Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Int J Mol Sci ; 24(15)2023 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-37569598

RESUMEN

One of the leading causes of death worldwide, in both men and women, is cancer. Despite the significant development in therapeutic strategies, the inevitable emergence of drug resistance limits the success and impedes the curative outcome. Intrinsic and acquired resistance are common mechanisms responsible for cancer relapse. Several factors crucially regulate tumourigenesis and resistance, including physical barriers, tumour microenvironment (TME), heterogeneity, genetic and epigenetic alterations, the immune system, tumour burden, growth kinetics and undruggable targets. Moreover, transforming growth factor-beta (TGF-ß), Notch, epidermal growth factor receptor (EGFR), integrin-extracellular matrix (ECM), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), phosphoinositol-3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR), wingless-related integration site (Wnt/ß-catenin), Janus kinase/signal transducers and activators of transcription (JAK/STAT) and RAS/RAF/mitogen-activated protein kinase (MAPK) signalling pathways are some of the key players that have a pivotal role in drug resistance mechanisms. To guide future cancer treatments and improve results, a deeper comprehension of drug resistance pathways is necessary. This review covers both intrinsic and acquired resistance and gives a comprehensive overview of recent research on mechanisms that enable cancer cells to bypass barriers put up by treatments, and, like "satellite navigation", find alternative routes by which to carry on their "journey" to cancer progression.


Asunto(s)
Antineoplásicos , Fosfatidilinositol 3-Quinasas , Masculino , Humanos , Femenino , Recurrencia Local de Neoplasia , Transducción de Señal , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Resistencia a Medicamentos , Microambiente Tumoral
2.
J Pathol ; 232(5): 566-77, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24407904

RESUMEN

Genomic changes affecting tumour suppressor genes are fundamental to cancer. We applied SNP array analysis to a panel of testicular germ cell tumours to search for novel tumour suppressor genes and identified a frequent small deletion on 6q25.3 affecting just one gene, ZDHHC14. The expression of ZDHHC14, a putative protein palmitoyltransferase with unknown cellular function, was decreased at both RNA and protein levels in testicular germ cell tumours. ZDHHC14 expression was also significantly decreased in a panel of prostate cancer samples and cell lines. In addition to our findings of genetic and protein expression changes in clinical samples, inducible overexpression of ZDHHC14 led to reduced cell viability and increased apoptosis through the classic caspase-dependent apoptotic pathway and heterozygous knockout of ZDHHC14 increased [CORRECTED] cell colony formation ability. Finally, we confirmed our in vitro findings of the tumour suppressor role of ZDHHC14 in a mouse xenograft model, showing that overexpression of ZDHHC14 inhibits tumourigenesis. Thus, we have identified a novel tumour suppressor gene that is commonly down-regulated in testicular germ cell tumours and prostate cancer, as well as given insight into the cellular functional role of ZDHHC14, a potential protein palmitoyltransferase that may play a key protective role in cancer.


Asunto(s)
Aciltransferasas/genética , Genes Supresores de Tumor , Neoplasias de Células Germinales y Embrionarias/genética , Neoplasias de la Próstata/genética , Neoplasias Testiculares/genética , Aciltransferasas/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Regulación hacia Abajo , Eliminación de Gen , Perfilación de la Expresión Génica/métodos , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias de Células Germinales y Embrionarias/enzimología , Neoplasias de Células Germinales y Embrionarias/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo de Nucleótido Simple , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/prevención & control , Interferencia de ARN , Neoplasias Testiculares/enzimología , Neoplasias Testiculares/patología , Factores de Tiempo , Transfección , Carga Tumoral
3.
Clin Sci (Lond) ; 127(4): 217-31, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24780002

RESUMEN

FGFR (fibroblast growth factor receptor) signalling plays critical roles in embryogensis, adult physiology, tissue repair and many pathologies. Of particular interest over recent years, it has been implicated in a wide range of cancers, and concerted efforts are underway to target different aspects of FGFR signalling networks. A major focus has been identifying the canonical downstream signalling pathways in cancer cells, and these are now relatively well understood. In the present review, we focus on two distinct but emerging hot topics in FGF biology: its role in stromal cross-talk during cancer progression and the potential roles of FGFR signalling in the nucleus. These neglected areas are proving to be of great interest clinically and are intimately linked, at least in pancreatic cancer. The importance of the stroma in cancer is well accepted, both as a conduit/barrier for treatment and as a target in its own right. Nuclear receptors are less acknowledged as targets, largely due to historical scepticism as to their existence or importance. However, increasing evidence from across the receptor tyrosine kinase field is now strong enough to make the study of nuclear growth factor receptors a major area of interest.


Asunto(s)
Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/fisiología , Animales , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Humanos , Neoplasias Pancreáticas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Microambiente Tumoral/fisiología
4.
FEBS J ; 289(12): 3440-3456, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34951738

RESUMEN

Cervical cancer is one of the most frequently diagnosed cancers in women worldwide. While cervical cancer is caused by human papillomavirus (HPV), not all females infected with HPV develop the disease, suggesting that other factors might facilitate its progression. Growing evidence supports the involvement of the fibroblast growth factor receptor (FGFR) axis in several cancers, including gynecological. However, for cervical cancer, the molecular mechanisms that underpin the disease remain poorly understood, including the role of FGFR signaling. The aim of this study was to investigate FGF(R) signaling in cervical cancer through bioinformatic analysis of cell line and patient data and through detailed expression profiling, manipulation of the FGFR axis, and downstream phenotypic analysis in cell lines (HeLa, SiHa, and CaSki). Expression (protein and mRNA) analysis demonstrated that FGFR1b/c, FGFR2b/c, FGFR4, FGF2, FGF4, and FGF7 were expressed in all three lines. Interestingly, FGFR1 and 2 localized to the nucleus, supporting that nuclear FGFRs could act as transcription factors. Importantly, 2D and 3D cell cultures demonstrated that FGFR activation can facilitate cell functions correlated with invasive disease. Collectively, this study supports an association between FGFR signaling and cervical cancer progression, laying the foundations for the development of therapeutic approaches targeting FGFR in this disease.


Asunto(s)
Factores de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos , Neoplasias del Cuello Uterino , Femenino , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Infecciones por Papillomavirus , Procesamiento Proteico-Postraduccional , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Neoplasias del Cuello Uterino/genética
5.
Cancers (Basel) ; 13(22)2021 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-34830836

RESUMEN

The pleiotropic effects of fibroblast growth factors (FGFs), the widespread expression of all seven signalling FGF receptors (FGFRs) throughout the body, and the dramatic phenotypes shown by many FGF/R knockout mice, highlight the diversity, complexity and functional importance of FGFR signalling. The FGF/R axis is critical during normal tissue development, homeostasis and repair. Therefore, it is not surprising that substantial evidence also pinpoints the involvement of aberrant FGFR signalling in disease, including tumourigenesis. FGFR aberrations in cancer include mutations, gene fusions, and amplifications as well as corrupted autocrine/paracrine loops. Indeed, many clinical trials on cancer are focusing on targeting the FGF/FGFR axis, using selective FGFR inhibitors, nonselective FGFR tyrosine kinase inhibitors, ligand traps, and monoclonal antibodies and some have already been approved for the treatment of cancer patients. The heterogeneous tumour microenvironment and complexity of FGFR signalling may be some of the factors responsible for the resistance or poor response to therapy with FGFR axis-directed therapeutic agents. In the present review we will focus on the structure and function of FGF(R)s, their common irregularities in cancer and the therapeutic value of targeting their function in cancer.

6.
Methods Mol Biol ; 1487: 255-267, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27924573

RESUMEN

Organotypic models are 3D in vitro representations of an in vivo environment. Their complexity can range from an epidermal replica to the establishment of a cancer microenvironment. These models have been used for many years, in an attempt to mimic the structure and function of cells and tissues found inside the body. Methods for developing 3D organotypic models differ according to the tissue of interest and the experimental design. For example, cultures may be grown submerged in culture medium and or at an air-liquid interface. Our group is focusing on an air-liquid interface 3D organotypic model. These cultures are grown on a nylon membrane-covered metal grid with the cells embedded in a Collagen-Matrigel gel. This allows cells to grow in an air-liquid interface to enable diffusion and nourishment from the medium below. Subsequently, the organotypic cultures can be used for immunohistochemical staining of various components of ERK signaling, which is a key player in mediating communication between cells and their microenvironment.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Modelos Biológicos , Técnicas de Cultivo de Órganos , Transducción de Señal , Células Cultivadas , Células Epiteliales , Humanos , Inmunohistoquímica
7.
Clin Cancer Res ; 11(15): 5381-9, 2005 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16061851

RESUMEN

PURPOSE: Ion channel activity is involved in several basic cellular behaviors that are integral to metastasis (e.g., proliferation, motility, secretion, and invasion), although their contribution to cancer progression has largely been ignored. The purpose of this study was to investigate voltage-gated Na(+) channel (VGSC) expression and its possible role in human breast cancer. EXPERIMENTAL DESIGN: Functional VGSC expression was investigated in human breast cancer cell lines by patch clamp recording. The contribution of VGSC activity to directional motility, endocytosis, and invasion was evaluated by in vitro assays. Subsequent identification of the VGSC alpha-subunit(s) expressed in vitro was achieved using reverse transcription-PCR, immunocytochemistry, and Western blot techniques and used to investigate VGSCalpha expression and its association with metastasis in vivo. RESULTS: VGSC expression was significantly up-regulated in metastatic human breast cancer cells and tissues, and VGSC activity potentiated cellular directional motility, endocytosis, and invasion. Reverse transcription-PCR revealed that Na(v)1.5, in its newly identified "neonatal" splice form, was specifically associated with strong metastatic potential in vitro and breast cancer progression in vivo. An antibody specific for this form confirmed up-regulation of neonatal Na(v)1.5 protein in breast cancer cells and tissues. Furthermore, a strong correlation was found between neonatal Na(v)1.5 expression and clinically assessed lymph node metastasis. CONCLUSIONS: Up-regulation of neonatal Na(v)1.5 occurs as an integral part of the metastatic process in human breast cancer and could serve both as a novel marker of the metastatic phenotype and a therapeutic target.


Asunto(s)
Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica , Canales de Sodio/biosíntesis , Canales de Sodio/fisiología , Secuencia de Aminoácidos , Biopsia , Western Blotting , Mama/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Electrofisiología , Endocitosis , Células Epiteliales/citología , Humanos , Inmunohistoquímica , Técnicas In Vitro , Iones , Metástasis Linfática , Datos de Secuencia Molecular , Canal de Sodio Activado por Voltaje NAV1.5 , Invasividad Neoplásica , Metástasis de la Neoplasia , Técnicas de Placa-Clamp , Fenotipo , Isoformas de Proteínas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tetrodotoxina/farmacología , Regulación hacia Arriba
8.
J Neurosci Methods ; 147(2): 88-98, 2005 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-16111763

RESUMEN

Voltage-gated Na(+) channel (VGSC) diversity is achieved through a number of mechanisms: multiple subunits, multiple genes encoding the pore-forming VGSC alpha-subunit and multiple gene isoforms generated by alternative splicing. A major type of VGSCalpha alternative splicing is in D1:S3, which has been proposed to be developmentally regulated. We recently reported a D1:S3 spliced form of Na(v)1.5 in human metastatic breast cancer cells. This novel 'neonatal' isoform differs from the counterpart 'adult' form at seven amino acids (in the extracellular loop between S3-S4 of D1). Here, we generated an anti-peptide polyclonal antibody, named NESOpAb, which specifically recognised 'neonatal' but not 'adult' Na(v)1.5 when tested on cells specifically over-expressing one or other of these Na(v)1.5 spliced forms. The antibody was used to investigate developmental expression of 'neonatal' Na(v)1.5 (nNa(v)1.5) in a range of mouse tissues by immunohistochemistry. Overall, the results were consistent with nNa(v)1.5 protein being more abundantly expressed in selected tissues (particularly heart and brain) from neonate as compared to adult animals. Importantly, NESOpAb blocked functional nNa(v)1.5 ion conductance when applied extracellularly at concentrations as low as 0.05 ng/ml. Possible biological and clinical applications of NESOpAb are discussed.


Asunto(s)
Empalme Alternativo/fisiología , Anticuerpos/inmunología , Especificidad de Anticuerpos , Proteínas Musculares/inmunología , Canales de Sodio/inmunología , Animales , Animales Recién Nacidos , Western Blotting/métodos , Línea Celular , Clonación Molecular , Estimulación Eléctrica/métodos , Humanos , Inmunohistoquímica/métodos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Potenciales de la Membrana/fisiología , Ratones , Proteínas Musculares/química , Proteínas Musculares/genética , Mutagénesis/fisiología , Canal de Sodio Activado por Voltaje NAV1.5 , Técnicas de Placa-Clamp/métodos , Isoformas de Proteínas/química , Isoformas de Proteínas/inmunología , ARN Mensajero/biosíntesis , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Canales de Sodio/química , Canales de Sodio/genética , Transfección/métodos
9.
FEBS Lett ; 569(1-3): 191-4, 2004 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-15225632

RESUMEN

Whole-cell patch-clamp recordings showed that a sub-population (10%) of Jurkat cells, a model of human T-cells, expressed a functional voltage-gated sodium channel, which was tetrodotoxin (TTX)-resistant. Expression of voltage-gated sodium channel protein was confirmed by western blots. Semi-quantitative PCR analysis revealed that mRNAs for the alpha-subunits of multiple voltage-gated sodium channel subtypes were present but indicated that Na(v)1.5 was the predominant subtype, consistent with the TTX-resistant nature of the recorded currents. Importantly, 10 microM TTX reduced the number of Jurkat cells invading a Matrigel basement membrane by 93.0+/-5.5%. Since similar sodium channels have also been detected in normal human T-lymphocytes, it is concluded that the activity of voltage-gated sodium channels could represent a novel mechanism potentiating the invasive capacity of these cells.


Asunto(s)
Canales de Sodio/fisiología , Linfocitos T/fisiología , Humanos , Células Jurkat , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Invasividad Neoplásica , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , ARN Neoplásico/genética , ARN Neoplásico/aislamiento & purificación , Canales de Sodio/genética , Linfocitos T/inmunología , Tetrodotoxina/farmacología
10.
EMBO Mol Med ; 6(4): 467-81, 2014 04.
Artículo en Inglés | MEDLINE | ID: mdl-24503018

RESUMEN

Pancreatic cancer is characterised by desmoplasia, driven by activated pancreatic stellate cells (PSCs). Over-expression of FGFs and their receptors is a feature of pancreatic cancer and correlates with poor prognosis, but whether their expression impacts on PSCs is unclear. At the invasive front of human pancreatic cancer, FGF2 and FGFR1 localise to the nucleus in activated PSCs but not cancer cells. In vitro, inhibiting FGFR1 and FGF2 in PSCs, using RNAi or chemical inhibition, resulted in significantly reduced cell proliferation, which was not seen in cancer cells. In physiomimetic organotypic co-cultures, FGFR inhibition prevented PSC as well as cancer cell invasion. FGFR inhibition resulted in cytoplasmic localisation of FGFR1 and FGF2, in contrast to vehicle-treated conditions where PSCs with nuclear FGFR1 and FGF2 led cancer cells to invade the underlying extra-cellular matrix. Strikingly, abrogation of nuclear FGFR1 and FGF2 in PSCs abolished cancer cell invasion. These findings suggest a novel therapeutic approach, where preventing nuclear FGF/FGFR mediated proliferation and invasion in PSCs leads to disruption of the tumour microenvironment, preventing pancreatic cancer cell invasion.


Asunto(s)
Núcleo Celular/enzimología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Neoplasias Pancreáticas/enzimología , Células Estrelladas Pancreáticas/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proliferación Celular , Factor 2 de Crecimiento de Fibroblastos/genética , Humanos , Invasividad Neoplásica , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/fisiopatología , Células Estrelladas Pancreáticas/enzimología , Transporte de Proteínas , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Microambiente Tumoral
11.
J Cell Biol ; 197(6): 801-17, 2012 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-22665522

RESUMEN

FGF-10 and its receptors, FGFR1 and FGFR2, have been implicated in breast cancer susceptibility and progression, suggesting that fibroblast growth factor (FGF) signaling may be co-opted by breast cancer cells. We identify a novel pathway downstream of FGFR1 activation, whereby the receptor is cleaved and traffics to the nucleus, where it can regulate specific target genes. We confirm Granzyme B (GrB) as the protease responsible for cleavage and show that blocking GrB activity stopped FGFR1 trafficking to the nucleus and abrogates the promigratory effect of FGF stimulation. We confirm the in vivo relevance of our findings, showing that FGFR1 localized to the nucleus specifically in invading cells in both clinical material and a three-dimensional model of breast cancer. We identify target genes for FGFR1, which exert significant effects on cell migration and may represent an invasive signature. Our experiments identify a novel mechanism by which FGF signaling can regulate cancer cell behavior and provide a novel therapeutic target for treatment of invasive breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Núcleo Celular/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , División Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Transporte de Proteínas , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/análisis , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética
12.
PLoS One ; 7(6): e39436, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22737238

RESUMEN

Fibroblast Growth Factors play critical roles during development, tissue homeostasis and repair by controlling cell proliferation, survival, migration and differentiation. Of the 22 mammalian FGFs, FGF22, a member of the FGF7/10/22 subfamily, has been shown to have a clear role in synaptogenesis, but its roles in other tissues have not been studied. We have investigated the in vivo functions of FGF22 in mice. Fgf22 null animals were viable, fertile and did not display any obvious abnormalities. Despite the known expression profile of FGF22 in the skin, no differences in either skin or pelage were observed, demonstrating that FGF22 is dispensable during embryogenesis and in unchallenged adult skin. Mice lacking FGF22 were able to heal acute wounds just as efficiently as wild type mice. However, classical two-step skin carcinogenesis challenge revealed that FGF22 null mice developed fewer papillomas than wild type controls, suggesting a potential pro-oncogenic role for FGF22 in the skin.


Asunto(s)
Factores de Crecimiento de Fibroblastos/fisiología , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Animales , Femenino , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Heterocigoto , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Neoplasias/metabolismo , Papiloma/metabolismo , Piel/patología , Cicatrización de Heridas
13.
Int J Biochem Cell Biol ; 42(2): 346-58, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19948241

RESUMEN

Voltage-gated Na(+) channels (VGSCs) are expressed in excitable cells (e.g. neurons and muscles), as well as in some classically 'non-excitable' cells (e.g. fibroblasts), and in carcinomas. In general, functional expression of VGSCs in plasma membrane (PM) is hierarchical and dynamic. Previously, we have shown that an activity-dependent positive feedback mechanism involving cAMP-dependent protein kinase A (PKA) plays a significant role in upregulation of VGSCs in strongly metastatic rat prostate cancer Mat-LyLu cells expressing Nav1.7. Here, we investigated the possible role of PKA in VGSC regulation and its functional consequences in strongly metastatic human breast cancer (BCa) MDA-MB-231 cells, where the neonatal splice form of Nav1.5 (nNav1.5) is the predominant VGSC present. Treatment with the PKA activator forskolin for 24h increased mRNA and PM protein levels of nNav1.5, without changing the total VGSC protein level. Opposite effects were obtained by application of the PKA inhibitor KT5720 or the highly specific VGSC blocker tetrodotoxin (TTX), the latter implying activity-dependent upregulation. We tested the possibility, therefore, that the activity dependence of VGSC (nNav1.5) expression involved PKA. Indeed, TTX pretreatment reduced the level of phosphorylated PKA and eliminated basal and PKA-stimulated cellular migration. These data suggested that activity-dependent positive feedback mediated by PKA plays an important role in the functional expression of nNav1.5 in BCa, and in turn, this enhances the cells' metastatic potential.


Asunto(s)
Neoplasias de la Mama/patología , Movimiento Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Retroalimentación Fisiológica , Regulación Neoplásica de la Expresión Génica , Proteínas Musculares/genética , Canales de Sodio/genética , Animales , Neoplasias de la Mama/genética , Carbazoles/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Colforsina/farmacología , Humanos , Recién Nacido , Canal de Sodio Activado por Voltaje NAV1.5 , Invasividad Neoplásica , Fosforilación/efectos de los fármacos , Pirroles/farmacología , Ratas , Células del Estroma/patología , Tetrodotoxina/farmacología , Regulación hacia Arriba/efectos de los fármacos
14.
Eur J Cell Biol ; 88(7): 371-84, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19410332

RESUMEN

FGF-10 plays an important role in development and disease, acting as the key ligand for FGFR2B to regulate cell proliferation, migration and differentiation. Aberrant FGF signalling is implicated in tumourigenesis, with several cancer studies reporting FGF-10 or FGFR2B upregulation or identifying activating mutations in Fgfr2. We used 5' RACE to identify a novel transcription start site for murine Fgf-10. Conventional in silico analysis predicted multiple binding sites for the transcription factor PEA3 upstream of this site. Binding was confirmed by chromatin immunopreciptation, and functional significance was studied by both RNAi knockdown and transient over-expression of PEA3. Knockdown of PEA3 message led to increased Fgf-10 expression, whereas overexpression of PEA3 resulted in decreased Fgf-10 expression. Thus, we have identified PEA3 as a negative regulator of Fgf-10 expression in a murine cell line and confirmed that activity also is seen in human breast cancer cell lines (MCF-7 and MDA-MB-231). Furthermore, over-expression of PEA3 in these cells resulted in impaired cell migration, which was rescued by treatment with FGF-10. Thus, PEA3 can regulate the transcription of Fgf-10 and such modulation can control breast cancer cell behaviour.


Asunto(s)
Factor 10 de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular , Movimiento Celular , Elementos de Facilitación Genéticos , Femenino , Factor 10 de Crecimiento de Fibroblastos/genética , Humanos , Ratones , Regiones Promotoras Genéticas , Interferencia de ARN , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Transcripción/genética , Sitio de Iniciación de la Transcripción
15.
Int J Biochem Cell Biol ; 41(5): 1216-27, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19041953

RESUMEN

Voltage-gated Na(+) channels (VGSCs), predominantly the 'neonatal' splice form of Na(v)1.5 (nNa(v)1.5), are upregulated in metastatic breast cancer (BCa) and potentiate metastatic cell behaviours. VGSCs comprise one pore-forming alpha subunit and one or more beta subunits. The latter modulate VGSC expression and gating, and can function as cell adhesion molecules of the immunoglobulin superfamily. The aims of this study were (1) to determine which beta subunits were expressed in weakly metastatic MCF-7 and strongly metastatic MDA-MB-231 human BCa cells, and (2) to investigate the possible role of beta subunits in adhesion and migration. In both cell lines, the beta subunit mRNA expression profile was SCN1B (encoding beta1)>>SCN4B (encoding beta4)>SCN2B (encoding beta2); SCN3B (encoding beta3) was not detected. MCF-7 cells had much higher levels of all beta subunit mRNAs than MDA-MB-231 cells, and beta1 mRNA was the most abundant. Similarly, beta1 protein was strongly expressed in MCF-7 and barely detectable in MDA-MB-231 cells. In MCF-7 cells transfected with siRNA targeting beta1, adhesion was reduced by 35%, while migration was increased by 121%. The increase in migration was reversed by tetrodotoxin (TTX). In addition, levels of nNa(v)1.5 mRNA and protein were increased following beta1 down-regulation. Stable expression of beta1 in MDA-MB-231 cells increased functional VGSC activity, process length and adhesion, and reduced lateral motility and proliferation. We conclude that beta1 is a novel cell adhesion molecule in BCa cells and can control VGSC (nNa(v)1.5) expression and, concomitantly, cellular migration.


Asunto(s)
Neoplasias de la Mama/metabolismo , Moléculas de Adhesión Celular/metabolismo , Activación del Canal Iónico/fisiología , Canales de Sodio/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Moléculas de Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular/fisiología , Femenino , Humanos , Activación del Canal Iónico/genética , Canales de Sodio/genética , Células Tumorales Cultivadas
16.
Fibrogenesis Tissue Repair ; 1(1): 8, 2008 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-19077226

RESUMEN

The advent of co-culture approaches has allowed researchers to more accurately model the behaviour of epithelial cells in cell culture studies. The initial work on epidermal modelling allowed the development of reconstituted epidermis, growing keratinocytes on top of fibroblasts seeded in a collagen gel at an air-liquid interface to generate terminally differentiated 'skin equivalents'. In addition to developing ex vivo skin sheets for the treatment of burns victims, such cultures have also been used as a means of investigating both the development and repair of the epidermis, in more relevant conditions than simple two-dimensional culture, but without the use of animals. More recently, by varying the cell types used and adjusting the composition of the matrix components, this physiological system can be adapted to allow the study of interactions between tumour cells and their surrounding stroma, particularly with regards to how such interactions regulate invasion. Here we provide a summary of the major themes involved in tumour progression and consider the evolution of the approaches used to study cancer cell behaviour. Finally, we review how organotypic models have facilitated the study of several key pathways in cancer development and invasion, and speculate on the exciting future roles for these models in cancer research.

17.
Breast Cancer Res Treat ; 101(2): 149-60, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16838113

RESUMEN

Upregulation of functional voltage-gated Na+ channels (VGSCs) occurs in metastatic human breast cancer (BCa) in vitro and in vivo. The present study aimed to ascertain the specific involvement of the "neonatal" splice variant of Nav1.5 (nNav1.5), thought to be predominant, in the VGSC-dependent invasive behaviour of MDA-MB-231 cells. Functional activity of nNav1.5 was suppressed by two different methods targeting nNav1.5: (i) small interfering RNA (siRNA), and (ii) a polyclonal antibody (NESO-pAb); effects upon migration and invasion were determined. nNav1.5 mRNA, protein and signalling were measured using real-time PCR, Western blotting, and patch clamp recording, respectively. Treatment with the siRNA rapidly reduced (by approximately 90%) the level of nNav1.5 (but not adult Nav1.5) mRNA, but the protein reduction was much smaller (approximately 30%), even after 13 days. Nevertheless, the siRNA reduced peak VGSC current density by 33%, and significantly increased the cells' sensitivity to nanomolar tetrodotoxin (TTX). Importantly, the siRNA suppressed in vitro migration by 43%, and eliminated the normally inhibitory effect of TTX. Migrated MDA-MB-231 cells expressed more nNav1.5 protein at the plasma membrane than non-migrated cells. Furthermore, NESO-pAb reduced migration by up to 42%, in a dose-dependent manner. NESO-pAb also reduced Matrigel invasion without affecting proliferation. TTX had no effect on cells already treated with NESO-pAb. It was concluded that nNav1.5 is primarily responsible for the VGSC-dependent enhancement of invasive behaviour in MDA-MB-231 cells. Accordingly, targeting nNav1.5 expression/activity may be useful in clinical management of metastatic BCa.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteínas Musculares/metabolismo , Invasividad Neoplásica , Canales de Sodio/metabolismo , Western Blotting , Neoplasias de la Mama/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Técnicas In Vitro , Recién Nacido , Microscopía Confocal , Proteínas Musculares/efectos de los fármacos , Proteínas Musculares/genética , Canal de Sodio Activado por Voltaje NAV1.5 , Invasividad Neoplásica/genética , Técnicas de Placa-Clamp , Isoformas de Proteínas , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/efectos de los fármacos , Canales de Sodio/genética , Tetrodotoxina/farmacología
18.
EMBO J ; 26(5): 1268-78, 2007 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-17304214

RESUMEN

The epithelial isoform of fibroblast growth factor receptor 2 (Fgfr2b) is essential for embryogenesis, and Fgfr2b-null mice die at birth. Using Cre-Lox transgenics to delete Fgfr2b in cells expressing keratin 5, we show that mice lacking epidermal Fgfr2b survive into adulthood but display striking abnormalities in hair and sebaceous gland development. Epidermal hyperthickening develops with age, and 10% of mutant mice develop spontaneous papillomas, demonstrating the role of Fgfr2b in post-natal skin development and in adult skin homeostasis. Mice lacking epithelial Fgfr2b show great sensitivity to chemical carcinogenic insult, displaying several oncogenic ha-ras mutations with dramatic development of papillomas and squamous cell carcinomas. Mutant mice have increased inflammation in the skin, with increased numbers of macrophages and gammadeltaT cells with abnormal morphology. Mutant skin shows several changes in gene expression, including enhanced expression of the pro-inflammatory cytokine interleukin 18 and decreased expression of Serpin a3b, a potential tumor suppressor. Thus we describe a novel role of Fgfr2b and provide the first evidence of a tyrosine kinase receptor playing a tumor suppressive role in the skin.


Asunto(s)
Homeostasis/fisiología , Neoplasias Experimentales/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/fisiología , Piel/metabolismo , Animales , Carcinoma de Células Escamosas/química , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Epidermis/metabolismo , Epidermis/patología , Femenino , Cabello/metabolismo , Cabello/patología , Folículo Piloso/metabolismo , Folículo Piloso/patología , Inmunohistoquímica , Queratina-5/genética , Queratina-5/metabolismo , Masculino , Ratones , Ratones Noqueados , Mutación , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Papiloma/metabolismo , Papiloma/patología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Glándulas Sebáceas/metabolismo , Glándulas Sebáceas/patología , Piel/patología , Piel/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA