Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
J Biomol Screen ; 12(3): 351-60, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17379859

ABSTRACT

CAG-triplet repeat extension, translated into polyglutamines within the coding frame of otherwise unrelated gene products, causes 9 incurable neurodegenerative disorders, including Huntington's disease. Although an expansion in the CAG repeat length is the autosomal dominant mutation that causes the fully penetrant neurological phenotypes, the repeat length is inversely correlated with the age of onset. The precise molecular mechanism(s) of neurodegeneration remains elusive, but compelling evidence implicates the protein or its proteolytic fragments as the cause for the gain of novel pathological function(s). The authors sought to identify small molecules that target the selective clearance of polypeptides containing pathological polyglutamine extension. In a high-throughput chemical screen, they identified compounds that facilitate the clearance of a small huntingtin fragment with extended polyglutamines fused to green fluorescent protein reporter. Identified hits were validated in dose-response and toxicity tests. Compounds have been further tested in an assay for clearance of a larger huntingtin fragment, containing either pathological or normal polyglutamine repeats. In this assay, the authors identified compounds selectively targeting the clearance of mutant but not normal huntingtin fragments. These compounds were subjected to a functional assay, which yielded a lead compound that rescues cells from induced mutant polyglutamine toxicity.


Subject(s)
Drug Evaluation, Preclinical , Mutant Proteins/metabolism , Nerve Tissue Proteins/metabolism , Peptide Fragments/metabolism , Animals , Dose-Response Relationship, Drug , Green Fluorescent Proteins/metabolism , Molecular Weight , PC12 Cells , Peptides , Rats , Reproducibility of Results , Sensitivity and Specificity , Substrate Specificity
3.
Hum Mol Genet ; 15(6): 965-77, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16467349

ABSTRACT

Huntington's disease (HD) pathology is well understood at a histological level but a comprehensive molecular analysis of the effect of the disease in the human brain has not previously been available. To elucidate the molecular phenotype of HD on a genome-wide scale, we compared mRNA profiles from 44 human HD brains with those from 36 unaffected controls using microarray analysis. Four brain regions were analyzed: caudate nucleus, cerebellum, prefrontal association cortex [Brodmann's area 9 (BA9)] and motor cortex [Brodmann's area 4 (BA4)]. The greatest number and magnitude of differentially expressed mRNAs were detected in the caudate nucleus, followed by motor cortex, then cerebellum. Thus, the molecular phenotype of HD generally parallels established neuropathology. Surprisingly, no mRNA changes were detected in prefrontal association cortex, thereby revealing subtleties of pathology not previously disclosed by histological methods. To establish that the observed changes were not simply the result of cell loss, we examined mRNA levels in laser-capture microdissected neurons from Grade 1 HD caudate compared to control. These analyses confirmed changes in expression seen in tissue homogenates; we thus conclude that mRNA changes are not attributable to cell loss alone. These data from bona fide HD brains comprise an important reference for hypotheses related to HD and other neurodegenerative diseases.


Subject(s)
Brain/metabolism , Gene Expression Profiling , Huntington Disease/genetics , Huntington Disease/metabolism , Adult , Aged , Axons/metabolism , Brain/pathology , Cell Death/genetics , Female , Humans , Huntington Disease/pathology , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , RNA, Messenger/biosynthesis , Signal Transduction/genetics
4.
Proc Natl Acad Sci U S A ; 101(10): 3498-503, 2004 Mar 09.
Article in English | MEDLINE | ID: mdl-14993615

ABSTRACT

Huntington's disease (HD) is an autosomal dominant neurodegenerative disease caused by a triplet (CAG) expansion mutation. The length of the triplet repeat is the most important factor in determining age of onset of HD, although substantial variability remains after controlling for repeat length. The Venezuelan HD kindreds encompass 18,149 individuals spanning 10 generations, 15,409 of whom are living. Of the 4,384 immortalized lymphocyte lines collected, 3,989 DNAs were genotyped for their HD alleles, representing a subset of the population at greatest genetic risk. There are 938 heterozygotes, 80 people with variably penetrant alleles, and 18 homozygotes. Analysis of the 83 kindreds that comprise the Venezuelan HD kindreds demonstrates that residual variability in age of onset has both genetic and environmental components. We created a residual age of onset phenotype from a regression analysis of the log of age of onset on repeat length. Familial correlations (correlation +/- SE) were estimated for sibling (0.40 +/- 0.09), parent-offspring (0.10 +/- 0.11), avuncular (0.07 +/- 0.11), and cousin (0.15 +/- 0.10) pairs, suggesting a familial origin for the residual variance in onset. By using a variance-components approach with all available familial relationships, the additive genetic heritability of this residual age of onset trait is 38%. A model, including shared sibling environmental effects, estimated the components of additive genetic (0.37), shared environment (0.22), and nonshared environment (0.41) variances, confirming that approximately 40% of the variance remaining in onset age is attributable to genes other than the HD gene and 60% is environmental.


Subject(s)
Huntington Disease/etiology , Huntington Disease/genetics , Adolescent , Adult , Age of Onset , Aged , Child , Child, Preschool , Environment , Female , Humans , Huntington Disease/epidemiology , Male , Middle Aged , Models, Genetic , Phenotype , Trinucleotide Repeat Expansion , Venezuela/epidemiology
5.
Brain Res Mol Brain Res ; 102(1-2): 118-28, 2002 Jun 15.
Article in English | MEDLINE | ID: mdl-12191502

ABSTRACT

The diminished expression of D1 and D2 dopamine receptors is a well-documented hallmark of Huntington's disease (HD), but relatively little is known about how these changes in receptor populations affect the dopaminergic responses of striatal neurons. Using transgenic mice expressing an N-terminal portion of mutant huntingtin (R6/2 mice), we have examined immediate early gene (IEG) expression as an index of dopaminergic signal transduction. c-fos, jun B, zif268, and N10 mRNA levels and expression patterns were analyzed using quantitative in situ hybridization histochemistry following intraperitoneal administration of selective D1 and D2 family pharmacological agents (SKF-82958 and eticlopride). Basal IEG levels were generally lower in the dorsal subregion of R6/2 striata relative to wild-type control striata at 10-11 weeks of age, a finding in accord with previously reported decreases in D1 and adenosine A2A receptors. D2-antagonist-stimulated IEG expression was significantly reduced in the striata of transgenic animals. In contrast, D1-agonist-induced striatal R6/2 IEG mRNA levels were either equivalent or significantly enhanced relative to control levels, an unexpected result given the reduced level of D1 receptors in R6/2 animals. Understanding the functional bases for these effects may further elucidate the complex pathophysiology of Huntington's disease.


Subject(s)
Gene Expression Regulation/genetics , Genes, Immediate-Early/genetics , Huntington Disease/genetics , Neostriatum/metabolism , Neurons/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Animals , Disease Models, Animal , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Female , Gene Expression Regulation/drug effects , Genes, Immediate-Early/drug effects , Huntingtin Protein , Huntington Disease/metabolism , Huntington Disease/physiopathology , Male , Mice , Mice, Transgenic , Neostriatum/drug effects , Neostriatum/physiopathology , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurons/drug effects , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , RNA, Messenger/metabolism , Receptors, Dopamine D1/drug effects , Receptors, Dopamine D1/genetics , Receptors, Dopamine D2/drug effects , Receptors, Dopamine D2/genetics , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...