Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
PLoS Biol ; 20(12): e3001900, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36469503

RESUMEN

How progenitor cells can attain a distinct differentiated cell identity is a challenging problem given the fluctuating signaling environment in which cells exist and that critical transcription factors are often not unique to a differentiation process. Here, we test the hypothesis that a unique differentiated cell identity can result from a core component of the differentiated state doubling up as a signaling protein that also drives differentiation. Using live single-cell imaging in the adipocyte differentiation system, we show that progenitor fat cells (preadipocytes) can only commit to terminally differentiate after up-regulating FABP4, a lipid buffer that is highly enriched in mature adipocytes. Upon induction of adipogenesis in mouse preadipocyte cells, we show that after a long delay, cells first abruptly start to engage a positive feedback between CEBPA and PPARG before then engaging, after a second delay, a positive feedback between FABP4 and PPARG. These sequential positive feedbacks both need to engage in order to drive PPARG levels past the threshold for irreversible differentiation. In the last step before commitment, PPARG transcriptionally increases FABP4 expression while fatty acid-loaded FABP4 increases PPARG activity. Together, our study suggests a control principle for robust cell identity whereby a core component of the differentiated state also promotes differentiation from its own progenitor state.


Asunto(s)
Adipogénesis , PPAR gamma , Ratones , Animales , PPAR gamma/genética , PPAR gamma/metabolismo , Diferenciación Celular/fisiología , Adipocitos/metabolismo , Factores de Transcripción/metabolismo
2.
Nucleic Acids Res ; 48(13): 7279-7297, 2020 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-32463448

RESUMEN

In order to maintain cellular protein homeostasis, ribosomes are safeguarded against dysregulation by myriad processes. Remarkably, many cell types can withstand genetic lesions of certain ribosomal protein genes, some of which are linked to diverse cellular phenotypes and human disease. Yet the direct and indirect consequences from these lesions are poorly understood. To address this knowledge gap, we studied in vitro and cellular consequences that follow genetic knockout of the ribosomal proteins RPS25 or RACK1 in a human cell line, as both proteins are implicated in direct translational control. Prompted by the unexpected detection of an off-target ribosome alteration in the RPS25 knockout, we closely interrogated cellular phenotypes. We found that multiple RPS25 knockout clones display viral- and toxin-resistance phenotypes that cannot be rescued by functional cDNA expression, suggesting that RPS25 loss elicits a cell state transition. We characterized this state and found that it underlies pleiotropic phenotypes and has a common rewiring of gene expression. Rescuing RPS25 expression by genomic locus repair failed to correct for the phenotypic and expression hysteresis. Our findings illustrate how the elasticity of cells to a ribosome perturbation can drive specific phenotypic outcomes that are indirectly linked to translation and suggests caution in the interpretation of ribosomal protein gene mutation data.


Asunto(s)
Mutación con Pérdida de Función , Fenotipo , Proteínas Ribosómicas/genética , Línea Celular Tumoral , Células HEK293 , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteostasis , Receptores de Cinasa C Activada/genética , Receptores de Cinasa C Activada/metabolismo , Proteínas Ribosómicas/metabolismo , Ribosomas/genética , Ribosomas/metabolismo
3.
Mol Syst Biol ; 14(5): e7997, 2018 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-29759982

RESUMEN

Due to noise in the synthesis and degradation of proteins, the concentrations of individual vertebrate signaling proteins were estimated to vary with a coefficient of variation (CV) of approximately 25% between cells. Such high variation is beneficial for population-level regulation of cell functions but abolishes accurate single-cell signal transmission. Here, we measure cell-to-cell variability of relative protein abundance using quantitative proteomics of individual Xenopus laevis eggs and cultured human cells and show that variation is typically much lower, in the range of 5-15%, compatible with accurate single-cell transmission. Focusing on bimodal ERK signaling, we show that variation and covariation in MEK and ERK expression improves controllability of the percentage of activated cells, demonstrating how variation and covariation in expression enables population-level control of binary cell-fate decisions. Together, our study argues for a control principle whereby low expression variation enables accurate control of analog single-cell signaling, while increased variation, covariation, and numbers of pathway components are required to widen the stimulus range over which external inputs regulate binary cell activation to enable precise control of the fraction of activated cells in a population.


Asunto(s)
Regulación de la Expresión Génica , Variación Genética , Transducción de Señal , Animales , Diferenciación Celular , Células Cultivadas , Simulación por Computador , Estudios de Evaluación como Asunto , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Modelos Moleculares , Óvulo , Proteómica , Xenopus laevis
4.
Cell Syst ; 14(12): 1074-1086.e7, 2023 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-37995680

RESUMEN

Hypoxia-induced upregulation of HIF1α triggers adipose tissue dysfunction and insulin resistance in obese patients. HIF1α closely interacts with PPARγ, the master regulator of adipocyte differentiation and lipid accumulation, but there are conflicting results regarding how this interaction controls the excessive lipid accumulation that drives adipocyte dysfunction. To directly address these conflicts, we established a differentiation system that recapitulated prior seemingly opposing observations made across different experimental settings. Using single-cell imaging and coarse-grained mathematical modeling, we show how HIF1α can both promote and repress lipid accumulation during adipogenesis. Our model predicted and our experiments confirmed that the opposing roles of HIF1α are isolated from each other by the positive-feedback-mediated upregulation of PPARγ that drives adipocyte differentiation. Finally, we identify three factors: strength of the differentiation cue, timing of hypoxic perturbation, and strength of HIF1α expression changes that, when considered together, provide an explanation for many of the previous conflicting reports.


Asunto(s)
Adipocitos , PPAR gamma , Humanos , PPAR gamma/metabolismo , Retroalimentación , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Lípidos
5.
Cell Rep ; 31(11): 107769, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32553172

RESUMEN

Terminal differentiation is essential for the development and maintenance of tissues in all multi-cellular organisms and is associated with permanent exit from the cell cycle. Failure to permanently exit the cell cycle can result in cancer and disease. However, the molecular mechanisms and timing that coordinate differentiation commitment and cell cycle exit are not yet understood. Using live, single-cell imaging of cell cycle progression and differentiation commitment during adipogenesis, we show that a rapid switch mechanism engages exclusively in G1 to trigger differentiation commitment simultaneously with permanent exit from the cell cycle. We identify a molecular competition in G1 between when the differentiation switch is triggered and when the proliferative window closes that allows mitogen and differentiation stimuli to control the balance between terminally differentiating cells produced and progenitor cells kept in reserve, a parameter of critical importance for enabling proper development of tissue domains and organs.


Asunto(s)
Ciclo Celular/fisiología , Diferenciación Celular/fisiología , División Celular/fisiología , Células Madre/citología , Adipogénesis/fisiología , Animales , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos
6.
Cell Metab ; 27(4): 854-868.e8, 2018 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-29617644

RESUMEN

Glucocorticoid and other adipogenic hormones are secreted in mammals in circadian oscillations. Loss of this circadian oscillation pattern correlates with obesity in humans, raising the intriguing question of how hormone secretion dynamics affect adipocyte differentiation. Using live, single-cell imaging of the key adipogenic transcription factors CEBPB and PPARG, endogenously tagged with fluorescent proteins, we show that pulsatile circadian hormone stimuli are rejected by the adipocyte differentiation control system. In striking contrast, equally strong persistent signals trigger maximal differentiation. We identify the mechanism of how hormone oscillations are filtered as a combination of slow and fast positive feedback centered on PPARG. Furthermore, we confirm in mice that flattening of daily glucocorticoid oscillations significantly increases the mass of subcutaneous and visceral fat pads. Together, our study provides a molecular mechanism for why stress, Cushing's disease, and other conditions for which glucocorticoid secretion loses its pulsatility may lead to obesity.


Asunto(s)
Adipocitos/citología , Adipogénesis/genética , Ritmo Circadiano/genética , Glucocorticoides/metabolismo , PPAR gamma/metabolismo , Células del Estroma/metabolismo , Transcripción Genética , Células 3T3-L1 , Adipocitos/metabolismo , Animales , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de la Célula Individual , Células del Estroma/citología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda