Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 30(8): 706-715, 2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-33772540

RESUMEN

Spinocerebellar Ataxia Type 1 (SCA1) is an autosomal dominant neurodegenerative disorder caused by a polyglutamine expansion in the ataxin-1 protein. Recent genetic correlational studies have implicated DNA damage repair pathways in modifying the age at onset of disease symptoms in SCA1 and Huntington's Disease, another polyglutamine expansion disease. We demonstrate that both endogenous and transfected ataxin-1 localizes to sites of DNA damage, which is impaired by polyglutamine expansion. This response is dependent on ataxia-telangiectasia mutated (ATM) kinase activity. Further, we characterize an ATM phosphorylation motif within ataxin-1 at serine 188. We show reduction of the Drosophila ATM homolog levels in a ATXN1[82Q] Drosophila model through shRNA or genetic cross ameliorates motor symptoms. These findings offer a possible explanation as to why DNA repair was implicated in SCA1 pathogenesis by past studies. The similarities between the ataxin-1 and the huntingtin responses to DNA damage provide further support for a shared pathogenic mechanism for polyglutamine expansion diseases.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/genética , Ataxina-1/genética , Daño del ADN , Ataxias Espinocerebelosas/genética , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Ataxina-1/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Drosophila/genética , Drosophila/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Humanos , Microscopía Confocal , Mutación , Péptidos/genética , Homología de Secuencia de Aminoácido , Transducción de Señal/genética , Ataxias Espinocerebelosas/metabolismo , Ataxias Espinocerebelosas/patología , Repeticiones de Trinucleótidos/genética
2.
Mol Psychiatry ; 26(9): 5441-5463, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-32514103

RESUMEN

Huntington's disease (HD) is a monogenic neurodegenerative disorder resulting from a mutation in the huntingtin gene. This leads to the expression of the mutant huntingtin protein (mHTT) which provokes pathological changes in both the central nervous system (CNS) and periphery. Accumulating evidence suggests that mHTT can spread between cells of the CNS but here, we explored the possibility that mHTT could also propagate and cause pathology via the bloodstream. For this, we used a parabiosis approach to join the circulatory systems of wild-type (WT) and zQ175 mice. After surgery, we observed mHTT in the plasma and circulating blood cells of WT mice and post-mortem analyses revealed the presence of mHTT aggregates in several organs including the liver, kidney, muscle and brain. The presence of mHTT in the brain was accompanied by vascular abnormalities, such as a reduction of Collagen IV signal intensity and altered vessel diameter in the striatum, and changes in expression of Glutamic acid decarboxylase 65/67 (GAD65-67) in the cortex. Conversely, we measured reduced pathology in zQ175 mice by decreased mitochondrial impairments in peripheral organs, restored vessel diameter in the cortex and improved expression of Dopamine- and cAMP-regulated phosphoprotein 32 (DARPP32) in striatal neurons. Collectively, these results demonstrate that circulating mHTT can disseminate disease, but importantly, that healthy blood can dilute pathology. These findings have significant implications for the development of therapies in HD.


Asunto(s)
Enfermedad de Huntington , Animales , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Fosfoproteína 32 Regulada por Dopamina y AMPc , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/genética , Ratones , Ratones Transgénicos , Neuronas/metabolismo
3.
Proc Natl Acad Sci U S A ; 115(30): E7081-E7090, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-29987005

RESUMEN

The huntingtin N17 domain is a modulator of mutant huntingtin toxicity and is hypophosphorylated in Huntington's disease (HD). We conducted high-content analysis to find compounds that could restore N17 phosphorylation. One lead compound from this screen was N6-furfuryladenine (N6FFA). N6FFA was protective in HD model neurons, and N6FFA treatment of an HD mouse model corrects HD phenotypes and eliminates cortical mutant huntingtin inclusions. We show that N6FFA restores N17 phosphorylation levels by being salvaged to a triphosphate form by adenine phosphoribosyltransferase (APRT) and used as a phosphate donor by casein kinase 2 (CK2). N6FFA is a naturally occurring product of oxidative DNA damage. Phosphorylated huntingtin functionally redistributes and colocalizes with CK2, APRT, and N6FFA DNA adducts at sites of induced DNA damage. We present a model in which this natural product compound is salvaged to provide a triphosphate substrate to signal huntingtin phosphorylation via CK2 during low-ATP stress under conditions of DNA damage, with protective effects in HD model systems.


Asunto(s)
Adenina , Aductos de ADN/metabolismo , Daño del ADN , Enfermedad de Huntington/tratamiento farmacológico , Neuronas/metabolismo , Transducción de Señal/efectos de los fármacos , Adenina/análogos & derivados , Adenina/farmacocinética , Adenina/farmacología , Adenina Fosforribosiltransferasa/genética , Adenina Fosforribosiltransferasa/metabolismo , Animales , Quinasa de la Caseína II/genética , Quinasa de la Caseína II/metabolismo , Línea Celular Transformada , Aductos de ADN/genética , Modelos Animales de Enfermedad , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Ratones , Ratones Transgénicos , Neuronas/patología , Fosforilación/efectos de los fármacos , Fosforilación/genética , Transducción de Señal/genética
4.
J Biol Chem ; 294(6): 1915-1923, 2019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30538129

RESUMEN

Huntington's disease (HD) is a neurodegenerative, age-onset disorder caused by a CAG DNA expansion in exon 1 of the HTT gene, resulting in a polyglutamine expansion in the huntingtin protein. Nuclear accumulation of mutant huntingtin is a hallmark of HD, resulting in elevated mutant huntingtin levels in cell nuclei. Huntingtin is normally retained at the endoplasmic reticulum via its N17 amphipathic α-helix domain but is released by oxidation of Met-8 during reactive oxygen species (ROS) stress. Huntingtin enters the nucleus via an importin ß1- and 2-dependent proline-tyrosine nuclear localization signal (PY-NLS), which has a unique intervening sequence in huntingtin. Here, we have identified the high-mobility group box 1 (HMGB1) protein as an interactor of the intervening sequence within the PY-NLS. Nuclear levels of HMGB1 positively correlated with varying levels of nuclear huntingtin in both HD and normal human fibroblasts. We also found that HMGB1 interacts with the huntingtin N17 region and that this interaction is enhanced by the presence of ROS and phosphorylation of critical serine residues in the N17 region. We conclude that HMGB1 is a huntingtin N17/PY-NLS ROS-dependent interactor, and this protein bridging is essential for relaying ROS sensing by huntingtin to its nuclear entry during ROS stress. ROS may therefore be a critical age-onset stress that triggers nuclear accumulation of mutant huntington in Huntington's disease.


Asunto(s)
Transporte Activo de Núcleo Celular , Proteína HMGB1/fisiología , Proteína Huntingtina/metabolismo , Especies Reactivas de Oxígeno/farmacología , Sitios de Unión , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Proteína Huntingtina/efectos de los fármacos , Proteína Huntingtina/fisiología , Señales de Localización Nuclear , Proteínas Nucleares/metabolismo , Fosforilación , Unión Proteica
5.
Hum Mol Genet ; 26(2): 395-406, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-28017939

RESUMEN

Huntington's disease (HD) is an age-dependent neurodegenerative disease. DNA repair pathways have recently been implicated as the most predominant modifiers of age of onset in HD patients. We report that endogenous huntingtin protein directly participates in oxidative DNA damage repair. Using novel chromobodies to detect endogenous human huntingtin in live cells, we show that localization of huntingtin to DNA damage sites is dependent on the kinase activity of ataxia telangiectasia mutated (ATM) protein. Super-resolution microscopy and biochemical assays revealed that huntingtin co-localizes with and scaffolds proteins of the DNA damage response pathway in response to oxidative stress. In HD patient fibroblasts bearing typical clinical HD allele lengths, we demonstrate that there is deficient oxidative DNA damage repair. We propose that DNA damage in HD is caused by dysfunction of the mutant huntingtin protein in DNA repair, and accumulation of DNA oxidative lesions due to elevated reactive oxygen species may contribute to the onset of HD.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Estrés Oxidativo/genética , Alelos , Daño del ADN/genética , Reparación del ADN/genética , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/fisiopatología , Especies Reactivas de Oxígeno/metabolismo
6.
Lancet ; 391(10126): 1197-1204, 2018 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-29223694

RESUMEN

BACKGROUND: Atraumatic needles have been proposed to lower complication rates after lumbar puncture. However, several surveys indicate that clinical adoption of these needles remains poor. We did a systematic review and meta-analysis to compare patient outcomes after lumbar puncture with atraumatic needles and conventional needles. METHODS: In this systematic review and meta-analysis, we independently searched 13 databases with no language restrictions from inception to Aug 15, 2017, for randomised controlled trials comparing the use of atraumatic needles and conventional needles for any lumbar puncture indication. Randomised trials comparing atraumatic and conventional needles in which no dural puncture was done (epidural injections) or without a conventional needle control group were excluded. We screened studies and extracted data from published reports independently. The primary outcome of postdural-puncture headache incidence and additional safety and efficacy outcomes were assessed by random-effects and fixed-effects meta-analysis. This study is registered with the International Prospective Register of Systematic Reviews, number CRD42016047546. FINDINGS: We identified 20 241 reports; after exclusions, 110 trials done between 1989 and 2017 from 29 countries, including a total of 31 412 participants, were eligible for analysis. The incidence of postdural-puncture headache was significantly reduced from 11·0% (95% CI 9·1-13·3) in the conventional needle group to 4·2% (3·3-5·2) in the atraumatic group (relative risk 0·40, 95% CI 0·34-0·47, p<0·0001; I2=45·4%). Atraumatic needles were also associated with significant reductions in the need for intravenous fluid or controlled analgesia (0·44, 95% CI 0·29-0·64; p<0·0001), need for epidural blood patch (0·50, 0·33-0·75; p=0·001), any headache (0·50, 0·43-0·57; p<0·0001), mild headache (0·52, 0·38-0·70; p<0·0001), severe headache (0·41, 0·28-0·59; p<0·0001), nerve root irritation (0·71, 0·54-0·92; p=0·011), and hearing disturbance (0·25, 0·11-0·60; p=0·002). Success of lumbar puncture on first attempt, failure rate, mean number of attempts, and the incidence of traumatic tap and backache did not differ significantly between the two needle groups. Prespecified subgroup analyses of postdural-puncture headache revealed no interactions between needle type and patient age, sex, use of prophylactic intravenous fluid, needle gauge, patient position, indication for lumbar puncture, bed rest after puncture, or clinician specialty. These results were rated high-quality evidence as examined using the grading of recommendations assessment, development, and evaluation. INTERPRETATION: Among patients who had lumbar puncture, atraumatic needles were associated with a decrease in the incidence of postdural-puncture headache and in the need for patients to return to hospital for additional therapy, and had similar efficacy to conventional needles. These findings offer clinicians and stakeholders a comprehensive assessment and high-quality evidence for the safety and efficacy of atraumatic needles as a superior option for patients who require lumbar puncture. FUNDING: None.


Asunto(s)
Agujas , Punción Espinal/instrumentación , Humanos , Punción Espinal/efectos adversos
7.
Hum Mol Genet ; 25(18): 3937-3945, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27466181

RESUMEN

The N17 domain of the huntingtin protein is post-translationally modified and is the master regulator of huntingtin intracellular localization. In Huntington's disease (HD), mutant huntingtin is hypo-phosphorylated at serines 13 and 16 within N17, and increasing N17 phosphorylation has been shown to be protective in HD mouse models. Thus, N17 phosphorylation is defined as a sub-target of huntingtin for potential therapeutic intervention. We have previously shown that cellular stress can affect huntingtin nuclear entry and phosphorylation. Here, we demonstrate that huntingtin localization can be specifically affected by reactive oxygen species (ROS) stress. We have located the sensor of this stress to the N17 domain, specifically to a highly conserved methionine at position 8. In vitro, we show by circular dichroism spectroscopy structural studies that the alpha-helical structure of N17 changes in response to redox conditions and show that the consequence of this change is enhanced N17 phosphorylation and nuclear targeting of endogenous huntingtin. Using N17 substitution point mutants, we demonstrate that N17 sulphoxidation enhances N17 dissociation from the endoplasmic reticulum (ER) membrane. This enhanced solubility makes N17 a better substrate for phosphorylation and subsequent nuclear retention. This ability of huntingtin to sense ROS levels at the ER, with phosphorylation and nuclear localization as a response, suggests that ROS stress due to aging could be a critical molecular trigger of huntingtin functions and dysfunctions in HD and may explain the age-onset nature of the disorder.


Asunto(s)
Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Estrés Oxidativo/genética , Especies Reactivas de Oxígeno/metabolismo , Animales , Modelos Animales de Enfermedad , Retículo Endoplásmico/genética , Retículo Endoplásmico/patología , Humanos , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Ratones , Mutación , Fosforilación/genética , Dominios Proteicos
9.
Hum Mol Genet ; 24(2): 450-62, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25205111

RESUMEN

Cellular stress is a normal part of the aging process and is especially relevant in neurodegenerative disease. Canonical stress responses, such as the heat shock response, activate following exposure to stress and restore proteostasis through the action of isomerases and chaperones within the cytosol. Through live-cell imaging, we demonstrate involvement of the Huntington's disease (HD) protein, huntingtin, in a rapid cell stress response that lies temporally upstream of canonical stress responses. This response is characterized by the formation of distinct cytosolic puncta and reversible localization of huntingtin to early endosomes. The formation of these puncta, which we have termed huntingtin stress bodies (HSBs), is associated with arrest of early-to-recycling and early-to-late endosomal trafficking. The critical domains for this response have been mapped to two regions of huntingtin flanking the polyglutamine tract, and we observe polyglutamine-expanded huntingtin-expressing cells to be defective in their ability to recover from this stress response. We propose that HSB formation rapidly diverts high ATP use from vesicular trafficking during stress, thus mobilizing canonical stress responses without relying on increased energy metabolism, and that restoration from this response is defective in HD.


Asunto(s)
Endosomas/metabolismo , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/fisiopatología , Proteínas del Tejido Nervioso/metabolismo , Secuencias de Aminoácidos , Animales , Endosomas/genética , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Transporte de Proteínas , Estrés Fisiológico
11.
Hum Mol Genet ; 23(9): 2324-38, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24334607

RESUMEN

Huntington's disease (HD) is an autosomal dominant, neurodegenerative disorder that can be characterized by the presence of protein inclusions containing mutant huntingtin within a subset of neurons in the brain. Since their discovery, the relevance of inclusions to disease pathology has been controversial. We show using super-resolution fluorescence imaging and Förster resonance energy transfer (FRET) in live cells, that mutant huntingtin fragments can form two morphologically and conformationally distinct inclusion types. Using fluorescence recovery after photobleaching (FRAP), we demonstrate that the two huntingtin inclusion types have unique dynamic properties. The ability to form one or the other type of inclusion can be influenced by the phosphorylation state of serine residues at amino acid positions 13 and 16 within the huntingtin protein. We can define two types of inclusions: fibrillar, which are tightly packed, do not exchange protein with the soluble phase, and result from phospho-modification at serines 13 and 16 of the N17 domain, and globular, which are loosely packed, can readily exchange with the soluble phase, and are not phosphorylated in N17. We hypothesize that the protective effect of N17 phosphorylation or phospho-mimicry seen in animal models, at the level of protein inclusions with elevated huntingtin levels, is to induce a conformation of the huntingtin amino-terminus that causes fragments to form tightly packed inclusions that do not exit the insoluble phase, and hence exert less toxicity. The identification of these sub-types of huntingtin inclusions could allow for drug discovery to promote protective inclusions of mutant huntingtin protein in HD.


Asunto(s)
Enfermedad de Huntington/metabolismo , Cuerpos de Inclusión/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Animales , Recuperación de Fluorescencia tras Fotoblanqueo , Transferencia Resonante de Energía de Fluorescencia , Proteína Huntingtina , Ratones , Fosforilación , Técnicas de Cultivo de Tejidos
12.
Proc Natl Acad Sci U S A ; 110(36): 14610-5, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-23898200

RESUMEN

Huntington disease (HD) is a neurodegenerative disorder caused by a CAG expansion within the huntingtin gene that encodes a polymorphic glutamine tract at the amino terminus of the huntingtin protein. HD is one of nine polyglutamine expansion diseases. The clinical threshold of polyglutamine expansion for HD is near 37 repeats, but the mechanism of this pathogenic length is poorly understood. Using Förster resonance energy transfer, we describe an intramolecular proximity between the N17 domain and the downstream polyproline region that flanks the polyglutamine tract of huntingtin. Our data support the hypothesis that the polyglutamine tract can act as a flexible domain, allowing the flanking domains to come into close spatial proximity. This flexibility is impaired with expanded polyglutamine tracts, and we can detect changes in huntingtin conformation at the pathogenic threshold for HD. Altering the structure of N17, either via phosphomimicry or with small molecules, also affects the proximity between the flanking domains. The structural capacity of N17 to fold back toward distal regions within huntingtin requires an interacting protein, protein kinase C and casein kinase 2 substrate in neurons 1 (PACSIN1). This protein has the ability to bind both N17 and the polyproline region, stabilizing the interaction between these two domains. We also developed an antibody-based FRET assay that can detect conformational changes within endogenous huntingtin in wild-type versus HD fibroblasts. Therefore, we hypothesize that wild-type length polyglutamine tracts within huntingtin can form a flexible domain that is essential for proper functional intramolecular proximity, conformations, and dynamics.


Asunto(s)
Enfermedad de Huntington/genética , Proteínas del Tejido Nervioso/genética , Péptidos/genética , Expansión de Repetición de Trinucleótido/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Sitios de Unión/genética , Línea Celular , Células Cultivadas , Exones/genética , Femenino , Transferencia Resonante de Energía de Fluorescencia , Humanos , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Masculino , Ratones , Microscopía Fluorescente , Persona de Mediana Edad , Mutación , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Péptidos/metabolismo , Unión Proteica , Conformación Proteica
13.
Proc Natl Acad Sci U S A ; 109(9): 3528-33, 2012 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-22331905

RESUMEN

Huntington disease (HD) is a progressive neurodegenerative monogenic disorder caused by expansion of a polyglutamine stretch in the huntingtin (Htt) protein. Mutant huntingtin triggers neural dysfunction and death, mainly in the corpus striatum and cerebral cortex, resulting in pathognomonic motor symptoms, as well as cognitive and psychiatric decline. Currently, there is no effective treatment for HD. We report that intraventricular infusion of ganglioside GM1 induces phosphorylation of mutant huntingtin at specific serine amino acid residues that attenuate huntingtin toxicity, and restores normal motor function in already symptomatic HD mice. Thus, our studies have identified a potential therapy for HD that targets a posttranslational modification of mutant huntingtin with critical effects on disease pathogenesis.


Asunto(s)
Gangliósido G(M1)/uso terapéutico , Actividad Motora/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Animales , Codón/efectos de los fármacos , Cuerpo Estriado/metabolismo , Dimerización , Modelos Animales de Enfermedad , Fosfoproteína 32 Regulada por Dopamina y AMPc/biosíntesis , Fosfoproteína 32 Regulada por Dopamina y AMPc/genética , Evaluación Preclínica de Medicamentos , Gangliósido G(M1)/administración & dosificación , Proteína Huntingtina , Bombas de Infusión Implantables , Infusiones Parenterales , Ratones , Ratones Mutantes Neurológicos , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Fosfoserina/análisis , Desempeño Psicomotor/efectos de los fármacos
14.
J Cell Sci ; 125(Pt 17): 3977-88, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22623727

RESUMEN

Cofilin protein is involved in regulating the actin cytoskeleton during typical steady state conditions, as well as during cell stress conditions where cofilin saturates F-actin, forming cofilin-actin rods. Cofilin can enter the nucleus through an active nuclear localization signal (NLS), accumulating in nuclear actin rods during stress. Here, we characterize the active nuclear export of cofilin through a leptomycin-B-sensitive, CRM1-dependent, nuclear export signal (NES). We also redefine the NLS of cofilin as a bipartite NLS, with an additional basic epitope required for nuclear localization. Using fluorescence lifetime imaging microscopy (FLIM) and Förster resonant energy transfer (FRET) between cofilin moieties and actin, as well as automated image analysis in live cells, we have defined subtle mutations in the cofilin NLS that allow cofilin to bind actin in vivo and affect cofilin dynamics during stress. We further define the requirement of cofilin-actin rod formation in a system of cell stress by temporal live-cell imaging. We propose that cofilin nuclear shuttling is critical for the cofilin-actin rod stress response with cofilin dynamically communicating between the nucleus and cytoplasm during cell stress.


Asunto(s)
Actinas/metabolismo , Núcleo Celular/metabolismo , Cofilina 1/metabolismo , Estrés Fisiológico , Secuencia de Aminoácidos , Animales , Línea Celular , Cofilina 1/química , Secuencia Conservada , Técnicas de Silenciamiento del Gen , Humanos , Carioferinas/metabolismo , Ratones , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Mutación , Señales de Exportación Nuclear , Señales de Localización Nuclear/química , Señales de Localización Nuclear/metabolismo , Fenotipo , Unión Proteica , Transporte de Proteínas , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteína Exportina 1
15.
J Biol Chem ; 287(47): 39626-33, 2012 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-23012356

RESUMEN

Among the known pathways of protein nuclear import, the karyopherin ß2/transportin pathway is only the second to have a defined nuclear localization signal (NLS) consensus. Huntingtin, a 350-kDa protein, has defined roles in the nucleus, as well as a CRM1/exportin-dependent nuclear export signal; however, the NLS and exact pathway of import have remained elusive. Here, using a live cell assay and affinity chromatography, we show that huntingtin has a karyopherin ß2-dependent proline-tyrosine (PY)-NLS in the amino terminus of the protein. This NLS comprises three consensus components: a basic charged sequence, a downstream conserved arginine, and a PY sequence. Unlike the classic PY-NLS, which has an unstructured intervening sequence between the consensus components, we show that a ß sheet structured region separating the consensus elements is critical for huntingtin NLS function. The huntingtin PY-NLS is also capable of import through the importin/karyopherin ß1 pathway but was not functional in all cell types tested. We propose that this huntingtin PY-NLS may comprise a new class of multiple import factor-dependent NLSs with an internal structural component that may regulate NLS activity.


Asunto(s)
Proteínas del Tejido Nervioso/genética , Señales de Localización Nuclear/fisiología , Proteínas Nucleares/genética , beta Carioferinas/genética , Animales , Humanos , Proteína Huntingtina , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Mapeo Peptídico/métodos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , beta Carioferinas/metabolismo
16.
Hum Mol Genet ; 20(10): 1937-51, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21355047

RESUMEN

Huntington's disease (HD) is caused by an expanded CAG tract in the Interesting transcript 15 (IT15) gene encoding the 350 kDa huntingtin protein. Cellular stresses can trigger the release of huntingtin from the endoplasmic reticulum, allowing huntingtin nuclear entry. Here, we show that endogenous, full-length huntingtin localizes to nuclear cofilin-actin rods during stress and is required for the proper stress response involving actin remodeling. Mutant huntingtin induces a dominant, persistent nuclear rod phenotype similar to that described in Alzheimer's disease for cytoplasmic cofilin-actin rods. Using live cell temporal studies, we show that this stress response is similarly impaired when mutant huntingtin is present, or when normal huntingtin levels are reduced. In clinical lymphocyte samples from HD patients, we have quantitatively detected cross-linked complexes of actin and cofilin with complex formation varying in correlation with disease progression. By live cell fluorescence lifetime imaging measurement-Förster resonant energy transfer studies and western blot assays, we quantitatively observed that stress-activated tissue transglutaminase 2 (TG2) is responsible for the actin-cofilin covalent cross-linking observed in HD. These data support a direct role for huntingtin in nuclear actin re-organization, and describe a new pathogenic mechanism for aberrant TG2 enzymatic hyperactivity in neurodegenerative diseases.


Asunto(s)
Actinas/metabolismo , Proteínas de Unión al GTP/metabolismo , Respuesta al Choque Térmico/genética , Enfermedad de Huntington/enzimología , Enfermedad de Huntington/genética , Mutación/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Transglutaminasas/metabolismo , Factores Despolimerizantes de la Actina/metabolismo , Animales , Línea Celular , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al GTP/genética , Expresión Génica/genética , Calor , Humanos , Proteína Huntingtina , Espacio Intracelular/metabolismo , Linfocitos/metabolismo , Ratones , Modelos Biológicos , Células 3T3 NIH , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Unión Proteica , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transporte de Proteínas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transglutaminasas/genética
17.
Nat Chem Biol ; 7(7): 453-60, 2011 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-21623356

RESUMEN

Two serine residues within the first 17 amino acid residues of huntingtin (N17) are crucial for modulation of mutant huntingtin toxicity in cell and mouse genetic models of Huntington's disease. Here we show that the stress-dependent phosphorylation of huntingtin at Ser13 and Ser16 affects N17 conformation and targets full-length huntingtin to chromatin-dependent subregions of the nucleus, the mitotic spindle and cleavage furrow during cell division. Polyglutamine-expanded mutant huntingtin is hypophosphorylated in N17 in both homozygous and heterozygous cell contexts. By high-content screening in live cells, we identified kinase inhibitors that modulated N17 phosphorylation and hence huntingtin subcellular localization. N17 phosphorylation was reduced by casein kinase-2 inhibitors. Paradoxically, IKKß kinase inhibition increased N17 phosphorylation, affecting huntingtin nuclear and subnuclear localization. These data indicate that huntingtin phosphorylation at Ser13 and Ser16 can be modulated by small-molecule drugs, which may have therapeutic potential in Huntington's disease.


Asunto(s)
Quinasa de la Caseína II/antagonistas & inhibidores , Enfermedad de Huntington/metabolismo , Quinasa I-kappa B/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Inhibidores de Proteínas Quinasas/farmacología , Animales , Western Blotting , Línea Celular , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Retículo Endoplásmico/metabolismo , Técnica del Anticuerpo Fluorescente , Proteína Huntingtina , Enfermedad de Huntington/enzimología , Enfermedad de Huntington/genética , Ratones , Mutación , Fosforilación , Serina/genética , Huso Acromático/metabolismo , Transfección
18.
Res Sq ; 2023 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-36945419

RESUMEN

CRISPR enzymes require a defined protospacer adjacent motif (PAM) flanking a guide RNA-programmed target site, limiting their sequence accessibility for robust genome editing applications. In this study, we recombine the PAM-interacting domain of SpRY, a broad-targeting Cas9 possessing an NRN > NYN PAM preference, with the N-terminus of Sc++, a Cas9 with simultaneously broad, efficient, and accurate NNG editing capabilities, to generate a chimeric enzyme with highly flexible PAM preference: SpRYc. We demonstrate that SpRYc leverages properties of both enzymes to specifically edit diverse NNN PAMs and disease-related loci for potential therapeutic applications. In total, the unique approaches to generate SpRYc, coupled with its robust flexibility, highlight the power of integrative protein design for Cas9 engineering and motivate downstream editing applications that require precise genomic positioning.

19.
Nat Commun ; 14(1): 6175, 2023 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-37794046

RESUMEN

CRISPR enzymes require a defined protospacer adjacent motif (PAM) flanking a guide RNA-programmed target site, limiting their sequence accessibility for robust genome editing applications. In this study, we recombine the PAM-interacting domain of SpRY, a broad-targeting Cas9 possessing an NRN > NYN (R = A or G, Y = C or T) PAM preference, with the N-terminus of Sc + +, a Cas9 with simultaneously broad, efficient, and accurate NNG editing capabilities, to generate a chimeric enzyme with highly flexible PAM preference: SpRYc. We demonstrate that SpRYc leverages properties of both enzymes to specifically edit diverse PAMs and disease-related loci for potential therapeutic applications. In total, the approaches to generate SpRYc, coupled with its robust flexibility, highlight the power of integrative protein design for Cas9 engineering and motivate downstream editing applications that require precise genomic positioning.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Sistemas CRISPR-Cas/genética , Proteína 9 Asociada a CRISPR/genética , Proteína 9 Asociada a CRISPR/metabolismo , Genoma
20.
PLoS One ; 17(6): e0269743, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35731739

RESUMEN

The phenomenon of "publish-or-perish" in academia, spurred on by limited funding and academic positions, has led to increased competition and pressure on academics to publish. Publication pressure has been linked with multiple negative outcomes, including increased academic misconduct and researcher burnout. COVID-19 has disrupted research worldwide, leading to lost research time and increased anxiety amongst researchers. The objective of this study was to examine how COVID-19 has impacted perceived publication pressure amongst academic researchers in Canada. We used the revised Publication Pressure Questionnaire, in addition to Likert-type questions to discern respondents' beliefs and concerns about the impact of COVID-19 on academic publishing. We found that publication pressure increased across academic researchers in Canada following the pandemic, with respondents reporting increased stress, increased pessimism, and decreased access to support related to publishing. Doctoral students reported the highest levels of stress and pessimism, while principal investigators had the most access to publication support. There were no significant differences in publication pressure reported between different research disciplines. Women and non-binary or genderfluid respondents reported higher stress and pessimism than men. We also identified differences in perceived publication pressure based on respondents' publication frequency and other demographic factors, including disability and citizenship status. Overall, we document a snapshot of perceived publication pressure in Canada across researchers of different academic career stages and disciplines. This information can be used to guide the creation of researcher supports, as well as identify groups of researchers who may benefit from targeted resources.


Asunto(s)
COVID-19 , COVID-19/epidemiología , Femenino , Humanos , Masculino , Pandemias , Edición , Investigadores , Encuestas y Cuestionarios
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA