Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 1.055
Filtrar
Más filtros

Publication year range
1.
J Pathol ; 263(4-5): 466-481, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38924548

RESUMEN

The E3 ubiquitin ligase thyroid hormone receptor interacting protein 12 (TRIP12) has been implicated in pancreatic adenocarcinoma (PDAC) through its role in mediating the degradation of pancreas transcription factor 1a (PTF1a). PTF1a is a transcription factor essential for the acinar differentiation state that is notably diminished during the early steps of pancreatic carcinogenesis. Despite these findings, the direct involvement of TRIP12 in the onset of pancreatic cancer has yet to be established. In this study, we demonstrated that TRIP12 protein was significantly upregulated in human pancreatic preneoplastic lesions. Furthermore, we observed that TRIP12 overexpression varied within PDAC samples and PDAC-derived cell lines. We further demonstrated that TRIP12 was required for PDAC-derived cell growth and for the expression of E2F-targeted genes. Acinar-to-ductal cell metaplasia (ADM) is a reversible process that reflects the high plasticity of acinar cells. ADM becomes irreversible in the presence of oncogenic Kras mutations and leads to the formation of preneoplastic lesions. Using two genetically modified mouse models, we showed that a loss of TRIP12 prevented acini from developing ADM in response to pancreatic injury. With two additional mouse models, we further discovered that a depletion of TRIP12 prevented the formation of KrasG12D-induced preneoplastic lesions and impaired metastasis formation in the presence of mutated KrasG12D and Trp53R172H genes. In summary our study identified an overexpression of TRIP12 from the early stages of pancreatic carcinogenesis and proposed this E3 ubiquitin ligase as a novel regulator of acinar plasticity with an important dual role in initiation and metastatic steps of PDAC. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Células Acinares , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Ubiquitina-Proteína Ligasas , Animales , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/enzimología , Humanos , Células Acinares/patología , Células Acinares/metabolismo , Células Acinares/enzimología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/enzimología , Metaplasia/patología , Metaplasia/metabolismo , Plasticidad de la Célula , Carcinogénesis/genética , Carcinogénesis/metabolismo , Ratones , Línea Celular Tumoral , Proliferación Celular , Ratones Noqueados , Regulación Neoplásica de la Expresión Génica , Lesiones Precancerosas/patología , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/enzimología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Transformación Celular Neoplásica/metabolismo , Proteínas Portadoras
2.
Gastroenterology ; 162(3): 813-827.e8, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34767785

RESUMEN

BACKGROUND & AIMS: Because inflammatory bowel disease is increasing worldwide and can lead to colitis-associated carcinoma (CAC), new interventions are needed. We have shown that spermine oxidase (SMOX), which generates spermidine (Spd), regulates colitis. Here we determined whether Spd treatment reduces colitis and carcinogenesis. METHODS: SMOX was quantified in human colitis and associated dysplasia using quantitative reverse-transcription polymerase chain reaction and immunohistochemistry. We used wild-type (WT) and Smox-/- C57BL/6 mice treated with dextran sulfate sodium (DSS) or azoxymethane (AOM)-DSS as models of colitis and CAC, respectively. Mice with epithelial-specific deletion of Apc were used as a model of sporadic colon cancer. Animals were supplemented or not with Spd in the drinking water. Colonic polyamines, inflammation, tumorigenesis, transcriptomes, and microbiomes were assessed. RESULTS: SMOX messenger RNA levels were decreased in human ulcerative colitis tissues and inversely correlated with disease activity, and SMOX protein was reduced in colitis-associated dysplasia. DSS colitis and AOM-DSS-induced dysplasia and tumorigenesis were worsened in Smox-/- vs WT mice and improved in both genotypes with Spd. Tumor development caused by Apc deletion was also reduced by Spd. Smox deletion and AOM-DSS treatment were both strongly associated with increased expression of α-defensins, which was reduced by Spd. A shift in the microbiome, with reduced abundance of Prevotella and increased Proteobacteria and Deferribacteres, occurred in Smox-/- mice and was reversed with Spd. CONCLUSIONS: Loss of SMOX is associated with exacerbated colitis and CAC, increased α-defensin expression, and dysbiosis of the microbiome. Spd supplementation reverses these phenotypes, indicating that it has potential as an adjunctive treatment for colitis and chemopreventive for colon carcinogenesis.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Colitis/genética , Neoplasias del Colon/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Espermidina/uso terapéutico , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Azoximetano , Colitis/inducido químicamente , Colitis/enzimología , Colitis/prevención & control , Colitis Ulcerosa/enzimología , Colitis Ulcerosa/genética , Colon/enzimología , Colon/patología , Neoplasias del Colon/prevención & control , Sulfato de Dextran , Microbioma Gastrointestinal/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Mucosa Intestinal/enzimología , Mucosa Intestinal/patología , Masculino , Ratones , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Lesiones Precancerosas/enzimología , Factores Protectores , ARN Mensajero/metabolismo , Índice de Severidad de la Enfermedad , Espermidina/metabolismo , Espermidina/farmacología , Pérdida de Peso/efectos de los fármacos , alfa-Defensinas/genética , alfa-Defensinas/metabolismo , Poliamino Oxidasa
3.
Gastroenterology ; 161(6): 1907-1923.e26, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34391772

RESUMEN

BACKGROUND & AIMS: Metaplasia and dysplasia in the corpus are reportedly derived from de-differentiation of chief cells. However, the cellular origin of metaplasia and cancer remained uncertain. Therefore, we investigated whether pepsinogen C (PGC) transcript-expressing cells represent the cellular origin of metaplasia and cancer using a novel Pgc-specific CreERT2 recombinase mouse model. METHODS: We generated a Pgc-mCherry-IRES-CreERT2 (Pgc-CreERT2) knock-in mouse model. Pgc-CreERT2/+ and Rosa-EYFP mice were crossed to generate Pgc-CreERT2/Rosa-EYFP (Pgc-CreERT2/YFP) mice. Gastric tissues were collected, followed by lineage-tracing experiments and histologic and immunofluorescence staining. We further established Pgc-CreERT2;KrasG12D/+ mice and investigated whether PGC transcript-expressing cells are responsible for the precancerous state in gastric glands. To investigate cancer development from PGC transcript-expressing cells with activated Kras, inactivated Apc, and Trp53 signaling pathways, we crossed Pgc-CreERT2/+ mice with conditional KrasG12D, Apcflox, Trp53flox mice. RESULTS: Expectedly, mCherry mainly labeled chief cells in the Pgc-CreERT2 mice. However, mCherry was also detected throughout the neck cell and isthmal stem/progenitor regions, albeit at lower levels. In the Pgc-CreERT2;KrasG12D/+ mice, PGC transcript-expressing cells with KrasG12D/+ mutation presented pseudopyloric metaplasia. The early induction of proliferation at the isthmus may reflect the ability of isthmal progenitors to react rapidly to Pgc-driven KrasG12D/+ oncogenic mutation. Furthermore, Pgc-CreERT2;KrasG12D/+;Apcflox/flox mice presented intramucosal dysplasia/carcinoma and Pgc-CreERT2;KrasG12D/+;Apcflox/flox;Trp53flox/flox mice presented invasive and metastatic gastric carcinoma. CONCLUSIONS: The Pgc-CreERT2 knock-in mouse is an invaluable tool to study the effects of successive oncogenic activation in the mouse corpus. Time-course observations can be made regarding the responses of isthmal and chief cells to oncogenic insults. We can observe stomach-specific tumorigenesis from the beginning to metastatic development.


Asunto(s)
Proliferación Celular , Transformación Celular Neoplásica/genética , Células Principales Gástricas/enzimología , Integrasas/genética , Pepsinógeno C/genética , Lesiones Precancerosas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Neoplasias Gástricas/genética , Activación Transcripcional , Animales , Desdiferenciación Celular , Linaje de la Célula , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Principales Gástricas/patología , Regulación Neoplásica de la Expresión Génica , Genes APC , Predisposición Genética a la Enfermedad , Integrasas/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Metaplasia , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Pepsinógeno C/metabolismo , Fenotipo , Lesiones Precancerosas/enzimología , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Neoplasias Gástricas/enzimología , Neoplasias Gástricas/patología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteína Fluorescente Roja
4.
Gastroenterology ; 154(1): 140-153.e17, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28912017

RESUMEN

BACKGROUND & AIMS: Chronic gastrointestinal inflammation increases the risk of cancer by mechanisms that are not well understood. Indoleamine-2,3-dioxygenase 1 (IDO1) is a heme-binding enzyme that regulates the immune response via catabolization and regulation of tryptophan availability for immune cell uptake. IDO1 expression is increased during the transition from chronic inflammation to gastric metaplasia. We investigated whether IDO1 contributes to the inflammatory response that mediates loss of parietal cells leading to metaplasia. METHODS: Chronic gastric inflammation was induced in Ido1-/- and CB57BL/6 (control) mice by gavage with Helicobacter felis or overexpression of interferon gamma in gastric parietal cells. We also performed studies in Jh-/- mice, which are devoid of B cells. Gastric tissues were collected and analyzed by flow cytometry, immunostaining, and real-time quantitative polymerase chain reaction. Plasma samples were analyzed by enzyme-linked immunosorbent assay. Gastric tissues were obtained from 20 patients with gastric metaplasia and 20 patients without gastric metaplasia (controls) and analyzed by real-time quantitative polymerase chain reaction; gastric tissue arrays were analyzed by immunohistochemistry. We collected genetic information on gastric cancers from The Cancer Genome Atlas database. RESULTS: H felis gavage induced significantly lower levels of pseudopyloric metaplasia in Ido1-/- mice, which had lower frequencies of gastric B cells, than in control mice. Blood plasma from H felis-infected control mice had increased levels of autoantibodies against parietal cells, compared to uninfected control mice, but this increase was lower in Ido1-/- mice. Chronically inflamed stomachs of Ido1-/- mice had significantly lower frequencies of natural killer cells in contact with parietal cells, compared with stomachs of control mice. Jh-/- mice had lower levels of pseudopyloric metaplasia than control mice in response to H felis infection. Human gastric pre-neoplasia and carcinoma specimens had increased levels of IDO1 messenger RNA compared with control gastric tissues, and IDO1 protein colocalized with B cells. Co-clustering of IDO1 messenger RNA with B-cell markers was corroborated by The Cancer Genome Atlas database. CONCLUSIONS: IDO1 mediates gastric metaplasia by regulating the B-cell compartment. This process appears to be associated with type II hypersensitivity/autoimmunity. The role of autoimmunity in the progression of pseudopyloric metaplasia warrants further investigation.


Asunto(s)
Gastritis/etiología , Hipersensibilidad/etiología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Lesiones Precancerosas/enzimología , Neoplasias Gástricas/etiología , Animales , Linfocitos B/fisiología , Gastritis/enzimología , Gastritis/patología , Humanos , Hipersensibilidad/enzimología , Hipersensibilidad/patología , Metaplasia , Ratones , Ratones Endogámicos C57BL , Lesiones Precancerosas/patología , Neoplasias Gástricas/enzimología , Neoplasias Gástricas/patología
5.
Dig Dis Sci ; 64(1): 68-75, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30311151

RESUMEN

BACKGROUND: Esophageal squamous cell precursor lesions remain one of the most controversial topics in pathology and clinical management. AIMS: To analyze the dysregulation of human telomerase RNA component (hTERC) in esophageal squamous cell precursor lesions and the clinicopathological correlations with the characteristics of esophageal squamous cell precursor lesions. METHODS: Florescence in situ hybridization was performed to detect hTERC amplification in different gradings of esophageal squamous cell precursor lesions. With retrospective follow-up data, clinicopathological correlations between hTERC and esophageal squamous cell precursor lesions were subjected to logistic regression analysis. RESULTS: hTERC amplification gradually increased with upgrading of dysplasia, reaching the highest level in high-grade intraepithelial neoplasia, and there was a significant difference between the low-grade intraepithelial neoplasia group and the high-grade intraepithelial neoplasia group (P = 0.00). Logistic regression analysis showed that hTERC amplification was correlated with both dysplasia grading and ulcer characteristics of esophageal squamous cell precursor lesions (P < 0.05). CONCLUSIONS: hTERC amplification with increasing grading of esophageal squamous cell precursor lesions and the presence of ulcer characteristics might provide an important molecular and pathological marker for the diagnosis and clinical prognosis of esophageal squamous cell precursor lesions, especially for those ambiguous cases with more divergence in classification.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas de Esófago/genética , Amplificación de Genes , Lesiones Precancerosas/genética , ARN/genética , Telomerasa/genética , Adulto , Anciano , Anciano de 80 o más Años , Biopsia , Neoplasias Esofágicas/enzimología , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/enzimología , Carcinoma de Células Escamosas de Esófago/patología , Femenino , Predisposición Genética a la Enfermedad , Humanos , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Fenotipo , Lesiones Precancerosas/enzimología , Lesiones Precancerosas/patología , Estudios Retrospectivos , Factores de Riesgo
6.
Esophagus ; 15(1): 19-26, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29892805

RESUMEN

BACKGROUND: The relationship between thymidine phosphorylase (TP) and angiogenesis at the early stage of esophageal squamous cell carcinoma has been unclear. METHODS: Using 14 samples of normal squamous epithelium, 11 samples of low-grade intraepithelial neoplasia, and 64 samples of superficial esophageal cancer, microvessel density (MVD) was estimated using immunostaining for CD34 and CD105. TP expression was also evaluated in both cancer cells and stromal monocytic cells (SMCs). We then investigated the correlation between MVD and TP expression in both cancer cells and SMCs. RESULTS: On the basis of the above parameters, MVD was significantly higher in cancerous lesions than in normal squamous epithelium. In terms of CD34 and CD105 expression, MVD showed a gradual increase from normal squamous epithelium, to low-grade intraepithelial neoplasia, and then to M1 and M2 cancer, and M3 or deeper cancer. M1 and M2 cancer showed overexpression of TP in both cancer cells and SMCs. There was no significant correlation between TP expression in cancer cells and MVD estimated from CD34 (rS = 0.16, P = 0.21) or CD105 (rS = 0.05, P = 0.68) expression. Significant correlations were found between TP expression in SMCs and CD34-related (rS = 0.46, P < 0.001) and CD105-related (rS = 0.34, P < 0.01) MVD. In M3 or deeper cancers, there were no significant correlations between TP expression in cancer cells or SMCs and venous invasion, lymphatic invasion, and lymph node metastasis. CONCLUSION: TP expression is activated in both cancer cells and stromal monocytic cells at the very early stage of ESCC progression. TP expression in SMCs, rather than in cancer cells, is significantly correlated with angiogenesis.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Neoplasias Esofágicas/enzimología , Neovascularización Patológica/enzimología , Timidina Fosforilasa/fisiología , Antígenos CD34/metabolismo , Carcinoma de Células Escamosas/irrigación sanguínea , Progresión de la Enfermedad , Endoglina/metabolismo , Epitelio/irrigación sanguínea , Epitelio/enzimología , Neoplasias Esofágicas/irrigación sanguínea , Carcinoma de Células Escamosas de Esófago , Esófago/irrigación sanguínea , Esófago/enzimología , Humanos , Microvasos/patología , Lesiones Precancerosas/enzimología , Células del Estroma/enzimología , Timidina Fosforilasa/metabolismo
7.
Carcinogenesis ; 38(3): 329-335, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28203752

RESUMEN

Endometrial carcinomas are histologically classified as endometrioid, assumed to originate from hyperplastic endometrium, or non-endometrioid carcinomas, assumed to originate from atrophic endometrium. However, both on a histological and a molecular level there are indications that there are more carcinoma types and carcinogenetic pathways. This study aims to analyze endometrial carcinogenesis on a molecular level. The presence of known KRAS, PIK3CA, AKT1, CTNNB1, BRAF, EGFR and NRAS mutations was studied in proliferative, atrophic and hyperplastic endometrium, endometrioid and serous carcinomas, and the endometrium next to these carcinomas, using single molecule Molecular Inversion Probes. Mutations were found in 9 (15%) of the 62 non atypical, and in 6 (18%) of the 34 atypical hyperplasia cases. In comparison, mutations were found in 1 (3%) of the simple, and 8 (30%) of the 27 complex hyperplasia cases. In 12/22 (55%) endometrioid carcinomas, a mutation was found. The KRAS gene was most often mutated in carcinomas next to hyperplastic endometrium, whereas PIK3CA and CTNNB1 mutations were found in endometrioid carcinomas with adjacent atrophic endometrium. Complex hyperplasia rather than atypical hyperplasia appears to be the most important lesion in the carcinogenesis of endometrioid carcinomas, and KRAS, PIK3CA and CTNNB1 mutations appear to play an important role in this process. Carcinogenesis of endometrioid carcinomas next to hyperplasia seems to be different to that of those next to atrophia. The value of these findings in managing endometrial hyperplasia and carcinoma should be studied.


Asunto(s)
Carcinoma Endometrioide/genética , Neoplasias Endometriales/genética , Enfermedades Uterinas/genética , Carcinogénesis/genética , Carcinogénesis/patología , Carcinoma Endometrioide/enzimología , Carcinoma Endometrioide/patología , Fosfatidilinositol 3-Quinasa Clase I , ADN de Neoplasias/genética , Hiperplasia Endometrial/enzimología , Hiperplasia Endometrial/genética , Hiperplasia Endometrial/patología , Neoplasias Endometriales/enzimología , Neoplasias Endometriales/patología , Receptores ErbB/genética , Femenino , Humanos , Mutación , Fosfatidilinositol 3-Quinasas/genética , Lesiones Precancerosas/enzimología , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Enfermedades Uterinas/enzimología , Enfermedades Uterinas/patología
8.
Toxicol Mech Methods ; 27(2): 136-150, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28004603

RESUMEN

Asiatic acid (AA), a pentacyclic triterpenoid, derived from the tropical medicinal plant Centella asiatica is known to exhibit numerous pharmacological properties. We hypothesized that AA will have chemopreventive potential against 1,2-dimethylhydrazine (DMH)-induced experimental colon carcinogenesis in male Wistar rats. Rats were arbitrarily divided into six groups. Group I rats were processed as control. Group II rats received AA (8 mg/kg b.w., p.o.) and groups III-VI rats received subcutaneous injections of DMH (20 mg/kg b.w.) once a week, for the first four weeks. In addition, groups IV-VI rats received AA at the doses of 2, 4 and 8 mg/kg b.w., respectively, for 16 weeks. Our results discovered that supplementation with AA to the DMH-exposed rats significantly decreased the incidence of polyps and Aberrant crypt foci (ACF) as compared to the DMH-alone-exposed rats. Moreover, in the AA-supplemented DMH-exposed rats, we ascertained increased activities of the antioxidants and decreased levels of lipid peroxidation (LPO) in the liver and circulation and enhanced levels of both LPO and antioxidants in the colon, which were altered in the DMH-alone-exposed rats. Furthermore, we also observed altered activities of vitamins C and E and biotransforming enzymes in DMH-alone-exposed rats, which were reversed on AA supplementation. All the observations were supported by our histological findings. Thus, we can conclude that, AA could be used as an effective chemopreventive agent against DMH-induced colon carcinogenesis.


Asunto(s)
Anticarcinógenos/uso terapéutico , Colon/efectos de los fármacos , Neoplasias del Colon/prevención & control , Estrés Oxidativo/efectos de los fármacos , Triterpenos Pentacíclicos/uso terapéutico , Lesiones Precancerosas/prevención & control , 1,2-Dimetilhidrazina/farmacocinética , Animales , Anticarcinógenos/administración & dosificación , Ácido Ascórbico/metabolismo , Biotransformación , Catalasa/metabolismo , Colon/enzimología , Colon/patología , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Relación Dosis-Respuesta a Droga , Masculino , Triterpenos Pentacíclicos/administración & dosificación , Lesiones Precancerosas/inducido químicamente , Lesiones Precancerosas/enzimología , Lesiones Precancerosas/patología , Ratas Wistar , Superóxido Dismutasa/metabolismo , Vitamina E/metabolismo
9.
J Contemp Dent Pract ; 18(10): 943-946, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28989135

RESUMEN

OBJECTIVE: The purpose of the study is to highlight the use of gamma-glutamyl transpeptidase (GGT) as salivary enzyme tumor marker and assess the activity and concentration of GGT in precancerous and cancer patients and compare it with the normal control. MATERIALS AND METHODS: Individuals in the age group of above 20 years were included in the study. In this study, salivary GGT was analyzed in 75 cases. The selected patients were divided into three main groups as group I (controls with normal health), group II (patients with precancerous lesions and conditions), and group III (patients with oral cancer lesions). All the selected individuals were analyzed for salivary GGT. RESULTS: A significant difference was observed between control and precancerous groups with GGT values at 5% level of significance. The mean GGT value is significantly higher in precancerous group as compared with control group (p < 0.05). A significant difference was observed between control and cancerous groups with GGT values at 5% level of significance. The mean GGT value is significantly higher in cancerous group as compared with control group (p < 0.05). A significant difference was observed between precancerous and cancerous groups with GGT values at 5% level of significance. The mean GGT value is significantly higher in cancerous group as compared with precancerous group (p < 0.05). CONCLUSION: There is a remarkable increase in salivary GGT activity in both precancerous and cancerous conditions. The increased activity was more marked in cancerous conditions than in precancerous conditions. The GGT levels were two- to threefold increased in precancerous conditions as compared with control group. This finding was statistically significant and also suggested the strong correlation between GGT levels and presence of precancerous conditions. CLINICAL SIGNIFICANCE: Integration of all these observation strengthens our thinking that elevation of salivary GTT in pre-cancerous and cancerous conditions can be a reliable biomarker in early detection and prevention of oral malignancy.


Asunto(s)
Neoplasias/enzimología , Lesiones Precancerosas/enzimología , gamma-Glutamiltransferasa/metabolismo , Biomarcadores de Tumor/análisis , Estudios de Casos y Controles , Humanos , Saliva/enzimología , gamma-Glutamiltransferasa/análisis
10.
Toxicol Pathol ; 44(8): 1146-1159, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-28245158

RESUMEN

The medium-term multiorgan initiation-promotion chemical bioassay (diethylnitrosamine, methyl-nitrosourea, butyl-hydroxybutylnitrosamine, dihydroxypropylnitrosamine, dimethylhydrazine [DMBDD]) with the Fischer 344 rat was proposed as an alternative to the conventional 2-year carcinogenesis bioassay for regulatory purposes. The acronym DMBDD stands for the names of five genotoxic agents used for initiation of multiorgan carcinogenesis. The Brazilian Agency for the Environment officially recognized a variation of this assay (DMBDDb) as a valid method to assess the carcinogenic potential of agrochemicals. Different from the original protocol, this DMBDDb is 30-week long, uses Wistar rats and two positive control groups exposed to carcinogenesis promoters sodium phenobarbital (PB) or 2-acetylaminofluorene (2-AAF). This report presents the experience of an academic laboratory with the DMBDDb assay and contributes to the establishment of this alternative DMBDD bioassay in a different rat strain. Frequent lesions observed in positive groups to evaluate the promoting potential of pesticides and the immunohistochemical expressions of liver cytochrome P450 (CYP) 2B1/2B2 and CYP1A2 enzymes were assessed. Commonly affected organs were liver, kidney, intestines, urinary bladder, and thyroid. PB promoting activity was less evident than that of 2-AAF, especially in males. This study provides a repository of characteristic lesions occurring in positive control animals submitted to a modified alternative 2-stage multiorgan protocol for carcinogenesis in Wistar rat.


Asunto(s)
2-Acetilaminofluoreno/toxicidad , Pruebas de Carcinogenicidad/métodos , Carcinógenos/toxicidad , Neoplasias Experimentales/inducido químicamente , Fenobarbital/toxicidad , Lesiones Precancerosas/inducido químicamente , Animales , Hidrocarburo de Aril Hidroxilasas/biosíntesis , Bioensayo , Citocromo P-450 CYP1A2/biosíntesis , Citocromo P-450 CYP2B1/biosíntesis , Femenino , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Masculino , Neoplasias Experimentales/enzimología , Tamaño de los Órganos/efectos de los fármacos , Especificidad de Órganos , Lesiones Precancerosas/enzimología , Ratas Wistar , Esteroide Hidroxilasas/biosíntesis
11.
J Oral Pathol Med ; 45(10): 746-752, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26991907

RESUMEN

BACKGROUND: PI3K-AKT-mTOR signaling pathway is associated with several cellular functions and is frequently changed in several malignancies. The aim of this study was to characterize the immunohistochemical expression pattern of components in PI3K-AKT-mTOR signaling pathway in oral epithelial dysplasia (OED), comparing to oral squamous cell carcinoma (OSCC) and non-dysplastic oral tissues (NDOT). METHODS: A total of 186 cases of NDOT, OED and OSCC were retrieved. Nuclear staining and cytoplasmic staining of the keratinocytes were considered positive, and the percentage of positive cells was calculated. RESULTS: Increased immunoreactivity from NDOT to OED and OSCC was seen for all proteins. In NDOT cases, positivity was found only for pS6 (52.9%) and p4EBP1 (13.5%). In OED, immunoreactivity was observed for pAKT (62.2%), pmTOR (28.6%), pS6 (70.8%), and p4EBP1 (42.9%). In OSCC cases, immunoreactivity was found for pAKT (83.3%), pmTOR (50%), pS6 (77.4%), and p4EBP1 (50%). The pAKT and pmTOR expression was higher in OED (<0.001, Fisher's exact test) and OSCC (<0.001, Fisher's exact test). CONCLUSION: Our study demonstrated higher pAKT and pmTOR expression during carcinogenesis of oral mucosa, differing considerably among OED and OSCC specimens when compared to NDOT. These proteins can be considered potential diagnostic markers for early detection of cancer.


Asunto(s)
Carcinogénesis/metabolismo , Carcinoma de Células Escamosas/enzimología , Neoplasias de Cabeza y Cuello/enzimología , Neoplasias de la Boca/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Quinasas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Biomarcadores de Tumor/metabolismo , Biopsia , Carcinogénesis/patología , Carcinoma in Situ/enzimología , Carcinoma in Situ/metabolismo , Carcinoma in Situ/patología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Citoplasma/enzimología , Citoplasma/metabolismo , Citoplasma/patología , Femenino , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/metabolismo , Fosfatidilinositol 3-Quinasas/biosíntesis , Lesiones Precancerosas/enzimología , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Proteínas Quinasas/biosíntesis , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello , Serina-Treonina Quinasas TOR/biosíntesis
12.
Mol Cell ; 31(1): 21-32, 2008 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-18614044

RESUMEN

The Mre11 complex (Mre11, Rad50, and Nbs1) and Chk2 have been implicated in the DNA-damage response, an inducible process required for the suppression of malignancy. The Mre11 complex is predominantly required for repair and checkpoint activation in S phase, whereas Chk2 governs apoptosis. We examined the relationship between the Mre11 complex and Chk2 in the DNA-damage response via the establishment of Nbs1(DeltaB/DeltaB) Chk2(-/-) and Mre11(ATLD1/ATLD1) Chk2(-/-) mice. Chk2 deficiency did not modify the checkpoint defects or chromosomal instability of Mre11 complex mutants; however, the double-mutant mice exhibited synergistic defects in DNA-damage-induced p53 regulation and apoptosis. Nbs1(DeltaB/DeltaB) Chk2(-/-) and Mre11(ATLD1/ATLD1) Chk2(-/-) mice were also predisposed to tumors. In contrast, DNA-PKcs-deficient mice, in which G1-specific chromosome breaks are present, did not exhibit synergy with Chk2(-/-) mutants. These data suggest that Chk2 suppresses the oncogenic potential of DNA damage arising during S and G2 phases of the cell cycle.


Asunto(s)
Daño del ADN , Replicación del ADN , Lesiones Precancerosas/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Alelos , Animales , Apoptosis , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Quinasa de Punto de Control 2 , Inestabilidad Cromosómica , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Reparación del ADN , Enzimas Reparadoras del ADN/metabolismo , ADN Complementario/genética , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Exones/genética , Genoma/genética , Proteína Homóloga de MRE11 , Ratones , Mutación/genética , Proteínas Nucleares/metabolismo , Lesiones Precancerosas/patología , Proteínas Serina-Treonina Quinasas/deficiencia , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo
13.
Med Sci Monit ; 22: 4794-4802, 2016 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-27926694

RESUMEN

BACKGROUND Activated Cdc42 kinase1 (ACK1) is a non-receptor tyrosine kinase which is critical for cell survival, proliferation, and migration. Genomic amplification of ACK1 has been reported in multiple human cancers. We aimed to investigate ACK1 protein expression in colorectal mucosa with inflammation and neoplasm, and to evaluate its correlation with disease activity and severity. MATERIAL AND METHODS A total of 250 individuals who underwent total colonoscopy were collected randomly from January 2007 to May 2013 in Nanfang Hospital, Guangzhou, China. Colorectal mucosal biopsy specimens were obtained by endoscopy from 78 patients with ulcerative colitis (UC), 22 with Crohn's disease (CD), 20 with infectious colitis, 26 with non-IBD and noninfectious colitis, 16 with sporadic adenomas, 4 with dysplasia-associated lesions or masses, 10 with sporadic colorectal cancer (CRC), 4 with UC-related CRC, 10 with hyperplastic polyps, and 60 without colonic abnormalities. ACK1 protein levels were determined immunohistochemically. The correlations of ACK1 expression with disease activity and severity were also evaluated. RESULTS Significantly increased ACK1 expression was observed in epithelial cells of colorectal mucosa with inflammation and dysplasia compared to controls (P<0.05). ACK1 expression correlated with clinical activity in IBD (χ²=4.57, P=0.033 for UC; χ²=5.68, P=0.017 for CD), as well as grade of dysplasia in preneoplastic lesions (P<0.05). No significant differences in ACK1 expression were found between UC and CD, or between IBD and non-IBD conditions (P>0.05). CONCLUSIONS ACK1 protein is increased extensively in colitis and colorectal dysplasia. ACK1 overexpression may play a role in colorectal inflammation and neoplasms.


Asunto(s)
Colitis/enzimología , Neoplasias Colorrectales/enzimología , Proteínas Tirosina Quinasas/metabolismo , Adenoma/enzimología , Adenoma/genética , Adenoma/patología , Adulto , Biopsia , Colitis/genética , Colitis/patología , Colitis Ulcerosa/complicaciones , Colonoscopía , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Enfermedad de Crohn/patología , Activación Enzimática , Femenino , Humanos , Enfermedades Inflamatorias del Intestino/enzimología , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/patología , Masculino , Persona de Mediana Edad , Lesiones Precancerosas/enzimología , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Proteínas Tirosina Quinasas/genética
14.
Carcinogenesis ; 36(12): 1572-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26449252

RESUMEN

To investigate the role of cyclooxygenase (COX)-2/prostaglandin E2 (PGE2) in the process of Helicobacter pylori-induced gastric carcinogenesis, a prospective study based on an intervention trial was conducted in Linqu County, China. A total of 1401 subjects with histopathologic diagnosis were investigated at baseline, among those, 919 completed subsequent interventions (anti-H.pylori and/or celecoxib treatment). Expressions of COX-2 and Ki-67 were assessed by immunohistochemistry, and PGE2 levels were measured by enzyme immunoassay before and after interventions, respectively. We found a grade-response relationship between COX-2 expression level and risk of advanced gastric lesions at baseline. Stratified analysis indicated an additive interaction between COX-2 expression and H.pylori infection on the elevated risk of advanced gastric lesions. The odds ratios (ORs) for both factors combined were 9.31 [95% confidence interval (CI): 4.13-20.95] for chronic atrophic gastritis, 16.26 (95% CI: 7.29-36.24) for intestinal metaplasia and 21.13 (95% CI: 7.87-56.75) for dysplasia, respectively. After interventions, COX-2 expression and Ki-67 labeling index (LI) were decreased in anti-H.pylori group (OR: 1.65, 95% CI: 1.36-1.99 for COX-2; OR: 1.78, 95% CI: 1.49-2.12 for Ki-67) or anti-H.pylori followed by celecoxib group (OR: 1.41, 95% CI: 1.17-1.70 for COX-2; OR: 1.63, 95% CI: 1.37-1.94 for Ki-67). PGE2 levels were decreased in all treatment groups. Furthermore, the regression of gastric lesions was associated with the decrease of COX-2 expression or Ki-67 LI after interventions. Our findings indicate that H.pylori-induced COX-2/PGE2 pathways play an important role on the progression of precancerous gastric lesions in a Chinese population.


Asunto(s)
Celecoxib/uso terapéutico , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Ciclooxigenasa 2/metabolismo , Infecciones por Helicobacter/tratamiento farmacológico , Lesiones Precancerosas/tratamiento farmacológico , Neoplasias Gástricas/prevención & control , Adulto , Amoxicilina/uso terapéutico , Antibacterianos/uso terapéutico , China , Claritromicina/uso terapéutico , Dinoprostona/metabolismo , Método Doble Ciego , Femenino , Infecciones por Helicobacter/enzimología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/inmunología , Humanos , Masculino , Persona de Mediana Edad , Omeprazol/uso terapéutico , Lesiones Precancerosas/enzimología , Lesiones Precancerosas/microbiología , Estómago/enzimología , Estómago/inmunología , Estómago/microbiología , Neoplasias Gástricas/enzimología , Neoplasias Gástricas/microbiología
15.
Gastroenterology ; 146(7): 1739-51.e14, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24530706

RESUMEN

BACKGROUND & AIMS: The gastric cancer-causing pathogen Helicobacter pylori up-regulates spermine oxidase (SMOX) in gastric epithelial cells, causing oxidative stress-induced apoptosis and DNA damage. A subpopulation of SMOX(high) cells are resistant to apoptosis, despite their high levels of DNA damage. Because epidermal growth factor receptor (EGFR) activation can regulate apoptosis, we determined its role in SMOX-mediated effects. METHODS: SMOX, apoptosis, and DNA damage were measured in gastric epithelial cells from H. pylori-infected Egfr(wa5) mice (which have attenuated EGFR activity), Egfr wild-type mice, or in infected cells incubated with EGFR inhibitors or deficient in EGFR. A phosphoproteomic analysis was performed. Two independent tissue microarrays containing each stage of disease, from gastritis to carcinoma, and gastric biopsy specimens from Colombian and Honduran cohorts were analyzed by immunohistochemistry. RESULTS: SMOX expression and DNA damage were decreased, and apoptosis increased in H. pylori-infected Egfr(wa5) mice. H. pylori-infected cells with deletion or inhibition of EGFR had reduced levels of SMOX, DNA damage, and DNA damage(high) apoptosis(low) cells. Phosphoproteomic analysis showed increased EGFR and erythroblastic leukemia-associated viral oncogene B (ERBB)2 signaling. Immunoblot analysis showed the presence of a phosphorylated (p)EGFR-ERBB2 heterodimer and pERBB2; knockdown of ErbB2 facilitated apoptosis of DNA damage(high) apoptosis(low) cells. SMOX was increased in all stages of gastric disease, peaking in tissues with intestinal metaplasia, whereas pEGFR, pEGFR-ERBB2, and pERBB2 were increased predominantly in tissues showing gastritis or atrophic gastritis. Principal component analysis separated gastritis tissues from patients with cancer vs those without cancer. pEGFR, pEGFR-ERBB2, pERBB2, and SMOX were increased in gastric samples from patients whose disease progressed to intestinal metaplasia or dysplasia, compared with patients whose disease did not progress. CONCLUSIONS: In an analysis of gastric tissues from mice and patients, we identified a molecular signature (based on levels of pEGFR, pERBB2, and SMOX) for the initiation of gastric carcinogenesis.


Asunto(s)
Daño del ADN , Células Epiteliales/enzimología , Receptores ErbB/metabolismo , Mucosa Gástrica/enzimología , Infecciones por Helicobacter/enzimología , Helicobacter pylori/metabolismo , Receptor ErbB-2/metabolismo , Animales , Apoptosis , Línea Celular , Supervivencia Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Técnicas de Cocultivo , Colombia , Progresión de la Enfermedad , Activación Enzimática , Células Epiteliales/microbiología , Células Epiteliales/patología , Receptores ErbB/deficiencia , Receptores ErbB/genética , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Gastritis/enzimología , Gastritis/microbiología , Gastritis/patología , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/patología , Helicobacter pylori/patogenicidad , Honduras , Humanos , Metaplasia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Fosforilación , Lesiones Precancerosas/enzimología , Lesiones Precancerosas/microbiología , Lesiones Precancerosas/patología , Análisis de Componente Principal , Multimerización de Proteína , Receptor ErbB-2/genética , Transducción de Señal , Neoplasias Gástricas/enzimología , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología , Tennessee , Poliamino Oxidasa
16.
Clin Sci (Lond) ; 128(12): 863-75, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25818486

RESUMEN

The mechanism by which reactive oxygen species (ROS) are produced by tumour cells remained incompletely understood until the discovery over the last 15 years of the family of NADPH oxidases (NOXs 1-5 and dual oxidases DUOX1/2) which are structural homologues of gp91phox, the major membrane-bound component of the respiratory burst oxidase of leucocytes. Knowledge of the roles of the NOX isoforms in cancer is rapidly expanding. Recent evidence suggests that both NOX1 and DUOX2 species produce ROS in the gastrointestinal tract as a result of chronic inflammatory stress; cytokine induction (by interferon-γ, tumour necrosis factor α, and interleukins IL-4 and IL-13) of NOX1 and DUOX2 may contribute to the development of colorectal and pancreatic carcinomas in patients with inflammatory bowel disease and chronic pancreatitis, respectively. NOX4 expression is increased in pre-malignant fibrotic states which may lead to carcinomas of the lung and liver. NOX5 is highly expressed in malignant melanomas, prostate cancer and Barrett's oesophagus-associated adenocarcinomas, and in the last it is related to chronic gastro-oesophageal reflux and inflammation. Over-expression of functional NOX proteins in many tissues helps to explain tissue injury and DNA damage from ROS that accompany pre-malignant conditions, as well as elucidating the potential mechanisms of NOX-related damage that contribute to both the initiation and the progression of a wide range of solid and haematopoietic malignancies.


Asunto(s)
NADPH Oxidasas/metabolismo , Neoplasias/enzimología , Neoplasias Hematológicas/enzimología , Humanos , NADPH Oxidasas/genética , NADPH Oxidasas/fisiología , Lesiones Precancerosas/enzimología , Especies Reactivas de Oxígeno/metabolismo , Células Tumorales Cultivadas
17.
Gynecol Oncol ; 136(3): 549-53, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25451692

RESUMEN

OBJECTIVE: We previously found that Dual-specificity phosphatase 6 (Dusp6) over-expression enhanced the growth-promoting effect of estrogen in endometrial adenocarcinoma cells. The aim of this study was to explore the correlation of Dusp6 expression with progestin sensitivity in atypical endometrial hyperplasia (AEH) and earlier endometrial carcinomas (EC). METHODS: Using immunohistochemistry study, we analyzed the expression of Dusp6 protein in AEH. RESULTS: We found that progestin treatment was effective in 89% of AEH and 50% of EC. Before treatment, Dusp6 expression was significantly higher in progestin-sensitive AEH groups compared with progestin-resistant groups. After treatment, Dusp6 expression was significantly upregulated in progestin-sensitive groups, but not in progestin-resistant groups. Moreover, a high-dose of Dusp6 transfection significantly enhanced progestin-induced growth-inhibition in Ishikawa cells. CONCLUSIONS: Dusp6 could be a predicting marker for deciding the effectiveness of progestin therapy in AEH.


Asunto(s)
Antineoplásicos Hormonales/uso terapéutico , Fosfatasa 6 de Especificidad Dual/metabolismo , Hiperplasia Endometrial/tratamiento farmacológico , Neoplasias Endometriales/tratamiento farmacológico , Acetato de Medroxiprogesterona/uso terapéutico , Lesiones Precancerosas/tratamiento farmacológico , Progestinas/uso terapéutico , Biomarcadores/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos , Hiperplasia Endometrial/enzimología , Neoplasias Endometriales/enzimología , Femenino , Humanos , Inmunohistoquímica , Lesiones Precancerosas/enzimología , Resultado del Tratamiento , Regulación hacia Arriba
18.
J Biol Regul Homeost Agents ; 28(4): 767-73, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25620185

RESUMEN

Nitric Oxide (NO) has been linked to several cardiovascular, neurological and immunological physiological and pathological functions. Several studies have shown that the eNOS, nNOS and iNOS effects on cancer cell growth and proliferation are related to the upregulation of the Wnt pathway and have a central role during metastasis development. Recent studies suggest that cancer cells undergo metabolic reprogramming, which drives cancer cell growth and progression. The aim of this study was to observe the NOS activity in the pathogenesis of oral precancerous and cancerous lesions. The results showed changes in eNOS activity levels, which increased from healthy oral mucosa to oral squamous cell carcinoma SCC, through different dysplasia levels. The iNOS activity levels increased in precancerous lesions compared to healthy mucosa, where iNOS was absent, while it decreased in SCC lesions. Moreover, a gradual increase of nNOS activity together with the progression of the lesions was also found. These results may suggest how NO could play a critical role during pathogenesis, growth and development of precancerous lesions to cancer degeneration.


Asunto(s)
Neoplasias de la Boca/enzimología , Óxido Nítrico Sintasa/metabolismo , Lesiones Precancerosas/enzimología , Adulto , Anciano , Carcinoma de Células Escamosas/enzimología , Femenino , Humanos , Leucoplasia Bucal/enzimología , Masculino , Persona de Mediana Edad , Mucosa Bucal/enzimología , Neoplasias de la Boca/etiología , Óxido Nítrico/fisiología , Lesiones Precancerosas/etiología
19.
Gut ; 62(8): 1179-86, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22698650

RESUMEN

OBJECTIVES: Screening colonoscopy to monitor for early colitis-associated colon cancer (CAC) is difficult due to the aberrant mucosal patterns associated with long-standing colitis. The aim of this study was to develop a rapid fluorescent detection method for use during colonoscopy for improving the detection of CAC utilising a topically applied enzymatically activatable probe (gGlu-HMRG) which fluoresces in the presence of γ-glutamyltranspeptidase (GGT), an enzyme associated with cancer. METHODS: Expression of GGT in colon cell lines was examined with fluorescence microscopy and flow cytometry. A mouse model (azoxymethane/dextran sulphate sodium) of CAC was used and mice were examined with white light and fluorescence colonoscopy before and after topical gGlu-HMRG administration. RESULTS: Expression of GGT, although variable, was higher in human colon cancer cells than normal human colon cells. Using fluorescence colonoscopy in mice, gGlu-HMRG fluorescent lesions were detected 5 min after topical administration and fluorescence persisted for at least 30 min. Fluorescence guided biopsy revealed all fluorescent lesions that contained cancer or dysplasia (n=16), whereas three out of 12 non-fluorescent lesions contained low grade dysplasia and others did not contain neoplastic histology. Microscopic inflammatory infiltration also had variable fluorescence but in general was much lower (∼10-fold) in signal than cancer. Repeat fluorescence endoscopy allowed individual tumours to be monitored. CONCLUSION: These results suggest that gGlu-HMRG can improve endoscopic detection of CAC with a higher target to background ratio than conventional white light colonoscopy. This could be of benefit to patients with long-standing colitis who must undergo repeated screening colonoscopies.


Asunto(s)
Colitis/complicaciones , Neoplasias del Colon/diagnóstico , Neoplasias del Colon/etiología , Adenocarcinoma/diagnóstico , Adenocarcinoma/enzimología , Adenocarcinoma/etiología , Administración Tópica , Animales , Biomarcadores de Tumor/metabolismo , Biopsia , Colon/enzimología , Neoplasias del Colon/enzimología , Colonoscopía/métodos , Modelos Animales de Enfermedad , Detección Precoz del Cáncer/métodos , Colorantes Fluorescentes/administración & dosificación , Humanos , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente/métodos , Lesiones Precancerosas/diagnóstico , Lesiones Precancerosas/enzimología , Lesiones Precancerosas/etiología , Células Tumorales Cultivadas , gamma-Glutamiltransferasa/metabolismo
20.
Am J Pathol ; 180(6): 2462-78, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22546478

RESUMEN

Lesions displaying a variety of dysplastic changes precede invasive oral and epidermal squamous cell carcinoma (SCC); however, there are no histopathological criteria for either confirming or staging premalignancy. SCCs and dysplasias frequently contain cells that abnormally express the γ2 subunit of laminin-332. We developed cell culture models to investigate γ2 dysregulation. Normal human keratinocytes displayed density-dependent repression of γ2, whereas premalignant keratinocytes and SCC cells overexpressed γ2 and secreted laminin assembly intermediates. Neoplastic cells had hyperactive EGFR/MAPK(ERK) signaling coordinate with overexpressed γ2, and EGFR and MEK inhibitors normalized γ2 expression. Keratinocytes engineered to express HPV16 E6 or activated mutant HRAS, cRAF1, or MEK1 lost density repression of γ2 and shared with neoplastic cells signaling abnormalities downstream of ERK, including increased phosphorylation of S6 and eIF4 translation factors. Notably, qPCR results revealed that γ2 overexpression was not accompanied by increased γ2 mRNA levels, consistent with ERK-dependent, eIF4B-mediated translation initiation of the stem-looped, 5'-untranslated region of γ2 mRNA in neoplastic cells. Inhibitors of MEK, but not of TORC1/2, blocked S6 and eIF4B phosphorylation and γ2 overexpression. Immunostaining of oral dysplasias identified γ2 overexpression occurring within fields of basal cells that had elevated p-S6 levels. These results reveal a causal relationship between ERK-dependent translation factor activation and laminin γ2 dysregulation and identify new markers of preinvasive neoplastic change during progression to SCC.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Laminina/biosíntesis , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Neoplasias de la Boca/metabolismo , Lesiones Precancerosas/metabolismo , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/patología , Progresión de la Enfermedad , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Queratinocitos/metabolismo , Laminina/genética , Sistema de Señalización de MAP Quinasas/fisiología , Neoplasias de la Boca/enzimología , Neoplasias de la Boca/patología , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Lesiones Precancerosas/enzimología , Lesiones Precancerosas/patología , Modificación Traduccional de las Proteínas/fisiología , Transducción de Señal/fisiología , Células Tumorales Cultivadas , Quinasas raf/fisiología , Proteínas ras/fisiología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda