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










Base de datos
Intervalo de año de publicación
4.
Mod Pathol ; 36(8): 100195, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37100228

RESUMEN

Cell morphology is a fundamental feature used to evaluate patient specimens in pathologic analysis. However, traditional cytopathology analysis of patient effusion samples is limited by low tumor cell abundance coupled with the high background of nonmalignant cells, restricting the ability of downstream molecular and functional analyses to identify actionable therapeutic targets. We applied the Deepcell platform that combines microfluidic sorting, brightfield imaging, and real-time deep learning interpretations based on multidimensional morphology to enrich carcinoma cells from malignant effusions without cell staining or labels. Carcinoma cell enrichment was validated with whole genome sequencing and targeted mutation analysis, which showed a higher sensitivity for detection of tumor fractions and critical somatic variant mutations that were initially at low levels or undetectable in presort patient samples. Our study demonstrates the feasibility and added value of supplementing traditional morphology-based cytology with deep learning, multidimensional morphology analysis, and microfluidic sorting.


Asunto(s)
Líquidos Corporales , Carcinoma , Derrame Pleural Maligno , Humanos , Inteligencia Artificial , Derrame Pleural Maligno/diagnóstico , Derrame Pleural Maligno/patología
5.
Cell ; 183(3): 702-716.e14, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-33125890

RESUMEN

The cellular complexity and scale of the early liver have constrained analyses examining its emergence during organogenesis. To circumvent these issues, we analyzed 45,334 single-cell transcriptomes from embryonic day (E)7.5, when endoderm progenitors are specified, to E10.5 liver, when liver parenchymal and non-parenchymal cell lineages emerge. Our data detail divergence of vascular and sinusoidal endothelia, including a distinct transcriptional profile for sinusoidal endothelial specification by E8.75. We characterize two distinct mesothelial cell types as well as early hepatic stellate cells and reveal distinct spatiotemporal distributions for these populations. We capture transcriptional profiles for hepatoblast specification and migration, including the emergence of a hepatomesenchymal cell type and evidence for hepatoblast collective cell migration. Further, we identify cell-cell interactions during the organization of the primitive sinusoid. This study provides a comprehensive atlas of liver lineage establishment from the endoderm and mesoderm through to the organization of the primitive sinusoid at single-cell resolution.


Asunto(s)
Linaje de la Célula/genética , Hígado/citología , Hígado/metabolismo , Análisis de la Célula Individual , Transcriptoma/genética , Animales , Movimiento Celular , Embrión de Mamíferos/citología , Endotelio/citología , Mesodermo/citología , Ratones , Transducción de Señal , Células Madre/citología
6.
Nat Biotechnol ; 38(6): 747-755, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32518403

RESUMEN

Single-cell RNA sequencing (scRNA-seq) is the leading technique for characterizing the transcriptomes of individual cells in a sample. The latest protocols are scalable to thousands of cells and are being used to compile cell atlases of tissues, organs and organisms. However, the protocols differ substantially with respect to their RNA capture efficiency, bias, scale and costs, and their relative advantages for different applications are unclear. In the present study, we generated benchmark datasets to systematically evaluate protocols in terms of their power to comprehensively describe cell types and states. We performed a multicenter study comparing 13 commonly used scRNA-seq and single-nucleus RNA-seq protocols applied to a heterogeneous reference sample resource. Comparative analysis revealed marked differences in protocol performance. The protocols differed in library complexity and their ability to detect cell-type markers, impacting their predictive value and suitability for integration into reference cell atlases. These results provide guidance both for individual researchers and for consortium projects such as the Human Cell Atlas.


Asunto(s)
Análisis de Secuencia de ARN , Análisis de la Célula Individual , Animales , Benchmarking , Línea Celular , Bases de Datos Genéticas , Genómica/métodos , Genómica/normas , Humanos , Ratones , Análisis de Secuencia de ARN/métodos , Análisis de Secuencia de ARN/normas , Análisis de la Célula Individual/métodos , Análisis de la Célula Individual/normas
7.
Nature ; 571(7764): 205-210, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31270459

RESUMEN

The mammalian brain contains neurogenic niches that comprise neural stem cells and other cell types. Neurogenic niches become less functional with age, but how they change during ageing remains unclear. Here we perform single-cell RNA sequencing of young and old neurogenic niches in mice. The analysis of 14,685 single-cell transcriptomes reveals a decrease in activated neural stem cells, changes in endothelial cells and microglia, and an infiltration of T cells in old neurogenic niches. T cells in old brains are clonally expanded and are generally distinct from those in old blood, which suggests that they may experience specific antigens. T cells in old brains also express interferon-γ, and the subset of neural stem cells that has a high interferon response shows decreased proliferation in vivo. We find that T cells can inhibit the proliferation of neural stem cells in co-cultures and in vivo, in part by secreting interferon-γ. Our study reveals an interaction between T cells and neural stem cells in old brains, opening potential avenues through which to counteract age-related decline in brain function.


Asunto(s)
Envejecimiento/fisiología , Encéfalo/citología , Movimiento Celular , Células-Madre Neurales/citología , Neurogénesis , Análisis de la Célula Individual , Nicho de Células Madre/fisiología , Linfocitos T/citología , Animales , Sangre , Proliferación Celular , Células Clonales/citología , Técnicas de Cocultivo , Células Endoteliales/citología , Interferón gamma/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Análisis de Secuencia de ARN , Transducción de Señal , Linfocitos T/metabolismo , Transcriptoma/genética
8.
Nature ; 569(7756): 361-367, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30959515

RESUMEN

Here we delineate the ontogeny of the mammalian endoderm by generating 112,217 single-cell transcriptomes, which represent all endoderm populations within the mouse embryo until midgestation. We use graph-based approaches to model differentiating cells, which provides a spatio-temporal characterization of developmental trajectories and defines the transcriptional architecture that accompanies the emergence of the first (primitive or extra-embryonic) endodermal population and its sister pluripotent (embryonic) epiblast lineage. We uncover a relationship between descendants of these two lineages, in which epiblast cells differentiate into endoderm at two distinct time points-before and during gastrulation. Trajectories of endoderm cells were mapped as they acquired embryonic versus extra-embryonic fates and as they spatially converged within the nascent gut endoderm, which revealed these cells to be globally similar but retain aspects of their lineage history. We observed the regionalized identity of cells along the anterior-posterior axis of the emergent gut tube, which reflects their embryonic or extra-embryonic origin, and the coordinated patterning of these cells into organ-specific territories.


Asunto(s)
Endodermo/citología , Endodermo/embriología , Intestinos/citología , Intestinos/embriología , Análisis de la Célula Individual , Animales , Blastocisto/citología , Tipificación del Cuerpo , Diferenciación Celular , Linaje de la Célula , Femenino , Gastrulación , Masculino , Ratones
9.
Cell Stem Cell ; 21(1): 78-90.e6, 2017 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-28686870

RESUMEN

Several cell populations have been reported to possess intestinal stem cell (ISC) activity during homeostasis and injury-induced regeneration. Here, we explored inter-relationships between putative mouse ISC populations by comparative RNA-sequencing (RNA-seq). The transcriptomes of multiple cycling ISC populations closely resembled Lgr5+ ISCs, the most well-defined ISC pool, but Bmi1-GFP+ cells were distinct and enriched for enteroendocrine (EE) markers, including Prox1. Prox1-GFP+ cells exhibited sustained clonogenic growth in vitro, and lineage-tracing of Prox1+ cells revealed long-lived clones during homeostasis and after radiation-induced injury in vivo. Single-cell mRNA-seq revealed two subsets of Prox1-GFP+ cells, one of which resembled mature EE cells while the other displayed low-level EE gene expression but co-expressed tuft cell markers, Lgr5 and Ascl2, reminiscent of label-retaining secretory progenitors. Our data suggest that the EE lineage, including mature EE cells, comprises a reservoir of homeostatic and injury-inducible ISCs, extending our understanding of cellular plasticity and stemness.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Células Enteroendocrinas/metabolismo , Mucosa Intestinal/lesiones , Mucosa Intestinal/metabolismo , Yeyuno/lesiones , Yeyuno/metabolismo , Células Madre/metabolismo , Animales , Antígenos de Diferenciación/genética , Células Enteroendocrinas/patología , Regulación de la Expresión Génica , Mucosa Intestinal/patología , Yeyuno/patología , Ratones , Ratones Transgénicos , Células Madre/patología
11.
Cell Syst ; 4(4): 458-469.e5, 2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-28396000

RESUMEN

Signaling proteins display remarkable cell-to-cell heterogeneity in their dynamic responses to stimuli, but the consequences of this heterogeneity remain largely unknown. For instance, the contribution of the dynamics of the innate immune transcription factor nuclear factor κB (NF-κB) to gene expression output is disputed. Here we explore these questions by integrating live-cell imaging approaches with single-cell sequencing technologies. We used this approach to measure both the dynamics of lipopolysaccharide-induced NF-κB activation and the global transcriptional response in the same individual cell. Our results identify multiple, distinct cytokine expression patterns that are correlated with NF-κB activation dynamics, establishing a functional role for NF-κB dynamics in determining cellular phenotypes. Applications of this approach to other model systems and single-cell sequencing technologies have significant potential for discovery, as it is now possible to trace cellular behavior from the initial stimulus, through the signaling pathways, down to genome-wide changes in gene expression, all inside of a single cell.


Asunto(s)
Modelos Inmunológicos , FN-kappa B/fisiología , Animales , Citocinas/genética , Citocinas/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Inmunidad Innata/genética , Lipopolisacáridos/inmunología , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Células RAW 264.7 , Análisis de Secuencia de ARN , Transducción de Señal , Análisis de la Célula Individual , Activación Transcripcional , Transcriptoma
12.
Cell Rep ; 18(3): 777-790, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-28099854

RESUMEN

Neural stem cells (NSCs) in the adult mammalian brain serve as a reservoir for the generation of new neurons, oligodendrocytes, and astrocytes. Here, we use single-cell RNA sequencing to characterize adult NSC populations and examine the molecular identities and heterogeneity of in vivo NSC populations. We find that cells in the NSC lineage exist on a continuum through the processes of activation and differentiation. Interestingly, rare intermediate states with distinct molecular profiles can be identified and experimentally validated, and our analysis identifies putative surface markers and key intracellular regulators for these subpopulations of NSCs. Finally, using the power of single-cell profiling, we conduct a meta-analysis to compare in vivo NSCs and in vitro cultures, distinct fluorescence-activated cell sorting strategies, and different neurogenic niches. These data provide a resource for the field and contribute to an integrative understanding of the adult NSC lineage.


Asunto(s)
Células-Madre Neurales/metabolismo , Transcriptoma , Algoritmos , Animales , Astrocitos/citología , Astrocitos/metabolismo , Diferenciación Celular , Linaje de la Célula , Conexina 43/genética , Conexina 43/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Antígeno Ki-67/metabolismo , Ratones , Células-Madre Neurales/citología , Neurogénesis , Análisis de Componente Principal , ARN/química , ARN/genética , ARN/metabolismo , Análisis de Secuencia de ARN , Análisis de la Célula Individual , ATPasa Intercambiadora de Sodio-Potasio/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
Nat Biotechnol ; 34(7): 752-9, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27240197

RESUMEN

A promising therapeutic strategy for diverse genetic disorders involves transplantation of autologous stem cells that have been genetically corrected ex vivo. A major challenge in such approaches is a loss of stem cell potency during culture. Here we describe an artificial niche for maintaining muscle stem cells (MuSCs) in vitro in a potent, quiescent state. Using a machine learning method, we identified a molecular signature of quiescence and used it to screen for factors that could maintain mouse MuSC quiescence, thus defining a quiescence medium (QM). We also engineered muscle fibers that mimic the native myofiber of the MuSC niche. Mouse MuSCs maintained in QM on engineered fibers showed enhanced potential for engraftment, tissue regeneration and self-renewal after transplantation in mice. An artificial niche adapted to human cells similarly extended the quiescence of human MuSCs in vitro and enhanced their potency in vivo. Our approach for maintaining quiescence may be applicable to stem cells isolated from other tissues.


Asunto(s)
Técnicas de Cultivo Celular por Lotes/métodos , Proteínas Musculares/metabolismo , Mioblastos Esqueléticos/fisiología , Mioblastos Esqueléticos/trasplante , Nicho de Células Madre/fisiología , Conservación de Tejido/métodos , Animales , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Mioblastos Esqueléticos/citología , Trasplante de Células Madre/métodos , Resultado del Tratamiento
14.
Nucleic Acids Res ; 42(22): e172, 2014 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-25352556

RESUMEN

Rapid development of next generation sequencing technology has enabled the identification of genomic alterations from short sequencing reads. There are a number of software pipelines available for calling single nucleotide variants from genomic DNA but, no comprehensive pipelines to identify, annotate and prioritize expressed SNVs (eSNVs) from non-directional paired-end RNA-Seq data. We have developed the eSNV-Detect, a novel computational system, which utilizes data from multiple aligners to call, even at low read depths, and rank variants from RNA-Seq. Multi-platform comparisons with the eSNV-Detect variant candidates were performed. The method was first applied to RNA-Seq from a lymphoblastoid cell-line, achieving 99.7% precision and 91.0% sensitivity in the expressed SNPs for the matching HumanOmni2.5 BeadChip data. Comparison of RNA-Seq eSNV candidates from 25 ER+ breast tumors from The Cancer Genome Atlas (TCGA) project with whole exome coding data showed 90.6-96.8% precision and 91.6-95.7% sensitivity. Contrasting single-cell mRNA-Seq variants with matching traditional multicellular RNA-Seq data for the MD-MB231 breast cancer cell-line delineated variant heterogeneity among the single-cells. Further, Sanger sequencing validation was performed for an ER+ breast tumor with paired normal adjacent tissue validating 29 out of 31 candidate eSNVs. The source code and user manuals of the eSNV-Detect pipeline for Sun Grid Engine and virtual machine are available at http://bioinformaticstools.mayo.edu/research/esnv-detect/.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Variación Genética , Análisis de Secuencia de ARN/métodos , Neoplasias de la Mama/genética , Línea Celular , Línea Celular Tumoral , Exoma , Femenino , Humanos , Mutación , Polimorfismo de Nucleótido Simple , Alineación de Secuencia , Análisis de la Célula Individual , Programas Informáticos
15.
Cell Stem Cell ; 10(3): 327-36, 2012 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-22385659

RESUMEN

Pax3, a key myogenic regulator, is transiently expressed during activation of adult muscle stem cells, or satellite cells (SCs), and is also expressed in a subset of quiescent SCs (QSCs), but only in specific muscles. The mechanisms regulating these variations in expression are not well understood. Here we show that Pax3 levels are regulated by miR-206, a miRNA with a previously demonstrated role in myogenic differentiation. In most QSCs and activated SCs, miR-206 expression suppresses Pax3 expression. Paradoxically, QSCs that express high levels of Pax3 also express high levels of miR-206. In these QSCs, Pax3 transcripts are subject to alternative polyadenylation, resulting in transcripts with shorter 3' untranslated regions (3'UTRs) that render them resistant to regulation by miR-206. Similar alternate polyadenylation of the Pax3 transcript also occurs in myogenic progenitors during development. Our findings may reflect a general role of alternative polyadenylation in circumventing miRNA-mediated regulation of stem cell function.


Asunto(s)
MicroARNs/metabolismo , Mioblastos/metabolismo , Células Madre/metabolismo , Animales , Línea Celular , Células Cultivadas , Clonación Molecular , Embrión de Mamíferos , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Modelos Biológicos , Factores de Transcripción Paired Box/genética , Poliadenilación , Reacción en Cadena de la Polimerasa , Células Madre/citología
16.
Mol Cell ; 40(5): 749-61, 2010 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-21145483

RESUMEN

Pax3 plays critical roles during developmental and postnatal myogenesis. We have previously shown that levels of Pax3 protein are regulated by monoubiquitination and proteasomal degradation during postnatal myogenesis, but none of the key regulators of the monoubiquitination process were known. Here we show that Pax3 monoubiquitination is mediated by the ubiquitin-activating/conjugating activity of Taf1, a component of the core transcriptional machinery that was recently reported to be downregulated during myogenic differentiation. We show that Taf1 binds directly to Pax3 and overexpression of Taf1 increases the level of monoubiquitinated Pax3 and its degradation by the proteasome. A decrease of Taf1 results in a decrease in Pax3 monoubiquitination, an increase in the levels of Pax3 protein, and a concomitant increase in Pax3-mediated inhibition of myogenic differentiation and myoblast migration. These results suggest that Taf1 regulates Pax3 protein levels through its ability to mediate monoubiquitination, revealing a critical interaction between two proteins that are involved in distinct aspects of myogenic differentiation. Finally, these results suggest that the components of the core transcriptional are integrally involved in the process of myogenic differentiation, acting as nodal regulators of the differentiation program.


Asunto(s)
Factores de Transcripción Paired Box/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Factor de Transcripción TFIID/metabolismo , Ubiquitina/metabolismo , Animales , Células Cultivadas , Histona Acetiltransferasas , Ratones , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/genética , Factores Asociados con la Proteína de Unión a TATA/genética , Factor de Transcripción TFIID/genética , Ubiquitinación
18.
Cell ; 130(2): 349-62, 2007 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-17662948

RESUMEN

Pax3 and Pax7 play distinct but overlapping roles in developmental and postnatal myogenesis. The mechanisms involved in the differential regulation of these highly homologous proteins are unknown. We present evidence that Pax3, but not Pax7, is regulated by ubiquitination and proteasomal degradation during adult muscle stem cell activation. Intriguingly, only monoubiquitinated forms of Pax3 could be detected. Mutation of two specific lysine residues in the C-terminal region of Pax3 reduced the extent of its monoubiquitination and susceptibility to proteasomal degradation, whereas introduction of a key lysine into the C-terminal region of Pax7 rendered that protein susceptible to monoubiquitination and proteasomal degradation. Monoubiquitinated Pax3 was shuttled to the intrinsic proteasomal protein S5a by interacting specifically with the ubiquitin-binding protein Rad23B. Functionally, sustained expression of Pax3 proteins inhibited myogenic differentiation, demonstrating that Pax3 degradation is an essential step for the progression of the myogenic program. These results reveal an important mechanism of Pax3 regulation in muscle progenitors and an unrecognized role of protein monoubiquitination in mediating proteasomal degradation.


Asunto(s)
Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/metabolismo , Factores de Transcripción Paired Box/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional , Ubiquitina/metabolismo , Secuencia de Aminoácidos , Animales , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Lisina/metabolismo , Ratones , Datos de Secuencia Molecular , Desarrollo de Músculos , Mioblastos Esqueléticos/citología , Factor de Transcripción PAX3 , Factor de Transcripción PAX7/metabolismo , Factores de Transcripción Paired Box/química , Termodinámica
19.
J Cell Sci ; 117(Pt 19): 4469-79, 2004 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-15316066

RESUMEN

The regulation of the cytoskeleton is critical to normal cell function during tissue morphogenesis. Cell-matrix interactions mediated by integrins regulate cytoskeletal dynamics, but the signaling cascades that control these processes remain largely unknown. Here we show that myristoylated alanine-rich C-kinase substrate (MARCKS) a specific substrate of protein kinase C (PKC), is regulated by alpha5beta1 integrin-mediated activation of PKC and is critical to the regulation of actin stress fiber formation during muscle cell spreading. Using MARCKS mutants that are defective in membrane association or responsiveness to PKC-dependent phosphorylation, we demonstrate that the translocation of MARCKS from the membrane to the cytosol in a PKC-dependent manner permits the initial phases of cell adhesion. The dephosphorylation of MARCKS and its translocation back to the membrane permits the later stages of cell spreading during the polymerization and cross-linking of actin and the maturation of the cytoskeleton. All of these processes are directly dependent on the binding of alpha5beta1 integrin to its extracellular matrix receptor, fibronectin. These results demonstrate a direct biochemical pathway linking alpha5beta1 integrin signaling to cytoskeletal dynamics and involving bi-directional translocation of MARCKS during the dramatic changes in cellular morphology that occur during cell migration and tissue morphogenesis.


Asunto(s)
Membrana Celular/metabolismo , Forma de la Célula/fisiología , Integrina alfa5beta1/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteína Quinasa C/metabolismo , Actinas/metabolismo , Animales , Adhesión Celular/fisiología , Células Cultivadas , Citoesqueleto/metabolismo , Citosol/metabolismo , Fibronectinas/metabolismo , Humanos , Mioblastos/citología , Mioblastos/metabolismo , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada , Fosforilación , Transporte de Proteínas/fisiología , Transducción de Señal , Fibras de Estrés/metabolismo
20.
J Cell Sci ; 115(Pt 10): 2151-63, 2002 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-11973356

RESUMEN

To understand how muscle cell spreading and survival are mediated by integrins, we studied the signaling events initiated by the attachment of muscle cells to fibronectin (FN). We have previously demonstrated that muscle cell spreading on FN is mediated by alpha5beta1 integrin, is associated with rapid phosphorylation of focal adhesion kinase and is dependent on activation of protein kinase C (PKC). Here we investigated the role of individual PKC isozymes in these cellular processes. We show that alpha, delta and epsilonPKC are expressed in muscle cells and are activated upon integrin engagement with different kinetics - epsilonPKC was activated early, whereas alpha and deltaPKC were activated later. Using isozyme-specific inhibitors, we found that the activation of epsilonPKC was necessary for cell attachment to FN. However, using isozyme-specific activators, we found that activation of each of three isozymes was sufficient to promote the spreading of alpha5-integrin-deficient cells on FN. To investigate further the mechanism by which integrin signaling and PKC activation mediate cell spreading, we studied the effects of these processes on MARCKS, a substrate of PKC and a protein known to regulate actin dynamics. We found that MARCKS was localized to focal adhesion sites soon after cell adhesion and that MARCKS translocated from the membrane to the cytosol during the process of cell spreading. This translocation correlated with different phases of PKC activation and with reorganization of the actin cytoskeleton. Using MARCKS-antisense cDNA, we show that alpha5-expressing cells in which MARCKS expression is inhibited fail to spread on FN, providing evidence for the crucial role of MARCKS in muscle cell spreading. Together, the data suggest a model in which early activation of epsilonPKC is necessary for cell attachment; the later activation of alpha or deltaPKC may be necessary for the progression from attachment to spreading. The mechanism of PKC-mediated cell spreading may be via the phosphorylation of signaling proteins, such as MARCKS, that are involved in the reorganization of the actin cytoskeleton.


Asunto(s)
Integrinas/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana , Músculo Esquelético/citología , Músculo Esquelético/enzimología , Fosfoproteínas/metabolismo , Proteína Quinasa C/metabolismo , Transducción de Señal , Animales , Proteínas de Unión al Calcio , Adhesión Celular , Línea Celular , Movimiento Celular , Tamaño de la Célula , Activación Enzimática , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Glucosidasas , Humanos , Isoenzimas/metabolismo , Ratones , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA