Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
J Neurol ; 270(10): 4903-4913, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37347292

ABSTRACT

BACKGROUND: A declining cognitive performance is a hallmark of Huntington's disease (HD). The neuropsychological battery of the Unified HD Rating Scale (UHDRS'99) is commonly used for assessing cognition. However, there is a need to identify and minimize the impact of confounding factors, such as language, gender, age, and education level on cognitive decline. OBJECTIVES: Aim is to provide appropriate, normative data to allow clinicians to identify disease-associated cognitive decline in diverse HD populations by compensating for the impact of confounding factors METHODS: Sample data, N = 3267 (60.5% females; mean age of 46.9 years (SD = 14.61, range 18-86) of healthy controls were used to create a normative dataset. For each neuropsychological test, a Bayesian generalized additive model with age, education, gender, and language as predictors was constructed to appropriately stratify the normative dataset. RESULTS: With advancing age, there was a non-linear decline in cognitive performance. In addition, performance was dependent on educational levels and language in all tests. Gender had a more limited impact. Standardized scores have been calculated to ease the interpretation of an individual's test outcome. A web-based online tool has been created to provide free access to normative data. CONCLUSION: For defined neuropsychological tests, the impact of gender, age, education, and language as factors confounding disease-associated cognitive decline can be minimized at the level of a single patient examination.


Subject(s)
Huntington Disease , Female , Humans , Middle Aged , Male , Huntington Disease/complications , Huntington Disease/diagnosis , Bayes Theorem , Neuropsychological Tests , Educational Status , Cognition , Language
2.
Eur J Neurol ; 30(5): 1262-1271, 2023 05.
Article in English | MEDLINE | ID: mdl-36732902

ABSTRACT

BACKGROUND AND PURPOSE: Motor speech alterations are a prominent feature of clinically manifest Huntington's disease (HD). Objective acoustic analysis of speech can quantify speech alterations. It is currently unknown, however, at what stage of HD speech alterations can be reliably detected. We aimed to explore the patterns and extent of speech alterations using objective acoustic analysis in HD and to assess correlations with both rater-assessed phenotypical features and biological determinants of HD. METHODS: Speech samples were acquired from 44 premanifest (29 pre-symptomatic and 15 prodromal) and 25 manifest HD gene expansion carriers, and 25 matched healthy controls. A quantitative automated acoustic analysis of 10 speech dimensions was performed. RESULTS: Automated speech analysis allowed us to differentiate between participants with HD and controls, with areas under the curve of 0.74 for pre-symptomatic, 0.92 for prodromal, and 0.97 for manifest stages. In addition to irregular alternating motion rates and prolonged pauses seen only in manifest HD, both prodromal and manifest HD displayed slowed articulation rate, slowed alternating motion rates, increased loudness variability, and unstable steady-state position of articulators. In participants with premanifest HD, speech alteration severity was associated with cognitive slowing (r = -0.52, p < 0.001) and the extent of bradykinesia (r = 0.43, p = 0.004). Speech alterations correlated with a measure of exposure to mutant gene products (CAG-age-product score; r = 0.60, p < 0.001). CONCLUSION: Speech abnormalities in HD are associated with other motor and cognitive deficits and are measurable already in premanifest stages of HD. Therefore, automated speech analysis might represent a quantitative HD biomarker with potential for assessing disease progression.


Subject(s)
Cognition Disorders , Huntington Disease , Humans , Huntington Disease/complications , Huntington Disease/genetics , Huntington Disease/psychology , Speech , Cross-Sectional Studies , Cognition Disorders/complications , Biomarkers
3.
Nervenarzt ; 93(2): 179-190, 2022 Feb.
Article in German | MEDLINE | ID: mdl-34762178

ABSTRACT

Huntington disease (HD) is the most frequent monogenetic neurodegenerative disease and can be unequivocally diagnosed even in the preclinical stage, at least in all individuals in whom the CAG expansion mutation in the huntingtin gene (HTT) is in the range of full penetrance. Therefore, important preconditions for an intervention early in the disease process are met, rendering modification of the course of the disease in a clinically meaningful way possible. In this respect, HD can be viewed as a model disorder for exploring neuroprotective treatment approaches. In the past emphasis was placed on the compensation of a suspected neurotransmitter deficit (GABA) analogous to Parkinson's disease and on classical neuroprotective strategies to influence hypothetical common pathways in neurodegenerative diseases (e.g., excitotoxicity, mitochondrial dysfunction, oxidative stress). With the discovery of the causative HTT mutation in 1993, therapeutic research increasingly focused on intervening as proximally as possible in the chain of pathophysiological events. Currently, an important point of intervention is the HTT mRNA with the aim of reducing the continued production of mutant huntingtin gene products and thus relieving the body of their detrimental actions. To this end, various treatment modalities (single-stranded DNA and RNA, divalent RNA and zinc finger repressor complexes, orally available splice modulators) were developed and are currently in clinical trials (phases I-III) or in late stages of preclinical development. In addition, there is the notion that it may be possible to modify the length of the somatically unstable CAG mutation, i.e. its increase in the brain during the lifetime, thereby slowing the progression of HD.


Subject(s)
Huntington Disease , Neurodegenerative Diseases , Brain , Humans , Huntingtin Protein/genetics , Huntington Disease/diagnosis , Huntington Disease/drug therapy , Huntington Disease/genetics , Mutation/genetics , Oligonucleotides, Antisense
4.
Neurobiol Dis ; 158: 105476, 2021 10.
Article in English | MEDLINE | ID: mdl-34390835

ABSTRACT

The huntingtin-associated protein 40 (HAP40) is an abundant interactor of huntingtin (HTT). In complexes of these proteins, HAP40 tightly binds to HTT in a cleft formed by two larger domains rich in HEAT repeats, and a smaller bridge domain connecting the two. We show that HAP40 steady-state protein levels are directly dependent on HTT (both normal and mutant HTT) and that HAP40 is strongly stabilized by the interaction with HTT resulting in an at least 5-fold increase in HAP40's half-life when bound to HTT. Cellular HAP40 protein levels were reduced in primary fibroblasts and lymphoblasts of Huntington Disease (HD) patients and in brain tissue of a full-length HTT mouse model of HD, concomitant with decreased soluble HTT levels in these cell types. This data and our previous demonstration of coevolution between HTT and HAP40 and evolutionary conservation of their interaction suggest that HAP40 is an obligate interaction partner of HTT. Our observation of reduced HAP40 levels in HD invites further studies, whether HAP40 loss-of-function contributes to the pathophysiology of HD.


Subject(s)
Huntingtin Protein/metabolism , Huntington Disease/metabolism , Nuclear Proteins/metabolism , Animals , Cell Line , Fibroblasts/metabolism , Gene Knock-In Techniques , HEK293 Cells , Half-Life , Humans , Kinetics , Lymphocytes/metabolism , Mice
5.
J Huntingtons Dis ; 10(2): 259-268, 2021.
Article in English | MEDLINE | ID: mdl-33459656

ABSTRACT

BACKGROUND: Besides cognitive and psychiatric abnormalities, motor symptoms are the most prominent in Huntington's disease. The manifest disease is preceded by a prodromal phase with subtle changes such as fine motor disturbances or concentration problems. OBJECTIVE: Movement disorders show a high variation in their clinical manifestation depending on condition and external influences. Therefore, devices for continuous measurements, which patients use in their daily life and which can monitor motor abnormalities, in addition to the medical examination, might be useful. The aim of current scientific efforts is to find markers that reflect the prodromal phase in gene carriers. This is important for future interventional studies, as future therapies should be applied at the stage of neuronal dysfunction, i.e., before the clinical manifestation. METHODS: We performed a software-supported, continuous monitoring of keyboard typing on the participants' own computer to evaluate this method as a tool to assess the motor phenotype in HD. We included 40 participants and obtained sufficient data from 25 participants, 12 of whom were manifest HD patients, 7 HD gene expansion carriers (HDGEC) and 6 healthy controls. RESULTS: In a cross-sectional analysis we found statistically significant higher typing inconsistency in HD patients compared to controls. Typing inconsistency compared between HDGEC and healthy controls showed a trend to higher inconsistency levels in HDGEC. We found correlations between typing cadence and clinical scores: the UHDRS finger tapping item, the composite UHDRS and the CAP score. CONCLUSION: The typing cadence inconsistency is an appropriate marker to evaluate fine motor skills of HD patients and HDGEC and is correlated to established clinical measurements.


Subject(s)
Activities of Daily Living/classification , Huntington Disease , Motor Skills/classification , Adult , Computers , Female , Humans , Huntington Disease/classification , Huntington Disease/diagnosis , Huntington Disease/physiopathology , Male , Middle Aged , Phenotype
6.
Front Mol Neurosci ; 10: 156, 2017.
Article in English | MEDLINE | ID: mdl-28603486

ABSTRACT

Neurodegenerative diseases are characterized by distinct patterns of neuronal loss. In amyotrophic lateral sclerosis (ALS) upper and lower motoneurons degenerate whereas in Huntington's disease (HD) medium spiny neurons in the striatum are preferentially affected. Despite these differences the pathophysiological mechanisms and risk factors are remarkably similar. In addition, non-neuronal features, such as weight loss implicate a dysregulation in energy metabolism. Mammalian sirtuins, especially the mitochondrial NAD+ dependent sirtuin 3 (SIRT3), regulate mitochondrial function and aging processes. SIRT3 expression depends on the activity of the metabolic master regulator peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), a modifier of ALS and HD in patients and model organisms. This prompted us to systematically probe Sirt3 mRNA and protein levels in mouse models of ALS and HD and to correlate these with patient tissue levels. We found a selective reduction of Sirt3 mRNA levels and function in the cervical spinal cord of end-stage ALS mice (superoxide dismutase 1, SOD1G93A). In sharp contrast, a tendency to increased Sirt3 mRNA levels was found in the striatum in HD mice (R6/2). Cultured primary neurons express the highest levels of Sirt3 mRNA. In primary cells from PGC-1α knock-out (KO) mice the Sirt3 mRNA levels were highest in astrocytes. In human post mortem tissue increased mRNA and protein levels of Sirt3 were found in the spinal cord in ALS, while Sirt3 levels were unchanged in the human HD striatum. Based on these findings we conclude that SIRT3 mediates the different effects of PGC-1α during the course of transgenic (tg) ALS and HD and in the human conditions only partial aspects Sirt3 dysregulation manifest.

7.
PLoS One ; 12(4): e0175248, 2017.
Article in English | MEDLINE | ID: mdl-28406926

ABSTRACT

Alterations in mitochondrial respiration are an important hallmark of Huntington's disease (HD), one of the most common monogenetic causes of neurodegeneration. The ubiquitous expression of the disease causing mutant huntingtin gene raises the prospect that mitochondrial respiratory deficits can be detected in skeletal muscle. While this tissue is readily accessible in humans, transgenic animal models offer the opportunity to cross-validate findings and allow for comparisons across organs, including the brain. The integrated respiratory chain function of the human vastus lateralis muscle was measured by high-resolution respirometry (HRR) in freshly taken fine-needle biopsies from seven pre-manifest HD expansion mutation carriers and nine controls. The respiratory parameters were unaffected. For comparison skeletal muscle isolated from HD knock-in mice (HdhQ111) as well as a broader spectrum of tissues including cortex, liver and heart muscle were examined by HRR. Significant changes of mitochondrial respiration in the HdhQ knock-in mouse model were restricted to the liver and the cortex. Mitochondrial mass as quantified by mitochondrial DNA copy number and citrate synthase activity was stable in murine HD-model tissue compared to control. mRNA levels of key enzymes were determined to characterize mitochondrial metabolic pathways in HdhQ mice. We demonstrated the feasibility to perform high-resolution respirometry measurements from small human HD muscle biopsies. Furthermore, we conclude that alterations in respiratory parameters of pre-manifest human muscle biopsies are rather limited and mirrored by a similar absence of marked alterations in HdhQ skeletal muscle. In contrast, the HdhQ111 murine cortex and liver did show respiratory alterations highlighting the tissue specific nature of mutant huntingtin effects on respiration.


Subject(s)
Huntingtin Protein/genetics , Huntington Disease , Mitochondria, Muscle , Muscle, Skeletal/metabolism , Mutation , Oxygen Consumption , Adult , Aged , Animals , Biopsy, Fine-Needle , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Female , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , Male , Mice , Mice, Transgenic , Middle Aged , Mitochondria, Muscle/genetics , Mitochondria, Muscle/metabolism
8.
Neurobiol Dis ; 97(Pt A): 36-45, 2017 01.
Article in English | MEDLINE | ID: mdl-27818323

ABSTRACT

BACKGROUND: Monogenetic forms of amyotrophic lateral sclerosis (ALS) offer an opportunity for unraveling the molecular mechanisms underlying this devastating neurodegenerative disorder. In order to identify a link between ALS-related metabolic changes and neurodegeneration, we investigated whether ALS-causing mutations interfere with the peripheral and brain-specific expression and signaling of the metabolic master regulator PGC (PPAR gamma coactivator)-1α (PGC-1α). METHODS: We analyzed the expression of PGC-1α isoforms and target genes in two mouse models of familial ALS and validated the stimulated PGC-1α signaling in primary adipocytes and neurons of these animal models and in iPS derived motoneurons of two ALS patients harboring two different frame-shift FUS/TLS mutations. RESULTS: Mutations in SOD1 and FUS/TLS decrease Ppargc1a levels in the CNS whereas in muscle and brown adipose tissue Ppargc1a mRNA levels were increased. Probing the underlying mechanism in neurons, we identified the monocarboxylate lactate as a previously unrecognized potent and selective inducer of the CNS-specific PGC-1α isoforms. Lactate also induced genes like brain-derived neurotrophic factor, transcription factor EB and superoxide dismutase 3 that are down-regulated in PGC-1α deficient neurons. The lactate-induced CNS-specific PGC-1α signaling system is completely silenced in motoneurons derived from induced pluripotent stem cells obtained from two ALS patients harboring two different frame-shift FUS/TLS mutations. CONCLUSION: ALS mutations increase the canonical PGC-1α system in the periphery while inhibiting the CNS-specific isoforms. We identify lactate as an inducer of the neuronal PGC-1α system directly linking brain metabolism and neuroprotection. Changes in the PGC-1α system might be involved in the ALS accompanied metabolic changes and in neurodegeneration.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Brain/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , RNA-Binding Protein FUS/genetics , Superoxide Dismutase-1/genetics , Adipose Tissue, Brown/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Cell Line , Disease Models, Animal , Humans , Induced Pluripotent Stem Cells/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/metabolism , Mutation , Neurons/metabolism , Protein Isoforms , RNA, Messenger/metabolism , RNA-Binding Protein FUS/metabolism , Rats , Superoxide Dismutase-1/metabolism
9.
PLoS One ; 11(11): e0166106, 2016.
Article in English | MEDLINE | ID: mdl-27820862

ABSTRACT

Huntington´s disease (HD) is a hereditary neurodegenerative disease resulting from an expanded polyglutamine sequence (poly-Q) in the protein huntingtin (HTT). Various studies report atrophy and metabolic pathology of skeletal muscle in HD and suggest as part of the process a fast-to-slow fiber type transition that may be caused by the pathological changes in central motor control or/and by mutant HTT in the muscle tissue itself. To investigate muscle pathology in HD, we used R6/2 mice, a common animal model for a rapidly progressing variant of the disease expressing exon 1 of the mutant human gene. We investigated alterations in the extensor digitorum longus (EDL), a typical fast-twitch muscle, and the soleus (SOL), a slow-twitch muscle. We focussed on mechanographic measurements of excised muscles using single and repetitive electrical stimulation and on the expression of the various myosin isoforms (heavy and light chains) using dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) of whole muscle and single fiber preparations. In EDL of R6/2, the functional tests showed a left shift of the force-frequency relation and decrease in specific force. Moreover, the estimated relative contribution of the fastest myosin isoform MyHC IIb decreased, whereas the contribution of the slower MyHC IIx isoform increased. An additional change occurred in the alkali MyLC forms showing a decrease in 3f and an increase in 1f level. In SOL, a shift from fast MyHC IIa to the slow isoform I was detectable in male R6/2 mice only, and there was no evidence of isoform interconversion in the MyLC pattern. These alterations point to a partial remodeling of the contractile apparatus of R6/2 mice towards a slower contractile phenotype, predominantly in fast glycolytic fibers.


Subject(s)
Huntington Disease/metabolism , Muscle Contraction/physiology , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Slow-Twitch/metabolism , Myosin Heavy Chains/metabolism , Myosin Light Chains/metabolism , Animals , Disease Models, Animal , Electric Stimulation/methods , Exons/genetics , Female , Humans , Huntington Disease/physiopathology , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Slow-Twitch/physiology , Muscular Diseases/metabolism , Muscular Diseases/physiopathology , Myosins/metabolism , Protein Isoforms/metabolism
10.
J Gen Physiol ; 144(5): 393-413, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25348412

ABSTRACT

Huntington's disease (HD) is caused by an expanded CAG trinucleotide repeat within the gene encoding the protein huntingtin. The resulting elongated glutamine (poly-Q) sequence of mutant huntingtin (mhtt) affects both central neurons and skeletal muscle. Recent reports suggest that ryanodine receptor-based Ca(2+) signaling, which is crucial for skeletal muscle excitation-contraction coupling (ECC), is changed by mhtt in HD neurons. Consequently, we searched for alterations of ECC in muscle fibers of the R6/2 mouse, a mouse model of HD. We performed fluorometric recordings of action potentials (APs) and cellular Ca(2+) transients on intact isolated toe muscle fibers (musculi interossei), and measured L-type Ca(2+) inward currents on internally dialyzed fibers under voltage-clamp conditions. Both APs and AP-triggered Ca(2+) transients showed slower kinetics in R6/2 fibers than in fibers from wild-type mice. Ca(2+) removal from the myoplasm and Ca(2+) release flux from the sarcoplasmic reticulum were characterized using a Ca(2+) binding and transport model, which indicated a significant reduction in slow Ca(2+) removal activity and Ca(2+) release flux both after APs and under voltage-clamp conditions. In addition, the voltage-clamp experiments showed a highly significant decrease in L-type Ca(2+) channel conductance. These results indicate profound changes of Ca(2+) turnover in skeletal muscle of R6/2 mice and suggest that these changes may be associated with muscle pathology in HD.


Subject(s)
Calcium Signaling , Huntington Disease/metabolism , Muscle Fibers, Skeletal/metabolism , Action Potentials , Animals , Calcium Channels, L-Type/metabolism , Excitation Contraction Coupling , Huntington Disease/genetics , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/physiology , Sarcoplasmic Reticulum/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics
11.
PLoS One ; 9(8): e105556, 2014.
Article in English | MEDLINE | ID: mdl-25144457

ABSTRACT

The recent discovery of active brown fat in human adults has led to renewed interest in the role of this key metabolic tissue. This is particularly true for neurodegenerative conditions like Huntington disease (HD), an adult-onset heritable disorder with a prominent energy deficit phenotype. Current methods for imaging brown adipose tissue (BAT) are in limited use because they are equipment-wise demanding and often prohibitively expensive. This prompted us to explore how a standard MRI set-up can be modified to visualize BAT in situ by taking advantage of its characteristic fat/water content ratio to differentiate it from surrounding white fat. We present a modified MRI protocol for use on an 11.7 T small animal MRI scanner to visualize and quantify BAT in wild-type and disease model laboratory mice. In this application study using the R6/2 transgenic mouse model of HD we demonstrate a significantly reduced BAT volume in HD mice vs. matched controls (n = 5 per group). This finding provides a plausible structural explanation for the previously described temperature phenotype of HD mice and underscores the significance of peripheral tissue pathology for the HD phenotype. On a more general level, the results demonstrate the feasibility of MR-based BAT imaging in rodents and open the path towards transferring this imaging approach to human patients. Future studies are needed to determine if this method can be used to track disease progression in HD and other disease entities associated with BAT abnormalities, including metabolic conditions such as obesity, cachexia, and diabetes.


Subject(s)
Adipose Tissue, Brown/pathology , Huntington Disease/pathology , Magnetic Resonance Imaging , Animals , Disease Models, Animal , Female , Huntington Disease/diagnosis , Magnetic Resonance Imaging/methods , Mice , Mice, Transgenic
12.
Proc Natl Acad Sci U S A ; 109(38): E2523-32, 2012 Sep 18.
Article in English | MEDLINE | ID: mdl-22927399

ABSTRACT

Aberrant mitochondrial function, morphology, and transport are main features of neurodegenerative diseases. To date, mitochondrial transport within neurons is thought to rely mainly on microtubules, whereas actin might mediate short-range movements and mitochondrial anchoring. Here, we analyzed the impact of actin on neuronal mitochondrial size and localization. F-actin enhanced mitochondrial size and mitochondrial number in neurites and growth cones. In contrast, raising G-actin resulted in mitochondrial fragmentation and decreased mitochondrial abundance. Cellular F-actin/G-actin levels also regulate serum response factor (SRF)-mediated gene regulation, suggesting a possible link between SRF and mitochondrial dynamics. Indeed, SRF-deficient neurons display neurodegenerative hallmarks of mitochondria, including disrupted morphology, fragmentation, and impaired mitochondrial motility, as well as ATP energy metabolism. Conversely, constitutively active SRF-VP16 induced formation of mitochondrial networks and rescued huntingtin (HTT)-impaired mitochondrial dynamics. Finally, SRF and actin dynamics are connected via the actin severing protein cofilin and its slingshot phosphatase to modulate neuronal mitochondrial dynamics. In summary, our data suggest that the SRF-cofilin-actin signaling axis modulates neuronal mitochondrial function.


Subject(s)
Actins/metabolism , Cofilin 1/metabolism , Mitochondria/metabolism , Serum Response Factor/metabolism , Signal Transduction , Adenosine Triphosphate/metabolism , Animals , Herpes Simplex Virus Protein Vmw65/metabolism , Hippocampus/metabolism , Huntingtin Protein , Mice , Mice, Transgenic , Microtubules/metabolism , Models, Biological , Mutation , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Phosphoric Monoester Hydrolases/metabolism , Tissue Distribution
13.
J Gene Med ; 14(7): 468-81, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22700462

ABSTRACT

BACKGROUND: Neuronal degeneration, in particular in the striatum, and the formation of nuclear and cytoplasmic inclusions are characteristics of Huntington's disease (HD) as a result of the expansion of a polyglutamine tract located close to the N-terminus of huntingtin (htt). Because of the large (10-kb) size of the htt cDNA, expression of full-length htt in primary neurons has proved difficult in the past. METHODS: We generated a new chronic in vitro model that is based on high-capacity adenovirus vector-mediated transduction of primary murine striatal and cortical neurons. Because the vector has a large capacity for transport of foreign DNA, it was possible to quantitatively express in these primary cells normal and mutant full-length htt (designed as fusion proteins with enhanced green fluorescent protein) in addition to its truncated versions. Pathological changes caused by mutant htt were characterized. RESULTS: The model mimicked several features observed in HD patients: prominent nuclear inclusions in cortical but not in striatal neurons, preferential neuronal degeneration of striatal neurons and neurofilament fragmentation in this cell type. Compared with expressed truncated mutant htt, the expression of full-length mutant htt in neurons resulted in a much slower appearance of pathological changes. Different from cortical neurons, the vast majority of nuclei in striatal cells contained only diffusely distributed N-terminal htt fragments. Cytoplasmic inclusions in both cell types contained full-length mutant htt. CONCLUSIONS: This model and the adenovirus vectors used will be valuable for studying the function of htt and the pathogenesis of HD at molecular and cellular levels in different neuronal cell types.


Subject(s)
Adenoviridae/genetics , Corpus Striatum/pathology , Huntington Disease/pathology , Nerve Degeneration/pathology , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Animals , Cell Culture Techniques , Corpus Striatum/metabolism , Female , Genetic Vectors , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/metabolism , Inclusion Bodies/metabolism , Intranuclear Inclusion Bodies/metabolism , Mice , Mice, Inbred BALB C , Models, Neurological , Mutation , Nerve Degeneration/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Nuclear Proteins/metabolism , Peptides/genetics , Pregnancy
14.
Neurosci Lett ; 517(1): 7-12, 2012 May 23.
Article in English | MEDLINE | ID: mdl-22712074

ABSTRACT

Ataxia is a clinical feature of most polyglutamine disorders. Cerebellar neurodegeneration of Purkinje cells (PCs) in Huntington's Disease (HD) brain was described in the 1980s. PC death in the R6/2 transgenic model for HD was published by Turmaine et al. So far, PCs have not been examined on a single cell level. In order to begin to understand PC dysfunction and degeneration in HD we performed a gene expression study on laser-dissected PC based on a DNA microarray screening and quantitative real time PCR (Q-PCR). We demonstrate downregulation of the retinoid acid receptor-related orphan receptor (ROR) mRNA and ROR-mediated mRNAs, also seen by immunofluorescent staining. As ROR and ROR-dependent transcriptional dysregulation is not only found in the R6/2 model for HD but also in a model for spinocerebellar ataxia type 1 (SCA1) (Serra et al.) the data suggest common pathogenic mechanisms for both polyglutamine diseases.


Subject(s)
Huntington Disease/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Purkinje Cells/cytology , Purkinje Cells/metabolism , Animals , Cerebellum/metabolism , Cerebellum/pathology , Disease Models, Animal , Down-Regulation , Female , Huntington Disease/metabolism , Mice , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/metabolism
15.
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
16.
Mov Disord ; 22(11): 1637-40, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17534945

ABSTRACT

A semi professional marathon runner at risk for Huntington's disease (HD) (43 CAG repeats) developed signs of a slowly progressive myopathy with exercise-induced muscle fatigue, pain, elevated creatine kinase level, and worsening of his running performance many years before first signs of chorea were detected. Muscle biopsy displayed a mild myopathy with mitochondrial pathology including a complex IV deficiency and analysis of the patient's fibroblast culture demonstrated deficits in mitochondrial function. Challenging skeletal muscle by excessive training might have disclosed myopathy in HD even years before the appearance of other neurological symptoms.


Subject(s)
Huntington Disease/complications , Muscular Diseases/etiology , Adult , Disease Progression , Humans , Huntington Disease/genetics , Male , Mitochondria, Muscle/pathology , Muscle, Skeletal/pathology , Muscular Diseases/genetics , Muscular Diseases/pathology , Mutation , Oxygen Consumption/physiology , Proton Pumps/genetics , Running
17.
J Neurochem ; 98(2): 576-87, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16805848

ABSTRACT

Polyglutamine (polyQ) expansion in many proteins, including huntingtin and ataxin-3, is pathogenic and responsible for neuronal dysfunction and degeneration. Although at least nine neurodegenerative diseases are caused by expanded polyQ, the pathogenesis of these diseases is still not well understood. In the present study, we used Caenorhabditis elegans to study the molecular mechanism of polyQ-mediated toxicity. We expressed full-length and truncated ataxin-3 with different lengths of polyQ in the nervous system of C. elegans. We show that expanded polyQ interrupts synaptic transmission, and induces swelling and aberrant branching of neuronal processes. Using an ubiquitinated fluorescence reporter construct, we also showed that polyQ aggregates impair the ubiquitin-proteasome system in C. elegans. These results may provide information for further understanding the pathogenesis of polyQ diseases.


Subject(s)
Caenorhabditis elegans/physiology , Peptides/pharmacology , Proteasome Endopeptidase Complex/physiology , Synaptic Transmission/drug effects , Ubiquitin/physiology , Animals , Blotting, Western , Cell Death , Cell Line , Fluorescent Dyes , Humans , Immunohistochemistry , Larva/metabolism , Microscopy, Fluorescence , Motor Activity/drug effects , Neurons/drug effects , Transfection
18.
Ann Neurol ; 59(2): 407-11, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16437579

ABSTRACT

OBJECTIVE: The aim of the present work was the detection of Mitochondrial dysfunction of Huntington's disease (HD). METHODS: We investigated muscle and muscle mitochondria of 14- to 16-week-old R6/2 mice in comparison with wild-type mice. RESULTS: Atrophic fibers, increased fuchsinophilic aggregates, and reduced cytochrome c oxidase (15%) were found in HD muscle. With swelling measurements and Ca2+ accumulation experiments, a decreased stability of HD mitochondria against Ca2+-induced permeability transition was detected. Complex I-dependent respiration of HD mitochondria was more sensitive to inhibition by adding 10 microm Ca2+ than wild-type mitochondria. INTERPRETATION: Data suggest that the decreased stability of HD mitochondria against Ca2+ contributes to energetic depression and cell atrophy.


Subject(s)
Calcium/pharmacology , Huntington Disease/metabolism , Mitochondria, Muscle/drug effects , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Oxygen Consumption/drug effects , Respiration/drug effects , Time Factors , Trinucleotide Repeats/genetics
19.
Cardiovasc Res ; 66(1): 33-44, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15769446

ABSTRACT

OBJECTIVE: Most cardiac myosin binding protein C (cMyBP-C) gene mutations causing familial hypertrophic cardiomyopathy (FHC) result in C-terminal truncated proteins. However, truncated cMyBP-Cs were undetectable in myocardial tissue of FHC patients. In the present study, we investigated whether truncated cMyBP-Cs are subject to accelerated degradation by the lysosome or ubiquitin-proteasome system (UPS). METHODS AND RESULTS: By using an adenovirus-based approach, we analyzed expression and localization of myc-tagged truncated proteins (M6t 3%, M7t 80% truncation, both mutations have been identified in FHC patients) compared to wild type (WT) in neonatal rat cardiomyocytes. Despite similar mRNA levels, protein expression of M6t and M7t was markedly lower than WT (70+/-4% and 11+/-5% of WT, respectively, p<0.05). M6t exhibited weak incorporation in the sarcomere, whereas M7t was mis-incorporated at the Z-disk and formed ubiquitin-positive aggregates. The lysosome inhibitor bafilomycin only slightly raised the protein level of M7t, whereas the UPS inhibitors lactacystin or MG132 markedly raised M6t and M7t to WT level. Using an adenovirus encoding a fluorescent reporter of UPS activity, we demonstrate that mutant cMyBP-Cs impair the proteolytic capacity of the UPS. CONCLUSION: Truncated cMyBP-Cs are preferentially degraded by the UPS, which, in turn, may competitively inhibit breakdown of other UPS substrates. Since the UPS plays an important role in a variety of fundamental cellular processes, we propose impairment of this system by mutant cMyBP-Cs as a contributing factor to the pathogenesis of FHC.


Subject(s)
Cardiomyopathy, Hypertrophic/metabolism , Carrier Proteins/genetics , Mutation , Myocytes, Cardiac/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Adenoviridae/genetics , Animals , Animals, Newborn , Blotting, Northern/methods , Blotting, Western/methods , Cardiomyopathy, Hypertrophic/genetics , Carrier Proteins/metabolism , Cell Culture Techniques , Flow Cytometry , Genetic Vectors/pharmacology , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Rats , Rats, Wistar , Sarcomeres/metabolism , Transduction, Genetic , Ubiquitin/genetics
20.
Cell ; 119(1): 121-35, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15454086

ABSTRACT

PGC-1alpha is a coactivator of nuclear receptors and other transcription factors that regulates several metabolic processes, including mitochondrial biogenesis and respiration, hepatic gluconeogenesis, and muscle fiber-type switching. We show here that, while hepatocytes lacking PGC-1alpha are defective in the program of hormone-stimulated gluconeogenesis, the mice have constitutively activated gluconeogenic gene expression that is completely insensitive to normal feeding controls. C/EBPbeta is elevated in the livers of these mice and activates the gluconeogenic genes in a PGC-1alpha-independent manner. Despite having reduced mitochondrial function, PGC-1alpha null mice are paradoxically lean and resistant to diet-induced obesity. This is largely due to a profound hyperactivity displayed by the null animals and is associated with lesions in the striatal region of the brain that controls movement. These data illustrate a central role for PGC-1alpha in the control of energy metabolism but also reveal novel systemic compensatory mechanisms and pathogenic effects of impaired energy homeostasis.


Subject(s)
Brain/metabolism , Energy Metabolism/genetics , Gluconeogenesis/genetics , Hyperkinesis/genetics , Mitochondria/metabolism , Trans-Activators/genetics , Adaptation, Physiological/genetics , Animals , Appetite Regulation/genetics , Basal Ganglia Diseases/genetics , Basal Ganglia Diseases/metabolism , Basal Ganglia Diseases/pathology , Brain/physiopathology , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Corpus Striatum/metabolism , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Gene Expression Regulation/genetics , Glucose/metabolism , Hepatocytes/metabolism , Homeostasis/genetics , Hyperkinesis/pathology , Hyperkinesis/physiopathology , Liver/metabolism , Liver/physiopathology , Mice , Mice, Knockout , Mitochondria/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/metabolism , Obesity/genetics , Obesity/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Trans-Activators/deficiency , Transcription Factors , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...