Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(50): 31902-31913, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33257563

RESUMEN

Proteostasis collapse, the diminished ability to maintain protein homeostasis, has been established as a hallmark of nematode aging. However, whether proteostasis collapse occurs in humans has remained unclear. Here, we demonstrate that proteostasis decline is intrinsic to human senescence. Using transcriptome-wide characterization of gene expression, splicing, and translation, we found a significant deterioration in the transcriptional activation of the heat shock response in stressed senescent cells. Furthermore, phosphorylated HSF1 nuclear localization and distribution were impaired in senescence. Interestingly, alternative splicing regulation was also dampened. Surprisingly, we found a decoupling between different unfolded protein response (UPR) branches in stressed senescent cells. While young cells initiated UPR-related translational and transcriptional regulatory responses, senescent cells showed enhanced translational regulation and endoplasmic reticulum (ER) stress sensing; however, they were unable to trigger UPR-related transcriptional responses. This was accompanied by diminished ATF6 nuclear localization in stressed senescent cells. Finally, we found that proteasome function was impaired following heat stress in senescent cells, and did not recover upon return to normal temperature. Together, our data unraveled a deterioration in the ability to mount dynamic stress transcriptional programs upon human senescence with broad implications on proteostasis control and connected proteostasis decline to human aging.


Asunto(s)
Envejecimiento/fisiología , Senescencia Celular/fisiología , Proteostasis/genética , Activación Transcripcional/fisiología , Factor de Transcripción Activador 6/metabolismo , Línea Celular , Núcleo Celular/metabolismo , Estrés del Retículo Endoplásmico/genética , Respuesta al Choque Térmico/genética , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Biosíntesis de Proteínas , RNA-Seq , Respuesta de Proteína Desplegada/genética
2.
Int J Hyperthermia ; 38(1): 1495-1501, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34666607

RESUMEN

OBJECTIVES: Mutations in the human IQSEC2 gene are associated with drug-resistant epilepsy and severe behavioral dysfunction. We have focused on understanding one human IQSEC2 missense mutation (A350V) for which we have created a corresponding A350V IQSEC2 mouse model by CRISPR which demonstrates seizures when the mice are 15-20 days old and impaired social vocalizations in adulthood. We observed that a child with the A350V mutation stops having seizures when experiencing a fever of greater than 38 °C. In this study, we first sought to determine if we could recapitulate this phenomenon in A350V 15-20 day old mice using a previously established protocol to raise body temperature to 39 °C achieved by housing the mice at 37 °C. We then sought to determine if mice in whom seizure activity had been prevented as pups would develop social vocalization activity in adulthood. METHODS: 15-20 day old A350V male mice were housed either at 37 °C or 22 °C. Ultrasonic vocalizations of these mice were assessed at 8-10 weeks in response to a female stimulus. RESULTS: Housing of 15-20 day old A350V mice at 37 °C resulted in a reduction in lethal seizures to 2% (1/41) compared to 45% (48/108) in mice housed at 22 °C, p = 0.0001. Adult A350V mice who had been housed at 37 °C as pups displayed a significant improvement in the production of social vocalizations. CONCLUSION: Raising the body temperature by raising the ambient temperature might provide a means to reduce seizures associated with the A350V IQSEC2 mutation and thereby allow for an improved neurodevelopmental trajectory.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido , Convulsiones/prevención & control , Temperatura , Vocalización Animal , Animales , Femenino , Factores de Intercambio de Guanina Nucleótido/genética , Vivienda , Masculino , Ratones , Proteínas del Tejido Nervioso
3.
Biochim Biophys Acta Mol Cell Res ; 1864(12): 2449-2459, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28964849

RESUMEN

Although Hematopoietic Stem and Progenitor Cell (HSPC) proliferation, survival and expansion have been shown to be supported by the cooperative action of different cytokines, little is known about the intracellular signaling pathways that are activated by cytokines upon binding to their receptors. Our study showed that Growth factor receptor-bound protein 2 (Grb2) mRNAs are preferentially expressed in HSC compared to progenitors and differentiated cells of the myeloid and erythroid lineages. Conditional deletion of Grb2 induced a rapid decline of erythroid and myeloid progenitors and a progressive decline of HSC numbers in steady state conditions. We showed that when transplanted, Grb2 deleted bone marrow cells could not reconstitute irradiated recipients. Strinkingly, Grb2 deletion did not modify HSPC quiescence, but impaired LT-HSC and progenitors ability to respond a proliferative signal induced by 5FU in vivo and by various cytokines in vitro. We showed finally that Grb2 links IL3 signaling to the ERK/MAPK proliferative pathway and that both SH2 and SH3 domains of Grb2 are crucial for IL3 signaling in progenitor cells. Our findings position Grb2 as a key adaptor that integrates various cytokines response in cycling HSPC.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Proteína Adaptadora GRB2/genética , Células Madre Hematopoyéticas/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Proliferación Celular/genética , Células Eritroides/metabolismo , Técnicas de Inactivación de Genes , Células Madre Hematopoyéticas/citología , Ratones , Células Mieloides/metabolismo , Transducción de Señal
4.
Nat Commun ; 13(1): 516, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-35082301

RESUMEN

Protein aggregation is a hallmark of neurodegeneration. Here, we find that Huntington's disease-related HTT-polyQ aggregation induces a cellular proteotoxic stress response, while ALS-related mutant FUS (mutFUS) aggregation leads to deteriorated proteostasis. Further exploring chaperone function as potential modifiers of pathological aggregation in these contexts, we reveal divergent effects of naturally-occurring chaperone isoforms on different aggregate types. We identify a complex of the full-length (FL) DNAJB14 and DNAJB12, that substantially protects from mutFUS aggregation, in an HSP70-dependent manner. Their naturally-occurring short isoforms, however, do not form a complex, and lose their ability to preclude mutFUS aggregation. In contrast, DNAJB12-short alleviates, while DNAJB12-FL aggravates, HTT-polyQ aggregation. DNAJB14-FL expression increases the mobility of mutFUS aggregates, and restores the deteriorated proteostasis in mutFUS aggregate-containing cells and primary neurons. Our results highlight a maladaptive cellular response to pathological aggregation, and reveal a layer of chaperone network complexity conferred by DNAJ isoforms, in regulation of different aggregate types.


Asunto(s)
Proteínas del Choque Térmico HSP40/metabolismo , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Chaperonas Moleculares/metabolismo , Péptidos/metabolismo , Agregado de Proteínas , Proteína FUS de Unión a ARN/metabolismo , Células HEK293 , Proteínas del Choque Térmico HSP40/química , Humanos , Chaperonas Moleculares/química , Neuronas/metabolismo , Imagen Óptica , Isoformas de Proteínas/metabolismo , Proteostasis
5.
Epilepsy Res ; 182: 106907, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35344748

RESUMEN

IQSEC2 is an X-linked gene localized to the post synaptic density encoding a GTP exchange factor that regulates NMDA mediated changes in synaptic function. Mutations in the IQSEC2 gene are associated with drug resistant epilepsy, intellectual disability and autism. Precision medicine based therapeutics to treat IQSEC2 associated epilepsy requires the development and characterization of mutation specific animal models. To date no EEG recordings have been presented for any mouse model of any IQSEC2 mutation showing seizures. In this study we characterize the seizures and EEG brain wave abnormalities present in mice with a A350V IQSEC2 missense mutation that is associated with drug resistant epilepsy in man. We show that seizures are associated with a greater than 40% mortality rate in male mice and occur exclusively from post-natal day 16-20. EEG recordings of mouse pups during this window demonstrate seizures and the presence of spikes with a marked increase in delta waves. EEG recordings in adult male mice have persistent excessive slow frequency activity and spikes, but seizures were not recorded. RNAseq analysis of the hippocampi of mice prior to the development of seizures demonstrated marked abnormalities in canonical pathways involved in synaptogenesis and dendritic maturation with the most prominently dysregulated gene being that for TRH suggesting a potential target for therapy given the previous demonstration of TRH to decrease seizures in several forms of drug resistant epilepsy.


Asunto(s)
Epilepsia Refractaria , Medicina de Precisión , Animales , Modelos Animales de Enfermedad , Electroencefalografía , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Masculino , Ratones , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Convulsiones/tratamiento farmacológico , Convulsiones/genética
6.
Front Mol Neurosci ; 12: 43, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30842726

RESUMEN

We have recently described an A350V mutation in IQSEC2 associated with intellectual disability, autism and epilepsy. We sought to understand the molecular pathophysiology of this mutation with the goal of developing targets for drug intervention. We demonstrate here that the A350V mutation results in interference with the binding of apocalmodulin to the IQ domain of IQSEC2. We further demonstrate that this mutation results in constitutive activation of the guanine nucleotide exchange factor (GEF) activity of IQSEC2 resulting in increased production of the active form of Arf6. In a CRISPR generated mouse model of the A350V IQSEC2 mutation, we demonstrate that the surface expression of GluA2 AMPA receptors in mouse hippocampal tissue was significantly reduced in A350V IQSEC2 mutant mice compared to wild type IQSEC2 mice and that there is a significant reduction in basal synaptic transmission in the hippocampus of A350V IQSEC2 mice compared to wild type IQSEC2 mice. Finally, the A350V IQSEC2 mice demonstrated increased activity, abnormal social behavior and learning as compared to wild type IQSEC2 mice. These findings suggest a model of how the A350V mutation in IQSEC2 may mediate disease with implications for targets for drug therapy. These studies provide a paradigm for a personalized approach to precision therapy for a disease that heretofore has no therapy.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA