Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
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
2.
Hum Mol Genet ; 27(2): 239-253, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29121340

RESUMEN

Oxidative stress is a prominent feature of Huntington disease (HD), and we have shown previously that reduced levels of hace1 (HECT domain and Ankyrin repeat containing E3 ubiquitin protein ligase 1) in patient striatum may contribute to the pathogenesis of HD. Hace1 promotes the stability of Nrf2 and thus plays an important role in antioxidant response mechanisms, which are dysfunctional in HD. Moreover, hace1 overexpression mitigates mutant huntingtin (mHTT)-induced oxidative stress in vitro through promotion of the Nrf2 antioxidant response. Here, we show that the genetic ablation of hace1 in the YAC128 mouse model of HD accelerates motor deficits and exacerbates cognitive and psychiatric phenotypes in vivo. We find that both the expression of mHTT and the ablation of hace1 alone are sufficient to cause deficits in astrocytic mitochondrial respiration. We confirm the crucial role of hace1 in astrocytes in vivo, since its ablation is sufficient to cause dramatic astrogliosis in wild-type FVB/N mice. Astrogliosis is not observed in the presence of mHTT but a strong dysregulation in the expression of astrocytic markers in HACE1-/- x YAC128 striatum suggests an additive effect of mHTT expression and hace1 loss on this cell type. HACE1-/- x YAC128 mice and primary cells derived from these animals therefore provide model systems that will allow for the further dissection of Nrf2 pathways and astrocyte dysfunction in the context of HD.


Asunto(s)
Astrocitos/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Ratones , Mitocondrias/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Neostriado/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Estrés Oxidativo/fisiología
3.
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
4.
BMC Biol ; 14(1): 108, 2016 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-27927242

RESUMEN

BACKGROUND: Palmitoylation, the addition of palmitate to proteins by palmitoyl acyltransferases (PATs), is an important regulator of synaptic protein localization and function. Many palmitoylated proteins and PATs have been implicated in neuropsychiatric diseases, including Huntington disease, schizophrenia, amyotrophic lateral sclerosis, Alzheimer disease, and X-linked intellectual disability. HIP14/DHHC17 is the most conserved PAT that palmitoylates many synaptic proteins. Hip14 hypomorphic mice have behavioral and synaptic deficits. However, the phenotype is developmental; thus, a model of post-developmental loss of Hip14 was generated to examine the role of HIP14 in synaptic function in the adult. RESULTS: Ten weeks after Hip14 deletion (iHip14 Δ/Δ ), mice die suddenly from rapidly progressive paralysis. Prior to death the mice exhibit motor deficits, increased escape response during tests of anxiety, anhedonia, a symptom indicative of depressive-like behavior, and striatal synaptic deficits, including reduced probability of transmitter release and increased amplitude but decreased frequency of spontaneous post-synaptic currents. The mice also have increased brain weight due to microgliosis and astrogliosis in the cortex. CONCLUSIONS: Behavioral changes and electrophysiological measures suggest striatal dysfunction in iHip14 Δ/Δ mice, and increased cortical volume due to astrogliosis and microgliosis suggests a novel role for HIP14 in glia. These data suggest that HIP14 is essential for maintenance of life and neuronal integrity in the adult mouse.


Asunto(s)
Aciltransferasas/genética , Muerte Súbita , Eliminación de Gen , Aciltransferasas/metabolismo , Animales , Peso Corporal , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Lipoilación , Masculino , Ratones , Ratones Noqueados , Neuroglía/patología , Tamaño de los Órganos
5.
Hum Mol Genet ; 25(17): 3654-3675, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27378694

RESUMEN

Huntington disease (HD) model mice with heterozygous knock-in (KI) of an expanded CAG tract in exon 1 of the mouse huntingtin (Htt) gene homolog genetically recapitulate the mutation that causes HD, and might be favoured for preclinical studies. However, historically these mice have failed to phenotypically recapitulate the human disease. Thus, homozygous KI mice, which lack wildtype Htt, and are much less relevant to human HD, have been used. The zQ175 model was the first KI mouse to exhibit significant HD-like phenotypes when heterozygous. In an effort to exacerbate HD-like phenotypes and enhance preclinical utility, we have backcrossed zQ175 mice to FVB/N, a strain highly susceptible to neurodegeneration. These Q175F mice display significant HD-like phenotypes along with sudden early death from fatal seizures. The zQ175 KI allele retains a floxed neomycin resistance cassette upstream of the Htt gene locus and produces dramatically reduced mutant Htt as compared to the endogenous wildtype Htt allele. By intercrossing with mice expressing cre in germ line cells, we have excised the neo cassette from Q175F mice generating a new line, Q175FΔneo (Q175FDN). Removal of the neo cassette resulted in a ∼2 fold increase in mutant Htt and rescue of fatal seizures, indicating that the early death phenotype of Q175F mice is caused by Htt deficiency rather than by mutant Htt. Additionally, Q175FDN mice exhibit earlier onset and a greater variety and severity of HD-like phenotypes than Q175F mice or any previously reported KI HD mouse model, making them valuable for preclinical studies.


Asunto(s)
Técnicas de Sustitución del Gen/métodos , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Mutación , Animales , Conducta Animal , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Heterocigoto , Humanos , Enfermedad de Huntington/patología , Ratones , Fenotipo
6.
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
7.
Neurosci Lett ; 539: 16-21, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23347841

RESUMEN

Aberrant oxysterol biosynthesis is implicated in the pathogenesis of neurodegenerative diseases. During the present study we have investigated the effects of exogenously added 25-hydroxycholesterol (25-HC) on transcription of cholesterol biosynthetic genes, sterol-regulatory element binding protein (SREBP) processing and cholesterol biosynthesis in the murine CATH.a neuronal cell line. A single i.p. injection of lipopolysaccharide resulted in robust induction of cholesterol 25-hydroxylase mRNA and protein levels in brains of treated mice. In vitro, 25-HC upregulated the transcription of ATP-binding cassette transporter A1 (ABCA1) and (to a lesser extent) apolipoprotein E (apoE) in CATH.a neurons. Cholesterol biosynthetic gene expression (squalene synthase, HMG-CoA synthase, HMG-CoA reductase, and SREBP2) was downregulated by 25-HC. 25-HC also significantly attenuated proteolytic processing of SREBP2. Finally, 25-HC downregulated cholesterol biosynthesis in CATH.a neurons. Our results demonstrate that 25-HC is a potent effector oxysterol of neuronal cholesterol homeostasis.


Asunto(s)
Colesterol/biosíntesis , Hidroxicolesteroles/farmacología , Neuronas/efectos de los fármacos , Animales , Línea Celular , Colesterol/genética , Homeostasis , Hidroxicolesteroles/metabolismo , Lipopolisacáridos/farmacología , Ratones , Neuronas/metabolismo , ARN Mensajero/metabolismo , Esteroide Hidroxilasas/genética , Esteroide Hidroxilasas/metabolismo , Proteínas de Unión a los Elementos Reguladores de Esteroles/genética , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo , Transcripción Genética
8.
Free Radic Biol Med ; 49(11): 1655-65, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20807565

RESUMEN

Plasmalogens, 1-O-alk-1'-enyl-2-acyl-sn-glycerophospholipids, are significant constituents of cellular membranes and are essential for normal brain development. Plasmalogens, which contain a vinyl ether bond at the sn-1 position, are preferential targets for hypochlorous acid (HOCl), generated by myeloperoxidase (MPO) from H(2)O(2) and chloride ions. Because MPO is implicated in neurodegeneration, this study pursued two aims: (i) to investigate the reactivity of mouse brain plasmalogens toward HOCl in vitro and (ii) to obtain in vivo evidence for MPO-mediated brain plasmalogen modification. Liquid chromatography coupled to hybrid linear ion trap-Fourier transform-ion cyclotron resonance mass spectrometry revealed plasmalogen modification in mouse brain lipid extracts at lower HOCl concentrations as observed for diacylphospholipids, resulting in the generation of 2-chloro fatty aldehydes and lysophospholipids. Lysophosphatidylethanolamine accumulation was transient, whereas lysophosphatidylcholine species containing saturated acyl residues remained stable. In vivo, a single, systemic endotoxin injection resulted in upregulation of cerebral MPO mRNA levels to a range comparable to that observed for tumor necrosis factor-α and cyclooxygenase-2. This inflammatory response was accompanied by a significant decrease in several brain plasmalogen species and concomitant in vivo generation of 2-chlorohexadecanal. The present findings demonstrate that activation of the MPO-H(2)O(2)-chloride system under neuroinflammatory conditions results in oxidative attack of the total cerebral plasmalogen pool. As this lipid class is indispensable for normal neuronal function, HOCl-mediated plasmalogen modification is likely to compromise normal synaptic transmission.


Asunto(s)
Encéfalo/efectos de los fármacos , Ácido Hipocloroso/farmacología , Plasmalógenos/química , Plasmalógenos/metabolismo , Animales , Encéfalo/metabolismo , Química Encefálica/efectos de los fármacos , Células Cultivadas , Etanolaminas/análisis , Etanolaminas/metabolismo , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción , Fosfolípidos/análisis , Fosfolípidos/metabolismo , Fosforilcolina/análisis , Fosforilcolina/metabolismo , Transcripción Genética/efectos de los fármacos
9.
Free Radic Biol Med ; 48(12): 1588-600, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20226853

RESUMEN

Recent observations link myeloperoxidase (MPO) activation to neurodegeneration. In multiple sclerosis MPO is present in areas of active demyelination where the potent oxidant hypochlorous acid (HOCl), formed by MPO from H(2)O(2) and chloride ions, could oxidatively damage myelin-associated lipids. The purpose of this study was (i) to characterize reaction products of sphingomyelin (SM) formed in response to modification by HOCl, (ii) to define the impact of exogenously added SM and HOCl-modified SM (HOCl-SM) on viability parameters of a neuronal cell line (PC12), and (iii) to study alterations in the PC12 cell proteome in response to SM and HOCl-SM. MALDI-TOF-MS analyses revealed that HOCl, added as reagent or generated enzymatically, transforms SM into chlorinated species. On the cellular level HOCl-SM but not SM induced the formation of reactive oxygen species. HOCl-SM induced severely impaired cell viability, dissipation of the mitochondrial membrane potential, and activation of caspase-3 and DNA damage. Proteome analyses identified differential expression of specific subsets of proteins in response to SM and HOCl-SM. Our results demonstrate that HOCl modification of SM results in the generation of chlorinated lipid species with potent neurotoxic properties. Given the emerging connections between the MPO-H(2)O(2)-chloride axis and neurodegeneration, this chlorinating pathway might be implicated in neuropathogenesis.


Asunto(s)
Apoptosis/efectos de los fármacos , Ácido Hipocloroso/farmacología , Neuronas/metabolismo , Oxidantes/farmacología , Estrés Oxidativo/fisiología , Esfingomielinas/metabolismo , Animales , Apoptosis/fisiología , Western Blotting , Química Encefálica , Línea Celular , Dopamina/metabolismo , Electroforesis en Gel Bidimensional , Análisis de Fourier , Halogenación , Procesamiento de Imagen Asistido por Computador , Técnicas In Vitro , Lípidos/química , Potencial de la Membrana Mitocondrial , Ratones , Ratones Endogámicos C57BL , Proteoma/química , Proteoma/efectos de los fármacos , Proteoma/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Esfingomielinas/química
10.
J Neurochem ; 108(3): 707-18, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19046407

RESUMEN

Alpha-tocopherol (alphaTocH), a member of the vitamin E family, is essential for normal neurological function. Despite the importance of alphaTocH transport into the CNS, transfer mechanisms across the blood-brain barrier (BBB) are not entirely clear. We here investigate whether afamin, a known alphaTocH-binding protein, contributes to alphaTocH transport across an in vitro model of the BBB consisting of primary porcine brain capillary endothelial cells (BCEC) and basolaterally cultured astrocytoma cells. Exogenously added afamin had no adverse effects on BCEC viability or barrier function and was transported across BCEC Transwell cultures. Furthermore, alphaTocH transport across polarized BCEC cultures to astrocytoma cells is facilitated by afamin, though to a lesser extent than by high-density lipoprotein-mediated transport, an essential and in vivo operating alphaTocH import pathway at the cerebrovasculature. We also demonstrate that porcine BCEC endogenously synthesize afamin. In line with these in vitro findings, afamin was detected by immunohistochemistry in porcine, human postmortem, and mouse brain, where prominent staining was observed almost exclusively in the cerebrovasculature. The demonstration of afamin mRNA expression in isolated brain capillaries suggests that afamin might be a new family member of binding/transport proteins contributing to alphaTocH homeostasis at the BBB in vivo.


Asunto(s)
Barrera Hematoencefálica/fisiología , Proteínas Portadoras/biosíntesis , Circulación Cerebrovascular/fisiología , Células Endoteliales/metabolismo , Glicoproteínas/biosíntesis , Albúmina Sérica/biosíntesis , alfa-Tocoferol/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitoma/metabolismo , Transporte Biológico Activo , Western Blotting , Células CHO , Capilares/metabolismo , Técnicas de Cocultivo , Cricetinae , Cricetulus , Electroforesis en Gel de Poliacrilamida , Técnica del Anticuerpo Fluorescente , Humanos , Lipoproteínas HDL/biosíntesis , Lipoproteínas HDL/aislamiento & purificación , Ratones , Ratones Endogámicos C57BL , ARN/biosíntesis , ARN/aislamiento & purificación , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Albúmina Sérica Humana , Porcinos , Sales de Tetrazolio , Tiazoles
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA