Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
RNA Biol ; 21(1): 1-11, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38832821

RESUMEN

LncRNA is a group of transcripts with a length exceeding 200 nucleotides that contribute to tumour development. Our research group found that LINC00052 expression was repressed during the formation of breast cancer (BC) multicellular spheroids. Intriguingly, LINC00052 precise role in BC remains uncertain. We explored LINC00052 expression in BC patients` RNA samples (TCGA) in silico, as well as in an in-house patient cohort, and inferred its cellular and molecular mechanisms. In vitro studies evaluated LINC00052 relevance in BC cells viability, cell cycle and DNA damage. Results. Bioinformatic RNAseq analysis of BC patients showed that LINC00052 is overexpressed in samples from all BC molecular subtypes. A similar LINC00052 expression pattern was observed in an in-house patient cohort. In addition, higher LINC00052 levels are related to better BC patient´s overall survival. Remarkably, MCF-7 and ZR-75-1 cells treated with estradiol showed increased LINC00052 expression compared to control, while these changes were not observed in MDA-MB-231 cells. In parallel, bioinformatic analyses indicated that LINC00052 influences DNA damage and cell cycle. MCF-7 cells with low LINC00052 levels exhibited increased cellular protection against DNA damage and diminished growth capacity. Furthermore, in cisplatin-resistant MCF-7 cells, LINC00052 expression was downregulated. Conclusion. This work shows that LINC00052 expression is associated with better BC patient survival. Remarkably, LINC00052 expression can be regulated by Estradiol. Additionally, assays suggest that LINC00052 could modulate MCF-7 cells growth and DNA damage repair. Overall, this study highlights the need for further research to unravel LINC00052 molecular mechanisms and potential clinical applications in BC.


Asunto(s)
Neoplasias de la Mama , Regulación Neoplásica de la Expresión Génica , ARN Largo no Codificante , Femenino , Humanos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/genética , Biología Computacional/métodos , Daño del ADN , Resistencia a Antineoplásicos/genética , Perfilación de la Expresión Génica , Células MCF-7 , Pronóstico , ARN Largo no Codificante/genética
2.
Int J Mol Sci ; 25(7)2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38612643

RESUMEN

Breast cancer is a leading cause of cancer-related deaths among women. Cisplatin is used for treatment, but the development of resistance in cancer cells is a significant concern. This study aimed to investigate changes in the transcriptomes of cisplatin-resistant MCF7 cells. We conducted RNA sequencing of cisplatin-resistant MCF7 cells, followed by differential expression analysis and bioinformatic investigations to identify changes in gene expression and modified signal transduction pathways. We examined the size and quantity of extracellular vesicles. A total of 724 genes exhibited differential expression, predominantly consisting of protein-coding RNAs. Notably, two long non-coding RNAs (lncRNAs), NEAT1 and MALAT, were found to be dysregulated. Bioinformatic analysis unveiled dysregulation in processes related to DNA synthesis and repair, cell cycle regulation, immune response, and cellular communication. Additionally, modifications were observed in events associated with extracellular vesicles. Conditioned media from resistant cells conferred resistance to wild-type cells in vitro. Furthermore, there was an increase in the number of vesicles in cisplatin-resistant cells. Cisplatin-resistant MCF7 cells displayed differential RNA expression, including the dysregulation of NEAT1 and MALAT long non-coding RNAs. Key processes related to DNA and extracellular vesicles were found to be altered. The increased number of extracellular vesicles in resistant cells may contribute to acquired resistance in wild-type cells.


