Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Nucleic Acids Res ; 49(3): 1345-1363, 2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33434264

RESUMEN

Enhancers play important roles in controlling gene expression in a choreographed spatial and temporal manner during development. However, it is unclear how these regulatory regions are established during differentiation. Here we investigated the genome-wide binding profile of the forkhead transcription factor FOXK2 in human embryonic stem cells (ESCs) and downstream cell types. This transcription factor is bound to thousands of regulatory regions in human ESCs, and binding at many sites is maintained as cells differentiate to mesendodermal and neural precursor cell (NPC) types, alongside the emergence of new binding regions. FOXK2 binding is generally associated with active histone marks in any given cell type. Furthermore newly acquired, or retained FOXK2 binding regions show elevated levels of activating histone marks following differentiation to NPCs. In keeping with this association with activating marks, we demonstrate a role for FOXK transcription factors in gene activation during NPC differentiation. FOXK2 occupancy in ESCs is therefore an early mark for delineating the regulatory regions, which become activated in later lineages.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/metabolismo , Factores de Transcripción Forkhead/metabolismo , Activación Transcripcional , Linaje de la Célula/genética , Células Cultivadas , Cromatina/metabolismo , Células Madre Embrionarias/citología , Endodermo/citología , Elementos de Facilitación Genéticos , Código de Histonas , Humanos , Mesodermo/citología , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Factores de Transcripción/metabolismo
2.
Nucleic Acids Res ; 44(4): 1566-78, 2016 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-26578569

RESUMEN

Transcription factors belonging to the same transcription factor families contain very similar DNA binding domains and hence have the potential to bind to related DNA sequences. However, subtle differences in binding specificities can be detected in vitro with the potential to direct specific responses in vivo. Here, we have examined the binding properties of three Forkhead (FOX) transcription factors, FOXK2, FOXO3 and FOXJ3 in vivo. Extensive overlap in chromatin binding is observed, although underlying differential DNA binding specificity can dictate the recruitment of FOXK2 and FOXJ3 to chromatin. However, functionally, FOXO3-dependent gene regulation is generally mediated not through uniquely bound regions but through regions occupied by both FOXK2 and FOXO3 where both factors play a regulatory role. Our data point to a model whereby FOX transcription factors control gene expression through dynamically binding and generating partial occupancy of the same site rather than mutually exclusive binding derived by stable binding of individual FOX proteins.


Asunto(s)
Sitios de Unión/genética , Proteínas de Ciclo Celular/genética , Factores de Transcripción Forkhead/genética , Secuencia de Bases/genética , Proteínas de Ciclo Celular/metabolismo , Cromatina/genética , Cromatina/metabolismo , Proteínas de Unión al ADN , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Genoma Humano , Humanos
4.
Nucleic Acids Res ; 42(10): 6232-42, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24748658

RESUMEN

There are numerous forkhead transcription factors in mammalian cells but we know little about the molecular functions of the majority of these. FOXK2 is a ubiquitously expressed family member suggesting an important function across multiple cell types. Here, we show that FOXK2 binds to the SIN3A and PR-DUB complexes. The PR-DUB complex contains the important tumour suppressor protein, the deubiquitinase BAP1. FOXK2 recruits BAP1 to DNA, promotes local histone deubiquitination and causes changes in target gene activity. Our results therefore provide an important link between BAP1 and the transcription factor FOXK2 and demonstrate how BAP1 can be recruited to specific regulatory loci.


Asunto(s)
Cromatina/metabolismo , Factores de Transcripción Forkhead/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Línea Celular , Cromatina/enzimología , Factores de Transcripción Forkhead/química , Regulación de la Expresión Génica , Genoma , Humanos , Dominios y Motivos de Interacción de Proteínas , Proteínas Represoras/metabolismo , Complejo Correpresor Histona Desacetilasa y Sin3 , Proteínas Supresoras de Tumor/química , Ubiquitina Tiolesterasa/química
5.
Mol Cancer ; 14: 69, 2015 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-25889361

RESUMEN

BACKGROUND: Survival rates for oesophageal adenocarcinoma (OAC) remain disappointingly poor and current conventional treatment modalities have minimal impact on long-term survival. This is partly due to a lack of understanding of the molecular changes that occur in this disease. Previous studies have indicated that the transcription factor FOXM1 is commonly upregulated in this cancer type but the impact of this overexpression on gene expression in the context of OAC is largely unknown. FOXM1 does not function alone but works alongside the antagonistically-functioning co-regulatory MMB and DREAM complexes. METHODS: To establish how FOXM1 affects gene expression in OAC we have identified the FOXM1 target gene network in OAC-derived cells using ChIP-seq and determined the expression of both its coregulatory partners and members of this target gene network in OAC by digital transcript counting using the Nanostring gene expression assay. RESULTS: We find co-upregulation of FOXM1 with its target gene network in OAC. Furthermore, we find changes in the expression of its coregulatory partners, including co-upregulation of LIN9 and, surprisingly, reduced expression of LIN54. Mechanistically, we identify LIN9 as the direct binding partner for FOXM1 in the MMB complex. In the context of OAC, both coregulator (eg LIN54) and target gene (eg UHRF1) expression levels are predictive of disease stage. CONCLUSIONS: Together our data demonstrate that there are global changes to the FOXM1 regulatory network in OAC and the expression of components of this network help predict cancer prognosis.


Asunto(s)
Adenocarcinoma/genética , Neoplasias Esofágicas/genética , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Línea Celular Tumoral , Proteína Forkhead Box M1 , Humanos , Proteínas Nucleares/genética , Transactivadores/genética , Proteínas Supresoras de Tumor/genética , Regulación hacia Arriba/genética
6.
Proteomics ; 14(19): 2179-89, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24634419

RESUMEN

MBD5 and MBD6 are two members of the methyl-CpG-binding domain (MBD) family of proteins that are poorly characterized. Studies performed thus far have failed to show binding of the MBD5 and MBD6 MBD to methylated DNA. Here, we show that both MBD5 and MBD6 interact with the mammalian PR-DUB Polycomb protein complex in a mutually exclusive manner. Strikingly, the MBD of MBD5 and MBD6 is both necessary and sufficient to mediate this interaction. Chromatin immunoprecipitation analyses reveal that MBD6 and FOXK2/PR-DUB share a subset of genomic target genes, suggesting a functional interaction in vivo. Finally, we show that MBD6, but not MBD5, is recruited to sites of DNA damage in a PR-DUB independent manner. Our study thus implies a shared function for MBD5 and MBD6 through an interaction with PR-DUB, as well as an MBD6-specific recruitment to sites of DNA damage.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Secuencia de Aminoácidos , Cromatina , Daño del ADN , Metilación de ADN , Factores de Transcripción Forkhead , Células HEK293 , Células HeLa , Humanos , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo
7.
J Biol Chem ; 287(42): 35382-35396, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-22915589

RESUMEN

The GRB2-associated binder 1 (GAB1) docking/scaffold protein is a key mediator of the MET-tyrosine kinase receptor activated by hepatocyte growth factor/scatter factor (HGF/SF). Activated MET promotes recruitment and tyrosine phosphorylation of GAB1, which in turn recruits multiple proteins and mediates MET signaling leading to cell survival, motility, and morphogenesis. We previously reported that, without its ligand, MET is a functional caspase target during apoptosis, allowing the generation of a p40-MET fragment that amplifies apoptosis. In this study we established that GAB1 is also a functional caspase target by evidencing a caspase-cleaved p35-GAB1 fragment that contains the MET binding domain. GAB1 is cleaved by caspases before MET, and the resulting p35-GAB1 fragment is phosphorylated by MET upon HGF/SF binding and can interact with a subset of GAB1 partners, PI3K, and GRB2 but not with SHP2. This p35-GAB1 fragment favors cell survival by maintaining HGF/SF-induced MET activation of AKT and by hindering p40-MET pro-apoptotic function. These data demonstrate an anti-apoptotic role of caspase-cleaved GAB1 in HGF/SF-MET signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis/fisiología , Caspasas/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Proteolisis , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Caspasas/genética , Perros , Activación Enzimática/fisiología , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo , Células HeLa , Células Hep G2 , Factor de Crecimiento de Hepatocito/genética , Humanos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-met/genética
8.
Elife ; 122023 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-37934570

RESUMEN

ZMYM2 is a zinc finger transcriptional regulator that plays a key role in promoting and maintaining cell identity. It has been implicated in several diseases such as congenital anomalies of the kidney where its activity is diminished and cancer where it participates in oncogenic fusion protein events. ZMYM2 is thought to function through promoting transcriptional repression and here we provide more evidence to support this designation. Here we studied ZMYM2 function in human cells and demonstrate that ZMYM2 is part of distinct chromatin-bound complexes including the established LSD1-CoREST-HDAC1 corepressor complex. We also identify new functional and physical interactions with ADNP and TRIM28/KAP1. The ZMYM2-TRIM28 complex forms in a SUMO-dependent manner and is associated with repressive chromatin. ZMYM2 and TRIM28 show strong functional similarity and co-regulate a large number of genes. However, there are no strong links between ZMYM2-TRIM28 binding events and nearby individual gene regulation. Instead, ZMYM2-TRIM28 appears to regulate genes in a more regionally defined manner within TADs where it can directly regulate co-associated retrotransposon expression. We find that different types of ZMYM2 binding complex associate with and regulate distinct subclasses of retrotransposons, with ZMYM2-ADNP complexes at SINEs and ZMYM2-TRIM28 complexes at LTR elements. We propose a model whereby ZMYM2 acts directly through retrotransposon regulation, which may then potentially affect the local chromatin environment and associated coding gene expression.


Asunto(s)
Elementos Transponibles de ADN , Retroelementos , Humanos , Dedos de Zinc , Cromatina , Proteínas Co-Represoras , Proteínas de Unión al ADN , Factores de Transcripción
9.
J Biol Chem ; 285(46): 35728-39, 2010 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-20810654

RESUMEN

Several mammalian forkhead transcription factors have been shown to impact on cell cycle regulation and are themselves linked to cell cycle control systems. Here we have investigated the little studied mammalian forkhead transcription factor FOXK2 and demonstrate that it is subject to control by cell cycle-regulated protein kinases. FOXK2 exhibits a periodic rise in its phosphorylation levels during the cell cycle, with hyperphosphorylation occurring in mitotic cells. Hyperphosphorylation occurs in a cyclin-dependent kinase (CDK)·cyclin-dependent manner with CDK1·cyclin B as the major kinase complex, although CDK2 and cyclin A also appear to be important. We have mapped two CDK phosphorylation sites, serines 368 and 423, which play a role in defining FOXK2 function through regulating its stability and its activity as a transcriptional repressor protein. These two CDK sites appear vital for FOXK2 function because expression of a mutant lacking these sites cannot be tolerated and causes apoptosis.


Asunto(s)
Ciclo Celular , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Factores de Transcripción Forkhead/metabolismo , Secuencia de Aminoácidos , Apoptosis , Proteína Quinasa CDC2/genética , Proteína Quinasa CDC2/metabolismo , Línea Celular Tumoral , Ciclina B1/genética , Ciclina B1/metabolismo , Quinasas Ciclina-Dependientes/genética , Ciclinas/genética , Factores de Transcripción Forkhead/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Inmunoprecipitación , Espectrometría de Masas , Microscopía Fluorescente , Datos de Secuencia Molecular , Mutación , Fosforilación , Unión Proteica , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serina/genética , Serina/metabolismo , Transfección
11.
Biochem Biophys Res Commun ; 411(4): 780-5, 2011 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-21782801

RESUMEN

The GRB2 associated binder 1 (GAB1) is an essential docking/adaptor protein for transmitting intracellular signals of the MET tyrosine kinase receptor activated by hepatocyte growth factor/scatter factor (HGF/SF). We found that in response to hours of HGF/SF treatment, the GAB1 protein level is degraded by a mechanism involving MET activity and the proteasomal machinery. We also showed that GAB1 is both multi- and poly-ubiquitinated in a CBL-dependent manner. A long term exposure to HGF/SF caused a more sustained down-regulation of GAB1 than of MET, associated with a loss of reactivation of the ERK MAP kinases to subsequent acute ligand treatment. These data demonstrate that GAB1 is ubiquitinated by CBL and degraded by the proteasome, and plays a role in negative-feedback regulation of HGF/SF-MET signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Ubiquitinación , Células HEK293 , Células HeLa , Humanos , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Transducción de Señal , Ubiquitina/metabolismo
13.
Nucleic Acids Res ; 35(15): 5203-12, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17670796

RESUMEN

The combinatorial control of gene expression by the association of members of different families of transcription factors is a common theme in eukaryotic transcriptional control. The MADS-box transcription factors SRF and Mcm1 represent paradigms for such regulation through their interaction with numerous partner proteins. For example, in Saccharomyces cerevisiae, Mcm1 interacts with the forkhead transcription factor Fkh2. Here, we identify a novel interaction between SRF and the Forkhead transcription factor FOXK1 in human cells. The importance of this interaction is shown for the regulation of the SRF target genes SM alpha-actin and PPGB. The binding of FOXK1 to the SM alpha-actin and PPGB promoters requires the presence of SRF on the promoter. FOXK1 acts as a transcriptional repressor and it represses SM alpha-actin and PPGB expression. Thus FOXK1 represents an additional member of the growing repertoire of transcription factors that can interact with SRF and modulate the transcriptional output from SRF-regulated promoters.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Factor de Respuesta Sérica/metabolismo , Actinas/genética , Línea Celular Tumoral , ADN/metabolismo , Humanos , Regiones Promotoras Genéticas , Proteínas Represoras/metabolismo , Factor de Respuesta Sérica/antagonistas & inhibidores
14.
Biochem Biophys Res Commun ; 367(3): 573-7, 2008 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-18187039

RESUMEN

The MET tyrosine kinase receptor activated by its ligand HGF/SF, induces several cellular responses, including survival. Nonetheless, the MET receptor is cleaved in stress conditions by caspases within its intracellular region, generating a 40kDa fragment, p40 MET, with pro-apoptotic properties. Here, we established that this cleavage splits the receptor at the juxtamembrane ESVD site, causing the concomitant generation of p100 MET, corresponding to the entire extracellular region of the MET receptor still spanning the membrane. This fragment is able to bind HGF/SF and to prevent HGF-dependent signaling downstream of full MET, demonstrating its function as a decoy receptor.


Asunto(s)
Caspasas/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Fragmentos de Péptidos/biosíntesis , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Apoptosis/efectos de los fármacos , Fraccionamiento Celular , Células Cultivadas , Perros , Técnicas de Transferencia de Gen , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Factor de Crecimiento de Hepatocito/química , Humanos , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/farmacología , Proteínas Proto-Oncogénicas c-met/química , Proteínas Proto-Oncogénicas c-met/genética , Transducción de Señal/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología
15.
Oncogenesis ; 7(3): 29, 2018 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-29540677

RESUMEN

The forkhead transcription factor FOXK2 plays a critical role in suppressing tumorigenesis and mediating cytotoxic drug action in breast cancer. However, the mechanism by which the biological function of FOXK2 is regulated remains poorly understood. Here, we investigated the role of SUMOylation in modulating FOXK2-mediated drug sensitivity. We identified SUMOylation consensus motifs within the FOXK2 sequence and constructed two SUMOylation-defective double mutants by converting lysine 527 and 633 to arginines and glutamic acid 529 and 635 to alanines, respectively. We found that both the FOXK2 SUMOylation-deficient (K527/633 R) and (E529/635 A) mutants were ineffective in mediating the cytotoxic function of paclitaxel when compared to the wild-type (WT) FOXK2. When overexpressed, unlike the wild-type (WT) FOXK2, the K527/633 R mutant had little effect on the sensitivity of MCF-7 and MDA-MB-231 cells to paclitaxel, as examined by cell viability and clonogenic assays. Our results also showed that MCF-7 cells overexpressing the K527/633 R mutant form of FOXK2 or the empty expression vector have lower protein and mRNA levels of its tumour suppressive transcriptional target FOXO3 compared to the wild-type FOXK2. Consistently, ChIP assays revealed that unlike wild-type FOXK2, the SUMOylation-defective (K527/633 R) mutant is unable to bind to the FOXO3 promoter, despite expressing comparable levels of protein and having the same subcellular localization as the wild-type FOXK2 in MCF-7 cells. Interestingly, expression of neither the wild-type nor the K527/633 R mutant FOXK2 had any effect on the proliferation and paclitaxel sensitivity of the MCF-7 TaxR paclitaxel-resistant cells. In agreement, both the wild-type and the (K527/633 R) mutant FOXK2 failed to bind to the endogenous FOXO3 promoter in these cells. Collectively, our results suggest that SUMOylation positively regulates FOXK2 transcriptional activity and has a role in mediating the cytotoxic response to paclitaxel through the tumour suppressor FOXO3.

16.
Mol Cell Biol ; 24(23): 10328-39, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15542841

RESUMEN

The MET tyrosine kinase, the receptor of hepatocyte growth factor-scatter factor (HGF/SF), is known to be essential for normal development and cell survival. We report that stress stimuli induce the caspase-mediated cleavage of MET in physiological cellular targets, such as epithelial cells, embryonic hepatocytes, and cortical neurons. Cleavage occurs at aspartic residue 1000 within the SVD site of the juxtamembrane region, independently of the crucial docking tyrosine residues Y1001 or Y1347 and Y1354. This cleavage generates an intracellular 40-kDa MET fragment containing the kinase domain. The p40 MET fragment itself causes apoptosis of MDCK epithelial cells and embryonic cortical neurons, whereas its kinase-dead version is impaired in proapoptotic activity. Finally, HGF/SF treatment does not favor MET cleavage and apoptosis, confirming the known survival role of ligand-activated MET. Our results show that stress stimuli convert the MET survival receptor into a proapoptotic factor.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Proteínas Proto-Oncogénicas c-met/fisiología , Secuencia de Aminoácidos , Animales , Ácido Aspártico/química , Ácido Aspártico/metabolismo , Western Blotting , Caspasa 3 , Supervivencia Celular , Células Cultivadas , Citocinas/metabolismo , Perros , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Hepatocitos/metabolismo , Etiquetado Corte-Fin in Situ , Ligandos , Ratones , Microscopía Fluorescente , Datos de Secuencia Molecular , Neuronas/metabolismo , Plásmidos/metabolismo , Biosíntesis de Proteínas , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Recombinantes/química , Transfección , Tirosina/química
17.
Ann N Y Acad Sci ; 1090: 188-202, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17384262

RESUMEN

Hepatocyte growth factor/scatter factor (HGF/SF) induces scattering, morphogenesis, and survival of epithelial cells through activation of the MET tyrosine kinase receptor. HGF/SF and MET are involved in normal development and tumor progression of many tissues and organs, including the mammary gland. In order to find target genes of HGF/SF involved in its survival function, we used an oligonucleotide microarray representing 1,920 genes known to be involved in apoptosis, transcriptional regulation, and signal transduction. MCF-10A human mammary epithelial cells were grown in the absence of serum and treated or not with HGF/SF for 2 h. Total RNA was reverse-transcribed to cDNA in the presence of fluorescent Cy3-dUTP or Cy5-dUTP to generate fluorescently labeled cDNA probes. Microarrays were performed and the ratios of Cy5/Cy3 fluorescence were determined. The expression of three apoptotic genes was modified by HGF/SF, with A20 being upregulated, and DAXX and SMAC being downregulated. These changes of expression were confirmed by real-time quantitative PCR. According to current-knowledge, A20 is antiapoptotic and SMAC is proapoptotic, while a pro- or antiapoptotic function of DAXX is controversial. The fact that HGF/SF upregulates an antiapoptotic gene (A20) and downregulates a proapoptotic gene (SMAC) is in agreement with its survival effect in MCF-10A cells. This study identified novel apoptotic genes regulated by HGF/SF, which can contribute to its survival effect.


Asunto(s)
Apoptosis/genética , Regulación de la Expresión Génica/fisiología , Factor de Crecimiento de Hepatocito/fisiología , Glándulas Mamarias Humanas/metabolismo , Secuencia de Bases , Western Blotting , Línea Celular , Cartilla de ADN , Células Epiteliales/metabolismo , Humanos , Glándulas Mamarias Humanas/citología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Cancer Res ; 63(12): 3257-62, 2003 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12810656

RESUMEN

In this study, a novel approach to antitumor therapy was devised by generating a chimeric tumor-targeted killer protein, referred to as immunocasp-3, that comprises a single-chain anti-erbB2/HER2 antibody with a NH(2)-terminal signal sequence, a Pseudomonas exotoxin A translocation domain, and a constitutively active caspase-3 molecule. In principle, cells transfected with the immunocasp-3 gene would express and secrete the chimeric protein, which then binds to HER2-overexpressing tumor cells. Subsequent cleavage of the constitutively active capase-3 domain from the immunocasp-3 molecule and its release from internalized vesicles would lead to apoptotic tumor cell death. To test this strategy, we transduced human lymphoma Jurkat cells with a chimeric immunocasp-3 gene expression vector and showed that they not only expressed and secreted the fusion protein but also selectively killed tumor cells overexpressing HER2 in vitro. i.v. injection of the transduced Jurkat cells led to tumor regression in a mouse xenograft model because of continuous secretion of immunocasp-3 by the transduced cells. The growth of HER2-positive tumor cells in this model was inhibited by i.m. as well as intratumor injection of immunocasp-3 expression plasmid DNA, indicating that the immunocasp-3 molecules secreted by transfected cells have systematic antitumor activity. We conclude that the immunocasp-3 molecule, combining the properties of a tumor-specific antibody with the proapoptotic activity of a caspase, has potent and selective antitumor activity, either as cell-based therapy or as a DNA vaccine. These findings provide a compelling rationale for therapeutic protocols designed for erbB2/HER2-positive tumors.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antineoplásicos/uso terapéutico , Antígenos de Neoplasias/inmunología , Apoptosis/efectos de los fármacos , Caspasas/uso terapéutico , Inmunoconjugados/uso terapéutico , Región Variable de Inmunoglobulina/uso terapéutico , Proteínas de Neoplasias/inmunología , Receptor ErbB-2/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Antineoplásicos/farmacología , Caspasa 3 , Caspasas/farmacología , Activación Enzimática , Inducción Enzimática , Femenino , Células HeLa , Humanos , Inmunoconjugados/farmacología , Región Variable de Inmunoglobulina/farmacología , Células Jurkat/metabolismo , Células Jurkat/trasplante , Ratones , Datos de Secuencia Molecular , Neoplasias Ováricas/patología , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Zhonghua Wai Ke Za Zhi ; 41(9): 641-5, 2003 Sep.
Artículo en Zh | MEDLINE | ID: mdl-14680558

RESUMEN

OBJECTIVE: To express human osteoprotegerin (OPG) in E. Coli and analyze its bioactivity in vitro. METHODS: Synthetic oligonucleotides were used to amplify human OPG gene by RT-PCR from total RNA of human osteosarcoma cell line MG63. The OPG cDNA coding for 380 amino acid residues was inserted into prokaryotic expression vector pRSET-A, transformed into competent E. Coli BL21, and induced by 0.1 mmol/l IPTG. SDS-PAGE and Western blot were performed to identify OPG-6His fusion protein. After purified by affinity chromatography, 1,000 microg/L or 1,500 microg/L of OPG-6His were added into the mouse bone marrow cells culture medium. The number of tartrate-resistant acid phophatase (TRAP)-positive multinucleated cells and resorption pits were counted to assess the bioactivity of expression products. RESULTS: The sequence of OPG mature peptide encoding cDNA obtained in this experiment was as same as reported. SDS-PAGE showed 24% of total bacterial protein was of OPG-6His fusion protein. Western blot assay demonstrated that the molecular weight of recombinant protein was about 46 KD and could react specifically with human anti-OPG antibody. The mouse bone marrow cells were induced by 1alpha, 25-dihydroxyvitaminD3 (10(-8) mol/L) and Dexamethasone (10(-7) mol/L) to form osteoclastic-like multinucleated cells. 1,500 microg/L of purified OPG-6His protein could decrease the number of resorption pits and TRAP-positive multinucleated cells in vitro (P < 0.05), but it didn't show the same effects when the concentration of OPG-6His fusion protein was of 1,000 microg/L. CONCLUSIONS: Human OPG-6His fusion protein is expressed and purified in E. Coli. The expression products have moderate inhibitory effects on osteoclast differentiation and bone resorption in vitro only when excessive amount of proteins are added into the culture medium, indicating that prokaryotic expression of fuctionalal OPG protein awaits further investigation.


Asunto(s)
Escherichia coli/genética , Glicoproteínas/genética , Receptores Citoplasmáticos y Nucleares/genética , Proteínas Recombinantes de Fusión/biosíntesis , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Clonación Molecular , Glicoproteínas/biosíntesis , Humanos , Osteoclastos/efectos de los fármacos , Osteoclastos/fisiología , Osteoprotegerina , Receptores Citoplasmáticos y Nucleares/biosíntesis , Receptores del Factor de Necrosis Tumoral , Proteínas Recombinantes de Fusión/farmacología
20.
Mol Cell Biol ; 32(2): 385-98, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22083952

RESUMEN

The transcriptional control circuitry in eukaryotic cells is complex and is orchestrated by combinatorially acting transcription factors. Forkhead transcription factors often function in concert with heterotypic transcription factors to specify distinct transcriptional programs. Here, we demonstrate that FOXK2 participates in combinatorial transcriptional control with the AP-1 transcription factor. FOXK2 binding regions are widespread throughout the genome and are often coassociated with AP-1 binding motifs. FOXK2 acts to promote AP-1-dependent gene expression changes in response to activation of the AP-1 pathway. In this context, FOXK2 is required for the efficient recruitment of AP-1 to chromatin. Thus, we have uncovered an important new molecular mechanism that controls AP-1-dependent gene expression.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Factor de Transcripción AP-1/genética , Activación Transcripcional , Secuencia de Bases , Sitios de Unión , Línea Celular , Cromatina/genética , Cromatina/metabolismo , Factores de Transcripción Forkhead/genética , Genoma Humano , Humanos , Unión Proteica , ARN Interferente Pequeño/genética , Factor de Transcripción AP-1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA