Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Exp Clin Cancer Res ; 42(1): 301, 2023 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-37957685

RESUMO

BACKGROUND: Inherited defects in the base-excision repair gene MBD4 predispose individuals to adenomatous polyposis and colorectal cancer, which is characterized by an accumulation of C > T transitions resulting from spontaneous deamination of 5'-methylcytosine. METHODS: Here, we have investigated the potential role of MBD4 in regulating DNA methylation levels using genome-wide transcriptome and methylome analyses. Additionally, we have elucidated its function through a series of in vitro experiments. RESULTS: Here we show that the protein MBD4 is required for DNA methylation maintenance and G/T mismatch repair. Transcriptome and methylome analyses reveal a genome-wide hypomethylation of promoters, gene bodies and repetitive elements in the absence of MBD4 in vivo. Methylation mark loss is accompanied by a broad transcriptional derepression phenotype affecting promoters and retroelements with low methylated CpG density. MBD4 in vivo forms a complex with the mismatch repair proteins (MMR), which exhibits high bi-functional glycosylase/AP-lyase endonuclease specific activity towards methylated DNA substrates containing a G/T mismatch. Experiments using recombinant proteins reveal that the association of MBD4 with the MMR protein MLH1 is required for this activity. CONCLUSIONS: Our data identify MBD4 as an enzyme specifically designed to repair deaminated 5-methylcytosines and underscores its critical role in safeguarding against methylation damage. Furthermore, it illustrates how MBD4 functions in normal and pathological conditions.


Assuntos
Reparo do DNA , Retroelementos , Humanos , Reparo de Erro de Pareamento de DNA , Proteínas Recombinantes/genética , Metilação de DNA , Endodesoxirribonucleases/genética , Endodesoxirribonucleases/metabolismo
2.
Science ; 372(6549)2021 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-34324427

RESUMO

The Rett syndrome protein MeCP2 was described as a methyl-CpG-binding protein, but its exact function remains unknown. Here we show that mouse MeCP2 is a microsatellite binding protein that specifically recognizes hydroxymethylated CA repeats. Depletion of MeCP2 alters chromatin organization of CA repeats and lamina-associated domains and results in nucleosome accumulation on CA repeats and genome-wide transcriptional dysregulation. The structure of MeCP2 in complex with a hydroxymethylated CA repeat reveals a characteristic DNA shape, with considerably modified geometry at the 5-hydroxymethylcytosine, which is recognized specifically by Arg133, a key residue whose mutation causes Rett syndrome. Our work identifies MeCP2 as a microsatellite DNA binding protein that targets the 5hmC-modified CA-rich strand and maintains genome regions nucleosome-free, suggesting a role for MeCP2 dysfunction in Rett syndrome.


Assuntos
Repetições de Dinucleotídeos , Proteína 2 de Ligação a Metil-CpG/metabolismo , Repetições de Microssatélites , Nucleossomos/metabolismo , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/química , 5-Metilcitosina/metabolismo , Animais , Células Cultivadas , Cromatina/química , Cromatina/metabolismo , Cromatina/ultraestrutura , Citosina/química , Citosina/metabolismo , Metilação de DNA , Células-Tronco Embrionárias/metabolismo , Fibroblastos , Lobo Frontal/metabolismo , Proteína 2 de Ligação a Metil-CpG/química , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Neurônios/metabolismo , Conformação de Ácido Nucleico , Oxirredução , Ligação Proteica , Domínios Proteicos , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Transcrição Gênica
3.
J Mol Biol ; 433(6): 166659, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33010306

RESUMO

Epigenetic modifications and nucleosome positioning play an important role in modulating gene expression. However, how the patterns of epigenetic modifications and nucleosome positioning are established around promoters is not well understood. Here, we have addressed these questions in a series of genome-wide experiments coupled to a novel bioinformatic analysis approach. Our data reveal a clear correlation between CpG density, promoter activity and accumulation of active or repressive histone marks. CGI boundaries define the chromatin promoter regions that will be epigenetically modified. CpG-rich promoters are targeted by histone modifications and histone variants, while CpG-poor promoters are regulated by DNA methylation. CGIs boundaries, but not transcriptional activity, are essential determinants of H2A.Z positioning in vicinity of the promoters, suggesting that the presence of H2A.Z is not related to transcriptional control. Accordingly, H2A.Z depletion has no impact on gene expression of arrested mouse embryonic fibroblasts. Therefore, the underlying DNA sequence, the promoter CpG density and, to a lesser extent, transcriptional activity, are key factors implicated in promoter chromatin architecture.


Assuntos
Ilhas de CpG , Epigênese Genética , Epigenoma , Histonas/genética , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional , Animais , Cromatina/metabolismo , Cromatina/ultraestrutura , Montagem e Desmontagem da Cromatina , Biologia Computacional/métodos , Metilação de DNA , Embrião de Mamíferos , Fibroblastos/citologia , Fibroblastos/metabolismo , Histonas/química , Histonas/deficiência , Histonas/metabolismo , Camundongos , Camundongos Knockout , Cultura Primária de Células , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
4.
Nat Commun ; 10(1): 4436, 2019 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-31570711

RESUMO

CENP-A is an essential histone H3 variant that epigenetically marks the centromeric region of chromosomes. Here we show that CENP-A nucleosomes form characteristic clusters during the G1 phase of the cell cycle. 2D and 3D super-resolution microscopy and segmentation analysis reveal that these clusters encompass a globular rosette-like structure, which evolves into a more compact structure in late G1. The rosette-like clusters contain numerous CENP-A molecules and form a large cellular structure of ∼250-300 nm diameter with remarkably similar shapes for each centromere. Co-localization analysis shows that HJURP, the CENP-A chaperone, is located in the center of the rosette and serves as a nucleation point. The discovery of an HJURP-mediated CENP-A nucleation in human cells and its structural description provide important insights into the mechanism of CENP-A deposition and the organization of CENP-A chromatin in the centromeric region.


Assuntos
Proteína Centromérica A/metabolismo , Proteína Centromérica A/ultraestrutura , Proteínas de Ligação a DNA/metabolismo , Fase G1/fisiologia , Nucleossomos/metabolismo , Animais , Ciclo Celular/fisiologia , Linhagem Celular , Centrômero/metabolismo , Centrômero/ultraestrutura , Cromatina , Montagem e Desmontagem da Cromatina/fisiologia , Proteínas de Ligação a DNA/química , Epigenômica , Células HeLa , Humanos , Imageamento Tridimensional , Camundongos , Camundongos Endogâmicos C57BL/embriologia , Chaperonas Moleculares/química , Nucleossomos/ultraestrutura , Imagem Óptica
5.
Genome Res ; 27(6): 934-946, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28348165

RESUMO

DNA methylation is an essential epigenetic modification, present in both unique DNA sequences and repetitive elements, but its exact function in repetitive elements remains obscure. Here, we describe the genome-wide comparative analysis of the 5mC, 5hmC, 5fC, and 5caC profiles of repetitive elements in mouse embryonic fibroblasts and mouse embryonic stem cells. We provide evidence for distinct and highly specific DNA methylation/oxidation patterns of the repetitive elements in both cell types, which mainly affect CA repeats and evolutionarily conserved mouse-specific transposable elements including IAP-LTRs, SINEs B1m/B2m, and L1Md-LINEs. DNA methylation controls the expression of these retroelements, which are clustered at specific locations in the mouse genome. We show that TDG is implicated in the regulation of their unique DNA methylation/oxidation signatures and their dynamics. Our data suggest the existence of a novel epigenetic code for the most recently acquired evolutionarily conserved repeats that could play a major role in cell differentiation.


Assuntos
Metilação de DNA , Epigênese Genética , Fibroblastos/metabolismo , Genoma , Células-Tronco Embrionárias Murinas/metabolismo , Sequências Repetitivas de Ácido Nucleico , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Animais , Diferenciação Celular , Citosina/análogos & derivados , Citosina/metabolismo , Elementos de DNA Transponíveis , Fibroblastos/citologia , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Cultura Primária de Células , Timina DNA Glicosilase/genética , Timina DNA Glicosilase/metabolismo
6.
Nat Struct Mol Biol ; 23(4): 309-16, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26974126

RESUMO

H2A.Z, a widely conserved histone variant, is evicted from chromatin by the histone chaperone ANP32E. However, to date, no deposition chaperone for H2A.Z is known in metazoans. Here, we identify YL1 as a specific H2A.Z-deposition chaperone. The 2.7-Å-resolution crystal structure of the human YL1-H2A.Z-H2B complex shows that YL1 binding, similarly to ANP32E binding, triggers an extension of the H2A.Z αC helix. The interaction with YL1 is, however, more extensive and includes both the extended acidic patch and the entire DNA-binding surface of H2A.Z-H2B. Substitution of only four amino acid residues of H2A is sufficient for the formation of an H2A.Z-like interface specifically recognized by YL1. Collectively, our data reveal the molecular basis of H2A.Z-specific recognition by YL1 and shed light on the mechanism of H2A.Z transfer to the nucleosome by the ATP-dependent chromatin-remodeling complexes SRCAP and P400-TIP60.


Assuntos
Chaperonas de Histonas/química , Chaperonas de Histonas/metabolismo , Histonas/metabolismo , Proteínas Repressoras/química , Proteínas Repressoras/metabolismo , Sequência de Aminoácidos , Cristalografia por Raios X , Células HeLa , Histonas/química , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mapas de Interação de Proteínas , Estrutura Secundária de Proteína
7.
Nature ; 505(7485): 648-53, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-24463511

RESUMO

H2A.Z is an essential histone variant implicated in the regulation of key nuclear events. However, the metazoan chaperones responsible for H2A.Z deposition and its removal from chromatin remain unknown. Here we report the identification and characterization of the human protein ANP32E as a specific H2A.Z chaperone. We show that ANP32E is a member of the presumed H2A.Z histone-exchange complex p400/TIP60. ANP32E interacts with a short region of the docking domain of H2A.Z through a new motif termed H2A.Z interacting domain (ZID). The 1.48 Å resolution crystal structure of the complex formed between the ANP32E-ZID and the H2A.Z/H2B dimer and biochemical data support an underlying molecular mechanism for H2A.Z/H2B eviction from the nucleosome and its stabilization by ANP32E through a specific extension of the H2A.Z carboxy-terminal α-helix. Finally, analysis of H2A.Z localization in ANP32E(-/-) cells by chromatin immunoprecipitation followed by sequencing shows genome-wide enrichment, redistribution and accumulation of H2A.Z at specific chromatin control regions, in particular at enhancers and insulators.


Assuntos
Cromatina/química , Cromatina/metabolismo , Histonas/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Núcleo Celular/química , Núcleo Celular/metabolismo , Cromatina/genética , Imunoprecipitação da Cromatina , Cristalografia por Raios X , DNA/genética , DNA/metabolismo , Genoma Humano/genética , Histonas/química , Histonas/isolamento & purificação , Humanos , Modelos Moleculares , Chaperonas Moleculares/química , Dados de Sequência Molecular , Proteínas Nucleares/química , Nucleossomos/química , Nucleossomos/metabolismo , Fosfoproteínas/química , Ligação Proteica , Conformação Proteica , Especificidade por Substrato
8.
J Cell Sci ; 126(Pt 23): 5500-12, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-24105263

RESUMO

Inter-organelle membrane contacts sites (MCSs) are specific subcellular regions favoring the exchange of metabolites and information. We investigated the potential role of the late-endosomal membrane-anchored proteins StAR related lipid transfer domain-3 (STARD3) and STARD3 N-terminal like (STARD3NL) in the formation of MCSs involving late-endosomes (LEs). We demonstrate that both STARD3 and STARD3NL create MCSs between LEs and the endoplasmic reticulum (ER). STARD3 and STARD3NL use a conserved two phenylalanines in an acidic tract (FFAT)-motif to interact with ER-anchored VAP proteins. Together, they form an LE-ER tethering complex allowing heterologous membrane apposition. This LE-ER tethering complex affects organelle dynamics by altering the formation of endosomal tubules. An in situ proximity ligation assay between STARD3, STARD3NL and VAP proteins identified endogenous LE-ER MCS. Thus, we report here the identification of proteins involved in inter-organellar interaction.


Assuntos
Proteínas de Transporte/metabolismo , Retículo Endoplasmático/metabolismo , Endossomos/metabolismo , Membranas Intracelulares/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Motivos de Aminoácidos , Animais , Transporte Biológico , Proteínas de Transporte/genética , Retículo Endoplasmático/ultraestrutura , Endossomos/ultraestrutura , Regulação da Expressão Gênica , Células HeLa , Humanos , Membranas Intracelulares/ultraestrutura , Proteínas de Membrana/genética , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Proteínas de Transporte Vesicular/genética
9.
Int J Cancer ; 127(6): 1347-55, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20209494

RESUMO

MMP11 expression is a poor prognosis factor in human carcinomas. Although it has been shown to favor primary tumor development, its role in metastatic processes remains unclear. We studied the hematogenous metastatic activity of C26 mouse colon cancer cells injected into the tail vain of wild-type or MMP11-deficient mice during 2 months. Using X-ray computed tomography to image metastasis development in recipient living mice, lung metastases were found to occur earlier and to grow faster in wild-type mice. Histological analyses of the lung, liver, kidney, adrenal gland, mammary gland, ovary and salivary gland, performed at the end of experiment, also showed lower numbers of metastases in wild-type mice, regardless of organ. Lung metastases showed similar Factor VIII-positive vascular networks regardless of the mouse MMP11 status. However, those found in MMP11-deficient mice also exhibited vessel-like structures that did not express Factor VIII, Lyve-1 and vimentin, and were not stained with PAS. Consequently, they did not correspond to vascular or lymphatic vessels or to vascular mimicry channels. Collectively, these results revealed significant spatio-temporal variability that is dependent on host MMP11 status. Furthermore, they point-out the paradoxical role of MMP11 in favoring the onset and growth of lung metastases but limiting lung foci number, and inhibiting the cancer cell dissemination to other organs. These data highlight the complexity of the metastatic process in which the same factor can play activator or repressor functions depending on the metastatic step.


Assuntos
Metaloproteinase 11 da Matriz/metabolismo , Metástase Neoplásica , Animais , Camundongos , Camundongos Endogâmicos BALB C , Tomografia Computadorizada por Raios X
10.
PLoS One ; 3(10): e3518, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18953416

RESUMO

BACKGROUND: Despite numerous in vivo evidences that Tumor Necrosis Factor Receptor-Associated Factor 4 (TRAF4) plays a key biological function, how it works at the cellular and molecular level remains elusive. METHODOLOGY/PRINCIPAL FINDINGS: In the present study, we show using immunofluorescence and immuohistochemistry that TRAF4 is a novel player at the tight junctions (TJs). TRAF4 is connected to assembled TJs in confluent epithelial cells, but accumulates in the cytoplasm and/or nucleus when TJs are open in isolated cells or EGTA-treated confluent cells. In vivo, TRAF4 is consistently found at TJs in normal human mammary epithelia as well as in well-differentiated in situ carcinomas. In contrast, TRAF4 is never localized at the plasma membrane of poorly-differentiated invasive carcinomas devoid of correct TJs, but is observed in the cytoplasm and/or nucleus of the cancer cells. Moreover, TRAF4 TJ subcellular localization is remarkably dynamic. Fluorescence recovery after photobleaching (FRAP) experiments show that TRAF4 is highly mobile and shuttles between TJs and the cytoplasm. Finally, we show that intracellular TRAF4 potentiates ERK1/2 phosphorylation in proliferating HeLa cells, an epithelial cell line known to be devoid of TJs. CONCLUSIONS/SIGNIFICANCE: Collectively, our data strongly support the new concept of TJs as a dynamic structure. Moreover, our results implicate TRAF4 in one of the emerging TJ-dependent signaling pathways that responds to cell polarity by regulating the cell proliferation/differentiation balance, and subsequently epithelium homeostasis. Drastic phenotypes or lethality in TRAF4-deficient mice and drosophila strongly argue in favor of such a function.


Assuntos
Homeostase , Glândulas Mamárias Humanas/metabolismo , Fator 4 Associado a Receptor de TNF/fisiologia , Junções Íntimas/metabolismo , Animais , Comunicação Celular/fisiologia , Diferenciação Celular/fisiologia , Linhagem Celular Transformada , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Polaridade Celular/fisiologia , Proliferação de Células , Células Cultivadas , Cães , Células HeLa , Homeostase/fisiologia , Humanos , Glândulas Mamárias Humanas/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Modelos Biológicos , Transporte Proteico/fisiologia , Transdução de Sinais/fisiologia , Fator 4 Associado a Receptor de TNF/metabolismo
11.
Cancer Res ; 65(23): 10862-71, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16322233

RESUMO

The initial invasive processes during cancer development remain largely unknown. Stromelysin-3/matrix metalloproteinase 11 (ST3/MMP11) is associated with tumor invasion and poor prognosis. We present novel evidence that adipocytes present at human breast tumor invasive front are induced by cancer cells to express ST3. Using mouse syngeneic model, light and electron microscopy showed that in ST3-deficient mice but not in wild-type mice, forced cancer cell-adipocyte interaction/crosstalk results in adipocyte membrane alteration, allowing cancer cell fat infiltration and death. Thus, adipocytes are involved in initial cancer cell survival into connective tissue, and this effect is ST3 mediated. This suggested that ST3 might play a role in adipocyte metabolism. Accordingly, ST3-deficient mice exhibited fat excess and increased mRNA levels of peroxisome proliferator-activated receptor gamma (PPARgamma) and adipocyte protein 2 (aP2) adipogenic markers, indicating that, in vivo, ST3 negatively regulates fat homeostasis. Moreover, ST3-deficient mouse embryonic fibroblasts exhibited a dramatic enhanced potential to differentiate into adipocytes associated with increased PPARgamma and aP2 expression, and recombinant ST3 treatment reverted their differentiation. Thus, in vitro, ST3 reduces adipocyte differentiation in an autocrine manner. High fibroblasts/adipocytes ratio is a stroma feature, and peritumoral fibroblast origin remains debated. Our results support the concept that invading cancer cells aberrantly restore the negative ST3 function on adipogenesis into proximal adipocytes/preadipocytes, leading to the accumulation/maintenance of a particular peritumoral fibroblast subpopulation. Accordingly, in human breast tumors, we observed that ST3-expressing peritumoral fibroblasts are distinct from alpha-smooth muscle actin-expressing myofibroblasts. This constitutes the first report of implication of a MMP in cancer cell-adipocyte interaction/crosstalk during early steps of connective tissue invasion.


Assuntos
Adipócitos/citologia , Adipogenia/fisiologia , Neoplasias da Mama/patologia , Comunicação Celular/fisiologia , Metaloendopeptidases/fisiologia , Adipócitos/metabolismo , Animais , Neoplasias da Mama/metabolismo , Diferenciação Celular/fisiologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Embrião de Mamíferos , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Masculino , Metaloproteinase 11 da Matriz , Metaloendopeptidases/biossíntese , Metaloendopeptidases/deficiência , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica
12.
J Biol Chem ; 279(32): 33702-15, 2004 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-15166247

RESUMO

MLN51 is a nucleocytoplasmic shuttling protein that is overexpressed in breast cancer. The function of MLN51 in mammals remains elusive. Its fly homolog, named barentsz, as well as the proteins mago nashi and tsunagi have been shown to be required for proper oskar mRNA localization to the posterior pole of the oocyte. Magoh and Y14, the human homologs of mago nashi and tsunagi, are core components of the exon junction complex (EJC). The EJC is assembled on spliced mRNAs and plays important roles in post-splicing events including mRNA export, nonsense-mediated mRNA decay, and translation. In the present study, we show that human MLN51 is an RNA-binding protein present in ribonucleo-protein complexes. By co-immunoprecipitation assays, endogenous MLN51 protein is found to be associated with EJC components, including Magoh, Y14, and NFX1/TAP, and subcellular localization studies indicate that MLN51 transiently co-localizes with Magoh in nuclear speckles. Moreover, we demonstrate that MLN51 specifically associates with spliced mRNAs in co-precipitation experiments, both in the nucleus and in the cytoplasm, at the position where the EJC is deposited. Most interesting, we have identified a region within MLN51 sufficient to bind RNA, to interact with Magoh and spliced mRNA, and to address the protein to nuclear speckles. This conserved region of MLN51 was therefore named SELOR for speckle localizer and RNA binding module. Altogether our data demonstrate that MLN51 associates with EJC in the nucleus and remains stably associated with mRNA in the cytoplasm, suggesting that its overexpression might alter mRNA metabolism in cancer.


Assuntos
Núcleo Celular/metabolismo , Éxons , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , RNA/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Núcleo Celular/química , Núcleo Celular/ultraestrutura , Sequência Conservada , Citoplasma/química , Escherichia coli/genética , Peixes , Imunofluorescência , Células HeLa , Humanos , Técnicas de Imunoadsorção , Camundongos , Dados de Sequência Molecular , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Splicing de RNA , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA , Proteínas Recombinantes de Fusão , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonucleoproteínas/química , Ribonucleoproteínas/metabolismo , Alinhamento de Sequência , Transfecção , Xenopus , Peixe-Zebra
13.
Cancer Res ; 63(18): 5844-9, 2003 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-14522908

RESUMO

In human carcinomas, stromelysin-3/matrix metalloproteinase 11 (ST3, MMP11) expression by nonmalignant fibroblastic cells located in the immediate vicinity of cancer cells is a bad prognostic factor. Using mouse models of primary tumors, it has been demonstrated that ST3 is a key player during local invasion, favoring cancer cell survival in connective tissue through an antiapoptotic function. To investigate the ST3 impact on additional phases of cancer cell invasion, we developed mammary gland cancer prone MMTV-ras transgenic mice in wild-type (ras+/+;ST3+/+) or ST3-deficient (ras+/+;ST3-/-) genotype and studied their whole natural cancer history. The tumor-free survival and delay between the first ras oncogenic hit and primary tumor appearance increased in ras+/+;ST3-/- mice (P < 0.000001 and <0.0000007, respectively). A systematic search for occult primary tumors and metastases revealed, in addition to a lower total number and size of primary tumors (P < 0.02), an unexpected higher number of metastases (P < 0.01) in ras+/+;ST3-/- mice. Moreover, for a similar number and size of primary invasive tumors, ras+/+;ST3-/- mice developed more metastases, indicating that the cancer cells evolving in ST3-deficient stroma have an increased potential to hematogenous dissemination. We conclude that the ST3 microenvironment is a consistently active partner of invading cancer cells but that its function differs throughout cancer progression, being tumor enhancer or repressor in processes leading to local or distal invasion. Such a dual effect for an MMP might shed light, at least partially, for the absence of survival benefit for patients included in anti-MMP clinical trials.


Assuntos
Transformação Celular Viral/fisiologia , Genes ras/fisiologia , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/patologia , Vírus do Tumor Mamário do Camundongo/fisiologia , Metaloendopeptidases/fisiologia , Animais , Feminino , Neoplasias Hepáticas Experimentais/enzimologia , Neoplasias Hepáticas Experimentais/secundário , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/secundário , Masculino , Neoplasias Mamárias Experimentais/virologia , Metaloproteinase 11 da Matriz , Metaloendopeptidases/deficiência , Camundongos , Camundongos Transgênicos
14.
Oncogene ; 22(24): 3770-80, 2003 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-12802284

RESUMO

MLN64, is invariably coamplified and coexpressed with erbB-2 in breast cancers. The human MLN64 and ERBB2 genes are positioned at less than 50 kb from each other, on chromosome 17q12. To understand the molecular basis of MLN64 overexpression in cancer, the genomic region containing the MLN64 and ERBB2 genes was isolated and mapped. The two genes, DARPP32 and Telethonin, flanking MLN64 respectively on its centromeric and telomeric sides, although coamplified, are not overexpressed in breast cancer cells, indicating that gene amplification is not sufficient to allow overexpression. The MLN64 minimal promoter was isolated and found to be a housekeeping gene promoter containing four potential Sp1 binding elements. Using Sp1-deficient Drosophila SL2 cells, MLN64 promoter activity was induced in a dose-dependent manner by exogenous Sp1 addition. Furthermore, mutation of each individual Sp1 element resulted in a significant decrease in reporter gene activity, indicating that all the Sp1 binding elements are functional and act together to promote gene expression. Since the ERBB2 promoter is also positively regulated by Sp1, this study indicates that MLN64 and ERBB2 genes share common transcriptional controls together with a physical link on chromosome 17q. We speculate that, in addition to the oncogenic potential of erbB-2 overexpression, the unbalanced action of MLN64 contributes to the poor clinical outcome of breast tumors bearing this amplified region.


Assuntos
Neoplasias da Mama/genética , Proteínas de Transporte , Regulação Neoplásica da Expressão Gênica , Proteínas de Membrana/genética , Fator de Transcrição Sp1/fisiologia , Sequência de Aminoácidos , Sequência de Bases , Sítios de Ligação , Proteína beta Intensificadora de Ligação a CCAAT/fisiologia , Linhagem Celular , Sequência Conservada , Feminino , Amplificação de Genes , Genes erbB-2 , Humanos , Proteínas de Membrana/química , Dados de Sequência Molecular , Regiões Promotoras Genéticas
15.
J Biol Chem ; 277(28): 25527-36, 2002 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-12006591

RESUMO

Human stromelysin 3 (ST3) is a matrix metalloproteinase (MMP) that has been implicated in cancer progression and in various tissue remodeling processes. Unlike most MMPs, ST3 is characterized by a distinct substrate specificity and a specific regulation and is not directly involved in extracellular matrix degradation. In the present study, we have identified an additional ST3 gene promoter that is accessible to nuclear factors such as C/EBP and retinoic acid receptors. This human specific promoter is inducible and controls the expression of a novel ST3 transcript called the beta-ST3 that is expressed in cultured cells and in placenta. This transcript encodes a 40-kDa ST3 isoform that lacks both the signal peptide common to all secreted MMPs and the prodomain that normally maintains enzyme latency. Consistent with the lack of a signal peptide, the beta-ST3 was found to be intracellular. The relative amount of the extracellular alpha-ST3 isoform was about 20-fold higher than that of the intracellular ST3 isoforms, as estimated by Western blot analysis. Furthermore, recombinant beta-ST3 produced in Escherichia coli exhibits a proteolytic activity against alpha1-proteinase inhibitor, a substrate previously shown to be inactivated by the alpha-ST3. Therefore, although it was thought that all MMPs were synthesized as inactive zymogens and functioned extracellularly, this is the first MMP isoform reported that is generated by alternative promoter usage and directly translated as an active enzyme. Although the intracellular function of the beta-ST3 remains to be investigated, these data support the idea that the functions of MMPs are not restricted to the extracellular space.


Assuntos
Processamento Alternativo , Isoenzimas/genética , Metaloendopeptidases/genética , Regiões Promotoras Genéticas , Biossíntese de Proteínas , Sequência de Aminoácidos , Sequência de Bases , DNA Complementar , Humanos , Metaloproteinase 11 da Matriz , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Células Tumorais Cultivadas
16.
J Cell Biol ; 157(5): 761-70, 2002 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-12034770

RESUMO

Trefoil factor (TFF)1 is synthesized and secreted by the normal stomach mucosa and by the gastrointestinal cells of injured tissues. The link between mouse TFF1 inactivation and the fully penetrant antropyloric tumor phenotype prompted the classification of TFF1 as a gastric tumor suppressor gene. Accordingly, altered expression, deletion, and/or mutations of the TFF1 gene are frequently observed in human gastric carcinomas. The present study was undertaken to address the nature of the cellular and molecular mechanisms targeted by TFF1 signalling. TFF1 effects were investigated in IEC18, HCT116, and AGS gastrointestinal cells treated with recombinant human TFF1, and in stably transfected HCT116 cells synthesizing constitutive or doxycycline-induced human TFF1. We observed that TFF1 triggers two types of cellular responses. On one hand, TFF1 lowers cell proliferation by delaying G1-S cell phase transition. This results from a TFF1-mediated increase in the levels of cyclin-dependent kinase inhibitors of both the INK4 and CIP subfamilies, leading to lower E2F transcriptional activity. On the other hand, TFF1 protects cells from chemical-, anchorage-free-, or Bad-induced apoptosis. In this process, TFF1 signalling targets the active form of caspase-9. Together, these results provide the first evidence of a dual antiproliferative and antiapoptotic role for TFF1. Similar paradoxical functions have been reported for tumor suppressor genes involved in cell differentiation, a function consistent with TFF1.


Assuntos
Apoptose/fisiologia , Proteínas de Ligação a DNA , Fase G1/fisiologia , Mucosa Intestinal/citologia , Proteínas/metabolismo , Fase S/fisiologia , Animais , Caspase 3 , Caspase 6 , Caspase 8 , Caspase 9 , Caspases/metabolismo , Contagem de Células , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/fisiologia , Neoplasias do Colo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p27 , Fatores de Transcrição E2F , Precursores Enzimáticos/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Células HeLa , Humanos , Mucosa Intestinal/metabolismo , Células Jurkat , Proteínas/genética , Ratos , Proteínas Recombinantes/farmacologia , Proteína do Retinoblastoma/metabolismo , Neoplasias Gástricas , Fatores de Transcrição/metabolismo , Transfecção , Fator Trefoil-1 , Proteínas Supressoras de Tumor/metabolismo
17.
Proc Natl Acad Sci U S A ; 99(8): 5585-90, 2002 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-11943846

RESUMO

TRAF4 belongs to the tumor necrosis factor receptor-associated factor (TRAF) family of proteins but, unlike other family members, has not yet been clearly associated to any specific receptor or signaling pathway. To investigate the biological function of TRAF4, we have generated traf4-deficient mice by gene disruption. The traf4 gene mutation is embryonic lethal but with great individual variation, as approximately one third of the homozygous mutant embryos died in utero around embryonic day 14, whereas the others reach adulthood. Surviving mutant mice manifest numerous developmental abnormalities; notably, 100% of homozygous mutant mice suffer respiratory disorder and wheezing caused by tracheal ring disruption. Additional malformations concern mainly the axial skeleton, as the ribs, sternum, tail, and vertebral arches are affected, with various degrees of penetrance. Traf4-deficient mice also exhibit a high incidence of spina bifida, a defect likened to neural tube defects (NTD) that are common congenital malformations in humans. Altogether, our results demonstrate that TRAF4 is required during embryogenesis in key biological processes including the formation of the trachea, the development of the axial skeleton, and the closure of the neural tube. Considering the normal expression pattern of TRAF4 in neural tissues, we can conclude that TRAF4 participates in neurulation in vivo.


Assuntos
Osso e Ossos/anormalidades , Crista Neural/anormalidades , Crista Neural/embriologia , Proteínas/genética , Proteínas/fisiologia , Traqueia/anormalidades , Animais , Western Blotting , DNA Complementar/metabolismo , Éxons , Homozigoto , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Genéticos , Mutagênese Sítio-Dirigida , Neurônios/citologia , Neurônios/metabolismo , Reação em Cadeia da Polimerase , Disrafismo Espinal/genética , Fator 4 Associado a Receptor de TNF , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...