Asunto(s)
Cisplatino , Transcriptoma , Femenino , Humanos , Cisplatino/farmacología , Células MCF-7 , Perfilación de la Expresión Génica , ADN
3.
Cancer Biomark ; 30(4): 429-446, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33492282

RESUMEN

BACKGROUND: Smac/DIABLO is a proapoptotic protein deregulated in breast cancer, with a controversial role as a tumor marker, possibly due to a lack of correlative mRNA and protein analyses. OBJECTIVE: To investigate the association of Smac/DIABLO gene and protein levels with clinical variables in breast cancer patients. METHODS: Smac/DIABLO mRNA expression was analyzed by qPCR in 57 frozen tissues, whereas protein levels were assessed by immunohistochemistry in 82 paraffin-embedded tissues. Survivin mRNA levels were also measured. In vitro assays were performed to investigate possible regulators of Smac/DIABLO. RESULTS: Higher levels of Smac/DIABLO mRNA and protein were found in estrogen receptor (ER)-positive samples (p= 0.0054 and p= 0.0043, respectively) in comparison to ER-negative tumors. A negligible positive association was found between Smac/DIABLO and survivin expression. In vitro assays showed that Smac/DIABLO is not regulated by ER and, conversely, it does not participate in ER expression modulation. CONCLUSIONS: mRNA and protein levels of Smac/DIABLO were increased in ER-positive breast tumors in comparison with ER-negative samples, although the mechanism of this regulation is still unknown. Public databases showed a possible clinical relevance for this association.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Neoplasias de la Mama/metabolismo , Proteínas Mitocondriales/genética , Receptores de Estrógenos/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Persona de Mediana Edad , Transfección
4.
Cancer Biomark ; 30(4): 365-379, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33361583

RESUMEN

BACKGROUND: Long-non-coding RNAs, a class of transcripts with lengths > 200 nt, play key roles in tumour progression. Previous reports revealed that LINC00052 (long intergenic non-coding RNA 00052) was strongly downregulated during breast cancer multicellular spheroids formation and suggested a role in cell migration and oxidative metabolism. OBJECTIVE: To examine the function of LINC00052 in MCF-7 breast cancer cells. METHODS: Loss-of-function studies were performed to evaluate LINC00052 role on MCF-7 breast cancer cells. Microarray expression assays were performed to determine genes and cellular functions modified after LINC00052 knockdown. Next, the impact of LINC00052 depletion on MCF-7 cell respiration and migration was evaluated. RESULTS: 1,081 genes were differentially expressed upon LINC00052 inhibition. Gene set enrichment analysis, Gene Ontology and Key Pathway Advisor analysis showed that signalling networks related to cell migration and oxidative phosphorylation were enriched. However, whereas LINC00052 knockdown in MCF-7 cells revealed marginal difference in oxygen consumption rates when compared with control cells, LINC00052 inhibition enhanced cell migration in vitro and in vivo, as observed using a Zebrafish embryo xenotransplant model. CONCLUSION: Our data show that LINC00052 modulates MCF-7 cell migration. Genome-wide microarray experiments suggest that cancer cell migration is affected by LINC00052 through cytoskeleton modulation and Notch/ß-catenin/NF-κB signalling pathways.


Asunto(s)
Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica/genética , ARN Largo no Codificante/genética , Animales , Neoplasias de la Mama/patología , Movimiento Celular , Femenino , Humanos , Pez Cebra
5.
Carcinogenesis ; 41(11): 1485-1497, 2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-32710610

RESUMEN

Multicellular tumor spheroids (MCTSs) constitute a three-dimensional culture system that recapitulates the in vivo tumor microenvironment. Tumor cells cultured as MCTSs present antineoplastic resistance due to the effect of microenvironmental signals acting upon them. In this work, we evaluated the biological function of a new microenvironment-regulated long non-coding RNA, lncMat2B, in breast cancer. In MCTSs, the expression of lncMat2B presented an increase and a zonal heterogeneity, as it was expressed principally in quiescent cells of hypoxic regions of the MCTSs. As expected, functional assays supported the role of severe hypoxia in the regulation of lncMat2B. Moreover, gain- and loss-of-function assays using a transcriptional silencing CRISPR/Cas9 system and gBlock revealed that lncMAT2B regulates the tumor-initiating phenotype. Interestingly, lncMat2B is overexpressed in a cisplatin-resistant MCF-7 cell line, and its ectopic expression in wild type MCF-7 cells increased survival to cisplatin exposure by reducing DNA damage and reactive oxygen species accumulation. lncMAT2B is a possible link between severe hypoxia, tumor-initiating phenotype and drug resistance in breast cancer cells.


Asunto(s)
Neoplasias de la Mama/patología , Cisplatino/farmacología , Reparación del ADN , Resistencia a Antineoplásicos , Hipoxia/fisiopatología , Células Madre Neoplásicas/patología , ARN Largo no Codificante/genética , Animales , Antineoplásicos/farmacología , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proliferación Celular , Daño del ADN , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Metionina Adenosiltransferasa/genética , Invasividad Neoplásica , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Especies Reactivas de Oxígeno , Esferoides Celulares , Células Tumorales Cultivadas , Microambiente Tumoral , Pez Cebra
6.
Biochim Biophys Acta Mol Cell Res ; 1866(12): 118523, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31401107

RESUMEN

Multicellular Tumor Spheroids culture (MCTS) is an in vitro model mimicking the characteristics of the tumor microenvironment, such as hypoxia and acidosis, resulting in the presence of both proliferating and quiescent cell populations. lncRNA's is a novel group of regulatory molecules that participates in the acquisition of tumorigenic phenotypes. In the present work we evaluated the oncogenic association of an uncharacterized lncRNA (lncRNA-HAL) in the tumorigenic phenotype induced by the MCTS microenvironment. We measured lncRNA-HAL expression level in MCF-7-MCTS populations and under different hypoxic conditions by RT-qPCR. Afterwards, we silenced lncRNA-HAL expression by shRNAs and evaluated its effect in MCF-7 transcriptome (by RNAseq) and validated the modified cellular processes by proliferation, migration, and stem cells assays. Finally, we analyzed which proteins interacts with lncRNA-HAL by ChIRP assay, to propose a possible molecular mechanism for this lncRNA. We found that lncRNA-HAL is overexpressed in the internal quiescent populations (p27 positive populations) of MCF-7-MCTS, mainly in the quiescent stem cell population, being hypoxia one of the microenvironmental cues responsible of its overexpression. Transcriptome analysis of lncRNA-HAL knockdown MCF7 cells revealed that lncRNA-HAL effect is associated with proliferation, migration and cell survival mechanisms; moreover, lncRNA-HAL silencing increased cell proliferation and impaired cancer stem cell proportion and function, resulting in decreased tumor grafting in vivo. In addition, we found that this lncRNA was overexpressed in triple-negative breast cancer patients. Analysis by ChIRP assay showed that this nuclear lncRNA binds to histones and hnRNPs suggesting a participation at the chromatin level and transcriptional regulation. The results obtained in the present work suggest that the function of lncRNA-HAL is associated with quiescent stem cell populations, which in turn is relevant due to its implications in cancer cell survival and resistance against treatment in vivo. Altogether, our data highlights a new lncRNA whose expression is regulated by the tumor microenvironment and associated to stemness in breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , ARN Largo no Codificante/genética , Microambiente Tumoral/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclo Celular , Silenciador del Gen , Humanos , Células MCF-7 , Fenotipo , ARN Largo no Codificante/metabolismo , Células Tumorales Cultivadas
7.
Biochem Biophys Res Commun ; 516(4): 1258-1264, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31301772

RESUMEN

BACKGROUND: Multicellular tumor spheroids mimic the functional organization of tumors in vivo, providing biological readouts that predict the behavior of cancer cells more accurately. The current study aimed to evaluate the transcriptome (mRNAs and long non-coding RNAs) of multicellular tumor spheroids from breast cancer cells. METHODS: MCF-7 cell spheroids were used; the transcriptome was analyzed using RNAseq and RNA microarrays; the secretion of macrophage migration inhibitor (MIF), a cytokine exported by the cholesterol efflux regulatory protein, was measured by ELISA. Linc00052 was inhibited using short-hairpin RNAs (shRNAs). RESULTS: We found several differentially regulated mRNAs and lncRNAs in MCF-7 cell spheroids. We also found significant enrichment of the Wnt/B-catenin death receptor and the cholesterol metabolic processes. Interestingly, we also found an increased concentration of MIF. Further, at 12 and 20 days of 3D culture we found 221 and 1146 dysregulated lncRNAs, respectively; including linc00052 (long intergenic non-protein coding RNA 52), which has been involved in breast cancer. Linc00052 knock-down experiments suggest that it could be a key regulator of cholesterol pathways in breast cancer. CONCLUSIONS: Our data shows that tumor spheroids can induce changes in the transcriptome of the cultured cells, including both mRNAs and ncRNA. One of the major changes included the deregulation of cholesterol pathways, of which linc00052 is apparently a key regulator.


Asunto(s)
Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , ARN Largo no Codificante/genética , Esferoides Celulares/metabolismo , Transcriptoma , Hipoxia de la Célula , Línea Celular Tumoral , Movimiento Celular , Colesterol/metabolismo , Femenino , Perfilación de la Expresión Génica , Silenciador del Gen , Humanos , Oxidorreductasas Intramoleculares/genética , Cinética , Células MCF-7 , Factores Inhibidores de la Migración de Macrófagos/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN
9.
EMBO Rep ; 17(5): 648-58, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27113763

RESUMEN

Cancer stem cells (CSCs) are linked to metastasis. Moreover, a discrete group of miRNAs (metastamiRs) has been shown to promote metastasis. Accordingly, we propose that miRNAs that function as metastatic promoters may influence the CSC phenotype. To study this issue, we compared the expression of 353 miRNAs in CSCs enriched from breast cancer cell lines using qRT-PCR analysis. One of the most altered miRNAs was miR-10b, which is a reported promoter of metastasis and migration. Stable overexpression of miR-10b in MCF-7 cells (miR-10b-OE cells) promoted higher self-renewal and expression of stemness and epithelial-mesenchymal transition (EMT) markers. In agreement with these results, inhibiting miR-10b expression using synthetic antisense RNAs resulted in a decrease in CSCs self-renewal. Bioinformatics analyses identified several potential miR-10b mRNA targets, including phosphatase and tensin homolog (PTEN), a key regulator of the PI3K/AKT pathway involved in metastasis, cell survival, and self-renewal. The targeting of PTEN by miR-10b was confirmed using a luciferase reporter, qRT-PCR, and Western blot analyses. Lower PTEN levels were observed in CSCs, and miR-10b depletion not only increased PTEN mRNA and protein expression but also decreased the activity of AKT, a downstream PTEN target kinase. Correspondingly, PTEN knockdown increased stem cell markers, whereas AKT inhibitors compromised the self-renewal ability of CSCs and breast cancer cell lines overexpressing miR-10b. In conclusion, miR-10b regulates the self-renewal of the breast CSC phenotype by inhibiting PTEN and maintaining AKT pathway activation.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Autorrenovación de las Células/genética , MicroARNs/genética , Células Madre Neoplásicas/metabolismo , Fosfohidrolasa PTEN/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/genética , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Xenoinjertos , Humanos , Ratones , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , ARN Mensajero/genética , Transducción de Señal , Transcriptoma
10.
Mol Carcinog ; 55(12): 1952-1961, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-26618609

RESUMEN

Tissue inhibitor of metalloproteinase-4 (TIMP-4) belongs to a family of extracellular matrix (ECM) metalloproteinases inhibitors that are overexpressed in several cancers. However, the role of TIMP-4 during carcinogenesis is poorly understood. To evaluate TIMP-4 functions in carcinogenesis, stably transfected cells overexpressing this tissue inhibitor were used. Xenograft tumor growth, stem cell enrichment, colony formation, and gene regulation were investigated. Microarrays and in silico analysis were carried out to elucidate TIMP-4 molecular mechanisms. In the present report, we show that in nude mice, cervical cancer cells that overexpress TIMP-4 formed tumors faster than control cell-derived tumors. Furthermore, in vivo limiting dilution assays showed that fewer TIMP-4 overexpressing cells are needed for tumor formation. In vitro analyses demonstrated that TIMP-4 overexpression or exposure to human recombinant TIMP-4 (hrTIMP4) caused an enrichment of the tumor progenitor cell (TPC) population. Accordingly, genome-wide expression and signaling pathway analyses showed that hrTIMP-4 modulated cell survival, cell proliferation, inflammation, and epithelial-mesenchymal transition (EMT) signaling networks. Notably, NFκB signaling pathway appeared to be globally activated upon hrTIMP-4 treatment. Overall, this report provides the first example that TIMP-4 regulates carcinogenesis through enriching the TPC population in cervical cancer cells. Understanding TIMP-4 effects on tumorigenesis may provide clues for future therapies design. © 2015 Wiley Periodicals, Inc.


Asunto(s)
Cuello del Útero/patología , Regulación Neoplásica de la Expresión Génica , Células Madre Neoplásicas/patología , Inhibidores Tisulares de Metaloproteinasas/genética , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología , Animales , Cuello del Útero/metabolismo , Femenino , Células HeLa , Humanos , Ratones , FN-kappa B/metabolismo , Células Madre Neoplásicas/metabolismo , Transducción de Señal , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Regulación hacia Arriba , Neoplasias del Cuello Uterino/metabolismo , Inhibidor Tisular de Metaloproteinasa-4
11.
PLoS One ; 10(8): e0135929, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26291714

RESUMEN

Tissue inhibitor of metalloproteinase-4 (TIMP-4) is a member of extracellular matrix (ECM) metalloproteinases inhibitors that has pleiotropic functions. However, TIMP-4 roles in carcinogenesis are not well understood. Cell viability and flow cytometer assays were employed to evaluate cell death differences between H-Vector and H-TIMP-4 cell lines. Immunobloting and semi-quantitative RT-PCR were used to evaluate the expression of apoptosis regulators. We showed that TIMP-4 has apoptosis-sensitizing effects towards several death stimuli. Consistent with these findings, regulators of apoptosis from Inhibitors of Apoptosis Proteins (IAP), FLICE-like inhibitor proteins (FLIP) and Bcl-2 family members were modulated by TIMP-4. In addition, TIMP-4 knockdown resulted in cell survival increase after serum deprivation, as assessed by clonogenic cell analyses. This report shows that TIMP-4 regulates carcinogenesis through apoptosis activation in cervical cancer cells. Understanding TIMP-4 effects in tumorigenesis may provide clues for future therapies.


Asunto(s)
Apoptosis/fisiología , Inhibidores Tisulares de Metaloproteinasas/fisiología , Neoplasias del Cuello Uterino/fisiopatología , Carcinogénesis/metabolismo , Caspasas/metabolismo , Femenino , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Inhibidor Tisular de Metaloproteinasa-4
12.
Hum Cell ; 26(2): 47-55, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23494744

RESUMEN

B cell leukemia-3 (Bcl-3) has been defined as an anti-apoptotic gene; however, the exact mechanisms through which Bcl-3 influences apoptosis have been elusive. To determine the specific role of Bcl-3 in apoptosis, we evaluated the effect of its silencing on the expression of proteins involved in either the extrinsic or intrinsic apoptotic pathways induced by ultraviolet light B-mediated DNA damage. We found that, in Bcl-3-silenced cells, caspase-3, caspase-8 and caspase-9 activation is accelerated and tBid mitochondrial content is increased. It is important to note that, although mitochondrial Smac levels were reduced after UV exposure, the rate of reduction was slightly higher in Bcl-3 silenced cells than in control cells. Additionally, p53 levels diminished in Bcl-3 silenced cells compared to control cells, as did those of DNA-PK, a DNA repair protein. Altogether, our data indicate that Bcl-3 protects cells from apoptosis by regulating both apoptotic pathways.


Asunto(s)
Apoptosis/genética , Apoptosis/efectos de la radiación , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/metabolismo , Rayos Ultravioleta/efectos adversos , Proteínas Reguladoras de la Apoptosis , Proteínas del Linfoma 3 de Células B , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Caspasas/metabolismo , Daño del ADN/efectos de la radiación , Silenciador del Gen , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
13.
Proc Natl Acad Sci U S A ; 108(5): 1943-8, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21245302

RESUMEN

Cancer cells use different modes of migration, including integrin-dependent mesenchymal migration of elongated cells along elements of the 3D matrix as opposed to low-adhesion-, contraction-based amoeboid motility of rounded cells. We report that MDA-MB-231 human breast adenocarcinoma cells invade 3D Matrigel with a characteristic rounded morphology and with F-actin and myosin-IIa accumulating at the cell rear in a uropod-like structure. MDA-MB-231 cells display neither lamellipodia nor bleb extensions at the leading edge and do not require Arp2/3 complex activity for 3D invasion in Matrigel. Accumulation of phospho-MLC and blebbing activity were restricted to the uropod as reporters of actomyosin contractility, and velocimetric analysis of fluorescent beads embedded within the 3D matrix showed that pulling forces exerted to the matrix are restricted to the side and rear of cells. Inhibition of actomyosin contractility or ß1 integrin function interferes with uropod formation, matrix deformation, and invasion through Matrigel. These findings support a model whereby actomyosin-based uropod contractility generates traction forces on the ß1 integrin adhesion system to drive cell propulsion within the 3D matrix, with no contribution of lamellipodia extension or blebbing to movement.


Asunto(s)
Adenocarcinoma/patología , Neoplasias de la Mama/patología , Colágeno , Laminina , Invasividad Neoplásica , Proteoglicanos , Línea Celular Tumoral , Movimiento Celular , Combinación de Medicamentos , Femenino , Humanos , Integrina beta1/metabolismo , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Miosina Tipo II/metabolismo
14.
Eur J Cell Biol ; 90(2-3): 128-35, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-20970878

RESUMEN

Invasion across tissue boundaries by metastatic tumor cells depends on the proteolytic degradation of the extracellular matrix, initiated by the formation of invadopodia, actin-driven membrane protrusions with matrix-degradative activity. Yet, mechanisms underlying invadopodia formation remain largely unknown. In this report, we examined the role of the histone deacetylase HDAC6 in invadopodia formation and invasion by breast cancer cells. Using small interfering RNA silencing of protein expression in highly invasive MDA-MB-231 breast adenocarcinoma cells, we show that HDAC6 is required for two-dimensional matrix proteolysis. In addition, we demonstrate that HDAC6 acts as a tubulin and cortactin deacetylase. We also report that the inhibition of HDAC6 by siRNA or treatment with HDAC inhibitor TSA results in a decreased invasion capacity of a three-dimensional type I collagen matrix by MDA-MB-231 cells. These data identify HDAC6 as a critical component of the invasive apparatus of tumor cells, in both two- and three-dimensional matrices.


Asunto(s)
Adenocarcinoma/enzimología , Adenocarcinoma/patología , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Extensiones de la Superficie Celular/enzimología , Histona Desacetilasas/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/ultraestructura , Membrana Basal/enzimología , Membrana Basal/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/ultraestructura , Línea Celular Tumoral , Extensiones de la Superficie Celular/patología , Colágeno Tipo I/metabolismo , Cortactina/metabolismo , Matriz Extracelular/enzimología , Matriz Extracelular/patología , Femenino , Histona Desacetilasa 6 , Histona Desacetilasas/biosíntesis , Histona Desacetilasas/genética , Humanos , Invasividad Neoplásica , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Transfección
15.
Cancer Res ; 70(19): 7710-22, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20841469

RESUMEN

Loss of NM23-H1 expression correlates with the degree of metastasis and with unfavorable clinical prognosis in several types of human carcinoma. However, the mechanistic basis for the metastasis suppressor function of NM23-H1 is obscure. We silenced NM23-H1 expression in human hepatoma and colon carcinoma cells and methodologically investigated effects on cell-cell adhesion, migration, invasion, and signaling linked to cancer progression. NM23-H1 silencing disrupted cell-cell adhesion mediated by E-cadherin, resulting in ß-catenin nuclear translocation and T-cell factor/lymphoid-enhancing factor-1 transactivation. Further, NM23-H1 silencing promoted cellular scattering, motility, and extracellular matrix invasion by promoting invadopodia formation and upregulating several matrix metalloproteinases (MMP), including membrane type 1 MMP. In contrast, silencing the related NM23-H2 gene was ineffective at promoting invasion. NM23-H1 silencing activated proinvasive signaling pathways involving Rac1, mitogen-activated protein kinases, phosphatidylinositol 3-kinase (PI3K)/Akt, and src kinase. Conversely, NM23-H1 was dispensable for cancer cell proliferation in vitro and liver regeneration in NM23-M1 null mice, instead inducing cellular resistance to chemotherapeutic drugs in vitro. Analysis of NM23-H1 expression in clinical specimens revealed high expression in premalignant lesions (liver cirrhosis and colon adenoma) and the central body of primary liver or colon tumors, but downregulation at the invasive front of tumors. Our findings reveal that NM23-H1 is critical for control of cell-cell adhesion and cell migration at early stages of the invasive program in epithelial cancers, orchestrating a barrier against conversion of in situ carcinoma into invasive malignancy.


Asunto(s)
Uniones Adherentes/genética , Nucleósido Difosfato Quinasas NM23/genética , Neoplasias/genética , Actinas/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Uniones Adherentes/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/genética , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Citoesqueleto/metabolismo , Citoesqueleto/patología , Silenciador del Gen , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Metaloproteinasa 14 de la Matriz/metabolismo , Nucleósido Difosfato Quinasas NM23/biosíntesis , Invasividad Neoplásica , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Wnt/metabolismo
16.
J Cell Sci ; 122(Pt 17): 3015-24, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19692588

RESUMEN

When migrating away from a primary tumour, cancer cells interact with and remodel the extracellular matrix (ECM). Matrix metalloproteinases (MMPs), and in particular the transmembrane MT1-MMP (also known as MMP-14), are key enzymes in tumour-cell invasion. Results from recent in vitro studies highlight that MT1-MMP is implicated both in the breaching of basement membranes by tumour cells and in cell invasion through interstitial type-I collagen tissues. Remarkably, MT1-MMP accumulates at invadopodia, which are specialized ECM-degrading membrane protrusions of invasive cells. Here we review current knowledge about MT1-MMP trafficking and its importance for the regulation of protease activity at invadopodia. In invasive cells, endocytosis of MT1-MMP by clathrin- and caveolae-dependent pathways can be counteracted by several mechanisms, which leads to protease stabilization at the cell surface and increased pericellular degradation of the matrix. Furthermore, the recent identification of cellular components that control delivery of MT1-MMP to invadopodia brings new insight into mechanisms of cancer-cell invasion and reveals potential pharmacological targets.


Asunto(s)
Estructuras de la Membrana Celular/metabolismo , Matriz Extracelular/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , Invasividad Neoplásica , Neoplasias/metabolismo , Animales , Estructuras de la Membrana Celular/genética , Matriz Extracelular/genética , Humanos , Metaloproteinasa 14 de la Matriz/genética , Neoplasias/genética , Neoplasias/patología , Transporte de Proteínas
17.
Cancer Res ; 69(7): 2792-800, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19276357

RESUMEN

Proteolytic degradation of the extracellular matrix by metastatic tumor cells is initiated by the formation of invadopodia, i.e., actin-driven filopodia-like membrane protrusions endowed with matrix-degradative activity. A signaling cascade involving neural Wiskott-Aldrich syndrome protein and the Arp2/3 actin nucleating complex is involved in actin assembly at invadopodia. Yet, the mechanism of invadopodia formation is poorly understood. Based on their role as actin nucleators in cytoskeletal rearrangements, including filopodia formation, we examined the function of Diaphanous-related formins (DRF) in invadopodia formation and invasion by breast tumor cells. Using small interfering RNA silencing of protein expression in highly invasive MDA-MB-231 breast adenocarcinoma cells, we show that three members of the DRF family (DRF1-DRF3) are required for invadopodia formation and two-dimensional matrix proteolysis. We also report that invasion of a three-dimensional Matrigel matrix involves filopodia-like protrusions enriched for invadopodial proteins, including membrane type 1 matrix metalloproteinase, which depend on DRFs for their formation. These data identify DRFs as critical components of the invasive apparatus of tumor cells in two-dimensional and three-dimensional matrices and suggest that different types of actin nucleators cooperate during the formation of invadopodia.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenocarcinoma/patología , Neoplasias de la Mama/patología , Actinas/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/ultraestructura , Membrana Basal/metabolismo , Membrana Basal/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/ultraestructura , Línea Celular Tumoral , Cortactina/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Forminas , Humanos , Metaloproteinasa 14 de la Matriz/metabolismo , Microscopía Electrónica de Transmisión , Invasividad Neoplásica
18.
Anticancer Res ; 25(1B): 623-7, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15816637

RESUMEN

BACKGROUND: Tissue Inhibitors of Metalloproteinases (TIMPs) play a critical role in extracellular matrix remodeling, which is involved in tumor growth and metastasis. Elevated TIMP levels are reported in association with cancer progression. In particular, it has been shown that TIMP-1 and -2 levels are increased in cervical cancer patients. We analyzed, for the first time, TIMP-4 expression in cervical tumor samples. MATERIALS AND METHODS: Semiquantitative RT-PCR was performed in 26 tumor and 6 normal cervical samples. RESULTS: The study included 32 samples, 7 IB samples, 9 IIB samples, 10 IIIB samples and a control group (n =6) of normal cervical squamous epithelial tissues. Whereas none of the control samples expressed TIMP-4, 24 (88%) of the 26 cervical cancer samples expressed the inhibitor. Higher TIMP-4 levels were found in advanced stage disease (p=0.016, Chi-square test). CONCLUSION: TIMP-4 is expressed de novo in cervical cancer. Higher inhibitor expression levels were found in stages II and III.


Asunto(s)
Inhibidores Tisulares de Metaloproteinasas/biosíntesis , Neoplasias del Cuello Uterino/enzimología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor , Línea Celular Tumoral , ADN Complementario/metabolismo , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Humanos , Persona de Mediana Edad , Metástasis de la Neoplasia , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Distribución Tisular , Transfección , Células Tumorales Cultivadas , Inhibidor Tisular de Metaloproteinasa-4
19.
Int J Biochem Cell Biol ; 36(8): 1655-63, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15147743

RESUMEN

Tissue inhibitors of metalloproteinases (TIMPs) are pleiotropic factors that function as key regulators of extracellular matrix remodeling. They exhibit multifunctional roles including cell growth-stimulating activities and protection from apoptosis. In the present study, we showed that human recombinant TIMP-2 (hrTIMP-2) promotes growth of A549 lung cells. This effect was accompanied by increase in nuclear factor-kappa B (NF-kappaB) activity 24h after exposure as determined by electrophoretic mobility shift assay (EMSA) analysis. This effect was correlated with downregulation of IkappaBalpha and beta proteins and later increases in Bcl-3, IkappaB, and cyclin D1 proteins. Blocking induction of NF-kappaB activity using a dominant-negative mutated version of IkappaBalpha abrogated NF-kappaB activation and cell proliferation.


Asunto(s)
Proliferación Celular , FN-kappa B/fisiología , Mucosa Respiratoria/citología , Inhibidor Tisular de Metaloproteinasa-2/fisiología , Proteínas del Linfoma 3 de Células B , Línea Celular , Ciclina D1/genética , Regulación de la Expresión Génica/fisiología , Humanos , Quinasa I-kappa B , Proteínas I-kappa B , Pulmón/citología , Pulmón/metabolismo , Inhibidor NF-kappaB alfa , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Mucosa Respiratoria/metabolismo , Factores de Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA