Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Genes Chromosomes Cancer ; 60(5): 303-313, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-32734664

RESUMO

In vitro assays for clustered DNA lesions will facilitate the analysis of the mechanisms underlying complex genome rearrangements such as chromothripsis, including the recruitment of repair factors to sites of DNA double-strand breaks (DSBs). We present a novel method generating localized DNA DSBs using UV irradiation with photomasks. The size of the damage foci and the spacing between lesions are fully adjustable, making the assay suitable for different cell types and targeted areas. We validated this setup with genomically stable epithelial cells, normal fibroblasts, pluripotent stem cells, and patient-derived primary cultures. Our method does not require a specialized device such as a laser, making it accessible to a broad range of users. Sensitization by 5-bromo-2-deoxyuridine incorporation is not required, which enables analyzing the DNA damage response in post-mitotic cells. Irradiated cells can be cultivated further, followed by time-lapse imaging or used for downstream biochemical analyses, thanks to the high throughput of the system. Importantly, we showed genome rearrangements in the irradiated cells, providing a proof of principle for the induction of structural variants by localized DNA lesions.


Assuntos
Quebras de DNA de Cadeia Dupla , Mutagênese , Linhagem Celular , Células Cultivadas , Células Epiteliais/metabolismo , Células Epiteliais/efeitos da radiação , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Humanos , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/efeitos da radiação , Raios Ultravioleta
2.
Nucleic Acids Res ; 46(16): 8299-8310, 2018 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-29986092

RESUMO

Mammalian DNA replication is a highly organized and regulated process. Large, Mb-sized regions are replicated at defined times along S-phase. Replication Timing (RT) is thought to play a role in shaping the mammalian genome by affecting mutation rates. Previous analyses relied on somatic RT profiles. However, only germline mutations are passed on to offspring and affect genomic composition. Therefore, germ cell RT information is necessary to evaluate the influences of RT on the mammalian genome. We adapted the RT mapping technique for limited amounts of cells, and measured RT from two stages in the mouse germline - primordial germ cells (PGCs) and spermatogonial stem cells (SSCs). RT in germline cells exhibited stronger correlations to both mutation rate and recombination hotspots density than those of RT in somatic tissues, emphasizing the importance of using correct tissues-of-origin for RT profiling. Germline RT maps exhibited stronger correlations to additional genetic features including GC-content, transposable elements (SINEs and LINEs), and gene density. GC content stratification and multiple regression analysis revealed independent contributions of RT to SINE, gene, mutation, and recombination hotspot densities. Together, our results establish a central role for RT in shaping multiple levels of mammalian genome composition.


Assuntos
Período de Replicação do DNA/genética , Replicação do DNA/genética , Genoma/genética , Células Germinativas/metabolismo , Células-Tronco/metabolismo , Animais , Composição de Bases/genética , Linhagem Celular Tumoral , Células Cultivadas , Elementos de DNA Transponíveis/genética , Feminino , Células Germinativas/citologia , Mutação em Linhagem Germinativa , Masculino , Mamíferos/genética , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Elementos Nucleotídeos Curtos e Dispersos/genética , Células-Tronco/citologia
3.
Nat Commun ; 15(1): 3270, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38627364

RESUMO

Epigenetic defects caused by hereditary or de novo mutations are implicated in various human diseases. It remains uncertain whether correcting the underlying mutation can reverse these defects in patient cells. Here we show by the analysis of myotonic dystrophy type 1 (DM1)-related locus that in mutant human embryonic stem cells (hESCs), DNA methylation and H3K9me3 enrichments are completely abolished by repeat excision (CTG2000 expansion), whereas in patient myoblasts (CTG2600 expansion), repeat deletion fails to do so. This distinction between undifferentiated and differentiated cells arises during cell differentiation, and can be reversed by reprogramming of gene-edited myoblasts. We demonstrate that abnormal methylation in DM1 is distinctively maintained in the undifferentiated state by the activity of the de novo DNMTs (DNMT3b in tandem with DNMT3a). Overall, the findings highlight a crucial difference in heterochromatin maintenance between undifferentiated (sequence-dependent) and differentiated (sequence-independent) cells, thus underscoring the role of differentiation as a locking mechanism for repressive epigenetic modifications at the DM1 locus.


Assuntos
Distrofia Miotônica , Humanos , Distrofia Miotônica/genética , Heterocromatina/genética , Diferenciação Celular/genética , Metilação de DNA , Epigênese Genética
4.
Nat Commun ; 14(1): 3359, 2023 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-37291192

RESUMO

Human trophoblast stem cells (hTSCs) can be derived from embryonic stem cells (hESCs) or be induced from somatic cells by OCT4, SOX2, KLF4 and MYC (OSKM). Here we explore whether the hTSC state can be induced independently of pluripotency, and what are the mechanisms underlying its acquisition. We identify GATA3, OCT4, KLF4 and MYC (GOKM) as a combination of factors that can generate functional hiTSCs from fibroblasts. Transcriptomic analysis of stable GOKM- and OSKM-hiTSCs reveals 94 hTSC-specific genes that are aberrant specifically in OSKM-derived hiTSCs. Through time-course-RNA-seq analysis, H3K4me2 deposition and chromatin accessibility, we demonstrate that GOKM exert greater chromatin opening activity than OSKM. While GOKM primarily target hTSC-specific loci, OSKM mainly induce the hTSC state via targeting hESC and hTSC shared loci. Finally, we show that GOKM efficiently generate hiTSCs from fibroblasts that harbor knockout for pluripotency genes, further emphasizing that pluripotency is dispensable for hTSC state acquisition.


Assuntos
Reprogramação Celular , Células-Tronco Pluripotentes Induzidas , Humanos , Reprogramação Celular/genética , Trofoblastos , Fibroblastos , Células-Tronco Embrionárias , Cromatina/genética , Fator 3 de Transcrição de Octâmero/genética
5.
Cell Signal ; 97: 110395, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35752351

RESUMO

The potential impact of Vav1 on human cancer was only recently acknowledged, as it is detected as a mutant or an overexpressed gene in various cancers, including lung cancer. Vav1, which is normally and exclusively expressed in the hematopoietic system functions as a specific GDP/GTP nucleotide exchange factor (GEF), strictly regulated by tyrosine phosphorylation. To investigate whether Vav1 plays a causative or facilitating role in-vivo in lung cancer development and to examine whether it co-operates with other oncogenes, such as mutant K-Ras, we generated novel mouse strains that express: Vav1 or K-RasG12D in type II pneumocytes, as well as a transgenic mouse line that expresses both Vav1 and K-RasG12D in these cells. Coexpression of Vav1 and K-RasG12D in the lungs dramatically increased malignant lung cancer lesions, and did so significantly faster than K-RasG12D alone, strongly suggesting that these two oncogenes synergize to enhance lung tumor development. Vav1 expression alone had no apparent effects on lung tumorigenesis. The increase in lung cancer in K-RasG12D/Vav1 mice was accompanied by an increase in B-cell, T-cells, and monocyte infiltration in the tumor microenvironment. Concomitantly, ERK phosphorylation was highly elevated in the lungs of K-RasG12 D/Vav1 mice. Also, several cytokines such as IL-4 and IL-13 which play a significant role in the immune system, were elevated in lungs of Vav1 and K-RasG12 D/Vav1 mice. Our findings emphasize the contribution of Vav1 to lung tumor development through its signaling properties.


Assuntos
Neoplasias Pulmonares , Microambiente Tumoral , Animais , Transformação Celular Neoplásica/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-vav/metabolismo , Transdução de Sinais/genética
6.
Cells ; 11(6)2022 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-35326399

RESUMO

Vav1 is normally and exclusively expressed in the hematopoietic system where it functions as a specific GDP/GTP nucleotide exchange factor (GEF), firmly regulated by tyrosine phosphorylation. Mutations and overexpression of Vav1 in hematopoietic malignancies, and in human cancers of various histologic origins, are well documented. To reveal whether overexpression of Vav1 in different tissues suffices for promoting the development of malignant lesions, we expressed Vav1 in transgenic mice by using the ubiquitous ROSA26 promoter (Rosa Vav1). We detected Vav1 expression in epithelial tissues of various organs including pancreas, liver, and lung. While carcinomas did not develop in these organs, surprisingly, we noticed the development of B-cell lymphomas. Rac1-GTP levels did not change in tissues from Rosa Vav1 mice expressing the transgenic Vav1, while ERK phosphorylation increased in the lymphomas, suggesting that signaling pathways are evoked. One of the growth factors analyzed by us as a suspect candidate to mediate paracrine stimulation in the lymphocytes was CSF-1, which was highly expressed in the epithelial compartment of Rosa Vav1 mice. The expression of its specific receptor, CSF-1R, was found to be highly expressed in the B-cell lymphomas. Taken together, our results suggest a potential cross-talk between epithelial cells expressing Vav1, that secrete CSF-1, and the lymphocytes that express CSF-1R, thus leading to the generation of B-cell lymphomas. Our findings provide a novel mechanism by which Vav1 contributes to tumor propagation.


Assuntos
Linfoma de Células B , Linfoma , Animais , Guanosina Trifosfato , Humanos , Linfoma de Células B/genética , Fator Estimulador de Colônias de Macrófagos/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-vav/genética , Proteínas Proto-Oncogênicas c-vav/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo
7.
Nat Commun ; 13(1): 3475, 2022 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-35715410

RESUMO

Following fertilization, it is only at the 32-64-cell stage when a clear segregation between cells of the inner cell mass and trophectoderm is observed, suggesting a 'T'-shaped model of specification. Here, we examine whether the acquisition of these two states in vitro, by nuclear reprogramming, share similar dynamics/trajectories. Using a comparative parallel multi-omics analysis (i.e., bulk RNA-seq, scRNA-seq, ATAC-seq, ChIP-seq, RRBS and CNVs) on cells undergoing reprogramming to pluripotency and TSC state we show that each reprogramming system exhibits specific trajectories from the onset of the process, suggesting 'V'-shaped model. We describe in detail the various trajectories toward the two states and illuminate reprogramming stage-specific markers, blockers, facilitators and TSC subpopulations. Finally, we show that while the acquisition of the TSC state involves the silencing of embryonic programs by DNA methylation, during the acquisition of pluripotency these regions are initially defined but retain inactive by the elimination of H3K27ac.


Assuntos
Blastocisto , Reprogramação Celular , Blastocisto/metabolismo , Células Cultivadas , Reprogramação Celular/genética , Metilação de DNA
8.
Dev Cell ; 56(13): 1900-1916.e5, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34197726

RESUMO

Stem cells (SCs) play a key role in homeostasis and repair. While many studies have focused on SC self-renewal and differentiation, little is known regarding the molecular mechanism regulating SC elimination and compensation upon loss. Here, we report that Caspase-9 deletion in hair follicle SCs (HFSCs) attenuates the apoptotic cascade, resulting in significant temporal delays. Surprisingly, Casp9-deficient HFSCs accumulate high levels of cleaved caspase-3 and are improperly cleared due to an essential caspase-3/caspase-9 feedforward loop. These SCs are retained in an apoptotic-engaged state, serving as mitogenic signaling centers by continuously releasing Wnt3 and instructing proliferation. Investigating the underlying mechanism, we reveal a caspase-3/Dusp8/p38 module responsible for Wnt3 induction, which operates in both normal and Casp9-deleted HFSCs. Notably, Casp9-deleted mice display accelerated wound repair and de novo hair follicle regeneration. Taken together, we demonstrate that apoptotic cells represent a dynamic SC niche, from which emanating signals drive SC proliferation and tissue regeneration.


Assuntos
Caspase 3/genética , Caspase 9/genética , Fosfatases de Especificidade Dupla/genética , Regeneração/genética , Proteína Wnt3/genética , Animais , Apoptose/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Autorrenovação Celular/genética , Folículo Piloso/crescimento & desenvolvimento , Folículo Piloso/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Nicho de Células-Tronco/genética , Células-Tronco/metabolismo , Cicatrização/genética
9.
Life Sci Alliance ; 3(5)2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32277014

RESUMO

To explore the contribution of Vav1, a hematopoietic signal transducer, to pancreatic ductal adenocarcinoma (PDAC) development, we generated transgenic mouse lines expressing, Vav1, K-RasG12D, or both K-RasG12D and Vav1 in pancreatic acinar cells. Co-expression of Vav1 and K-RasG12D synergistically enhanced acinar-to-ductal metaplasia (ADM) formation, far exceeding the number of lesions developed in K-RasG12D mice. Mice expressing only Vav1 did not develop ADM. Moreover, the incidence of PDAC in K-RasG12D/Vav1 was significantly higher than in K-RasG12D mice. Discontinuing Vav1 expression in K-RasG12D/Vav1 mice elicited a marked regression of malignant lesions in the pancreas, demonstrating Vav1 is required for generation and maintenance of ADM. Rac1-GTP levels in the K-RasG12D/Vav1 mice pancreas clearly demonstrated an increase in Rac1 activity. Treatment of K-RasG12D and K-RasG12D/Vav1 mice with azathioprine, an immune-suppressor drug which inhibits Vav1's activity as a GDP/GTP exchange factor, dramatically reduced the number of malignant lesions. These results suggest that Vav1 plays a role in the development of PDAC when co-expressed with K-RasG12D via its activity as a GEF for Rac1GTPase.


Assuntos
Carcinoma Ductal Pancreático/genética , Proteínas Proto-Oncogênicas c-vav/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Células Acinares/metabolismo , Animais , Carcinoma Ductal Pancreático/metabolismo , Transformação Celular Neoplásica/genética , Genes ras/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pâncreas/metabolismo , Neoplasias Pancreáticas/genética , Lesões Pré-Cancerosas/genética , Proteínas Proto-Oncogênicas c-vav/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Transdução de Sinais/genética , Neoplasias Pancreáticas
10.
Virchows Arch ; 454(1): 71-9, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19015874

RESUMO

Lysyl oxidase-like enzymes (LOXL) are expressed in various cancers. We analyzed the expression of LOXL2, LOXL3, and LOXL4 in cancers involving the serosal cavities-breast carcinoma, ovarian carcinoma, and malignant mesothelioma using reverse-transcriptase polymerase chain reaction. We discovered two new alternative splice variants of LOXL4. The spliced segments were exon 9 (splice variant 1) or both exons 8 and 9 (splice variant 2). In ovarian carcinoma, splice variant 1 was significantly elevated in effusions compared to solid lesions (p < 0.001). Splice variant 2 appeared only in effusions. In breast carcinoma, LOXL4 was expressed only in the effusion samples. In malignant mesothelioma, LOXL4 and its splice variants were expressed at all sites. Breast carcinoma effusions showed significantly higher LOXL2 (p = 0.003) and lower LOXL3 (p < 0.001) expression compared to primary carcinomas. Our data show differences in LOXL messenger RNA expression as a function of anatomic site and tumor type in cancers affecting the serosal cavities.


Assuntos
Processamento Alternativo/genética , Aminoácido Oxirredutases/genética , Aminoácido Oxirredutases/metabolismo , Neoplasias da Mama/metabolismo , Mesotelioma/metabolismo , Neoplasias Ovarianas/metabolismo , RNA Mensageiro/genética , Adenocarcinoma de Células Claras/metabolismo , Adenocarcinoma de Células Claras/patologia , Adulto , Idoso , Neoplasias da Mama/patologia , Carcinoma Endometrioide/metabolismo , Carcinoma Endometrioide/patologia , Éxons/genética , Feminino , Humanos , Mesotelioma/patologia , Pessoa de Meia-Idade , Neoplasias Ovarianas/patologia , Proteína-Lisina 6-Oxidase , RNA Neoplásico/genética , Estudos Retrospectivos
11.
Cell Stem Cell ; 24(6): 983-994.e7, 2019 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-31031139

RESUMO

Following fertilization, totipotent cells undergo asymmetric cell divisions, resulting in three distinct cell types in the late pre-implantation blastocyst: epiblast (Epi), primitive endoderm (PrE), and trophectoderm (TE). Here, we aim to understand whether these three cell types can be induced from fibroblasts by one combination of transcription factors. By utilizing a sophisticated fluorescent knockin reporter system, we identified a combination of five transcription factors, Gata3, Eomes, Tfap2c, Myc, and Esrrb, that can reprogram fibroblasts into induced pluripotent stem cells (iPSCs), induced trophoblast stem cells (iTSCs), and induced extraembryonic endoderm stem cells (iXENs), concomitantly. In-depth transcriptomic, chromatin, and epigenetic analyses provide insights into the molecular mechanisms that underlie the reprogramming process toward the three cell types. Mechanistically, we show that the interplay between Esrrb and Eomes during the reprogramming process determines cell fate, where high levels of Esrrb induce a XEN-like state that drives pluripotency and high levels of Eomes drive trophectodermal fate.


Assuntos
Blastocisto/fisiologia , Endoderma/fisiologia , Fibroblastos/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Trofoblastos/fisiologia , Animais , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Reprogramação Celular , Implantação do Embrião , Camundongos , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/metabolismo
12.
Curr Opin Genet Dev ; 46: 37-43, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28662446

RESUMO

How the first cell fate decision of an embryo occurs is one of the most fascinating biological questions examined over the last few decades, with numerous in vivo models proposed and many factors tested for their role in the process. In this review, we will primarily focus on the mouse model and discuss the role that transcription factors play during establishment and maintenance of the first lineage segregation in the embryo, towards inner cell mass or trophectoderm. We will also overview recent developments in somatic nuclear reprogramming into induced pluripotent stem cells, the inner cell mass (epiblast) equivalent, and into induced trophoblast stem cells, the trophectoderm equivalent, and discuss potential correspondences between the in vivo and in vitro models.


Assuntos
Diferenciação Celular/genética , Reprogramação Celular/genética , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes , Animais , Blastocisto/citologia , Massa Celular Interna do Blastocisto/citologia , Linhagem da Célula/genética , Camadas Germinativas/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos
13.
Trends Cell Biol ; 26(1): 65-75, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26437595

RESUMO

The generation of induced pluripotent stem cells (iPSCs) and directly converted cells holds great promise in regenerative medicine. However, after in-depth studies of the murine system, we know that the current methodologies to produce these cells are not ideal and mostly yield cells of poor quality that might hold a risk in therapeutic applications. In this review we address the duality found in the literature regarding the use of 'quality' as a criterion for the clinic. We discuss the elements that influence reprogramming quality, and provide evidence that safety and functionality are directly linked to cell quality. Finally, because most of the available data come from murine systems, we speculate about what aspects can be applied to human cells.


Assuntos
Reprogramação Celular , Fatores de Transcrição/fisiologia , Animais , Técnicas de Cultura de Células , Células Cultivadas , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Medicina Regenerativa
14.
Cell Stem Cell ; 17(5): 543-56, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26412562

RESUMO

Induced pluripotent stem cells (iPSCs) undergo extensive nuclear reprogramming and are generally indistinguishable from embryonic stem cells (ESCs) in their functional capacity and transcriptome and DNA methylation profiles. However, direct conversion of cells from one lineage to another often yields incompletely reprogrammed, functionally compromised cells, raising the question of whether pluripotency is required to achieve a high degree of nuclear reprogramming. Here, we show that transient expression of Gata3, Eomes, and Tfap2c in mouse fibroblasts induces stable, transgene-independent trophoblast stem-like cells (iTSCs). iTSCs possess transcriptional profiles highly similar to blastocyst-derived TSCs, with comparable methylation and H3K27ac patterns and genome-wide H2A.X deposition. iTSCs generate trophoectodermal lineages upon differentiation, form hemorrhagic lesions, and contribute to developing placentas in chimera assays, indicating a high degree of nuclear reprogramming, with no evidence of passage through a transient pluripotent state. Together, these data demonstrate that extensive nuclear reprogramming can be achieved independently of pluripotency.


Assuntos
Linhagem da Célula , Núcleo Celular/metabolismo , Reprogramação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Trofoblastos/citologia , Animais , Células Cultivadas , Camundongos , Camundongos Transgênicos , Trofoblastos/metabolismo
15.
Oncotarget ; 5(19): 9214-26, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25313137

RESUMO

Vav1 is a signal transducer that functions as a scaffold protein and a regulator of cytoskeleton organization in the hematopoietic system, where it is exclusively expressed. Recently, Vav1 was shown to be involved in diverse human cancers, including lung cancer. We demonstrate that lung cancer cells that abnormally express Vav1 secrete growth factors in a Vav1-dependent manner. Transcriptome analysis demonstrated that Vav1 depletion results in a marked reduction in the expression of colony-stimulating-factor-1 (CSF1), a hematopoietic growth factor. The association between Vav1 expression and CSF1 was further supported by signal transduction experiments, supporting involvement of Vav1 in regulating lung cancer secretome. Blocking of ERK phosphorylation, led to a decrease in CSF1 transcription, thus suggesting a role for ERK, a downstream effector of Vav1, in CSF1 expression. CSF1-silenced cells exhibited reduced focus formation, proliferation abilities, and growth in NOD/SCID mice. CSF1-silenced H358 cells resulted in significantly smaller tumors, showing increased fibrosis and a decrease in tumor infiltrating macrophages. Finally, immunohistochemical analysis of primary human lung tumors revealed a positive correlation between Vav1 and CSF1 expression, which was associated with tumor grade. Additional results presented herein suggest a potential cross-talk between cancer cells and the microenvironment controlled by CSF1/Vav1 signaling pathways.


Assuntos
Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Fator Estimulador de Colônias de Macrófagos/metabolismo , Proteínas Proto-Oncogênicas c-vav/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Perfilação da Expressão Gênica , Humanos , Fator Estimulador de Colônias de Macrófagos/biossíntese , Fator Estimulador de Colônias de Macrófagos/genética , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fosforilação , Proteínas Proto-Oncogênicas c-vav/biossíntese , Proteínas Proto-Oncogênicas c-vav/genética , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais/genética , Transcrição Gênica , Microambiente Tumoral
16.
Clin Exp Metastasis ; 30(1): 103-17, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22806361

RESUMO

We previously found LOXL4 to be alternatively spliced in an anatomic site-specific manner in tumors involving the serosal cavities. LOXL4 splice variants were predominantly or exclusively expressed in effusion specimens from ovarian and breast carcinoma patients, and were absent in primary carcinomas. In the present study, LOXL4 full-length or splice variants were overexpressed in ES-2 and MDA-MB-231 cells and their invasive and metastatic potential and microRNA expression profile were evaluated. ES-2 cells were further injected into SCID mice ovaries and the extent of tumor progression and metastases formation were compared. We show that both splice variants have a positive effect on the metastatic potential of cells in vitro and on tumor progression in vivo. In contrast, full-length LOXL4 is not pro-metastatic, and may even be considered as a tumor suppressor. In addition, we show that LOXL4 is a possible splicing target of the oncogenic splicing factors SRSF1 and hnRNP A1. In conclusion, our results point to a significant role for LOXL4 alternative splicing in tumor progression.


Assuntos
Processamento Alternativo , Aminoácido Oxirredutases/genética , Neoplasias da Mama/genética , Neoplasias Ovarianas/genética , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Neoplasias da Mama/secundário , Feminino , Perfilação da Expressão Gênica , Ribonucleoproteína Nuclear Heterogênea A1 , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/metabolismo , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Isoenzimas , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos SCID , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Ovarianas/secundário , Mutação Puntual/genética , Complexo de Endopeptidases do Proteassoma/química , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteína-Lisina 6-Oxidase , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Processamento de Serina-Arginina , Esferoides Celulares , Células Tumorais Cultivadas
17.
PLoS One ; 8(1): e54321, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23342133

RESUMO

Vav1 functions as a signal transducer protein in the hematopoietic system, where it is exclusively expressed. Vav1 was recently implicated in several human cancers, including lung, pancreatic and neuroblasoma. In this study, we analyzed the expression and function of Vav1 in human breast tumors and breast cancer cell lines. Immunohistochemical analysis of primary human breast carcinomas indicated that Vav1 is expressed in 62% of 65 tumors tested and is correlated positively with estrogen receptor expression. Based on published gene profiling of 50 breast cancer cell lines, several Vav1-expressing cell lines were identified. RT-PCR confirmed Vav1 mRNA expression in several of these cell lines, yet no detectable levels of Vav1 protein were observed due to cbl-c proteasomal degradation. We used two of these lines, MCF-7 (Vav1 mRNA negative) and AU565 (Vav1 mRNA positive), to explore the effect of Vav1 expression on breast cell phenotype and function. Vav1 expression had opposite effects on function in these two lines: it reduced proliferation and enhanced cell death in MCF-7 cells but enhanced proliferation in AU565 cells. Consistent with these findings, transcriptome analysis revealed an increase in expression of proliferation-related genes in Vav1-expressing AU565 cells compared to controls, and an increase in apoptosis-related genes in Vav1-expressing MCF-7 cells compared with controls. TUNEL and γ-H2AX foci assays confirmed that expression of Vav1 increased apoptosis in MCF-7 cells but not AU565 cells and shRNA experiments revealed that p53 is required for this pro-apoptotic effect of Vav1 in these cells. These results highlight for the first time the potential role of Vav1 as an oncogenic stress activator in cancer and the p53 dependence of its pro-apoptotic effect in breast cells.


Assuntos
Apoptose/fisiologia , Neoplasias da Mama/metabolismo , Proteínas Proto-Oncogênicas c-vav/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Apoptose/genética , Neoplasias da Mama/genética , Linhagem Celular , Proliferação de Células , Feminino , Humanos , Imuno-Histoquímica , Imunoprecipitação , Marcação In Situ das Extremidades Cortadas , Proteínas Proto-Oncogênicas c-vav/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA