Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Nat Commun ; 14(1): 6464, 2023 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-37833256

RESUMEN

Regulation of global transcription output is important for normal development and disease, but little is known about the mechanisms involved. DNA topoisomerase I (TOP1) is an enzyme well-known for its role in relieving DNA supercoils for enabling transcription. Here, we report a non-enzymatic function of TOP1 that downregulates RNA synthesis. This function is dependent on specific DNA-interacting residues located on a conserved protein surface. A loss-of-function knock-in mutation on this surface, R548Q, is sufficient to cause hypertranscription and alter differentiation outcomes in mouse embryonic stem cells (mESCs). Hypertranscription in mESCs is accompanied by reduced TOP1 chromatin binding and change in genomic supercoiling. Notably, the mutation does not impact TOP1 enzymatic activity; rather, it diminishes TOP1-DNA binding and formation of compact protein-DNA structures. Thus, TOP1 exhibits opposing influences on transcription through distinct activities which are likely to be coordinated. This highlights TOP1 as a safeguard of appropriate total transcription levels in cells.


Asunto(s)
ADN-Topoisomerasas de Tipo I , Transcripción Genética , Animales , Ratones , ADN-Topoisomerasas de Tipo I/metabolismo , Replicación del ADN , Mutación , ADN/genética
2.
Nat Commun ; 14(1): 2439, 2023 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-37117180

RESUMEN

Cancer cells undergo transcriptional reprogramming to drive tumor progression and metastasis. Using cancer cell lines and patient-derived tumor organoids, we demonstrate that loss of the negative elongation factor (NELF) complex inhibits breast cancer development through downregulating epithelial-mesenchymal transition (EMT) and stemness-associated genes. Quantitative multiplexed Rapid Immunoprecipitation Mass spectrometry of Endogenous proteins (qPLEX-RIME) further reveals a significant rewiring of NELF-E-associated chromatin partners as a function of EMT and a co-option of NELF-E with the key EMT transcription factor SLUG. Accordingly, loss of NELF-E leads to impaired SLUG binding on chromatin. Through integrative transcriptomic and genomic analyses, we identify the histone acetyltransferase, KAT2B, as a key functional target of NELF-E-SLUG. Genetic and pharmacological inactivation of KAT2B ameliorate the expression of EMT markers, phenocopying NELF ablation. Elevated expression of NELF-E and KAT2B is associated with poorer prognosis in breast cancer patients, highlighting the clinical relevance of our findings. Taken together, we uncover a crucial role of the NELF-E-SLUG-KAT2B epigenetic axis in breast cancer carcinogenesis.


Asunto(s)
Neoplasias de la Mama , Femenino , Humanos , Neoplasias de la Mama/patología , Carcinogénesis/genética , Línea Celular Tumoral , Cromatina , Epigénesis Genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Factores de Transcripción p300-CBP/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Factores de Transcripción/metabolismo
3.
Nat Cell Biol ; 22(2): 175-186, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31932739

RESUMEN

Mouse embryonic stem cells (ESCs) sporadically transit into an early embryonic-like state characterized by the expression of 2-cell (2C) stage-restricted transcripts. Here, we identify a maternal factor-negative elongation factor A (NELFA)-whose heterogeneous expression in mouse ESCs is coupled to 2C gene upregulation and expanded developmental potential in vivo. We show that NELFA partners with Top2a in an interaction specific to the 2C-like state, and that it drives the expression of Dux-a key 2C regulator. Accordingly, loss of NELFA and/or Top2a suppressed Dux activation. Further characterization of 2C-like cells uncovered reduced glycolytic activity; remarkably, mere chemical suppression of glycolysis was sufficient to promote a 2C-like fate, obviating the need for genetic manipulation. Global chromatin state analysis on NELFA-induced cells revealed decommissioning of ESC-specific enhancers, suggesting ESC-state impediments to 2C reversion. Our study positions NELFA as one of the earliest drivers of the 2C-like state and illuminates factors and processes that govern this transition.


Asunto(s)
Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Células Madre Embrionarias de Ratones/metabolismo , Factores de Transcripción/genética , Animales , Diferenciación Celular , Cromatina/química , Cromatina/metabolismo , ADN-Topoisomerasas de Tipo II/genética , ADN-Topoisomerasas de Tipo II/metabolismo , Embrión de Mamíferos , Femenino , Glucólisis/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Patrón de Herencia , Masculino , Ratones , Ratones Transgénicos , Células Madre Embrionarias de Ratones/citología , Familia de Multigenes , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo
4.
J Biol Chem ; 283(3): 1679-1691, 2008 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-17974561

RESUMEN

The Sprouty (Spry) proteins function as inhibitors of the Ras-ERK pathway downstream of various receptor tyrosine kinases. In this study, we have identified Tesk1 (testicular protein kinase 1) as a novel regulator of Spry2 function. Endogenous Tesk1 and Spry2 exist in a complex in cell lines and mouse tissues. Tesk1 coexpression relocalizes Spry2 to vesicles including endosomes, inhibiting its translocation to membrane ruffles upon growth factor stimulation. Independent of its kinase activity, Tesk1 binding leads to a loss of Spry2 function as an inhibitor of ERK phosphorylation and reverses inhibition of basic fibroblast growth factor (bFGF)- and nerve growth factor-induced neurite outgrowth in PC12 cells by Spry2. Furthermore, depletion of endogenous Tesk1 in PC12 cells leads to a reduction in neurite outgrowth induced by bFGF. Tesk1 nullifies the inhibitory effect of Spry2 by abrogating its interaction with the adaptor protein Grb2 and interfering with its serine dephosphorylation upon bFGF and FGF receptor 1 stimulation by impeding its binding to the catalytic subunit of protein phosphatase 2A. A construct of Tesk1 that binds to Spry2 but does not localize to the vesicles does not interfere with its function, highlighting the importance of subcellular localization of Tesk1 in this context. Conversely, Tesk1 does not affect interaction of Spry2 with the E3 ubiquitin ligase, c-Cbl, and consequently, does not affect its inhibition of Cbl-mediated ubiquitination of the epidermal growth factor receptor. By selectively modulating the downstream effects of Spry2, Tesk1 may thus serve as a molecular determinant of the signaling outcome.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales , Animales , Vesículas Citoplasmáticas/metabolismo , Regulación hacia Abajo , Receptores ErbB/metabolismo , Proteína Adaptadora GRB2/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Neuritas/metabolismo , Células PC12 , Fosforilación , Unión Proteica , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Ratas , Proteínas Represoras/metabolismo , Serina/metabolismo , Fracciones Subcelulares , Ubiquitinación , Proteínas ras/metabolismo
5.
Stem Cells ; 25(2): 529-36, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17068185

RESUMEN

Mesenchymal stem cells have been implicated as playing an important role in stem cell engraftment. Recently, a new pluripotent population of umbilical cord blood (UCB) cells, unrestricted somatic stem cells (USSCs), with intrinsic and directable potential to develop into mesodermal, endodermal, and ectodermal fates, has been identified. In this study, we evaluated the capacity of ex vivo expanded USSCs to influence the homing of UCB-derived CD34(+) cells into the marrow and spleen of nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice. USSCs induced a significant enhancement of CD34(+) cell homing to both bone marrow and spleen (2.2 +/- 0.3- and 2.4 +/- 0.6-fold, respectively; p < .05), with a magnitude similar to that induced by USSCs that had been thawed prior to transplantation. The effect of USSCs was dose-dependent and detectable at USSC:CD34(+) ratios of 1:1 and above. Enhanced marrow homing by USSCs was unaltered by extensive culture passaging of the cells, as similar enhancement was observed for both early-passage (passage 5 [p5]) and late-passage (p10) USSCs. The homing effect of USSCs was also reflected in an increased proportion of NOD/SCID mice exhibiting significant human cell engraftment 6 weeks after transplantation, with a similar distribution of myeloid and lymphoid components. USSCs enhanced the homing of cellular products of ex vivo expanded UCB lineage-negative (lin(-)) cells, generated in 14-day cultures by Selective Amplification. The relative proportion of homing CD34(+) cells within the culture-expanded cell population was unaltered by USSC cotransplantation. Production of stromal-derived factor-1 (SDF-1) by USSCs was detected by both gene expression and protein released into culture media of these cells. Knockdown of SDF-1 production by USSCs using lentiviral-SiRNA led to a significant (p < .05) reduction in USSC-mediated enhancement of CD34(+) homing. Our findings thus suggest a clinical potential for using USSCs in facilitating homing and engraftment for cord blood transplant recipients.


Asunto(s)
Antígenos CD34/inmunología , Trasplante de Células Madre de Sangre del Cordón Umbilical , Sangre Fetal/citología , Sangre Fetal/trasplante , Células Madre/citología , Animales , Células de la Médula Ósea/citología , Separación Celular , Células Cultivadas , Quimiocina CXCL12 , Quimiocinas CXC/genética , Quimiocinas CXC/metabolismo , Regulación de la Expresión Génica , Humanos , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo
6.
J Biol Chem ; 281(40): 29993-30000, 2006 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-16893902

RESUMEN

Because the Sprouty (Spry) proteins were shown to be inhibitors of the mainstream Ras/ERK pathway, there has been considerable interest in ascertaining their mechanism of action especially since a possible role as tumor suppressors for these inhibitory proteins has been suggested. We compared the ability of the mammalian Spry isoforms to inhibit the Ras/ERK pathway in the context of fibroblast growth factor receptor (FGFR) signaling. Spry2 is considerably more inhibitory than Spry1 or Spry4, and this correlates with the binding to Grb2 via a C-terminal proline-rich sequence that is found exclusively on Spry2. This PXXPXR motif binds directly to the N-terminal Src homology domain 3 of Grb2, and when added onto the C terminus of Spry4 the resultant chimera inhibits the Ras/ERK pathway. The ability to inhibit neurite outgrowth in PC-12 cells correlates with the propensity of Spry isoforms and engineered constructs to inhibit the phosphorylation of ERK1/2. The PXXPXR motif is cryptic in unstimulated cells, and it is postulated that Spry2 undergoes a conformational change following FGFR stimulation, enabling the subsequent interaction with Grb2. We present evidence that Spry2 can compete with the RasGEF (guanine nucleotide exchange factor) SOS1 for binding to Grb2, resulting in the inhibition of phosphorylation of ERK1/2.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Fragmentos de Péptidos/fisiología , Proteínas/fisiología , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Transducción de Señal/fisiología , Proteínas ras/antagonistas & inhibidores , Dominios Homologos src/fisiología , Animales , Línea Celular , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana , Células PC12 , Fosforilación , Unión Proteica , Ratas
7.
Cancer Res ; 64(17): 6127-36, 2004 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15342396

RESUMEN

Sprouty (Spry) proteins were found to be endogenous inhibitors of the Ras/mitogen-activated protein kinase pathway that play an important role in the remodeling of branching tissues. We investigated Spry expression levels in various cancers and found that Spry1 and Spry2 were down-regulated consistently in breast cancers. Such prevalent patterns of down-regulation may herald the later application of these isoforms as tumor markers that are breast cancer specific and more profound than currently characterized markers. Spry1 and 2 were expressed specifically in the luminal epithelial cells of breast ducts, with higher expression during stages of tissue remodeling when the epithelial ducts are forming and branching. These findings suggest that Sprys might be involved as a modeling counterbalance and surveillance against inappropriate epithelial expansion. The abrogation of endogenous Spry activity in MCF-7 cells by the overexpression of a previously characterized dominant-negative mutant of Spry, hSpry2Y55F resulted in enhanced cell proliferation in vitro. The hSpry2Y55F stably expressing cells also formed larger and greater number of colonies in the soft-agar assay. An in vivo nude mice assay showed a dramatic increase in the tumorigenic potential of hSpry2Y55F stable cells. The consistent down-regulation of Spry1 and 2 in breast cancer and the experimental evidence using a dominant-negative hSpry2Y55F indicate that Spry proteins may actively maintain tissue integrity that runs amok when their expression is decreased below normal threshold levels. This alludes to a previously unrecognized role for Sprys in cancer development.


Asunto(s)
Neoplasias de la Mama/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas de la Membrana/biosíntesis , Fosfoproteínas/biosíntesis , Biosíntesis de Proteínas , Proteínas Adaptadoras Transductoras de Señales , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , División Celular/fisiología , Línea Celular Tumoral , Regulación hacia Abajo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/fisiología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinasas , Proteínas/antagonistas & inhibidores , Proteínas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Trasplante Heterólogo
8.
J Biol Chem ; 278(35): 33456-64, 2003 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-12815057

RESUMEN

Mammalian Sprouty (Spry) proteins are now established as receptor tyrosine kinase-induced modulators of the Ras/mitogen-activated protein kinase pathway. Specifically, hSpry2 inhibits the fibroblast growth factor receptor (FGFR)-induced mitogen-activated protein kinase pathway but conversely prolongs activity of the same pathway following epidermal growth factor (EGF) stimulation, where activated EGF receptors are retained on the cell surface. In this study it is demonstrated that hSpry2 is tyrosine-phosphorylated upon stimulation by either FGFR or EGF and subsequently binds endogenous c-Cbl with high affinity. A conserved motif on hSpry2, together with phosphorylation on tyrosine 55, is required for its enhanced interaction with the SH2-like domain of c-Cbl. A hSpry2 mutant (Y55F) that did not exhibit an enhanced binding with c-Cbl failed to retain EGF receptors on the cell surface. Furthermore, individually mutating hSpry2 residues 52-59 to alanine indicated a tight correlation between their affinity for c-Cbl binding and their inhibition of ERK2 activity in the FGFR pathway. We postulate that tyrosine phosphorylation "activates" hSpry2 by enhancing its interaction with c-Cbl and that this interaction is critical for its physiological function in a signal-specific context.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Tirosina/metabolismo , Ubiquitina-Proteína Ligasas , Alanina/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Western Blotting , Células COS , Línea Celular , Membrana Celular/metabolismo , Células Cultivadas , ADN/metabolismo , ADN Complementario/metabolismo , Regulación hacia Abajo , Humanos , Sistema de Señalización de MAP Quinasas , Microscopía Confocal , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Mutación , Fosforilación , Plásmidos/metabolismo , Pruebas de Precipitina , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-cbl , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Homología de Secuencia de Aminoácido , Factores de Tiempo , Tirosina/química , Dominios Homologos src
9.
J Biol Chem ; 277(5): 3195-201, 2002 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11698404

RESUMEN

Several genetic studies in Drosophila have shown that the dSprouty (dSpry) protein inhibits the Ras/mitogen-activated protein (MAP) kinase pathway induced by various activated receptor tyrosine kinase receptors, most notably those of the epidermal growth factor receptor (EGFR) and fibroblast growth factor receptor (FGFR). Currently, the mode of action of dSpry is unknown, and the point of inhibition remains controversial. There are at least four mammalian Spry isoforms that have been shown to co-express preferentially with FGFRs as compared with EGFRs. In this study, we investigated the effects of the various mammalian Spry isoforms on the Ras/MAP kinase pathway in cells overexpressing constitutively active FGFR1. hSpry2 was significantly more potent than mSpry1 or mSpry4 in inhibiting the Ras/MAP kinase pathway. Additional experiments indicated that full-length hSpry2 was required for its full potency. hSpry2 had no inhibitory effect on either the JNK or the p38 pathway and displayed no inhibition of FRS2 phosphorylation, Akt activation, and Ras activation. Constitutively active mutants of Ras, Raf, and Mek were employed to locate the prospective point of inhibition of hSpry2 downstream of activated Ras. Results from this study indicated that hSpry2 exerted its inhibitory effect at the level of Raf, which was verified in a Raf activation assay in an FGF signaling context.


Asunto(s)
Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-raf/antagonistas & inhibidores , Animales , Línea Celular , Clonación Molecular , Drosophila , Humanos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas del Tejido Nervioso/genética , Plásmidos , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA