Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
ACS Omega ; 6(34): 22073-22102, 2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34497901

RESUMEN

Following the identification of thieno[2,3-d]pyrimidine-based selective and potent inhibitors of MCL-1, we explored the effect of core swapping at different levels of advancement. During hit-to-lead optimization, X-ray-guided S-N replacement in the core provided a new vector, whose exploration led to the opening of the so-called deep-S2 pocket of MCL-1. Unfortunately, the occupation of this region led to a plateau in affinity and had to be abandoned. As the project approached selection of a clinical candidate, a series of core swap analogues were also prepared. The affinity and cellular activity of these compounds showed a significant dependence on the core structure. In certain cases, we also observed an increased and accelerated epimerization of the atropoisomers. The most potent core replacement analogues showed considerable in vivo PD response. One compound was progressed into efficacy studies and inhibited tumor growth.

2.
J Med Chem ; 63(22): 13762-13795, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33146521

RESUMEN

Myeloid cell leukemia 1 (Mcl-1) has emerged as an attractive target for cancer therapy. It is an antiapoptotic member of the Bcl-2 family of proteins, whose upregulation in human cancers is associated with high tumor grade, poor survival, and resistance to chemotherapy. Here we report the discovery of our clinical candidate S64315, a selective small molecule inhibitor of Mcl-1. Starting from a fragment derived lead compound, we have conducted structure guided optimization that has led to a significant (3 log) improvement of target affinity as well as cellular potency. The presence of hindered rotation along a biaryl axis has conferred high selectivity to the compounds against other members of the Bcl-2 family. During optimization, we have also established predictive PD markers of Mcl-1 inhibition and achieved both efficient in vitro cell killing and tumor regression in Mcl-1 dependent cancer models. The preclinical candidate has drug-like properties that have enabled its development and entry into clinical trials.


Asunto(s)
Antineoplásicos/química , Descubrimiento de Drogas/métodos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/antagonistas & inhibidores , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/química , Animales , Antineoplásicos/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Células HCT116 , Células HeLa , Humanos , Ratones , Ratones SCID , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
3.
J Med Chem ; 62(15): 6913-6924, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31339316

RESUMEN

Myeloid cell leukemia 1 (Mcl-1), an antiapoptotic member of the Bcl-2 family of proteins, whose upregulation when observed in human cancers is associated with high tumor grade, poor survival, and resistance to chemotherapy, has emerged as an attractive target for cancer therapy. Here, we report the discovery of selective small molecule inhibitors of Mcl-1 that inhibit cellular activity. Fragment screening identified thienopyrimidine amino acids as promising but nonselective hits that were optimized using nuclear magnetic resonance and X-ray-derived structural information. The introduction of hindered rotation along a biaryl axis has conferred high selectivity to the compounds, and cellular activity was brought on scale by offsetting the negative charge of the anchoring carboxylate group. The obtained compounds described here exhibit nanomolar binding affinity and mechanism-based cellular efficacy, caspase induction, and growth inhibition. These early research efforts illustrate drug discovery optimization from thienopyrimidine hits to a lead compound, the chemical series leading to the identification of our more advanced compounds S63845 and S64315.


Asunto(s)
Supervivencia Celular/fisiología , Descubrimiento de Drogas/métodos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/antagonistas & inhibidores , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Pirimidinas/química , Pirimidinas/metabolismo , Tiofenos/química , Tiofenos/metabolismo , Supervivencia Celular/efectos de los fármacos , Células HCT116 , Células HeLa , Humanos , Estructura Terciaria de Proteína , Pirimidinas/farmacología , Relación Estructura-Actividad , Tiofenos/farmacología
4.
Leukemia ; 33(4): 905-917, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30214012

RESUMEN

Improving outcomes in acute myeloid leukemia (AML) remains a major clinical challenge. Overexpression of pro-survival BCL-2 family members rendering transformed cells resistant to cytotoxic drugs is a common theme in cancer. Targeting BCL-2 with the BH3-mimetic venetoclax is active in AML when combined with low-dose chemotherapy or hypomethylating agents. We now report the pre-clinical anti-leukemic efficacy of a novel BCL-2 inhibitor S55746, which demonstrates synergistic pro-apoptotic activity in combination with the MCL1 inhibitor S63845. Activity of the combination was caspase and BAX/BAK dependent, superior to combination with standard cytotoxic AML drugs and active against a broad spectrum of poor risk genotypes, including primary samples from patients with chemoresistant AML. Co-targeting BCL-2 and MCL1 was more effective against leukemic, compared to normal hematopoietic progenitors, suggesting a therapeutic window of activity. Finally, S55746 combined with S63845 prolonged survival in xenograft models of AML and suppressed patient-derived leukemia but not normal hematopoietic cells in bone marrow of engrafted mice. In conclusion, a dual BH3-mimetic approach is feasible, highly synergistic, and active in diverse models of human AML. This approach has strong clinical potential to rapidly suppress leukemia, with reduced toxicity to normal hematopoietic precursors compared to chemotherapy.


Asunto(s)
Biomimética , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Pirimidinas/farmacología , Sulfonamidas/farmacología , Tiofenos/farmacología , Animales , Antineoplásicos/farmacología , Quimioterapia Combinada , Femenino , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Fragmentos de Péptidos , Proteínas Proto-Oncogénicas , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Oncotarget ; 9(28): 20075-20088, 2018 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-29732004

RESUMEN

Escape from apoptosis is one of the major hallmarks of cancer cells. The B-cell Lymphoma 2 (BCL-2) gene family encodes pro-apoptotic and anti-apoptotic proteins that are key regulators of the apoptotic process. Overexpression of the pro-survival member BCL-2 is a well-established mechanism contributing to oncogenesis and chemoresistance in several cancers, including lymphoma and leukemia. Thus, BCL-2 has become an attractive target for therapeutic strategy in cancer, as demonstrated by the recent approval of ABT-199 (Venclexta™) in relapsed or refractory Chronic Lymphocytic Leukemia with 17p deletion. Here, we describe a novel orally bioavailable BCL-2 selective and potent inhibitor called S55746 (also known as BCL201). S55746 occupies the hydrophobic groove of BCL-2. Its selectivity profile demonstrates no significant binding to MCL-1, BFL-1 (BCL2A1/A1) and poor affinity for BCL-XL. Accordingly, S55746 has no cytotoxic activity on BCL-XL-dependent cells, such as platelets. In a panel of hematological cell lines, S55746 induces hallmarks of apoptosis including externalization of phosphatidylserine, caspase-3 activation and PARP cleavage. Ex vivo, S55746 induces apoptosis in the low nanomolar range in primary Chronic Lymphocytic Leukemia and Mantle Cell Lymphoma patient samples. Finally, S55746 administered by oral route daily in mice demonstrated robust anti-tumor efficacy in two hematological xenograft models with no weight lost and no change in behavior. Taken together, these data demonstrate that S55746 is a novel, well-tolerated BH3-mimetic targeting selectively and potently the BCL-2 protein.

6.
Nat Struct Mol Biol ; 24(3): 279-289, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28165510

RESUMEN

Type I interferons (IFNs) are multifunctional cytokines that regulate immune responses and cellular functions but also can have detrimental effects on human health. A tight regulatory network therefore controls IFN signaling, which in turn may interfere with medical interventions. The JAK-STAT signaling pathway transmits the IFN extracellular signal to the nucleus, thus resulting in alterations in gene expression. STAT2 is a well-known essential and specific positive effector of type I IFN signaling. Here, we report that STAT2 is also a previously unrecognized, crucial component of the USP18-mediated negative-feedback control in both human and mouse cells. We found that STAT2 recruits USP18 to the type I IFN receptor subunit IFNAR2 via its constitutive membrane-distal STAT2-binding site. This mechanistic coupling of effector and negative-feedback functions of STAT2 may provide novel strategies for treatment of IFN-signaling-related human diseases.


Asunto(s)
Endopeptidasas/metabolismo , Interferón Tipo I/metabolismo , Factor de Transcripción STAT2/metabolismo , Transducción de Señal , Animales , Línea Celular Tumoral , Retroalimentación Fisiológica , Humanos , Immunoblotting , Ratones , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Unión Proteica , Dominios Proteicos , Receptor de Interferón alfa y beta/metabolismo , Factor de Transcripción STAT2/química , Técnicas del Sistema de Dos Híbridos , Ubiquitina Tiolesterasa
7.
Nature ; 538(7626): 477-482, 2016 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-27760111

RESUMEN

Avoidance of apoptosis is critical for the development and sustained growth of tumours. The pro-survival protein myeloid cell leukemia 1 (MCL1) is overexpressed in many cancers, but the development of small molecules targeting this protein that are amenable for clinical testing has been challenging. Here we describe S63845, a small molecule that specifically binds with high affinity to the BH3-binding groove of MCL1. Our mechanistic studies demonstrate that S63845 potently kills MCL1-dependent cancer cells, including multiple myeloma, leukaemia and lymphoma cells, by activating the BAX/BAK-dependent mitochondrial apoptotic pathway. In vivo, S63845 shows potent anti-tumour activity with an acceptable safety margin as a single agent in several cancers. Moreover, MCL1 inhibition, either alone or in combination with other anti-cancer drugs, proved effective against several solid cancer-derived cell lines. These results point towards MCL1 as a target for the treatment of a wide range of tumours.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Modelos Biológicos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Tiofenos/farmacología , Tiofenos/uso terapéutico , Animales , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Femenino , Humanos , Leucemia/tratamiento farmacológico , Leucemia/metabolismo , Leucemia/patología , Linfoma/tratamiento farmacológico , Linfoma/metabolismo , Linfoma/patología , Masculino , Ratones , Modelos Moleculares , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/química , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Neoplasias/metabolismo , Pirimidinas/administración & dosificación , Tiofenos/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo
8.
Cancer Cell ; 27(4): 547-60, 2015 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-25873176

RESUMEN

Many types of human cancers having hyperactivated Wnt signaling display no causative alterations in known effectors of this pathway. Here, we report a function of TGIF in Wnt signaling. TGIF associates with and diverts Axin1 and Axin2 from the ß-catenin destruction complex, therefore allowing ß-catenin accrual. Intriguingly, activation of Wnt signaling induces the expression of TGIF, which unveils a feed-forward loop that ensures effective integration of Wnt signaling. In triple-negative breast cancers (TNBC), elevated levels of TGIF correlate with high Wnt signaling and poor survival of patients. Moreover, genetic experiments revealed that Tgif1 ablation impeded mammary tumor development in MMTV-Wnt1 mice, further underscoring a requirement of TGIF for oncogenic Wnt signaling.


Asunto(s)
Proteínas de Homeodominio/fisiología , Neoplasias Mamarias Experimentales/metabolismo , Proteínas Represoras/fisiología , Vía de Señalización Wnt , Transporte Activo de Núcleo Celular , Animales , Proteína Axina/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Modelos Biológicos , Proteínas Represoras/metabolismo , beta Catenina/metabolismo
9.
Cell Rep ; 4(3): 530-41, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23911286

RESUMEN

The homeodomain protein TGIF (TG-interacting factor) restricts TGF-ß/Smad cytostatic signaling by interfering with the nucleocytoplasmic transit of the tumor suppressor cPML. Here, we identify PHRF1 as a ubiquitin ligase that enforces TGIF decay by driving its ubiquitination at lysine 130. In so doing, PHRF1 ensures redistribution of cPML into the cytoplasm, where it associates with SARA and coordinates activation of Smad2 by the TGF-ß receptor. The PHRF1 gene resides within the tumor suppressor locus 11p15.5, which displays frequent loss in a wide variety of malignancies, including breast cancer. Remarkably, we found that the PHRF1 gene is deleted or silenced in a high proportion of human breast cancer samples and cancer cell lines. Reconstitution of PHRF1 into deficient cells impeded their propensity to form tumors in vivo, most likely because of the reemergence of TGF-ß responsiveness. These findings unveil a paradigm behind inactivation of the cPML tumor suppressor network in human malignancies.


Asunto(s)
Neoplasias de la Mama/genética , Factor 7 Regulador del Interferón/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Supresoras de Tumor/genética , Animales , Neoplasias de la Mama/metabolismo , Perros , Femenino , Genes Supresores de Tumor , Células Hep G2 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Factor 7 Regulador del Interferón/metabolismo , Células de Riñón Canino Madin Darby , Proteínas Nucleares/genética , Fosforilación , Proteína de la Leucemia Promielocítica , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Transcripción Genética , Transfección , Factor de Crecimiento Transformador beta/genética , Proteínas Supresoras de Tumor/metabolismo , Ubiquitinación
10.
Chem Biol ; 19(4): 467-77, 2012 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-22520753

RESUMEN

The human USP7 deubiquitinating enzyme was shown to regulate many proteins involved in the cell cycle, as well as tumor suppressors and oncogenes. Thus, USP7 offers a promising, strategic target for cancer therapy. Using biochemical assays and activity-based protein profiling in living systems, we identified small-molecule antagonists of USP7 and demonstrated USP7 inhibitor occupancy and selectivity in cancer cell lines. These compounds bind USP7 in the active site through a covalent mechanism. In cancer cells, these active-site-targeting inhibitors were shown to regulate the level of several USP7 substrates and thus recapitulated the USP7 knockdown phenotype that leads to G1 arrest in colon cancer cells. The data presented in this report provide proof of principle that USP7 inhibitors may be a valuable therapeutic for cancer. In addition, the discovery of such molecules offers interesting tools for studying deubiquitination.


Asunto(s)
Inhibidores Enzimáticos/química , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/farmacología , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Células HCT116 , Humanos , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Ubiquitina Tiolesterasa/metabolismo , Peptidasa Específica de Ubiquitina 7
11.
Mol Microbiol ; 82(1): 68-86, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21801243

RESUMEN

The definition of bacterial cell shape is a complex process requiring the participation of multiple components of an intricate macromolecular machinery. We aimed at characterizing the determinants involved in cell shape of the helical bacterium Helicobacter pylori. Using a yeast two-hybrid screen with the key cell elongation protein PBP2 as bait, we identified an interaction between PBP2 and MreC. The minimal region of MreC required for this interaction ranges from amino acids 116 to 226. Using recombinant proteins, we showed by affinity and size exclusion chromatographies and surface plasmon resonance that PBP2 and MreC form a stable complex. In vivo, the two proteins display a similar spatial localization and their complex has an apparent 1:1 stoichiometry; these results were confirmed in vitro by analytical ultracentrifugation and chemical cross-linking. Small angle X-ray scattering analyses of the PBP2 : MreC complex suggest that MreC interacts directly with the C-terminal region of PBP2. Depletion of either PBP2 or MreC leads to transition into spherical cells that lose viability. Finally, the specific expression in trans of the minimal interacting domain of MreC with PBP2 in the periplasmic space leads to cell rounding, suggesting that the PBP2/MreC complex formation in vivo is essential for cell morphology.


Asunto(s)
Proteínas Bacterianas/metabolismo , Helicobacter pylori/citología , Helicobacter pylori/metabolismo , Proteínas de Unión a las Penicilinas/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Helicobacter pylori/química , Helicobacter pylori/genética , Viabilidad Microbiana , Datos de Secuencia Molecular , Proteínas de Unión a las Penicilinas/química , Proteínas de Unión a las Penicilinas/genética , Unión Proteica , Estructura Terciaria de Proteína , Alineación de Secuencia , Técnicas del Sistema de Dos Híbridos
12.
Future Oncol ; 7(5): 619-32, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21568678

RESUMEN

Ubiquitin-specific proteases are deubiquitinating enzymes involved in the removal of ubiquitin from specific protein substrates resulting in protein salvage from proteasome degradation, regulation of protein localization or activation. DNA alteration and overexpression in different cancer types, as well as involvement in many cancer-associated pathways, make ubiquitin-specific proteases attractive for the cancer drug discovery purposes. Their proteolytic function associated to available structural biology data reinforce their potential for pharmacological interference. Here, we review this class of enzymes as cancer drug targets in terms of validation and druggability.


Asunto(s)
Endopeptidasas/metabolismo , Neoplasias/enzimología , Animales , Antineoplásicos/uso terapéutico , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/tratamiento farmacológico , Proteasas Ubiquitina-Específicas
13.
Proc Natl Acad Sci U S A ; 107(40): 17333-8, 2010 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-20855622

RESUMEN

Listeria monocytogenes is an intracellular pathogen responsible for severe foodborne infections. It can replicate in both phagocytic and nonphagocytic mammalian cells. The infectious process at the cellular level has been studied extensively, but how the bacterium overcomes early host innate immune responses remains largely unknown. Here we show that InlC, a member of the internalin family, is secreted intracellularly and directly interacts with IKKα, a subunit of the IκB kinase complex critical for the phosphorylation of IκB and activation of NF-κB, the major regulator of innate immune responses. Infection experiments with WT Listeria or the inlC-deletion mutant and transfection of cells with InlC reveal that InlC expression impairs phosphorylation and consequently delays IκB degradation normally induced by TNF-α, a classical NF-κB stimulator. Moreover, infection of RAW 264.7 macrophages by the inlC mutant leads to increased production of proinflammatory cytokines compared with that obtained with the WT. Finally, in a peritonitis mouse model, we show that infection with the inlC mutant induces increased production of chemokines and increased recruitment of neutrophils in the peritoneal cavity compared with infection with WT. Together, these results demonstrate that InlC, by interacting with IKKα, dampens the host innate response induced by Listeria during the infection process.


Asunto(s)
Proteínas Bacterianas/inmunología , Quinasa I-kappa B/metabolismo , Inmunidad Innata , Subunidades de Proteína/metabolismo , Animales , Línea Celular , Humanos , Quinasa I-kappa B/genética , Listeria monocytogenes/inmunología , Listeria monocytogenes/patogenicidad , Ratones , Regiones Promotoras Genéticas , Subunidades de Proteína/genética , Factor de Necrosis Tumoral alfa/metabolismo , Técnicas del Sistema de Dos Híbridos
14.
ChemMedChem ; 5(4): 552-8, 2010 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-20186914

RESUMEN

High-throughput screening highlighted 9-oxo-9H-indeno[1,2-b]pyrazine-2,3-dicarbonitrile (1) as an active inhibitor of ubiquitin-specific proteases (USPs), a family of hydrolytic enzymes involved in the removal of ubiquitin from protein substrates. The chemical behavior of compound 1 was examined. Moreover, the synthesis and in vitro evaluation of new compounds, analogues of 1, led to the identification of potent and selective inhibitors of the deubiquitinating enzyme USP8.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/antagonistas & inhibidores , Inhibidores Enzimáticos/síntesis química , Indenos/química , Pirazinas/química , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Cristalografía por Rayos X , Evaluación Preclínica de Medicamentos , Endopeptidasas/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento , Humanos , Indenos/farmacología , Conformación Molecular , Pirazinas/síntesis química , Pirazinas/farmacología , Ubiquitina Tiolesterasa/metabolismo , Peptidasa Específica de Ubiquitina 7
15.
Biochem Soc Trans ; 38(Pt 1): 137-43, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20074048

RESUMEN

Proteases play a key role in various pathological processes and several protease inhibitors are already available for treatment. DUBs (deubiquitinating enzymes) constitute one of the largest classes of human proteases and are key effectors of the ubiquitin-proteasome system. This pathway regulating cellular protein turnover has been implicated in the pathogenesis of many human diseases, including neurodegenerative disorders, viral diseases and cancer. The therapeutic efficacy of the proteasome inhibitor Velcade (bortezomib) for treating multiple myeloma and mantle cell lymphoma establishes this system as a valid target for cancer treatment. A promising alternative to targeting the proteasome itself would be to target the upstream, ubiquitin conjugation/deconjugation system, to generate more specific, less toxic anticancer agents. Advances in small molecule-based inhibitors specifically targeting DUBs are presented in this review.


Asunto(s)
Endopeptidasas/metabolismo , Inhibidores de Proteasas/uso terapéutico , Ubiquitina/metabolismo , Animales , Ácidos Borónicos/uso terapéutico , Bortezomib , Endopeptidasas/química , Humanos , Estructura Molecular , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Inhibidores de Proteasas/química , Inhibidores de Proteasas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Conformación Proteica , Pirazinas/uso terapéutico , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/metabolismo , Peptidasa Específica de Ubiquitina 7 , Ubiquitinación , Virus/metabolismo , Virus/patogenicidad
16.
Exp Cell Res ; 316(4): 667-75, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19909739

RESUMEN

The SYK non-receptor tyrosine kinase is a key effector of immune receptors signaling in hematopoietic cells. Here, we identified and characterized a novel interaction between SYK and the ubiquitin-specific protease 25 (USP25). We report that the second SH2 domain of SYK physically interacts with a tyrosine-rich, C-terminal region of USP25 independently of tyrosine phosphorylation. Moreover, we showed that SYK specifically phosphorylates USP25 and alters its cellular levels. This study thus uncovers a new SYK substrate and reveals a novel SYK function, namely the regulation of USP25 cellular levels.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Mapeo Cromosómico , Vectores Genéticos/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Mutación/genética , Fosforilación , Plásmidos/genética , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/genética , Quinasa Syk , Técnicas del Sistema de Dos Híbridos , Ubiquitina Tiolesterasa/genética
17.
Mol Cancer Ther ; 8(8): 2286-95, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19671755

RESUMEN

Deregulation of the ubiquitin/proteasome system has been implicated in the pathogenesis of many human diseases, including cancer. Ubiquitin-specific proteases (USP) are cysteine proteases involved in the deubiquitination of protein substrates. Functional connections between USP7 and essential viral proteins and oncogenic pathways, such as the p53/Mdm2 and phosphatidylinositol 3-kinase/protein kinase B networks, strongly suggest that the targeting of USP7 with small-molecule inhibitors may be useful for the treatment of cancers and viral diseases. Using high-throughput screening, we have discovered HBX 41,108, a small-molecule compound that inhibits USP7 deubiquitinating activity with an IC(50) in the submicromolar range. Kinetics data indicate an uncompetitive reversible inhibition mechanism. HBX 41,108 was shown to affect USP7-mediated p53 deubiquitination in vitro and in cells. As RNA interference-mediated USP7 silencing in cancer cells, HBX 41,108 treatment stabilized p53, activated the transcription of a p53 target gene without inducing genotoxic stress, and inhibited cancer cell growth. Finally, HBX 41,108 induced p53-dependent apoptosis as shown in p53 wild-type and null isogenic cancer cell lines. We thus report the identification of the first lead-like inhibitor against USP7, providing a structural basis for the development of new anticancer drugs.


Asunto(s)
Indenos/farmacología , Inhibidores de Proteasas/farmacología , Pirazinas/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Proteína p53 Supresora de Tumor/genética , Ubiquitina Tiolesterasa/metabolismo , Peptidasa Específica de Ubiquitina 7
18.
Dev Cell ; 16(4): 551-62, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19386264

RESUMEN

The human Lkb1 kinase, encoded by the ortholog of the invertebrate Par4 polarity gene, is mutated in Peutz-Jeghers cancer syndrome. Lkb1 activity requires complex formation with the pseudokinase Strad and the adaptor protein Mo25. The complex can induce complete polarization in a single isolated intestinal epithelial cell. We describe an interaction between Mo25alpha and a human serine/threonine kinase termed Mst4. A homologous interaction occurs in the yeast Schizosaccharomyces pombe in the control of polar tip growth. Human Mst4 translocates from the Golgi to the subapical membrane compartment upon activation of Lkb1. Inhibition of Mst4 activity inhibits Lkb1-induced brush border formation, whereas other aspects of polarity such as the formation of lateral junctions remain unaffected. As an essential event in brush border formation, Mst4 phosphorylates the regulatory T567 residue of Ezrin. These data define a brush border induction pathway downstream of the Lkb1/Strad/Mo25 polarization complex, yet separate from other polarity events.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Proteínas de Unión al Calcio/metabolismo , Polaridad Celular , Proteínas del Citoesqueleto/metabolismo , Microvellosidades/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Humanos , Microvellosidades/ultraestructura , Modelos Biológicos , Fosfoproteínas/metabolismo , Fosforilación , Unión Proteica , Transporte de Proteínas
19.
EMBO J ; 27(13): 1804-15, 2008 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-18511908

RESUMEN

The TGIF homoeodomain protein functions as an important negative regulator in the TGF-beta signalling pathway. The inhibitory function of TGIF is executed in part through its ability to sequester the tumour suppressor cytoplasmic promyelocytic leukaemia (cPML) in the nucleus, thereby preventing the phosphorylation of Smad2 by the activated TGF-beta type I receptor. Here, we report on the identification of PCTA (PML competitor for TGIF association), a TGIF antagonist that promotes TGF-beta-induced transcriptional and cytostatic responses. We provide evidence that PCTA functions in TGF-beta signalling by relieving the suppression of Smad2 phosphorylation by TGIF. Furthermore, we demonstrate that PCTA selectively competes with cPML for TGIF association, resulting in the accumulation of cPML in the cytoplasm, where it associates with SARA and coordinates the access of Smad2 for phosphorylation by the activated TGF-beta type I receptor. Thus, our findings on the mode of action of PCTA provide new and important insights into the molecular mechanism underlying the antagonistic interplay between TGIF and cPML in the TGF-beta signalling network.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteína Smad2/metabolismo , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Línea Celular , ADN Complementario , Perros , Femenino , Biblioteca de Genes , Humanos , Fosforilación , Placenta/metabolismo , Proteína de la Leucemia Promielocítica , Técnicas del Sistema de Dos Híbridos , Ubiquitina-Proteína Ligasas
20.
Appl Environ Microbiol ; 74(7): 2095-102, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18245237

RESUMEN

The Escherichia coli-Helicobacter pylori shuttle vector pHeL2 was modified to introduce the inducible LacI(q)-pTac system of E. coli, in which the promoters were engineered to be under the control of H. pylori RNA polymerase. The amiE gene promoter of H. pylori was taken to constitutively express the LacI(q) repressor. Expression of the reporter gene lacZ was driven by either pTac (pILL2150) or a modified version of the ureI gene promoter in which one or two LacI-binding sites and/or mutated nucleotides between the ribosomal binding site and the ATG start codon (pILL2153 and pILL2157) were introduced. Promoter activity was evaluated by measuring beta-galactosidase activity. pILL2150 is a tightly regulated expression system suitable for the analysis of genes with low-level expression, while pILL2157 is well adapted for the controlled expression of genes encoding recombinant proteins in H. pylori. To exemplify the usefulness of these tools, we constructed conditional mutants of the putative essential pbp1 and ftsI genes encoding penicillin-binding proteins 1 and 3 of H. pylori, respectively. Both genes were cloned into pILL2150 and introduced in the parental H. pylori strain N6. The chromosomally harbored pbp1 and ftsI genes were then inactivated by replacing them with a nonpolar kanamycin cassette. Inactivation was strictly dependent upon addition of isopropyl-beta-d-thiogalactopyranoside. Hence, we were able to construct the first conditional mutants of H. pylori. Finally, we demonstrated that following in vitro methylation of the recombinant plasmids, these could be introduced into a large variety of H. pylori isolates with different genetic backgrounds.


Asunto(s)
Genes Esenciales , Ingeniería Genética , Vectores Genéticos , Helicobacter pylori/genética , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Helicobacter pylori/fisiología , Datos de Secuencia Molecular , Mutagénesis , Regiones Promotoras Genéticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA