Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
STAR Protoc ; 4(1): 102109, 2023 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-36853730

RESUMO

Transduction with lentiviral vectors is a useful approach to study the molecular function of specific genes in mammalian cells. Here, we present a calcium phosphate-based transfection protocol that guarantees highly efficient production and delivery of lentiviral vectors in adherent cultured cells. We also describe in detail a direct lysis technique to measure protein expression, an optimized sulforhodamine B proliferation assay, and a step-by-step chromatin immunoprecipitation procedure to verify the binding of ETV5 to E2F1 first intron in SYO-1 sarcoma cells. For complete details on the use and execution of this protocol, please refer to Kingston et al. (2003),1 Ireton et al. (2002),2 Brown et al. (2009),3 DeSalvo et al. (2021),4 Vichai and Kirtikara (2006),5 and Boyer et al. (2005).6.


Assuntos
Mamíferos , Animais , Morte Celular , Íntrons , Imunoprecipitação da Cromatina , Proliferação de Células/genética
2.
Dis Markers ; 2022: 7729968, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35198082

RESUMO

Chondrosarcoma is a group of primary bone cancers that arise from transformed cells of chondrocytic lineage. Tumor recurrence and metastasis are devastating for patients with chondrosarcoma since there are no effective treatment options. IDH mutations occur in over 50% of tumors from patients with conventional or dedifferentiated chondrosarcomas and represent an attractive target for therapy. However, their role in the pathogenesis of chondrosarcoma remains largely unknown. In this study, we sought to determine the association of IDH mutation and HIF-1α in chondrosarcoma. We used the chondrosarcoma JJ012 cell line and its derived CRISPR/Cas9 mutant IDH1 (IDH1mut) knockout (KO) cells. RNA-Seq data analysis revealed downregulation of several HIF-1α target genes upon loss of IDH1mut. This was associated with reduced HIF-1α levels in the IDH1mut KO cells and tumors. Loss of IDH1mut also attenuated the expression of angiogenic markers in tumor tissues and abrogated the angiogenic capacity of JJ012 cells. Moreover, we observed that exogenous expression of HIF-1α significantly promoted anchorage-independent colony-formation by IDH1mut KO cells. These results suggest IDH1 mutation confers angiogenic and tumorigenic properties of JJ012 cells by inducing HIF-1α. Thus, the HIF pathway represents a promising candidate for combinatorial regimens to target IDH1 mutated chondrosarcomas.


Assuntos
Neoplasias Ósseas/genética , Condrossarcoma/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Isocitrato Desidrogenase/genética , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Condrossarcoma/metabolismo , Condrossarcoma/patologia , Regulação para Baixo , Humanos , Isocitrato Desidrogenase/metabolismo , Mutação , Neovascularização Patológica , Análise de Sequência de RNA
3.
J Clin Invest ; 131(13)2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33983905

RESUMO

Synovial sarcoma is an aggressive malignancy with no effective treatments for patients with metastasis. The synovial sarcoma fusion SS18-SSX, which recruits the SWI/SNF-BAF chromatin remodeling and polycomb repressive complexes, results in epigenetic activation of FGF receptor (FGFR) signaling. In genetic FGFR-knockout models, culture, and xenograft synovial sarcoma models treated with the FGFR inhibitor BGJ398, we show that FGFR1, FGFR2, and FGFR3 were crucial for tumor growth. Transcriptome analyses of BGJ398-treated cells and histological and expression analyses of mouse and human synovial sarcoma tumors revealed prevalent expression of two ETS factors and FGFR targets, ETV4 and ETV5. We further demonstrate that ETV4 and ETV5 acted as drivers of synovial sarcoma growth, most likely through control of the cell cycle. Upon ETV4 and ETV5 knockdown, we observed a striking upregulation of DUX4 and its transcriptional targets that activate the zygotic genome and drive the atrophy program in facioscapulohumeral dystrophy patients. In addition to demonstrating the importance of inhibiting all three FGFRs, the current findings reveal potential nodes of attack for the cancer with the discovery of ETV4 and ETV5 as appropriate biomarkers and molecular targets, and activation of the embryonic DUX4 pathway as a promising approach to block synovial sarcoma tumors.


Assuntos
Proteínas Proto-Oncogênicas c-ets/metabolismo , Sarcoma Sinovial/metabolismo , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Epigênese Genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Xenoenxertos , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Compostos de Fenilureia/farmacologia , Proteínas Proto-Oncogênicas c-ets/genética , Pirimidinas/farmacologia , Receptores de Fatores de Crescimento de Fibroblastos/deficiência , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Sarcoma Sinovial/genética , Sarcoma Sinovial/patologia , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
Cancers (Basel) ; 12(1)2020 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-31935911

RESUMO

Chondrosarcomas are a heterogeneous group of malignant bone tumors that produce hyaline cartilaginous matrix. Mutations in isocitrate dehydrogenase enzymes (IDH1/2) were recently described in several cancers, including conventional and dedifferentiated chondrosarcomas. These mutations lead to the inability of IDH to convert isocitrate into α-ketoglutarate (α-KG). Instead, α-KG is reduced into D-2-hydroxyglutarate (D-2HG), an oncometabolite. IDH mutations and D-2HG are thought to contribute to tumorigenesis due to the role of D-2HG as a competitive inhibitor of α-KG-dependent dioxygenases. However, the function of IDH mutations in chondrosarcomas has not been clearly defined. In this study, we knocked out mutant IDH1 (IDH1mut) in two chondrosarcoma cell lines using the CRISPR/Cas9 system. We observed that D-2HG production, anchorage-independent growth, and cell migration were significantly suppressed in the IDH1mut knockout cells. Loss of IDH1mut also led to a marked attenuation of chondrosarcoma formation and D-2HG production in a xenograft model. In addition, RNA-Seq analysis of IDH1mut knockout cells revealed downregulation of several integrin genes, including those of integrin alpha 5 (ITGA5) and integrin beta 5 (ITGB5). We further demonstrated that deregulation of integrin-mediated processes contributed to the tumorigenicity of IDH1-mutant chondrosarcoma cells. Our findings showed that IDH1mut knockout abrogates chondrosarcoma genesis through modulation of integrins. This suggests that integrin molecules are appealing candidates for combinatorial regimens with IDH1mut inhibitors for chondrosarcomas that harbor this mutation.

5.
BioDrugs ; 31(5): 379-392, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28840584

RESUMO

Cellular metabolism reprogramming is an emerging hallmark of cancer, which provides tumor cells with not only necessary energy but also crucial materials to support growth. Exploiting the unique features of cancer metabolism is promising in cancer therapies. The growing interest in this field has led to numerous inhibitors being developed against key molecules in metabolic pathways, though most of them are still in preclinical development. Potential targeted cancer cell metabolic pathways under investigation include glycolysis, tricarboxylic acid (TCA) cycle, oxidative phosphorylation (OXPHOS), glutaminolysis, pentose phosphate pathway (PPP), lipid synthesis, amino acid and nucleotide metabolism. Sarcoma is a type of cancer that arises from transformed cells of mesenchymal origin, in contrast to carcinoma which originates from epithelial cells. Compared with carcinoma, progress towards harnessing the therapeutic potential of targeting sarcoma cell metabolism has been relatively slow. Recently however, with the discovery of cancer-specific mutations in metabolic enzymes such as isocitrate dehydrogenase (IDH) and succinate dehydrogenase (SDH) in certain sarcoma types, cancer cellular metabolism has been considered more as a source of new targets for treating sarcoma. In this article, we review metabolic enzymes currently tested for cancer therapies and describe the therapeutic potential of targeting IDH mutations and SDH deficiency in sarcomas.


Assuntos
Antineoplásicos/farmacologia , Enzimas/metabolismo , Sarcoma/metabolismo , Animais , Reprogramação Celular/efeitos dos fármacos , Desenho de Fármacos , Enzimas/genética , Humanos , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/metabolismo , Terapia de Alvo Molecular , Mutação , Sarcoma/tratamento farmacológico , Sarcoma/enzimologia , Succinato Desidrogenase/genética , Succinato Desidrogenase/metabolismo
6.
Semin Cancer Biol ; 32: 18-31, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24938913

RESUMO

Mesenchymal stem cells (MSCs) originate from embryonic mesoderm and give rise to the multiple lineages of connective tissues. Transformed MSCs develop into aggressive sarcomas, some of which are initiated by specific chromosomal translocations that generate fusion proteins with potent oncogenic properties. The sarcoma oncogenes typically prime MSCs through aberrant reprogramming. They dictate commitment to a specific lineage but prevent mature differentiation, thus locking the cells in a state of proliferative precursors. Deregulated expression of lineage-specific transcription factors and controllers of chromatin structure play a central role in MSC reprogramming and sarcoma pathogenesis. This suggests that reversing the epigenetic aberrancies created by the sarcoma oncogenes with differentiation-related reagents holds great promise as a beneficial addition to sarcoma therapies.


Assuntos
Reprogramação Celular/genética , Regulação Neoplásica da Expressão Gênica/genética , Células-Tronco Mesenquimais , Oncogenes/genética , Sarcoma/genética , Diferenciação Celular/genética , Humanos , Fatores de Transcrição/genética
7.
Cancer Discov ; 3(11): 1286-301, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23921231

RESUMO

UNLABELLED: Synovial sarcoma is an aggressive soft-tissue malignancy of children and young adults, with no effective systemic therapies. Its specific oncogene, SYT-SSX (SS18-SSX), drives sarcoma initiation and development. The exact mechanism of SYT-SSX oncogenic function remains unknown. In an SYT-SSX2 transgenic model, we show that a constitutive Wnt/ß-catenin signal is aberrantly activated by SYT-SSX2, and inhibition of Wnt signaling through the genetic loss of ß-catenin blocks synovial sarcoma tumor formation. In a combination of cell-based and synovial sarcoma tumor xenograft models, we show that inhibition of the Wnt cascade through coreceptor blockade and the use of small-molecule CK1α activators arrests synovial sarcoma tumor growth. We find that upregulation of the Wnt/ß-catenin cascade by SYT-SSX2 correlates with its nuclear reprogramming function. These studies reveal the central role of Wnt/ß-catenin signaling in SYT-SSX2-induced sarcoma genesis, and open new venues for the development of effective synovial sarcoma curative agents. SIGNIFICANCE: Synovial sarcoma is an aggressive soft-tissue cancer that afflicts children and young adults, and for which there is no effective treatment. The current studies provide critical insight into our understanding of the pathogenesis of SYT­SSX-dependent synovial sarcoma and pave the way for the development of effective therapeutic agents for the treatment of the disease in humans.


Assuntos
Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Sarcoma Sinovial/genética , Sarcoma Sinovial/patologia , Via de Sinalização Wnt/efeitos dos fármacos , Adolescente , Adulto , Animais , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Camundongos , Camundongos Nus , Camundongos Transgênicos , Compostos de Pirvínio/farmacologia , Sarcoma Experimental , Sarcoma Sinovial/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Adulto Jovem
8.
BMC Genomics ; 13: 189, 2012 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-22594313

RESUMO

BACKGROUND: SYT-SSX is the oncogene associated with synovial sarcoma (SS), a stem cell disease. SYT-SSX is thought to be responsible for sarcoma initiation and development. It interacts with components of Polycomb and SWI/SNF complexes, the two epigenetic controllers that maintain the heritable status of differentiation-specific genes in the stem/progenitor cell. Through these associations SYT-SSX is thought to alter gene expression programs by epigenetic mechanisms. Recently, we reported that SYT-SSX2 reprograms mesenchymal stem cells and myoblasts by dictating their commitment to the neural lineage while disrupting their normal differentiation. This reprogramming was due to the direct occupancy of proneural genes by the SYT-SSX2 nuclear complex. To gain a clear understanding of SYT-SSX2 control of gene expression networks, we conducted a thorough genome-wide analysis to determine the mechanism of its recruitment and identify signature sets of epigenetic markers that would predict its targeting and transcriptional activity. RESULTS: SYT-SSX2 was recruited to distinct loci across all chromosomes, and an overwhelming number of Polycomb-modified sites enriched with the trimethylated histone H3 on lysine 27 (H3K27me3) formed the main recruiting module for SYT-SSX2. Not all SYT-SSX2/H3K27me3-occupied genes had altered expression, denoting the requirement for additional signals upon oncogene binding. Differential binding and epigenetic patterns distinguished upregulated and downregulated genes. Most activated genes had SYT-SSX2 sites enriched with H3K27me3 within their body or near their transcription start site (TSS) whereas a majority of downregulated genes were characterized by SYT-SSX2/H3K27me3-rich regions at long-range, or by modifications associated with transcription activation within the gene body or near the TSS. Hierarchical and functional clustering identified H3K27me3 as the dominant epigenetic marker associated with SYT-SSX2 binding and gene expression. Notably, this analysis revealed a cluster of upregulated neuronal genes densely covered by H3K27me3, consistent with programming toward the neural lineage by SYT-SSX2 observed previously. CONCLUSIONS: The data analysis revealed that Polycomb complexes or their modified chromatin and their stably silenced differentiation programs seem to be the main target for SYT-SSX2, suggesting that their perturbation is at the center of tumorigenesis driven by the oncogene. Further research into this mechanism is crucial to the full understanding of SS biology.


Assuntos
Cromatina/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Proteínas do Grupo Polycomb/metabolismo , Sarcoma Sinovial/metabolismo , Animais , Linhagem Celular , Epigênese Genética/genética , Histonas/metabolismo , Humanos , Metilação , Camundongos , Ligação Proteica , Transporte Proteico
9.
PLoS One ; 4(4): e5060, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19337376

RESUMO

This study demonstrates deregulation of polycomb activity by the synovial sarcoma-associated SYT-SSX2 oncogene, also known as SS18-SSX2. Synovial sarcoma is a soft tissue cancer associated with a recurrent t(X:18) translocation event that generates one of two fusion proteins, SYT-SSX1 or SYT-SSX2. The role of the translocation products in this disease is poorly understood. We present evidence that the SYT-SSX2 fusion protein interacts with the polycomb repressive complex and modulates its gene silencing activity. SYT-SSX2 causes destabilization of the polycomb subunit Bmi1, resulting in impairment of polycomb-associated histone H2A ubiquitination and reactivation of polycomb target genes. Silencing by polycomb complexes plays a vital role in numerous physiological processes. In recent years, numerous reports have implicated gain of polycomb silencing function in several cancers. This study provides evidence that, in the appropriate context, expression of the SYT-SSX2 oncogene leads to loss of polycomb function. It challenges the notion that cancer is solely associated with an increase in polycomb function and suggests that any imbalance in polycomb activity could drive the cell toward oncogenesis. These findings provide a mechanism by which the SYT-SSX2 chimera may contribute to synovial sarcoma pathogenesis.


Assuntos
Proteínas Nucleares/antagonistas & inibidores , Proteínas de Fusão Oncogênica/genética , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Repressoras/antagonistas & inibidores , Sarcoma Sinovial/genética , Sequência de Bases , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Primers do DNA , Técnica Indireta de Fluorescência para Anticorpo , Histonas/metabolismo , Humanos , Complexo Repressor Polycomb 1 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ubiquitinação
10.
Exp Cell Res ; 314(19): 3551-62, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18835266

RESUMO

The SYT proto-oncoprotein (also known as SS18) is a gene expression regulator conserved across species. Although its biological function is still unknown, the importance of SYT as a housekeeping protein is illustrated by the lethal phenotype of SYT-null embryos. Notably, SYT is a component of the synovial sarcoma-associated translocation product, the SYT-SSX oncogene. SYT was previously reported as a mediator of cell adhesion. In the present study we show that SYT possesses distinct domains that control MDCK cyst formation in three-dimensional collagen cultures. While the carboxy-half of SYT, the QPGY domain, is required for cyst growth, the amino-terminal region appears to exert on this process a regulatory effect. Further analysis suggested that the purinergic G protein-coupled P2Y receptor signaling is involved in SYT-induced cystogenesis. Activation of this cascade is due to facilitation of ATP release in the extracellular space of polarized MDCK cells by SYT. These studies allow us to begin to understand the vital role of SYT in controlling epithelial morphogenesis and might explain the lethality of its loss in the developing embryo.


Assuntos
Trifosfato de Adenosina/metabolismo , Células Epiteliais/citologia , Proteínas Proto-Oncogênicas/metabolismo , Animais , Polaridade Celular , Células Cultivadas , Cistos/metabolismo , Cães , Células Epiteliais/metabolismo , Espaço Extracelular/metabolismo , Humanos , Microscopia Confocal , Modelos Biológicos , Receptores Purinérgicos P2/metabolismo , Proteínas Repressoras/metabolismo
11.
Biomed Microdevices ; 10(2): 243-9, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17899384

RESUMO

DC-Dielectrophoresis (DC-DEP), the induced motion of the dielectric particles in a spatially non-uniform DC electric field, is applied to separate biological cells by size. The locally non-uniform electric field is generated by an insulating hurdle fabricated within a PDMS microchannel. The cells experience a negative DEP (accordingly a repulsive) force at the corners of the hurdle where the gradient of local electric-field strength is the strongest. The DC-DEP force acting on the cells is proportional to the cells' size. Thus the moving cells deviate from the streamlines and the degree of deviation is dependent on the cell size. In this paper, we demonstrated by using this method that, combined with the electroosmotic flow, mixed biological cells of a few to tens of micrometers difference in diameter can be continuously separated into different collecting wells. For separating target cells of a specific size, all that is required is to adjust the voltage outputs of the electrodes.


Assuntos
Técnicas de Cultura de Células/instrumentação , Separação Celular/instrumentação , Eletroforese/instrumentação , Citometria de Fluxo/instrumentação , Técnicas Analíticas Microfluídicas/instrumentação , Técnicas de Cultura de Células/métodos , Separação Celular/métodos , Tamanho Celular , Células Cultivadas , Eletroforese/métodos , Desenho de Equipamento , Análise de Falha de Equipamento , Citometria de Fluxo/métodos , Humanos , Leucócitos/citologia , Técnicas Analíticas Microfluídicas/métodos
12.
Mol Biol Cell ; 18(10): 4003-12, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17686994

RESUMO

Synovial sarcoma is a soft tissue cancer associated with a recurrent t(X:18) translocation that generates one of two fusion proteins, SYT-SSX1 or SYT-SSX2. In this study, we demonstrate that SYT-SSX2 is a unique oncogene. Rather than confer enhanced proliferation on its target cells, SYT-SSX2 instead causes a profound alteration of their architecture. This aberrant morphology included elongation of the cell body and formation of neurite-like extensions. We also observed that cells transduced with SYT-SSX2 often repulsed one another. Notably, cell repulsion is a known component of ephrin signaling. Further analysis of SYT-SSX2-infected cells revealed significant increases in the expression and activation of Eph/ephrin pathway components. On blockade of EphB2 signaling SYT-SSX2 infectants demonstrated significant reversion of the aberrant cytoskeletal phenotype. In addition, we discovered, in parallel, that SYT-SSX2 induced stabilization of the microtubule network accompanied by accumulation of detyrosinated Glu tubulin and nocodazole resistance. Glu tubulin regulation was independent of ephrin signaling. The clinical relevance of these studies was confirmed by abundant expression of both EphB2 and Glu tubulin in SYT-SSX2-positive synovial sarcoma tissues. These results indicate that SYT-SSX2 exerts part of its oncogenic effect by altering cytoskeletal architecture in an Eph-dependent manner and cytoskeletal stability through a concurrent and distinct pathway.


Assuntos
Citoesqueleto/metabolismo , Efrinas/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Sarcoma Sinovial/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Efrinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Células NIH 3T3 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor EphB2/metabolismo , Retroviridae/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Tirfostinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...