Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 57(9): 2216-2231.e11, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39151426

RESUMEN

Microglia are the resident macrophages of the central nervous system (CNS). Their phagocytic activity is central during brain development and homeostasis-and in a plethora of brain pathologies. However, little is known about the composition, dynamics, and function of human microglial phagosomes under homeostatic and pathological conditions. Here, we developed a method for rapid isolation of pure and intact phagosomes from human pluripotent stem cell-derived microglia under various in vitro conditions, and from human brain biopsies, for unbiased multiomic analysis. Phagosome profiling revealed that microglial phagosomes were equipped to sense minute changes in their environment and were highly dynamic. We detected proteins involved in synapse homeostasis, or implicated in brain pathologies, and identified the phagosome as the site where quinolinic acid was stored and metabolized for de novo nicotinamide adenine dinucleotide (NAD+) generation in the cytoplasm. Our findings highlight the central role of phagosomes in microglial functioning in the healthy and diseased brain.


Asunto(s)
Microglía , Fagocitosis , Fagosomas , Humanos , Microglía/metabolismo , Fagosomas/metabolismo , Encéfalo/metabolismo , Encéfalo/citología , Células Cultivadas , Células Madre Pluripotentes/metabolismo , Proteómica/métodos
2.
Cell ; 154(1): 61-74, 2013 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-23827675

RESUMEN

The recent discovery that normal and neoplastic epithelial cells re-enter the stem cell state raised the intriguing possibility that the aggressiveness of carcinomas derives not from their existing content of cancer stem cells (CSCs) but from their proclivity to generate new CSCs from non-CSC populations. Here, we demonstrate that non-CSCs of human basal breast cancers are plastic cell populations that readily switch from a non-CSC to CSC state. The observed cell plasticity is dependent on ZEB1, a key regulator of the epithelial-mesenchymal transition. We find that plastic non-CSCs maintain the ZEB1 promoter in a bivalent chromatin configuration, enabling them to respond readily to microenvironmental signals, such as TGFß. In response, the ZEB1 promoter converts from a bivalent to active chromatin configuration, ZEB1 transcription increases, and non-CSCs subsequently enter the CSC state. Our findings support a dynamic model in which interconversions between low and high tumorigenic states occur frequently, thereby increasing tumorigenic and malignant potential.


Asunto(s)
Neoplasias de la Mama/patología , Cromatina/metabolismo , Proteínas de Homeodominio/metabolismo , Células Madre Neoplásicas/patología , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , Animales , Neoplasias de la Mama/genética , Células Epiteliales/patología , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/genética , Humanos , Receptores de Hialuranos/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
3.
Cell ; 150(3): 549-62, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22863008

RESUMEN

Heat-Shock Factor 1 (HSF1), master regulator of the heat-shock response, facilitates malignant transformation, cancer cell survival, and proliferation in model systems. The common assumption is that these effects are mediated through regulation of heat-shock protein (HSP) expression. However, the transcriptional network that HSF1 coordinates directly in malignancy and its relationship to the heat-shock response have never been defined. By comparing cells with high and low malignant potential alongside their nontransformed counterparts, we identify an HSF1-regulated transcriptional program specific to highly malignant cells and distinct from heat shock. Cancer-specific genes in this program support oncogenic processes: cell-cycle regulation, signaling, metabolism, adhesion and translation. HSP genes are integral to this program, however, many are uniquely regulated in malignancy. This HSF1 cancer program is active in breast, colon and lung tumors isolated directly from human patients and is strongly associated with metastasis and death. Thus, HSF1 rewires the transcriptome in tumorigenesis, with prognostic and therapeutic implications.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Neoplasias/metabolismo , Factores de Transcripción/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Células Cultivadas , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Genoma Humano , Factores de Transcripción del Choque Térmico , Humanos , Neoplasias/patología , Factores de Transcripción/análisis , Factores de Transcripción/genética
4.
Cell ; 148(5): 1015-28, 2012 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-22385965

RESUMEN

Regulatory networks orchestrated by key transcription factors (TFs) have been proposed to play a central role in the determination of stem cell states. However, the master transcriptional regulators of adult stem cells are poorly understood. We have identified two TFs, Slug and Sox9, that act cooperatively to determine the mammary stem cell (MaSC) state. Inhibition of either Slug or Sox9 blocks MaSC activity in primary mammary epithelial cells. Conversely, transient coexpression of exogenous Slug and Sox9 suffices to convert differentiated luminal cells into MaSCs with long-term mammary gland-reconstituting ability. Slug and Sox9 induce MaSCs by activating distinct autoregulatory gene expression programs. We also show that coexpression of Slug and Sox9 promotes the tumorigenic and metastasis-seeding abilities of human breast cancer cells and is associated with poor patient survival, providing direct evidence that human breast cancer stem cells are controlled by key regulators similar to those operating in normal murine MaSCs.


Asunto(s)
Neoplasias de la Mama/metabolismo , Glándulas Mamarias Humanas/citología , Factor de Transcripción SOX9/metabolismo , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Glándulas Mamarias Humanas/metabolismo , Ratones , Factor de Transcripción SOX9/genética , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética
5.
Cell ; 145(6): 926-40, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21663795

RESUMEN

The epithelial-mesenchymal transition (EMT) has been associated with the acquisition of motility, invasiveness, and self-renewal traits. During both normal development and tumor pathogenesis, this change in cell phenotype is induced by contextual signals that epithelial cells receive from their microenvironment. The signals that are responsible for inducing an EMT and maintaining the resulting cellular state have been unclear. We describe three signaling pathways, involving transforming growth factor (TGF)-ß and canonical and noncanonical Wnt signaling, that collaborate to induce activation of the EMT program and thereafter function in an autocrine fashion to maintain the resulting mesenchymal state. Downregulation of endogenously synthesized inhibitors of autocrine signals in epithelial cells enables the induction of the EMT program. Conversely, disruption of autocrine signaling by added inhibitors of these pathways inhibits migration and self-renewal in primary mammary epithelial cells and reduces tumorigenicity and metastasis by their transformed derivatives.


Asunto(s)
Comunicación Autocrina , Neoplasias de la Mama/metabolismo , Mama/citología , Células Madre Neoplásicas/metabolismo , Comunicación Paracrina , Células Madre/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/patología , Movimiento Celular , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Femenino , Humanos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/efectos de los fármacos , Mesodermo/metabolismo , Ratones , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Wnt/metabolismo
6.
Proc Natl Acad Sci U S A ; 120(16): e2220576120, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-37036962

RESUMEN

Across species, sperm maturation involves the dramatic reconfiguration of chromatin into highly compact nuclei that enhance hydrodynamic ability and ensure paternal genomic integrity. This process is mediated by the replacement of histones by sperm nuclear basic proteins, also referred to as protamines. In humans, a carefully balanced dosage between two known protamine genes is required for optimal fertility. However, it remains unknown how their proper balance is regulated and how defects in balance may lead to compromised fertility. Here, we show that a nucleolar protein, modulo, a homolog of nucleolin, mediates the histone-to-protamine transition during Drosophila spermatogenesis. We find that modulo mutants display nuclear compaction defects during late spermatogenesis due to decreased expression of autosomal protamine genes (including Mst77F) and derepression of Y-linked multicopy Mst77F homologs (Mst77Y), leading to the mutant's known sterility. Overexpression of Mst77Y in a wild-type background is sufficient to cause nuclear compaction defects, similar to modulo mutant, indicating that Mst77Y is a dominant-negative variant interfering with the process of histone-to-protamine transition. Interestingly, ectopic overexpression of Mst77Y caused decompaction of X-bearing spermatids nuclei more frequently than Y-bearing spermatid nuclei, although this did not greatly affect the sex ratio of offspring. We further show that modulo regulates these protamine genes at the step of transcript polyadenylation. We conclude that the regulation of protamines mediated by modulo, ensuring the expression of functional ones while repressing dominant-negative ones, is critical for male fertility.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Humanos , Animales , Masculino , Drosophila melanogaster/metabolismo , Histonas/genética , Histonas/metabolismo , Protaminas/genética , Protaminas/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Semen/metabolismo , Espermatozoides/metabolismo , Cromatina/metabolismo , Espermatogénesis/genética , Drosophila/genética
7.
Cell ; 136(5): 964-77, 2009 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-19269371

RESUMEN

Induced pluripotent stem cells (iPSCs) derived from somatic cells of patients represent a powerful tool for biomedical research and may provide a source for replacement therapies. However, the use of viruses encoding the reprogramming factors represents a major limitation of the current technology since even low vector expression may alter the differentiation potential of the iPSCs or induce malignant transformation. Here, we show that fibroblasts from five patients with idiopathic Parkinson's disease can be efficiently reprogrammed and subsequently differentiated into dopaminergic neurons. Moreover, we derived hiPSCs free of reprogramming factors using Cre-recombinase excisable viruses. Factor-free hiPSCs maintain a pluripotent state and show a global gene expression profile, more closely related to hESCs than to hiPSCs carrying the transgenes. Our results indicate that residual transgene expression in virus-carrying hiPSCs can affect their molecular characteristics and that factor-free hiPSCs therefore represent a more suitable source of cells for modeling of human disease.


Asunto(s)
Enfermedad de Parkinson/metabolismo , Células Madre Pluripotentes/patología , Diferenciación Celular , Reprogramación Celular , Dopamina/metabolismo , Fibroblastos/metabolismo , Humanos , Neuronas/metabolismo
8.
Nature ; 560(7717): E26, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29849139

RESUMEN

In Fig. 4e of this Article, the labels for 'Control' and 'HFD' were reversed ('Control' should have been labelled blue rather than purple, and 'HFD' should have been labelled purple rather than blue). Similarly, in Fig. 4f of this Article, the labels for 'V' and 'GW' were reversed ('V' should have been labelled blue rather than purple, and 'GW' should have been labelled purple instead of blue). The original figure has been corrected online.

9.
Breast Cancer Res ; 25(1): 52, 2023 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-37147680

RESUMEN

BACKGROUND: In light of the growing appreciation for the role of collective cell motility in metastasis, a deeper understanding of the underlying signaling pathways will be critical to translating these observations to the treatment of advanced cancers. Here, we examine the contribution of Wnt/planar cell polarity (Wnt/PCP), one of the non-canonical Wnt signaling pathways and defined by the involvement of the tetraspanin-like proteins Vangl1 and Vangl2, to breast tumor cell motility, collective cell invasiveness and mammary tumor metastasis. METHODS: Vangl1 and Vangl2 knockdown and overexpression and Wnt5a stimulation were employed to manipulate Wnt/PCP signaling in a battery of breast cancer cell lines representing all breast cancer subtypes, and in tumor organoids from MMTV-PyMT mice. Cell migration was assessed by scratch and organoid invasion assays, Vangl protein subcellular localization was assessed by confocal fluorescence microscopy, and RhoA activation was assessed in real time by fluorescence imaging with an advanced FRET biosensor. The impact of Wnt/PCP suppression on mammary tumor growth and metastasis was assessed by determining the effect of conditional Vangl2 knockout on the MMTV-NDL mouse mammary tumor model. RESULTS: We observed that Vangl2 knockdown suppresses the motility of all breast cancer cell lines examined, and overexpression drives the invasiveness of collectively migrating MMTV-PyMT organoids. Vangl2-dependent RhoA activity is localized in real time to a subpopulation of motile leader cells displaying a hyper-protrusive leading edge, Vangl protein is localized to leader cell protrusions within leader cells, and actin cytoskeletal regulator RhoA is preferentially activated in the leader cells of a migrating collective. Mammary gland-specific knockout of Vangl2 results in a striking decrease in lung metastases in MMTV-NDL mice, but does not impact primary tumor growth characteristics. CONCLUSIONS: We conclude that Vangl-dependent Wnt/PCP signaling promotes breast cancer collective cell migration independent of breast tumor subtype and facilitates distant metastasis in a genetically engineered mouse model of breast cancer. Our observations are consistent with a model whereby Vangl proteins localized at the leading edge of leader cells in a migrating collective act through RhoA to mediate the cytoskeletal rearrangements required for pro-migratory protrusion formation.


Asunto(s)
Neoplasias , Vía de Señalización Wnt , Animales , Ratones , Polaridad Celular/fisiología , Neoplasias/patología , Movimiento Celular/genética
10.
Cell ; 134(1): 62-73, 2008 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-18614011

RESUMEN

The p53 tumor suppressor is a key mediator of cellular responses to various stresses. Here, we show that under conditions of basal physiologic and cell-culture stress, p53 inhibits expression of the CD44 cell-surface molecule via binding to a noncanonical p53-binding sequence in the CD44 promoter. This interaction enables an untransformed cell to respond to stress-induced, p53-dependent cytostatic and apoptotic signals that would otherwise be blocked by the actions of CD44. In the absence of p53 function, the resulting derepressed CD44 expression is essential for the growth and tumor-initiating ability of highly tumorigenic mammary epithelial cells. In both tumorigenic and nontumorigenic cells, CD44's expression is positively regulated by p63, a paralogue of p53. Our data indicate that CD44 is a key tumor-promoting agent in transformed tumor cells lacking p53 function. They also suggest that the derepression of CD44 resulting from inactivation of p53 can potentially aid the survival of immortalized, premalignant cells.


Asunto(s)
Receptores de Hialuranos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Células Epiteliales/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Proteína p53 Supresora de Tumor/genética
11.
Nature ; 531(7592): 53-8, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26935695

RESUMEN

Little is known about how pro-obesity diets regulate tissue stem and progenitor cell function. Here we show that high-fat diet (HFD)-induced obesity augments the numbers and function of Lgr5(+) intestinal stem cells of the mammalian intestine. Mechanistically, a HFD induces a robust peroxisome proliferator-activated receptor delta (PPAR-δ) signature in intestinal stem cells and progenitor cells (non-intestinal stem cells), and pharmacological activation of PPAR-δ recapitulates the effects of a HFD on these cells. Like a HFD, ex vivo treatment of intestinal organoid cultures with fatty acid constituents of the HFD enhances the self-renewal potential of these organoid bodies in a PPAR-δ-dependent manner. Notably, HFD- and agonist-activated PPAR-δ signalling endow organoid-initiating capacity to progenitors, and enforced PPAR-δ signalling permits these progenitors to form in vivo tumours after loss of the tumour suppressor Apc. These findings highlight how diet-modulated PPAR-δ activation alters not only the function of intestinal stem and progenitor cells, but also their capacity to initiate tumours.


Asunto(s)
Transformación Celular Neoplásica/efectos de los fármacos , Neoplasias del Colon/patología , Dieta Alta en Grasa/efectos adversos , Intestinos/patología , Células Madre/efectos de los fármacos , Células Madre/patología , Animales , Recuento de Células , Autorrenovación de las Células/efectos de los fármacos , Femenino , Genes APC , Humanos , Masculino , Ratones , Obesidad/inducido químicamente , Obesidad/patología , Organoides/efectos de los fármacos , Organoides/metabolismo , Organoides/patología , PPAR delta/metabolismo , Transducción de Señal/efectos de los fármacos , Nicho de Células Madre/efectos de los fármacos , Células Madre/metabolismo , beta Catenina/metabolismo
12.
Proc Natl Acad Sci U S A ; 116(50): 25293-25303, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31772018

RESUMEN

Microglia are essential for maintenance of normal brain function, with dysregulation contributing to numerous neurological diseases. Protocols have been developed to derive microglia-like cells from human induced pluripotent stem cells (hiPSCs). However, primary microglia display major differences in morphology and gene expression when grown in culture, including down-regulation of signature microglial genes. Thus, in vitro differentiated microglia may not accurately represent resting primary microglia. To address this issue, we transplanted microglial precursors derived in vitro from hiPSCs into neonatal mouse brains and found that the cells acquired characteristic microglial morphology and gene expression signatures that closely resembled primary human microglia. Single-cell RNA-sequencing analysis of transplanted microglia showed similar cellular heterogeneity as primary human cells. Thus, hiPSCs-derived microglia transplanted into the neonatal mouse brain assume a phenotype and gene expression signature resembling that of resting microglia residing in the human brain, making chimeras a superior tool to study microglia in human disease.


Asunto(s)
Encéfalo/fisiología , Células Madre Pluripotentes Inducidas/trasplante , Microglía/trasplante , Animales , Encéfalo/metabolismo , Encéfalo/cirugía , Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Microglía/metabolismo , Fenotipo
13.
Genes Dev ; 26(9): 988-1002, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22549959

RESUMEN

Planarians are capable of regenerating any missing body part and present an attractive system for molecular investigation of regeneration initiation. The gene activation program that occurs at planarian wounds to coordinate regenerative responses remains unknown. We identified a large set of wound-induced genes during regeneration initiation in planarians. Two waves of wound-induced gene expression occurred in differentiated tissues. The first wave includes conserved immediate early genes. Many second-wave genes encode conserved patterning factors required for proper regeneration. Genes of both classes were generally induced by wounding, indicating that a common initial gene expression program is triggered regardless of missing tissue identity. Planarian regeneration uses a population of regenerative cells (neoblasts), including pluripotent stem cells. A class of wound-induced genes was activated directly within neoblasts, including the Runx transcription factor-encoding runt-1 gene. runt-1 was required for specifying different cell types during regeneration, promoting heterogeneity in neoblasts near wounds. Wound-induced gene expression in neoblasts, including that of runt-1, required SRF (serum response factor) and sos-1. Taken together, these data connect wound sensation to the activation of specific cell type regeneration programs in neoblasts. Most planarian wound-induced genes are conserved across metazoans, and identified genes and mechanisms should be important broadly for understanding wound signaling and regeneration initiation.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Regulación de la Expresión Génica , Planarias/genética , Planarias/fisiología , Cicatrización de Heridas/genética , Animales , Ojo/crecimiento & desarrollo , Expresión Génica , Neuronas/fisiología , Biosíntesis de Proteínas/genética , Factor de Respuesta Sérica
14.
BMC Genomics ; 19(1): 623, 2018 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-30134926

RESUMEN

BACKGROUND: Genomic regions repressed for DNA replication, resulting in either delayed replication in S phase or underreplication in polyploid cells, are thought to be controlled by inhibition of replication origin activation. Studies in Drosophila polytene cells, however, raised the possibility that impeding replication fork progression also plays a major role. RESULTS: We exploited genomic regions underreplicated (URs) with tissue specificity in Drosophila polytene cells to analyze mechanisms of replication repression. By localizing the Origin Recognition Complex (ORC) in the genome of the larval fat body and comparing this to ORC binding in the salivary gland, we found that sites of ORC binding show extensive tissue specificity. In contrast, there are common domains nearly devoid of ORC in the salivary gland and fat body that also have reduced density of ORC binding sites in diploid cells. Strikingly, domains lacking ORC can still be replicated in some polytene tissues, showing absence of ORC and origins is insufficient to repress replication. Analysis of the width and location of the URs with respect to ORC position indicates that whether or not a genomic region lacking ORC is replicated is controlled by whether replication forks formed outside the region are inhibited. CONCLUSIONS: These studies demonstrate that inhibition of replication fork progression can block replication across genomic regions that constitutively lack ORC. Replication fork progression can be inhibited in both tissue-specific and genome region-specific ways. Consequently, when evaluating sources of genome instability it is important to consider altered control of replication forks in response to differentiation.


Asunto(s)
Diferenciación Celular/genética , Estructuras Cromosómicas , Replicación del ADN/genética , Organogénesis/genética , Complejo de Reconocimiento del Origen/metabolismo , Origen de Réplica/fisiología , Animales , Sitios de Unión , Estructuras Cromosómicas/química , Estructuras Cromosómicas/genética , Estructuras Cromosómicas/metabolismo , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Embrión no Mamífero , Larva , Especificidad de Órganos/genética
15.
Mol Cell ; 38(5): 675-88, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20542000

RESUMEN

Polycomb proteins maintain cell identity by repressing the expression of developmental regulators specific for other cell types. Polycomb repressive complex-2 (PRC2) catalyzes trimethylation of histone H3 lysine-27 (H3K27me3). Although repressed, PRC2 targets are generally associated with the transcriptional initiation marker H3K4me3, but the significance of this remains unclear. Here, we identify a class of short RNAs, approximately 50-200 nucleotides in length, transcribed from the 5' end of polycomb target genes in primary T cells and embryonic stem cells. Short RNA transcription is associated with RNA polymerase II and H3K4me3, occurs in the absence of mRNA transcription, and is independent of polycomb activity. Short RNAs form stem-loop structures resembling PRC2 binding sites in Xist, interact with PRC2 through SUZ12, cause gene repression in cis, and are depleted from polycomb target genes activated during cell differentiation. We propose that short RNAs play a role in the association of PRC2 with its target genes.


Asunto(s)
ARN/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética , Animales , Secuencia de Bases , Células Cultivadas , Cromatina/genética , Cromatina/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Histonas/genética , Histonas/metabolismo , Lisina/metabolismo , Ratones , Datos de Secuencia Molecular , Neuronas/citología , Neuronas/fisiología , Conformación de Ácido Nucleico , Proteínas del Grupo Polycomb , Regiones Promotoras Genéticas , ARN/química , ARN/genética , Proteínas Represoras/genética , Linfocitos T/citología , Linfocitos T/fisiología
16.
Nucleic Acids Res ; 44(D1): D560-6, 2016 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-26626150

RESUMEN

The oncogenic transformation of normal cells into malignant, rapidly proliferating cells requires major alterations in cell physiology. For example, the transformed cells remodel their metabolic processes to supply the additional demand for cellular building blocks. We have recently demonstrated essential metabolic processes in tumor progression through the development of a methodological analysis of gene expression. Here, we present the Metabolic gEne RApid Visualizer (MERAV, http://merav.wi.mit.edu), a web-based tool that can query a database comprising ∼4300 microarrays, representing human gene expression in normal tissues, cancer cell lines and primary tumors. MERAV has been designed as a powerful tool for whole genome analysis which offers multiple advantages: one can search many genes in parallel; compare gene expression among different tissue types as well as between normal and cancer cells; download raw data; and generate heatmaps; and finally, use its internal statistical tool. Most importantly, MERAV has been designed as a unique tool for analyzing metabolic processes as it includes matrixes specifically focused on metabolic genes and is linked to the Kyoto Encyclopedia of Genes and Genomes pathway search.


Asunto(s)
Bases de Datos Genéticas , Perfilación de la Expresión Génica , Neoplasias/genética , Programas Informáticos , Línea Celular , Línea Celular Tumoral , Perfilación de la Expresión Génica/normas , Humanos , Internet , Redes y Vías Metabólicas/genética , Neoplasias/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos/normas
17.
Proc Natl Acad Sci U S A ; 112(42): E5679-88, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26438848

RESUMEN

Reactive oxygen species (ROS) such as hydrogen peroxide (H2O2) govern cellular homeostasis by inducing signaling. H2O2 modulates the activity of phosphatases and many other signaling molecules through oxidation of critical cysteine residues, which led to the notion that initiation of ROS signaling is broad and nonspecific, and thus fundamentally distinct from other signaling pathways. Here, we report that H2O2 signaling bears hallmarks of a regular signal transduction cascade. It is controlled by hierarchical signaling events resulting in a focused response as the results place the mitochondrial respiratory chain upstream of tyrosine-protein kinase Lyn, Lyn upstream of tyrosine-protein kinase SYK (Syk), and Syk upstream of numerous targets involved in signaling, transcription, translation, metabolism, and cell cycle regulation. The active mediators of H2O2 signaling colocalize as H2O2 induces mitochondria-associated Lyn and Syk phosphorylation, and a pool of Lyn and Syk reside in the mitochondrial intermembrane space. Finally, the same intermediaries control the signaling response in tissues and species responsive to H2O2 as the respiratory chain, Lyn, and Syk were similarly required for H2O2 signaling in mouse B cells, fibroblasts, and chicken DT40 B cells. Consistent with a broad role, the Syk pathway is coexpressed across tissues, is of early metazoan origin, and displays evidence of evolutionary constraint in the human. These results suggest that H2O2 signaling is under control of a signal transduction pathway that links the respiratory chain to the mitochondrial intermembrane space-localized, ubiquitous, and ancient Syk pathway in hematopoietic and nonhematopoietic cells.


Asunto(s)
Transporte de Electrón , Peróxido de Hidrógeno/metabolismo , Membranas Mitocondriales/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Pollos , Activación Enzimática , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Quinasa Syk , Tirosina/metabolismo
18.
J Biol Chem ; 290(41): 24784-92, 2015 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-26304118

RESUMEN

In both physiological and cell culture systems, EGF-stimulated ERK activity occurs in discrete pulses within individual cells. Many feedback loops are present in the EGF receptor (EGFR)-ERK network, but the mechanisms driving pulsatile ERK kinetics are unknown. Here, we find that in cells that respond to EGF with frequency-modulated pulsatile ERK activity, stimulation through a heterologous TrkA receptor system results in non-pulsatile, amplitude-modulated activation of ERK. We further dissect the kinetics of pulse activity using a combination of FRET- and translocation-based reporters and find that EGFR activity is required to maintain ERK activity throughout the 10-20-minute lifetime of pulses. Together, these data indicate that feedbacks operating within the core Ras-Raf-MEK-ERK cascade are insufficient to drive discrete pulses of ERK activity and instead implicate mechanisms acting at the level of EGFR.


Asunto(s)
Factor de Crecimiento Epidérmico/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Humanos , Cinética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Receptor trkA/metabolismo
19.
Proc Natl Acad Sci U S A ; 110(23): 9368-73, 2013 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-23613587

RESUMEN

Throughout the plant and animal kingdoms specific cell types become polyploid, increasing their DNA content to attain a large cell size. In mammals, megakaryocytes (MKs) become polyploid before fragmenting into platelets. The mammalian trophoblast giant cells (TGCs) exploit their size to form a barrier between the maternal and embryonic tissues. The mechanism of polyploidization has been investigated extensively in Drosophila, in which a modified cell cycle--the endocycle, consisting solely of alternating S and gap phases--produces polyploid tissues. During S phase in the Drosophila endocycle, heterochromatin and specific euchromatic regions are underreplicated and reduced in copy number. Here we investigate the properties of polyploidization in murine MKs and TGCs. We induced differentiation of primary MKs and directly microdissected TGCs from embryonic day 9.5 implantation sites. The copy number across the genome was analyzed by array-based comparative genome hybridization. In striking contrast to Drosophila, the genome was uniformly and integrally duplicated in both MKs and TGCs. This was true even for heterochromatic regions analyzed by quantitative PCR. Underreplication of specific regions in polyploid cells is proposed to be due to a slower S phase, resulting from low expression of S-phase genes, causing failure to duplicate late replicating genomic intervals. We defined the transcriptome of TGCs and found robust expression of S-phase genes. Similarly, S-phase gene expression is not repressed in MKs, providing an explanation for the distinct endoreplication parameters compared with Drosophila. Consistent with TGCs endocycling rather than undergoing endomitosis, they have low expression of M-phase genes.


Asunto(s)
Células Gigantes/citología , Megacariocitos/citología , Poliploidía , Fase S/fisiología , Trofoblastos/citología , Animales , Diferenciación Celular/fisiología , Hibridación Genómica Comparativa , Variaciones en el Número de Copia de ADN , Cartilla de ADN/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/fisiología , Heterocromatina/metabolismo , Ratones , Microdisección , Reacción en Cadena en Tiempo Real de la Polimerasa
20.
Proc Natl Acad Sci U S A ; 110(24): 9776-81, 2013 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-23720309

RESUMEN

Ectodomain cleavage of cell-surface proteins by A disintegrin and metalloproteinases (ADAMs) is highly regulated, and its dysregulation has been linked to many diseases. ADAM10 and ADAM17 cleave most disease-relevant substrates. Broad-spectrum metalloprotease inhibitors have failed clinically, and targeting the cleavage of a specific substrate has remained impossible. It is therefore necessary to identify signaling intermediates that determine substrate specificity of cleavage. We show here that phorbol ester or angiotensin II-induced proteolytic release of EGF family members may not require a significant increase in ADAM17 protease activity. Rather, inducers activate a signaling pathway using PKC-α and the PKC-regulated protein phosphatase 1 inhibitor 14D that is required for ADAM17 cleavage of TGF-α, heparin-binding EGF, and amphiregulin. A second pathway involving PKC-δ is required for neuregulin (NRG) cleavage, and, indeed, PKC-δ phosphorylation of serine 286 in the NRG cytosolic domain is essential for induced NRG cleavage. Thus, signaling-mediated substrate selection is clearly distinct from regulation of enzyme activity, an important mechanism that offers itself for application in disease.


Asunto(s)
Proteínas ADAM/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador alfa/metabolismo , Proteínas ADAM/genética , Proteína ADAM17 , Anfirregulina , Angiotensina II/farmacología , Western Blotting , Línea Celular Tumoral , Familia de Proteínas EGF , Activación Enzimática/efectos de los fármacos , Citometría de Flujo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Isoenzimas/metabolismo , Células Jurkat , Ligandos , Fosforilación , Proteína Quinasa C/metabolismo , Proteínas/metabolismo , Proteolisis/efectos de los fármacos , Interferencia de ARN , Serina/genética , Serina/metabolismo , Especificidad por Sustrato , Acetato de Tetradecanoilforbol/farmacología , Factor de Crecimiento Transformador alfa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA