Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 18 de 18
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Mol Cancer Ther ; : OF1-OF13, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38904222

RÉSUMÉ

KRAS is the most frequently mutated oncogene in human cancer and facilitates uncontrolled growth through hyperactivation of the receptor tyrosine kinase (RTK)/mitogen-activated protein kinase (MAPK) pathway. The Son of Sevenless homolog 1 (SOS1) protein functions as a guanine nucleotide exchange factor (GEF) for the RAS subfamily of small GTPases and represents a druggable target in the pathway. Using a structure-based drug discovery approach, MRTX0902 was identified as a selective and potent SOS1 inhibitor that disrupts the KRAS:SOS1 protein-protein interaction to prevent SOS1-mediated nucleotide exchange on KRAS and translates into an anti-proliferative effect in cancer cell lines with genetic alterations of the KRAS-MAPK pathway. MRTX0902 augmented the antitumor activity of the KRAS G12C inhibitor adagrasib when dosed in combination in eight out of 12 KRAS G12C-mutant human non-small cell lung cancer and colorectal cancer xenograft models. Pharmacogenomic profiling in preclinical models identified cell cycle genes and the SOS2 homolog as genetic co-dependencies and implicated tumor suppressor genes (NF1 and PTEN) in resistance following combination treatment. Lastly, combined vertical inhibition of RTK/MAPK pathway signaling by MRTX0902 with inhibitors of EGFR or RAF/MEK led to greater downregulation of pathway signaling and improved antitumor responses in KRAS-MAPK pathway-mutant models. These studies demonstrate the potential clinical application of dual inhibition of SOS1 and KRAS G12C and additional SOS1 combination strategies that will aide in the understanding of SOS1 and RTK/MAPK biology in targeted cancer therapy.

2.
J Med Chem ; 67(1): 774-781, 2024 01 11.
Article de Anglais | MEDLINE | ID: mdl-38156904

RÉSUMÉ

SOS1 and SOS2 are guanine nucleotide exchange factors that mediate RTK-stimulated RAS activation. Selective SOS1:KRAS PPI inhibitors are currently under clinical investigation, whereas there are no reports to date of SOS2:KRAS PPI inhibitors. SOS2 activity is implicated in MAPK rebound when divergent SOS1 mutant cell lines are treated with the SOS1 inhibitor BI-3406; therefore, SOS2:KRAS inhibitors are of therapeutic interest. In this report, we detail a fragment-based screening strategy to identify X-ray cocrystal structures of five diverse fragment hits bound to SOS2.


Sujet(s)
Furanes , Facteurs d'échange de nucléotides guanyliques , Protéines proto-oncogènes p21(ras) , Quinazolines , Rayons X , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme , Facteurs d'échange de nucléotides guanyliques/métabolisme , Lignée cellulaire , Protéine SOS1/métabolisme
3.
Bioinformation ; 19(13): 1301-1306, 2023.
Article de Anglais | MEDLINE | ID: mdl-38415045

RÉSUMÉ

It is of interest to compare 0.2% chlorhexidine gel, 0.2% chlorhexidine chip, minocycline microspheres and slow-release doxycycline gel and tetracycline fibers as drug delivery systems in the management of peri-implantitis. The study comprised of 105 Indian participants who had a minimum of one dental implant with a probing depth of 4 mm, along with exudate and/or bleeding upon probing along with the presence of potentially harmful germs. The use of minocycline microspheres and 0.2% chlorhexidine gel resulted in significant improvements in probing depths at 1 month, 3 months and 6 months and all treatments showed decline in the indicator bacteria. Thus, minocycline microspheres and 0.2% chlorhexidine gel is useful as an adjuvant for mechanical debridement in management of peri-implantitis.

4.
J Med Chem ; 65(14): 9678-9690, 2022 07 28.
Article de Anglais | MEDLINE | ID: mdl-35833726

RÉSUMÉ

SOS1 is one of the major guanine nucleotide exchange factors that regulates the ability of KRAS to cycle through its "on" and "off" states. Disrupting the SOS1:KRASG12C protein-protein interaction (PPI) can increase the proportion of GDP-loaded KRASG12C, providing a strong mechanistic rationale for combining inhibitors of the SOS1:KRAS complex with inhibitors like MRTX849 that target GDP-loaded KRASG12C. In this report, we detail the design and discovery of MRTX0902─a potent, selective, brain-penetrant, and orally bioavailable SOS1 binder that disrupts the SOS1:KRASG12C PPI. Oral administration of MRTX0902 in combination with MRTX849 results in a significant increase in antitumor activity relative to that of either single agent, including tumor regressions in a subset of animals in the MIA PaCa-2 tumor mouse xenograft model.


Sujet(s)
Encéphale , Protéines proto-oncogènes p21(ras) , Acétonitriles , Animaux , Lignée cellulaire tumorale , Humains , Souris , Mutation , Pipérazines , Protéines proto-oncogènes p21(ras)/génétique , Pyrimidines , Protéine SOS1/métabolisme
5.
J Med Chem ; 63(19): 10773-10781, 2020 10 08.
Article de Anglais | MEDLINE | ID: mdl-32667203

RÉSUMÉ

Visceral leishmaniasis is responsible for up to 30,000 deaths every year. Current treatments have shortcomings that include toxicity and variable efficacy across endemic regions. Previously, we reported the discovery of GNF6702, a selective inhibitor of the kinetoplastid proteasome, which cleared parasites in murine models of leishmaniasis, Chagas disease, and human African trypanosomiasis. Here, we describe the discovery and characterization of LXE408, a structurally related kinetoplastid-selective proteasome inhibitor currently in Phase 1 human clinical trials. Furthermore, we present high-resolution cryo-EM structures of the Leishmania tarentolae proteasome in complex with LXE408, which provides a compelling explanation for the noncompetitive mode of binding of this novel class of inhibitors of the kinetoplastid proteasome.


Sujet(s)
Antiprotozoaires/composition chimique , Antiprotozoaires/pharmacologie , Leishmaniose viscérale/traitement médicamenteux , Oxazoles/composition chimique , Oxazoles/pharmacologie , Inhibiteurs du protéasome/composition chimique , Inhibiteurs du protéasome/pharmacologie , Pyrimidines/composition chimique , Pyrimidines/pharmacologie , Animaux , Antiprotozoaires/usage thérapeutique , Chiens , Humains , Leishmania donovani/effets des médicaments et des substances chimiques , Leishmania donovani/isolement et purification , Leishmania major/effets des médicaments et des substances chimiques , Leishmania major/isolement et purification , Leishmaniose viscérale/parasitologie , Foie/parasitologie , Macaca fascicularis , Souris , Souris de lignée BALB C , Oxazoles/usage thérapeutique , Inhibiteurs du protéasome/usage thérapeutique , Pyrimidines/usage thérapeutique , Rats , Rat Sprague-Dawley , Triazoles/composition chimique
6.
Nat Chem Biol ; 13(6): 624-632, 2017 06.
Article de Anglais | MEDLINE | ID: mdl-28346406

RÉSUMÉ

Obesity-associated insulin resistance plays a central role in type 2 diabetes. As such, tyrosine phosphatases that dephosphorylate the insulin receptor (IR) are potential therapeutic targets. The low-molecular-weight protein tyrosine phosphatase (LMPTP) is a proposed IR phosphatase, yet its role in insulin signaling in vivo has not been defined. Here we show that global and liver-specific LMPTP deletion protects mice from high-fat diet-induced diabetes without affecting body weight. To examine the role of the catalytic activity of LMPTP, we developed a small-molecule inhibitor with a novel uncompetitive mechanism, a unique binding site at the opening of the catalytic pocket, and an exquisite selectivity over other phosphatases. This inhibitor is orally bioavailable, and it increases liver IR phosphorylation in vivo and reverses high-fat diet-induced diabetes. Our findings suggest that LMPTP is a key promoter of insulin resistance and that LMPTP inhibitors would be beneficial for treating type 2 diabetes.


Sujet(s)
Diabète de type 2/traitement médicamenteux , Antienzymes/pharmacologie , Antienzymes/usage thérapeutique , Protein Tyrosine Phosphatases/antagonistes et inhibiteurs , Protein Tyrosine Phosphatases/génétique , Bibliothèques de petites molécules , Animaux , Sites de fixation , Diabète de type 2/enzymologie , Diabète de type 2/génétique , Évaluation préclinique de médicament , Activation enzymatique/effets des médicaments et des substances chimiques , Délétion de gène , Concentration inhibitrice 50 , Souris , Souris knockout , Souris obèse , Modèles biologiques , Structure moléculaire , Masse moléculaire , Bibliothèques de petites molécules/pharmacologie , Relation structure-activité
7.
Nature ; 537(7619): 229-233, 2016 09 08.
Article de Anglais | MEDLINE | ID: mdl-27501246

RÉSUMÉ

Chagas disease, leishmaniasis and sleeping sickness affect 20 million people worldwide and lead to more than 50,000 deaths annually. The diseases are caused by infection with the kinetoplastid parasites Trypanosoma cruzi, Leishmania spp. and Trypanosoma brucei spp., respectively. These parasites have similar biology and genomic sequence, suggesting that all three diseases could be cured with drugs that modulate the activity of a conserved parasite target. However, no such molecular targets or broad spectrum drugs have been identified to date. Here we describe a selective inhibitor of the kinetoplastid proteasome (GNF6702) with unprecedented in vivo efficacy, which cleared parasites from mice in all three models of infection. GNF6702 inhibits the kinetoplastid proteasome through a non-competitive mechanism, does not inhibit the mammalian proteasome or growth of mammalian cells, and is well-tolerated in mice. Our data provide genetic and chemical validation of the parasite proteasome as a promising therapeutic target for treatment of kinetoplastid infections, and underscore the possibility of developing a single class of drugs for these neglected diseases.


Sujet(s)
Maladie de Chagas/traitement médicamenteux , Kinetoplastida/effets des médicaments et des substances chimiques , Kinetoplastida/enzymologie , Leishmaniose/traitement médicamenteux , Proteasome endopeptidase complex/effets des médicaments et des substances chimiques , Inhibiteurs du protéasome/pharmacologie , Inhibiteurs du protéasome/usage thérapeutique , Pyrimidines/pharmacologie , Triazoles/pharmacologie , Maladie du sommeil/traitement médicamenteux , Animaux , Maladie de Chagas/parasitologie , Chymotrypsine/antagonistes et inhibiteurs , Chymotrypsine/métabolisme , Modèles animaux de maladie humaine , Femelle , Humains , Concentration inhibitrice 50 , Leishmaniose/parasitologie , Souris , Structure moléculaire , Thérapie moléculaire ciblée , Inhibiteurs du protéasome/effets indésirables , Inhibiteurs du protéasome/classification , Pyrimidines/effets indésirables , Pyrimidines/composition chimique , Pyrimidines/usage thérapeutique , Spécificité d'espèce , Triazoles/effets indésirables , Triazoles/composition chimique , Triazoles/usage thérapeutique , Maladie du sommeil/parasitologie
8.
J Int Oral Health ; 7(8): 89-92, 2015 Aug.
Article de Anglais | MEDLINE | ID: mdl-26464547

RÉSUMÉ

BACKGROUND: Denture repair involves joining two parts of a fractured denture with a denture repair material. Hence, a substantial repairing system for denture base fracture should be there to elude frequent fracture. MATERIALS AND METHODS: Surface treatment of conventional heat cure denture base resin with different surface treatments (chemical ethyl acetate, and mechanical roughening with bur), with control group formed without surface treatment. Specimens were repaired with auto polymerizing acrylic resin using sprinkle on technique. The testing of the transverse strength of the repaired specimens was evaluated with three-point bending test on universal testing machine. RESULTS: The study revealed that surface chemical treatment with ethyl acetate improved the transverse strength of repaired heat cure denture base when compared with mechanical and control group. A two-way analysis of variance revealed that there was statistically significant difference in mean strengths of the three groups. CONCLUSION: Surface chemical treatment with ethyl acetate improved the transverse strength of the repaired heat cure denture base when compared with mechanical roughening with bur and group without surface treatment.

9.
Antimicrob Agents Chemother ; 59(10): 6385-94, 2015 Oct.
Article de Anglais | MEDLINE | ID: mdl-26239982

RÉSUMÉ

Two CYP51 inhibitors, posaconazole and the ravuconazole prodrug E1224, were recently tested in clinical trials for efficacy in indeterminate Chagas disease. The results from these studies show that both drugs cleared parasites from the blood of infected patients at the end of the treatment but that parasitemia rebounded over the following months. In the current study, we sought to identify a dosing regimen of posaconazole that could permanently clear Trypanosoma cruzi from mice with experimental Chagas disease. Infected mice were treated with posaconazole or benznidazole, an established Chagas disease drug, and parasitological cure was defined as an absence of parasitemia recrudescence after immunosuppression. Twenty-day therapy with benznidazole (10 to 100 mg/kg of body weight/day) resulted in a dose-dependent increase in antiparasitic activity, and the 100-mg/kg regimen effected parasitological cure in all treated mice. In contrast, all mice remained infected after a 25-day treatment with posaconazole at all tested doses (10 to 100 mg/kg/day). Further extension of posaconazole therapy to 40 days resulted in only a marginal improvement of treatment outcome. We also observed similar differences in antiparasitic activity between benznidazole and posaconazole in acute T. cruzi heart infections. While benznidazole induced rapid, dose-dependent reductions in heart parasite burdens, the antiparasitic activity of posaconazole plateaued at low doses (3 to 10 mg/kg/day) despite increasing drug exposure in plasma. These observations are in good agreement with the outcomes of recent phase 2 trials with posaconazole and suggest that the efficacy models combined with the pharmacokinetic analysis employed here will be useful in predicting clinical outcomes of new drug candidates.


Sujet(s)
Inhibiteurs de la 14-alpha déméthylase/pharmacologie , Maladie de Chagas/traitement médicamenteux , Nitroimidazoles/pharmacologie , Parasitémie/traitement médicamenteux , Triazoles/pharmacologie , Trypanocides/pharmacologie , Inhibiteurs de la 14-alpha déméthylase/pharmacocinétique , Administration par voie orale , Animaux , Maladie de Chagas/enzymologie , Maladie de Chagas/immunologie , Maladie de Chagas/parasitologie , Essais cliniques de phase II comme sujet , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Calendrier d'administration des médicaments , Coeur/effets des médicaments et des substances chimiques , Coeur/parasitologie , Humains , Immunosuppression thérapeutique , Souris , Cellules NIH 3T3 , Nitroimidazoles/pharmacocinétique , Parasitémie/enzymologie , Parasitémie/immunologie , Parasitémie/parasitologie , Récidive , Sterol 14-demethylase/métabolisme , Triazoles/pharmacocinétique , Trypanocides/pharmacocinétique , Trypanosoma cruzi/effets des médicaments et des substances chimiques , Trypanosoma cruzi/pathogénicité , Trypanosoma cruzi/physiologie
10.
PLoS Pathog ; 11(7): e1005058, 2015 Jul.
Article de Anglais | MEDLINE | ID: mdl-26186534

RÉSUMÉ

Unbiased phenotypic screens enable identification of small molecules that inhibit pathogen growth by unanticipated mechanisms. These small molecules can be used as starting points for drug discovery programs that target such mechanisms. A major challenge of the approach is the identification of the cellular targets. Here we report GNF7686, a small molecule inhibitor of Trypanosoma cruzi, the causative agent of Chagas disease, and identification of cytochrome b as its target. Following discovery of GNF7686 in a parasite growth inhibition high throughput screen, we were able to evolve a GNF7686-resistant culture of T. cruzi epimastigotes. Clones from this culture bore a mutation coding for a substitution of leucine by phenylalanine at amino acid position 197 in cytochrome b. Cytochrome b is a component of complex III (cytochrome bc1) in the mitochondrial electron transport chain and catalyzes the transfer of electrons from ubiquinol to cytochrome c by a mechanism that utilizes two distinct catalytic sites, QN and QP. The L197F mutation is located in the QN site and confers resistance to GNF7686 in both parasite cell growth and biochemical cytochrome b assays. Additionally, the mutant cytochrome b confers resistance to antimycin A, another QN site inhibitor, but not to strobilurin or myxothiazol, which target the QP site. GNF7686 represents a promising starting point for Chagas disease drug discovery as it potently inhibits growth of intracellular T. cruzi amastigotes with a half maximal effective concentration (EC50) of 0.15 µM, and is highly specific for T. cruzi cytochrome b. No effect on the mammalian respiratory chain or mammalian cell proliferation was observed with up to 25 µM of GNF7686. Our approach, which combines T. cruzi chemical genetics with biochemical target validation, can be broadly applied to the discovery of additional novel drug targets and drug leads for Chagas disease.


Sujet(s)
Antifongiques/pharmacologie , Maladie de Chagas/traitement médicamenteux , Maladie de Chagas/microbiologie , Cytochromes b/métabolisme , Trypanosoma cruzi/effets des médicaments et des substances chimiques , Animaux , Antimycine A/métabolisme , Maladie de Chagas/génétique , Cytochromes b/génétique , Transport d'électrons/effets des médicaments et des substances chimiques , Transport d'électrons/immunologie , Génomique , Souris , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mutation , Consommation d'oxygène/effets des médicaments et des substances chimiques , Trypanosoma cruzi/isolement et purification , Trypanosoma cruzi/métabolisme
11.
J Biomol Screen ; 20(1): 101-11, 2015 Jan.
Article de Anglais | MEDLINE | ID: mdl-25281737

RÉSUMÉ

Chagas disease affects 8 million people worldwide and remains a main cause of death due to heart failure in Latin America. The number of cases in the United States is now estimated to be 300,000, but there are currently no Food and Drug Administration (FDA)-approved drugs available for patients with Chagas disease. To fill this gap, we have established a public-private partnership between the University of California, San Francisco and the Genomics Institute of the Novartis Research Foundation (GNF) with the goal of delivering clinical candidates to treat Chagas disease. The discovery phase, based on the screening of more than 160,000 compounds from the GNF Academic Collaboration Library, led to the identification of new anti-Chagas scaffolds. Part of the screening campaign used and compared two screening methods, including a colorimetric-based assay using Trypanosoma cruzi expressing ß-galactosidase and an image-based, high-content screening (HCS) assay using the CA-I/72 strain of T. cruzi. Comparing molecules tested in both assays, we found that ergosterol biosynthesis inhibitors had greater potency in the colorimetric assay than in the HCS assay. Both assays were used to inform structure-activity relationships for antiparasitic efficacy and pharmacokinetics. A new anti-T. cruzi scaffold derived from xanthine was identified, and we describe its development as lead series.


Sujet(s)
Découverte de médicament/méthodes , Tests de criblage à haut débit , Trypanocides/pharmacologie , Trypanosoma cruzi/effets des médicaments et des substances chimiques , Animaux , Lignée cellulaire , Maladie de Chagas/traitement médicamenteux , Colorimétrie/méthodes , Modèles animaux de maladie humaine , Évaluation préclinique de médicament , Humains , Souris , Maladies négligées/traitement médicamenteux , Bibliothèques de petites molécules , Trypanocides/composition chimique , Xanthine/composition chimique , Xanthine/pharmacologie
13.
Autoimmunity ; 46(1): 21-31, 2013 Feb.
Article de Anglais | MEDLINE | ID: mdl-23039363

RÉSUMÉ

Modifications of both DNA and protein by methylation are key factors in normal T and B cell immune responses as well as in the development of autoimmune disease. For example, the failure to maintain the methylation status of CpG dinucleotides in DNA triggers T cell autoreactivity. Methylated proteins are known targets of autoimmunity, including the symmetrical dimethylarginine residues of SmD1 and SmD3 in SLE. Herein, we demonstrate that altering the metabolism of S-adenosylmethionine (SAM), the major methyl donor for transmethylation reactions, can suppress T cell immunity. A by-product of SAM metabolism, 5'-deoxy-5'-methylthioadenosine (MTA), and an indirect inhibitor of methyltransferases, inhibits T cell responses including T cell activation markers, Th1/Th2 cytokines and TCR-related signaling events. Moreover, treatment of the lupus-prone MRL/lpr mouse with MTA markedly ameliorates splenomegaly, lymphadenopathy, autoantibody titers as well as IgG deposition and cellular infiltration in the kidney. Incubation of cells with SAM, which increases intracellular MTA levels, inhibits both TCR-mediated T cell proliferation and BCR (anti-IgM)-triggered B cell proliferation in a dose-dependent manner. These studies define the central role of MTA and SAM in immune responses and provide a simple approach to altering lymphocyte transmethylation and T cell mediated autoimmune syndromes.


Sujet(s)
Auto-immunité/immunologie , Lymphocytes B/immunologie , Lupus érythémateux disséminé/immunologie , Activation des lymphocytes/immunologie , Adémétionine/immunologie , Lymphocytes T/immunologie , Animaux , Prolifération cellulaire , Cytokines/sang , Cytokines/immunologie , Femelle , Humains , Méthylation , Souris , Souris de lignée MRL lpr , Souris transgéniques , Adémétionine/métabolisme , Lymphocytes auxiliaires Th1/immunologie , Lymphocytes auxiliaires Th2/immunologie
14.
PLoS One ; 7(1): e29984, 2012.
Article de Anglais | MEDLINE | ID: mdl-22279556

RÉSUMÉ

The nematode Caenorhabditis elegans arrests development at the first larval stage if food is not present upon hatching. Larvae in this stage provide an excellent model for studying stress responses during development. We found that supplementing starved larvae with ethanol markedly extends their lifespan within this L1 diapause. The effects of ethanol-induced lifespan extension can be observed when the ethanol is added to the medium at any time between 0 and 10 days after hatching. The lowest ethanol concentration that extended lifespan was 1 mM (0.005%); higher concentrations to 68 mM (0.4%) did not result in increased survival. In spite of their extended survival, larvae did not progress to the L2 stage. Supplementing starved cultures with n-propanol and n-butanol also extended lifespan, but methanol and isopropanol had no measurable effect. Mass spectrometry analysis of nematode fatty acids and amino acids revealed that L1 larvae can incorporate atoms from ethanol into both types of molecules. Based on these data, we suggest that ethanol supplementation may extend the lifespan of L1 larvae by either serving as a carbon and energy source and/or by inducing a stress response.


Sujet(s)
Caenorhabditis elegans/physiologie , Éthanol/pharmacologie , Privation alimentaire/physiologie , Longévité/effets des médicaments et des substances chimiques , Stress physiologique/physiologie , Acides aminés/métabolisme , Animaux , Caenorhabditis elegans/génétique , Caenorhabditis elegans/métabolisme , Dépresseurs du système nerveux central/pharmacologie , Relation dose-effet des médicaments , Acides gras/métabolisme , Glutamine/analogues et dérivés , Larve/génétique , Larve/métabolisme , Larve/physiologie , Longévité/génétique , Longévité/physiologie , Mutation , Facteurs temps
15.
PLoS One ; 6(6): e20850, 2011.
Article de Anglais | MEDLINE | ID: mdl-21695191

RÉSUMÉ

The protein L-isoaspartyl-O-methyltransferase functions to initiate the repair of isomerized aspartyl and asparaginyl residues that spontaneously accumulate with age in a variety of organisms. Caenorhabditis elegans nematodes lacking the pcm-1 gene encoding this enzyme display a normal lifespan and phenotype under standard laboratory growth conditions. However, significant defects in development, egg laying, dauer survival, and autophagy have been observed in pcm-1 mutant nematodes when deprived of food and when exposed to oxidative stress. Interestingly, overexpression of this repair enzyme in both Drosophila and C. elegans extends adult lifespan under thermal stress. In this work, we show the involvement of the insulin/insulin-like growth factor-1 signaling (IIS) pathway in PCM-1-dependent lifespan extension in C. elegans. We demonstrate that reducing the levels of the DAF-16 downstream transcriptional effector of the IIS pathway by RNA interference reduces the lifespan extension resulting from PCM-1 overexpression. Using quantitative real-time PCR analysis, we show the up-regulation of DAF-16-dependent stress response genes in the PCM-1 overexpressor animals compared to wild-type and pcm-1 mutant nematodes under mild thermal stress conditions. Additionally, similar to other long-lived C. elegans mutants in the IIS pathway, including daf-2 and age-1 mutants, PCM-1 overexpressor adult animals display increased resistance to severe thermal stress, whereas pcm-1 mutant animals survive less long under these conditions. Although we observe a higher accumulation of damaged proteins in pcm-1 mutant nematodes, the basal level of isoaspartyl residues detected in wild-type animals was not reduced by PCM-1 overexpression. Our results support a signaling role for the protein L-isoaspartyl methyltransferase in lifespan extension that involves the IIS pathway, but that may be independent of its function in overall protein repair.


Sujet(s)
Caenorhabditis elegans/cytologie , Caenorhabditis elegans/physiologie , Facteur de croissance IGF-I/métabolisme , Insuline/métabolisme , Longévité , Protein D-aspartate-L-isoaspartate methyltransferase/métabolisme , Transduction du signal , Animaux , Caenorhabditis elegans/enzymologie , Caenorhabditis elegans/génétique , Protéines de Caenorhabditis elegans/génétique , Protéines de Caenorhabditis elegans/métabolisme , Facteurs de transcription Forkhead , Régulation de l'expression des gènes codant pour des enzymes , Réaction de choc thermique/génétique , Longévité/génétique , Longévité/physiologie , Protein D-aspartate-L-isoaspartate methyltransferase/génétique , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Transcription génétique
16.
Mech Ageing Dev ; 130(10): 670-80, 2009 Oct.
Article de Anglais | MEDLINE | ID: mdl-19682488

RÉSUMÉ

We have shown that Caenorhabditis elegans lacking the PCM-1 protein repair l-isoaspartyl methyltransferase are more sensitive to oxidative stress than wild-type nematodes. Exposure to the redox-cycling quinone juglone upon exit from dauer diapause results in defective egg-laying (Egl phenotype) in the pcm-1 mutants only. Treatment with paraquat, a redox-cycling dipyridyl, causes a more severe developmental delay at the second larval stage in pcm-1 mutants than in wild-type nematodes. Finally, exposure to homocysteine and homocysteine thiolactone, molecules that can induce oxidative stress via distinct mechanisms, results in a more pronounced delay in development at the first larval stage in pcm-1 mutants than in wild-type animals. Homocysteine treatment also induced the Egl phenotype in mutant but not wild-type nematodes. All of the effects of these agents were reversed upon addition of vitamin C, indicating that the developmental delay and egg-laying defects result from oxidative stress. Furthermore, we have demonstrated that a mutation in the gene encoding the insulin-like receptor DAF-2 suppresses the Egl phenotype in pcm-1 mutants treated with juglone. Our results support a role of PCM-1 in the cellular responses mediated by the DAF-2 insulin-like signaling pathway in C. elegans for optimal protection against oxidative stress.


Sujet(s)
Caenorhabditis elegans/enzymologie , Methyltransferases/déficit , Stress oxydatif , Animaux , Antioxydants/pharmacologie , Acide ascorbique/pharmacologie , Caenorhabditis elegans/effets des médicaments et des substances chimiques , Caenorhabditis elegans/embryologie , Caenorhabditis elegans/génétique , Protéines de Caenorhabditis elegans/génétique , Protéines de Caenorhabditis elegans/métabolisme , Protéines du cycle cellulaire/génétique , Génotype , Homocystéine/analogues et dérivés , Homocystéine/métabolisme , Homocystéine/toxicité , Larve/métabolisme , Métamorphose biologique , Methyltransferases/génétique , Mutation , Naphtoquinones/toxicité , Oxydants/toxicité , Stress oxydatif/effets des médicaments et des substances chimiques , Stress oxydatif/génétique , Paraquat/toxicité , Phénotype , Récepteur à l'insuline/génétique , Récepteur à l'insuline/métabolisme
17.
J Clin Invest ; 118(12): 3917-29, 2008 Dec.
Article de Anglais | MEDLINE | ID: mdl-19033662

RÉSUMÉ

The role of autophagy in oncogenesis remains ambiguous, and mechanisms that induce autophagy and regulate its outcome in human cancers are poorly understood. The maternally imprinted Ras-related tumor suppressor gene aplasia Ras homolog member I (ARHI; also known as DIRAS3) is downregulated in more than 60% of ovarian cancers, and here we show that re-expression of ARHI in multiple human ovarian cancer cell lines induces autophagy by blocking PI3K signaling and inhibiting mammalian target of rapamycin (mTOR), upregulating ATG4, and colocalizing with cleaved microtubule-associated protein light chain 3 (LC3) in autophagosomes. Furthermore, ARHI is required for spontaneous and rapamycin-induced autophagy in normal and malignant cells. Although ARHI re-expression led to autophagic cell death when SKOv3 ovarian cancer cells were grown in culture, it enabled the cells to remain dormant when they were grown in mice as xenografts. When ARHI levels were reduced in dormant cells, xenografts grew rapidly. However, inhibition of ARHI-induced autophagy with chloroquine dramatically reduced regrowth of xenografted tumors upon reduction of ARHI levels, suggesting that autophagy contributed to the survival of dormant cells. Further analysis revealed that autophagic cell death was reduced when cultured human ovarian cancer cells in which ARHI had been re-expressed were treated with growth factors (IGF-1, M-CSF), angiogenic factors (VEGF, IL-8), and matrix proteins found in xenografts. Thus, ARHI can induce autophagic cell death, but can also promote tumor dormancy in the presence of factors that promote survival in the cancer microenvironment.


Sujet(s)
Autophagie , Tumeurs de l'ovaire/métabolisme , Protéines suppresseurs de tumeurs/métabolisme , Protéines G rho/métabolisme , Animaux , Antibiotiques antinéoplasiques/pharmacologie , Antirhumatismaux/pharmacologie , Autophagie/effets des médicaments et des substances chimiques , Autophagie/génétique , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/génétique , Chloroquine/pharmacologie , Cysteine endopeptidases/génétique , Cysteine endopeptidases/métabolisme , Femelle , Empreinte génomique/effets des médicaments et des substances chimiques , Empreinte génomique/génétique , Humains , Protéines et peptides de signalisation intercellulaire/pharmacologie , Souris , Souris de lignée BALB C , Souris nude , Protéines associées aux microtubules/génétique , Protéines associées aux microtubules/métabolisme , Transplantation tumorale , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Phagosomes/génétique , Phagosomes/métabolisme , Phosphatidylinositol 3-kinases/génétique , Phosphatidylinositol 3-kinases/métabolisme , Protein kinases/génétique , Protein kinases/métabolisme , Sirolimus/pharmacologie , Sérine-thréonine kinases TOR , Transplantation hétérologue , Protéines suppresseurs de tumeurs/génétique , Protéines G rho/génétique
18.
J Gen Appl Microbiol ; 47(6): 307-312, 2001 Dec.
Article de Anglais | MEDLINE | ID: mdl-12483605

RÉSUMÉ

Several facultative anaerobes tolerant to high levels of chromate (>400 mg/ml) were isolated from tannery effluents. These isolates displayed varying degrees of Cr(VI) reduction under aerobic and anaerobic conditions at room temperature (24+/-2 degrees C). Interestingly, eight isolates were efficient in reducing 70% Cr(VI) anaerobically. This includes 5 isolates of genus Aerococcus, two isolates of Micrococcus and single isolate of genus Aeromonas. These isolates were subjected to further characterization for possible use in Cr(VI) detoxification of industrial wastes. This is the first report of Aerococcus sp. capable of Cr(VI) reduction >70% anaerobically. These bacteria were further checked for tolerance to a variety of other heavy metals. Our study indicates the possible use of these bacteria in environmental clean up.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE