Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Immunity ; 47(1): 183-198.e6, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28723550

RESUMEN

Tissue macrophages arise during embryogenesis from yolk-sac (YS) progenitors that give rise to primitive YS macrophages. Until recently, it has been impossible to isolate or derive sufficient numbers of YS-derived macrophages for further study, but data now suggest that induced pluripotent stem cells (iPSCs) can be driven to undergo a process reminiscent of YS-hematopoiesis in vitro. We asked whether iPSC-derived primitive macrophages (iMacs) can terminally differentiate into specialized macrophages with the help of growth factors and organ-specific cues. Co-culturing human or murine iMacs with iPSC-derived neurons promoted differentiation into microglia-like cells in vitro. Furthermore, murine iMacs differentiated in vivo into microglia after injection into the brain and into functional alveolar macrophages after engraftment in the lung. Finally, iPSCs from a patient with familial Mediterranean fever differentiated into iMacs with pro-inflammatory characteristics, mimicking the disease phenotype. Altogether, iMacs constitute a source of tissue-resident macrophage precursors that can be used for biological, pathophysiological, and therapeutic studies.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Hematopoyesis , Macrófagos/fisiología , Neuronas/fisiología , Células Madre Pluripotentes/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Embrión de Mamíferos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neurogénesis
2.
J Neurochem ; 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38898700

RESUMEN

Studies of the pathophysiology of fragile X syndrome (FXS) have predominantly focused on synaptic and neuronal disruptions in the disease. However, emerging studies highlight the consistency of white matter abnormalities in the disorder. Recent investigations using animal models of FXS have suggested a role for the fragile X translational regulator 1 protein (FMRP) in the development and function of oligodendrocytes, the myelinating cells of the central nervous system. These studies are starting to uncover FMRP's involvement in the regulation of myelin-related genes, such as myelin basic protein, and its influence on the maturation and functionality of oligodendrocyte precursor cells and oligodendrocytes. Here, we consider evidence of white matter abnormalities in FXS, review our current understanding of FMRP's role in oligodendrocyte development and function, and highlight gaps in our knowledge of the pathogenic mechanisms that may contribute to white matter abnormalities in FXS. Addressing these gaps may help identify new therapeutic strategies aimed at enhancing outcomes for individuals affected by FXS.

4.
Hum Mol Genet ; 29(16): 2788-2802, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32898862

RESUMEN

Huntington disease (HD) is a neurodegenerative disorder that is caused by a CAG repeat expansion in HTT. The length of this repeat, however, only explains a proportion of the variability in age of onset in patients. Genome-wide association studies have identified modifiers that contribute toward a proportion of the observed variance. By incorporating tissue-specific transcriptomic information with these results, additional modifiers can be identified. We performed a transcriptome-wide association study assessing heritable differences in genetically determined expression in diverse tissues, with genome-wide data from over 4000 patients. Functional validation of prioritized genes was undertaken in isogenic HD stem cells and patient brains. Enrichment analyses were performed with biologically relevant gene sets to identify the core pathways. HD-associated gene coexpression modules were assessed for associations with neurological phenotypes in an independent cohort and to guide drug repurposing analyses. Transcriptomic analyses identified genes that were associated with age of HD onset and displayed colocalization with gene expression signals in brain tissue (FAN1, GPR161, PMS2, SUMF2), with supporting evidence from functional experiments. This included genes involved in DNA repair, as well as novel-candidate modifier genes that have been associated with other neurological conditions. Further, cortical coexpression modules were also associated with cognitive decline and HD-related traits in a longitudinal cohort. In summary, the combination of population-scale gene expression information with HD patient genomic data identified novel modifier genes for the disorder. Further, these analyses expanded the pathways potentially involved in modifying HD onset and prioritized candidate therapeutics for future study.


Asunto(s)
Estudio de Asociación del Genoma Completo , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Transcriptoma/genética , Adulto , Edad de Inicio , Anciano , Reparación del ADN/genética , Endodesoxirribonucleasas/genética , Exodesoxirribonucleasas/genética , Femenino , Regulación de la Expresión Génica/genética , Genoma/genética , Genómica , Humanos , Enfermedad de Huntington/epidemiología , Enfermedad de Huntington/patología , Masculino , Persona de Mediana Edad , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto/genética , Enzimas Multifuncionales/genética , Especificidad de Órganos/genética , Polimorfismo de Nucleótido Simple/genética , Receptores Acoplados a Proteínas G/genética , Sulfatasas/genética , Expansión de Repetición de Trinucleótido/genética
5.
N Engl J Med ; 380(15): 1433-1441, 2019 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-30970188

RESUMEN

We report an inborn error of metabolism caused by an expansion of a GCA-repeat tract in the 5' untranslated region of the gene encoding glutaminase (GLS) that was identified through detailed clinical and biochemical phenotyping, combined with whole-genome sequencing. The expansion was observed in three unrelated patients who presented with an early-onset delay in overall development, progressive ataxia, and elevated levels of glutamine. In addition to ataxia, one patient also showed cerebellar atrophy. The expansion was associated with a relative deficiency of GLS messenger RNA transcribed from the expanded allele, which probably resulted from repeat-mediated chromatin changes upstream of the GLS repeat. Our discovery underscores the importance of careful examination of regions of the genome that are typically excluded from or poorly captured by exome sequencing.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/genética , Ataxia/genética , Discapacidades del Desarrollo/genética , Glutaminasa/deficiencia , Glutaminasa/genética , Glutamina/metabolismo , Repeticiones de Microsatélite , Mutación , Atrofia/genética , Cerebelo/patología , Preescolar , Femenino , Genotipo , Glutamina/análisis , Humanos , Masculino , Fenotipo , Reacción en Cadena de la Polimerasa , Secuenciación Completa del Genoma
6.
Proc Natl Acad Sci U S A ; 116(19): 9622-9627, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-31015293

RESUMEN

White matter abnormalities are a nearly universal pathological feature of neurodegenerative disorders including Huntington disease (HD). A long-held assumption is that this white matter pathology is simply a secondary outcome of the progressive neuronal loss that manifests with advancing disease. Using a mouse model of HD, here we show that white matter and myelination abnormalities are an early disease feature appearing before the manifestation of any behavioral abnormalities or neuronal loss. We further show that selective inactivation of mutant huntingtin (mHTT) in the NG2+ oligodendrocyte progenitor cell population prevented myelin abnormalities and certain behavioral deficits in HD mice. Strikingly, the improvements in behavioral outcomes were seen despite the continued expression of mHTT in nonoligodendroglial cells including neurons, astrocytes, and microglia. Using RNA-seq and ChIP-seq analyses, we implicate a pathogenic mechanism that involves enhancement of polycomb repressive complex 2 (PRC2) activity by mHTT in the intrinsic oligodendroglial dysfunction and myelination deficits observed in HD. Our findings challenge the long-held dogma regarding the etiology of white matter pathology in HD and highlight the contribution of epigenetic mechanisms to the observed intrinsic oligodendroglial dysfunction. Our results further suggest that ameliorating white matter pathology and oligodendroglial dysfunction may be beneficial for HD.


Asunto(s)
Conducta Animal , Enfermedades Desmielinizantes , Proteína Huntingtina , Enfermedad de Huntington , Mutación , Oligodendroglía , Animales , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Ratones , Ratones Mutantes , Oligodendroglía/metabolismo , Oligodendroglía/patología , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Sustancia Blanca/metabolismo , Sustancia Blanca/patología
7.
Dev Biol ; 458(1): 98-105, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31682806

RESUMEN

Attempts to constitutively knockout HTT in rodents resulted in embryonic lethality, curtailing efforts to study HTT function later in development. Here we show that HTT is dispensable for early zebrafish development, contrasting published zebrafish morpholino experiment results. Homozygous HTT knockouts were embryonically viable and appeared developmentally normal through juvenile stages. Comparison of adult fish revealed significant reduction in body size and fitness in knockouts compared to hemizygotes and wildtype fish, indicating an important role for wildtype HTT in postnatal development. Our zebrafish model provides an opportunity to understand the function of wildtype HTT later in development.


Asunto(s)
Modelos Animales , Proteínas del Tejido Nervioso/fisiología , Proteínas de Pez Cebra/fisiología , Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Tamaño Corporal , Sistemas CRISPR-Cas , Secuencia Conservada , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/embriología , Edición Génica , Técnicas de Inactivación de Genes , Estudios de Asociación Genética , Aptitud Genética , Humanos , Proteína Huntingtina/química , Morfolinos/farmacología , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Neurulación/genética , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Pez Cebra/embriología , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
8.
Glia ; 69(12): 2947-2962, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34427356

RESUMEN

The function of astrocytes intertwines with the extracellular matrix, whose neuron and glial cell-derived components shape neuronal plasticity. Astrocyte abnormalities have been reported in the brain of the mouse model for fragile X syndrome (FXS), the most common cause of inherited intellectual disability, and a monogenic cause of autism spectrum disorder. We compared human FXS and control astrocytes generated from human induced pluripotent stem cells and we found increased expression of urokinase plasminogen activator (uPA), which modulates degradation of extracellular matrix. Several pathways associated with uPA and its receptor function were activated in FXS astrocytes. Levels of uPA were also increased in conditioned medium collected from FXS hiPSC-derived astrocyte cultures and correlated inversely with intracellular Ca2+ responses to activation of L-type voltage-gated calcium channels in human astrocytes. Increased uPA augmented neuronal phosphorylation of TrkB within the docking site for the phospholipase-Cγ1 (PLCγ1), indicating effects of uPA on neuronal plasticity. Gene expression changes during neuronal differentiation preceding astrogenesis likely contributed to properties of astrocytes with FXS-specific alterations that showed specificity by not affecting differentiation of adenosine triphosphate (ATP)-responsive astrocyte population. To conclude, our studies identified uPA as an important regulator of astrocyte function and demonstrated that increased uPA in human FXS astrocytes modulated astrocytic responses and neuronal plasticity.


Asunto(s)
Trastorno del Espectro Autista , Síndrome del Cromosoma X Frágil , Células Madre Pluripotentes Inducidas , Animales , Astrocitos/metabolismo , Trastorno del Espectro Autista/metabolismo , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
9.
Neurobiol Dis ; 158: 105479, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34390831

RESUMEN

Huntington disease (HD) is a neurodegenerative disorder caused by a CAG expansion in the HTT gene that codes for an elongated polyglutamine tract in the huntingtin (HTT) protein. HTT is subject to multiple post-translational modifications (PTMs) that regulate its cellular function. Mutating specific PTM sites within mutant HTT (mHTT) in HD mouse models can modulate disease phenotypes, highlighting the key role of HTT PTMs in the pathogenesis of HD. These findings have led to increased interest in developing small molecules to modulate HTT PTMs in order to decrease mHTT toxicity. However, the therapeutic efficacy of pharmacological modulation of HTT PTMs in preclinical HD models remains largely unknown. HTT is palmitoylated at cysteine 214 by the huntingtin-interacting protein 14 (HIP14 or ZDHHC17) and 14-like (HIP14L or ZDHHC13) acyltransferases. Here, we assessed if HTT palmitoylation should be regarded as a therapeutic target to treat HD by (1) investigating palmitoylation dysregulation in rodent and human HD model systems, (2) measuring the impact of mHTT-lowering therapy on brain palmitoylation, and (3) evaluating if HTT palmitoylation can be pharmacologically modulated. We show that palmitoylation of mHTT and some HIP14/HIP14L-substrates is decreased early in multiple HD mouse models, and that mHTT palmitoylation decreases further with aging. Lowering mHTT in the brain of YAC128 mice is not sufficient to rescue aberrant palmitoylation. However, we demonstrate that mHTT palmitoylation can be normalized in COS-7 cells, in YAC128 cortico-striatal primary neurons and HD patient-derived lymphoblasts using an acyl-protein thioesterase (APT) inhibitor. Moreover, we show that modulating palmitoylation reduces mHTT aggregation and mHTT-induced cytotoxicity in COS-7 cells and YAC128 neurons.


Asunto(s)
Proteína Huntingtina/genética , Proteína Huntingtina/toxicidad , Lipoilación/efectos de los fármacos , Lipoilación/genética , Aciltransferasas/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Cisteína/química , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Femenino , Humanos , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Masculino , Ratones , Mutación , Proteínas del Tejido Nervioso/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas
10.
Acta Neuropathol ; 142(5): 791-806, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34448021

RESUMEN

Huntington disease (HD) is a fatal neurodegenerative disorder caused by an expanded CAG repeat in the huntingtin (HTT) gene. The typical motor symptoms have been associated with basal ganglia pathology. However, psychiatric and cognitive symptoms often precede the motor component and may be due to changes in the limbic system. Recent work has indicated pathology in the hypothalamus in HD but other parts of the limbic system have not been extensively studied. Emerging evidence suggests that changes in HD also include white matter pathology. Here we investigated if the main white matter tract of the limbic system, the fornix, is affected in HD. We demonstrate that the fornix is 34% smaller already in prodromal HD and 41% smaller in manifest HD compared to controls using volumetric analyses of MRI of the IMAGE-HD study. In post-mortem fornix tissue from HD cases, we confirm the smaller fornix volume in HD which is accompanied by signs of myelin breakdown and reduced levels of the transcription factor myelin regulating factor but detect no loss of oligodendrocytes. Further analyses using RNA-sequencing demonstrate downregulation of oligodendrocyte identity markers in the fornix of HD cases. Analysis of differentially expressed genes based on transcription-factor/target-gene interactions also revealed enrichment for binding sites of SUZ12 and EZH2, components of the Polycomb Repressive Complex 2, as well as RE1 Regulation Transcription Factor. Taken together, our data show that there is early white matter pathology of the fornix in the limbic system in HD likely due to a combination of reduction in oligodendrocyte genes and myelin break down.


Asunto(s)
Fórnix/patología , Enfermedad de Huntington/patología , Sistema Límbico/patología , Sustancia Blanca/patología , Adulto , Anciano , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Vaina de Mielina/patología , Oligodendroglía/patología
11.
Neurobiol Dis ; 135: 104744, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31931139

RESUMEN

Structural and molecular myelination deficits represent early pathological features of Huntington disease (HD). Recent evidence from germ-free (GF) animals suggests a role for microbiota-gut-brain bidirectional communication in the regulation of myelination. In this study, we aimed to investigate the impact of microbiota on myelin plasticity and oligodendroglial population dynamics in the mixed-sex BACHD mouse model of HD. Ultrastructural analysis of myelin in the corpus callosum revealed alterations of myelin thickness in BACHD GF compared to specific-pathogen free (SPF) mice, whereas no differences were observed between wild-type (WT) groups. In contrast, myelin compaction was altered in all groups when compared to WT SPF animals. Levels of myelin-related proteins were generally reduced, and the number of mature oligodendrocytes was decreased in the prefrontal cortex under GF compared to SPF conditions, regardless of genotype. Minor differences in commensal bacteria at the family and genera levels were found in the gut microbiota of BACHD and WT animals housed in standard living conditions. Our findings indicate complex effects of a germ-free status on myelin-related characteristics, and highlight the adaptive properties of myelination as a result of environmental manipulation.


Asunto(s)
Enfermedad de Huntington/microbiología , Proteínas de la Mielina/metabolismo , Vaina de Mielina/patología , Sustancia Blanca/microbiología , Animales , Bacterias/aislamiento & purificación , Cuerpo Calloso/metabolismo , Cuerpo Calloso/microbiología , Modelos Animales de Enfermedad , Enfermedad de Huntington/patología , Ratones Transgénicos , Vaina de Mielina/metabolismo , Plasticidad Neuronal/fisiología , Oligodendroglía/metabolismo , Corteza Prefrontal/metabolismo , Sustancia Blanca/patología
12.
PLoS Biol ; 15(2): e1002597, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28207742

RESUMEN

Obesity develops when caloric intake exceeds metabolic needs. Promoting energy expenditure represents an attractive approach in the prevention of this fast-spreading epidemic. Here, we report a novel pharmacological strategy in which a natural compound, narciclasine (ncls), attenuates diet-induced obesity (DIO) in mice by promoting energy expenditure. Moreover, ncls promotes fat clearance from peripheral metabolic tissues, improves blood metabolic parameters in DIO mice, and protects these mice from the loss of voluntary physical activity. Further investigation suggested that ncls achieves these beneficial effects by promoting a shift from glycolytic to oxidative muscle fibers in the DIO mice thereby enhancing mitochondrial respiration and fatty acid oxidation (FAO) in the skeletal muscle. Moreover, ncls strongly activates AMPK signaling specifically in the skeletal muscle. The beneficial effects of ncls treatment in fat clearance and AMPK activation were faithfully reproduced in vitro in cultured murine and human primary myotubes. Mechanistically, ncls increases cellular cAMP concentration and ADP/ATP ratio, which further lead to the activation of AMPK signaling. Blocking AMPK signaling through a specific inhibitor significantly reduces FAO in myotubes. Finally, ncls also enhances mitochondrial membrane potential and reduces the formation of reactive oxygen species in cultured myotubes.


Asunto(s)
Alcaloides de Amaryllidaceae/farmacología , Alcaloides de Amaryllidaceae/uso terapéutico , Dieta/efectos adversos , Músculo Esquelético/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Fenantridinas/farmacología , Fenantridinas/uso terapéutico , Proteínas Quinasas Activadas por AMP/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Biomarcadores/metabolismo , Respiración de la Célula/efectos de los fármacos , Células Cultivadas , AMP Cíclico/metabolismo , Dieta Alta en Grasa , Metabolismo Energético/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Ácidos Grasos/metabolismo , Humanos , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares de Contracción Lenta/efectos de los fármacos , Fibras Musculares de Contracción Lenta/metabolismo , Músculo Esquelético/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Condicionamiento Físico Animal , Sustancias Protectoras/farmacología , Sustancias Protectoras/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
13.
Neurobiol Dis ; 127: 65-75, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30802499

RESUMEN

Structural and molecular myelination deficits represent early pathological features of Huntington disease (HD). Recent evidence from germ-free (GF) animals suggests a role for microbiota-gut-brain bidirectional communication in the regulation of myelination. In this study, we aimed to investigate the impact of microbiota on myelin plasticity and oligodendroglial population dynamics in the mixed-sex BACHD mouse model of HD. Ultrastructural analysis of myelin in the corpus callosum revealed alterations of myelin thickness in BACHD GF compared to specific-pathogen free (SPF) mice, whereas no differences were observed between wild-type (WT) groups. In contrast, myelin compaction was altered in all groups when compared to WT SPF animals. Levels of myelin-related proteins were generally reduced, and the number of mature oligodendrocytes was decreased in the prefrontal cortex under GF compared to SPF conditions, regardless of genotype. Minor differences in commensal bacteria at the family and genera levels were found in the gut microbiota of BACHD and WT animals housed in standard living conditions. Our findings indicate complex effects of a germ-free status on myelin-related characteristics, and highlight the adaptive properties of myelination as a result of environmental manipulation.


Asunto(s)
Cuerpo Calloso/patología , Microbioma Gastrointestinal/fisiología , Enfermedad de Huntington/microbiología , Vaina de Mielina/patología , Plasticidad Neuronal/fisiología , Sustancia Blanca/patología , Animales , Modelos Animales de Enfermedad , Enfermedad de Huntington/patología , Ratones
14.
Hum Mol Genet ; 26(6): 1115-1132, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28104789

RESUMEN

Huntington disease (HD) is a neurodegenerative disease caused by a mutation in the huntingtin (HTT) gene. HTT is a large protein, interacts with many partners and is involved in many cellular pathways, which are perturbed in HD. Therapies targeting HTT directly are likely to provide the most global benefit. Thus there is a need for preclinical models of HD recapitulating human HTT genetics. We previously generated a humanized mouse model of HD, Hu97/18, by intercrossing BACHD and YAC18 mice with knockout of the endogenous mouse HD homolog (Hdh). Hu97/18 mice recapitulate the genetics of HD, having two full-length, genomic human HTT transgenes heterozygous for the HD mutation and polymorphisms associated with HD in populations of Caucasian descent. We have now generated a companion model, Hu128/21, by intercrossing YAC128 and BAC21 mice on the Hdh-/- background. Hu128/21 mice have two full-length, genomic human HTT transgenes heterozygous for the HD mutation and polymorphisms associated with HD in populations of East Asian descent and in a minority of patients from other ethnic groups. Hu128/21 mice display a wide variety of HD-like phenotypes that are similar to YAC128 mice. Additionally, both transgenes in Hu128/21 mice match the human HTT exon 1 reference sequence. Conversely, the BACHD transgene carries a floxed, synthetic exon 1 sequence. Hu128/21 mice will be useful for investigations of human HTT that cannot be addressed in Hu97/18 mice, for developing therapies targeted to exon 1, and for preclinical screening of personalized HTT lowering therapies in HD patients of East Asian descent.


Asunto(s)
Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Mutación/genética , Alelos , Animales , Modelos Animales de Enfermedad , Exones/genética , Heterocigoto , Humanos , Enfermedad de Huntington/patología , Ratones , Ratones Transgénicos , Fenotipo
15.
Hum Mol Genet ; 25(8): 1600-18, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26908611

RESUMEN

Caspase-6 (CASP6) has emerged as an important player in Huntington disease (HD), Alzheimer disease (AD) and cerebral ischemia, where it is activated early in the disease process. CASP6 also plays a key role in axonal degeneration, further underscoring the importance of this protease in neurodegenerative pathways. As a protein's function is modulated by its protein-protein interactions, we performed a high-throughput yeast-2-hybrid (Y2H) screen against ∼17,000 human proteins to gain further insight into the function of CASP6. We identified a high-confidence list of 87 potential CASP6 interactors. From this list, 61% are predicted to contain a CASP6 recognition site. Of nine candidate substrates assessed, six are cleaved by CASP6. Proteins that did not contain a predicted CASP6 recognition site were assessed using a LUMIER assay approach, and 51% were further validated as interactors by this method. Of note, 54% of the high-confidence interactors identified show alterations in human HD brain at the mRNA level, and there is a significant enrichment for previously validated huntingtin (HTT) interactors. One protein of interest, STK3, a pro-apoptotic kinase, was validated biochemically to be a CASP6 substrate. Furthermore, our results demonstrate that in striatal cells expressing mutant huntingtin (mHTT), an increase in full length and fragment levels of STK3 are observed. We further show that caspase-3 is not essential for the endogenous cleavage of STK3. Characterization of the interaction network provides important new information regarding key pathways of interactors of CASP6 and highlights potential novel therapeutic targets for HD, AD and cerebral ischemia.


Asunto(s)
Caspasa 6/metabolismo , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Mapas de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/metabolismo , Sitios de Unión , Línea Celular , Regulación de la Expresión Génica , Humanos , Proteína Huntingtina/genética , Modelos Biológicos , Procesamiento Proteico-Postraduccional , Serina-Treonina Quinasa 3 , Técnicas del Sistema de Dos Híbridos
16.
Hum Mol Genet ; 25(13): 2621-2632, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27126634

RESUMEN

White matter (WM) atrophy is a significant feature of Huntington disease (HD), although its aetiology and early pathological manifestations remain poorly defined. In this study, we aimed to characterize WM-related features in the transgenic YAC128 and BACHD models of HD. Using diffusion tensor magnetic resonance imaging (DT-MRI), we demonstrate that microstructural WM abnormalities occur from an early age in YAC128 mice. Similarly, electron microscopy analysis of myelinated fibres of the corpus callosum indicated that myelin sheaths are thinner in YAC128 mice as early as 1.5 months of age, well before any neuronal loss can be detected. Transcript levels of myelin-related genes in striatal and cortical tissues were significantly lower in YAC128 mice from 2 weeks of age, and these findings were replicated in differentiated primary oligodendrocytes from YAC128 mice, suggesting a possible mechanistic explanation for the observed structural deficits. Concordant with these observations, we demonstrate reduced expression of myelin-related genes at 3 months of age and WM microstructural abnormalities using DT-MRI at 12 months of age in the BACHD rats. These findings indicate that WM deficits in HD are an early phenotype associated with cell-intrinsic effects of mutant huntingtin on myelin-related transcripts in oligodendrocytes, and raise the possibility that WM abnormalities may be an early contributing factor to the pathogenesis of HD.


Asunto(s)
Enfermedad de Huntington/genética , Vaina de Mielina/fisiología , Sustancia Blanca/fisiopatología , Animales , Atrofia/patología , Encéfalo/metabolismo , Cuerpo Calloso/metabolismo , Cuerpo Estriado/metabolismo , Imagen de Difusión Tensora/métodos , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Enfermedad de Huntington/etiología , Ratones , Ratones Transgénicos , Vaina de Mielina/genética , Vaina de Mielina/metabolismo , Neostriado/metabolismo , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Oligodendroglía/metabolismo , Ratas
17.
NMR Biomed ; 31(12): e4007, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30260561

RESUMEN

Recent studies suggest that neurodegenerative diseases could affect brain structure and function in disease-specific network patterns; however, how spontaneous activity affects structural covariance network (SC) is not clear. We hypothesized that hyper-excitability in Huntington disease (HD) disrupts the coordinated structural and functional connectivity, and treatment with memantine helps to reduce excitotoxicity and normalize the connectivity. MRI was conducted to measure somatosensory activation, resting-state functional-connectivity (rsFC), SC, amplitude of low frequency fluctuation (ALFF) and ALFF covariance (ALFFC) in the YAC128 mouse model of HD. We found somatosensory activation was unchanged but the subcortical ALFF was increased in HD mice, indicating subcortical but not cortical hyperactivity. The reduced sensorimotor rsFC but spared hippocampal and default mode networks in the HD mice was consistent with the more pronounced impairment in motor function compared with cognitive performance. The disease suppressed SC globally and reduced ALFFC in the basal ganglia network as well as its anti-correlation with the default mode network. By comparing these connectivity measures, we found that the originally coupled rsFC-SC relationship was impaired whereas SC-ALFFC correlation was increased by HD, suggesting disease facilitated covariation of brain volume and activity amplitude but not neural synchrony. The comparison with mono-synaptic axonal projection supports the hypothesis that rsFC, but not SC or ALFFC, is highly dependent on structural connectivity under healthy conditions. Treatment with memantine had a strong effect on normalizing the SC and reducing ALFF while slightly increasing other connectivity measures and restoring the rsFC-SC coupling, which is consistent with its effect on alleviating hyper-excitability and improving the coordinated neural growth. These results indicate that HD affects the cerebral structure-function relationship which could be partially reverted by NMDA antagonism. These connectivity measures provide unique insights into pathological and pharmaceutical effects in brain circuitry, and could be translatable biomarkers for evaluating drug effect and refining its efficacy.


Asunto(s)
Conectoma , Enfermedad de Huntington/patología , Enfermedad de Huntington/fisiopatología , Imagen por Resonancia Magnética , Animales , Axones/patología , Conducta Animal , Cognición , Modelos Animales de Enfermedad , Estimulación Eléctrica , Humanos , Masculino , Memantina , Ratones , Actividad Motora , Red Nerviosa/fisiopatología , Oxígeno/sangre , Descanso , Corteza Somatosensorial/patología , Corteza Somatosensorial/fisiopatología , Relación Estructura-Actividad
18.
Nat Rev Neurosci ; 14(10): 708-21, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24052178

RESUMEN

Since the identification of the causative gene in Huntington's disease (HD), a number of animal models of this disorder have been developed. A frequently asked question is: which of these models most closely recapitulates the human disease? In this Review, we provide an overview of the currently available animal models of HD in the context of the clinical features of the disease. In doing so, we highlight their strengths and limitations for modelling specific symptoms of the disease. This should highlight the animal model that is best suited to address a particular question of interest and, ultimately, to expedite the discovery of treatments that will prevent or slow the progression of HD.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedad de Huntington , Animales , Animales Modificados Genéticamente , Humanos , Proteína Huntingtina , Enfermedad de Huntington/clasificación , Enfermedad de Huntington/genética , Enfermedad de Huntington/fisiopatología , Proteínas del Tejido Nervioso/genética , Especificidad de la Especie , Repeticiones de Trinucleótidos/genética
19.
Neurobiol Dis ; 76: 24-36, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25583186

RESUMEN

Huntington Disease (HD) is a progressive neurodegenerative disease caused by an elongated CAG repeat in the huntingtin (HTT) gene that encodes a polyglutamine tract in the HTT protein. Proteolysis of the mutant HTT protein (mHTT) has been detected in human and murine HD brains and is implicated in the pathogenesis of HD. Of particular importance is the site at amino acid (aa) 586 that contains a caspase-6 (Casp6) recognition motif. Activation of Casp6 occurs presymptomatically in human HD patients and the inhibition of mHTT proteolysis at aa586 in the YAC128 mouse model results in the full rescue of HD-like phenotypes. Surprisingly, Casp6 ablation in two different HD mouse models did not completely prevent the generation of this fragment, and therapeutic benefits were limited, questioning the role of Casp6 in the disease. We have evaluated the impact of the loss of Casp6 in the YAC128 mouse model of HD. Levels of the mHTT-586 fragment are reduced but not absent in the absence of Casp6 and we identify caspase 8 as an alternate enzyme that can generate this fragment. In vivo, the ablation of Casp6 results in a partial rescue of body weight gain, normalized IGF-1 levels, a reversal of the depression-like phenotype and decreased HTT levels. In the YAC128/Casp6-/- striatum there is a concomitant reduction in p62 levels, a marker of autophagic activity, suggesting increased autophagic clearance. These results implicate the HTT-586 fragment as a key contributor to certain features of HD, irrespective of the enzyme involved in its generation.


Asunto(s)
Caspasa 6/metabolismo , Enfermedad de Huntington/enzimología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Animales , Peso Corporal , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Caspasa 6/genética , Cuerpo Estriado/metabolismo , Depresión/metabolismo , Modelos Animales de Enfermedad , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ratones , Ratones Transgénicos , Actividad Motora
20.
Neurobiol Dis ; 76: 46-56, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25662335

RESUMEN

Huntington disease (HD) is an inherited, fatal neurodegenerative disease with no disease-modifying therapy currently available. In addition to characteristic motor deficits and atrophy of the caudate nucleus, signature hallmarks of HD include behavioral abnormalities, immune activation, and cortical and white matter loss. The identification and validation of novel therapeutic targets that contribute to these degenerative cellular processes may lead to new interventions that slow or even halt the course of this insidious disease. Semaphorin 4D (SEMA4D) is a transmembrane signaling molecule that modulates a variety of processes central to neuroinflammation and neurodegeneration including glial cell activation, neuronal growth cone collapse and apoptosis of neural precursors, as well as inhibition of oligodendrocyte migration, differentiation and process formation. Therefore, inhibition of SEMA4D signaling could reduce CNS inflammation, increase neuronal outgrowth and enhance oligodendrocyte maturation, which may be of therapeutic benefit in the treatment of several neurodegenerative diseases, including HD. To that end, we evaluated the preclinical therapeutic efficacy of an anti-SEMA4D monoclonal antibody, which prevents the interaction between SEMA4D and its receptors, in the YAC128 transgenic HD mouse model. Anti-SEMA4D treatment ameliorated neuropathological signatures, including striatal atrophy, cortical atrophy, and corpus callosum atrophy and prevented testicular degeneration in YAC128 mice. In parallel, a subset of behavioral symptoms was improved in anti-SEMA4D treated YAC128 mice, including reduced anxiety-like behavior and rescue of cognitive deficits. There was, however, no discernible effect on motor deficits. The preservation of brain gray and white matter and improvement in behavioral measures in YAC128 mice treated with anti-SEMA4D suggest that this approach could represent a viable therapeutic strategy for the treatment of HD. Importantly, this work provides in vivo demonstration that inhibition of pathways initiated by SEMA4D constitutes a novel approach to moderation of neurodegeneration.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos CD/inmunología , Enfermedad de Huntington/terapia , Semaforinas/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Encéfalo/metabolismo , Encéfalo/patología , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/terapia , Modelos Animales de Enfermedad , Enfermedad de Huntington/complicaciones , Inmunoterapia , Ratones , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda