Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
PLoS One ; 12(4): e0174264, 2017.
Article in English | MEDLINE | ID: mdl-28388629

ABSTRACT

Tubulin alpha 8 (Tuba8) is the most divergent member of the highly conserved alpha tubulin family, and uniquely lacks two key post-translational modification sites. It is abundantly expressed in testis and muscle, with lower levels in the brain. We previously identified homozygous hypomorphic TUBA8 mutations in human subjects with a polymicrogyria (PMG) syndrome, suggesting its involvement in development of the cerebral cortex. We have now generated and characterized a Tuba8 knockout mouse model. Homozygous mice were confirmed to lack Tuba8 protein in the testis, but did not display PMG and appeared to be neurologically normal. In response to this finding, we re-analyzed the human PMG subjects using whole exome sequencing. This resulted in identification of an additional homozygous loss-of-function mutation in SNAP29, suggesting that SNAP29 deficiency, rather than TUBA8 deficiency, may underlie most or all of the neurodevelopmental anomalies in these subjects. Nonetheless, in the mouse brain, Tuba8 specifically localised to the cerebellar Purkinje cells, suggesting that the human mutations may affect or modify motor control. In the testis, Tuba8 localisation was cell-type specific. It was restricted to spermiogenesis with a strong acrosomal localization that was gradually replaced by cytoplasmic distribution and was absent from spermatozoa. Although the knockout mice were fertile, the localisation pattern indicated that Tuba8 may have a role in spermatid development during spermatogenesis, rather than as a component of the mature microtubule-rich flagellum itself.


Subject(s)
Brain/embryology , Spermatogenesis/genetics , Tubulin/genetics , Animals , Exome , Homozygote , Mice , Mice, Knockout
2.
PLoS One ; 7(9): e44498, 2012.
Article in English | MEDLINE | ID: mdl-22973455

ABSTRACT

Histone deacetylase (HDAC) inhibitors have received considerable attention as potential therapeutics for a variety of cancers and neurological disorders. Recent publications on a class of pimelic diphenylamide HDAC inhibitors have highlighted their promise in the treatment of the neurodegenerative diseases Friedreich's ataxia and Huntington's disease, based on efficacy in cell and mouse models. These studies' authors have proposed that the unique action of these compounds compared to hydroxamic acid-based HDAC inhibitors results from their unusual slow-on/slow-off kinetics of binding, preferentially to HDAC3, resulting in a distinctive pharmacological profile and reduced toxicity. Here, we evaluate the HDAC subtype selectivity, cellular activity, absorption, distribution, metabolism and excretion (ADME) properties, as well as the central pharmacodynamic profile of one such compound, HDACi 4b, previously described to show efficacy in vivo in the R6/2 mouse model of Huntington's disease. Based on our data reported here, we conclude that while the in vitro selectivity and binding mode are largely in agreement with previous reports, the physicochemical properties, metabolic and p-glycoprotein (Pgp) substrate liability of HDACi 4b render this compound suboptimal to investigate central Class I HDAC inhibition in vivo in mouse per oral administration. A drug administration regimen using HDACi 4b dissolved in drinking water was used in the previous proof of concept study, casting doubt on the validation of CNS HDAC3 inhibition as a target for the treatment of Huntington's disease. We highlight physicochemical stability and metabolic issues with 4b that are likely intrinsic liabilities of the benzamide chemotype in general.


Subject(s)
Central Nervous System/metabolism , Friedreich Ataxia/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Huntington Disease/drug therapy , Pimelic Acids/pharmacology , Administration, Oral , Animals , Caco-2 Cells , Chromatography, High Pressure Liquid , Dogs , Friedreich Ataxia/enzymology , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/pharmacokinetics , Histone Deacetylase Inhibitors/therapeutic use , Humans , Huntington Disease/enzymology , Madin Darby Canine Kidney Cells , Mice , Microsomes, Liver/metabolism , Pimelic Acids/administration & dosage , Pimelic Acids/chemical synthesis , Pimelic Acids/pharmacokinetics , Pimelic Acids/therapeutic use , Tandem Mass Spectrometry
3.
PLoS One ; 6(11): e27746, 2011.
Article in English | MEDLINE | ID: mdl-22140466

ABSTRACT

Huntington's disease (HD) is a progressive neurological disorder for which there are no disease-modifying treatments. Transcriptional dysregulation is a major molecular feature of HD, which significantly contributes to disease progression. Therefore, the development of histone deacetylase (HDAC) inhibitors as therapeutics for HD has been energetically pursued. Suberoylanilide hydroxamic acid (SAHA) - a class I HDAC as well an HDAC6 inhibitor, improved motor impairment in the R6/2 mouse model of HD. Recently it has been found that SAHA can also promote the degradation of HDAC4 and possibly other class IIa HDACs at the protein level in various cancer cell lines. To elucidate whether SAHA is a potent modifier of HDAC protein levels in vivo, we performed two independent mouse trials. Both WT and R6/2 mice were chronically treated with SAHA and vehicle. We found that prolonged SAHA treatment causes the degradation of HDAC4 in cortex and brain stem, but not hippocampus, without affecting its transcript levels in vivo. Similarly, SAHA also decreased HDAC2 levels without modifying the expression of its mRNA. Consistent with our previous data, SAHA treatment diminishes Hdac7 transcript levels in both wild type and R6/2 brains and unexpectedly was found to decrease Hdac11 in R6/2 but not wild type. We investigated the effects of SAHA administration on well-characterised molecular readouts of disease progression. We found that SAHA reduces SDS-insoluble aggregate load in the cortex and brain stem but not in the hippocampus of the R6/2 brains, and that this was accompanied by restoration of Bdnf cortical transcript levels.


Subject(s)
Histone Deacetylase 2/metabolism , Histone Deacetylases/metabolism , Huntington Disease/drug therapy , Huntington Disease/enzymology , Hydroxamic Acids/therapeutic use , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Exons/genetics , Histone Deacetylase 2/genetics , Histone Deacetylases/genetics , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/pharmacology , Mice , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vorinostat
4.
J Clin Invest ; 121(8): 3306-19, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21785217

ABSTRACT

Huntington disease (HD) is a devastating neurodegenerative disorder for which there are no disease-modifying treatments. Previous studies have proposed that activation of the heat shock response (HSR) via the transcription factor heat shock factor 1 (HSF1) may be of therapeutic benefit. However, the effect of disease progression on the HSR and the therapeutic potential of this pathway are currently unknown. Here, we used a brain-penetrating HSP90 inhibitor and physiological, molecular, and behavioral readouts to demonstrate that pharmacological activation of HSF1 improves huntingtin aggregate load, motor performance, and other HD-related phenotypes in the R6/2 mouse model of HD. However, the beneficial effects of this treatment were transient and diminished with disease progression. Molecular analyses to understand the transient nature of these effects revealed altered chromatin architecture, reduced HSF1 binding, and impaired HSR accompanied disease progression in both the R6/2 transgenic and HdhQ150 knockin mouse models of HD. Taken together, our findings reveal that the HSR, a major inducible regulator of protein homeostasis and longevity, is disrupted in HD. Consequently, pharmacological induction of HSF1 as a therapeutic approach to HD is more complex than was previously anticipated.


Subject(s)
Chromatin/chemistry , DNA-Binding Proteins/physiology , Heat-Shock Response/genetics , Huntington Disease/metabolism , Transcription Factors/physiology , Animals , Cell Nucleus/metabolism , Chromatin/metabolism , Cytoplasm/metabolism , Disease Models, Animal , Female , HSP90 Heat-Shock Proteins/metabolism , Heat Shock Transcription Factors , Huntington Disease/genetics , Male , Mice , Mice, Transgenic , Phenotype , Up-Regulation
5.
ACS Chem Biol ; 6(6): 540-6, 2011 Jun 17.
Article in English | MEDLINE | ID: mdl-21370928

ABSTRACT

Sirtuin 2 (SIRT2) deacetylase-dependent inhibition mediates neuroprotective reduction of cholesterol biosynthesis in an in vitro Huntington's disease model. This study sought to identify the first brain-permeable SIRT2 inhibitor and to characterize its cholesterol-reducing properties in neuronal models. Using biochemical sirtuin deacetylation assays, we screened a brain-permeable in silico compound library, yielding 3-(1-azepanylsulfonyl)-N-(3-bromphenyl)benzamide as the most potent and selective SIRT2 inhibitor. Pharmacokinetic studies demonstrated brain-permeability but limited metabolic stability of the selected candidate. In accordance with previous observations, this SIRT2 inhibitor stimulated cytoplasmic retention of sterol regulatory element binding protein-2 and subsequent transcriptional downregulation of cholesterol biosynthesis genes, resulting in reduced total cholesterol in primary striatal neurons. Furthermore, the identified inhibitor reduced cholesterol in cultured naïve neuronal cells and brain slices from wild-type mice. The outcome of this study provides a clear opportunity for lead optimization and drug development, targeting metabolic dysfunctions in CNS disorders where abnormal cholesterol homeostasis is implicated.


Subject(s)
Brain/metabolism , Cholesterol/biosynthesis , Enzyme Inhibitors/pharmacology , Neurons/drug effects , Neurons/metabolism , Sirtuin 2/antagonists & inhibitors , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Mice , Models, Neurological , Molecular Structure , Neurons/enzymology , Permeability , Sirtuin 2/metabolism , Small Molecule Libraries , Stereoisomerism , Structure-Activity Relationship , Tumor Cells, Cultured
6.
Hum Mol Genet ; 19(9): 1756-65, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20147317

ABSTRACT

Huntingtin protein (Htt) is ubiquitously expressed, yet Huntington's disease (HD), a fatal neurologic disorder produced by expansion of an Htt polyglutamine tract, is characterized by neurodegeneration that occurs primarily in the striatum and cerebral cortex. Such discrepancies between sites of expression and pathology occur in multiple neurodegenerative disorders associated with expanded polyglutamine tracts. One possible reason is that disease-modifying factors are tissue-specific. Here, we show that the striatum-enriched protein, CalDAG-GEFI, is severely down-regulated in the striatum of mouse HD models and is down-regulated in HD individuals. In the R6/2 transgenic mouse model of HD, striatal neurons with the largest aggregates of mutant Htt have the lowest levels of CalDAG-GEFI. In a brain-slice explant model of HD, knock-down of CalDAG-GEFI expression rescues striatal neurons from pathology induced by transfection of polyglutamine-expanded Htt exon 1. These findings suggest that the striking down-regulation of CalDAG-GEFI in HD could be a protective mechanism that mitigates Htt-induced degeneration.


Subject(s)
Corpus Striatum/metabolism , Down-Regulation , Guanine Nucleotide Exchange Factors/metabolism , Huntington Disease/metabolism , Animals , Corpus Striatum/pathology , Humans , Huntingtin Protein , Immunoblotting , Immunohistochemistry , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Rats , Rats, Sprague-Dawley
7.
J Biol Chem ; 285(12): 8808-23, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20086007

ABSTRACT

Huntingtin proteolysis has been implicated in the molecular pathogenesis of Huntington disease (HD). Despite an intense effort, the identity of the pathogenic smallest N-terminal fragment has not been determined. Using a panel of anti-huntingtin antibodies, we employed an unbiased approach to generate proteolytic cleavage maps of mutant and wild-type huntingtin in the HdhQ150 knock-in mouse model of HD. We identified 14 prominent N-terminal fragments, which, in addition to the full-length protein, can be readily detected in cytoplasmic but not nuclear fractions. These fragments were detected at all ages and are not a consequence of the pathogenic process. We demonstrated that the smallest fragment is an exon 1 huntingtin protein, known to contain a potent nuclear export signal. Prior to the onset of behavioral phenotypes, the exon 1 protein, and possibly other small fragments, accumulate in neuronal nuclei in the form of a detergent insoluble complex, visualized as diffuse granular nuclear staining in tissue sections. This methodology can be used to validate the inhibition of specific proteases as therapeutic targets for HD by pharmacological or genetic approaches.


Subject(s)
Huntington Disease/metabolism , Mutation , Nerve Tissue Proteins/genetics , Neurons/metabolism , Nuclear Proteins/genetics , Animals , COS Cells , Calpain/chemistry , Cell Nucleus/metabolism , Chlorocebus aethiops , Cytoplasm/metabolism , Disease Models, Animal , Exons , Genotype , Huntingtin Protein , Mice , Protein Structure, Tertiary
8.
Hum Mol Genet ; 19(1): 65-78, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19825844

ABSTRACT

Huntington's disease (HD) is a late-onset neurodegenerative disorder that is characterized neuropathologically by the presence of neuropil aggregates and nuclear inclusions. However, the profile of aggregate structures that are present in the brains of HD patients or of HD mouse models and the relative contribution of specific aggregate structures to disease pathogenesis is unknown. We have used the Seprion ligand to develop a highly sensitive enzyme-linked immunosorbent assay (ELISA)-based method for quantifying aggregated polyglutamine in tissues from HD mouse models. We used a combination of electron microscopy, atomic force microscopy (AFM) and sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) to investigate the aggregate structures isolated by the ligand. We found that the oligomeric, proto-fibrillar and fibrillar aggregates extracted from the brains of R6/2 and HdhQ150 knock-in mice were remarkably similar. Using AFM, we determined that the nanometre globular oligomers isolated from the brains of both mouse models have dimensions identical to those generated from recombinant huntingtin exon 1 proteins. Finally, antibodies that detect exon 1 Htt epitopes differentially recognize the ligand-captured material on SDS-PAGE gels. The Seprion-ligand ELISA provides an assay with good statistical power for use in preclinical pharmacodynamic therapeutic trials or to assess the effects of the genetic manipulation of potential therapeutic targets on aggregate load. This, together with the ability to identify a spectrum of aggregate species in HD mouse tissues, will contribute to our understanding of how these structures relate to the pathogenesis of HD and whether their formation can be manipulated for therapeutic benefit.


Subject(s)
Brain/pathology , Gene Knock-In Techniques , Huntington Disease/pathology , Neuropil Threads/pathology , Animals , Biological Assay , Brain/ultrastructure , Disease Models, Animal , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Exons/genetics , Ligands , Mice , Microscopy, Atomic Force , Microscopy, Immunoelectron , Neuropil Threads/ultrastructure , Peptides/metabolism , Phenotype , Protein Structure, Quaternary , Serotonin Plasma Membrane Transport Proteins/ultrastructure
9.
PLoS One ; 4(11): e8025, 2009 Nov 30.
Article in English | MEDLINE | ID: mdl-19956633

ABSTRACT

BACKGROUND: Huntington's disease (HD) is an inherited progressive neurodegenerative disorder caused by a CAG repeat expansion in the ubiquitously expressed HD gene resulting in an abnormally long polyglutamine repeat in the huntingtin protein. Polyglutamine inclusions are a hallmark of the neuropathology of HD. We have previously shown that inclusion pathology is also present in the peripheral tissues of the R6/2 mouse model of HD which expresses a small N-terminal fragment of mutant huntingtin. To determine whether this peripheral pathology is a consequence of the aberrant expression of this N-terminal fragment, we extend this analysis to the genetically precise knock-in mouse model of HD, HdhQ150, which expresses mutant mouse huntingtin. METHODOLOGY/PRINCIPAL FINDINGS: We have previously standardized the CAG repeat size and strain background of the R6/2 and HdhQ150 knock-in mouse models and found that they develop a comparable and widespread neuropathology. To determine whether HdhQ150 knock-in mice also develop peripheral inclusion pathology, homozygous Hdh(Q150/Q150) mice were perfusion fixed at 22 months of age, and tissues were processed for histology and immunohistochemistry with the anti-huntingtin antibody S830. The peripheral inclusion pathology was almost identical to that found in R6/2 mice at 12 weeks of age with minor differences in inclusion abundance. CONCLUSIONS/SIGNIFICANCE: The highly comparable peripheral inclusion pathology that is present in both the R6/2 and HdhQ150 knock-in models of HD indicates that the presence of peripheral inclusions in R6/2 mice is not a consequence of the aberrant expression of an N-terminal huntingtin protein. It remains to be determined whether peripheral inclusions are a pathological feature of the human disease. Both mouse models carry CAG repeats that cause childhood disease in humans, and therefore, inclusion pathology may be a feature of the childhood rather than the adult forms of HD. It is important to establish the extent to which peripheral pathology causes the peripheral symptoms of HD from the perspective of a mechanistic understanding and future treatment options.


Subject(s)
Huntington Disease/genetics , Peptides/metabolism , Adrenal Glands/metabolism , Animals , Cell Nucleus/metabolism , Disease Models, Animal , Gastric Mucosa/metabolism , Huntingtin Protein , Insulin-Secreting Cells/metabolism , Mice , Mice, Transgenic , Models, Biological , Muscle, Skeletal/metabolism , Mutation , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Protein Structure, Tertiary
10.
PLoS One ; 4(6): e5747, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19484127

ABSTRACT

Huntington's disease (HD) is an inherited, progressive neurological disorder caused by a CAG/polyglutamine repeat expansion, for which there is no effective disease modifying therapy. In recent years, transcriptional dysregulation has emerged as a pathogenic process that appears early in disease progression. Administration of histone deacetylase (HDAC) inhibitors such as suberoylanilide hydroxamic acid (SAHA) have consistently shown therapeutic potential in models of HD, at least partly through increasing the association of acetylated histones with down-regulated genes and by correcting mRNA abnormalities. The HDAC enzyme through which SAHA mediates its beneficial effects in the R6/2 mouse model of HD is not known. Therefore, we have embarked on a series of genetic studies to uncover the HDAC target that is relevant to therapeutic development for HD. HDAC7 is of interest in this context because SAHA has been shown to decrease HDAC7 expression in cell culture systems in addition to inhibiting enzyme activity. After confirming that expression levels of Hdac7 are decreased in the brains of wild type and R6/2 mice after SAHA administration, we performed a genetic cross to determine whether genetic reduction of Hdac7 would alleviate phenotypes in the R6/2 mice. We found no improvement in a number of physiological or behavioral phenotypes. Similarly, the dysregulated expression levels of a number of genes of interest were not improved suggesting that reduction in Hdac7 does not alleviate the R6/2 HD-related transcriptional dysregulation. Therefore, we conclude that the beneficial effects of HDAC inhibitors are not predominantly mediated through the inhibition of HDAC7.


Subject(s)
Histone Deacetylases/genetics , Huntington Disease/genetics , Huntington Disease/physiopathology , Animals , Brain/metabolism , Disease Models, Animal , Down-Regulation , Female , Genotype , Heterozygote , Male , Mice , Mice, Inbred C57BL , Models, Genetic , Phenotype , Transcription, Genetic
11.
Mol Cell Proteomics ; 8(4): 720-34, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19043139

ABSTRACT

Huntington disease (HD) is fatal in humans within 15-20 years of symptomatic disease. Although late stage HD has been studied extensively, protein expression changes that occur at the early stages of disease and during disease progression have not been reported. In this study, we used a large two-dimensional gel/mass spectrometry-based proteomics approach to investigate HD-induced protein expression alterations and their kinetics at very early stages and during the course of disease. The murine HD model R6/2 was investigated at 2, 4, 6, 8, and 12 weeks of age, corresponding to absence of disease and early, intermediate, and late stage HD. Unexpectedly the most HD stage-specific protein changes (71-100%) as well as a drastic alteration (almost 6% of the proteome) in protein expression occurred already as early as 2 weeks of age. Early changes included mainly the up-regulation of proteins involved in glycolysis/gluconeogenesis and the down-regulation of the actin cytoskeleton. This suggests a period of highly variable protein expression that precedes the onset of HD phenotypes. Although an up-regulation of glycolysis/gluconeogenesis-related protein alterations remained dominant during HD progression, late stage alterations at 12 weeks showed an up-regulation of proteins involved in proteasomal function. The early changes in HD coincide with a peak in protein alteration during normal mouse development at 2 weeks of age that may be responsible for these massive changes. Protein and mRNA data sets showed a large overlap on the level of affected pathways but not single proteins/mRNAs. Our observations suggest that HD is characterized by a highly dynamic disease pathology not represented by linear protein concentration alterations over the course of disease.


Subject(s)
Huntington Disease/metabolism , Huntington Disease/pathology , Proteins/metabolism , Animals , Disease Models, Animal , Disease Progression , Embryonic Development , Energy Metabolism , Female , Gene Expression Regulation , Huntington Disease/genetics , Kinetics , Male , Metabolic Networks and Pathways , Mice , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors , Tissue Extracts
12.
J Exp Med ; 205(8): 1869-77, 2008 Aug 04.
Article in English | MEDLINE | ID: mdl-18625748

ABSTRACT

Huntington's disease (HD) is an inherited neurodegenerative disorder characterized by both neurological and systemic abnormalities. We examined the peripheral immune system and found widespread evidence of innate immune activation detectable in plasma throughout the course of HD. Interleukin 6 levels were increased in HD gene carriers with a mean of 16 years before the predicted onset of clinical symptoms. To our knowledge, this is the earliest plasma abnormality identified in HD. Monocytes from HD subjects expressed mutant huntingtin and were pathologically hyperactive in response to stimulation, suggesting that the mutant protein triggers a cell-autonomous immune activation. A similar pattern was seen in macrophages and microglia from HD mouse models, and the cerebrospinal fluid and striatum of HD patients exhibited abnormal immune activation, suggesting that immune dysfunction plays a role in brain pathology. Collectively, our data suggest parallel central nervous system and peripheral pathogenic pathways of immune activation in HD.


Subject(s)
Huntington Disease/immunology , Animals , Case-Control Studies , Central Nervous System/immunology , Central Nervous System/pathology , Cytokines/blood , Cytokines/cerebrospinal fluid , Gene Expression , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/pathology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Transgenic , Microglia/immunology , Microglia/metabolism , Models, Immunological , Monocytes/immunology , Monocytes/metabolism , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Trinucleotide Repeat Expansion
13.
Proc Natl Acad Sci U S A ; 105(9): 3467-72, 2008 Mar 04.
Article in English | MEDLINE | ID: mdl-18299573

ABSTRACT

Huntington's disease (HD) is caused by a CAG repeat expansion that is unstable upon germ-line transmission and exhibits mosaicism in somatic tissues. We show that region-specific CAG repeat mosaicism profiles are conserved between several mouse models of HD and therefore develop in a predetermined manner. Furthermore, we demonstrate that these synchronous, radical changes in CAG repeat size occur in terminally differentiated neurons. In HD this ongoing mutation of the repeat continuously generates genetically distinct neuronal populations in the adult brain of mouse models and HD patients. The neuronal population of the striatum is particularly distinguished by a high rate of CAG repeat allele instability and expression driving the repeat upwards and would be expected to enhance its toxicity. In both mice and humans, neurons are distinguished from nonneuronal cells by expression of MSH3, which provides a permissive environment for genetic instability independent of pathology. The neuronal mutations described here accumulate to generate genetically discrete populations of cells in the absence of selection. This is in contrast to the traditional view in which genetically discrete cellular populations are generated by the sequence of random variation, selection, and clonal proliferation. We are unaware of any previous demonstration that mutations can occur in terminally differentiated neurons and provide a proof of principle that, dependent on a specific set of conditions, functional DNA polymorphisms can be produced in adult neurons.


Subject(s)
Huntington Disease/etiology , Neurons/pathology , Trinucleotide Repeat Expansion , Animals , Brain/pathology , Cell Differentiation , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Mice , Mitosis , Mosaicism
14.
Neurobiol Dis ; 29(1): 41-51, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17920283

ABSTRACT

Huntington's disease (HD) is a hereditary disorder characterized by personality changes, chorea, dementia and weight loss. The cause of this weight loss is unknown. The aim of this study was to examine body weight changes and weight-regulating factors in HD using the R6/2 mouse model as a tool. We found that R6/2 mice started losing weight at 9 weeks of age. Total locomotor activity was unaltered and caloric intake was not decreased until 11 weeks of age, which led us to hypothesize that increased metabolism might underlie the weight loss. Indeed, oxygen consumption in R6/2 mice was elevated from 6 weeks of age, indicative of an increased metabolism. Several organ systems that regulate weight and metabolism, including the hypothalamus, the stomach and adipose tissue displayed abnormalities in R6/2 mice. Together, these data demonstrate that weight loss in R6/2 mice is associated with increased metabolism and changes in several weight-regulating factors.


Subject(s)
Disease Models, Animal , Huntington Disease/metabolism , Age Factors , Analysis of Variance , Animals , Behavior, Animal , Body Temperature , Body Weight/genetics , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Oxygen Consumption , Rotarod Performance Test/methods , Trinucleotide Repeat Expansion/genetics
15.
J Neurochem ; 104(3): 846-58, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17986219

ABSTRACT

A CAG-repeat gene expansion translated into a pathogenic polyglutamine stretch at the N-terminus of huntingtin triggers Huntington's Disease. Mutated huntingtin is predicted to adopt toxic properties mainly if aggregation-prone N-terminal fragments are released by proteolysis. Huntingtin-aggregates are indeed a major hallmark of this disorder and could represent useful markers of disease-onset or progression. We designed a simple method for qualitative and quantitative characterization of aggregates. For this, we analyzed samples from in vitro and in vivo Huntington's Disease models by agarose gel electrophoresis and showed that in the brain of transgenic mice huntingtin-aggregates became larger as a function of disease progression. This appears to be a property of cytoplasmic but not nuclear aggregates. In cell cultures, treatment with Congo Red inhibited aggregate growth but not total load. Finally, we showed that in primary striatal neurons and in brains of R6/2 and HdhQ150 mice, the presence of aggregates preceded initiation of any other functional deficits. This observation argues for a pathogenic role of huntingtin-aggregation in Huntington's Disease. Our results emphasize that thorough analysis of huntingtin metabolism and aggregation is now feasible, thus significantly improving the power of studies assessing therapies designed to lower huntingtin levels or to interfere with its aggregation.


Subject(s)
Huntington Disease/pathology , Nerve Tissue Proteins/metabolism , Neurons/physiology , Nuclear Proteins/metabolism , Age Factors , Animals , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Cells, Cultured , Corpus Striatum/pathology , Disease Models, Animal , Electrophoresis, Gel, Two-Dimensional/methods , Embryo, Mammalian , Female , Huntingtin Protein , Huntington Disease/metabolism , Huntington Disease/physiopathology , Mice , Mice, Transgenic , Mutation/physiology , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Pregnancy , Subcellular Fractions/metabolism , Time Factors , Transfection/methods , Trinucleotide Repeat Expansion/genetics
16.
Nature ; 448(7154): 704-8, 2007 Aug 09.
Article in English | MEDLINE | ID: mdl-17687326

ABSTRACT

Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder caused by expansion of CAG triplet repeats in the huntingtin (HTT) gene (also called HD) and characterized by accumulation of aggregated fragments of polyglutamine-expanded HTT protein in affected neurons. Abnormal enrichment of HD inclusion bodies with ubiquitin, a diagnostic characteristic of HD and many other neurodegenerative disorders including Alzheimer's and Parkinson's diseases, has suggested that dysfunction in ubiquitin metabolism may contribute to the pathogenesis of these diseases. Because modification of proteins with polyubiquitin chains regulates many essential cellular processes including protein degradation, cell cycle, transcription, DNA repair and membrane trafficking, disrupted ubiquitin signalling is likely to have broad consequences for neuronal function and survival. Although ubiquitin-dependent protein degradation is impaired in cell-culture models of HD and of other neurodegenerative diseases, it has not been possible to evaluate the function of the ubiquitin-proteasome system (UPS) in HD patients or in animal models of the disease, and a functional role for UPS impairment in neurodegenerative disease pathogenesis remains controversial. Here we exploit a mass-spectrometry-based method to quantify polyubiquitin chains and demonstrate that the abundance of these chains is a faithful endogenous biomarker of UPS function. Lys 48-linked polyubiquitin chains accumulate early in pathogenesis in brains from the R6/2 transgenic mouse model of HD, from a knock-in model of HD and from human HD patients, establishing that UPS dysfunction is a consistent feature of HD pathology. Lys 63- and Lys 11-linked polyubiquitin chains, which are not typically associated with proteasomal targeting, also accumulate in the R6/2 mouse brain. Thus, HD is linked to global changes in the ubiquitin system to a much greater extent than previously recognized.


Subject(s)
Huntington Disease/metabolism , Ubiquitin/metabolism , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Humans , Huntington Disease/pathology , Inclusion Bodies/metabolism , Lysine/metabolism , Mice , Mice, Transgenic , Polyubiquitin/metabolism , Proteasome Endopeptidase Complex/metabolism
17.
Brain Res Bull ; 72(2-3): 83-97, 2007 Apr 30.
Article in English | MEDLINE | ID: mdl-17352931

ABSTRACT

The identification of the Huntington's disease (HD) mutation as a CAG/polyglutamine repeat expansion enabled the generation of transgenic rodent models and gene-targeted mouse models of HD. Of these, mice that are transgenic for an N-terminal huntingtin fragment have been used most extensively because they develop phenotypes with relatively early ages of onset and rapid disease progression. Although the fragment models have led to novel insights into the pathophysiology of HD, it is important that models expressing a mutant version of the full-length protein are analysed in parallel. We have generated congenic C57BL/6 and CBA strains for the HdhQ150 knock-in mouse model of HD so that homozygotes can be analysed on an F1 hybrid background. Although a significant impairment in grip strength could be detected from a very early age, the performance of these mice in the quantitative behavioural tests most frequently used in preclinical efficacy trials indicates that they are unlikely to be useful for preclinical screening using a battery of conventional tests. However, at 22 months of age, the Hdh(Q150/Q150) homozygotes showed unexpected widespread aggregate deposition throughout the brain, transcriptional dysregulation in the striatum and cerebellum and decreased levels of specific chaperones, all well-characterised molecular phenotypes present in R6/2 mice aged 12 weeks. Therefore, when strain background and CAG repeat length are controlled for, the knock-in and fragment models develop comparable phenotypes. This supports the continued use of the more high-throughput fragment models to identify mechanisms of pathogenesis and for preclinical screening.


Subject(s)
Brain/pathology , Disease Models, Animal , Huntington Disease/genetics , Huntington Disease/physiopathology , Mice , Animals , Blotting, Western , Exons , Female , Huntingtin Protein , Immunohistochemistry , Male , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Trinucleotide Repeat Expansion
18.
Hum Mol Genet ; 16(9): 1078-90, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17360721

ABSTRACT

Huntington's disease (HD) is caused by an expanded polyglutamine tract in the huntingtin protein. Mitochondrial dysfunction and free radical damage occur in both R6/2 mice and HD patient brains and might play a role in disease pathogenesis. In cell culture systems, heat-shock protein 27 (Hsp27), a small molecular chaperone, suppresses mutant huntingtin-induced reactive oxygen species formation and cell death. To investigate this in vivo, we conducted an extensive phenotypic characterization of mice arising from a cross between R6/2 mice and Hsp27 transgenic mice but did not observe an improvement of the R6/2 phenotype. Hsp27 overexpression had no effect in reducing oxidative stress in the R6/2 brain, assessed by measuring striatal aconitase activity and protein carbonylation levels. Native protein gel analysis revealed that transgenic Hsp27 forms active, large oligomeric species in heat-shocked brain lysates, demonstrating that it is efficiently activated upon stress. In contrast, Hsp27 in double transgenic brains exists predominantly as a low molecular weight, inactive species. This suggests that Hsp27, which is otherwise activatable upon heat shock, remains inactive in the R6/2 model of chronic neurodegeneration. Hsp27 transgenics had been previously shown to be protected from acute stresses such as kainate administration, ischemia/reperfusion heart injury and neonatal nerve injury. Our study is the first to suggest a differential modulation of Hsp27 activation in vivo and, importantly, it illustrates the diverse effect of Hsp27 on acute versus chronic models of disease.


Subject(s)
Heat-Shock Proteins/genetics , Huntington Disease/genetics , Nerve Degeneration/genetics , Aconitate Hydratase/metabolism , Animals , Behavior, Animal , Blotting, Western , Brain/metabolism , Brain/pathology , Disease Models, Animal , Female , Gene Expression , Genotype , Heat-Shock Proteins/metabolism , Heat-Shock Response/genetics , Heat-Shock Response/physiology , Huntington Disease/metabolism , Huntington Disease/physiopathology , Immunohistochemistry , Immunoprecipitation , Inclusion Bodies/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Mice, Transgenic , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Oxidative Stress , Phenotype , Transglutaminases/genetics , Transglutaminases/metabolism
19.
Neurobiol Dis ; 23(1): 190-7, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16697652

ABSTRACT

The brain levels of the endogenous excitotoxin quinolinic acid (QUIN) and its bioprecursor, the free radical generator 3-hydroxykynurenine (3-HK), are elevated in early stage Huntington disease (HD). We now examined the status of these metabolites in three mouse models of HD. In R6/2 mice, 3-HK levels were significantly and selectively elevated in the striatum, cortex and cerebellum starting at 4 weeks of age. In contrast, both 3-HK and QUIN levels were increased in the striatum and cortex of the full-length HD models, beginning at 8 months (YAC128) and 15 months (Hdh(Q92) and Hdh(Q111)), respectively. No changes were seen in 13-month-old shortstop mice, which show no signs of motor or cognitive dysfunction or selective neuropathology. These results demonstrate both important parallels and intriguing differences in the progressive neurochemical changes in these HD mouse models and support the hypothesis that QUIN may play a role in the striatal and cortical neurodegeneration of HD.


Subject(s)
Huntington Disease/metabolism , Kynurenine/analogs & derivatives , Kynurenine/metabolism , Quinolinic Acid/analysis , Age Factors , Animals , Brain Chemistry , Chromatography, Gas , Chromatography, High Pressure Liquid , Disease Models, Animal , Humans , Kynurenic Acid/metabolism , Kynurenine/analysis , Mice , Mice, Transgenic , Quinolinic Acid/metabolism
20.
Neurobiol Dis ; 23(1): 44-53, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16651003

ABSTRACT

Huntington Disease (HD) is characterized by choreic involuntary movements and striatal vulnerability. A2A receptors expressed on GABAergic striatal neurons have been suggested to play a pathogenetic role. Previous data demonstrated the presence of an aberrant alteration of A2A receptor-dependent adenylyl cyclase in an in vitro model of the disease (striatal cells expressing mutant huntingtin) and in peripheral circulating cells of HD patients. Here, we investigated whether this dysfunction is present in the R6/2 HD transgenic mouse model, by analyzing striatal A2A receptor-binding and adenylyl cyclase activity at different developmental stages in comparison with age-matched wild type animals. A transient increase in A2A receptor density (Bmax) and A2A receptor-dependent cAMP production at early presymptomatic ages (7-14 postnatal days) was found. Both alterations normalized to control values starting from postnatal day 21. In contrast, A2A receptor mRNA, as detected by real time PCR, dramatically decreased starting from PND21 until late symptomatic stages (12 weeks of age). The discrepancy between A2A receptor expression and density suggests compensatory mechanisms. These data, reproducing ex vivo the previous observations in vitro, support the hypothesis that an alteration of A2A receptor signaling is present in HD and might represent an interesting target for neuroprotective therapies.


Subject(s)
Basal Ganglia/metabolism , Huntington Disease/metabolism , Receptors, Adenosine A2/metabolism , Signal Transduction/physiology , Adenylyl Cyclases/metabolism , Animals , Cyclic AMP/metabolism , Disease Models, Animal , Mice , Mice, Transgenic , RNA, Messenger/analysis , Receptors, Adenosine A2/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Triazines/pharmacology , Triazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...