Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
J Periodontal Res ; 48(4): 405-10, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23050778

RESUMEN

BACKGROUND AND OBJECTIVE: Wingless-type MMTV integration site family (Wnt)/ß-catenin signaling plays an essential role in cellular differentiation and matrix formation during skeletal development. However, little is known about its role in tooth-root formation. In a previous study, we found excessive formation of dentin and cementum in mice with constitutive ß-catenin stabilization in the dental mesenchyme. In the present study we analyzed the molar roots of these mice to investigate the role of Wnt/ß-catenin signaling in root formation in more detail. MATERIAL AND METHODS: We generated OC-Cre:Catnb(+/lox(ex3)) mice by intercrossing Catnb(+/lox(ex3)) and OC-Cre mice, and we analyzed their mandibular molars using radiography, histomorphometry and immunohistochemistry. RESULTS: OC-Cre:Catnb(+/lox(ex3)) mice showed impaired root formation. At the beginning of root formation in mutant molars, dental papilla cells did not show normal differentiation into odontoblasts; rather, they were prematurely differentiated and had a disorganized arrangement. Interestingly, SMAD family member 4 was upregulated in premature odontoblasts. In 4-wk-old mutant mice, molar roots were about half the length of those in their wild-type littermates. In contrast to excessively formed dentin in crown, root dentin was thin and hypomineralized in mutant mice. Biglycan and dentin sialophosphoprotein were downregulated in root dentin of mutant mice, whereas dentin matrix protein 1 and Dickkopf-related protein 1 were upregulated. Additionally, ectonucleotide pyrophosphatase/phosphodiesterase 1 was significantly downregulated in the cementoblasts of mutant molars. Finally, in the cementum of mutant mice, bone sialoprotein was downregulated but Dickkopf-related protein 2 was upregulated. CONCLUSION: These results suggest that temporospatial regulation of Wnt/ß-catenin signaling plays an important role in cell differentiation and matrix formation during root and cementum formation.


Asunto(s)
Odontogénesis/fisiología , Raíz del Diente/crecimiento & desarrollo , Vía de Señalización Wnt/fisiología , Animales , Biglicano/análisis , Diferenciación Celular/fisiología , Polaridad Celular/fisiología , Cementogénesis/fisiología , Cemento Dental/patología , Papila Dental/patología , Dentina/patología , Dentinogénesis/fisiología , Regulación hacia Abajo , Proteínas de la Matriz Extracelular/análisis , Sialoproteína de Unión a Integrina/análisis , Péptidos y Proteínas de Señalización Intercelular/análisis , Ratones , Ratones Mutantes , Diente Molar/crecimiento & desarrollo , Mutación/genética , Odontoblastos/patología , Fosfoproteínas/análisis , Hidrolasas Diéster Fosfóricas , Pirofosfatasas , Sialoglicoproteínas/análisis , Transducción de Señal/fisiología , Proteína Smad4/análisis , Regulación hacia Arriba , Vía de Señalización Wnt/genética
2.
Nat Med ; 7(9): 1048-51, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11533709

RESUMEN

Arachidonic acid is metabolized to prostaglandin H(2) (PGH(2)) by cyclooxygenase (COX). COX-2, the inducible COX isozyme, has a key role in intestinal polyposis. Among the metabolites of PGH(2), PGE(2) is implicated in tumorigenesis because its level is markedly elevated in tissues of intestinal adenoma and colon cancer. Here we show that homozygous deletion of the gene encoding a cell-surface receptor of PGE(2), EP2, causes decreases in number and size of intestinal polyps in Apc(Delta 716) mice (a mouse model for human familial adenomatous polyposis). This effect is similar to that of COX-2 gene disruption. We also show that COX-2 expression is boosted by PGE(2) through the EP2 receptor via a positive feedback loop. Homozygous gene knockout for other PGE(2) receptors, EP1 or EP3, did not affect intestinal polyp formation in Apc(Delta 716) mice. We conclude that EP2 is the major receptor mediating the PGE2 signal generated by COX-2 upregulation in intestinal polyposis, and that increased cellular cAMP stimulates expression of more COX-2 and vascular endothelial growth factor in the polyp stroma.


Asunto(s)
Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/fisiopatología , Isoenzimas/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Receptores de Prostaglandina E/genética , Adenoma/genética , Adenoma/patología , Poliposis Adenomatosa del Colon/patología , Animales , Ciclooxigenasa 2 , Dinoprostona/metabolismo , Factores de Crecimiento Endotelial/genética , Homocigoto , Isoenzimas/genética , Linfocinas/genética , Ratones , Ratones Noqueados , Prostaglandina-Endoperóxido Sintasas/genética , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/metabolismo , Receptores de Prostaglandina E/metabolismo , Subtipo EP1 de Receptores de Prostaglandina E , Subtipo EP2 de Receptores de Prostaglandina E , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
3.
J Cell Biol ; 151(3): 563-72, 2000 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-11062258

RESUMEN

To investigate the expression and biological roles of cytokeratin 19 (K19) in development and in adult tissues, we inactivated the mouse K19 gene (Krt1-19) by inserting a bacterial beta-galactosidase gene (lacZ) by homologous recombination in embryonic stem cells, and established germ line mutant mice. Both heterozygous and homozygous mutant mice were viable, fertile, and appeared normal. By 7.5-8.0 days post coitum (dpc), heterozygous mutant embryos expressed lacZ in the notochordal plate and hindgut diverticulum, reflecting the fact that the notochord and the gut endoderm are derived from the axial mesoderm-originated cells. In the adult mutant, lacZ was expressed mainly in epithelial tissues. To investigate the possible functional cooperation and synergy between K19 and K8, we then constructed compound homozygous mutants, whose embryos died approximately 10 dpc. The lethality resulted from defects in the placenta where both K19 and K8 are normally expressed. As early as 9. 5 dpc, the compound mutant placenta had an excessive number of giant trophoblasts, but lacked proper labyrinthine trophoblast or spongiotrophoblast development, which apparently caused flooding of the maternal blood into the embryonic placenta. These results indicate that K19 and K8 cooperate in ensuring the normal development of placental tissues.


Asunto(s)
Queratinas/metabolismo , Placenta/embriología , Placenta/metabolismo , Animales , Cruzamientos Genéticos , Embrión de Mamíferos/irrigación sanguínea , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Femenino , Muerte Fetal , Retardo del Crecimiento Fetal , Técnica del Anticuerpo Fluorescente , Galactosidasas/genética , Galactosidasas/metabolismo , Eliminación de Gen , Marcación de Gen , Genes Reporteros , Genotipo , Mutación de Línea Germinal/genética , Hibridación in Situ , Queratinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Fenotipo , Placenta/irrigación sanguínea , Placenta/patología , Circulación Placentaria , Embarazo , ARN Mensajero/análisis , ARN Mensajero/genética , Recombinación Genética , Células Madre/metabolismo , Trofoblastos/metabolismo , Trofoblastos/patología
4.
Oncogene ; 26(32): 4679-88, 2007 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-17297455

RESUMEN

Chemokines and their receptors are essential for leukocyte trafficking, and also implicated in cancer metastasis to specific organs. We have recently demonstrated that CXCR3 plays a critical role in metastasis of mouse melanoma cells to lymph nodes. Here, we show that some human colon cancer cell lines express CXCR3 constitutively. We constructed cells that expressed CXCR3 cDNA ('DLD-1-CXCR3'), and compared with nonexpressing controls by rectal transplantation in nude mice. Although both cell lines disseminated to lymph nodes at similar frequencies at 2 weeks, DLD-1-CXCR3 expanded more rapidly than the control in 4 weeks. In 6 weeks, 59% of mice inoculated with DLD1-CXCR3 showed macroscopic metastasis in para-aortic lymph nodes, whereas only 14% of those with the control (P<0.05). In contrast, metastasis to the liver or lung was rare, and unaffected by CXCR3 expression. In clinical colon cancer samples, we found expression of CXCR3 in 34% cases, most of which had lymph node metastasis. Importantly, patients with CXCR3-positive cancer showed significantly poorer prognosis than those without CXCR3, or those expressing CXCR4 or CCR7. These results indicate that activation of CXCR3 with its ligands stimulates colon cancer metastasis preferentially to the draining lymph nodes with poorer prognosis.


Asunto(s)
Neoplasias del Colon/patología , Ganglios Linfáticos/patología , Receptores de Quimiocina/fisiología , Animales , Movimiento Celular , ADN Complementario/genética , Humanos , Ligandos , Metástasis Linfática , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Receptores CXCR3 , Receptores de Quimiocina/genética
5.
Oncogene ; 26(24): 3511-20, 2007 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-17160019

RESUMEN

Adenomatous polyposis coli (APC/Apc) gene encodes a key tumor suppressor whose mutations activate beta-catenin/T-cell factor (TCF)-mediated transcription (canonical Wnt signaling). Here, we show that Wnt signaling can cause chromosomal instability (CIN). As an indicator of CIN, we scored anaphase bridge index (ABI) in mouse polyps and ES cells where Wnt signaling was activated by Apc or beta-catenin mutations. We found three to nine times higher ABI than in wild-type controls. Furthermore, karyotype analysis confirmed that the Wnt signal-activated ES cells produced new chromosomal aberrations at higher rates; hence CIN. Consistently, expression of dominant-negative TCFs in these cells reduced their ABI. We also found that Wnt signal activation increased phosphorylation of Cdc2 (Cdk1) that inhibited its activity, and suppressed apoptosis upon exposure of the cells to nocodazole or colcemid. The data suggest that Wnt signaling stimulates the cells to escape from mitotic arrest and apoptosis, resulting in CIN. In human gastric cancer tissues with nuclear beta-catenin, ABI was significantly higher than in those without. These results collectively indicate that beta-catenin/TCF-mediated transcription itself increases CIN through dysregulation of G2/M progression.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Inestabilidad Cromosómica , Mutación , Factores de Transcripción TCF/genética , beta Catenina/genética , Adenoma/genética , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , División Celular/genética , Supervivencia Celular/genética , Células Cultivadas , Aberraciones Cromosómicas , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Células Madre Embrionarias , Fase G2/genética , Humanos , Neoplasias Intestinales/genética , Pólipos Intestinales/genética , Ratones , Microtúbulos/metabolismo , Transducción de Señal , Factores de Transcripción TCF/metabolismo , Transcripción Genética , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
6.
Oncogene ; 25(57): 7522-30, 2006 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-17143296

RESUMEN

The canonical Wnt signaling plays important roles in embryonic development and tumorigenesis. For the latter, induced mutations in mice have greatly contributed to our understanding of the molecular mechanisms of cancer initiation and progression. Here, I will review recent reports on gastrointestinal cancer model mice, with an emphasis on the roles of the Wnt signal pathway. They include: mouse models for familial adenomatous polyposis; modifying factors that affect mouse intestinal polyposis, including the genes that help cancer progression; Wnt target genes that affect mouse intestinal polyposis; and a mouse model of gastric cancer that mimics Helicobacter pyroli infection.


Asunto(s)
Neoplasias Gastrointestinales/metabolismo , Proteínas Wnt/fisiología , Poliposis Adenomatosa del Colon/genética , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Neoplasias Gastrointestinales/genética , Humanos , Ratones , Ratones Transgénicos , Modelos Biológicos , Mutación , Transducción de Señal , Proteínas Wnt/genética
7.
Oncogene ; 25(12): 1816-20, 2006 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-16278673

RESUMEN

Germline mutations in the LKB1 gene are responsible for Peutz-Jeghers syndrome (PJS), which is characterized by gastrointestinal hamartomas and increasing risk of cancer. Mice with Lkb1(+/-) mutation develop gastric hamartomas after >20 weeks of age, and hepatocellular adenomas and carcinomas >30 weeks. It has been reported that, in PJS patients, carcinomas progressed from hamartomas contain p53 mutations, and that LKB1 regulates p53-dependent apoptosis. To investigate the roles of LKB1 and p53 mutations in tumorigenesis, we constructed compound mutant mice of Lkb1 and p53 genes. In the Lkb1(+/-)p53(-/-) mice, formation of gastric hamartomas and hepatic tumors was accelerated. However, histopathology of hamartomas was similar between Lkb1(+/-)p53(-/-) and Lkb1(+/-) mice, and Lkb1 genotype remained heterozygous, suggesting that the p53 mutation affected hamartoma initiation. Contrary to the heterozygous hamartomas in the stomach and duodenum, the hepatic adenomas in Lkb1(+/-)p53(-/-) mice showed loss of Lkb1 heterozygosity (LOH), suggesting that lack of p53 stimulated Lkb1 LOH and tumor initiation in the liver. Taken together, these results indicate that lack of p53 causes earlier onsets of gastric hamartomas and hepatic tumors in Lkb1(+/-)p53(-/-) mice.


Asunto(s)
Adenoma/genética , Hamartoma/genética , Neoplasias Hepáticas/genética , Proteínas Serina-Treonina Quinasas/genética , Gastropatías/genética , Proteína p53 Supresora de Tumor/genética , Proteínas Quinasas Activadas por AMP , Animales , Western Blotting , Modelos Animales de Enfermedad , Femenino , Pérdida de Heterocigocidad , Masculino , Ratones , Ratones Transgénicos , Síndrome de Peutz-Jeghers/genética
8.
Cytokine Growth Factor Rev ; 11(1-2): 147-57, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10708962

RESUMEN

The SMAD4 gene plays a key role in the TGF-beta signaling pathway. We inactivated its mouse homolog Smad4. The homozygous mutants were embryonically lethal, whereas the heterozygotes were viable and fertile. Although young heterozygotes appeared normal, old mice developed gastric and duodenal polyps similar to human juvenile polyps characterized by abundant stroma and eosinophilic infiltrations. These data are consistent with the reports that a subset of human juvenile polyposis kindreds carry germline mutations in the SMAD4 gene. We then introduced the Smad4 mutation into the Apc(Delta716) knockout mice, a model for human familial adenomatous polyposis. Because both Apc and Smad4 are located on mouse chromosome 18, we constructed by meiotic recombination compound heterozygotes carrying both mutations on the same chromosome. In such mice, intestinal polyps developed into more malignant tumors than those in the simple Apc(Delta716) heterozygotes, showing an extensive stromal cell proliferation and strong submucosal invasion. These results indicate that mutations in SMAD4 play a significant role in the malignant progression of colorectal tumors.


Asunto(s)
Proteínas de Unión al ADN/genética , Neoplasias Gastrointestinales/etiología , Transactivadores/genética , Proteína de la Poliposis Adenomatosa del Colon , Animales , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN/metabolismo , Neoplasias Gastrointestinales/patología , Heterocigoto , Ratones , Ratones Noqueados , Ratones Mutantes , Proteínas de Neoplasias/genética , Fenotipo , Proteína Smad4 , Transactivadores/metabolismo
9.
Oncogene ; 36(46): 6480-6489, 2017 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-28759045

RESUMEN

Mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) is frequently activated in cancers and can be counteracted with the clinical mTORC1 inhibitors everolimus and temsirolimus. Although mTORC1 and dual mTORC1/2 inhibitors are currently under development to treat various malignancies, the emergence of drug resistance has proven to be a major complication. Using the cis-Apc/Smad4 mouse model of locally invasive intestinal adenocarcinoma, we show that administration of everolimus or the dual mTORC1/2 inhibitor AZD8055 significantly reduces the growth of intestinal tumors. In contrast, although everolimus treatment at earlier phase of tumor progression delayed invasion of the tumors, both inhibitors exhibited little effect on blocking invasion of the tumors when administered later in their progression. Biochemical and immunohistochemical analyses revealed that treatment of cis-Apc/Smad4 mice with everolimus or AZD8055 induced marked increases in epidermal growth factor receptor (EGFR) and MEK/ERK signaling in tumor epithelial and stromal cells, respectively. Notably, co-administration of AZD8055 and the EGFR inhibitor erlotinib or the MEK inhibitor trametinib was sufficient to suppress tumor invasion in cis-Apc/Smad4 mice. These data indicate that mTOR inhibitor resistance in invasive intestinal tumors involves feedback signaling from both cancer epithelial and stromal cells, highlighting the role of tumor microenvironment in drug resistance, and support that simultaneous inhibition of mTOR and EGFR or MEK may be more effective in treating colon cancer.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Neoplasias Intestinales/tratamiento farmacológico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Microambiente Tumoral/efectos de los fármacos , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Modelos Animales de Enfermedad , Receptores ErbB/metabolismo , Clorhidrato de Erlotinib/farmacología , Everolimus/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Células HT29 , Humanos , Neoplasias Intestinales/genética , Neoplasias Intestinales/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Morfolinas/farmacología , Invasividad Neoplásica , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo
10.
Oncogene ; 36(42): 5885-5896, 2017 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-28628120

RESUMEN

Tumor suppressor TP53 is frequently mutated in colorectal cancer (CRC), and most mutations are missense type. Although gain-of-functions by mutant p53 have been demonstrated experimentally, the precise mechanism for malignant progression in in vivo tumors remains unsolved. We generated ApcΔ716 Trp53LSL•R270H villin-CreER compound mice, in which mutant p53R270H was expressed in the intestinal epithelia upon tamoxifen treatment, and examined the intestinal tumor phenotypes and tumor-derived organoids. Mutant Trp53R270H, but not Trp53-null mutation accelerated submucosal invasion with generation of desmoplastic microenvironment. The nuclear accumulation of p53 was evident in ApcΔ716 Trp53R270H/R270H homozygous tumors like human CRC. Although p53 was distributed to the cytoplasm in ApcΔ716 Trp53+/R270H heterozygous tumors, it accumulated in the nuclei at the invasion front, suggesting a regulation mechanism for p53 localization by the microenvironment. Importantly, mutant p53 induced drastic morphological changes in the tumor organoids to complex glandular structures, which was associated with the acquisition of invasiveness. Consistently, the branching scores of human CRC that carry TP53 mutations at codon 273 significantly increased in comparison with those of TP53 wild-type tumors. Moreover, allografted ApcΔ716 Trp53R270H/R270H organoid tumors showed a malignant histology with an increased number of myofibroblasts in the stroma. These results indicate that nuclear-accumulated mutant p53R270H induces malignant progression of intestinal tumors through complex tumor gland formation and acquisition of invasiveness. Furthermore, RNA sequencing analyses revealed global gene upregulation by mutant p53R270H, which was associated with the activation of inflammatory and innate immune pathways. Accordingly, it is possible that mutant p53R270H induces CRC progression, not only by a cell intrinsic mechanism, but also by the generation or activation of the microenvironment, which may synergistically contribute to the acceleration of submucosal invasion. Therefore, the present study indicates that nuclear-accumulated mutant p53R270H is a potential therapeutic target for the treatment of advanced CRCs.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias Intestinales/patología , Neoplasias Hepáticas/secundario , Mutación , Proteína p53 Supresora de Tumor/genética , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Humanos , Neoplasias Intestinales/genética , Neoplasias Intestinales/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Invasividad Neoplásica , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Microambiente Tumoral , Proteína p53 Supresora de Tumor/metabolismo
11.
J Natl Cancer Inst ; 90(21): 1609-20, 1998 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-9811310

RESUMEN

The rate-limiting step in arachidonate metabolism is mediated by enzymes known as cyclooxygenases (COXs). These enzymes catalyze the biosynthesis of prostaglandin H2, the precursor of molecules such as prostaglandins, prostacyclin, and thromboxanes. The COX enzyme family consists of the classical COX-1 enzyme, which is constitutively expressed in many tissues, and a second isozyme, i.e., COX-2, which is induced by various stimuli, such as mitogens and cytokines, and is involved in many inflammatory reactions. Because nonsteroidal anti-inflammatory drugs (NSAIDs) inhibit both COX-1 and COX-2, these drugs also cause unwanted side effects, exemplified by gastrointestinal bleeding. Accumulating evidence indicates that NSAIDs can reduce the incidence of colorectal cancers in human and experimental animals and can reduce the number and size of polyps in patients with familial adenomatous polyposis. This Part II (of a two-part review) focuses on the growing clinical and experimental evidence that NSAIDS and COX-2 inhibitors can influence the risk of colon (and possibly of other) cancers.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/prevención & control , Inhibidores de la Ciclooxigenasa/farmacología , Isoenzimas/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Animales , Ensayos Clínicos como Asunto , Ciclooxigenasa 1 , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inducción Enzimática/efectos de los fármacos , Humanos , Isoenzimas/efectos de los fármacos , Isoenzimas/genética , Proteínas de la Membrana , Neoplasias/enzimología , Neoplasias/prevención & control , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/prevención & control , Prostaglandina-Endoperóxido Sintasas/efectos de los fármacos , Prostaglandina-Endoperóxido Sintasas/genética , Prostaglandinas/metabolismo , Sulindac/farmacología
12.
J Natl Cancer Inst ; 90(20): 1529-36, 1998 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-9790545

RESUMEN

The rate-limiting enzyme in arachidonate metabolism is mediated by enzymes known as cyclooxygenases (COXs). These enzymes catalyze the biosynthesis of prostaglandin H2, the precursor of molecules, such as prostaglandins, prostacyclin, and thromboxanes. The COX enzyme family consists of the classical COX-1 enzyme, which is constitutively expressed in many tissues, and a second enzyme, i.e., COX-2, which is induced by various stimuli, such as mitogens and cytokines, and is involved in many inflammatory reactions. Because nonsteroidal anti-inflammatory drugs inhibit both COX-1 and COX-2, these drugs also cause unwanted side effects, exemplified by gastrointestinal bleeding. Accumulating evidence indicates that nonsteroidal anti-inflammatory drugs can reduce the incidence of colorectal cancers in human and experimental animals and can reduce the polyp number and size in patients with familial adenomatous polyposis. This Part I (of a two-part review) focuses on the discovery of the COXs; their biochemical, molecular, and structural properties; and on the discovery of isozyme-specific inhibitors of COX activity.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/prevención & control , Inhibidores de la Ciclooxigenasa/farmacología , Isoenzimas/química , Prostaglandina-Endoperóxido Sintasas/química , Animales , Antiinflamatorios no Esteroideos/química , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Modelos Animales de Enfermedad , Humanos , Proteínas de la Membrana
13.
Cancer Res ; 57(9): 1644-9, 1997 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-9135000

RESUMEN

Mutations in the human adenomatous polyposis coli (APC) gene are responsible for not only familial adenomatous polyposis but also many sporadic cancers of the digestive tract. Using homologous recombination in embryonic stem cells, we recently constructed Apc gene knockout mice that contained a truncation mutation at codon 716 (Apc(delta716)). The heterozygous mice developed numerous intestinal polyps. All microadenomas dissected from nascent polyps had already lost the wild-type allele, indicating the loss of heterozygosity (M. Oshima et al., Proc. Natl. Acad. Sci. USA, 92: 4482-4486, 1995). We also demonstrated that cyclooxygenase 2 is induced in the polyps at an early stage and plays a key role in polyp development (M. Oshima et al., Cell 87: 803-809, 1996). We have analyzed the process of polyp development in these mice both at morphological and molecular levels. A small intestinal microadenoma is initiated as an outpocketing pouch in a single crypt and develops into the inner (lacteal) side of a neighboring villus forming a double-layer nascent polyp. The microadenoma then enlarges and gets folded inside the villus. When it fills the intravillous space, it expands downward and extends into adjoining villi, rather than rupturing into the intestinal lumen. During this course of development, the basement membrane remains intact, and the labeling index of the microadenoma cells is similar to that of the normal crypt epithelium. As in the crypt cells, neither transforming growth factor beta1 nor its receptor type II is expressed in the microadenoma cells. No hot spot mutations in the K-ras gene are found in the microadenoma tissue during these early stages of polyp development. Essentially, the same results have been obtained for the colonic polyps as well. These results suggest that early adenomas in the Apc(delta716) polyps are very similar to the normal proliferating cells of the crypt except for the lack of directed migration along the crypt-villus axis.


Asunto(s)
Receptores de Activinas Tipo I , Proteínas del Citoesqueleto/deficiencia , Pólipos Intestinales/patología , Adenoma/genética , Adenoma/patología , Proteína de la Poliposis Adenomatosa del Colon , Animales , División Celular , Heterocigoto , Neoplasias Intestinales/genética , Neoplasias Intestinales/patología , Pólipos Intestinales/genética , Ratones , Ratones Noqueados , Lesiones Precancerosas/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo
14.
Cancer Res ; 55(13): 2719-22, 1995 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-7796392

RESUMEN

Mutations in the adenomatous polyposis coli (APC) gene are responsible for not only familial adenomatous polyposis but also many sporadic cancers of the digestive tract. Most mutations found in familial adenomatous polyposis patients are of the truncation type, and the phenotype is affected by the mutation sites in the gene. Truncated APC proteins can associate with the wild-type protein. Accordingly, it has been proposed that the polyposis is caused by a dominant negative mechanism. To test this possibility, we constructed transgenic mice that contained mutant minigenes. They expressed the APC protein truncated either at codon 716 (Apc delta 716) or 1287 (Apc delta 1287) at high levels in the intestinal epithelium. Contrary to our expectation, no intestinal polyps or tumors were found in any of such mice, even after 7 months. These results rule out any dominant negative mechanisms in which the truncated APC protein is directly involved in the formation of intestinal polyps in the mouse.


Asunto(s)
Proteínas del Citoesqueleto/genética , Pólipos Intestinales/genética , Proteína de la Poliposis Adenomatosa del Colon , Secuencia de Aminoácidos , Animales , Genes Dominantes , Genes Supresores de Tumor , Humanos , Pólipos Intestinales/patología , Intestino Delgado/patología , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Mutación , Péptidos/química , Eliminación de Secuencia
15.
Cancer Res ; 59(24): 6113-7, 1999 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-10626800

RESUMEN

The SMAD4 (DPC4) gene was initially isolated as a candidate tumor suppressor from the convergent site of homozygous deletions on 18q in a panel of pancreatic carcinoma cell lines. It encodes a common cytoplasmic signaling molecule shared by the transforming growth factor-beta, activin, and bone morphogenic pathways. We recently inactivated its mouse homologue Smad4 and demonstrated its role in the malignant progression of benign adenomas to invasive adenocarcinomas by analyzing mice with Apc and Smad4 compound mutations. Although simple Smad4 homozygotes were embryonically lethal, the heterozygotes were fertile and appeared normal up to the age of 1 year. Upon further investigation, however, they have developed inflammatory polyps in the glandular stomach and duodenum. By PCR genotyping and immunohistochemical staining, the wild-type Smad4 allele has been lost in the polyp epithelial cells, ie., loss of heterozygosity. On the other hand, we have not found any mutations in such genes as K-Ras, H-Ras, N-Ras, p53, or PTEN. Histologically, the polyps are similar to human juvenile polyps showing moderate stromal cell proliferation and infiltrations by eosinophils and plasma cells. In addition, foci of adenocarcinoma with signet ring cells are also found. These results are consistent with a recent report that germ-line SMAD4 mutations are found in a subset of familial juvenile polyposis.


Asunto(s)
Proteínas de Unión al ADN/genética , Neoplasias Duodenales/genética , Pólipos Intestinales/genética , Pólipos/genética , Neoplasias Gástricas/genética , Transactivadores/genética , Animales , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/metabolismo , Neoplasias Duodenales/patología , Genes Supresores de Tumor , Heterocigoto , Humanos , Pólipos Intestinales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pólipos/patología , Proteína Smad4 , Neoplasias Gástricas/patología , Transactivadores/deficiencia , Transactivadores/metabolismo
16.
Cancer Res ; 59(12): 2965-70, 1999 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-10383162

RESUMEN

To determine the biological role of caudal-like homeobox gene CDX2, we constructed knockout mice in which its mouse homologue Cdx2 was inactivated by homologous recombination, placing a bacterial lacZ gene under the control of the Cdx2 promoter. Although the homozygous mutants died in utero around implantation, the heterozygotes were viable and fertile and expressed lacZ in the caudal region in early embryos and in the gut tissues in adults. The heterozygotes developed cecal and colonic villi by anteriorization and formed hamartomatous polyps in the proximal colon. The hamartoma started to develop at 11.5 days of gestation as an outpocket of the gut epithelium, which ceased to express the remaining Cdx2 allele. The outpocket then expanded as a partially duplicated gut but was contained as a hamartoma after birth. In adult mice, these hamartomas grew very slowly and took a benign course. None of them progressed into invasive adenocarcinomas, even at 1.5 years of age. Whereas the cecal and colonic villi expressed lacZ, the hamartoma epithelium did not, nor did it express Cdx2 mRNA from the wild-type allele. However, genomic DNA analysis of the polyp epithelium did not show a loss of heterozygosity of the Cdx2 gene, suggesting a mechanism of biallelic Cdx2 inactivation other than loss of heterozygosity. These results indicate that the Cdx2 haploin-sufficiency caused cecal and colonic villi, whereas the biallelic inactivation of Cdx2 triggered anomalous duplications of the embryonic gut epithelium, which were contained as hamartomas after birth.


Asunto(s)
Enfermedades del Colon/genética , Hamartoma/genética , Proteínas de Homeodominio/genética , Animales , Factor de Transcripción CDX2 , Enfermedades del Ciego/genética , Enfermedades del Ciego/patología , Vellosidades Coriónicas/patología , Mapeo Cromosómico , Enfermedades del Colon/embriología , Enfermedades del Colon/patología , Epitelio/metabolismo , Hamartoma/embriología , Hamartoma/patología , Proteínas de Homeodominio/metabolismo , Operón Lac , Ratones , Ratones Noqueados , Transactivadores
17.
Cancer Res ; 58(21): 4823-6, 1998 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-9809985

RESUMEN

Nonsteroidal anti-inflammatory drugs reduce the incidence and mortality of colorectal carcinoma. Their chemopreventive effects appear to be due to inhibition of cyclooxygenase (COX)-2. Here, we have studied the relationship between the COX-2 mRNA levels and pathological characteristics in 43 primary colorectal carcinomas. COX-2 levels were significantly higher in tumors with larger sizes and in those with deeper invasions but were not correlated with whether the patients had metastasis or not. These results suggest that larger carcinomas produce more COX-2 to support their own growth and that COX-2 inhibitors may be effective agents of carcinoma growth suppression.


Asunto(s)
Neoplasias Colorrectales/enzimología , Inhibidores de la Ciclooxigenasa/uso terapéutico , Isoenzimas/biosíntesis , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Bases , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/prevención & control , Ciclooxigenasa 1 , Femenino , Humanos , Isoenzimas/genética , Masculino , Proteínas de la Membrana , Persona de Mediana Edad , Datos de Secuencia Molecular , Invasividad Neoplásica , Prostaglandina-Endoperóxido Sintasas/genética
18.
Cancer Res ; 61(4): 1733-40, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11245490

RESUMEN

Mutations in the human adenomatous polyposis (APC) gene are causative for familial adenomatous polyposis (FAP), a rare condition in which numerous colonic polyps arise during puberty and, if left untreated, lead to colon cancer. The APC gene is a tumor suppressor that has been termed the "gatekeeper gene" for colon cancer. In addition to the 100% mutation rate in FAP patients, the APC gene is mutated in >80% of sporadic colon and intestinal cancers. The Apc gene in mice has been mutated either by chemical carcinogenesis, resulting in the Min mouse Apcdelta850, or by heterologous recombination, resulting in the Apcdelta716 or Apedelta1368 mice (M. Oshima et al., Proc. Natl. Acad. Sci. USA, 92: 4482-4486, 1995). Although homozygote Apc-/- mice are embryonically lethal, the heterozygotes are viable but develop numerous intestinal polyps with loss of Apc heterozygosity within the polyps (M. Oshima et al., Proc. Natl. Acad. Sci. USA, 92: 4482-4486, 1995). The proinflammatory, prooncogenic protein cyclooxygenase (COX)-2 has been shown to be markedly induced in the Apcdelta716 polyps at an early stage of polyp development (M. Oshima et al., Cell, 87: 803-809, 1996). We demonstrate here that treatment with the specific COX-2 inhibitor rofecoxib results in a dose-dependent reduction in the number and size of intestinal and colonic polyps in the Apcdelta716 mouse. The plasma concentration of rofecoxib that resulted in a 55% inhibition of polyp number and an 80% inhibition of polyps > 1 mm in size is comparable with the human clinical steady-state concentration of 25 mg rofecoxib (Vioxx) taken once daily (A. Porras et al., Clin. Pharm. Ther., 67: 137, 2000). Polyps from both untreated and rofecoxib- or sulindac-treated Apcdelta716 mice expressed COX-1 and -2, whereas normal epithelium from all mice expressed COX-1 but minimal amounts of COX-2. Polyps from either rofecoxib- or sulindac-treated mice had lower rates of DNA replication, expressed less proangiogenic vascular endothelial-derived growth factor and more membrane-bound beta-catenin, but showed unchanged nuclear localization of this transcription factor. This study showing the inhibition of polyposis in the Apcdelta716 mouse suggests that the specific COX-2 inhibitor rofecoxib (Vioxx) has potential as a chemopreventive agent in human intestinal and colon cancer.


Asunto(s)
Anticarcinógenos/farmacología , Inhibidores de la Ciclooxigenasa/farmacología , Genes APC/genética , Neoplasias Intestinales/prevención & control , Pólipos Intestinales/prevención & control , Isoenzimas/antagonistas & inhibidores , Lactonas/farmacología , Transactivadores , Animales , Anticarcinógenos/farmacocinética , Núcleo Celular/metabolismo , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/farmacocinética , Proteínas del Citoesqueleto/metabolismo , Replicación del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Neoplasias Intestinales/enzimología , Neoplasias Intestinales/genética , Pólipos Intestinales/enzimología , Pólipos Intestinales/genética , Isoenzimas/biosíntesis , Lactonas/farmacocinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Sulfonas , Sulindac/análogos & derivados , Sulindac/farmacocinética , Sulindac/farmacología , beta Catenina
19.
Cell Death Dis ; 7: e2136, 2016 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-26962686

RESUMEN

Recent studies have reported the role of Wnt/ß-catenin signaling in hair cell (HC) development, regeneration, and differentiation in the mouse cochlea; however, the role of Wnt/ß-catenin signaling in HC protection remains unknown. In this study, we took advantage of transgenic mice to specifically knockout or overactivate the canonical Wnt signaling mediator ß-catenin in HCs, which allowed us to investigate the role of Wnt/ß-catenin signaling in protecting HCs against neomycin-induced damage. We first showed that loss of ß-catenin in HCs made them more vulnerable to neomycin-induced injury, while constitutive activation of ß-catenin in HCs reduced HC loss both in vivo and in vitro. We then showed that loss of ß-catenin in HCs increased caspase-mediated apoptosis induced by neomycin injury, while ß-catenin overexpression inhibited caspase-mediated apoptosis. Finally, we demonstrated that loss of ß-catenin in HCs led to increased expression of forkhead box O3 transcription factor (Foxo3) and Bim along with decreased expression of antioxidant enzymes; thus, there were increased levels of reactive oxygen species (ROS) after neomycin treatment that might be responsible for the increased aminoglycoside sensitivity of HCs. In contrast, ß-catenin overexpression reduced Foxo3 and Bim expression and ROS levels, suggesting that ß-catenin is protective against neomycin-induced HC loss. Our findings demonstrate that Wnt/ß-catenin signaling has an important role in protecting HCs against neomycin-induced HC loss and thus might be a new therapeutic target for the prevention of HC death.


Asunto(s)
Células Ciliadas Auditivas/metabolismo , Pérdida Auditiva/prevención & control , Neomicina/efectos adversos , Vía de Señalización Wnt/efectos de los fármacos , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , Muerte Celular , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Células Ciliadas Auditivas/patología , Pérdida Auditiva/inducido químicamente , Pérdida Auditiva/genética , Pérdida Auditiva/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Neomicina/farmacología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Vía de Señalización Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
20.
Oncogene ; 19(33): 3739-49, 2000 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-10949928

RESUMEN

The transcription factor Bach2, a member of the BTB-basic region leucine zipper (bZip) factor family, binds to a 12-O-tetradecanoylphorbol-13-acetate (TPA)-responsive element and the related Maf-recognition element (MARE) by forming homodimers or heterodimers with Maf-related transcription factors. Bach2 regulates transcription by binding to these elements. To understand the function in hematopoiesis, we isolated a cDNA clone for human Bach2 (BACH2) encoding a protein of 841 amino acid residues with a deduced amino acid sequence having 89.5% identity to mouse homolog. Among human hematopoietic cell lines, BACH2 is expressed abundantly only in some B-lymphocytic cell lines. RT-PCR analysis of hematopoietic cells revealed that BACH2 mRNA is expressed in primary B-cells. Enforced expression of BACH2 in a human Burkitt cell line, RAJI that does not express endogenous BACH2, resulted in marked reduction of clonogenic activity, indicating that BACH2 possesses an inhibitory effect on cell proliferation. By fluorescent in situ hybridization, the BACH2 gene was localized to chromosome 6q15. Because deletion of the long arm of chromosome 6 (6q) is one of the commonest chromosomal alterations in human B-cell lymphoma, we examined for the loss of heterozygosity (LOH) of the BACH2 gene in human B-cell non-Hodgkin's lymphomas (NHL). Among 25 informative cases, five (20%) showed LOH. These results indicate that BACH2 plays important roles in regulation of B cell development.


Asunto(s)
Linfocitos B/metabolismo , Cromosomas Humanos Par 6 , Leucina Zippers , Linfoma de Células B/genética , Linfoma no Hodgkin/genética , Factores de Transcripción/genética , Adulto , Secuencia de Aminoácidos , Animales , Linfocitos B/citología , Secuencia de Bases , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , Células Cultivadas , Mapeo Cromosómico , Clonación Molecular , ADN Complementario , Expresión Génica , Frecuencia de los Genes , Humanos , Células K562 , Pérdida de Heterocigocidad , Linfoma de Células B/patología , Linfoma no Hodgkin/patología , Ratones , Datos de Secuencia Molecular , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda