Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Neurobiol Dis ; 134: 104711, 2020 02.
Article in English | MEDLINE | ID: mdl-31841677

ABSTRACT

Cerebral palsy (CP) is the most common cause of childhood motor disability, manifesting most often as spasticity and/or dystonia. Spasticity and dystonia are often co-morbid clinically following severe injury at term gestation. Currently available animal CP models have not demonstrated or differentiated between these two motor phenotypes, limiting their clinical relevance. We sought to develop an animal CP model displaying objectively identifiable spasticity and dystonia. We exposed rat pups at post-natal day 7-8 (equivalent to human 37 post-conceptional weeks) to global hypoxia. Since spasticity and dystonia can be difficult to differentiate from each other in CP, objective electrophysiologic markers of motor phenotypes were assessed. Spasticity was inferred using an electrophysiologic measure of hyperreflexia: soleus Hoffman reflex suppression with 2 Hz tibial nerve stimulation. Dystonia was assessed during voluntary isometric hindlimb withdrawal at different levels of arousal by calculating tibialis anterior and triceps surae electromyographic co-activation as a surrogate of overflow muscle activity. Hypoxia affected spasticity and dystonia measures in a sex-dependent manner. Males had attenuated Hoffman reflex suppression suggestive of spasticity but no change in antagonist muscle co-activation. In contrast, females demonstrated increased co-activation suggestive of dystonia but no change in Hoffman reflex suppression. Therefore, there was an unexpected segregation of electrophysiologically-defined motor phenotypes based on sex with males predominantly demonstrating spasticity and females predominantly demonstrating dystonia. These results require human clinical confirmation but suggest that sex could play a critical role in the motor manifestations of neonatal brain injury.


Subject(s)
Cerebral Palsy/physiopathology , Disease Models, Animal , Dystonia/physiopathology , Muscle Spasticity/physiopathology , Animals , Cerebral Palsy/complications , Dystonia/complications , Electromyography , Female , Male , Muscle Spasticity/complications , Muscle, Skeletal/physiopathology , Phenotype , Rats, Sprague-Dawley
2.
J Huntingtons Dis ; 8(4): 435-441, 2019.
Article in English | MEDLINE | ID: mdl-31381523

ABSTRACT

BACKGROUND: There is limited understanding of the feasibility of conducting long-term research among undiagnosed (pre-symptomatic) adults at risk to develop Huntington disease (HD), while protecting their emotional well-being and safety. OBJECTIVE: To assess pre-specified events pertaining to emotional well-being, safety, and feasibility among healthy consenting adults at risk for developing HD who have chosen not to undergo genetic testing. METHODS: PHAROS research participants prospectively reported the occurrence of events pertaining to psychological distress (psychiatric evaluations, depression, suicidality) and feasibility (maintaining confidentiality, study attrition). PHAROS enrolled 1001 participants. RESULTS: Events pertaining to psychological distress were reported by 35% of participants. The most common events included heightened suicide risk (26%), new onset depression (12%), and new mental health evaluation (9%); all occurred significantly more frequently among participants with expanded trinucleotide CAG repeats (≥37). Five deaths occurred, none related to suicide. Forty-one percent of participants reported self-disclosure of their HD at-risk status, and 15% reported that someone else (usually a family member) had done so. Confidentiality of CAG test results was maintained by investigators. The withdrawal rate was largely uniform over the study period and did not differ significantly by gender or CAG status. CONCLUSIONS: The potentially vulnerable research participants in PHAROS showed good emotional tolerability and safety. Individual CAG data were not disclosed, and confidentiality about disclosure of at-risk HD status was well maintained by others (family, friends, etc.). Long-term research participation of adults at risk for HD who choose not to undergo pre-symptomatic DNA testing is well tolerated, safe and feasible.


Subject(s)
Depression/psychology , Huntington Disease , Mental Disorders/psychology , Personal Satisfaction , Stress, Psychological/psychology , Suicide/psychology , Adult , Confidentiality , Feasibility Studies , Female , Genetic Testing , Humans , Huntington Disease/diagnosis , Huntington Disease/genetics , Huntington Disease/psychology , Male , Middle Aged , Observational Studies as Topic , Patient Selection , Prospective Studies , Risk , Self Disclosure
3.
Ann Clin Transl Neurol ; 6(6): 1046-1052, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31211168

ABSTRACT

OBJECTIVE: To examine phenotype-genotype discrepancies (PGDs) wherein genotype-concealed and prospective judgments of the motor onset of Huntington disease (HD) occurred among at-risk adults who had nonexpanded (<37) cytosine-adenine-guanine (CAG) trinucleotide DNA repeats. METHODS: We examined the prospective clinical assessments of investigators who were kept unaware of individual CAG lengths in the Prospective Huntington At-Risk Observational Study (PHAROS) who enrolled and followed undiagnosed adults at risk for HD who chose not to learn their gene status. Subjects (n = 1001) at 43 Huntington Study Group research sites in the US and Canada were evaluated prospectively and systematically between 1999 and 2009. At each site, an investigator was designated to perform comprehensive clinic assessments and another investigator to rate only the motor examination. Phenoconversion from a "premanifest" status to a confidently "manifest" status was based on investigator judgment (diagnostic confidence level) of the extrapyramidal motor features of HD. RESULTS: There were 20 PGDs that over time had less severe motor scores than the 101 phenoconversions with CAG ≥37, but more severe motor scores than nonconversions. Following conversion, subjects with CAG ≥37 expansions worsened more motorically and cognitively than PGD subjects in the < 37 group. PGDs were concentrated among three sites and a few investigators, especially raters who only assessed the motor examination. INTERPRETATION: The ability to detect the clinical onset of HD in a timely and reliable fashion remains the key for developing experimental treatments aimed at postponing the clinical onset of HD. Comprehensive clinical evaluation is a more accurate and reliable basis for determining HD clinical onset than sole reliance on judging the extrapyramidal features of HD.


Subject(s)
Huntington Disease/diagnosis , Huntington Disease/genetics , Adolescent , Adult , Canada , Female , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Movement Disorders , Phenotype , Prospective Studies , Risk Factors , Trinucleotide Repeats/genetics , United States , Young Adult
4.
Adv Neurobiol ; 15: 129-161, 2017.
Article in English | MEDLINE | ID: mdl-28674980

ABSTRACT

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder characterised by extensive neuronal loss in the striatum and cerebral cortex, and a triad of clinical symptoms affecting motor, cognitive/behavioural and mood functioning. The mutation causing HD is an expansion of a CAG tract in exon 1 of the HTT gene. This chapter provides a multifaceted overview of the clinical complexity of HD. We explore recent directions in molecular genetics including the identification of loci that are genetic modifiers of HD that could potentially reveal therapeutic targets beyond the HTT gene transcript and protein. The variability of clinical symptomatology in HD is considered alongside recent findings of variability in cellular and neurochemical changes in the striatum and cerebral cortex in human brain. We review evidence from structural neuroimaging methods of progressive changes of striatum, cerebral cortex and white matter in pre-symptomatic and symptomatic HD, with a particular focus on the potential identification of neuroimaging biomarkers that could be used to test promising disease-specific and modifying treatments. Finally we provide an overview of completed clinical trials in HD and future therapeutic developments.


Subject(s)
Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Genes, Modifier/genetics , Huntingtin Protein/genetics , Huntington Disease/genetics , Affect , Cerebral Cortex/diagnostic imaging , Cognition , Cognition Disorders/physiopathology , Corpus Striatum/diagnostic imaging , Humans , Huntington Disease/diagnostic imaging , Huntington Disease/physiopathology , Huntington Disease/psychology , Mental Disorders/physiopathology , Molecular Biology , Molecular Targeted Therapy , Movement , Movement Disorders/physiopathology , Neuroimaging
5.
J Huntingtons Dis ; 5(4): 389-394, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27983561

