Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Nucleic Acids Res ; 47(19): 10247-10266, 2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31504805

RESUMEN

The anti-apoptotic BAG-1 protein isoforms are known to be overexpressed in colorectal tumors and are considered to be potential therapeutic targets. The isoforms are derived from alternative translation initiations occuring at four in-frame start codons of a single mRNA transcript. Its 5'UTR also contains an internal ribosome entry site (IRES) regulating the cap-independent translation of the transcript. An RNA G-quadruplex (rG4) is located at the 5'end of the BAG-1 5'UTR, upstream of the known cis-regulatory elements. Herein, we observed that the expression of BAG-1 isoforms is post-transcriptionally regulated in colorectal cancer cells and tumors, and that stabilisation of the rG4 by small molecules ligands reduces the expression of endogenous BAG-1 isoforms. We demonstrated a critical role for the rG4 in the control of both cap-dependent and independent translation of the BAG-1 mRNA in colorectal cancer cells. Additionally, we found an upstream ORF that also represses BAG-1 mRNA translation. The structural probing of the complete 5'UTR showed that the rG4 acts as a steric block which controls the initiation of translation at each start codon of the transcript and also maintains the global 5'UTR secondary structure required for IRES-dependent translation.


Asunto(s)
Proteínas de Unión al ADN/genética , G-Cuádruplex , Biosíntesis de Proteínas , Factores de Transcripción/genética , Regiones no Traducidas 5'/genética , Apoptosis/genética , Codón Iniciador/genética , Proteínas de Unión al ADN/química , Regulación de la Expresión Génica/genética , Humanos , Sitios Internos de Entrada al Ribosoma/genética , Ligandos , Iniciación de la Cadena Peptídica Traduccional/genética , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Estructura Secundaria de Proteína , Proteínas de Unión a Caperuzas de ARN/genética , ARN Mensajero/genética , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción/química
2.
J Cell Sci ; 131(13)2018 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-29898915

RESUMEN

HNF4α is a key nuclear receptor for regulating gene expression in the gut. Although both P1 and P2 isoform classes of HNF4α are expressed in colonic epithelium, specific inhibition of P1 isoforms is commonly found in colorectal cancer. Previous studies have suggested that P1 and P2 isoforms might regulate different cellular functions. Despite these advances, it remains unclear whether these isoform classes are functionally divergent in the context of human biology. Here, the consequences of specific inhibition of P1 or P2 isoform expression was measured in a human colorectal cancer cell transcriptome. Results indicate that P1 isoforms were specifically associated with the control of cell metabolism, whereas P2 isoforms globally supported aberrant oncogenic signalization, promoting cancer cell survival and progression. P1 promoter-driven isoform expression was found to be repressed by ß-catenin, one of the earliest oncogenic pathways to be activated during colon tumorigenesis. These findings identify a novel cascade by which the expression of P1 isoforms is rapidly shut down in the early stages of colon tumorigenesis, allowing a change in HNF4α-dependent transcriptome, thereby promoting colorectal cancer progression.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Factor Nuclear 4 del Hepatocito/genética , Regiones Promotoras Genéticas , beta Catenina/genética , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 4 del Hepatocito/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transcriptoma , beta Catenina/metabolismo
3.
J Cell Physiol ; 234(5): 6731-6745, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30273442

RESUMEN

The Ras/mitogen-activated protein kinase (MAPK) pathway controls fundamental cellular processes such as proliferation, differentiation, and apoptosis. The dual-specificity phosphatase 6 (DUSP6) regulates cytoplasmic MAPK signaling by dephosphorylating and inactivating extracellular signal-regulated kinase (ERK1/2) MAPK. To determine the role of DUSP6 in the maintenance of intestinal homeostasis, we characterized the intestinal epithelial phenotype of Dusp6 knockout (KO) mice under normal, oncogenic, and proinflammatory conditions. Our results show that loss of Dusp6 increased crypt depth and epithelial cell proliferation without altering colonic architecture. Crypt regeneration capacity was also enhanced, as revealed by ex vivo Dusp6 KO organoid cultures. Additionally, loss of Dusp6 induced goblet cell expansion without affecting enteroendocrine and absorptive cell differentiation. Our data also demonstrate that Dusp6 KO mice were protected from acute dextran sulfate sodium-induced colitis, as opposed to wild-type mice. In addition, Dusp6 gene deletion markedly enhanced tumor load in Apc Min/+ mice. Decreased DUSP6 expression by RNA interference in HT29 colorectal cancer cells enhanced ERK1/2 activation levels and promoted both anchorage-independent growth in soft agar as well as invasion through Matrigel. Finally, DUSP6 mRNA expression in human colorectal tumors was decreased in advanced stage tumors compared with paired normal tissues. These results demonstrate that DUSP6 phosphatase, by controlling ERK1/2 activation, regulates colonic inflammatory responses, and protects the intestinal epithelium against oncogenic stress.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Transformación Celular Neoplásica/metabolismo , Colon/patología , Neoplasias Colorrectales/metabolismo , Fosfatasa 6 de Especificidad Dual/metabolismo , Animales , Apoptosis/fisiología , Colitis/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Sulfato de Dextran , Fosfatasa 6 de Especificidad Dual/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Mucosa Intestinal/metabolismo , Ratones Transgénicos
4.
Biochim Biophys Acta Mol Basis Dis ; 1864(5 Pt A): 1539-1551, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29454075

RESUMEN

Colorectal tumors are immersed in an array of tumor-promoting factors including extracellular nucleotides such as uridine 5'­diphosphate (UDP). UDP is the endogenous agonist of the G protein-coupled P2Y6 receptor (P2Y6R), which may contribute to the formation of a tumor-promoting microenvironment by coordinating resistance to apoptosis. Colorectal cancer (CRC) was chemically induced in P2ry6 knockout (P2ry6-/-) mice using azoxymethane and dextran sulfate sodium challenges. Mice were euthanatized and their tumor load determined. Fixed tissues were stained for histological and immunohistochemistry analysis. Tumoroids were also prepared from CRC tumors resected from P2ry6+/+ mice to determine the role of P2Y6R in resistance to apoptosis, whereas HT29 carcinoma cells were used to elucidate the signaling mechanism involved in P2Y6R anti-apoptotic effect. P2ry6-/- mice developed a reduced number of colorectal tumors with apparent tumors having smaller volumes. Overall dysplastic score was significantly lower in P2ry6-/- animals. Stimulation of P2Y6R with the selective agonist MRS2693 protected HT-29 cells from TNFα-induced apoptosis. This protective effect was mediated by the stabilizing phosphorylation of the X-linked inhibitor of apoptosis protein (XIAP) by AKT. Using CRC-derived tumoroids, P2Y6R activation was found to contribute to chemoresistance since addition of the P2Y6R agonist MRS2693 significantly prevented the cytotoxic effect of 5-fluorouracil. The present study shows that sustained activation of P2Y6R may contribute to intestinal tumorigenesis by blocking the apoptotic process and by contributing to chemoresistance, a substantial concern in the treatment of patients with CRC. These results suggest that P2Y6R may represent a prime target for reducing colorectal carcinogenesis.


Asunto(s)
Apoptosis , Transformación Celular Neoplásica/metabolismo , Neoplasias Colorrectales/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores Purinérgicos P2/metabolismo , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Receptores Purinérgicos P2/genética , Factor de Necrosis Tumoral alfa/metabolismo
5.
FASEB J ; 31(8): 3512-3526, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28465325

RESUMEN

Shp-1 (Src homology region 2 domain-containing protein tyrosine phosphatase-1) is a phosphatase that is highly expressed in hematopoietic and epithelial cells. Whereas its function is largely characterized in hematopoietic cells, its role in epithelial cells, such as intestinal epithelial cells (IECs), is not well known. Here, we generated mice with an IEC-specific knockout of Shp-1 (Src homology region 2 domain-containing phosphatase-1; Shp-1IEC-KO). We showed that the loss of epithelial Shp-1 leads to an intestinalomegaly that is associated with an increase in epithelial cell proliferation and size. Histologic analysis demonstrates significant perturbation of the crypt-villus architecture with an apparent increase in the number of goblet and Paneth cells and increased expression of their respective markers {Muc2 (mucin 2), αDef, and Sox9 [SRY (sex determining region Y)-box 9]}. Expansion of intermediate cells-common progenitors of goblet and Paneth cell lineages-is also observed in Shp-1IEC-KO mice. Although sustained activation of Wnt/ß-catenin and PI3K/Akt/mammalian target of rapamycin signaling is observed, Shp-1IEC-KO mice fail to develop any intestinal tumors after 15 mo; however, the loss of Shp-1 in IECs markedly enhances tumor load ApcMin/+ mice. These findings show a novel role for Shp-1 in the regulation of IEC growth and secretory lineage allocation, possibly via modulation of PI3K/Akt-dependent signaling pathways. Finally, Shp-1 does not function as a classic tumor suppressor gene in the intestinal epithelium.-Leblanc, C., Langlois, M.-J., Coulombe, G., Vaillancourt-Lavigueur, V., Jones, C., Carrier, J. C., Boudreau, F., Rivard, N. Epithelial Src homology region 2 domain-containing phosphatase-1 restrains intestinal growth, secretory cell differentiation, and tumorigenesis.


Asunto(s)
Neoplasias del Colon/metabolismo , Regulación de la Expresión Génica/fisiología , Intestinos/crecimiento & desarrollo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Animales , Cateninas/genética , Cateninas/metabolismo , Células Epiteliales/fisiología , Humanos , Mucosa Intestinal/metabolismo , Intestinos/citología , Ratones , Ratones Noqueados , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteínas Proto-Oncogénicas c-akt , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
6.
Mol Carcinog ; 55(5): 671-87, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-25808857

RESUMEN

Cathepsin B is a cysteine proteinase that primarily functions as an endopeptidase within endolysosomal compartments in normal cells. However, during tumoral expansion, the regulation of cathepsin B can be altered at multiple levels, thereby resulting in its overexpression and export outside of the cell. This may suggest a possible role of cathepsin B in alterations leading to cancer progression. The aim of this study was to determine the contribution of intracellular and extracellular cathepsin B in growth, tumorigenesis, and invasion of colorectal cancer (CRC) cells. Results show that mRNA and activated levels of cathepsin B were both increased in human adenomas and in CRCs of all stages. Treatment of CRC cells with the highly selective and non-permeant cathepsin B inhibitor Ca074 revealed that extracellular cathepsin B actively contributed to the invasiveness of human CRC cells while not essential for their growth in soft agar. Cathepsin B silencing by RNAi in human CRC cells inhibited their growth in soft agar, as well as their invasion capacity, tumoral expansion, and metastatic spread in immunodeficient mice. Higher levels of the cell cycle inhibitor p27(Kip1) were observed in cathepsin B-deficient tumors as well as an increase in cyclin B1. Finally, cathepsin B colocalized with p27(Kip1) within the lysosomes and efficiently degraded the inhibitor. In conclusion, the present data demonstrate that cathepsin B is a significant factor in colorectal tumor development, invasion, and metastatic spreading and may, therefore, represent a potential pharmacological target for colorectal tumor therapy.


Asunto(s)
Carcinogénesis/genética , Catepsina B/genética , Catepsina B/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Animales , Células CACO-2 , Carcinogénesis/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Dipéptidos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Trasplante de Neoplasias
7.
Carcinogenesis ; 35(6): 1217-27, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24403311

RESUMEN

The integrin α6 subunit pre-messenger RNA undergoes alternative splicing to generate two different splice variants, named α6A and α6B, having distinct cytoplasmic domains. In the human colonic gland, these splice variants display different patterns of expression suggesting specific functions for each variant. We have previously found an up-regulation of the α6ß4 integrin in colon adenocarcinomas as well as an increase in the α6A/α6B ratio, but little is known about the involvement of α6Aß4 versus α6Bß4 in this context. The aim of this study was to elucidate the function of the α6Aß4 integrin in human colorectal cancer (CRC) cells. Expression studies on a panel of primary CRCs confirmed that the up-regulation of the α6 subunit in CRC is a direct consequence of the increase of the α6A variant. To investigate the functional significance of an α6A up-regulation in CRC, we specifically knocked down its expression in well-established CRC cell lines using a small-hairpin RNA approach. Results showed a growth rate reduction in all α6A knockdown CRC cell lines studied. The α6A silencing was also found to be associated with a significant repression of a number of Wnt/ß-catenin pathway end points. Moreover, it was accompanied by a reduction in the capacity of these cells to develop tumours in xenografts. Taken together, these results demonstrate that the α6A variant is a pro-proliferative form of the α6 integrin subunit in CRC cells and appears to mediate its effects through the Wnt/ß-catenin pathway.


Asunto(s)
Empalme Alternativo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Integrina alfa6/genética , Vía de Señalización Wnt , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Proteínas Dishevelled , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Xenoinjertos , Humanos , Integrina alfa6/metabolismo , Espacio Intracelular/metabolismo , Ratones , Fosfoproteínas/metabolismo , Subunidades de Proteína/genética , Transporte de Proteínas , Carga Tumoral
8.
J Cell Sci ; 125(Pt 14): 3454-63, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22467857

RESUMEN

The crypt-villus axis constitutes the functional unit of the small intestine, where mature absorptive cells are confined to the villi, and stem cells and transit amplifying and differentiating cells are restricted to the crypts. The polycomb group (PcG) proteins repress differentiation and promote self-renewal in embryonic stem cells. PcGs prevent transcriptional activity by catalysing epigenetic modifications, such as the covalent addition of methyl groups on histone tails, through the action of the polycomb repressive complex 2 (PRC2). Although a role for PcGs in the preservation of stemness characteristics is now well established, recent evidence suggests that they may also be involved in the regulation of differentiation. Using intestinal epithelial cell models that recapitulate the enterocytic differentiation programme, we generated a RNAi-mediated stable knockdown of SUZ12, which constitutes a cornerstone for PRC2 assembly and functionality, in order to analyse intestinal cell proliferation and differentiation. Expression of SUZ12 was also investigated in human intestinal tissues, revealing the presence of SUZ12 in most proliferative epithelial cells of the crypt and an increase in its expression in colorectal cancers. Moreover, PRC2 disruption led to a significant precocious expression of a number of terminal differentiation markers in intestinal cell models. Taken together, our data identified a mechanism whereby PcG proteins participate in the repression of the enterocytic differentiation program, and suggest that a similar mechanism exists in situ to slow down terminal differentiation in the transit amplifying cell population.


Asunto(s)
Tracto Gastrointestinal/citología , Complejo Represivo Polycomb 2/fisiología , Células CACO-2 , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Células Cultivadas , Tracto Gastrointestinal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Histonas/genética , Histonas/metabolismo , Humanos , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Regiones Promotoras Genéticas , Transcripción Genética
9.
Am J Pathol ; 183(1): 266-76, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23680656

RESUMEN

Colorectal cancer is the second leading cause of death from cancer. Osteopontin (OPN) is a component of tumor extracellular matrix identified as a key marker of cancer progression. The estrogen-related receptor α (ERRα) has been implicated in endocrine-related cancer development and progression, possibly through modulation of cellular energy metabolism. Previous reports that ERRα regulates OPN expression in bone prompted us to investigate whether ERRα controls OPN expression in human colorectal cancer. Using a tissue microarray containing 83 tumor-normal tissue pairs of colorectal cancer samples, we found that tumor epithelial cells displayed higher staining for ERRα than normal mucosa, in correlation with elevated OPN expression. In addition, knocking down endogenous ERRα led to reduced OPN expression in HT29 colon cancer cells. Promoter analysis, inhibition of ERRα activity, and expression and mutation of potential ERRα response elements in the proximal promoter of human OPN showed that ERRα and its obligate co-activator, peroxisome proliferator-activated receptor γ co-activator-1 α, positively control human OPN promoter activity. Furthermore, chromatin immunoprecipitation experiments confirmed in vivo occupancy of the OPN promoter by ERRα in HT29 cells, suggesting that OPN is a direct target of ERRα in colorectal cancer. These findings suggest an additional mechanism by which ERRα participates in the development and progression of colorectal cancer, further supporting the relevance of targeting ERRα with antagonists as anticancer agents.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/metabolismo , Osteopontina/metabolismo , Receptores de Estrógenos/metabolismo , Anciano , Estudios de Casos y Controles , Inmunoprecipitación de Cromatina , Neoplasias Colorrectales/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Células HT29 , Humanos , Immunoblotting , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Matrices Tisulares , Regulación hacia Arriba , Receptor Relacionado con Estrógeno ERRalfa
10.
Carcinogenesis ; 34(10): 2253-61, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23720198

RESUMEN

The estrogen-related receptor alpha (ERRα) is a nuclear receptor that acts primarily as a regulator of metabolic processes, particularly in tissues subjected to high-energy demand. In addition to its control of energy metabolism and mitochondrial biogenesis, ERRα has recently been associated with cancer progression. Notably, increased expression of ERRα has been shown in several cancerous tissues, including breast, ovary and colon. However, additional studies are required to gain insight into the action of ERRα in cancer biology, particularly in non-endocrine-related cancers. Therefore, using a short hairpin RNA-mediated approach, we investigated whether ERRα is required for the rapid growth of colon cancer cells and to maintain their neoplastic metabolic state. Results show that silencing ERRα significantly impaired colon cancer cell proliferation and colony formation in vitro as well as their in vivo tumorigenic capacity. A pronounced delay in G1-to-S cell cycle phase transition was observed in ERRα-depleted cells in association with reduced cyclin-dependent kinase 2 activity and hyperphosphorylated state of the retinoblastoma protein along with disturbed expression of several cell cycle regulators, including p15 and p27. Interestingly, ERRα-depleted HCT116 cells also displayed significant reduction in expression of a large set of key genes to glycolysis, tricarboxylic acid cycle and lipid synthesis. Furthermore, using (14)C isotope tracer analysis, ERRα depletion in colon cancer cells resulted in reduced glucose incorporation and glucose-mediated lipogenesis in these cells. These findings suggest that ERRα coordinates colon cancer cell proliferation and tumorigenic capacity with energy metabolism. Thus, ERRα could represent a promising therapeutic target in colon cancer.


Asunto(s)
Neoplasias del Colon/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Estrógenos/metabolismo , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Neoplasias del Colon/genética , Metabolismo Energético/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Glucosa/metabolismo , Glucólisis/genética , Células HCT116 , Humanos , Lípidos/biosíntesis , Lipogénesis/genética , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Estrógenos/genética , Ensayo de Tumor de Célula Madre , Receptor Relacionado con Estrógeno ERRalfa
11.
BMC Cell Biol ; 14: 33, 2013 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-23919615

RESUMEN

BACKGROUND: The transcription factor E2F4 controls proliferation of normal and cancerous intestinal epithelial cells. E2F4 localization in normal human intestinal epithelial cells (HIEC) is cell cycle-dependent, being cytoplasmic in quiescent differentiated cells but nuclear in proliferative cells. However, the intracellular signaling mechanisms regulating such E2F4 localization remain unknown. RESULTS: Treatment of quiescent HIEC with serum induced ERK1/2 activation, E2F4 phosphorylation, E2F4 nuclear translocation and G1/S phase transition while inhibition of MEK/ERK signaling by U0126 prevented these events. Stimulation of HIEC with epidermal growth factor (EGF) also led to the activation of ERK1/2 but, in contrast to serum or lysophosphatidic acid (LPA), EGF failed to induce E2F4 phosphorylation, E2F4 nuclear translocation and G1/S phase transition. Furthermore, Akt and GSK3ß phosphorylation levels were markedly enhanced in serum- or LPA-stimulated HIEC but not by EGF. Importantly, E2F4 phosphorylation, E2F4 nuclear translocation and G1/S phase transition were all observed in response to EGF when GSK3 activity was concomitantly inhibited by SB216763. Finally, E2F4 was found to be overexpressed, phosphorylated and nuclear localized in epithelial cells from human colorectal adenomas exhibiting mutations in APC and KRAS or BRAF genes, known to deregulate GSK3/ß-catenin and MEK/ERK signaling, respectively. CONCLUSIONS: The present results indicate that MEK/ERK activation and GSK3 inhibition are both required for E2F4 phosphorylation as well as its nuclear translocation and S phase entry in HIEC. This finding suggests that dysregulated E2F4 nuclear localization may be an instigating event leading to hyperproliferation and hence, of tumor initiation and promotion in the colon and rectum.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Factor de Transcripción E2F4/efectos de los fármacos , Factor de Transcripción E2F4/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Sistema de Señalización de MAP Quinasas/fisiología , Mitógenos/farmacología , Transcripción Genética/efectos de los fármacos , Adenoma/metabolismo , Adenoma/patología , Butadienos/farmacología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Línea Celular , Células Cultivadas , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Factor de Transcripción E2F4/genética , Inhibidores Enzimáticos/farmacología , Factor de Crecimiento Epidérmico/farmacología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/efectos de los fármacos , Humanos , Mucosa Intestinal/efectos de los fármacos , Lisofosfolípidos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Nitrilos/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Transcripción Genética/fisiología
12.
Am J Physiol Gastrointest Liver Physiol ; 300(6): G1065-79, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21415412

RESUMEN

Bone morphogenetic protein (BMP) signaling within the gastrointestinal tract is complex. BMP ligands and their receptors are expressed in both epithelial and mesenchymal compartments, suggesting bidirectional signaling between these two entities. Despite an increasing interest in BMP signaling in gut physiology and pathologies, the distinct contribution of BMP signaling in the epithelium vs. the mesenchyme in gastrointestinal homeostasis remains to be established. We aimed to investigate the role of epithelial BMP signaling in gastric organogenesis, gland morphogenesis, and maintenance of epithelial cell functions. Using the Cre/loxP system, we generated a mouse model with an early deletion during development of BMP receptor 1A (Bmpr1a) exclusively in the foregut endoderm. Bmpr1a(ΔGEC) mice showed no severe abnormalities in gastric organogenesis, gland epithelial proliferation, or morphogenesis, suggesting only a minor role for epithelial BMP signaling in these processes. However, early loss of BMP signaling in foregut endoderm did impact on gastric patterning, leading to an anteriorization of the stomach. In addition, numbers of parietal cells were reduced in Bmpr1a(ΔGEC) mice. Epithelial BMP deletion significantly increased the numbers of chromogranin A-, ghrelin-, somatostatin-, gastrin-, and serotonin-expressing gastric endocrine cells. Cancer never developed in young adult (<100 days) Bmpr1a-inactivated mice although a marker of spasmolytic polypeptide-expressing metaplasia was upregulated. Using this model, we have uncovered that BMP signaling negatively regulates the proliferation and commitment of endocrine precursor cells. Our data also indicate that loss of BMP signaling in epithelial gastric cells alone is not sufficient to induce gastric neoplasia.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Linaje de la Célula , Células Enteroendocrinas/metabolismo , Células Epiteliales/metabolismo , Mucosa Gástrica/metabolismo , Transducción de Señal , Envejecimiento , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/deficiencia , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Diferenciación Celular , Proliferación Celular , Duodeno/embriología , Duodeno/metabolismo , Células Enteroendocrinas/patología , Células Epiteliales/patología , Mucosa Gástrica/embriología , Mucosa Gástrica/patología , Factor Nuclear 3-gamma del Hepatocito/genética , Factor Nuclear 3-gamma del Hepatocito/metabolismo , Hiperplasia , Integrasas/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Morfogénesis , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
13.
Mol Carcinog ; 50(5): 359-69, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21480389

RESUMEN

Epidemiological and experimental evidence suggests that estrogen replacement therapy reduces the risk of colon cancer in postmenopausal women. Estrogen receptor beta (ERß) is thought to be the principal mediator of the estrogen effect in the colon. Recent studies by our team suggested positive regulation of the transforming growth factor (TGF)ß pathway by estrogen in mice colonocytes. We therefore wanted to investigate the effects of ERß agonist treatment on intestinal tumorigenesis in Apc(Min/+) mice. Weaned Apc(Min/+) mice were injected subcutaneously three times a week for 12 wk with vehicle or ERß-selective agonist, diarylpropionitrile (DPN, 5 mg/kg). DPN administration resulted in a significant reduction in small intestinal polyp multiplicity in both Apc(Min/+) male and female mice. Furthermore, the mean diameter of small intestinal polyps was lower in DPN-treated than vehicle-treated males, along with lower BrdU incorporation indices in jejunal and colon epithelial cells of both sexes. DPN treatment also increased apoptosis in colon epithelium as measured by TUNEL assay and cleaved caspase 3 quantification. The effect of DPN on various components of the TGFß pathway was also studied in colonocytes. DPN treatment increased expression of TGFß1 and TGFß3 transcripts, levels of nuclear and phosphorylated Smad2 as well as p27 cell-cycle inhibitor, a TGFß pathway target gene. Our results demonstrate that DPN treatment reduces intestinal tumorigenesis in Apc(Min/+) mice. Furthermore, we suggest that positive regulation of the TGFß pathway by ERß activation could contribute to the protective role of estrogen in intestinal tumor development.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Receptor beta de Estrógeno/agonistas , Neoplasias Intestinales/prevención & control , Nitrilos/farmacología , Propionatos/farmacología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Caspasa 3/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular , Colon/efectos de los fármacos , Colon/metabolismo , Colon/patología , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Epitelio/patología , Femenino , Expresión Génica/efectos de los fármacos , Etiquetado Corte-Fin in Situ , Inyecciones Subcutáneas , Neoplasias Intestinales/genética , Neoplasias Intestinales/patología , Pólipos Intestinales/genética , Pólipos Intestinales/patología , Pólipos Intestinales/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Nitrilos/administración & dosificación , Fosforilación/efectos de los fármacos , Propionatos/administración & dosificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/genética
14.
Mol Cancer ; 9: 271, 2010 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-20942929

RESUMEN

BACKGROUND: Among the most harmful of all genetic abnormalities that appear in colorectal cancer (CRC) development are mutations of KRAS and its downstream effector BRAF as they result in abnormal extracellular signal-related kinase (ERK) signaling. In a previous report, we had shown that expression of a constitutive active mutant of MEK1 (caMEK) in normal rat intestinal epithelial cells (IECs) induced morphological transformation associated with epithelial to mesenchymal transition, growth in soft agar, invasion and metastases in nude mice. Results from microarrays comparing control to caMEK-expressing IECs identified the gene encoding for serpinE2, a serine protease inhibitor, as a potential target of activated MEK1. RESULTS: 1- RT-PCR and western blot analyses confirmed the strong up-regulation of serpinE2 expression and secretion by IECs expressing oncogenic MEK, Ras or BRAF. 2- Interestingly, serpinE2 mRNA and protein were also markedly enhanced in human CRC cells exhibiting mutation in KRAS and BRAF. 3- RNAi directed against serpinE2 in caMEK-transformed rat IECs or in human CRC cell lines HCT116 and LoVo markedly decreased foci formation, anchorage-independent growth in soft agarose, cell migration and tumor formation in nude mice. 4- Treatment of CRC cell lines with U0126 markedly reduced serpinE2 mRNA levels, indicating that expression of serpinE2 is likely dependent of ERK activity. 5- Finally, Q-PCR analyses demonstrated that mRNA levels of serpinE2 were markedly increased in human adenomas in comparison to healthy adjacent tissues and in colorectal tumors, regardless of tumor stage and grade. CONCLUSIONS: Our data indicate that serpinE2 is up-regulated by oncogenic activation of Ras, BRAF and MEK1 and contributes to pro-neoplastic actions of ERK signaling in intestinal epithelial cells. Hence, serpinE2 may be a potential therapeutic target for colorectal cancer treatment.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Serpina E2/metabolismo , Animales , Western Blotting , Butadienos/farmacología , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/genética , Células HCT116 , Humanos , Técnicas In Vitro , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/metabolismo , Ratones , Ratones Desnudos , Nitrilos/farmacología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Interferencia de ARN , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serpina E2/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteínas ras/genética , Proteínas ras/metabolismo
15.
FASEB J ; 23(6): 1835-44, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19168705

RESUMEN

Phosphatase and tensin homolog (PTEN), a negative regulator of the phosphatidylinositol 3-kinase/Akt pathway, is one of the most frequently mutated/deleted tumor suppressor genes in human cancers. The aim of this study was to gain insight into the role played by PTEN in intestinal homeostasis and epithelial cell function. Using the Cre/loxP system, we have generated a mouse with a conditional intestinal epithelial Pten deficiency. Pten mutant mice and controls were sacrificed for histology, immunofluorescence, Western blot, and quantitative polymerase chain reaction analysis. Our results show that loss of epithelial Pten leads to an intestinalomegaly associated with an increase in epithelial cell proliferation. Histological analysis demonstrated significant perturbation of the crypt-villus architecture, a marked increase in goblet cells and a decrease in enteroendocrine cells, suggesting a role for Pten in the commitment of the multipotential-secretory precursor cell. Loss of epithelial Pten does not result in induction of nuclear beta-catenin protein levels, nor is it sufficient to promote tumorigenesis initiation. However, it severely enhances intestinal tumor load in Apc(Min/+) mice, in which c-Myc is already deregulated. These results reveal an unknown function for Pten signaling in the commitment of multipotential-secretory progenitor cells and suggest that epithelial Pten functions as a modifier gene in intestinal neoplasia.


Asunto(s)
Neoplasias Intestinales , Intestinos/anatomía & histología , Fosfohidrolasa PTEN/metabolismo , Animales , Células Epiteliales/citología , Células Epiteliales/fisiología , Eliminación de Gen , Expresión Génica , Genes APC , Células Caliciformes/citología , Células Caliciformes/metabolismo , Homeostasis , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/patología , Neoplasias Intestinales/genética , Neoplasias Intestinales/patología , Intestinos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/fisiología , beta Catenina/genética , beta Catenina/metabolismo
16.
BMC Infect Dis ; 10: 363, 2010 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-21192802

RESUMEN

BACKGROUND: Oral vancomycin (125 mg qid) is recommended as treatment of severe Clostridium difficile infection (CDI). Higher doses (250 or 500 mg qid) are sometimes recommended for patients with very severe CDI, without supporting clinical evidence. We wished to determine to what extent faecal levels of vancomycin vary according to diarrhoea severity and dosage, and whether it is rational to administer high-dose vancomycin to selected patients. METHODS: We recruited hospitalized adults suspected to have CDI for whom oral vancomycin (125, 250 or 500 mg qid) had been initiated. Faeces were collected up to 3 times/day and levels were measured with the AxSYM fluorescence polarization immunoassay. RESULTS: Fifteen patients (9 with confirmed CDI) were treated with oral vancomycin. Patients with ≥ 4 stools daily presented lower faecal vancomycin levels than those with a lower frequency. Higher doses of oral vancomycin (250 mg or 500 mg qid) led to consistently higher faecal levels (> 2000 mg/L), which were 3 orders of magnitude higher than the MIC90 of vancomycin against C. difficile. One patient receiving 125 mg qid had levels below 50 mg/L during the first day of treatment. CONCLUSIONS: Faecal levels of vancomycin are proportional to the dosage administered and, even in patients with increased stool frequency, much higher than the MIC90. Patients given the standard 125 mg qid dosage might have low faecal levels during the first day of treatment. A loading dose of 250 mg or 500 mg qid during the first 24-48 hours followed by the standard dosage should be evaluated in larger studies, since it might be less disruptive to the colonic flora and save unnecessary costs.


Asunto(s)
Antibacterianos/administración & dosificación , Antibacterianos/farmacocinética , Infecciones por Clostridium/tratamiento farmacológico , Heces/química , Vancomicina/administración & dosificación , Vancomicina/farmacocinética , Administración Oral , Adulto , Anciano , Anciano de 80 o más Años , Antibacterianos/análisis , Clostridioides difficile/aislamiento & purificación , Infecciones por Clostridium/microbiología , Femenino , Humanos , Masculino , Pruebas de Sensibilidad Microbiana , Persona de Mediana Edad , Vancomicina/análisis
17.
J Cell Physiol ; 221(2): 350-8, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19562678

RESUMEN

The generation of knock-out mice for E2F4 gene expression has suggested a role for this transcription factor in establishing and/or maintaining the intestinal crypt compartment. Having previously demonstrated that E2F4 is cytoplasmic in quiescent-differentiated cells but nuclear in growth factor-stimulated proliferative cells, the present study was aimed at determining the role of E2F4 in the control of human intestinal epithelial proliferation. Results herein demonstrate that lentiviral infection of an shRNA which specifically knocked-down E2F4 expression slowed down G1/S phase transition and the proliferation rate of normal human intestinal epithelial cells (HIEC) and of colon cancer cells. Protein expression of Cdk2, cyclins D1 and A, Cdc25A and c-myc was markedly down-regulated in shE2F4-expressing cells; by contrast, expression of the cell cycle inhibitors p21(Cip/Waf) and p27(Kip1) was increased. In addition, the expression of many genes involved in DNA synthesis was down-regulated in shE2F4-expressing cells, whereas no modulation in E2F1 expression was observed. A decrease in E2F4 in colon cancer cell lines also resulted in a reduction in soft-agar growth capacity. Immunofluorescence experiments in human fetal intestine revealed that cells expressing high nuclear levels of E2F4 also expressed cyclin A protein. Lastly, E2F4 and its target cyclin A were up-regulated and mostly nuclear in human colorectal tumor cells in comparison to the corresponding benign epithelium. These results indicate that nuclear E2F4 may be determinant in the promotion of proliferation of human intestinal epithelial crypt cells and colorectal cancer cells.


Asunto(s)
Ciclo Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Factor de Transcripción E2F4/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/citología , Agar , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular , Neoplasias Colorrectales/genética , Ciclina A/metabolismo , ADN/biosíntesis , Regulación hacia Abajo , Factor de Transcripción E2F4/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Fase G1 , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Transporte de Proteínas , Fase S
18.
Int J Cancer ; 123(2): 303-311, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18464259

RESUMEN

Clinical evidence suggests that estradiol replacement therapy reduces colon cancer risk in 'post'menopausal women. In colon epithelial cells, the estrogen receptor beta (ERbeta) is the predominant ER subtype and is thought to mediate the genomic effect of estrogens. The first aim of this study was to investigate the consequence of ERbeta deficiency on intestinal tumorigenesis in the Apc(Min/+) mouse model. Furthermore, to explore the biological mechanisms by which estrogens may influence the pathogenesis of colorectal cancer, we performed gene expression profiles in colonocytes from ovariectomized wild-type (WT) vs. ERbeta(-/-) mice, treated with estradiol (E(2)) or vehicle. Specifically in female, ERbeta deficiency was found to be associated with higher adenoma multiplicity in the small intestine, but not in the colon. Furthermore, tumors from ERbeta(-/-)Apc(Min/+) female mice were on average significantly larger than those from control Apc(Min/+) mice. Higher steady-state proliferation in epithelial cells of the jejunum and colon from ERbeta(-/-)Apc(Min/+) vs. Apc(Min/+) female mice was confirmed by BrdU incorporation assay. Interestingly, functional categorization of microarray results revealed the TGFbeta signaling pathway to be modulated in colonocytes, especially for the WT + E(2) vs. WT + Vehicle and the ERbeta(-/-) + E(2) vs. WT + E(2) comparisons. Using quantitative PCR analysis, we observed transcripts from ligands of the TGFbeta pathway to be upregulated in colonocytes from E(2)-treated WT and ERbeta(-/-) mice and downregulated in ERbeta-deficient mice, mostly in an E(2)-independent manner. Therefore, our results demonstrate that ERbeta deficiency enhances small intestinal tumorigenesis and suggest that modulation of the TGFbeta signaling pathway could contribute to the protective role of estrogens on intestinal tumorigenesis.


Asunto(s)
Adenoma/metabolismo , Receptor beta de Estrógeno/deficiencia , Neoplasias Intestinales/metabolismo , Intestino Delgado , Factor de Crecimiento Transformador beta/metabolismo , Adenoma/etiología , Animales , Bromodesoxiuridina , Neoplasias del Colon/metabolismo , Modelos Animales de Enfermedad , Receptor beta de Estrógeno/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias Intestinales/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Análisis de Secuencia por Matrices de Oligonucleótidos , Ovariectomía , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
19.
Methods Mol Biol ; 1765: 43-56, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29589300

RESUMEN

Proximal promoters are located upstream of the transcription start sites of genes, and they contain regulatory sequences on which bind different transcription factors for promoting colorectal cancer progression. Here we describe the comprehensive methodology used previously for the identification and functional characterization of MYC-responsive elements in the integrin α1 subunit (ITGA1) gene using a combination of in silico analysis, site-directed mutagenesis, and chromatin immunoprecipitation.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Neoplasias Colorrectales/genética , Integrina alfa1/genética , Mutagénesis Sitio-Dirigida/métodos , Elementos de Respuesta/genética , Sitios de Unión , Carcinogénesis/genética , Inmunoprecipitación de Cromatina/instrumentación , Simulación por Computador , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Mutagénesis Sitio-Dirigida/instrumentación , Proteínas Proto-Oncogénicas c-myc/metabolismo , Análisis de Secuencia de ADN/métodos , Sitio de Iniciación de la Transcripción
20.
Cancers (Basel) ; 9(8)2017 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-28933766

RESUMEN

Integrins are a family of heterodimeric glycoproteins involved in bidirectional cell signaling that participate in the regulation of cell shape, adhesion, migration, survival and proliferation. The integrin α1ß1 is known to be involved in RAS/ERK proliferative pathway activation and plays an important role in fibroblast proliferation. In the small intestine, the integrin α1 subunit is present in the crypt proliferative compartment and absent in the villus. We have recently shown that the integrin α1 protein and transcript (ITGA1) are present in a large proportion of colorectal cancers (CRC) and that their expression is controlled by the MYC oncogenic factor. Considering that α1 subunit/ITGA1 expression is correlated with MYC in more than 70% of colon adenocarcinomas, we postulated that the integrin α1ß1 has a pro-tumoral contribution to CRC. In HT29, T84 and SW480 CRC cells, α1 subunit/ITGA1 knockdown resulted in a reduction of cell proliferation associated with an impaired resistance to anoikis and an altered cell migration in HT29 and T84 cells. Moreover, tumor development in xenografts was reduced in HT29 and T84 sh-ITGA1 cells, associated with extensive necrosis, a low mitotic index and a reduced number of blood vessels. Our results show that α1ß1 is involved in tumor cell proliferation, survival and migration. This finding suggests that α1ß1 contributes to CRC progression.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA