Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Br J Cancer ; 107(8): 1337-44, 2012 Oct 09.
Article in English | MEDLINE | ID: mdl-22976799

ABSTRACT

BACKGROUND: Metabolites released by the gut microbiota may influence host metabolism and immunity. We have tested the hypothesis that inulin-type fructans (ITF), by promoting microbial production of short-chain fatty acids (SCFA), influence cancer cell proliferation outside the gut. METHODS: Mice transplanted with Bcr-Abl-transfected BaF3 cells, received ITF in their drinking water. Gut microbiota was analysed by 16S rDNA polymerase chain reaction (PCR)-denaturing gradient gel electrophoresis (DGGE) and qPCR. Serum Short-chain fatty acids were quantified by UHPLC-MS. Cell proliferation was evaluated in vivo, by molecular biology and histology, and in vitro. RESULTS: Inulin-type fructans treatment reduces hepatic BaF3 cell infiltration, lessens inflammation and increases portal propionate concentration. In vitro, propionate reduces BaF3 cell growth through a cAMP level-dependent pathway. Furthermore, the activation of free fatty acid receptor 2 (FFA2), a Gi/Gq-protein-coupled receptor also known as GPR43 and that binds propionate, lessens the proliferation of BaF3 and other human cancer cell lines. CONCLUSION: We show for the first time that the fermentation of nutrients such as ITF into propionate can counteract malignant cell proliferation in the liver tissue. Our results support the interest of FFA2 activation as a new strategy for cancer therapeutics. This study highlights the importance of research focusing on gut microbes-host interactions for managing systemic and severe diseases such as leukaemia.


Subject(s)
Fructans/administration & dosage , Intestines/microbiology , Leukemia/metabolism , Liver/pathology , Metagenome/immunology , Propionates/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Proliferation , Diet , Disease Models, Animal , Fatty Acids, Volatile/metabolism , Female , Fructans/metabolism , Fructans/pharmacology , Metagenome/drug effects , Mice , Mice, Inbred BALB C , Prebiotics
2.
iScience ; 25(6): 104435, 2022 Jun 17.
Article in English | MEDLINE | ID: mdl-35707720

ABSTRACT

Lactate sits at the crossroad of metabolism, immunity, and inflammation. The expression of cellular lactate transporter MCT1 (known as Slc16a1) increases during immune cell activation to cope with the metabolic reprogramming. We investigated the impact of MCT1 deficiency on CD8+ T cell function during obesity-related inflammatory conditions. The absence of MCT1 impaired CD8+ T cell proliferation with a shift of ATP production to mitochondrial oxidative phosphorylation. In Slc16a1 f/f Tcell cre mice fed a high-fat diet, a reduction in the number of CD8+ T cells, which infiltrated epididymal visceral adipose tissue (epiWAT) or subcutaneous adipose tissue, was observed. Adipose tissue weight and adipocyte area were significantly reduced together with downregulation of adipogenic genes only in the epiWAT. Our findings highlight a distinct effect of MCT1 deficiency in CD8+ T cells in the crosstalk with adipocytes and reinforce the concept that targeting immunometabolic reprogramming in lymphocyte could impact the immune-adipose tissue axis in obesity.

3.
Oncogene ; 36(19): 2637-2642, 2017 05 11.
Article in English | MEDLINE | ID: mdl-27797377

ABSTRACT

The Warburg effect and its accompanying metabolic features (anaplerosis, cataplerosis) are presented in textbooks and reviews as a hallmark (general characteristic): the metabolic map of cancer. On the other hand, research articles on specific tumors since a few years emphasize various biological features of different cancers, different cells in a cancer and the dynamic heterogeneity of these cells. We have analysed the research literature of the subject and show the generality of a dynamic, evolving biological and metabolic, spatial and temporal heterogeneity of individual cancers. We conclude that there is no one metabolic map of cancer but several and describe the two extremes of a panel from the hypoxic to the normoxic state. The implications for the significance of general 'omic' studies, and on therapeutic conclusions drawn from them and for the diagnostic use of fractional biopsies is discussed.


Subject(s)
Genetic Heterogeneity , Metabolic Networks and Pathways/genetics , Neoplasms/metabolism , Epithelial-Mesenchymal Transition/genetics , Glycolysis , Humans , Neoplasms/genetics
4.
Circ Res ; 89(10): 866-73, 2001 Nov 09.
Article in English | MEDLINE | ID: mdl-11701613

ABSTRACT

3-Hydroxy-3-methylglutaryl (HMG)-coenzyme A reductase inhibitors or statins exert direct beneficial effects on the endothelium in part through an increase in nitric oxide (NO) production. Here, we examined whether posttranslational modifications of the endothelial NO synthase (eNOS) could account for the proangiogenic effects of statins. We used endothelial cells (ECs) isolated from cardiac microvasculature, aorta, and umbilical veins, as well as dissected microvessels and aortic rings, that were cultured on reconstituted basement membrane matrix (Matrigel). Tube or precapillary formation was evaluated after statin treatment, in parallel with immunoblotting and immunoprecipitation experiments. Atorvastatin stimulated NO-dependent angiogenesis from both isolated and outgrowing (vessel-derived) ECs, independently of changes in eNOS expression. We found that in macro- but not microvascular ECs, atorvastatin stabilized tube formation through a decrease in caveolin abundance and its inhibitory interaction with eNOS. We also identified the chaperone protein hsp90 as a key target for the proangiogenic effects of statins. Using geldanamycin, an inhibitor of hsp90 function, and overexpression of recombinant hsp90, we documented that the statin-induced phosphorylation of eNOS on Ser1177 was directly dependent on the ability of hsp90 to recruit Akt in the eNOS complex. Finally, we showed that statin promoted the tyrosine phosphorylation of hsp90 and the direct interaction of hsp90 with Akt, which further potentiated the NO-dependent angiogenic processes. Our study provides new mechanistic insights into the NO-mediated angiogenic effects of statins and underscores the potential of these drugs and other modulators of hsp90 and caveolin abundance to promote neovascularization in disease states associated or not with atherosclerosis.


Subject(s)
Caveolins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neovascularization, Physiologic/drug effects , Nitric Oxide/metabolism , Protein Serine-Threonine Kinases , Animals , Atorvastatin , Benzoquinones , Biological Assay/methods , Cattle , Caveolin 1 , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , Heptanoic Acids/pharmacology , Humans , Immunoblotting , Lactams, Macrocyclic , Mice , Models, Cardiovascular , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Processing, Post-Translational , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Pyrroles/pharmacology , Quinones/pharmacology , Rats , Transfection
5.
Endocrinology ; 157(6): 2545-59, 2016 06.
Article in English | MEDLINE | ID: mdl-27035650

ABSTRACT

Iodine deficiency (ID) induces TSH-independent microvascular activation in the thyroid via the reactive oxygen species/nitric oxide-hypoxia-inducible factor-1α/vascular endothelial growth factor (VEGF) pathway. We hypothesized the additional involvement of mammalian target of rapamycin (mTOR) as a positive regulator of this pathway and AMP-activated protein kinase (AMPK) as a negative feedback regulator to explain the transient nature of ID-induced microvascular changes under nonmalignant conditions. mTOR and AMPK involvement was investigated using an in vitro model (human thyrocytes in primary cultures) and 2 murine models of goitrogenesis (normal NMRI and RET-PTC mice [a papillary thyroid cancer model]). In NMRI mice, ID had no effect on the phosphorylation of ribosomal S6 kinase (p70S6K), a downstream target of mTOR. However, rapamycin inhibited ID-induced thyroid blood flow and VEGF protein expression. In the RET-PTC model, ID strongly increased the phosphorylation of p70S6K, whereas rapamycin completely inhibited the ID-induced increase in p70S6K phosphorylation, thyroid blood flow, and VEGF-A expression. In vitro, although ID increased p70S6K phosphorylation, the ID-stimulated hypoxia-inducible factor/VEGF pathway was inhibited by rapamycin. Activation of AMPK by metformin inhibited ID effects both in vivo and in vitro. In AMPK-α1 knockout mice, the ID-induced increase in thyroid blood flow and VEGF-A protein expression persisted throughout the treatment, whereas both parameters returned to control values in wild-type mice after 4 days of ID. In conclusion, mTOR is required for early ID-induced thyroid microvascular activation. AMPK negatively regulates this pathway, which may account for the transient nature of ID-induced TSH-independent vascular effects under benign conditions.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Iodine/deficiency , TOR Serine-Threonine Kinases/metabolism , Thyroid Gland/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Cell Proliferation/genetics , Cell Proliferation/physiology , Female , Humans , In Vitro Techniques , Male , Metformin/metabolism , Mice , Mice, Knockout , Phosphorylation/drug effects , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/genetics , Thyroid Gland/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
6.
Biochim Biophys Acta ; 1440(2-3): 266-74, 1999 Sep 22.
Article in English | MEDLINE | ID: mdl-10521710

ABSTRACT

The presence of CB(2) receptors was reported in the rat basophilic cell line RBL-2H3 and N-palmitoylethanolamide was proposed as an endogenous, potent agonist of this receptor. We synthesized a series of 10 N-palmitoylethanolamide homologues and analogues, varying by the elongation of the fatty acid chain from caproyl to stearoyl and by the nature of the amide substituent, respectively, and evaluated the affinity of these compounds to cannabinoid receptors in the rat spleen, RBL-2H3 cells and CHO-CB(1) and CHO-CB(2) receptor-transfected cells. In rat spleen slices, CB(2) receptors were the predominant form of the cannabinoid receptors. No binding of [(3)H]SR141716A was observed. [(3)H]CP-55,940 binding was displaced by WIN 55,212-2 and anandamide. No displacement of [(3)H]CP-55,940 or [(3)H]WIN 55,212-2 by palmitoylethanolamide derivatives was observed in rat spleen slices. In RBL-2H3 cells, no binding of [(3)H]CP-55,940 or [(3)H]WIN 55,212-2 could be observed and conversely, no inhibitory activity of N-palmitoylethanolamide derivatives and analogues was measurable. These compounds do not recognize the human CB(1) and CB(2) receptors expressed in CHO cells. In conclusion, N-palmitoylethanolamide was, in our preparations, a weak ligand while its synthesized homologues or analogues were essentially inactive. Therefore, it seems unlikely that N-palmitoylethanolamide is an endogenous agonist of the CB(2) receptors but it may be a compound with potential therapeutic applications since it may act via other mechanisms than cannabinoid CB(1)-CB(2) receptor interactions.


Subject(s)
Cannabinoids/pharmacology , Ethanolamines/pharmacology , Receptor, Cannabinoid, CB2 , Receptors, Drug/drug effects , Animals , Benzoxazines , Binding, Competitive , CHO Cells , Cricetinae , Cyclohexanols/pharmacology , Humans , Molecular Structure , Morpholines/pharmacology , Naphthalenes/pharmacology , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Receptors, Cannabinoid , Receptors, Drug/antagonists & inhibitors , Receptors, Drug/genetics , Rimonabant , Spleen/drug effects , Spleen/metabolism , Transfection , Tumor Cells, Cultured/drug effects
7.
Endocrinology ; 156(2): 707-20, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25406019

ABSTRACT

Iodine deficiency (ID) induces microvascular changes in the thyroid gland via a TSH-independent reactive oxygen species-hypoxia inducible factor (HIF)-1α-vascular endothelial growth factor (VEGF) pathway. The involvement of nitric oxide (NO) in this pathway and the role of calcium (Ca(2+)) and of ryanodine receptors (RYRs) in NO synthase 3 (NOS3) activation were investigated in a murine model of goitrogenesis and in 3 in vitro models of ID, including primary cultures of human thyrocytes. ID activated NOS3 and the production of NO in thyrocytes in vitro and increased the thyroid blood flow in vivo. Using bevacizumab (a blocking antibody against VEGF-A) in mice, it appeared that NOS3 is activated upstream of VEGF-A. L-nitroarginine methyl ester (a NOS inhibitor) blocked the ID-induced increase in thyroid blood flow in vivo and NO production in vitro, as well as ID-induced VEGF-A mRNA and HIF-1α expression in vitro, whereas S-nitroso-acetyl-penicillamine (a NO donor) did the opposite. Ca(2+) is involved in this pathway as intracellular Ca(2+) flux increased after ID, and thapsigargin activated NOS3 and increased VEGF-A mRNA expression. Two of the 3 known mammalian RYR isoforms (RYR1 and RYR2) were shown to be expressed in thyrocytes. RYR inhibition using ryanodine at 10µM decreased ID-induced NOS3 activation, HIF-1α, and VEGF-A expression, whereas RYR activation with ryanodine at 1nM increased NOS3 activation and VEGF-A mRNA expression. In conclusion, during the early phase of TSH-independent ID-induced microvascular activation, ID sequentially activates RYRs and NOS3, thereby supporting ID-induced activation of the NO/HIF-1α/VEGF-A pathway in thyrocytes.


Subject(s)
Iodine/deficiency , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Thyroid Gland/blood supply , Animals , Calcium/metabolism , Cell Line , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Rats , Reactive Oxygen Species/metabolism , Vascular Endothelial Growth Factor A/metabolism
8.
Oncogene ; 33(31): 4060-8, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-24166504

ABSTRACT

The glycolytic end-product lactate is a pleiotropic tumor growth-promoting factor. Its activities primarily depend on its uptake, a process facilitated by the lactate-proton symporter monocarboxylate transporter 1 (MCT1). Therefore, targeting the transporter or its chaperon protein CD147/basigin, itself involved in the aggressive malignant phenotype, is an attractive therapeutic option for cancer, but basic information is still lacking regarding the regulation of the expression, interaction and activities of both proteins. In this study, we found that glucose deprivation dose-dependently upregulates MCT1 and CD147 protein expression and their interaction in oxidative tumor cells. While this posttranslational induction could be recapitulated using glycolysis inhibition, hypoxia, oxidative phosphorylation (OXPHOS) inhibitor rotenone or hydrogen peroxide, it was blocked with alternative oxidative substrates and specific antioxidants, pointing out at a mitochondrial control. Indeed, we found that the stabilization of MCT1 and CD147 proteins upon glucose removal depends on mitochondrial impairment and the associated generation of reactive oxygen species. When glucose was a limited resource (a situation occurring naturally or during the treatment of many tumors), MCT1-CD147 heterocomplexes accumulated, including in cell protrusions of the plasma membrane. It endowed oxidative tumor cells with increased migratory capacities towards glucose. Migration increased in cells overexpressing MCT1 and CD147, but it was inhibited in glucose-starved cells provided with an alternative oxidative fuel, treated with an antioxidant, lacking MCT1 expression, or submitted to pharmacological MCT1 inhibition. While our study identifies the mitochondrion as a glucose sensor promoting tumor cell migration, MCT1 is also revealed as a transducer of this response, providing a new rationale for the use of MCT1 inhibitors in cancer.


Subject(s)
Basigin/metabolism , Cell Movement , Glucose/metabolism , Mitochondria/metabolism , Monocarboxylic Acid Transporters/metabolism , Reactive Oxygen Species/metabolism , Symporters/metabolism , Uterine Cervical Neoplasms/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Glycolysis/physiology , HeLa Cells , Humans , Mitochondria/genetics , Monocarboxylic Acid Transporters/genetics , Symporters/genetics
9.
Contrast Media Mol Imaging ; 7(3): 302-7, 2012.
Article in English | MEDLINE | ID: mdl-22539400

ABSTRACT

MRI cell tracking is a promising technique to track various cell types (stem cells, tumor cells, etc.) in living animals. Usually, cells are incubated with iron oxides (T(2) contrast agent) in order to take up the particles before being injected in vivo. Iron oxide quantification is important in such studies for validating the labeling protocols and assessing the dilution of the particles with cell proliferation. We here propose to implement electron paramagnetic resonance (EPR) as a very sensitive method to quantify iron oxide concentration in cells. Iron oxide particles exhibit a unique EPR spectrum, which directly reflects the number of particles in a sample. In order to compare EPR with existing methods (Perls's Prussian blue reaction, ICP-MS and fluorimetry), we labeled tumor cells (melanoma and renal adenocarcinoma cell lines) and fibroblasts with fluorescent iron oxide particles, and determined the limits of detection of the different techniques. We show that EPR is a very sensitive technique and is specific for iron oxide quantification as measurements are not affected by endogenous iron. As a consequence, EPR is well adapted to perform ex vivo analysis of tissues after cell tracking experiments in order to confirm MRI results.


Subject(s)
Adenocarcinoma/chemistry , Electron Spin Resonance Spectroscopy , Ferric Compounds/analysis , Fibroblasts/chemistry , Kidney Neoplasms/chemistry , Magnetic Resonance Imaging , Melanoma, Experimental/chemistry , Adenocarcinoma/pathology , Animals , Cells, Cultured , Ferric Compounds/metabolism , Fibroblasts/cytology , Kidney Neoplasms/pathology , Kinetics , Limit of Detection , Luciferases/metabolism , Mass Spectrometry , Melanoma, Experimental/pathology , Mice , Microscopy, Fluorescence
10.
Int J Hyperthermia ; 22(4): 263-73, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16754348

ABSTRACT

PURPOSE: It has been well established that inadequate blood supply combined with high metabolic rates of oxygen consumption results in areas of low oxygen tension (<1%) within malignant tumours and that elevating tumour temperatures above 39 degrees Celsius results in significant improvement in tumour oxygenation. Macrophages play a dual role in tumour initiation and progression having both pro-tumour and anti-tumour effects. However, the response of macrophages to heat within a hypoxic environment has not yet been clearly defined. METHODS: Raw 264.7 murine macrophages were incubated under normoxia and chronic hypoxia at temperatures ranging from 37-43 degrees Celsius. Under normoxia at 41 degrees Celsius, macrophages start to release significant levels of superoxide. The combination of heat with hypoxia constitutes an additional stimulus leading to increased respiratory burst of macrophages. RESULTS: The high levels of superoxide were found to be associated with changes in macrophage production of pro-angiogenic cytokines. While hypoxia alone (37 degrees Celsius) increased levels of hypoxia inducible factor-1alpha (HIF-1alpha) in macrophages, the combination of hypoxia and mild hyperthermia (39-41 degrees Celsius) induced a strong reduction in HIF-1alpha expression. The HIF-regulated vascular endothelial growth factor (VEGF) decreased simultaneously, revealing that heat inhibits both HIF-1alpha stabilization and transcriptional activity. CONCLUSION: The data suggest that temperatures which are readily achievable in the clinic (39-41 degrees Celsius) might be optimal for maximizing hyperthermic response. At higher temperatures, these effects are reversed, thereby limiting the therapeutic benefits of more severe hyperthermic exposure.


Subject(s)
Hot Temperature , Hyperthermia, Induced , Hypoxia/metabolism , Macrophages/metabolism , Neovascularization, Physiologic/physiology , Reactive Oxygen Species/metabolism , Animals , Cell Line , Cytokines/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Mice , Neoplasms/metabolism , Neoplasms/therapy , Oxygen Consumption/physiology , Respiratory Burst/physiology , Superoxides/metabolism , Temperature , Vascular Endothelial Growth Factor A/metabolism
11.
J Biol Chem ; 276(35): 32663-9, 2001 Aug 31.
Article in English | MEDLINE | ID: mdl-11425855

ABSTRACT

Vascular endothelial growth factor (VEGF) exerts its angiogenic effects partly through the activation of endothelial nitric-oxide synthase (eNOS). Association with heat shock protein 90 (hsp90) and phosphorylation by Akt were recently shown to separately activate eNOS upon VEGF stimulation in endothelial cells. Here, we examined the interplay between these different mechanisms in VEGF-exposed endothelial cells. We documented that hsp90 binding to eNOS is, in fact, the crucial event triggering the transition from the Ca(2+)-dependent activation of eNOS to the phosphorylation-mediated potentiation of its activity by VEGF. Accordingly, we showed that early VEGF stimulation first leads to the Ca(2+)/calmodulin disruption of the caveolin-eNOS complex and promotes the association between eNOS and hsp90. eNOS-bound hsp90 can then recruit VEGF-activated (phosphorylated) Akt to the complex, which in turn can phosphorylate eNOS. Further experiments in transfected COS cells expressing either wild-type or S1177A mutant eNOS led us to identify the serine 1177 as the critical residue for the hsp90-dependent Akt-mediated activation of eNOS. Finally, we documented that although the VEGF-induced phosphorylation of eNOS leads to a sustained production of NO independently of a maintained increase in [Ca(2+)](i), this late stage of eNOS activation is strictly conditional on the initial VEGF-induced Ca(2+)-dependent stimulation of the enzyme. These data establish the critical temporal sequence of events leading to the sustained activation of eNOS by VEGF and suggest new ways of regulating the production of NO in response to this cytokine through the ubiquitous chaperone protein, hsp90.


Subject(s)
Calcium/metabolism , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/metabolism , HSP90 Heat-Shock Proteins/metabolism , Lymphokines/pharmacology , Nitric Oxide Synthase/metabolism , Protein Serine-Threonine Kinases , Animals , Animals, Newborn , COS Cells , Calcimycin/pharmacology , Caveolin 1 , Caveolins/metabolism , Cells, Cultured , Chlorocebus aethiops , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme Activation , HSP90 Heat-Shock Proteins/isolation & purification , Humans , Insulin/pharmacology , Kinetics , Mice , Nitric Oxide Synthase/isolation & purification , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Nitrites/metabolism , Phosphorylation , Protein Binding , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Recombinant Proteins/metabolism , Transfection , Umbilical Veins , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL