Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Curr Alzheimer Res ; 17(9): 858-867, 2020.
Article in English | MEDLINE | ID: mdl-33272184

ABSTRACT

BACKGROUND: We have recently identified Huntingtin (Htt), the pathogenic protein in Huntington's disease, as a mediator of Alzheimer's disease (AD) pathology in an amyloid precursor protein (APP) knock-in mouse model of AD. That finding prompted us to examine if Htt is accumulated in the brains of AD patients and in which cell type Htt is present in the AD brain. OBJECTIVE: To investigate whether location and levels of Htt are affected in hippocampus and frontal cortex in AD. METHODS: Brains from AD patients (n=11) and controls (n=11) were stained for Htt using immunohistochemistry and signal intensity of Htt was quantified and localized in subregions and neurons. Confocal microscopy was used to characterize neuronal Htt localisation and its relationship with tau tangles and astrocytes. RESULTS: Htt levels were increased in neuronal cells in the granular layer of the dentate gyrus, in CA1 and CA3 in hippocampus and in layer III of the frontal cortex. Htt was found in the soma, perinuclear space, thin neurites and nucleus of pyramidal neurons. Htt was present in neurons containing tau tangles but did not colocalize with astrocytes. CONCLUSION: Htt accumulates in pyramidal neuron-rich areas including hippocampal subregions associated with memory and frontal cortex layer III. The accumulation of Htt in AD shows distinct cellular and morphological patterns and is not present in astrocytes. Clearly, further research is warranted to elucidate the role of Htt as a mediator of AD pathology and the potential use of Htt as a target in future therapeutic strategies.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Hippocampus/metabolism , Hippocampus/pathology , Huntingtin Protein/metabolism , Aged , Aged, 80 and over , Autopsy/methods , Female , Frontal Lobe/metabolism , Frontal Lobe/pathology , Humans , Huntingtin Protein/analysis , Male
2.
J Med Chem ; 63(15): 8608-8633, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32662649

ABSTRACT

Mutant huntingtin (mHTT) protein carrying the elongated N-terminal polyglutamine (polyQ) tract misfolds and forms protein aggregates characteristic of Huntington's disease (HD) pathology. A high-affinity ligand specific for mHTT aggregates could serve as a positron emission tomography (PET) imaging biomarker for HD therapeutic development and disease progression. To identify such compounds with binding affinity for polyQ aggregates, we embarked on systematic structural activity studies; lead optimization of aggregate-binding affinity, unbound fractions in brain, permeability, and low efflux culminated in the discovery of compound 1, which exhibited target engagement in autoradiography (ARG) studies in brain slices from HD mouse models and postmortem human HD samples. PET imaging studies with 11C-labeled 1 in both HD mice and WT nonhuman primates (NHPs) demonstrated that the right-hand-side labeled ligand [11C]-1R (CHDI-180R) is a suitable PET tracer for imaging of mHTT aggregates. [11C]-1R is now being advanced to human trials as a first-in-class HD PET radiotracer.


Subject(s)
Huntingtin Protein/analysis , Huntington Disease/diagnostic imaging , Positron-Emission Tomography/methods , Protein Aggregation, Pathological/diagnostic imaging , Animals , Disease Models, Animal , Dogs , Female , Humans , Huntingtin Protein/genetics , Huntington Disease/genetics , Ligands , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Inbred C57BL , Mutation , Peptides/genetics , Protein Aggregation, Pathological/genetics , Radiopharmaceuticals/analysis , Rats, Sprague-Dawley
3.
Nutr Neurosci ; 23(3): 183-189, 2020 Mar.
Article in English | MEDLINE | ID: mdl-29973113

ABSTRACT

Objectives: Green tea infusion contains a complex mixture of polyphenolic compounds that were shown to provide health benefits. It was previously demonstrated that (-)-epigallocatechin-3-gallate, one of the major polyphenols present in green tea, has a suppressing effect on various aspects of pathogenesis in models of Huntington's disease (HD), an inherited neurodegenerative disorder. In this study, we aimed to investigate, whether green tea infusion prepared as for human consumption has similar positive effects.Methods: We used a transgenic Drosophila model of HD to study the effects of green tea on mutant Huntingtin induced phenotypes. We tested the effects of green tea infusion on mutant Huntingtin induced neurodegeneration, impaired motor performance, reduced viability and lifespan by pseudopupil assay, climbing assay, eclosion and survival tests, respectively. We used immunoblots to measure Huntingtin protein levels and tested generic health benefits of green tea by longevity analysis.Results: We found that green tea supplementation reduced mutant Huntingtin induced neurodegeneration in Drosophila and positively impacted the longevity of mutant Huntingtin expressing flies. However, green tea did not rescue reduced viability of Drosophila expressing mutant Huntingtin or increased longevity of wild-type fruit flies.Discussion: Our results indicate that green tea consumption might have a modest positive effect on symptoms of HD.


Subject(s)
Animals, Genetically Modified , Drosophila/genetics , Huntingtin Protein/genetics , Nerve Degeneration/drug therapy , Plant Extracts/administration & dosage , Tea , Animals , Female , Gene Expression , Huntingtin Protein/analysis , Huntington Disease/drug therapy , Huntington Disease/physiopathology , Longevity/drug effects , Male
4.
Biochem Biophys Res Commun ; 521(3): 549-554, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31677786

ABSTRACT

Huntington's disease (HD) is a progressive neurodegenerative disorder caused by an expansion of a CAG triplet repeat (encoding for a polyglutamine tract) within the first exon of the huntingtin gene. Expression of the mutant huntingtin (mHTT) protein can result in the production of N-terminal fragments with a robust propensity to form oligomers and aggregates, which may be causally associated with HD pathology. Several lines of evidence indicate that N17 phosphorylation or pseudophosphorylation at any of the residues T3, S13 or S16, alone or in combination, modulates mHTT aggregation, subcellular localization and toxicity. Consequently, increasing N17 phosphorylation has been proposed as a potential therapeutic approach. However, developing genetic/pharmacological tools to quantify these phosphorylation events is necessary in order to subsequently develop tool modulators, which is difficult given the transient and incompletely penetrant nature of such post-translational modifications. Here we describe the first ultrasensitive sandwich immunoassay that quantifies HTT phosphorylated at residue S13 and demonstrate its utility for specific analyte detection in preclinical models of HD.


Subject(s)
Huntingtin Protein/analysis , Animals , Cells, Cultured , Gene Knock-In Techniques , HEK293 Cells , Humans , Huntingtin Protein/genetics , Huntington Disease/genetics , Mice , Mutation , Neurons/chemistry , Neurons/metabolism , Phosphorylation , Protein Aggregates , Protein Processing, Post-Translational
5.
ACS Chem Neurosci ; 10(1): 472-481, 2019 01 16.
Article in English | MEDLINE | ID: mdl-30149694

ABSTRACT

Huntington's disease is a neurodegenerative disorder resulting from an expanded polyglutamine (polyQ) repeat of the Huntingtin (Htt) protein. Affected tissues often contain aggregates of the N-terminal Htt exon 1 (Htt-Ex1) fragment. The N-terminal N17 domain proximal to the polyQ tract is key to enhance aggregation and modulate Htt toxicity. Htt-Ex1 is intrinsically disordered, yet it has been postulated that under physiological conditions membranes induce the N17 to adopt an α-helical structure, which then plays a key role in regulating Htt protein aggregation. The present study leverages the recently available assignment of NMR peaks in an N17Q17 construct, in order to provide a look into the changes occurring in vitro upon exposing this fragment to various brain extract fragments as well as to synthetic bilayers. Residue-specific changes were observed by 3D HNCO NMR, whose nature was further clarified with ancillary CD and aggregation studies, as well as with molecular dynamic calculations. From this combination of measurements and computations, a unified picture emerges, whereby transient structures consisting of α-helices spanning a fraction of the N17 residues form during N17Q17-membrane interactions. These interactions are fairly dynamic, but they qualitatively mimic more rigid variants that have been discussed in the literature. The nature of these interactions and their potential influence on the aggregation process of these kinds of constructs under physiological conditions are briefly assessed.


Subject(s)
Cell Membrane/chemistry , Cell Membrane/genetics , Huntingtin Protein/chemistry , Huntingtin Protein/genetics , Magnetic Resonance Spectroscopy/methods , Protein Aggregates/genetics , Animals , Humans , Huntingtin Protein/analysis , Huntington Disease/genetics , Mice
6.
J Vis Exp ; (141)2018 11 28.
Article in English | MEDLINE | ID: mdl-30582582

ABSTRACT

For the investigation of protein localization and trafficking using live cell imaging, researchers often rely on fusing their protein of interest to a fluorescent reporter. The constantly evolving list of genetically encoded fluorescent proteins (FPs) presents users with several alternatives when it comes to fluorescent fusion design. Each FP has specific optical and biophysical properties that can affect the biochemical, cellular, and functional properties of the resulting fluorescent fusions. For instance, several FPs tend to form nonspecific oligomers that are susceptible to impede on the function of the fusion partner. Unfortunately, only a few methods exist to test the impact of FPs on the behavior of the fluorescent reporter. Here, we describe a simple method that enables the rapid assessment of the impact of FPs using polyglutamine (polyQ) toxicity assays in the budding yeast Saccharomyces cerevisiae. PolyQ-expanded huntingtin proteins are associated with the onset of Huntington's disease (HD), where the expanded huntingtin aggregates into toxic oligomers and inclusion bodies. The aggregation and toxicity of polyQ expansions in yeast are highly dependent on the sequences flanking the polyQ region, including the presence of fluorescent tags, thus providing an ideal experimental platform to study the impact of FPs on the behavior of their fusion partner.


Subject(s)
Fluorescent Dyes/metabolism , Huntingtin Protein/metabolism , Peptides/toxicity , Saccharomyces cerevisiae Proteins/metabolism , Fluorescent Dyes/analysis , Humans , Huntingtin Protein/analysis , Huntingtin Protein/genetics , Huntington Disease/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/analysis
7.
eNeuro ; 5(4)2018.
Article in English | MEDLINE | ID: mdl-30310861

ABSTRACT

The neurodegenerative Huntington's disease (HD) is caused by a polyglutamine (polyQ) amplification in the huntingtin protein (HTT). Currently there is no effective therapy available for HD; however, several efforts are directed to develop and optimize HTT-lowering methods to improve HD phenotypes. To validate these approaches, there is an immediate need for reliable, sensitive, and easily accessible methods to quantify HTT expression. Using the AlphaLISA platform, we developed two novel sensitive and robust assays for quantification of HTT in biological samples using commercially available antibodies. The first, a polyQ-independent assay, measures the total pool of HTT, while the second, a polyQ-dependent assay, preferentially detects the mutant form of HTT. Using purified HTT protein standards and brain homogenates from an HD mouse model, we determine a lower limit of quantification of 1 and 3 pm and optimal reproducibility with CV values lower than 7% for intra- and 20% for interassay. In addition, we used the assays to quantify HTT in neural stem cells generated from patient-derived induced pluripotent stem cells in vitro and in human brain tissue lysates. Finally, we could detect changes in HTT levels in a mouse model where mutant HTT was conditionally deleted in neural tissue, verifying the potential to monitor the outcome of HTT-lowering strategies. This analytical platform is ideal for high-throughput screens and thus has an added value for the HD community as a tool to optimize novel therapeutic approaches aimed at modulating HTT protein levels.


Subject(s)
Huntingtin Protein/analysis , Huntington Disease/diagnosis , Immunoassay/standards , Animals , Disease Models, Animal , HEK293 Cells , Humans , Immunoassay/methods , Mice , Mutation , Neural Stem Cells , Reproducibility of Results
8.
Methods Mol Biol ; 1780: 31-40, 2018.
Article in English | MEDLINE | ID: mdl-29856013

ABSTRACT

N-terminal mutant huntingtin (mHTT) fragments with pathogenic polyglutamine (polyQ) tracts spontaneously form stable, amyloidogenic protein aggregates with a fibrillar morphology. Such structures are detectable in brains of Huntington's disease (HD) patients and various model organisms, suggesting that they play a critical role in pathogenesis. Heat-stable, fibrillar mHTT aggregates can be detected and quantified in cells and tissues using a denaturing filter retardation assay (FRA). Here, we describe step-by-step protocols and experimental procedures for the investigation of mHTT aggregates in complex biosamples using FRAs. The methods are illustrated with examples from studies in cellular, transgenic fly, and mouse models of HD, but can be adapted for any disease-relevant protein with amyloidogenic polyQ tracts.


Subject(s)
Brain/pathology , Huntingtin Protein/analysis , Huntington Disease/diagnosis , Immunoblotting/methods , Protein Aggregation, Pathological/diagnosis , Animals , Animals, Genetically Modified , Disease Models, Animal , Drosophila melanogaster , Hot Temperature , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/pathology , Immunoblotting/instrumentation , Mice , Mutation , Protein Aggregates , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology
9.
Methods Mol Biol ; 1780: 329-396, 2018.
Article in English | MEDLINE | ID: mdl-29856027

ABSTRACT

Huntington's disease (HD) is a chronic progressive neurodegenerative condition where new markers of disease progression are needed. So far no disease-modifying interventions have been found, and few interventions have been proven to alleviate symptoms. This may be partially explained by the lack of reliable indicators of disease severity, progression, and phenotype.Biofluid biomarkers may bring advantages in addition to clinical measures, such as reliability, reproducibility, price, accuracy, and direct quantification of pathobiological processes at the molecular level; and in addition to empowering clinical trials, they have the potential to generate useful hypotheses for new drug development.In this chapter we review biofluid biomarker reports in HD, emphasizing those we feel are likely to be closest to clinical applicability.


Subject(s)
Body Fluids/chemistry , Huntingtin Protein/analysis , Huntington Disease/diagnosis , Biomarkers/analysis , Brain/cytology , Brain/metabolism , Brain/pathology , Drug Development , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/pathology , Inflammation Mediators/analysis , Inflammation Mediators/metabolism , Neurons/pathology , Neurotransmitter Agents/analysis , Neurotransmitter Agents/metabolism , Oxidative Stress , Proteomics/instrumentation , Proteomics/methods , Transglutaminases/analysis , Transglutaminases/metabolism
10.
Nat Commun ; 9(1): 1881, 2018 05 14.
Article in English | MEDLINE | ID: mdl-29760420

ABSTRACT

Understanding and reshaping cellular behaviors with synthetic gene networks requires the ability to sense and respond to changes in the intracellular environment. Intracellular proteins are involved in almost all cellular processes, and thus can provide important information about changes in cellular conditions such as infections, mutations, or disease states. Here we report the design of a modular platform for intrabody-based protein sensing-actuation devices with transcriptional output triggered by detection of intracellular proteins in mammalian cells. We demonstrate reporter activation response (fluorescence, apoptotic gene) to proteins involved in hepatitis C virus (HCV) infection, human immunodeficiency virus (HIV) infection, and Huntington's disease, and show sensor-based interference with HIV-1 downregulation of HLA-I in infected T cells. Our method provides a means to link varying cellular conditions with robust control of cellular behavior for scientific and therapeutic applications.


Subject(s)
Apoptosis Regulatory Proteins/analysis , Biosensing Techniques , Gene Regulatory Networks , Genetic Engineering/methods , Hepacivirus/genetics , Huntingtin Protein/analysis , Antibodies/chemistry , Antibodies/metabolism , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Endopeptidases/chemistry , Genes, Reporter , HEK293 Cells , HIV-1/genetics , HIV-1/metabolism , HLA Antigens/genetics , HLA Antigens/immunology , Hepacivirus/metabolism , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Jurkat Cells , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Optical Imaging/methods , Plasmids/chemistry , Plasmids/metabolism
12.
Free Radic Biol Med ; 120: 311-316, 2018 05 20.
Article in English | MEDLINE | ID: mdl-29609019

ABSTRACT

Targeted expression of a therapeutic agent is a major bottleneck in designing a drug delivery system. Protein aggregation and elevated oxidative stress are associated with the onset of many neurodegenerative disorders, including Huntington's disease (HD). An oxidative stress-inducible promoter, i.e. Thioredoxin 2, was employed to design a sensor for protein aggregation. RNA aptamers specific for mutant huntingtin were expressed only in cells where aggregation of mutant huntingtin occurred. A nine-fold increase in RNA expression was seen when aptamer sequences were cloned under the Trx2 promoter. Expression of aptamer resulted in reduced protein aggregation and decreased oxidative stress, which, in turn, reduced the expression of aptamers by two-fold. Reduction in aggregation led to increased cell survival. The aptamers were not expressed in cells expressing wild-type huntingtin in the soluble form. This rational and simple design will allow the use of this construct for the targeted expression of other therapeutic nucleic acid molecules as well.


Subject(s)
Aptamers, Nucleotide , Huntingtin Protein/analysis , Oxidative Stress , Protein Aggregation, Pathological/prevention & control , Aptamers, Nucleotide/genetics , Humans , Huntingtin Protein/genetics , Mutant Proteins/genetics , Promoter Regions, Genetic , Saccharomyces cerevisiae Proteins/genetics , Thioredoxins/genetics
13.
Biochim Biophys Acta Mol Basis Dis ; 1863(3): 793-800, 2017 03.
Article in English | MEDLINE | ID: mdl-28040507

ABSTRACT

Most neurodegenerative diseases such as Alzheimer's, Parkinson's and Huntington's disease are hallmarked by aggregate formation of disease-related proteins. In various of these diseases transfer of aggregation-prone proteins between neurons and between neurons and glial cells has been shown, thereby initiating aggregation in neighboring cells and so propagating the disease phenotype. Whereas this prion-like transfer is well studied in Alzheimer's and Parkinson's disease, only a few studies have addressed this potential mechanism in Huntington's disease. Here, we present an overview of in vitro and in vivo methodologies to study release, intercellular transfer and uptake of aggregation-prone protein fragments in Huntington's disease models.


Subject(s)
Huntingtin Protein/metabolism , Huntington Disease/metabolism , Neuroglia/metabolism , Neurons/metabolism , Prions/metabolism , Protein Aggregation, Pathological/metabolism , Animals , Humans , Huntingtin Protein/analysis , Huntingtin Protein/genetics , Huntington Disease/genetics , Huntington Disease/pathology , Mutation , Neuroglia/pathology , Neurons/pathology , Prions/analysis , Prions/genetics , Protein Aggregates , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Protein Transport
14.
Elife ; 52016 08 03.
Article in English | MEDLINE | ID: mdl-27484239

ABSTRACT

The presumptive altered dynamics of transient molecular interactions in vivo contributing to neurodegenerative diseases have remained elusive. Here, using single-molecule localization microscopy, we show that disease-inducing Huntingtin (mHtt) protein fragments display three distinct dynamic states in living cells - 1) fast diffusion, 2) dynamic clustering and 3) stable aggregation. Large, stable aggregates of mHtt exclude chromatin and form 'sticky' decoy traps that impede target search processes of key regulators involved in neurological disorders. Functional domain mapping based on super-resolution imaging reveals an unexpected role of aromatic amino acids in promoting protein-mHtt aggregate interactions. Genome-wide expression analysis and numerical simulation experiments suggest mHtt aggregates reduce transcription factor target site sampling frequency and impair critical gene expression programs in striatal neurons. Together, our results provide insights into how mHtt dynamically forms aggregates and disrupts the finely-balanced gene control mechanisms in neuronal cells.


Subject(s)
Huntingtin Protein/analysis , Protein Aggregation, Pathological , Single Molecule Imaging , Transcription, Genetic , Animals , Cells, Cultured , Mice , Mouse Embryonic Stem Cells , Mutant Proteins/analysis , Neurons/pathology , Transcription Factors/antagonists & inhibitors , Visual Cortex/pathology
15.
Prion ; 10(3): 221-7, 2016 05 03.
Article in English | MEDLINE | ID: mdl-27220690

ABSTRACT

Proteins with expanded polyglutamine (polyQ) regions are prone to form amyloids, which can cause diseases in humans and toxicity in yeast. Recently, we showed that in yeast non-toxic amyloids of Q-rich proteins can induce aggregation and toxicity of wild type huntingtin (Htt) with a short non-pathogenic polyglutamine tract. Similarly to mutant Htt with an elongated N-terminal polyQ sequence, toxicity of its wild type counterpart was mediated by induced aggregation of the essential Sup35 protein, which contains a Q-rich region. Notably, polymerization of Sup35 was not caused by the initial benign amyloids and, therefore, aggregates of wild type Htt acted as intermediaries in seeding Sup35 polymerization. This exemplifies a protein polymerization cascade which can generate a network of interdependent polymers. Here we discuss cross-seeded protein polymerization as a possible mechanism underlying known interrelations between different polyQ diseases. We hypothesize that similar mechanisms may enable proteins, which possess expanded Q-rich tracts but are not associated with diseases, to promote the development of polyQ diseases.


Subject(s)
Amyloidosis/metabolism , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Peptides/metabolism , Protein Aggregation, Pathological/metabolism , Saccharomyces cerevisiae/metabolism , Amyloidosis/genetics , Humans , Huntingtin Protein/analysis , Huntingtin Protein/genetics , Huntington Disease/genetics , Mutation , Peptide Termination Factors/analysis , Peptide Termination Factors/metabolism , Peptides/analysis , Peptides/genetics , Protein Aggregation, Pathological/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/analysis , Saccharomyces cerevisiae Proteins/metabolism
16.
J Huntingtons Dis ; 5(1): 33-8, 2016.
Article in English | MEDLINE | ID: mdl-27003665

ABSTRACT

BACKGROUND: Silencing mutant huntingtin mRNA by RNA interference (RNAi) is a therapeutic strategy for Huntington's disease. RNAi induces specific endonucleolytic cleavage of the target HTT mRNA, followed by exonucleolytic processing of the cleaved mRNA fragments. OBJECTIVES: We investigated the clearance of huntingtin mRNA cleavage products following RNAi, to find if particular huntingtin mRNA sequences persist. We especially wanted to find out if the expanded CAG increased production of a toxic mRNA species by impeding degradation of human mutant huntingtin exon 1 mRNA. METHODS: Mice expressing the human mutant HTT transgene with 128 CAG repeats (YAC128 mice) were injected in the striatum with self-complementary AAV9 vectors carrying a miRNA targeting exon 48 of huntingtin mRNA (scAAV-U6-miRNA-HTT-GFP). Transgenic huntingtin mRNA levels were measured in striatal lysates after two weeks. For qPCR, we used species specific primer-probe combinations that together spanned 6 positions along the open reading frame and untranslated regions of the human huntingtin mRNA. Knockdown was also measured in the liver following tail vein injection. RESULTS: Two weeks after intrastriatal administration of scAAV9-U6-miRNA-HTT-GFP, we measured transgenic mutant huntingtin in striatum using probes targeting six different sites along the huntingtin mRNA. Real time PCR showed a reduction of 29% to 36% in human HTT. There was no significant difference in knockdown measured at any of the six sites, including exon 1. In liver, we observed a more pronounced HTT mRNA knockdown of 70% to 76% relative to the untreated mice, and there were also no significant differences among sites. CONCLUSIONS: Our results demonstrate that degradation is equally distributed across the human mutant huntingtin mRNA following RNAi-induced cleavage.


Subject(s)
Huntingtin Protein/genetics , Huntington Disease/genetics , Mutation/genetics , RNA Interference , RNA, Messenger/genetics , Trinucleotide Repeats/genetics , Animals , Corpus Striatum/metabolism , Disease Models, Animal , Exons/genetics , Gene Knockdown Techniques , Huntingtin Protein/analysis , Huntingtin Protein/metabolism , Liver/metabolism , Mice , RNA, Messenger/analysis , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...