Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Nature ; 566(7744): 403-406, 2019 02.
Article in English | MEDLINE | ID: mdl-30728499

ABSTRACT

Most tumours have an aberrantly activated lipid metabolism1,2 that enables them to synthesize, elongate and desaturate fatty acids to support proliferation. However, only particular subsets of cancer cells are sensitive to approaches that target fatty acid metabolism and, in particular, fatty acid desaturation3. This suggests that many cancer cells contain an unexplored plasticity in their fatty acid metabolism. Here we show that some cancer cells can exploit an alternative fatty acid desaturation pathway. We identify various cancer cell lines, mouse hepatocellular carcinomas, and primary human liver and lung carcinomas that desaturate palmitate to the unusual fatty acid sapienate to support membrane biosynthesis during proliferation. Accordingly, we found that sapienate biosynthesis enables cancer cells to bypass the known fatty acid desaturation pathway that is dependent on stearoyl-CoA desaturase. Thus, only by targeting both desaturation pathways is the in vitro and in vivo proliferation of cancer cells that synthesize sapienate impaired. Our discovery explains metabolic plasticity in fatty acid desaturation and constitutes an unexplored metabolic rewiring in cancers.


Subject(s)
Fatty Acids/chemistry , Fatty Acids/metabolism , Metabolic Networks and Pathways , Neoplasms/metabolism , Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation , Fatty Acid Desaturases/metabolism , Female , HEK293 Cells , Humans , Male , Mice , Oleic Acids/metabolism , Palmitates/metabolism , Palmitic Acids/metabolism , Stearoyl-CoA Desaturase/metabolism
2.
Handb Exp Pharmacol ; 233: 321-53, 2016.
Article in English | MEDLINE | ID: mdl-25912014

ABSTRACT

Targeting cancer metabolism has the potential to lead to major advances in tumor therapy. Numerous promising metabolic drug targets have been identified. Yet, it has emerged that there is no singular metabolism that defines the oncogenic state of the cell. Rather, the metabolism of cancer cells is a function of the requirements of a tumor. Hence, the tissue of origin, the (epi)genetic drivers, the aberrant signaling, and the microenvironment all together define these metabolic requirements. In this chapter we discuss in light of (epi)genetic, signaling, and environmental factors the diversity in cancer metabolism based on triple-negative and estrogen receptor-positive breast cancer, early- and late-stage prostate cancer, and liver cancer. These types of cancer all display distinct and partially opposing metabolic behaviors (e.g., Warburg versus reverse Warburg metabolism). Yet, for each of the cancers, their distinct metabolism supports the oncogenic phenotype. Finally, we will assess the therapeutic potential of metabolism based on the concepts of metabolic normalization and metabolic depletion.


Subject(s)
Neoplasms/metabolism , Breast Neoplasms/metabolism , Female , Humans , Liver Neoplasms/metabolism , Male , Neoplasms/drug therapy , Organ Specificity , Prostatic Neoplasms/metabolism , Tumor Microenvironment
3.
Int J Cancer ; 135(6): 1487-96, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24347491

ABSTRACT

Regorafenib, a novel multikinase inhibitor, has recently demonstrated overall survival benefits in metastatic colorectal cancer (CRC) patients. Our study aimed to gain further insight into the molecular mechanisms of regorafenib and to assess its potential in combination therapy. Regorafenib was tested alone and in combination with irinotecan in patient-derived (PD) CRC models and a murine CRC liver metastasis model. Mechanism of action was investigated using in vitro functional assays, immunohistochemistry and correlation with CRC-related oncogenes. Regorafenib demonstrated significant inhibition of growth-factor-mediated vascular endothelial growth factor receptor (VEGFR) 2 and VEGFR3 autophosphorylation, and intracellular VEGFR3 signaling in human umbilical vascular endothelial cells (HuVECs) and lymphatic endothelial cells (LECs), and also blocked migration of LECs. Furthermore, regorafenib inhibited proliferation in 19 of 25 human CRC cell lines and markedly slowed tumor growth in five of seven PD xenograft models. Combination of regorafenib with irinotecan significantly delayed tumor growth after extended treatment in four xenograft models. Reduced CD31 staining indicates that the antiangiogenic effects of regorafenib contribute to its antitumor activity. Finally, regorafenib significantly delayed disease progression in a murine CRC liver metastasis model by inhibiting the growth of established liver metastases and preventing the formation of new metastases in other organs. In addition, our results suggest that regorafenib displays antimetastatic activity, which may contribute to its efficacy in patients with metastatic CRC. Combination of regorafenib and irinotecan demonstrated an increased antitumor effect and could provide a future treatment option for CRC patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colorectal Neoplasms/drug therapy , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , Animals , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Cell Growth Processes/drug effects , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm , Female , Humans , Irinotecan , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/secondary , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Metastasis , Organoplatinum Compounds/pharmacology , Oxaliplatin , Phenylurea Compounds/administration & dosage , Pyridines/administration & dosage , Random Allocation , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-3/metabolism , Xenograft Model Antitumor Assays
4.
Cancer Res ; 81(8): 1988-2001, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33687947

ABSTRACT

Hepatic fat accumulation is associated with diabetes and hepatocellular carcinoma (HCC). Here, we characterize the metabolic response that high-fat availability elicits in livers before disease development. After a short term on a high-fat diet (HFD), otherwise healthy mice showed elevated hepatic glucose uptake and increased glucose contribution to serine and pyruvate carboxylase activity compared with control diet (CD) mice. This glucose phenotype occurred independently from transcriptional or proteomic programming, which identifies increased peroxisomal and lipid metabolism pathways. HFD-fed mice exhibited increased lactate production when challenged with glucose. Consistently, administration of an oral glucose bolus to healthy individuals revealed a correlation between waist circumference and lactate secretion in a human cohort. In vitro, palmitate exposure stimulated production of reactive oxygen species and subsequent glucose uptake and lactate secretion in hepatocytes and liver cancer cells. Furthermore, HFD enhanced the formation of HCC compared with CD in mice exposed to a hepatic carcinogen. Regardless of the dietary background, all murine tumors showed similar alterations in glucose metabolism to those identified in fat exposed nontransformed mouse livers, however, particular lipid species were elevated in HFD tumor and nontumor-bearing HFD liver tissue. These findings suggest that fat can induce glucose-mediated metabolic changes in nontransformed liver cells similar to those found in HCC. SIGNIFICANCE: With obesity-induced hepatocellular carcinoma on a rising trend, this study shows in normal, nontransformed livers that fat induces glucose metabolism similar to an oncogenic transformation.


Subject(s)
Carcinoma, Hepatocellular/etiology , Diet, High-Fat , Dietary Fats/metabolism , Glucose/metabolism , Hepatocytes/metabolism , Liver Neoplasms/etiology , Animals , Carcinoma, Hepatocellular/metabolism , Cell Transformation, Neoplastic , Citric Acid Cycle/physiology , Fatty Acids/metabolism , Glucose Tolerance Test , Humans , Lactic Acid/biosynthesis , Lipid Metabolism , Liver Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Obesity/complications , Palmitates/pharmacology , Peroxisomes/metabolism , Proteomics , Pyruvate Carboxylase/metabolism , Random Allocation , Reactive Oxygen Species/metabolism , Serine/metabolism , Transcriptional Activation
5.
Cell Rep ; 17(3): 837-848, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27732858

ABSTRACT

Cellular proliferation depends on refilling the tricarboxylic acid (TCA) cycle to support biomass production (anaplerosis). The two major anaplerotic pathways in cells are pyruvate conversion to oxaloacetate via pyruvate carboxylase (PC) and glutamine conversion to α-ketoglutarate. Cancers often show an organ-specific reliance on either pathway. However, it remains unknown whether they adapt their mode of anaplerosis when metastasizing to a distant organ. We measured PC-dependent anaplerosis in breast-cancer-derived lung metastases compared to their primary cancers using in vivo 13C tracer analysis. We discovered that lung metastases have higher PC-dependent anaplerosis compared to primary breast cancers. Based on in vitro analysis and a mathematical model for the determination of compartment-specific metabolite concentrations, we found that mitochondrial pyruvate concentrations can promote PC-dependent anaplerosis via enzyme kinetics. In conclusion, we show that breast cancer cells proliferating as lung metastases activate PC-dependent anaplerosis in response to the lung microenvironment.


Subject(s)
Breast Neoplasms/pathology , Citric Acid Cycle , Lung Neoplasms/enzymology , Lung Neoplasms/secondary , Pyruvate Carboxylase/metabolism , Acetyl Coenzyme A/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Carbon Isotopes , Cell Compartmentation , Cell Line, Tumor , Cytosol/metabolism , Female , Humans , Isotope Labeling , Mitochondria/metabolism , Pyruvic Acid/metabolism , Tumor Microenvironment
6.
Cell Metab ; 23(2): 280-91, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26774962

ABSTRACT

The oxygen-sensing prolyl hydroxylase domain proteins (PHDs) regulate cellular metabolism, but their role in neuronal metabolism during stroke is unknown. Here we report that PHD1 deficiency provides neuroprotection in a murine model of permanent brain ischemia. This was not due to an increased collateral vessel network. Instead, PHD1(-/-) neurons were protected against oxygen-nutrient deprivation by reprogramming glucose metabolism. Indeed, PHD1(-/-) neurons enhanced glucose flux through the oxidative pentose phosphate pathway by diverting glucose away from glycolysis. As a result, PHD1(-/-) neurons increased their redox buffering capacity to scavenge oxygen radicals in ischemia. Intracerebroventricular injection of PHD1-antisense oligonucleotides reduced the cerebral infarct size and neurological deficits following stroke. These data identify PHD1 as a regulator of neuronal metabolism and a potential therapeutic target in ischemic stroke.


Subject(s)
Brain Ischemia/prevention & control , Cellular Reprogramming , Gene Deletion , Neurons/metabolism , Oxygen/metabolism , Procollagen-Proline Dioxygenase/metabolism , Stroke/prevention & control , Animals , Brain/blood supply , Brain/drug effects , Brain/pathology , Brain Ischemia/complications , Carbon/metabolism , Cellular Reprogramming/drug effects , Free Radical Scavengers/metabolism , Hydroxylation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Injections, Intraventricular , Mice, Knockout , Neurons/drug effects , Neuroprotection/drug effects , Oligonucleotides/administration & dosage , Oligonucleotides/pharmacology , Oxidation-Reduction/drug effects , Pentose Phosphate Pathway/drug effects , Phenotype , Procollagen-Proline Dioxygenase/deficiency , Reactive Oxygen Species/metabolism , Stroke/complications
7.
Neoplasia ; 15(10): 1161-71, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24204195

ABSTRACT

OBJECTIVE: The objectives of the study were to evaluate the allosteric mitogen-activated protein kinase kinase (MEK) inhibitor BAY 86-9766 in monotherapy and in combination with sorafenib in orthotopic and subcutaneous hepatocellular carcinoma (HCC) models with different underlying etiologies in two species. DESIGN: Antiproliferative potential of BAY 86-9766 and synergistic effects with sorafenib were studied in several HCC cell lines. Relevant pathway signaling was studied in MH3924a cells. For in vivo testing, the HCC cells were implanted subcutaneously or orthotopically. Survival and mode of action (MoA) were analyzed. RESULTS: BAY 86-9766 exhibited potent antiproliferative activity in HCC cell lines with half-maximal inhibitory concentration values ranging from 33 to 762 nM. BAY 86-9766 was strongly synergistic with sorafenib in suppressing tumor cell proliferation and inhibiting phosphorylation of the extracellular signal-regulated kinase (ERK). BAY 86-9766 prolonged survival in Hep3B xenografts, murine Hepa129 allografts, and MH3924A rat allografts. Additionally, tumor growth, ascites formation, and serum alpha-fetoprotein levels were reduced. Synergistic effects in combination with sorafenib were shown in Huh-7, Hep3B xenografts, and MH3924A allografts. On the signaling pathway level, the combination of BAY 86-9766 and sorafenib led to inhibition of the upregulatory feedback loop toward MEK phosphorylation observed after BAY 86-9766 monotreatment. With regard to the underlying MoA, inhibition of ERK phosphorylation, tumor cell proliferation, and microvessel density was observed in vivo. CONCLUSION: BAY 86-9766 shows potent single-agent antitumor activity and acts synergistically in combination with sorafenib in preclinical HCC models. These results support the ongoing clinical development of BAY 86-9766 and sorafenib in advanced HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Diphenylamine/analogs & derivatives , Liver Neoplasms/drug therapy , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Sulfonamides/pharmacology , Allografts , Allosteric Regulation , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Drug Synergism , Female , Heterografts , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Niacinamide/pharmacology , Niacinamide/therapeutic use , Phenylurea Compounds/therapeutic use , Rats , Sorafenib , Sulfonamides/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL