Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 9(1): 1208, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29572438

ABSTRACT

Lactate exchange between glycolytic and oxidative cancer cells is proposed to optimize tumor growth. Blocking lactate uptake through monocarboxylate transporter 1 (MCT1) represents an attractive therapeutic strategy but may stimulate glucose consumption by oxidative cancer cells. We report here that inhibition of mitochondrial pyruvate carrier (MPC) activity fulfils the tasks of blocking lactate use while preventing glucose oxidative metabolism. Using in vitro 13C-glucose and in vivo hyperpolarized 13C-pyruvate, we identify 7ACC2 as a potent inhibitor of mitochondrial pyruvate transport which consecutively blocks extracellular lactate uptake by promoting intracellular pyruvate accumulation. Also, while in spheroids MCT1 inhibition leads to cytostatic effects, MPC activity inhibition induces cytotoxic effects together with glycolysis stimulation and uncompensated inhibition of mitochondrial respiration. Hypoxia reduction obtained with 7ACC2 is further shown to sensitize tumor xenografts to radiotherapy. This study positions MPC as a control point for lactate metabolism and expands on the anticancer potential of MPC inhibition.


Subject(s)
Lactic Acid/pharmacokinetics , Mitochondria/metabolism , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/physiology , Pyruvic Acid/metabolism , Symporters/genetics , Symporters/physiology , Animals , Antineoplastic Agents/pharmacology , Biological Transport , Cell Line, Tumor , Female , Gene Silencing , Glucose/chemistry , Glycolysis/drug effects , Humans , Ion Transport/drug effects , Lactic Acid/chemistry , MCF-7 Cells , Male , Mice , Mice, Inbred C57BL , Muscle Proteins/genetics , Muscle Proteins/physiology , Neoplasm Transplantation , Oxygen/chemistry , RNA, Small Interfering/metabolism , Radiation-Sensitizing Agents/pharmacology , Rats , Thiophenes/chemistry , Uracil/analogs & derivatives , Uracil/chemistry , Xenopus laevis
2.
Open Biol ; 7(12)2017 12.
Article in English | MEDLINE | ID: mdl-29263247

ABSTRACT

Endothelial cell (EC) metabolism has lately emerged as a novel and promising therapeutic target to block vascular dysregulation associated with diseases like cancer and blinding eye disease. Glycolysis, fatty acid oxidation (FAO) and, more recently, glutamine/asparagine metabolism emerged as key regulators of EC metabolism, able to impact angiogenesis in health and disease. ECs are highly glycolytic as they require ATP and biomass for vessel sprouting. Notably, a regulator of the glycolytic pathway, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3, controls vessel sprouting during the angiogenic switch and its inhibition in tumour ECs leads to vessel normalization, thereby reducing metastasis and ameliorating chemotherapy. Moreover, FAO promotes EC proliferation through DNA synthesis, and plays an essential role in lymphangiogenesis via epigenetic regulation of histone acetylation. Pathological angiogenesis was decreased upon blockade of carnitine palmitoyltransferase 1, a regulator of FAO in ECs. More recently, metabolism of glutamine, in conjunction with asparagine, was reported to maintain EC sprouting through TCA anaplerosis, redox homeostasis, mTOR activation and endoplasmic stress control. Inactivation or blockade of glutaminase 1, which hydrolyses glutamine into ammonia and glutamate, impairs angiogenesis in health and disease, while silencing of asparagine synthetase reduces vessel sprouting in vitro In this review, we summarize recent insights into EC metabolism and discuss therapeutic implications of targeting EC metabolism.


Subject(s)
Endothelium, Vascular/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Energy Metabolism , Humans , Neovascularization, Pathologic/drug therapy , Oxidative Stress
3.
Curr Opin Hematol ; 24(3): 240-247, 2017 May.
Article in English | MEDLINE | ID: mdl-28212191

ABSTRACT

PURPOSE OF REVIEW: Endothelial cell metabolism has recently emerged as an important coregulator of angiogenesis and is therefore a promising new target in various angiogenesis-associated illnesses, like cancer. In this review, we discuss recent insights in endothelial cell metabolism in both physiological and pathological conditions and discuss possible translational implications. RECENT FINDINGS: Two metabolic pathways that determine the performance of endothelial cells are glycolysis and fatty acid oxidation (FAO). Glycolysis is essential as endothelial cells primarily rely on this pathway for ATP production. 6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) is a key regulator of glycolysis in endothelial cells. As endothelial cells increase glycolysis even further during angiogenesis, PFKFB3 also controls vessel sprouting and promotes endothelial cell migration. Moreover, in tumors, additional PFKFB3 upregulation leads to a more immature and dysfunctional vasculature. PFKFB3 blockade therefore results in tumor vessel normalization, with beneficial therapeutic effects on reduced metastasis and improved chemotherapy. Also, FAO stimulates endothelial cell proliferation through affecting DNA synthesis, and is critical for lymphangiogenesis, in part through epigenetic changes in histone acetylation. As FAO is controlled by carnitine palmitoyltransferase 1a, inhibition of this key enzyme decreases pathological angiogenesis. SUMMARY: Both PFKFB3 and carnitine palmitoyltransferase 1a are key metabolic regulators of vessel sprouting and promising new therapeutic targets in diseases associated with pathological angiogenesis.


Subject(s)
Endothelial Cells/metabolism , Animals , Biomarkers , Blood Vessels/metabolism , Cell Communication , Energy Metabolism , Gene Expression Regulation , Homeostasis , Humans , Metabolic Networks and Pathways , Neoplasms/etiology , Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic , Signal Transduction
4.
Int J Pharm ; 476(1-2): 9-15, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25245548

ABSTRACT

The purpose of this study was to develop vitamin E-based micelles loaded with Doxorubicin (DOX) (DOX-TOS-TPGS), taking advantages of the anti-cancer activity of vitamin E derivatives: Tocopherol succinate (TOS) and D-α-tocopherol polyethylene2000 succinate (TPGS). Therefore, we developed micelles consisting in a mixture of TOS (as solubilizer) and TPGS2000 (as stabilizer) (1:1). DOX-TOS-TPGS micelles exhibited a size of 78 nm and a ζ potential of -7 mV. High drug loading (40% w/w) was achieved. The critical micellar concentration was determined at 14 µg/ml. In vitro, after 24 h, DOX-TOS-TPGS micelles exhibited higher cytotoxicity than free-DOX (IC50 on MCF-7 cells, at 24 h, 58 vs 5 µg/ml). In vivo anti-tumor efficacy, performed on two tumor models (CT26 and MCF-7), demonstrated a 100% long-term survival of mice when treated with DOX-TOS-TPGS compared to DOX-free. Interestingly, the survival time of mice treated with unloaded TOS-TPGS micelles was similar to DOX-free, indicating an anti-cancer activity of vitamin E derivatives. Based on these results, it can be concluded that the formulations developed in this work may be considered as an effective DOX delivery system for cancer chemotherapy.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Drug Delivery Systems , Vitamin E/analogs & derivatives , Animals , Antibiotics, Antineoplastic/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Doxorubicin/administration & dosage , Excipients/chemistry , Female , Humans , Inhibitory Concentration 50 , MCF-7 Cells , Mice , Mice, Nude , Micelles , Polyethylene Glycols/chemistry , Survival Rate , Vitamin E/chemistry , alpha-Tocopherol/chemistry
5.
Cancer Res ; 74(19): 5507-19, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25085245

ABSTRACT

Extracellular tumor acidosis largely results from an exacerbated glycolytic flux in cancer and cancer-associated cells. Conversely, little is known about how tumor cells adapt their metabolism to acidosis. Here, we demonstrate that long-term exposure of cancer cells to acidic pH leads to a metabolic reprogramming toward glutamine metabolism. This switch is triggered by the need to reduce the production of protons from glycolysis and further maintained by the NAD(+)-dependent increase in SIRT1 deacetylase activity to ensure intracellular pH homeostasis. A consecutive increase in HIF2α activity promotes the expression of various transporters and enzymes supporting the reductive and oxidative glutamine metabolism, whereas a reduction in functional HIF1α expression consolidates the inhibition of glycolysis. Finally, in vitro and in vivo experiments document that acidosis accounts for a net increase in tumor sensitivity to inhibitors of SIRT1 and glutaminase GLS1. These findings highlight the influence that tumor acidosis and metabolism exert on each other.


Subject(s)
Acidosis/metabolism , Basic Helix-Loop-Helix Transcription Factors/physiology , Glutamine/metabolism , Neoplasms/therapy , Sirtuin 1/physiology , Cell Line, Tumor , Chronic Disease , Glucose/metabolism , Glycolysis , Humans , Neoplasms/metabolism
6.
Mol Cancer Ther ; 13(6): 1410-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24672058

ABSTRACT

High lactate concentration in tumors is associated with bad prognosis. Lactate is released by glycolytic cells in tumors and recaptured by oxidative cancer cells to feed the tricarboxylic acid (TCA) cycle after conversion into pyruvate. Monocarboxylate transporters (MCT) mediate these fluxes of proton-linked lactate and represent attractive targets to interrupt lactate shuttle and to inhibit tumor growth. Here, we investigated the properties of 7-aminocarboxycoumarins (7ACC) developed to selectively interfere with lactate fluxes in the lactate-rich tumor microenvironment. The pharmacologic properties of two compounds of this family, including their effects on lactate influx and efflux and antitumor activity, were investigated using human cancer cell lines and mouse xenograft models. Contrary to the reference MCT1 inhibitor AR-C155858, 7ACC unexpectedly inhibited lactate influx but not efflux in tumor cells expressing MCT1 and MCT4 transporters. 7ACC delayed the growth of cervix SiHa tumors, colorectal HCT116 tumors, and orthoptopic MCF-7 breast tumors. MCT target engagement was confirmed by the lack of activity of 7ACC on bladder UM-UC-3 carcinoma that does not express functional MCT. 7ACC also inhibited SiHa tumor relapse after treatment with cisplatin. Finally, we found that contrary to AR-C155858, 7ACC did not prevent the cell entry of the substrate-mimetic drug 3-bromopyruvate (3BP) through MCT1, and contributed to the inhibition of tumor relapse after 3BP treatment. In conclusion, our results indicate that 7ACC selectively affects a single part of the MCT symporter translocation cycle, leading to strict inhibition of lactate influx. This singular activity is associated with antitumor effects less prone to resistance and side effects.


Subject(s)
Breast Neoplasms/drug therapy , Colorectal Neoplasms/drug therapy , Coumarins/administration & dosage , Neoplasm Recurrence, Local/drug therapy , Animals , Breast Neoplasms/pathology , Colorectal Neoplasms/pathology , Coumarins/chemistry , Female , HCT116 Cells , Humans , Lactic Acid , MCF-7 Cells , Mice , Neoplasm Recurrence, Local/pathology , Prognosis , Thiophenes/administration & dosage , Uracil/administration & dosage , Uracil/analogs & derivatives
7.
Bioorg Med Chem ; 21(22): 7107-17, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24095010

ABSTRACT

Under hypoxia, cancer cells consume glucose and release lactate at a high rate. Lactate was recently documented to be recaptured by oxygenated cancer cells to fuel the TCA cycle and thereby to support tumor growth. Monocarboxylate transporters (MCT) are the main lactate carriers and therefore represent potential therapeutic targets to limit cancer progression. In this study, we have developed and implemented a stepwise in vitro screening procedure on human cancer cells to identify new potent MCT inhibitors. Various 7-substituted carboxycoumarins and quinolinone derivatives were synthesized and pharmacologically evaluated. Most active compounds were obtained using a palladium-catalyzed Buchwald-Hartwig type coupling reaction, which proved to be a quick and efficient method to obtain aminocarboxycoumarin derivatives. Inhibition of lactate flux revealed that the most active compound 19 (IC50 11 nM) was three log orders more active than the CHC reference compound. Comparison with warfarin, a conventional anticoagulant coumarin, further showed that compound 19 did not influence the prothrombin time which, together with a good in vitro ADME profile, supports the potential of this new family of compounds to act as anticancer drugs through inhibition of lactate flux.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Coumarins/chemical synthesis , Coumarins/pharmacology , Lactates/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Half-Life , Humans , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/metabolism , Quinolones/chemistry
8.
Dis Model Mech ; 4(6): 727-32, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22065843

ABSTRACT

Hypoxia and oncogene expression both stimulate glycolytic metabolism in tumors, thereby leading to lactate production. However, lactate is more than merely a by-product of glycolysis: it can be used as a metabolic fuel by oxidative cancer cells. This phenomenon resembles processes that have been described for skeletal muscle and brain that involve what are known as cell-cell and intracellular lactate shuttles. Two control points regulate lactate shuttles: the lactate dehydrogenase (LDH)-dependent conversion of lactate into pyruvate (and back), and the transport of lactate into and out of cells through specific monocarboxylate transporters (MCTs). In tumors, MCT4 is largely involved in hypoxia-driven lactate release, whereas the uptake of lactate into both tumor cells and tumor endothelial cells occurs via MCT1. Translating knowledge of lactate shuttles to the cancer field offers new perspectives to therapeutically target the hypoxic tumor microenvironment and to tackle tumor angiogenesis.


Subject(s)
Antineoplastic Agents/therapeutic use , Lactic Acid/metabolism , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Biological Transport/drug effects , Humans , L-Lactate Dehydrogenase/metabolism , Monocarboxylic Acid Transporters/metabolism , Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL