Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
2.
J Huntingtons Dis ; 3(2): 159-74, 2014.
Article in English | MEDLINE | ID: mdl-25062859

ABSTRACT

BACKGROUND: Increasing mutant huntingtin (mHTT) clearance through the autophagy pathway may be a way to treat Huntington's disease (HD). Tools to manipulate and measure autophagy flux in brain in vivo are not well established. OBJECTIVE: To examine the in vivo pharmacokinetics and pharmacodynamics of the lysosomal inhibitor chloroquine (CQ) and the levels of selected autophagy markers to determine usefulness of CQ as a tool to study autophagy flux in brain. METHODS: Intraperitoneal injections of CQ were administered to WT and HD(Q175/Q175) mice. CQ levels were measured by LC-MS/MS in WT brain, muscle and blood at 4 to 24 hours after the last dose. Two methods of tissue preparation were used to detect by Western blot levels of the macroautophagy markers LC3 II and p62, the chaperone mediated autophagy receptor LAMP-2A and the late endosome/lysosomal marker RAB7. RESULTS: Following peripheral administration, CQ levels were highest in muscle and declined rapidly between 4 and 24 hours. In the brain, CQ levels were greater in the cortex than striatum, and levels persisted up to 24 hours post-injection. CQ treatment induced changes in LC3 II and p62 that were variable across regions and tissue preparations. HD(Q175/Q175) mice exposed to CQ had variable but diminished levels of LC3 II, p62 and LAMP-2A, and increased levels of RAB7. Higher levels of mHTT were found in the membrane compartment of CQ treated HD mice. CONCLUSION: Our findings suggest that the response of brain to CQ treatment, a blocker of autophagy flux, is variable and not as robust as it has been demonstrated in vitro, suggesting that CQ treatment has limitations for modulating autophagy flux in vivo. Alternative methods, compounds, and technologies need to be developed to further investigate autophagy flux in vivo, especially in the brain.


Subject(s)
Autophagy/drug effects , Brain/drug effects , Chloroquine/pharmacology , Huntington Disease/drug therapy , Animals , Antimalarials/pharmacokinetics , Antimalarials/pharmacology , Brain/metabolism , Brain/pathology , Chloroquine/pharmacokinetics , Disease Models, Animal , Gene Knock-In Techniques , Huntingtin Protein , Huntington Disease/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factor TFIIH , Transcription Factors/metabolism , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
3.
Hum Mol Genet ; 22(6): 1112-31, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23223017

ABSTRACT

A mutation in the huntingtin (Htt) gene produces mutant Htt and Huntington's disease (HD), a neurodegenerative disorder. HD patients have oxidative damage in the brain, but the causes are unclear. Compared with controls, we found brain levels of NADPH oxidase (NOX) activity, which produces reactive oxygen species (ROS), elevated in human HD postmortem cortex and striatum and highest in striatum of presymptomatic individuals. Synaptosome fractions from cortex and striatum of HD(140Q/140Q) mice had elevated NOX activity at 3 months of age and a further rise at 6 and 12 months compared with synaptosomes of age-matched wild-type (WT) mice. High NOX activity in primary cortical and striatal neurons of HD(140Q/140Q) mice correlated with more ROS and neurite swellings. These features and neuronal cell death were markedly reduced by treatment with NOX inhibitors such as diphenyleneiodonium (DPI), apocynin (APO) and VAS2870. The rise in ROS levels in mitochondria of HD(140Q/140Q) neurons followed the rise in NOX activity and inhibiting only mitochondrial ROS was not neuroprotective. Mutant Htt colocalized at plasma membrane lipid rafts with gp91-phox, a catalytic subunit for the NOX2 isoform. Assembly of NOX2 components at lipid rafts requires activation of Rac1 which was also elevated in HD(140Q/140Q) neurons. HD(140Q/140Q) mice bred to gp91-phox knock-out mice had lower NOX activity in the brain and in primary neurons, and neurons had normal ROS levels and significantly improved survival. These findings suggest that increased NOX2 activity at lipid rafts is an early and major source of oxidative stress and cell death in HD(140Q/140Q) neurons.


Subject(s)
Huntington Disease/enzymology , Huntington Disease/physiopathology , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Oxidative Stress , Animals , Cell Death , Disease Models, Animal , Female , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/metabolism , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NADPH Oxidase 2 , NADPH Oxidases/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/enzymology , Neurons/metabolism , Reactive Oxygen Species/metabolism , Up-Regulation
4.
J Huntingtons Dis ; 2(4): 459-75, 2013.
Article in English | MEDLINE | ID: mdl-24696705

ABSTRACT

BACKGROUND: Synaptic connections are disrupted in patients with Huntington's disease (HD). Synaptosomes from postmortem brain are ideal for synaptic function studies because they are enriched in pre- and post-synaptic proteins important in vesicle fusion, vesicle release, and neurotransmitter receptor activation. OBJECTIVE: To examine striatal synaptosomes from 3, 6 and 12 month old WT and Hdh140Q/140Q knock-in mice for levels of synaptic proteins, methionine oxidation, and glutamate release. METHODS: We used Western blot analysis, glutamate release assays, and liquid chromatography tandem mass spectrometry (LC-MS/MS). RESULTS: Striatal synaptosomes of 6 month old Hdh140Q/140Q mice had less DARPP32, syntaxin 1 and calmodulin compared to WT. Striatal synaptosomes of 12 month old Hdh140Q/140Q mice had lower levels of DARPP32, alpha actinin, HAP40, Na+/K+-ATPase, PSD95, SNAP-25, TrkA and VAMP1, VGlut1 and VGlut2, increased levels of VAMP2, and modifications in actin and calmodulin compared to WT. More glutamate released from vesicles of depolarized striatal synaptosomes of 6 month old Hdh140Q/140Q than from age matched WT mice but there was no difference in glutamate release in synaptosomes of 3 and 12 month old WT and Hdh140Q/140Q mice. LC-MS/MS of 6 month old Hdh140Q/140Q mice striatal synaptosomes revealed that about 4% of total proteins detected (>600 detected) had novel sites of methionine oxidation including proteins involved with vesicle fusion, trafficking, and neurotransmitter function (synaptophysin, synapsin 2, syntaxin 1, calmodulin, cytoplasmic actin 2, neurofilament, and tubulin). Altered protein levels and novel methionine oxidations were also seen in cortical synaptosomes of 12 month old Hdh140Q/140Q mice. CONCLUSIONS: Findings provide support for early synaptic dysfunction in Hdh140Q/140Q knock-in mice arising from altered protein levels, oxidative damage, and impaired glutamate neurotransmission and suggest that study of synaptosomes could be of value for evaluating HD therapies.


Subject(s)
Glutamic Acid/metabolism , Huntington Disease/metabolism , Methionine/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Synaptosomes/metabolism , Animals , Blotting, Western , Chromatography, Liquid , Corpus Striatum/metabolism , Disease Models, Animal , Gene Knock-In Techniques , Huntingtin Protein , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Tandem Mass Spectrometry
5.
Biochem Biophys Res Commun ; 421(4): 727-30, 2012 May 18.
Article in English | MEDLINE | ID: mdl-22542623

ABSTRACT

Huntington's disease (HD) is a progressive neurodegenerative disorder caused by a CAG repeat expansion in the huntingtin gene. Positron emission tomography studies have revealed a decline in glucose metabolism in the brain of patients with HD by a mechanism that has not been established. We examined glucose utilization in embryonic primary cortical neurons of wild-type (WT) and HD knock-in mice, which have 140 CAG repeats inserted in the endogenous mouse huntingtin gene (HD(140Q/140Q)). Primary HD(140Q/140Q) cortical neurons took up significantly less glucose than did WT neurons. Expression of permanently inactive and permanently active forms of Rab11 correspondingly altered glucose uptake in WT neurons, suggesting that normal activity of Rab11 is needed for neuronal uptake of glucose. It is known that Rab11 activity is diminished in HD(140Q/140Q) neurons. Expression of dominant active Rab11 to enhance the activity of Rab11 normalized glucose uptake in HD(140Q/140Q) neurons. These results suggest that deficient activity of Rab11 is a novel mechanism for glucose hypometabolism in HD.


Subject(s)
Glucose/metabolism , Huntington Disease/metabolism , Neurons/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Gene Knock-In Techniques , Mice , Mice, Mutant Strains , rab GTP-Binding Proteins/genetics
6.
Acta Pharmacol Sin ; 33(6): 743-51, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22543707

ABSTRACT

AIM: Huntingtin protein (Htt) was a neuropathological hallmark in human Huntington's Disease. The study aimed to investigate whether the macroautophagy regulator, Beclin1, was involved in the degradation of Htt. METHODS: PC12 cells and primary cultured brain neurons of rats were examined. pDC316 adenovirus shuttle plasmid was used to mediate the expression of wild-type Htt-18Q-552 or mutant Htt-100Q-552 in PC12 cells. The expression of the autophagy-related proteins LC3 II and Beclin1, as well as the lysosome-associated enzymes Cathepsin B and L was evaluated using Western blotting. The locations of Beclin1 and Htt were observed with immunofluorescence and confocal microscope. RESULTS: Htt552 expression increased the expression of LC3 II, Beclin1, cathepsin B and L in autophagy/lysosomal degradation pathway. Treatment with the autophagy inhibitor 3-MA or the proteasome inhibitors lactacystin and MG-132 increased Htt552 levels in PC12 cells infected with Ad-Htt-18Q-552 or Ad-Htt-100Q-552. The proteasome inhibitor caused a higher accumulation of Htt552-18Q than Htt552-100Q, and the autophagy inhibitor resulted in a higher accumulation of Htt552-100Q than Htt552-18Q. Similar results were observed in primary cultured neurons infected with adenovirus. In Htt552-expressing cells, Beclin1 was redistributed from the nucleus to the cytoplasm. Htt siRNA prevented Beclin1 redistribution in starvation conditions. Blockade of Beclin1 nuclear export by leptomycin B or Beclin1 deficiency caused by RNA interference induced the formation of mHtt552 aggregates. CONCLUSION: Beclin1 regulates the accumulation of Htt via macroautophagy.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Huntington Disease/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Active Transport, Cell Nucleus , Animals , Apoptosis Regulatory Proteins/analysis , Apoptosis Regulatory Proteins/genetics , Autophagy , Beclin-1 , Brain/cytology , Brain/metabolism , Brain/pathology , Cells, Cultured , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/pathology , Mutation , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , Neurons/pathology , Nuclear Proteins/analysis , Nuclear Proteins/genetics , PC12 Cells , RNA, Small Interfering/genetics , Rats , Up-Regulation
7.
Mol Cell Neurosci ; 50(1): 70-81, 2012 May.
Article in English | MEDLINE | ID: mdl-22508027

ABSTRACT

Neural stem (NS) cells are a limitless resource, and thus superior to primary neurons for drug discovery provided they exhibit appropriate disease phenotypes. Here we established NS cells for cellular studies of Huntington's disease (HD). HD is a heritable neurodegenerative disease caused by a mutation resulting in an increased number of glutamines (Q) within a polyglutamine tract in Huntingtin (Htt). NS cells were isolated from embryonic wild-type (Htt(7Q/7Q)) and "knock-in" HD (Htt(140Q/140Q)) mice expressing full-length endogenous normal or mutant Htt. NS cells were also developed from mouse embryonic stem cells that were devoid of Htt (Htt(-/-)), or knock-in cells containing human exon1 with an N-terminal FLAG epitope tag and with 7Q or 140Q inserted into one of the mouse alleles (Htt(F7Q/7Q) and Htt(F140Q/7Q)). Compared to Htt(7Q/7Q) NS cells, HD Htt(140Q/140Q) NS cells showed significantly reduced levels of cholesterol, increased levels of reactive oxygen species (ROS), and impaired motility. The heterozygous Htt(F140Q/7Q) NS cells had increased ROS and decreased motility compared to Htt(F7Q/7Q). These phenotypes of HD NS cells replicate those seen in HD patients or in primary cell or in vivo models of HD. Huntingtin "knock-out" NS cells (Htt(-/-)) also had impaired motility, but in contrast to HD cells had increased cholesterol. In addition, Htt(140Q/140Q) NS cells had higher phospho-AKT/AKT ratios than Htt(7Q/7Q) NS cells in resting conditions and after BDNF stimulation, suggesting mutant htt affects AKT dependent growth factor signaling. Upon differentiation, the Htt(7Q/7Q) and Htt(140Q/140Q) generated numerous Beta(III)-Tubulin- and GABA-positive neurons; however, after 15 days the cellular architecture of the differentiated Htt(140Q/140Q) cultures changed compared to Htt(7Q/7Q) cultures and included a marked increase of GFAP-positive cells. Our findings suggest that NS cells expressing endogenous mutant Htt will be useful for study of mechanisms of HD and drug discovery.


Subject(s)
Cholesterol/metabolism , Nerve Tissue Proteins/genetics , Neural Stem Cells/metabolism , Nuclear Proteins/genetics , Animals , Cell Differentiation/physiology , Cell Movement , Disease Models, Animal , Embryonic Stem Cells/metabolism , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/metabolism , Mice , Mice, Mutant Strains , Mice, Transgenic , Mutagenesis, Insertional , Mutation , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Nuclear Proteins/metabolism , Phenotype , Reactive Oxygen Species/metabolism
8.
J Biol Chem ; 287(16): 13487-99, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22375012

ABSTRACT

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


Subject(s)
Huntington Disease/genetics , Huntington Disease/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Adult , Animals , Blotting, Western , Detergents/pharmacology , Electrophoresis, Gel, Two-Dimensional , Humans , Huntingtin Protein , Huntington Disease/epidemiology , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/chemistry , Neurons/cytology , Nuclear Proteins/chemistry , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Prevalence , Primary Cell Culture , Protein Denaturation , Protein Folding , Rabbits , Reticulocytes/cytology , Solubility , Subcellular Fractions/chemistry , Subcellular Fractions/metabolism , Tissue Banks
9.
Neuroreport ; 23(1): 10-5, 2012 Jan 04.
Article in English | MEDLINE | ID: mdl-22045254

ABSTRACT

Patients with Huntington's disease suffer severe neuronal loss and signs of oxidative damage in the brain. Previously we found that primary neurons from embryonic cortex of mice bearing the Huntington's disease mutation (140 glutamines inserted into exon 1 of huntingtin) showed higher levels of reactive oxygen species before cell death. Here, we treated mutant neurons with known neuroprotective agents and determined the effects on neuronal survival and levels of reactive oxygen species. Primary neurons were exposed to the neurotrophin, brain derived neurotrophic factor, the antioxidant N-acetyl-cysteine or a specific inhibitor of glycogen synthase kinase 3-ß, SB216763. Each reagent increased the survival of the mutant neurons compared with untreated mutant neurons and also reduced the levels of reactive oxygen species to levels of wild-type neurons. These results suggest that reducing the levels of reactive oxygen species may be necessary to protect neurons with the Huntington's disease mutation from cell death.


Subject(s)
Huntington Disease/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Animals , Antioxidants/pharmacology , Brain-Derived Neurotrophic Factor/pharmacology , Cell Death , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Huntington Disease/genetics , Huntington Disease/pathology , Indicators and Reagents , Male , Mice , Mice, Transgenic , Mutation , Neurons/drug effects , Neurons/metabolism
10.
Mol Neurodegener ; 5: 58, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21156064

ABSTRACT

BACKGROUND: The mutation in Huntington's disease is a polyglutamine expansion near the N-terminus of huntingtin. Huntingtin expressed in immortalized neurons is cleaved near the N-terminus to form N-terminal polypeptides known as cleavage products A and B (cpA and cpB). CpA and cpB with polyglutamine expansion form inclusions in the nucleus and cytoplasm, respectively. The formation of cpA and cpB in primary neurons has not been established and the proteases involved in the formation of these fragments are unknown. RESULTS: Delivery of htt cDNA into the mouse striatum using adeno-associated virus or into primary cortical neurons using lentivirus generated cpA and cpB, indicating that neurons in brain and in vitro can form these fragments. A screen of small molecule protease inhibitors introduced to clonal striatal X57 cells and HeLa cells identified compounds that reduced levels of cpA and are inhibitors of the aspartyl proteases cathepsin D and cathepsin E. The most effective compound, P1-N031, is a transition state mimetic for aspartyl proteases. By western blot analysis, cathepsin D was easily detected in clonal striatal X57 cells, mouse brain and primary neurons, whereas cathepsin E was only detectible in clonal striatal X57 cells. In primary neurons, levels of cleavage product A were not changed by the same compounds that were effective in clonal striatal cells or by mRNA silencing to partially reduce levels of cathepsin D. Instead, treating primary neurons with compounds that are known to inhibit gamma secretase activity either indirectly (Imatinib mesylate, Gleevec) or selectively (LY-411,575 or DAPT) reduced levels of cpA. LY-411,575 or DAPT also increased survival of primary neurons expressing endogenous full-length mutant huntingtin. CONCLUSION: We show that cpA and cpB are produced from a larger huntingtin fragment in vivo in mouse brain and in primary neuron cultures. The aspartyl protease involved in forming cpA has cathepsin-D like properties in immortalized neurons and gamma secretase-like properties in primary neurons, suggesting that cell type may be a critical factor that specifies the aspartyl protease responsible for cpA. Since gamma secretase inhibitors were also protective in primary neurons, further study of the role of gamma-secretase activity in HD neurons is justified.

11.
J Neurosci ; 30(13): 4552-61, 2010 Mar 31.
Article in English | MEDLINE | ID: mdl-20357106

ABSTRACT

Oxidative stress contributes to neurodegeneration in Huntington's disease (HD). However, the origins of oxidative stress in HD remain unclear. Studies in HD transgenic models suggest involvement of mitochondrial dysfunction, which would lead to overproduction of reactive oxygen species (ROS). Impaired mitochondria complexes occur in late stages of HD but not in presymptomatic or early-stage HD patients. Thus, other mechanisms may account for the earliest source of oxidative stress caused by endogenous mutant huntingtin. Here, we report that decreased levels of a major intracellular antioxidant glutathione coincide with accumulation of ROS in primary HD neurons prepared from embryos of HD knock-in mice (HD(140Q/140Q)), which have human huntingtin exon 1 with 140 CAG repeats inserted into the endogenous mouse huntingtin gene. Uptake of extracellular cysteine through the glutamate/cysteine transporter EAAC1 is required for de novo synthesis of glutathione in neurons. We found that, compared with wild-type neurons, HD neurons had lower cell surface levels of EAAC1 and were deficient in taking up cysteine. Constitutive trafficking of EAAC1 from recycling endosomes relies on Rab11 activity, which is defective in the brain of HD(140Q/140Q) mice. Enhancement of Rab11 activity by expression of a dominant-active Rab11 mutant in primary HD neurons ameliorated the deficit in cysteine uptake, increased levels of intracellular glutathione, normalized clearance of ROS, and improved neuronal survival. Our data support a novel mechanism for oxidative stress in HD: Rab11 dysfunction slows trafficking of EAAC1 to the cell surface and impairs cysteine uptake, thereby leading to deficient synthesis of glutathione.


Subject(s)
Excitatory Amino Acid Transporter 3/metabolism , Huntington Disease/metabolism , Neurons/metabolism , Oxidative Stress , rab GTP-Binding Proteins/physiology , Animals , Cell Death , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cysteine/metabolism , Gene Knock-In Techniques , Glutathione/metabolism , Humans , Huntingtin Protein , Huntington Disease/pathology , Mice , Nerve Tissue Proteins/genetics , Neurons/pathology , Nuclear Proteins/genetics , Protein Transport , Reactive Oxygen Species/metabolism , rab GTP-Binding Proteins/genetics
12.
J Neurosci Res ; 88(1): 179-90, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19642201

ABSTRACT

Patients with Huntington's disease have an expanded polyglutamine tract in huntingtin and suffer severe brain atrophy and neurodegeneration. Because membrane dysfunction can occur in Huntington's disease, we addressed whether mutant huntingtin in brain and primary neurons is present in lipid rafts, which are cholesterol-enriched membrane domains that mediate growth and survival signals. Biochemical analysis of detergent-resistant membranes from brains and primary neurons of wild-type and presymptomatic Huntington's disease knock-in mice showed that wild-type and mutant huntingtin were recovered in lipid raft-enriched detergent-resistant membranes. The association with lipid rafts was stronger for mutant huntingtin than wild-type huntingtin. Lipid rafts extracted from Huntington's disease mice had normal levels of lipid raft markers (G(alphaq), Ras, and flotillin) but significantly more glycogen synthase kinase 3-beta. Increases in glycogen synthase kinase 3-beta have been associated with apoptotic cell death. Treating Huntington's disease primary neurons with inhibitors of glycogen synthase kinase 3-beta reduced neuronal death. We speculate that accumulation of mutant huntingtin and glycogen synthase kinase 3-beta in lipid rafts of presymptomatic Huntington's disease mouse neurons contributes to neurodegeneration in Huntington's disease.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Huntington Disease/metabolism , Membrane Microdomains/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Analysis of Variance , Animals , Blotting, Western , Cell Fractionation , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Glycogen Synthase Kinase 3/genetics , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/pathology , Indoles/pharmacology , Maleimides/pharmacology , Membrane Microdomains/genetics , Membrane Microdomains/pathology , Mice , Mice, Transgenic , Microscopy, Confocal , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Tissue Proteins/genetics , Neurons/drug effects , Neurons/pathology , Nuclear Proteins/genetics
13.
PLoS Curr ; 2: RRN1205, 2010 Dec 29.
Article in English | MEDLINE | ID: mdl-21278900

ABSTRACT

A means for measuring levels of soluble huntingtin proteins in clinical samples is essential for assessing the biological effects of potential mutant huntingtin (mtHtt) modifying treatments being developed for Huntington's disease (HD). We have optimized a previously described cell-based Homogeneous Time Resolved Fluorescence method that can measure soluble mtHtt and its ratio to the total Htt (tHtt) in blood buffy coats [1]. The results of the optimization and assay qualification indicate the assay to be specific for mtHtt in HD compared to Control subjects, highly sensitive, and technically and biologically reproducible. We therefore generated a Good Laboratory Practice Standard Operating Procedure which we validated, using 30 HD and 8 control buffy coat samples in which significant differences in mtHtt levels were found. We intend to deploy the assay to evaluate sample sets from observational and therapeutic studies enrolling HD subjects to further validate soluble mtHtt measurement by HTRF as a biomarker for HD and to explore its potential uses.

14.
Mol Cell Biol ; 29(22): 6106-16, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19752198

ABSTRACT

Huntingtin (Htt) localizes to endosomes, but its role in the endocytic pathway is not established. Recently, we found that Htt is important for the activation of Rab11, a GTPase involved in endosomal recycling. Here we studied fibroblasts of healthy individuals and patients with Huntington's disease (HD), which is a movement disorder caused by polyglutamine expansion in Htt. The formation of endocytic vesicles containing transferrin at plasma membranes was the same in control and HD patient fibroblasts. However, HD fibroblasts were delayed in recycling biotin-transferrin back to the plasma membrane. Membranes of HD fibroblasts supported less nucleotide exchange on Rab11 than did control membranes. Rab11-positive vesicular and tubular structures in HD fibroblasts were abnormally large, suggesting that they were impaired in forming vesicles. We used total internal reflection fluorescence imaging of living fibroblasts to monitor fluorescence-labeled transferrin-carrying transport intermediates that emerged from recycling endosomes. HD fibroblasts had fewer small vesicles and more large vesicles and long tubules than did control fibroblasts. Dominant active Rab11 expressed in HD fibroblasts normalized the recycling of biotin-transferrin. We propose a novel mechanism for cellular dysfunction by the HD mutation arising from the inhibition of Rab11 activity and a deficit in vesicle formation at recycling endosomes.


Subject(s)
Endocytosis , Endosomes/metabolism , Mutant Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , rab GTP-Binding Proteins/metabolism , Adolescent , Adult , Biotin/metabolism , Cells, Cultured , Child , Clathrin-Coated Vesicles/metabolism , Endosomes/enzymology , Endosomes/pathology , Enzyme Activation , Fibroblasts/enzymology , Fibroblasts/pathology , Genes, Dominant , Humans , Huntingtin Protein , Microscopy, Fluorescence , Models, Biological , Protein Transport , Receptors, Transferrin/metabolism , Staining and Labeling , Transferrin/metabolism
15.
Neurobiol Dis ; 36(2): 374-83, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19699304

ABSTRACT

The Huntington's disease (HD) mutation causes polyglutamine expansion in huntingtin (Htt) and neurodegeneration. Htt interacts with a complex containing Rab11GDP and is involved in activation of Rab11, which functions in endosomal recycling and neurite growth and long-term potentiation. Like other Rab proteins, Rab11GDP undergoes nucleotide exchange to Rab11GTP for its activation. Here we show that striatal membranes of HD(140Q/140Q) knock-in mice are impaired in supporting conversion of Rab11GDP to Rab11GTP. Dominant negative Rab11 expressed in the striatum and cortex of normal mice caused neuropathology and motor dysfunction, suggesting that a deficiency in Rab11 activity is pathogenic in vivo. Primary cortical neurons from HD(140Q/140Q) mice were delayed in recycling transferrin receptors back to the plasma membrane. Partial rescue from glutamate-induced cell death occurred in HD neurons expressing dominant active Rab11. We propose a novel mechanism of HD pathogenesis arising from diminished Rab11 activity at recycling endosomes.


Subject(s)
Disease Models, Animal , Gene Knock-In Techniques , Huntington Disease/genetics , Huntington Disease/metabolism , rab GTP-Binding Proteins/deficiency , rab GTP-Binding Proteins/genetics , Animals , Cell Cycle/genetics , Cell Line , Cells, Cultured , Endosomes/genetics , Endosomes/metabolism , Gene Expression Regulation , Huntington Disease/etiology , Mice , Mice, Neurologic Mutants , rab GTP-Binding Proteins/metabolism
16.
Biochem Biophys Res Commun ; 387(3): 472-5, 2009 Sep 25.
Article in English | MEDLINE | ID: mdl-19607813

ABSTRACT

An expanded polyglutamine (Q) tract (>37Q) in huntingtin (htt) causes Huntington disease. Htt associates with membranes and polyglutamine expansion in htt may alter membrane function in Huntington disease through a mechanism that is not known. Here we used differential scanning calorimetry to examine the effects of polyQ expansion in htt on its insertion into lipid bilayers. We prepared synthetic lipid vesicles composed of phosphatidylcholine and phosphatidylethanolamine and tested interactions of htt amino acids 1-89 with 20Q, 32Q or 53Q with the vesicles. GST-htt1-89 with 53Q inserted into synthetic lipid vesicles significantly more than GST-htt1-89 with 20Q or 32Q. We speculate that by inserting more into cell membranes, mutant huntingtin could increase disorder within the lipid bilayer and thereby disturb cellular membrane function.


Subject(s)
Cell Membrane/metabolism , Huntington Disease/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Peptides/metabolism , Cell Membrane/chemistry , Humans , Huntingtin Protein , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Peptides/chemistry , Phosphatidylcholines/chemistry , Phosphatidylethanolamines/chemistry
17.
J Neurochem ; 110(5): 1585-97, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19566678

ABSTRACT

Huntingtin has an expanded polyglutamine tract in patients with Huntington's disease. Huntingtin localizes to intracellular and plasma membranes but the function of huntingtin at membranes is unknown. Previously we reported that exogenously expressed huntingtin bound pure phospholipids using protein-lipid overlays. Here we show that endogenous huntingtin from normal (Hdh(7Q/7Q)) mouse brain and mutant huntingtin from Huntington's disease (Hdh(140Q/140Q)) mouse brain bound to large unilamellar vesicles containing phosphoinositol (PI) PI 3,4-bisphosphate, PI 3,5-bisphosphate, and PI 3,4,5-triphosphate [PI(3,4,5)P3]. Huntingtin interactions with multivalent phospholipids were similar to those of dynamin. Mutant huntingtin associated more with phosphatidylethanolamine and PI(3,4,5)P3 than did wild-type huntingtin, and associated with other phospholipids not recognized by wild-type huntingtin. Wild-type and mutant huntingtin also bound to large unilamellar vesicles containing cardiolipin, a phospholipid specific to mitochondrial membranes. Maximal huntingtin-phospholipid association required inclusion of huntingtin amino acids 171-287. Endogenous huntingtin recruited to the plasma membrane in cells that incorporated exogenous PI 3,4-bisphosphate and PI(3,4,5)P3 or were stimulated by platelet-derived growth factor or insulin growth factor 1, which both activate PI 3-kinase. These data suggest that huntingtin interacts with membranes through specific phospholipid associations and that mutant huntingtin may disrupt membrane trafficking and signaling at membranes.


Subject(s)
Huntington Disease/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Peptides/metabolism , Phospholipids/metabolism , Animals , Cardiolipins/metabolism , Cell Line, Transformed , Cells, Cultured , Humans , Huntingtin Protein , Huntington Disease/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Peptides/genetics , Phosphatidylinositol 3-Kinases/physiology , Phospholipids/genetics , Protein Binding/physiology
18.
Neuroreport ; 19(16): 1643-7, 2008 Oct 29.
Article in English | MEDLINE | ID: mdl-18845944

ABSTRACT

Huntingtin is ubiquitously expressed and enriched in the brain. Deletion of the huntingtin gene in mice is lethal during early embryonic development. The function of huntingtin is, however, not clear. Here, we report that huntingtin is important for the function of Rab11, a critical GTPase in regulating membrane traffic from recycling endosomes to the plasma membrane. In huntingtin-null embryonic stem cells, the levels of Rab11 on membranes and nucleotide exchange activity on Rab11 were significantly reduced compared with normal embryonic stem cells. In brain membranes, an antibody against huntingtin immunoprecipitated a nucleotide exchange activity on Rab11 and huntingtin was coprecipitated with Rab11 in the presence of guanosine diphosphate. These data suggest a role for huntingtin in a complex that activates Rab11.


Subject(s)
Embryonic Stem Cells/metabolism , Guanine Nucleotides/metabolism , Serotonin Plasma Membrane Transport Proteins/physiology , rab GTP-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Cells, Cultured , Embryonic Stem Cells/cytology , Endocytosis/physiology , Immunoprecipitation , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice , Mice, Knockout , Molecular Sequence Data , Protein Transport/physiology , Sequence Homology, Amino Acid , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , rab GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism
19.
Neurobiol Dis ; 22(2): 346-56, 2006 May.
Article in English | MEDLINE | ID: mdl-16423528

ABSTRACT

N-terminal mutant huntingtin (N-mhtt) fragments form inclusions and cause cell death in vitro. Mutant htt expression stimulates autophagy and increases levels of lysosomal proteases. Here, we show that lysosomal proteases, cathepsins D, B and L, affected mhtt processing and levels of cleavage products (cp) known as A and B, which form inclusions. Adding inhibitors of cathepsin D, B and L to clonal striatal cells reduced mhtt, especially mhtt fragment cp A. Mutant htt fully degraded in cathepsin-L-treated lysates but formed stable N-mhtt fragments upon exposure to cathepsin D. Mutagenesis analysis of htt cDNA suggested that cathepsin D and the protease for cp A may cleave htt in the same region. Brain lysates from HD knock-in mice expressed N-mhtt fragments that accumulated with cathepsin D treatment and declined with aspartyl protease inhibition. Findings implicate lysosomal proteases in formation of N-mhtt fragments and clearance of mhtt.


Subject(s)
Cathepsins/metabolism , Huntington Disease/enzymology , Lysosomes/enzymology , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Peptide Fragments/metabolism , Peptide Hydrolases/metabolism , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/metabolism , Binding Sites/physiology , Cathepsin D/antagonists & inhibitors , Cathepsin D/metabolism , Cathepsins/antagonists & inhibitors , Cell Line, Transformed , Corpus Striatum/enzymology , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Enzyme Inhibitors/pharmacology , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/physiopathology , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurons/enzymology , Neurons/pathology , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Peptide Hydrolases/drug effects , Protein Structure, Tertiary/physiology
20.
J Biol Chem ; 280(43): 36464-73, 2005 Oct 28.
Article in English | MEDLINE | ID: mdl-16085648

ABSTRACT

We have identified a domain in the N terminus of huntingtin that binds to membranes. A three-dimensional homology model of the structure of the binding domain predicts helical HEAT repeats, which emanate a positive electrostatic potential, consistent with a charge-based mechanism for membrane association. An amphipathic helix capable of inserting into pure lipid bilayers may serve to anchor huntingtin to the membrane. In cells, N-terminal huntingtin fragments targeted to regions of plasma membrane enriched in phosphatidylinositol 4,5-bisphosphate, receptor bound-transferrin, and endogenous huntingtin. N-terminal huntingtin fragments with an expanded polyglutamine tract aberrantly localized to intracellular regions instead of plasma membrane. Our data support a new model in which huntingtin directly binds membranes through electrostatic interactions with acidic phospholipids.


Subject(s)
Cell Membrane/metabolism , Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , Phospholipids/chemistry , Amino Acid Sequence , Animals , Blotting, Western , COS Cells , Calorimetry, Differential Scanning , Cell Line, Tumor , Chlorocebus aethiops , DNA, Complementary/metabolism , Endoplasmic Reticulum/metabolism , Glutathione Transferase/metabolism , Humans , Huntingtin Protein , Immunohistochemistry , Immunoprecipitation , Lipid Bilayers/chemistry , Microscopy, Fluorescence , Models, Molecular , Molecular Sequence Data , Mutation , Nerve Tissue Proteins/chemistry , Nuclear Proteins/chemistry , Peptides/chemistry , Phosphatidylinositol 4,5-Diphosphate/chemistry , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Software , Static Electricity , Subcellular Fractions , Temperature , Transfection , Transferrin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...