Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
BMC Cancer ; 18(1): 797, 2018 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-30086728

RESUMEN

BACKGROUND: Hormone sensitive lipase (HSL) is a neutral lipase that preferentially catalyzes the hydrolysis of diacylglycerol contributing to triacylglycerol breakdown in the adipose tissue. HSL has been implicated to play a role in tumor cachexia, a debilitating syndrome characterized by progressive loss of adipose tissue. Consequently, pharmacological inhibitors of HSL have been proposed for the treatment of cancer-associated cachexia. In the present study we used the conditional KrasG12D (KC) mouse model of pancreatic ductal adenocarcinoma (PDAC) with a deficiency in HSL to determine the impact of HSL suppression on the development of PDAC. METHODS: KC;Hsl+/+ and KC;Hsl-/- mice were fed standard rodent chow for 20 weeks. At sacrifice, the incidence of PDAC was determined and inflammation in the mesenteric adipose tissue and pancreas was assessed histologically and by immunofluorescence. To determine statistical significance, ANOVA and two-tailed Student's t-tests were performed. To compare PDAC incidence, a two-sided Fisher's exact test was used. RESULTS: Compared to KC;Hsl+/+ mice, KC;Hsl-/- mice gained similar weight and displayed adipose tissue and pancreatic inflammation. In addition, KC;Hsl-/- mice had reduced levels of plasma insulin and leptin. Importantly, the increased adipose tissue and pancreatic inflammation was associated with a significant increase in PDAC incidence in KC;Hsl-/- mice. CONCLUSIONS: HSL deficiency is associated with adipose tissue and pancreatic inflammation and accelerates PDAC development in the KC mouse model.


Asunto(s)
Neoplasias Pancreáticas , Esterol Esterasa , Animales , Femenino , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Transgénicos , Páncreas/metabolismo , Páncreas/patología , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Esterol Esterasa/deficiencia , Esterol Esterasa/genética , Esterol Esterasa/metabolismo
2.
Sci Rep ; 8(1): 5899, 2018 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-29651002

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a particularly deadly disease. Chronic conditions, including obesity and type-2 diabetes are risk factors, thus making PDAC amenable to preventive strategies. We aimed to characterize the chemo-preventive effects of metformin, a widely used anti-diabetic drug, on PDAC development using the KrasG12D mouse model subjected to a diet high in fats and calories (HFCD). LSL-KrasG12D/+;p48-Cre (KC) mice were given control diet (CD), HFCD, or HFCD with 5 mg/ml metformin in drinking water for 3 or 9 months. After 3 months, metformin prevented HFCD-induced weight gain, hepatic steatosis, depletion of intact acini, formation of advanced PanIN lesions, and stimulation of ERK and mTORC1 in pancreas. In addition to reversing hepatic and pancreatic histopathology, metformin normalized HFCD-induced hyperinsulinemia and hyperleptinemia among the 9-month cohort. Importantly, the HFCD-increased PDAC incidence was completely abrogated by metformin (p < 0.01). The obesogenic diet also induced a marked increase in the expression of TAZ in pancreas, an effect abrogated by metformin. In conclusion, administration of metformin improved the metabolic profile and eliminated the promoting effects of diet-induced obesity on PDAC formation in KC mice. Given the established safety profile of metformin, our findings have a strong translational potential for novel chemo-preventive strategies for PDAC.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Carcinoma Ductal Pancreático/prevención & control , Hígado Graso/prevención & control , Hiperinsulinismo/prevención & control , Metformina/farmacología , Obesidad/prevención & control , Neoplasias Pancreáticas/prevención & control , Aciltransferasas , Administración Oral , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Carcinoma Ductal Pancreático/etiología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Quimioprevención/métodos , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Agua Potable , Hígado Graso/etiología , Hígado Graso/genética , Hígado Graso/patología , Femenino , Regulación de la Expresión Génica , Humanos , Hiperinsulinismo/etiología , Hiperinsulinismo/genética , Hiperinsulinismo/patología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Transgénicos , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Obesidad/etiología , Obesidad/genética , Obesidad/patología , Neoplasias Pancreáticas/etiología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/deficiencia , Proteínas Proto-Oncogénicas p21(ras)/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Aumento de Peso/efectos de los fármacos
3.
PLoS One ; 12(9): e0184455, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28886117

RESUMEN

Epidemiologic data has linked obesity to a higher risk of pancreatic cancer, but the underlying mechanisms are poorly understood. To allow for detailed mechanistic studies in a relevant model mimicking diet-induced obesity and pancreatic cancer, a high-fat, high-calorie diet (HFCD) was given to P48+/Cre;LSL-KRASG12D (KC) mice carrying a pancreas-specific oncogenic Kras mutation. The mice were randomly allocated to a HFCD or control diet (CD). Cohorts were sacrificed at 3, 6, and 9 months and tissues were harvested for further analysis. Compared to CD-fed mice, HFCD-fed animals gained significantly more weight. Importantly, the cancer incidence was remarkably increased in HFCD-fed KC mice, particularly in male KC mice. In addition, KC mice fed the HFCD showed more extensive inflammation and fibrosis, and more advanced PanIN lesions in the pancreas, compared to age-matched CD-fed animals. Interestingly, we found that the HFCD reduced autophagic flux in PanIN lesions in KC mice. Further, exome sequencing of isolated murine PanIN lesions identified numerous genetic variants unique to the HFCD. These data underscore the role of sustained inflammation and dysregulated autophagy in diet-induced pancreatic cancer development and suggest that diet-induced genetic alterations may contribute to this process. Our findings provide a better understanding of the mechanisms underlying the obesity-cancer link in males and females, and will facilitate the development of interventions targeting obesity-associated pancreatic cancer.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Ingestión de Energía , Mutación , Neoplasias Pancreáticas/etiología , Proteínas Proto-Oncogénicas p21(ras)/genética , Sustitución de Aminoácidos , Animales , Autofagia/genética , Peso Corporal , Codón , Biología Computacional/métodos , Modelos Animales de Enfermedad , Exoma , Matriz Extracelular/metabolismo , Femenino , Fibrosis , Variación Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Inflamación/etiología , Inflamación/patología , Masculino , Ratones , Neoplasias Pancreáticas/patología
4.
Am J Physiol Gastrointest Liver Physiol ; 311(4): G675-G687, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27609771

RESUMEN

Epidemiological studies support strong links between obesity, diabetes, and pancreatic disorders including pancreatitis and pancreatic adenocarcinoma (PDAC). Type 2 diabetes (T2DM) is associated with insulin resistance, hyperglycemia, and hyperinsulinemia, the latter due to increased insulin secretion by pancreatic beta-cells. We reported that high-fat diet-induced PDAC progression in mice is associated with hyperglycemia, hyperinsulinemia, and activation of pancreatic stellate cells (PaSC). We investigated here the effects of high concentrations of insulin and glucose on mouse and human PaSC growth and fibrosing responses. We found that compared with normal, pancreata from T2DM patients displayed extensive collagen deposition and activated PaSC in islet and peri-islet exocrine pancreas. Mice fed a high-fat diet for up to 12 mo similarly displayed increasing peri-islet fibrosis compared with mice fed control diet. Both quiescent and activated PaSC coexpress insulin (IR; mainly A type) and IGF (IGF-1R) receptors, and both insulin and glucose modulate receptor expression. In cultured PaSC, insulin induced rapid tyrosine autophosphorylation of IR/IGF-1R at specific kinase domain activation loop sites, activated Akt/mTOR/p70S6K signaling, and inactivated FoxO1, a transcription factor that restrains cell growth. Insulin did not promote activation of quiescent PaSC in either 5 mM or 25 mM glucose containing media. However, in activated PaSC, insulin enhanced cell proliferation and augmented production of extracellular matrix proteins, and these effects were abolished by specific inhibition of mTORC1 and mTORC2. In conclusion, our data support the concept that increased local glucose and insulin concentrations associated with obesity and T2DM promote PaSC growth and fibrosing responses.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Diabetes Mellitus Tipo 2/patología , Fibrosis/patología , Glucosa/farmacología , Insulina/farmacología , Células Estrelladas Pancreáticas/efectos de los fármacos , Animales , Células Cultivadas , Colágeno/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa , Femenino , Fibrosis/metabolismo , Humanos , Ratones , Persona de Mediana Edad , Páncreas Exocrino/metabolismo , Páncreas Exocrino/patología , Células Estrelladas Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/patología , Fosforilación/efectos de los fármacos , Receptor IGF Tipo 1/metabolismo
5.
Pancreas ; 45(3): 458-65, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26495779

RESUMEN

OBJECTIVES: Obesity increases the incidence of multiple types of cancer. Our previous work has shown that a high-fat, high-calorie diet (HFCD) leads to visceral obesity, pancreatic inflammation, and accelerated pancreatic neoplasia in KrasG12D (KC) mice. In this study, we aimed to investigate the effects of an HFCD on visceral adipose inflammation with emphasis on potential differences between distinct visceral adipose depots. METHODS: We examined the weight and visceral obesity in both wild-type and KC mice on either control diet (CD) or HFCD. After 3 months, mice were killed for histological examination. Multiplex assays were also performed to obtain cytokine profiles between different adipose depots. RESULTS: Both wild-type and KC mice on an HFCD exhibited significantly increased inflammation in the visceral adipose tissue, particularly in the peripancreatic fat (PPF), compared with animals on a CD. This was associated with significantly increased inflammation in the pancreas. Cytokine profiles were different between visceral adipose depots and between mice on the HFCD and CD. CONCLUSIONS: Our results clearly demonstrate that an HFCD leads to obesity and inflammation in the visceral adipose tissue, particularly the PPF. These data suggest that obesity-associated inflammation in PPF may accelerate pancreatic neoplasia in KC mice.


Asunto(s)
Inflamación/genética , Grasa Intraabdominal/metabolismo , Obesidad/genética , Páncreas/metabolismo , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Citocinas/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Humanos , Inflamación/metabolismo , Grasa Intraabdominal/patología , Ratones Noqueados , Ratones Transgénicos , Obesidad/etiología , Obesidad/metabolismo , Páncreas/patología , Neoplasias Pancreáticas/etiología , Neoplasias Pancreáticas/metabolismo
6.
Am J Physiol Cell Physiol ; 309(10): C639-49, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26310818

RESUMEN

Obesity, a known risk factor for pancreatic cancer, is associated with inflammation and insulin resistance. Proinflammatory prostaglandin E2 (PGE2) and elevated insulin-like growth factor type 1 (IGF-1), related to insulin resistance, are shown to play critical roles in pancreatic cancer progression. We aimed to explore a potential cross talk between PGE2 signaling and the IGF-1/Akt/mammalian target of rapamycin complex 1 (mTORC1) pathway in pancreatic cancer, which may be a key to unraveling the obesity-cancer link. In PANC-1 human pancreatic cancer cells, we showed that PGE2 stimulated mTORC1 activity independently of Akt, as evaluated by downstream signaling events. Subsequently, using pharmacological and genetic approaches, we demonstrated that PGE2-induced mTORC1 activation is mediated by the EP4/cAMP/PKA pathway, as well as an EP1/Ca(2+)-dependent pathway. The cooperative roles of the two pathways were supported by the maximal inhibition achieved with the combined pharmacological blockade, and the coexistence of highly expressed EP1 (mediating the Ca(2+) response) and EP2 or EP4 (mediating the cAMP/PKA pathway) in PANC-1 cells and in the prostate cancer line PC-3, which also robustly exhibited PGE2-induced mTORC1 activation, as identified from a screen in various cancer cell lines. Importantly, we showed a reinforcing interaction between PGE2 and IGF-1 on mTORC1 signaling, with an increase in IL-23 production as a cellular outcome. Our data reveal a previously unrecognized mechanism of PGE2-stimulated mTORC1 activation mediated by EP4/cAMP/PKA and EP1/Ca(2+) signaling, which may be of great importance in elucidating the promoting effects of obesity in pancreatic cancer. Ultimately, a precise understanding of these molecular links may provide novel targets for efficacious interventions devoid of adverse effects.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Dinoprostona/farmacología , Complejos Multiproteicos/metabolismo , Subtipo EP1 de Receptores de Prostaglandina E/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Calcio/metabolismo , Línea Celular Tumoral , AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Dinoprostona/metabolismo , Regulación de la Expresión Génica/fisiología , Humanos , Subunidad p19 de la Interleucina-23/genética , Subunidad p19 de la Interleucina-23/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/genética , Subtipo EP1 de Receptores de Prostaglandina E/genética , Subtipo EP2 de Receptores de Prostaglandina E/genética , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/genética , Serina-Treonina Quinasas TOR/genética
7.
Surgery ; 158(6): 1564-72, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26297056

RESUMEN

BACKGROUND: The epithelial-mesenchymal transition (EMT) is critical in the development of invasive epithelial malignancies. EMT is accelerated by inflammation and results in decreased E-cadherin expression. Diet-induced obesity is an inflammatory state that accelerates pancreatic carcinogenesis; its effect on EMT and E-cadherin expression in the development of pancreatic ductal adenocarcinoma is unclear. METHODS: Conditional Kras(G12D) mice were fed a control diet or a high-fat, high-calorie diet for 3 or 9 months (n = 10 each). Immunohistochemistry with anti-E-cadherin antibody was performed. E-cadherin expression was characterized by staining intensity, location, and proportion of positive cells. In vitro expression of E-cadherin and Slug in primary pancreatic intraepithelial neoplasia (PanIN) and cancer cells was determined by Western blot. RESULTS: The HFCD led to increased weight gain in both 3- (15.8 vs 5.6 g, P < .001) and 9-month (19.8 vs 12.9 g, P = .007) mice. No differences in E-cadherin expression among various stages of preinvasive PanIN lesions were found--regardless of age or diet. In invasive cancer, E-cadherin expression was aberrant, with loss of membranous staining and prominent cytoplasmic staining, associated with strong, cytoplasmic expression of ß-catenin. In vitro expression of E-cadherin was greatest in primary PanIN cells, accompanied by absent Slug expression. Cancer cell lines demonstrated significantly decreased E-cadherin expression in the presence of upregulated Slug. CONCLUSION: Despite increased pancreatic inflammation and accelerated carcinogenesis, the high-fat, high-calorie diet did not induce changes in E-cadherin expression in PanIN lesions of all stages. Invasive lesions demonstrated aberrant cytoplasmic E-cadherin staining. Loss of normal membranous localization may reflect a functional loss of E-cadherin.


Asunto(s)
Adenocarcinoma/metabolismo , Cadherinas/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Obesidad/complicaciones , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Cadherinas/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Células Cultivadas , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Ingestión de Energía , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica/genética , Técnicas In Vitro , Ratones , Ratones Mutantes , Mutación/genética , Estadificación de Neoplasias , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Nutr Cancer ; 65(8): 1232-44, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24127740

RESUMEN

Ellagic acid is a polyphenolic phytochemical present in many fruits and nuts with anticancer properties demonstrated in experimental tumor studies. Embelin is a benzoquinone phytochemical isolated from the Japanese herb Ardisiae Japonicae and has been shown to induce apoptosis in cancer cells. We found that ellagic acid and embelin each dose-dependently increased apoptosis and inhibited proliferation in human pancreatic cancer cells, MIA PaCa-2 and HPAF-II cells, and in pancreatic stellate cells, which are progenitors of pancreatic cancer desmoplasia. In each of these cell types, combinations of ellagic acid and embelin at low micromolar concentrations (0.5-3 µM) induced synergistic increases in apoptosis and decreases in proliferation. Ellagic acid decreased NF-κB transcriptional activity, whereas embelin decreased STAT-3 phosphorylation and protein expression of its downstream target survivin in cancer cells. In vivo dietary ellagic acid alone or in combination with embelin decreased tumor size and tumor cellularity in a subcutaneous xenograft mouse model of pancreatic cancer. These results show that ellagic acid and embelin interact with divergent intracellular signaling pathways resulting in augmentation of apoptosis and inhibition of proliferation at low micromolar concentrations for the key cellular components of pancreatic adenocarcinoma.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Benzoquinonas/farmacología , Ácido Elágico/farmacología , Neoplasias Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Humanos , Masculino , Ratones , Ratones Desnudos , FN-kappa B/genética , FN-kappa B/metabolismo , Fosforilación , Fitoquímicos/farmacología , Extractos Vegetales/farmacología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Neoplasias Pancreáticas
9.
Cancer Prev Res (Phila) ; 6(10): 1064-73, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23943783

RESUMEN

There is epidemiologic evidence that obesity increases the risk of cancers. Several underlying mechanisms, including inflammation and insulin resistance, are proposed. However, the driving mechanisms in pancreatic cancer are poorly understood. The goal of the present study was to develop a model of diet-induced obesity and pancreatic cancer development in a state-of-the-art mouse model, which resembles important clinical features of human obesity, for example, weight gain and metabolic disturbances. Offspring of Pdx-1-Cre and LSL-KrasG12D mice were allocated to either a high-fat, high-calorie diet (HFCD; ∼4,535 kcal/kg; 40% of calories from fats) or control diet (∼3,725 kcal/kg; 12% of calories from fats) for 3 months. Compared with control animals, mice fed with the HFCD significantly gained more weight and developed hyperinsulinemia, hyperglycemia, hyperleptinemia, and elevated levels of insulin-like growth factor I (IGF-I). The pancreas of HFCD-fed animals showed robust signs of inflammation with increased numbers of infiltrating inflammatory cells (macrophages and T cells), elevated levels of several cytokines and chemokines, increased stromal fibrosis, and more advanced PanIN lesions. Our results show that a diet high in fats and calories leads to obesity and metabolic disturbances similar to humans and accelerates early pancreatic neoplasia in the conditional KrasG12D mouse model. This model and findings will provide the basis for more robust studies attempting to unravel the mechanisms underlying the cancer-promoting properties of obesity, as well as to evaluate dietary- and chemopreventive strategies targeting obesity-associated pancreatic cancer development.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Dieta Alta en Grasa/efectos adversos , Genes ras , Neoplasias Pancreáticas/genética , Proteínas ras/genética , Proteínas ras/metabolismo , Actinas/metabolismo , Animales , Peso Corporal , Carcinoma Ductal Pancreático/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ingestión de Energía , Femenino , Fibronectinas/metabolismo , Genotipo , Inmunohistoquímica , Inflamación , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Neoplasias Pancreáticas/metabolismo
10.
Biochem Biophys Res Commun ; 439(1): 6-11, 2013 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-23973710

RESUMEN

Small non-coding RNAs, microRNAs (miRNA), inhibit the translation or accelerate the degradation of message RNA (mRNA) by targeting the 3'-untranslated region (3'-UTR) in regulating growth and survival through gene suppression. Deregulated miRNA expression contributes to disease progression in several cancers types, including pancreatic cancers (PaCa). PaCa tissues and cells exhibit decreased miRNA, elevated cyclooxygenase (COX)-2 and increased prostaglandin E2 (PGE2) resulting in increased cancer growth and metastases. Human PaCa cell lines were used to demonstrate that restoration of miRNA-143 (miR-143) regulates COX-2 and inhibits cell proliferation. miR-143 were detected at fold levels of 0.41 ± 0.06 in AsPC-1, 0.20 ± 0.05 in Capan-2 and 0.10 ± 0.02 in MIA PaCa-2. miR-143 was not detected in BxPC-3, HPAF-II and Panc-1 which correlated with elevated mitogen-activated kinase (MAPK) and MAPK kinase (MEK) activation. Treatment with 10 µM of MEK inhibitor U0126 or PD98059 increased miR-143, respectively, by 187 ± 18 and 152 ± 26-fold in BxPC-3 and 182 ± 7 and 136 ± 9-fold in HPAF-II. miR-143 transfection diminished COX-2 mRNA stability at 60 min by 2.6 ± 0.3-fold in BxPC-3 and 2.5 ± 0.2-fold in HPAF-II. COX-2 expression and cellular proliferation in BxPC-3 and HPAF-II inversely correlated with increasing miR-143. PGE2 levels decreased by 39.3 ± 5.0% in BxPC-3 and 48.0 ± 3.0% in HPAF-II transfected with miR-143. Restoration of miR-143 in PaCa cells suppressed of COX-2, PGE2, cellular proliferation and MEK/MAPK activation, implicating this pathway in regulating miR-143 expression.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Neoplasias Pancreáticas/metabolismo , Estabilidad del ARN , Butadienos/farmacología , Línea Celular Tumoral , Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Dinoprostona/metabolismo , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Humanos , Quinasas Quinasa Quinasa PAM/metabolismo , Nitrilos/farmacología , Neoplasias Pancreáticas/genética , ARN Mensajero/metabolismo , Transducción de Señal , Factores de Tiempo , Factores de Transcripción/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
J Biol Chem ; 288(37): 26888-97, 2013 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-23880764

RESUMEN

Recent studies have demonstrated that discriminatory salivary biomarkers can be readily detected upon the development of systemic diseases such as pancreatic cancer, breast cancer, lung cancer, and ovarian cancer. However, the utility of salivary biomarkers for the detection of systemic diseases has been undermined due to the absence of the biological and mechanistic rationale as to why distal diseases from the oral cavity would lead to the development of discriminatory biomarkers in saliva. Here, we examine the hypothesis that pancreatic tumor-derived exosomes are mechanistically involved in the development of pancreatic cancer-discriminatory salivary transcriptomic biomarkers. We first developed a pancreatic cancer mouse model that yielded discriminatory salivary biomarkers by implanting the mouse pancreatic cancer cell line Panc02 into the pancreas of the syngeneic host C57BL/6. The role of pancreatic cancer-derived exosomes in the development of discriminatory salivary biomarkers was then tested by engineering a Panc02 cell line that is suppressed for exosome biogenesis, implanting into the C56BL/6 mouse, and examining whether the discriminatory salivary biomarker profile was ablated or disrupted. Suppression of exosome biogenesis results in the ablation of discriminatory salivary biomarker development. This study supports that tumor-derived exosomes provide a mechanism in the development of discriminatory biomarkers in saliva and distal systemic diseases.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Exosomas/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Pancreáticas/metabolismo , Saliva/metabolismo , Acetilcolina/metabolismo , Animales , Línea Celular Tumoral , Esterasas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Trasplante de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Páncreas/metabolismo , Transcriptoma
12.
Pancreas ; 42(5): 781-5, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23462329

RESUMEN

OBJECTIVE: Pancreatic ductal adenocarcinoma is a devastating disease, with an overall 5-year survival rate of only 3% to 5%. As the current therapies offer very limited survival benefits, novel therapeutic strategies are urgently required to treat this disease. Here, we determined whether metformin administration inhibits the growth of PANC-1 and MiaPaCa-2 tumor xenografts in vivo. METHODS: Different xenograft models, including orthotopic implantation, were used to determine whether intraperitoneal or oral administration of metformin inhibits the growth of pancreatic cancer in vivo. RESULTS: We demonstrate that metformin given once daily intraperitoneally at various doses (50-250 mg/kg) to nude mice inhibited the growth of PANC-1 xenografts in a dose-dependent manner. A significant effect of metformin was obtained at 50 mg/kg and maximal effect at 200 mg/kg. Metformin administration also caused a significant reduction in the phosphorylation of ribosomal S6 protein and ERK in these xenografts. Metformin also inhibited the growth of pancreatic cancer xenografts when administered orally (2.5 mg/mL) either before or after tumor implantation. Importantly, oral administration of metformin also inhibited the growth of MiaPaCa-2 tumors xenografted orthotopically. CONCLUSIONS: The studies presented here provide further evidence indicating that metformin offers a potential novel approach for pancreatic ductal adenocarcinoma prevention and therapy.


Asunto(s)
Metformina/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Administración Oral , Animales , Western Blotting , Peso Corporal/efectos de los fármacos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/farmacología , Inyecciones Intraperitoneales , Masculino , Metformina/administración & dosificación , Ratones , Ratones Desnudos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neoplasias Pancreáticas/patología , Fosforilación/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas/metabolismo , Factores de Tiempo
13.
Pancreas ; 42(2): 223-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23000892

RESUMEN

OBJECTIVES: The flavonoid quercetin holds promise as an antitumor agent in several preclinical animal models. However, the efficacy of oral administration of quercetin in a pancreatic cancer mouse model is unknown. METHODS: The antiproliferative effects of quercetin alone or in combination with gemcitabine were tested in 2 human pancreatic cancer cell lines using cell count and MTT assays. Apoptosis was evaluated by flow cytometry. Tumor growth in vivo was investigated in an orthotopic pancreatic cancer animal model using bioluminescence. Quercetin was administered orally in the diet. RESULTS: Quercetin inhibited the growth of pancreatic cancer cell lines, which was caused by an induction of apoptosis. In addition, dietary supplementation of quercetin attenuated the growth of orthotopically transplanted pancreatic xenografts. The combination of gemcitabine and quercetin had no additional effect compared with quercetin alone. In vivo quercetin caused significant apoptosis and reduced tumor cell proliferation. CONCLUSIONS: Our data provide evidence that oral administration of quercetin was capable of inhibiting growth of orthotopic pancreatic tumors in a nude mouse model. These data suggest a possible benefit of quercetin in patients with pancreatic cancer.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Neoplasias Pancreáticas/tratamiento farmacológico , Quercetina/farmacología , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Relación Dosis-Respuesta a Droga , Humanos , Luciferasas de Luciérnaga/biosíntesis , Luciferasas de Luciérnaga/genética , Mediciones Luminiscentes , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Quercetina/administración & dosificación , Factores de Tiempo , Transducción Genética , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
14.
J Chromatogr Sep Tech ; 4(1)2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24482742

RESUMEN

Rottlerin is a natural polyphenolic ketone isolated from the pericarps of Mallotus phillippinensis. In previous studies we showed that parenteral administration of rottlerin reduced tumor growth in murine xenograft models of pancreatic cancer. The aim of this study was to develop a simple and validated method for the quantitative determination of rottlerin in plasma and tumor tissues of mice fed a rottlerin diet. A xenograft model of pancreatic cancer was prepared by injection of 2×106 HPAF-II cells subcutaneously into nude mice. One week before tumor implantation, mice were randomly allocated to standard diet (AIN76A) and standard diet supplement with 0.012% rottlerin (n=6 per group). Mice were sacrificed after 6 weeks on diets. Rottlerin was extracted from the plasma and tissues using protein precipitation-extraction and analyzed by reverse-phase HPLC-DAD method. The same HPLC method was also applied to determine rottlerin levels in conditioned culture media and in cell lysates from HPAF-II cells exposed to 25 µM concentration of rottlerin. A substantial amount of rottlerin was detected in tumor (2.11 ± 0.25 nmol/g tissue) and plasma (2.88 ± 0.41 µM) in mice fed rottlerin diet. In addition, significant levels of rottlerin (57.4 ± 5.4 nmol/mg protein) were detected in cell lysates from rottlerin-treated HPAF-II cells. These data indicate that rottlerin is efficiently absorbed in cells and tissues both in vivo and in vitro and suggest a strong potential for rottlerin as a preventive or adjuvant supplement for pancreatic cancer.

15.
J Nutr Biochem ; 23(11): 1537-42, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22405694

RESUMEN

It has been demonstrated in various animal models that the oral administration of green tea (GT) extracts in drinking water can inhibit tumor growth, but the effects of brewed GT on factors promoting tumor growth, including oxidant damage of DNA and protein, angiogenesis and DNA methylation, have not been tested in an animal model. To explore these potential mechanisms, brewed GT was administered instead of drinking water to male severe combined immunodeficiency (SCID) mice with androgen-dependent human LAPC4 prostate cancer cell subcutaneous xenografts. Tumor volume was decreased significantly in mice consuming GT, and tumor size was significantly correlated with GT polyphenol (GTP) content in tumor tissue. There was a significant reduction in hypoxia-inducible factor 1-alpha and vascular endothelial growth factor protein expression. GT consumption significantly reduced oxidative DNA and protein damage in tumor tissue as determined by 8-hydroxydeoxyguanosine/deoxyguanosine ratio and protein carbonyl assay, respectively. Methylation is known to inhibit antioxidative enzymes such as glutathione S-transferase pi to permit reactive oxygen species promotion of tumor growth. GT inhibited tumor 5-cytosine DNA methyltransferase 1 mRNA and protein expression significantly, which may contribute to the inhibition of tumor growth by reactivation of antioxidative enzymes. This study advances our understanding of tumor growth inhibition by brewed GT in an animal model by demonstrating tissue localization of GTPs in correlation with inhibition of tumor growth. Our results suggest that the inhibition of tumor growth is due to GTP-mediated inhibition of oxidative stress and angiogenesis in the LAPC4 xenograft prostate tumor in SCID mice.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neovascularización Patológica/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Polifenoles/farmacología , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/tratamiento farmacológico , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Camellia sinensis , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , Desoxiguanosina/análogos & derivados , Desoxiguanosina/análisis , Desoxiguanosina/metabolismo , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/patología , Masculino , Ratones , Ratones SCID , Extractos Vegetales/farmacología , Polifenoles/farmacocinética , Neoplasias de la Próstata/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Prostate ; 72(10): 1052-9, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22314496

RESUMEN

BACKGROUND: The "New Western-style Diet" (NWD) characterized by high in fat and low in fiber, vitamin D, calcium, and methyl donors--are considered as a risk factor for prostate cancer. Previous studies have shown that premalignant lesions of human prostate have decreased expression of the Retinoid X Receptor alpha (RXRα). This study was to determine the effect of diet in RXRα knockout mice in developing high-grade prostate intraepithelial neoplasia (mPIN). METHODS: Male mice (n = 54) with or without the RXRα prostate null mutation were fed either NWD or AIN-76A control diet for 10 months; prostates were harvested at 11 months of age and examined for prostate mPIN. RESULTS: mPIN was seen in 79% of RXRα prostate null mice fed NWD (n = 19), 30.8% RXRα prostate null mice fed AIN-76A (n = 13), 42.9% RXRα wild-type mice fed NWD (n = 14), and 12.5% RXRα wild-type mice fed AIN-76A (n = 8). Unconditional Logistic analysis showed a significant joint effect of NWD and RXRα status in developing mPIN 26.3 (95% CI: 2.5-280), but interaction was not significant owing to the small sample size 1.6 (0.09-27.7, P = 0.7441). CONCLUSION: This study provides preliminary data to support a joint RXRα-diet effect in prostate carcinogenesis.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Neoplasia Intraepitelial Prostática/metabolismo , Neoplasias de la Próstata/metabolismo , Receptor alfa X Retinoide/deficiencia , Animales , Masculino , Ratones , Ratones Noqueados , Neoplasia Intraepitelial Prostática/etiología , Neoplasia Intraepitelial Prostática/genética , Neoplasias de la Próstata/etiología , Neoplasias de la Próstata/genética , Distribución Aleatoria , Receptor alfa X Retinoide/genética
17.
Biochim Biophys Acta ; 1823(2): 593-604, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22227579

RESUMEN

Pancreatic cancer is an exceedingly lethal disease with a five-year survival that ranks among the lowest of gastrointestinal malignancies. Part of its lethality is attributable to a generally poor response to existing chemotherapeutic regimens. New therapeutic approaches are urgently needed. We aimed to elucidate the anti-neoplastic mechanisms of apigenin-an abundant, naturally-occurring plant flavonoid-with a particular focus on p53 function. Pancreatic cancer cells (BxPC-3, MiaPaCa-2) experienced dose and time-dependent growth inhibition and increased apoptosis with apigenin treatment. p53 post-translational modification, nuclear translocation, DNA binding, and upregulation of p21 and PUMA were all enhanced by apigenin treatment despite mutated p53 in both cell lines. Transcription-dependent p53 activity was reversed by pifithrin-α, a specific DNA binding inhibitor of p53, but not growth inhibition or apoptosis suggesting transcription-independent p53 activity. This was supported by immunoprecipitation assays which demonstrated disassociation of p53/BclXL and PUMA/BclXL and formation of complexes with Bak followed by cytochrome c release. Treated animals grew smaller tumors with increased cellular apoptosis than those fed control diet. These results suggest that despite deactivating mutation, p53 retains some of its function which is augmented following treatment with apigenin. Cell cycle arrest and apoptosis induction may be mediated by transcription-independent p53 function via interactions with BclXL and PUMA. Further study of flavonoids as chemotherapeutics is warranted.


Asunto(s)
Apigenina/metabolismo , Mutación , Neoplasias Pancreáticas/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apigenina/farmacología , Apigenina/uso terapéutico , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Benzotiazoles/metabolismo , Línea Celular Tumoral , Suplementos Dietéticos , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/terapia , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Tolueno/análogos & derivados , Tolueno/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
18.
Pancreas ; 41(4): 571-6, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22158070

RESUMEN

OBJECTIVES: Scutellaria baicalensis has been a subject of research interest due to its potential multiple therapeutic benefits. This study was to examine the distribution of baicalein, wogonin, oroxylin A and their glucuronide/sulfate-conjugated metabolites in plasma, colon, small intestine, lung, liver, pancreas, kidney, and prostate tissues and in pancreatic tumor in a xenograft animal model. In addition, we examined metabolic stability of baicalin in these tissues. METHODS: A mouse xenograft model was prepared by injection of 3 × 10 human pancreatic cancer MiaPaCa-2 cells subcutaneously into nude mice. Mice were randomly allocated to control diet (AIN-76A) and 1% S. baicalensis diet (n = 8 per group) for 13 weeks. Levels of baicalein, wogonin, oroxylin A, and their conjugates in mouse tissues were measured by high-pressure liquid chromatography after enzymatic hydrolysis and then extraction. RESULTS: A substantial amount of baicalin (34%-63%) was methylated to oroxylin A and its conjugates in various organs during absorption. Whereas plasma contained predominantly conjugates of baicalein, wogonin, and oroxylin A, both aglycones and conjugates were found in all other tissues investigated and in tumor. CONCLUSIONS: Substantial accumulation of bioactive metabolites are found in target tissues, suggesting strong potential for S. baicalensis use as a preventive or adjuvant supplement for pancreatic cancer.


Asunto(s)
Flavanonas/metabolismo , Flavonoides/metabolismo , Páncreas/metabolismo , Neoplasias Pancreáticas/metabolismo , Extractos Vegetales/metabolismo , Scutellaria baicalensis/metabolismo , Animales , Disponibilidad Biológica , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Dieta , Flavanonas/farmacocinética , Flavonoides/farmacocinética , Ratones , Ratones Desnudos , Extractos Vegetales/administración & dosificación , Extractos Vegetales/farmacocinética , Raíces de Plantas , Distribución Aleatoria
19.
Am J Pathol ; 178(3): 1340-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21356384

RESUMEN

Epithelial neutrophil-activating peptide-78 (CXCL5), a member of the CXC chemokine family, has been shown to be involved in angiogenesis, tumor growth, and metastasis. The objective of this study was to determine the relationship between CXCL5 expression and tumor progression in human pancreatic cancer and to elucidate the mechanism underlying CXCL5-mediated tumor angiogenesis and cancer growth. We report herein that CXCL5 is overexpressed in human pancreatic cancer compared with paired normal pancreas tissue. Overexpression of CXCL5 is significantly correlated with poorer tumor differentiation, advanced clinical stage, and shorter patient survival. Patients with pancreatic cancer and CXCL5 overexpression who underwent resection of cancer had a mean survival time 25.5 months shorter than that of patients who did not overexpress CXCL5. Blockade of CXCL5 or its receptor CXCR2 by small-interfering RNA knockdown or antibody neutralization attenuated human pancreatic cancer growth in a nude mouse model. Finally, we demonstrated that CXCL5 mediates pancreatic cancer-derived angiogenesis through activation of several signaling pathways, including protein kinase B (Akt), extracellular signal-regulated kinase (ERK), and signal transducer and activator of transcription (STAT) in human endothelial cells. These data suggest that CXCL5 is an important mediator of tumor-derived angiogenesis and that it may serve as a survival factor for pancreatic cancer. Blockade of either CXCL5 or CXCR2 may be a critical adjunct antiangiogenic therapy against pancreatic cancer.


Asunto(s)
Quimiocina CXCL5/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Quimiocina CXCL5/genética , Progresión de la Enfermedad , Células Endoteliales/enzimología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Desnudos , Neovascularización Patológica/metabolismo , Pruebas de Neutralización , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Interleucina-8B/metabolismo , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Cancer Prev Res (Phila) ; 4(4): 608-15, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21278313

RESUMEN

Prostate cancer is a slowly developing but very common cancer in males that may be amenable to preventive strategies that are not toxic. Chinese red yeast rice (RYR), a food herb made by fermenting Monascus purpureus Went yeast on white rice, contains a mixture of eight different monacolins that inhibit cholesterogenesis in addition to red pigments with antioxidant properties. Monacolin K is identical to lovastatin (LV), but LV unlike RYR can be used in individuals intolerant to statins due to muscle pain. Both LV and RYR inhibit de novo cholesterogenesis, which is critical to the growth of tumor cells. Long-term use of statin drugs has been associated with a reduced risk of prostate cancer. We have previously shown that RYR inhibited androgen-dependent and androgen receptor-overexpressing androgen-independent prostate cancer cell proliferation in vitro. This study was designed to determine whether RYR and LV inhibit prostate tumor growth in SCID mice. RYR significantly reduced tumor volumes of androgen-dependent and androgen-independent prostate xenograft tumors compared with animals receiving vehicle alone (P < 0.05). Inhibition by RYR was greater than that observed with LV at the dose found in RYR, showing that other compounds in RYR contributed to the antiproliferative effect. There was a significant correlation of tumor volume to serum cholesterol (P < 0.001). RYR decreased gene expression of androgen synthesizing enzymes (HSD3B2, AKR1C3, and SRD5A1) in both type of tumors (P < 0.05). Clinical studies of RYR for prostate cancer prevention in the increasing population of men undergoing active surveillance should be considered.


Asunto(s)
Antineoplásicos/farmacología , Productos Biológicos/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Colesterol/sangre , Ensayo de Inmunoadsorción Enzimática , Humanos , Lovastatina/farmacología , Masculino , Ratones , Ratones SCID , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA