Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
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
3.
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
4.
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.

5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...