Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Tissue Res ; 370(3): 441-449, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28856432

RESUMEN

Kruppel-like factor 4 (KLF4) is a zinc finger transcription factor that plays crucial roles during the development and maintenance of multiple organs. We and others have previously shown that KLF4 is involved in bone modeling and remodeling but roles played by KLF4 during skeletogenesis are still not fully understood. Here, we show that KLF4 is expressed in the epiphyseal growth plate and articular chondrocytes. Most articular chondrocytes expressed KLF4 in embryos but it localized only in a subset of superficial zone cells in postnatal mice. When KLF4 was overexpressed in chondrocytes in vitro, it severely repressed chondrocytic gene expressions. Global gene expression profiling of KLF4-transduced chondrocytes revealed matrix degrading proteinases of the matrix metalloproteinase and disintegrin and metalloproteinase with thrombospondin-1 domain families within the group of upregulated genes. Proteinase induction by KLF4 was alleviated by Trichostatin A treatment suggesting the possible involvement of epigenetic mechanisms on proteinase induction by KLF4. These results indicate the possible involvement of KLF4 in physiological and pathological aspects during cartilage development and maintenance.


Asunto(s)
Cartílago Articular/metabolismo , Condrocitos/metabolismo , Endopeptidasas/biosíntesis , Factores de Transcripción de Tipo Kruppel/metabolismo , Metaloproteinasas de la Matriz/biosíntesis , Trombospondina 1/biosíntesis , Animales , Células Cultivadas , Endopeptidasas/genética , Regulación del Desarrollo de la Expresión Génica , Ácidos Hidroxámicos/farmacología , Factor 4 Similar a Kruppel , Masculino , Metaloproteinasas de la Matriz/genética , Ratones , Ratones Endogámicos ICR , Inhibidores de la Síntesis de la Proteína/farmacología , Trombospondina 1/genética
2.
Dev Biol ; 387(2): 191-202, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24440658

RESUMEN

Krüppel-like factor 5 (KLF5) is a pro-proliferative transcriptional regulator primarily expressed in the intestinal crypt epithelial cells. Constitutive intestine-specific deletion of Klf5 is neonatal lethal suggesting a crucial role for KLF5 in intestinal development and homeostasis. We have previously shown Klf5 to play an active role regulating intestinal tumorigenesis. Here we examine the effect of inducible intestine-specific deletion of Klf5 in adult mice. Klf5 is lost from the intestine beginning at day 3 after the start of a 5-day treatment with the inducer tamoxifen. Although the mice have no significant weight loss or lethality, the colonic tissue shows signs of epithelial distress starting at day 3 following induction. Accompanying the morphological changes is a significant loss of proliferative crypt epithelial cells as revealed by BrdU or Ki67 staining at days 3 and 5 after start of tamoxifen. We also observed a loss of goblet cells from the colon and Paneth cells from the small intestine upon induced deletion of Klf5. In addition, loss of Klf5 from the colonic epithelium is accompanied by a regenerative response that coincides with an expansion in the zone of Sox9 expression along the crypt axis. At day 11, both proliferation and Sox9 expression return to baseline levels. Microarray and quantitative PCR analyses reveal an up-regulation of several regeneration-associated genes (Reg1A, Reg3G and Reg3B) and down-regulation of many Klf5 targets (Ki-67, cyclin B, Cdc2 and cyclin D1). Sox9 and Reg1A protein levels are also increased upon Klf5 loss. Lentiviral-mediated knockdown of KLF5 and exogenous expression of KLF5 in colorectal cancer cell lines confirm that Sox9 expression is negatively regulated by KLF5. Furthermore, ChIP assays reveal a direct association of KLF5 with both the Sox9 and Reg1A promoters. We have shown that disruption of epithelial homeostasis due to Klf5 loss from the adult colon is followed by a regenerative response led by Sox9 and the Reg family of proteins. Our study demonstrates that adult mouse colonic tissue undergoes acute physiological changes to accommodate the loss of Klf5 withstanding epithelial damage further signifying importance of Klf5 in colonic homeostasis.


Asunto(s)
Colon/fisiología , Factores de Transcripción de Tipo Kruppel/genética , Regeneración/genética , Animales , Antineoplásicos Hormonales/farmacología , Proteína Quinasa CDC2/metabolismo , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Ciclina B/metabolismo , Ciclina D1/metabolismo , Regulación hacia Abajo , Células Caliciformes/efectos de los fármacos , Células HCT116 , Células HEK293 , Humanos , Antígeno Ki-67/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Litostatina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Pancreatitis , Células de Paneth/efectos de los fármacos , Regiones Promotoras Genéticas , Proteínas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Factor de Transcripción SOX9/metabolismo , Eliminación de Secuencia , Transducción de Señal/genética , Tamoxifeno/farmacología , Regulación hacia Arriba
3.
Am J Physiol Gastrointest Liver Physiol ; 308(2): G121-38, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25414097

RESUMEN

Gut radiation-induced injury is a concern during treatment of patients with cancer. Krüppel-like factor 4 (KLF4) is expressed in differentiated villous epithelial cells of the small intestine. We previously showed that KLF4 protects cells from apoptosis following γ-irradiation in vitro. We sought to determine whether KLF4 mediates the small intestinal response to γ-irradiation in vivo. Mice with intestinal epithelium-specific deletion of Klf4 (Klf4(ΔIS)) and control (Klf4(fl/fl)) mice were irradiated with total-body γ-radiation. Following irradiation, the Klf4(ΔIS) mice had significantly increased mortality compared with irradiated Klf4(fl/fl) mice. Immunohistochemistry and immunofluorescence staining were used to assess the morphological changes, levels of proliferation, and apoptosis in the intestinal epithelium. At 96 h following irradiation, there was a regenerative response manifested by an expansion of the proliferative zone in both mouse groups, with the control mice having a higher proliferative activity than the Klf4(ΔIS) group. In addition, there was a significant increase in the number of Klf4/Ki67-copositive cells in the irradiated control mice compared with unirradiated mice. Also, the irradiated Klf4(ΔIS) mice had a significantly higher number of crypt cells positive for apoptosis, p53, and p21 compared with irradiated Klf4(fl/fl) mice. Taken together, our data suggest that Klf4 may function as a radioprotective factor against gastrointestinal syndrome in mice following γ-irradiation by inhibiting apoptosis in the acute response to irradiation and contributing to crypt regeneration.


Asunto(s)
Mucosa Intestinal/lesiones , Mucosa Intestinal/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Animales , Apoptosis/fisiología , Línea Celular Tumoral , Daño del ADN/efectos de la radiación , Modelos Animales de Enfermedad , Rayos gamma , Intestinos/efectos de la radiación , Factor 4 Similar a Kruppel , Ratones , Factores de Transcripción/genética
4.
Mol Cancer ; 12: 89, 2013 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-23919723

RESUMEN

BACKGROUND: Krüppel-like factor 4 (KLF4) is a member of the KLF family of transcription factors and regulates proliferation, differentiation, apoptosis and somatic cell reprogramming. Evidence also suggests that KLF4 is a tumor suppressor in certain cancers including colorectal cancer. We previously showed that KLF4 inhibits cell cycle progression following DNA damage and that mouse embryonic fibroblasts (MEFs) null for Klf4 are genetically unstable, as evidenced by increased rates of cell proliferation, and the presence of DNA double strand breaks (DSBs), centrosome amplification, chromosome aberrations and aneuploidy. METHODS: To determine whether re-expression of Klf4 corrects the observed genetic instability in MEFs null for Klf4 (Klf4(-/-)), we transfected Klf4(-/-)MEFs with Klf4-expressing plasmids and compared the results to wild type (Klf4(+/+)) and untransfected or mock-transfected Klf4(-/-)MEFs. RESULTS: We show that overexpression of Klf4 in Klf4(-/-)MEFs reduced cell proliferation rates and the proportion of cells with DSBs, abnormal centrosome numbers, aneuploidy and micronuclei. In addition, Klf4-transfected Klf4(-/-)MEFs exhibited a more robust DNA damage repair response as demonstrated by the greater rate in disappearance of γ-H2AX and 53BP1 foci following γ-irradiation. CONCLUSION: Taken together these findings provide evidence that KLF4 plays a crucial role in the maintenance of genetic stability by modulating the DNA damage response and repair processes.


Asunto(s)
Fibroblastos/metabolismo , Inestabilidad Genómica , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Aneuploidia , Animales , Línea Celular , Proliferación Celular , Centrosoma/metabolismo , Daño del ADN/genética , Daño del ADN/efectos de la radiación , Expresión Génica , Técnicas de Inactivación de Genes , Factor 4 Similar a Kruppel , Ratones , Micronúcleos con Defecto Cromosómico , Transfección
5.
Dev Biol ; 349(2): 310-20, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21070761

RESUMEN

The zinc finger transcription factor, Krüppel-like factor 4 (KLF4), is expressed in the post-mitotic, differentiated epithelial cells lining the intestinal tract and exhibits a tumor suppressive effect on intestinal tumorigenesis. Here we report a role for KLF4 in maintaining homeostasis of intestinal epithelial cells. Mice with conditional ablation of the Klf4 gene from the intestinal epithelium were viable. However, both the rates of proliferation and migration of epithelial cells were increased in the small intestine of mutant mice. In addition, the brush-border alkaline phosphatase was reduced as was expression of ephrine-B1 in the small intestine, resulting in mispositioning of Paneth cells to the upper crypt region. In the colon of mutant mice, there was a reduction of the differentiation marker, carbonic anhydrase-1, and failure of differentiation of goblet cells. Mechanistically, deletion of Klf4 from the intestine resulted in activation of genes in the Wnt pathway and reduction in expression of genes encoding regulators of differentiation. Taken together, these data provide new insights into the function of KLF4 in regulating postnatal proliferation, migration, differentiation, and positioning of intestinal epithelial cells and demonstrate an essential role for KLF4 in maintaining normal intestinal epithelial homeostasis in vivo.


Asunto(s)
Diferenciación Celular/fisiología , Células Epiteliales/citología , Regulación del Desarrollo de la Expresión Génica/fisiología , Homeostasis/fisiología , Mucosa Intestinal/fisiología , Factores de Transcripción de Tipo Kruppel/deficiencia , Fosfatasa Alcalina/metabolismo , Animales , Western Blotting , Bromodesoxiuridina , Anhidrasa Carbónica I/metabolismo , Movimiento Celular , Proliferación Celular , Efrina-B1/metabolismo , Técnica del Anticuerpo Fluorescente , Eliminación de Gen , Técnicas Histológicas , Inmunohistoquímica , Mucosa Intestinal/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Reacción en Cadena de la Polimerasa
6.
Int J Cancer ; 131(6): 1435-44, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22052467

RESUMEN

Germline mutation of the tumor suppressor gene, adenomatous polyposis coli (APC), is responsible for familial adenomatous polyposis (FAP) with nearly 100% risk for colon cancer at an early age. Although FAP is involved in only 1% of all colon cancer cases, over 80% of sporadic cancers harbor somatic mutations of APC. We show here that bromo-noscapine (EM011), a rationally designed synthetic derivative of a natural nontoxic tubulin-binding alkaloid-noscapine, that reduces the dynamics of microtubules, causes a reversible G(2) /M arrest in wild type murine embryonic fibroblasts (MEFs), but an aberrant exit from a brief mitotic block, followed by apoptosis in MEFs after APC deletion with small interfering RNA. Furthermore, both ß-catenin levels and activity fell to half the original levels with a concomitant reduction of cell proliferation-inducing cyclin D1, c-Myc, and induction of cytostatic protein p21 before caspase-3 activation. Additionally, we show a statistically significant reduction in the number of newly emerging intestinal polyps (to 35% compared with untreated mice) as well as the mean size of polyps (to 42% compared with untreated mice) in EM011-treated Apc(Min/+) mice as compared to their sham-treated control littermates. The remaining polyps in the EM011 treated group of Apc(Min/+) mice showed evidence of elevated apoptosis as revealed by immunohistochemistry. We failed to detect any evidence of histopathological and hematological toxicities following EM011 treatment. Taken together, our data are persuasive that a clinical trial of EM011 is possible for the prevention/amelioration of polyposis in FAP patients.


Asunto(s)
Poliposis Adenomatosa del Colon/prevención & control , Anticarcinógenos/uso terapéutico , Dioxoles/uso terapéutico , Genes APC/fisiología , Isoquinolinas/uso terapéutico , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/fisiología , Femenino , Células HCT116 , Humanos , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción 4 , beta Catenina/fisiología
7.
Gastroenterology ; 141(4): 1302-13, 1313.e1-6, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21763241

RESUMEN

BACKGROUND & AIMS: Krüppel-like factor 5 (KLF5) is transcription factor that is expressed by dividing epithelial cells of the intestinal epithelium. KLF5 promotes proliferation in vitro and in vivo and is induced by mitogens and various stress stimuli. To study the role of KLF5 in intestinal epithelial homeostasis, we examined the phenotype of mice with conditional deletion of Klf5 in the gut. METHODS: Mice were generated with intestinal-specific deletion of Klf5 (Vil-Cre;Klf5fl/fl). Morphologic changes in the small intestine and colon were examined by immunohistochemistry, immunoblotting, and real-time polymerase chain reaction. RESULTS: Klf5 mutant mice were born at a normal Mendelian ratio but had high mortality compared with controls. Complete deletion of Klf5 from the intestinal mucosa resulted in neonatal lethality that corresponded with an absence of epithelial proliferation. Variegated intestinal-specific deletion of Klf5 in adult mice resulted in morphologic changes that included a regenerative phenotype, impaired barrier function, and inflammation. Adult mutant mice exhibited defects in epithelial differentiation and migration. These changes were associated with reduced expression of Caudal type homeobox (Cdx) 1, Cdx2, and Eph and ephrin signaling proteins. Concomitantly, Wnt signaling to ß-catenin was reduced. Proliferation in regenerative crypts was associated with increased expression of the progenitor cell marker Sox9. CONCLUSIONS: Deletion of Klf5 in the gut epithelium of mice demonstrated that KLF5 maintains epithelial proliferation, differentiation, and cell positioning along the crypt radial axis. Morphologic changes that occur with deletion of Klf5 are associated with disruption of canonical Wnt signaling and increased expression of Sox9.


Asunto(s)
Colon/metabolismo , Células Epiteliales/metabolismo , Íleon/metabolismo , Absorción Intestinal , Mucosa Intestinal/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Animales , Western Blotting , Factor de Transcripción CDX2 , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Colon/patología , Efrinas/metabolismo , Células Epiteliales/patología , Genotipo , Proteínas de Homeodominio/metabolismo , Íleon/patología , Inmunohistoquímica , Mucosa Intestinal/patología , Factores de Transcripción de Tipo Kruppel/deficiencia , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Permeabilidad , Fenotipo , Reacción en Cadena de la Polimerasa , Receptores de la Familia Eph/metabolismo , Regeneración , Factor de Transcripción SOX9/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
8.
Gastroenterology ; 141(4): 1381-92, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21723221

RESUMEN

BACKGROUND & AIMS: Inflammatory bowel disease increases the risks of colon cancer and colitis-associated cancer (CAC). Epithelial cell-derived matrix metalloproteinase (MMP)-9 mediates inflammation during acute colitis and the cleavage and activation of the transcription factor Notch1, which prevents differentiation of progenitor cells into goblet cells. However, MMP-9 also protects against the development of CAC and acts as a tumor suppressor. We investigated the mechanisms by which MMP-9 protects against CAC in mice. METHODS: C57/B6 wild-type mice were given a single dose of azoxymethane and 2 cycles of dextran sulfate sodium (DSS). Mice were also given the γ-secretase inhibitor difluorophenacetyl-l-alanyl-S-phenylglycine t-butyl ester (DAPT) or dimethyl sulfoxide (control) during each DSS cycle; they were killed on day 56. We analyzed embryonic fibroblasts isolated from wild-type and MMP-9-/- mice and HCT116 cells that were stably transfected with MMP-9. RESULTS: Wild-type mice were more susceptible to CAC following inhibition of Notch1 by DAPT, shown by increased numbers of tumors and level of dysplasia compared with controls. Inhibition of Notch1 signaling significantly reduced protein levels of active Notch1, p53, p21WAF1/Cip1, Bax-1, active caspase-3, as well as apoptosis, compared with controls. Similar results were observed in transgenic HCT116 cells and embryonic fibroblasts from MMP-9-/- mice on γ-radiation-induced damage of DNA. CONCLUSIONS: MMP-9 mediates Notch1 signaling via p53 to regulate apoptosis, cell cycle arrest, and inflammation. By these mechanisms, it might prevent CAC.


Asunto(s)
Colitis/enzimología , Colon/enzimología , Neoplasias del Colon/enzimología , Metaloproteinasa 9 de la Matriz/metabolismo , Receptor Notch1/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Apoptosis , Azoximetano , Caspasa 3/metabolismo , Colitis/inducido químicamente , Colitis/patología , Colon/efectos de los fármacos , Colon/inmunología , Colon/patología , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Citocinas/genética , Citocinas/metabolismo , Daño del ADN , Sulfato de Dextran , Dipéptidos/farmacología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Fibroblastos/enzimología , Fibroblastos/efectos de la radiación , Rayos gamma , Células HCT116 , Humanos , Metaloproteinasa 9 de la Matriz/deficiencia , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/metabolismo , Receptor Notch1/antagonistas & inhibidores , Transducción de Señal , Factores de Tiempo , Transfección , Proteína p53 Supresora de Tumor/metabolismo
9.
Mol Cancer ; 9: 63, 2010 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-20298593

RESUMEN

BACKGROUND: Both mutational inactivation of the adenomatous polyposis coli (APC) tumor suppressor gene and activation of the KRAS oncogene are implicated in the pathogenesis of colorectal cancer. Mice harboring a germline ApcMin mutation or intestine-specific expression of the KRASV12 gene have been developed. Both mouse strains develop spontaneous intestinal tumors, including adenoma and carcinoma, though at a different age. The zinc finger transcription factor Krüppel-like factor 5 (KLF5) has previously been shown to promote proliferation of intestinal epithelial cells and modulate intestinal tumorigenesis. Here we investigated the in vivo effect of Klf5 heterozygosity on the propensity of ApcMin/KRASV12 double transgenic mice to develop intestinal tumors. RESULTS: At 12 weeks of age, ApcMin/KRASV12 mice had three times as many intestinal tumors as ApcMin mice. This increase in tumor number was reduced by 92% in triple transgenic ApcMin/KRASV12/Klf5+/- mice. The reduction in tumor number in ApcMin/KRASV12/Klf5+/- mice was also statistically significant compared to ApcMin mice alone, with a 75% decrease. Compared with ApcMin/KRASV12, tumors from both ApcMin/KRASV12/Klf5+/- and ApcMin mice were smaller. In addition, tumors from ApcMin mice were more distally distributed in the intestine as contrasted by the more proximal distribution in ApcMin/KRASV12 and ApcMin/KRASV12/Klf5+/- mice. Klf5 levels in the normal-appearing intestinal mucosa were higher in both ApcMin and ApcMin/KRASV12 mice but were attenuated in ApcMin/KRASV12/Klf5+/- mice. The levels of beta-catenin, cyclin D1 and Ki-67 were also reduced in the normal-appearing intestinal mucosa of ApcMin/KRASV12/Klf5+/- mice when compared to ApcMin/KRASV12 mice. Levels of pMek and pErk1/2 were elevated in the normal-appearing mucosa of ApcMin/KRASV12 mice and modestly reduced in ApcMin/KRASV12/Klf5+/- mice. Tumor tissues displayed higher levels of both Klf5 and beta-catenin, irrespective of the mouse genotype from which tumors were derived. CONCLUSIONS: Results of the current study confirm the cumulative effect of Apc loss and oncogenic KRAS activation on intestinal tumorigenesis. The drastic reduction in tumor number and size due to Klf5 heterozygosity in ApcMin/KRASV12 mice indicate a critical function of KLF5 in modulating intestinal tumor initiation and progression.


Asunto(s)
Transformación Celular Neoplásica/genética , Neoplasias Colorrectales/genética , Genes APC , Factores de Transcripción de Tipo Kruppel/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Transgénicos , Mutación , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
10.
Gastroenterology ; 136(5): 1711-20, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19328876

RESUMEN

BACKGROUND & AIMS: Chronic inflammation is a risk factor for colon cancer (CC). Lysophosphatidic acid (LPA), a naturally produced phospholipid, mediates multiple effects that are vital to disease process, including inflammation and cancer. The expression of LPA receptor 2 (LPA2) is up-regulated in several types of cancer, including ovarian and colon cancer, but the importance of LPA and LPA2 in the development and progression of CC is unclear. In this study, we sought to determine whether LPA and LPA2 regulate the progression of CC in vivo. METHODS: We examined the potential role of LPA in CC progression by administering LPA to mice heterozygous for the adenomatous polyposis coli (Apc) allele. We determined the loss of LPA2 function in tumorigenesis in the colon by treating mice with genetic deletion of LPA2 (LPA2-/-) with azoxymethane and dextran sulfate sodium. RESULTS: We found that LPA increased tumor incidence in Apc(min/+) mice. LPA2-/- mice showed reduced mucosal damage and fewer tumors than wild-type (WT) mice. Reduced epithelial cell proliferation and decreases in beta-catenin, Krüppel-like factor 5, and cyclooxygenase-2 expression were observed in LPA2-/- mice. Unlike WT mice, induction of monocyte chemoattractant protein-1 and macrophage migration inhibitory factor was significantly attenuated in LPA2-/- mice with reduced infiltration by macrophages. CONCLUSIONS: These results show that LPA is capable of promoting tumorigenesis in the colon. The absence of LPA2 attenuates several effects that contribute to cancer progression in vivo, and, hence, the current study identifies LPA2 as an important modulator of CC.


Asunto(s)
Colitis/complicaciones , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Receptores del Ácido Lisofosfatídico/metabolismo , Animales , Azoximetano , Proliferación Celular/efectos de los fármacos , Quimiocina CCL2/metabolismo , Neoplasias del Colon/etiología , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Progresión de la Enfermedad , Genes APC , Factores de Transcripción de Tipo Kruppel/metabolismo , Lipopolisacáridos/farmacología , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , beta Catenina/metabolismo
11.
Mol Cancer Res ; 6(12): 1920-7, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19074836

RESUMEN

The zinc finger-containing transcription factor, Krüppel-like factor 4 (KLF4), inhibits cell proliferation. An in vivo tumor-suppressive role for KLF4 is shown by the recent finding that Klf4 haploinsufficiency in Apc(Min/+) mice promotes intestinal tumorigenesis. Studies also show that KLF4 is required for the terminal differentiation of goblet cells in the mouse intestine. The Notch signaling pathway suppresses goblet cell formation and is up-regulated in intestinal tumors. Here, we investigated the relationship between Notch signaling and KLF4 expression in intestinal epithelial cells. The rate of proliferation of HT29 human colon cancer cells was reduced when treated with the gamma-secretase inhibitor dibenzazepine to inhibit Notch signaling or small interfering RNA directed against Notch. KLF4 levels were increased in dibenzazepine-treated or Notch small interfering RNA-treated cells. Conversely, overexpression of Notch in HT29 cells reduced KLF4 levels, suppressed KLF4 promoter activity, and increased proliferation rate. Treatment of Apc(Min/+) mice with dibenzazepine resulted in a 50% reduction in the number of intestinal adenomas compared with the vehicle-treated group (P < 0.001). Both the normal-appearing intestinal mucosa and adenomas obtained from dibenzazepine-treated Apc(Min/+) mice had increased goblet cell numbers and Klf4 staining accompanied by reduced cyclin D1 and Ki-67 staining when compared with those from vehicle-treated mice. Results of these studies indicate that Notch signaling suppresses KLF4 expression in intestinal tumors and colorectal cancer cells. Inhibition of Notch signaling increases KLF4 expression and goblet cell differentiation and reduces proliferation and tumor formation. KLF4 is therefore a potential mediator for the antitumor effect of Notch inhibitors such as dibenzazepine.


Asunto(s)
Adenoma/fisiopatología , Neoplasias Colorrectales/fisiopatología , Células Caliciformes/fisiología , Mucosa Intestinal/citología , Factores de Transcripción de Tipo Kruppel/genética , Receptor Notch1/metabolismo , Adenoma/metabolismo , Adenoma/patología , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , División Celular/fisiología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Dibenzazepinas/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/fisiología , Células Caliciformes/citología , Células HT29 , Humanos , Mucosa Intestinal/fisiología , Factor 4 Similar a Kruppel , Ratones , Ratones Endogámicos , Ratones Mutantes , Receptor Notch1/antagonistas & inhibidores , Receptor Notch1/genética , Transducción de Señal/fisiología
12.
Gastroenterology ; 134(1): 120-30, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18054006

RESUMEN

BACKGROUND & AIMS: Krüppel-like factor 5 (KLF5) is a zinc finger-transcription factor that regulates cell proliferation. Oncogenic KRAS mutations are commonly found in colorectal cancers. We aimed to determine whether KLF5 mediates KRAS functions during intestinal tumorigenesis. METHODS: The effects of KLF5 on proliferation and transformation were examined in IEC-6 intestinal epithelial cells stably transfected with inducible KRAS(V12G). KLF5 expression was examined in intestinal tumors derived from transgenic mice expressing KRAS(V12G) under villin promoter and in human colorectal cancers with mutated KRAS. RESULTS: Induction of KRAS(V12G) in IEC-6 cells resulted in increased expression of KLF5, accompanied by increased rates of proliferation and anchorage-independent growth. Inhibition of KLF5 expression by mitogen-activated protein kinase/extracellular signal-regulated kinase (MEK) inhibitors or KLF5-specific small interfering RNA reduced proliferation and anchorage-independent growth despite KRAS(V12G) induction. Human colorectal cancer cell lines with mutated KRAS contained high levels of KLF5 and reduction of KLF5 by MEK inhibitors or KLF5 small interfering RNA also led to reduced proliferation and transformation. In vivo, both intestinal tumors derived from mice transgenic for villin-KRAS(V12G) and human primary colorectal cancers with mutated KRAS contained high levels of KLF5 and increased staining of the proliferative marker Ki67. CONCLUSIONS: Elevated levels of KLF5 protein are strongly correlated with activating KRAS mutations in intestinal tumors in vitro and in vivo. Inhibition of KLF5 expression in tumor cells resulted in significantly reduced rates of proliferation and transforming activities. We conclude that KLF5 is an important mediator of oncogenic KRAS transforming functions during intestinal tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica/patología , Neoplasias Colorrectales/etiología , Células Epiteliales/fisiología , Mucosa Intestinal/patología , Factores de Transcripción de Tipo Kruppel/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas ras/fisiología , Animales , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/patología , Humanos , Ratones , Proteínas Proto-Oncogénicas p21(ras)
13.
Cancer Res ; 67(8): 3862-70, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17440101

RESUMEN

We have previously discovered the naturally occurring antitussive alkaloid noscapine as a tubulin-binding agent that attenuates microtubule dynamics and arrests mammalian cells at mitosis via activation of the c-Jun NH(2)-terminal kinase pathway. It is well established that the p53 protein plays a crucial role in the control of tumor cell response to chemotherapeutic agents and DNA-damaging agents; however, the relationship between p53-driven genes and drug sensitivity remains controversial. In this study, we compared chemosensitivity, cell cycle distribution, and apoptosis on noscapine treatment in four cell lines derived from the colorectal carcinoma HCT116 cells: p53(+/+) (p53-wt), p53(-/-) (p53-null), p21(-/-) (p21-null), and BAX(-/-) (BAX-null). Using these isogenic variants, we investigated the roles of p53, BAX, and p21 in the cellular response to treatment with noscapine. Our results show that noscapine treatment increases the expression of p53 over time in cells with wild-type p53 status. This increase in p53 is associated with an increased apoptotic BAX/Bcl-2 ratio consistent with increased sensitivity of these cells to apoptotic stimuli. Conversely, loss of p53 and p21 alleles had a counter effect on both BAX and Bcl-2 expression and the p53-null and p21-null cells were significantly resistant to the antiproliferative and apoptotic effects of noscapine. All but the p53-null cells displayed p53 protein accumulation in a time-dependent manner on noscapine treatment. Interestingly, despite increased levels of p53, p21-null cells were resistant to apoptosis, suggesting a proapoptotic role of p21 and implying that p53 is a necessary but not sufficient condition for noscapine-mediated apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Noscapina/farmacología , Proteína p53 Supresora de Tumor/biosíntesis , Alelos , Apoptosis/fisiología , División Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Fase G2/efectos de los fármacos , Células HCT116 , Humanos , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Proteína X Asociada a bcl-2/metabolismo
14.
Cancer Res ; 67(15): 7147-54, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17671182

RESUMEN

The zinc finger transcription factor Krüppel-like factor 4 (KLF4) is frequently down-regulated in colorectal cancer. Previous studies showed that the expression of KLF4 was activated by the colorectal cancer tumor suppressor adenomatous polyposis coli (APC) and that KLF4 repressed the Wnt/beta-catenin pathway. Here, we examined whether KLF4 plays a role in modulating intestinal tumorigenesis by comparing the tumor burdens in mice heterozygous for the Apc(Min) allele (Apc(Min/+)) and those heterozygous for both the Apc(Min) and Klf4 alleles (Klf4(+/-)/Apc(Min/+)). Between 10 and 20 weeks of age, Klf4(+/-)/Apc(Min/+) mice developed, on average, 59% more intestinal adenomas than Apc(Min/+) mice (P < 0.0001). Immunohistochemical staining showed that Klf4 protein levels were lower in the normal-appearing intestinal tissues of Klf4(+/-)/Apc(Min/+) mice compared with wild-type, Klf4(+/-), or Apc(Min/+) mice. In contrast, the levels of beta-catenin and cyclin D1 were higher in the normal-appearing intestinal tissues of Klf4(+/-)/Apc(Min/+) mice compared with the other three genotypes. Klf4 levels were further decreased in adenomas from both Apc(Min/+) and Klf4(+/-)/Apc(Min/+) mice compared with their corresponding normal-appearing tissues. Reverse transcription-PCR showed an inverse correlation between adenoma size and Klf4 mRNA levels in both Klf4(+/-)/Apc(Min/+) and Apc(Min/+) mice. There was also a progressive loss of heterozygosity of the wild-type Apc allele in adenomas with increasing size from Klf4(+/-)/Apc(Min/+) and Apc(Min/+) mice. Results from this study show that KLF4 plays an important role in promoting the development of intestinal adenomas in the presence of Apc(Min) mutation.


Asunto(s)
Poliposis Adenomatosa del Colon/metabolismo , Transformación Celular Neoplásica/metabolismo , Neoplasias Intestinales/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Adenoma/genética , Adenoma/metabolismo , Adenoma/patología , Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/patología , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Ciclina D , Ciclinas/genética , Ciclinas/metabolismo , Femenino , Técnicas para Inmunoenzimas , Neoplasias Intestinales/genética , Neoplasias Intestinales/patología , Pólipos Intestinales/genética , Pólipos Intestinales/metabolismo , Pólipos Intestinales/patología , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Pérdida de Heterocigocidad , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , beta Catenina/metabolismo
15.
Artículo en Inglés | MEDLINE | ID: mdl-32566755

RESUMEN

AIM: Colorectal cancer (CRC) is the third leading cancer-related cause of death due to its propensity to metastasize. Epithelial-mesenchymal transition (EMT) is a multistep process important for invasion and metastasis of CRC. Krüppel-like factor 4 (KLF4) is a zinc finger transcription factor highly expressed in differentiated cells of the intestinal epithelium. KLF4 has been shown to play a tumor suppressor role during CRC tumorigenesis - its loss accelerates development and progression of cancer. The present study examined the relationship between KLF4 and markers of EMT in CRC. METHODS: Immunofluorescence staining for KLF4 and EMT markers was performed on archived patient samples after colorectal cancer resection and on colonic tissues of mice with colitis-associated cancer. RESULTS: We found that KLF4 expression is lost in tumor sections obtained from CRC patients and in those of mouse colon following azoxymethane and dextran sodium sulfate (AOM/DSS) treatment when compared to their respective normal appearing mucosa. Importantly, in CRC patient tumor sections, we observed a negative correlation between KLF4 levels and mesenchymal markers including TWIST, ß-catenin, claudin-1, N-cadherin, and vimentin. Similarly, in tumor tissues from AOM/DSS-treated mice, KLF4 levels were negatively correlated with mesenchymal markers including SNAI2, ß-catenin, and vimentin and positively correlated with the epithelial marker E-cadherin. CONCLUSION: These findings suggest that the loss of KLF4 expression is a potentially significant indicator of EMT in CRC.

16.
Gene ; 611: 27-37, 2017 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-28237823

RESUMEN

Krüppel-like factor 4 (KLF4) is an evolutionarily conserved zinc finger-containing transcription factor that regulates diverse cellular processes such as cell growth, proliferation, and differentiation. Since its discovery in 1996, KLF4 has been gaining a lot of attention, particularly after it was shown in 2006 as one of four factors involved in the induction of pluripotent stem cells (iPSCs). Here we review the current knowledge about the different functions and roles of KLF4 in various tissue and organ systems.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular/genética , Regulación de la Expresión Génica , Células Madre Pluripotentes Inducidas/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Animales , Secuencia de Bases , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Homología de Secuencia de Aminoácido
17.
Oncogene ; 24(25): 4017-25, 2005 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-15806166

RESUMEN

Centrosome duplication is a carefully controlled process in the cell cycle. Previous studies indicate that the tumor suppressor, p53, regulates centrosome duplication. Here, we present evidence for the involvement of the mammalian Krüppel-like transcription factor, KLF4, in preventing centrosome amplification following DNA damage caused by gamma-irradiation. The colon cancer cell line HCT116, which contains wild-type p53 alleles (HCT116 p53+/+), displayed stable centrosome numbers following gamma-irradiation. In contrast, HCT116 cells null for the p53 alleles (HCT116 p53-/-) exhibited centrosome amplification after irradiation. In the latter cell line, KLF4 was not activated following gamma-irradiation due to the absence of p53. However, centrosome amplification could be suppressed in irradiated HCT116 p53-/- cells by conditional induction of exogenous KLF4. Conversely, in a HCT116 p53+/+ cell line stably transfected with small hairpin RNA (shRNA) designed to specifically inhibit KLF4, gamma-irradiation induced centrosome amplification. In these cells, the inability of KLF4 to become activated in response to DNA damage was directly associated with an increase in cyclin E level and Cdk2 activity, both essential for regulating centrosome duplication. Cotransfection experiments showed that KLF4 overexpression suppressed the promoter activity of the cyclin E gene. The results of this study demonstrated that KLF4 is both necessary and sufficient in preventing centrosome amplification following gamma-radiation-induced DNA damage and does so by transcriptionally suppressing cyclin E expression.


Asunto(s)
Centrosoma/fisiología , Daño del ADN/efectos de la radiación , Proteínas de Unión al ADN/fisiología , Rayos gamma , Factores de Transcripción/fisiología , Línea Celular Tumoral , Neoplasias del Colon , Ciclina E/genética , Eliminación de Gen , Genes p53 , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel , ARN/genética , Transfección
18.
Microbes Infect ; 8(9-10): 2367-75, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16849041

RESUMEN

In vitro experiments on adult Schistosoma mansoni worms have shown the existence of mutual attraction between males and females mediated by factor(s) released by both sexes. Males were attracted to female-released factor(s), which was found in <30 kDa fractions. A combination of bioassays, size fractionations, chromatographic and chemical and enzymatic treatments were used to identify female-released factor(s) that attracted males. Boiling reduced the attraction of males towards female incubate. Male-attractant factor(s) was not in the female vomitus, and TLC showed the absence of lipids and carbohydrates in the female incubate. Male-attractant factor(s) was found to be sensitive to proteases and to be hydrophilic in nature. Using RP-HPLC, a 3260 Da peptide released by S. mansoni adult females that plays a role in attracting the males was identified, purified and partially characterized. A partial amino acid sequence of that peptide was obtained using MALDI peptide-MS and MALDI-TOF/TOF.


Asunto(s)
Péptidos/química , Schistosoma mansoni/química , Atractivos Sexuales/química , Secuencia de Aminoácidos , Animales , Cromatografía Líquida de Alta Presión/métodos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Datos de Secuencia Molecular , Péptidos/aislamiento & purificación , Péptidos/fisiología , Schistosoma mansoni/fisiología , Atractivos Sexuales/aislamiento & purificación , Atractivos Sexuales/fisiología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos
19.
Gene Expr ; 13(2): 85-96, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17017123

RESUMEN

Krüppel-like factor 4 (KLF4; also known as gut-enriched Krüppel-like factor or GKLF) is known to exhibit checkpoint function during the G1/S and G2/M transitions of the cell cycle. The mechanism by which KLF4 exerts these effects is not fully established. Here we investigated the expression profile of KLF4 in an inducible system over a time course of 24 h. Using oligonucleotide microarrays, we determined that the fold changes relative to control in expression levels of KLF4 exhibited a time-dependent increase from 3- to 20-fold between 4 and 24 h following KLF4 induction. During this period and among a group of 473 cell cycle regulatory genes examined, 96 were positively correlated and 86 were negatively correlated to KLF4's expression profile. Examples of upregulated cell cycle genes include those encoding tumor suppressors such as MCC and FHIT, and cell cycle inhibitors such as CHES1 and CHEK1. Examples of downregulated genes include those that promote the cell cycle including several cyclins and those required for DNA replication. Unexpectedly, several groups of genes involved in macromolecular synthesis, including protein biosynthesis, transcription, and cholesterol biosynthesis, were also significantly inhibited by KLF4. Thus, KLF4 exerts a global inhibitory effect on macromolecular biosynthesis that is beyond its established role as a cell cycle inhibitor.


Asunto(s)
Ciclo Celular/genética , Perfilación de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/genética , Transcripción Genética , Línea Celular Tumoral , Colesterol/biosíntesis , Neoplasias del Colon , Regulación de la Expresión Génica , Humanos , Cinética , Factor 4 Similar a Kruppel , Análisis de Secuencia por Matrices de Oligonucleótidos , Transfección
20.
Mol Cancer Res ; 14(4): 385-96, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26839262

RESUMEN

UNLABELLED: The zinc finger transcription factor Krüppel-like factor 4 (KLF4) is frequently downregulated in colorectal cancer. Previous studies showed that KLF4 is a tumor suppressor in the intestinal tract and plays an important role in DNA damage-repair mechanisms. Here, the in vivo effects of Klf4 deletion were examined from the mouse intestinal epithelium (Klf4(ΔIS)) in a genetic or pharmacological setting of colonic tumorigenesis:Apc(Min/⁺) mutation or carcinogen treatment with azoxymethane (AOM), respectively.Klf4 (ΔIS)/Apc (Min/⁺) mice developed significantly more colonic adenomas with 100% penetrance as compared with Apc(Min/⁺) mice with intact Klf4 (Klf4(fl/fl)/Apc (Min/⁺)). The colonic epithelium of Klf4 (ΔIS)/Apc (Min/⁺)mice showed increased mTOR pathway activity, together with dysregulated epigenetic mechanism as indicated by altered expression of HDAC1 and p300. Colonic adenomas from both genotypes stained positive for γH2AX, indicating DNA double-strand breaks. InKlf4 (ΔIS)/Apc (Min/+) mice, this was associated with reduced nonhomologous end joining (NHEJ) repair and homologous recombination repair (HRR) mechanisms as indicated by reduced Ku70 and Rad51 staining, respectively. In a separate model, following treatment with AOM, Klf4 (ΔIS) mice developed significantly more colonic tumors than Klf4 (fl/fl) mice, with more Klf4 (ΔIS) mice harboring K-Rasmutations than Klf4 (fl/fl)mice. Compared with AOM-treated Klf4 (fl/fl)mice, adenomas of treated Klf4 (ΔIS) mice had suppressed NHEJ and HRR mechanisms, as indicated by reduced Ku70 and Rad51 staining. This study highlights the important role of KLF4 in suppressing the development of colonic neoplasia under different tumor-promoting conditions. IMPLICATIONS: The study demonstrates that KLF4 plays a significant role in the pathogenesis of colorectal neoplasia.


Asunto(s)
Azoximetano/efectos adversos , Neoplasias del Colon/patología , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Mutación , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Células Cultivadas , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/genética , Epigénesis Genética , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Factor 4 Similar a Kruppel , Ratones , Neoplasias Experimentales , Serina-Treonina Quinasas TOR/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA