Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cell ; 187(14): 3652-3670.e40, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38843833

RESUMEN

While ultraviolet (UV) radiation damages DNA, eliciting the DNA damage response (DDR), it also damages RNA, triggering transcriptome-wide ribosomal collisions and eliciting a ribotoxic stress response (RSR). However, the relative contributions, timing, and regulation of these pathways in determining cell fate is unclear. Here we use time-resolved phosphoproteomic, chemical-genetic, single-cell imaging, and biochemical approaches to create a chronological atlas of signaling events activated in cells responding to UV damage. We discover that UV-induced apoptosis is mediated by the RSR kinase ZAK and not through the DDR. We identify two negative-feedback modules that regulate ZAK-mediated apoptosis: (1) GCN2 activation limits ribosomal collisions and attenuates ZAK-mediated RSR and (2) ZAK activity leads to phosphodegron autophosphorylation and its subsequent degradation. These events tune ZAK's activity to collision levels to establish regimes of homeostasis, tolerance, and death, revealing its key role as the cellular sentinel for nucleic acid damage.


Asunto(s)
Apoptosis , Daño del ADN , Rayos Ultravioleta , Rayos Ultravioleta/efectos adversos , Apoptosis/efectos de la radiación , Fosforilación/efectos de la radiación , Humanos , Transducción de Señal/efectos de la radiación , Proteínas Serina-Treonina Quinasas/metabolismo , Estrés Fisiológico/efectos de la radiación , Ribosomas/metabolismo , Muerte Celular/efectos de la radiación
2.
Science ; 384(6695): eadi2421, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38696576

RESUMEN

Cell cycle events are coordinated by cyclin-dependent kinases (CDKs) to ensure robust cell division. CDK4/6 and CDK2 regulate the growth 1 (G1) to synthesis (S) phase transition of the cell cycle by responding to mitogen signaling, promoting E2F transcription and inhibition of the anaphase-promoting complex. We found that this mechanism was still required in G2-arrested cells to prevent cell cycle exit after the S phase. This mechanism revealed a role for CDK4/6 in maintaining the G2 state, challenging the notion that the cell cycle is irreversible and that cells do not require mitogens after passing the restriction point. Exit from G2 occurred during ribotoxic stress and was actively mediated by stress-activated protein kinases. Upon relief of stress, a significant fraction of cells underwent a second round of DNA replication that led to whole-genome doubling.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina , Quinasa 6 Dependiente de la Ciclina , Endorreduplicación , Puntos de Control de la Fase G2 del Ciclo Celular , Estrés Fisiológico , Humanos , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/genética , Factores de Transcripción E2F/metabolismo , Factores de Transcripción E2F/genética , Fase S , Línea Celular
3.
Dev Cell ; 59(13): 1724-1736.e4, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38640927

RESUMEN

Whole-genome duplication (WGD) is a frequent event in cancer evolution that fuels chromosomal instability. WGD can result from mitotic errors or endoreduplication, yet the molecular mechanisms that drive WGD remain unclear. Here, we use live single-cell analysis to characterize cell-cycle dynamics upon aberrant Ras-ERK signaling. We find that sustained ERK signaling in human cells leads to reactivation of the APC/C in G2, resulting in tetraploid G0-like cells that are primed for WGD. This process is independent of DNA damage or p53 but dependent on p21. Transcriptomics analysis and live-cell imaging showed that constitutive ERK activity promotes p21 expression, which is necessary and sufficient to inhibit CDK activity and which prematurely activates the anaphase-promoting complex (APC/C). Finally, either loss of p53 or reduced ERK signaling allowed for endoreduplication, completing a WGD event. Thus, sustained ERK signaling-induced G2 cell cycle exit represents an alternative path to WGD.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Proteína p53 Supresora de Tumor , Humanos , Sistema de Señalización de MAP Quinasas/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Ciclosoma-Complejo Promotor de la Anafase/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Genoma Humano , Duplicación de Gen , Fase G2/genética , Daño del ADN/genética
4.
Nat Cell Biol ; 26(6): 859-867, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38689013

RESUMEN

Tissue regeneration and maintenance rely on coordinated stem cell behaviours. This orchestration can be impaired by oncogenic mutations leading to cancer. However, it is largely unclear how oncogenes perturb stem cells' orchestration to disrupt tissue. Here we used intravital imaging to investigate the mechanisms by which oncogenic Kras mutation causes tissue disruption in the hair follicle. Through longitudinally tracking hair follicles in live mice, we found that KrasG12D, a mutation that can lead to squamous cell carcinoma, induces epithelial tissue deformation in a spatiotemporally specific manner, linked with abnormal cell division and migration. Using a reporter mouse capture real-time ERK signal dynamics at the single-cell level, we discovered that KrasG12D, but not a closely related mutation HrasG12V, converts ERK signal in stem cells from pulsatile to sustained. Finally, we demonstrated that interrupting sustained ERK signal reverts KrasG12D-induced tissue deformation through modulating specific features of cell migration and division.


Asunto(s)
Movimiento Celular , Folículo Piloso , Mutación , Proteínas Proto-Oncogénicas p21(ras) , Animales , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Ratones , Folículo Piloso/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Ratones Transgénicos , Células Madre/metabolismo , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Humanos , Femenino , Activación Enzimática
5.
Cell Stem Cell ; 30(7): 962-972.e6, 2023 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-37419106

RESUMEN

The ability of stem cells to build and replenish tissues depends on support from their niche. Although niche architecture varies across organs, its functional importance is unclear. During hair follicle growth, multipotent epithelial progenitors build hair via crosstalk with their remodeling fibroblast niche, the dermal papilla, providing a powerful model to functionally interrogate niche architecture. Through mouse intravital imaging, we show that dermal papilla fibroblasts remodel individually and collectively to form a morphologically polarized, structurally robust niche. Asymmetric TGF-ß signaling precedes morphological niche polarity, and loss of TGF-ß signaling in dermal papilla fibroblasts leads them to progressively lose their stereotypic architecture, instead surrounding the epithelium. The reorganized niche induces the redistribution of multipotent progenitors but nevertheless supports their proliferation and differentiation. However, the differentiated lineages and hairs produced by progenitors are shorter. Overall, our results reveal that niche architecture optimizes organ efficiency but is not absolutely essential for organ function.


Asunto(s)
Folículo Piloso , Cabello , Ratones , Animales , Diferenciación Celular , Epitelio , Factor de Crecimiento Transformador beta
6.
Cell Syst ; 13(11): 885-894.e4, 2022 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-36356576

RESUMEN

The classic network of mitogen-activated protein kinases (MAPKs) is highly interconnected and controls a diverse array of biological processes. In multicellular eukaryotes, the MAPKs ERK, JNK, and p38 control opposing cell behaviors but are often activated simultaneously, raising questions about how input-output specificity is achieved. Here, we use multiplexed MAPK activity biosensors to investigate how cell fate control emerges from the connectivity and dynamics of the MAPK network. Through chemical and genetic perturbation, we systematically explore the outputs and functions of all the MAP3 kinases encoded in the human genome and show that MAP3Ks control cell fate by triggering unique combinations of MAPK activity. We show that these MAPK activity combinations explain the paradoxical dual role of JNK signaling as pro-apoptotic or pro-proliferative kinase. Overall, our integrative analysis indicates that the MAPK network operates as a unit to control cell fate and shifts the focus from MAPKs to MAP3Ks to better understand signaling-mediated control of cell fate.


Asunto(s)
Proteínas Quinasas JNK Activadas por Mitógenos , Sistema de Señalización de MAP Quinasas , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Fosforilación , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Transducción de Señal
7.
Nat Commun ; 12(1): 3175, 2021 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-34039988

RESUMEN

Antagonistic pleiotropy is a foundational theory that predicts aging-related diseases are the result of evolved genetic traits conferring advantages early in life. Here we examine CaMKII, a pluripotent signaling molecule that contributes to common aging-related diseases, and find that its activation by reactive oxygen species (ROS) was acquired more than half-a-billion years ago along the vertebrate stem lineage. Functional experiments using genetically engineered mice and flies reveal ancestral vertebrates were poised to benefit from the union of ROS and CaMKII, which conferred physiological advantage by allowing ROS to increase intracellular Ca2+ and activate transcriptional programs important for exercise and immunity. Enhanced sensitivity to the adverse effects of ROS in diseases and aging is thus a trade-off for positive traits that facilitated the early and continued evolutionary success of vertebrates.


Asunto(s)
Envejecimiento/fisiología , Evolución Biológica , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Vertebrados/fisiología , Animales , Animales Modificados Genéticamente , Sistemas CRISPR-Cas/genética , Señalización del Calcio/fisiología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Femenino , Edición Génica , Técnicas de Sustitución del Gen , Masculino , Ratones , Modelos Animales , Oxidación-Reducción , Filogenia , Aptitud Física/fisiología , Mutación Puntual
8.
STAR Protoc ; 2(2): 100446, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-33899025

RESUMEN

Determining how signaling dynamics relate to gene expression and cell fate is essential to understanding multicellular development. We present a unified live imaging and lineage analysis method that allows integrated analysis of both techniques in the same mouse embryos. This protocol describes the embryo isolation, confocal imaging, immunofluorescence, and in silico alignment required to connect time-lapse and endpoint measurements. By utilizing different biosensors and fixed readouts, this method allows interrogation of signaling dynamics that specify cell fates in developing embryos. For complete details on the use and execution of this protocol, please refer to Pokrass et al. (2020).


Asunto(s)
Blastocisto , Microscopía Confocal/métodos , Técnicas de Sonda Molecular , Imagen de Lapso de Tiempo/métodos , Animales , Blastocisto/citología , Blastocisto/metabolismo , Blastocisto/fisiología , Células Cultivadas , Femenino , Imagenología Tridimensional , Masculino , Ratones , Transducción de Señal/fisiología
9.
Nat Commun ; 12(1): 1836, 2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33758175

RESUMEN

To prevent damage to the host or its commensal microbiota, epithelial tissues must match the intensity of the immune response to the severity of a biological threat. Toll-like receptors allow epithelial cells to identify microbe associated molecular patterns. However, the mechanisms that mitigate biological noise in single cells to ensure quantitatively appropriate responses remain unclear. Here we address this question using single cell and single molecule approaches in mammary epithelial cells and primary organoids. We find that epithelial tissues respond to bacterial microbe associated molecular patterns by activating a subset of cells in an all-or-nothing (i.e. digital) manner. The maximum fraction of responsive cells is regulated by a bimodal epigenetic switch that licenses the TLR2 promoter for transcription across multiple generations. This mechanism confers a flexible memory of inflammatory events as well as unique spatio-temporal control of epithelial tissue-level immune responses. We propose that epigenetic licensing in individual cells allows for long-term, quantitative fine-tuning of population-level responses.


Asunto(s)
Bacterias/inmunología , Células Epiteliales/inmunología , Inmunidad Innata , Lipopéptidos/inmunología , FN-kappa B/metabolismo , Receptor Toll-Like 2/metabolismo , Animales , Bacterias/metabolismo , Línea Celular , Citocinas/metabolismo , Citocinas/farmacología , Metilación de ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Flagelina/farmacología , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/genética , Hibridación Fluorescente in Situ , Glándulas Mamarias Animales , Ratones , Organoides/efectos de los fármacos , Organoides/inmunología , Organoides/metabolismo , Regiones Promotoras Genéticas , RNA-Seq , Transducción de Señal/inmunología , Análisis de la Célula Individual , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 2/genética , Receptores Toll-Like/agonistas , Receptores Toll-Like/metabolismo
10.
Dev Cell ; 55(3): 328-340.e5, 2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-33091369

RESUMEN

Despite the noisy nature of single cells, multicellular organisms robustly generate different cell types from one zygote. This process involves dynamic cross regulation between signaling and gene expression that is difficult to capture with fixed-cell approaches. To study signaling dynamics and fate specification during preimplantation development, we generated a transgenic mouse expressing the ERK kinase translocation reporter and measured ERK activity in single cells of live embryos. Our results show primarily active ERK in both the inner cell mass and trophectoderm cells due to fibroblast growth factor (FGF) signaling. Strikingly, a subset of mitotic events results in a short pulse of ERK inactivity in both daughter cells that correlates with elevated endpoint NANOG levels. Moreover, endogenous tagging of Nanog in embryonic stem cells reveals that ERK inhibition promotes enhanced stabilization of NANOG protein after mitosis. Our data show that cell cycle, signaling, and differentiation are coordinated during preimplantation development.


Asunto(s)
Blastocisto/citología , Blastocisto/enzimología , Ciclo Celular , Linaje de la Célula , Sistema de Señalización de MAP Quinasas , Mamíferos/embriología , Animales , Estratos Germinativos/citología , Humanos , Ratones , Mitosis , Modelos Biológicos , Proteína Homeótica Nanog/metabolismo , Estabilidad Proteica , Reproducibilidad de los Resultados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA