Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 13 de 13
Filtrer
1.
Cell Chem Biol ; 2024 Mar 19.
Article de Anglais | MEDLINE | ID: mdl-38537632

RÉSUMÉ

This study describes the identification and target deconvolution of small molecule inhibitors of oncogenic Yes-associated protein (YAP1)/TAZ activity with potent anti-tumor activity in vivo. A high-throughput screen (HTS) of 3.8 million compounds was conducted using a cellular YAP1/TAZ reporter assay. Target deconvolution studies identified the geranylgeranyltransferase-I (GGTase-I) complex as the direct target of YAP1/TAZ pathway inhibitors. The small molecule inhibitors block the activation of Rho-GTPases, leading to subsequent inactivation of YAP1/TAZ and inhibition of cancer cell proliferation in vitro. Multi-parameter optimization resulted in BAY-593, an in vivo probe with favorable PK properties, which demonstrated anti-tumor activity and blockade of YAP1/TAZ signaling in vivo.

2.
J Med Chem ; 63(15): 8025-8042, 2020 08 13.
Article de Anglais | MEDLINE | ID: mdl-32338514

RÉSUMÉ

Inhibition of monopolar spindle 1 (MPS1) kinase represents a novel approach to cancer treatment: instead of arresting the cell cycle in tumor cells, cells are driven into mitosis irrespective of DNA damage and unattached/misattached chromosomes, resulting in aneuploidy and cell death. Starting points for our optimization efforts with the goal to identify MPS1 inhibitors were two HTS hits from the distinct chemical series "triazolopyridines" and "imidazopyrazines". The major initial issue of the triazolopyridine series was the moderate potency of the HTS hits. The imidazopyrazine series displayed more than 10-fold higher potencies; however, in the early project phase, this series suffered from poor metabolic stability. Here, we outline the evolution of the two hit series to clinical candidates BAY 1161909 and BAY 1217389 and reveal how both clinical candidates bind to the ATP site of MPS1 kinase, while addressing different pockets utilizing different binding interactions, along with their synthesis and preclinical characterization in selected in vivo efficacy models.


Sujet(s)
Antinéoplasiques/métabolisme , Protéines du cycle cellulaire/métabolisme , Systèmes de délivrance de médicaments/méthodes , Découverte de médicament/méthodes , Points de contrôle de la phase M du cycle cellulaire/effets des médicaments et des substances chimiques , Protein-Serine-Threonine Kinases/métabolisme , Protein-tyrosine kinases/métabolisme , Appareil du fuseau/effets des médicaments et des substances chimiques , Animaux , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Chiens , Femelle , Cellules HT29 , Cellules HeLa , Humains , Points de contrôle de la phase M du cycle cellulaire/physiologie , Mâle , Microsomes du foie/effets des médicaments et des substances chimiques , Microsomes du foie/métabolisme , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Structure tertiaire des protéines , Protein-tyrosine kinases/antagonistes et inhibiteurs , Rats , Rat Wistar , Appareil du fuseau/métabolisme , Résultat thérapeutique
3.
Clin Cancer Res ; 25(4): 1404-1414, 2019 02 15.
Article de Anglais | MEDLINE | ID: mdl-30429199

RÉSUMÉ

PURPOSE: The catalytic function of BUB1 is required for chromosome arm resolution and positioning of the chromosomal passenger complex for resolution of spindle attachment errors and plays only a minor role in spindle assembly checkpoint activation. Here, we present the identification and preclinical pharmacologic profile of the first BUB1 kinase inhibitor with good bioavailability. EXPERIMENTAL DESIGN: The Bayer compound library was screened for BUB1 kinase inhibitors and medicinal chemistry efforts to improve target affinity and physicochemical and pharmacokinetic parameters resulting in the identification of BAY 1816032 were performed. BAY 1816032 was characterized for kinase selectivity, inhibition of BUB1 signaling, and inhibition of tumor cell proliferation alone and in combination with taxanes, ATR, and PARP inhibitors. Effects on tumor growth in vivo were evaluated using human triple-negative breast xenograft models. RESULTS: The highly selective compound BAY 1816032 showed long target residence time and induced chromosome mis-segregation upon combination with low concentrations of paclitaxel. It was synergistic or additive in combination with paclitaxel or docetaxel, as well as with ATR or PARP inhibitors in cellular assays. Tumor xenograft studies demonstrated a strong and statistically significant reduction of tumor size and excellent tolerability upon combination of BAY 1816032 with paclitaxel or olaparib as compared with the respective monotherapies. CONCLUSIONS: Our findings suggest clinical proof-of-concept studies evaluating BAY 1816032 in combination with taxanes or PARP inhibitors to enhance their efficacy and potentially overcome resistance.


Sujet(s)
Résistance aux médicaments antinéoplasiques/génétique , Tumeurs/traitement médicamenteux , Inhibiteurs de protéines kinases/pharmacologie , Protein-Serine-Threonine Kinases/génétique , Animaux , Protéines mutées dans l'ataxie-télangiectasie/antagonistes et inhibiteurs , Protéines mutées dans l'ataxie-télangiectasie/génétique , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Cellules HeLa , Humains , Souris , Tumeurs/génétique , Tumeurs/anatomopathologie , Phtalazines/pharmacologie , Pipérazines/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Taxoïdes/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe
4.
J Med Chem ; 62(2): 928-940, 2019 01 24.
Article de Anglais | MEDLINE | ID: mdl-30563338

RÉSUMÉ

The availability of a chemical probe to study the role of a specific domain of a protein in a concentration- and time-dependent manner is of high value. Herein, we report the identification of a highly potent and selective ERK5 inhibitor BAY-885 by high-throughput screening and subsequent structure-based optimization. ERK5 is a key integrator of cellular signal transduction, and it has been shown to play a role in various cellular processes such as proliferation, differentiation, apoptosis, and cell survival. We could demonstrate that inhibition of ERK5 kinase and transcriptional activity with a small molecule did not translate into antiproliferative activity in different relevant cell models, which is in contrast to the results obtained by RNAi technology.


Sujet(s)
Mitogen-Activated Protein Kinase 7/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/composition chimique , Pyridines/composition chimique , Pyrimidines/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Sites de fixation , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cristallographie aux rayons X , Évaluation préclinique de médicament , Période , Humains , Mitogen-Activated Protein Kinase 7/métabolisme , Simulation de docking moléculaire , Inhibiteurs de protéines kinases/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Structure tertiaire des protéines , Pyridines/métabolisme , Pyridines/pharmacologie , Pyrimidines/métabolisme , Pyrimidines/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Relation structure-activité , Transcription génétique/effets des médicaments et des substances chimiques
5.
Cell Death Dis ; 8(3): e2709, 2017 03 30.
Article de Anglais | MEDLINE | ID: mdl-28358364

RÉSUMÉ

Owing to lagging or insufficient neo-angiogenesis, hypoxia is a feature of most solid tumors. Hypoxic tumor regions contribute to resistance against antiproliferative chemotherapeutics, radiotherapy and immunotherapy. Targeting cells in hypoxic tumor areas is therefore an important strategy for cancer treatment. Most approaches for targeting hypoxic cells focus on the inhibition of hypoxia adaption pathways but only a limited number of compounds with the potential to specifically target hypoxic tumor regions have been identified. By using tumor spheroids in hypoxic conditions as screening system, we identified a set of compounds, including the phenothiazine antipsychotic Fluphenazine, as hits with novel mode of action. Fluphenazine functionally inhibits acid sphingomyelinase and causes cellular sphingomyelin accumulation, which induces cancer cell death specifically in hypoxic tumor spheroids. Moreover, we found that functional inhibition of acid sphingomyelinase leads to overactivation of hypoxia stress-response pathways and that hypoxia-specific cell death is mediated by the stress-responsive transcription factor ATF4. Taken together, the here presented data suggest a novel, yet unexplored mechanism in which induction of sphingolipid stress leads to the overactivation of hypoxia stress-response pathways and thereby promotes their pro-apoptotic tumor-suppressor functions to specifically kill cells in hypoxic tumor areas.


Sujet(s)
Tumeurs du côlon/enzymologie , Fluphénazine/pharmacologie , Protéines tumorales/antagonistes et inhibiteurs , Sphingomyeline phosphodiesterase/antagonistes et inhibiteurs , Facteur de transcription ATF-4/génétique , Facteur de transcription ATF-4/métabolisme , Mort cellulaire/effets des médicaments et des substances chimiques , Hypoxie cellulaire/effets des médicaments et des substances chimiques , Hypoxie cellulaire/génétique , Lignée cellulaire tumorale , Tumeurs du côlon/génétique , Tumeurs du côlon/anatomopathologie , Humains , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Sphingomyeline phosphodiesterase/génétique
6.
J Biol Chem ; 291(21): 11252-67, 2016 05 20.
Article de Anglais | MEDLINE | ID: mdl-27030009

RÉSUMÉ

The spindle assembly checkpoint (SAC) is an essential safeguarding mechanism devised to ensure equal chromosome distribution in daughter cells upon mitosis. The proteins Bub3 and BubR1 are key components of the mitotic checkpoint complex, an essential part of the molecular machinery on which the SAC relies. In the present work we have performed a detailed functional and biochemical characterization of the interaction between human Bub3 and BubR1 in cells and in vitro Our results demonstrate that genetic knockdown of Bub3 abrogates the SAC, promotes apoptosis, and inhibits the proliferation of human cancer cells. We also show that the integrity of the human mitotic checkpoint complex depends on the specific recognition between BubR1 and Bub3, for which the BubR1 Gle2 binding sequence motif is essential. This 1:1 binding event is high affinity, enthalpy-driven and with slow dissociation kinetics. The affinity, kinetics, and thermodynamic parameters of the interaction are differentially modulated by small regions in the N and C termini of the Gle2 binding domain sequence, suggesting the existence of "hotspots" for this protein-protein interaction. Furthermore, we show that specific disruption of endogenous BubR1·Bub3 complexes in human cancer cells phenocopies the effects observed in gene targeting experiments. Our work enhances the current understanding of key members of the SAC and paves the road for the pursuit of novel targeted cancer therapies based on SAC inhibition.


Sujet(s)
Protéines du cycle cellulaire/composition chimique , Protéines du cycle cellulaire/métabolisme , Points de contrôle de la phase M du cycle cellulaire/physiologie , Protein-Serine-Threonine Kinases/composition chimique , Protein-Serine-Threonine Kinases/métabolisme , Appareil du fuseau/métabolisme , Apoptose , Protéines du cycle cellulaire/génétique , Lignée cellulaire , Lignée cellulaire tumorale , Prolifération cellulaire , Techniques de knock-down de gènes , Cellules HeLa , Humains , Cinétique , Points de contrôle de la phase M du cycle cellulaire/génétique , Cellules MCF-7 , Modèles moléculaires , Protéines liant le poly-adp-ribose , Motifs et domaines d'intéraction protéique , Protein-Serine-Threonine Kinases/génétique , ARN messager/génétique , ARN messager/métabolisme , Appareil du fuseau/génétique , Thermodynamique
7.
Mol Cancer Ther ; 15(4): 583-92, 2016 04.
Article de Anglais | MEDLINE | ID: mdl-26832791

RÉSUMÉ

Monopolar spindle 1 (Mps1) has been shown to function as the key kinase that activates the spindle assembly checkpoint (SAC) to secure proper distribution of chromosomes to daughter cells. Here, we report the structure and functional characterization of two novel selective Mps1 inhibitors, BAY 1161909 and BAY 1217389, derived from structurally distinct chemical classes. BAY 1161909 and BAY 1217389 inhibited Mps1 kinase activity with IC50 values below 10 nmol/L while showing an excellent selectivity profile. In cellular mechanistic assays, both Mps1 inhibitors abrogated nocodazole-induced SAC activity and induced premature exit from mitosis ("mitotic breakthrough"), resulting in multinuclearity and tumor cell death. Both compounds efficiently inhibited tumor cell proliferation in vitro (IC50 nmol/L range). In vivo, BAY 1161909 and BAY 1217389 achieved moderate efficacy in monotherapy in tumor xenograft studies. However, in line with its unique mode of action, when combined with paclitaxel, low doses of Mps1 inhibitor reduced paclitaxel-induced mitotic arrest by the weakening of SAC activity. As a result, combination therapy strongly improved efficacy over paclitaxel or Mps1 inhibitor monotreatment at the respective MTDs in a broad range of xenograft models, including those showing acquired or intrinsic paclitaxel resistance. Both Mps1 inhibitors showed good tolerability without adding toxicity to paclitaxel monotherapy. These preclinical findings validate the innovative concept of SAC abrogation for cancer therapy and justify clinical proof-of-concept studies evaluating the Mps1 inhibitors BAY 1161909 and BAY 1217389 in combination with antimitotic cancer drugs to enhance their efficacy and potentially overcome resistance. Mol Cancer Ther; 15(4); 583-92. ©2016 AACR.


Sujet(s)
Antinéoplasiques/pharmacologie , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/pharmacologie , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protein-tyrosine kinases/antagonistes et inhibiteurs , Animaux , Antinéoplasiques/composition chimique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Découverte de médicament , Évaluation préclinique de médicament , Activation enzymatique/effets des médicaments et des substances chimiques , Femelle , Humains , Mâle , Souris , Mitose/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/composition chimique , Rats , Charge tumorale/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
8.
Exp Cell Res ; 339(1): 35-43, 2015 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-26475730

RÉSUMÉ

Invasion processes underlie or accompany several pathological processes but only a limited number of high-throughput capable phenotypic models exist to test anti-invasive compounds in vitro. We here evaluated 3D co-cultures as a high-content phenotypic screening system for fibrotic invasive processes. 3D multicellular spheroids were used as living tissue surrogates in co-culture with fluorescently labeled lung fibroblasts to monitor invasion processes by automated microscopy. This setup was used to screen a compound library containing 480 known bioactive substances. Identified hits prevented fibroblast invasion and could be subdivided into two hit classes. First, Prostaglandins were shown to prevent fibroblast invasion, most likely mediated by the prostaglandin EP2 receptor and generation of cAMP. Additionally, Rho-associated protein kinase (ROCK) inhibitors prevented fibroblast invasion, possibly by inactivation of myosin II. Importantly, both Prostaglandins and ROCK inhibitors are potential treatment options shown to be effective in in vitro and in vivo models of fibrotic diseases. This validates the presented novel phenotypic screening approach for the evaluation of potential inhibitors and the identification of novel compounds with activity in diseases that are associated with fibroblast invasion.


Sujet(s)
Tumeurs du sein/anatomopathologie , Techniques de culture cellulaire , Fibroblastes/anatomopathologie , Tests de criblage à haut débit , Prostaglandines/pharmacologie , Inhibiteurs de protéines kinases/pharmacologie , Sphéroïdes de cellules/anatomopathologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Femelle , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Humains , Traitement d'image par ordinateur , Techniques immunoenzymatiques , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Sphéroïdes de cellules/métabolisme , Cellules cancéreuses en culture , rho-Associated Kinases/antagonistes et inhibiteurs , rho-Associated Kinases/métabolisme
9.
J Biomol Screen ; 20(2): 190-201, 2015 Feb.
Article de Anglais | MEDLINE | ID: mdl-25409661

RÉSUMÉ

EZH2 inhibition can decrease global histone H3 lysine 27 trimethylation (H3K27me3) and thereby reactivates silenced tumor suppressor genes. Inhibition of EZH2 is regarded as an option for therapeutic cancer intervention. To identify novel small-molecule (SMOL) inhibitors of EZH2 in drug discovery, trustworthy cellular assays amenable for phenotypic high-throughput screening (HTS) are crucial. We describe a reliable approach that quantifies changes in global levels of histone modification marks using high-content analysis (HCA). The approach was validated in different cell lines by using small interfering RNA and SMOL inhibitors. By automation and miniaturization from a 384-well to 1536-well plate, we demonstrated its utility in conducting phenotypic HTS campaigns and assessing structure-activity relationships (SAR). This assay enables screening of SMOL EZH2 inhibitors and can advance the mechanistic understanding of H3K27me3 suppression, which is crucial with regard to epigenetic therapy. We observed that a decrease in global H3K27me3, induced by EZH2 inhibition, comprises two distinct mechanisms: (1) inhibition of de novo DNA methylation and (II) inhibition of dynamic, replication-independent H3K27me3 turnover. This report describes an HCA assay for primary HTS to identify, profile, and optimize cellular active SMOL inhibitors targeting histone methyltransferases, which could benefit epigenetic drug discovery.


Sujet(s)
Découverte de médicament , Tests de criblage à haut débit , Histone/métabolisme , Microscopie , Complexe répresseur Polycomb-2/antagonistes et inhibiteurs , Bibliothèques de petites molécules , Laboratoire automatique , Cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Relation dose-effet des médicaments , Protéine-2 homologue de l'activateur de Zeste , Techniques de knock-down de gènes , Histone/antagonistes et inhibiteurs , Histone/génétique , Humains , Concentration inhibitrice 50 , Méthylation/effets des médicaments et des substances chimiques , Interférence par ARN , Relation structure-activité
10.
Exp Cell Res ; 323(1): 131-143, 2014 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-24480576

RÉSUMÉ

Cancer cells in poorly vascularized tumor regions need to adapt to an unfavorable metabolic microenvironment. As distance from supplying blood vessels increases, oxygen and nutrient concentrations decrease and cancer cells react by stopping cell cycle progression and becoming dormant. As cytostatic drugs mainly target proliferating cells, cancer cell dormancy is considered as a major resistance mechanism to this class of anti-cancer drugs. Therefore, substances that target cancer cells in poorly vascularized tumor regions have the potential to enhance cytostatic-based chemotherapy of solid tumors. With three-dimensional growth conditions, multicellular tumor spheroids (MCTS) reproduce several parameters of the tumor microenvironment, including oxygen and nutrient gradients as well as the development of dormant tumor regions. We here report the setup of a 3D cell culture compatible high-content screening system and the identification of nine substances from two commercially available drug libraries that specifically target cells in inner MCTS core regions, while cells in outer MCTS regions or in 2D cell culture remain unaffected. We elucidated the mode of action of the identified compounds as inhibitors of the respiratory chain and show that induction of cell death in inner MCTS core regions critically depends on extracellular glucose concentrations. Finally, combinational treatment with cytostatics showed increased induction of cell death in MCTS. The data presented here shows for the first time a high-content based screening setup on 3D tumor spheroids for the identification of substances that specifically induce cell death in inner tumor spheroid core regions. This validates the approach to use 3D cell culture screening systems to identify substances that would not be detectable by 2D based screening in otherwise similar culture conditions.


Sujet(s)
Antinéoplasiques/isolement et purification , Antienzymes/isolement et purification , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Techniques de culture cellulaire , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tests de criblage d'agents antitumoraux/méthodes , Transport d'électrons/effets des médicaments et des substances chimiques , Antienzymes/pharmacologie , Femelle , Glucose/métabolisme , Humains , Staurosporine/pharmacologie , Cellules cancéreuses en culture , Microenvironnement tumoral/physiologie
11.
Comb Chem High Throughput Screen ; 11(3): 216-30, 2008 Mar.
Article de Anglais | MEDLINE | ID: mdl-18336214

RÉSUMÉ

High-Content Analysis (HCA) has developed into an established tool and is used in a wide range of academic laboratories and pharmaceutical research groups. HCA is now routinely proving to be effective in providing functionally relevant results. It is essential to select the appropriate HCA application with regard to the targeted compound's cellular function. The cellular impact and compound specificity as revealed by HCA analysis facilitates reaching definitive conclusions at an early stage in the drug discovery process. This technology therefore has the potential to substantially improve the efficiency of pharmaceutical research. Recent advances in fluorescent probes have significantly boosted the success of HCA. Auto-fluorescent proteins which minimally hinder the functioning of the living cell have been playing a decisive role in cell biology research. For companies the severely restricted license conditions regarding auto-fluorescent proteins hamper their general use in pharmaceutical research. This has opened the field for other solutions such as self-labeling protein technology, which could potentially replace the well established methods that utilize auto-fluorescent proteins. In addition, direct labeling techniques have improved considerably and may supersede many of the approaches based on fusion proteins. Following sample preparation, treated cells are imaged and the resulting multiple fluorescent signals are subjected to contextual and statistical analysis. The extraordinary advantage of HCA is that it enables the large-scale and simultaneous quantification and correlation of multiple phenotypic responses and physiological reactions using sophisticated software solutions that permit assay-specific image analysis. Hence, HCA once more has demonstrated its outstanding potential to significantly support establishing effective pharmaceutical research processes in order to both advance research projects and cut costs.


Sujet(s)
Évaluation préclinique de médicament/méthodes , Cytométrie à balayage laser/méthodes , Animaux , Biologie informatique , Évaluation préclinique de médicament/instrumentation , Humains , Traitement d'image par ordinateur/méthodes , Cytométrie à balayage laser/instrumentation , Bibliothèques de petites molécules
12.
Mol Biol Cell ; 18(10): 4024-36, 2007 Oct.
Article de Anglais | MEDLINE | ID: mdl-17671160

RÉSUMÉ

Polo-like kinase 1 (Plk1) is a key regulator of mitotic progression and cell division in eukaryotes. It is highly expressed in tumor cells and considered a potential target for cancer therapy. Here, we report the discovery and application of a novel potent small-molecule inhibitor of mammalian Plk1, ZK-Thiazolidinone (TAL). We have extensively characterized TAL in vitro and addressed TAL specificity within cells by studying Plk1 functions in sister chromatid separation, centrosome maturation, and spindle assembly. Moreover, we have used TAL for a detailed analysis of Plk1 in relation to PICH and PRC1, two prominent interaction partners implicated in spindle assembly checkpoint function and cytokinesis, respectively. Specifically, we show that Plk1, when inactivated by TAL, spreads over the arms of chromosomes, resembling the localization of its binding partner PICH, and that both proteins are mutually dependent on each other for correct localization. Finally, we show that Plk1 activity is essential for cleavage furrow formation and ingression, leading to successful cytokinesis.


Sujet(s)
Dérivés de l'aniline/pharmacologie , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Protéines du cycle cellulaire/métabolisme , Mitose/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/pharmacologie , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protein-Serine-Threonine Kinases/métabolisme , Protéines proto-oncogènes/antagonistes et inhibiteurs , Protéines proto-oncogènes/métabolisme , Thiazolidines/pharmacologie , Anaphase/effets des médicaments et des substances chimiques , Dérivés de l'aniline/composition chimique , Animaux , Lignée cellulaire tumorale , Centrosome/effets des médicaments et des substances chimiques , Centrosome/enzymologie , Chromatides/effets des médicaments et des substances chimiques , Chromatides/enzymologie , Cytocinèse/effets des médicaments et des substances chimiques , Helicase , Activation enzymatique/effets des médicaments et des substances chimiques , Humains , Souris , Index mitotique , Inhibiteurs de protéines kinases/composition chimique , Appareil du fuseau/effets des médicaments et des substances chimiques , Appareil du fuseau/enzymologie , Thiazolidines/composition chimique ,
13.
Proc Natl Acad Sci U S A ; 99(18): 11808-12, 2002 Sep 03.
Article de Anglais | MEDLINE | ID: mdl-12189207

RÉSUMÉ

Certain IFN regulatory factor (IRF) transcription factors indirectly influence T helper (Th) cell differentiation by regulating the production of IL-12. Here, we show that IRF4 directly regulates Th cell differentiation in vitro and in vivo during murine leishmaniasis. In the absence of IRF4, IL-12-induced Th1 cell differentiation was compromised, while IL-4 failed to induce Th2 cell differentiation. Instead, IL-4 tended to induce Th1 cells, defined by production of IFN-gamma and TNF. Although early IL-4 signaling was normal in IRF4(-/-) Th cells, the protein GATA-3, a transcription factor critical for Th2 development, was not up-regulated following IL-4 treatment. Retroviral overexpression of GATA-3 rescued Th2 differentiation. Therefore, IRF4 deficiency manifests itself as severely dysregulated Th cell differentiation.


Sujet(s)
Différenciation cellulaire/physiologie , Protéines de liaison à l'ADN/physiologie , Lymphocytes T auxiliaires/cytologie , Facteurs de transcription/physiologie , Animaux , Technique de Western , Protéines de liaison à l'ADN/génétique , Facteur de transcription GATA-3 , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Facteurs de régulation d'interféron , Interleukine-4/pharmacologie , Leishmaniose/immunologie , Leishmaniose/anatomopathologie , Souris , Souris de lignée C57BL , Phosphorylation , Facteur de transcription STAT-6 , Lymphocytes T auxiliaires/effets des médicaments et des substances chimiques , Transactivateurs/génétique , Transactivateurs/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...