Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 591(7850): 431-437, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33505021

RESUMEN

Lysosomes have fundamental physiological roles and have previously been implicated in Parkinson's disease1-5. However, how extracellular growth factors communicate with intracellular organelles to control lysosomal function is not well understood. Here we report a lysosomal K+ channel complex that is activated by growth factors and gated by protein kinase B (AKT) that we term lysoKGF. LysoKGF consists of a pore-forming protein TMEM175 and AKT: TMEM175 is opened by conformational changes in, but not the catalytic activity of, AKT. The minor allele at rs34311866, a common variant in TMEM175, is associated with an increased risk of developing Parkinson's disease and reduces channel currents. Reduction in lysoKGF function predisposes neurons to stress-induced damage and accelerates the accumulation of pathological α-synuclein. By contrast, the minor allele at rs3488217-another common variant of TMEM175, which is associated with a decreased risk of developing Parkinson's disease-produces a gain-of-function in lysoKGF during cell starvation, and enables neuronal resistance to damage. Deficiency in TMEM175 leads to a loss of dopaminergic neurons and impairment in motor function in mice, and a TMEM175 loss-of-function variant is nominally associated with accelerated rates of cognitive and motor decline in humans with Parkinson's disease. Together, our studies uncover a pathway by which extracellular growth factors regulate intracellular organelle function, and establish a targetable mechanism by which common variants of TMEM175 confer risk for Parkinson's disease.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Lisosomas/metabolismo , Complejos Multiproteicos/metabolismo , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Canales de Potasio/metabolismo , Potasio/metabolismo , Animales , Biocatálisis , Neuronas Dopaminérgicas/metabolismo , Femenino , Mutación con Ganancia de Función , Células HEK293 , Humanos , Mutación con Pérdida de Función , Masculino , Ratones , Ratones Noqueados , Destreza Motora , Complejos Multiproteicos/química , Complejos Multiproteicos/deficiencia , Complejos Multiproteicos/genética , Enfermedad de Parkinson/genética , Canales de Potasio/química , Canales de Potasio/deficiencia , Canales de Potasio/genética , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , alfa-Sinucleína/metabolismo
2.
Nat Chem Biol ; 19(11): 1372-1383, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37592155

RESUMEN

RNA molecules with the expanded CAG repeat (eCAGr) may undergo sol-gel phase transitions, but the functional impact of RNA gelation is completely unknown. Here, we demonstrate that the eCAGr RNA may form cytoplasmic gel-like foci that are rapidly degraded by lysosomes. These RNA foci may significantly reduce the global protein synthesis rate, possibly by sequestering the translation elongation factor eEF2. Disrupting the eCAGr RNA gelation restored the global protein synthesis rate, whereas enhanced gelation exacerbated this phenotype. eEF2 puncta were significantly enhanced in brain slices from a knock-in mouse model and from patients with Huntington's disease, which is a CAG expansion disorder expressing eCAGr RNA. Finally, neuronal expression of the eCAGr RNA by adeno-associated virus injection caused significant behavioral deficits in mice. Our study demonstrates the existence of RNA gelation inside the cells and reveals its functional impact, providing insights into repeat expansion diseases and functional impacts of RNA phase transition.


Asunto(s)
Enfermedad de Huntington , Expansión de Repetición de Trinucleótido , Humanos , Ratones , Animales , ARN/genética , ARN/metabolismo , Biosíntesis de Proteínas , Enfermedad de Huntington/genética , Modelos Animales de Enfermedad , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo
3.
Nature ; 575(7781): 203-209, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31666698

RESUMEN

Accumulation of mutant proteins is a major cause of many diseases (collectively called proteopathies), and lowering the level of these proteins can be useful for treatment of these diseases. We hypothesized that compounds that interact with both the autophagosome protein microtubule-associated protein 1A/1B light chain 3 (LC3)1 and the disease-causing protein may target the latter for autophagic clearance. Mutant huntingtin protein (mHTT) contains an expanded polyglutamine (polyQ) tract and causes Huntington's disease, an incurable neurodegenerative disorder2. Here, using small-molecule-microarray-based screening, we identified four compounds that interact with both LC3 and mHTT, but not with the wild-type HTT protein. Some of these compounds targeted mHTT to autophagosomes, reduced mHTT levels in an allele-selective manner, and rescued disease-relevant phenotypes in cells and in vivo in fly and mouse models of Huntington's disease. We further show that these compounds interact with the expanded polyQ stretch and could lower the level of mutant ataxin-3 (ATXN3), another disease-causing protein with an expanded polyQ tract3. This study presents candidate compounds for lowering mHTT and potentially other disease-causing proteins with polyQ expansions, demonstrating the concept of lowering levels of disease-causing proteins using autophagosome-tethering compounds.


Asunto(s)
Alelos , Evaluación Preclínica de Medicamentos/métodos , Proteína Huntingtina/antagonistas & inhibidores , Proteína Huntingtina/genética , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/genética , Mutación/genética , Animales , Ataxina-3/genética , Autofagosomas/metabolismo , Autofagia , Modelos Animales de Enfermedad , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Femenino , Humanos , Proteína Huntingtina/química , Proteína Huntingtina/metabolismo , Masculino , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación/efectos de los fármacos , Neuronas/citología , Péptidos/genética , Fenotipo , Reproducibilidad de los Resultados
4.
Proc Natl Acad Sci U S A ; 119(10): e2114303119, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35238684

RESUMEN

Identifying inhibitors of pathogenic proteins is the major strategy of targeted drug discoveries. This strategy meets challenges in targeting neurodegenerative disorders such as Huntington's disease (HD), which is mainly caused by the mutant huntingtin protein (mHTT), an "undruggable" pathogenic protein with unknown functions. We hypothesized that some of the chemical binders of mHTT may change its conformation and/or stability to suppress its downstream toxicity, functioning similarly to an "inhibitor" under a broader definition. We identified 21 potential mHTT selective binders through a small-molecule microarray­based screening. We further tested these compounds using secondary phenotypic screens for their effects on mHTT-induced toxicity and revealed four potential mHTT-binding compounds that may rescue HD-relevant phenotypes. Among them, a Food and Drug Administration­approved drug, desonide, was capable of suppressing mHTT toxicity in HD cellular and animal models by destabilizing mHTT through enhancing its polyubiquitination at the K6 site. Our study reveals the therapeutic potential of desonide for HD treatment and provides the proof of principle for a drug discovery pipeline: target-binder screens followed by phenotypic validation and mechanistic studies.


Asunto(s)
Desonida , Proteína Huntingtina , Enfermedad de Huntington , Mutación , Animales , Desonida/química , Desonida/farmacología , Modelos Animales de Enfermedad , Proteína Huntingtina/química , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Ratones , Ratones Transgénicos , Estabilidad Proteica/efectos de los fármacos
5.
Chem Soc Rev ; 51(21): 8832-8876, 2022 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-36218065

RESUMEN

Targeted protein degradation (TPD) provides unprecedented opportunities for drug discovery. While the proteolysis-targeting chimera (PROTAC) technology has already entered clinical trials and changed the landscape of small-molecule drugs, new degrader technologies harnessing alternative degradation machineries, especially lysosomal pathways, have emerged and broadened the spectrum of degradable targets. We have recently proposed the concept of autophagy-tethering compounds (ATTECs) that hijack the autophagy protein microtubule-associated protein 1A/1B light chain 3 (LC3) for targeted degradation. Other groups also reported degrader technologies engaging lysosomal pathways through different mechanisms including AUTACs, AUTOTACs, LYTACs and MoDE-As. In this review, we analyse and discuss ATTECs along with other lysosomal-relevant degrader technologies. Finally, we will briefly summarize the current status of these degrader technologies and envision possible future studies.


Asunto(s)
Descubrimiento de Drogas , Proteínas , Proteolisis , Proteínas/metabolismo , Autofagia , Lisosomas/metabolismo
6.
Sensors (Basel) ; 23(21)2023 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-37960410

RESUMEN

Smart agriculture utilizes Internet of Things (IoT) technologies to enable low-cost electrical conductivity (EC) sensors to support farming intelligence. Due to aging and changes in weather and soil conditions, EC sensors are prone to long-term drift over years of operation. Therefore, regular recalibration is necessary to ensure data accuracy. In most existing solutions, an EC sensor is calibrated by using the standard sensor to build the calibration table. This paper proposes SensorTalk3, an ensemble approach of machine learning models including XGBOOST and Random Forest, which can be executed at an edge device (e.g., Raspberry Pi) without GPU acceleration. Our study indicates that the soil information (both temperature and moisture sensor data) plays an important role in SensorTalk3, which significantly outperforms the existing calibration approaches. The MAPE of SensorTalk3 can be as low as 1.738%, compared to the 7.792% error of the original sensor. Our study indicates that when the errors of uncalibrated moisture and temperature sensors are not larger than 8.3%, SensorTalk3 can accurately calibrate EC. SensorTalk3 can perform model training during data collection at the edge node. When all training data are collected, AI training is also finished at the edge node. Such an AI training approach has not been found in existing edge AI approaches. We also proposed the dual-sensor detection solution to determine when to conduct recalibration. The overhead of this solution is less than twice the optimal detection scenario (which cannot be achieved practically). If the two non-standard sensors are homogeneous and stable, then the optimal detection scenario can be approached. Conventional methods require training calibration AI models in the cloud. However, SensorTalk3 introduces a significant advancement by enabling on-site transfer learning in the edge node. Given the abundance of farming sensors deployed in the fields, performing local transfer learning using low-cost edge nodes proves to be a more cost-effective solution for farmers.

7.
Trends Biochem Sci ; 43(6): 424-435, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29636213

RESUMEN

Expanded polyglutamine (polyQ) stretches within endogenous proteins cause at least nine human diseases. The structural basis of polyQ pathogenesis is the key to understanding fundamental mechanisms of these diseases, but it remains unclear and controversial due to a lack of polyQ protein structures at the single-atom level. Various hypotheses have been proposed to explain the structure-cytotoxicity relationship of pathogenic proteins with polyQ expansion, largely based on indirect evidence. Here we review these hypotheses and their supporting evidence, along with additional insights from recent structural biology and chemical biology studies, with a focus on Huntingtin (HTT), the most extensively studied polyQ disease protein. Lastly, we propose potential novel strategies that may further clarify the conformation-cytotoxicity relationship of polyQ proteins.


Asunto(s)
Proteína Huntingtina/química , Péptidos/química , Humanos , Proteína Huntingtina/metabolismo , Péptidos/metabolismo , Conformación Proteica
8.
Hum Mol Genet ; 29(2): 216-227, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31813995

RESUMEN

Huntington's disease (HD) is a neurodegenerative disorder caused by an expanded polyglutamine tract in the huntingtin (HTT) protein. Mutant HTT (mHTT) toxicity is caused by its aggregation/oligomerization. The striatum is the most vulnerable region, although all brain regions undergo neuronal degeneration in the disease. Here we show that the levels of Bim, a BH3-only protein, are significantly increased in HD human post-mortem and HD mouse striata, correlating with neuronal death. Bim reduction ameliorates mHTT neurotoxicity in HD cells. In the HD mouse model, heterozygous Bim knockout significantly mitigates mHTT accumulation and neuronal death, ameliorating disease-associated phenotypes and lifespan. Therefore, Bim could contribute to the progression of HD.


Asunto(s)
Proteína 11 Similar a Bcl2/metabolismo , Cuerpo Estriado/metabolismo , Proteína Huntingtina/genética , Enfermedad de Huntington/metabolismo , Neuronas/patología , Anciano , Animales , Proteína 11 Similar a Bcl2/genética , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Técnicas de Inactivación de Genes , Heterocigoto , Humanos , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/mortalidad , Enfermedad de Huntington/patología , Masculino , Ratones , Persona de Mediana Edad , Neuronas/metabolismo , Fenotipo , Agregado de Proteínas/genética , ARN Interferente Pequeño
10.
Acta Pharmacol Sin ; 42(10): 1556-1566, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33495516

RESUMEN

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by toxic aggregates of mutant huntingtin protein (mHTT) in the brain. Decreasing mHTT is a potential strategy for therapeutic purpose of HD. Valosin-containing protein (VCP/p97) is a crucial regulator of proteostasis, which regulates the degradation of damaged protein through proteasome and autophagy pathway. Since VCP has been implicated in pathogenesis of HD as well as other neurodegenerative diseases, small molecules that specifically regulate the activity of VCP may be of therapeutic benefits for HD patients. In this study we established a high-throughput screening biochemical assay for VCP ATPase activity measurement and identified gossypol, a clinical approved drug in China, as a novel modulator of VCP. Gossypol acetate dose-dependently inhibited the enzymatic activity of VCP in vitro with IC50 of 6.53±0.6 µM. We further demonstrated that gossypol directly bound to the interface between the N and D1 domains of VCP. Gossypol acetate treatment not only lowered mHTT levels and rescued HD-relevant phenotypes in HD patient iPS-derived Q47 striatal neurons and HD knock-in mouse striatal cells, but also improved motor function deficits in both Drosophila and mouse HD models. Taken together, gossypol acetate acted through a gain-of-function way to induce the formation of VCP-LC3-mHTT ternary complex, triggering autophagic degradation of mHTT. This study reveals a new strategy for treatment of HD and raises the possibility that an existing drug can be repurposed as a new treatment of neurodegenerative diseases.


Asunto(s)
Autofagia/efectos de los fármacos , Gosipol/uso terapéutico , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Animales , Drosophila , Inhibidores Enzimáticos/uso terapéutico , Femenino , Células HEK293 , Células HeLa , Humanos , Proteína Huntingtina/química , Proteína Huntingtina/genética , Masculino , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Mutación , Multimerización de Proteína/efectos de los fármacos , Proteolisis/efectos de los fármacos , Proteína que Contiene Valosina/antagonistas & inhibidores , Proteína que Contiene Valosina/metabolismo
11.
RNA Biol ; 17(4): 500-516, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31928144

RESUMEN

Huntington's Disease (HD) is a monogenetic neurodegenerative disorder mainly caused by the cytotoxicity of the mutant HTT protein (mHTT) encoded by the mutant HTT gene. Lowering HTT mRNA has been extensively studied as a potential therapeutic strategy, but how its level is regulated endogenously has been unclear. Here we report that the RNA-binding protein (RBP) HuR interacts with and stabilizes HTT mRNA in an mHTT-dependent manner. In HD cells but not wild-type cells, siRNA knockdown or CRISPR-induced heterozygous knockout of HuR decreased HTT mRNA stability. HuR interacted with HTT mRNA at a conserved site in exon 11 rather than the 3'-UTR region of the mRNA. Interestingly, this interaction was dependent on the presence of mHTT, likely via the activation of MAPK11, which enhanced cytosolic localization of the HuR protein. Thus, mHTT, MAPK11 and HuR may form a positive feedback loop that stabilizes HTT mRNA and enhances mHTT accumulation, which may contribute to HD progression. Our data reveal a novel regulatory mechanism of HTT mRNA via non-canonical binding of HuR.


Asunto(s)
Proteína 1 Similar a ELAV/metabolismo , Proteína Huntingtina/química , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Mutación , Regiones no Traducidas 3' , Animales , Sitios de Unión , Sistemas CRISPR-Cas , Línea Celular , Proteína 1 Similar a ELAV/genética , Exones , Retroalimentación Fisiológica , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Enfermedad de Huntington/metabolismo , Ratones , Estabilidad del ARN
12.
Nat Chem Biol ; 13(11): 1152-1154, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28869595

RESUMEN

Protein misfolding is a common theme in neurodegenerative disorders including Huntington's disease (HD). The HD-causing mutant huntingtin protein (mHTT) has an expanded polyglutamine (polyQ) stretch that may adopt multiple conformations, and the most toxic of these is the one recognized by antibody 3B5H10. Here we show that the 3B5H10-recognized mHTT species has a slower degradation rate due to its resistance to selective autophagy in human cells and brains, revealing mechanisms of its higher toxicity.


Asunto(s)
Autofagia , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/patología , Mutación , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Péptidos/genética , Poliubiquitina/metabolismo , Conformación Proteica , Proteolisis , Secuencias Repetitivas de Ácidos Nucleicos , Ubiquitinación
13.
Brain ; 141(6): 1782-1798, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29608652

RESUMEN

See Huang and Gitler (doi:10.1093/brain/awy112) for a scientific commentary on this article.Lowering the levels of disease-causing proteins is an attractive treatment strategy for neurodegenerative disorders, among which Huntington's disease is an appealing disease for testing this strategy because of its monogenetic nature. Huntington's disease is mainly caused by cytotoxicity of the mutant HTT protein with an expanded polyglutamine repeat tract. Lowering the soluble mutant HTT may reduce its downstream toxicity and provide potential treatment for Huntington's disease. This is hard to achieve by small-molecule compound drugs because of a lack of effective targets. Here we demonstrate Gpr52, an orphan G protein-coupled receptor, as a potential Huntington's disease drug target. Knocking-out Gpr52 significantly reduces mutant HTT levels in the striatum and rescues Huntington's disease-associated behavioural phenotypes in a knock-in Huntington's disease mouse model expressing endogenous mutant Htt. Importantly, a novel Gpr52 antagonist E7 reduces mutant HTT levels and rescues Huntington's disease-associated phenotypes in cellular and mouse models. Our study provides an entry point for Huntington's disease drug discovery by targeting Gpr52.


Asunto(s)
Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Mutación/genética , Receptores Acoplados a Proteínas G/deficiencia , Factores de Edad , Animales , Benzamidas/uso terapéutico , Cuerpo Estriado/metabolismo , AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Conducta Exploratoria/fisiología , Marcha/fisiología , Células HEK293 , Humanos , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/fisiopatología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Ratones Transgénicos , Neuronas/patología , Fenotipo , Quinoxalinas/uso terapéutico , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Tiofenos/uso terapéutico , Caminata/fisiología
14.
Acta Pharmacol Sin ; 37(10): 1307-1314, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27264314

RESUMEN

AIM: The accumulation of disease-causing proteins is a common hallmark of many neurodegenerative disorders. Measuring the degradation of such proteins using high-throughput-compatible assays is highly desired for the identification of genetic and chemical modulators of degradation. For example, Huntington's disease (HD) is an incurable hereditary neurodegenerative disorder caused by the cytotoxicity of mutant huntingtin protein (mHTT). The high-throughput measurement of mHTT degradation is important in HD drug discovery and research. Existing methods for such purposes have limitations due to their dependence on protein tags or pan protein synthesis inhibitors. Here, we report a high-throughput-compatible pulse-chase method (CH-chase) for the measurement of endogenous tag-free huntingtin protein (HTT) degradation based on Click chemistry and Homogeneous Time Resolved Fluorescence (HTRF) technologies. METHODS: The pulsed-labeled proteins were conjugated with biotin using the click reaction strain-promoted alkyne-azide cycloaddition (SPAAC), and the chase signals were calculated by measuring the reduction percentage of the HTT HTRF signals after pull-down with streptavidin beads. RESULTS: We validated that the signals were within the linear detection range and were HTT-specific. We successfully measured the degradation of endogenous HTT in a high-throughput-compatible format using 96-well plates. The predicted changes of HTT degradation by known modifiers were observed, which confirmed that the assay is suitable for the identification of HTT degradation modifiers. CONCLUSION: We have established the first high-throughput-compatible assay capable of measuring endogenous, tag-free HTT degradation, providing a valuable tool for HD research and drug discovery. The method could be applied to other proteins and can facilitate research on other neurodegenerative disorders and proteinopathies.


Asunto(s)
Proteína Huntingtina/metabolismo , Animales , Línea Celular , Química Clic , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Proteolisis
15.
Nature ; 457(7230): 741-4, 2009 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-19092807

RESUMEN

Several neurotransmitters act through G-protein-coupled receptors to evoke a 'slow' excitation of neurons. These include peptides, such as substance P and neurotensin, as well as acetylcholine and noradrenaline. Unlike the fast (approximately millisecond) ionotropic actions of small-molecule neurotransmitters, the slow excitation is not well understood at the molecular level, but can be mainly attributed to suppressing K(+) currents and/or activating a non-selective cation channel. The molecular identity of this cation channel has yet to be determined; similarly, how the channel is activated and its relative contribution to neuronal excitability induced by the neuropeptides are unknown. Here we show that, in the mouse hippocampal and ventral tegmental area neurons, substance P and neurotensin activate a channel complex containing NALCN and a large previously unknown protein UNC-80. The activation by substance P through TACR1 (a G-protein-coupled receptor for substance P) occurs by means of a unique mechanism: it does not require G-protein activation but is dependent on Src family kinases. These findings identify NALCN as the cation channel activated by substance P receptor, and suggest that UNC-80 and Src family kinases, rather than a G protein, are involved in the coupling from receptor to channel.


Asunto(s)
Canales Iónicos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurotransmisores/farmacología , Animales , Línea Celular , Conductividad Eléctrica , Guanosina Trifosfato/metabolismo , Proteínas de Unión al GTP Heterotriméricas , Hipocampo/citología , Humanos , Canales Iónicos/agonistas , Canales Iónicos/genética , Proteínas de la Membrana , Ratones , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Neurotensina/farmacología , Receptores de Neuroquinina-1/metabolismo , Sustancia P/farmacología , Transfección , Área Tegmental Ventral/citología , Familia-src Quinasas/metabolismo
16.
FASEB J ; 27(5): 1820-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23325320

RESUMEN

Most neurodegenerative diseases are linked to aberrant accumulation of aggregation-prone proteins. Among them, Huntington's disease (HD) is caused by an expanded polyglutamine repeat stretch in the N terminus of the mutant huntingtin protein (mHTT), which gets cleaved and aggregates in the brain. Recently established human induced pluripotent stem cell-derived HD neurons exhibit some disease-relevant phenotypes and provide tools for HD research. However, they have limitations such as genetic heterogeneity and an absence of mHTT aggregates and lack a robust neurodegeneration phenotype. In addition, the relationship between the phenotype and mHTT levels has not been elucidated. Herein, we present a human embryonic stem cell (hESC)-derived HD neuronal model expressing HTTexon1 fragments, which addresses the deficiencies enumerated above. The wild-type and HD lines are derived from an isogenic background and exhibit insoluble mHTT aggregates and neurodegeneration. We also demonstrate a quantitative relationship between neurodegeneration and soluble monomeric (but not oligomeric or aggregated) mHTT levels. Reduction of ~10% of mHTT is sufficient to prevent toxicity, whereas ~90% reduction of wild-type HTT is safe and well-tolerated in these cells. A known HD toxicity modifier (Rhes) showed expected rescue of neurodegeneration. Therefore, the hESC-derived neuronal models complement existing induced pluripotent stem cell-derived neuronal models and provide valuable tools for HD research.


Asunto(s)
Células Madre Embrionarias/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Proteínas del Tejido Nervioso/genética , Línea Celular , Proteínas de Unión al GTP/genética , Técnicas de Silenciamiento del Gen , Humanos , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Modelos Biológicos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/fisiopatología , Neuronas/metabolismo , Neuronas/patología , Transfección
17.
Autophagy ; 20(2): 451-453, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37876230

RESUMEN

Phase transitions (PT) of biomolecules are heavily involved in neurodegenerative disorders. Almost all previous studies were focusing on the PT of misfolded proteins whereas RNA molecules containing expanded repeats such as the CAG repeats are also able to undergo PT in vitro, a process called RNA gelation. Meanwhile, the expanded CAG repeat (eCAGr) RNA forms condensates that are largely observed only in the nuclei and exhibit liquid-like properties without obvious gelation. Thus, whether eCAGr RNA gelation occurs in cells and what function it is involved in remained elusive. We recently discovered that eCAGr RNA forms solid-like RNA gels in the cytoplasm, but they are rapidly cleared by the lysosomes via an autophagy-independent but LAMP2C-depdent pathway, making their presence in the cytoplasm difficult to be observed. We further revealed that these RNA gels sequester EEF2 in the cells and thus suppress global protein synthesis. In vivo expression of eCAGr RNA alone without detectable protein expression in the mouse model led to neurodegeneration-relevant electrophysiological and behavioral phenotypes, demonstrating its possible pathogenic roles.


Asunto(s)
Enfermedad de Huntington , ARN , Ratones , Animales , ARN/metabolismo , Expansión de Repetición de Trinucleótido/genética , Enfermedad de Huntington/metabolismo , Autofagia/genética , Lisosomas/metabolismo , Geles , Proteína Huntingtina/metabolismo
18.
Cell Death Differ ; 31(3): 348-359, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38332048

RESUMEN

Hereditary spastic paraplegia (HSP) is a group of inherited neurodegenerative disorders characterized by progressive lower limb spasticity and weakness. One subtype of HSP, known as SPG54, is caused by biallelic mutations in the DDHD2 gene. The primary pathological feature observed in patients with SPG54 is the massive accumulation of lipid droplets (LDs) in the brain. However, the precise mechanisms and roles of DDHD2 in regulating lipid homeostasis are not yet fully understood. Through Affinity Purification-Mass Spectroscopy (AP-MS) analysis, we identify that DDHD2 interacts with multiple members of the ATG8 family proteins (LC3, GABARAPs), which play crucial roles in lipophagy. Mutational analysis reveals the presence of two authentic LIR motifs in DDHD2 protein that are essential for its binding to LC3/GABARAPs. We show that DDHD2 deficiency leads to LD accumulation, while enhanced DDHD2 expression reduces LD formation. The LC3/GABARAP-binding capacity of DDHD2 and the canonical autophagy pathway both contribute to its LD-eliminating activity. Moreover, DDHD2 enhances the colocalization between LC3B and LDs to promote lipophagy. LD·ATTEC, a small molecule that tethers LC3 to LDs to enhance their autophagic clearance, effectively counteracts DDHD2 deficiency-induced LD accumulation. These findings provide valuable insights into the regulatory roles of DDHD2 in LD catabolism and offer a potential therapeutic approach for treating SPG54 patients.


Asunto(s)
Fosfolipasas , Paraplejía Espástica Hereditaria , Humanos , Autofagia/genética , Familia de las Proteínas 8 Relacionadas con la Autofagia , Mutación/genética , Fosfolipasas/genética , Paraplejía Espástica Hereditaria/genética , Paraplejía Espástica Hereditaria/patología
19.
Life Sci ; 339: 122378, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38142737

RESUMEN

AIMS: The paucity of functional annotations on hundreds of KCNQ2 variants impedes the diagnosis and treatment of KCNQ2-related disorders. The aims of this work were to determine the functional properties of 331 clinical KCNQ2 variants, interpreted the pathogenicity of 331 variants using functional data,and explored the association between homomeric channel functions and phenotypes. MAIN METHODS: We collected 145 KCNQ2 variants from 232 epilepsy patients and 186 KCNQ2 missense variants from the ClinVar database. Whole-cell patch-clamp recording was used to classify the function of 331 variants. Subsequently, we proposed 24 criteria for the pathogenicity interpretation of KCNQ2 variants and used them to assess pathogenicity of 331 variants. Finally, we analyzed the clinical phenotypes of patients carrying these variants, and explored the correlations between functional mechanisms and phenotypes. KEY FINDINGS: In the homozygous state, 287 were classified as loss-of-function and 14 as gain-of-function. In the more clinically relative heterozygous state, 200 variants exhibited functional impairment, 121 of which showed dominant-negative effects on wild-type KCNQ2 subunits. After introducing functional data as strong-level evidence to interpret pathogenicity, over half of variants (169/331) were reclassified and 254 were classified as pathogenic/likely pathogenic. Moreover, dominant-negative effect and haploinsufficiency were identified as primary mechanisms in DEE/ID and SeLNE, respectively. The degree of impairment of channel function correlated with the phenotype severity. SIGNIFICANCE: Our study reveals the possible cause of KCNQ2-related disorders at the molecular level, provides compelling evidence for clinical classification of KCNQ2 variants, and expands the knowledge of correlations between functional mechanisms and phenotypes.


Asunto(s)
Epilepsia , Humanos , Virulencia , Epilepsia/genética , Mutación Missense , Fenotipo , Heterocigoto , Canal de Potasio KCNQ2/genética
20.
J Cell Biol ; 223(8)2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-38913026

RESUMEN

The double-stranded RNA-binding protein Staufen1 (STAU1) regulates a variety of physiological and pathological events via mediating RNA metabolism. STAU1 overabundance was observed in tissues from mouse models and fibroblasts from patients with neurodegenerative diseases, accompanied by enhanced mTOR signaling and impaired autophagic flux, while the underlying mechanism remains elusive. Here, we find that endogenous STAU1 forms dynamic cytoplasmic condensate in normal and tumor cell lines, as well as in mouse Huntington's disease knockin striatal cells. STAU1 condensate recruits target mRNA MTOR at its 5'UTR and promotes its translation both in vitro and in vivo, and thus enhanced formation of STAU1 condensate leads to mTOR hyperactivation and autophagy-lysosome dysfunction. Interference of STAU1 condensate normalizes mTOR levels, ameliorates autophagy-lysosome function, and reduces aggregation of pathological proteins in cellular models of neurodegenerative diseases. These findings highlight the importance of balanced phase separation in physiological processes, suggesting that modulating STAU1 condensate may be a strategy to mitigate the progression of neurodegenerative diseases with STAU1 overabundance.


Asunto(s)
Autofagia , Biosíntesis de Proteínas , Proteínas de Unión al ARN , Serina-Treonina Quinasas TOR , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Serina-Treonina Quinasas TOR/metabolismo , Serina-Treonina Quinasas TOR/genética , Animales , Humanos , Autofagia/genética , Ratones , Proteínas del Citoesqueleto/metabolismo , Proteínas del Citoesqueleto/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/genética , Lisosomas/metabolismo , Lisosomas/genética , Transducción de Señal , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Enfermedad de Huntington/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA