Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Hum Mol Genet ; 25(8): 1619-36, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26908599

ABSTRACT

Identifying molecular drivers of pathology provides potential therapeutic targets. Differentiating between drivers and coincidental molecular alterations presents a major challenge. Variation unrelated to pathology further complicates transcriptomic, proteomic and metabolomic studies which measure large numbers of individual molecules. To overcome these challenges towards the goal of determining drivers of Huntington's disease (HD), we generated an allelic series of HD knock-in mice with graded levels of phenotypic severity for comparison with molecular alterations. RNA-sequencing analysis of this series reveals high numbers of transcripts with level alterations that do not correlate with phenotypic severity. These discorrelated molecular changes are unlikely to be drivers of pathology allowing an exclusion-based strategy to provide a short list of driver candidates. Further analysis of the data shows that a majority of transcript level changes in HD knock-in mice involve alteration of the rate of mRNA processing and/or degradation rather than solely being due to alteration of transcription rate. The overall strategy described can be applied to assess the influence of any molecular change on pathology for diseases where different mutations cause graded phenotypic severity.


Subject(s)
Gene Expression Regulation , Gene Knock-In Techniques/methods , Huntington Disease/pathology , RNA, Messenger/metabolism , Alleles , Animals , Disease Models, Animal , Humans , Huntington Disease/genetics , Mice , Phenotype , Sequence Analysis, RNA
2.
Am J Physiol Gastrointest Liver Physiol ; 306(4): G265-77, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24356880

ABSTRACT

Chronic ethanol consumption increases sensitivity of the mitochondrial permeability transition (MPT) pore induction in liver. Ca(2+) promotes MPT pore opening, and genetic ablation of cyclophilin D (CypD) increases the Ca(2+) threshold for the MPT. We used wild-type (WT) and CypD-null (CypD(-/-)) mice fed a control or an ethanol-containing diet to investigate the role of the MPT in ethanol-mediated liver injury. Ca(2+)-mediated induction of the MPT and mitochondrial respiration were measured in isolated liver mitochondria. Steatosis was present in WT and CypD(-/-) mice fed ethanol and accompanied by increased terminal deoxynucleotidyl transferase dUTP-mediated nick-end label-positive nuclei. Autophagy was increased in ethanol-fed WT mice compared with ethanol-fed CypD(-/-) mice, as reflected by an increase in the ratio of microtubule protein 1 light chain 3B II to microtubule protein 1 light chain 3B I. Higher levels of p62 were measured in CypD(-/-) than WT mice. Ethanol decreased mitochondrial respiratory control ratios and select complex activities in WT and CypD(-/-) mice. Ethanol also increased CypD protein in liver of WT mice. Mitochondria from control- and ethanol-fed WT mice were more sensitive to Ca(2+)-mediated MPT pore induction than mitochondria from their CypD(-/-) counterparts. Mitochondria from ethanol-fed CypD(-/-) mice were also more sensitive to Ca(2+)-induced swelling than mitochondria from control-fed CypD(-/-) mice but were less sensitive than mitochondria from ethanol-fed WT mice. In summary, CypD deficiency was associated with impaired autophagy and did not prevent ethanol-mediated steatosis. Furthermore, increased MPT sensitivity was observed in mitochondria from ethanol-fed WT and CypD(-/-) mice. We conclude that chronic ethanol consumption likely lowers the threshold for CypD-regulated and -independent characteristics of the ethanol-mediated MPT pore in liver mitochondria.


Subject(s)
Ethanol , Liver Diseases, Alcoholic/metabolism , Liver/metabolism , Mitochondria, Liver/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Animals , Autophagy , Calcium Signaling , Cell Respiration , Peptidyl-Prolyl Isomerase F , Cyclophilins/deficiency , Cyclophilins/genetics , Disease Models, Animal , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/metabolism , Genotype , Liver/pathology , Liver Diseases, Alcoholic/etiology , Liver Diseases, Alcoholic/genetics , Liver Diseases, Alcoholic/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Mitochondria, Liver/pathology , Mitochondrial Permeability Transition Pore , Mitochondrial Swelling , Phenotype , Time Factors
3.
Neurobiol Dis ; 58: 192-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23756199

ABSTRACT

Huntington's disease (HD) causes preferential loss of a subset of neurons in the brain although the huntingtin protein is expressed broadly in various neural cell types, including astrocytes. Glutamate-mediated excitotoxicity is thought to cause selective neuronal injury, and brain astrocytes have a central role in regulating extracellular glutamate. To determine whether full-length mutant huntingtin expression causes a cell-autonomous phenotype and perturbs astrocyte gliotransmitter release, we studied cultured cortical astrocytes from BACHD mice. Here, we report augmented glutamate release through Ca(2+)-dependent exocytosis from BACHD astrocytes. Although such release is usually dependent on cytosolic Ca(2+) levels, surprisingly, we found that BACHD astrocytes displayed Ca(2+) dynamics comparable to those in wild type astrocytes. These results point to a possible involvement of other factors in regulating Ca(2+)-dependent/vesicular release of glutamate from astrocytes. We found a biochemical footprint that would lead to increased availability of cytosolic glutamate in BACHD astrocytes: i) augmented de novo glutamate synthesis due to an increase in the level of the astrocyte specific mitochondrial enzyme pyruvate carboxylase; and ii) unaltered conversion of glutamate to glutamine, as there were no changes in the expression level of the astrocyte specific enzyme glutamine synthetase. This work identifies a new mechanism in astrocytes that could lead to increased levels of extracellular glutamate in HD and thus may contribute to excitotoxicity in this devastating disease.


Subject(s)
Astrocytes/cytology , Astrocytes/metabolism , Calcium/metabolism , Glutamic Acid/metabolism , Huntington Disease/pathology , Amino Acid Transport Systems, Acidic/genetics , Amino Acid Transport Systems, Acidic/metabolism , Animals , Animals, Newborn , Cells, Cultured , Cerebral Cortex/pathology , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Huntingtin Protein , Huntington Disease/genetics , Mice , Mutation/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Physical Stimulation , Pyruvate Carboxylase/metabolism , Subcellular Fractions/metabolism , Transfection
4.
Exp Neurol ; 237(1): 78-89, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22698685

ABSTRACT

Huntington's disease (HD) is a devastating autosomal-dominant neurodegenerative disorder initiated by an abnormally expanded polyglutamine in the huntingtin protein. Determining the contribution of specific factors to the pathogenesis of HD should provide rational targets for therapeutic intervention. One suggested contributor is the type 2 transglutaminase (TG2), a multifunctional calcium dependent enzyme. A role for TG2 in HD has been suggested because a polypeptide-bound glutamine is a rate-limiting factor for a TG2-catalyzed reaction, and TG2 can cross-link mutant huntingtin in vitro. Further, TG2 is up regulated in brain areas affected in HD. The objective of this study was to further examine the contribution of TG2 as a potential modifier of HD pathogenesis and its validity as a therapeutic target in HD. In particular our goal was to determine whether an increase in TG2 level, as documented in human HD brains, modulates the well-characterized phenotype of the R6/2 HD mouse model. To accomplish this objective a genetic cross was performed between R6/2 mice and an established transgenic mouse line that constitutively expresses human TG2 (hTG2) under control of the prion promoter. Constitutive expression of hTG2 did not affect the onset and progression of the behavioral and neuropathological HD phenotype of R6/2 mice. We found no alterations in body weight changes, rotarod performances, grip strength, overall activity, and no significant effect on the neuropathological features of R6/2 mice. Overall the results of this study suggest that an increase in hTG2 expression does not significantly modify the pathology of HD.


Subject(s)
Huntington Disease/enzymology , Huntington Disease/genetics , Phenotype , Transglutaminases/biosynthesis , Transglutaminases/genetics , Age of Onset , Animals , Behavior, Animal/physiology , Disease Models, Animal , Disease Progression , Female , GTP-Binding Proteins , Humans , Huntington Disease/pathology , Male , Mice , Mice, Inbred C57BL , Protein Glutamine gamma Glutamyltransferase 2 , Random Allocation , Transglutaminases/physiology
5.
PLoS One ; 6(7): e21720, 2011.
Article in English | MEDLINE | ID: mdl-21799745

ABSTRACT

Disruption of 14-3-3 function by alpha-synuclein has been implicated in Parkinson's disease. As 14-3-3s are important regulators of cell death pathways, disruption of 14-3-3s could result in the release of pro-apoptotic factors, such as Bax. We have previously shown that overexpression of 14-3-3θ reduces cell loss in response to rotenone and MPP(+) in dopaminergic cell culture and reduces cell loss in transgenic C. elegans that overexpress alpha-synuclein. In this study, we investigate the mechanism for 14-3-3θ's neuroprotection against rotenone toxicity. While 14-3-3s can inhibit many pro-apoptotic factors, we demonstrate that inhibition of one factor in particular, Bax, is important to 14-3-3s' protection against rotenone toxicity in dopaminergic cells. We found that 14-3-3θ overexpression reduced Bax activation and downstream signaling events, including cytochrome C release and caspase 3 activation. Pharmacological inhibition or shRNA knockdown of Bax provided protection against rotenone, comparable to 14-3-3θ's neuroprotective effects. A 14-3-3θ mutant incapable of binding Bax failed to protect against rotenone. These data suggest that 14-3-3θ's neuroprotective effects against rotenone are at least partially mediated by Bax inhibition and point to a potential therapeutic role of 14-3-3s in Parkinson's disease.


Subject(s)
14-3-3 Proteins/metabolism , Apoptosis/drug effects , Neurotoxins/toxicity , Parkinson Disease/pathology , Rotenone/toxicity , bcl-2-Associated X Protein/antagonists & inhibitors , Animals , Cell Line , Gene Expression Regulation/drug effects , Immunoprecipitation , Membrane Potential, Mitochondrial/drug effects , Parkinson Disease/metabolism , Signal Transduction/drug effects , bcl-2-Associated X Protein/metabolism
6.
Neurobiol Dis ; 41(1): 11-22, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20732421

ABSTRACT

The presence of aggregates of abnormally expanded polyglutamine (polyQ)-containing proteins are a pathological hallmark of a number of neurodegenerative diseases including Huntington's disease (HD) and spinocerebellar ataxia-3 (SCA3). Previous studies in cellular, Drosophila, and mouse models of HD and SCA have shown that neurodegeneration can be prevented by manipulations that inhibit polyQ aggregation. We have shown that the UL97 kinase of the human cytomegalovirus (HCMV) prevents aggregation of the pp71 and pp65 viral tegument proteins. To explore whether UL97 may act as a general antiaggregation factor, we examined whether UL97 prevents aggregation of cellular non-polyQ and polyQ proteins. We report that UL97 prevents the deposition of aggregates of two non-polyQ proteins: a protein chimera (GFP170*) composed of the green fluorescent protein and a fragment of the Golgi Complex protein (GCP-170) and a chimera composed of the red fluorescent protein (RFP) fused to the Werner syndrome protein (WRN), a RecQ helicase and exonuclease involved in DNA repair. Furthermore, we show that UL97 inhibits aggregate deposition in cellular models of HD and SCA3. UL97 prevents the deposition of aggregates of the mutant huntingtin exon 1 containing 82 glutamine repeats (HttExon1-Q82) or full length ataxin-3 containing a 72 polyQ track (AT3-72Q). The kinase activity of UL97 appears critical, as the kinase-dead UL97 mutant (K335M) fails to prevent aggregate formation. We further show that UL97 disrupts nuclear PML bodies and decreases p53-mediated transcription. The universality of the antiaggregation effect of UL97 suggests that UL97 targets a key cellular factor that regulates cellular aggregation mechanisms. Our results identify UL97 as a novel means to modulate polyQ aggregation and suggest that UL97 can serve as a novel tool to probe the cellular mechanisms that contribute to the formation of aggregates in polyglutamine disorders.


Subject(s)
Cytomegalovirus/enzymology , Huntington Disease/virology , Neurons/metabolism , Peptides/antagonists & inhibitors , Peptides/metabolism , Phosphotransferases (Alcohol Group Acceptor)/physiology , Spinocerebellar Ataxias/virology , Cytomegalovirus/genetics , HeLa Cells , Humans , Huntingtin Protein , Huntington Disease/enzymology , Huntington Disease/metabolism , Mutation , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/enzymology , Neurons/virology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peptides/genetics , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/genetics , Spinocerebellar Ataxias/enzymology , Spinocerebellar Ataxias/metabolism
7.
Cell Calcium ; 49(1): 23-34, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21163525

ABSTRACT

Immunophilins are receptors for immunosuppressive drugs such as the macrolides cyclosporin A (CsA) and FK506; correspondingly these immunophilins are referred to as cyclophilins and FK506-binding proteins (FKBPs). In particular, CsA targets cyclophilin D (CypD), which can modulate mitochondrial Ca(2+) dynamics. Since mitochondria have been implicated in the regulation of astrocytic cytosolic Ca(2+) (Ca(cyt)(2+)) dynamics and consequential Ca(2+)-dependent exocytotic release of glutamate, we investigated the role of CypD in this process. Cortical astrocytes isolated from CypD deficient mice Ppif(-/-) displayed reduced mechanically induced Ca(cyt)(2+) increases, even though these cells showed augmented exocytotic release of glutamate, when compared to responses obtained from astrocytes isolated from wild-type mice. Furthermore, acute treatment with CsA to inhibit CypD modulation of mitochondrial Ca(2+) buffering, or with FK506 to inhibit FKBP12 interaction with inositol-trisphosphate receptor of the endoplasmic reticulum, led to similar reductive effects on astrocytic Ca(cyt)(2+) dynamics, but also to an enhanced Ca(2+)-dependent exocytotic release of glutamate in wild-type astrocytes. These findings point to a possible role of immunophilin signal transduction pathways in astrocytic modulation of neuronal activity at the tripartite synapse.


Subject(s)
Astrocytes/metabolism , Calcium/metabolism , Cerebral Cortex/cytology , Cyclophilins/deficiency , Glutamic Acid/metabolism , Animals , Astrocytes/drug effects , Buffers , Cells, Cultured , Peptidyl-Prolyl Isomerase F , Cyclophilins/metabolism , Cyclosporine/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Tacrolimus/pharmacology
8.
Am J Physiol Gastrointest Liver Physiol ; 299(4): G954-66, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20651005

ABSTRACT

Chronic ethanol consumption increases mitochondrial oxidative stress and sensitivity to form the mitochondrial permeability transition pore (MPTP). The mechanism responsible for increased MPTP sensitivity in ethanol-exposed mitochondria and its relation to mitochondrial Ca(2+) handling is unknown. Herein, we investigated whether increased sensitivity to MPTP induction in liver mitochondria from ethanol-fed rats compared with controls is related to an ethanol-dependent change in mitochondrial Ca(2+) accumulation. Liver mitochondria were isolated from control and ethanol-fed rats, and Ca(2+)-mediated induction of the MPTP and mitochondrial Ca(2+) retention capacity were measured. Levels of proposed MPTP proteins as well as select pro- and antiapoptotic proteins were measured along with gene expression. We observed increased steatosis and TUNEL-stained nuclei in liver of ethanol-fed rats compared with controls. Liver mitochondria from ethanol-fed rats had increased levels of proapoptotic Bax protein and reduced Ca(2+) retention capacity compared with control mitochondria. We observed increased cyclophilin D (Cyp D) gene expression in liver and protein in mitochondria from ethanol-fed animals compared with controls, whereas there was no change in the adenine nucleotide translocase and voltage-dependent anion channel. Together, these results suggest that enhanced sensitivity to Ca(2+)-mediated MPTP induction may be due, in part, to higher Cyp D levels in liver mitochondria from ethanol-fed rats. Therefore, therapeutic strategies aimed at normalizing Cyp D levels may be beneficial in preventing ethanol-dependent mitochondrial dysfunction and liver injury.


Subject(s)
Calcium/metabolism , Cyclophilins/metabolism , Ethanol/adverse effects , Liver/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Animals , Peptidyl-Prolyl Isomerase F , Gene Expression Regulation/drug effects , Liver/metabolism , Male , Mitochondria, Liver/drug effects , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/physiology , Mitochondrial Permeability Transition Pore , Permeability , Rats , Rats, Sprague-Dawley
9.
Hum Mol Genet ; 19(19): 3702-20, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20616151

ABSTRACT

The aggregation of mutant polyglutamine (polyQ) proteins has sparked interest in the role of protein quality-control pathways in Huntington's disease (HD) and related polyQ disorders. Employing a novel knock-in HD mouse model, we provide in vivo evidence of early, sustained alterations of autophagy in response to mutant huntingtin (mhtt). The HdhQ200 knock-in model, derived from the selective breeding of HdhQ150 knock-in mice, manifests an accelerated and more robust phenotype than the parent line. Heterozygous HdhQ200 mice accumulate htt aggregates as cytoplasmic aggregation foci (AF) as early as 9 weeks of age and striatal neuronal intranuclear inclusions (NIIs) by 20 weeks. By 40 weeks, striatal AF are perinuclear and immunoreactive for ubiquitin and the autophagosome marker LC3. Striatal NIIs accumulate earlier in HdhQ200 mice than in HdhQ150 mice. The earlier appearance of aggregate pathology in HdhQ200 mice is paralleled by earlier and more rapidly progressive motor deficits: progressive imbalance and decreased motor coordination by 50 weeks, gait deficits by 60 weeks and gross motor impairment by 80 weeks of age. At 80 weeks, heterozygous HdhQ200 mice exhibit striatal and cortical astrogliosis and a approximately 50% reduction in striatal dopamine receptor binding. Increased LC3-II protein expression, which is noted early and sustained throughout the disease course, is paralleled by increased expression of the autophagy-related protein, p62. Early and sustained expression of autophagy-related proteins in this genetically precise mouse model of HD suggests that the alteration of autophagic flux is an important and early component of the neuronal response to mhtt.


Subject(s)
Autophagy , Gene Knock-In Techniques , Huntington Disease/genetics , Huntington Disease/pathology , Animals , Biomarkers/metabolism , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Health , Heterozygote , Huntington Disease/physiopathology , Mice , Microtubule-Associated Proteins/metabolism , Motor Activity , Mutation/genetics , Neostriatum/pathology , Neostriatum/physiopathology , Neostriatum/ultrastructure , Neurons/pathology , Neurons/ultrastructure , Protein Structure, Quaternary , Protein Transport , Receptors, Dopamine/metabolism , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/genetics , Ubiquitin/metabolism
10.
Hum Mol Genet ; 19(17): 3354-71, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20558522

ABSTRACT

Huntington's disease (HD) is an incurable autosomal-dominant neurodegenerative disorder initiated by an abnormally expanded polyglutamine domain in the huntingtin protein. It is proposed that abnormal mitochondrial Ca2+ capacity results in an increased susceptibility to mitochondrial permeability transition (MPT) induction that may contribute significantly to HD pathogenesis. The in vivo contribution of these hypothesized defects remains to be elucidated. In this proof-of-principle study, we examined whether increasing mitochondrial Ca2+ capacity could ameliorate the well-characterized phenotype of the R6/2 transgenic mouse model. Mouse models lacking cyclophilin D demonstrate convincingly that cyclophilin D is an essential component and a key regulator of MPT induction. Mitochondria of cyclophilin D knockout mice are particularly resistant to Ca2+ overload. We generated R6/2 mice with normal, reduced or absent cyclophilin D expression and examined the effect of increasing mitochondrial Ca2+ capacity on the behavioral and neuropathological features of the R6/2 model. A predicted outcome of this approach was the finding that cyclophilin D deletion enhanced the R6/2 brain mitochondria Ca2+ capacity significantly. Increased neuronal mitochondrial Ca2+ capacity failed to ameliorate either the behavioral and neuropathological features of R6/2 mice. We found no alterations in body weight changes, lifespan, RotaRod performances, grip strength, overall activity and no significant effect on the neuropathological features of R6/2 mice. The results of this study demonstrate that increasing neuronal mitochondrial Ca2+-buffering capacity is not beneficial in the R6/2 mouse model of HD.


Subject(s)
Calcium/metabolism , Disease Models, Animal , Huntington Disease/metabolism , Mice , Mitochondria/metabolism , Animals , Biological Transport , Peptidyl-Prolyl Isomerase F , Cyclophilins/genetics , Cyclophilins/metabolism , Female , Humans , Huntington Disease/drug therapy , Huntington Disease/genetics , Huntington Disease/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitochondria/genetics , Neurons/metabolism
11.
J Neurosci Res ; 85(8): 1774-88, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17455294

ABSTRACT

Huntington's disease (HD) is a progressive, autosomal dominant neurodegenerative disease caused by an abnormally expanded CAG repeat in the HD gene. Ubiquitylated aggregates containing mutant huntingtin protein in neurons are hallmarks of HD. Misfolded mutant huntingtin monomers, oligomers, or aggregates may be a result of, and cause, ubiquitin- proteasome dysfunction. To investigate the ubiquitin-proteasome system we designed a series of firefly luciferase reporters targeted selectively to different points along this pathway. These reporters were used to monitor ubiquitin-proteasome system function in a striatal cell culture model of HD. Ubiquitylation processes were not reduced in mutant huntingtin cells but recognition or degradation of ubiquitylated substrates was decreased. We also found mutant huntingtin expressing cells had slight but significant decreases in chymotrypsin-like and caspase-like activities, and an unexpected increase in trypsin-like activity of the proteasome core. General proteasome core inhibitors, as well as selective caspase-like activity inhibitors, were less effective in mutant cells. Finally, treatment with 3-nitropropionic acid, a succinate dehydrogenase inhibitor, had opposite effects on the ubiquitin-proteasome system with activation in wild-type and decreased activity in mutant huntingtin expressing cells. The results of these experiments show clearly selective disruption of the ubiquitin-proteasome system in this cell culture model of HD. The high throughput tools that we have designed and optimized will also be useful in identifying compounds that alter ubiquitin-proteasome system function and to investigate other neurodegenerative diseases such Alzheimer's disease and Parkinson's disease.


Subject(s)
Corpus Striatum/enzymology , Huntington Disease/enzymology , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Cell Line , Corpus Striatum/pathology , Fluorescent Dyes/metabolism , Genes, Reporter , Humans , Huntingtin Protein , Huntington Disease/pathology , Luciferases, Firefly/genetics , Mutation , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nitro Compounds/pharmacology , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Oligopeptides/metabolism , Propionates/pharmacology , Proteasome Endopeptidase Complex/genetics , Proteasome Inhibitors , Succinate Dehydrogenase/antagonists & inhibitors , Ubiquitin/genetics
12.
Eur J Neurosci ; 23(7): 1701-10, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16623826

ABSTRACT

Abstract Cystamine significantly improved motor deficits and extended survival in mouse models of Huntington's disease (HD); however, the precise mechanism(s) by which cystamine and the related compound cysteamine are beneficial remain to be elucidated. Using clonal striatal cell lines from wild-type (STHdhQ7/HdhQ7) and mutant huntingtin knock-in (STHdhQ111/HdhQ111) mice, we have tested the hypothesis that cystamine and cysteamine could be beneficial by preventing the depolarization of mitochondria in cell cultures. Treatment with 3-nitroproprionic acid (3-NP), a mitochondrial complex II inhibitor, induces mitochondrial depolarization and cell death of mutant HD striatal cells but not of wild-type cells. The 3-NP-mediated decrease in the mitochondrial membrane potential was attenuated by 50 microm cystamine and completely inhibited by 250 microm cystamine. Similar results were obtained using cysteamine (50-500 microm). In addition, both cystamine and cysteamine significantly attenuated the 3-NP-induced cell death. Treatment of mutant HD striatal cells with 3-NP resulted in a robust decrease in the cellular and mitochondrial levels of glutathione (GSH) compared with cells exposed to the vehicle alone. Pre-treatment of the cells with cystamine and cysteamine completely prevented the 3-NP-mediated decrease in cellular and mitochondrial GSH levels. Incubation with L-buthionine (S,R) sulfoximine (BSO) 250 microm in combination with cystamine (250 microm) or cysteamine (250 microm) prior to being treated with 3-NP completely prevented the beneficial effects of cystamine and cysteamine on the 3-NP-mediated mitochondrial depolarization. These results demonstrate that cystamine and cysteamine prevent the 3-NP-induced mitochondrial depolarization of HD striatal cell cultures.


Subject(s)
Cystamine/pharmacology , Cysteamine/pharmacology , Electron Transport Complex II/antagonists & inhibitors , Mitochondria/drug effects , Nerve Tissue Proteins/genetics , Nitro Compounds/pharmacology , Nuclear Proteins/genetics , Propionates/pharmacology , Animals , Cell Death/drug effects , Cell Line , Clone Cells , Corpus Striatum/cytology , Glutathione/metabolism , Huntingtin Protein , Membrane Potentials , Mice , Mice, Mutant Strains , Mitochondria/metabolism , Mitochondria/physiology , Mitochondrial Membranes/physiology , Mutation
13.
Hum Mol Genet ; 14(19): 2871-80, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16115812

ABSTRACT

The 'expanded' HD CAG repeat that causes Huntington's disease (HD) encodes a polyglutamine tract in huntingtin, which first targets the death of medium-sized spiny striatal neurons. Mitochondrial energetics, related to N-methyl-d-aspartate (NMDA) Ca2+-signaling, has long been implicated in this neuronal specificity, implying an integral role for huntingtin in mitochondrial energy metabolism. As a genetic test of this hypothesis, we have looked for a relationship between the length of the HD CAG repeat, expressed in endogenous huntingtin, and mitochondrial ATP production. In STHdhQ111 knock-in striatal cells, a juvenile onset HD CAG repeat was associated with low mitochondrial ATP and decreased mitochondrial ADP-uptake. This metabolic inhibition was associated with enhanced Ca2+-influx through NMDA receptors, which when blocked resulted in increased cellular [ATP/ADP]. We then evaluated [ATP/ADP] in 40 human lymphoblastoid cell lines, bearing non-HD CAG lengths (9-34 units) or HD-causing alleles (35-70 units). This analysis revealed an inverse association with the longer of the two allelic HD CAG repeats in both the non-HD and HD ranges. Thus, the polyglutamine tract in huntingtin appears to regulate mitochondrial ADP-phosphorylation in a Ca2+-dependent process that fulfills the genetic criteria for the HD trigger of pathogenesis, and it thereby determines a fundamental biological parameter--cellular energy status, which may contribute to the exquisite vulnerability of striatal neurons in HD. Moreover, the evidence that this polymorphism can determine energy status in the non-HD range suggests that it should be tested as a potential physiological modifier in both health and disease.


Subject(s)
Calcium Signaling , DNA Repeat Expansion , Huntington Disease/genetics , Huntington Disease/metabolism , Mitochondria/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Age of Onset , Alleles , Cells, Cultured , Energy Metabolism/genetics , Humans , Huntingtin Protein , Peptides/genetics , Phosphorylation , Polymorphism, Genetic , Receptors, N-Methyl-D-Aspartate/metabolism
14.
Neurosci Lett ; 386(1): 63-8, 2005 Sep 23.
Article in English | MEDLINE | ID: mdl-15993538

ABSTRACT

Huntington's disease (HD) is a progressive neurodegenerative disease characterized by a severe neuronal loss that occurs primarily in the neostriatum. It has been postulated that mitochondria dysfunction and oxidative stress may play significant roles in the etiology of the disease. Indeed, markers of oxidative stress damage have been detected in the brains of HD patients and in mouse models of HD. In this study, we evaluate the changes in the levels of the potent, endogenous antioxidant glutathione and enzymes involved in its metabolism or recycling in the cortex and striatum of an extensively studied HD mouse model (R6/2). In both cortex and striatum, the levels of cellular glutathione were not significantly different in the R6/2 mice when compared with littermate wild type controls. Remarkably, the levels of glutathione were significantly increased in mitochondria isolated from the cortex and striatum of R6/2 mice when compared with wild type control mice. This specific increase in the levels of glutathione in mitochondria suggests that a compensatory mechanism is induced in the R6/2 mice to protect against an increase in oxidative stress in mitochondria.


Subject(s)
Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Glutathione/metabolism , Huntington Disease/metabolism , Mitochondria/metabolism , Oxidative Stress/physiology , Adaptation, Physiological/physiology , Animals , Cerebral Cortex/physiopathology , Corpus Striatum/physiopathology , Disease Models, Animal , Energy Metabolism/physiology , Female , Huntington Disease/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Up-Regulation/physiology
15.
J Neurosci Methods ; 144(1): 11-7, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15848234

ABSTRACT

The inheritance of a long CAG repeat causes several late onset neurological disorders including Huntington's disease (HD). Longer CAG repeats correlate with earlier onset of HD suggesting an increased toxicity for the products of long repeat alleles. PCR based data has been used to show that HD CAG repeat expansion beyond the inherited length occurs in affected tissues indicating a possible role for somatic instability in the disease process. PCR, however, is prone to artifacts resulting from expansion of repeat sequences during amplification. We describe a method to distinguish between CAG repeat expansions that exist in vivo and those that potentially occur during PCR. The method involves size fractionation of genomic restriction fragments containing the expanded repeats followed by PCR amplification. The application of this method confirms the presence of somatic expansions in the brains of a knock-in mouse model of HD.


Subject(s)
Huntington Disease/genetics , Polymerase Chain Reaction/methods , Trinucleotide Repeat Expansion/genetics , Animals , Brain/metabolism , Chemical Fractionation/methods , DNA/isolation & purification , Disease Models, Animal , Mice , Mice, Transgenic , RNA, Messenger/analysis
16.
Hum Mol Genet ; 13(14): 1407-20, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15163634

ABSTRACT

Huntington's disease (HD) is initiated by an abnormally expanded polyglutamine stretch in the huntingtin protein, conferring a novel property on the protein that leads to the loss of striatal neurons. Defects in mitochondrial function have been implicated in the pathogenesis of HD. Here, we have examined the hypothesis that the mutant huntingtin protein may directly interact with the mitochondrion and affect its function. In human neuroblastoma cells and clonal striatal cells established from HdhQ7 (wild-type) and HdhQ111 (mutant) homozygote mouse knock-in embryos, huntingtin was present in a purified mitochondrial fraction. Subfractionation of the mitochondria and limited trypsin digestion of the organelle demonstrated that huntingtin was associated with the outer mitochondrial membrane. We further demonstrated that a recombinant truncated mutant huntingtin protein, but not a wild-type, directly induced mitochondrial permeability transition (MPT) pore opening in isolated mouse liver mitochondria, an effect that was prevented completely by cyclosporin A (CSA) and ATP. Importantly, the mutant huntingtin protein significantly decreased the Ca2+ threshold necessary to trigger MPT pore opening. We found a similar increased susceptibility to the calcium-induced MPT in liver mitochondria isolated from a knock-in HD mouse model. The mutant huntingtin protein-induced MPT pore opening was accompanied by a significant release of cytochrome c, an effect completely inhibited by CSA. These findings suggest that the development of specific MPT inhibitors may be an interesting therapeutic avenue to delay the onset of HD.


Subject(s)
Calcium/metabolism , Cytochromes c/metabolism , Ion Channels/metabolism , Mitochondria, Liver/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line , Cyclosporine/pharmacology , Humans , Huntingtin Protein , Huntington Disease/genetics , Intracellular Membranes/metabolism , Mice , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Mutation , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Recombinant Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics
17.
Hum Mol Genet ; 13(7): 669-81, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-14962977

ABSTRACT

Extensive striatal neuronal loss occurs in Huntington's disease (HD), which is caused by an expanded polyglutamine tract in huntingtin (htt). Evidence suggests that mutant htt directly or indirectly compromises mitochondrial function, contributing to the neuronal loss. To determine the role of compromised mitochondrial function in the neuronal cell death in HD, immortalized striatal cells established from Hdh(Q7) (wild-type) and Hdh(Q111) (mutant) mouse knock-in embryos were treated with 3-nitropropionic acid (3-NP), a mitochondrial complex II toxin. 3-NP treatment caused significantly greater cell death in mutant striatal cells compared with wild-type cells. In contrast, the extent of cell death induced by rotenone, a complex I inhibitor, was similar in both cell lines. Although evidence of apoptosis was present in 3-NP-treated wild-type striatal cells, it was absent in 3-NP-treated mutant cells. 3-NP treatment caused a greater loss of mitochondrial membrane potential (deltapsim) in mutant striatal cells compared with wild-type cells. Cyclosporine A, an inhibitor of mitochondrial permeability transition pore (PTP), and ruthenium red, an inhibitor of the mitochondrial calcium uniporter, both rescued mutant striatal cells from 3-NP-induced cell death and prevented the loss of deltapsim. These data show that mutant htt specifically increases cell vulnerability to mitochondrial complex II inhibition and further switched the type of cell death induced by complex II inhibition from apoptosis to a non-apoptotic form, caused by mitochondrial membrane depolarization, probably initiated by mitochondrial calcium overload and subsequent PTP opening. These findings suggest that impaired mitochondrial complex II function in HD may contribute to non-apoptotic neuronal cell death.


Subject(s)
Apoptosis , Electron Transport Complex II/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Animals , Antihypertensive Agents/pharmacology , Caspase 3 , Caspase 9 , Caspases/metabolism , Cell Death , Cell Line, Tumor , Cell Survival , Chromatin/metabolism , Coloring Agents/pharmacology , Cyclosporine/pharmacology , Cytochromes c/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Humans , Huntingtin Protein , Immunoblotting , Immunosuppressive Agents/pharmacology , L-Lactate Dehydrogenase/metabolism , Membrane Potentials , Mice , Mice, Transgenic , Mitochondria/metabolism , Mutation , Neurons/pathology , Nitro Compounds , Peptides/metabolism , Propionates/pharmacology , Rotenone/pharmacology , Ruthenium Red/pharmacology , Subcellular Fractions , Time Factors , Toxins, Biological , Uncoupling Agents/pharmacology
18.
J Biol Chem ; 278(6): 3825-30, 2003 Feb 07.
Article in English | MEDLINE | ID: mdl-12458211

ABSTRACT

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by an abnormally expended polyglutamine domain. There is no effective treatment for HD; however, inhibition of caspase activity or prevention of mitochondria dysfunction delays disease progression in HD mouse models. Similarly administration of cystamine, which can inhibit transglutaminase, prolonged survival of HD mice, suggesting that inhibition of transglutaminase might provide a new treatment strategy. However, it has been suggested that cystamine may inhibit other thiol-dependent enzymes in addition to transglutaminase. In this study we show that cystamine inhibits recombinant active caspase-3 in a concentration-dependent manner. At low concentrations cystamine is an uncompetitive inhibitor of caspase-3 activity, becoming a non-competitive inhibitor at higher concentrations. The IC(50) for cystamine-mediated inhibition of caspase-3 activity in vitro was 23.6 microm. In situ cystamine inhibited in a concentration-dependent manner the activation of caspase-3 by different pro-apoptotic agents. Additionally, cystamine inhibited caspase-3 activity to the same extent in cell lines stably overexpressing wild type tissue transglutaminase (tTG), a mutant inactive tTG, or an antisense for tTG, demonstrating that cystamine inhibits caspase activity independently of any effects it may have on the transamidating activity of tTG. Finally, treatment with cystamine resulted in a robust increase in the levels of glutathione. These findings demonstrate that cystamine may prolong neuronal survival and delay the onset of HD by inhibiting caspases and increasing the level of antioxidants such as glutathione.


Subject(s)
Caspase Inhibitors , Cystamine/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Huntington Disease/drug therapy , Peptides/metabolism , Caspase 3 , Caspases/metabolism , Cystamine/therapeutic use , Cysteine Proteinase Inhibitors/therapeutic use , Enzyme Activation , Humans , Huntington Disease/enzymology , Hydrogen Peroxide/pharmacology , Tumor Cells, Cultured
19.
Neuroreport ; 13(18): 2543-6, 2002 Dec 20.
Article in English | MEDLINE | ID: mdl-12499864

ABSTRACT

Human neuroblastoma SH-SY5Y cell lines stably expressing mutant truncated huntingtin with 82 (mutant) glutamine repeats (N63-82Q) were briefly exposed to hyperosmotic conditions which decrease cell volume and therefore transiently increased the concentration of N63-82Q, as well as activating specific stress-induced pathways. Transient hyperosmotic treatment significantly increased the number of cells with aggregates. When the N63-82Q cells were subsequently returned to iso-osmotic medium after the treatment, the number of cells with aggregates remained constant up to 12 h. However, between 12 and 24 h another significant increase in aggregate frequency was observed, with approximately 55% of the cells containing aggregates after 24 h. This may be due in part to the formation of microaggregates during hyperosmotic conditions that act as seeds for the aggregate formation. Further, treatment of cells with geldanamycin, which activates a heat shock response, significantly attenuated the hyperosmotic-induced increase in aggregate formation.


Subject(s)
Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuroblastoma , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Water-Electrolyte Balance/physiology , Benzoquinones , Enzyme Inhibitors/pharmacology , Heat-Shock Response/drug effects , Heat-Shock Response/physiology , Humans , Huntingtin Protein , Hypertonic Solutions/pharmacology , Lactams, Macrocyclic , Mutagenesis/physiology , Osmotic Pressure , Quinones/pharmacology , Tumor Cells, Cultured
20.
FEBS Lett ; 515(1-3): 61-5, 2002 Mar 27.
Article in English | MEDLINE | ID: mdl-11943195

ABSTRACT

It has been postulated that neuronal inclusions composed of mutant huntingtin may play a causative role in the pathogenesis of Huntington's disease. To study the putative role of aggregates in modulating apoptotic vulnerability, SH-SY5Y cell lines stably expressing truncated huntingtin with 18 (wild-type) (N63-18Q) or 82 (mutant) (N63-82Q) glutamine repeats were established. Aggregates were observed in approximately 13% of the N63-82Q cells; no aggregates were observed in the N63-18Q cells. In response to apoptotic stimuli such as staurosporine or hyperosmotic stress, caspase-3 activity was significantly greater in the N63-82Q cells compared to the N63-18Q cells. However, double immunostaining for huntingtin and active caspase-3 revealed that the presence of aggregates did not correlate with the presence of active caspase-3, indicating that aggregates do not contribute to the increase in apoptosis in the N63-82Q cells.


Subject(s)
Apoptosis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Apoptosis/drug effects , Caspase 3 , Caspases/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Huntingtin Protein , Huntington Disease/etiology , Immunoblotting , Macromolecular Substances , Nerve Tissue Proteins/pharmacology , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Neuroblastoma/pathology , Nuclear Proteins/pharmacology , Osmolar Concentration , Poly(ADP-ribose) Polymerases/metabolism , Sorbitol/pharmacology , Staurosporine/pharmacology , Transgenes , Trinucleotide Repeat Expansion/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...