Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
BMC Biol ; 16(1): 29, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29510700

RESUMEN

BACKGROUND: Cell migration is essential for development and tissue repair, but it also contributes to disease. Rho GTPases regulate cell migration, but a comprehensive analysis of how each Rho signalling component affects migration has not been carried out. RESULTS: Through an RNA interference screen, and using a prostate cancer cell line, we find that approximately 25% of Rho network components alter migration. Some genes enhance migration while others decrease basal and/or hepatocyte growth factor-stimulated migration. Surprisingly, we identify RhoH as a screen hit. RhoH expression is normally restricted to haematopoietic cells, but we find it is expressed in multiple epithelial cancer cell lines. High RhoH expression in samples from prostate cancer patients correlates with earlier relapse. RhoH depletion reduces cell speed and persistence and decreases migratory polarity. Rac1 activity normally localizes to the front of migrating cells at areas of dynamic membrane movement, but in RhoH-depleted cells active Rac1 is localised around the whole cell periphery and associated with membrane regions that are not extending or retracting. RhoH interacts with Rac1 and with several p21-activated kinases (PAKs), which are Rac effectors. Similar to RhoH depletion, PAK2 depletion increases cell spread area and reduces cell migration. In addition, RhoH depletion reduces lamellipodium extension induced by PAK2 overexpression. CONCLUSIONS: We describe a novel role for RhoH in prostate cancer cell migration. We propose that RhoH promotes cell migration by coupling Rac1 activity and PAK2 to membrane protrusion. Our results also suggest that RhoH expression levels correlate with prostate cancer progression.


Asunto(s)
Movimiento Celular/genética , Pruebas Genéticas/métodos , Neoplasias de la Próstata/genética , Interferencia de ARN/fisiología , Factores de Transcripción/genética , Proteína de Unión al GTP rac1/genética , Proteínas de Unión al GTP rho/genética , Animales , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/genética , Células COS , Chlorocebus aethiops , Detección Precoz del Cáncer/métodos , Células HT29 , Humanos , Células MCF-7 , Masculino , Neoplasias de la Próstata/diagnóstico , Factores de Transcripción/análisis , Proteína de Unión al GTP rac1/análisis , Proteínas de Unión al GTP rho/análisis
2.
J Cell Sci ; 125(Pt 24): 6020-9, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23038771

RESUMEN

Phagocytosis is the force-dependent complex cellular process by which immune cells engulf particles. Although there has been considerable progress in understanding ligand-receptor-induced actin polymerisation in pushing the membrane around the particle, significantly less is known about how localised contractile activities regulate cup closure in coordination with the actin cytoskeleton. Herein, we show that the unconventional class-I myosin, myosin 1G (Myo1G) is localised at phagocytic cups following Fcγ-receptor (FcγR) ligation in macrophages. This progressive recruitment is dependent on the activity of phosphoinositide 3-kinase and is particularly important for engulfment of large particles. Furthermore, point mutations in the conserved pleckstrin homology-like domain of Myo1G abolishes the localisation of the motor protein at phagocytic cups and inhibits engulfment downstream of FcγR. Binding of Myo1G to both F-actin and phospholipids might enable cells to transport phospholipids towards the leading edge of cups and to facilitate localised contraction for cup closure.


Asunto(s)
Miosina Tipo I/metabolismo , Fagocitosis/fisiología , Receptores de IgG/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animales , Células COS , Chlorocebus aethiops , Fagocitosis/genética
3.
J Med Chem ; 66(8): 5892-5906, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-37026591

RESUMEN

B-cell lymphoma 6 (BCL6) is a transcriptional repressor and oncogenic driver of diffuse large B-cell lymphoma (DLBCL). Here, we report the optimization of our previously reported tricyclic quinolinone series for the inhibition of BCL6. We sought to improve the cellular potency and in vivo exposure of the non-degrading isomer, CCT373567, of our recently published degrader, CCT373566. The major limitation of our inhibitors was their high topological polar surface areas (TPSA), leading to increased efflux ratios. Reducing the molecular weight allowed us to remove polarity and decrease TPSA without considerably reducing solubility. Careful optimization of these properties, as guided by pharmacokinetic studies, led to the discovery of CCT374705, a potent inhibitor of BCL6 with a good in vivo profile. Modest in vivo efficacy was achieved in a lymphoma xenograft mouse model after oral dosing.


Asunto(s)
Linfoma de Células B Grandes Difuso , Quinolonas , Animales , Humanos , Ratones , Línea Celular Tumoral , Modelos Animales de Enfermedad , Linfoma de Células B Grandes Difuso/patología , Proteínas Proto-Oncogénicas c-bcl-6/química , Factores de Transcripción
4.
Mol Microbiol ; 80(6): 1420-38, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21488979

RESUMEN

The human pathogens enteropathogenic and enterohemorrhagic Escherichia coli (EPEC and EHEC) share a unique mechanism of colonization that results from the concerted action of effector proteins translocated into the host cell by a type III secretion system (T3SS). EPEC and EHEC not only induce characteristic attaching and effacing (A/E) lesions, but also subvert multiple host cell signalling pathways during infection. Our understanding of the mechanisms by which A/E pathogens hijack host cell signalling has advanced dramatically in recent months with the identification of novel activities for many effectors. In addition to further characterization of established effectors (Tir, EspH and Map), new effectors have emerged as important mediators of virulence through activities such as mimicry of Rho guanine nucleotide exchange factors (Map and EspM), inhibition of apoptosis (NleH and NleD), interference with inflammatory signalling pathways (NleB, NleC, NleE and NleH) and phagocytosis (EspF, EspH and EspJ). The findings have highlighted the multifunctional nature of the effectors and their ability to participate in redundant, synergistic or antagonistic relationships, acting in a co-ordinated spatial and temporal manner on different host organelles and cellular pathways during infection.


Asunto(s)
Escherichia coli Enterohemorrágica/metabolismo , Escherichia coli Enteropatógena/metabolismo , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/metabolismo , Animales , Escherichia coli Enterohemorrágica/genética , Escherichia coli Enterohemorrágica/patogenicidad , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/patogenicidad , Infecciones por Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Humanos , Transducción de Señal
5.
J Med Chem ; 65(12): 8191-8207, 2022 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-35653645

RESUMEN

The transcriptional repressor BCL6 is an oncogenic driver found to be deregulated in lymphoid malignancies. Herein, we report the optimization of our previously reported benzimidazolone molecular glue-type degrader CCT369260 to CCT373566, a highly potent probe suitable for sustained depletion of BCL6 in vivo. We observed a sharp degradation SAR, where subtle structural changes conveyed the ability to induce degradation of BCL6. CCT373566 showed modest in vivo efficacy in a lymphoma xenograft mouse model following oral dosing.


Asunto(s)
Carcinogénesis , Regulación Neoplásica de la Expresión Génica , Animales , Humanos , Ratones , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo
6.
J Med Chem ; 65(12): 8169-8190, 2022 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-35657291

RESUMEN

To identify new chemical series with enhanced binding affinity to the BTB domain of B-cell lymphoma 6 protein, we targeted a subpocket adjacent to Val18. With no opportunities for strong polar interactions, we focused on attaining close shape complementarity by ring fusion onto our quinolinone lead series. Following exploration of different sized rings, we identified a conformationally restricted core which optimally filled the available space, leading to potent BCL6 inhibitors. Through X-ray structure-guided design, combined with efficient synthetic chemistry to make the resulting novel core structures, a >300-fold improvement in activity was obtained by the addition of seven heavy atoms.


Asunto(s)
Dominio BTB-POZ , Unión Proteica , Proteínas Proto-Oncogénicas c-bcl-6
7.
J Med Chem ; 64(23): 17079-17097, 2021 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-34846884

RESUMEN

We describe the optimization of modestly active starting points to potent inhibitors of BCL6 by growing into a subpocket, which was occupied by a network of five stably bound water molecules. Identifying potent inhibitors required not only forming new interactions in the subpocket but also perturbing the water network in a productive, potency-increasing fashion while controlling the physicochemical properties. We achieved this goal in a sequential manner by systematically probing the pocket and the water network, ultimately achieving a 100-fold improvement of activity. The most potent compounds displaced three of the five initial water molecules and formed hydrogen bonds with the remaining two. Compound 25 showed a promising profile for a lead compound with submicromolar inhibition of BCL6 in cells and satisfactory pharmacokinetic (PK) properties. Our work highlights the importance of finding productive ways to perturb existing water networks when growing into solvent-filled protein pockets.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Proto-Oncogénicas c-bcl-6/antagonistas & inhibidores , Antineoplásicos/química , Cristalografía por Rayos X , Diseño de Fármacos , Humanos , Enlace de Hidrógeno , Solubilidad , Relación Estructura-Actividad
8.
J Med Chem ; 63(8): 4047-4068, 2020 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-32275432

RESUMEN

Deregulation of the transcriptional repressor BCL6 enables tumorigenesis of germinal center B-cells, and hence BCL6 has been proposed as a therapeutic target for the treatment of diffuse large B-cell lymphoma (DLBCL). Herein we report the discovery of a series of benzimidazolone inhibitors of the protein-protein interaction between BCL6 and its co-repressors. A subset of these inhibitors were found to cause rapid degradation of BCL6, and optimization of pharmacokinetic properties led to the discovery of 5-((5-chloro-2-((3R,5S)-4,4-difluoro-3,5-dimethylpiperidin-1-yl)pyrimidin-4-yl)amino)-3-(3-hydroxy-3-methylbutyl)-1-methyl-1,3-dihydro-2H-benzo[d]imidazol-2-one (CCT369260), which reduces BCL6 levels in a lymphoma xenograft mouse model following oral dosing.


Asunto(s)
Bencimidazoles/administración & dosificación , Bencimidazoles/química , Sistemas de Liberación de Medicamentos/métodos , Descubrimiento de Drogas/métodos , Proteínas Proto-Oncogénicas c-bcl-6/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Animales , Línea Celular Tumoral , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
9.
Cell Signal ; 44: 127-137, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29329780

RESUMEN

Cancer cells are able to survive under conditions that cause endoplasmic reticulum stress (ER-stress), and can adapt to this stress by upregulating cell-survival signalling pathways and down-regulating apoptotic pathways. The cellular response to ER-stress is controlled by the unfolded protein response (UPR). Small Rho family GTPases are linked to many cell responses including cell growth and apoptosis. In this study, we investigate the function of small GTPases in cell survival under ER-stress. Using siRNA screening we identify that RAC1 promotes cell survival under ER-stress in cells with an oncogenic N92I RAC1 mutation. We uncover a novel connection between the UPR and N92I RAC1, whereby RAC1 attenuates phosphorylation of EIF2S1 under ER-stress and drives over-expression of ATF4 in basal conditions. Interestingly, the UPR connection does not drive resistance to ER-stress, as knockdown of ATF4 did not affect this. We further investigate cancer-associated kinase signalling pathways and show that RAC1 knockdown reduces the activity of AKT and ERK, and using a panel of clinically important kinase inhibitors, we uncover a role for MEK/ERK, but not AKT, in cell viability under ER-stress. A known major activator of ERK phosphorylation in cancer is oncogenic NRAS and we show that knockdown of NRAS in cells, which bear a Q61 NRAS mutation, sensitises to ER-stress. These findings highlight a novel mechanism for resistance to ER-stress through oncogenic activation of MEK/ERK signalling by small GTPases.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Estrés del Retículo Endoplásmico , GTP Fosfohidrolasas/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Línea Celular Tumoral , Humanos , Transducción de Señal , Respuesta de Proteína Desplegada
10.
Clin Cancer Res ; 24(10): 2395-2407, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29391353

RESUMEN

Purpose: Myeloma is a plasma cell malignancy characterized by the overproduction of immunoglobulin, and is therefore susceptible to therapies targeting protein homeostasis. We hypothesized that heat shock factor 1 (HSF1) was an attractive therapeutic target for myeloma due to its direct regulation of transcriptional programs implicated in both protein homeostasis and the oncogenic phenotype. Here, we interrogate HSF1 as a therapeutic target in myeloma using bioinformatic, genetic, and pharmacologic means.Experimental Design: To assess the clinical relevance of HSF1, we analyzed publicly available patient myeloma gene expression datasets. Validation of this novel target was conducted in in vitro experiments using shRNA or inhibitors of the HSF1 pathway in human myeloma cell lines and primary cells as well as in in vivo human myeloma xenograft models.Results: Expression of HSF1 and its target genes were associated with poorer myeloma patient survival. ShRNA-mediated knockdown or pharmacologic inhibition of the HSF1 pathway with a novel chemical probe, CCT251236, or with KRIBB11, led to caspase-mediated cell death that was associated with an increase in EIF2α phosphorylation, CHOP expression and a decrease in overall protein synthesis. Importantly, both CCT251236 and KRIBB11 induced cytotoxicity in human myeloma cell lines and patient-derived primary myeloma cells with a therapeutic window over normal cells. Pharmacologic inhibition induced tumor growth inhibition and was well-tolerated in a human myeloma xenograft murine model with evidence of pharmacodynamic biomarker modulation.Conclusions: Taken together, our studies demonstrate the dependence of myeloma cells on HSF1 for survival and support the clinical evaluation of pharmacologic inhibitors of the HSF1 pathway in myeloma. Clin Cancer Res; 24(10); 2395-407. ©2018 AACRSee related commentary by Parekh, p. 2237.


Asunto(s)
Biomarcadores de Tumor , Supervivencia Celular/genética , Factores de Transcripción del Choque Térmico/genética , Mieloma Múltiple/genética , Animales , Antineoplásicos/farmacología , Apoptosis/genética , Caspasas/metabolismo , Línea Celular Tumoral , Biología Computacional/métodos , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Factores de Transcripción del Choque Térmico/antagonistas & inhibidores , Factores de Transcripción del Choque Térmico/metabolismo , Humanos , Estimación de Kaplan-Meier , Ratones , Terapia Molecular Dirigida , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Mieloma Múltiple/mortalidad , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda