the nuclear envelope. These results implicate the disruption of nuclear envelope integrity as a possible initiating event in tauopathies and reveal targets for therapeutic intervention.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Enfermedad de Alzheimer/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Fosforilación , Tauopatías/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
2.
PLoS Genet ; 9(10): e1003930, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24204323

RESUMEN

The Huntington's disease gene (HTT) CAG repeat mutation undergoes somatic expansion that correlates with pathogenesis. Modifiers of somatic expansion may therefore provide routes for therapies targeting the underlying mutation, an approach that is likely applicable to other trinucleotide repeat diseases. Huntington's disease Hdh(Q111) mice exhibit higher levels of somatic HTT CAG expansion on a C57BL/6 genetic background (B6.Hdh(Q111) ) than on a 129 background (129.Hdh(Q111) ). Linkage mapping in (B6x129).Hdh(Q111) F2 intercross animals identified a single quantitative trait locus underlying the strain-specific difference in expansion in the striatum, implicating mismatch repair (MMR) gene Mlh1 as the most likely candidate modifier. Crossing B6.Hdh(Q111) mice onto an Mlh1 null background demonstrated that Mlh1 is essential for somatic CAG expansions and that it is an enhancer of nuclear huntingtin accumulation in striatal neurons. Hdh(Q111) somatic expansion was also abolished in mice deficient in the Mlh3 gene, implicating MutLγ (MLH1-MLH3) complex as a key driver of somatic expansion. Strikingly, Mlh1 and Mlh3 genes encoding MMR effector proteins were as critical to somatic expansion as Msh2 and Msh3 genes encoding DNA mismatch recognition complex MutSß (MSH2-MSH3). The Mlh1 locus is highly polymorphic between B6 and 129 strains. While we were unable to detect any difference in base-base mismatch or short slipped-repeat repair activity between B6 and 129 MLH1 variants, repair efficiency was MLH1 dose-dependent. MLH1 mRNA and protein levels were significantly decreased in 129 mice compared to B6 mice, consistent with a dose-sensitive MLH1-dependent DNA repair mechanism underlying the somatic expansion difference between these strains. Together, these data identify Mlh1 and Mlh3 as novel critical genetic modifiers of HTT CAG instability, point to Mlh1 genetic variation as the likely source of the instability difference in B6 and 129 strains and suggest that MLH1 protein levels play an important role in driving of the efficiency of somatic expansions.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Portadoras/genética , Enfermedad de Huntington/genética , Proteínas Nucleares/genética , Expansión de Repetición de Trinucleótido/genética , Animales , Modelos Animales de Enfermedad , Estudios de Asociación Genética , Estudio de Asociación del Genoma Completo , Inestabilidad Genómica , Humanos , Enfermedad de Huntington/patología , Ratones , Homólogo 1 de la Proteína MutL , Proteínas MutL , ARN Mensajero
3.
PLoS One ; 7(9): e44273, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22970194

RESUMEN

The CAG trinucleotide repeat mutation in the Huntington's disease gene (HTT) exhibits age-dependent tissue-specific expansion that correlates with disease onset in patients, implicating somatic expansion as a disease modifier and potential therapeutic target. Somatic HTT CAG expansion is critically dependent on proteins in the mismatch repair (MMR) pathway. To gain further insight into mechanisms of somatic expansion and the relationship of somatic expansion to the disease process in selectively vulnerable MSNs we have crossed HTT CAG knock-in mice (HdhQ111) with mice carrying a conditional (floxed) Msh2 allele and D9-Cre transgenic mice, in which Cre recombinase is expressed specifically in MSNs within the striatum. Deletion of Msh2 in MSNs eliminated Msh2 protein in those neurons. We demonstrate that MSN-specific deletion of Msh2 was sufficient to eliminate the vast majority of striatal HTT CAG expansions in HdhQ111 mice. Furthermore, MSN-specific deletion of Msh2 modified two mutant huntingtin phenotypes: the early nuclear localization of diffusely immunostaining mutant huntingtin was slowed; and the later development of intranuclear huntingtin inclusions was dramatically inhibited. Therefore, Msh2 acts within MSNs as a genetic enhancer both of somatic HTT CAG expansions and of HTT CAG-dependent phenotypes in mice. These data suggest that the selective vulnerability of MSNs may be at least in part contributed by the propensity for somatic expansion in these neurons, and imply that intervening in the expansion process is likely to have therapeutic benefit.


Asunto(s)
Enfermedad de Huntington/patología , Proteína 2 Homóloga a MutS/metabolismo , Proteínas Mutantes/metabolismo , Neostriado/metabolismo , Neostriado/patología , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Expansión de Repetición de Trinucleótido/genética , Alelos , Animales , Núcleo Celular/metabolismo , Modelos Animales de Enfermedad , Eliminación de Gen , Técnicas de Sustitución del Gen , Proteína Huntingtina , Enfermedad de Huntington/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Fenotipo
4.
PLoS One ; 7(6): e38310, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22701626

RESUMEN

Cln3(Δex7/8) mice harbor the most common genetic defect causing juvenile neuronal ceroid lipofuscinosis (JNCL), an autosomal recessive disease involving seizures, visual, motor and cognitive decline, and premature death. Here, to more thoroughly investigate the manifestations of the common JNCL mutation, we performed a broad phenotyping study of Cln3(Δex7/8) mice. Homozygous Cln3(Δex7/8) mice, congenic on a C57BL/6N background, displayed subtle deficits in sensory and motor tasks at 10-14 weeks of age. Homozygous Cln3(Δex7/8) mice also displayed electroretinographic changes reflecting cone function deficits past 5 months of age and a progressive decline of retinal post-receptoral function. Metabolic analysis revealed increases in rectal body temperature and minimum oxygen consumption in 12-13 week old homozygous Cln3(Δex7/8) mice, which were also seen to a lesser extent in heterozygous Cln3(Δex7/8) mice. Heart weight was slightly increased at 20 weeks of age, but no significant differences were observed in cardiac function in young adults. In a comprehensive blood analysis at 15-16 weeks of age, serum ferritin concentrations, mean corpuscular volume of red blood cells (MCV), and reticulocyte counts were reproducibly increased in homozygous Cln3(Δ) (ex7/8) mice, and male homozygotes had a relative T-cell deficiency, suggesting alterations in hematopoiesis. Finally, consistent with findings in JNCL patients, vacuolated peripheral blood lymphocytes were observed in homozygous Cln3(Δ) (ex7/8) neonates, and to a greater extent in older animals. Early onset, severe vacuolation in clear cells of the epididymis of male homozygous Cln3(Δ) (ex7/8) mice was also observed. These data highlight additional organ systems in which to study CLN3 function, and early phenotypes have been established in homozygous Cln3(Δ) (ex7/8) mice that merit further study for JNCL biomarker development.


Asunto(s)
Modelos Animales de Enfermedad , Glicoproteínas de Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Lipofuscinosis Ceroideas Neuronales/patología , Fenotipo , Degeneración Retiniana/patología , Análisis de Varianza , Animales , Temperatura Corporal , Encéfalo/patología , Electrorretinografía , Conducta Exploratoria/fisiología , Femenino , Ferritinas/sangre , Genotipo , Corazón/crecimiento & desarrollo , Inmunohistoquímica , Linfocitos/patología , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Chaperonas Moleculares/genética , Lipofuscinosis Ceroideas Neuronales/complicaciones , Lipofuscinosis Ceroideas Neuronales/genética , Lipofuscinosis Ceroideas Neuronales/metabolismo , Tamaño de los Órganos , Consumo de Oxígeno/fisiología , Degeneración Retiniana/etiología
5.
BMC Syst Biol ; 4: 29, 2010 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-20302627

RESUMEN

BACKGROUND: In Huntington's disease (HD), an expanded CAG repeat produces characteristic striatal neurodegeneration. Interestingly, the HD CAG repeat, whose length determines age at onset, undergoes tissue-specific somatic instability, predominant in the striatum, suggesting that tissue-specific CAG length changes could modify the disease process. Therefore, understanding the mechanisms underlying the tissue specificity of somatic instability may provide novel routes to therapies. However progress in this area has been hampered by the lack of sensitive high-throughput instability quantification methods and global approaches to identify the underlying factors. RESULTS: Here we describe a novel approach to gain insight into the factors responsible for the tissue specificity of somatic instability. Using accurate genetic knock-in mouse models of HD, we developed a reliable, high-throughput method to quantify tissue HD CAG repeat instability and integrated this with genome-wide bioinformatic approaches. Using tissue instability quantified in 16 tissues as a phenotype and tissue microarray gene expression as a predictor, we built a mathematical model and identified a gene expression signature that accurately predicted tissue instability. Using the predictive ability of this signature we found that somatic instability was not a consequence of pathogenesis. In support of this, genetic crosses with models of accelerated neuropathology failed to induce somatic instability. In addition, we searched for genes and pathways that correlated with tissue instability. We found that expression levels of DNA repair genes did not explain the tissue specificity of somatic instability. Instead, our data implicate other pathways, particularly cell cycle, metabolism and neurotransmitter pathways, acting in combination to generate tissue-specific patterns of instability. CONCLUSION: Our study clearly demonstrates that multiple tissue factors reflect the level of somatic instability in different tissues. In addition, our quantitative, genome-wide approach is readily applicable to high-throughput assays and opens the door to widespread applications with the potential to accelerate the discovery of drugs that alter tissue instability.


Asunto(s)
Enfermedad de Huntington/genética , Modelos Genéticos , Expansión de Repetición de Trinucleótido , Repeticiones de Trinucleótidos/genética , Animales , Biología Computacional/métodos , ADN/metabolismo , Femenino , Masculino , Ratones , Ratones Transgénicos , Enfermedades Neurodegenerativas/genética , Neurotransmisores/metabolismo , Análisis de Regresión , Distribución Tisular
6.
Neurobiol Dis ; 33(1): 37-47, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18930147

RESUMEN

Modifying the length of the Huntington's disease (HD) CAG repeat, the major determinant of age of disease onset, is an attractive therapeutic approach. To explore this we are investigating mechanisms of intergenerational and somatic HD CAG repeat instability. Here, we have crossed HD CAG knock-in mice onto backgrounds deficient in mismatch repair genes, Msh3 and Msh6, to discern the effects on CAG repeat size and disease pathogenesis. We find that different mechanisms predominate in inherited and somatic instability, with Msh6 protecting against intergenerational contractions and Msh3 required both for increasing CAG length and for enhancing an early disease phenotype in striatum. Therefore, attempts to decrease inherited repeat size may entail a full understanding of Msh6 complexes, while attempts to block the age-dependent increases in CAG size in striatal neurons and to slow the disease process will require a full elucidation of Msh3 complexes and their function in CAG repeat instability.


Asunto(s)
Reparación del ADN/genética , Modelos Animales de Enfermedad , Inestabilidad Genómica , Enfermedad de Huntington/genética , Animales , Cuerpo Estriado/metabolismo , Cruzamientos Genéticos , Proteínas de Unión al ADN/genética , Femenino , Proteína Huntingtina , Enfermedad de Huntington/fisiopatología , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Proteína 2 Homóloga a MutS/genética , Proteína 3 Homóloga de MutS , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Proteínas Nucleares/genética , Fenotipo , Proteínas/genética , Expansión de Repetición de Trinucleótido
7.
J Infect Dis ; 194(5): 661-5, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16897665

RESUMEN

The influence of CD4(+) T cell nadirs on human immunodeficiency virus (HIV)-specific immune responses in subjects with apparently normal CD4(+) T cell counts is not known. We evaluated the frequency of HIV-1-specific immune responses in a cohort of patients with complete viral suppression (HIV-1 RNA load, <50 copies/mL) who were receiving highly active antiretroviral therapy and had a wide range of CD4(+) T cell nadirs. We found positive associations between CD4(+) T cell nadirs and the magnitude of HIV-specific CD8(+) T cell responses (P=.02) and of T cell helper responses (P=.04). These data show the CD4(+) T cell nadir to be an independent predictor of HIV-specific CD4(+) and CD8(+) T cell responses in HIV-1-infected subjects with optimal suppression of viremia.


Asunto(s)
Terapia Antirretroviral Altamente Activa , Recuento de Linfocito CD4 , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/inmunología , Linfocitos T CD4-Positivos/inmunología , Activación de Linfocitos , Valor Predictivo de las Pruebas , ARN Viral/sangre , Resultado del Tratamiento , Carga Viral
8.
Hum Mol Genet ; 15(12): 2015-24, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16687439

RESUMEN

Genetically precise models of Huntington's disease (HD), Hdh CAG knock-in mice, are powerful systems in which phenotypes associated with expanded HD CAG repeats are studied. To dissect the genetic pathways that underlie such phenotypes, we have generated Hdh(Q111) knock-in mouse lines that are congenic for C57BL/6, FVB/N and 129Sv inbred genetic backgrounds and investigated four Hdh(Q111) phenotypes in these three genetic backgrounds: the intergenerational instability of the HD CAG repeat and the striatal-specific somatic HD CAG repeat expansion, nuclear mutant huntingtin accumulation and intranuclear inclusion formation. Our results reveal increased intergenerational and somatic instability of the HD CAG repeat in C57BL/6 and FVB/N backgrounds compared with the 129Sv background. The accumulation of nuclear mutant huntingtin and the formation of intranuclear inclusions were fastest in the C57BL/6 background, slowest in the 129Sv background and intermediate in the FVB/N background. Inbred strain-specific differences were independent of constitutive HD CAG repeat size and did not correlate with Hdh mRNA levels. These data provide evidence for genetic modifiers of both intergenerational HD CAG repeat instability and striatal-specific phenotypes. Different relative contributions of C57BL/6 and 129Sv genetic backgrounds to the onset of nuclear mutant huntingtin and somatic HD CAG repeat expansion predict that the initiation of each of these two phenotypes is modified by different genes. Our findings set the stage for defining disease-related genetic pathways that will ultimately provide insight into disease mechanism.


Asunto(s)
Expansión de las Repeticiones de ADN , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Animales , Núcleo Celular/metabolismo , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Femenino , Proteína Huntingtina , Enfermedad de Huntington/patología , Cuerpos de Inclusión Intranucleares/metabolismo , Cuerpos de Inclusión Intranucleares/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Mutación , Neuronas/ultraestructura , Repeticiones de Trinucleótidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...