Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Brain ; 143(6): 1746-1765, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32516804

RESUMO

TOR1A/TorsinA mutations cause two incurable diseases: a recessive congenital syndrome that can be lethal, and a dominantly-inherited childhood-onset dystonia (DYT-TOR1A). TorsinA has been linked to phosphatidic acid lipid metabolism in Drosophila melanogaster. Here we evaluate the role of phosphatidic acid phosphatase (PAP) enzymes in TOR1A diseases using induced pluripotent stem cell-derived neurons from patients, and mouse models of recessive Tor1a disease. We find that Lipin PAP enzyme activity is abnormally elevated in human DYT-TOR1A dystonia patient cells and in the brains of four different Tor1a mouse models. Its severity also correlated with the dosage of Tor1a/TOR1A mutation. We assessed the role of excess Lipin activity in the neurological dysfunction of Tor1a disease mouse models by interbreeding these with Lpin1 knock-out mice. Genetic reduction of Lpin1 improved the survival of recessive Tor1a disease-model mice, alongside suppressing neurodegeneration, motor dysfunction, and nuclear membrane pathology. These data establish that TOR1A disease mutations cause abnormal phosphatidic acid metabolism, and suggest that approaches that suppress Lipin PAP enzyme activity could be therapeutically useful for TOR1A diseases.


Assuntos
Chaperonas Moleculares/metabolismo , Fosfatidato Fosfatase/metabolismo , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Distonia/genética , Distonia/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Chaperonas Moleculares/genética , Mutação , Neurônios/metabolismo , Fosfatidato Fosfatase/genética , Fosfatidato Fosfatase/fisiologia
2.
EMBO Mol Med ; 11(1)2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30552094

RESUMO

Dopamine D2 receptor signaling is central for striatal function and movement, while abnormal activity is associated with neurological disorders including the severe early-onset DYT1 dystonia. Nevertheless, the mechanisms that regulate D2 receptor signaling in health and disease remain poorly understood. Here, we identify a reduced D2 receptor binding, paralleled by an abrupt reduction in receptor protein level, in the striatum of juvenile Dyt1 mice. This occurs through increased lysosomal degradation, controlled by competition between ß-arrestin 2 and D2 receptor binding proteins. Accordingly, we found lower levels of striatal RGS9-2 and spinophilin. Further, we show that genetic depletion of RGS9-2 mimics the D2 receptor loss of DYT1 dystonia striatum, whereas RGS9-2 overexpression rescues both receptor levels and electrophysiological responses in Dyt1 striatal neurons. This work uncovers the molecular mechanism underlying D2 receptor downregulation in Dyt1 mice and in turn explains why dopaminergic drugs lack efficacy in DYT1 patients despite significant evidence for striatal D2 receptor dysfunction. Our data also open up novel avenues for disease-modifying therapeutics to this incurable neurological disorder.


Assuntos
Corpo Estriado/patologia , Distonia Muscular Deformante/patologia , Distonia Muscular Deformante/fisiopatologia , Chaperonas Moleculares/genética , Proteínas RGS/análise , Receptores de Dopamina D2/análise , Transdução de Sinais , Animais , Modelos Animais de Doenças , Expressão Gênica , Técnicas de Silenciamento de Genes , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/análise , Proteínas do Tecido Nervoso/análise , Proteínas RGS/genética
3.
Front Immunol ; 9: 2234, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30323813

RESUMO

The nucleopore is an essential structure of the eukaryotic cell, regulating passage between the nucleus and cytoplasm. While individual functions of core nucleopore proteins have been identified, the role of other components, such as Nup210, are poorly defined. Here, through the use of an unbiased ENU mutagenesis screen for mutations effecting the peripheral T cell compartment, we identified a Nup210 mutation in a mouse strain with altered CD4/CD8 T cell ratios. Through the generation of Nup210 knockout mice we identified Nup210 as having a T cell-intrinsic function in the peripheral homeostasis of T cells. Remarkably, despite the deep evolutionary conservation of this key nucleopore complex member, no other major phenotypes developed, with viable and healthy knockout mice. These results identify Nup210 as an important nucleopore complex component for peripheral T cells, and raise further questions of why this nucleopore component shows deep evolutionary conservation despite seemingly redundant functions in most cell types.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Animais , Feminino , Homeostase , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Mutação , Poro Nuclear/metabolismo , Estatísticas não Paramétricas , Células Th1/metabolismo , Células Th2/metabolismo , Timócitos/metabolismo , Timo/citologia
4.
Mov Disord ; 33(1): 73, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29266403
5.
EMBO Rep ; 18(11): 1905-1921, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29074503

RESUMO

Endoplasmic reticulum (ER)-localized enzymes synthesize the vast majority of cellular lipids. The ER therefore has a major influence on cellular lipid biomass and balances the production of different lipid categories, classes, and species. Signals from outside and inside the cell are directed to ER-localized enzymes, and lipid enzyme activities are defined by the integration of internal, homeostatic, and external information. This allows ER-localized lipid synthesis to provide the cell with membrane lipids for growth, proliferation, and differentiation-based changes in morphology and structure, and to maintain membrane homeostasis across the cell. ER enzymes also respond to physiological signals to drive carbohydrates and nutritionally derived lipids into energy-storing triglycerides. In this review, we highlight some key regulatory mechanisms that control ER-localized enzyme activities in animal cells. We also discuss how they act in concert to maintain cellular lipid homeostasis, as well as how their dysregulation contributes to human disease.


Assuntos
Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Lipídeos/biossíntese , Lipogênese/genética , Triglicerídeos/biossíntese , Animais , Membrana Celular/química , Retículo Endoplasmático/ultraestrutura , Células Eucarióticas/citologia , Células Eucarióticas/metabolismo , Retroalimentação Fisiológica , Regulação da Expressão Gênica , Homeostase/genética , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas de Ligação a Elemento Regulador de Esterol/genética , Proteínas de Ligação a Elemento Regulador de Esterol/metabolismo , Resposta a Proteínas não Dobradas
6.
Neurobiol Dis ; 108: 128-139, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28823931

RESUMO

Striatal dysfunction is implicated in many movement disorders. However, the precise nature of defects often remains uncharacterized, which hinders therapy development. Here we examined striatal function in a mouse model of the incurable movement disorder, myoclonus dystonia, caused by SGCE mutations. Using RNAseq we found surprisingly normal gene expression, including normal levels of neuronal subclass markers to strongly suggest that striatal microcircuitry is spared by the disease insult. We then functionally characterized Sgce mutant medium spiny projection neurons (MSNs) and cholinergic interneurons (ChIs). This revealed normal intrinsic electrophysiological properties and normal responses to basic excitatory and inhibitory neurotransmission. Nevertheless, high-frequency stimulation in Sgce mutants failed to induce normal long-term depression (LTD) at corticostriatal glutamatergic synapses. We also found that pharmacological manipulation of MSNs by inhibiting adenosine 2A receptors (A2AR) restores LTD, again pointing to structurally intact striatal circuitry. The fact that Sgce loss specifically inhibits LTD implicates this neurophysiological defect in myoclonus dystonia, and emphasizes that neurophysiological changes can occur in the absence of broad striatal dysfunction. Further, the positive effect of A2AR antagonists indicates that this drug class be tested in DYT11/SGCE dystonia.


Assuntos
Antagonistas do Receptor A2 de Adenosina/farmacologia , Corpo Estriado/efeitos dos fármacos , Distúrbios Distônicos/tratamento farmacológico , Plasticidade Neuronal/efeitos dos fármacos , Animais , Corpo Estriado/fisiopatologia , Modelos Animais de Doenças , Distúrbios Distônicos/fisiopatologia , Feminino , Ácido Glutâmico/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Plasticidade Neuronal/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Técnicas de Patch-Clamp , RNA Mensageiro/metabolismo , Receptor A2A de Adenosina/metabolismo , Sarcoglicanas/genética , Sarcoglicanas/metabolismo , Técnicas de Cultura de Tecidos
7.
J Virol ; 91(19)2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28724767

RESUMO

Herpesvirus capsids assemble in the nucleus, while final virion maturation proceeds in the cytoplasm. This requires that newly formed nucleocapsids cross the nuclear envelope (NE), which occurs by budding at the inner nuclear membrane (INM), release of the primary enveloped virion into the perinuclear space (PNS), and subsequent rapid fusion with the outer nuclear membrane (ONM). During this process, the NE remains intact, even at late stages of infection. In addition, the spacing between the INM and ONM is maintained, as is that between the primary virion envelope and nuclear membranes. The linker of nucleoskeleton and cytoskeleton (LINC) complex consists of INM proteins with a luminal SUN (Sad1/UNC-84 homology) domain connected to ONM proteins with a KASH (Klarsicht, ANC-1, SYNE homology) domain and is thought to be responsible for spacing the nuclear membranes. To investigate the role of the LINC complex during herpesvirus infection, we generated cell lines constitutively expressing dominant negative (dn) forms of SUN1 and SUN2. Ultrastructural analyses revealed a significant expansion of the PNS and the contiguous intracytoplasmic lumen, most likely representing endoplasmic reticulum (ER), especially in cells expressing dn-SUN2. After infection, primary virions accumulated in these expanded luminal regions, also very distant from the nucleus. The importance of the LINC complex was also confirmed by reduced progeny virus titers in cells expressing dn-SUN2. These data show that the intact LINC complex is required for efficient nuclear egress of herpesviruses, likely acting to promote fusion of primary enveloped virions with the ONM.IMPORTANCE While the viral factors for primary envelopment of nucleocapsids at the inner nuclear membrane are known to the point of high-resolution structures, the roles of cellular components and regulators remain enigmatic. Furthermore, the machinery responsible for fusion with the outer nuclear membrane is unsolved. We show here that dominant negative SUN2 interferes with efficient herpesvirus nuclear egress, apparently by interfering with fusion between the primary virion envelope and outer nuclear membrane. This identifies a new cellular component important for viral egress and implicates LINC complex integrity in nonconventional nuclear membrane trafficking.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Citoesqueleto/metabolismo , Herpesviridae/crescimento & desenvolvimento , Membrana Nuclear/metabolismo , Matriz Nuclear/metabolismo , Transporte Ativo do Núcleo Celular/genética , Animais , Linhagem Celular , Herpesviridae/metabolismo , Proteínas de Membrana/genética , Proteínas Associadas aos Microtúbulos/genética , Proteínas Nucleares/metabolismo , Coelhos , Suínos , Montagem de Vírus/fisiologia , Liberação de Vírus/fisiologia
8.
Mov Disord ; 32(3): 371-381, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27911022

RESUMO

Heterozygosity for a 3-base pair deletion (ΔGAG) in TOR1A/torsinA is one of the most common causes of hereditary dystonia. In this review, we highlight current understanding of how this mutation causes disease from research spanning structural biochemistry, cell science, neurobiology, and several model organisms. We now know that homozygosity for ΔGAG has the same effects as Tor1aKO , implicating a partial loss of function mechanism in the ΔGAG/+ disease state. In addition, torsinA loss specifically affects neurons in mice, even though the gene is broadly expressed, apparently because of differential expression of homologous torsinB. Furthermore, certain neuronal subtypes are more severely affected by torsinA loss. Interestingly, these include striatal cholinergic interneurons that display abnormal responses to dopamine in several Tor1a animal models. There is also progress on understanding torsinA molecular cell biology. The structural basis of how ΔGAG inhibits torsinA ATPase activity is defined, although mutant torsinAΔGAG protein also displays some characteristics suggesting it contributes to dystonia by a gain-of-function mechanism. Furthermore, a consistent relationship is emerging between torsin dysfunction and membrane biology, including an evolutionarily conserved regulation of lipid metabolism. Considered together, these findings provide major advances toward understanding the molecular, cellular, and neurobiological pathologies of DYT1/TOR1A dystonia that can hopefully be exploited for new approaches to treat this disease. © 2016 International Parkinson and Movement Disorder Society.


Assuntos
Distonia Muscular Deformante/metabolismo , Chaperonas Moleculares/metabolismo , Animais , Distonia Muscular Deformante/genética , Humanos , Chaperonas Moleculares/genética
9.
Dev Cell ; 38(3): 235-47, 2016 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-27453503

RESUMO

Torsins are developmentally essential AAA+ proteins, and mutation of human torsinA causes the neurological disease DYT1 dystonia. They localize in the ER membranes, but their cellular function remains unclear. We now show that dTorsin is required in Drosophila adipose tissue, where it suppresses triglyceride levels, promotes cell growth, and elevates membrane lipid content. We also see that human torsinA at the inner nuclear membrane is associated with membrane expansion and elevated cellular lipid content. Furthermore, the key lipid metabolizing enzyme, lipin, is mislocalized in dTorsin-KO cells, and dTorsin increases levels of the lipin substrate, phosphatidate, and reduces the product, diacylglycerol. Finally, genetic suppression of dLipin rescues dTorsin-KO defects, including adipose cell size, animal growth, and survival. These findings identify that torsins are essential regulators of cellular lipid metabolism and implicate disturbed lipid biology in childhood-onset DYT1 dystonia.


Assuntos
Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Metabolismo dos Lipídeos , Chaperonas Moleculares/metabolismo , Membrana Nuclear/metabolismo , Fosfatidato Fosfatase/metabolismo , Tecido Adiposo/metabolismo , Animais , Células Cultivadas , Diglicerídeos/metabolismo , Drosophila melanogaster/genética , Retículo Endoplasmático/metabolismo , Feminino , Humanos , Masculino , Lipídeos de Membrana/metabolismo , Chaperonas Moleculares/genética , Fosfolipídeos/metabolismo
10.
J Cell Sci ; 128(15): 2854-65, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26092934

RESUMO

TorsinA (also known as torsin-1A) is a membrane-embedded AAA+ ATPase that has an important role in the nuclear envelope lumen. However, most torsinA is localized in the peripheral endoplasmic reticulum (ER) lumen where it has a slow mobility that is incompatible with free equilibration between ER subdomains. We now find that nuclear-envelope-localized torsinA is present on the inner nuclear membrane (INM) and ask how torsinA reaches this subdomain. The ER system contains two transmembrane proteins, LAP1 and LULL1 (also known as TOR1AIP1 and TOR1AIP2, respectively), that reversibly co-assemble with and activate torsinA. Whereas LAP1 localizes on the INM, we show that LULL1 is in the peripheral ER and does not enter the INM. Paradoxically, interaction between torsinA and LULL1 in the ER targets torsinA to the INM. Native gel electrophoresis reveals torsinA oligomeric complexes that are destabilized by LULL1. Mutations in torsinA or LULL1 that inhibit ATPase activity reduce the access of torsinA to the INM. Furthermore, although LULL1 binds torsinA in the ER lumen, its effect on torsinA localization requires cytosolic-domain-mediated oligomerization. These data suggest that LULL1 oligomerizes to engage and transiently disassemble torsinA oligomers, and is thereby positioned to transduce cytoplasmic signals to the INM through torsinA.


Assuntos
Proteínas de Transporte/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas de Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Membrana Nuclear/metabolismo , Células 3T3 , Adenosina Trifosfatases/metabolismo , Animais , Células CHO , Proteínas de Transporte/genética , Linhagem Celular , Cricetulus , Proteínas de Membrana/genética , Camundongos , Complexos Multiproteicos/genética , Proteínas Nucleares/metabolismo , Ligação Proteica
11.
Trends Neurosci ; 36(12): 717-25, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24144882

RESUMO

Primary dystonia is a poorly understood but common movement disorder. Recently, several new primary dystonia genes were identified that provide new insight into dystonia pathogenesis. The GNAL dystonia gene is central for striatal responses to dopamine (DA) and is a component of a molecular pathway already implicated in DOPA-responsive dystonia (DRD). Furthermore, this pathway is also dysfunctional and pathogenically linked to mTOR signaling in L-DOPA-induced dyskinesias (LID). These new data suggest that striatal DA responses are central to primary dystonia, even when symptoms do not benefit from DA therapies. Here we integrate these new findings with current understanding of striatal microcircuitry and other dystonia-causing insults to develop new ideas on the pathophysiology of this incapacitating movement disorder.


Assuntos
Dopamina/metabolismo , Distonia/genética , Distonia/fisiopatologia , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Distonia/induzido quimicamente , Distonia/patologia , Humanos , Levodopa/efeitos adversos , Chaperonas Moleculares/genética , Mutação/genética , Serina-Treonina Quinases TOR/genética
12.
BMC Cell Biol ; 12: 24, 2011 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-21627841

RESUMO

BACKGROUND: DYT1 dystonia is an autosomal dominant neurological condition caused by a mutation that removes a single glutamic acid residue (ΔE) from the torsinA (torA) AAA+ protein. TorA appears to possess a nuclear envelope (NE) localized activity that requires Lamina-Associated-Polypeptide 1 (LAP1), which is an inner nuclear membrane localized torA-binding partner. Although hypoactive, the DYT1 dystonia torA-ΔE isoform often concentrates in the NE, suggesting that torA-ΔE also interacts with an NE-localized binding partner. RESULTS: We confirm that NE-localized torA-ΔE does not co-immunoprecipitate with LAP1, and find that torA-ΔE continues to concentrate in the NE of cells that lack LAP1. Instead, we find that variability in torA-ΔE localization correlates with the presence of the SUN-domain and Nesprin proteins that assemble into the LINC complex. We also find that siRNA depletion of SUN1, but not other LINC complex components, removes torA-ΔE from the NE. In contrast, the LAP1-dependent NE-accumulation of an ATP-locked torA mutant is unaffected by loss of LINC complex proteins. This SUN1 dependent torA-ΔE localization requires the torA membrane association domain, as well as a putative substrate-interaction residue, Y147, neither of which are required for torA interaction with LAP1. We also find that mutation of these motifs, or depletion of SUN1, decreases the amount of torA-WT that colocalizes with NE markers, indicating that each also underlies a normal NE-localized torA binding interaction. CONCLUSIONS: These data suggest that the disease causing ΔE mutation promotes an association between torA and SUN1 that is distinct to the interaction between LAP1 and ATP-bound torA. This evidence for two NE-localized binding partners suggests that torA may act on multiple substrates and/or possesses regulatory co-factor partners. In addition, finding that the DYT1 mutation causes abnormal association with SUN1 implicates LINC complex dysfunction in DYT1 dystonia pathogenesis, and suggests a gain-of-function activity contributes to this dominantly inherited disease.


Assuntos
Proteínas Associadas aos Microtúbulos/metabolismo , Chaperonas Moleculares/análise , Membrana Nuclear/metabolismo , Animais , Núcleo Celular/metabolismo , Distonia/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/genética , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Mutação , Células NIH 3T3 , Domínios e Motivos de Interação entre Proteínas , Interferência de RNA , RNA Interferente Pequeno/metabolismo
13.
J Neurosci ; 28(27): 6872-83, 2008 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-18596162

RESUMO

Neuregulin-1 (Nrg1)/erbB signaling regulates neuronal development, migration, myelination, and synaptic maintenance. The Nrg1 gene is a schizophrenia susceptibility gene. To understand the contribution of Nrg1 signaling to adult brain structure and behaviors, we studied the regulation of type III Nrg1 expression and evaluated the effect of decreased expression of the type III Nrg1 isoforms. Type III Nrg1 is transcribed by a promoter distinct from those for other Nrg1 isoforms and, in the adult brain, is expressed in the medial prefrontal cortex, ventral hippocampus, and ventral subiculum, regions involved in the regulation of sensorimotor gating and short-term memory. Adult heterozygous mutant mice with a targeted disruption for type III Nrg1 (Nrg1(tm1.1Lwr+/-)) have enlarged lateral ventricles and decreased dendritic spine density on subicular pyramidal neurons. Magnetic resonance imaging of type III Nrg1 heterozygous mice revealed hypofunction in the medial prefrontal cortex and the hippocampal CA1 and subiculum regions. Type III Nrg1 heterozygous mice also have impaired performance on delayed alternation memory tasks, and deficits in prepulse inhibition (PPI). Chronic nicotine treatment eliminated differences in PPI between type III Nrg1 heterozygous mice and their wild-type littermates. Our findings demonstrate a role of type III Nrg1 signaling in the maintenance of corticostriatal components and in the neural circuits involved in sensorimotor gating and short-term memory.


Assuntos
Corpo Estriado/anormalidades , Hipocampo/anormalidades , Transtornos da Memória/genética , Proteínas do Tecido Nervoso/genética , Córtex Pré-Frontal/anormalidades , Transtornos de Sensação/genética , Animais , Atrofia/genética , Atrofia/metabolismo , Atrofia/fisiopatologia , Corpo Estriado/metabolismo , Corpo Estriado/fisiopatologia , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento/genética , Heterozigoto , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Ventrículos Laterais/anormalidades , Transtornos da Memória/metabolismo , Transtornos da Memória/fisiopatologia , Memória de Curto Prazo/fisiologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/metabolismo , Malformações do Sistema Nervoso/fisiopatologia , Inibição Neural/genética , Vias Neurais/anormalidades , Vias Neurais/metabolismo , Vias Neurais/fisiopatologia , Neuregulina-1 , Agonistas Nicotínicos/farmacologia , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/fisiopatologia , Regiões Promotoras Genéticas/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transtornos de Sensação/metabolismo , Transtornos de Sensação/fisiopatologia
14.
Neuron ; 48(6): 923-32, 2005 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-16364897

RESUMO

An enigmatic feature of many genetic diseases is that mutations in widely expressed genes cause tissue-specific illness. One example is DYT1 dystonia, a neurodevelopmental disease caused by an in-frame deletion (Deltagag) in the gene encoding torsinA. Here we show that neurons from both torsinA null (Tor1a(-/-)) and homozygous disease mutant "knockin" mice (Tor1a(Deltagag/Deltagag)) contain severely abnormal nuclear membranes, although non-neuronal cell types appear normal. These membrane abnormalities develop in postmigratory embryonic neurons and subsequently worsen with further neuronal maturation, a finding evocative of the developmental dependence of DYT1 dystonia. These observations demonstrate that neurons have a unique requirement for nuclear envelope localized torsinA function and suggest that loss of this activity is a key molecular event in the pathogenesis of DYT1 dystonia.


Assuntos
Encéfalo/anormalidades , Encéfalo/metabolismo , Distonia Muscular Deformante/metabolismo , Chaperonas Moleculares/genética , Neurônios/metabolismo , Membrana Nuclear/metabolismo , Animais , Encéfalo/fisiopatologia , Proteínas de Transporte/metabolismo , Diferenciação Celular/genética , Movimento Celular/genética , Células Cultivadas , Modelos Animais de Doenças , Distonia Muscular Deformante/genética , Distonia Muscular Deformante/fisiopatologia , Predisposição Genética para Doença/genética , Proteínas de Choque Térmico HSC70/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Mutação/genética , Neurônios/patologia , Neurônios/ultraestrutura , Membrana Nuclear/patologia , Membrana Nuclear/ultraestrutura
15.
J Cell Biol ; 168(6): 855-62, 2005 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-15767459

RESUMO

A glutamic acid deletion (DeltaE) in the AAA+ protein torsinA causes DYT1 dystonia. Although the majority of torsinA resides within the endoplasmic reticulum (ER), torsinA binds a substrate in the lumen of the nuclear envelope (NE), and the DeltaE mutation enhances this interaction. Using a novel cell-based screen, we identify lamina-associated polypeptide 1 (LAP1) as a torsinA-interacting protein. LAP1 may be a torsinA substrate, as expression of the isolated lumenal domain of LAP1 inhibits the NE localization of "substrate trap" EQ-torsinA and EQ-torsinA coimmunoprecipitates with LAP1 to a greater extent than wild-type torsinA. Furthermore, we identify a novel transmembrane protein, lumenal domain like LAP1 (LULL1), which also appears to interact with torsinA. Interestingly, LULL1 resides in the main ER. Consequently, torsinA interacts directly or indirectly with a novel class of transmembrane proteins that are localized in different subdomains of the ER system, either or both of which may play a role in the pathogenesis of DYT1 dystonia.


Assuntos
Retículo Endoplasmático/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Transporte/metabolismo , Linhagem Celular , Sequência Conservada , Cricetinae , Retículo Endoplasmático/química , Recuperação de Fluorescência Após Fotodegradação , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Choque Térmico HSC70 , Proteínas de Choque Térmico HSP70/química , Proteínas de Choque Térmico HSP70/genética , Células HeLa , Humanos , Cinética , Proteínas de Membrana/metabolismo , Camundongos , Microscopia Confocal , Dados de Sequência Molecular , Mutação , Células NIH 3T3 , Membrana Nuclear/metabolismo , Testes de Precipitina , Estrutura Terciária de Proteína , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Deleção de Sequência , Homologia de Sequência de Aminoácidos , Transfecção
16.
Proc Natl Acad Sci U S A ; 101(3): 847-52, 2004 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-14711988

RESUMO

Primary dystonia is a disease characterized by involuntary twisting movements caused by CNS dysfunction without underlying histopathology. DYT1 dystonia is a form of primary dystonia caused by an in-frame GAG deletion (DeltaE302/3) in the TOR1A gene that encodes the endoplasmic reticulum luminal protein torsinA. We show that torsinA is also present in the nuclear envelope (NE), where it appears to interact with substrate, and that the DeltaE302/3 mutation causes a striking redistribution of torsinA from the endoplasmic reticulum to the NE. In addition, DeltaE302/3-torsinA recruits WT torsinA to the NE, potentially providing insight into an understanding of the dominant inheritance of the disease. DYT1 dystonia appears to be a previously uncharacterized NE disease and the first, to our knowledge, to selectively affect CNS function.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Distonia Muscular Deformante/genética , Distonia Muscular Deformante/metabolismo , Chaperonas Moleculares , Mutação , Membrana Nuclear/metabolismo , Animais , Linhagem Celular , Cricetinae , Distonia Muscular Deformante/patologia , Retículo Endoplasmático/metabolismo , Genes Dominantes , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia Imunoeletrônica , Membrana Nuclear/ultraestrutura , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Deleção de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA