Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
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.
Int J Mol Sci ; 23(19)2022 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-36232741

RESUMEN

Pancreatic cancer is a pathology with a high mortality rate since it is detected at advanced stages, so the search for early-stage diagnostic biomarkers is essential. Liquid biopsies are currently being explored for this purpose and educated platelets are a good candidate, since they are known to present a bidirectional interaction with tumor cells. In this work, we analyzed the effects of platelets on cancer cells' viability, as determined by MTT, migration using transwell assays, clonogenicity in soft agar and stemness by dilution assays and stem markers' expression. We found that the co-culture of platelets and pancreatic cancer cells increased the proliferation and migration capacity of BXCP3 cells, augmented clonogenicity and induced higher levels of Nanog, Sox2 and Oct4 expression. As platelets can provide horizontal transfer of microRNAs, we also determined the differential expression of miRNAs in platelets obtained from a small cohort of pancreatic cancer patients and healthy subjects. We found clear differences in the expression of several miRNAs between platelets of patients with cancer healthy subjects. Moreover, when we analyzed microRNAs from the platelets of the pancreatic juice and blood derived from each of the cancer patients, interestingly we find differences between the blood- and pancreatic juice-derived platelets suggesting the presence of different subpopulations of platelets in cancer patients, which warrant further analysis.


Asunto(s)
MicroARNs , Neoplasias Pancreáticas , Agar , Plaquetas/metabolismo , Línea Celular Tumoral , Humanos , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas
4.
Int J Mol Sci ; 22(12)2021 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-34203589

RESUMEN

Pancreatic ductal adenocarcinoma is one of the deadliest tumors. This neoplasia is characterized by an important cellular and phenotypic heterogeneity. In particular, it has been shown that at least two subtypes can be found: basal-like, which presents stem-like properties, and classical. Cancer stem cells have been isolated and characterized from these tumors, showing their dependance on general and tissue-specific stem transcription factors and signaling pathways. Nevertheless, little is known about their tissue microenvironment and cell non-autonomous regulators, such as long-non-coding RNAs. (lncRNAs). In this review, we summarize the current knowledge about the positive and negative effects of lncRNAs in the stemness phenotype of pancreatic ductal adenocarcinoma cancer (PDAC).


Asunto(s)
Adenocarcinoma/genética , Carcinoma Ductal Pancreático/genética , Células Madre Neoplásicas/patología , Neoplasias Pancreáticas/genética , ARN Largo no Codificante/metabolismo , Adenocarcinoma/patología , Animales , Carcinoma Ductal Pancreático/patología , Humanos , Células Madre Neoplásicas/metabolismo , Neoplasias Pancreáticas/patología , Fenotipo , ARN Largo no Codificante/genética , Neoplasias Pancreáticas
5.
Int J Mol Sci ; 22(9)2021 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-33946652

RESUMEN

Leucine-rich repeats containing G protein-coupled receptor 4 (LGR4) is a receptor that belongs to the superfamily of G protein-coupled receptors that can be activated by R-spondins (RSPOs), Norrin, circLGR4, and the ligand of the receptor activator of nuclear factor kappa-B (RANKL) ligands to regulate signaling pathways in normal and pathological processes. LGR4 is widely expressed in different tissues where it has multiple functions such as tissue development and maintenance. LGR4 mainly acts through the Wnt/ß-catenin pathway to regulate proliferation, survival, and differentiation. In cancer, LGR4 participates in tumor progression, invasion, and metastasis. Furthermore, recent evidence reveals that LGR4 is essential for the regulation of the cancer stem cell population by controlling self-renewal and regulating stem cell properties. This review summarizes the function of LGR4 and its ligands in normal and malignant processes.


Asunto(s)
Neoplasias/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Carcinogénesis , Femenino , Humanos , Ligandos , Masculino , Ratones , MicroARNs/genética , Modelos Biológicos , Neoplasias/etiología , Neoplasias/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Distribución Tisular , Vía de Señalización Wnt
6.
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
7.
Mol Biol Rep ; 47(7): 5175-5184, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32583281

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers in humans, with less than 5% 5-year survival rate. PDAC is characterized by a small number of recurrent mutations, including KRAS, CDKN2A, TP53, and SMAD4 and a long "tail" of infrequent mutated genes. Most of the studies have been performed in US and European populations, so new studies are needed to describe the mutational landscape of these tumors in other cohorts. The present study analyzed the exome and transcriptome of four PDAC tumors from Mexican patients. We found a paucity of the previously described recurrent mutations, with mutations in only three genes (HERC2, CNTNAP2 and HMCN1) previously reported in PDAC with a frequency > 1%. In addition, we discovered several recurrent putative copy number aberrations in SKP2, BRAF, CSSF1R, FOXE1, JAK2 and MET genes and in genes previously reported as putative drivers in PDAC, including KRAS, SF3B1, BRAF, MYC and MET. Although a larger cohort is needed to validate these findings, our results could be pointing toward potential differences in contributing factors for PDAC in Latin-American populations.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma Ductal Pancreático/genética , Mutación , Neoplasias Pancreáticas/genética , Adulto , Anciano , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Femenino , Frecuencia de los Genes , Humanos , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína Smad4/genética , Proteína p53 Supresora de Tumor/genética
9.
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
10.
Mediators Inflamm ; 2019: 2056085, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31360118

RESUMEN

Colitis-associated colorectal cancer (CRC) development has been shown to be related to chronically enhanced inflammation. Macrophage migration inhibitory factor (MIF) is an inflammatory mediator that favors inflammatory cytokine production and has chemotactic properties for the recruitment of macrophages (Møs) and T cells. Here, we investigated the role of MIF in the inflammatory response and recruitment of immune cells in a murine model of chemical carcinogenesis to establish the impact of MIF on CRC genesis and malignancy. We used BALB/c MIF-knockout (MIF-/-) and wild-type (WT) mice to develop CRC by administering intraperitoneal (i.p.) azoxymethane and dextran sodium sulfate in drinking water. Greater tumor burdens were observed in MIF-/- mice than in WT mice. Tumors from MIF-/- mice were histologically identified to be more aggressive than tumors from WT mice. The localization of MIF suggests that it is also involved in cell differentiation. The relative gene expression of il-17, measured by real-time PCR, was higher in MIF-/- CRC mice, compared to the WT CRC and healthy MIF-/- mice. Importantly, compared to the WT intestinal epithelium, lower percentages of tumor-associated Møs were found in the MIF-/- intestinal epithelium. These results suggest that MIF plays a role in controlling the initial development of CRC by attracting Møs to the tumor, which is a condition that favors the initial antitumor responses.


Asunto(s)
Colitis/metabolismo , Colitis/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Macrófagos/metabolismo , Linfocitos T/metabolismo , Animales , Progresión de la Enfermedad , Femenino , Citometría de Flujo , Inmunohistoquímica , Factores Inhibidores de la Migración de Macrófagos/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados
11.
Biochim Biophys Acta Mol Cell Res ; 1864(10): 1642-1655, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28576513

RESUMEN

Multicellular Tumor Spheroids develop a heterogeneous micromilieu and different cell populations, thereby constituting a cancer model with intermediate characteristics between in vitro bi-dimensional cultures and in vivo tumors. Multicellular Tumor Spheroids also acquire tumor aggressiveness features due to transcription modulation of coding and non-coding RNA. Utilizing microarray analyses, we evaluated the microRNAs expression profile in MCF-7 breast cancer cells cultured as Multicellular Tumor Spheroids. The expression data was used to predict associated cellular and molecular functions using different software tools. The biological importance of two dysregulated miRNAs (miR-221-3p and miR-187) was studied by functional assays. Finally, the clinical relevance of these dysregulated miRNAs was explored using previously reported data. Thirty-three dysregulated microRNAs were found in MCF-7 Multicellular Tumor Spheroids. miRNA expression changes were closely linked with growth, proliferation, and cell development. miRNA-221-3p and miR-187 were implicated in the acquisition of migration/invasion capacities, sensitivity to the deprivation of growth factors, cell cycle phase regulation, and cell death. A panel of 5 miRNAs, including miR-187, showed a good predictive value in discriminating between low and high-risk groups of breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , MicroARNs/genética , Esferoides Celulares/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Análisis de Secuencia por Matrices de Oligonucleótidos , Esferoides Celulares/patología
12.
J Biol Chem ; 292(33): 13745-13757, 2017 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-28655776

RESUMEN

Messenger RNA alternative splicing (AS) regulates the expression of a variety of genes involved in both physiological and pathological processes. AS of the anti-apoptotic and proliferation-associated survivin (BIRC5) gene generates six isoforms, which regulate key aspects of cancer initiation and progression. One of the isoforms is survivin DEx3, in which the exclusion of exon 3 generates a unique carboxyl terminus with specific anti-apoptotic functions. This isoform is highly expressed in advanced stages of breast and cervical tumors. Therefore, understanding the mechanisms that regulate survivin DEx3 mRNA AS is clearly important. To this end, we designed a minigene (M), and in combination with a series of deletions and site-directed mutations, we determined that the first 22 bp of exon 3 contain cis-acting elements that enhance the exclusion of exon 3 to generate the survivin DEx3 mRNA isoform. Furthermore, using pulldown assays, we discovered that Sam68 is a possible trans-acting factor that binds to this region and regulates exon 3 splicing. This result was corroborated using a cell line in which the Sam68 binding site in the survivin gene was mutated with the CRISPR/Cas system. This work provides the first clues regarding the regulation of survivin DEx3 mRNA splicing.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Empalme Alternativo , Proteínas de Unión al ADN/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteínas de Neoplasias/metabolismo , ARN Mensajero/metabolismo , ARN Neoplásico/metabolismo , Proteínas de Unión al ARN/metabolismo , Elementos de Respuesta , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Sistemas CRISPR-Cas , Células Clonales , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Embrión no Mamífero , Exones , Eliminación de Gen , Células HeLa , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Mutagénesis Sitio-Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Trasplante de Neoplasias/patología , Mutación Puntual , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferencia de ARN , ARN Mensajero/química , ARN Neoplásico/química , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Survivin , Trasplante Heterólogo , Carga Tumoral , Pez Cebra
13.
Breast Cancer Res Treat ; 171(2): 247-259, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29948402

RESUMEN

BACKGROUND: MicroRNAs constitute a large family of non-coding RNAs, which actively participate in tumorigenesis by regulating a set of mRNAs of distinct signaling pathways. An altered expression of these molecules has been found in different tumorigenic processes of breast cancer, the most common type of cancer in the female population worldwide. PURPOSE: The objective of this review is to discuss how miRNAs become master regulators in breast tumorigenesis. METHODS: An integrative review of miRNAs and breast cancer literature from the last 5 years was done on PubMed. We summarize recent works showing that the defects on the biogenesis of miRNAs are associated with different breast cancer characteristics. Then, we show several examples that demonstrate the link between cellular processes regulated by miRNAs and the hallmarks of breast cancer. Finally, we examine the complexity in the regulation of these molecules as they are modulated by other non-coding RNAs and the clinical applications of miRNAs as they could serve as good diagnostic and classification tools. CONCLUSION: The information presented in this review is important to encourage new directed studies that consider microRNAs as a good tool to improve the diagnostic and treatment alternatives in breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Animales , Biomarcadores de Tumor , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Transformación Celular Neoplásica/genética , Femenino , Variación Genética , Inestabilidad Genómica , Humanos , Invasividad Neoplásica , Células Madre Neoplásicas/metabolismo , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Interferencia de ARN , ARN no Traducido/genética , Investigación , Transducción de Señal , Investigación Biomédica Traslacional
14.
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
15.
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
16.
Cancer Invest ; 34(3): 155-66, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26963048

RESUMEN

We used both in vitro cultures of neuroblastoma cell lines and nude-mice xenotransplants to explore the effects of co-administration of cisplatin and probenecid. Probenecid sensitized neuroblastoma cells, including tumor cells with stem features, to the effects of cisplatin, both in vitro and in vivo. This effect was mediated by an increase in the apoptotic cell death and a concomitant decrease in cell proliferation. This effect is accompanied by modulation of the mRNA and protein of the drug efflux transporters MDR1, MRP2, and BCRP. The co-administration of probenecid with cisplatin should be explored as a possible therapeutic strategy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Neuroblastoma/tratamiento farmacológico , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/administración & dosificación , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Transición Epitelial-Mesenquimal , Femenino , Expresión Génica , Humanos , Ratones Desnudos , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/fisiología , Neuroblastoma/patología , Probenecid/administración & dosificación , Células de Población Lateral/efectos de los fármacos , Células de Población Lateral/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Exp Cell Res ; 335(2): 207-15, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25999146

RESUMEN

Tissue inhibitors of metalloproteases (TIMPs) are multifunctional proteins that inhibit matrix metalloproteases (MMPs). The latest described member of the family, TIMP-4, is expressed mainly in adipose tissue, with detectable levels in the brain and heart. Besides its high expression in fat, the role of this inhibitor in adipose tissue is unknown. In order to study the role of TIMP-4 during adipogenesis in vitro, 3T3-L1 cells were stably transfected with a TIMP-4 specific shRNA or a control shRNA. Unexpectedly, upon TIMP-4 knockdown, 3T3-L1 cells differentiated faster into mature adipocytes. To get better insight of TIMP-4's role in adipogenesis, microarray expression analyses were performed. Network enrichment analyses uncovered 25 significant upstream signaling pathways, among which the NFκB cascade was found. Previous works have shown that NFκB is a key regulator of adipogenesis. In accordance, we found that TIMP-4 knockdown decreased NFκB activity during adipogenesis. The present work suggests that TIMP-4 might act as a negative regulator of adipogenesis through NFκB cascade modulation.


Asunto(s)
Adipogénesis , Inhibidores Tisulares de Metaloproteinasas/fisiología , Células 3T3-L1 , Adipocitos/fisiología , Animales , Técnicas de Silenciamiento del Gen , Ratones , FN-kappa B/metabolismo , Transducción de Señal , Transcriptoma , Inhibidor Tisular de Metaloproteinasa-4
18.
Am J Physiol Lung Cell Mol Physiol ; 308(6): L511-22, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25575513

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive and usually lethal interstitial lung disease of unknown etiology characterized by aberrant activation of epithelial cells that induce the migration, proliferation and activation of fibroblasts. The resulting distinctive fibroblastic/myofibroblastic foci are responsible for the excessive extracellular matrix (ECM) production and abnormal lung remodeling. We have recently found that matrix metalloproteinase 19 (MMP-19)-deficient (Mmp19-/-) mice develop an exaggerated bleomycin-induced lung fibrosis, but the mechanisms are unclear. In this study, we explored the effect of MMP-19 deficiency on fibroblast gene expression and cell behavior. Microarray analysis of Mmp19-/- lung fibroblasts revealed the dysregulation of several profibrotic pathways, including ECM formation, migration, proliferation, and autophagy. Functional studies confirmed these findings. Compared with wild-type mice, Mmp19-/- lung fibroblasts showed increased α1 (I) collagen gene and collagen protein production at baseline and after transforming growth factor-ß treatment and increased smooth muscle-α actin expression (P < 0.05). Likewise, Mmp19-deficient lung fibroblasts showed a significant increase in proliferation (P < 0.01) and in transmigration and locomotion over Boyden chambers coated with type I collagen or with Matrigel (P < 0.05). These findings suggest that, in lung fibroblasts, MMP-19 has strong regulatory effects on the synthesis of key ECM components, on fibroblast to myofibroblast differentiation, and in migration and proliferation.


Asunto(s)
Diferenciación Celular , Movimiento Celular , Proliferación Celular , Metaloproteinasas de la Matriz Secretadas/deficiencia , Miofibroblastos/enzimología , Fibrosis Pulmonar/enzimología , Animales , Autoantígenos/biosíntesis , Autoantígenos/genética , Células Cultivadas , Matriz Extracelular/enzimología , Matriz Extracelular/genética , Matriz Extracelular/patología , Regulación de la Expresión Génica/genética , Ratones , Ratones Noqueados , Miofibroblastos/patología , Colágenos no Fibrilares/biosíntesis , Colágenos no Fibrilares/genética , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Colágeno Tipo XVII
19.
Oncol Lett ; 28(5): 522, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39268158

RESUMEN

Cancer is a multifactorial disease characterized by the loss of control in the expression of genes known as cancer driver genes. Cancer driver genes trigger uncontrolled cell replication, which leads to the development of malignant tumors. A cluster of signal transduction pathways that contain cancer driver genes involved in cellular processes, such as cell proliferation, differentiation, apoptosis and dysregulated organ growth, are associated with cancer initiation and progression. In the present study, three signal transduction pathways involved in cervical cancer (CC) development were analyzed: The Hippo pathway (FAT atypical cadherin, yes-associated protein 1, SMAD4 and TEA domain family member 2), the Notch pathway [cellular-MYC, cAMP response element-binding binding protein (CREBBP), E1A-associated cellular p300 transcriptional co-activator protein and F-Box and WD repeat domain containing 7] and the nuclear factor erythroid 2-related factor 2 (NRF2) pathway [NRF2, kelch-like ECH-associated protein 1 (KEAP1), AKT and PIK3-catalytic subunit α]. Tumor samples from patients diagnosed with various stages of CC, including cervical intraepithelial neoplasia (CIN) 1, CIN 2, CIN 3, in situ CC and invasive CC, were analyzed. The mRNA expression levels were analyzed using reverse transcription-quantitative PCR assays, whereas protein expression levels were assessed through immunohistochemical tissue microarrays. High mRNA expression levels of c-MYC and AKT and low expression levels of NRF2 and KEAP1 were associated with a decreased survival time of patients with CC. Additionally, increased expression levels of c-MYC were detected in the invasive CC stage. At the protein level, increased NRF2 expression levels were observed in all five stages of CC samples compared with those in the cancer-free control samples. AKT1 was found to be dysregulated in the CIN 1 and CIN 2 stages, PI3K in the in situ and invasive stages, and CREBBP in the CIN 3 and in situ stages. In summary, the present study demonstrated significant changes in proteins of the Notch and NRF2 pathways in CC. NRF2 was overexpressed in all cervical cancer stages (cervical intraepithelial neoplasia, in situ CC and invasive CC). The present study makes an important contribution to the possible biomarker proteins to be analyzed for the presence of premalignant and malignant lesions in the cervix.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA