Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters

Publication year range
1.
BMC Cancer ; 18(1): 797, 2018 Aug 07.
Article in English | MEDLINE | ID: mdl-30086728

ABSTRACT

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.


Subject(s)
Pancreatic Neoplasms , Sterol Esterase , Animals , Female , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Transgenic , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Sterol Esterase/deficiency , Sterol Esterase/genetics , Sterol Esterase/metabolism
2.
Am J Physiol Gastrointest Liver Physiol ; 311(4): G675-G687, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27609771

ABSTRACT

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.


Subject(s)
Cell Proliferation/drug effects , Diabetes Mellitus, Type 2/pathology , Fibrosis/pathology , Glucose/pharmacology , Insulin/pharmacology , Pancreatic Stellate Cells/drug effects , Animals , Cells, Cultured , Collagen/metabolism , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat , Female , Fibrosis/metabolism , Humans , Mice , Middle Aged , Pancreas, Exocrine/metabolism , Pancreas, Exocrine/pathology , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , Phosphorylation/drug effects , Receptor, IGF Type 1/metabolism
3.
Am J Physiol Cell Physiol ; 309(10): C639-49, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26310818

ABSTRACT

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.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Dinoprostone/pharmacology , Multiprotein Complexes/metabolism , Receptors, Prostaglandin E, EP1 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , TOR Serine-Threonine Kinases/metabolism , Calcium/metabolism , Cell Line, Tumor , Cyclic AMP/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Dinoprostone/metabolism , Gene Expression Regulation/physiology , Humans , Interleukin-23 Subunit p19/genetics , Interleukin-23 Subunit p19/metabolism , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/genetics , Receptors, Prostaglandin E, EP1 Subtype/genetics , Receptors, Prostaglandin E, EP2 Subtype/genetics , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/genetics , TOR Serine-Threonine Kinases/genetics
4.
J Biol Chem ; 288(37): 26888-97, 2013 Sep 13.
Article in English | MEDLINE | ID: mdl-23880764

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor/metabolism , Exosomes/metabolism , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/metabolism , Saliva/metabolism , Acetylcholine/metabolism , Animals , Cell Line, Tumor , Esterases/metabolism , Humans , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Pancreas/metabolism , Transcriptome
5.
Biochim Biophys Acta ; 1823(2): 593-604, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22227579

ABSTRACT

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.


Subject(s)
Apigenin/metabolism , Mutation , Pancreatic Neoplasms/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Apigenin/pharmacology , Apigenin/therapeutic use , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Benzothiazoles/metabolism , Cell Line, Tumor , Dietary Supplements , Humans , Male , Mice , Mice, Nude , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/therapy , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Toluene/analogs & derivatives , Toluene/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , bcl-X Protein/genetics , bcl-X Protein/metabolism
6.
Biochem Biophys Res Commun ; 439(1): 6-11, 2013 Sep 13.
Article in English | MEDLINE | ID: mdl-23973710

ABSTRACT

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.


Subject(s)
Cyclooxygenase 2/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Pancreatic Neoplasms/metabolism , RNA Stability , Butadienes/pharmacology , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins/metabolism , Dinoprostone/metabolism , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , MAP Kinase Kinase Kinases/metabolism , Nitriles/pharmacology , Pancreatic Neoplasms/genetics , RNA, Messenger/metabolism , Signal Transduction , Time Factors , Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Nutr Cancer ; 65(8): 1232-44, 2013.
Article in English | MEDLINE | ID: mdl-24127740

ABSTRACT

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.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Benzoquinones/pharmacology , Ellagic Acid/pharmacology , Pancreatic Neoplasms/metabolism , Pancreatic Stellate Cells/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Humans , Male , Mice , Mice, Nude , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation , Phytochemicals/pharmacology , Plant Extracts/pharmacology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Pancreatic Neoplasms
8.
Prostate ; 72(10): 1052-9, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22314496

ABSTRACT

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.


Subject(s)
Diet, High-Fat/adverse effects , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Neoplasms/metabolism , Retinoid X Receptor alpha/deficiency , Animals , Male , Mice , Mice, Knockout , Prostatic Intraepithelial Neoplasia/etiology , Prostatic Intraepithelial Neoplasia/genetics , Prostatic Neoplasms/etiology , Prostatic Neoplasms/genetics , Random Allocation , Retinoid X Receptor alpha/genetics
9.
Am J Pathol ; 178(3): 1340-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21356384

ABSTRACT

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.


Subject(s)
Chemokine CXCL5/metabolism , Pancreatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Chemokine CXCL5/genetics , Disease Progression , Endothelial Cells/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/metabolism , Neutralization Tests , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interleukin-8B/metabolism , Survival Analysis , Xenograft Model Antitumor Assays
10.
Prostate ; 70(14): 1547-54, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20687227

ABSTRACT

BACKGROUND: Dietary lycopene combined with other constituents from whole tomatoes was previously found to have greater chemopreventive effects against prostate cancer as compared to pure lycopene provided in a beadlet formulation. We hypothesized that tomato paste would have greater chemopreventive effects in transgenic adenocarcinoma of the mouse prostate (TRAMP) mice relative to equivalent lycopene doses provided from lycopene beadlets. METHODS: Fifty-nine TRAMP mice were randomized to a control diet or to diets providing 28 mg lycopene per kg diet from tomato paste (TP) or from lycopene beadlet (LB), and sacrificed at 20 weeks. Prostate histopathology, prostate weight and serum levels of IGF-I and IGF binding protein-3 were evaluated. RESULTS: The incidence of prostate cancer was significantly decreased in the LB group relative to the control group (60% vs. 95%, respectively, P = 0.0197) whereas the difference between the TP and control groups was not statistically significant (80% vs. 95%, P = 0.34). There was no difference in prostate weights between the groups. Total lycopene levels in the serum and prostate tissue were similarly elevated in the LB and TP groups relative to the control group. The ratio of 5-cis-lycopene to trans-lycopene in the serum was significantly greater in the LB group relative to the TP group (P = 0.0001). Oxidative DNA damage was significantly reduced in the livers of mice fed LB and TP diets relative to the control group. CONCLUSIONS: This preclinical trial suggests significant chemopreventive activity with a lycopene beadlet-enriched diet. The chemopreventive effects of lycopene from beadlets versus whole tomato products requires further testing in preclinical and clinical models of prostate cancer.


Subject(s)
Animal Feed , Anticarcinogenic Agents/therapeutic use , Carotenoids/therapeutic use , Prostatic Neoplasms/prevention & control , Animals , Carotenoids/administration & dosage , Carotenoids/blood , DNA Damage , Diet , Disease Models, Animal , Humans , Insulin-Like Growth Factor Binding Protein 3/blood , Insulin-Like Growth Factor I/metabolism , Lycopene , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Size , Prostate/anatomy & histology , Random Allocation , Vitamin E/administration & dosage , Vitamin E/blood , Vitamin E/therapeutic use , Vitamins/administration & dosage , Vitamins/therapeutic use
11.
Mol Cancer Ther ; 7(9): 2662-71, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18790748

ABSTRACT

Constitutive nuclear factor-kappaB (NF-kappaB) activation is observed in androgen-independent prostate cancer and represents a predictor for biochemical recurrence after radical prostatectomy. Dietary agents such as pomegranate extract (PE) have received increasing attention as potential agents to prevent the onset or progression of many malignancies, including prostate cancer. Here, we show that PE inhibited NF-kappaB and cell viability of prostate cancer cell lines in a dose-dependent fashion in vitro. Importantly, maximal PE-induced apoptosis was dependent on PE-mediated NF-kappaB blockade. In the LAPC4 xenograft model, PE delayed the emergence of LAPC4 androgen-independent xenografts in castrated mice through an inhibition of proliferation and induction of apoptosis. Moreover, the observed increase in NF-kappaB activity during the transition from androgen dependence to androgen independence in the LAPC4 xenograft model was abrogated by PE. Our study represents the first description of PE as a promising dietary agent for the prevention of the emergence of androgen independence that is driven in part by heightened NF-kappaB activity.


Subject(s)
Lythraceae/metabolism , NF-kappa B/metabolism , Phytotherapy , Plant Extracts/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Androgens/metabolism , Animals , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Male , Mice , NF-kappa B/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Xenograft Model Antitumor Assays
12.
Int J Oncol ; 32(2): 475-80, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18202771

ABSTRACT

Angiogenesis is critical to tumor growth and is stimulated by tissue hypoxia due to poor oxygen delivery. In turn, cellular hypoxia leads to angiogenesis via the induction of hypoxia-inducible factor-1alpha (HIF-1alpha) and vascular endothelial growth factor (VEGF) at a cellular level. Pomegranate juice and extracts, which are rich sources of ellagitannins, have been shown to have chemopreventive potential against prostate cancer, but there have been no studies on the effects of an ellagitannin-rich pomegranate extract on angiogenesis. Human prostate cancer cells (LNCaP) and human umbilical vein endothelial cells (HUVEC) were incubated with a pomegranate extract standardized to ellagitannin content (POMx), under normoxic and hypoxic conditions in vitro. Human prostate cancer cells (LAPC4) were injected subcutaneously into severe combined immunodeficient (SCID) mice and the effects of oral administration of POMx on tumor growth, microvessel density, and HIF-1alpha and VEGF expression were determined after 4 weeks of treatment. POMx inhibited the proliferation of LNCaP and HUVEC cells significantly under both normoxic and hypoxic conditions. HIF-1alpha and VEGF protein levels were also reduced by POMx under hypoxic conditions. POMx decreased prostate cancer xenograft size, tumor vessel density, VEGF peptide levels and HIF-1alpha expression after 4 weeks of treatment in SCID mice. These results demonstrate that an ellagitannin-rich pomegranate extract can inhibit tumor-associated angiogenesis as one of several potential mechanisms for slowing the growth of prostate cancer in chemopreventive applications. Further studies in humans are needed to confirm that angiogenesis can be inhibited by an ellagitannin-rich pomegranate extract administered orally as a dietary supplement.


Subject(s)
Gene Expression Regulation, Neoplastic , Hydrolyzable Tannins/metabolism , Lythraceae/metabolism , Neovascularization, Pathologic , Plant Extracts/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Administration, Oral , Animals , Cell Line, Tumor , Humans , Hypoxia , In Vitro Techniques , Male , Mice , Mice, SCID , Neoplasm Transplantation
13.
Sci Rep ; 8(1): 5899, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29651002

ABSTRACT

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.


Subject(s)
Carcinogenesis/drug effects , Carcinoma, Pancreatic Ductal/prevention & control , Fatty Liver/prevention & control , Hyperinsulinism/prevention & control , Metformin/pharmacology , Obesity/prevention & control , Pancreatic Neoplasms/prevention & control , Acyltransferases , Administration, Oral , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Pancreatic Ductal/etiology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Chemoprevention/methods , Diet, High-Fat/adverse effects , Disease Models, Animal , Drinking Water , Fatty Liver/etiology , Fatty Liver/genetics , Fatty Liver/pathology , Female , Gene Expression Regulation , Humans , Hyperinsulinism/etiology , Hyperinsulinism/genetics , Hyperinsulinism/pathology , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Obesity/etiology , Obesity/genetics , Obesity/pathology , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/deficiency , Proto-Oncogene Proteins p21(ras)/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Weight Gain/drug effects
14.
J Agric Food Chem ; 55(19): 7732-7, 2007 Sep 19.
Article in English | MEDLINE | ID: mdl-17722872

ABSTRACT

Our group has shown in a phase II clinical trial that pomegranate juice (PJ) increases prostate specific antigen (PSA) doubling time in prostate cancer (CaP) patients with a rising PSA. Ellagitannins (ETs) are the most abundant polyphenols present in PJ and contribute greatly towards its reported biological properties. On consumption, ETs hydrolyze to release ellagic acid (EA), which is then converted by gut microflora to 3,8-dihydroxy-6H-dibenzo[b, d]pyran-6-one (urolithin A, UA) derivatives. Despite the accumulating knowledge of ET metabolism in animals and humans, there is no available data on the pharmacokinetics and tissue disposition of urolithins. Using a standardized ET-enriched pomegranate extract (PE), we sought to further define the metabolism and tissue distribution of ET metabolites. PE and UA (synthesized in our laboratory) were administered to C57BL/6 wild-type male mice, and metabolite levels in plasma and tissues were determined over 24 h. ET metabolites were concentrated at higher levels in mouse prostate, colon, and intestinal tissues as compared to other tissues after administration of PE or UA. We also evaluated the effects of PE on CaP growth in severe combined immunodeficient (SCID) mice injected subcutaneously with human CaP cells (LAPC-4). PE significantly inhibited LAPC-4 xenograft growth in SCID mice as compared to vehicle control. Finally, EA and several synthesized urolithins were shown to inhibit the growth of human CaP cells in vitro. The chemopreventive potential of pomegranate ETs and localization of their bioactive metabolites in mouse prostate tissue suggest that pomegranate may play a role in CaP treatment and chemoprevention. This warrants future human tissue bioavailability studies and further clinical studies in men with CaP.


Subject(s)
Fruit/chemistry , Hydrolyzable Tannins/metabolism , Hydrolyzable Tannins/pharmacology , Lythraceae/chemistry , Prostate/metabolism , Prostatic Neoplasms/pathology , Animals , Cell Division/drug effects , Coumarins/metabolism , Coumarins/pharmacology , Humans , Hydrolyzable Tannins/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Neoplasm Transplantation , Plant Extracts/administration & dosage , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/prevention & control
15.
PLoS One ; 12(9): e0184455, 2017.
Article in English | MEDLINE | ID: mdl-28886117

ABSTRACT

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.


Subject(s)
Diet, High-Fat/adverse effects , Energy Intake , Mutation , Pancreatic Neoplasms/etiology , Proto-Oncogene Proteins p21(ras)/genetics , Amino Acid Substitution , Animals , Autophagy/genetics , Body Weight , Codon , Computational Biology/methods , Disease Models, Animal , Exome , Extracellular Matrix/metabolism , Female , Fibrosis , Genetic Variation , High-Throughput Nucleotide Sequencing , Inflammation/etiology , Inflammation/pathology , Male , Mice , Pancreatic Neoplasms/pathology
16.
Pancreas ; 45(3): 458-65, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26495779

ABSTRACT

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.


Subject(s)
Inflammation/genetics , Intra-Abdominal Fat/metabolism , Obesity/genetics , Pancreas/metabolism , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Cytokines/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Humans , Inflammation/metabolism , Intra-Abdominal Fat/pathology , Mice, Knockout , Mice, Transgenic , Obesity/etiology , Obesity/metabolism , Pancreas/pathology , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/metabolism
17.
Surgery ; 158(6): 1564-72, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26297056

ABSTRACT

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.


Subject(s)
Adenocarcinoma/metabolism , Cadherins/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Gene Expression Regulation, Neoplastic/physiology , Obesity/complications , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Cadherins/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cells, Cultured , Diet, High-Fat/adverse effects , Disease Models, Animal , Energy Intake , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic/genetics , In Vitro Techniques , Mice , Mice, Mutant Strains , Mutation/genetics , Neoplasm Staging , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Pancreas ; 42(5): 781-5, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23462329

ABSTRACT

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.


Subject(s)
Metformin/pharmacology , Pancreatic Neoplasms/drug therapy , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Administration, Oral , Animals , Blotting, Western , Body Weight/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Injections, Intraperitoneal , Male , Metformin/administration & dosage , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Pancreatic Neoplasms/pathology , Phosphorylation/drug effects , Ribosomal Protein S6 Kinases/metabolism , Time Factors
19.
J Chromatogr Sep Tech ; 4(1)2013 Jan 01.
Article in English | MEDLINE | ID: mdl-24482742

ABSTRACT

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.

20.
Pancreas ; 42(2): 223-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23000892

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Pancreatic Neoplasms/drug therapy , Quercetin/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Dose-Response Relationship, Drug , Humans , Luciferases, Firefly/biosynthesis , Luciferases, Firefly/genetics , Luminescent Measurements , Mice , Mice, Nude , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Quercetin/administration & dosage , Time Factors , Transduction, Genetic , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL