Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
2.
Nat Cell Biol ; 26(4): 613-627, 2024 Apr.
Article En | MEDLINE | ID: mdl-38429478

The ability of tumour cells to thrive in harsh microenvironments depends on the utilization of nutrients available in the milieu. Here we show that pancreatic cancer-associated fibroblasts (CAFs) regulate tumour cell metabolism through the secretion of acetate, which can be blocked by silencing ATP citrate lyase (ACLY) in CAFs. We further show that acetyl-CoA synthetase short-chain family member 2 (ACSS2) channels the exogenous acetate to regulate the dynamic cancer epigenome and transcriptome, thereby facilitating cancer cell survival in an acidic microenvironment. Comparative H3K27ac ChIP-seq and RNA-seq analyses revealed alterations in polyamine homeostasis through regulation of SAT1 gene expression and enrichment of the SP1-responsive signature. We identified acetate/ACSS2-mediated acetylation of SP1 at the lysine 19 residue that increased SP1 protein stability and transcriptional activity. Genetic or pharmacologic inhibition of the ACSS2-SP1-SAT1 axis diminished the tumour burden in mouse models. These results reveal that the metabolic flexibility imparted by the stroma-derived acetate enabled cancer cell survival under acidosis via the ACSS2-SP1-SAT1 axis.


Cancer-Associated Fibroblasts , Pancreatic Neoplasms , Animals , Mice , Cancer-Associated Fibroblasts/metabolism , Cell Line, Tumor , Acetates/pharmacology , Acetates/metabolism , Pancreatic Neoplasms/genetics , Polyamines , Tumor Microenvironment
3.
Proc Natl Acad Sci U S A ; 121(14): e2315509121, 2024 Apr 02.
Article En | MEDLINE | ID: mdl-38547055

Dysregulation of polyamine metabolism has been implicated in cancer initiation and progression; however, the mechanism of polyamine dysregulation in cancer is not fully understood. In this study, we investigated the role of MUC1, a mucin protein overexpressed in pancreatic cancer, in regulating polyamine metabolism. Utilizing pancreatic cancer patient data, we noted a positive correlation between MUC1 expression and the expression of key polyamine metabolism pathway genes. Functional studies revealed that knockdown of spermidine/spermine N1-acetyltransferase 1 (SAT1), a key enzyme involved in polyamine catabolism, attenuated the oncogenic functions of MUC1, including cell survival and proliferation. We further identified a regulatory axis whereby MUC1 stabilized hypoxia-inducible factor (HIF-1α), leading to increased SAT1 expression, which in turn induced carbon flux into the tricarboxylic acid cycle. MUC1-mediated stabilization of HIF-1α enhanced the promoter occupancy of the latter on SAT1 promoter and corresponding transcriptional activation of SAT1, which could be abrogated by pharmacological inhibition of HIF-1α or CRISPR/Cas9-mediated knockout of HIF1A. MUC1 knockdown caused a significant reduction in the levels of SAT1-generated metabolites, N1-acetylspermidine and N8-acetylspermidine. Given the known role of MUC1 in therapy resistance, we also investigated whether inhibiting SAT1 would enhance the efficacy of FOLFIRINOX chemotherapy. By utilizing organoid and orthotopic pancreatic cancer mouse models, we observed that targeting SAT1 with pentamidine improved the efficacy of FOLFIRINOX, suggesting that the combination may represent a promising therapeutic strategy against pancreatic cancer. This study provides insights into the interplay between MUC1 and polyamine metabolism, offering potential avenues for the development of treatments against pancreatic cancer.


Antineoplastic Combined Chemotherapy Protocols , Pancreatic Neoplasms , Mice , Animals , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Polyamines/metabolism , Signal Transduction , Acetyltransferases/genetics , Acetyltransferases/metabolism , Mucin-1
4.
Cancer Discov ; 14(1): 176-193, 2024 01 12.
Article En | MEDLINE | ID: mdl-37931287

Nutritional factors play crucial roles in immune responses. The tumor-caused nutritional deficiencies are known to affect antitumor immunity. Here, we demonstrate that pancreatic ductal adenocarcinoma (PDAC) cells can suppress NK-cell cytotoxicity by restricting the accessibility of vitamin B6 (VB6). PDAC cells actively consume VB6 to support one-carbon metabolism, and thus tumor cell growth, causing VB6 deprivation in the tumor microenvironment. In comparison, NK cells require VB6 for intracellular glycogen breakdown, which serves as a critical energy source for NK-cell activation. VB6 supplementation in combination with one-carbon metabolism blockage effectively diminishes tumor burden in vivo. Our results expand the understanding of the critical role of micronutrients in regulating cancer progression and antitumor immunity, and open new avenues for developing novel therapeutic strategies against PDAC. SIGNIFICANCE: The nutrient competition among the different tumor microenvironment components drives tumor growth, immune tolerance, and therapeutic resistance. PDAC cells demand a high amount of VB6, thus competitively causing NK-cell dysfunction. Supplying VB6 with blocking VB6-dependent one-carbon metabolism amplifies the NK-cell antitumor immunity and inhibits tumor growth in PDAC models. This article is featured in Selected Articles from This Issue, p. 5.


Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Vitamin B 6 , Tumor Microenvironment , Killer Cells, Natural , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Carbon
5.
Cancer Lett ; 552: 215981, 2023 01 01.
Article En | MEDLINE | ID: mdl-36341997

Inhibitors of dihydroorotate dehydrogenase (DHODH), a key enzyme for de novo synthesis of pyrimidine nucleotides, have failed in clinical trials for various cancers despite robust efficacy in preclinical animal models. To probe for druggable mediators of DHODH inhibitor resistance, we performed a combination screen with a small molecule library against pancreatic cancer cell lines that are highly resistant to the DHODH inhibitor brequinar (BQ). The screen revealed that CNX-774, a preclinical Bruton tyrosine kinase (BTK) inhibitor, sensitizes resistant cell lines to BQ. Mechanistic studies showed that this effect is independent of BTK and instead results from inhibition of equilibrative nucleoside transporter 1 (ENT1) by CNX-774. We show that ENT1 mediates BQ resistance by taking up extracellular uridine, which is salvaged to generate pyrimidine nucleotides in a DHODH-independent manner. In BQ-resistant cell lines, BQ monotherapy slowed proliferation and caused modest pyrimidine nucleotide depletion, whereas combination treatment with BQ and CNX-774 led to profound cell viability loss and pyrimidine starvation. We also identify N-acetylneuraminic acid accumulation as a potential marker of the therapeutic efficacy of DHODH inhibitors. In an aggressive, immunocompetent pancreatic cancer mouse model, combined targeting of DHODH and ENT1 dramatically suppressed tumor growth and prolonged mouse survival. Overall, our study defines CNX-774 as a previously uncharacterized ENT1 inhibitor and provides strong proof of concept support for dual targeting of DHODH and ENT1 in pancreatic cancer.


Oxidoreductases Acting on CH-CH Group Donors , Pancreatic Neoplasms , Mice , Animals , Dihydroorotate Dehydrogenase , Equilibrative Nucleoside Transporter 1/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Pyrimidines/pharmacology , Enzyme Inhibitors/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pyrimidine Nucleotides , Pancreatic Neoplasms
6.
Oncogene ; 41(7): 971-982, 2022 02.
Article En | MEDLINE | ID: mdl-35001076

Metabolic alterations regulate cancer aggressiveness and immune responses. Given the poor response of pancreatic ductal adenocarcinoma (PDAC) to conventional immunotherapies, we investigated the link between metabolic alterations and immunosuppression. Our metabolic enzyme screen indicated that elevated expression of CD73, an ecto-5'-nucleotidase that generates adenosine, correlates with increased aggressiveness. Correspondingly, we observed increased interstitial adenosine levels in tumors from spontaneous PDAC mouse models. Diminishing CD73 by genetic manipulations ablated in vivo tumor growth, and decreased myeloid-derived suppressor cells (MDSC) in orthotopic mouse models of PDAC. A high-throughput cytokine profiling demonstrated decreased GM-CSF in mice implanted with CD73 knockdowns. Furthermore, we noted increased IFN-γ expression by intratumoral CD4+ and CD8+ T cells in pancreatic tumors with CD73 knockdowns. Depletion of CD4+ T cells, but not CD8+ T cells abrogated the beneficial effects of decreased CD73. We also observed that splenic MDSCs from Nt5e knockdown tumor-bearing mice were incompetent in suppressing T cell activation in the ex vivo assays. Replenishing GM-CSF restored tumor growth in Nt5e knockout tumors, which was reverted by MDSC depletion. Finally, anti-CD73 antibody treatment significantly improved gemcitabine efficacy in orthotopic models. Thus, targeting the adenosine axis presents a novel therapeutic opportunity for improving the anti-tumoral immune response against PDAC.


Myeloid-Derived Suppressor Cells
7.
Gastroenterology ; 161(5): 1584-1600, 2021 11.
Article En | MEDLINE | ID: mdl-34245764

BACKGROUND & AIMS: SIRT5 plays pleiotropic roles via post-translational modifications, serving as a tumor suppressor, or an oncogene, in different tumors. However, the role SIRT5 plays in the initiation and progression of pancreatic ductal adenocarcinoma (PDAC) remains unknown. METHODS: Published datasets and tissue arrays with SIRT5 staining were used to investigate the clinical relevance of SIRT5 in PDAC. Furthermore, to define the role of SIRT5 in the carcinogenesis of PDAC, we generated autochthonous mouse models with conditional Sirt5 knockout. Moreover, to examine the mechanistic role of SIRT5 in PDAC carcinogenesis, SIRT5 was knocked down in PDAC cell lines and organoids, followed by metabolomics and proteomics studies. A novel SIRT5 activator was used for therapeutic studies in organoids and patient-derived xenografts. RESULTS: SIRT5 expression negatively regulated tumor cell proliferation and correlated with a favorable prognosis in patients with PDAC. Genetic ablation of Sirt5 in PDAC mouse models promoted acinar-to-ductal metaplasia, precursor lesions, and pancreatic tumorigenesis, resulting in poor survival. Mechanistically, SIRT5 loss enhanced glutamine and glutathione metabolism via acetylation-mediated activation of GOT1. A selective SIRT5 activator, MC3138, phenocopied the effects of SIRT5 overexpression and exhibited antitumor effects on human PDAC cells. MC3138 also diminished nucleotide pools, sensitizing human PDAC cell lines, organoids, and patient-derived xenografts to gemcitabine. CONCLUSIONS: Collectively, we identify SIRT5 as a key tumor suppressor in PDAC, whose loss promotes tumorigenesis through increased noncanonic use of glutamine via GOT1, and that SIRT5 activation is a novel therapeutic strategy to target PDAC.


Carcinoma, Pancreatic Ductal/enzymology , Energy Metabolism , Pancreatic Neoplasms/enzymology , Proto-Oncogene Proteins p21(ras)/metabolism , Sirtuins/deficiency , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Aspartate Aminotransferase, Cytoplasmic/genetics , Aspartate Aminotransferase, Cytoplasmic/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Disease Progression , Energy Metabolism/drug effects , Enzyme Activation , Enzyme Activators/pharmacology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mutation , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction , Sirtuins/genetics , Tumor Burden , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Gemcitabine
9.
J Exp Med ; 217(7)2020 07 06.
Article En | MEDLINE | ID: mdl-32441762

Approximately one third of cancer patients die due to complexities related to cachexia. However, the mechanisms of cachexia and the potential therapeutic interventions remain poorly studied. We observed a significant positive correlation between SIRT1 expression and muscle fiber cross-sectional area in pancreatic cancer patients. Rescuing Sirt1 expression by exogenous expression or pharmacological agents reverted cancer cell-induced myotube wasting in culture conditions and mouse models. RNA-seq and follow-up analyses showed cancer cell-mediated SIRT1 loss induced NF-κB signaling in cachectic muscles that enhanced the expression of FOXO transcription factors and NADPH oxidase 4 (Nox4), a key regulator of reactive oxygen species production. Additionally, we observed a negative correlation between NOX4 expression and skeletal muscle fiber cross-sectional area in pancreatic cancer patients. Knocking out Nox4 in skeletal muscles or pharmacological blockade of Nox4 activity abrogated tumor-induced cachexia in mice. Thus, we conclude that targeting the Sirt1-Nox4 axis in muscles is an effective therapeutic intervention for mitigating pancreatic cancer-induced cachexia.


Cachexia/complications , Cachexia/metabolism , NADPH Oxidase 4/metabolism , Neoplasms/complications , Neoplasms/metabolism , Signal Transduction , Sirtuin 1/metabolism , Adipose Tissue/pathology , Animals , Cell Line , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Forkhead Transcription Factors/metabolism , HEK293 Cells , Humans , Metabolome/drug effects , Mice , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , NF-kappa B/metabolism , Oxidation-Reduction , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Stability/drug effects , Reactive Oxygen Species/metabolism , Resveratrol/pharmacology , Signal Transduction/drug effects , Wasting Syndrome/pathology
10.
Nat Med ; 25(4): 628-640, 2019 04.
Article En | MEDLINE | ID: mdl-30833752

Pancreatic ductal adenocarcinoma (PDAC) is characterized by KRAS- and autophagy-dependent tumorigenic growth, but the role of KRAS in supporting autophagy has not been established. We show that, to our surprise, suppression of KRAS increased autophagic flux, as did pharmacological inhibition of its effector ERK MAPK. Furthermore, we demonstrate that either KRAS suppression or ERK inhibition decreased both glycolytic and mitochondrial functions. We speculated that ERK inhibition might thus enhance PDAC dependence on autophagy, in part by impairing other KRAS- or ERK-driven metabolic processes. Accordingly, we found that the autophagy inhibitor chloroquine and genetic or pharmacologic inhibition of specific autophagy regulators synergistically enhanced the ability of ERK inhibitors to mediate antitumor activity in KRAS-driven PDAC. We conclude that combinations of pharmacologic inhibitors that concurrently block both ERK MAPK and autophagic processes that are upregulated in response to ERK inhibition may be effective treatments for PDAC.


Autophagy , Chloroquine/pharmacology , MAP Kinase Signaling System , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , HEK293 Cells , Humans , MAP Kinase Signaling System/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mutation/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Pancreatic Neoplasms
11.
Metabolomics ; 14(12): 156, 2018 11 27.
Article En | MEDLINE | ID: mdl-30830412

INTRODUCTION: Gemcitabine is an important component of pancreatic cancer clinical management. Unfortunately, acquired gemcitabine resistance is widespread and there are limitations to predicting and monitoring therapeutic outcomes. OBJECTIVE: To investigate the potential of metabolomics to differentiate pancreatic cancer cells that develops resistance or respond to gemcitabine treatment. RESULTS: We applied 1D 1H and 2D 1H-13C HSQC NMR methods to profile the metabolic signature of pancreatic cancer cells. 13C6-glucose labeling identified 30 key metabolites uniquely altered between wild-type and gemcitabine-resistant cells upon gemcitabine treatment. Gemcitabine resistance was observed to reprogram glucose metabolism and to enhance the pyrimidine synthesis pathway. Myo-inositol, taurine, glycerophosphocholine and creatinine phosphate exhibited a "binary switch" in response to gemcitabine treatment and acquired resistance. CONCLUSION: Metabolic differences between naïve and resistant pancreatic cancer cells and, accordingly, their unique responses to gemcitabine treatment were revealed, which may be useful in the clinical setting for monitoring a patient's therapeutic response.


Antimetabolites, Antineoplastic/pharmacology , Biomarkers/metabolism , Deoxycytidine/analogs & derivatives , Drug Monitoring/methods , Drug Resistance, Neoplasm , Metabolome , Pancreatic Neoplasms/metabolism , Deoxycytidine/pharmacology , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Tumor Cells, Cultured , Gemcitabine
13.
J Proteome Res ; 16(10): 3536-3546, 2017 10 06.
Article En | MEDLINE | ID: mdl-28809118

Pancreatic cancer cells overexpressing Mucin 1 (MUC1) rely on aerobic glycolysis and, correspondingly, are dependent on glucose for survival. Our NMR metabolomics comparative analysis of control (S2-013.Neo) and MUC1-overexpressing (S2-013.MUC1) cells demonstrates that MUC1 reprograms glutamine metabolism upon glucose limitation. The observed alteration in glutamine metabolism under glucose limitation was accompanied by a relative decrease in the proliferation of MUC1-overexpressing cells compared with steady-state conditions. Moreover, glucose limitation induces G1 phase arrest where S2-013.MUC1 cells fail to enter S phase and synthesize DNA because of a significant disruption in pyrimidine nucleotide biosynthesis. Our metabolomics analysis indicates that glutamine is the major source of oxaloacetate in S2-013.Neo and S2-013.MUC1 cells, where oxaloacetate is converted to aspartate, an important metabolite for pyrimidine nucleotide biosynthesis. However, glucose limitation impedes the flow of glutamine carbons into the pyrimidine nucleotide rings and instead leads to a significant accumulation of glutamine-derived aspartate in S2-013.MUC1 cells.


Glucose/metabolism , Glutamine/metabolism , Mucin-1/genetics , Pancreatic Neoplasms/metabolism , Aspartic Acid , Cell Line, Tumor , Cell Proliferation/genetics , Citric Acid Cycle , DNA Replication/genetics , Glucose/genetics , Glutamine/genetics , Glycolysis/genetics , Humans , Magnetic Resonance Spectroscopy , Metabolomics , Mucin-1/metabolism , Oxaloacetic Acid/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology
14.
Cancer Res ; 77(20): 5503-5517, 2017 10 15.
Article En | MEDLINE | ID: mdl-28811332

Pancreatic adenocarcinoma is moderately responsive to gemcitabine-based chemotherapy, the most widely used single-agent therapy for pancreatic cancer. Although the prognosis in pancreatic cancer remains grim in part due to poor response to therapy, previous attempts at identifying and targeting the resistance mechanisms have not been very successful. By leveraging The Cancer Genome Atlas dataset, we identified lipid metabolism as the metabolic pathway that most significantly correlated with poor gemcitabine response in pancreatic cancer patients. Furthermore, we investigated the relationship between alterations in lipogenesis pathway and gemcitabine resistance by utilizing tissues from the genetically engineered mouse model and human pancreatic cancer patients. We observed a significant increase in fatty acid synthase (FASN) expression with increasing disease progression in spontaneous pancreatic cancer mouse model, and a correlation of high FASN expression with poor survival in patients and poor gemcitabine responsiveness in cell lines. We observed a synergistic effect of FASN inhibitors with gemcitabine in pancreatic cancer cells in culture and orthotopic implantation models. Combination of gemcitabine and the FASN inhibitor orlistat significantly diminished stemness, in part due to induction of endoplasmic reticulum (ER) stress that resulted in apoptosis. Moreover, direct induction of ER stress with thapsigargin caused a similar decrease in stemness and showed synergistic activity with gemcitabine. Our in vivo studies with orthotopic implantation models demonstrated a robust increase in gemcitabine responsiveness upon inhibition of fatty acid biosynthesis with orlistat. Altogether, we demonstrate that fatty acid biosynthesis pathway manipulation can help overcome the gemcitabine resistance in pancreatic cancer by regulating ER stress and stemness. Cancer Res; 77(20); 5503-17. ©2017 AACR.


Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Endoplasmic Reticulum Stress/physiology , Lipids/biosynthesis , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Adenocarcinoma/pathology , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm , Endoplasmic Reticulum Stress/drug effects , Female , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/pathology , Random Allocation , Signal Transduction , Xenograft Model Antitumor Assays , Gemcitabine
15.
Clin Cancer Res ; 23(19): 5881-5891, 2017 Oct 01.
Article En | MEDLINE | ID: mdl-28720669

Purpose:MUC1, an oncogene overexpressed in multiple solid tumors, including pancreatic cancer, reduces overall survival and imparts resistance to radiation and chemotherapies. We previously identified that MUC1 facilitates growth-promoting metabolic alterations in pancreatic cancer cells. The present study investigates the role of MUC1-mediated metabolism in radiation resistance of pancreatic cancer by utilizing cell lines and in vivo models.Experimental Design: We used MUC1-knockdown and -overexpressed cell line models for evaluating the role of MUC1-mediated metabolism in radiation resistance through in vitro cytotoxicity, clonogenicity, DNA damage response, and metabolomic evaluations. We also investigated whether inhibition of glycolysis could revert MUC1-mediated metabolic alterations and radiation resistance by using in vitro and in vivo models.Results: MUC1 expression diminished radiation-induced cytotoxicity and DNA damage in pancreatic cancer cells by enhancing glycolysis, pentose phosphate pathway, and nucleotide biosynthesis. Such metabolic reprogramming resulted in high nucleotide pools and radiation resistance in in vitro models. Pretreatment with the glycolysis inhibitor 3-bromopyruvate abrogated MUC1-mediated radiation resistance both in vitro and in vivo, by reducing glucose flux into nucleotide biosynthetic pathways and enhancing DNA damage, which could again be reversed by pretreatment with nucleoside pools.Conclusions: MUC1-mediated nucleotide metabolism plays a key role in facilitating radiation resistance in pancreatic cancer and targeted effectively through glycolytic inhibition. Clin Cancer Res; 23(19); 5881-91. ©2017 AACR.


DNA Damage/radiation effects , Mucin-1/genetics , Pancreatic Neoplasms/radiotherapy , Radiation Tolerance/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/radiation effects , Gene Expression Regulation, Neoplastic/radiation effects , Gene Knockdown Techniques , Glucose/metabolism , Glycolysis/radiation effects , Humans , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Signal Transduction/radiation effects , Xenograft Model Antitumor Assays
16.
Cancer Cell ; 32(1): 71-87.e7, 2017 07 10.
Article En | MEDLINE | ID: mdl-28697344

Poor response to cancer therapy due to resistance remains a clinical challenge. The present study establishes a widely prevalent mechanism of resistance to gemcitabine in pancreatic cancer, whereby increased glycolytic flux leads to glucose addiction in cancer cells and a corresponding increase in pyrimidine biosynthesis to enhance the intrinsic levels of deoxycytidine triphosphate (dCTP). Increased levels of dCTP diminish the effective levels of gemcitabine through molecular competition. We also demonstrate that MUC1-regulated stabilization of hypoxia inducible factor-1α (HIF-1α) mediates such metabolic reprogramming. Targeting HIF-1α or de novo pyrimidine biosynthesis, in combination with gemcitabine, strongly diminishes tumor burden. Finally, reduced expression of TKT and CTPS, which regulate flux into pyrimidine biosynthesis, correlates with better prognosis in pancreatic cancer patients on fluoropyrimidine analogs.


Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , Glucose/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mucin-1/metabolism , Pancreatic Neoplasms/drug therapy , Carbon/metabolism , Deoxycytidine/therapeutic use , Digoxin/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pentose Phosphate Pathway , Prognosis , Pyrimidines/biosynthesis , Signal Transduction , Gemcitabine
17.
PLoS One ; 12(6): e0179098, 2017.
Article En | MEDLINE | ID: mdl-28570622

[This corrects the article DOI: 10.1371/journal.pone.0176820.].

18.
PLoS One ; 12(5): e0176820, 2017.
Article En | MEDLINE | ID: mdl-28464016

BACKGROUND: Mucin1 (MUC1), a glycoprotein associated with chemoresistance and an aggressive cancer phenotype, is aberrantly overexpressed in triple-negative breast cancer (TNBC). Recent studies suggest that MUC1 plays a role in modulating cancer cell metabolism and thereby supports tumor growth. Herein, we examined the role of MUC1 in metabolic reprogramming in TNBC. METHODS: MUC1 was stably overexpressed in MDA-MB-231 TNBC cells and stably knocked down in MDA-MB-468 cells. We performed liquid chromatography-coupled tandem mass spectrometry-assisted metabolomic analyses and physiological assays, which indicated significant alterations in the metabolism of TNBC cells due to MUC1 expression. RESULTS: Differential analyses identified significant differences in metabolic pathways implicated in cancer cell growth. In particular, MUC1 expression altered glutamine dependency of the cells, which can be attributed in part to the changes in the expression of genes that regulate glutamine metabolism, as observed by real-time PCR analysis. Furthermore, MUC1 expression altered the sensitivity of cells to transaminase inhibitor aminooxyacetate (AOA), potentially by altering glutamine metabolism. CONCLUSIONS: Collectively, these results suggest that MUC1 serves as a metabolic regulator in TNBC, facilitating the metabolic reprogramming of glutamine utilization that influences TNBC tumor growth.


Cellular Reprogramming/physiology , Metabolome , Mucin-1/metabolism , Triple Negative Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Cell Survival/physiology , Chromatography, Liquid , Gene Expression Regulation, Neoplastic/physiology , Gene Knockdown Techniques , Glucose/metabolism , Glutamine/metabolism , Humans , Mucin-1/genetics , Nitrogen/metabolism , Real-Time Polymerase Chain Reaction , Tandem Mass Spectrometry
19.
Oncotarget ; 6(38): 41146-61, 2015 Dec 01.
Article En | MEDLINE | ID: mdl-26510913

Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer-related deaths in the US. Cancer-associated cachexia is present in up to 80% of PDAC patients and is associated with aggressive disease and poor prognosis. In the present studies we evaluated an anti-cancer natural product silibinin for its effectiveness in targeting pancreatic cancer aggressiveness and the cachectic properties of pancreatic cancer cells and tumors. Our results demonstrate that silibinin inhibits pancreatic cancer cell growth in a dose-dependent manner and reduces glycolytic activity of cancer cells. Our LC-MS/MS based metabolomics data demonstrates that silibinin treatment induces global metabolic reprogramming in pancreatic cancer cells. Silibinin treatment diminishes c-MYC expression, a key regulator of cancer metabolism. Furthermore, we observed reduced STAT3 signaling in silibinin-treated cancer cells. Overexpression of constitutively active STAT3 was sufficient to substantially revert the silibinin-induced downregulation of c-MYC and the metabolic phenotype. Our in vivo investigations demonstrate that silibinin reduces tumor growth and proliferation in an orthotopic mouse model of pancreatic cancer and prevents the loss of body weight and muscle. It also improves physical activity including grip strength and latency to fall in tumor-bearing mice. In conclusion, silibinin-induced metabolic reprogramming diminishes cell growth and cachectic properties of pancreatic cancer cells and animal models.


Cachexia/prevention & control , Metabolome/drug effects , Pancreatic Neoplasms/drug therapy , Silymarin/pharmacology , Xenograft Model Antitumor Assays , Animals , Antioxidants/pharmacology , Cachexia/etiology , Cachexia/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Glycolysis/drug effects , Humans , Immunoblotting , Interleukin-6/genetics , Interleukin-6/metabolism , Mice, Nude , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Silybin , Tumor Burden/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
20.
Oncotarget ; 6(22): 19118-31, 2015 Aug 07.
Article En | MEDLINE | ID: mdl-26046375

MUC16, a transmembrane mucin, facilitates pancreatic adenocarcinoma progression and metastasis. In the current studies, we observed that MUC16 knockdown pancreatic cancer cells exhibit reduced glucose uptake and lactate secretion along with reduced migration and invasion potential, which can be restored by supplementing the culture media with lactate, an end product of aerobic glycolysis. MUC16 knockdown leads to inhibition of mTOR activity and reduced expression of its downstream target c-MYC, a key player in cellular growth, proliferation and metabolism. Ectopic expression of c-MYC in MUC16 knockdown pancreatic cancer cells restores the altered cellular physiology. Our LC-MS/MS based metabolomics studies indicate global metabolic alterations in MUC16 knockdown pancreatic cancer cells, as compared to the controls. Specifically, glycolytic and nucleotide metabolite pools were significantly decreased. We observed similar metabolic alterations that correlated with MUC16 expression in primary tumor tissue specimens from human pancreatic adenocarcinoma cancer patients. Overall, our results demonstrate that MUC16 plays an important role in metabolic reprogramming of pancreatic cancer cells by increasing glycolysis and enhancing motility and invasiveness.


CA-125 Antigen/metabolism , Membrane Proteins/metabolism , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , TOR Serine-Threonine Kinases/metabolism , CA-125 Antigen/genetics , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Gene Knockdown Techniques , Glucose/metabolism , Humans , Lactic Acid/metabolism , Membrane Proteins/genetics , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , TOR Serine-Threonine Kinases/genetics , Transfection
...