Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
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.
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
4.
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
5.
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
6.
Cancer Lett ; 491: 70-77, 2020 10 28.
Article En | MEDLINE | ID: mdl-32735910

Cancer cachexia patients experience significant muscle wasting, which impairs the quality of life and treatment efficacy for patients. Skeletal muscle protein turnover is imparted by increased expression of ubiquitin-proteasome pathway components. Mitogen-activated protein kinases p38 and ERK have been shown to augment E3 ubiquitin ligase expression. Utilizing reverse-phase protein arrays, we identified pancreatic cancer cell-conditioned media-induced activation of JNK signaling in myotubes differentiated from C2C12 myoblasts. Inhibition of JNK signaling with SP600125 reduced cancer cell-conditioned media-induced myotube atrophy, myosin heavy chain protein turnover, and mRNA expression of cachexia-specific ubiquitin ligases Trim63 and Fbxo32. Furthermore, utilizing an orthotopic pancreatic cancer cachexia mouse model, we demonstrated that treatment of tumor-bearing mice with SP600125 improved longitudinal measurements of forelimb grip strength. Post-necropsy measurements demonstrated that SP600125 treatment rescued body weight, carcass weight, and gastrocnemius muscle weight loss without impacting tumor growth. JNK inhibitor treatment also rescued myofiber degeneration and reduced the muscle expression of Trim63 and Fbxo32. These data demonstrate that JNK signaling contributes to muscle wasting in cancer cachexia, and its inhibition has the potential to be utilized as an anti-cachectic therapy.


Cachexia/etiology , JNK Mitogen-Activated Protein Kinases/physiology , MAP Kinase Signaling System/physiology , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Pancreatic Neoplasms/complications , Animals , Anthracenes/pharmacology , Anthracenes/therapeutic use , Cachexia/drug therapy , Cachexia/metabolism , Cell Line, Tumor , Female , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mice , Muscle Fibers, Skeletal/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism
7.
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
8.
Methods Mol Biol ; 1882: 321-333, 2019.
Article En | MEDLINE | ID: mdl-30378066

Cachexia, a complex metabolic syndrome, is characterized by involuntary weight loss along with muscle wasting and fat depletion leading to poor quality of life of patients. About 80% of pancreatic cancer patients exhibit cachectic phenotype at the time of diagnosis. Here, we present the several molecular and physiological parameters, which we utilize to study the pancreatic cancer-induced cachexia in in vitro models and preclinical mice models of pancreatic cancer. We have described myotube and adipocyte-based in vitro models of muscle and fat wasting, including methods of cell culture, differentiation, and treatment with cancer cell-conditioned medium. Furthermore, we have explained the methods of evaluation of key cachectic markers for muscles. Next, we have detailed the orthotopic implantation mouse models of pancreatic cancer and evaluation of different physiological parameters, including body weight, food intake, body composition analysis, glucose tolerance test, insulin resistance test, grip strength measurement, and rotarod performance test. We have also explained morphological parameters and molecular markers to evaluate the muscle wasting in pancreatic cancer-induced cachexia.


Cachexia/pathology , Cell Culture Techniques/methods , Pancreatic Neoplasms/complications , Xenograft Model Antitumor Assays/methods , 3T3-L1 Cells , Absorptiometry, Photon , Adipocytes/physiology , Animals , Cachexia/diagnosis , Cachexia/etiology , Cachexia/physiopathology , Cell Culture Techniques/instrumentation , Cell Differentiation/drug effects , Cell Line, Tumor , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Humans , Mice , Mice, Nude , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Pancreatic Neoplasms/pathology , Rotarod Performance Test/instrumentation , Rotarod Performance Test/methods , Xenograft Model Antitumor Assays/instrumentation
9.
Methods Mol Biol ; 1742: 107-113, 2018.
Article En | MEDLINE | ID: mdl-29330794

Most solid tumors are hypoxic in nature due to the limited supply of oxygen to internal tissues. Hypoxia plays an important role in metabolic adaptations of tumors that contribute significantly to cancer pathogenesis. Among the several metabolic alterations induced by hypoxia, hypoxia-mediated increased glucose uptake serves as the hallmark of metabolic reprogramming. Hypoxia-mediated stabilization of hypoxia-inducible factor-1 alpha (HIF-1α) transcription factor leads to altered expression of several glycolytic genes and glucose transporters, which results in increased glucose uptake by tumor cells. Here we describe an easy and simple way of measuring the hypoxia-mediated tumor glucose uptake in vivo. The method is based on fluorescent imaging probe, RediJect 2-DG, which is a nonradioactive fluorescent-tagged glucose molecule. We have discussed orthotopic tumor implantation of HIF-1α knockdown and control pancreatic cancer cells and glucose uptake measurement in vivo by using IVIS imaging system along with reagent preparations.


Glucose/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Pancreatic Neoplasms/metabolism , Animals , Cell Hypoxia , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Regulatory Networks , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Optical Imaging , Pancreatic Neoplasms/genetics
10.
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
...