Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
ACS Med Chem Lett ; 10(11): 1524-1529, 2019 Nov 14.
Article in English | MEDLINE | ID: mdl-31749905

ABSTRACT

Small molecules that inhibit the metabolic enzyme NAMPT have emerged as potential therapeutics in oncology. As part of our effort in this area, we took a scaffold morphing approach and identified 3-pyridyl azetidine ureas as a potent NAMPT inhibiting motif. We explored the SAR of this series, including 5 and 6 amino pyridines, using a convergent synthetic strategy. This lead optimization campaign yielded multiple compounds with excellent in vitro potency and good ADME properties that culminated in compound 27.

2.
Bioorg Med Chem Lett ; 28(3): 365-370, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29275937

ABSTRACT

Nicotinamide phosphoribosyltransferase is a key metabolic enzyme that is a potential target for oncology. Utilizing publicly available crystal structures of NAMPT and in silico docking of our internal compound library, a NAMPT inhibitor, 1, obtained from a phenotypic screening effort was replaced with a more synthetically tractable scaffold. This compound then provided an excellent foundation for further optimization using crystallography driven structure based drug design. From this approach, two key motifs were identified, the (S,S) cyclopropyl carboxamide and the (S)-1-N-phenylethylamide that endowed compounds with excellent cell based potency. As exemplified by compound 27e such compounds could be useful tools to explore NAMPT biology in vivo.


Subject(s)
Amides/pharmacology , Cyclopropanes/pharmacology , Cytokines/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/pharmacology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Adenosine/analogs & derivatives , Amides/chemical synthesis , Amides/chemistry , Crystallography, X-Ray , Cyclopropanes/chemical synthesis , Cyclopropanes/chemistry , Cytokines/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Docking Simulation , Molecular Structure , Nicotinamide Phosphoribosyltransferase/metabolism , Phenotype , Structure-Activity Relationship
3.
Sci Rep ; 7: 42728, 2017 02 16.
Article in English | MEDLINE | ID: mdl-28205648

ABSTRACT

Chemogenomic profiling is a powerful and unbiased approach to elucidate pharmacological targets and the mechanism of bioactive compounds. Until recently, genome-wide, high-resolution experiments of this nature have been limited to fungal systems due to lack of mammalian genome-wide deletion collections. With the example of a novel nicotinamide phosphoribosyltransferase (NAMPT) inhibitor, we demonstrate that the CRISPR/Cas9 system enables the generation of transient homo- and heterozygous deletion libraries and allows for the identification of efficacy targets and pathways mediating hypersensitivity and resistance relevant to the compound mechanism of action.


Subject(s)
CRISPR-Cas Systems , Drug Discovery/methods , Enzyme Inhibitors/pharmacology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Cells, Cultured , Enzyme Inhibitors/chemistry , Gene Deletion , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , Pharmacogenomic Testing/methods
4.
J Biomol Screen ; 18(4): 407-19, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23150017

ABSTRACT

Translation initiation is a fine-tuned process that plays a critical role in tumorigenesis. The use of small molecules that modulate mRNA translation provides tool compounds to explore the mechanism of translational initiation and to further validate protein synthesis as a potential pharmaceutical target for cancer therapeutics. This report describes the development and use of a click beetle, dual luciferase cell-based assay multiplexed with a measure of compound toxicity using resazurin to evaluate the differential effect of natural products on cap-dependent or internal ribosome entry site (IRES)-mediated translation initiation and cell viability. This screen identified a series of cardiac glycosides as inhibitors of IRES-mediated translation using, in particular, the oncogene mRNA c-Myc IRES. Treatment of c-Myc-dependent cancer cells with these compounds showed a decrease in c-Myc protein associated with a significant modulation of cell viability. These findings suggest that inhibition of IRES-mediated translation initiation may be a strategy to inhibit c-Myc-driven tumorigenesis.


Subject(s)
Cardiac Glycosides/analysis , Cardiac Glycosides/pharmacology , Drug Evaluation, Preclinical , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Ribosomes/metabolism , Apoptosis/drug effects , Base Sequence , Biological Assay , Cardiac Glycosides/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cymarine/chemistry , Cymarine/pharmacology , DNA Damage , Genes, Reporter , HEK293 Cells , Humans , Inhibitory Concentration 50 , Protein Synthesis Inhibitors/analysis , Protein Synthesis Inhibitors/chemistry , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Ribosomes/drug effects , Vascular Endothelial Growth Factor A/metabolism
5.
Hum Mol Genet ; 19(18): 3502-15, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20584926

ABSTRACT

Early-onset torsion dystonia is the most severe heritable form of dystonia, a human movement disorder that typically starts during a developmental window in early adolescence. Deletion in the DYT1 gene, encoding the torsinA protein, is responsible for this dominantly inherited disorder, which is non-degenerative and exhibits reduced penetrance among carriers. Here, we explore the hypothesis that deficits in torsinA function result in an increased vulnerability to stress associated with protein folding and processing in the endoplasmic reticulum (ER), where torsinA is located. Using an in vivo quantitative readout for the ER stress response, we evaluated the consequences of torsinA mutations in transgenic nematodes expressing variants of human torsinA. This analysis revealed that, normally, torsinA serves a protective function to maintain a homeostatic threshold against ER stress. Furthermore, we show that the buffering capacity of torsinA is greatly diminished by the DYT1-associated deletion or mutations that prevent its translocation to the ER, block ATPase activity, or increase the levels of torsinA in the nuclear envelope versus ER. Combinations of transgenic Caenorhabditis elegans designed to mimic clinically relevant genetic modifiers of disease susceptibility also exhibit a direct functional correlation to changes in the ER stress response. Furthermore, using mouse embryonic fibroblasts (MEFs) from torsinA knockout mice, we demonstrated that loss of endogenous torsinA results in enhanced sensitivity to ER stress. This study extends our understanding of molecular mechanisms underlying dystonia, and establishes a new functional paradigm to evaluate therapeutic strategies to compensate for reduced torsinA activity in the ER as a means to restore homeostatic balance and neuronal function.


Subject(s)
Dystonia/metabolism , Endoplasmic Reticulum/metabolism , Homeostasis , Molecular Chaperones/metabolism , Age of Onset , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Cells, Cultured , Disease Models, Animal , Dystonia/genetics , Dystonia/physiopathology , Endoplasmic Reticulum/genetics , Fibroblasts/metabolism , Humans , Mice , Mice, Knockout , Molecular Chaperones/genetics , Protein Transport , Stress, Physiological
6.
Dis Model Mech ; 3(5-6): 386-96, 2010.
Article in English | MEDLINE | ID: mdl-20223934

ABSTRACT

Movement disorders represent a significant societal burden for which therapeutic options are limited and focused on treating disease symptomality. Early-onset torsion dystonia (EOTD) is one such disorder characterized by sustained and involuntary muscle contractions that frequently cause repetitive movements or abnormal postures. Transmitted in an autosomal dominant manner with reduced penetrance, EOTD is caused in most cases by the deletion of a glutamic acid (DeltaE) in the DYT1 (also known as TOR1A) gene product, torsinA. Although some patients respond well to anticholingerics, therapy is primarily limited to either neurosurgery or chemodenervation. As mutant torsinA (DeltaE) expression results in decreased torsinA function, therapeutic strategies directed toward enhancement of wild-type (WT) torsinA activity in patients who are heterozygous for mutant DYT1 may restore normal cellular functionality. Here, we report results from the first-ever screen for candidate small molecule therapeutics for EOTD, using multiple activity-based readouts for torsinA function in Caenorhabditis elegans, subsequent validation in human DYT1 patient fibroblasts, and behavioral rescue in a mouse model of DYT1 dystonia. We exploited the nematode to rapidly discern chemical effectors of torsinA and identified two classes of antibiotics, quinolones and aminopenicillins, which enhance WT torsinA activity in two separate in vivo assays. Representative molecules were assayed in EOTD patient fibroblasts for improvements in torsinA-dependent secretory function, which was improved significantly by ampicillin. Furthermore, a behavioral defect associated with an EOTD mouse knock-in model was also rescued following administration of ampicillin. These combined data indicate that specific small molecules that enhance torsinA activity represent a promising new approach toward therapeutic development for EOTD, and potentially for other diseases involving the processing of mutant proteins.


Subject(s)
Disease Models, Animal , Dystonia Musculorum Deformans/metabolism , Molecular Chaperones/metabolism , Small Molecule Libraries/pharmacology , Age of Onset , Ampicillin/chemistry , Ampicillin/pharmacology , Animals , Animals, Genetically Modified , Behavior, Animal/drug effects , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/genetics , Drug Evaluation, Preclinical , Dystonia Musculorum Deformans/genetics , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Mice , Models, Molecular , Neuroprotective Agents/pharmacology , Reproducibility of Results , Small Molecule Libraries/analysis , Structure-Activity Relationship
7.
J Cell Sci ; 121(Pt 20): 3476-86, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18827015

ABSTRACT

A specific mutation (DeltaE) in torsinA underlies most cases of the dominantly inherited movement disorder, early-onset torsion dystonia (DYT1). TorsinA, a member of the AAA+ ATPase superfamily, is located within the lumen of the nuclear envelope (NE) and endoplasmic reticulum (ER). We investigated an association between torsinA and nesprin-3, which spans the outer nuclear membrane (ONM) of the NE and links it to vimentin via plectin in fibroblasts. Mouse nesprin-3alpha co-immunoprecipitated with torsinA and this involved the C-terminal region of torsinA and the KASH domain of nesprin-3alpha. This association with human nesprin-3 appeared to be stronger for torsinADeltaE than for torsinA. TorsinA also associated with the KASH domains of nesprin-1 and -2 (SYNE1 and 2), which link to actin. In the absence of torsinA, in knockout mouse embryonic fibroblasts (MEFs), nesprin-3alpha was localized predominantly in the ER. Enrichment of yellow fluorescent protein (YFP)-nesprin-3 in the ER was also seen in the fibroblasts of DYT1 patients, with formation of YFP-positive globular structures enriched in torsinA, vimentin and actin. TorsinA-null MEFs had normal NE structure, but nuclear polarization and cell migration were delayed in a wound-healing assay, as compared with wild-type MEFs. These studies support a role for torsinA in dynamic interactions between the KASH domains of nesprins and their protein partners in the lumen of the NE, with torsinA influencing the localization of nesprins and associated cytoskeletal elements and affecting their role in nuclear and cell movement.


Subject(s)
Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Molecular Chaperones/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Actins/genetics , Actins/metabolism , Animals , Cell Line , Cell Movement/genetics , Cytoskeletal Proteins , Dystonia Musculorum Deformans/genetics , Dystonia Musculorum Deformans/metabolism , Embryo, Mammalian/metabolism , Endoplasmic Reticulum/genetics , Fibroblasts/metabolism , Humans , Membrane Proteins/genetics , Mice , Mice, Knockout , Microfilament Proteins/genetics , Molecular Chaperones/genetics , Mutation , Nerve Tissue Proteins/genetics , Nuclear Envelope/genetics , Nuclear Proteins/genetics , Plectin/genetics , Plectin/metabolism , Protein Structure, Tertiary/physiology , Vimentin/genetics , Vimentin/metabolism , Wound Healing/genetics
8.
Hum Mol Genet ; 17(10): 1436-45, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18258738

ABSTRACT

Most cases of the dominantly inherited movement disorder, early onset torsion dystonia (DYT1) are caused by a mutant form of torsinA lacking a glutamic acid residue in the C-terminal region (torsinADeltaE). TorsinA is an AAA+ protein located predominantly in the lumen of the endoplasmic reticulum (ER) and nuclear envelope apparently involved in membrane structure/movement and processing of proteins through the secretory pathway. A reporter protein Gaussia luciferase (Gluc) shows a reduced rate of secretion in primary fibroblasts from DYT1 patients expressing endogenous levels of torsinA and torsinADeltaE when compared with control fibroblasts expressing only torsinA. In this study, small interfering RNA (siRNA) oligonucleotides were identified, which downregulate the levels of torsinA or torsinADeltaE mRNA and protein by over 65% following transfection. Transfection of siRNA for torsinA message in control fibroblasts expressing Gluc reduced levels of luciferase secretion compared with the same cells non-transfected or transfected with a non-specific siRNA. Transfection of siRNA selectively inhibiting torsinADeltaE message in DYT fibroblasts increased luciferase secretion when compared with cells non-transfected or transfected with a non-specific siRNA. Further, transduction of DYT1 cells with a lentivirus vector expressing torsinA, but not torsinB, also increased secretion. These studies are consistent with a role for torsinA as an ER chaperone affecting processing of proteins through the secretory pathway and indicate that torsinADeltaE acts to inhibit this torsinA activity. The ability of allele-specific siRNA for torsinADeltaE to normalize secretory function in DYT1 patient cells supports its potential role as a therapeutic agent in early onset torsion dystonia.


Subject(s)
Dystonia Musculorum Deformans/genetics , Gene Silencing , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Mutation , Protein Processing, Post-Translational , RNA, Small Interfering/genetics , Base Sequence , Cell Culture Techniques , Cells, Cultured , Dystonia Musculorum Deformans/metabolism , Dystonia Musculorum Deformans/pathology , Fibroblasts/metabolism , Genes, Reporter , Humans , Luciferases/metabolism , Molecular Chaperones/chemistry , Molecular Sequence Data , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection
9.
PLoS One ; 2(6): e571, 2007 Jun 27.
Article in English | MEDLINE | ID: mdl-17593970

ABSTRACT

BACKGROUND: The secretory pathway is a critical index of the capacity of cells to incorporate proteins into cellular membranes and secrete proteins into the extracellular space. Importantly it is disrupted in response to stress to the endoplasmic reticulum that can be induced by a variety of factors, including expression of mutant proteins and physiologic stress. Activation of the ER stress response is critical in the etiology of a number of diseases, such as diabetes and neurodegeneration, as well as cancer. We have developed a highly sensitive assay to monitor processing of proteins through the secretory pathway and endoplasmic reticulum (ER) stress in real-time based on the naturally secreted Gaussia luciferase (Gluc). METHODOLOGY/PRINCIPLE FINDINGS: An expression cassette for Gluc was delivered to cells, and its secretion was monitored by measuring luciferase activity in the conditioned medium. Gluc secretion was decreased down to 90% when these cells were treated with drugs that interfere with the secretory pathway at different steps. Fusing Gluc to a fluorescent protein allowed quantitation and visualization of the secretory pathway in real-time. Expression of this reporter protein did not itself elicit an ER stress response in cells; however, Gluc proved very sensitive at sensing this type of stress, which is associated with a temporary decrease in processing of proteins through the secretory pathway. The Gluc secretion assay was over 20,000-fold more sensitive as compared to the secreted alkaline phosphatase (SEAP), a well established assay for monitoring of protein processing and ER stress in mammalian cells. CONCLUSIONS/SIGNIFICANCE: The Gluc assay provides a fast, quantitative and sensitive technique to monitor the secretory pathway and ER stress and its compatibility with high throughput screening will allow discovery of drugs for treatment of conditions in which the ER stress is generally induced.


Subject(s)
Biological Assay , Endoplasmic Reticulum/physiology , Luciferases/metabolism , Secretory Pathway , Alkaline Phosphatase/metabolism , Animals , Blotting, Western , Copepoda/enzymology , Endoplasmic Reticulum/drug effects , Humans , Immunoenzyme Techniques , Kidney/cytology , Kidney/metabolism , Lentivirus/genetics , Luciferases/genetics , Luminescent Measurements , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
10.
Proc Natl Acad Sci U S A ; 104(17): 7271-6, 2007 Apr 24.
Article in English | MEDLINE | ID: mdl-17428918

ABSTRACT

TorsinA is an AAA(+) protein located predominantly in the lumen of the endoplasmic reticulum (ER) and nuclear envelope responsible for early onset torsion dystonia (DYT1). Most cases of this dominantly inherited movement disorder are caused by deletion of a glutamic acid in the carboxyl terminal region of torsinA. We used a sensitive reporter, Gaussia luciferase (Gluc) to evaluate the role of torsinA in processing proteins through the ER. In primary fibroblasts from controls and DYT1 patients most Gluc activity (95%) was released into the media and processed through the secretory pathway, as confirmed by inhibition with brefeldinA and nocodazole. Fusion of Gluc to a fluorescent protein revealed coalignment and fractionation with ER proteins and association of Gluc with torsinA. Notably, fibroblasts from DYT1 patients were found to secrete markedly less Gluc activity as compared with control fibroblasts. This decrease in processing of Gluc in DYT1 cells appear to arise, at least in part, from a loss of torsinA activity, because mouse embryonic fibroblasts lacking torsinA also had reduced secretion as compared with control cells. These studies demonstrate the exquisite sensitivity of this reporter system for quantitation of processing through the secretory pathway and support a role for torsinA as an ER chaperone protein.


Subject(s)
Dystonia/metabolism , Dystonia/pathology , Molecular Chaperones/metabolism , Mutant Proteins/metabolism , Protein Processing, Post-Translational , Animals , Cell Count , Endoplasmic Reticulum/metabolism , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Immunoprecipitation , Luciferases/metabolism , Mice , Protein Disulfide-Isomerases/metabolism , Recombinant Fusion Proteins/metabolism , Reproducibility of Results , Solubility , Time Factors
11.
Neurobiol Dis ; 22(1): 98-111, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16361107

ABSTRACT

Early onset torsion dystonia is a movement disorder inherited as an autosomal dominant syndrome with reduced penetrance. Symptoms appear to result from altered neuronal circuitry within the brain with no evidence of neuronal loss. Most cases are caused by loss of a glutamic acid residue in the AAA+ chaperone protein, torsinA, encoded in the DYT1 gene. In this study, torsinA was found to move in conjunction with vimentin in three cell culture paradigms-recovery from microtubule depolymerization, expression of a dominant-negative form of kinesin light chain and respreading after trypsinization. Co-immune precipitation studies revealed association between vimentin and torsinA in a complex including other cytoskeletal elements, actin and tubulin, as well as two proteins previously shown to interact with torsinA-the motor protein, kinesin light chain 1, and the nuclear envelope protein, LAP1. Morphologic and functional differences related to vimentin were noted in primary fibroblasts from patients carrying this DYT1 mutation as compared with controls, including an increased perinuclear concentration of vimentin and a delayed rate of adhesion to the substratum. Overexpression of mutant torsinA inhibited neurite extension in human neuroblastoma cells, with torsinA and vimentin immunoreactivity enriched in the perinuclear region and in cytoplasmic inclusions. Collectively, these studies suggest that mutant torsinA interferes with cytoskeletal events involving vimentin, possibly by restricting movement of these particles/filaments, and hence may affect development of neuronal pathways in the brain.


Subject(s)
Brain/metabolism , Cytoskeleton/metabolism , Molecular Chaperones/metabolism , Mutation/physiology , Neurites/metabolism , Vimentin/metabolism , Actins/metabolism , Brain/physiopathology , Cell Adhesion/physiology , Cell Line , Cell Line, Tumor , Cell Shape/physiology , Cells, Cultured , Cytoplasm/metabolism , Cytoplasm/ultrastructure , Cytoplasmic Streaming/physiology , Dystonia Musculorum Deformans/metabolism , Dystonia Musculorum Deformans/physiopathology , Fibroblasts/metabolism , Fibroblasts/ultrastructure , HSC70 Heat-Shock Proteins/metabolism , Humans , Kinesins , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Molecular Chaperones/genetics , Neurites/ultrastructure , Tubulin/metabolism
12.
J Neurochem ; 89(5): 1186-94, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15147511

ABSTRACT

The torsins comprise a four-member family of AAA+ chaperone proteins, including torsinA, torsinB, torp2A and torp3A in humans. Mutations in torsinA underlie early onset torsion dystonia, an autosomal dominant, neurologically based movement disorder. TorsinB is highly homologous to torsinA with its gene adjacent to that for torsinA on human chromosome 9q34. Antibodies have been generated which can distinguish torsinA and torsinB from each other, and from the torps in human and rodent cells. TorsinB (approximately MW 38 kDa), like torsinA ( approximately MW 37 kDa), is an N-glycosylated protein and both reside primarily in the endoplasmic reticulum (ER) and nuclear envelope in cultured cells. Immunoprecipitation studies in cultured cells and human brain tissue indicate that torsinA and torsinB are associated with each other in cells. Overexpression of both wild-type torsinB and mutant torsinA lead to enrichment of the protein in the nuclear envelope and formation of large cytoplasmic inclusions. We conclude that torsinB and torsinA are localized in overlapping cell compartments within the same protein complex, and thus may carry out related functions in vivo.


Subject(s)
Brain/metabolism , Carrier Proteins/metabolism , Molecular Chaperones/metabolism , Neuroblastoma/metabolism , Nuclear Envelope/metabolism , Animals , Antibody Specificity , Blotting, Western , Brain/cytology , Brain Chemistry , Carrier Proteins/analysis , Cell Compartmentation/physiology , Endoplasmic Reticulum/metabolism , Glycosylation , Humans , Immunohistochemistry , Kidney/cytology , Kidney/metabolism , Mice , Molecular Chaperones/analysis , Neuroblastoma/pathology , Nuclear Envelope/ultrastructure , Precipitin Tests
13.
J Biol Chem ; 279(19): 19882-92, 2004 May 07.
Article in English | MEDLINE | ID: mdl-14970196

ABSTRACT

Early onset dystonia is a movement disorder caused by loss of a glutamic acid residue (Glu(302/303)) in the carboxyl-terminal portion of the AAA+ protein, torsinA. We identified the light chain subunit (KLC1) of kinesin-I as an interacting partner for torsinA, with binding occurring between the tetratricopeptide repeat domain of KLC1 and the carboxyl-terminal region of torsinA. Coimmunoprecipitation analysis demonstrated that wild-type torsinA and kinesin-I form a complex in vivo. In cultured cortical neurons, both proteins co-localized along processes with enrichment at growth cones. Wild-type torsinA expressed in CAD cells co-localized with endogenous KLC1 at the distal end of processes, whereas mutant torsinA remained confined to the cell body. Subcellular fractionation of adult rat brain revealed torsinA and KLC associated with cofractionating membranes, and both proteins were co-immunoprecipitated after cross-linking cytoplasmically oriented proteins on isolated rat brain membranes. These studies suggest that wild-type torsinA undergoes anterograde transport along microtubules mediated by kinesin and may act as a molecular chaperone regulating kinesin activity and/or cargo binding.


Subject(s)
Carrier Proteins/chemistry , Microtubule-Associated Proteins/chemistry , Molecular Chaperones/chemistry , Adenine/analogs & derivatives , Adenine/metabolism , Amino Acid Sequence , Animals , Biotinylation , Brain/metabolism , Carrier Proteins/metabolism , Cell Line , Cross-Linking Reagents/pharmacology , Cytoplasm/metabolism , Dystonia/metabolism , Glutathione Transferase/metabolism , Humans , Kinesins/chemistry , Kinetin , Microscopy, Fluorescence , Microtubule-Associated Proteins/metabolism , Molecular Chaperones/metabolism , Molecular Sequence Data , Mutation , Neurons/cytology , Neurons/metabolism , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Isoforms , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Streptavidin/pharmacology , Subcellular Fractions/metabolism , Transfection , Two-Hybrid System Techniques , beta-Galactosidase/metabolism
14.
Brain Res ; 986(1-2): 12-21, 2003 Oct 03.
Article in English | MEDLINE | ID: mdl-12965225

ABSTRACT

We have examined the distribution and ultrastructural localization of torsinA, the protein product of the TOR1A gene, in the normal adult human and Macaque brain. TorsinA immunoreactivity was visualized using a monoclonal antibody raised against a fusion protein encoding exon 4 of human torsinA. Western blot analysis of brain homogenates revealed a major species of about 39 kDa, consistent with the predicted size of glycosylated torsinA protein. By light microscopy, torsinA like-immunoreactivity was enriched in gray matter in all brain regions examined. Immunoreactivity was concentrated in the neuropil and immunopositive cell bodies were not observed. Structures particularly enriched in torsinA like-immunoreactivity included the cerebral cortex, the caudate-putamen, globus pallidus, the hippocampal formation, the thalamus, the substantia nigra and molecular cell layer of the cerebellar cortex. Cell bodies of pigmented dopamine neurons in the substantia nigra pars compacta were immunonegative. Biochemical fractionation of the human striata revealed a concentration of torsinA immunoreactivity in particulate fractions. Ultrastructural studies of the human and Macaque striata further revealed an association of torsinA immunostaining with small vesicles within axons and presynaptic terminals forming symmetric synapses. These ultrastructural studies are consistent with a pre-synaptic localization of torsinA protein in the adult striatum and are consistent with a role of torsinA in modulating striatal signaling, although the widespread localization of the protein suggests it probably also participates in signaling in other regions.


Subject(s)
Brain/metabolism , Carrier Proteins/metabolism , Molecular Chaperones , Presynaptic Terminals/metabolism , Animals , Brain/ultrastructure , Dendrites/metabolism , Dendrites/ultrastructure , Humans , Immunohistochemistry , Macaca fascicularis , Microscopy, Electron , Neostriatum/metabolism , Neostriatum/ultrastructure , Presynaptic Terminals/ultrastructure , Recombinant Fusion Proteins/immunology , Synaptic Transmission/physiology , Synaptic Vesicles/metabolism , Synaptic Vesicles/ultrastructure
15.
J Neurosci Res ; 72(2): 158-68, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12671990

ABSTRACT

Most cases of early-onset torsion dystonia are caused by deletion of GAG in the coding region of the DYT1 gene encoding torsinA. This autosomal dominant neurologic disorder is characterized by abnormal movements, believed to originate from neuronal dysfunction in the basal ganglia of the human brain. The torsins (torsinA and torsinB) are members of the "ATPases associated with a variety of cellular activities" (AAA(+)) superfamily of proteins that mediate chaperone and other functions involved in conformational modeling of proteins, protection from stress, and targeting of proteins to cellular organelles. In this study, the intracellular localization and levels of endogenous torsin were evaluated in rat pheochromocytoma PC12 cells following differentiation and stress. TorsinA, apparent MW 37 kDa, cofractionates with markers for the microsomal/endoplasmic reticulum (ER) compartment and appears to reside primarily within the ER lumen based on protease resistance. TorsinA immunoreactivity colocalizes with the lumenal ER protein protein disulfide isomerase (PDI) and extends throughout neurites. Levels of torsinA did not increase notably in response to nerve growth factor-induced differentiation. None of the stress conditions tested, including heat shock and the unfolded protein response, affected torsinA, except for oxidative stress, which resulted in an increase in the apparent MW of torsinA and redistribution to protrusions from the cell surface. These findings are consistent with a relatively rapid covalent modification of torsinA in response to oxidative stress causing a change in state. Mutant torsinA may interfere with and/or compromise ER functions, especially in dopaminergic neurons, which have high levels of torsinA and are intrinsically vulnerable to oxidative stress.


Subject(s)
Carrier Proteins/metabolism , Endoplasmic Reticulum/metabolism , Molecular Chaperones , PC12 Cells/metabolism , Animals , Blotting, Northern , Blotting, Western , Carrier Proteins/genetics , Cell Differentiation/physiology , Cytoplasm/metabolism , Heat-Shock Response , Immunohistochemistry , Nerve Growth Factor/metabolism , Oxidative Stress/physiology , PC12 Cells/cytology , Rats , Tumor Cells, Cultured
16.
Neurobiol Dis ; 12(1): 11-24, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12609485

ABSTRACT

Familial, early onset, generalized torsion dystonia is the most common and severe primary dystonia. Most cases are caused by a 3-bp deletion (GAG) in the coding region of the TOR1A (DYT1) gene, which is widely expressed in human brain and encodes the protein torsinA. This study compares neuropathology and torsinA expression in the normal human brain with that in dystonia cases with and without the GAG deletion. TorsinA-like protein was expressed in neuronal cytoplasm throughout the human brain, including cerebellum, substantia nigra, hippocampus, and neostriatum, with higher levels in specific neurons. This immunostaining pattern was not discernibly different in dystonia and normal brains in midbrain and neostriatal regions. However, nigral dopaminergic neurons appeared to be larger in both GAG-deletion and non-GAG-deletion dystonia brains compared to normal, and may be more closely spaced in GAG-deletion brains. Beyond these apparent changes in neuronal size and spacing in dystonia brains, there was no indication of neuron loss, inflammation, DNA strand breaks, or altered distribution of torsin-like immunoreactivity, supporting a functional rather than degenerative etiology of early onset torsion dystonia.


Subject(s)
Brain/metabolism , Brain/pathology , Carrier Proteins/metabolism , Dystonia Musculorum Deformans/metabolism , Dystonia Musculorum Deformans/pathology , Gene Deletion , Molecular Chaperones , Neurons/metabolism , Neurons/pathology , Adolescent , Adult , Aged , Brain/physiopathology , Carrier Proteins/genetics , Dystonia Musculorum Deformans/genetics , Female , Genotype , Humans , Immunohistochemistry , Infant , Male , Middle Aged , Mutation/genetics
17.
J Neurochem ; 83(4): 846-54, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12421356

ABSTRACT

TorsinA, a protein with homology to yeast heat shock protein104, has previously been demonstrated to colocalize with alpha-synuclein in Lewy bodies, the pathological hallmark of Parkinson's disease. Heat shock proteins are a family of chaperones that are both constitutively expressed and induced by stressors, and that serve essential functions for protein refolding and/or degradation. Here, we demonstrate that, like torsinA, specific molecular chaperone heat shock proteins colocalize with alpha-synuclein in Lewy bodies. In addition, using a cellular model of alpha-synuclein aggregation, we demonstrate that torsinA and specific heat shock protein molecular chaperones colocalize with alpha-synuclein immunopositive inclusions. Further, overexpression of torsinA and specific heat shock proteins suppress alpha-synuclein aggregation in this cellular model, whereas mutant torsinA has no effect. These data suggest that torsinA has chaperone-like activity and that the disease-associated GAG deletion mutant has a loss-of-function phenotype. Moreover, these data support a role for chaperone proteins, including torsinA and heat shock proteins, in cellular responses to neurodegenerative inclusions.


Subject(s)
Carrier Proteins/metabolism , Heat-Shock Proteins/metabolism , Lewy Body Disease/pathology , Molecular Chaperones/metabolism , Nerve Tissue Proteins/metabolism , Alzheimer Disease/pathology , Carrier Proteins/genetics , Cell Line , Gene Expression , Heat-Shock Proteins/genetics , Humans , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Lewy Bodies/metabolism , Lewy Bodies/pathology , Macromolecular Substances , Molecular Chaperones/genetics , Nerve Tissue Proteins/genetics , Protein Binding/physiology , Protein Folding , Substantia Nigra/pathology , Synucleins , Transfection , alpha-Synuclein
SELECTION OF CITATIONS
SEARCH DETAIL
...