Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mediators Inflamm ; 2023: 2899271, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36926280

RESUMEN

Toll-like receptors (TLRs) are the most studied receptors among the pattern recognition receptors (PRRs). They act as microbial sensors, playing major roles in the regulation of the innate immune system. TLRs mediate their cellular functions through the activation of MyD88-dependent or MyD88-independent signaling pathways. Myd88, or myeloid differentiation primary response 88, is a cytosolic adaptor protein essential for the induction of proinflammatory cytokines by all TLRs except TLR3. While the crucial role of Myd88 is well described, the contribution of other adaptors in mediating TLR signaling and function has been underestimated. In this review, we highlight important results demonstrating that TIRAP and TRAM adaptors are also required for full signaling activity and responses induced by most TLRs.


Asunto(s)
Factor 88 de Diferenciación Mieloide , Receptor Toll-Like 4 , Receptor Toll-Like 3 , Receptores Toll-Like , Transducción de Señal/fisiología , Proteínas Adaptadoras Transductoras de Señales
2.
Cell Commun Signal ; 19(1): 10, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33494775

RESUMEN

Over the past 2 decades, pattern recognition receptors (PRRs) have been shown to be on the front line of many illnesses such as autoimmune, inflammatory, and neurodegenerative diseases as well as allergies and cancer. Among PRRs, toll-like receptors (TLRs) are the most studied family. Dissecting TLRs signaling turned out to be advantageous to elaborate efficient treatments to cure autoimmune and chronic inflammatory disorders. However, a broad understanding of TLR effectors is required to propose a better range of cures. In addition to kinases and E3 ubiquitin ligases, phosphatases emerge as important regulators of TLRs signaling mediated by NF-κB, type I interferons (IFN I) and Mitogen-Activated Protein Kinases signaling pathways. Here, we review recent knowledge on TLRs signaling modulation by different classes and subclasses of phosphatases. Thus, it becomes more and more evident that phosphatases could represent novel therapeutic targets to control pathogenic TLRs signaling. Video Abstract.


Asunto(s)
Monoéster Fosfórico Hidrolasas/metabolismo , Receptores Toll-Like/metabolismo , Animales , COVID-19/metabolismo , Humanos , SARS-CoV-2 , Transducción de Señal
3.
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
4.
J Pathol ; 247(1): 135-146, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30376595

RESUMEN

The Src homology-2 domain-containing tyrosine phosphatase 2 (SHP-2) regulates many cellular processes, including proliferation, differentiation and survival. Polymorphisms in the gene encoding SHP-2 are associated with an increased susceptibility to develop ulcerative colitis. We recently reported that intestinal epithelial cell (IEC)-specific deletion of Shp-2 in mice (Shp-2IEC-KO ) leads to chronic colitis and colitis-associated cancer. This suggests that SHP-2-dependent signaling protects the colonic epithelium against inflammation and colitis-associated cancer development. To verify this hypothesis, we generated mice expressing the Shp-2 E76K activated form specifically in IEC. Our results showed that sustained Shp-2 activation in IEC increased intestine and crypt length, correlating with increased cell proliferation and migration. Crypt regeneration capacity was also markedly enhanced, as revealed by ex vivo organoid culture. Shp-2 activation alters the secretory cell lineage, as evidenced by increased goblet cell numbers and mucus secretion. Notably, these mice also demonstrated elevated ERK signaling in IEC and exhibited resistance against both chemical- and Citrobacter rodentium-induced colitis. In contrast, mice with IEC-specific Shp-2 deletion displayed reduced ERK signaling and rapidly developed chronic colitis. Remarkably, expression of an activated form of Braf in Shp-2-deficient mice restored ERK activation, goblet cell production and prevented colitis. Altogether, our results indicate that chronic activation of Shp-2/ERK signaling in the colonic epithelium confers resistance to mucosal erosion and colitis. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Movimiento Celular , Proliferación Celular , Colitis/prevención & control , Colon/enzimología , Células Caliciformes/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Regeneración , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Colitis/enzimología , Colitis/genética , Colitis/patología , Colon/patología , Modelos Animales de Enfermedad , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células Caliciformes/patología , Ratones Transgénicos , Fenotipo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Transducción de Señal , Técnicas de Cultivo de Tejidos , Cicatrización de Heridas
5.
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
6.
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
7.
J Cell Physiol ; 231(2): 436-48, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26174178

RESUMEN

The intestinal epithelium responds to and transmits signals from the microbiota and the mucosal immune system to insure intestinal homeostasis. These interactions are in part conveyed by epigenetic modifications, which respond to environmental changes. Protein acetylation is an epigenetic signal regulated by histone deacetylases, including Hdac1 and Hdac2. We have previously shown that villin-Cre-inducible intestinal epithelial cell (IEC)-specific Hdac1 and Hdac2 deletions disturb intestinal homeostasis. To determine the role of Hdac1 and Hdac2 in the regulation of IEC function and the establishment of the dual knockout phenotype, we have generated villin-Cre murine models expressing one Hdac1 allele without Hdac2, or one Hdac2 allele without Hdac1. We have also investigated the effect of short-term deletion of both genes in naphtoflavone-inducible Ah-Cre and tamoxifen-inducible villin-Cre(ER) mice. Mice with one Hdac1 allele displayed normal tissue architecture, but increased sensitivity to DSS-induced colitis. In contrast, mice with one Hdac2 allele displayed intestinal architecture defects, increased proliferation, decreased goblet cell numbers as opposed to Paneth cells, increased immune cell infiltration associated with fibrosis, and increased sensitivity to DSS-induced colitis. In comparison to dual knockout mice, intermediary activation of Notch, mTOR, and Stat3 signaling pathways was observed. While villin-Cre(ER) Hdac1 and Hdac2 deletions led to an impaired epithelium and differentiation defects, Ah-Cre-mediated deletion resulted in blunted proliferation associated with the induction of a DNA damage response. Our results suggest that IEC determination and intestinal homeostasis are highly dependent on Hdac1 and Hdac2 activity levels, and that changes in the IEC acetylome may alter the mucosal environment.


Asunto(s)
Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Mucosa Intestinal/enzimología , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proliferación Celular/genética , Proliferación Celular/fisiología , Colitis/enzimología , Colitis/genética , Colitis/patología , Daño del ADN , Modelos Animales de Enfermedad , Células Epiteliales/enzimología , Células Caliciformes/citología , Células Caliciformes/enzimología , Histona Desacetilasa 1/deficiencia , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/deficiencia , Histona Desacetilasa 2/genética , Homeostasis , Inmunidad Mucosa , Mucosa Intestinal/anomalías , Mucosa Intestinal/citología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Notch/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
8.
J Cell Physiol ; 231(11): 2529-40, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27100271

RESUMEN

Polymorphisms in the PTPN11 gene encoding for the tyrosine phosphatase SHP-2 were described in patients with ulcerative colitis. We have recently demonstrated that mice with an intestinal epithelial cell-specific deletion of SHP-2 (SHP-2(IEC-KO) ) develop severe colitis 1 month after birth. However, the mechanisms by which SHP-2 deletion induces colonic inflammation remain to be elucidated. We generated SHP-2(IEC-KO) mice lacking Myd88 exclusively in the intestinal epithelium. The colonic phenotype was histologically analyzed and cell differentiation was determined by electron microscopy and lysozyme or Alcian blue staining. Microbiota composition was analyzed by 16S sequencing. Results show that innate defense genes including those specific to Paneth cells were strongly up-regulated in SHP-2-deficient colons. Expansion of intermediate cells (common progenitors of the Goblet and Paneth cell lineages) was found in the colon of SHP-2(IEC-KO) mice whereas Goblet cell number was clearly diminished. These alterations in Goblet/intermediate cell ratio were noticed 2 weeks after birth, before the onset of inflammation and were associated with significant alterations in microbiota composition. Indeed, an increase in Enterobacteriaceae and a decrease in Firmicutes were observed in the colon of these mice, indicating that dysbiosis also occurred prior to inflammation. Importantly, loss of epithelial Myd88 expression inhibited colitis development in SHP-2(IEC-KO) mice, rescued Goblet/intermediate cell ratio, and prevented NFκB hyperactivation and inflammation. These data indicate that SHP-2 is functionally important for the maintenance of appropriate barrier function and host-microbiota homeostasis in the large intestine. J. Cell. Physiol. 231: 2529-2540, 2016. © 2016 The Authors. Journal of Cellular Physiology published by Wiley Periodicals, Inc.


Asunto(s)
Diferenciación Celular , Colon/patología , Homeostasis , Inflamación/patología , Inflamación/prevención & control , Microbiota , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Animales , Animales Recién Nacidos , Péptidos Catiónicos Antimicrobianos/metabolismo , Biomarcadores/metabolismo , Células Caliciformes/metabolismo , Células Caliciformes/patología , Inflamación/genética , Ratones Endogámicos C57BL , Muramidasa/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Células de Paneth/metabolismo , Células de Paneth/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/deficiencia , Regulación hacia Arriba/genética
9.
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
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.
Int J Cancer ; 133(1): 58-66, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23280881

RESUMEN

Promyelocytic leukemia zinc-finger (PLZF) is a transcriptional repressor that regulates proliferation, differentiation and apoptosis among various cellular origins. PLZF expression is upregulated in colorectal cancer cell lines but its putative functional role in this context is unknown. Here, we report the identification of a novel p65 PLZF isoform that results from the usage of an evolutionarily conserved alternative translational initiation site. This isoform is devoid of the classical BTB/POZ domain required for nuclear localization and transcriptional repression. Depletion of p65 PLZF expression in colorectal cancer cell lines results in reduction of cell growth, loss of cell anchorage and increase in cell apoptosis. Overall, these results indicate that p65 PLZF is crucial to maintain colorectal cancer cell adhesion as well as survival and must occur independently of the traditionally viewed transcriptional role of PLZF in the course of these biological processes.


Asunto(s)
Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Factores de Transcripción de Tipo Kruppel/metabolismo , Animales , Adhesión Celular , Línea Celular Tumoral , Supervivencia Celular , Transformación Celular Neoplásica/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Inmunohistoquímica , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Ratones Desnudos , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Isoformas de Proteínas/metabolismo , Trasplante Heterólogo , Células Tumorales Cultivadas
13.
Gastroenterology ; 142(2): 316-25.e1-12, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22079593

RESUMEN

BACKGROUND & AIMS: Copolymers of hydroxyethyl methacrylate and styrene sulfonate complex with isolated gliadin (the toxic fraction of gluten) and prevent damage to the intestinal barrier in HLA-HCD4/DQ8 mice. We studied the activity toward gluten and hordein digestion and biologic effects of poly(hydroxyethyl methacrylate-co-styrene sulfonate (P(HEMA-co-SS)). We also investigated the effect of gliadin complex formation in intestinal biopsy specimens from patients with celiac disease. METHODS: We studied the ability of P(HEMA-co-SS) to reduce digestion of wheat gluten and barley hordein into immunotoxic peptides using liquid chromatography-mass spectrometry. The biodistribution and pharmacokinetic profile of orally administered P(HEMA-co-SS) was established in rodents using tritium-labeled polymer. We assessed the capacity of P(HEMA-co-SS) to prevent the immunologic and intestinal effects induced by a gluten-food mixture in gluten-sensitized HLA-HCD4/DQ8 mice after short-term and long-term administration. We measured the effects of gliadin complex formation on cytokine release ex vivo using intestinal biopsy specimens from patients with celiac disease. RESULTS: P(HEMA-co-SS) reduced digestion of wheat gluten and barley hordein in vitro, thereby decreasing formation of toxic peptides associated with celiac disease. After oral administration to rodents, P(HEMA-co-SS) was predominantly excreted in feces, even in the presence of low-grade mucosal inflammation and increased intestinal permeability. In gluten-sensitized mice, P(HEMA-co-SS) reduced paracellular permeability, normalized anti-gliadin immunoglobulin A in intestinal washes, and modulated the systemic immune response to gluten in a food mixture. Furthermore, incubation of P(HEMA-co-SS) with mucosal biopsy specimens from patients with celiac disease showed that secretion of tumor necrosis factor-α was reduced in the presence of partially digested gliadin. CONCLUSIONS: The copolymer P(HEMA-co-SS) reduced digestion of wheat gluten and barley hordein and attenuated the immune response to gluten in a food mixture in rodents. It might be developed to prevent or reduce gluten-induced disorders in humans.


Asunto(s)
Enfermedad Celíaca/metabolismo , Digestión/efectos de los fármacos , Glútenes/metabolismo , Mucosa Intestinal/efectos de los fármacos , Polihidroxietil Metacrilato/análogos & derivados , Poliestirenos/farmacología , Estirenos/farmacología , Animales , Enfermedad Celíaca/tratamiento farmacológico , Enfermedad Celíaca/inmunología , Cromatografía Liquida , Femenino , Gliadina/metabolismo , Gliadina/toxicidad , Glútenes/toxicidad , Humanos , Absorción Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Yeyuno/efectos de los fármacos , Yeyuno/inmunología , Yeyuno/patología , Masculino , Espectrometría de Masas , Ratones , Permeabilidad/efectos de los fármacos , Polihidroxietil Metacrilato/farmacocinética , Polihidroxietil Metacrilato/farmacología , Polihidroxietil Metacrilato/uso terapéutico , Poliestirenos/farmacocinética , Poliestirenos/uso terapéutico , Unión Proteica , Distribución Aleatoria , Ratas , Estirenos/farmacocinética , Estirenos/uso terapéutico
14.
Anat Rec (Hoboken) ; 306(5): 1165-1183, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36196983

RESUMEN

The effect of Western diets in the gastrointestinal system is largely mediated by their ability to promote alterations in the immunity and physiology of the intestinal epithelium, and to affect the composition of the commensal microbiota. To investigate the response of the colonic epithelium to high-fat/high-cholesterol diets (HFHCDs), we evaluated the synthesis of host defense factors involved in the maintenance of the colonic homeostasis. C57BL/6 mice were fed an HFHCD for 3 weeks and their colons were evaluated for histopathology, gene expression, and microbiota composition. In addition, intestinal permeability and susceptibility to Citrobacter rodentium were also studied. HFHCD caused colonic hyperplasia, loss of goblet cells, thinning of the mucus layer, moderate changes in the composition of the intestinal microbiota, and an increase in intestinal permeability. Gene expression analyses revealed significant drops in the transcript levels of Muc1, Muc2, Agr2, Atoh1, Spdef, Ang4, Camp, Tff3, Dmbt1, Fcgbp, Saa3, and Retnlb. The goblet cell granules of HFHCD-fed mice were devoid of Relmß and Tff3, indicating defective production of those two factors critical for intestinal epithelial defense and homeostasis. In correspondence with these defects, colonic bacteria were in close contact with, and invading the epithelium. Fecal shedding of C. rodentium showed an increased bacterial burden in HFHCD-fed animals accompanied by increased epithelial damage. Collectively, our results show that HFHCD perturbs the synthesis of colonic host defense factors, which associate with alterations in the commensal microbiota, the integrity of the intestinal barrier, and the host's susceptibility to enteric infections.


Asunto(s)
Colon , Mucosa Intestinal , Ratones , Animales , Ratones Endogámicos C57BL , Colon/metabolismo , Células Caliciformes/metabolismo , Dieta
15.
BMC Cell Biol ; 12: 55, 2011 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-22185284

RESUMEN

BACKGROUND: The human homologue of the Drosophila Discs-large tumor suppressor protein, hDlg, is a multi-domain cytoplasmic protein that localizes to the membrane at intercellular junction sites. At both synaptic junctions and epithelia cell-cell junctions, hDlg is known to recruit several signaling proteins into macromolecular complexes. hDlg is also found at the midbody, a small microtubule-rich structure bridging the two daughter cells during cytokinesis, but its function at this site is not clear. RESULTS: Here we describe the interaction of hDlg with the activated form of MEK2 of the canonical RAF/MEK/ERK pathway, a protein that is found at the midbody during cytokinesis. We show that both proteins localize to a sub-structure of the midbody, the midbody ring, and that the interaction between the PDZ domains of hDlg and the C-terminal portion of MEK2 is dependent on the phosphorylation of MEK2. Finally, we found that E-cadherin also localizes to the midbody and that its expression is required for the isoform-specific recruitment of hDlg, but not activated MEK2, to that structure. CONCLUSION: Our results suggest that like at other cell-cell junction sites, hDlg is part of a macromolecular complex of structural and signaling proteins at the midbody.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cadherinas/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Adaptadoras Transductoras de Señales/análisis , Secuencia de Aminoácidos , Animales , Línea Celular , Citocinesis , Homólogo 1 de la Proteína Discs Large , Humanos , MAP Quinasa Quinasa 2/química , Proteínas de la Membrana/análisis , Datos de Secuencia Molecular , Dominios PDZ , Unión Proteica , Alineación de Secuencia
16.
Am J Physiol Gastrointest Liver Physiol ; 301(4): G719-30, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21737780

RESUMEN

The Ras/Raf/MEK/ERK cascade regulates intestinal epithelial cell proliferation. Indeed, while barely detectable in differentiated cells of the villi, ERK1/2-activated forms are detected in the nucleus of undifferentiated human intestinal crypt cells. In addition, we and others have reported that ERKs are selectively inactivated during enterocyte differentiation. However, whether inactivation of the ERK pathway is necessary for inhibition of both proliferation and induction of differentiation of intestinal epithelial cells is unknown. Human Caco-2/15 cells, undifferentiated crypt IEC-6 cells, and differentiating Cdx3-expressing IEC-6 cells were infected with retroviruses encoding either a hemagglutinin (HA)-tagged MEK1 wild type (wtMEK) or a constitutively active S218D/S222D MEK1 mutant (caMEK). Protein and gene expression was assessed by Western blotting, semiquantitative RT-PCR, and real-time PCR. Morphology was analyzed by transmission electron microscopy. We found that 1) IEC-6/Cdx3 cells formed multicellular layers after confluence and differentiated after 30 days in culture, as assessed by increased polarization, microvilli formation, expression of differentiation markers, and ERK1/2 inhibition; 2) while activated MEK prevented neither the inhibition of ERK1/2 activities nor the differentiation process in postconfluent Caco-2/15 cells, caMEK expression prevented ERK inhibition in postconfluent IEC-6/Cdx3 cells, thus leading to maintenance of elevated ERK1/2 activities; 3) caMEK-expressing IEC-6/Cdx3 cells exhibited altered multicellular structure organization, poorly defined tight junctions, reduced number of microvilli on the apical surface, and decreased expression of the hepatocyte nuclear factor 1α transcription factor and differentiation markers, namely apolipoprotein A-4, fatty acid-binding protein, calbindin-3, mucin 2, alkaline phosphatase, and sucrase-isomaltase; and 4) increased Cdx3 phosphorylation on serine-60 (S60) in IEC-6/Cdx3 cells expressing caMEK led to decreased Cdx2 transactivation potential. These results indicate that inactivation of the ERK pathway is required to ensure the full Cdx2/3 transcriptional activity necessary for intestinal epithelial cell terminal differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Mucosa Intestinal/citología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Animales , Factor de Transcripción CDX2 , Células CACO-2 , Células Cultivadas , Células Epiteliales/metabolismo , Proteínas de Homeodominio/fisiología , Humanos , MAP Quinasa Quinasa 1/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Ratas , Transactivadores/fisiología
17.
Cells ; 10(7)2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34359960

RESUMEN

Intestinal epithelial self-renewal is tightly regulated by signaling pathways controlling stem cell proliferation, determination and differentiation. In particular, Wnt/ß-catenin signaling controls intestinal crypt cell division, survival and maintenance of the stem cell niche. Most colorectal cancers are initiated by mutations activating the Wnt/ß-catenin pathway. Wnt signals are transduced through Frizzled receptors and LRP5/LRP6 coreceptors to downregulate GSK3ß activity, resulting in increased nuclear ß-catenin. Herein, we explored if LRP6 expression is required for maintenance of intestinal homeostasis, regeneration and oncogenesis. Mice with an intestinal epithelial cell-specific deletion of Lrp6 (Lrp6IEC-KO) were generated and their phenotype analyzed. No difference in intestinal architecture nor in proliferative and stem cell numbers was found in Lrp6IEC-KO mice in comparison to controls. Nevertheless, using ex vivo intestinal organoid cultures, we found that LRP6 expression was critical for crypt cell proliferation and stem cell maintenance. When exposed to dextran sodium sulfate, Lrp6IEC-KO mice developed more severe colitis than control mice. However, loss of LRP6 did not affect tumorigenesis in ApcMin/+ mice nor growth of human colorectal cancer cells. By contrast, Lrp6 silencing diminished anchorage-independent growth of BRafV600E-transformed intestinal epithelial cells (IEC). Thus, LRP6 controls intestinal stem cell functionality and is necessary for BRAF-induced IEC oncogenesis.


Asunto(s)
Células Epiteliales/metabolismo , Homeostasis/fisiología , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Células Madre/citología , Animales , Carcinogénesis/metabolismo , Transformación Celular Neoplásica/genética , Homeostasis/genética , Humanos , Mucosa Intestinal/metabolismo , Intestinos , Ratones , Organoides/metabolismo , Regeneración/fisiología
18.
Cancers (Basel) ; 13(17)2021 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-34503224

RESUMEN

NCOR1 is a corepressor that mediates transcriptional repression through its association with nuclear receptors and specific transcription factors. Some evidence supports a role for NCOR1 in neonatal intestinal epithelium maturation and the maintenance of epithelial integrity during experimental colitis in mice. We hypothesized that NCOR1 could control colorectal cancer cell proliferation and tumorigenicity. Conditional intestinal epithelial deletion of Ncor1 in ApcMin/+ mice resulted in a significant reduction in polyposis. RNAi targeting of NCOR1 in Caco-2/15 and HT-29 cell lines led to a reduction in cell growth, characterized by cellular senescence associated with a secretory phenotype. Tumor growth of HT-29 cells was reduced in the absence of NCOR1 in the mouse xenografts. RNA-seq transcriptome profiling of colon cancer cells confirmed the senescence phenotype in the absence of NCOR1 and predicted the occurrence of a pro-migration cellular signature in this context. SOX2, a transcription factor essential for pluripotency of embryonic stem cells, was induced under these conditions. In conclusion, depletion of NCOR1 reduced intestinal polyposis in mice and caused growth arrest, leading to senescence in human colorectal cell lines. The acquisition of a pro-metastasis signature in the absence of NCOR1 could indicate long-term potential adverse consequences of colon-cancer-induced senescence.

19.
Oncogene ; 40(2): 452-464, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33177649

RESUMEN

Interleukin-17 receptor D (IL-17RD), also known as similar expression to Fgf genes (SEF), is proposed to act as a signaling hub that negatively regulates mitogenic signaling pathways, like the ERK1/2 MAP kinase pathway, and innate immune signaling. The expression of IL-17RD is downregulated in certain solid tumors, which has led to the hypothesis that it may exert tumor suppressor functions. However, the role of IL-17RD in tumor biology remains to be studied in vivo. Here, we show that genetic disruption of Il17rd leads to the increased formation of spontaneous tumors in multiple tissues of aging mice. Loss of IL-17RD also promotes tumor development in a model of colitis-associated colorectal cancer, associated with an exacerbated inflammatory response. Colon tumors from IL-17RD-deficient mice are characterized by a strong enrichment in inflammation-related gene signatures, elevated expression of pro-inflammatory tumorigenic cytokines, such as IL-17A and IL-6, and increased STAT3 tyrosine phosphorylation. We further show that RNAi depletion of IL-17RD enhances Toll-like receptor and IL-17A signaling in colon adenocarcinoma cells. No change in the proliferation of normal or tumor intestinal epithelial cells was observed upon genetic inactivation of IL-17RD. Our findings establish IL-17RD as a tumor suppressor in mice and suggest that the protein exerts its function mainly by limiting the extent and duration of inflammation.


Asunto(s)
Carcinogénesis/patología , Colitis/complicaciones , Neoplasias del Colon/patología , Inflamación/complicaciones , Receptores de Interleucina/fisiología , Animales , Carcinogénesis/metabolismo , Proliferación Celular , Neoplasias del Colon/etiología , Neoplasias del Colon/metabolismo , Citocinas/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Factor de Transcripción STAT3/metabolismo , Transcriptoma , Tirosina/metabolismo
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA