Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep ; 35(11): 109252, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34133926

ABSTRACT

Heme is an iron-containing porphyrin of vital importance for cell energetic metabolism. High rates of heme synthesis are commonly observed in proliferating cells. Moreover, the cell-surface heme exporter feline leukemia virus subgroup C receptor 1a (FLVCR1a) is overexpressed in several tumor types. However, the reasons why heme synthesis and export are enhanced in highly proliferating cells remain unknown. Here, we illustrate a functional axis between heme synthesis and heme export: heme efflux through the plasma membrane sustains heme synthesis, and implementation of the two processes down-modulates the tricarboxylic acid (TCA) cycle flux and oxidative phosphorylation. Conversely, inhibition of heme export reduces heme synthesis and promotes the TCA cycle fueling and flux as well as oxidative phosphorylation. These data indicate that the heme synthesis-export system modulates the TCA cycle and oxidative metabolism and provide a mechanistic basis for the observation that both processes are enhanced in cells with high-energy demand.


Subject(s)
Citric Acid Cycle , Heme/biosynthesis , Oxidative Phosphorylation , Animals , Biological Transport , Caco-2 Cells , Heme/metabolism , Humans , Membrane Transport Proteins/metabolism , Mice, Inbred C57BL , Mice, SCID , Receptors, Virus/metabolism , Xenograft Model Antitumor Assays
2.
Nat Commun ; 11(1): 6343, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33311467

ABSTRACT

D-mannose is a monosaccharide approximately a hundred times less abundant than glucose in human blood. Previous studies demonstrated that supraphysiological levels of D-mannose inhibit tumour growth and stimulate regulatory T cell differentiation. It is not known whether D-mannose metabolism affects the function of non-proliferative cells, such as inflammatory macrophages. Here, we show that D-mannose suppresses LPS-induced macrophage activation by impairing IL-1ß production. In vivo, mannose administration improves survival in a mouse model of LPS-induced endotoxemia as well as decreases progression in a mouse model of DSS-induced colitis. Phosphomannose isomerase controls response of LPS-activated macrophages to D-mannose, which impairs glucose metabolism by raising intracellular mannose-6-phosphate levels. Such alterations result in the suppression of succinate-mediated HIF-1α activation, imposing a consequent reduction of LPS-induced Il1b expression. Disclosing an unrecognized metabolic hijack of macrophage activation, our study points towards safe D-mannose utilization as an effective intervention against inflammatory conditions.


Subject(s)
Interleukin-1beta/metabolism , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Mannose/metabolism , Mannose/pharmacology , Animals , Cell Differentiation/drug effects , Cell Line , Colitis/metabolism , Colitis/pathology , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/metabolism , Interleukin-1beta/genetics , Lipopolysaccharides/adverse effects , Mannosephosphates/metabolism , Metabolic Networks and Pathways/drug effects , Metabolomics , Monocytes/metabolism
3.
Cells ; 9(2)2020 02 21.
Article in English | MEDLINE | ID: mdl-32098202

ABSTRACT

(1) Background: Extracellular nicotinamide phosphoribosyltrasferase (eNAMPT) is released by various cell types with pro-tumoral and pro-inflammatory properties. In cancer, eNAMPT regulates tumor growth through the activation of intracellular pathways, suggesting that it acts through a putative receptor, although its nature is still elusive. It has been shown, using surface plasma resonance, that eNAMPT binds to the C-C chemokine receptor type 5 (CCR5), although the physiological meaning of this finding is unknown. The aim of the present work was to characterize the pharmacodynamics of eNAMPT on CCR5. (2) Methods: HeLa CCR5-overexpressing stable cell line and B16 melanoma cells were used. We focused on some phenotypic effects of CCR5 activation, such as calcium release and migration, to evaluate eNAMPT actions on this receptor. (3) Results: eNAMPT did not induce ERK activation or cytosolic Ca2+-rises alone. Furthermore, eNAMPT prevents CCR5 internalization mediated by Rantes. eNAMPT pretreatment inhibits CCR5-mediated PKC activation and Rantes-dependent calcium signaling. The effect of eNAMPT on CCR5 was specific, as the responses to ATP and carbachol were unaffected. This was strengthened by the observation that eNAMPT inhibited Rantes-induced Ca2+-rises and Rantes-induced migration in a melanoma cell line. (4) Conclusions: Our work shows that eNAMPT binds to CCR5 and acts as a natural antagonist of this receptor.


Subject(s)
Cytokines/metabolism , Melanoma/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Receptors, CCR5/metabolism , Animals , Calcium/metabolism , Calcium Signaling/genetics , Cell Movement/genetics , Chemokine CCL5/metabolism , HEK293 Cells , HeLa Cells , Humans , Melanoma/pathology , Mice , Protein Binding/genetics , Protein Kinase C/metabolism , Receptors, CCR5/genetics , Recombinant Proteins/metabolism , Signal Transduction/genetics , Transfection
4.
J Biol Chem ; 295(11): 3635-3651, 2020 03 13.
Article in English | MEDLINE | ID: mdl-31988240

ABSTRACT

All cells require sustained intracellular energy flux, which is driven by redox chemistry at the subcellular level. NAD+, its phosphorylated variant NAD(P)+, and its reduced forms NAD(P)/NAD(P)H are all redox cofactors with key roles in energy metabolism and are substrates for several NAD-consuming enzymes (e.g. poly(ADP-ribose) polymerases, sirtuins, and others). The nicotinamide salvage pathway, constituted by nicotinamide mononucleotide adenylyltransferase (NMNAT) and nicotinamide phosphoribosyltransferase (NAMPT), mainly replenishes NAD+ in eukaryotes. However, unlike NMNAT1, NAMPT is not known to be a nuclear protein, prompting the question of how the nuclear NAD+ pool is maintained and how it is replenished upon NAD+ consumption. In the present work, using human and murine cells; immunoprecipitation, pulldown, and surface plasmon resonance assays; and immunofluorescence, small-angle X-ray scattering, and MS-based analyses, we report that GAPDH and NAMPT form a stable complex that is essential for nuclear translocation of NAMPT. This translocation furnishes NMN to replenish NAD+ to compensate for the activation of NAD-consuming enzymes by stressful stimuli induced by exposure to H2O2 or S-nitrosoglutathione and DNA damage inducers. These results indicate that by forming a complex with GAPDH, NAMPT can translocate to the nucleus and thereby sustain the stress-induced NMN/NAD+ salvage pathway.


Subject(s)
Cell Nucleus/enzymology , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , NAD/metabolism , Nicotinamide Mononucleotide/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Stress, Physiological , Animals , Cell Line, Tumor , HeLa Cells , Humans , Kinetics , Melanoma, Experimental/enzymology , Melanoma, Experimental/pathology , Mice , NIH 3T3 Cells , Nicotinamide Mononucleotide/chemistry , Nicotinamide Phosphoribosyltransferase/chemistry , Protein Binding , Protein Multimerization , Protein Transport
5.
Bioorg Med Chem ; 24(2): 140-52, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26705144

ABSTRACT

Curcumin is a unique blend of pharmacophores responsible for the pleiotropy of this natural pigment. In the present study we have replaced the 1,3-dicarbonyl moiety with a 1,2,3-triazole ring to furnish a new class of triazole-curcuminoids as a possible strategy to generate new compounds with different potency and selectivity compared to curcumin. We obtained a proof-of-principle library of 28 compounds tested for their cytotoxicity (SY-SY5Y and HeLa cells) and for their ability to inhibit NF-κB. Furthermore, we also generated 1,3-dicarbonyl curcuminoids of selected click compounds. Triazole-curcuminoids lost their ability to be Michael's acceptors, yet maintained some of the features of the parent compounds and disclosed new ones. In particular, we found that some compounds were able to inhibit NF-κB without showing cytotoxicity, while others, unlike curcumin, activated NF-κB signalling. This validates the hypothesis that click libraries can be used to investigate the biological activities of curcumin as well as generate analogs with selected features.


Subject(s)
Curcumin/pharmacology , NF-kappa B/antagonists & inhibitors , Triazoles/pharmacology , Cell Cycle/drug effects , Cell Survival/drug effects , Curcumin/chemistry , Dose-Response Relationship, Drug , HeLa Cells , Humans , Molecular Structure , NF-kappa B/metabolism , Structure-Activity Relationship , Triazoles/chemistry , Tumor Cells, Cultured
6.
Pigment Cell Melanoma Res ; 28(6): 718-29, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26358657

ABSTRACT

High plasma levels of nicotinamide phosphoribosyltransferase (NAMPT), traditionally considered an intracellular enzyme with a key role in NAD synthesis, have been reported in several oncological, inflammatory and metabolic diseases. We now show that eNAMPT can be actively released by melanoma cells in vitro. We analysed the mechanisms of its release, and we found both classical and non-classical pathway involvement. eNAMPT released by melanoma cells, in our hands, has paracrine and autocrine effects: it activates MAPK, AKT and NF-κB pathways and increases colony formation in anchorage-independent conditions. eNAMPT also induces M1 polarization in human monocytes. Last, we demonstrate, for the first time in any cancer type, that eNAMPT levels in plasma of tumour-bearing mice increase and that this increase can be reconducted to the tumour itself. This provides an important cue on previous observations that eNAMPT is increased in patients with cancer. Moreover, silencing NAMPT in melanoma cells leads to a reduction in the tumour growth rate. Our findings extend the basis to consider eNAMPT as a cytokine involved in tumour progression.


Subject(s)
Cytokines/metabolism , Melanoma/enzymology , Nicotinamide Phosphoribosyltransferase/metabolism , Skin Neoplasms/enzymology , Animals , Autocrine Communication/drug effects , Cell Hypoxia/drug effects , Cell Line, Tumor , Cytokines/blood , Extracellular Space/enzymology , Humans , Hydrogen Peroxide/pharmacology , Melanoma/pathology , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/metabolism , Nicotinamide Phosphoribosyltransferase/blood , Paracrine Communication/drug effects , Secretory Vesicles/metabolism , Skin Neoplasms/pathology
7.
J Med Chem ; 58(3): 1345-57, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25584687

ABSTRACT

In the present manuscript, starting from the 1,4-benzodiazepin-2-one nucleus, a privileged structure in medicinal chemistry, we have synthesized a novel class of cis-locked combretastatins named combreatabenzodiazepines. They show similar cytotoxic and antitubulin activity compared to combretastatin A-4 in neuroblastoma cells, showing a better pharmacokinetic profile. This class of compounds has therefore the potential for further development as antitubulin agents.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Benzodiazepinones/pharmacology , Drug Design , Tubulin Modulators/chemical synthesis , Tubulin Modulators/pharmacology , Tubulin/metabolism , Antineoplastic Agents/chemistry , Benzodiazepinones/chemical synthesis , Benzodiazepinones/chemistry , Cell Proliferation/drug effects , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Tubulin Modulators/chemistry , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...