Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Mol Cell Neurosci ; 83: 103-112, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28743452

RESUMEN

Huntington's disease is caused by polyglutamine (polyQ)-expansion mutations in the CAG tandem repeat of the Huntingtin gene. The central feature of Huntington's disease pathology is the aggregation of mutant Huntingtin (Htt) protein into micrometer-sized inclusion bodies. Soluble mutant Htt states are most proteotoxic and trigger an enhanced risk of death whereas inclusions confer different changes to cellular health, and may even provide adaptive responses to stress. Yet the molecular mechanisms underpinning these changes remain unclear. Using the flow cytometry method of pulse-shape analysis (PulSA) to sort neuroblastoma (Neuro2a) cells enriched with mutant or wild-type Htt into different aggregation states, we clarified which transcriptional signatures were specifically attributable to cells before versus after inclusion assembly. Dampened CREB signalling was the most striking change overall and invoked specifically by soluble mutant Httex1 states. Toxicity could be rescued by stimulation of CREB signalling. Other biological processes mapped to different changes before and after aggregation included NF-kB signalling, autophagy, SUMOylation, transcription regulation by histone deacetylases and BRD4, NAD+ biosynthesis, ribosome biogenesis and altered HIF-1 signalling. These findings open the path for therapeutic strategies targeting key molecular changes invoked prior to, and subsequently to, Httex1 aggregation.


Asunto(s)
Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Mutación , Agregación Patológica de Proteínas/metabolismo , Transducción de Señal , Transcriptoma , Animales , Línea Celular Tumoral , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Exones , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Ratones , Agregación Patológica de Proteínas/genética
2.
Traffic ; 15(5): 572-82, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24612275

RESUMEN

Current methods for the quantitation of membrane protein trafficking rely heavily on microscopy, which has limited quantitative capacity for analyses of cell populations and is cumbersome to perform. Here we describe a simple flow cytometry-based method that circumvents these limitations. The method utilizes fluorescent pulse-width measurements as a highly sensitive indicator to monitor the changes in intracellular distributions of a fluorescently labelled molecule in a cell. Pulse-width analysis enabled us to discriminate cells with target proteins in different intracellular locations including Golgi, lyso-endosomal network and the plasma membrane, as well as detecting morphological changes in organelles such as Golgi perturbation. The movement of endogenous and exogenous retrograde cargo was tracked from the plasma membrane-to-endosomes-to-Golgi, by decreasing pulse-width values. A block in transport upon RNAi-mediated ablation of transport machinery was readily quantified, demonstrating the versatility of this technique to identify pathway inhibitors. We also showed that pulse-width can be exploited to sort and recover cells based on different intracellular staining patterns, e.g. early endosomes and Golgi, opening up novel downstream applications. Overall, the method provides new capabilities for viewing membrane transport in thousands of cells per minute, unbiased analysis of the trafficking of cargo, and the potential for rapid screening of inhibitors of trafficking pathways.


Asunto(s)
Endosomas/metabolismo , Citometría de Flujo/métodos , Aparato de Golgi/metabolismo , Lisosomas/metabolismo , Proteínas de la Membrana/metabolismo , Transporte de Proteínas/fisiología , Línea Celular Tumoral , Membrana Celular/metabolismo , Membrana Celular/fisiología , Endosomas/fisiología , Aparato de Golgi/fisiología , Células HeLa , Humanos , Lisosomas/fisiología
3.
J Biol Chem ; 289(10): 6669-6680, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24425868

RESUMEN

Protein aggregation into intracellular inclusions is a key feature of many neurodegenerative disorders. A common theme has emerged that inappropriate self-aggregation of misfolded or mutant polypeptide sequences is detrimental to cell health. Yet protein quality control mechanisms may also deliberately cluster them together into distinct inclusion subtypes, including the insoluble protein deposit (IPOD) and the juxtanuclear quality control (JUNQ). Here we investigated how the intrinsic oligomeric state of three model systems of disease-relevant mutant protein and peptide sequences relates to the IPOD and JUNQ patterns of aggregation using sedimentation velocity analysis. Two of the models (polyalanine (37A) and superoxide dismutase 1 (SOD1) mutants A4V and G85R) accumulated into the same JUNQ-like inclusion whereas the other, polyglutamine (72Q), formed spatially distinct IPOD-like inclusions. Using flow cytometry pulse shape analysis (PulSA) to separate cells with inclusions from those without revealed the SOD1 mutants and 37A to have abruptly altered oligomeric states with respect to the nonaggregating forms, regardless of whether cells had inclusions or not, whereas 72Q was almost exclusively monomeric until inclusions formed. We propose that mutations leading to JUNQ inclusions induce a constitutively "misfolded" state exposing hydrophobic side chains that attract and ultimately overextend protein quality capacity, which leads to aggregation into JUNQ inclusions. Poly(Q) is not misfolded in this same sense due to universal polar side chains, but is highly prone to forming amyloid fibrils that we propose invoke a different engagement mechanism with quality control.


Asunto(s)
Péptidos/química , Pliegue de Proteína , Superóxido Dismutasa/química , Amiloide/química , Humanos , Cuerpos de Inclusión/química , Mutación , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Péptidos/genética , Superóxido Dismutasa/genética , Superóxido Dismutasa-1
4.
Nat Methods ; 9(5): 467-70, 2012 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-22426490

RESUMEN

We applied pulse-shape analysis (PulSA) to monitor protein localization changes in mammalian cells by flow cytometry. PulSA enabled high-throughput tracking of protein aggregation, translocation from the cytoplasm to the nucleus and trafficking from the plasma membrane to the Golgi as well as stress-granule formation. Combining PulSA with tetracysteine-based oligomer sensors in a cell model of Huntington's disease enabled further separation of cells enriched with monomers, oligomers and inclusion bodies.


Asunto(s)
Citometría de Flujo/métodos , Enfermedad de Huntington/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Aparato de Golgi/metabolismo , Humanos , Proteína Huntingtina , Cuerpos de Inclusión/metabolismo , Transporte de Proteínas
5.
J Biol Chem ; 288(52): 37192-203, 2013 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-24196953

RESUMEN

Our capacity for tracking how misfolded proteins aggregate inside a cell and how different aggregation states impact cell biology remains enigmatic. To address this, we built a new toolkit that enabled the high throughput tracking of individual cells enriched with polyglutamine-expanded Htt exon 1 (Httex1) monomers, oligomers, and inclusions using biosensors of aggregation state and flow cytometry pulse shape analysis. Supplemented with gel filtration chromatography and fluorescence-adapted sedimentation velocity analysis of cell lysates, we collated a multidimensional view of Httex1 aggregation in cells with respect to time, polyglutamine length, expression levels, cell survival, and overexpression of protein quality control chaperones hsp40 (DNAJB1) and hsp70 (HSPA1A). Cell death rates trended higher for Neuro2a cells containing Httex1 in inclusions than with Httex1 dispersed through the cytosol at time points of expression over 2 days. hsp40 stabilized monomers and suppressed inclusion formation but did not otherwise change Httex1 toxicity. hsp70, however, had no major effect on aggregation of Httex1 but increased the survival rate of cells with inclusions. hsp40 and hsp70 also increased levels of a second bicistronic reporter of Httex1 expression, mKate2, and increased total numbers of cells in culture, suggesting these chaperones partly rectify Httex1-induced deficiencies in quality control and growth rates. Collectively, these data suggest that Httex1 overstretches the protein quality control resources and that the defects can be partly rescued by overexpression of hsp40 and hsp70. Importantly, these effects occurred in a pronounced manner for soluble Httex1, which points to Httex1 aggregation occurring subsequently to more acute impacts on the cell.


Asunto(s)
Amiloide/metabolismo , Exones , Regulación de la Expresión Génica , Proteínas del Choque Térmico HSP40/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Amiloide/genética , Animales , Muerte Celular , Línea Celular , Supervivencia Celular , Proteínas del Choque Térmico HSP40/agonistas , Proteínas HSP70 de Choque Térmico/genética , Humanos , Proteína Huntingtina , Ratones , Proteínas del Tejido Nervioso/agonistas
6.
J Biol Chem ; 285(28): 21807-16, 2010 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-20444706

RESUMEN

Huntington disease is caused by expanded polyglutamine sequences in huntingtin, which procures its aggregation into intracellular inclusion bodies (IBs). Aggregate intermediates, such as soluble oligomers, are predicted to be toxic to cells, yet because of a lack of quantitative methods, the kinetics of aggregation in cells remains poorly understood. We used sedimentation velocity analysis to define and compare the heterogeneity and flux of purified huntingtin with huntingtin expressed in mammalian cells under non-denaturing conditions. Non-pathogenic huntingtin remained as hydrodynamically elongated monomers in vitro and in cells. Purified polyglutamine-expanded pathogenic huntingtin formed elongated monomers (2.4 S) that evolved into a heterogeneous aggregate population of increasing size over time (100-6,000 S). However, in cells, mutant huntingtin formed three major populations: monomers (2.3 S), oligomers (mode s(20,w) = 140 S) and IBs (mode s(20,w) = 320,000 S). Strikingly, the oligomers did not change in size heterogeneity or in their proportion of total huntingtin over 3 days despite continued monomer conversion to IBs, suggesting that oligomers are rate-limiting intermediates to IB formation. We also determined how a chaperone known to modulate huntingtin toxicity, Hsc70, influences in-cell huntingtin partitioning. Hsc70 decreased the pool of 140 S oligomers but increased the overall flux of monomers to IBs, suggesting that Hsc70 reduces toxicity by facilitating transfer of oligomers into IBs. Together, our data suggest that huntingtin aggregation is streamlined in cells and is consistent with the 140 S oligomers, which remain invariant over time, as a constant source of toxicity to cells irrespective of total load of insoluble aggregates.


Asunto(s)
Enfermedad de Huntington/genética , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Amiloide/química , Animales , Biofisica/métodos , Clonación Molecular , Exones , Humanos , Proteína Huntingtina , Cinética , Mutagénesis , Péptidos/química , Péptidos/genética , Desnaturalización Proteica , Pliegue de Proteína , Ultracentrifugación
7.
J Mol Biol ; 430(10): 1442-1458, 2018 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-29627459

RESUMEN

Soluble huntingtin exon 1 (Httex1) with expanded polyglutamine (polyQ) engenders neurotoxicity in Huntington's disease. To uncover the physical basis of this toxicity, we performed structural studies of soluble Httex1 for wild-type and mutant polyQ lengths. Nuclear magnetic resonance experiments show evidence for conformational rigidity across the polyQ region. In contrast, hydrogen-deuterium exchange shows absence of backbone amide protection, suggesting negligible persistence of hydrogen bonds. The seemingly conflicting results are explained by all-atom simulations, which show that Httex1 adopts tadpole-like structures with a globular head encompassing the N-terminal amphipathic and polyQ regions and the tail encompassing the C-terminal proline-rich region. The surface area of the globular domain increases monotonically with polyQ length. This stimulates sharp increases in gain-of-function interactions in cells for expanded polyQ, and one of these interactions is with the stress-granule protein Fus. Our results highlight plausible connections between Httex1 structure and routes to neurotoxicity.


Asunto(s)
Mutación con Ganancia de Función , Proteína Huntingtina/química , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Péptidos/genética , Línea Celular , Medición de Intercambio de Deuterio , Exones , Humanos , Proteína Huntingtina/metabolismo , Enlace de Hidrógeno , Dominios Proteicos , Estructura Secundaria de Proteína , Proteína FUS de Unión a ARN/metabolismo , Proteínas de Unión al ARN/metabolismo
8.
Cell Rep ; 19(5): 919-927, 2017 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-28467905

RESUMEN

Competing models exist in the literature for the relationship between mutant Huntingtin exon 1 (Httex1) inclusion formation and toxicity. In one, inclusions are adaptive by sequestering the proteotoxicity of soluble Httex1. In the other, inclusions compromise cellular activity as a result of proteome co-aggregation. Using a biosensor of Httex1 conformation in mammalian cell models, we discovered a mechanism that reconciles these competing models. Newly formed inclusions were composed of disordered Httex1 and ribonucleoproteins. As inclusions matured, Httex1 reconfigured into amyloid, and other glutamine-rich and prion domain-containing proteins were recruited. Soluble Httex1 caused a hyperpolarized mitochondrial membrane potential, increased reactive oxygen species, and promoted apoptosis. Inclusion formation triggered a collapsed mitochondrial potential, cellular quiescence, and deactivated apoptosis. We propose a revised model where sequestration of soluble Httex1 inclusions can remove the trigger for apoptosis but also co-aggregate other proteins, which curtails cellular metabolism and leads to a slow death by necrosis.


Asunto(s)
Amiloide/metabolismo , Apoptosis , Proteína Huntingtina/genética , Exones , Células HEK293 , Células HeLa , Humanos , Proteína Huntingtina/metabolismo , Cuerpos de Inclusión/metabolismo , Potencial de la Membrana Mitocondrial , Mutación , Especies Reactivas de Oxígeno/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo
9.
Methods Mol Biol ; 1270: 227-38, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25702121

RESUMEN

Pulse shape analysis (PulSA) is a flow cytometry-based method that involves the measurement of the pulse width and height of a fluorescently labeled molecule simultaneously, enabling a multidimensional analysis of protein localization in a cell at high speed and throughput. We have used the method to detect morphological changes in organelles such as Golgi fragmentation, track protein trafficking from the cell surface, and also discriminate cells with different target protein localizations such as the Golgi, lyso-endosomal network, and the plasma membrane. Here, we describe the basic experimental setup and analytical methods for performing PulSA to examine membrane trafficking processes. We illustrate in particular the application of PulSA for monitoring the trafficking of the membrane-bound enzyme furin and morphological changes to the Golgi caused by Brefeldin A.


Asunto(s)
Membrana Celular/metabolismo , Citometría de Flujo/métodos , Espacio Intracelular/metabolismo , Proteínas/metabolismo , Animales , Línea Celular , Furina/metabolismo , Aparato de Golgi/metabolismo , Humanos , Transporte de Proteínas
10.
ACS Chem Biol ; 9(7): 1426-31, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24828008

RESUMEN

We developed a new approach to distinguish distinct protein conformations in live cells. The method, exposable tetracysteine (XTC), involved placing an engineered tetracysteine motif into a target protein that has conditional access to biarsenical dye binding by conformational state. XTC was used to distinguish open and closed regulatory conformations of Src family kinases. Substituting just four residues with cysteines in the conserved SH2 domain of three Src-family kinases (c-Src, Lck, Lyn) enabled open and closed conformations to be monitored on the basis of binding differences to biarsenical dyes FlAsH or ReAsH. Fusion of the kinases with a fluorescent protein tracked the kinase presence, and the XTC approach enabled simultaneous assessment of regulatory state. The c-Src XTC biosensor was applied in a boutique screen of kinase inhibitors, which revealed six compounds to induce conformational closure. The XTC approach demonstrates new potential for assays targeting conformational changes in key proteins in disease and biology.


Asunto(s)
Técnicas Biosensibles/métodos , Cisteína/química , Familia-src Quinasas/química , Secuencia de Aminoácidos , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Colorantes/química , Colorantes/metabolismo , Cisteína/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Humanos , Modelos Moleculares , Conformación Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Dominios Homologos src/efectos de los fármacos , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
11.
Methods Mol Biol ; 1017: 85-93, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23719909

RESUMEN

Pulse shape analysis (PulSA) is a flow cytometry-based method that can be used to study protein localization patterns in cells. Examples for its use include tracking the formation of inclusion bodies of polyglutamine-expanded proteins and other aggregating proteins. The method can also be used for phenomena relating to protein movements in cells such as translocation from the cytoplasm to the nucleus, trafficking from the plasma membrane to the Golgi, and stress granule formation. An attractive feature is its capacity to quantify these parameters in whole-cell populations very quickly and in high throughput. We describe the basic experimental details for performing PulSA using expression of GFP-tagged proteins, endogenous proteins labelled immunofluorescently, and organelle dyes.


Asunto(s)
Núcleo Celular/metabolismo , Citometría de Flujo/métodos , Aparato de Golgi/metabolismo , Cuerpos de Inclusión Intranucleares/metabolismo , Péptidos/metabolismo , Transporte Activo de Núcleo Celular , Animales , Línea Celular , Núcleo Celular/genética , Núcleo Celular/patología , Aparato de Golgi/genética , Aparato de Golgi/patología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Cuerpos de Inclusión Intranucleares/genética , Cuerpos de Inclusión Intranucleares/patología , Péptidos/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
12.
J Vis Exp ; (54)2011 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-21897361

RESUMEN

Fluorescent proteins and dyes are essential tools for the study of protein trafficking, localization and function in cells. While fluorescent proteins such as green fluorescence protein (GFP) have been extensively used as fusion partners to proteins to track the properties of a protein of interest, recent developments with smaller tags enable new functionalities of proteins to be examined in cells such as conformational change and protein-association. One small tag system involves a tetracysteine motif (CCXXCC) genetically inserted into a target protein, which binds to biarsenical dyes, ReAsH (red fluorescent) and FlAsH (green fluorescent), with high specificity even in live cells. The TC/biarsenical dye system offers far less steric constraints to the host protein than fluorescent proteins which has enabled several new approaches to measure conformational change and protein-protein interactions. We recently developed a novel application of TC tags as sensors of oligomerization in cells expressing mutant huntingtin, which when mutated aggregates in neurons in Huntington disease. Huntingtin was tagged with two fluorescent dyes, one a fluorescent protein to track protein location, and the second a TC tag which only binds biarsenical dyes in monomers. Hence, changes in colocalization between protein and biarsenical dye reactivity enabled submicroscopic oligomer content to be spatially mapped within cells. Here, we describe how to label TC-tagged proteins fused to a fluorescent protein (Cherry, GFP or CFP) with FlAsH or ReAsH in live mammalian cells and how to quantify the two color fluorescence (Cherry/FlAsH, CFP/FlAsH or GFP/ReAsH combinations).


Asunto(s)
Arsenicales/química , Colorantes/química , Cisteína/análogos & derivados , Proteínas Fluorescentes Verdes/química , Proteínas Luminiscentes/química , Proteínas/química , Cisteína/química , Técnicas Citológicas/métodos , Humanos , Proteína Huntingtina , Microscopía Confocal/métodos , Proteínas del Tejido Nervioso/química , Proteínas Nucleares/química , Proteína Fluorescente Roja
13.
Chem Biol ; 17(4): 371-9, 2010 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-20416508

RESUMEN

Proteins prone to misfolding form large macroscopic deposits in many neurodegenerative diseases. Yet the in situ aggregation kinetics remains poorly understood because of an inability to demarcate precursor oligomers from monomers. We developed a strategy for mapping the localization of soluble oligomers and monomers directly in live cells. Sensors for mutant huntingtin, which forms aggregates in Huntington's disease, were made by introducing a tetracysteine motif into huntingtin that becomes occluded from binding biarsenical fluorophores in oligomers, but not monomers. Up to 70% of the diffusely distributed huntingtin molecules appeared as submicroscopic oligomers in individual neuroblastoma cells expressing mutant huntingtin. We anticipate the sensors to enable insight into cellular mechanisms mediated by oligomers and monomers and for the approach to be adaptable more generally in the study of protein self-association.


Asunto(s)
Enfermedad de Huntington/metabolismo , Proteínas del Tejido Nervioso/análisis , Animales , Línea Celular , Humanos , Mutación , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Conformación Proteica , Pliegue de Proteína , Multimerización de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA