Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 21
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Chembiochem ; 25(4): e202300712, 2024 02 16.
Article de Anglais | MEDLINE | ID: mdl-38015747

RÉSUMÉ

Chemically induced proximity (CIP) refers to co-opting naturally occurring biological pathways using synthetic molecules to recruit neosubstrates that are not normally encountered or to enhance the affinity of naturally occurring interactions. Leveraging proximity biology through CIPs has become a rapidly evolving field and has garnered considerable interest in basic research and drug discovery. PROteolysis TArgeting Chimera (PROTAC) is a well-established CIP modality that induces the proximity between a target protein and an E3 ubiquitin ligase, causing target protein degradation via the ubiquitin-proteasome system. Inspired by PROTACs, several other induced proximity modalities have emerged to modulate both proteins and RNA over recent years. In this review, we summarize the critical advances and opportunities in the field, focusing on protein degraders, RNA degraders and non-degrader modalities such as post-translational modification (PTM) and protein-protein interaction (PPI) modulators. We envision that these emerging proximity-based drug modalities will be valuable resources for both biological research and therapeutic discovery in the future.


Sujet(s)
Tics , Humains , Protéines/métabolisme , Ubiquitin-protein ligases/métabolisme , Protéolyse , Découverte de médicament , ARN/métabolisme , Biologie , Ligands
2.
J Am Chem Soc ; 143(28): 10571-10575, 2021 07 21.
Article de Anglais | MEDLINE | ID: mdl-34236858

RÉSUMÉ

We hypothesized that the proximity-driven ubiquitylation of E3-interacting small molecules could affect the degradation of E3 ubiquitin ligases. A series of XIAP BIR2 domain-binding small molecules was modified to append a nucleophilic primary amine. This modification transforms XIAP binders into inducers of XIAP degradation. The degradation of XIAP is E1- and proteasome-dependent, dependent on the ligase function of XIAP, and is rescued by subtle modifications of the small molecule that would obviate ubiquitylation. We demonstrate in vitro ubiquitylation of the small molecule that is dependent on its interaction with XIAP. Taken together, these results demonstrate the designed ubiquitylation of an engineered small molecule and a novel approach for the degradation of E3 ubiquitin ligases.


Sujet(s)
Amines/pharmacologie , Bibliothèques de petites molécules/pharmacologie , Protéine inhibitrice de l'apoptose liée au chromosome X/antagonistes et inhibiteurs , Amines/composition chimique , Cristallographie aux rayons X , Humains , Modèles moléculaires , Structure moléculaire , Bibliothèques de petites molécules/composition chimique , Protéine inhibitrice de l'apoptose liée au chromosome X/métabolisme
3.
Nat Chem Biol ; 16(11): 1157-1158, 2020 11.
Article de Anglais | MEDLINE | ID: mdl-32747810

Sujet(s)
Cyclines
4.
Mol Cell ; 77(5): 1092-1106.e9, 2020 03 05.
Article de Anglais | MEDLINE | ID: mdl-31973889

RÉSUMÉ

Co-opting Cullin4 RING ubiquitin ligases (CRL4s) to inducibly degrade pathogenic proteins is emerging as a promising therapeutic strategy. Despite intense efforts to rationally design degrader molecules that co-opt CRL4s, much about the organization and regulation of these ligases remains elusive. Here, we establish protein interaction kinetics and estimation of stoichiometries (PIKES) analysis, a systematic proteomic profiling platform that integrates cellular engineering, affinity purification, chemical stabilization, and quantitative mass spectrometry to investigate the dynamics of interchangeable multiprotein complexes. Using PIKES, we show that ligase assemblies of Cullin4 with individual substrate receptors differ in abundance by up to 200-fold and that Cand1/2 act as substrate receptor exchange factors. Furthermore, degrader molecules can induce the assembly of their cognate CRL4, and higher expression of the associated substrate receptor enhances degrader potency. Beyond the CRL4 network, we show how PIKES can reveal systems level biochemistry for cellular protein networks important to drug development.


Sujet(s)
Chromatographie en phase liquide à haute performance , Protéomique/méthodes , Spectrométrie de masse ESI , Spectrométrie de masse en tandem , Ubiquitin-protein ligases/métabolisme , Cullines/génétique , Cullines/métabolisme , Cellules HEK293 , Humains , Cinétique , Protéines du muscle/génétique , Protéines du muscle/métabolisme , Protéine NEDD8/génétique , Protéine NEDD8/métabolisme , Cartes d'interactions protéiques , Protéolyse , Transduction du signal , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Ubiquitin-protein ligases/génétique
5.
Bioorg Med Chem Lett ; 30(4): 126907, 2020 02 15.
Article de Anglais | MEDLINE | ID: mdl-31902710

RÉSUMÉ

Chimeric molecules which effect intracellular degradation of target proteins via E3 ligase-mediated ubiquitination (e.g., PROTACs) are currently of high interest in medicinal chemistry. However, these entities are relatively large compounds that often possess molecular characteristics which may compromise oral bioavailability, solubility, and/or in vivo pharmacokinetic properties. Accordingly, we explored whether conjugation of chimeric degraders to monoclonal antibodies using technologies originally developed for cytotoxic payloads might provide alternate delivery options for these novel agents. In this report we describe the construction of several degrader-antibody conjugates comprised of two distinct ERα-targeting degrader entities and three independent ADC linker modalities. We subsequently demonstrate the antigen-dependent delivery to MCF7-neo/HER2 cells of the degrader payloads that are incorporated into these conjugates. We also provide evidence for efficient intracellular degrader release from one of the employed linkers. In addition, preliminary data are described which suggest that reasonably favorable in vivo stability properties are associated with the linkers utilized to construct the degrader conjugates.


Sujet(s)
Anticorps monoclonaux/immunologie , Vecteurs de médicaments/composition chimique , Récepteur alpha des oestrogènes/immunologie , Anticorps monoclonaux/composition chimique , Antinéoplasiques/composition chimique , Antinéoplasiques/immunologie , Antinéoplasiques/pharmacologie , Conception de médicament , Récepteur alpha des oestrogènes/métabolisme , Humains , Immunoconjugués/composition chimique , Immunoconjugués/immunologie , Immunoconjugués/pharmacologie , Cellules MCF-7 , Protéolyse/effets des médicaments et des substances chimiques , Récepteur ErbB-2/métabolisme
6.
Chemistry ; 24(19): 4830-4834, 2018 Apr 03.
Article de Anglais | MEDLINE | ID: mdl-29493023

RÉSUMÉ

A novel strategy to attach indole-containing payloads to antibodies through a carbamate moiety and a self-immolating, disulfide-based linker is described. This new strategy was employed to connect a selective estrogen receptor down-regulator (SERD) to various antibodies in a site-selective manner. The resulting conjugates displayed potent, antigen-dependent down-regulation of estrogen receptor levels in MCF7-neo/HER2 and MCF7-hB7H4 cells. They also exhibited similar antigen-dependent modulation of the estrogen receptor in tumors when administered intravenously to mice bearing MCF7-neo/HER2 tumor xenografts. The indole-carbamate moiety present in the new linker was stable in whole blood from various species and also exhibited good in vivo stability properties in mice.


Sujet(s)
Indoles/composition chimique , Animaux , Anticorps monoclonaux/composition chimique , Lignée cellulaire tumorale , Humains , Immunoconjugués/administration et posologie , Cellules MCF-7 , Souris
7.
Sci Rep ; 6: 18846, 2016 Jan 06.
Article de Anglais | MEDLINE | ID: mdl-26733354

RÉSUMÉ

Ribonucleotide reductase small subunit B (RRM2B) is a stress response protein that protects normal human fibroblasts from oxidative stress. However, the underlying mechanism that governs this function is not entirely understood. To identify factors that interact with RRM2B and mediate anti-oxidation function, large-scale purification of human Flag-tagged RRM2B complexes was performed. Pyrroline-5-carboxylate reductase 1 and 2 (PYCR1, PYCR2) were identified by mass spectrometry analysis as components of RRM2B complexes. Silencing of both PYCR1 and PYCR2 by expressing short hairpin RNAs induced defects in cell proliferation, partial fragmentation of the mitochondrial network, and hypersensitivity to oxidative stress in hTERT-immortalized human foreskin fibroblasts (HFF-hTERT). Moderate overexpression of RRM2B, comparable to stress-induced level, protected cells from oxidative stress. Silencing of both PYCR1 and PYCR2 completely abolished anti-oxidation activity of RRM2B, demonstrating a functional collaboration of these metabolic enzymes in response to oxidative stress.


Sujet(s)
Protéines du cycle cellulaire/métabolisme , Stress oxydatif , Pyrroline carboxylate reductases/métabolisme , Ribonucleotide reductases/métabolisme , Animaux , Antioxydants/métabolisme , Points de contrôle du cycle cellulaire , Protéines du cycle cellulaire/génétique , Lignée cellulaire , Techniques de knock-down de gènes , Extinction de l'expression des gènes , Humains , Isoenzymes , Spectrométrie de masse/méthodes , Mitochondries/génétique , Mitochondries/métabolisme , Complexes multiprotéiques/métabolisme , Liaison aux protéines , Cartographie d'interactions entre protéines , Transport des protéines , Pyrroline carboxylate reductases/génétique , Protéines de fusion recombinantes , Ribonucleotide reductases/génétique , Transduction du signal , Telomerase/génétique , Telomerase/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Danio zébré ,
8.
Nat Chem Biol ; 11(8): 611-7, 2015 Aug.
Article de Anglais | MEDLINE | ID: mdl-26075522

RÉSUMÉ

The current predominant therapeutic paradigm is based on maximizing drug-receptor occupancy to achieve clinical benefit. This strategy, however, generally requires excessive drug concentrations to ensure sufficient occupancy, often leading to adverse side effects. Here, we describe major improvements to the proteolysis targeting chimeras (PROTACs) method, a chemical knockdown strategy in which a heterobifunctional molecule recruits a specific protein target to an E3 ubiquitin ligase, resulting in the target's ubiquitination and degradation. These compounds behave catalytically in their ability to induce the ubiquitination of super-stoichiometric quantities of proteins, providing efficacy that is not limited by equilibrium occupancy. We present two PROTACs that are capable of specifically reducing protein levels by >90% at nanomolar concentrations. In addition, mouse studies indicate that they provide broad tissue distribution and knockdown of the targeted protein in tumor xenografts. Together, these data demonstrate a protein knockdown system combining many of the favorable properties of small-molecule agents with the potent protein knockdown of RNAi and CRISPR.


Sujet(s)
Antinéoplasiques/pharmacologie , Tumeurs du sein/traitement médicamenteux , Protéines tumorales/antagonistes et inhibiteurs , Receptor-Interacting Protein Serine-Threonine Kinase 2/antagonistes et inhibiteurs , Récepteurs des oestrogènes/antagonistes et inhibiteurs , Bibliothèques de petites molécules/pharmacologie , Animaux , Sites de fixation , Biocatalyse , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Femelle , Humains , Cellules MCF-7 , Souris , Modèles moléculaires , Thérapie moléculaire ciblée , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Transplantation tumorale , Proteasome endopeptidase complex/métabolisme , Liaison aux protéines , Protéolyse , Receptor-Interacting Protein Serine-Threonine Kinase 2/génétique , Receptor-Interacting Protein Serine-Threonine Kinase 2/métabolisme , Récepteurs des oestrogènes/génétique , Récepteurs des oestrogènes/métabolisme , Ubiquitine/génétique , Ubiquitine/métabolisme , Ubiquitination , Protéine Von Hippel-Lindau supresseur de tumeur/génétique , Protéine Von Hippel-Lindau supresseur de tumeur/métabolisme ,
9.
J Biol Chem ; 289(28): 19789-98, 2014 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-24855649

RÉSUMÉ

Because the deubiquitinating enzyme USP33 is involved in several important cellular processes (ß-adrenergic receptor recycling, centrosome amplification, RalB signaling, and cancer cell migration), its levels must be carefully regulated. Using quantitative mass spectrometry, we found that the intracellular level of USP33 is highly sensitive to the activity of p97. Knockdown or chemical inhibition of p97 causes robust accumulation of USP33 due to inhibition of its degradation. The p97 adaptor complex involved in this function is the Ufd1-Npl4 heterodimer. Furthermore, we identified HERC2, a HECT domain-containing E3 ligase, as being responsible for polyubiquitination of USP33. Inhibition of p97 causes accumulation of polyubiquitinated USP33, suggesting that p97 is required for postubiquitination processing. Thus, our study has identified several key molecules that control USP33 degradation within the ubiquitin-proteasome system.


Sujet(s)
Adenosine triphosphatases/métabolisme , Protéines du cycle cellulaire/métabolisme , Facteurs d'échange de nucléotides guanyliques/métabolisme , Protéolyse , Ubiquitin thiolesterase/métabolisme , Ubiquitination/physiologie , Adenosine triphosphatases/génétique , Animaux , Protéines du cycle cellulaire/génétique , Facteurs d'échange de nucléotides guanyliques/génétique , Cellules HeLa , Humains , Souris , Cellules NIH 3T3 , Proteasome endopeptidase complex/génétique , Proteasome endopeptidase complex/métabolisme , Ubiquitine/génétique , Ubiquitine/métabolisme , Ubiquitin thiolesterase/génétique , Ubiquitin-protein ligases , Protéine contenant la valosine
10.
Elife ; 3: e01856, 2014.
Article de Anglais | MEDLINE | ID: mdl-24448410

RÉSUMÉ

Proteasome inhibition elicits an evolutionarily conserved response wherein proteasome subunit mRNAs are upregulated, resulting in recovery (i.e., 'bounce-back') of proteasome activity. We previously demonstrated that the transcription factor Nrf1/NFE2L1 mediates this homeostatic response in mammalian cells. We show here that Nrf1 is initially translocated into the lumen of the ER, but is rapidly and efficiently retrotranslocated to the cytosolic side of the membrane in a manner that depends on p97/VCP. Normally, retrotranslocated Nrf1 is degraded promptly by the proteasome and active species do not accumulate. However, in cells with compromised proteasomes, retrotranslocated Nrf1 escapes degradation and is cleaved N-terminal to Leu-104 to yield a fragment that is no longer tethered to the ER membrane. Importantly, this cleavage event is essential for Nrf1-dependent activation of proteasome gene expression upon proteasome inhibition. Our data uncover an unexpected role for p97 in activation of a transcription factor by relocalizing it from the ER lumen to the cytosol. DOI: http://dx.doi.org/10.7554/eLife.01856.001.


Sujet(s)
Adenosine triphosphatases/métabolisme , Protéines du cycle cellulaire/métabolisme , Réticulum endoplasmique/métabolisme , Facteur nucléaire-1 respiratoire/métabolisme , Proteasome endopeptidase complex/effets des médicaments et des substances chimiques , Inhibiteurs du protéasome/pharmacologie , Adenosine triphosphatases/génétique , Animaux , Protéines du cycle cellulaire/génétique , Cellules HEK293 , Humains , Souris , Mutation , Cellules NIH 3T3 , Facteur nucléaire-1 respiratoire/génétique , Proteasome endopeptidase complex/génétique , Proteasome endopeptidase complex/métabolisme , Maturation post-traductionnelle des protéines , Transport des protéines , Protéolyse , Interférence par ARN , Transcription génétique , Activation de la transcription , Transfection , Protéine contenant la valosine
11.
Nat Struct Mol Biol ; 19(5): 511-6, S1, 2012 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-22466964

RÉSUMÉ

The AAA+ ATPase p97 and its UBA-UBX cofactors are thought to extract ubiquitinated proteins from membranes or protein complexes as a prelude to their degradation. However, for many cofactors ubiquitinated targets have not yet been identified, leaving their biological function unclear. Previous analysis has linked the p97 pathway to cullin-RING ubiquitin ligases (CRLs); here we demonstrate that the human p97 cofactor UBXD7 mediates the p97-CRL interaction through its conserved ubiquitin-interacting motif (UIM). UBXD7 and its yeast ortholog, Ubx5, associate only with the active, NEDD8- or Rub1-modified form of cullins. Disruption of the Ubx5 UIM results in a loss of CRL binding and consequently impedes degradation of a Cul3 substrate. These results uncover an unexpected and conserved role for NEDD8 in linking CRL ubiquitin ligase function to the p97 pathway.


Sujet(s)
Adenosine triphosphatases/métabolisme , Protéines de transport/métabolisme , Protéines nucléaires/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitines/métabolisme , Protéines adaptatrices de la transduction du signal , Protéines de transport/composition chimique , Lignée cellulaire , Cullines/métabolisme , Protéines de liaison à l'ADN/métabolisme , Humains , Modèles moléculaires , Protéine NEDD8 , Complexe répresseur Polycomb-1 , Motifs et domaines d'intéraction protéique , Cartes d'interactions protéiques , Ubiquitine/métabolisme
12.
Mol Cell Proteomics ; 11(6): M111.016444, 2012 Jun.
Article de Anglais | MEDLINE | ID: mdl-22337587

RÉSUMÉ

UBXD1 is a member of the poorly understood subfamily of p97 adaptors that do not harbor a ubiquitin association domain or bind ubiquitin-modified proteins. Of clinical importance, p97 mutants found in familial neurodegenerative conditions Inclusion Body Myopathy Paget's disease of the bone and/or Frontotemporal Dementia and Amyotrophic Lateral Sclerosis are defective at interacting with UBXD1, indicating that functions regulated by a p97-UBXD1 complex are altered in these diseases. We have performed liquid chromatography-mass spectrometric analysis of UBXD1-interacting proteins to identify pathways in which UBXD1 functions. UBXD1 displays prominent association with ERGIC-53, a hexameric type I integral membrane protein that functions in protein trafficking. The UBXD1-ERGIC-53 interaction requires the N-terminal 10 residues of UBXD1 and the C-terminal cytoplasmic 12 amino acid tail of ERGIC-53. Use of p97 and E1 enzyme inhibitors indicate that complex formation between UBXD1 and ERGIC-53 requires the ATPase activity of p97, but not ubiquitin modification. We also performed SILAC-based quantitative proteomic profiling to identify ERGIC-53 interacting proteins. This analysis identified known (e.g. COPI subunits) and novel (Rab3GAP1/2 complex involved in the fusion of vesicles at the cell membrane) interactions that are also mediated through the C terminus of the protein. Immunoprecipitation and Western blotting analysis confirmed the proteomic interaction data and it also revealed that an UBXD1-Rab3GAP association requires the ERGIC-53 binding domain of UBXD1. Localization studies indicate that UBXD1 modules the sub-cellular trafficking of ERGIC-53, including promoting movement to the cell membrane. We propose that p97-UBXD1 modulates the trafficking of ERGIC-53-containing vesicles by controlling the interaction of transport factors with the cytoplasmic tail of ERGIC-53.


Sujet(s)
Protéines de transport/métabolisme , Lectines liant le mannose/métabolisme , Protéines membranaires/métabolisme , Protéines adaptatrices de la transduction du signal , Protéines adaptatrices du transport vésiculaire , Adenosine triphosphatases/antagonistes et inhibiteurs , Adenosine triphosphatases/métabolisme , Protéines associées à l'autophagie , Benzoates/pharmacologie , Protéines de transport/composition chimique , Lignée cellulaire tumorale , Furanes/pharmacologie , Humains , Lectines liant le mannose/composition chimique , Protéines membranaires/composition chimique , Protéines nucléaires/antagonistes et inhibiteurs , Protéines nucléaires/métabolisme , Liaison aux protéines , Motifs et domaines d'intéraction protéique , Cartographie d'interactions entre protéines , Transport des protéines , Pyrazoles/pharmacologie , Quinazolines/pharmacologie , Vésicules de sécrétion/métabolisme , Ubiquitin-activating enzymes/antagonistes et inhibiteurs , Ubiquitin-activating enzymes/métabolisme , Protéines G rab3/métabolisme
13.
Cell Cycle ; 7(23): 3737-46, 2008 Dec.
Article de Anglais | MEDLINE | ID: mdl-19029828

RÉSUMÉ

Cyclin(-D-)-dependent kinase (Cdk) inhibitors of the Ink4 family specifically bind to Cdk4 and Cdk6, but not to other Cdks. Ink4c and Ink4d mRNAs are maximally and periodically expressed during the G(2)/M phase of the cell division cycle, but the abundance of their encoded proteins is regulated through distinct mechanisms. Both proteins undergo polyubiquitination, but the half life of p18(Ink4c) (approximately 10 hours) is much longer than that of p19(Ink4d) (approximately 2.5 hours). Lysines 46 and 112 are preferred sites of ubiquitin conjugation in p18(Ink4c), although substitution of these and other lysine residues with arginine, particularly in combination, triggers protein misfolding and accelerates p18(Ink4c) degradation. When tethered to either catalytically active or inactive Cdk4 or Cdk6, polyubiquitination of p18(Ink4c) is inhibited, and the protein is further stabilized. Conversely, in competing with p18(Ink4c) for binding to Cdks, cyclin D1 accelerates p18(Ink4c) turnover. In direct contrast, polyubiquitination of p19(Ink4d) is induced by its association with Cdks, whereas cyclin D1 overexpression retards p19(Ink4d) degradation. Although it has been generally assumed that p18(Ink4c) and p19(Ink4d) are biochemically similar Cdk inhibitors, the major differences in their stability and turnover are likely key to understanding their distinct biological functions.


Sujet(s)
Inhibiteur p18 de kinase cycline-dépendante/génétique , Inhibiteur p19 de kinase cycline-dépendante/génétique , Régulation de l'expression des gènes , Transcription génétique , Animaux , Cycle cellulaire , Cycline D1/métabolisme , Kinase-4 cycline-dépendante/métabolisme , Humains , Cinétique , Lysine/métabolisme , Souris , Cellules NIH 3T3 , Neurones/métabolisme , Polyubiquitine/métabolisme , Liaison aux protéines , Stabilité protéique , Ubiquitination
14.
Cell Cycle ; 7(21): 3378-87, 2008 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-18948745

RÉSUMÉ

The stabilization and subcellular localization of the p19(Arf) tumor suppressor protein and the SUMO-2/3 deconjugating protease Senp3 each depend upon their binding to the abundant nucleolar protein nucleophosmin (Npm/B23). Senp3 and p19(Arf) antagonize each other's functions in regulating the SUMOylation of target proteins including Npm itself. The p19(Arf) protein triggers the sequential phosphorylation, polyubiquitination and rapid proteasomal degradation of Senp3, and this ability of p19(Arf) to accelerate Senp3 turnover also depends on the presence of Npm. In turn, endogenous p19(Arf) and Senp3 are both destabilized in viable Npm-null mouse embryo fibroblasts (that also lack p53), and reintroduction of the human NPM protein into these cells reverses this phenotype. NPM mutants that retain their acidic and oligomerization domains can re-stabilize both p19(Arf) and Senp3 in this setting, but the nucleolar localization of NPM is not strictly required for these effects. Knockdown of Senp3 with shRNAs mimics the antiproliferative functions of p19(Arf) in cells that lack p53 alone or in triple knock-out cells that lack the Arf, Mdm2 and p53 genes. These findings reinforce the hypothesis that the p53-independent tumor suppressive functions of p19(Arf) may be mediated by its ability to antagonize Senp3, thereby inducing cell cycle arrest by abnormally elevating the cellular levels of SUMOylated proteins.


Sujet(s)
Nucléole/métabolisme , Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Protéines nucléaires/métabolisme , Peptide hydrolases/métabolisme , Petites protéines modificatrices apparentées à l'ubiquitine/métabolisme , Ubiquitines/métabolisme , Animaux , Prolifération cellulaire , Cysteine endopeptidases , Régulation négative , Humains , Souris , Cellules NIH 3T3 , Protéines nucléaires/composition chimique , Nucléophosmine , Phosphorylation , Polyubiquitine/métabolisme , Proteasome endopeptidase complex/métabolisme , Liaison aux protéines , Stabilité protéique , Structure tertiaire des protéines
15.
Genes Dev ; 21(18): 2283-7, 2007 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-17761812

RÉSUMÉ

Retroviral transduction of the BCR-ABL kinase into primary mouse bone marrow cells lacking the Arf tumor suppressor rapidly generates polyclonal populations of continuously self-renewing pre-B cells, virtually all of which have leukemic potential. Intravenous infusion of 20 such cells into healthy syngeneic mice induces rapidly fatal, transplantable lymphoblastic leukemias that resist imatinib therapy. Introduction of BCR-ABL into Arf-null severe combined immunodeficient (SCID) bone marrow progenitors lacking the cytokine receptor common gamma-chain yields leukemogenic pre-B cells that exhibit greater sensitivity to imatinib in vivo. Hence, salutary cytokines in the hematopoietic microenvironment can facilitate leukemic proliferation and confer resistance to targeted therapy.


Sujet(s)
Inhibiteur p16 de kinase cycline-dépendante/génétique , Cytokines/pharmacologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Protéines de fusion bcr-abl/génétique , Pipérazines/usage thérapeutique , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Pyrimidines/usage thérapeutique , Animaux , Antinéoplasiques/usage thérapeutique , Benzamides , Inhibiteur p16 de kinase cycline-dépendante/physiologie , Résistance aux médicaments antinéoplasiques/génétique , Cellules souches hématopoïétiques/anatomopathologie , Mésilate d'imatinib , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Souris SCID , Modèles biologiques , Transplantation tumorale/anatomopathologie , Échec thérapeutique
16.
Isr Med Assoc J ; 8(4): 249-51, 2006 Apr.
Article de Anglais | MEDLINE | ID: mdl-16671360

RÉSUMÉ

The Ink4a-Arf locus, which encodes two distinct tumor suppressor proteins, is inactivated in many cancers. Whereas p16Ink4a is an inhibitor of cyclin D-dependent kinases, p19Arf (p14ARF in humans) antagonizes the E3 ubiquitin protein ligase activity of Mdm2 to activate p53. We now recognize that Arf functions in both p53-dependent and -independent modes to counteract hyper-proliferative signals originating from proto-oncogene activation, but its p53-independent activities remain poorly understood. Arf proteins are highly basic (> 20% arginine content, pl > 12) and predominantly localize within nucleoli in physical association with an abundant acidic protein, nucleophosmin (NPM/B23). When bound to NPM, Arf proteins are relatively stable with half-lives of 6-8 hours. Although mouse p19Arf contains only a single lysine residue and human p14ARF has none, both proteins are N-terminally ubiquitinated and degraded in proteasomes. Through as yet uncharacterized mechanisms, p19Arf induces p53-independent sumoylation of a variety of cellular target proteins with which it interacts, including both Mdm2 and NPM. A naturally occurring NPM mutant (NPMc) expressed in myeloid leukemia cells redirects both wild-type NPM and p19Arf to the cytoplasm, inhibits Arf-induced sumoylation, and attenuates p53 activity. Thus, ubiquitination and sumoylation can each influence Arf tumor suppressor activity.


Sujet(s)
Inhibiteur p16 de kinase cycline-dépendante/physiologie , Protéine p14(ARF) suppresseur de tumeur/physiologie , Protéine p53 suppresseur de tumeur/physiologie , Ubiquitine/physiologie , Prolifération cellulaire , Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Humains , Leucémie myéloïde , Protéines nucléaires/physiologie , Nucléophosmine , Proto-oncogène Mas , Cellules cancéreuses en culture , Protéine p14(ARF) suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme
17.
Cell Cycle ; 4(11): 1593-8, 2005 Nov.
Article de Anglais | MEDLINE | ID: mdl-16205118

RÉSUMÉ

Nucleophosmin (NPM or B23) plays key roles in ribosome biogenesis, centrosome duplication, and maintenance of genomic integrity. Mutations affecting the carboxylterminal domain of NPM occur in a significant percentage of adult patients with acute myeloid leukemia (AML), and these alterations create an additional nuclear export signal that relocalizes much of the protein from its normal nucleolar stores to the cytoplasm. When induced by oncogenic stress, the Arf tumor suppressor protein accumulates within the nucleolus, where it is physically associated with, and stabilized by, NPM. Ectopic overexpression of an NPM cytoplasmic mutant (NPMc) relocalized p19Arf and the endogenous NPM protein to the cytoplasm. NPMc-dependent export of p19Arf from the nucleus inhibited its functional interaction with the p53 negative regulator, Mdm2, and blunted Arf-induced activation of the p53 transcriptional program. Cytoplasmic NPM relocalization also attenuated Arf-induced sumoylation of Mdm2 and NPM and prevented wild type NPM from inhibiting p19Arf protein turnover. However, despite the ability of NPMc to interfere with these p53-dependent and independent activities of Arf, NPMc exhibited anti-proliferative activity in Arf-null NIH-3T3 cells. Overexpression of wild type NPM, but not NPMc, overcame premature senescence of Atm-null cells, a phenotype that can be rescued by inactivation of Arf or p53. Therefore, perturbation of Arf function appears to be insufficient to explain the oncogenic effects of the NPMc mutation. We favor the idea that NPMc also contributes to AML by dominantly perturbing other functions of the wild type NPM protein.


Sujet(s)
Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/métabolisme , Mutagenèse par insertion , Protéines nucléaires/génétique , Protéines oncogènes/génétique , Protéine p14(ARF) suppresseur de tumeur/antagonistes et inhibiteurs , Protéine p53 suppresseur de tumeur/physiologie , Animaux , Cytoplasme/génétique , Cytoplasme/métabolisme , Cytoplasme/physiologie , Humains , Souris , Cellules NIH 3T3 , Protéines nucléaires/antagonistes et inhibiteurs , Protéines nucléaires/métabolisme , Nucléophosmine , Protéines oncogènes/physiologie , Transport des protéines/génétique , Transport des protéines/physiologie , Transfection , Protéine p14(ARF) suppresseur de tumeur/biosynthèse , Protéine p14(ARF) suppresseur de tumeur/physiologie , Protéine p53 suppresseur de tumeur/antagonistes et inhibiteurs
18.
Cell Cycle ; 3(11): 1367-9, 2004 Nov.
Article de Anglais | MEDLINE | ID: mdl-15467472

RÉSUMÉ

Ubiquitin-dependent proteolysis by proteasomes generally depends upon the conjugation of polyubiquitin chains to lysine epsilon-NH(2) groups within the targeted proteins. However, engineered lysine-less mutants of certain viral and cellular proteins can undergo polyubiquitination at their N-termini. Is N-terminal polyubiquitination a physiologic process, and how many cellular proteins can be targeted for proteasomal degradation through this mechanism? Recent work indicates that the turnover of the endogenous lysine-less human ARF tumor suppressor protein and its mouse Arf counterpart (containing a single, non-conserved lysine residue) is regulated in this manner.


Sujet(s)
Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Polyubiquitine/métabolisme , Maturation post-traductionnelle des protéines , Inhibiteur p16 de kinase cycline-dépendante/composition chimique , Inhibiteur p16 de kinase cycline-dépendante/génétique , Lysine/composition chimique , Mutagenèse dirigée , Proteasome endopeptidase complex/métabolisme
19.
Genes Dev ; 18(15): 1862-74, 2004 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-15289458

RÉSUMÉ

Unknown mechanisms govern degradation of the p19Arf tumor suppressor, an activator of p53 and inhibitor of ribosomal RNA processing. Kinetic metabolic labeling of cells with [3H]-leucine indicated that p19Arf is a relatively stable protein (half-life approximately 6 h) whose degradation depends upon the ubiquitin-proteasome pathway. Although p19Arf binds to the Mdm2 E3 ubiquitin protein ligase to activate p53, neither of these molecules regulates p19Arf turnover. In contrast, the nucleolar protein nucleophosmin/B23, which binds to p19Arf with high stoichiometry, retards its turnover, and Arf mutants that do not efficiently associate with nucleophosmin/B23 are unstable and functionally impaired. Mouse p19Arf, although highly basic (22% arginine content), contains only a single lysine residue absent from human p14ARF, and substitution of arginine for lysine in mouse p19Arf had no effect on its rate of degradation. Mouse p19Arf (either wild-type or lacking lysine) and human p14ARF undergo N-terminal polyubiquitination, a process that has not as yet been documented in naturally occurring lysine-less proteins. Re-engineering of the p19Arf N terminus to provide consensus sequences for N-acetylation limited Arf ubiquitination and decelerated its turnover.


Sujet(s)
Cysteine endopeptidases/métabolisme , Complexes multienzymatiques/métabolisme , Protéine p14(ARF) suppresseur de tumeur/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitine/métabolisme , Acétylation , Séquence d'acides aminés , Animaux , Inhibiteur p16 de kinase cycline-dépendante , Gènes suppresseurs de tumeur/physiologie , Gènes p16/physiologie , Humains , Leucine/métabolisme , Souris , Données de séquences moléculaires , Mutation/génétique , Cellules NIH 3T3 , Protéines nucléaires/métabolisme , Nucléophosmine , Proteasome endopeptidase complex , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes c-mdm2 , Protéine p53 suppresseur de tumeur/métabolisme
20.
Cancer Res ; 63(5): 1046-53, 2003 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-12615721

RÉSUMÉ

The tumor suppressor p19(Arf) (p14(ARF) in humans), encoded by the Ink4a/Arf locus, is mutated, deleted, or silenced in many forms of cancer. p19(Arf) induces growth arrest by antagonizing the activity of the p53-negative regulator, Mdm2, thereby inducing a p53 transcriptional response. p19(Arf) can also inhibit cell cycle progression of mouse embryo fibroblasts lacking Cip1 or lacking both Mdm2 and p53, although in the absence of p53, arrest occurs more slowly. Profiling with high-density oligonucleotide GeneChips and cDNA microarrays was used to interrogate mouse genes, the expression of which was induced or suppressed by a conditionally regulated Arf gene. Cluster analysis of temporal gene expression patterns and validation of the results by RNA analysis identified Arf-responsive genes whose induction was both p53-dependent and -independent. The latter included four members of the B-cell translocation gene family (Btg1, Btg2, Btg3, and Tob1) that were demonstrated to inhibit cell proliferation in primary mouse embryo fibroblasts expressing or lacking functional p53. Together, the results indicate that p19(Arf) induces a broad spectrum of proteins that likely act in concert to arrest cell proliferation.


Sujet(s)
Protéines nucléaires , Protéine p14(ARF) suppresseur de tumeur/physiologie , Protéine p53 suppresseur de tumeur/physiologie , Cellules 3T3 , Animaux , Cycle cellulaire/génétique , Division cellulaire/génétique , Inhibiteur p16 de kinase cycline-dépendante , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes/génétique , Souris , Séquençage par oligonucléotides en batterie , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes c-mdm2 , Ovis , Activation de la transcription/génétique , Transfection , Protéine p14(ARF) suppresseur de tumeur/biosynthèse , Protéine p14(ARF) suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...