Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 336
Filtrer
1.
Mol Biol Cell ; 31(17): 1835-1845, 2020 08 01.
Article de Anglais | MEDLINE | ID: mdl-32583743

RÉSUMÉ

Fig4 is a phosphoinositide phosphatase that converts PI3,5P2 to PI3P. Paradoxically, mutation of Fig4 results in lower PI3,5P2, indicating that Fig4 is also required for PI3,5P2 production. Fig4 promotes elevation of PI3,5P2, in part, through stabilization of a protein complex that includes its opposing lipid kinase, Fab1, and the scaffold protein Vac14. Here we show that multiple regions of Fig4 contribute to its roles in the elevation of PI3,5P2: its catalytic site, an N-terminal disease-related surface, and a C-terminal region. We show that mutation of the Fig4 catalytic site enhances the formation of the Fab1-Vac14-Fig4 complex, and reduces the ability to elevate PI3,5P2. This suggests that independent of its lipid phosphatase function, the active site plays a role in the Fab1-Vac14-Fig4 complex. We also show that the N-terminal disease-related surface contributes to the elevation of PI3,5P2 and promotes Fig4 association with Vac14 in a manner that requires the Fig4 C-terminus. We find that the Fig4 C-terminus alone interacts with Vac14 in vivo and retains some functions of full-length Fig4. Thus, a subset of Fig4 functions are independent of its phosphatase domain and at least three regions of Fig4 play roles in the function of the Fab1-Vac14-Fig4 complex.


Sujet(s)
Flavoprotéines/métabolisme , Phosphoric monoester hydrolases/métabolisme , Phosphotransferases (Alcohol Group Acceptor)/métabolisme , Protéines de Saccharomyces cerevisiae/métabolisme , Flavoprotéines/physiologie , Protéines et peptides de signalisation intracellulaire/métabolisme , Lipides/physiologie , Protéines membranaires/métabolisme , Phosphates phosphatidylinositol/métabolisme , Phosphoinositide phosphatases/métabolisme , Phosphoric monoester hydrolases/physiologie , Phosphotransferases (Alcohol Group Acceptor)/physiologie , Liaison aux protéines , Saccharomyces cerevisiae/métabolisme , Protéines de Saccharomyces cerevisiae/physiologie
2.
Proc Natl Acad Sci U S A ; 115(51): 12967-12972, 2018 12 18.
Article de Anglais | MEDLINE | ID: mdl-30510005

RÉSUMÉ

Ferredoxin-dependent thioredoxin reductase was identified 35 y ago in the fermentative bacterium Clostridium pasteurianum [Hammel KE, Cornwell KL, Buchanan BB (1983) Proc Natl Acad Sci USA 80:3681-3685]. The enzyme, a flavoprotein, was strictly dependent on ferredoxin as reductant and was inactive with either NADPH or NADH. This early work has not been further pursued. We have recently reinvestigated the problem and confirmed that the enzyme, here designated ferredoxin-dependent flavin thioredoxin reductase (FFTR), is a flavoprotein. The enzyme differs from ferredoxin-thioredoxin reductase (FTR), which has a signature [4Fe-4S] cluster, but shows structural similarities to NADP-dependent thioredoxin reductase (NTR). Comparative amino acid sequence analysis showed that FFTR is present in a number of clostridial species, some of which lack both FTR and an archetypal NTR. We have isolated, crystallized, and determined the structural properties of FFTR from a member of this group, Clostridium acetobutylicum, both alone and in complex with Trx. The structures showed an elongated FFTR homodimer, each monomer comprising two Rossmann domains and a noncovalently bound FAD cofactor that exposes the isoalloxazine ring to the solvent. The FFTR structures revealed an alternative domain organization compared with NTR that enables the enzyme to accommodate Fdx rather than NADPH. The results suggest that FFTR exists in a range of conformations with varying degrees of domain separation in solution and that the stacking between the two redox-active groups for the transfer of reducing equivalents results in a profound structural reorganization. A mechanism in accord with the findings is proposed.


Sujet(s)
Clostridium acetobutylicum/enzymologie , Ferrédoxines/composition chimique , Flavoprotéines/composition chimique , Cristallographie aux rayons X , Flavoprotéines/métabolisme , Flavoprotéines/physiologie , Modèles moléculaires , NADP/composition chimique , Oxydoréduction , Conformation des protéines , Analyse de séquence de protéine , Similitude de séquences
3.
Brain ; 139(Pt 12): 3121-3136, 2016 12.
Article de Anglais | MEDLINE | ID: mdl-27797811

RÉSUMÉ

SEE PLUCHINO AND PERUZZOTTI-JAMETTI DOI101093/AWW266 FOR A SCIENTIFIC COMMENTARY ON THIS ARTICLE: Myelin regeneration (remyelination) is a spontaneous process that occurs following central nervous system demyelination. However, for reasons that remain poorly understood, remyelination fails in the progressive phase of multiple sclerosis. Emerging evidence indicates that alternatively activated macrophages in central nervous system lesions are required for oligodendrocyte progenitor differentiation into remyelinating oligodendrocytes. Here, we show that an alternatively activated macrophage secreted enzyme, interleukin-four induced one (IL4I1), is upregulated at the onset of inflammation resolution and remyelination in mouse central nervous system lesions after lysolecithin-induced focal demyelination. Focal demyelination in mice lacking IL4I1 or interleukin 4 receptor alpha (IL4Rα) results in increased proinflammatory macrophage density, remyelination impairment, and axonal injury in central nervous system lesions. Conversely, recombinant IL4I1 administration into central nervous system lesions reduces proinflammatory macrophage density, enhances remyelination, and rescues remyelination impairment in IL4Rα deficient mice. We find that IL4I1 does not directly affect oligodendrocyte differentiation, but modulates inflammation by reducing interferon gamma and IL17 expression in lesioned central nervous system tissues, and in activated T cells from splenocyte cultures. Remarkably, intravenous injection of IL4I1 into mice with experimental autoimmune encephalomyelitis at disease onset significantly reversed disease severity, resulting in recovery from hindlimb paralysis. Analysis of post-mortem tissues reveals reduced axonal dystrophy in spinal cord, and decreased CD4+ T cell populations in spinal cord and spleen tissues. These results indicate that IL4I1 modulates inflammation by regulating T cell expansion, thereby permitting the formation of a favourable environment in the central nervous system tissue for remyelination. Therefore, IL4I1 is a potentially novel therapeutic for promoting central nervous system repair in multiple sclerosis.


Sujet(s)
Axones/métabolisme , Lymphocytes T CD4+/métabolisme , Encéphalomyélite auto-immune expérimentale/prévention et contrôle , Flavoprotéines/physiologie , Inflammation/métabolisme , Macrophages/métabolisme , Gaine de myéline/métabolisme , Régénération nerveuse/physiologie , Animaux , Femelle , Flavoprotéines/pharmacologie , Inflammation/traitement médicamenteux , L-Amino acid oxidase , Mâle , Souris , Souris de lignée C57BL , Régénération nerveuse/effets des médicaments et des substances chimiques
4.
Biochem Biophys Res Commun ; 440(2): 342-7, 2013 Oct 18.
Article de Anglais | MEDLINE | ID: mdl-24070605

RÉSUMÉ

The phosphoinositide 5-kinase PIKfyve and 5-phosphatase Sac3 are scaffolded by ArPIKfyve in the PIKfyve-ArPIKfyve-Sac3 (PAS) regulatory complex to trigger a unique loop of PtdIns3P-PtdIns(3,5)P2 synthesis and turnover. Whereas the metabolizing enzymes of the other 3-phosphoinositides have already been implicated in breast cancer, the role of the PAS proteins and the PtdIns3P-PtdIns(3,5)P2 conversion is unknown. To begin elucidating their roles, in this study we monitored the endogenous levels of the PAS complex proteins in cell lines derived from hormone-receptor positive (MCF7 and T47D) or triple-negative breast cancers (TNBC) (BT20, BT549 and MDA-MB-231) as well as in MCF10A cells derived from non-tumorigenic mastectomy. We report profound upregulation of Sac3 and ArPIKfyve in the triple negative vs. hormone-sensitive breast cancer or non-tumorigenic cells, with BT cell lines showing the highest levels. siRNA-mediated knockdown of Sac3, but not that of PIKfyve, significantly inhibited proliferation of BT20 and BT549 cells. In these cells, knockdown of ArPIKfyve had only a minor effect, consistent with a primary role for Sac3 in TNBC cell proliferation. Intriguingly, steady-state levels of PtdIns(3,5)P2 in BT20 and T47D cells were similar despite the 6-fold difference in Sac3 levels between these cell lines. However, steady-state levels of PtdIns3P and PtdIns5P, both regulated by the PAS complex, were significantly reduced in BT20 vs. T47D or MCF10A cell lines, consistent with elevated Sac3 affecting directly or indirectly the homeostasis of these lipids in TNBC. Together, our results uncover an unexpected role for Sac3 phosphatase in TNBC cell proliferation. Database analyses, discussed herein, reinforce the involvement of Sac3 in breast cancer pathogenesis.


Sujet(s)
Flavoprotéines/physiologie , Protéines membranaires/physiologie , Phosphatidylinositol 3-kinases/physiologie , Phosphates phosphatidylinositol/métabolisme , Tumeurs du sein triple-négatives/physiopathologie , Femelle , Humains , Protéines et peptides de signalisation intracellulaire , Phosphoric monoester hydrolases , Tumeurs du sein triple-négatives/génétique
5.
J Cardiovasc Pharmacol ; 61(5): 369-77, 2013 May.
Article de Anglais | MEDLINE | ID: mdl-23318991

RÉSUMÉ

Cardiac mitochondria and the sarcolemmal (sarc)KATP channels contribute to cardioprotective signaling of anesthetic-induced preconditioning. Changes in mitochondrial bioenergetics influence the sarcolemmal ATP-sensitive K (sarcKATP) channel function, but whether this channel has impacts on mitochondria is uncertain. We used the mouse model with deleted pore-forming Kir6.2 subunit of sarcKATP channel (Kir6.2 KO) to investigate whether the functional sarcKATP channels are necessary for isoflurane activation of mitochondrial protective mechanisms. Ventricular cardiomyocytes were isolated from C57Bl6 wild-type (WT) and Kir6.2 KO mouse hearts. Flavoprotein autofluorescence, mitochondrial reactive oxygen species production, and mitochondrial membrane potential were monitored by laser-scanning confocal microscopy in intact cardiomyocytes. Cell survival was assessed using H2O2-induced stress. Isoflurane (0.5 mM) increased flavoprotein fluorescence to 180% ± 14% and 190% ± 15% and reactive oxygen species production to 118% ± 2% and 124% ± 6% of baseline in WT and Kir6.2 KO myocytes, respectively. Tetramethylrhodamine ethyl ester fluorescence decreased to 84% ± 6% in WT and to 86% ± 4% in Kir6.2 KO myocytes. This effect was abolished by 5HD. Pretreatment with isoflurane decreased the stress-induced cell death from 31% ± 1% to 21% ± 1% in WT and from 44% ± 2% to 35% ± 2% in Kir6.2 KO myocytes. In conclusion, Kir6.2 deletion increases the sensitivity of intact cardiomyocytes to oxidative stress, but does not alter the isoflurane-elicited protective mitochondrial mechanisms, suggesting independent roles for cardiac mitochondria and sarcKATP channels in anesthetic-induced preconditioning by isoflurane.


Sujet(s)
Anesthésiques par inhalation/pharmacologie , Préconditionnement ischémique myocardique , Isoflurane/pharmacologie , Mitochondries du myocarde/effets des médicaments et des substances chimiques , Myocytes cardiaques/effets des médicaments et des substances chimiques , Canaux potassiques rectifiants entrants/métabolisme , Sarcolemme/effets des médicaments et des substances chimiques , Animaux , Survie cellulaire , Flavoprotéines/effets des médicaments et des substances chimiques , Flavoprotéines/physiologie , Fluorescence , Ventricules cardiaques/cytologie , Mâle , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Souris , Souris de lignée C57BL , Microscopie confocale , Mitochondries du myocarde/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme
6.
Curr Pharm Des ; 19(14): 2540-51, 2013.
Article de Anglais | MEDLINE | ID: mdl-23116393

RÉSUMÉ

Renalase is a flavoprotein recently discovered in humans, preferentially expressed in the proximal tubules of the kidney and secreted in blood and urine. It is highly conserved in vertebrates, with homologs identified in eukaryotic and prokaryotic organisms. Several genetic, epidemiological, clinical and experimental studies show that renalase plays a role in the modulation of the functions of the cardiovascular system, being particularly active in decreasing the catecholaminergic tone, in lowering blood pressure and in exerting a protective action against myocardial ischemic damage. Deficient renalase synthesis might be the cause of the high occurrence of hypertension and adverse cardiac events in kidney disease patients. Very recently, recombinant human renalase has been structurally and functionally characterized in vitro. Results show that it belongs to the p-hydroxybenzoate hydroxylase structural family of flavoenzymes, contains non-covalently bound FAD with redox features suggestive of a dehydrogenase activity, and is not a catecholamine-degrading enzyme,either through oxidase or NAD(P)H-dependent monooxygenase reactions. The biochemical data now available will hopefully provide the basis for a systematic and rational quest toward the identification of the reaction catalyzed by renalase and of the molecular mechanism of its physiological action, which in turn are expected to favor the development of novel therapeutic tools for the treatment of kidney and cardiovascular diseases.


Sujet(s)
Catécholamines/métabolisme , Flavoprotéines/physiologie , Monoamine oxidase/physiologie , Transduction du signal/physiologie , Animaux , Sites de fixation , Maladies cardiovasculaires/enzymologie , Maladies cardiovasculaires/épidémiologie , Maladies cardiovasculaires/génétique , Découverte de médicament , Flavoprotéines/composition chimique , Flavoprotéines/génétique , Humains , Maladies du rein/enzymologie , Maladies du rein/épidémiologie , Maladies du rein/génétique , Monoamine oxidase/composition chimique , Monoamine oxidase/génétique , Polymorphisme de nucléotide simple , Conformation des protéines , Facteurs de risque
7.
Biochem Biophys Res Commun ; 430(1): 137-43, 2013 Jan 04.
Article de Anglais | MEDLINE | ID: mdl-23159617

RÉSUMÉ

Nitric oxide (NO) is a ubiquitous signaling molecule involved in the regulation of a large number of cellular functions. In the unicellular eukaryote yeast, NO may be involved in stress response pathways, but its role is poorly understood due to the lack of mammalian NO synthase (NOS) orthologues. Previously, we have proposed the oxidative stress-induced l-arginine synthesis and its physiological role under stress conditions in yeast Saccharomyces cerevisiae. Here, our experimental results indicated that increased conversion of l-proline into l-arginine led to NO production in response to elevated temperature. We also showed that the flavoprotein Tah18, which was previously reported to transfer electrons to the Fe-S cluster protein Dre2, was involved in NO synthesis in yeast. Gene knockdown analysis demonstrated that Tah18-dependent NO synthesis confers high-temperature stress tolerance on yeast cells. As it appears that such a unique cell protection mechanism is specific to yeasts and fungi, it represents a promising target for antifungal activity.


Sujet(s)
Flavoprotéines/physiologie , Température élevée , Monoxyde d'azote/biosynthèse , Stress oxydatif/physiologie , Oxidoreductases/physiologie , Protéines de Saccharomyces cerevisiae/physiologie , Saccharomyces cerevisiae/physiologie , Arginine/métabolisme , Flavoprotéines/génétique , Techniques de knock-down de gènes , Ferrosulfoprotéines/métabolisme , Stress oxydatif/génétique , Oxidoreductases/génétique , Proline/métabolisme , Saccharomyces cerevisiae/génétique , Protéines de Saccharomyces cerevisiae/génétique , Protéines de Saccharomyces cerevisiae/métabolisme
8.
J Neurosci ; 32(46): 16141-8, 2012 Nov 14.
Article de Anglais | MEDLINE | ID: mdl-23152598

RÉSUMÉ

Flavoprotein autofluorescence imaging was used to examine auditory cortical synaptic responses in aged animals with behavioral evidence of tinnitus and hearing loss. Mice were exposed to noise trauma at 1-3 months of age and were assessed for behavioral evidence of tinnitus and hearing loss immediately after the noise trauma and again at ~24-30 months of age. Within 2 months of the final behavioral assessment, auditory cortical synaptic transmission was examined in brain slices using electrical stimulation of putative thalamocortical afferents, and flavoprotein autofluorescence imaging was used to measure cortical activation. Noise-exposed animals showed a 68% increase in amplitude of cortical activation compared with controls (p = 0.008), and these animals showed a diminished sensitivity to GABA(A)ergic blockade (p = 0.008, using bath-applied 200 nm SR 95531 [6-Imino-3-(4-methoxyphenyl)-1(6H)-p yridazinebutanoic acid hydrobromide]). The strength of cortical activation was significantly correlated to the degree of tinnitus behavior, assessed via a loss of gap detection in a startle paradigm. The decrease in GABA(A) sensitivity was greater in the regions of the cortex farther away from the stimulation site, potentially reflecting a greater sensitivity of corticocortical versus thalamocortical projections to the effects of noise trauma. Finally, there was no relationship between auditory cortical activation and activation of the somatosensory cortex in the same slices, suggesting that the increases in auditory cortical activation were not attributable to a generalized hyperexcitable state in noise-exposed animals. These data suggest that noise trauma can cause long-lasting changes in the auditory cortical physiology and may provide specific targets to ameliorate the effects of chronic tinnitus.


Sujet(s)
Cortex cérébral/physiopathologie , Inhibition nerveuse/physiologie , Acouphène/physiopathologie , Stimulation acoustique , Vieillissement/physiologie , Animaux , Cortex auditif/physiologie , Seuil auditif/physiologie , Maladie chronique , Modèles animaux de maladie humaine , Potentiels évoqués auditifs du tronc cérébral/physiologie , Flavoprotéines/physiologie , Antagonistes GABA/pharmacologie , Surdité due au bruit/physiopathologie , Mâle , Souris , Souris de lignée CBA , N-Méthyl-aspartate/physiologie , Pyridazines/pharmacologie , Récepteurs GABA-A/effets des médicaments et des substances chimiques , Réflexe de sursaut/physiologie , Acide gamma-amino-butyrique/physiologie
10.
FEBS J ; 279(18): 3449-61, 2012 Sep.
Article de Anglais | MEDLINE | ID: mdl-22329503

RÉSUMÉ

Purple bacteria derive energy from aerobic respiration or photosynthesis depending on the availability of oxygen and light. Under aerobic conditions, photosynthesis genes are specifically repressed by the PpsR protein. In Rhodobacter sphaeroides, the repressive action of PpsR is antagonized by the blue-light and redox-sensitive flavoprotein AppA, which sequesters PpsR under anaerobic conditions into transcriptionally inactive complexes. However, under semi-aerobic conditions, blue-light excitation of AppA causes the AppA-PpsR complexes to dissociate, again leading to a repression of photosynthesis genes. We have recently developed a simple mathematical model suggesting that this phenotype arises from the formation of a maximum in the response curve of reduced PpsR at intermediate oxygen concentrations. However, this model focused mainly on the oxygen-dependent interactions whereas light regulation was only implemented in a simplified manner. In the present study, we incorporate a more detailed mechanism for the light-dependent interaction between AppA and PpsR, which now allows for a direct comparison with experiments. Specifically, we take into account that, upon blue-light excitation, AppA undergoes a conformational change, creating a long-lived signalling state causing the dissociation of the AppA-PpsR complexes. The predictions of the extended model are found to be in good agreement with experimental results on the light-dependent repression of photosynthesis genes under semi-aerobic conditions. We also identify the potential kinetic and stoichiometric constraints that the interplay between light and redox regulation imposes on the functionality of the AppA/PpsR system, especially with respect to a possible bistable response.


Sujet(s)
Protéines bactériennes/physiologie , Flavoprotéines/physiologie , Photorécepteurs microbiens/physiologie , Photosynthèse/génétique , Complexe protéique du centre réactionnel de la photosynthèse/métabolisme , Protéines de répression/physiologie , Rhodobacter sphaeroides/génétique , Simulation numérique , Régulation de l'expression des gènes bactériens , Cinétique , Lumière , Modèles biologiques , Oxydoréduction , Rhodobacter sphaeroides/métabolisme
11.
Proc Natl Acad Sci U S A ; 109(9): 3481-6, 2012 Feb 28.
Article de Anglais | MEDLINE | ID: mdl-22308473

RÉSUMÉ

Legionella pneumophila directs the formation of a specialized vacuole within host cells, dependent on protein substrates of the Icm/Dot translocation system. Survival of the host cell is essential for intracellular replication of L. pneumophila. Strains lacking the translocated substrate SdhA are defective for intracellular replication and activate host cell death pathways in primary macrophages. To understand how SdhA promotes evasion of death pathways, we performed a mutant hunt to identify bacterial suppressors of the ΔsdhA growth defect. We identified the secreted phospholipase PlaA as key to activation of death pathways by the ΔsdhA strain. Based on homology between PlaA and SseJ, a Salmonella protein associated with vacuole degradation, we determined the roles of SdhA and PlaA in controlling vacuole integrity. In the absence of sdhA, the Legionella-containing vacuole was unstable, resulting in access to the host cytosol. Both vacuole disruption and host cell death were largely dependent on PlaA. Consistent with these observations, the ΔsdhA strain colocalized with galectin-3, a marker of vacuole rupture, in a PlaA-dependent process. Access of ΔsdhA strains to the macrophage cytosol triggered multiple responses in the host cell, including degradation of bacteria, induction of the type I IFN response, and activation of inflammasomes. Therefore, we have demonstrated that the Legionella-containing vacuole is actively stabilized by the SdhA protein during intracellular replication. This vacuolar niche affords the bacterium protection from cytosolic host factors that degrade bacteria and initiate immune responses.


Sujet(s)
Protéines bactériennes/physiologie , Flavoprotéines/physiologie , Legionella pneumophila/physiologie , Macrophages/microbiologie , Phospholipases/physiologie , Vacuoles/microbiologie , Séquence d'acides aminés , Animaux , Protéines bactériennes/génétique , Marqueurs biologiques , Mort cellulaire , Cytosol/microbiologie , Flagelline/génétique , Flagelline/métabolisme , Flavoprotéines/génétique , Galectine -3/analyse , Délétion de gène , Interactions hôte-pathogène , Humains , Legionella pneumophila/génétique , Macrophages/ultrastructure , Souris , Souris de lignée A , Données de séquences moléculaires , Mutagenèse dirigée , Phospholipases/génétique , Alignement de séquences , Similitude de séquences d'acides aminés , Cellules U937
12.
Mol Pharmacol ; 81(1): 31-40, 2012 Jan.
Article de Anglais | MEDLINE | ID: mdl-21984255

RÉSUMÉ

The clinical agent PR-104 is converted systemically to PR-104A, a nitrogen mustard prodrug designed to target tumor hypoxia. Reductive activation of PR-104A is initiated by one-electron oxidoreductases in a process reversed by oxygen. The identity of these oxidoreductases is unknown, with the exception of cytochrome P450 reductase (POR). To identify other hypoxia-selective PR-104A reductases, nine candidate oxidoreductases were expressed in HCT116 cells. Increased PR-104A-cytotoxicity was observed in cells expressing methionine synthase reductase (MTRR), novel diflavin oxidoreductase 1 (NDOR1), and inducible nitric-oxide synthase (NOS2A), in addition to POR. Plasmid-based expression of these diflavin oxidoreductases also enhanced bioreductive metabolism of PR-104A in an anoxia-specific manner. Diflavin oxidoreductase-dependent PR-104A metabolism was suppressed >90% by pan-flavoenzyme inhibition with diphenyliodonium chloride. Antibodies were used to quantify endogenous POR, MTRR, NDOR1, and NOS2A expression in 23 human tumor cell lines; however, only POR protein was detectable and its expression correlated with anoxic PR-104A reduction (r(2) = 0.712). An anti-POR monoclonal antibody was used to probe expression using human tissue microarrays; 13 of 19 cancer types expressed detectable POR with 21% of cores (185 of 874) staining positive; this heterogeneity suggests that POR is a useful biomarker for PR-104A activation. Immunostaining for carbonic anhydrase 9 (CAIX), reportedly an endogenous marker of hypoxia, revealed only moderate coexpression (9.6%) of both CAIX and POR across a subset of five cancer types.


Sujet(s)
Flavines/physiologie , Flavoprotéines/physiologie , Moutardes à l'azote/métabolisme , Oxidoreductases/physiologie , Promédicaments/pharmacologie , Animaux , Hypoxie cellulaire/effets des médicaments et des substances chimiques , Hypoxie cellulaire/physiologie , Flavoprotéines/métabolisme , Cellules HCT116 , Humains , Souris , Souris nude , NADPH-ferrihemoprotéine reductase/physiologie , Moutardes à l'azote/pharmacologie , Oxydoréduction , Oxidoreductases/métabolisme
13.
Neuroreport ; 21(13): 861-4, 2010 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-20647961

RÉSUMÉ

Two classes of thalamic nuclei project to either middle layers or upper layers, including layer 1, of the neocortex, and are referred to as 'specific' and 'nonspecific' nuclei, respectively. The electrophysiological properties of the nonspecific nuclei have not been investigated, largely because of the paucity of in vitro slice preparations containing intact nonspecific pathways. In this study, we used flavoprotein autofluorescence imaging to show intact thalamocortical connectivity of nonspecific nuclei in slice preparations of the somatosensory and auditory systems. These preparations will enable the elucidation of electrophysiological properties of nonspecific pathways.


Sujet(s)
Cortex auditif/physiologie , Voies nerveuses/physiologie , Cortex somatosensoriel/physiologie , Thalamus/physiologie , Animaux , Cortex auditif/anatomie et histologie , Stimulation électrique , Flavoprotéines/métabolisme , Flavoprotéines/physiologie , Fluorescence , Traitement d'image par ordinateur , Techniques in vitro , Souris , Souris de lignée BALB C , Voies nerveuses/anatomie et histologie , Cortex somatosensoriel/anatomie et histologie , Thalamus/anatomie et histologie
15.
FEBS J ; 276(16): 4313-24, 2009 Aug.
Article de Anglais | MEDLINE | ID: mdl-19624732

RÉSUMÉ

Quinone reductases are ubiquitous soluble enzymes found in bacteria, fungi, plants and animals. These enzymes utilize a reduced nicotinamide such as NADH or NADPH to reduce the flavin cofactor (either FMN or FAD), which then affords two-electron reduction of cellular quinones. Although the chemical nature of the quinone substrate is still a matter of debate, the reaction appears to play a pivotal role in quinone detoxification by preventing the generation of potentially harmful semiquinones. In recent years, an additional role of quinone reductases as regulators of proteasomal degradation of transcription factors and possibly intrinsically unstructured protein has emerged. To fulfil this role, quinone reductase binds to the core particle of the proteasome and recruits certain transcription factors such as p53 and p73alpha to the complex. The latter process appears to be governed by the redox state of the flavin cofactor of the quinone reductase, thus linking the stability of transcription factors to cellular events such as oxidative stress. Here, we review the current evidence for protein complex formation between quinone reductase and the 20S proteasome in eukaryotic cells and describe the regulatory role of this complex in stabilizing transcription factors by acting as inhibitors of their proteasomal degradation.


Sujet(s)
Flavoprotéines/physiologie , Proteasome endopeptidase complex/métabolisme , Quinone reductases/physiologie , Flavoprotéines/métabolisme , Oxydoréduction , Stabilité protéique , Quinone reductases/métabolisme , Facteurs de transcription/composition chimique , Facteurs de transcription/métabolisme
16.
FEBS J ; 276(16): 4304-12, 2009 Aug.
Article de Anglais | MEDLINE | ID: mdl-19624733

RÉSUMÉ

Lysine-specific demethylase 1 (LSD1) is an enzyme that removes methyl groups from mono- and dimethylated Lys4 of histone H3, a post-translational modification associated with gene activation. Human LSD1 was the first histone demethylase to be discovered and this enzymatic activity is conserved among eukaryotes. LSD1 has been identified in a number of chromatin-remodeling complexes that control gene transcription and its demethylase activity has also been linked to pathological processes including tumorigenesis. The 852-residue sequence of LSD1 comprises an amine oxidase domain which identifies a family of enzymes that catalyze the FAD-dependent oxidation of amine substrates ranging from amino acids to aromatic neurotransmitters. Among these proteins, LSD1 is peculiar in that it acts on a protein substrate in the nuclear environment of chromatin-remodeling complexes. This functional divergence occurred during evolution from the eubacteria to eukaryotes by acquisition of additional domains such as the SWIRM domain. The N-terminal part of LSD1, predicted to be disordered, contains linear motifs that might represent functional sites responsible for the association of this enzyme with a variety of transcriptional protein complexes. LSD1 shares structural features with other flavin amine oxidases, including the overall fold of the amine oxidase domain region and details in the active site that are relevant for amine substrate oxidation.


Sujet(s)
Chromatine/métabolisme , Flavoprotéines/physiologie , Régulation de l'expression des gènes , Oxidoreductases, (N-demethylating)/physiologie , Animaux , Protéines bactériennes , Protéines fongiques , Histone Demethylases , Humains , Méthylation , Oxidoreductases acting on CH-NH2 group donors , Protéines végétales
17.
FEBS J ; 276(16): 4290-303, 2009 Aug.
Article de Anglais | MEDLINE | ID: mdl-19624734

RÉSUMÉ

Exploring enzymatic mechanisms at a molecular level is one of the major challenges in modern biophysics. Based on enzyme structure data, as obtained by X-ray crystallography or NMR spectroscopy, one can suggest how substrates and products bind for catalysis. However, from the 3D structure alone it is very rarely possible to identify how intermediates are formed and how they are interconverted. Molecular spectroscopy can provide such information and thus supplement our knowledge on the specific enzymatic reaction under consideration. In the case of enzymatic processes in which paramagnetic molecules play a role, EPR and related methods such as electron-nuclear double resonance (ENDOR) are powerful techniques to unravel important details, e.g. the electronic structure or the protonation state of the intermediate(s) carrying (the) unpaired electron spin(s). Here, we review recent EPR/ENDOR studies of blue-light active flavoproteins with emphasis on photolyases that catalyze the enzymatic repair of UV damaged DNA, and on cryptochrome blue-light photoreceptors that act in several species as central components of the circadian clock.


Sujet(s)
Spectroscopie de résonance de spin électronique/méthodes , Flavoprotéines/composition chimique , Flavoprotéines/physiologie , Rythme circadien , Réparation de l'ADN , Deoxyribodipyrimidine photo-lyase/composition chimique , Deoxyribodipyrimidine photo-lyase/physiologie , Lumière
19.
Proc Natl Acad Sci U S A ; 106(31): 13106-11, 2009 Aug 04.
Article de Anglais | MEDLINE | ID: mdl-19620714

RÉSUMÉ

Organelle movement is essential for proper function of living cells. In plants, these movements generally depend on actin filaments, but the underlying mechanism is unknown. Here, in Arabidopsis, we identify associations of short actin filaments along the chloroplast periphery on the plasma membrane side associated with chloroplast photorelocation and anchoring to the plasma membrane. We have termed these chloroplast-actin filaments (cp-actin filaments). Cp-actin filaments emerge from the chloroplast edge and exhibit rapid turnover. The presence of cp-actin filaments depends on an actin-binding protein, chloroplast unusual positioning1 (CHUP1), localized on the chloroplast envelope. chup1 mutant lacked cp-actin filaments but showed normal cytoplasmic actin filaments. When irradiated with blue light to induce chloroplast movement, cp-actin filaments relocalize to the leading edge of chloroplasts before and during photorelocation and are regulated by 2 phototropins, phot1 and phot2. Our findings suggest that plants evolved a unique actin-based mechanism for organelle movement.


Sujet(s)
Actines/physiologie , Arabidopsis/physiologie , Chloroplastes/physiologie , Protéines d'Arabidopsis/physiologie , Membrane cellulaire/composition chimique , Protéines chloroplastiques , Cryptochromes , Flavoprotéines/physiologie , Fluorescence , Protéines à fluorescence verte , Lumière , Protéines des microfilaments/physiologie , Microtubules/physiologie , Mouvement
20.
J Biol ; 8(6): 55, 2009.
Article de Anglais | MEDLINE | ID: mdl-19591650

RÉSUMÉ

The annual migration of the Monarch butterfly (Danaus plexippus) from eastern North America to central Mexico is one of nature's most inspiring spectacles. Recent studies including one in BMC Biology, have begun to dissect the molecular and neurogenetic basis for this most complex behavior.


Sujet(s)
Migration animale , Horloges biologiques/génétique , Papillons/physiologie , Flavoprotéines/physiologie , Animaux , Cryptochromes , Périodicité
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...