ABSTRACT

BACKGROUND: Suicidal ideation (SI) and attempts are increased in Huntington's disease (HD), making risk factor assessment a priority. OBJECTIVE: To determine whether, hopelessness, irritability, aggression, anxiety, CAG expansion status, depression, and motor signs/symptoms were associated with Suicidal Ideation (SI) in those at risk for HD. METHODS: Behavioral and neurological data were collected from subjects in an observational study. Subject characteristics were calculated by CAG status and SI. Logistic regression models were adjusted for demographics. Separate logistic regressions were used to compare SI and non-SI subjects. A combined logistic regression model, including 4 pre-specified predictors, (hopelessness, irritability, aggression, anxiety) was used to assess the relationship of SI to these predictors. RESULTS: 801 subjects were assessed, 40 were classified as having SI, 6.3% of CAG mutation expansion carriers had SI, compared with 4.3% of non- CAG mutation expansion carriers (p = 0.2275). SI subjects had significantly increased depression (p < 0.0001), hopelessness (p < 0.0001), irritability (p < 0.0001), aggression (p = 0.0089), and anxiety (p < 0.0001), and an elevated motor score (p = 0.0098). Impulsivity, assessed in a subgroup of subjects, was also associated with SI (p = 0.0267). Hopelessness and anxiety remained significant in combined model (p < 0.001; p < 0.0198, respectively) even when motor score was included. CONCLUSIONS: Behavioral symptoms were significantly higher in those reporting SI. Hopelessness and anxiety showed a particularly strong association with SI. Risk identification could assist in assessment of suicidality in this group.


Subject(s)
Genetic Predisposition to Disease/psychology , Huntington Disease/genetics , Huntington Disease/psychology , Suicidal Ideation , Adult , Cohort Studies , Female , Humans , Logistic Models , Male , Middle Aged , Psychiatric Status Rating Scales , Risk Factors , Self Report
6.
J Huntingtons Dis ; 5(4): 347-355, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27983565

ABSTRACT

BACKGROUND: Modulation of gene transcription by HDAC inhibitors has been shown repeatedly to be neuroprotective in cellular, invertebrate, and rodent models of Huntington's disease (HD). It has been difficult to translate these treatments to the clinic, however, because existing compounds have limited potency or brain bioavailability. OBJECTIVE: In the present study, we assessed the therapeutic potential of LBH589, an orally bioavailable hydroxamic acid-derived nonselective HDAC inhibitor in mouse models of HD. METHOD: The efficacy of LBH589 is tested in two HD mouse models using various biochemical, behavioral and neuropathological outcome measures. RESULTS: We show that LBH589 crosses the blood brain barrier; induces histone hyperacetylation and prevents striatal neuronal shrinkage in R6/2 HD mice. In full-length knock-in HD mice LBH589-treatment improves motor performance and reduces neuronal atrophy. CONCLUSIONS: Our efficacious results of LBH589 in fragment and full-length mouse models of HD suggest that LBH589 is a promising candidate for clinical assessment in HD patients and provides confirmation that non-selective HDAC inhibitors can be viable clinical candidates.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Huntington Disease/drug therapy , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Neuroprotective Agents/pharmacology , Animals , Atrophy/drug therapy , Atrophy/metabolism , Atrophy/pathology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Female , Gene Knock-In Techniques , Histone Deacetylase Inhibitors/pharmacokinetics , Histones/metabolism , Huntington Disease/pathology , Huntington Disease/physiopathology , Hydroxamic Acids/pharmacokinetics , Indoles/pharmacokinetics , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Motor Activity/drug effects , Neurons/drug effects , Neurons/pathology , Neurons/physiology , Neuroprotective Agents/pharmacokinetics , Panobinostat
8.
Neurology ; 81(13): 1134-40, 2013 Sep 24.
Article in English | MEDLINE | ID: mdl-23966247

ABSTRACT

OBJECTIVE: We measured the levels of mutant huntingtin (mtHtt) and total huntingtin (tHtt) in blood leukocytes from Prospective Huntington At-Risk Observational Study (PHAROS) subjects at 50% risk of carrying the Huntington disease mutation using a homogeneous time-resolved fluorescence (HTRF) assay to assess its potential as a biomarker. METHODS: Peripheral blood mononuclear cells from consenting PHAROS subjects were analyzed by HTRF using antibodies that simultaneously measured mtHtt and tHtt. mtHtt levels were normalized to tHtt, double-stranded DNA, or protein and analyzed according to cytosine-adenine-guanine repeat length (CAGn), demographics, predicted time to clinical onset or known time since clinical onset, and available clinical measures. RESULTS: From 363 assayed samples, 342 met quality control standards. Levels of mtHtt and mt/tHtt were higher in 114 subjects with expanded CAG repeats (CAG ≥ 37) compared with 228 subjects with nonexpanded CAG repeats (CAG <37) (p < 0.0001). Analysis of relationships to predicted time to onset or to phenoconversion suggested that the HTRF signal could mark changes during the Huntington disease prodrome or after clinical onset. CONCLUSIONS: The HTRF assay can effectively measure mtHtt in multicenter sample sets and may be useful in trials of therapies targeting huntingtin.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Huntington Disease/blood , Huntington Disease/pathology , Leukocytes, Mononuclear/metabolism , Nerve Tissue Proteins/metabolism , Observation , Adult , Clinical Trials as Topic , Double-Blind Method , Female , Frontal Lobe/metabolism , Frontal Lobe/pathology , Humans , Huntingtin Protein , Huntington Disease/genetics , Longitudinal Studies , Male , Middle Aged , Mutation/genetics , Nerve Tissue Proteins/genetics , Postmortem Changes , Retrospective Studies , Trinucleotide Repeat Expansion/genetics
10.
Neurology ; 80(22): 2022-7, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23624566

ABSTRACT

OBJECTIVES: We aimed to describe the clinical phenotype conferred by the intermediate-length huntingtin allele CAG repeat expansion in a population-based study. METHODS: The Prospective Huntington At Risk Observational Study (PHAROS) enrolled adults at risk for Huntington disease (HD). They were assessed approximately every 9 months with the Unified Huntington's Disease Rating Scale (UHDRS) by investigators unaware of participants' gene status. UHDRS scores were compared according to the Huntingtin gene CAG repeat number: expanded >36, intermediate 27-35, and nonexpanded controls <26. RESULTS: Fifty (5.1%) of the 983 participants had an intermediate allele (IA). They were similar to controls on UHDRS motor, cognitive, and functional measures, but significantly worse behaviorally on apathy and suicidal ideation. On 5 of the 9 other behavioral items and on total behavior, the IA group's scores were worse than those of controls and expanded participants, who themselves scored significantly worse than controls on 6 behavioral measures. Retention rates at 4 years were 48% for the IA group compared to 58% and 60% for the expanded and control groups. CONCLUSIONS: In a cohort at risk for HD, the IA was associated with significant behavioral abnormalities but normal motor and cognition. This behavioral phenotype may represent a prodromal stage of HD, with the potential for subsequent clinical manifestations, or be part of a distinct phenotype conferred by pathology independent of the CAG expansion length.


Subject(s)
Huntington Disease/genetics , Huntington Disease/physiopathology , Nerve Tissue Proteins/genetics , Phenotype , Trinucleotide Repeat Expansion/genetics , Adult , Alleles , Cognition/physiology , Cohort Studies , Humans , Huntingtin Protein , Huntington Disease/classification , Male , Middle Aged , Motor Activity/genetics , Prospective Studies , Psychiatric Status Rating Scales , Risk
11.
J Biol Chem ; 287(16): 13487-99, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22375012

ABSTRACT

Huntington disease (HD) is caused by polyglutamine expansion in the N terminus of huntingtin (htt). Analysis of human postmortem brain lysates by SDS-PAGE and Western blot reveals htt as full-length and fragmented. Here we used Blue Native PAGE (BNP) and Western blots to study native htt in human postmortem brain. Antisera against htt detected a single band broadly migrating at 575-850 kDa in control brain and at 650-885 kDa in heterozygous and Venezuelan homozygous HD brains. Anti-polyglutamine antisera detected full-length mutant htt in HD brain. There was little htt cleavage even if lysates were pretreated with trypsin, indicating a property of native htt to resist protease cleavage. A soluble mutant htt fragment of about 180 kDa was detected with anti-htt antibody Ab1 (htt-(1-17)) and increased when lysates were treated with denaturants (SDS, 8 M urea, DTT, or trypsin) before BNP. Wild-type htt was more resistant to denaturants. Based on migration of in vitro translated htt fragments, the 180-kDa segment terminated ≈htt 670-880 amino acids. If second dimension SDS-PAGE followed BNP, the 180-kDa mutant htt was absent, and 43-50 kDa htt fragments appeared. Brain lysates from two HD mouse models expressed native full-length htt; a mutant fragment formed if lysates were pretreated with 8 M urea + DTT. Native full-length mutant htt in embryonic HD(140Q/140Q) mouse primary neurons was intact during cell death and when cell lysates were exposed to denaturants before BNP. Thus, native mutant htt occurs in brain and primary neurons as a soluble full-length monomer.


Subject(s)
Huntington Disease/genetics , Huntington Disease/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Adult , Animals , Blotting, Western , Detergents/pharmacology , Electrophoresis, Gel, Two-Dimensional , Humans , Huntingtin Protein , Huntington Disease/epidemiology , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/chemistry , Neurons/cytology , Nuclear Proteins/chemistry , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Prevalence , Primary Cell Culture , Protein Denaturation , Protein Folding , Rabbits , Reticulocytes/cytology , Solubility , Subcellular Fractions/chemistry , Subcellular Fractions/metabolism , Tissue Banks
12.
J Neurosci Methods ; 204(2): 221-6, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22155385

ABSTRACT

The availability of high quality, well-characterized antibodies for molecular and cellular neuroscience studies is important. However, not all available antibodies are rigorously evaluated, nor are limitations of particular antibodies often reported. We have examined a panel of currently available mGluR1 antibodies and have identified which ones are selective for use by western blots and immunocytochemistry. We have also specifically determined whether the antibodies cross-react to recognize mGluR5, by examining (1) tissue from both mGluR1 and mGluR5 knock-out mice and (2) primary cortical cultures, in which mGluR5 is widely expressed but mGluR1 is not. Together, these data provide a baseline characterization of antibodies that can and cannot be reliably used in these types of studies, and will hopefully facilitate and positively impact the research efforts of others studying mGluR1.


Subject(s)
Antibodies/metabolism , Antibody Specificity , Neurons/metabolism , Receptors, Metabotropic Glutamate/immunology , Animals , Animals, Newborn , Cells, Cultured , Cerebral Cortex/cytology , Gene Expression/genetics , Immunohistochemistry , Indoles , Mice , Mice, Knockout , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/deficiency
13.
Mov Disord ; 26(6): 1127-33, 2011 May.
Article in English | MEDLINE | ID: mdl-21626556

ABSTRACT

There have been extraordinary advances in our knowledge of the underlying gene, the protein it encodes, various models of disease, and potential targets for effective therapies for Huntington disease. Huntington disease research has increased exponentially in the past 25 years, and we now understand many of the molecular mechanisms underlying the disease. Still, more work needs to be done before we have a full understanding of the pathophysiology of the disease. Clinical research on biomarkers and clinical trials on potential neuroprotective agents are underway. Here we review our progress in these areas over the last 25 years and speculate on what the next 25 years may hold.


Subject(s)
Huntington Disease/epidemiology , Huntington Disease/history , Animals , Biomedical Research , Brain/pathology , Genetic Testing , History, 19th Century , History, 20th Century , History, 21st Century , Humans , Huntington Disease/genetics , Huntington Disease/therapy
14.
PLoS One ; 5(11): e13861, 2010 Nov 10.
Article in English | MEDLINE | ID: mdl-21085660

ABSTRACT

BACKGROUND: Dyskinesias associated with involuntary movements and painful muscle contractions are a common and severe complication of standard levodopa (L-DOPA, L-3,4-dihydroxyphenylalanine) therapy for Parkinson's disease. Pathologic neuroplasticity leading to hyper-responsive dopamine receptor signaling in the sensorimotor striatum is thought to underlie this currently untreatable condition. METHODOLOGY/PRINCIPAL FINDINGS: Quantitative real-time polymerase chain reaction (PCR) was employed to evaluate the molecular changes associated with L-DOPA-induced dyskinesias in Parkinson's disease. With this technique, we determined that thyrotropin releasing hormone (TRH) was greatly increased in the dopamine-depleted striatum of hemi-parkinsonian rats that developed abnormal movements in response to L-DOPA therapy, relative to the levels measured in the contralateral non-dopamine-depleted striatum, and in the striatum of non-dyskinetic control rats. ProTRH immunostaining suggested that TRH peptide levels were almost absent in the dopamine-depleted striatum of control rats that did not develop dyskinesias, but in the dyskinetic rats, proTRH immunostaining was dramatically up-regulated in the striatum, particularly in the sensorimotor striatum. This up-regulation of TRH peptide affected striatal medium spiny neurons of both the direct and indirect pathways, as well as neurons in striosomes. CONCLUSIONS/SIGNIFICANCE: TRH is not known to be a key striatal neuromodulator, but intrastriatal injection of TRH in experimental animals can induce abnormal movements, apparently through increasing dopamine release. Our finding of a dramatic and selective up-regulation of TRH expression in the sensorimotor striatum of dyskinetic rat models suggests a TRH-mediated regulatory mechanism that may underlie the pathologic neuroplasticity driving dopamine hyper-responsivity in Parkinson's disease.


Subject(s)
Corpus Striatum/drug effects , Dyskinesia, Drug-Induced/metabolism , Levodopa/toxicity , Parkinson Disease, Secondary/drug therapy , Thyrotropin-Releasing Hormone/metabolism , Analysis of Variance , Animals , Antiparkinson Agents/toxicity , Behavior, Animal/drug effects , Corpus Striatum/metabolism , Dopamine/metabolism , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/genetics , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Thyrotropin-Releasing Hormone/genetics
15.
Sci Transl Med ; 2(52): 52ra73, 2010 Oct 06.
Article in English | MEDLINE | ID: mdl-20926834

ABSTRACT

Parkinson's disease affects 5 million people worldwide, but the molecular mechanisms underlying its pathogenesis are still unclear. Here, we report a genome-wide meta-analysis of gene sets (groups of genes that encode the same biological pathway or process) in 410 samples from patients with symptomatic Parkinson's and subclinical disease and healthy controls. We analyzed 6.8 million raw data points from nine genome-wide expression studies, and 185 laser-captured human dopaminergic neuron and substantia nigra transcriptomes, followed by two-stage replication on three platforms. We found 10 gene sets with previously unknown associations with Parkinson's disease. These gene sets pinpoint defects in mitochondrial electron transport, glucose utilization, and glucose sensing and reveal that they occur early in disease pathogenesis. Genes controlling cellular bioenergetics that are expressed in response to peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) are underexpressed in Parkinson's disease patients. Activation of PGC-1α results in increased expression of nuclear-encoded subunits of the mitochondrial respiratory chain and blocks the dopaminergic neuron loss induced by mutant α-synuclein or the pesticide rotenone in cellular disease models. Our systems biology analysis of Parkinson's disease identifies PGC-1α as a potential therapeutic target for early intervention.


Subject(s)
Early Diagnosis , Genome-Wide Association Study , Heat-Shock Proteins , Parkinson Disease/genetics , Parkinson Disease/therapy , Transcription Factors , Adult , Aged , Aged, 80 and over , Computational Biology/methods , Databases, Genetic , Dopamine/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Male , Middle Aged , Neurons/metabolism , Neurons/pathology , Parkinson Disease/diagnosis , Parkinson Disease/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Transcription Factors/genetics , Transcription Factors/metabolism , alpha-Synuclein/metabolism
16.
J Neuropathol Exp Neurol ; 69(9): 880-95, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20720508

ABSTRACT

Motor dysfunction, cognitive impairment, and regional cortical atrophy indicate cerebral cortical involvement in Huntington disease (HD). To address the hypothesis that abnormal corticostriatal connectivity arises from polyglutamine-related alterations in cortical gene expression, we isolated layer 5 cortical neurons by laser-capture microdissection and analyzed transcriptome-wide mRNA changes in them. Enrichment of transcription factor mRNAs including foxp2, tbr1, and neuroD6, and neurotransmission- and plasticity-related RNAs including sema5A, pclo, ntrk2, cntn1, and Lin7b were observed. Layer 5 motor cortex neurons of transgenic R6/2 HD mice also demonstrated numerous transcriptomic changes, including decreased expression of mRNAs encoding the Lin7 homolog b ([Lin7b] also known as veli-2 and mals2). Decreases in LIN7B and CNTN1 RNAs were also detected in human HD layer 5 motor cortex neurons. Lin7 homolog b, a scaffold protein implicated in synaptic plasticity, neurite outgrowth, and cellular polarity, was decreased at the protein level in layer 5 cortical neurons in R6/2 mice and human HD brains. Decreases in Lin7b and Lin7a mRNAs were detected in R6/2 cortex as early as 6 weeks of age, suggesting that this is an early pathogenetic event. Thus, decreased cortical LIN7 expression may contribute to abnormal corticostriatal connectivity in HD.


Subject(s)
Cerebral Cortex , Huntington Disease , Membrane Proteins/metabolism , Neural Pathways , Neurons , Vesicular Transport Proteins/metabolism , Animals , Cerebral Cortex/cytology , Cerebral Cortex/pathology , Cerebral Cortex/physiology , Female , Humans , Huntington Disease/pathology , Huntington Disease/physiopathology , Membrane Proteins/genetics , Mice , Mice, Transgenic , Microarray Analysis , Neural Pathways/pathology , Neural Pathways/physiopathology , Neurons/cytology , Neurons/metabolism , Synaptic Transmission/physiology , Vesicular Transport Proteins/genetics
18.
J Neurochem ; 110(2): 557-69, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19457112

ABSTRACT

Phosphorylation of neurotransmitter receptors can modify their activity and regulate neuronal excitability. Cyclin-dependent kinase 5 (cdk5) is a proline-directed serine/threonine kinase involved not only in neuronal development, but also in synaptic function and plasticity. Here we demonstrate that group I metabotropic glutamate receptors (mGluRs), which modulate post-synaptic signaling by coupling to intracellular signal transduction pathways, are phosphorylated by cdk5. In vitro kinase assays reveal that cdk5 phosphorylates mGluR5 within the domain of the receptor that interacts with the scaffolding protein homer. Using a novel phosphospecific mGluR antibody, we show that the homer-binding domain of both mGluR1 and mGluR5 are phosphorylated in vivo, and that inhibition of cdk5 with siRNA decreases the amount of phosphorylated receptor. Furthermore, kinetic binding analysis, by surface plasmon resonance, indicates that phosphorylation of mGluR5 enhances its association with homer. Homer protein complexes in the post-synaptic density, and their disruption by an activity-dependent short homer 1a isoform, have been shown to regulate the trafficking and signaling of the mGluRs and impact many neuroadaptive processes. Phosphorylation of the mGluR homer-binding domain, in contrast to homer 1a induction, provides a novel mechanism for potentially regulating a subset of homer interactions.


Subject(s)
Carrier Proteins/metabolism , Cyclin-Dependent Kinase 5/metabolism , Receptors, Metabotropic Glutamate/metabolism , Amino Acid Sequence , Animals , COS Cells , Carrier Proteins/chemistry , Cells, Cultured , Chlorocebus aethiops , Cyclin-Dependent Kinase 5/chemistry , Homer Scaffolding Proteins , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Phosphorylation/physiology , Protein Binding/physiology , Rats , Rats, Sprague-Dawley , Receptors, AMPA/metabolism , Receptors, Kainic Acid/metabolism , Receptors, Metabotropic Glutamate/chemistry
19.
Proc Natl Acad Sci U S A ; 106(8): 2892-6, 2009 Feb 24.
Article in English | MEDLINE | ID: mdl-19171906

ABSTRACT

Voluntary movement difficulties in Parkinson's disease are initially relieved by l-DOPA therapy, but with disease progression, the repeated l-DOPA treatments can produce debilitating motor abnormalities known as l-DOPA-induced dyskinesias. We show here that 2 striatum-enriched regulators of the Ras/Rap/ERK MAP kinase signal transduction cascade, matrix-enriched CalDAG-GEFI and striosome-enriched CalDAG-GEFII (also known as RasGRP), are strongly and inversely dysregulated in proportion to the severity of abnormal movements induced by l-DOPA in a rat model of parkinsonism. In the dopamine-depleted striatum, the l-DOPA treatments produce down-regulation of CalDAG-GEFI and up-regulation of CalDAG-GEFII mRNAs and proteins, and quantification of the mRNA levels shows that these changes are closely correlated with the severity of the dyskinesias. As these CalDAG-GEFs control ERK cascades, which are implicated in l-DOPA-induced dyskinesias, and have differential compartmental expression patterns in the striatum, we suggest that they may be key molecules involved in the expression of the dyskinesias. They thus represent promising new therapeutic targets for limiting the motor complications induced by l-DOPA therapy.


Subject(s)
Antiparkinson Agents/adverse effects , DNA-Binding Proteins/physiology , Guanine Nucleotide Exchange Factors/physiology , Motor Activity , Animals , DNA-Binding Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , Immunohistochemistry , Male , Polymerase Chain Reaction , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley
20.
Proc Natl Acad Sci U S A ; 104(42): 16685-9, 2007 Oct 16.
Article in English | MEDLINE | ID: mdl-17925440

ABSTRACT

Huntington's disease (HD) is a progressive neurodegenerative disease caused by a glutamine expansion within huntingtin protein. The exact pathological mechanisms determining disease onset and progression remain unclear. However, aggregates of insoluble mutant huntingtin (mhtt), a hallmark of HD, are readily detected within neurons in HD brain. Although aggregated polyglutamines may not be inherently toxic, they constitute a biomarker for mutant huntingtin useful for developing therapeutics. We previously reported that the small molecule, C2-8, inhibits polyglutamine aggregation in cell culture and brain slices and rescues degeneration of photoreceptors in a Drosophila model of HD. In this study, we assessed the therapeutic potential of C2-8 in the R6/2 mouse model of HD, which has been used to provide proof-of-concept data in considering whether to advance therapies to human HD. We show that, at nontoxic doses, C2-8 penetrates the blood-brain barrier and is present in brain at a high concentration. C2-8-treated mice showed improved motor performance and reduced neuronal atrophy and had smaller huntingtin aggregates. There have been no prior drug-like, non-toxic, brain-penetrable aggregation inhibitors to arise from cell-based high-throughput screens for reducing huntingtin aggregation that is efficacious in preclinical in vivo models. C2-8 provides an essential tool to help elucidate mechanisms of neurodegeneration in HD and a therapeutic lead for further optimization and development.


Subject(s)
Anilides/therapeutic use , Huntington Disease/drug therapy , Sulfonamides/therapeutic use , Anilides/pharmacokinetics , Anilides/pharmacology , Animals , Atrophy/drug therapy , Blood-Brain Barrier/metabolism , Drug Evaluation, Preclinical , Female , Huntingtin Protein , Huntington Disease/pathology , Mice , Mice, Transgenic , Motor Activity/drug effects , Neostriatum/chemistry , Neostriatum/drug effects , Neostriatum/pathology , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/pathology , Nuclear Proteins/analysis , Nuclear Proteins/metabolism , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...