Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Biol Chem ; 299(11): 105328, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37806493

RESUMEN

The receptor tyrosine kinase MET is activated by hepatocyte growth factor binding, followed by phosphorylation of the intracellular kinase domain (KD) mainly within the activation loop (A-loop) on Y1234 and Y1235. Dysregulation of MET can lead to both tumor growth and metastatic progression of cancer cells. Tepotinib is a highly selective, potent type Ib MET inhibitor and approved for treatment of non-small cell lung cancer harboring METex14 skipping alterations. Tepotinib binds to the ATP site of unphosphorylated MET with critical π-stacking contacts to Y1230 of the A-loop, resulting in a high residence time. In our study, we combined protein crystallography, biophysical methods (surface plasmon resonance, differential scanning fluorimetry), and mass spectrometry to clarify the impacts of A-loop conformation on tepotinib binding using different recombinant MET KD protein variants. We solved the first crystal structures of MET mutants Y1235D, Y1234E/1235E, and F1200I in complex with tepotinib. Our biophysical and structural data indicated a linkage between reduced residence times for tepotinib and modulation of A-loop conformation either by mutation (Y1235D), by affecting the overall Y1234/Y1235 phosphorylation status (L1195V and F1200I) or by disturbing critical π-stacking interactions with tepotinib (Y1230C). We corroborated these data with target engagement studies by fluorescence cross-correlation spectroscopy using KD constructs in cell lysates or full-length receptors from solubilized cellular membranes as WT or activated mutants (Y1235D and Y1234E/1235E). Collectively, our results provide further insight into the MET A-loop structural determinants that affect the binding of the selective inhibitor tepotinib.


Asunto(s)
Antineoplásicos , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Proteínas Proto-Oncogénicas c-met , Humanos , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , Mutación , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Antineoplásicos/farmacología
2.
J Biol Chem ; 294(11): 4012-4026, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30635397

RESUMEN

The neutral amino acid transporter solute carrier family 1 member 5 (SLC1A5 or ASCT2) is overexpressed in many cancers. To identify its roles in tumors, we employed 143B osteosarcoma cells and HCC1806 triple-negative breast cancer cells with or without ASCT2 deletion. ASCT2ko 143B cells grew well in standard culture media, but ASCT2 was required for optimal growth at <0.5 mm glutamine, with tumor spheroid growth and monolayer migration of 143B ASCT2ko cells being strongly impaired at lower glutamine concentrations. However, the ASCT2 deletion did not affect matrix-dependent invasion. ASCT2ko 143B xenografts in nude mice exhibited a slower onset of growth and a higher number of small tumors than ASCT2wt 143B xenografts, but did not differ in average tumor size 25 days after xenotransplantation. ASCT2 deficiency was compensated by increased levels of sodium neutral amino acid transporter 1 (SNAT1 or SLC38A1) and SNAT2 (SLC38A2) in ASCT2ko 143B cells, mediated by a GCN2 EIF2α kinase (GCN2)-dependent pathway, but this compensation was not observed in ASCT2ko HCC1806 cells. Combined SNAT1 silencing and GCN2 inhibition significantly inhibited growth of ASCT2ko HCC1806 cells, but not of ASCT2ko 143B cells. Similarly, pharmacological inhibition of l-type amino acid transporter 1 (LAT1) and GCN2 significantly inhibited growth of ASCT2ko HCC1806 cells, but not of ASCT2ko 143B cells. We conclude that cancer cells with reduced transporter plasticity are more vulnerable to disruption of amino acid homeostasis than cells with a full capacity to up-regulate redundant transporters by an integrated stress response.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/genética , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Antígenos de Histocompatibilidad Menor/genética , Osteosarcoma/genética , Osteosarcoma/patología , Sistema de Transporte de Aminoácidos ASC/deficiencia , Sistema de Transporte de Aminoácidos ASC/metabolismo , Animales , Neoplasias Óseas/metabolismo , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Ratones , Ratones Noqueados , Antígenos de Histocompatibilidad Menor/metabolismo , Mutación , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Osteosarcoma/metabolismo , Células Tumorales Cultivadas
3.
J Chem Inf Model ; 60(11): 5457-5474, 2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-32813975

RESUMEN

Accurate ranking of compounds with regards to their binding affinity to a protein using computational methods is of great interest to pharmaceutical research. Physics-based free energy calculations are regarded as the most rigorous way to estimate binding affinity. In recent years, many retrospective studies carried out both in academia and industry have demonstrated its potential. Here, we present the results of large-scale prospective application of the FEP+ method in active drug discovery projects in an industry setting at Merck KGaA, Darmstadt, Germany. We compare these prospective data to results obtained on a new diverse, public benchmark of eight pharmaceutically relevant targets. Our results offer insights into the challenges faced when using free energy calculations in real-life drug discovery projects and identify limitations that could be tackled by future method development. The new public data set we provide to the community can support further method development and comparative benchmarking of free energy calculations.


Asunto(s)
Descubrimiento de Drogas , Ligandos , Estudios Prospectivos , Estudios Retrospectivos , Termodinámica
4.
ChemMedChem ; 18(8): e202200615, 2023 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-36749883

RESUMEN

Herein, we describe a systematic SAR- and SPR-investigation of the peptidomimetic hydroxy-proline based VHL-ligand VH032, from which most to-date published VHL-targeting PROTACs have been derived. This study provides for the first time a consistent data set which allows for direct comparison of structural variations including those which were so far hidden in patent literature. The gained knowledge about improved VHL binders was used to design a small library of highly potent BRD4-degraders comprising different VHL exit vectors. Newly designed degraders showed favorable molecular properties and significantly improved degradation potency compared to MZ1.


Asunto(s)
Proteínas Nucleares , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Ligandos , Proteínas Nucleares/metabolismo , Proteolisis , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
5.
Biosensors (Basel) ; 13(10)2023 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-37887106

RESUMEN

Modern drug discovery relies on combinatorial screening campaigns to find drug molecules targeting specific disease-associated proteins. The success of such campaigns often relies on functional and structural information of the selected therapeutic target, only achievable once its purification is mastered. With the aim of bypassing the protein purification process to gain insights on the druggability, ligand binding, and/or characterization of protein-protein interactions, herein, we describe the Extract2Chip method. This approach builds on the immobilization of site-specific biotinylated proteins of interest, directly from cellular extracts, on avidin-coated sensor chips to allow for the characterization of molecular interactions via surface plasmon resonance (SPR). The developed method was initially validated using Cyclophilin D (CypD) and subsequently applied to other drug discovery projects in which the targets of interest were difficult to express, purify, and crystallize. Extract2Chip was successfully applied to the characterization of Yes-associated protein (YAP): Transcriptional enhancer factor TEF (TEAD1) protein-protein interaction inhibitors, in the validation of a ternary complex assembly composed of Dyskerin pseudouridine synthase 1 (DKC1) and RuvBL1/RuvBL2, and in the establishment of a fast-screening platform to select the most suitable NUAK family SNF1-like kinase 2 (NUAK2) surrogate for binding and structural studies. The described method paves the way for a potential revival of the many drug discovery campaigns that have failed to deliver due to the lack of suitable and sufficient protein supply.


Asunto(s)
Descubrimiento de Drogas , Resonancia por Plasmón de Superficie , Resonancia por Plasmón de Superficie/métodos , Descubrimiento de Drogas/métodos , Proteínas , Cromatografía de Afinidad , Unión Proteica
6.
J Med Chem ; 66(13): 8666-8686, 2023 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-37403966

RESUMEN

Hypoxia-inducible factors (HIFs) are heterodimeric transcription factors induced in diverse pathophysiological settings. Inhibition of HIF-2α has become a strategy for cancer treatment since the discovery that small molecules, upon binding into a small cavity of the HIF-2α PAS B domain, can alter its conformation and disturb the activity of the HIF dimer complex. Herein, the design, synthesis, and systematic SAR exploration of cycloalkyl[c]thiophenes as novel HIF-2α inhibitors are described, providing the first chemotype featuring an alkoxy-aryl scaffold. X-ray data confirmed the ability of these inhibitors to induce perturbation of key amino acids by appropriately presenting key pharmacophoric elements in the hydrophobic cavity. Selected compounds showed inhibition of VEGF-A secretion in cancer cells and prevention of Arg1 expression and activity in IL4-stimulated macrophages. Moreover, in vivo target gene modulation was demonstrated with compound 35r. Thus, the disclosed HIF-2α inhibitors represent valuable tools for investigating selective HIF-2α inhibition and its effect on tumor biology.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Tiofenos , Humanos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Tiofenos/farmacología , Factores de Transcripción , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia
7.
Anal Biochem ; 421(1): 138-51, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22085444

RESUMEN

Targeting of the epidermal growth factor receptor (EGFR) with monoclonal antibodies has become an established antitumor strategy in clinical use or in late stages of drug development. The mAbs effector mechanisms have been widely analyzed based on in vivo or cell studies. Hereby we intend to complement these functional studies by investigating the mAb-EGFR interactions on a molecular level. Surface plasmon resonance, isothermal titration calorimetry, and static light scattering were employed to characterize the interactions of matuzumab, cetuximab, and panitumumab with the extracellular soluble form ecEGFR. The kinetic and thermodynamic determinants dissected the differences in mAbs binding mechanism toward ecEGFR. The quantitative stoichiometric data clearly demonstrated the bivalent binding of the mAbs to two ecEGFR molecules. Our results complement earlier studies on simultaneous binding of cetuximab and matuzumab. The antibodies retain their bivalent binding mode achieving a 1:2:1 complex formation. Interestingly the binding parameters remain nearly constant for the individual antibodies in this ternary assembly. In contrast the binding of panitumumab is almost exclusive either by directly blocking the accessibility for the second antibody or by negative allosteric modulation. Overall we provide a comprehensive biophysical dataset on binding parameters, the complex assembly, and relative epitope accessibility for therapeutic anti-EGFR antibodies.


Asunto(s)
Anticuerpos Monoclonales/química , Receptores ErbB/química , Receptores ErbB/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados/química , Sitios de Unión , Fenómenos Biofísicos , Calorimetría , Cetuximab , Receptores ErbB/antagonistas & inhibidores , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Cinética , Ligandos , Luz , Ratones , Peso Molecular , Panitumumab , Dominios y Motivos de Interacción de Proteínas , Dispersión de Radiación , Resonancia por Plasmón de Superficie
8.
Bioorg Med Chem Lett ; 22(13): 4396-403, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22632933

RESUMEN

Inhibitors of the Hsp90 molecular chaperone are showing considerable promise as potential molecular therapeutic agents for the treatment of cancer. Here we describe the identification of novel small molecular weight inhibitors of Hsp90 using a fragment based approach. Fragments were selected by docking, tested in a biochemical assay and the confirmed hits were crystallized. Information gained from X-ray structures of these fragments and other chemotypes was used to drive the fragment evolution process. Optimization of these high µM binders resulted in 3-benzylindazole derivatives with significantly improved affinity and anti-proliferative effects in different human cancer cell lines.


Asunto(s)
Amidas/química , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Indazoles/química , Bibliotecas de Moléculas Pequeñas/química , Amidas/toxicidad , Sitios de Unión , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Simulación por Computador , Cristalografía por Rayos X , Evaluación Preclínica de Medicamentos , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Estructura Terciaria de Proteína , Bibliotecas de Moléculas Pequeñas/toxicidad , Relación Estructura-Actividad
9.
J Immunother Cancer ; 10(7)2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35858707

RESUMEN

BACKGROUND: Bintrafusp alfa (BA) is a bifunctional fusion protein designed for colocalized, simultaneous inhibition of two immunosuppressive pathways, transforming growth factor-ß (TGF-ß) and programmed death-ligand 1 (PD-L1), within the tumor microenvironment (TME). We hypothesized that targeting PD-L1 to the tumor by BA colocalizes the TGF-ß trap (TGF-ßRII) to the TME, enabling it to sequester TGF-ß in the tumor more effectively than systemic TGF-ß blockade, thereby enhancing antitumor activity. METHODS: Multiple technologies were used to characterize the TGF-ß trap binding avidity. BA versus combinations of anti-PD-L1 and TGF-ß trap or the pan-TGF-ß antibody fresolimumab were compared in proliferation and two-way mixed lymphocyte reaction assays. Immunophenotyping of tumor-infiltrating lymphocytes (TILs) and RNA sequencing (RNAseq) analysis assessing stromal and immune landscape following BA or the combination therapy were performed in MC38 tumors. TGF-ß and PD-L1 co-expression and their associated gene signatures in MC38 tumors and human lung carcinoma tissue were studied with single-cell RNAseq (scRNAseq) and immunostaining. BA-induced internalization, degradation, and depletion of TGF-ß were investigated in vitro. RESULTS: BA and fresolimumab had comparable intrinsic binding to TGF-ß1, but there was an ~80× avidity-based increase in binding affinity with BA. BA inhibited cell proliferation in TGF-ß-dependent and PD-L1-expressing cells more potently than TGF-ß trap or fresolimumab. Compared with the combination of anti-PD-L1 and TGF-ß trap or fresolimumab, BA enhanced T cell activation in vitro and increased TILs in MC38 tumors, which correlated with efficacy. BA induced distinct gene expression in the TME compared with the combination therapy, including upregulation of immune-related gene signatures and reduced activities in TGF-ß-regulated pathways, such as epithelial-mesenchymal transition, extracellular matrix deposition, and fibrosis. Regulatory T cells, macrophages, immune cells of myeloid lineage, and fibroblasts were key PD-L1/TGF-ß1 co-expressing cells in the TME. scRNAseq analysis suggested BA modulation of the macrophage phenotype, which was confirmed by histological assessment. PD-L1/TGF-ß1 co-expression was also seen in human tumors. Finally, BA induced TGF-ß1 internalization and degradation in the lysosomes. CONCLUSION: BA more effectively blocks TGF-ß by targeting TGF-ß trap to the tumor via PD-L1 binding. Such colocalized targeting elicits distinct and superior antitumor responses relative to single agent combination therapy.


Asunto(s)
Neoplasias Pulmonares , Factor de Crecimiento Transformador beta , Antígeno B7-H1 , Humanos , Factores Inmunológicos , Receptor de Muerte Celular Programada 1 , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1 , Microambiente Tumoral
10.
J Med Chem ; 64(14): 10371-10392, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34255518

RESUMEN

Constitutive activation of the canonical Wnt signaling pathway, in most cases driven by inactivation of the tumor suppressor APC, is a hallmark of colorectal cancer. Tankyrases are druggable key regulators in these malignancies and are considered as attractive targets for therapeutic interventions, although no inhibitor has been progressed to clinical development yet. We continued our efforts to develop tankyrase inhibitors targeting the nicotinamide pocket with suitable drug-like properties for investigating effects of Wnt pathway inhibition on tumor growth. Herein, the identification of a screening hit series and its optimization through scaffold hopping and SAR exploration is described. The systematic assessment delivered M2912, a compound with an optimal balance between excellent TNKS potency, exquisite PARP selectivity, and a predicted human PK compatible with once daily oral dosing. Modulation of cellular Wnt pathway activity and significant tumor growth inhibition was demonstrated with this compound in colorectal xenograft models in vivo.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Tanquirasas/antagonistas & inhibidores , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Femenino , Humanos , Ratones , Ratones SCID , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Relación Estructura-Actividad , Tanquirasas/metabolismo
11.
J Med Chem ; 64(16): 11904-11933, 2021 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-34382802

RESUMEN

Due to increased lactate production during glucose metabolism, tumor cells heavily rely on efficient lactate transport to avoid intracellular lactate accumulation and acidification. Monocarboxylate transporter 4 (MCT4/SLC16A3) is a lactate transporter that plays a central role in tumor pH modulation. The discovery and optimization of a novel class of MCT4 inhibitors (hit 9a), identified by a cellular screening in MDA-MB-231, is described. Direct target interaction of the optimized compound 18n with the cytosolic domain of MCT4 was shown after solubilization of the GFP-tagged transporter by fluorescence cross-correlation spectroscopy and microscopic studies. In vitro treatment with 18n resulted in lactate efflux inhibition and reduction of cellular viability in MCT4 high expressing cells. Moreover, pharmacokinetic properties of 18n allowed assessment of lactate modulation and antitumor activity in a mouse tumor model. Thus, 18n represents a valuable tool for investigating selective MCT4 inhibition and its effect on tumor biology.


Asunto(s)
Antineoplásicos/uso terapéutico , Transportadores de Ácidos Monocarboxílicos/antagonistas & inhibidores , Proteínas Musculares/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Ácidos Picolínicos/uso terapéutico , Sulfonamidas/uso terapéutico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Células HEK293 , Humanos , Ácido Láctico/metabolismo , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones SCID , Estructura Molecular , Ácidos Picolínicos/síntesis química , Ácidos Picolínicos/farmacología , Relación Estructura-Actividad , Sulfonamidas/síntesis química , Sulfonamidas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
J Med Chem ; 62(24): 11119-11134, 2019 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-31725285

RESUMEN

The recently disclosed next generation of reversible, selective, and potent MetAP-2 inhibitors introduced a cyclic tartronic diamide scaffold. However, the lead compound 1a suffered from enterohepatic circulation, preventing further development. Nevertheless, 1a served as a starting point for further optimization. Maintaining potent antiproliferation activity, while improving other compound properties, enabled the generation of an attractive array of new MetAP-2 inhibitors. The most promising derivatives were identified by a multiparameter analysis of the compound properties. Essential for the efficient selection of candidates with in vivo activity was the identification of molecules with a long residence time on the target protein, high permeability, and low efflux ratio not only in Caco-2 but also in the MDR-MDCK cell line. Orally bioavailable, potent, and reversible MetAP-2 inhibitors impede the growth of primary endothelial cells and demonstrated antitumoral activity in mouse models. This assessment led to the nomination of the clinical development compound M8891, which is currently in phase I clinical testing in oncology patients.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Glioma/tratamiento farmacológico , Indoles/farmacología , Metionil Aminopeptidasas/antagonistas & inhibidores , Células A549 , Animales , Antineoplásicos/química , Apoptosis , Células CACO-2 , Proliferación Celular , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Inhibidores Enzimáticos/química , Femenino , Glioma/metabolismo , Glioma/patología , Humanos , Indoles/química , Ratones , Ratones Desnudos , Modelos Moleculares , Relación Estructura-Actividad , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Med Chem ; 62(17): 7643-7655, 2019 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-31368705

RESUMEN

Bruton's tyrosine kinase (BTK) inhibitors such as ibrutinib hold a prominent role in the treatment of B cell malignancies. However, further refinement is needed to this class of agents, particularly in terms of adverse events (potentially driven by kinase promiscuity), which preclude their evaluation in nononcology indications. Here, we report the discovery and preclinical characterization of evobrutinib, a potent, obligate covalent inhibitor with high kinase selectivity. Evobrutinib displayed sufficient preclinical pharmacokinetic and pharmacodynamic characteristics which allowed for in vivo evaluation in efficacy models. Moreover, the high selectivity of evobrutinib for BTK over epidermal growth factor receptor and other Tec family kinases suggested a low potential for off-target related adverse effects. Clinical investigation of evobrutinib is ongoing in several autoimmune diseases, including multiple sclerosis, rheumatoid arthritis, and systemic lupus erythematosus.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Descubrimiento de Drogas , Enfermedades del Sistema Inmune/tratamiento farmacológico , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Administración Oral , Agammaglobulinemia Tirosina Quinasa/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Enfermedades del Sistema Inmune/metabolismo , Estructura Molecular , Piperidinas/administración & dosificación , Piperidinas/química , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/química , Pirimidinas/administración & dosificación , Pirimidinas/química , Relación Estructura-Actividad
14.
J Mol Biol ; 330(5): 1203-13, 2003 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-12860139

RESUMEN

Sensory rhodopsin II, a repellent phototaxis receptor from Natronobacterium pharaonis (NpSRII) forms a tight complex with its cognate transducer (NpHtrII). Light excitation of the receptor triggers conformational changes in both proteins, thereby activating the cellular two-component signalling cascade. In membranes, the two proteins form a 2:2 complex, which dissociates to a 1:1 heterodimer in micelles. Complexed to the transducer sensory rhodopsin II is no longer capable of light-driven proton pumping. In order to elucidate the dimerisation and the size of the receptor-binding domain of the transducer, isothermal titration calorimetry and electrophysiological experiments have been carried out. It is shown, that an N-terminal sequence of 114 amino acid residues is sufficient for tight binding (K(d)=240nM; DeltaH=-17.6kJmol(-1)) and for inhibiting the proton transfer. These data and results obtained from selected site-directed mutants indicate a synergistic interplay of transducer transmembrane domain (1-82) and cytoplasmic peptide (83-114) leading to an optimal and specific interaction between receptor and transducer.


Asunto(s)
Proteínas Arqueales/química , Carotenoides/química , Halorrodopsinas , Rodopsinas Sensoriales , Animales , Calorimetría , Dimerización , Electroforesis en Gel de Poliacrilamida , Electrofisiología , Concentración de Iones de Hidrógeno , Cinética , Luz , Micelas , Modelos Moleculares , Oocitos , Péptidos/química , Plásmidos/metabolismo , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Protones , ARN Mensajero/metabolismo , Transducción de Señal , Temperatura , Termodinámica , Factores de Tiempo , Xenopus
15.
J Mol Biol ; 425(2): 309-33, 2013 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-23154170

RESUMEN

Syk is an essential non-receptor tyrosine kinase in intracellular immunological signaling, and the control of Syk kinase function is considered as a valuable target for pharmacological intervention in autoimmune or inflammation diseases. Upon immune receptor stimulation, the kinase activity of Syk is regulated by binding of phosphorylated immune receptor tyrosine-based activating motifs (pITAMs) to the N-terminal tandem Src homology 2 (tSH2) domain and by autophosphorylation with consequences for the molecular structure of the Syk protein. Here, we present the first crystal structures of full-length Syk (fl-Syk) as wild type and as Y348F,Y352F mutant forms in complex with AMP-PNP revealing an autoinhibited conformation. The comparison with the crystal structure of the truncated Syk kinase domain in complex with AMP-PNP taken together with ligand binding studies by surface plasmon resonance (SPR) suggests conformational differences in the ATP sites of autoinhibited and activated Syk forms. This hypothesis was corroborated by studying the thermodynamic and kinetic interaction of three published Syk inhibitors with isothermal titration calorimetry and SPR, respectively. We further demonstrate the modulation of inhibitor binding affinities in the presence of pITAM and discuss the observed differences of thermodynamic and kinetic signatures. The functional relevance of pITAM binding to fl-Syk was confirmed by a strong stimulation of in vitro autophosphorylation. A structural feedback mechanism on the kinase domain upon pITAM binding to the tSH2 domain is discussed in analogy of the related family kinase ZAP-70 (Zeta-chain-associated protein kinase 70). Surprisingly, we observed distinct conformations of the tSH2 domain and the activation switch including Tyr348 and Tyr352 in the interdomain linker of Syk in comparison to ZAP-70.


Asunto(s)
Adenosina Trifosfato/metabolismo , Péptidos y Proteínas de Señalización Intracelular/química , Fosfotirosina/química , Proteínas Tirosina Quinasas/química , Proteína Tirosina Quinasa ZAP-70/química , Adenilil Imidodifosfato/metabolismo , Cristalografía por Rayos X , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Cinética , Modelos Moleculares , Mutación/genética , Fosforilación , Fosfotirosina/metabolismo , Unión Proteica , Conformación Proteica , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Resonancia por Plasmón de Superficie , Quinasa Syk , Termodinámica , Tirosina/metabolismo , Proteína Tirosina Quinasa ZAP-70/metabolismo
16.
PLoS One ; 8(11): e78443, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24265689

RESUMEN

The molecular chaperones of the Hsp70 family have been recognized as targets for anti-cancer therapy. Since several paralogs of Hsp70 proteins exist in cytosol, endoplasmic reticulum and mitochondria, we investigated which isoform needs to be down-regulated for reducing viability of cancer cells. For two recently identified small molecule inhibitors, VER-155008 and 2-phenylethynesulfonamide (PES), which are proposed to target different sites in Hsp70s, we analyzed the molecular mode of action in vitro. We found that for significant reduction of viability of cancer cells simultaneous knockdown of heat-inducible Hsp70 (HSPA1) and constitutive Hsc70 (HSPA8) is necessary. The compound VER-155008, which binds to the nucleotide binding site of Hsp70, arrests the nucleotide binding domain (NBD) in a half-open conformation and thereby acts as ATP-competitive inhibitor that prevents allosteric control between NBD and substrate binding domain (SBD). Compound PES interacts with the SBD of Hsp70 in an unspecific, detergent-like fashion, under the conditions tested. None of the two inhibitors investigated was isoform-specific.


Asunto(s)
Proteínas del Choque Térmico HSC70/antagonistas & inhibidores , Nucleósidos de Purina/farmacología , Sulfonamidas/farmacología , Adenosina Trifosfatasas/antagonistas & inhibidores , Adenosina Trifosfato/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Diseño de Fármacos , Proteínas del Choque Térmico HSC70/química , Proteínas del Choque Térmico HSC70/metabolismo , Humanos , Hidrólisis/efectos de los fármacos , Luciferasas/química , Conformación Molecular , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Replegamiento Proteico/efectos de los fármacos , Estructura Terciaria de Proteína
18.
Anal Chem ; 77(7): 1957-62, 2005 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15801724

RESUMEN

A combination of electrospray mass spectrometry (ESI-MS) and element mass spectrometry (ICPMS) with phosphorus detection was used to characterize histidine phosphorylation (His-48) of the chemotaxis protein CheA quantitatively. The phosphorylation at His-48 was found to be responsible for a stabilization of the protein. For this investigation, the acceptor domain and the kinase domain of the bacterial chemotaxis protein CheA were recombinantly expressed as single proteins. Using in vitro kinase assay conditions, the acceptor domain CheA-H was phosphorylated by the kinase domain CheA-C. The degree of histidine phosphorylation was determined by nanoelectrospray mass spectrometry of intact CheA-H, and was found to be limited to a maximum value of approximately 50%. The site specificity of CheA-H phosphorylation was controlled by nanoESI-MS/MS of the [M + 16H](16+) ion of intact (pHis)-CheA-H and allowed localization of the pHis residue to the region between residues 32 and 86, containing candidates His-48 and His-67, for which His-48 phosphorylation has been described. Analysis of the tryptic digest of in vitro histidine-phosphorylated CheA-H by capillary chromatography coupled to ESI-MS and to ICPMS with phosphorus detection revealed a truncated (pHis)-CheA-H protein as the only phosphorus-containing analyte. Since the truncated (pHis)-CheA-H in the digest was found to exhibit a higher degree of phosphorylation than could be generated by in vitro phosphorylation without trypsin treatment, it is concluded that histidine phosphorylation at His-48 strongly interferes with structural properties of the CheA-H domain in particular with respect to proteolytic degradation by trypsin.


Asunto(s)
Proteínas Bacterianas/metabolismo , Histidina/metabolismo , Proteínas de la Membrana/metabolismo , Espectrometría de Masa por Ionización de Electrospray/métodos , Secuencia de Aminoácidos , Proteínas Bacterianas/análisis , Proteínas Bacterianas/genética , Cromatografía Liquida/métodos , Proteínas de la Membrana/análisis , Proteínas de la Membrana/genética , Proteínas Quimiotácticas Aceptoras de Metilo , Datos de Secuencia Molecular , Nanotecnología/métodos , Fosforilación , Proteínas Recombinantes/análisis , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
19.
J Biol Chem ; 280(46): 38767-75, 2005 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-16157581

RESUMEN

Sensory rhodopsin II, the photophobic receptor from Natronomonas pharaonis (NpSRII)5, forms a 2:2 complex with its cognate transducer (N. pharaonis halobacterial transducer of rhodopsins II (NpHtrII)) in lipid membranes. Light activation of NpSRII leads to a displacement of helix F, which in turn triggers a rotation/screw-like motion of TM2 in NpHtrII. This conformational change is thought to be transmitted through the membrane adjacent conserved signal transduction domain in histidine kinases, adenylyl cyclases, methyl-accepting chemotaxis proteins, and phosphatases (HAMP domain) to the cytoplasmic signaling domain of the transducer. The architecture and function of the HAMP domain are still unknown. In order to obtain information on the structure and dynamics of this region, EPR experiments on a truncated transducer (NpHtrII(157)) and NpSRII, site-directed spin-labeled and reconstituted into purple membrane lipids, have been carried out. A nitroxide scanning involving residues in the transducer helix TM2, in the predicted AS-1 region, and at selected positions in the following connector and AS-2 regions of the HAMP domain has been performed. Accessibility and dynamics data allowed us to identify a helical region up to residue Ala(94) in the AS-1 amphipathic sequence, followed by a highly dynamic domain protruding into the water phase. Additionally, transducer-transducer and transducer-receptor proximity relations revealed the overall architecture of the AS-1 sequences in the 2:2 complex, which are suggested to form a molten globular type of a coiled-coil bundle.


Asunto(s)
Halorrodopsinas/química , Rodopsinas Sensoriales/química , Secuencia de Aminoácidos , Archaea/metabolismo , Membrana Celular/metabolismo , Cisteína/química , Espectroscopía de Resonancia por Spin del Electrón , Escherichia coli/metabolismo , Histidina Quinasa , Luz , Lípidos/química , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Óxido Nítrico/química , Unión Proteica , Conformación Proteica , Proteínas Quinasas/metabolismo , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Transducción de Señal , Temperatura , Termodinámica
20.
Biophys J ; 84(2 Pt 1): 1208-17, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12547800

RESUMEN

The photocycle of the photophobic receptor from Natronobacterium pharaonis, NpSRII, is studied by static and time-resolved step-scan Fourier transform infrared spectroscopy. Both low-temperature static and time-resolved spectra resolve a K-like intermediate, and the corresponding spectra show little difference within the noise of the time-resolved data. As compared to intermediate K of bacteriorhodopsin, relatively large amide I bands indicate correspondingly larger distortions of the protein backbone. The time-resolved spectra identify an intermediate L-like state with surprisingly small additional molecular alterations. With the formation of intermediate M, the Schiff-base proton is transferred to the counterion Asp-75. This state is characterized by larger amide bands indicating larger distortions of the protein. We can identify a second M state that differs only in small-protein bands. Reisomerization of the chromophore to all-trans occurs with the formation of intermediate O. The accelerated decay of intermediate M caused by azide results in another red-shifted intermediate with a protonated Schiff base. The chromophore in this state, however, still has 13-cis geometry. Nevertheless, the reisomerization is still as slow as under the conditions without azide. The results are discussed with respect to mechanisms of the observed proton pumping and the possible roles of the intermediates in receptor activation.


Asunto(s)
Proteínas Arqueales/química , Carotenoides/química , Halorrodopsinas , Protones , Rodopsinas Sensoriales , Espectroscopía Infrarroja por Transformada de Fourier/métodos , Azidas/química , Transferencia de Energía , Isomerismo , Natronobacterium/química , Fotoquímica/métodos , Conformación Proteica/efectos de la radiación , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA