Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 13 de 13
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Cancer Rep (Hoboken) ; 5(2): e1459, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34245130

RÉSUMÉ

BACKGROUND: Data are steadily accruing that demonstrate that intestinal tumors are frequently derived from multiple founding cells, resulting in tumors comprised of distinct ancestral clones that might cooperate or alternatively compete, thereby potentially impacting different phases of the disease process. AIM: We sought to determine whether tumors with a multi-ancestral architecture involving at least two distinct clones show increased tumor number, growth, progression, or resistance to drug intervention. METHODS: Mice carrying the Min allele of Apc were generated that were mosaic with only a subset of cells in the intestinal epithelium expressing an activated form of PI3K, a key regulatory kinase affecting several important cellular processes. These cells were identifiable as they fluoresced green, whereas all other cells fluoresced red. RESULTS: Cell lineage tracing revealed that many intestinal tumors from our mouse model were derived from at least two founding cells, those expressing the activated PI3K (green) and those which did not (red). Heterotypic tumors with a multi-ancestral architecture as evidenced by a mixture of green and red cells exhibited increased tumor growth and invasiveness. Clonal architecture also had an impact on tumor response to low-dose aspirin. Aspirin treatment resulted in a greater reduction of heterotypic tumors derived from multiple founding cells as compared to tumors derived from a single founding cell. CONCLUSION: These data indicate that genetically distinct tumor-founding cells can contribute to early intratumoral heterogeneity. The coevolution of the founding cells and their progeny enhances colon tumor progression and impacts the response to aspirin. These findings are important to a more complete understanding of tumorigenesis with consequences for several distinct models of tumor evolution. They also have practical implications to the clinic. Mouse models with heterogenous tumors are likely better for predicting drug efficacy as compared to models in which the tumors are highly homogeneous. Moreover, understanding how interactions among different populations in a single heterotypic tumor with a multi-ancestral architecture impact response to a single agent and combination therapies are necessary to fully develop personalized medicine.


Sujet(s)
Transformation cellulaire néoplasique/génétique , Tumeurs de l'intestin/génétique , Animaux , Antinéoplasiques/pharmacologie , Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Transformation cellulaire néoplasique/anatomopathologie , Modèles animaux de maladie humaine , Évolution de la maladie , Résistance aux médicaments antinéoplasiques/génétique , Tumeurs de l'intestin/traitement médicamenteux , Tumeurs de l'intestin/anatomopathologie , Souris , Souris transgéniques
2.
Int J Radiat Biol ; 97(8): 1140-1151, 2021.
Article de Anglais | MEDLINE | ID: mdl-33720813

RÉSUMÉ

PURPOSE: Estimating cancer risk associated with interplanetary space travel is complicated. Human exposure data to high atomic number, high-energy (HZE) radiation is lacking, so data from low linear energy transfer (low-LET) γ-ray radiation is used in risk models, with the assumption that HZE and γ-ray radiation have comparable biological effects. This assumption has been challenged by reports indicating that HZE radiation might produce more aggressive tumors. The goal of this research is to test whether high-LET HZE radiation induced tumors are more aggressive. MATERIALS AND METHODS: Murine models of mammary and liver cancer were used to compare the impact of exposure to 0.2Gy of 300MeV/n silicon ions, 3 Gy of γ-rays or no radiation. Numerous measures of tumor aggressiveness were assessed. RESULTS: For the mammary cancer models, there was no significant change in the tumor latency or metastasis in silicon-irradiated mice compared to controls. For the liver cancer models, we observed an increase in tumor incidence but not tumor aggressiveness in irradiated mice. CONCLUSION: Tumors in the HZE-irradiated mice were not more aggressive than those arising from exposure to low-LET γ-rays or spontaneously. Thus, enhanced aggressiveness does not appear to be a uniform characteristic of all tumors in HZE-irradiated animals.


Sujet(s)
Carcinome hépatocellulaire/anatomopathologie , Tumeurs du foie/anatomopathologie , Tumeurs expérimentales de la mamelle/anatomopathologie , Animaux , Relation dose-effet des rayonnements , Femelle , Humains , Transfert linéique d'énergie , Souris
3.
Physiol Rep ; 8(5): e14391, 2020 03.
Article de Anglais | MEDLINE | ID: mdl-32170841

RÉSUMÉ

Cancer cachexia is the loss of lean muscle mass with or without loss of fat mass that is often highlighted by a progressive loss of skeletal muscle mass and function. The mechanisms behind the cachexia-related loss of skeletal muscle are poorly understood, including cachexia-related muscle functional impairments. Existing models have revealed some potential mechanisms, but appear limited to how the cancer develops and the type of tumors that form. We studied the C57BL6/J (B6) ApcMin/+ Tg::Fabp1-Cre TG::PIK3ca* (CANCER) mouse. In this model, mice develop highly aggressive intestinal cancers. We tested whether CANCER mice develop cancer cachexia, if muscle function is altered and if sex differences are present. Both female and male mice, B6 (CONTROL) and CANCER mice, were analyzed to determine body weight, hindlimb muscle mass, protein concentration, specific force, and fatigability. Female CANCER mice had reduced body weight and hindlimb muscle mass compared with female CONTROL mice, but lacked changes in protein concentration and specific force. Male CANCER mice had reduced protein concentration and reduced specific force, but lacked altered body weight and muscle mass. There were no changes in fatigability in either group. Our study demonstrates that CANCER mice present an early stage of cachexia, have reduced specific force in male CANCER mice and develop a sex-dependent cachexia phenotype. However, CANCER mice lack certain aspects of the syndrome seen in the human scenario and, therefore, using the CANCER mice as a preclinical model does not seem sufficient in order to maximize the translation of preclinical findings to humans.


Sujet(s)
Cachexie/anatomopathologie , Tumeurs colorectales/anatomopathologie , Muscles squelettiques/anatomopathologie , Animaux , Cachexie/complications , Cachexie/physiopathologie , Tumeurs colorectales/complications , Tumeurs colorectales/physiopathologie , Modèles animaux de maladie humaine , Femelle , Mâle , Souris de lignée C57BL , Souris transgéniques , Muscles squelettiques/physiopathologie , Caractères sexuels
4.
Radiat Res ; 193(1): 88-94, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31738662

RÉSUMÉ

Radiation-induced cancer is an ongoing and significant problem, with sources that include clinics worldwide in which 3.1 billion radiology exams are performed each year, as well as a variety of other scenarios such as space travel and nuclear cleanup. These radiation exposures are typically anticipated, and the exposure is typically well below 1 Gy. When radiation-induced (actually ROS-induced) DNA mutation is prevented, then so too are downstream radiation-induced cancers. Currently, there is no protection available against the effects of such <1 Gy radiation exposures. In this study, we address whether the new PrC-210 ROS-scavenger is effective in protecting p53-deficient (p53-/-) mice against X-ray-induced accelerated tumor mortality; this is the most sensitive radiation tumorigenesis model currently known. Six-day-old p53-/- pups received a single intraperitoneal PrC-210 dose [0.5 maximum tolerated dose (MTD)] or vehicle, and 25 min later, pups received 4.0 Gy X-ray irradiation. At 5 min postirradiation, blood was collected to quantify white blood cell c-H2AX foci. Over the next 250 days, tumor-associated deaths were recorded. Findings revealed that when administered 25 min before 4 Gy X-ray irradiation, PrC-210 reduced DNA damage (c-H2AX foci) by 40%, and in a notable coincidence, caused a 40% shift in tumor latency/incidence, and the 0.5 MTD PrC210 dose had no discernible toxicities in these p53-/- mice. Essentially, the moles of PrC-210 thiol within a single 0.5 MTD PrC-210 dose suppressed the moles of ROS generated by 40% of the 4 Gy X-ray dose administered to p53-/- pups, and in doing so, eliminated the lifetime leukemia/lymphoma risk normally residing "downstream" of that 40% of the 4 Gy dose. In conclusion: 1. PrC-210 is readily tolerated by the 6-day-old p53-/- mice, with no discernible lifetime toxicities; 2. PrC-210 does not cause the nausea, emesis or hypotension that preclude clinical use of earlier aminothiols; and 3. PrC-210 significantly increased survival after 4 Gy irradiation in the p53-/- mouse model.


Sujet(s)
Diamines/pharmacologie , Tumeurs radio-induites/mortalité , Radioprotecteurs/pharmacologie , Thiols/pharmacologie , Protéine p53 suppresseur de tumeur/déficit , Animaux , Carcinogenèse/effets des médicaments et des substances chimiques , Carcinogenèse/effets des radiations , Altération de l'ADN , Diamines/sang , Femelle , Humains , Nouveau-né , Mâle , Souris , Tumeurs radio-induites/génétique , Tumeurs radio-induites/anatomopathologie , Tumeurs radio-induites/prévention et contrôle , Radioprotecteurs/métabolisme , Thiols/sang
5.
Int J Cancer ; 145(11): 3022-3032, 2019 12 01.
Article de Anglais | MEDLINE | ID: mdl-31018249

RÉSUMÉ

The normal colon epithelium is transformed into its neoplastic counterpart through a series of genetic alterations in driver genes including activating mutations in PIK3CA. Treatment often involves surgery followed by 5-fluorouracil (5-FU) based therapy, which has limited efficiency and serious side effects. We sought to determine whether fisetin, a dietary flavonoid, alone or in combination with 5-FU affected tumorigenesis in the mammalian intestine. We first determined the effect of fisetin, 5-FU or their combination on PIK3CA-mutant and PIK3CA wild-type colon cancer cells by assessing cell viability, colony formation, apoptosis and effects on PI3K/AKT/mTOR signaling. Treatment of PIK3CA-mutant cells with fisetin and 5-FU reduced the expression of PI3K, phosphorylation of AKT, mTOR, its target proteins, constituents of mTOR signaling complex and this treatment increased the phosphorylation of AMPKα. We then determined whether fisetin and 5-FU together or singly affected tumorigenesis in ApcMin/+ mice that also express constitutively active PI3K in the distal small intestine and colon. Tumor incidence was markedly lower in fisetin-treated FC1 3K1 ApcMin/+ mice that also express constitutively active PI3K in distal small intestine and colon, as compared to control animals, indicating that fisetin is a strong preventive agent. In addition, the combination of fisetin and 5-FU also reduced the total number of intestinal tumors. Fisetin could be used as a preventive agent plus an adjuvant with 5-FU for the treatment of PIK3CA-mutant colorectal cancer.


Sujet(s)
Phosphatidylinositol 3-kinases de classe I/génétique , Tumeurs colorectales/traitement médicamenteux , Flavonoïdes/administration et posologie , Fluorouracil/administration et posologie , Mutation , Animaux , Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Tumeurs colorectales/génétique , Synergie des médicaments , Flavonoïdes/pharmacologie , Flavonols , Fluorouracil/pharmacologie , Cellules HCT116 , Cellules HT29 , Humains , Souris , Phosphorylation/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
6.
Gut ; 66(12): 2132-2140, 2017 12.
Article de Anglais | MEDLINE | ID: mdl-27609830

RÉSUMÉ

OBJECTIVE AND DESIGN: The goal of the study was to determine whether the mutational profile of early colorectal polyps correlated with growth behaviour. The growth of small polyps (6-9 mm) that were first identified during routine screening of patients was monitored over time by interval imaging with CT colonography. Mutations in these lesions with known growth rates were identified by targeted next-generation sequencing. The timing of mutational events was estimated using computer modelling and statistical inference considering several parameters including allele frequency and fitness. RESULTS: The mutational landscape of small polyps is varied both within individual polyps and among the group as a whole but no single alteration was correlated with growth behaviour. Polyps carried 0-3 pathogenic mutations with the most frequent being in APC, KRAS/NRAS, BRAF, FBXW7 and TP53. In polyps with two or more pathogenic mutations, allele frequencies were often variable, indicating the presence of multiple populations within a single tumour. Based on computer modelling, detectable mutations occurred at a mean polyp size of 30±35 crypts, well before the tumour is of a clinically detectable size. CONCLUSIONS: These data indicate that small colon polyps can have multiple pathogenic mutations in crucial driver genes that arise early in the existence of a tumour. Understanding the molecular pathway of tumourigenesis and clonal evolution in polyps that are at risk for progressing to invasive cancers will allow us to begin to better predict which polyps are more likely to progress into adenocarcinomas and which patients are at greater risk of developing advanced disease.


Sujet(s)
Polypes coliques/génétique , Tumeurs colorectales/génétique , Mutation , Allèles , Transformation cellulaire néoplasique , Polypes coliques/imagerie diagnostique , Polypes coliques/anatomopathologie , Coloscopie virtuelle par tomodensitométrie , Tumeurs colorectales/imagerie diagnostique , Tumeurs colorectales/anatomopathologie , Évolution de la maladie , Femelle , Régulation de l'expression des gènes tumoraux , Séquençage nucléotidique à haut débit , Humains , Mâle , Instabilité des microsatellites , Adulte d'âge moyen , Modèles génétiques , Modèles statistiques , Stadification tumorale , Phénotype
7.
PLoS One ; 11(2): e0150170, 2016.
Article de Anglais | MEDLINE | ID: mdl-26919712

RÉSUMÉ

A widely accepted paradigm in the field of cancer biology is that solid tumors are uni-ancestral being derived from a single founder and its descendants. However, data have been steadily accruing that indicate early tumors in mice and humans can have a multi-ancestral origin in which an initiated primogenitor facilitates the transformation of neighboring co-genitors. We developed a new mouse model that permits the determination of clonal architecture of intestinal tumors in vivo and ex vivo, have validated this model, and then used it to assess the clonal architecture of adenomas, intramucosal carcinomas, and invasive adenocarcinomas of the intestine. The percentage of multi-ancestral tumors did not significantly change as tumors progressed from adenomas with low-grade dysplasia [40/65 (62%)], to adenomas with high-grade dysplasia [21/37 (57%)], to intramucosal carcinomas [10/23 (43%]), to invasive adenocarcinomas [13/19 (68%)], indicating that the clone arising from the primogenitor continues to coexist with clones arising from co-genitors. Moreover, neoplastic cells from distinct clones within a multi-ancestral adenocarcinoma have even been observed to simultaneously invade into the underlying musculature [2/15 (13%)]. Thus, intratumoral heterogeneity arising early in tumor formation persists throughout tumorigenesis.


Sujet(s)
Adénocarcinome/anatomopathologie , Adénomes/anatomopathologie , Épithélioma in situ/anatomopathologie , Transformation cellulaire néoplasique , Tumeurs de l'intestin/anatomopathologie , Cellules souches tumorales/anatomopathologie , Adénocarcinome/génétique , Adénomes/génétique , Animaux , Épithélioma in situ/génétique , Lignage cellulaire , Transformation cellulaire néoplasique/génétique , Clones cellulaires/anatomopathologie , Modèles animaux de maladie humaine , Évolution de la maladie , Évolution moléculaire , Protéines de liaison aux acides gras/génétique , Femelle , Régulation de l'expression des gènes tumoraux , Gènes APC , Gènes rapporteurs , Integrases/génétique , Muqueuse intestinale/anatomopathologie , Tumeurs de l'intestin/génétique , Protéines luminescentes/analyse , Protéines luminescentes/génétique , Mâle , Souris , Souris de lignée C57BL , Modèles biologiques , Mosaïcisme , Invasion tumorale , ARN non traduit/génétique , Rats , Transgènes , Microenvironnement tumoral
8.
Cancer Prev Res (Phila) ; 8(10): 952-61, 2015 Oct.
Article de Anglais | MEDLINE | ID: mdl-26276752

RÉSUMÉ

Human colorectal cancers often possess multiple mutations, including three to six driver mutations per tumor. The timing of when these mutations occur during tumor development and progression continues to be debated. More advanced lesions carry a greater number of driver mutations, indicating that colon tumors might progress from adenomas to carcinomas through the stepwise accumulation of mutations following tumor initiation. However, mutations that have been implicated in tumor progression have been identified in normal-appearing epithelial cells of the colon, leaving the possibility that these mutations might be present before the initiation of tumorigenesis. We utilized mouse models of colon cancer to investigate whether tumorigenesis still occurs through the adenoma-to-carcinoma sequence when multiple mutations are present at the time of tumor initiation. To create a model in which tumors could concomitantly possess mutations in Apc, Kras, and Pik3ca, we developed a novel minimally invasive technique to administer an adenovirus expressing Cre recombinase to a focal region of the colon. Here, we demonstrate that the presence of these additional driver mutations at the time of tumor initiation results in increased tumor multiplicity and an increased rate of progression to invasive adenocarcinomas. These cancers can even metastasize to retroperitoneal lymph nodes or the liver. However, despite having as many as three concomitant driver mutations at the time of initiation, these tumors still proceed through the adenoma-to-carcinoma sequence.


Sujet(s)
Adénocarcinome/génétique , Adénomes/génétique , Tumeurs du côlon/génétique , Tumeurs du côlon/anatomopathologie , Gènes APC , Phosphatidylinositol 3-kinases/génétique , Protéines proto-oncogènes p21(ras)/génétique , Adénocarcinome/anatomopathologie , Adénomes/anatomopathologie , Animaux , Phosphatidylinositol 3-kinases de classe I , Modèles animaux de maladie humaine , Évolution de la maladie , Immunohistochimie , Souris , Souris de lignée C57BL , Souris transgéniques , Mutation
9.
PLoS One ; 10(8): e0132727, 2015.
Article de Anglais | MEDLINE | ID: mdl-26252492

RÉSUMÉ

Microsatellite instability (MSI) occurs in over 90% of Lynch syndrome cancers and is considered a hallmark of the disease. MSI is an early event in colon tumor development, but screening polyps for MSI remains controversial because of reduced sensitivity compared to more advanced neoplasms. To increase sensitivity, we investigated the use of a novel type of marker consisting of long mononucleotide repeat (LMR) tracts. Adenomas from 160 patients, ranging in age from 29-55 years old, were screened for MSI using the new markers and compared with current marker panels and immunohistochemistry standards. Overall, 15 tumors were scored as MSI-High using the LMRs compared to 9 for the NCI panel and 8 for the MSI Analysis System (Promega). This difference represents at least a 1.7-fold increase in detection of MSI-High lesions over currently available markers. Moreover, the number of MSI-positive markers per sample and the size of allelic changes were significantly greater with the LMRs (p = 0.001), which increased confidence in MSI classification. The overall sensitivity and specificity of the LMR panel for detection of mismatch repair deficient lesions were 100% and 96%, respectively. In comparison, the sensitivity and specificity of the MSI Analysis System were 67% and 100%; and for the NCI panel, 75% and 97%. The difference in sensitivity between the LMR panel and the other panels was statistically significant (p<0.001). The increased sensitivity for detection of MSI-High phenotype in early colorectal lesions with the new LMR markers indicates that MSI screening for the early detection of Lynch syndrome might be feasible.


Sujet(s)
Tumeurs colorectales/génétique , Dépistage précoce du cancer/méthodes , Instabilité des microsatellites , Adulte , Allèles , Marqueurs biologiques tumoraux/génétique , Réparation de mésappariement de l'ADN/génétique , Humains , Immunohistochimie , Adulte d'âge moyen , Protéines proto-oncogènes B-raf/génétique , Sensibilité et spécificité
10.
PLoS One ; 9(10): e109668, 2014.
Article de Anglais | MEDLINE | ID: mdl-25286226

RÉSUMÉ

The treatment of localized colorectal cancer (CRC) depends on resection of the primary tumor with adequate margins and sufficient lymph node sampling. A novel imaging agent that accumulates in CRCs and the associated lymph nodes is needed. Cellectar Biosciences has developed a phospholipid ether analog platform that is both diagnostic and therapeutic. CLR1502 is a near-infrared fluorescent molecule, whereas 124/131I-CLR1404 is under clinical investigation as a PET tracer/therapeutic agent imaged by SPECT. We investigated the use of CLR1502 for the detection of intestinal cancers in a murine model and 131I-CLR1404 in a patient with metastatic CRC. Mice that develop multiple intestinal tumors ranging from adenomas to locally advanced adenocarcinomas were utilized. After 96 hours post CLR1502 injection, the intestinal tumors were analyzed using a Spectrum IVIS (Perkin Elmer) and a Fluobeam (Fluoptics). The intensity of the fluorescent signal was correlated with the histological characteristics for each tumor. Colon adenocarcinomas demonstrated increased accumulation of CLR1502 compared to non-invasive lesions (total radiant efficiency: 1.76×10(10) vs 3.27×10(9) respectively, p = 0.006). Metastatic mesenteric tumors and uninvolved lymph nodes were detected with CLR1502. In addition, SPECT imaging with 131I-CLR1404 was performed as part of a clinical trial in patients with advanced solid tumors. 131I-CLR1404 was shown to accumulate in metastatic tumors in a patient with colorectal adenocarcinoma. Together, these compounds might enhance our ability to properly resect CRCs through better localization of the primary tumor and improved lymph node identification as well as detect distant disease.


Sujet(s)
Tumeurs colorectales/imagerie diagnostique , Indoles , Iodobenzènes , Éther-phospholipides , Phosphoryl-choline , Adénocarcinome/imagerie diagnostique , Animaux , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Femelle , Humains , Indoles/métabolisme , Tumeurs de l'intestin/imagerie diagnostique , Iodobenzènes/métabolisme , Métastase lymphatique , Souris , Invasion tumorale , Éther-phospholipides/métabolisme , Phosphoryl-choline/métabolisme , Tomographie par émission monophotonique
11.
Cancer Prev Res (Phila) ; 7(1): 105-13, 2014 Jan.
Article de Anglais | MEDLINE | ID: mdl-24196829

RÉSUMÉ

Colorectal cancer often arises from adenomatous colonic polyps. Polyps can grow and progress to cancer, but may also remain static in size, regress, or resolve. Predicting which polyps progress and which remain benign is difficult. We developed a novel long-lived murine model of colorectal cancer with tumors that can be followed by colonoscopy. Our aim was to assess whether these tumors have similar growth patterns and histologic fates to human colorectal polyps to identify features to aid in risk stratification of colonic tumors. Long-lived Apc(Min/+) mice were treated with dextran sodium sulfate to promote colonic tumorigenesis. Tumor growth patterns were characterized by serial colonoscopy with biopsies obtained for immunohistochemistry and gene expression profiling. Tumors grew, remained static, regressed, or resolved over time with different relative frequencies. Newly developed tumors demonstrated higher rates of growth and resolution than more established tumors that tended to remain static in size. Colonic tumors were hyperplastic lesions (3%), adenomas (73%), intramucosal carcinomas (20%), or adenocarcinomas (3%). Interestingly, the level of ß-catenin was higher in adenomas that became intratumoral carcinomas than those that failed to progress. In addition, differentially expressed genes between adenomas and intramucosal carcinomas were identified. This novel murine model of intestinal tumorigenesis develops colonic tumors that can be monitored by serial colonoscopy, mirror growth patterns seen in human colorectal polyps, and progress to colorectal cancer. Further characterization of cellular and molecular features is needed to determine which features can be used to risk-stratify polyps for progression to colorectal cancer and potentially guide prevention strategies.


Sujet(s)
Polypes coliques/anatomopathologie , Tumeurs colorectales/anatomopathologie , Modèles animaux de maladie humaine , Protéine de la polypose adénomateuse colique/génétique , Polypes adénomateux/anatomopathologie , Animaux , Coloscopie , Évolution de la maladie , Femelle , Régulation de l'expression des gènes tumoraux , Génotype , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Séquençage par oligonucléotides en batterie , Facteurs temps
12.
Cancer Res ; 72(12): 2931-6, 2012 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-22525701

RÉSUMÉ

Aberrations in the phosphoinositide 3-kinase (PI3K) signaling pathway play a key role in the pathogenesis of numerous cancers by altering cellular growth, metabolism, proliferation, and apoptosis. Mutations in the catalytic domain of PI3K that generate a dominantly active kinase are commonly found in human colorectal cancers and have been thought to drive tumor progression but not initiation. However, the effects of constitutively activated PI3K upon the intestinal mucosa have not been previously studied in animal models. Here, we show that the expression of a dominantly active form of the PI3K protein in the mouse intestine results in hyperplasia and advanced neoplasia. Mice expressing constitutively active PI3K in the epithelial cells of the distal small bowel and colon rapidly developed invasive adenocarcinomas in the colon that spread into the mesentery and adjacent organs. The histologic characteristics of these tumors were strikingly similar to invasive mucinous colon cancers in humans. Interestingly, these tumors formed without a benign polypoid intermediary, consistent with the lack of aberrant WNT signaling observed. Together, our findings indicate a noncanonical mechanism of colon tumor initiation that is mediated through activation of PI3K. This unique model has the potential to further our understanding of human disease and facilitate the development of therapeutics through pharmacologic screening and biomarker identification.


Sujet(s)
Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Phosphatidylinositol 3-kinases/génétique , Phosphatidylinositol 3-kinases/métabolisme , Adénocarcinome/génétique , Adénocarcinome/métabolisme , Adénocarcinome/anatomopathologie , Animaux , Apoptose , Marqueurs biologiques tumoraux , Prolifération cellulaire , Transformation cellulaire néoplasique , Tumeurs colorectales/métabolisme , Activation enzymatique , Génotype , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Souris , Souris de lignée C57BL , Souris transgéniques , Protéines proto-oncogènes c-akt/métabolisme , Voie de signalisation Wnt
13.
Arch Biochem Biophys ; 515(1-2): 64-71, 2011 Nov.
Article de Anglais | MEDLINE | ID: mdl-21907701

RÉSUMÉ

Epidemiological studies indicate that sunlight exposure and vitamin D are each associated with a lower risk of colon cancer. The few controlled supplementation trials testing vitamin D in humans reported to date show conflicting results. We have used two genetic models of familial colon cancer, the Apc(Pirc/+) (Pirc) rat and the Apc(Min/+) (Min) mouse, to investigate the effect of 25-hydroxyvitamin D(3) [25(OH)D(3)] and two analogs of vitamin D hormone on colonic tumors. Longitudinal endoscopic monitoring allowed us to test the efficacy of these compounds in preventing newly arising colonic tumors and in affecting established colonic tumors. 25(OH)D(3) and two analogs of vitamin D hormone each failed to reduce tumor multiplicities or alter the growth patterns of colonic tumors in the Pirc rat or the Min mouse.


Sujet(s)
Tumeurs du côlon/prévention et contrôle , Vitamine D/administration et posologie , Animaux , Tumeurs du côlon/métabolisme , Compléments alimentaires , Modèles animaux de maladie humaine , Femelle , Mâle , Souris , Rats , Vitamine D/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...