Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Mol Psychiatry ; 20(4): 459-71, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25070536

ABSTRACT

Human mutations in PQBP1, a molecule involved in transcription and splicing, result in a reduced but architecturally normal brain. Examination of a conditional Pqbp1-knockout (cKO) mouse with microcephaly failed to reveal either abnormal centrosomes or mitotic spindles, increased neurogenesis from the neural stem progenitor cell (NSPC) pool or increased cell death in vivo. Instead, we observed an increase in the length of the cell cycle, particularly for the M phase in NSPCs. Corresponding to the developmental expression of Pqbp1, the stem cell pool in vivo was decreased at E10 and remained at a low level during neurogenesis (E15) in Pqbp1-cKO mice. The expression profiles of NSPCs derived from the cKO mouse revealed significant changes in gene groups that control the M phase, including anaphase-promoting complex genes, via aberrant transcription and RNA splicing. Exogenous Apc4, a hub protein in the network of affected genes, recovered the cell cycle, proliferation, and cell phenotypes of NSPCs caused by Pqbp1-cKO. These data reveal a mechanism of brain size control based on the simple reduction of the NSPC pool by cell cycle time elongation. Finally, we demonstrated that in utero gene therapy for Pqbp1-cKO mice by intraperitoneal injection of the PQBP1-AAV vector at E10 successfully rescued microcephaly with preserved cortical structures and improved behavioral abnormalities in Pqbp1-cKO mice, opening a new strategy for treating this intractable developmental disorder.


Subject(s)
Genetic Therapy , Microcephaly/genetics , Microcephaly/therapy , Neural Stem Cells/physiology , Nuclear Proteins/deficiency , Adenoviridae/genetics , Animals , Apc4 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Apoptosis/genetics , Brain/pathology , Carrier Proteins/genetics , Cell Adhesion Molecules/metabolism , Cell Cycle , Cell Proliferation , DNA-Binding Proteins , Disease Models, Animal , Embryo, Mammalian , Female , Humans , Male , Mice , Mice, Knockout , Microcephaly/pathology , Nestin/genetics , Nestin/metabolism , Neurogenesis , Nuclear Proteins/genetics , Synapsins/genetics , Synapsins/metabolism
2.
Hum Mol Genet ; 20(24): 4916-31, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-21933836

ABSTRACT

The polyglutamine-binding protein 1 (PQBP1) has been linked to several X-linked intellectual disability disorders and progressive neurodegenerative diseases. While it is currently known that PQBP1 localizes in nuclear speckles and is engaged in transcription and splicing, we have now identified a cytoplasmic pool of PQBP1. Analysis of PQBP1 complexes revealed six novel interacting proteins, namely the RNA-binding proteins KSRP, SFPQ/PSF, DDX1 and Caprin-1, and two subunits of the intracellular transport-related dynactin complex, p150(Glued) and p27. PQBP1 protein complex formation is dependent on the presence of RNA. Immunofluorescence studies revealed that in primary neurons, PQBP1 co-localizes with its interaction partners in specific cytoplasmic granules, which stained positive for RNA. Our results suggest that PQBP1 plays a role in cytoplasmic mRNA metabolism. This is further supported by the partial co-localization and interaction of PQBP1 with the fragile X mental retardation protein (FMRP), which is one of the best-studied proteins found in RNA granules. In further studies, we show that arsenite-induced oxidative stress caused relocalization of PQBP1 to stress granules (SGs), where PQBP1 co-localizes with the new binding partners as well as with FMRP. Additional results indicated that the cellular distribution of PQBP1 plays a role in SG assembly. Together these data demonstrate a role for PQBP1 in the modulation of SGs and suggest its involvement in the transport of neuronal RNA granules, which are of critical importance for the development and maintenance of neuronal networks, thus illuminating a route by which PQBP1 aberrations might influence cognitive function.


Subject(s)
Chromosomes, Human, X/genetics , Cytoplasmic Granules/metabolism , Genes, X-Linked/genetics , Intellectual Disability/genetics , Neurons/metabolism , Oligopeptides/genetics , RNA/metabolism , Animals , Cells, Cultured , Dynactin Complex , Fragile X Mental Retardation Protein/metabolism , Humans , Mice , Microtubule-Associated Proteins/metabolism , Models, Biological , Oligopeptides/metabolism , Protein Binding , Protein Transport , RNA-Binding Proteins/metabolism , Ribosomes/metabolism , T-Cell Intracellular Antigen-1
3.
J Cell Biol ; 130(1): 29-39, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7790375

ABSTRACT

A cDNA encoding the 180-kD canine ribosome receptor (RRp) was cloned and sequenced. The deduced primary structure indicates three distinct domains: an NH2-terminal stretch of 28 uncharged amino acids representing the membrane anchor, a basic region (pI = 10.74) comprising the remainder of the NH2-terminal half and an acidic COOH-terminal half (pI = 4.99). The most striking feature of the amino acid sequence is a 10-amino acid consensus motif, NQGKKAEGAP, repeated 54 times in tandem without interruption in the NH2-terminal positively charged region. We postulate that this repeated sequence represents a ribosome binding domain which mediates the interaction between the ribosome and the ER membrane. To substantiate this hypothesis, recombinant full-length ribosome receptor and two truncated versions of this protein, one lacking the potential ribosome binding domain, and one lacking the COOH terminus, were expressed in Saccharomyces cerevisiae. Morphological and biochemical analyses showed all proteins were targeted to, and oriented correctly in the ER membrane. In vitro ribosome binding assays demonstrated that yeast microsomes containing the full-length canine receptor or one lacking the COOH-terminal domain were able to bind two to four times as many human ribosomes as control membranes lacking a recombinant protein or microsomes containing a receptor lacking the NH2-terminal basic domain. Electron micrographs of these cells revealed that the expression of all receptor constructs led to a proliferation of perinuclear ER membranes known as "karmellae." Strikingly, in those strains which expressed cDNAs encoding a receptor containing the putative ribosome binding domain, the induced ER membranes (examined in situ) were richly studded with ribosomes. In contrast, karmellae resulting from the expression of receptor cDNA lacking the putative ribosome binding domain were uniformly smooth and free of ribosomes. Cell fractionation and biochemical analyses corroborated the morphological characterization. Taken together these data provide further evidence that RRp functions as a ribosome receptor in vitro, provide new evidence indicating its functionality in vivo, and in both cases indicate that the NH2-terminal basic domain is essential for ribosome binding.


Subject(s)
Endoplasmic Reticulum/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Ribosomes/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA Primers/chemistry , Dogs , Gene Expression , HeLa Cells , Humans , Intracellular Membranes/metabolism , Molecular Sequence Data , RNA, Messenger/genetics , Repetitive Sequences, Nucleic Acid , Saccharomyces cerevisiae/metabolism
4.
Mol Biol Cell ; 12(5): 1393-407, 2001 May.
Article in English | MEDLINE | ID: mdl-11359930

ABSTRACT

The huntingtin exon 1 proteins with a polyglutamine repeat in the pathological range (51 or 83 glutamines), but not with a polyglutamine tract in the normal range (20 glutamines), form aggresome-like perinuclear inclusions in human 293 Tet-Off cells. These structures contain aggregated, ubiquitinated huntingtin exon 1 protein with a characteristic fibrillar morphology. Inclusion bodies with truncated huntingtin protein are formed at centrosomes and are surrounded by vimentin filaments. Inhibition of proteasome activity resulted in a twofold increase in the amount of ubiquitinated, SDS-resistant aggregates, indicating that inclusion bodies accumulate when the capacity of the ubiquitin-proteasome system to degrade aggregation-prone huntingtin protein is exhausted. Immunofluorescence and electron microscopy with immunogold labeling revealed that the 20S, 19S, and 11S subunits of the 26S proteasome, the molecular chaperones BiP/GRP78, Hsp70, and Hsp40, as well as the RNA-binding protein TIA-1, the potential chaperone 14-3-3, and alpha-synuclein colocalize with the perinuclear inclusions. In 293 Tet-Off cells, inclusion body formation also resulted in cell toxicity and dramatic ultrastructural changes such as indentations and disruption of the nuclear envelope. Concentration of mitochondria around the inclusions and cytoplasmic vacuolation were also observed. Together these findings support the hypothesis that the ATP-dependent ubiquitin-proteasome system is a potential target for therapeutic interventions in glutamine repeat disorders.


Subject(s)
Acetylcysteine/analogs & derivatives , Heat-Shock Proteins , Inclusion Bodies/metabolism , Mutation , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Peptide Fragments/metabolism , Proteins , 14-3-3 Proteins , Acetylcysteine/pharmacology , Carrier Proteins/metabolism , Cell Line , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Endoplasmic Reticulum Chaperone BiP , Exons , Humans , Huntingtin Protein , Huntington Disease/metabolism , Immunoblotting , Inclusion Bodies/ultrastructure , Membrane Proteins/metabolism , Microscopy, Fluorescence , Models, Biological , Molecular Chaperones/metabolism , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Peptide Fragments/genetics , Poly(A)-Binding Proteins , Proteasome Endopeptidase Complex , RNA-Binding Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Synucleins , T-Cell Intracellular Antigen-1 , Transgenes , Tyrosine 3-Monooxygenase/metabolism , Vimentin/metabolism , alpha-Synuclein
5.
FEBS Lett ; 579(18): 3913-9, 2005 Jul 18.
Article in English | MEDLINE | ID: mdl-15987638

ABSTRACT

Mutations in the parkin gene encoding an E3 ligase are responsible for autosomal recessive Parkinson's disease. Putative parkin substrates and interacting partners have been identified, but the molecular mechanism underlying parkin-related neurodegeneration is still unclear. We have identified the 20S proteasomal subunit alpha4 (synonyms: PSMA7, XAPC7, subunit alpha type 7) as a new interacting partner of parkin. The C-terminal IBR-RING domain of parkin and the C-terminal part of alpha4 were essential for the interaction. Biochemical studies revealed that alpha4 was not a substrate for parkin-dependent ubiquitylation. Putative functions of the interaction might therefore be substrate presentation to the proteasome or regulation of proteasomal activity. Full-length parkin and parkin lacking the N-terminal ubiquitin-like domain slightly increased the proteasomal activity in HEK 293T cells, in line with the latter hypothesis.


Subject(s)
Cysteine Endopeptidases/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Line , Cysteine Endopeptidases/chemistry , DNA, Complementary/metabolism , Humans , Immunoprecipitation , Models, Genetic , Multienzyme Complexes/chemistry , Mutation , PC12 Cells , Plasmids/metabolism , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Structure, Tertiary , Rats , Signal Transduction , Two-Hybrid System Techniques , Ubiquitin/chemistry , Ubiquitin-Protein Ligases/chemistry
6.
Cell Death Differ ; 22(9): 1425-36, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25678324

ABSTRACT

Impairment of synaptic function can lead to neuropsychiatric disorders collectively referred to as synaptopathies. The SNARE protein SNAP-25 is implicated in several brain pathologies and, indeed, brain areas of psychiatric patients often display reduced SNAP-25 expression. It has been recently found that acute downregulation of SNAP-25 in brain slices impairs long-term potentiation; however, the processes through which this occurs are still poorly defined. We show that in vivo acute downregulation of SNAP-25 in CA1 hippocampal region affects spine number. Consistently, hippocampal neurons from SNAP-25 heterozygous mice show reduced densities of dendritic spines and defective PSD-95 dynamics. Finally, we show that, in brain, SNAP-25 is part of a molecular complex including PSD-95 and p140Cap, with p140Cap being capable to bind to both SNAP-25 and PSD-95. These data demonstrate an unexpected role of SNAP-25 in controlling PSD-95 clustering and open the possibility that genetic reductions of the protein levels - as occurring in schizophrenia - may contribute to the pathology through an effect on postsynaptic function and plasticity.


Subject(s)
Dendritic Spines/physiology , Guanylate Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Synaptosomal-Associated Protein 25/metabolism , Animals , Dendritic Spines/metabolism , Disks Large Homolog 4 Protein , HEK293 Cells , Hippocampus/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morphogenesis , Neuronal Plasticity/physiology , Synapses/metabolism , Transfection
7.
J Biomol Struct Dyn ; 17(3): 409-27, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10636078

ABSTRACT

Folding of oligoglutamine chains of different lengths is of crucial interest for exploring the molecular mechanisms of Huntington's disease. A simple oligoglutamine model based upon the Flory-Huggins theory of polymer solutions demonstrates a random coil instability in chains containing more than 40 glutamine residues with respect to beta-sheet formation. This is in striking quantitative agreement with biochemical results on the chain length dependence of polyglutamine aggregation in vivo and in vitro, as well as with clinical data on the polyglutamine chain length dependence of the onset of Huntington's disease. Furthermore, a detailed molecular-mechanical investigation of a polypeptide chain carrying 40 glutamine residues was performed. Two possible folding modes of such an oligoglutamine chain were revealed: a) a beta-hairpin and b) a highly compact random coil entity stabilized by a wealth of H-bonds among the glutamine side chains. A possible role of these folding modes in polyglutamine aggregation, as well as in the onset of Huntington's disease, is discussed.


Subject(s)
Glutamine , Oligopeptides/chemistry , Peptides/chemistry , Protein Folding , Protein Structure, Secondary , Amino Acid Sequence , Models, Molecular , Molecular Sequence Data , Software , Thermodynamics
8.
Dialogues Clin Neurosci ; 3(1): 17-23, 2001 Mar.
Article in English | MEDLINE | ID: mdl-22034471

ABSTRACT

Huntington's disease (HD) is a progressive, late-onset neurodegenerative illness with autosomal dominant inheritance that affects one in 10 000 individuals in Western Europe. The disease is caused by a polyglutamine repeat expansion located in the N-terminal region of the huntingtin protein. The mutation is likely to act by a gain of function, but the molecular mechanisms by which it leads to neuronal dysfunction and cell death are not yet known. The normal function of huntingtin in cell metabolism is also unclear. There is no therapy for HD. Research on HD should help elucidate the pathogenetic mechanism of this illness in order to develop successful treatments to prevent or slow down symptoms. This article presents new results in HD research focusing on in vivo and in vitro model systems, potential molecular mechanisms of HD, and the development of therapeutic strategies.

11.
J Neural Transm (Vienna) ; 113(8): 1081-90, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16835690

ABSTRACT

Protein interaction networks are useful resources for the functional annotation of proteins. Recently, we have generated a highly connected protein-protein interaction network for Huntington's disease (HD) by automated yeast two-hybrid (Y2H) screening (Goehler et al., 2004). The network included several novel direct interaction partners for the disease protein huntingtin (htt). Some of these interactions, however, have not been validated by independent methods. Here we describe the verification of the interaction between htt and GASP2 (G protein-coupled receptor associated sorting protein 2), a protein involved in membrane receptor degradation. Using membrane-based and classical coimmunoprecipitation assays we demonstrate that htt and GASP2 form a complex in cotransfected mammalian cells. Moreover, we show that the two proteins colocalize in SH-SY5Y cells, raising the possibility that htt and GASP2 interact in neurons. As the GASP protein family plays a role in G protein-coupled receptor sorting, our data suggest that htt might influence receptor trafficking via the interaction with GASP2.


Subject(s)
Carrier Proteins/metabolism , Huntington Disease/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Humans , Huntingtin Protein , Intracellular Signaling Peptides and Proteins , Neuroblastoma , Two-Hybrid System Techniques
12.
Mol Med Today ; 6(10): 387-91, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11006527

ABSTRACT

The accumulation of highly insoluble intracellular protein aggregates in neuronal inclusions is a hallmark of Huntington's disease (HD) and Parkinson's disease (PD) as well as several other late-onset neurodegenerative disorders. The aggregates formed in vitro and in vivo generally have a fibrillar morphology, consist of individual beta-strands and are resistant to proteolytic degradation. Although the causal relationship between aggregate formation and disease remains to be proven, the gradual deposition of mutant protein in neurons is consistent with the late-onset and progressive nature of symptoms. Recently, circumstantial evidence from mouse and Drosophila model systems suggests that abnormal protein folding and aggregation play a key role in the pathogenesis of both HD and PD. Therefore, a detailed understanding of the molecular mechanisms of protein aggregation and its effects on neuronal cell death could open new opportunities for therapy.


Subject(s)
Huntington Disease/physiopathology , Huntington Disease/therapy , Nerve Tissue Proteins/metabolism , Neurons/physiology , Parkinson Disease/physiopathology , Parkinson Disease/therapy , Animals , Drosophila , Humans , Huntington Disease/pathology , Mice , Models, Animal , Nerve Tissue Proteins/chemistry , Neurons/pathology , Parkinson Disease/pathology
13.
Biol Chem ; 381(9-10): 937-42, 2000.
Article in English | MEDLINE | ID: mdl-11076024

ABSTRACT

The formation of insoluble protein aggregates is a hallmark of Huntington's disease (HD) and related neurodegenerative disorders, such as dentatorubral pallidoluysian atrophy (DRPLA), spinal bulbar muscular atrophy (SBMA) and the spinocerebellar ataxia (SCA) type 1, 2, 3, 6 and 7. These disorders are caused by an expanded polyglutamine (polyQ) tract in otherwise unrelated proteins. They are characterized by late-onset, selective neuropathology, a pathogenic polyQ threshold and a relationship between polyQ length and disease progression. Thus, molecular models of HD and related glutamine-repeat disorders must account for these characteristic features. During the last three years, considerable effort has been invested in the development of in vitro and in vivo model systems to study the mechanisms of protein aggregation in glutamine-repeat disorders and its potential effects on disease progression and neurodegeneration. A selection of these studies is reviewed here. Furthermore, the correlation between aggregate formation and development of HD is discussed.


Subject(s)
Huntington Disease/metabolism , Nerve Tissue Proteins/metabolism , Animals , Humans , Huntington Disease/pathology , Nerve Tissue Proteins/chemistry
14.
Philos Trans R Soc Lond B Biol Sci ; 354(1386): 991-4, 1999 Jun 29.
Article in English | MEDLINE | ID: mdl-10434297

ABSTRACT

We have shown previously by electron microscopy that the purified glutathione S-transferase (GST)-Huntington's disease (HD) exon 1 fusion protein with 51 glutamine residues (GST-HD51) is an oligomer, and that site-specific proteolytic cleavage of this fusion protein results in the formation of insoluble more highly ordered protein aggregates with a fibrillar or ribbon-like morphology (E. Scherzinger et al. (1997) Cell 90, 549-558). Here we report that a truncated GST HD exon 1 fusion protein with 51 glutamine residues, which lacks the proline-rich region C-terminal to the polyglutamine (polyQ) tract (GST-HD51 delta P) self-aggregates into high-molecular-mass protein aggregates without prior proteolytic cleavage. Electron micrographs of these protein aggregates revealed thread-like fibrils with a uniform diameter of ca. 25 nm. In contrast, proteolytic cleavage of GST-HD51 delta P resulted in the formation of numerous clusters of high-molecular-mass fibrils with a different, ribbon-like morphology. These structures were reminiscent of prion rods and beta-amyloid fibrils in Alzheimer's disease. In agreement with our previous results with full-length GST-HD exon 1, the truncated fusion proteins GST-HD20 delta P and GST-HD30 delta P did not show any tendency to form more highly ordered structures, either with or without protease treatment.


Subject(s)
Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Trinucleotide Repeat Expansion/genetics , Cloning, Molecular , Escherichia coli , Exons , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Huntingtin Protein , Huntington Disease/genetics , Microscopy, Electron , Nerve Tissue Proteins/ultrastructure , Nuclear Proteins/ultrastructure , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/ultrastructure , Sequence Deletion
15.
Proc Natl Acad Sci U S A ; 99(8): 5596-600, 2002 Apr 16.
Article in English | MEDLINE | ID: mdl-11960015

ABSTRACT

The exon-1 peptide of huntingtin has 51 Gln repeats and produces the symptoms of Huntington's disease in transgenic mice. Aggregation of the yeast Sup35 protein into prions has been attributed to its glutamine-rich and asparagine-rich domain. Here, we show that poly-L-asparagine forms polar zippers similar to those of poly-L-glutamine. In solution at acid pH, the glutamine-rich and asparagine-rich 18-residue Sup35 peptide, rendered soluble by the addition of two aspartates at the amino end and two lysines at the carboxyl end, gives a beta-sheet CD spectrum; it aggregates at neutral pH. A poly-alanine peptide D(2)A(10)K(2) gives an alpha-helical CD spectrum at all pHs and does not aggregate; a peptide with the sequence of the C-terminal helix of the alpha-chain of human hemoglobin, preceded by two aspartates and followed by two lysines, exhibits a random coil spectrum and does not aggregate either. Alignment of several beta-strands with the sequence of the 42-residue Alzheimer's amyloid beta-peptide shows that they can be linked together by a network of salt bridges. We also asked why single amino acid replacements can so destabilize the native structures of proteins that they unfold and form amyloids. The difference in free energy of a protein molecule between its native, fully ordered structure and an amorphous mixture of randomly coiled chains is only of the order of 10 kcal/mol. Theory shows that destabilization of the native structure by no more than 2 kcal/mol can increase the probability of nucleation of disordered aggregates from which amyloids could grow 130,000-fold.


Subject(s)
Alanine/chemistry , Amyloid beta-Peptides/chemistry , Asparagine/chemistry , Fungal Proteins/metabolism , Glutamine/chemistry , Peptide Fragments/chemistry , Prions , Saccharomyces cerevisiae Proteins , Amino Acid Sequence , Amino Acids/chemistry , Animals , Circular Dichroism , Exons , Hemoglobins/chemistry , Humans , Hydrogen-Ion Concentration , Light , Lysine/chemistry , Mice , Mice, Transgenic , Molecular Sequence Data , Peptide Termination Factors , Peptides/chemistry , Protein Binding , Protein Structure, Tertiary , Scattering, Radiation , Thermodynamics , Ultraviolet Rays , X-Ray Diffraction
16.
Mamm Genome ; 9(1): 26-31, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9434941

ABSTRACT

Huntington's disease (HD) is a devastating central nervous system disorder. Even though the gene responsible has been positionally cloned recently, its etiology has remained largely unclear. To investigate potential disease mechanisms, we conducted a search for binding partners of the HD-protein huntingtin. With the yeast two-hybrid system, one such interacting factor, the huntingtin interacting protein-1 (HIP-1), was identified (Wanker et al. 1997; Kalchman et al. 1997) and the human gene mapped to 7q11.2. In this paper we demonstrate the localization of the HIP1 mouse homologue (Hip1) into a previously identified region of human-mouse synteny on distal mouse Chromosome (Chr) 5, both employing an IRS-PCR-based mapping strategy and traditional fluorescent in situ hybridization (FISH) mapping.


Subject(s)
Chromosome Mapping , DNA-Binding Proteins , Nerve Tissue Proteins/genetics , Polymerase Chain Reaction/methods , Animals , Cloning, Molecular , Cosmids , Crosses, Genetic , Genome , In Situ Hybridization, Fluorescence , Mice , Polymorphism, Genetic
17.
Proc Natl Acad Sci U S A ; 97(14): 7841-6, 2000 Jul 05.
Article in English | MEDLINE | ID: mdl-10859365

ABSTRACT

The deposition of protein aggregates in neurons is a hallmark of neurodegenerative diseases caused by polyglutamine (polyQ) proteins. We analyzed the effects of the heat shock protein (Hsp) 70 chaperone system on the aggregation of fragments of huntingtin (htt) with expanded polyQ tracts. In vitro, Hsp70 and its cochaperone Hsp40 suppressed the assembly of htt into detergent-insoluble amyloid-like fibrils in an ATP-dependent manner and caused the formation of amorphous, detergent-soluble aggregates. The chaperones were most active in preventing fibrillization when added during the lag phase of the polymerization reaction. Similarly, coexpression of Hsp70 or Hsp40 with htt in yeast inhibited the formation of large, detergent-insoluble polyQ aggregates, resulting in the accumulation of detergent-soluble inclusions. Thus, the recently established potency of Hsp70 and Hsp40 to repress polyQ-induced neurodegeneration may be based on the ability of these chaperones to shield toxic forms of polyQ proteins and to direct them into nontoxic aggregates.


Subject(s)
Amyloid/metabolism , Chaperonins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Peptides/metabolism , Amino Acid Sequence , Amyloid/ultrastructure , Chaperonin 60/metabolism , Exons , HSP40 Heat-Shock Proteins , Heat-Shock Proteins/metabolism , Humans , Huntingtin Protein , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/ultrastructure , Nuclear Proteins/genetics , Nuclear Proteins/ultrastructure , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/ultrastructure , Peptides/genetics , Protein Binding , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Trinucleotide Repeat Expansion
18.
Proc Natl Acad Sci U S A ; 97(12): 6739-44, 2000 Jun 06.
Article in English | MEDLINE | ID: mdl-10829068

ABSTRACT

The accumulation of insoluble protein aggregates in intra and perinuclear inclusions is a hallmark of Huntington's disease (HD) and related glutamine-repeat disorders. A central question is whether protein aggregation plays a direct role in the pathogenesis of these neurodegenerative diseases. Here we show by using a filter retardation assay that the mAb 1C2, which specifically recognizes the elongated polyglutamine (polyQ) stretch in huntingtin, and the chemical compounds Congo red, thioflavine S, chrysamine G, and Direct fast yellow inhibit HD exon 1 protein aggregation in a dose-dependent manner. On the other hand, potential inhibitors of amyloid-beta formation such as thioflavine T, gossypol, melatonin, and rifampicin had little or no inhibitory effect on huntingtin aggregation in vitro. The results obtained by the filtration assay were confirmed by electron microscopy, SDS/PAGE, and MS. Furthermore, cell culture studies revealed that the Congo red dye at micromolar concentrations reduced the extent of HD exon 1 aggregation in transiently transfected COS cells. Together, these findings contribute to a better understanding of the mechanism of huntingtin fibrillogenesis in vitro and provide the basis for the development of new huntingtin aggregation inhibitors that may be effective in treating HD.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Huntington Disease/therapy , Nerve Tissue Proteins/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Peptides/antagonists & inhibitors , Animals , Benzoates/pharmacology , Benzothiazoles , Biphenyl Compounds/pharmacology , COS Cells , Congo Red/pharmacology , Gossypol/pharmacology , Humans , Huntingtin Protein , Melatonin/pharmacology , Rifampin/pharmacology , Thiazoles/pharmacology
19.
Exp Neurol ; 156(1): 92-9, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10192780

ABSTRACT

Intranuclear inclusion bodies are a shared pathological feature of Huntington's disease (HD) and its transgenic mouse model. Using a panel of antibodies spanning the entire huntingtin molecule, we have investigated the pattern of immunoreactivity within the intranuclear inclusions in the frontal cortex and striatum of patients with HD. The intranuclear inclusions reacted with anti-ubiquitin and antibodies against the N-terminal portion of huntingtin (CAG53b, HP1), but not with HD1 and the 1C2 antibodies that detect the expanded polyglutamine tract nor the more C-terminal antibodies. However, the 1C2, HP1, CAG53b, and HD1 antibodies detected granular cytoplasmic deposits in cortical and striatal neurons that also contained intranuclear N-terminal huntingtin immunoreactivity. These data show a differential intracellular location of truncated huntingtin in the HD brain. Both the cytoplasmic and the nuclear aggregates of the protein fragments could be neurotoxic. The frequency of the cortical intranuclear inclusions correlated with the size of CAG expansion and was inversely related to the age at onset and death. No such correlations were detected for the striatum, which most likely reflects a more advanced neuronal loss accrued by the time of death.


Subject(s)
Huntington Disease/metabolism , Inclusion Bodies/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Adult , Aged , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Epitopes , Female , Humans , Huntingtin Protein , Huntington Disease/pathology , Immunohistochemistry , Inclusion Bodies/pathology , Male , Middle Aged , Nerve Tissue Proteins/immunology , Neurites/metabolism , Neurites/pathology , Nuclear Proteins/immunology , Trinucleotide Repeats
20.
Hum Mol Genet ; 10(21): 2425-35, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11689489

ABSTRACT

Huntington's disease (HD) is a progressive neurological disorder caused by a CAG/polyglutamine repeat expansion. We have previously generated the R6/2 mouse model that expresses exon 1 of the human HD gene containing CAG repeats in excess of 150. These mice develop a progressive neurological phenotype with a rapid onset and progression. We show here that it is impossible to establish fibroblast lines from these mice at 12 weeks of age, whilst this can be achieved without difficulty at 6 and 9 weeks. Cultures derived from mice at 12 weeks contained a high frequency of dysmorphic cells, including cells with an aberrant nuclear morphology and a high frequency of micronuclei and large vacuoles. All of these features were also present in a line derived from a juvenile HD patient. Fibroblast lines derived from R6/2 mice and from HD patients were found to have a high frequency of multiple centrosomes which could account for all of the observed phenotypes including a reduced mitotic index, high frequency of aneuploidy and persistence of the midbody. We were unable to detect large insoluble polyglutamine aggregates in either the mouse or human lines. We have identified a novel progressive HD pathology that occurs in cells of non-central nervous system origin. An investigation of the pathological consequences of the HD mutation in these cells will provide insight into cellular basis of the disease.


Subject(s)
Centrosome/metabolism , Fibroblasts/metabolism , Huntington Disease/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Aneuploidy , Animals , Blotting, Western , Brain/metabolism , Cell Line , Cell Nucleus/metabolism , Cellular Senescence/genetics , Cytoplasm/metabolism , Cytoskeleton/metabolism , DNA Replication/genetics , Endocytosis , Endosomes/metabolism , Female , Fibroblasts/cytology , Humans , Huntingtin Protein , Huntington Disease/pathology , Lysosomes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Microscopy, Fluorescence , Mitotic Index , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Trinucleotide Repeat Expansion/genetics , Trinucleotide Repeats/genetics
SELECTION OF CITATIONS
SEARCH DETAIL