Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
Add more filters

Publication year range
1.
Nature ; 490(7419): 187-91, 2012 Oct 11.
Article in English | MEDLINE | ID: mdl-23060188

ABSTRACT

The US National Institute of Neurological Disorders and Stroke convened major stakeholders in June 2012 to discuss how to improve the methodological reporting of animal studies in grant applications and publications. The main workshop recommendation is that at a minimum studies should report on sample-size estimation, whether and how animals were randomized, whether investigators were blind to the treatment, and the handling of data. We recognize that achieving a meaningful improvement in the quality of reporting will require a concerted effort by investigators, reviewers, funding agencies and journal editors. Requiring better reporting of animal studies will raise awareness of the importance of rigorous study design to accelerate scientific progress.


Subject(s)
Publishing/standards , Research Design/standards , Animals , Publishing/trends , Random Allocation , Sample Size , Statistics as Topic
2.
PLoS Biol ; 12(6): e1001895, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24960609

ABSTRACT

The Wnt receptor Ryk is an evolutionary-conserved protein important during neuronal differentiation through several mechanisms, including γ-secretase cleavage and nuclear translocation of its intracellular domain (Ryk-ICD). Although the Wnt pathway may be neuroprotective, the role of Ryk in neurodegenerative disease remains unknown. We found that Ryk is up-regulated in neurons expressing mutant huntingtin (HTT) in several models of Huntington's disease (HD). Further investigation in Caenorhabditis elegans and mouse striatal cell models of HD provided a model in which the early-stage increase of Ryk promotes neuronal dysfunction by repressing the neuroprotective activity of the longevity-promoting factor FOXO through a noncanonical mechanism that implicates the Ryk-ICD fragment and its binding to the FOXO co-factor ß-catenin. The Ryk-ICD fragment suppressed neuroprotection by lin-18/Ryk loss-of-function in expanded-polyQ nematodes, repressed FOXO transcriptional activity, and abolished ß-catenin protection of mutant htt striatal cells against cell death vulnerability. Additionally, Ryk-ICD was increased in the nucleus of mutant htt cells, and reducing γ-secretase PS1 levels compensated for the cytotoxicity of full-length Ryk in these cells. These findings reveal that the Ryk-ICD pathway may impair FOXO protective activity in mutant polyglutamine neurons, suggesting that neurons are unable to efficiently maintain function and resist disease from the earliest phases of the pathogenic process in HD.


Subject(s)
Forkhead Transcription Factors/metabolism , Huntington Disease/etiology , Neurons/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Wnt/metabolism , Aged , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Line , Female , Humans , Huntington Disease/metabolism , Male , Mice , Mice, Transgenic , Middle Aged , Oligonucleotide Array Sequence Analysis , Presenilin-1/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Wnt Signaling Pathway
3.
J Neurosci ; 33(6): 2313-25, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23392662

ABSTRACT

We previously demonstrated that sodium butyrate is neuroprotective in Huntington's disease (HD) mice and that this therapeutic effect is associated with increased expression of mitogen-activated protein kinase/dual-specificity phosphatase 1 (MKP-1/DUSP1). Here we show that enhancing MKP-1 expression is sufficient to achieve neuroprotection in lentiviral models of HD. Wild-type MKP-1 overexpression inhibited apoptosis in primary striatal neurons exposed to an N-terminal fragment of polyglutamine-expanded huntingtin (Htt171-82Q), blocking caspase-3 activation and significantly reducing neuronal cell death. This neuroprotective effect of MKP-1 was demonstrated to be dependent on its enzymatic activity, being ablated by mutation of its phosphatase domain and being attributed to inhibition of specific MAP kinases (MAPKs). Overexpression of MKP-1 prevented the polyglutamine-expanded huntingtin-induced activation of c-Jun N-terminal kinases (JNKs) and p38 MAPKs, whereas extracellular signal-regulated kinase (ERK) 1/2 activation was not altered by either polyglutamine-expanded Htt or MKP-1. Moreover, mutants of MKP-1 that selectively prevented p38 or JNK binding confirmed the important dual contributions of p38 and JNK regulation to MKP-1-mediated neuroprotection. These results demonstrate additive effects of p38 and JNK MAPK inhibition by MKP-1 without consequence to ERK activation in this striatal neuron-based paradigm. MKP-1 also provided neuroprotection in vivo in a lentiviral model of HD neuropathology in rat striatum. Together, these data extend previous evidence that JNK- and p38-mediated pathways contribute to HD pathogenesis and, importantly, show that therapies simultaneously inhibiting both JNK and p38 signaling pathways may lead to improved neuroprotective outcomes.


Subject(s)
Dual Specificity Phosphatase 1/biosynthesis , Huntington Disease/enzymology , Huntington Disease/prevention & control , MAP Kinase Kinase 4/antagonists & inhibitors , Neuroprotective Agents/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Cells, Cultured , Female , MAP Kinase Kinase 4/metabolism , Mice , Rats , Rats, Wistar , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Proc Natl Acad Sci U S A ; 108(41): 17141-6, 2011 Oct 11.
Article in English | MEDLINE | ID: mdl-21969577

ABSTRACT

Huntington disease (HD) is a progressive neurodegenerative disease that affects 30,000 individuals in North America. Treatments that slow its relentless course are not yet available, and biomarkers that can reliably measure disease activity and therapeutic response are urgently needed to facilitate their development. Here, we interrogated 119 human blood samples for transcripts associated with HD. We found that the dynamic regulator of chromatin plasticity H2A histone family, member Y (H2AFY) is specifically overexpressed in the blood and frontal cortex of patients with HD compared with controls. This association precedes the onset of clinical symptoms, was confirmed in two mouse models, and was independently replicated in cross-sectional and longitudinal clinical studies comprising 142 participants. A histone deacetylase inhibitor that suppresses neurodegeneration in animal models reduces H2AFY levels in a randomized phase II clinical trial. This study identifies the chromatin regulator H2AFY as a potential biomarker associated with disease activity and pharmacodynamic response that may become useful for enabling disease-modifying therapeutics for HD.


Subject(s)
Histones/genetics , Histones/metabolism , Huntington Disease/genetics , Huntington Disease/metabolism , Adult , Aged , Animals , Case-Control Studies , Cross-Sectional Studies , Disease Models, Animal , Double-Blind Method , Female , Frontal Lobe/metabolism , Gene Expression , Histone Deacetylase Inhibitors/pharmacology , Histones/blood , Humans , Huntington Disease/blood , Longitudinal Studies , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Middle Aged , Nerve Degeneration/drug therapy , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
Neurobiol Dis ; 55: 26-35, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23537713

ABSTRACT

Caspase-mediated cell death contributes to the pathogenesis of motor neuron degeneration in the mutant SOD1(G93A) transgenic mouse model of amyotrophic lateral sclerosis (ALS), along with other factors such as inflammation and oxidative damage. By screening a drug library, we found that melatonin, a pineal hormone, inhibited cytochrome c release in purified mitochondria and prevented cell death in cultured neurons. In this study, we evaluated whether melatonin would slow disease progression in SOD1(G93A) mice. We demonstrate that melatonin significantly delayed disease onset, neurological deterioration and mortality in ALS mice. ALS-associated ventral horn atrophy and motor neuron death were also inhibited by melatonin treatment. Melatonin inhibited Rip2/caspase-1 pathway activation, blocked the release of mitochondrial cytochrome c, and reduced the overexpression and activation of caspase-3. Moreover, for the first time, we determined that disease progression was associated with the loss of both melatonin and the melatonin receptor 1A (MT1) in the spinal cord of ALS mice. These results demonstrate that melatonin is neuroprotective in transgenic ALS mice, and this protective effect is mediated through its effects on the caspase-mediated cell death pathway. Furthermore, our data suggest that melatonin and MT1 receptor loss may play a role in the pathological phenotype observed in ALS. The above observations indicate that melatonin and modulation of Rip2/caspase-1/cytochrome c or MT1 pathways may be promising therapeutic approaches for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Antioxidants/therapeutic use , Cell Death/drug effects , Cell Death/ethics , Melatonin/therapeutic use , Signal Transduction/drug effects , Amyotrophic Lateral Sclerosis/genetics , Analysis of Variance , Animals , Caspase 3/metabolism , Cytochromes c/metabolism , Disease Models, Animal , Disease Progression , Enzyme-Linked Immunosorbent Assay , Mice , Mice, Transgenic , Receptor, Melatonin, MT1/metabolism , Superoxide Dismutase/genetics
6.
J Neurosci ; 31(41): 14496-507, 2011 Oct 12.
Article in English | MEDLINE | ID: mdl-21994366

ABSTRACT

Melatonin mediates neuroprotection in several experimental models of neurodegeneration. It is not yet known, however, whether melatonin provides neuroprotection in genetic models of Huntington's disease (HD). We report that melatonin delays disease onset and mortality in a transgenic mouse model of HD. Moreover, mutant huntingtin (htt)-mediated toxicity in cells, mice, and humans is associated with loss of the type 1 melatonin receptor (MT1). We observe high levels of MT1 receptor in mitochondria from the brains of wild-type mice but much less in brains from HD mice. Moreover, we demonstrate that melatonin inhibits mutant htt-induced caspase activation and preserves MT1 receptor expression. This observation is critical, because melatonin-mediated protection is dependent on the presence and activation of the MT1 receptor. In summary, we delineate a pathologic process whereby mutant htt-induced loss of the mitochondrial MT1 receptor enhances neuronal vulnerability and potentially accelerates the neurodegenerative process.


Subject(s)
Huntington Disease/metabolism , Melatonin/pharmacology , Mutation/genetics , Nerve Tissue Proteins/genetics , Neurons/drug effects , Neuroprotective Agents/pharmacology , Nuclear Proteins/genetics , Receptor, Melatonin, MT1/metabolism , Analysis of Variance , Animals , Brain/cytology , Brain/drug effects , Brain/metabolism , Caspase 3/analysis , Caspase 3/metabolism , Caspase 9/analysis , Caspase 9/metabolism , Cell Death/drug effects , Cell Death/genetics , Cells, Cultured , Disease Models, Animal , Embryo, Mammalian , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Humans , Huntingtin Protein , Huntington Disease/drug therapy , Huntington Disease/pathology , Hydrogen Peroxide/toxicity , Male , Melatonin/therapeutic use , Mice , Mice, Mutant Strains , Middle Aged , Mitochondria/drug effects , Mitochondria/metabolism , Nerve Tissue Proteins/metabolism , Neurons/ultrastructure , Nuclear Proteins/metabolism , Postmortem Changes , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Rats , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT2/genetics , Receptor, Melatonin, MT2/metabolism , Statistics, Nonparametric , Time Factors , Transfection/methods
7.
Hum Mol Genet ; 19(20): 3919-35, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20660112

ABSTRACT

Although a direct causative pathway from the gene mutation to the selective neostriatal neurodegeneration remains unclear in Huntington's disease (HD), one putative pathological mechanism reported to play a prominent role in the pathogenesis of this neurological disorder is mitochondrial dysfunction. We examined mitochondria in preferentially vulnerable striatal calbindin-positive neurons in moderate-to-severe grade HD patients, using antisera against mitochondrial markers of COX2, SOD2 and cytochrome c. Combined calbindin and mitochondrial marker immunofluorescence showed a significant and progressive grade-dependent reduction in the number of mitochondria in spiny striatal neurons, with marked alteration in size. Consistent with mitochondrial loss, there was a reduction in COX2 protein levels using western analysis that corresponded with disease severity. In addition, both mitochondrial transcription factor A, a regulator of mtDNA, and peroxisome proliferator-activated receptor-co-activator gamma-1 alpha, a key transcriptional regulator of energy metabolism and mitochondrial biogenesis, were also significantly reduced with increasing disease severity. Abnormalities in mitochondrial dynamics were observed, showing a significant increase in the fission protein Drp1 and a reduction in the expression of the fusion protein mitofusin 1. Lastly, mitochondrial PCR array profiling in HD caudate nucleus specimens showed increased mRNA expression of proteins involved in mitochondrial localization, membrane translocation and polarization and transport that paralleled mitochondrial derangement. These findings reveal that there are both mitochondrial loss and altered mitochondrial morphogenesis with increased mitochondrial fission and reduced fusion in HD. These findings provide further evidence that mitochondrial dysfunction plays a critical role in the pathogenesis of HD.


Subject(s)
Huntington Disease/metabolism , Huntington Disease/pathology , Mitochondria/metabolism , Mitochondria/pathology , Neostriatum/metabolism , Neostriatum/ultrastructure , Calbindins , Cytochromes c/analysis , Cytochromes c/immunology , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/metabolism , Dynamins , Electron Transport Complex IV/analysis , Energy Metabolism , Fluorescent Antibody Technique , GTP Phosphohydrolases/metabolism , Gene Expression , Gene Expression Profiling , Humans , Huntingtin Protein , Huntington Disease/genetics , Membrane Potential, Mitochondrial , Membrane Transport Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Mitochondria/genetics , Mitochondrial Membrane Transport Proteins , Mitochondrial Proteins/metabolism , Nerve Tissue Proteins/genetics , Neurons/chemistry , Neurons/pathology , Nuclear Proteins/genetics , Peroxisome Proliferator-Activated Receptors/metabolism , Polymerase Chain Reaction , S100 Calcium Binding Protein G/analysis , Superoxide Dismutase/analysis , Superoxide Dismutase/immunology , Transcription Factors/metabolism
8.
Hum Mol Genet ; 19(15): 3053-67, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20494921

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder previously thought to be of primary neuronal origin, despite ubiquitous expression of mutant huntingtin (mHtt). We tested the hypothesis that mHtt expressed in astrocytes may contribute to the pathogenesis of HD. To better understand the contribution of astrocytes in HD in vivo, we developed a novel mouse model using lentiviral vectors that results in selective expression of mHtt into striatal astrocytes. Astrocytes expressing mHtt developed a progressive phenotype of reactive astrocytes that was characterized by a marked decreased expression of both glutamate transporters, GLAST and GLT-1, and of glutamate uptake. These effects were associated with neuronal dysfunction, as observed by a reduction in DARPP-32 and NR2B expression. Parallel studies in brain samples from HD subjects revealed early glial fibrillary acidic protein expression in striatal astrocytes from Grade 0 HD cases. Astrogliosis was associated with morphological changes that increased with severity of disease, from Grades 0 through 4 and was more prominent in the putamen. Combined immunofluorescence showed co-localization of mHtt in astrocytes in all striatal HD specimens, inclusive of Grade 0 HD. Consistent with the findings from experimental mice, there was a significant grade-dependent decrease in striatal GLT-1 expression from HD subjects. These findings suggest that the presence of mHtt in astrocytes alters glial glutamate transport capacity early in the disease process and may contribute to HD pathogenesis.


Subject(s)
Astrocytes/metabolism , Glutamic Acid/metabolism , Huntington Disease/metabolism , Neostriatum/pathology , Peptides/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Trinucleotide Repeat Expansion/genetics , Aged , Amino Acid Transport System X-AG/metabolism , Animals , Astrocytes/pathology , Biological Transport , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Down-Regulation , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/metabolism , Humans , Huntington Disease/pathology , Lentivirus/genetics , Mice , Middle Aged , Mutant Proteins/metabolism , Neostriatum/metabolism , Neurons/metabolism , Neurons/pathology , Phenotype , Receptors, N-Methyl-D-Aspartate/metabolism , Time Factors
9.
Bioorg Med Chem ; 20(2): 1029-45, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22189273

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive loss of motor neurons. Currently, there is only one FDA-approved treatment for ALS (riluzole), and that drug only extends life, on average, by 2-3 months. Mutations in Cu/Zn superoxide dismutase (SOD1) are found in familial forms of the disease and have played an important role in the study of ALS pathophysiology. On the basis of their activity in a PC12-G93A-YFP high-throughput screening assay, several bioactive compounds have been identified and classified as cyclohexane-1,3-dione (CHD) derivatives. A concise and efficient synthetic route has been developed to provide diverse CHD analogs. The structural modification of the CHD scaffold led to the discovery of a more potent analog (26) with an EC(50) of 700 nM having good pharmacokinetic properties, such as high solubility, low human and mouse metabolic potential, and relatively good plasma stability. It was also found to efficiently penetrate the blood-brain barrier. However, compound 26 did not exhibit any significant life span extension in the ALS mouse model. It was found that, although 26 was active in PC12 cells, it had poor activity in other cell types, including primary cortical neurons, indicating that it can penetrate into the brain, but is not active in neuronal cells, potentially due to poor selective cell penetration. Further structural modification of the CHD scaffold was aimed at improving global cell activity as well as maintaining potency. Two new analogs (71 and 73) were synthesized, which had significantly enhanced cortical neuronal cell permeability, as well as similar potency to that of 26 in the PC12-G93A assay. These CHD analogs are being investigated further as novel therapeutic candidates for ALS.


Subject(s)
Cyclohexanones/chemistry , Cyclohexanones/pharmacology , Cyclopropanes/chemistry , Phenyl Ethers/chemistry , Superoxide Dismutase/antagonists & inhibitors , Amino Acid Substitution , Amyotrophic Lateral Sclerosis/drug therapy , Animals , Blood-Brain Barrier/metabolism , Cyclohexanones/therapeutic use , Cyclohexanones/toxicity , Cyclopropanes/therapeutic use , Cyclopropanes/toxicity , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Mutation , Neurons/drug effects , PC12 Cells , Phenyl Ethers/therapeutic use , Phenyl Ethers/toxicity , Rats , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
10.
Biochim Biophys Acta ; 1802(7-8): 673-81, 2010.
Article in English | MEDLINE | ID: mdl-20460152

ABSTRACT

A major goal of current clinical research in Huntington's disease (HD) has been to identify preclinical and manifest disease biomarkers, as these may improve both diagnosis and the power for therapeutic trials. Although the underlying biochemical alterations and the mechanisms of neuronal degeneration remain unknown, energy metabolism defects in HD have been chronicled for many years. We report that the brain isoenzyme of creatine kinase (CK-BB), an enzyme important in buffering energy stores, was significantly reduced in presymptomatic and manifest disease in brain and blood buffy coat specimens in HD mice and HD patients. Brain CK-BB levels were significantly reduced in R6/2 mice by approximately 18% to approximately 68% from 21 to 91 days of age, while blood CK-BB levels were decreased by approximately 14% to approximately 44% during the same disease duration. Similar findings in CK-BB levels were observed in the 140 CAG mice from 4 to 12 months of age, but not at the earliest time point, 2 months of age. Consistent with the HD mice, there was a grade-dependent loss of brain CK-BB that worsened with disease severity in HD patients from approximately 28% to approximately 63%, as compared to non-diseased control patients. In addition, CK-BB blood buffy coat levels were significantly reduced in both premanifest and symptomatic HD patients by approximately 23% and approximately 39%, respectively. The correlation of CK-BB as a disease biomarker in both CNS and peripheral tissues from HD mice and HD patients may provide a powerful means to assess disease progression and to predict the potential magnitude of therapeutic benefit in this disorder.


Subject(s)
Central Nervous System/metabolism , Creatine Kinase, BB Form/blood , Creatine Kinase, BB Form/metabolism , Huntington Disease/blood , Huntington Disease/metabolism , Aged , Animals , Biomarkers/analysis , Biomarkers/blood , Biomarkers/metabolism , Case-Control Studies , Down-Regulation , Female , Humans , Huntington Disease/diagnosis , Huntington Disease/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Middle Aged , Postmortem Changes
11.
Amyotroph Lateral Scler ; 12(2): 87-96, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21073276

ABSTRACT

The underlying cause of amyotrophic lateral sclerosis (ALS), a progressive neurodegenerative disorder, remains unknown. However, there is strong evidence that one pathophysiological mechanism, toxic protein misfolding and/or aggregation, may trigger motor neuron dysfunction and loss. Since the clinical and pathological features of sporadic and familial ALS are indistinguishable, all forms of the disease may be better understood and ultimately treated by studying pathogenesis and therapy in models expressing mutant forms of SOD1. We developed a cellular model in which cell death depended on the expression of G93A-SOD1, a mutant form of superoxide dismutase found in familial ALS patients that produces toxic protein aggregates. This cellular model was optimized for high throughput screening to identify protective compounds from a >50,000 member chemical library. Three novel chemical scaffolds were selected for further study following screen implementation, counter-screening and secondary testing, including studies with purchased analogs. All three scaffolds blocked SOD1 aggregation in high content screening assays and data on the optimization and further characterization of these compounds will be reported separately. These data suggest that optimization of these chemicals scaffolds may produce therapeutic candidates for ALS patients.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Drug Design , Superoxide Dismutase/metabolism , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Benzoquinones/pharmacology , Cell Death/drug effects , Cytoprotection , Drug Evaluation, Preclinical , High-Throughput Screening Assays , Humans , Lactams, Macrocyclic/pharmacology , Leupeptins/pharmacology , Macrolides/pharmacology , Mutant Proteins/metabolism , PC12 Cells , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Small Molecule Libraries , Superoxide Dismutase/genetics
12.
Bioorg Med Chem ; 19(1): 613-22, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21095130

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an orphan neurodegenerative disease currently without a cure. Mutations in copper/zinc superoxide dismutase 1 (SOD1) have been implicated in the pathophysiology of this disease. Using a high-throughput screening assay expressing mutant G93A SOD1, two bioactive chemical hit compounds (1 and 2), identified as arylsulfanyl pyrazolones, were identified. The structural optimization of this scaffold led to the generation of a more potent analogue (19) with an EC(50) of 170nM. To determine the suitability of this class of compounds for further optimization, 1 was subjected to a battery of pharmacokinetic assays; most of the properties of 1 were good for a screening hit, except it had a relatively rapid clearance and short microsomal half-life stability. Compound 2 was found to be blood-brain barrier penetrating with a brain/plasma ratio=0.19. The optimization of this class of compounds could produce novel therapeutic candidates for ALS patients.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Enzyme Inhibitors/pharmacology , Pyrazolones/pharmacology , Superoxide Dismutase/antagonists & inhibitors , Animals , Humans , Magnetic Resonance Spectroscopy , Mice , Spectrometry, Mass, Electrospray Ionization , Superoxide Dismutase/genetics
13.
Biochim Biophys Acta ; 1792(6): 506-20, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19362590

ABSTRACT

Huntington's disease (HD) is an autosomal dominant, progressive, and fatal neurodegenerative disorder caused by an expanded polyglutamine cytosine-adenine-guanine repeat in the gene coding for the protein huntingtin. Despite great progress, a direct causative pathway from the HD gene mutation to neuronal dysfunction and death has not yet been established. One important advance in understanding the pathogenic mechanisms of this disease has been the development of multiple murine models that replicate many of the clinical, neuropathological, and molecular events in HD patients. These models have played an important role in providing accurate and experimentally accessible systems to study multiple aspects of disease pathogenesis and to test potential therapeutic treatment strategies. Understanding how disease processes interrelate has become important in identifying a pharmacotherapy in HD and in the design of clinical trials. A review of the current state of HD mouse models and their successes in elucidating disease pathogenesis are discussed. There is no clinically proven treatment for HD that can halt or ameliorate the inexorable disease progression. As such, a guide to assessing studies in mouse models and salient issues related to translation from mice to humans are included.


Subject(s)
Disease Models, Animal , Huntington Disease/etiology , Animals , Biomarkers/analysis , Drug Evaluation, Preclinical , Energy Metabolism , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/therapy , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurotoxins/metabolism , Nuclear Proteins/genetics , Treatment Outcome
14.
Hum Mol Genet ; 17(12): 1774-82, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18319327

ABSTRACT

Chromatin remodeling is tightly controlled under physiological conditions. Alterations in chromatin structure are involved in the pathogenesis of neuronal systems. We found that the monoallelic deletion of CREB binding protein (CBP) results in the induction of ERG-associated protein with SET domain (ESET) and increases trimethylation of histone H3 (K9) and condensation of pericentromeric heterochromatin structure in neurons. Nested deletion and mutational analysis of the ESET promoter further demonstrated that the Ets-2 transcription factor regulates transcriptional activity of the ESET gene. In CBP+/- mice, Ets-2 occupancy in the ESET promoter DNA was markedly elevated. Our results suggest that CBP is a transcriptional repressor of ESET gene expression by limiting Ets-2 transcriptional activity, while CBP siRNA enhances basal and Ets-2-dependent ESET transcriptional activity. Altered expression of the ESET gene and hypertrimethylation of H3 (K9) correlate with striatal neuron atrophy and dysfunction in CBP+/- mice. These results establish an alternative pathway that loss of CBP leads to the pericentric heterochromatin condensation through ESET expression and trimethylation of H3 (K9).


Subject(s)
CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Heterochromatin/metabolism , Histones/metabolism , Neurons/metabolism , Protein Methyltransferases/metabolism , Animals , Gene Deletion , Histone-Lysine N-Methyltransferase , Methylation , Mice , Mice, Inbred C57BL
15.
Mov Disord ; 25(12): 1924-8, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20669312

ABSTRACT

Coenzyme Q10 (CoQ(10)), a potential neuroprotective compound, was previously investigated at a dosage of 600 mg/day in Huntington's disease (HD) patients and demonstrated a trend toward slowing disease progression. Higher CoQ(10) dosages may prove beneficial. We investigated the tolerability and blood levels associated with 1,200, 2,400, and 3,600 mg/day of CoQ(10) in HD and healthy subjects. Twenty-eight subjects (20 HD, 8 healthy) enrolled in a 20-week open-label trial. Subjects started on 1,200 mg/day of CoQ(10), increasing every 4 weeks by 1,200 mg to a maximum dosage of 3,600 mg/day. Monthly evaluations included review of adverse events and CoQ(10) blood levels. Twenty-three subjects (82%) achieved the target dosage of 3,600 mg/day. Six subjects (2 healthy, 4 HD) withdrew prematurely (gastrointestinal (GI) symptoms in 3, worsening HD in 2, and 1 because of a fall). All three serious adverse events occurred in a single subject, and were deemed unrelated to CoQ(10). The most common adverse events seen were GI symptoms. Mean (± SD) CoQ10 blood levels achieved over the course of the trial were as follows: 1.26 ± 1.27 µg/mL (baseline, n = 28), 5.59 ± 2.24 µg/mL (1,200 mg/day, week 4, n = 26), 6.38 ± 3.25 µg/mL (2,400 mg/day, week 8, n = 25), 7.49 ± 4.09 µg/mL (3,600 mg/day, week 12, n = 23), and 6.78 ± 3.36 µg/mL (3,600 mg/day, week 20, n = 20). CoQ(10) was well tolerated with over 80% of subjects achieving the target dosage. Dosages of 2,400 mg/day may provide the best balance between tolerability and blood level achieved. Further studies examining the efficacy of 2,400 mg/day are planned.


Subject(s)
Huntington Disease/drug therapy , Ubiquinone/analogs & derivatives , Analysis of Variance , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug-Related Side Effects and Adverse Reactions , Female , Humans , Male , Treatment Outcome , Ubiquinone/administration & dosage , Ubiquinone/adverse effects , Ubiquinone/therapeutic use
16.
FASEB J ; 23(6): 1739-49, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19179380

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an adult-onset neurodegenerative disease characterized by selective degeneration of motor neurons and glial activation. Cell-specific transcriptional regulation induced by oxidative stress may contribute to the survival and activation of astrocytes in the face of motor neuron death. In the present study, we demonstrate an age-dependent increase in Bcl-xL and Ets-2 immunoreactivity that correlates with an increase of glial fibrillary acidic protein (GFAP)-positive cells in the ventral horn of the spinal cord in both ALS transgenic mice [mutant SOD1 (G93A)] and affected humans. Chromatin immunoprecipitation (ChIP) analysis verified that Ets-2 preferentially occupies the Ets-2 binding element in the promoter of Bcl-xL in primary astrocytes under oxidative stress conditions as well as in G93A spinal cords. Ets-2 small-interfering RNA down-regulated the transcriptional activity of Bcl-xL. In primary glial cultures, Bcl-xL overexpression and mutant SOD1 (G93A) both conferred resistance to oxidative stress-induced cell death. Our findings suggest that Ets-2 transcription factor activation of Bcl-xL gene may protect glia from constitutive oxidative stress that is thought to be a key mechanism contributing to the pathogenesis of ALS. This survival pathway may contribute to the glial survival and activation seen in the spinal cord of ALS patients.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Cell Survival/physiology , Neuroglia/physiology , Proto-Oncogene Protein c-ets-2/metabolism , bcl-X Protein/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Male , Mice , Mice, Transgenic , Motor Neurons/cytology , Motor Neurons/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Neuroglia/cytology , Oxidative Stress , Promoter Regions, Genetic , Proto-Oncogene Protein c-ets-2/genetics , RNA Interference , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , bcl-X Protein/genetics
17.
Amyotroph Lateral Scler ; 11(6): 520-30, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20565334

ABSTRACT

There is strong evidence from studies in humans and animal models to suggest the involvement of energy metabolism defects in neurodegenerative diseases. Uridine, a pyrimidine nucleoside, has been suggested to be neuroprotective in neurological disorders by improving bioenergetic effects, increasing ATP levels and enhancing glycolytic energy production. We assessed whether uridine treatment extended survival and improved the behavioral and neuropathological phenotype observed in G93A-ALS mice. In vitro and in vivo pharmacokinetic analyses in mutant SOD models provided optimal dose and assurance that uridine entered the brain. A dose-ranging efficacy trial in G93A mice was performed using survival, body weight, open-field analysis, and neuropathology as outcome measures. Urinary levels of 8-hydroxy-2'-deoxyguanosine, identifying DNA oxidative damage, were measured and used as a pharmacodynamic biomarker. Uridine administration significantly extended survival in a dose-dependent manner in G93A mice, while improving the behavioral and neuropathological phenotype. Uridine increased survival by 17.4%, ameliorated body weight loss, enhanced motor performance, reduced gross lumbar and ventral horn atrophy, attenuated lumbar ventral horn neuronal cell death, and decreased reactive astrogliosis. Consistent with a therapeutic effect, uridine significantly reduced urinary 8-hydroxy-2'-deoxyguanosine in G93A mice. These data suggest that uridine may be a therapeutic candidate in ALS patients.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Neuroprotective Agents/therapeutic use , Superoxide Dismutase/metabolism , Uridine/therapeutic use , 8-Hydroxy-2'-Deoxyguanosine , Amyotrophic Lateral Sclerosis/genetics , Animals , Anterior Horn Cells/drug effects , Anterior Horn Cells/metabolism , Anterior Horn Cells/pathology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Body Weight/drug effects , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/urine , Disease Models, Animal , Dose-Response Relationship, Drug , Energy Metabolism/physiology , Humans , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroprotective Agents/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley , Spinal Cord/cytology , Spinal Cord/drug effects , Spinal Cord/pathology , Superoxide Dismutase/genetics , Survival Rate , Uridine/pharmacology
18.
J Neurosci ; 28(38): 9473-85, 2008 Sep 17.
Article in English | MEDLINE | ID: mdl-18799679

ABSTRACT

Release of mitochondrial cytochrome c resulting in downstream activation of cell death pathways has been suggested to play a role in neurologic diseases featuring cell death. However, the specific biologic importance of cytochrome c release has not been demonstrated in Huntington's disease (HD). To evaluate the role of cytochrome c release, we screened a drug library to identify new inhibitors of cytochrome c release from mitochondria. Drugs effective at the level of purified mitochondria were evaluated in a cellular model of HD. As proof of principle, one drug was chosen for in depth evaluation in vitro and a transgenic mouse model of HD. Our findings demonstrate the utility of mitochondrial screening to identify inhibitors of cell death and provide further support for the important functional role of cytochrome c release in HD. Given that many of these compounds have been approved by the Food and Drug Administration for clinical usage and cross the blood-brain barrier, these drugs may lead to trials in patients.


Subject(s)
Brain/drug effects , Cytochromes c/antagonists & inhibitors , Huntington Disease/drug therapy , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Animals , Brain/metabolism , Brain/physiopathology , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrase Inhibitors/therapeutic use , Caspases/drug effects , Caspases/metabolism , Cell Death/drug effects , Cell Death/physiology , Cell Line, Transformed , Cytochromes c/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Huntington Disease/metabolism , Huntington Disease/physiopathology , Longevity/drug effects , Longevity/physiology , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Methazolamide/pharmacology , Methazolamide/therapeutic use , Mice , Mice, Transgenic , Mitochondria/metabolism , Neuroprotective Agents/therapeutic use , Treatment Outcome
19.
Biochim Biophys Acta ; 1782(3): 151-62, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18206128

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder for which there is no current therapy preventing cumulative neuronal loss. There is substantial evidence that mitochondrial dysfunction, oxidative stress, and associated caspase activity underlie the neurodegeneration observed. One potential drug therapy is the potent free radical scavenger and antioxidant cystamine, which has demonstrated significant clinical potential in models of neurodegenerative disorders and human neurological disease. This study examined the oral efficacy of cystamine in the MPTP and 6-hydroxydopamine neurotoxin models of PD. The neuroprotective effects of cystamine treatment significantly ameliorated nigral neuronal loss, preserved striatal dopaminergic projections, and improved striatal dopamine and metabolite levels, as compared to MPTP alone. Cystamine normalized striatal 8-hydroxy-2'-deoxyguanosine levels and ATP concentrations, consistent with reduced oxidative stress and improved mitochondrial function. Cystamine also protected against MPTP-induced mitochondrial loss, as identified by mitochondrial heat shock protein 70 and superoxide dismutase 2, with concomitant reductions in cytochrome c and caspase-3 activities. The neuroprotective value of cystamine was confirmed in the 6-hydroxydopamine model. Together these findings show cystamine's therapeutic benefit to reduce neuronal loss through attenuation of oxidative stress and mitochondrial dysfunction, providing the rationale for human clinical trials in PD patients.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Cystamine/therapeutic use , Disease Models, Animal , Mitochondrial Diseases/drug therapy , Neurotoxins , Oxidative Stress/drug effects , Oxidopamine , Parkinson Disease/drug therapy , Animals , Brain/cytology , Brain/metabolism , Drug Evaluation, Preclinical , Male , Parkinson Disease/etiology , Parkinson Disease/pathology , Parkinson Disease/physiopathology
20.
J Neurochem ; 109(5): 1427-39, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19476553

ABSTRACT

Coenzyme Q(10) (CoQ(10)) and creatine are promising agents for neuroprotection in neurodegenerative diseases via their effects on improving mitochondrial function and cellular bioenergetics and their properties as antioxidants. We examined whether a combination of CoQ(10) with creatine can exert additive neuroprotective effects in a MPTP mouse model of Parkinson's disease, a 3-NP rat model of Huntington's disease (HD) and the R6/2 transgenic mouse model of HD. The combination of the two agents produced additive neuroprotective effects against dopamine depletion in the striatum and loss of tyrosine hydroxylase neurons in the substantia nigra pars compacta (SNpc) following chronic subcutaneous administration of MPTP. The combination treatment resulted in significant reduction in lipid peroxidation and pathologic alpha-synuclein accumulation in the SNpc neurons of the MPTP-treated mice. We also observed additive neuroprotective effects in reducing striatal lesion volumes produced by chronic subcutaneous administration of 3-NP to rats. The combination treatment showed significant effects on blocking 3-NP-induced impairment of glutathione homeostasis and reducing lipid peroxidation and DNA oxidative damage in the striatum. Lastly, the combination of CoQ(10) and creatine produced additive neuroprotective effects on improving motor performance and extending survival in the transgenic R6/2 HD mice. These findings suggest that combination therapy using CoQ(10) and creatine may be useful in the treatment of neurodegenerative diseases such as Parkinson's disease and HD.


Subject(s)
Creatine/therapeutic use , Huntington Disease/drug therapy , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Ubiquinone/analogs & derivatives , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , 8-Hydroxy-2'-Deoxyguanosine , Analysis of Variance , Animals , Chromatography, High Pressure Liquid/methods , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Disease Models, Animal , Dopamine/metabolism , Drug Therapy, Combination , Glutathione/metabolism , Glutathione Disulfide/metabolism , Huntington Disease/chemically induced , Lipid Peroxidation/drug effects , Male , Malondialdehyde/metabolism , Mice , Mice, Inbred C57BL , Nitro Compounds , Parkinson Disease/etiology , Propionates , Rats , Rats, Inbred Lew , Tyrosine 3-Monooxygenase/metabolism , Ubiquinone/therapeutic use , alpha-Synuclein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL