Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 140
Filtrar
1.
Int J Mol Sci ; 22(15)2021 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-34361052

RESUMEN

Colon cancer (CC) is considered a high-risk cancer in developed countries. Its etiology is correlated with a high consumption of red meat and low consumption of plant-based foods, including whole grains. Sorghum bran is rich in polyphenols. This study aimed to determine whether different high-phenolic sorghum brans suppress tumor formation in a genetic CC rodent model and elucidate mechanisms. Tissue culture experiments used colorectal cancer cell lines SW480, HCT-116 and Caco-2 and measured protein expression, and protein activity. The animal model used in this study was APC Min+/mouse model combined with dextram sodium sulfate. High phenolic sorghum bran extract treatment resulted in the inhibition of proliferation and induced apoptosis in CC cell lines. Treatment with high phenolic sorghum bran extracts repressed TNF-α-stimulated NF-κB transactivation and IGF-1-stimulated PI3K/AKT pathway via the downregulation of ß-catenin transactivation. Furthermore, high-phenolic sorghum bran extracts activated AMPK and autophagy. Feeding with high-phenolic sorghum bran for 6 weeks significantly suppressed tumor formation in an APC Min/+ dextran sodium sulfate promoted CC mouse model. Our data demonstrates the potential application of high-phenolic sorghum bran as a functional food for the prevention of CC.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Neoplasias Colorrectales/tratamiento farmacológico , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Extractos Vegetales/farmacología , Sorghum/química , Animales , Apoptosis , Proliferación Celular , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Femenino , Humanos , Masculino , Ratones , Células Tumorales Cultivadas
2.
Blood ; 137(5): 610-623, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33538795

RESUMEN

This study was conducted to determine the dosage effect of c-Myc on hematopoiesis and its distinct role in mediating the Wnt/ß-catenin pathway in hematopoietic stem cell (HSC) and bone marrow niche cells. c-Myc haploinsufficiency led to ineffective hematopoiesis by inhibiting HSC self-renewal and quiescence and by promoting apoptosis. We have identified Nr4a1, Nr4a2, and Jmjd3, which are critical for the maintenance of HSC functions, as previously unrecognized downstream targets of c-Myc in HSCs. c-Myc directly binds to the promoter regions of Nr4a1, Nr4a2, and Jmjd3 and regulates their expression. Our results revealed that Nr4a1 and Nr4a2 mediates the function of c-Myc in regulating HSC quiescence, whereas all 3 genes contribute to the function of c-Myc in the maintenance of HSC survival. Adenomatous polyposis coli (Apc) is a negative regulator of the Wnt/ß-catenin pathway. We have provided the first evidence that Apc haploinsufficiency induces a blockage of erythroid lineage differentiation through promoting secretion of IL6 in bone marrow endothelial cells. We found that c-Myc haploinsufficiency failed to rescue defective function of Apc-deficient HSCs in vivo but it was sufficient to prevent the development of severe anemia in Apc-heterozygous mice and to significantly prolong the survival of those mice. Furthermore, we showed that c-Myc-mediated Apc loss induced IL6 secretion in endothelial cells, and c-Myc haploinsufficiency reversed the negative effect of Apc-deficient endothelial cells on erythroid cell differentiation. Our studies indicate that c-Myc has a context-dependent role in mediating the function of Apc in hematopoiesis.


Asunto(s)
Genes myc , Hematopoyesis/fisiología , Proteínas Proto-Oncogénicas c-myb/fisiología , Proteína de la Poliposis Adenomatosa del Colon/fisiología , Anemia/genética , Anemia/prevención & control , Animales , Apoptosis/fisiología , Trasplante de Médula Ósea , Autorrenovación de las Células/fisiología , Ensayo de Unidades Formadoras de Colonias , Células Endoteliales/patología , Células Eritroides/patología , Eliminación de Gen , Genes APC , Haploinsuficiencia , Hematopoyesis/genética , Células Madre Hematopoyéticas , Interleucina-6/fisiología , Histona Demetilasas con Dominio de Jumonji/fisiología , Ratones Mutantes , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/fisiología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/fisiología , Poli I-C/farmacología , Quimera por Radiación , Vía de Señalización Wnt/fisiología
3.
Int J Mol Sci ; 21(12)2020 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-32586050

RESUMEN

Pancreatic cancer (PC) is a highly lethal malignancy due to the cancer routinely being diagnosed late and having a limited response to chemotherapy. Pancreatic ductal adenocarcinoma (PDAC) is the most common form of pancreatic malignant tumor, representing more than 85% of all pancreatic cancers. In the present study, we characterized the phenotypes of concomitant P53 and APC mutations in pancreatic neoplasms driven by the oncogene KRAS in genetically modified mice (GEMM). In this GEMM setting, APC haploinsufficiency coupled with P53 deletion and KRASG12D activation resulted in an earlier appearance of pancreatic intraepithelial neoplasia (PanIN) lesions and progressed rapidly to highly invasive and metastatic PDAC. Through a microarray analysis of murine PDAC cells derived from our APC-deficient PDAC model, we observed that APC loss leads to upregulated CD34 expression in PDAC. CD34 is a member of a family of single-pass transmembrane proteins and is selectively expressed in hematopoietic progenitor cells, vascular endothelial cells, interstitial precursor cells, and various interstitial tumor cells. However, the functional roles of CD34 in pancreatic cancer remain unclear. Thus, in this study, we explored the mechanisms regarding how CD34 promotes the deterioration of pancreatic malignancy. Our results demonstrated that the increased expression of CD34 induced by APC inactivation promotes the invasion and migration of PDAC cells, which may relate to PDAC metastasis in vivo. Collectively, our study provides first-line evidence to delineate the association between CD34 and the APC/Wnt pathway in PDAC, and reveals the potential roles of CD34 in PDAC progression.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Antígenos CD34/metabolismo , Carcinoma Ductal Pancreático/secundario , Transición Epitelial-Mesenquimal , Células Madre Neoplásicas/patología , Neoplasias Pancreáticas/patología , Animales , Antígenos CD34/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Mutación , Células Madre Neoplásicas/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Fenotipo , Transducción de Señal
4.
Cell Death Differ ; 27(10): 2872-2887, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32355182

RESUMEN

Ribosome biogenesis inhibition causes cell cycle arrest and apoptosis through the activation of tumor suppressor-dependent surveillance pathways. These responses are exacerbated in cancer cells, suggesting that targeting ribosome synthesis may be beneficial to patients. Here, we characterize the effect of the loss-of-function of Notchless (Nle), an essential actor of ribosome biogenesis, on the intestinal epithelium undergoing tumor initiation due to acute Apc loss-of-function. We show that ribosome biogenesis dysfunction strongly alleviates Wnt-driven tumor initiation by restoring cell cycle exit and differentiation in Apc-deficient progenitors. Conversely Wnt hyperactivation attenuates the cellular responses to surveillance pathways activation induced by ribosome biogenesis dysfunction, as proliferation was maintained at control-like levels in the stem cells and progenitors of double mutants. Thus, our data indicate that, while ribosome biogenesis inhibition efficiently reduces cancer cell proliferation in the intestinal epithelium, enhanced resistance of Apc-deficient stem and progenitor cells to ribosome biogenesis defects may be an important concern when using a therapeutic strategy targeting ribosome production for the treatment of Wnt-dependent tumorigenesis.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Transformación Celular Neoplásica , Mucosa Intestinal , Proteínas de la Membrana/fisiología , Ribosomas/metabolismo , Vía de Señalización Wnt , Animales , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos C57BL , Biogénesis de Organelos
5.
Cancer Immunol Immunother ; 69(7): 1279-1292, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32185408

RESUMEN

The presence of activated T cells in colorectal cancer tissues is a strong predictor of patient survival. Our previous studies have shown that regulatory T cells (Treg) are able to reduce T cell transendothelial migration in vitro and accumulation of effector T cells in intestinal tumors in vivo in the murine APCMin/+ model for microsatellite stable intestinal tumors. In this study, we investigated the effect of Treg depletion on the density and effector functions of different TCRαß+ and TCRγδ+ T cell populations in intestinal tumors. We used the APCMin/+\DEREG mouse model, which harbor a diphtheria toxin receptor under the control of the FOXP3 promoter, to deplete Treg in tumor bearing mice. We found that the density of conventional TCRαß+CD8αß+ T cells was significantly increased in Treg-depleted tumors in comparison with Treg-proficient tumors. Furthermore, TCRαß+CD8αß+ T cells showed increased proliferation and activation as well as increased Granzyme B and IFN-γ production in Treg-depleted tumors. In sharp contrast, the densities and effector functions of TCRαß+CD8αα+ T cells and TCRγδ+ T cells remained unchanged by Treg depletion. We also documented a distinct population of IL-17A+TNF+ TCRγδ+CD8- T cells in tumors, which were not affected by Treg depletion. We conclude that Treg depletion affects only conventional TCRαß+CD8αß+ T cells in intestinal tumors, while unconventional T cells and T cells in unaffected tissue are not altered. Immunotherapies aimed at depleting Treg from tumors may thus be a viable option for reinvigoration of conventional cytotoxic T cells with a Th1 cytokine profile.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Antígenos CD8/inmunología , Linfocitos T CD8-positivos/inmunología , Neoplasias Intestinales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/metabolismo , Femenino , Humanos , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Activación de Linfocitos , Depleción Linfocítica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo
6.
Eur J Cancer Prev ; 29(6): 481-485, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-31833958

RESUMEN

HASPIN has been identified as a nuclear Ser/Thr kinase specifically expressed in haploid germ cells. HASPIN kinase inhibitors were recently isolated, and their antitumor activity reported. Colorectal cancer occurs with high incidence worldwide. In this study, we examined whether HASPIN inhibitor CHR-6494 suppresses cancer progression in Apc mice, a familial colon tumor disease model. Mice were treated by intraperitoneal injection of CHR-6494 for 50 days. Following the treatment period, intestinal polyps were counted and testosterone and spermatogenesis levels were observed. Intraperitoneal administration of CHR-6494 significantly inhibited intestinal polyp development and recovered body weight in Apc mice. Although spermatogenesis was inhibited with increasing age in Apc mice, CHR-6494 significantly improved blood testosterone levels and spermatogenesis. Our results suggest that HASPIN inhibitors may be useful as anti-cancer agents and for the treatment of hypogonadism in colorectal cancer patients.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Caquexia/tratamiento farmacológico , Hipogonadismo/tratamiento farmacológico , Indazoles/farmacología , Neoplasias Intestinales/tratamiento farmacológico , Pólipos Intestinales/tratamiento farmacológico , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Piridazinas/farmacología , Animales , Caquexia/etiología , Caquexia/metabolismo , Caquexia/patología , Femenino , Hipogonadismo/etiología , Hipogonadismo/metabolismo , Hipogonadismo/patología , Neoplasias Intestinales/etiología , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Pólipos Intestinales/etiología , Pólipos Intestinales/metabolismo , Pólipos Intestinales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
7.
Cancer Prev Res (Phila) ; 12(7): 433-448, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31088824

RESUMEN

Colorectal cancer is a leading cause of cancer deaths. The renin-angiotensin system (RAS) is upregulated in colorectal cancer, and epidemiologic studies suggest RAS inhibitors reduce cancer risk. Because vitamin D (VD) receptor negatively regulates renin, we examined anticancer efficacy of VD and losartan (L), an angiotensin receptor blocker. Control Apc+/LoxP mice and tumor-forming Apc+/LoxP Cdx2P-Cre mice were randomized to unsupplemented Western diet (UN), or diets supplemented with VD, L, or VD+L, the latter to assess additive or synergistic effects. At 6 months, mice were killed. Plasma Ca2+, 25(OH)D3, 1α, 25(OH)2D3, renin, and angiotensin II (Ang II) were quantified. Colonic transcripts were assessed by qPCR and proteins by immunostaining and blotting. Cancer incidence and tumor burden were significantly lower in Cre+ VD and Cre+ L, but not in the Cre+ VD+L group. In Apc+/LoxP mice, VD increased plasma 1,25(OH)2D3 and colonic VDR. In Apc+/LoxP-Cdx2P-Cre mice, plasma renin and Ang II, and colonic tumor AT1, AT2, and Cyp27B1 were increased and VDR downregulated. L increased, whereas VD decreased plasma renin and Ang II in Cre+ mice. VD or L inhibited tumor development, while exerting differential effects on plasma VD metabolites and RAS components. We speculate that AT1 is critical for tumor development, whereas RAS suppression plays a key role in VD chemoprevention. When combined with L, VD no longer increases active VD and colonic VDR in Cre- mice nor suppresses renin and Ang II in Cre+ mice, likely contributing to lack of chemopreventive efficacy of the combination.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Neoplasias del Colon/prevención & control , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Losartán/farmacología , Vitamina D/farmacología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Apoptosis , Proliferación Celular , Neoplasias del Colon/etiología , Neoplasias del Colon/patología , Quimioterapia Combinada , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Células Tumorales Cultivadas , Vitaminas/farmacología
8.
Sci Rep ; 8(1): 2735, 2018 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-29426940

RESUMEN

While the Wnt/ß-catenin pathway plays a critical role in the maintenance of the zonation of ammonia metabolizing enzymes in the adult liver, the mechanisms responsible for inducing zonation in the embryo are not well understood. Herein we address the spatiotemporal role of the Wnt/ß-catenin pathway in the development of zonation in embryonic mouse liver by conditional deletion of Apc and ß-catenin at different stages of mouse liver development. In normal development, the ammonia metabolising enzymes carbamoylphosphate synthetase I (CPSI) and Glutamine synthetase (GS) begin to be expressed in separate hepatoblasts from E13.5 and E15.5 respectively and gradually increase in number thereafter. Restriction of GS expression occurs at E18 and becomes increasingly limited to the terminal perivenous hepatocytes postnatally. Expression of nuclear ß-catenin coincides with the restriction of GS expression to the terminal perivenous hepatocytes. Conditional loss of Apc resulted in the expression of nuclear ß-catenin throughout the developing liver and increased number of cells expressing GS. Conversely, conditional loss of ß-catenin resulted in loss of GS expression. These data suggest that the Wnt pathway is critical to the development of zonation as well as maintaining the zonation in the adult liver.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Carbamoil-Fosfato Sintasa (Amoniaco)/metabolismo , Glutamato-Amoníaco Ligasa/metabolismo , Hepatocitos/metabolismo , Hígado/embriología , Vía de Señalización Wnt/fisiología , beta Catenina/fisiología , Proteína de la Poliposis Adenomatosa del Colon/genética , Amoníaco/metabolismo , Animales , Hepatocitos/citología , Hígado/metabolismo , Mutación con Pérdida de Función , Ratones , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/genética , beta Catenina/genética
9.
Curr Cancer Drug Targets ; 18(9): 905-911, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28786349

RESUMEN

BACKGROUND: Recent epidemiological and experimental studies have shown that obesity is a major risk factor for Colorectal Cancer (CRC). Regular intake of high fat-containing diet can promote obesity and metabolic syndrome by increasing the insulin resistance and inflammatory response which contribute to carcinogenesis. Previously, we have shown that inhibition of polyol pathway enzyme aldose reductase (AR) prevents carcinogens- and inflammatory growth factorsinduced CRC. However, the effect of AR inhibition on a high-fat diet (HFD)-induced formation of intestinal polyps in Apc-deficient Min (multiple intestinal neoplasia; ApcMin/+) mice is not known. METHODS: We examined the effect of AR inhibitor, fidarestat on the HFD-induced formation of preneoplastic intestinal polyps in ApcMin/+ mice which is an excellent model of colon cancer. RESULTS: APCMin/+ mice fed for 12 weeks of HFD caused a significant increase in the formation of polyps in the small and large intestines and fidarestat given along with the HFD prevented the number of intestinal polyps. Fidarestat also decreased the size of the polyps in the intestines of HFDtreated APC Min mice. Further, the expression levels of beta-catenin, PCNA, PKC-ß2, P-AKT, Pp65, COX-2, and iNOS in the small and large intestines of HFD-treated mice significantly increased, and AR inhibitor prevented it. CONCLUSION: Our results thus suggest that fidarestat could be used as a potential chemopreventive drug for intestinal cancers due to APC gene mutations.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Aldehído Reductasa/antagonistas & inhibidores , Neoplasias del Colon/prevención & control , Dieta Alta en Grasa/efectos adversos , Inhibidores Enzimáticos/farmacología , Imidazolidinas/farmacología , Pólipos Intestinales/prevención & control , Animales , Neoplasias del Colon/enzimología , Neoplasias del Colon/etiología , Femenino , Pólipos Intestinales/enzimología , Pólipos Intestinales/etiología , Masculino , Ratones , Ratones Endogámicos C57BL
10.
Genes Dev ; 31(16): 1679-1692, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28916710

RESUMEN

Adenomatous polyposis coli (APC) regulates the activity of ß-catenin, an integral component of Wnt signaling. However, the selective role of the APC-ß-catenin pathway in cerebral cortical development is unknown. Here we genetically dissected the relative contributions of APC-regulated ß-catenin signaling in cortical progenitor development, a necessary early step in cerebral cortical formation. Radial progenitor-specific inactivation of the APC-ß-catenin pathway indicates that the maintenance of appropriate ß-catenin-mediated Wnt tone is necessary for the orderly differentiation of cortical progenitors and the resultant formation of the cerebral cortex. APC deletion deregulates ß-catenin, leads to high Wnt tone, and disrupts Notch1 signaling and primary cilium maintenance necessary for radial progenitor functions. ß-Catenin deregulation directly disrupts cilium maintenance and signaling via Tulp3, essential for intraflagellar transport of ciliary signaling receptors. Surprisingly, deletion of ß-catenin or inhibition of ß-catenin activity in APC-null progenitors rescues the APC-null phenotype. These results reveal that APC-regulated ß-catenin activity in cortical progenitors sets the appropriate Wnt tone necessary for normal cerebral cortical development.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Corteza Cerebral/embriología , Células-Madre Neurales/metabolismo , Neurogénesis , Vía de Señalización Wnt , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Proliferación Celular , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Cilios/metabolismo , Proteínas Hedgehog/metabolismo , Ratones , Ratones Noqueados , Células-Madre Neurales/citología , Receptor Notch1/metabolismo , beta Catenina/fisiología
11.
Life Sci Space Res (Amst) ; 13: 45-50, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28554509

RESUMEN

Ionizing radiation (IR) is a recognized risk factor for colorectal cancer (CRC) and astronauts undertaking long duration space missions are expected to receive IR doses in excess of permissible limits with implications for colorectal carcinogenesis. Exposure to IR in outer space occurs at low doses and dose rates, and energetic heavy ions due to their high linear energy transfer (high-LET) characteristics remain a major concern for CRC risk in astronauts. Previously, we have demonstrated that intestinal tumorigenesis in a mouse model (APC1638N/+) of human colorectal cancer was significantly higher after exposure to high dose rate energetic heavy ions relative to low-LET γ radiation. The purpose of the current study was to compare intestinal tumorigenesis in APC1638N/+ mice after exposure to energetic heavy ions at high (50cGy/min) and relatively low (0.33cGy/min) dose rate. Male and female mice (6-8 weeks old) were exposed to either 10 or 50cGy of 28Si (energy: 300MeV/n; LET: 70keV/µm) or 56Fe (energy: 1000MeV/n; LET: 148keV/µm) ions at NASA Space Radiation Laboratory in Brookhaven National Laboratory. Mice (n=20 mice/group) were euthanized and intestinal and colon tumor frequency and size were counted 150days after radiation exposure. Intestinal tumorigenesis in male mice exposed to 56Fe was similar for high and low dose rate exposures. Although male mice showed a decreasing trend at low dose rate relative to high dose rate exposures, the differences in tumor frequency between the two types of exposures were not statistically significant after 28Si radiation. In female mice, intestinal tumor frequency was similar for both radiation type and dose rates tested. In both male and female mice intestinal tumor size was not different after high and low dose rate radiation exposures. Colon tumor frequency in male and female mice after high and low dose rate energetic heavy ions was also not significantly different. In conclusion, intestinal and colonic tumor frequency and size was similar irrespective of energetic heavy ion radiation dose rate suggesting that carcinogenic potential of energetic heavy ions is independent of dose rate.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Carcinogénesis/patología , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Radioterapia de Iones Pesados/efectos adversos , Neoplasias Intestinales/patología , Neoplasias Inducidas por Radiación/patología , Animales , Neoplasias del Colon/etiología , Relación Dosis-Respuesta en la Radiación , Femenino , Neoplasias Intestinales/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias Inducidas por Radiación/etiología , Exposición a la Radiación/efectos adversos
12.
BMC Cancer ; 16(1): 942, 2016 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-27927180

RESUMEN

BACKGROUND: Colorectal cancer is the second most common cause of cancer deaths for both men and women, and the third most common cause of cancer in the U.S. Toxicity of current chemotherapeutic agents for colorectal cancer, and emergence of drug resistance underscore the need to develop new, potentially less toxic alternatives. Our recent cross-sectional study in a large Appalachian population, showed a strong, inverse, dose-response association of serum perfluorooctane sulfonate (PFOS) levels to prevalent colorectal cancer, suggesting PFOS may have therapeutic potential in the prevention and/or treatment of colorectal cancer. In these preliminary studies using a mouse model of familial colorectal cancer, the APCmin mouse, and exposures comparable to those reported in human populations, we assess the efficacy of PFOS for reducing tumor burden, and evaluate potential dose-response effects. METHODS: At 5-6 weeks of age, APCmin mice were randomized to receive 0, 20, 250 mg PFOS/kg (females) or 0, 10, 50 and 200 mg PFOS/kg (males) via their drinking water. At 15 weeks of age, gastrointestinal tumors were counted and scored and blood PFOS levels measured. RESULTS: PFOS exposure was associated with a significant, dose-response reduction in total tumor number in both male and female mice. This inverse dose-response effect of PFOS exposure was particularly pronounced for larger tumors (r2 for linear trend = 0.44 for males, p's <0.001). CONCLUSIONS: The current study in a mouse model of familial adenomatous polyposis offers the first experimental evidence that chronic exposure to PFOS in drinking water can reduce formation of gastrointestinal tumors, and that these reductions are both significant and dose-dependent. If confirmed in further studies, these promising findings could lead to new therapeutic strategies for familial colorectal cancer, and suggest that PFOS testing in both preventive and therapeutic models for human colorectal cancer is warranted.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Poliposis Adenomatosa del Colon/tratamiento farmacológico , Ácidos Alcanesulfónicos/administración & dosificación , Modelos Animales de Enfermedad , Fluorocarburos/administración & dosificación , Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/patología , Administración Oral , Ácidos Alcanesulfónicos/farmacología , Animales , Relación Dosis-Respuesta a Droga , Femenino , Fluorocarburos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL
13.
Oncotarget ; 7(43): 69883-69902, 2016 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-27566565

RESUMEN

Two important protein-protein interactions establish E-cadherin (Cdh1) in the adhesion complex; homophilic binding via the extra-cellular (EC1) domain and cytoplasmic tail binding to ß-catenin. Here, we evaluate whether E-cadherin binding can inhibit ß-catenin when there is loss of Adenomatous polyposis coli (APC) from the ß-catenin destruction complex. Combined conditional loss of Cdh1 and Apc were generated in the intestine, intestinal adenoma and adenoma organoids. Combined intestinal disruption (Cdh1fl/flApcfl/flVil-CreERT2) resulted in lethality, breakdown of the intestinal barrier, increased Wnt target gene expression and increased nuclear ß-catenin localization, suggesting that E-cadherin inhibits ß-catenin. Combination with an intestinal stem cell Cre (Lgr5CreERT2) resulted in ApcΔ/Δ recombination and adenoma, but intact Cdh1fl/fl alleles. Cultured ApcΔ/ΔCdh1fl/fl adenoma cells infected with adenovirus-Cre induced Cdh1fl/fl recombination (Cdh1Δ/Δ), disruption of organoid morphology, nuclear ß-catenin localization, and cells with an epithelial-mesenchymal phenotype. Complementation with adenovirus expressing wild-type Cdh1 (Cdh1-WT) rescued adhesion and ß-catenin membrane localization, yet an EC1 specific double mutant defective in homophilic adhesion (Cdh1-MutW2A, S78W) did not. These data suggest that E-cadherin inhibits ß-catenin in the context of disruption of the APC-destruction complex, and that this function is also EC1 domain dependent. Both binding functions of E-cadherin may be required for its tumour suppressor activity.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Cadherinas/fisiología , Núcleo Celular/metabolismo , Transición Epitelial-Mesenquimal , beta Catenina/metabolismo , Adenoma/etiología , Animales , Antígenos CD , Cadherinas/química , Desarrollo Embrionario , Humanos , Neoplasias Intestinales/etiología , Células MCF-7 , Ratones , Organoides , Dominios Proteicos , Recombinación Genética , Tamoxifeno/farmacología
14.
J Cancer Res Clin Oncol ; 142(5): 913-25, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26702935

RESUMEN

PURPOSE: Naringin is a natural dietary flavonoid compound. We aimed to evaluate the effects of naringin on intestinal tumorigenesis in the adenomatous polyposis coli multiple intestinal neoplasia (Apc (Min/+)) mouse model. METHODS: Apc (Min/+) mice were given either naringin (150 mg/kg) or vehicle by p.o. gavage daily for 12 consecutive weeks. Mice were killed with ether, and blood samples were collected to assess the concentrations of IL-6 and PGE2. Total intestines were removed, and the number of polyps was examined. Tissue samples of intestinal polyps were subjected to the assays of histopathology, immunohistochemical analysis and Western blotting analysis. RESULTS: Apc (Min/+) mice fed with naringin developed less and smaller polyps in total intestines. Naringin prevented intestinal tumorigenesis without adverse effects. Histopathologic analysis revealed the reduction of dysplastic cells and dysplasia in the adenomatous polyps. The treatments' effects might arise from its anti-proliferation, induction of apoptosis and modulation of GSK-3ß and APC/ß-catenin signaling pathways. Naringin also exerted its effects on tumorigenesis through anti-chronic inflammation. CONCLUSION: Naringin prevented intestinal tumorigenesis likely through a collection of activities including anti-proliferation, induction of apoptosis, modulation of GSK-3ß and APC/ß-catenin pathways and anti-inflammation. Naringin is a potential chemopreventive agent for reducing the risk of colonic cancers.


Asunto(s)
Transformación Celular Neoplásica/efectos de los fármacos , Suplementos Dietéticos , Modelos Animales de Enfermedad , Flavanonas/farmacología , Neoplasias Intestinales/prevención & control , Pólipos Intestinales/prevención & control , Proteína de la Poliposis Adenomatosa del Colon/fisiología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Citocinas/metabolismo , Femenino , Humanos , Técnicas para Inmunoenzimas , Mediadores de Inflamación/metabolismo , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Pólipos Intestinales/metabolismo , Pólipos Intestinales/patología , Ratones , Ratones Endogámicos C57BL , Células Tumorales Cultivadas
15.
Mol Biol Cell ; 27(3): 466-82, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26658612

RESUMEN

Mutations in adenomatous polyposis coli (APC) disrupt regulation of Wnt signaling, mitosis, and the cytoskeleton. We describe a new role for APC in the transport of mitochondria. Silencing of wild-type APC by small interfering RNA caused mitochondria to redistribute from the cell periphery to the perinuclear region. We identified novel APC interactions with the mitochondrial kinesin-motor complex Miro/Milton that were mediated by the APC C-terminus. Truncating mutations in APC abolished its ability to bind Miro/Milton and reduced formation of the Miro/Milton complex, correlating with disrupted mitochondrial distribution in colorectal cancer cells that could be recovered by reconstitution of wild-type APC. Using proximity ligation assays, we identified endogenous APC-Miro/Milton complexes at mitochondria, and live-cell imaging showed that loss of APC slowed the frequency of anterograde mitochondrial transport to the membrane. We propose that APC helps drive mitochondria to the membrane to supply energy for cellular processes such as directed cell migration, a process disrupted by cancer mutations.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Proteínas Portadoras/metabolismo , Membrana Celular/ultraestructura , Mitocondrias/fisiología , Proteínas Mitocondriales/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/química , Animales , Transporte Biológico , Línea Celular Tumoral , Membrana Celular/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Microtúbulos/fisiología , Mutación , Células 3T3 NIH , Neoplasias/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica
16.
J Cell Biol ; 210(5): 717-26, 2015 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-26304725

RESUMEN

The tumor suppressor adenomatous polyposis coli (APC) is a crucial regulator of many stem cell types. In constantly cycling stem cells of fast turnover tissues, APC loss results in the constitutive activation of a Wnt target gene program that massively increases proliferation and leads to malignant transformation. However, APC function in skeletal muscle, a tissue with a low turnover rate, has never been investigated. Here we show that conditional genetic disruption of APC in adult muscle stem cells results in the abrogation of adult muscle regenerative potential. We demonstrate that APC removal in adult muscle stem cells abolishes cell cycle entry and leads to cell death. By using double knockout strategies, we further prove that this phenotype is attributable to overactivation of ß-catenin signaling. Our results demonstrate that in muscle stem cells, APC dampens canonical Wnt signaling to allow cell cycle progression and radically diverge from previous observations concerning stem cells in actively self-renewing tissues.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Células Madre Adultas/fisiología , Apoptosis/genética , Músculo Esquelético/fisiología , Regeneración/fisiología , Células Satélite del Músculo Esquelético/fisiología , Proteína de la Poliposis Adenomatosa del Colon/genética , Células Madre Adultas/citología , Animales , Ciclo Celular/genética , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proliferación Celular , Supervivencia Celular/genética , Femenino , Masculino , Ratones , Ratones Transgénicos , Desarrollo de Músculos/genética , Desarrollo de Músculos/fisiología , Músculo Esquelético/citología , Interferencia de ARN , ARN Interferente Pequeño/genética , Regeneración/genética , Células Satélite del Músculo Esquelético/citología , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/genética , Cicatrización de Heridas/genética , beta Catenina/metabolismo
17.
Biochim Biophys Acta ; 1853(3): 711-23, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25578398

RESUMEN

The Adenomatous Polyposis Coli (APC) tumor suppressor has been previously implicated in the control of apical-basal polarity; yet, the consequence of APC loss-of-function in epithelial polarization and morphogenesis has not been characterized. To test the hypothesis that APC is required for the establishment of normal epithelial polarity and morphogenesis programs, we generated APC-knockdown epithelial cell lines. APC depletion resulted in loss of polarity and multi-layering on permeable supports, and enlarged, filled spheroids with disrupted polarity in 3D culture. Importantly, these effects of APC knockdown were independent of Wnt/ß-catenin signaling, but were rescued with either full-length or a carboxy (c)-terminal segment of APC. Moreover, we identified a gene expression signature associated with APC knockdown that points to several candidates known to regulate cell-cell and cell-matrix communication. Analysis of epithelial tissues from mice and humans carrying heterozygous APC mutations further supports the importance of APC as a regulator of epithelial behavior and tissue architecture. These data also suggest that the initiation of epithelial-derived tumors as a result of APC mutation or gene silencing may be driven by loss of polarity and dysmorphogenesis.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Polaridad Celular/genética , Células Epiteliales/fisiología , Morfogénesis/genética , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Técnicas de Cultivo de Célula , Células Cultivadas , Perros , Técnicas de Silenciamiento del Gen , Genes Supresores de Tumor/fisiología , Células HEK293 , Humanos , Ratones , Mutación/fisiología
18.
J Steroid Biochem Mol Biol ; 148: 103-10, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25597951

RESUMEN

In colon cancer, adenomatous polyposis coli (APC) inactivating gene mutations increase nuclear ß-catenin levels and stimulate proliferation. In vitro, 1,25 dihydroxyvitamin D (1,25(OH)2D), suppresses ß-catenin-mediated gene transcription by inducing vitamin D receptor (VDR)-ß-catenin interactions. We examined whether acute treatment with 1,25(OH)2D could suppress ß-catenin-mediated gene transcription in the hyperplastic colonic lesions of mice with colon-specific deletion of both APC gene alleles (CAC; APC(Δ580/Δ580)). At four weeks of age, CAC; APC(Δ580/Δ580) and control mice were injected with vehicle or 1,25(OH)2D (1µg/kg body weight) once a day for three days and then killed six hours after the last injection. mRNA levels of ß-catenin target genes were elevated in the colon of CAC; APC(Δ580/Δ580) mice. 1,25(OH)2D increased 25 hydroxyvitamin D-24 hydroxylase mRNA levels in the colon of CAC; APC(Δ580/Δ580) and control mice indicating the treatments activated the VDR. However, 1,25(OH)2D had no effect on either ß-catenin target gene mRNA levels or the proliferation index in CAC; APC(Δ580/Δ580) or control mice. VDR mRNA and protein levels were lower (-65% and -90%) in the colon of CAC; APC(Δ580/Δ580) mice compared to control mice, suggesting loss of colon responsiveness to vitamin D. Consistent with this, vitamin D-induced expression of transient receptor potential cation channel, subfamily V, member 6 mRNA was reduced in the colon of CAC; APC(Δ580/Δ580) mice. Our data show that short term exposure to 1,25(OH)2D does not suppress colonic ß-catenin signaling in vivo. This article is part of a special issue entitled '17th Vitamin D Workshop'.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Calcitriol/farmacología , Colon/metabolismo , Enfermedades del Colon/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , ARN Mensajero/genética , Receptores de Calcitriol/genética , Vitamina D3 24-Hidroxilasa/genética , beta Catenina/genética , Animales , Colon/efectos de los fármacos , Colon/patología , Enfermedades del Colon/tratamiento farmacológico , Enfermedades del Colon/patología , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Vitaminas/farmacología
19.
Carcinogenesis ; 35(12): 2778-86, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25280562

RESUMEN

Sporadic and non-hereditary mutations account for the majority of colorectal cancers (CRC). After the loss of adenomatous polyposis coli (APC) function and activation of the ß-catenin/LEF signaling pathway, activating mutations in Kras are major drivers of sporadic CRC. Preventing the outgrowth of cells that develop sporadic mutations will decrease CRC. Resveratrol, a naturally occurring polyphenolic compound has anti-inflammatory, anti-oxidant and anti-cancer activities. We used a genetically engineered mouse model for sporadic CRC where the APC locus is knocked out and Kras is activated specifically in the distal colon to determine the effects of resveratrol on preventing and treating CRC. Feeding mice a diet supplemented with 150 or 300 ppm resveratrol (105 and 210mg daily human equivalent dose, respectively) before tumors were visible by colonoscopy resulted in a 60% inhibition of tumor production. In the 40% of mice that did develop tumors Kras expression was lost in the tumors. In a therapeutic assay where tumors were allowed to develop prior to treatment, feeding tumor bearing mice with resveratrol resulted in a complete remission in 33% of the mice and a 97% decrease in tumor size in the remaining mice. Analysis of miRNA expression in non-tumoral and tumoral colonic tissue of resveratrol treated mice showed an increased expression of miR-96, a miRNA previously shown to regulate Kras translation. These data indicate that resveratrol can prevent the formation and growth of colorectal tumors by downregulating Kras expression.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Anticarcinógenos/uso terapéutico , Transformación Celular Neoplásica/efectos de los fármacos , Neoplasias Colorrectales/prevención & control , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Estilbenos/uso terapéutico , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Noqueados , Mutación/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Resveratrol , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
20.
J Cell Sci ; 127(Pt 12): 2771-81, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24762815

RESUMEN

The APC tumor suppressor regulates diverse stem cell processes including gene regulation through Wnt-ß-catenin signaling and chromosome stability through microtubule interactions, but how the disparate functions of APC are controlled is not well understood. Acting as part of a Wnt-ß-catenin pathway that controls asymmetric cell division, Caenorhabditis elegans APC, APR-1, promotes asymmetric nuclear export of the ß-catenin WRM-1 by asymmetrically stabilizing microtubules. Wnt function also depends on a second ß-catenin, SYS-1, which binds to the C. elegans TCF POP-1 to activate gene expression. Here, we show that APR-1 regulates SYS-1 levels in asymmetric stem cell division, in addition to its known role in lowering nuclear levels of WRM-1. We demonstrate that SYS-1 is also negatively regulated by the C. elegans homolog of casein kinase 1α (CKIα), KIN-19. We show that KIN-19 restricts APR-1 localization, thereby regulating nuclear WRM-1. Finally, the polarity of APR-1 cortical localization is controlled by PRY-1 (C. elegans Axin), such that PRY-1 controls the polarity of both SYS-1 and WRM-1 asymmetries. We propose a model whereby Wnt signaling, through CKIα, regulates the function of two distinct pools of APC - one APC pool negatively regulates SYS-1, whereas the second pool stabilizes microtubules and promotes WRM-1 nuclear export.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/fisiología , Caenorhabditis elegans/citología , Proteínas del Citoesqueleto/metabolismo , Proteínas Quinasas/metabolismo , Factores de Transcripción/metabolismo , Transporte Activo de Núcleo Celular , Animales , División Celular Asimétrica , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Núcleo Celular/fisiología , Polaridad Celular , Proteínas del Citoesqueleto/genética , Microtúbulos/metabolismo , Transporte de Proteínas , Células Madre/fisiología , Factores de Transcripción/genética , Vía de Señalización Wnt
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...