Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Metabolites ; 14(5)2024 May 11.
Article in English | MEDLINE | ID: mdl-38786757

ABSTRACT

Mass spectrometry (MS)-based clinical metabolomics is very promising for the discovery of new biomarkers and diagnostics. However, poor data accuracy and reproducibility limit its true potential, especially when performing data analysis across multiple sample sets. While high-resolution mass spectrometry has gained considerable popularity for discovery metabolomics, triple quadrupole (QqQ) instruments offer several benefits for the measurement of known metabolites in clinical samples. These benefits include high sensitivity and a wide dynamic range. Here, we present the Olaris Global Panel (OGP), a HILIC LC-QqQ MS method for the comprehensive analysis of ~250 metabolites from all major metabolic pathways in clinical samples. For the development of this method, multiple HILIC columns and mobile phase conditions were compared, the robustness of the leading LC method assessed, and MS acquisition settings optimized for optimal data quality. Next, the effect of U-13C metabolite yeast extract spike-ins was assessed based on data accuracy and precision. The use of these U-13C-metabolites as internal standards improved the goodness of fit to a linear calibration curve from r2 < 0.75 for raw data to >0.90 for most metabolites across the entire clinical concentration range of urine samples. Median within-batch CVs for all metabolite ratios to internal standards were consistently lower than 7% and less than 10% across batches that were acquired over a six-month period. Finally, the robustness of the OGP method, and its ability to identify biomarkers, was confirmed using a large sample set.

3.
Nat Metab ; 4(6): 693-710, 2022 06.
Article in English | MEDLINE | ID: mdl-35760868

ABSTRACT

Elevated production of collagen-rich extracellular matrix is a hallmark of cancer-associated fibroblasts (CAFs) and a central driver of cancer aggressiveness. Here we find that proline, a highly abundant amino acid in collagen proteins, is newly synthesized from glutamine in CAFs to make tumour collagen in breast cancer xenografts. PYCR1 is a key enzyme for proline synthesis and highly expressed in the stroma of breast cancer patients and in CAFs. Reducing PYCR1 levels in CAFs is sufficient to reduce tumour collagen production, tumour growth and metastatic spread in vivo and cancer cell proliferation in vitro. Both collagen and glutamine-derived proline synthesis in CAFs are epigenetically upregulated by increased pyruvate dehydrogenase-derived acetyl-CoA levels. PYCR1 is a cancer cell vulnerability and potential target for therapy; therefore, our work provides evidence that targeting PYCR1 may have the additional benefit of halting the production of a pro-tumorigenic extracellular matrix. Our work unveils new roles for CAF metabolism to support pro-tumorigenic collagen production.


Subject(s)
Breast Neoplasms , Cancer-Associated Fibroblasts , Pyrroline Carboxylate Reductases/metabolism , Breast Neoplasms/metabolism , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Collagen/metabolism , Extracellular Matrix/metabolism , Female , Glutamine/metabolism , Humans , Proline , delta-1-Pyrroline-5-Carboxylate Reductase
4.
EMBO Mol Med ; 14(3): e14764, 2022 03 07.
Article in English | MEDLINE | ID: mdl-35014179

ABSTRACT

Despite the clinical benefit of androgen-deprivation therapy (ADT), the majority of patients with advanced prostate cancer (PCa) ultimately develop lethal castration-resistant prostate cancer (CRPC). In this study, we identified thioesterase superfamily member 6 (THEM6) as a marker of ADT resistance in PCa. THEM6 deletion reduces in vivo tumour growth and restores castration sensitivity in orthograft models of CRPC. Mechanistically, we show that the ER membrane-associated protein THEM6 regulates intracellular levels of ether lipids and is essential to trigger the induction of the ER stress response (UPR). Consequently, THEM6 loss in CRPC cells significantly alters ER function, reducing de novo sterol biosynthesis and preventing lipid-mediated activation of ATF4. Finally, we demonstrate that high THEM6 expression is associated with poor survival and correlates with high levels of UPR activation in PCa patients. Altogether, our results highlight THEM6 as a novel driver of therapy resistance in PCa as well as a promising target for the treatment of CRPC.


Subject(s)
Androgen Antagonists , Prostatic Neoplasms, Castration-Resistant , Androgen Antagonists/pharmacology , Androgen Antagonists/therapeutic use , Gene Expression Regulation, Neoplastic , Humans , Lipid Metabolism , Male , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/pathology
5.
Nat Metab ; 2(5): 432-446, 2020 05.
Article in English | MEDLINE | ID: mdl-32694660

ABSTRACT

Chronic inflammation is linked to diverse disease processes, but the intrinsic mechanisms that determine cellular sensitivity to inflammation are incompletely understood. Here, we show the contribution of glucose metabolism to inflammation-induced changes in the survival of pancreatic islet ß-cells. Using metabolomic, biochemical and functional analyses, we investigate the protective versus non-protective effects of glucose in the presence of pro-inflammatory cytokines. When protective, glucose metabolism augments anaplerotic input into the TCA cycle via pyruvate carboxylase (PC) activity, leading to increased aspartate levels. This metabolic mechanism supports the argininosuccinate shunt, which fuels ureagenesis from arginine and conversely diminishes arginine utilization for production of nitric oxide (NO), a chief mediator of inflammatory cytotoxicity. Activation of the PC-urea cycle axis is sufficient to suppress NO synthesis and shield cells from death in the context of inflammation and other stress paradigms. Overall, these studies uncover a previously unappreciated link between glucose metabolism and arginine-utilizing pathways via PC-directed ureagenesis as a protective mechanism.


Subject(s)
Arginine/metabolism , Glucose/metabolism , Glucose/pharmacology , Inflammation/prevention & control , Insulin-Secreting Cells/drug effects , Urea Cycle Disorders, Inborn/pathology , Urea/metabolism , Adolescent , Adult , Aged , Aspartic Acid/metabolism , Cell Survival , Citric Acid Cycle/drug effects , Female , Humans , Inflammation/pathology , Insulin-Secreting Cells/pathology , Male , Metabolomics , Middle Aged , Nitric Oxide/metabolism , Pyruvate Carboxylase/metabolism , Urea Cycle Disorders, Inborn/metabolism , Young Adult
6.
Nat Commun ; 11(1): 2508, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32427840

ABSTRACT

Despite the clinical success of Androgen Receptor (AR)-targeted therapies, reactivation of AR signalling remains the main driver of castration-resistant prostate cancer (CRPC) progression. In this study, we perform a comprehensive unbiased characterisation of LNCaP cells chronically exposed to multiple AR inhibitors (ARI). Combined proteomics and metabolomics analyses implicate an acquired metabolic phenotype common in ARI-resistant cells and associated with perturbed glucose and lipid metabolism. To exploit this phenotype, we delineate a subset of proteins consistently associated with ARI resistance and highlight mitochondrial 2,4-dienoyl-CoA reductase (DECR1), an auxiliary enzyme of beta-oxidation, as a clinically relevant biomarker for CRPC. Mechanistically, DECR1 participates in redox homeostasis by controlling the balance between saturated and unsaturated phospholipids. DECR1 knockout induces ER stress and sensitises CRPC cells to ferroptosis. In vivo, DECR1 deletion impairs lipid metabolism and reduces CRPC tumour growth, emphasizing the importance of DECR1 in the development of treatment resistance.


Subject(s)
Lipid Metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Prostatic Neoplasms, Castration-Resistant/enzymology , Androgen Receptor Antagonists/administration & dosage , Disease Progression , Homeostasis , Humans , Male , Mitochondria/enzymology , Mitochondria/genetics , Oxidoreductases Acting on CH-CH Group Donors/genetics , Phospholipids/metabolism , Prostate/enzymology , Prostate/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Receptors, Androgen/genetics , Receptors, Androgen/metabolism
7.
Cell Rep ; 30(8): 2729-2742.e4, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32101748

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) features a near-universal mutation in KRAS. Additionally, the tumor suppressor PTEN is lost in ∼10% of patients, and in mouse models, this dramatically accelerates tumor progression. While oncogenic KRAS and phosphatidylinositol 3-kinase (PI3K) cause divergent metabolic phenotypes individually, how they synergize to promote tumor metabolic alterations and dependencies remains unknown. We show that in KRAS-driven murine PDAC cells, loss of Pten strongly enhances both mTOR signaling and macropinocytosis. Protein scavenging alleviates sensitivity to mTOR inhibition by rescuing AKT phosphorylation at serine 473 and consequently cell proliferation. Combined inhibition of mTOR and lysosomal processing of internalized protein eliminates the macropinocytosis-mediated resistance. Our results indicate that mTORC2, rather than mTORC1, is an important regulator of protein scavenging and that protein-mediated resistance could explain the lack of effectiveness of mTOR inhibitors in certain genetic backgrounds. Concurrent inhibition of mTOR and protein scavenging might be a valuable therapeutic approach.


Subject(s)
Drug Resistance, Neoplasm , Pancreatic Neoplasms/pathology , Pinocytosis , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adenocarcinoma/pathology , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Death , Cell Line, Tumor , Cell Proliferation , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice, Inbred C57BL , Models, Biological , PTEN Phosphohydrolase/metabolism , Pancreatic Neoplasms/metabolism , Phosphorylation , Phosphoserine/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Up-Regulation , Pancreatic Neoplasms
8.
Nat Cancer ; 1(10): 998-1009, 2020 10.
Article in English | MEDLINE | ID: mdl-33479702

ABSTRACT

Metabolic reprogramming is a key hallmark of cancer, but less is known about metabolic plasticity of the same tumor at different sites. Here, we investigated the metabolic adaptation of leukemia in two different microenvironments, the bone marrow and the central nervous system (CNS). We identified a metabolic signature of fatty-acid synthesis in CNS leukemia, highlighting Stearoyl-CoA desaturase (SCD1) as a key player. In vivo SCD1 overexpression increases CNS disease, whilst genetic or pharmacological inhibition of SCD1 decreases CNS load. Overall, we demonstrated that leukemic cells dynamically rewire metabolic pathways to suit local conditions and that targeting these adaptations can be exploited therapeutically.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma , Stearoyl-CoA Desaturase , Central Nervous System/metabolism , Humans , Lipogenesis , Stearoyl-CoA Desaturase/genetics , Tumor Microenvironment
9.
Cancer Res ; 80(2): 175-188, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31562248

ABSTRACT

Statins are widely prescribed inhibitors of the mevalonate pathway, acting to lower systemic cholesterol levels. The mevalonate pathway is critical for tumorigenesis and is frequently upregulated in cancer. Nonetheless, reported effects of statins on tumor progression are ambiguous, making it unclear whether statins, alone or in combination, can be used for chemotherapy. Here, using advanced mass spectrometry and isotope tracing, we showed that statins only modestly affected cancer cholesterol homeostasis. Instead, they significantly reduced synthesis and levels of another downstream product, the mitochondrial electron carrier coenzyme Q, both in cultured cancer cells and tumors. This compromised oxidative phosphorylation, causing severe oxidative stress. To compensate, cancer cells upregulated antioxidant metabolic pathways, including reductive carboxylation, proline synthesis, and cystine import. Targeting cystine import with an xCT transporter-lowering MEK inhibitor, in combination with statins, caused profound tumor cell death. Thus, statin-induced ROS production in cancer cells can be exploited in a combinatorial regimen. SIGNIFICANCE: Cancer cells induce specific metabolic pathways to alleviate the increased oxidative stress caused by statin treatment, and targeting one of these pathways synergizes with statins to produce a robust antitumor response.See related commentary by Cordes and Metallo, p. 151.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Pancreatic Neoplasms , Humans , Mevalonic Acid , Oxidative Stress/drug effects , Ubiquinone
10.
Cell Metab ; 30(6): 1055-1074.e8, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31708446

ABSTRACT

Accumulation of lactate in the tissue microenvironment is a feature of both inflammatory disease and cancer. Here, we assess the response of immune cells to lactate in the context of chronic inflammation. We report that lactate accumulation in the inflamed tissue contributes to the upregulation of the lactate transporter SLC5A12 by human CD4+ T cells. SLC5A12-mediated lactate uptake into CD4+ T cells induces a reshaping of their effector phenotype, resulting in increased IL17 production via nuclear PKM2/STAT3 and enhanced fatty acid synthesis. It also leads to CD4+ T cell retention in the inflamed tissue as a consequence of reduced glycolysis and enhanced fatty acid synthesis. Furthermore, antibody-mediated blockade of SLC5A12 ameliorates the disease severity in a murine model of arthritis. Finally, we propose that lactate/SLC5A12-induced metabolic reprogramming is a distinctive feature of lymphoid synovitis in rheumatoid arthritis patients and a potential therapeutic target in chronic inflammatory disorders.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Inflammation/immunology , Lactic Acid/metabolism , Monocarboxylic Acid Transporters/physiology , Symporters/physiology , Animals , Cell Line , Fatty Acids/metabolism , Female , Glycolysis , Humans , Interleukin-17/immunology , Male , Mice , Mice, Knockout , Monocarboxylic Acid Transporters/genetics , Symporters/genetics
11.
Cancer Discov ; 9(5): 617-627, 2019 05.
Article in English | MEDLINE | ID: mdl-30837243

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) develops a pronounced stromal response reflecting an aberrant wound-healing process. This stromal reaction features transdifferentiation of tissue-resident pancreatic stellate cells (PSC) into activated cancer-associated fibroblasts, a process induced by PDAC cells but of unclear significance for PDAC progression. Here, we show that PSCs undergo a dramatic lipid metabolic shift during differentiation in the context of pancreatic tumorigenesis, including remodeling of the intracellular lipidome and secretion of abundant lipids in the activated, fibroblastic state. Specifically, stroma-derived lysophosphatidylcholines support PDAC cell synthesis of phosphatidylcholines, key components of cell membranes, and also facilitate production of the potent wound-healing mediator lysophosphatidic acid (LPA) by the extracellular enzyme autotaxin, which is overexpressed in PDAC. The autotaxin-LPA axis promotes PDAC cell proliferation, migration, and AKT activation, and genetic or pharmacologic autotaxin inhibition suppresses PDAC growth in vivo. Our work demonstrates how PDAC cells exploit the local production of wound-healing mediators to stimulate their own growth and migration. SIGNIFICANCE: Our work highlights an unanticipated role for PSCs in producing the oncogenic LPA signaling lipid and demonstrates how PDAC tumor cells co-opt the release of wound-healing mediators by neighboring PSCs to promote their own proliferation and migration.See related commentary by Biffi and Tuveson, p. 578.This article is highlighted in the In This Issue feature, p. 565.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Lysophosphatidylcholines/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Stellate Cells/metabolism , Phosphoric Diester Hydrolases/metabolism , Stromal Cells/metabolism , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Movement , Cell Proliferation , Disease Models, Animal , Disease Progression , Female , Humans , Male , Mice, Inbred C57BL , Mice, Nude , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/pathology , Signal Transduction , Stromal Cells/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Cell Rep ; 24(10): 2596-2605.e5, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30184495

ABSTRACT

Lipid droplets, which store triglycerides and cholesterol esters, are a prominent feature of clear cell renal cell carcinoma (ccRCC). Although their presence in ccRCC is critical for sustained tumorigenesis, their contribution to lipid homeostasis and tumor cell viability is incompletely understood. Here we show that disrupting triglyceride synthesis compromises the growth of both ccRCC tumors and ccRCC cells exposed to tumor-like conditions. Functionally, hypoxia leads to increased fatty acid saturation through inhibition of the oxygen-dependent stearoyl-CoA desaturase (SCD) enzyme. Triglycerides counter a toxic buildup of saturated lipids, primarily by releasing the unsaturated fatty acid oleate (the principal product of SCD activity) from lipid droplets into phospholipid pools. Disrupting this process derails lipid homeostasis, causing overproduction of toxic saturated ceramides and acyl-carnitines as well as activation of the NF-κB transcription factor. Our work demonstrates that triglycerides promote homeostasis by "buffering" specific fatty acids.


Subject(s)
Fatty Acids/metabolism , Hypoxia/metabolism , Triglycerides/metabolism , Animals , Carcinoma, Renal Cell/metabolism , Cell Line , Cell Survival/physiology , Ceramides/metabolism , Chromatography, Liquid , Fatty Acids/blood , Female , Flow Cytometry , Humans , Hypoxia/blood , Kidney Neoplasms/metabolism , Lipid Metabolism/physiology , Mass Spectrometry , Reverse Transcriptase Polymerase Chain Reaction , Stearoyl-CoA Desaturase/metabolism , Triglycerides/blood
13.
Cancer Res ; 78(12): 3321-3336, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29669760

ABSTRACT

The high mortality of pancreatic cancer demands that new therapeutic avenues be developed. The orally available small-molecule inhibitor AT13148 potently inhibits ROCK1 and ROCK2 kinases that regulate the actomyosin cytoskeleton. We previously reported that ROCK kinase expression increases with human and mouse pancreatic cancer progression and that conditional ROCK activation accelerates mortality in a genetically modified LSL-KrasG12D; LSL-p53R172H; Pdx1-Cre; (KPC) mouse pancreatic cancer model. In this study, we show that treatment of KPC mouse and human TKCC5 patient-derived pancreatic tumor cells with AT13148, as well as the ROCK-selective inhibitors Y27632 and H1152, act comparably in blocking ROCK substrate phosphorylation. AT13148, Y27632, and H1152 induced morphologic changes and reduced cellular contractile force generation, motility on pliable discontinuous substrates, and three-dimensional collagen matrix invasion. AT13148 treatment reduced subcutaneous tumor growth and blocked invasion of healthy pancreatic tissue by KPC tumor cells in vivo without affecting proliferation, suggesting a role for local tissue invasion as a contributor to primary tumor growth. These results suggest that AT13148 has antitumor properties that may be beneficial in combination therapies or in the adjuvant setting to reduce pancreatic cancer cell invasion and slow primary tumor growth. AT13148 might also have the additional benefit of enabling tumor resection by maintaining separation between tumor and healthy tissue boundaries.Significance: Preclinical evaluation of a small-molecule ROCK inhibitor reveals significant effects on PDAC invasion and tumor growth, further validating ROCK kinases as viable therapeutic targets in pancreatic cancer. Cancer Res; 78(12); 3321-36. ©2018 AACR.


Subject(s)
2-Hydroxyphenethylamine/analogs & derivatives , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , rho-Associated Kinases/antagonists & inhibitors , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 2-Hydroxyphenethylamine/pharmacology , 2-Hydroxyphenethylamine/therapeutic use , Amides/pharmacology , Amides/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor/transplantation , Cell Movement/drug effects , Disease Models, Animal , Female , HEK293 Cells , Humans , Male , Mice , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Pancreatic Neoplasms/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Signal Transduction/drug effects , rho-Associated Kinases/metabolism
14.
J Cell Sci ; 130(20): 3455-3466, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-28871044

ABSTRACT

Melanoma cells steer out of tumours using self-generated lysophosphatidic acid (LPA) gradients. The cells break down LPA, which is present at high levels around the tumours, creating a dynamic gradient that is low in the tumour and high outside. They then migrate up this gradient, creating a complex and evolving outward chemotactic stimulus. Here, we introduce a new assay for self-generated chemotaxis, and show that raising LPA levels causes a delay in migration rather than loss of chemotactic efficiency. Knockdown of the lipid phosphatase LPP3 - but not of its homologues LPP1 or LPP2 - diminishes the cell's ability to break down LPA. This is specific for chemotactically active LPAs, such as the 18:1 and 20:4 species. Inhibition of autotaxin-mediated LPA production does not diminish outward chemotaxis, but loss of LPP3-mediated LPA breakdown blocks it. Similarly, in both 2D and 3D invasion assays, knockdown of LPP3 diminishes the ability of melanoma cells to invade. Our results demonstrate that LPP3 is the key enzyme in the breakdown of LPA by melanoma cells, and confirm the importance of attractant breakdown in LPA-mediated cell steering.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Lysophospholipids/metabolism , Melanoma/metabolism , Phosphatidate Phosphatase/physiology , Skin Neoplasms/metabolism , Cell Line, Tumor , Chemotaxis , Humans , Melanoma/pathology , Neoplasm Invasiveness , Skin Neoplasms/pathology
15.
Clin Cancer Res ; 23(18): 5639-5647, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28611197

ABSTRACT

Purpose: To identify effective metabolic inhibitors to suppress the aggressive growth of pancreatic ductal adenocarcinoma (PDAC), we explored the in vivo antitumor efficacy of metabolic inhibitors, as single agents, in a panel of patient-derived PDAC xenograft models (PDX) and investigated whether genomic alterations of tumors correlate with the sensitivity to metabolic inhibitors.Experimental Design: Mice with established PDAC tumors from 6 to 13 individual PDXs were randomized and treated, once daily for 4 weeks, with either sterile PBS (vehicle) or the glutaminase inhibitor bis-2-(5-phenylacetamido-1,3,4-thiadiazol-2-yl)ethyl sulfide (BPTES), transaminase inhibitor aminooxyacetate (AOA), pyruvate dehydrogenase kinase inhibitor dichloroacetate (DCA), autophagy inhibitor chloroquine (CQ), and mitochondrial complex I inhibitor phenformin/metformin.Results: Among the agents tested, phenformin showed significant tumor growth inhibition (>30% compared with vehicle) in 5 of 12 individual PDXs. Metformin, at a fivefold higher dose, displayed significant tumor growth inhibition in 3 of 12 PDXs similar to BPTES (2/8 PDXs) and DCA (2/6 PDXs). AOA and CQ had the lowest response rates. Gene set enrichment analysis conducted using the baseline gene expression profile of pancreatic tumors identified a gene expression signature that inversely correlated with phenformin sensitivity, which is in agreement with the phenformin gene expression signature of NIH Library of Integrated Network-based Cellular Signatures (LINCS). The PDXs that were more sensitive to phenformin showed a baseline reduction in amino acids and elevation in oxidized glutathione. There was no correlation between phenformin response and genetic alterations in KRAS, TP53, SMAD4, or PTENConclusions: Phenformin treatment showed relatively higher antitumor efficacy against established PDAC tumors, compared with the efficacy of other metabolic inhibitors and metformin. Phenformin treatment significantly diminished PDAC tumor progression and prolonged tumor doubling time. Overall, our results serve as a foundation for further evaluation of phenformin as a therapeutic agent in pancreatic cancer. Clin Cancer Res; 23(18); 5639-47. ©2017 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Energy Metabolism/drug effects , Hypoglycemic Agents/pharmacology , Pancreatic Neoplasms/metabolism , Phenformin/pharmacology , Animals , Autophagy/drug effects , Biomarkers, Tumor , Cell Line, Tumor , Chloroquine/pharmacology , DNA Copy Number Variations , Disease Models, Animal , Energy Metabolism/genetics , Female , Genetic Variation , Glutamine/metabolism , Humans , Metabolic Networks and Pathways , Metabolomics/methods , Metformin/pharmacology , Mice , Mutation , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Xenograft Model Antitumor Assays
16.
Cell Rep ; 18(3): 647-658, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28099844

ABSTRACT

Acetyl-CoA is a key metabolic intermediate with an important role in transcriptional regulation. The nuclear-cytosolic acetyl-CoA synthetase 2 (ACSS2) was found to sustain the growth of hypoxic tumor cells. It generates acetyl-CoA from acetate, but exactly which pathways it supports is not fully understood. Here, quantitative analysis of acetate metabolism reveals that ACSS2 fulfills distinct functions depending on its cellular location. Exogenous acetate uptake is controlled by expression of both ACSS2 and the mitochondrial ACSS1, and ACSS2 supports lipogenesis. The mitochondrial and lipogenic demand for two-carbon acetyl units considerably exceeds the uptake of exogenous acetate, leaving it to only sparingly contribute to histone acetylation. Surprisingly, oxygen and serum limitation increase nuclear localization of ACSS2. We find that nuclear ACSS2 recaptures acetate released from histone deacetylation for recycling by histone acetyltransferases. Our work provides evidence for limited equilibration between nuclear and cytosolic acetyl-CoA and demonstrates that ACSS2 retains acetate to maintain histone acetylation.


Subject(s)
Acetate-CoA Ligase/metabolism , Acetates/metabolism , Cell Hypoxia , Histones/metabolism , Acetate-CoA Ligase/antagonists & inhibitors , Acetate-CoA Ligase/genetics , Acetates/chemistry , Acetyl Coenzyme A/metabolism , Acetylation , Carbon Isotopes/chemistry , Cell Line, Tumor , Cell Nucleus/enzymology , Chromatography, High Pressure Liquid , Culture Media/chemistry , Humans , Mass Spectrometry , Metabolome , Microscopy, Fluorescence , Mitochondria/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Serum/chemistry
18.
Curr Opin Biotechnol ; 43: 127-133, 2017 02.
Article in English | MEDLINE | ID: mdl-27915214

ABSTRACT

The lipidome comprises a large array of molecules with diverse physicochemical properties. Lipids are structural components of cells, act as a source of energy, and function as signaling mediators. Alterations in lipid metabolism are involved in the onset and progression of a variety of diseases, including metabolic syndrome and cancer. Because of this, interest in lipidomics, the comprehensive characterization of the lipidome by mass spectrometry, has intensified in recent years. However, obtaining a truly complete overview of all lipids in a sample has remained very challenging due to their enormous structural diversity. Here, we provide an overview of the collection of analytical approaches used to study various lipid classes, emphasizing innovations in sample preparation and liquid chromatography-mass spectrometry (LC-MS). Additionally, we provide practical suggestions for increasing the coverage of the lipidome.


Subject(s)
Lipids/analysis , Mass Spectrometry/methods , Animals , Humans , Lipid Metabolism
19.
Sci Adv ; 2(10): e1601273, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27819051

ABSTRACT

Serine catabolism to glycine and a one-carbon unit has been linked to the anabolic requirements of proliferating mammalian cells. However, genome-scale modeling predicts a catabolic role with one-carbon release as formate. We experimentally prove that in cultured cancer cells and nontransformed fibroblasts, most of the serine-derived one-carbon units are released from cells as formate, and that formate release is dependent on mitochondrial reverse 10-CHO-THF synthetase activity. We also show that in cancer cells, formate release is coupled to mitochondrial complex I activity, whereas in nontransformed fibroblasts, it is partially insensitive to inhibition of complex I activity. We demonstrate that in mice, about 50% of plasma formate is derived from serine and that serine starvation or complex I inhibition reduces formate synthesis in vivo. These observations transform our understanding of one-carbon metabolism and have implications for the treatment of diabetes and cancer with complex I inhibitors.

20.
J Cell Sci ; 129(18): 3367-73, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27635066

ABSTRACT

A defining hallmark of cancer is uncontrolled cell proliferation. This is initiated once cells have accumulated alterations in signaling pathways that control metabolism and proliferation, wherein the metabolic alterations provide the energetic and anabolic demands of enhanced cell proliferation. How these metabolic requirements are satisfied depends, in part, on the tumor microenvironment, which determines the availability of nutrients and oxygen. In this Cell Science at a Glance paper and the accompanying poster, we summarize our current understanding of cancer metabolism, emphasizing pathways of nutrient utilization and metabolism that either appear or have been proven essential for cancer cells. We also review how this knowledge has contributed to the development of anticancer therapies that target cancer metabolism.


Subject(s)
Neoplasms/metabolism , Animals , Fatty Acids/biosynthesis , Humans , Metabolic Networks and Pathways , Metabolome , Methylation , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...