Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 32(20): 3006-3025, 2023 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-37535888

RESUMEN

Huntington's disease (HD) is a progressive neurodegenerative disorder caused by a CAG repeat expansion in the HD gene, coding for huntingtin protein (HTT). Mechanisms of HD cellular pathogenesis remain undefined and likely involve disruptions in many cellular processes and functions presumably mediated by abnormal protein interactions of mutant HTT. We previously found HTT interaction with several protein arginine methyl-transferase (PRMT) enzymes. Protein arginine methylation mediated by PRMT enzymes is an important post-translational modification with an emerging role in neurodegeneration. We found that normal (but not mutant) HTT can facilitate the activity of PRMTs in vitro and the formation of arginine methylation complexes. These interactions appear to be disrupted in HD neurons. This suggests an additional functional role for HTT/PRMT interactions, not limited to substrate/enzyme relationship, which may result in global changes in arginine protein methylation in HD. Our quantitative analysis of striatal precursor neuron proteome indicated that arginine protein methylation is significantly altered in HD. We identified a cluster highly enriched in RNA-binding proteins with reduced arginine methylation, which is essential to their function in RNA processing and splicing. We found that several of these proteins interact with HTT, and their RNA-binding and localization are affected in HD cells likely due to a compromised arginine methylation and/or abnormal interactions with mutant HTT. These studies reveal a potential new mechanism for disruption of RNA processing in HD, involving a direct interaction of HTT with methyl-transferase enzymes and modulation of their activity and highlighting methylation of arginine as potential new therapeutic target for HD.

2.
Hum Mol Genet ; 31(10): 1651-1672, 2022 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-34888656

RESUMEN

Huntington's disease (HD) is an incurable neurodegenerative disorder caused by a CAG expansion in the huntingtin gene (HTT). Post-translational modifications of huntingtin protein (HTT), such as phosphorylation, acetylation and ubiquitination, have been implicated in HD pathogenesis. Arginine methylation/dimethylation is an important modification with an emerging role in neurodegeneration; however, arginine methylation of HTT remains largely unexplored. Here we report nearly two dozen novel arginine methylation/dimethylation sites on the endogenous HTT from human and mouse brain and human cells suggested by mass spectrometry with data-dependent acquisition. Targeted quantitative mass spectrometry identified differential arginine methylation at specific sites in HD patient-derived striatal precursor cell lines compared to normal controls. We found that HTT can interact with several type I protein arginine methyltransferases (PRMTs) via its N-terminal domain. Using a combination of in vitro methylation and cell-based experiments, we identified PRMT4 (CARM1) and PRMT6 as major enzymes methylating HTT at specific arginines. Alterations of these methylation sites had a profound effect on biochemical properties of HTT rendering it less soluble in cells and affected its liquid-liquid phase separation and phase transition patterns in vitro. We found that expanded HTT 1-586 fragment can form liquid-like assemblies, which converted into solid-like assemblies when the R200/205 methylation sites were altered. Methyl-null alterations increased HTT toxicity to neuronal cells, while overexpression of PRMT 4 and 6 was beneficial for neuronal survival. Thus, arginine methylation pathways that involve specific HTT-modifying PRMT enzymes and modulate HTT biochemical and toxic properties could provide targets for HD-modifying therapies.


Asunto(s)
Arginina , Enfermedad de Huntington , Animales , Arginina/genética , Arginina/metabolismo , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/patología , Metilación , Ratones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Procesamiento Proteico-Postraduccional/genética , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Solubilidad
3.
Hum Mol Genet ; 30(24): 2469-2487, 2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34296279

RESUMEN

We have previously established induced pluripotent stem cell (iPSC) models of Huntington's disease (HD), demonstrating CAG-repeat-expansion-dependent cell biological changes and toxicity. However, the current differentiation protocols are cumbersome and time consuming, making preparation of large quantities of cells for biochemical or screening assays difficult. Here, we report the generation of immortalized striatal precursor neurons (ISPNs) with normal (33) and expanded (180) CAG repeats from HD iPSCs, differentiated to a phenotype resembling medium spiny neurons (MSN), as a proof of principle for a more tractable patient-derived cell model. For immortalization, we used co-expression of the enzymatic component of telomerase hTERT and conditional expression of c-Myc. ISPNs can be propagated as stable adherent cell lines, and rapidly differentiated into highly homogeneous MSN-like cultures within 2 weeks, as demonstrated by immunocytochemical criteria. Differentiated ISPNs recapitulate major HD-related phenotypes of the parental iPSC model, including brain-derived neurotrophic factor (BDNF)-withdrawal-induced cell death that can be rescued by small molecules previously validated in the parental iPSC model. Proteome and RNA-seq analyses demonstrate separation of HD versus control samples by principal component analysis. We identified several networks, pathways, and upstream regulators, also found altered in HD iPSCs, other HD models, and HD patient samples. HD ISPN lines may be useful for studying HD-related cellular pathogenesis, and for use as a platform for HD target identification and screening experimental therapeutics. The described approach for generation of ISPNs from differentiated patient-derived iPSCs could be applied to a larger allelic series of HD cell lines, and to comparable modeling of other genetic disorders.


Asunto(s)
Enfermedad de Huntington , Células Madre Pluripotentes Inducidas , Diferenciación Celular/genética , Línea Celular , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/terapia , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/metabolismo
4.
Hum Mol Genet ; 29(8): 1340-1352, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32242231

RESUMEN

Nemo-like kinase (NLK), an evolutionarily conserved serine/threonine kinase, is highly expressed in the brain, but its function in the adult brain remains not well understood. In this study, we identify NLK as an interactor of huntingtin protein (HTT). We report that NLK levels are significantly decreased in HD human brain and HD models. Importantly, overexpression of NLK in the striatum attenuates brain atrophy, preserves striatal DARPP32 levels and reduces mutant HTT (mHTT) aggregation in HD mice. In contrast, genetic reduction of NLK exacerbates brain atrophy and loss of DARPP32 in HD mice. Moreover, we demonstrate that NLK lowers mHTT levels in a kinase activity-dependent manner, while having no significant effect on normal HTT protein levels in mouse striatal cells, human cells and HD mouse models. The NLK-mediated lowering of mHTT is associated with enhanced phosphorylation of mHTT. Phosphorylation defective mutation of serine at amino acid 120 (S120) abolishes the mHTT-lowering effect of NLK, suggesting that S120 phosphorylation is an important step in the NLK-mediated lowering of mHTT. A further mechanistic study suggests that NLK promotes mHTT ubiquitination and degradation via the proteasome pathway. Taken together, our results indicate a protective role of NLK in HD and reveal a new molecular target to reduce mHTT levels.


Asunto(s)
Atrofia/genética , Fosfoproteína 32 Regulada por Dopamina y AMPc/genética , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Proteínas Serina-Treonina Quinasas/genética , Animales , Atrofia/patología , Encéfalo/metabolismo , Encéfalo/patología , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Humanos , Enfermedad de Huntington/patología , Ratones , Neostriado/metabolismo , Neostriado/patología , Neuronas/metabolismo , Neuronas/patología , Fosforilación/genética , Complejo de la Endopetidasa Proteasomal/genética
5.
Hum Mol Genet ; 27(9): 1545-1555, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29462355

RESUMEN

Huntington's disease (HD) is caused by a cytosine-adenine-guanine (CAG) trinucleotide repeat expansion in the huntingtin (HTT) gene encoding an elongated polyglutamine tract within the N-terminal of the huntingtin protein (Htt) and leads to Htt misfolding, aberrant protein aggregation, and progressive appearance of disease symptoms. Chronic activation of endoplasmic reticulum (ER) stress by mutant Htt (mHtt) results in cellular dysfunction and ultimately cell death. Protein disulfide isomerase (PDI) is a chaperone protein located in the ER. Our previous studies demonstrated that mHtt caused PDI to accumulate at mitochondria-associated ER membranes and triggered cell death, and that modulating PDI activity using small molecules protected cells again mHtt toxicity in cell and brain slice models of HD. In this study, we demonstrated that PDI is upregulated in the HD human brain, in cell and mouse models. Chronic administration of a reversible, brain penetrable small molecule PDI modulator, LOC14 (20 mg/kg/day), significantly improved motor function, attenuated brain atrophy and extended survival in the N171-82Q HD mice. Moreover, LOC14 preserved medium spiny neuronal marker dopamine- and cyclic-AMP-regulated phosphoprotein of molecular weight 32 000 (DARPP32) levels in the striatum of HD mice. Mechanistic study revealed that LOC14 suppressed mHtt-induced ER stress, indicated by repressing the abnormally upregulated ER stress proteins in HD models. These findings suggest that LOC14 is promising to be further optimized for clinical trials of HD, and modulation of signaling pathways coping with ER stress may constitute an attractive approach to reduce mHtt toxicity and identify new therapeutic targets for treatment of HD.


Asunto(s)
Proteína Huntingtina/metabolismo , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Atrofia/tratamiento farmacológico , Atrofia/genética , Atrofia/metabolismo , Western Blotting , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/genética , Estrés del Retículo Endoplásmico/fisiología , Femenino , Enfermedad de Huntington/genética , Imagen por Resonancia Magnética , Masculino , Ratones , Mutación/genética , Proteína Disulfuro Isomerasas/antagonistas & inhibidores , Proteína Disulfuro Isomerasas/genética , Espectrometría de Masas en Tándem
6.
J Proteome Res ; 16(8): 2692-2708, 2017 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-28653853

RESUMEN

Post-translational modifications (PTMs) of proteins regulate various cellular processes. PTMs of polyglutamine-expanded huntingtin (Htt) protein, which causes Huntington's disease (HD), are likely modulators of HD pathogenesis. Previous studies have identified and characterized several PTMs on exogenously expressed Htt fragments, but none of them were designed to systematically characterize PTMs on the endogenous full-length Htt protein. We found that full-length endogenous Htt, which was immunoprecipitated from HD knock-in mouse and human post-mortem brain, is suitable for detection of PTMs by mass spectrometry. Using label-free and mass tag labeling-based approaches, we identified near 40 PTMs, of which half are novel (data are available via ProteomeXchange with identifier PXD005753). Most PTMs were located in clusters within predicted unstructured domains rather than within the predicted α-helical structured HEAT repeats. Using quantitative mass spectrometry, we detected significant differences in the stoichiometry of several PTMs between HD and WT mouse brain. The mass-spectrometry identification and quantitation were verified using phospho-specific antibodies for selected PTMs. To further validate our findings, we introduced individual PTM alterations within full-length Htt and identified several PTMs that can modulate its subcellular localization in striatal cells. These findings will be instrumental in further assembling the Htt PTM framework and highlight several PTMs as potential therapeutic targets for HD.


Asunto(s)
Proteína Huntingtina/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Encéfalo/metabolismo , Química Encefálica , Cuerpo Estriado/patología , Humanos , Proteína Huntingtina/química , Enfermedad de Huntington/patología , Espectrometría de Masas/métodos , Ratones , Proteínas del Tejido Nervioso/metabolismo , Péptido Hidrolasas/química , Fosforilación , Dominios Proteicos
7.
Hum Mol Genet ; 24(9): 2508-27, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25609071

RESUMEN

White matter abnormalities have been reported in premanifest Huntington's disease (HD) subjects before overt striatal neuronal loss, but whether the white matter changes represent a necessary step towards further pathology and the underlying mechanism of these changes remains unknown. Here, we characterized a novel knock-in mouse model that expresses mouse HD gene homolog (Hdh) with extended CAG repeat- HdhQ250, which was derived from the selective breeding of HdhQ150 mice. HdhQ250 mice manifest an accelerated and robust phenotype compared with its parent line. HdhQ250 mice exhibit progressive motor deficits, reduction in striatal and cortical volume, accumulation of mutant huntingtin aggregation, decreased levels of DARPP32 and BDNF and altered striatal metabolites. The abnormalities detected in this mouse model are reminiscent of several aspects of human HD. In addition, disturbed myelination was evident in postnatal Day 14 HdhQ250 mouse brain, including reduced levels of myelin regulatory factor and myelin basic protein, and decreased numbers of myelinated axons in the corpus callosum. Thinner myelin sheaths, indicated by increased G-ratio of myelin, were also detected in the corpus callosum of adult HdhQ250 mice. Moreover, proliferation of oligodendrocyte precursor cells is altered by mutant huntingtin both in vitro and in vivo. Our data indicate that this model is suitable for understanding comprehensive pathogenesis of HD in white matter and gray matter as well as developing therapeutics for HD.


Asunto(s)
Encéfalo/patología , Enfermedad de Huntington/patología , Enfermedad de Huntington/fisiopatología , Actividad Motora , Sustancia Blanca/patología , Alelos , Animales , Atrofia , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proliferación Celular , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Espectroscopía de Resonancia Magnética , Ratones , Ratones Transgénicos , Mutación , Vaina de Mielina/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Oligodendroglía/metabolismo , Tamaño de los Órganos , Agregación Patológica de Proteínas , Sustancia Blanca/metabolismo
8.
Hum Mol Genet ; 22(12): 2462-70, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23446639

RESUMEN

Huntington's disease (HD) is a fatal neurodegenerative disease characterized by abnormal motor coordination, cognitive decline and psychiatric disorders. This disease is caused by an expanded CAG trinucleotide repeat in the gene encoding the protein huntingtin. Reduced levels of brain-derived neurotrophic factor (BDNF) in the brain, which results from transcriptional inhibition and axonal transport deficits mediated by mutant huntingtin, have been suggested as critical factors underlying selective neurodegeneration in both HD patients and HD mouse models. BDNF activates its high-affinity receptor TrkB and promotes neuronal survival; restoring BDNF signaling is thus of particular therapeutic interest. In the present study, we evaluated the ability of a small-molecule TrkB agonist 7,8-dihydroxyflavone (7,8-DHF) and its synthetic derivative 4'-dimethylamino-7,8- dihydroxyflavone (4'-DMA-7,8-DHF) to protect neurons in the well-characterized N171-82Q HD mouse model. We found that chronic administration of 7, 8-DHF (5 mg/kg) or 4'-DMA-7,8-DHF (1 mg/kg) significantly improved motor deficits, ameliorated brain atrophy and extended survival in these N171-82Q HD mice. Moreover, 4'-DMA-7,8-DHF preserved DARPP32 levels in the striatum and rescued mutant huntingtin-induced impairment of neurogenesis in the N171-82Q HD mice. These data highlight consideration of TrkB as a therapeutic target in HD and suggest that small-molecule TrkB agonists that penetrate the brain have high potential to be further tested in clinical trials of HD.


Asunto(s)
Flavonas/administración & dosificación , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/mortalidad , Receptor trkB/agonistas , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Masculino , Ratones , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Receptor trkB/genética , Receptor trkB/metabolismo , Transducción de Señal/efectos de los fármacos , Sobrevida
9.
Mov Disord ; 29(11): 1366-74, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25124273

RESUMEN

The polyglutamine expansion within huntingtin is the causative factor in the pathogenesis of Huntington's disease (HD). Although the underlying mechanisms by which mutant huntingtin causes neuronal dysfunction and degeneration have not been fully elucidated, compelling evidence suggests that mitochondrial dysfunction and compromised energy metabolism are key players in HD pathogenesis. Longitudinal studies of HD subjects have shown reductions in glucose utilization before the disease clinical onset. Preferential striatal neurodegeneration, a hallmark of HD pathogenesis, also has been associated with interrupted energy metabolism. Data from genetic HD models indicate that mutant huntingtin disrupts mitochondrial bioenergetics and prevents adenosine triphosphate (ATP) generation, implying altered energy metabolism as an important component of HD pathogenesis. Here we revisit the evidence of abnormal energy metabolism in the central nervous system of HD patients, review our current understanding of the molecular mechanisms underlying abnormal metabolism induced by mutant huntingtin, and discuss the promising therapeutic development by halting abnormal metabolism in HD.


Asunto(s)
Enfermedad de Huntington/genética , Enfermedades Metabólicas/genética , Proteínas del Tejido Nervioso/genética , Péptidos/genética , Adenosina Trifosfato/metabolismo , Metabolismo Energético/genética , Humanos , Proteína Huntingtina , Enfermedad de Huntington/complicaciones , Enfermedades Metabólicas/etiología
10.
J Neurosci ; 32(1): 183-93, 2012 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-22219281

RESUMEN

Huntington's disease (HD) is caused by a polyglutamine expansion in the Huntingtin (Htt) protein. Proteolytic cleavage of Htt into toxic N-terminal fragments is believed to be a key aspect of pathogenesis. The best characterized putative cleavage event is at amino acid 586, hypothesized to be mediated by caspase 6. A corollary of the caspase 6 cleavage hypothesis is that the caspase 6 fragment should be a toxic fragment. To test this hypothesis, and further characterize the role of this fragment, we have generated transgenic mice expressing the N-terminal 586 aa of Htt with a polyglutamine repeat length of 82 (N586-82Q), under the control of the prion promoter. N586-82Q mice show a clear progressive rotarod deficit by 4 months of age, and are hyperactive starting at 5 months, later changing to hypoactivity before early mortality. MRI studies reveal widespread brain atrophy, and histologic studies demonstrate an abundance of Htt aggregates, mostly cytoplasmic, which are predominantly composed of the N586-82Q polypeptide. Smaller soluble N-terminal fragments appear to accumulate over time, peaking at 4 months, and are predominantly found in the nuclear fraction. This model appears to have a phenotype more severe than current full-length Htt models, but less severe than HD mouse models expressing shorter Htt fragments. These studies suggest that the caspase 6 fragment may be a transient intermediate, that fragment size is a factor contributing to the rate of disease progression, and that short soluble nuclear fragments may be most relevant to pathogenesis.


Asunto(s)
Caspasa 6/fisiología , Enfermedad de Huntington/metabolismo , Degeneración Nerviosa/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Fragmentos de Péptidos/genética , Animales , Atrofia , Modelos Animales de Enfermedad , Humanos , Proteína Huntingtina , Enfermedad de Huntington/patología , Enfermedad de Huntington/fisiopatología , Ratones , Ratones Endogámicos , Ratones Transgénicos , Degeneración Nerviosa/patología , Degeneración Nerviosa/fisiopatología , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/toxicidad , Proteínas Nucleares/metabolismo , Proteínas Nucleares/toxicidad , Fragmentos de Péptidos/biosíntesis , Fragmentos de Péptidos/toxicidad , Expansión de Repetición de Trinucleótido/fisiología
11.
J Biol Chem ; 287(29): 24460-72, 2012 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-22648412

RESUMEN

Huntington disease (HD) is an inherited neurodegenerative disorder caused by an abnormal polyglutamine expansion in the protein Huntingtin (Htt). Currently, no cure is available for HD. The mechanisms by which mutant Htt causes neuronal dysfunction and degeneration remain to be fully elucidated. Nevertheless, mitochondrial dysfunction has been suggested as a key event mediating mutant Htt-induced neurotoxicity because neurons are energy-demanding and particularly susceptible to energy deficits and oxidative stress. SIRT3, a member of sirtuin family, is localized to mitochondria and has been implicated in energy metabolism. Notably, we found that cells expressing mutant Htt displayed reduced SIRT3 levels. trans-(-)-ε-Viniferin (viniferin), a natural product among our 22 collected naturally occurring and semisynthetic stilbenic compounds, significantly attenuated mutant Htt-induced depletion of SIRT3 and protected cells from mutant Htt. We demonstrate that viniferin decreases levels of reactive oxygen species and prevents loss of mitochondrial membrane potential in cells expressing mutant Htt. Expression of mutant Htt results in decreased deacetylase activity of SIRT3 and further leads to reduction in cellular NAD(+) levels and mitochondrial biogenesis in cells. Viniferin activates AMP-activated kinase and enhances mitochondrial biogenesis. Knockdown of SIRT3 significantly inhibited viniferin-mediated AMP-activated kinase activation and diminished the neuroprotective effects of viniferin, suggesting that SIRT3 mediates the neuroprotection of viniferin. In conclusion, we establish a novel role for mitochondrial SIRT3 in HD pathogenesis and discovered a natural product that has potent neuroprotection in HD models. Our results suggest that increasing mitochondrial SIRT3 might be considered as a new therapeutic approach to counteract HD, as well as other neurodegenerative diseases with similar mechanisms.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Benzofuranos/farmacología , Enfermedad de Huntington/metabolismo , Mitocondrias/metabolismo , Sirtuina 3/metabolismo , Estilbenos/farmacología , Animales , Línea Celular Tumoral , Metabolismo Energético/efectos de los fármacos , Ratones , Ratas
12.
Neuroimage ; 49(3): 2340-51, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19850133

RESUMEN

Mouse models of human diseases play crucial roles in understanding disease mechanisms and developing therapeutic measures. Huntington's disease (HD) is characterized by striatal atrophy that begins long before the onset of motor symptoms. In symptomatic HD, striatal volumes decline predictably with disease course. Thus, imaging based volumetric measures have been proposed as outcomes for presymptomatic as well as symptomatic clinical trials of HD. Magnetic resonance imaging of the mouse brain structures is becoming widely available and has been proposed as one of the biomarkers of disease progression and drug efficacy testing. However, three-dimensional and quantitative morphological analyses of the brains are not straightforward. In this paper, we describe a tool for automated segmentation and voxel-based morphological analyses of the mouse brains. This tool was applied to a well-established mouse model of Huntington's disease, the R6/2 transgenic mouse strain. Comparison between the automated and manual segmentation results showed excellent agreement in most brain regions. The automated method was able to sensitively detect atrophy as early as 4 weeks of age and accurately follow disease progression. Comparison between ex vivo and in vivo MRI suggests that the ex vivo end-point measurement of brain morphology is also a valid approach except for the morphology of the ventricles. This is the first report of longitudinal characterization of brain atrophy in a mouse model of Huntington's disease by using automatic morphological analysis.


Asunto(s)
Encéfalo/patología , Enfermedad de Huntington/patología , Procesamiento de Imagen Asistido por Computador/métodos , Imagen por Resonancia Magnética , Animales , Atrofia/patología , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Transgénicos
13.
J Neurochem ; 114(2): 419-29, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20412383

RESUMEN

The E46K is a point mutation in alpha-synuclein (alpha-syn) that causes familial Parkinsonism with Lewy body dementia. We have now generated a cell model of Parkinsonism/Parkinson's disease (PD) and demonstrated cell toxicity after expression of E46K in the differentiated PC12 cells. E46K alpha-syn inhibited proteasome activity and induced mitochondrial depolarization in the cell model. Baicalein has been reported to inhibit fibrillation of wild type alpha-syn in vitro, and to protect neurons against several chemical-induced models of PD. We now report that baicalein significantly attenuated E46K-induced mitochondrial depolarization and proteasome inhibition, and protected cells against E46K-induced toxicity in a cell model of PD. Baicalein also reduced E46K fibrilization in vitro, with a concentration-dependent decrease in beta sheet conformation, though it increased some oligomeric species, and decreased formation of E46K alpha-syn-induced aggregates and rescued toxicity in N2A cells. Taken together, these data indicate that mitochondrial dysfunction, proteasome inhibition and specific aspects of abnormal E46K aggregation accompany E46K alpha-syn-induced cell toxicity, and baicalein can protect as well as altering aggregation properties. Baicalein has potential as a tool to understand the relation between different aggregation species and toxicity, and might be a candidate compound for further validation by using in vivo alpha-syn genetic PD models.


Asunto(s)
Flavanonas/farmacología , Trastornos Parkinsonianos/metabolismo , alfa-Sinucleína/genética , Animales , Muerte Celular , Diferenciación Celular , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mutación , Neuronas/metabolismo , Neuronas/ultraestructura , Células PC12 , Trastornos Parkinsonianos/genética , Inhibidores de Proteasoma , Ratas , alfa-Sinucleína/biosíntesis
14.
J Neurophysiol ; 102(6): 3251-9, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19776361

RESUMEN

Synaptic plasticity has been extensively studied in principal neurons of the neocortex, but less work has been done on GABAergic interneurons. Interneurons consist of multiple subtypes and their synaptic properties vary between subtypes. In the present study, we have examined long-term potentiation (LTP) of excitatory synapses on somatostatin (SS)-expressing interneurons in neocortex using transgenic mice that express enhanced green fluorescent protein in these interneurons. We found that a strong theta burst stimulation was required to induce LTP in SS interneurons. LTP was associated with a reduction in paired-pulse facilitation and was not blocked by an N-methyl-d-aspartate receptor (NMDAR) antagonist. LTP was not affected by chelating postsynaptic Ca(2+) with BAPTA, a fast Ca(2+) chelator, and blocking L-type voltage-dependent Ca(2+) channels with nimodipine. Application of forskolin, an activator of adenylate cyclase that increases cyclic adenosine monophosphate (cAMP) concentration, enhanced synaptic transmission and occluded subsequent induction of LTP. Finally, we found that LTP was blocked by protein kinase A (PKA) inhibitors. Our results suggest that excitatory synapses on SS interneurons express a presynaptic form of LTP that is not dependent on NMDARs or postsynaptic Ca(2+) rise but is dependent on the cAMP-PKA signaling pathway.


Asunto(s)
Interneuronas/fisiología , Potenciación a Largo Plazo/fisiología , Neocórtex/citología , Somatostatina/metabolismo , Sinapsis/fisiología , Animales , Animales Recién Nacidos , Biofisica , Estimulación Eléctrica/métodos , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Antagonistas del GABA/farmacología , Glutamato Descarboxilasa/genética , Proteínas Fluorescentes Verdes/genética , Técnicas In Vitro , Ratones , Ratones Transgénicos , Técnicas de Placa-Clamp/métodos , Picrotoxina/farmacología , Quinoxalinas/farmacología
15.
Neurobiol Dis ; 30(3): 293-302, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18395459

RESUMEN

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder characterized by chorea, incoordination, and shortened life-span, and by huntingtin inclusions and neurodegeneration. We previously screened the 1040 FDA-approved compounds from the NINDS compound library and found that a compound, nipecotic acid, significantly reduced mutant huntingtin aggregations and blocked cell toxicity in an inducible cell model of HD. Because nipecotic acid does not cross the blood-brain barrier (BBB), we studied its analogue, tiagabine, which is able to cross the BBB, in both N171-82Q and R6/2 transgenic mouse models of HD. Tiagabine was administered intraperitoneally at 2 and 5 mg/kg daily in HD mice. We found that tiagabine extended survival, improved motor performance, and attenuated brain atrophy and neurodegeneration in N171-82Q HD mice. These beneficial effects were further confirmed in R6/2 HD mice. The levels of tiagabine at effective doses in mouse serum are comparable to the levels in human patients treated with tiagabine. These results suggest that tiagabine may have beneficial effects in the treatment of HD. Because tiagabine is an FDA-approved drug, it may be a promising candidate for future clinical trials for the treatment of HD.


Asunto(s)
Enfermedad de Huntington/genética , Enfermedad de Huntington/prevención & control , Fármacos Neuroprotectores/uso terapéutico , Ácidos Nipecóticos/uso terapéutico , Animales , Arginina/genética , Asparagina/genética , Modelos Animales de Enfermedad , Femenino , Glutamina/genética , Enfermedad de Huntington/patología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Mutantes Neurológicos , Ratones Transgénicos , Células PC12 , Ratas , Tiagabina
16.
Sheng Li Xue Bao ; 60(2): 270-4, 2008 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-18425317

RESUMEN

Previous reports suggested that a novel stimulus pattern of multi-train stimulus at low-frequency (2-Hz or 5-Hz) could induce stable long-term depression (LTD) in the CA1 area of adult rat hippocampus. In the present study, in order to determine the mechanism in LTD induced by the two novel tetanus patterns, changes in the population spikes (PS) in the hippocampal CA1 area of adult rats following the multi-train stimulus in the presence of AP5 [antagonist of N-methyl-D-aspartate receptors (NMDARs)] or MCPG [antagonist of type I/II metabotropic glutamate receptors (mGluRs)] were recorded. The results showed that both AP5 and MCPG inhibited the LTD induced by 2-Hz multi-train stimulus. The mean amplitude of population spikes (PSA) normalized to the baseline was (96.0±3.5)% after applying AP5 (n=10) and (95.7±4.1)% after applying MCPG (n=8), respectively, measured at 20 min post-tetanus. While 5-Hz multi-train tetanus failed to induce LTD in the presence of MCPG. The mean PSA was (73.6±4.4)% (n=10) and (98.2±8.9)% (n=8) in the presence of AP5 and MCPG, respectively, measured at 35 min post-tetanus. So it is suggested that LTD induced by 2-Hz multi-train tetanus involves co-activation of NMDARs and mGluRs, while LTD induced by 5-Hz multi-train tetanus is only related to activation of mGluRs.


Asunto(s)
Región CA1 Hipocampal/fisiología , Depresión Sináptica a Largo Plazo , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , 2-Amino-5-fosfonovalerato/farmacología , Animales , Glicina/análogos & derivados , Glicina/farmacología , Ratas
17.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 39(4): 627-30, 2008 Jul.
Artículo en Zh | MEDLINE | ID: mdl-18798510

RESUMEN

OBJECTIVE: To test the impact of fetal growth restriction (FGR) on the spatial learning and memory abilities of the offspring of rats. METHODS: FGR Model of Sprague-Dawley rats was constructed according to the method of passive smoking. The offspring of the rats were divided into male FGR group, male control group, female FGR group and female control group. Within each group, the rats were randomly divided into three subgroups to be tested at 1, 2, and 4 months of age, respectively (n =10 for each subgroup). Morris water maze task was performed to assess the spatial learning and memory abilities of the rats. RESULTS: The escape latencies to find the platform were shortened with increased training times for all of the rats. At the age of 1 and 2 months, both male and female rats in the FGR group spent more time in finding the platform than their counterparts in the control group (P < 0.05). At the age of 4 months, significant prolonged latency was only found in the female rats. The rats in the FGR group, except the 4 months old male rats, were more likely to use non-effective strategies (random or marginal strategies) to find the platform than the efficient strategies (tendency or straight strategies). The rats in the FGR group stayed in the platform shorter than those in the control groups (P < 0.05). CONCLUSION: FGR can cause gender- and age-specific impairment of spatial learning and memory abilities to the offspring.


Asunto(s)
Retardo del Crecimiento Fetal/fisiopatología , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/fisiopatología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Animales , Femenino , Retardo del Crecimiento Fetal/etiología , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/etiología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Contaminación por Humo de Tabaco/efectos adversos
18.
Exp Neurol ; 293: 83-90, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28359739

RESUMEN

Huntington's disease (HD) is a neurodegenerative disorder caused by a CAG repeat expansion in the first exon of the gene huntingtin. There is no treatment to prevent or delay the disease course of HD currently. Oxidative stress and mitochondrial dysfunction have emerged as key determinants of the disease progression in HD. Therefore, counteracting mutant huntingtin (mHtt)-induced oxidative stress and mitochondrial dysfunction appears as a new approach to treat this devastating disease. Interestingly, mild mitochondrial uncoupling improves neuronal resistance to stress and facilitates neuronal survival. Mild mitochondrial uncoupling can be induced by the proper dose of 2,4-dinitrophenol (DNP), a proton ionophore that was previously used for weight loss. In this study, we evaluated the effects of chronic administration of DNP at three doses (0.5, 1, 5mg/kg/day) on mHtt-induced behavioral deficits and cellular abnormalities in the N171-82Q HD mouse model. DNP at a low dose (1mg/kg/day) significantly improved motor function and preserved medium spiny neuronal marker DARPP32 and postsynaptic protein PSD95 in the striatum of HD mice. Further mechanistic study suggests that DNP at this dose reduced oxidative stress in HD mice, which was indicated by reduced levels of F2-isoprostanes in the brain of HD mice treated with DNP. Our data indicated that DNP provided behavioral benefit and neuroprotective effect at a weight neutral dose in HD mice, suggesting that the potential value of repositioning DNP to HD treatment is warranted in well-controlled clinical trials in HD.


Asunto(s)
2,4-Dinitrofenol/farmacología , 2,4-Dinitrofenol/uso terapéutico , Enfermedad de Huntington/tratamiento farmacológico , Actividad Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Cuerpo Estriado/diagnóstico por imagen , Cuerpo Estriado/patología , Homólogo 4 de la Proteína Discs Large , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Guanilato-Quinasas/metabolismo , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Enfermedad de Huntington/fisiopatología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Actividad Motora/genética , Neuronas/metabolismo , Estrés Oxidativo/genética
19.
Sheng Li Xue Bao ; 58(3): 287-91, 2006 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-16786115

RESUMEN

Previous reports suggested that a low-frequency stimulus (LFS) of 1~2 Hz (600~900 pulses) induced a homosynaptic long-term depression (LTD) of synaptic efficacy in the hippocampal CA1 area of young rats (< 4-week old). However, these stimulation protocols often failed to induce LTD in the adult CA1 hippocampus. In the present study, we examined the effects of two novel tetanus patterns on LTD induction in adult rat hippocampal slices. We determined that these novel stimulation protocols induced LTD in the adult hippocampus, and that the characteristics of induced LTD were parameter-specific, including latency (period from the end of tetanus to a beginning of LTD) and the amplitude of LTD. These results suggest that LFS with certain patterns can induce LTD in the CA1 area of adult rat hippocampal slices, and that the multi-trains of 2-Hz protocol provided more effective response than the 5-Hz protocol.


Asunto(s)
Región CA1 Hipocampal/fisiología , Estimulación Eléctrica , Depresión Sináptica a Largo Plazo/fisiología , Animales , Técnicas In Vitro , Masculino , Ratas , Ratas Sprague-Dawley
20.
PLoS One ; 11(2): e0148839, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26859386

RESUMEN

Huntington's disease (HD) is caused by an expansion of the trinucleotide poly (CAG) tract located in exon 1 of the huntingtin (Htt) gene leading to progressive neurodegeneration in selected brain regions, and associated functional impairments in motor, cognitive, and psychiatric domains. Since the discovery of the gene mutation that causes the disease, mouse models have been developed by different strategies. Recently, a new model, the zQ175 knock-in (KI) line, was developed in an attempt to have the Htt gene in a context and causing a phenotype that more closely mimics HD in humans. The behavioral phenotype was characterized across the independent laboratories and important features reminiscent of human HD are observed in zQ175 mice. In the current study, we characterized the zQ175 model housed in an academic laboratory under reversed dark-light cycle, including motor function, in vivo longitudinal structural MRI imaging for brain volume, MRS for striatal metabolites, neuropathology, as well as a panel of key disease marker proteins in the striatum at different ages. Our results suggest that homozygous zQ175 mice exhibited significant brain atrophy before the motor deficits and brain metabolite changes. Altered striatal medium spiny neuronal marker, postsynaptic marker protein and complement component C1qC also characterized zQ175 mice. Our results confirmed that the zQ175 KI model is valuable in understanding of HD-like pathophysiology and evaluation of potential therapeutics. Our data also provide suggestions to select appropriate outcome measurements in preclinical studies using the zQ175 mice.


Asunto(s)
Encéfalo/metabolismo , Enfermedad de Huntington/genética , Ratones Transgénicos/genética , Animales , Atrofia , Western Blotting , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Técnicas de Sustitución del Gen , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Enfermedad de Huntington/fisiopatología , Enfermedad de Huntington/psicología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Transgénicos/metabolismo , Destreza Motora/fisiología , Neuroimagen , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA