Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
FEBS J ; 2023 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-38088212

RESUMEN

The B-cell lymphoma 2 (BCL2) family members, BCL2-associated protein X (BAX) and BCL2 homologous antagonist killer (BAK), are required for programmed cell death via the mitochondrial pathway. When cells are stressed, damaged or redundant, the balance of power between the BCL2 family of proteins shifts towards BAX and BAK, allowing their transition from an inactive, monomeric state to a membrane-active oligomeric form that releases cytochrome c from the mitochondrial intermembrane space. That oligomeric state has an essential intermediate, a symmetric homodimer of BAX or BAK. Here we describe crystal structures of dimers of the core domain of BAX, comprising its helices α2-α5. These structures provide an atomic resolution description of the interactions that drive BAX homo-dimerisation and insights into potential interaction between core domain dimers and membrane lipids. The previously identified BAK lipid-interacting sites are not conserved with BAX and are likely to determine the differences between them in their interactions with lipids. We also describe structures of heterodimers of BAK/BAX core domains, yielding further insight into the differences in lipid binding between BAX and BAK.

2.
Nat Commun ; 14(1): 3542, 2023 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-37336884

RESUMEN

PEAK pseudokinases regulate cell migration, invasion and proliferation by recruiting key signaling proteins to the cytoskeleton. Despite lacking catalytic activity, alteration in their expression level is associated with several aggressive cancers. Here, we elucidate the molecular details of key PEAK signaling interactions with the adapter proteins CrkII and Grb2 and the scaffold protein 14-3-3. Our findings rationalize why the dimerization of PEAK proteins has a crucial function in signal transduction and provide biophysical and structural data to unravel binding specificity within the PEAK interactome. We identify a conserved high affinity 14-3-3 motif on PEAK3 and demonstrate its role as a molecular switch to regulate CrkII binding and signaling via Grb2. Together, our studies provide a detailed structural snapshot of PEAK interaction networks and further elucidate how PEAK proteins, especially PEAK3, act as dynamic scaffolds that exploit adapter proteins to control signal transduction in cell growth/motility and cancer.


Asunto(s)
Proteínas 14-3-3 , Proteínas del Citoesqueleto , Transducción de Señal , Movimiento Celular , Proliferación Celular , Transducción de Señal/fisiología , Proteínas del Citoesqueleto/metabolismo , Proteínas 14-3-3/metabolismo
3.
Nat Commun ; 13(1): 5969, 2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-36216795

RESUMEN

Targeted protein degradation offers an alternative modality to classical inhibition and holds the promise of addressing previously undruggable targets to provide novel therapeutic options for patients. Heterobifunctional molecules co-recruit a target protein and an E3 ligase, resulting in ubiquitylation and proteosome-dependent degradation of the target. In the clinic, the oral route of administration is the option of choice but has only been achieved so far by CRBN- recruiting bifunctional degrader molecules. We aimed to achieve orally bioavailable molecules that selectively degrade the BAF Chromatin Remodelling complex ATPase SMARCA2 over its closely related paralogue SMARCA4, to allow in vivo evaluation of the synthetic lethality concept of SMARCA2 dependency in SMARCA4-deficient cancers. Here we outline structure- and property-guided approaches that led to orally bioavailable VHL-recruiting degraders. Our tool compound, ACBI2, shows selective degradation of SMARCA2 over SMARCA4 in ex vivo human whole blood assays and in vivo efficacy in SMARCA4-deficient cancer models. This study demonstrates the feasibility for broadening the E3 ligase and physicochemical space that can be utilised for achieving oral efficacy with bifunctional molecules.


Asunto(s)
Adenosina Trifosfatasas , Factores de Transcripción , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , ADN Helicasas/genética , ADN Helicasas/metabolismo , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteolisis , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
4.
Methods Enzymol ; 667: 1-35, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35525538

RESUMEN

The PEAK family of pseudokinases, which comprises PEAK1, PEAK2 and PEAK3, are newly identified scaffolds that dynamically assemble oncogenic signaling pathways known to contribute to the development of several aggressive cancers. A striking feature of this unique family of pseudokinase scaffolds is their large multi-domain structure, which allows them to achieve protein complex assemblies through their structural plasticity and functional versatility. Recent structural advances have begun to reveal the critical regulatory elements that control their function. Specifically, the dimer-dependent scaffolding activity of PEAK pseudokinases is emerging as a critical mechanism for their signaling function, in addition to their ability to hetero-associate to form higher-order regulatory networks to diversify and amplify their signaling output. Here, we present a suite of techniques that enable the efficient expression and purification of PEAK proteins for functional characterization.


Asunto(s)
Neoplasias , Transducción de Señal , Carcinogénesis , Humanos
5.
Sci Signal ; 15(722): eabj3554, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35192416

RESUMEN

The pseudokinase scaffolds PEAK1 and PEAK2 are implicated in cancer cell migration and metastasis. We characterized the regulation and role of the third family member PEAK3 in cell signaling. Similar to PEAK1 and PEAK2, PEAK3 formed both homotypic and heterotypic complexes. In addition, like PEAK1, it bound to the adaptors Grb2 and CrkII. However, unlike PEAK1 and PEAK2, homodimerized PEAK3 also interacted with the ARF GTPase-activating protein ASAP1, the E3 ubiquitin ligase Cbl, and the kinase PYK2. Dimerization and subsequent phosphorylation on Tyr24, likely by a Src family kinase, were required for the binding of PEAK3 to Grb2 and ASAP1. Interactions with Grb2, CrkII, ASAP1, Cbl, and PYK2 exhibited contrasting dynamics upon cell stimulation with epidermal growth factor (EGF), in part due to PEAK3 dephosphorylation mediated by the phosphatase PTPN12. Overexpressing PEAK3 in mesenchymal-like MDA-MB-231 breast cancer cells enhanced cell elongation in a manner dependent on PEAK3 dimerization, and manipulation of PEAK3 expression demonstrated a positive role for this scaffold in regulating cell migration. Overexpressing PEAK3 in PEAK1/2 double-knockout MCF-10A breast epithelial cells enhanced acinar growth, impaired basement membrane integrity, and promoted invasion in three-dimensional cultures, with the latter two effects dependent on the binding of PEAK3 to Grb2 and ASAP1. PEAK1 and PEAK2 quantitatively and temporally influenced PEAK3 function. These findings characterize PEAK3 as an integral, signal-diversifying member of the PEAK family with scaffolding roles that promote cell proliferation, migration, and invasion.


Asunto(s)
Proteínas Tirosina Quinasas , Transducción de Señal , Movimiento Celular , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Familia-src Quinasas/metabolismo
6.
Commun Biol ; 4(1): 1105, 2021 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-34545159

RESUMEN

Doublecortin-like kinase 1 (DCLK1) is an understudied bi-functional kinase with a proven role in tumour growth and development. However, the presence of tissue-specific spliced DCLK1 isoforms with distinct biological functions have challenged the development of effective strategies to understand the role of DCLK1 in oncogenesis. Recently, DCLK1-IN-1 was reported as a highly selective DCLK1 inhibitor, a powerful tool to dissect DCLK1 biological functions. Here, we report the crystal structures of DCLK1 kinase domain in complex with DCLK1-IN-1 and its precursors. Combined, our data rationalises the structure-activity relationship that informed the development of DCLK1-IN-1 and provides the basis for the high selectivity of DCLK1-IN-1, with DCLK1-IN-1 inducing a drastic conformational change of the ATP binding site. We demonstrate that DCLK1-IN-1 binds DCLK1 long isoforms but does not prevent DCLK1's Microtubule-Associated Protein (MAP) function. Together, our work provides an invaluable structural platform to further the design of isoform-specific DCLK1 modulators for therapeutic intervention.


Asunto(s)
Quinasas Similares a Doblecortina/antagonistas & inhibidores , Quinasas Similares a Doblecortina/genética , Sitios de Unión , Quinasas Similares a Doblecortina/metabolismo , Dominios Proteicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Relación Estructura-Actividad
7.
J Med Chem ; 64(9): 5447-5469, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33904752

RESUMEN

The BCL-2 family of proteins (including the prosurvival proteins BCL-2, BCL-XL, and MCL-1) is an important target for the development of novel anticancer therapeutics. Despite the challenges of targeting protein-protein interaction (PPI) interfaces with small molecules, a number of inhibitors (called BH3 mimetics) have entered the clinic and the BCL-2 inhibitor, ABT-199/venetoclax, is already proving transformative. For BCL-XL, new validated chemical series are desirable. Here, we outline the crystallography-guided development of a structurally distinct series of BCL-XL/BCL-2 inhibitors based on a benzoylurea scaffold, originally proposed as α-helix mimetics. We describe structure-guided exploration of a cryptic "p5" pocket identified in BCL-XL. This work yields novel inhibitors with submicromolar binding, with marked selectivity toward BCL-XL. Extension into the hydrophobic p2 pocket yielded the most potent inhibitor in the series, binding strongly to BCL-XL and BCL-2 (nanomolar-range half-maximal inhibitory concentration (IC50)) and displaying mechanism-based killing in cells engineered to depend on BCL-XL for survival.


Asunto(s)
Antineoplásicos/química , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Urea/análogos & derivados , Proteína bcl-X/antagonistas & inhibidores , Animales , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Sitios de Unión , Compuestos de Bifenilo/química , Compuestos de Bifenilo/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cristalografía por Rayos X , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Concentración 50 Inhibidora , Ratones , Simulación de Dinámica Molecular , Nitrofenoles/química , Nitrofenoles/metabolismo , Piperazinas/química , Piperazinas/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Relación Estructura-Actividad , Sulfonamidas/química , Sulfonamidas/metabolismo , Resonancia por Plasmón de Superficie , Urea/metabolismo , Urea/farmacología , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
8.
FEBS J ; 287(19): 4183-4197, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31599110

RESUMEN

The study of pseudokinases has uncovered that catalysis-independent functions play a critical role in cell signalling regulation. However, how pseudokinases dynamically assemble and regulate oncogenic signalling pathways remains, in most cases, unclear due to a limited knowledge of the structural determinants that are critical for their functions. Here, we review the recent progress made to unravel the role of the PEAK family of pseudokinases, which comprises SgK269, SgK223 and the recently identified PEAK3, in assembling specific oncogenic signalling pathways that contribute to the progression of several aggressive cancers. We focus on recent structural advances revealing that SgK269 and SgK223 can homo- and heteroassociate via a unique dimerisation domain, comprising conserved regulatory helices directly surrounding the pseudokinase domain, which is also conserved in PEAK3. We also highlight a potential oligomerisation mechanism driven by the pseudokinase domain. While it is likely that homo- or heterodimerisation and oligomerisation mechanisms contribute to the assembly of complex signalling hubs and provide a means to spatially and temporally modulate and diversify signalling outputs, the exact role that these oncogenic scaffolds play in regulating cell migration, invasion and morphology remains unclear. Here, we attempt to link their structural characteristics to their cellular functions by providing a thorough analysis of the signalling transduction pathways they are known to modulate.


Asunto(s)
Neoplasias/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal , Humanos , Neoplasias/patología
9.
Nat Chem Biol ; 15(8): 846, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31267096

RESUMEN

In the version of this article originally published, several lines of text in the last paragraph of the right column on page 1 of the PDF were transposed into the bottom paragraph of the left column. The affected text of the left column should read "The ATP-dependent activities of the BAF (SWI/SNF) chromatin remodeling complexes affect the positioning of nucleosomes on DNA and thereby many cellular processes related to chromatin structure, including transcription, DNA repair and decatenation of chromosomes during mitosis12,13." The affected text of the right column should read "SMARCA2/4BD inhibitors are thus precluded from use for the treatment of SMARCA4 mutant cancers but could provide attractive ligands for PROTAC conjugation. Small molecules binding to other bromodomains have been successfully converted into PROTACs by conjugating them with structures capable of binding to the E3 ligases von Hippel-Lindau (VHL) or cereblon5,6,10,11,25,26,27." The errors have been corrected in the PDF version of the paper.

10.
Nat Chem Biol ; 15(7): 672-680, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31178587

RESUMEN

Targeting subunits of BAF/PBAF chromatin remodeling complexes has been proposed as an approach to exploit cancer vulnerabilities. Here, we develop proteolysis targeting chimera (PROTAC) degraders of the BAF ATPase subunits SMARCA2 and SMARCA4 using a bromodomain ligand and recruitment of the E3 ubiquitin ligase VHL. High-resolution ternary complex crystal structures and biophysical investigation guided rational and efficient optimization toward ACBI1, a potent and cooperative degrader of SMARCA2, SMARCA4 and PBRM1. ACBI1 induced anti-proliferative effects and cell death caused by SMARCA2 depletion in SMARCA4 mutant cancer cells, and in acute myeloid leukemia cells dependent on SMARCA4 ATPase activity. These findings exemplify a successful biophysics- and structure-based PROTAC design approach to degrade high profile drug targets, and pave the way toward new therapeutics for the treatment of tumors sensitive to the loss of BAF complex ATPases.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Proteínas de Unión al ADN/genética , Leucemia Mieloide Aguda/genética , Proteínas Nucleares/genética , Proliferación Celular , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Humanos , Leucemia Mieloide Aguda/metabolismo , Estructura Molecular , Proteínas Nucleares/metabolismo
11.
J Med Chem ; 57(4): 1323-43, 2014 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-24456288

RESUMEN

The prosurvival BCL-2 proteins are attractive yet challenging targets for medicinal chemists. Their involvement in the initiation and progression of many, if not all, tumors makes them prime targets for developing new anticancer therapies. We present our approach based on de novo structure-based drug design. Using known structural information from complexes engaging opposing members of the BCL-2 family of proteins, we designed peptidomimetic compounds using a benzoylurea scaffold to reproduce key interactions between these proteins. A library stemming from the initial de novo designed scaffold led to the discovery of ligands with low micromolar potency (KD = 4 µM) and selectivity for BCL-XL. These compounds bind in the canonical BH3 binding groove in a binding mode distinct from previously known BCL-2 inhibitors. The results of our study provide insight into the design of a new class of antagonists targeting a challenging class of protein-protein interactions.


Asunto(s)
Urea/análogos & derivados , Proteína bcl-X/antagonistas & inhibidores , Cristalografía por Rayos X , Diseño de Fármacos , Espectroscopía de Resonancia Magnética , Imitación Molecular , Estructura Molecular , Espectrometría de Masa por Ionización de Electrospray , Relación Estructura-Actividad , Urea/síntesis química , Urea/farmacología , Proteína bcl-X/química
12.
J Anal Toxicol ; 30(1): 21-6, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16620527

RESUMEN

A rapid and highly sensitive gas chromatography-mass spectrometry (GC-MS) method for simultaneous determination of N,N-diethyl-m-toluamide (DEET) and permethrin with (2)H(10)-phenanthrene (98 atom %) as an internal standard and a separate external standard high-performance liquid chromatography (HPLC) method for pyridostigmine bromide (PB) determination in human plasma were developed and validated. The GC-MS method for DEET and permethrin quantification utilizes a one-step extraction with tert-butylmethylether. The HPLC method for PB quantification involves a solid-phase extraction and UV detection. The range of the analytical method for DEET and permethrin was 1 ng/mL to 100 ng/mL and for PB was 5 ng/mL to 100 ng/mL. Recovery from plasma proved to be more than 80%. The intraday precision ranged from 1.3% to 8% for DEET, from 2.1% to 11.4% for permethrin, and from 3.0% to 4.8% for PB. The interday precision was 3% for DEET, ranged from 5% to 9% for permethrin, and from 5% to 9% for PB. The accuracy for the limit of quantification was 92% +/- 8% relative standard deviation (RSD) for DEET, 112% +/- 11% RSD for permethrin, and 109% +/- 5% RSD for PB. All 3 compounds were stable in human plasma at -80 degrees C for at least 12 months and after 2 freeze-thaw cycles with RSD values ranging from 7.1% (DEET, 80 ng/mL) to 8.1% (DEET, 8 ng/mL), from 2.3% (permethrin, 80 ng/mL) to 11.6 % (permethrin, 8 ng/mL), and from 0.2% (PB, 80 ng/mL) to 3.6% (PB, 8 ng/mL). Both methods were successfully applied to pharmacokinetic/ pharmacodynamic studies of combined exposure of DEET (skin application), permethrin (treated uniforms), and PB (30 mg orally three times/day for four doses) in healthy volunteers (n = 81).


Asunto(s)
Cromatografía Líquida de Alta Presión , DEET/sangre , Cromatografía de Gases y Espectrometría de Masas/métodos , Repelentes de Insectos/sangre , Insecticidas/sangre , Permetrina/sangre , Bromuro de Piridostigmina/sangre , DEET/farmacocinética , Estabilidad de Medicamentos , Humanos , Repelentes de Insectos/farmacocinética , Insecticidas/farmacocinética , Medicina Militar , Permetrina/farmacocinética , Bromuro de Piridostigmina/farmacocinética , Reproducibilidad de los Resultados
13.
Mil Med ; 169(7): 570-3, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15291194

RESUMEN

A 24-year-old active duty male smoker presented with 3 days of fatigue, rhinorrhea, and sore throat. The diagnosis and management of pharyngitis, including a field friendly approach, are reviewed. The impact of the discontinuation of the adenovirus vaccine to military recruits is highlighted. The effects of smoking among military personnel are discussed, and smoking cessation measures are reviewed.


Asunto(s)
Tos/etiología , Fatiga/etiología , Personal Militar/psicología , Faringitis/etiología , Fumar/efectos adversos , Adulto , Diagnóstico Diferencial , Hospitales Militares , Humanos , Masculino , Cese del Hábito de Fumar/métodos , Prevención del Hábito de Fumar , Estados Unidos
14.
Mil Med ; 167(12): 986-9, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12502172

RESUMEN

OBJECTIVE: To report the occurrence of life-threatening polyneuropathy in association with renal cell carcinoma. METHODS: Case report and review of the medical literature. RESULTS: A 65-year-old man developed a fulminant, life-threatening sensory motor polyneuropathy several months before renal cell carcinoma was identified. Dramatic symptomatic improvement ensued after treatment with intravenous immunoglobulin, although the patient succumbed to complications of the tumor years later. CONCLUSIONS: Chronic sensory motor polyneuropathy is a known paraneoplastic syndrome associated with renal cell carcinoma. This report emphasizes that in rare conditions, the polyneuropathy could take an acute demyelinating form, which is important for clinicians to recognize.


Asunto(s)
Carcinoma de Células Renales/complicaciones , Neoplasias Renales/complicaciones , Polineuropatía Paraneoplásica/etiología , Anciano , Carcinoma de Células Renales/diagnóstico , Diagnóstico Diferencial , Progresión de la Enfermedad , Resultado Fatal , Humanos , Neoplasias Renales/diagnóstico , Masculino , Polineuropatía Paraneoplásica/diagnóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA