Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Chemphyschem ; 25(1): e202300636, 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-37955910

RESUMEN

The availability of high-resolution 3D structural information is crucial for investigating guest-host systems across a wide range of fields. In the context of drug discovery, the information is routinely used to establish and validate structure-activity relationships, grow initial hits from screening campaigns, and to guide molecular docking. For the generation of protein-ligand complex structural information, X-ray crystallography is the experimental method of choice, however, with limited information on protein flexibility. An experimentally verified structural model of the binding interface in the native solution-state would support medicinal chemists in their molecular design decisions. Here we demonstrate that protein-bound ligand 1 H NMR chemical shifts are highly sensitive and accurate probes for the immediate chemical environment of protein-ligand interfaces. By comparing the experimental ligand 1 H chemical shift values with those computed from the X-ray structure using quantum mechanics methodology, we identify significant disagreements for parts of the ligand between the two experimental techniques. We show that quantum mechanics/molecular mechanics (QM/MM) molecular dynamics (MD) ensembles can be used to refine initial X-ray co-crystal structures resulting in a better agreement with experimental 1 H ligand chemical shift values. Overall, our findings highlight the usefulness of ligand 1 H NMR chemical shift information in combination with a QM/MM MD workflow for generating protein-ligand ensembles that accurately reproduce solution structural data.


Asunto(s)
Imagen por Resonancia Magnética , Proteínas , Simulación del Acoplamiento Molecular , Ligandos , Espectroscopía de Resonancia Magnética/métodos , Proteínas/química
2.
Arch Pharm (Weinheim) ; 357(6): e2300649, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38396281

RESUMEN

Transcription factors are generally considered challenging, if not "undruggable", targets but they promise new therapeutic options due to their fundamental involvement in many diseases. In this study, we aim to assess the ligandability of the C-terminal Rel-homology domain of nuclear factor of activated T cells 1 (NFAT1), a TF implicated in T-cell regulation. Using a combination of experimental and computational approaches, we demonstrate that small molecule fragments can indeed bind to this protein domain. The newly identified binder is the first small molecule binder to NFAT1 validated with biophysical methods and an elucidated binding mode by X-ray crystallography. The reported eutomer/distomer pair provides a strong basis for potential exploration of higher potency binders on the path toward degrader or glue modalities.


Asunto(s)
Factores de Transcripción NFATC , Sitios de Unión , Cristalografía por Rayos X , Ligandos , Factores de Transcripción NFATC/metabolismo , Factores de Transcripción NFATC/química , Unión Proteica , Dominios Proteicos , Relación Estructura-Actividad
3.
Int J Colorectal Dis ; 37(5): 1107-1117, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35426079

RESUMEN

BACKGROUND: The impact of body mass index (BMI) on prognosis in patients with curatively resected stage I-III colon carcinoma was analyzed. METHODS: The prospectively collected data of 694 patients who underwent complete mesocolic excision between 2003 and 2014 were analyzed. BMI was classified into four categories: underweight (BMI < 18.5 kg/m2; n = 13), normal weight (BMI 18.5 to 24.9 kg/m2; n = 221), overweight (BMI 25.0 to 29.9 kg/m2; n = 309), and obese (BMI ≥ 30.0 kg/m2; n = 151). Univariate and multivariate analyses for comparison of prognosis were performed. RESULTS: The 5-year rate of locoregional recurrence in all 694 patients was 2.1%, and no differences were found with respect to BMI (p = 0.759). For distant metastasis, the 5-year rate for all patients was 13.4%, and BMI did not have a significant impact (p = 0.593). The 5-year rate of disease-free survival for all 694 patients was 72.4%. The differences with respect to BMI were not found to be significant in univariate analysis (p = 0.222). In multivariate Cox regression analysis, disease-free survival was significantly better in obese patients (HR 0.7; p = 0.034). Regarding overall survival, the 5-year rate for all patients was 78.1%. In univariate analyses, no significant differences were found for BMI (p = 0.094). In the Cox regression analysis, overweight and obese patients had significantly better survival (overweight: HR 0.7; p = 0.027; obese: HR 0.6; p = 0.019). CONCLUSION: The better survival of overweight and obese patients in multivariate analyses must be interpreted with caution. It is influenced by several factors and seems to correspond to the phenomenon of the obesity paradox.


Asunto(s)
Carcinoma , Neoplasias del Colon , Índice de Masa Corporal , Neoplasias del Colon/complicaciones , Neoplasias del Colon/cirugía , Humanos , Recurrencia Local de Neoplasia , Obesidad/complicaciones , Sobrepeso/complicaciones , Pronóstico , Estudios Retrospectivos
4.
Nat Chem Biol ; 15(8): 822-829, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31285596

RESUMEN

Here, we report the fragment-based discovery of BI-9321, a potent, selective and cellular active antagonist of the NSD3-PWWP1 domain. The human NSD3 protein is encoded by the WHSC1L1 gene located in the 8p11-p12 amplicon, frequently amplified in breast and squamous lung cancer. Recently, it was demonstrated that the PWWP1 domain of NSD3 is required for the viability of acute myeloid leukemia cells. To further elucidate the relevance of NSD3 in cancer biology, we developed a chemical probe, BI-9321, targeting the methyl-lysine binding site of the PWWP1 domain with sub-micromolar in vitro activity and cellular target engagement at 1 µM. As a single agent, BI-9321 downregulates Myc messenger RNA expression and reduces proliferation in MOLM-13 cells. This first-in-class chemical probe BI-9321, together with the negative control BI-9466, will greatly facilitate the elucidation of the underexplored biological function of PWWP domains.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Proteínas Nucleares/antagonistas & inhibidores , Sistemas CRISPR-Cas , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Regulación de la Expresión Génica/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Dominios Proteicos , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo
5.
Proc Natl Acad Sci U S A ; 115(44): E10505-E10514, 2018 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-30309962

RESUMEN

Human BCL-2-associated death promoter (hBAD) is an apoptosis-regulatory protein mediating survival signals to carcinoma cells upon phosphorylation of Ser99, among other residues. Herein, we screened multiple small-molecule databases queried in a Laplacian-modified naive Bayesian-based cheminformatics platform and identified a Petasis reaction product as a site-specific inhibitor for hBAD phosphorylation. Based on apoptotic efficacy against mammary carcinoma cells, N-cyclopentyl-3-((4-(2,3-dichlorophenyl) piperazin-1-yl) (2-hydroxyphenyl) methyl) benzamide (NPB) was identified as a potential lead compound. In vitro biochemical analyses demonstrated that NPB inhibited the phosphorylation of hBAD specifically on Ser99. NPB was observed to exert this effect independently of AKT and other kinase activities despite the demonstration of AKT-mediated BAD-Ser99 phosphorylation. Using a structure-based bioinformatics platform, we observed that NPB exhibited predicted interactions with hBAD in silico and verified the same by direct binding kinetics. NPB reduced phosphorylation of BAD-Ser99 and enhanced caspase 3/7 activity with associated loss of cell viability in various human cancer cell lines derived from mammary, endometrial, ovarian, hepatocellular, colon, prostatic, and pancreatic carcinoma. Furthermore, by use of a xenograft model, it was observed that NPB, as a single agent, markedly diminished BAD phosphorylation in tumor tissue and significantly inhibited tumor growth. Similar doses of NPB utilized in acute toxicity studies in mice did not exhibit significant effects. Hence, we report a site-specific inhibitor of BAD phosphorylation with efficacy in tumor models.


Asunto(s)
Antineoplásicos/farmacología , Benzamidas/farmacología , Supervivencia Celular/efectos de los fármacos , Piperazinas/farmacología , Serina/química , Proteína Letal Asociada a bcl/antagonistas & inhibidores , Antineoplásicos/química , Apoptosis , Benzamidas/química , Proliferación Celular , Bases de Datos Factuales , Sistemas de Liberación de Medicamentos , Descubrimiento de Drogas , Humanos , Células MCF-7 , Fosforilación , Piperazinas/química , Interferencia de ARN , Bibliotecas de Moléculas Pequeñas , Resonancia por Plasmón de Superficie
6.
J Comput Chem ; 41(10): 986-999, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-31930547

RESUMEN

Alchemically derived free energies are artifacted when the perturbed moiety has a nonzero net charge. The source of the artifacts lies in the effective treatment of the electrostatic interactions within and between the perturbed atoms and remaining (partial) charges in the simulated system. To treat the electrostatic interactions effectively, lattice-summation (LS) methods or cutoff schemes in combination with a reaction-field contribution are usually employed. Both methods render the charging component of the calculated free energies sensitive to essential parameters of the system like the cutoff radius or the box side lengths. Here, we discuss the results of three previously published studies of ligand binding. These studies presented estimates of binding free energies that were artifacted due to the charged nature of the ligands. We show that the size of the artifacts can be efficiently calculated and raw simulation data can be corrected. We compare the corrected results with experimental estimates and nonartifacted estimates from path-sampling methods. Although the employed correction scheme involves computationally demanding continuum-electrostatics calculations, we show that the correction estimate can be deduced from a small sample of configurations rather than from the entire ensemble. This observation makes the calculations of correction terms feasible for complex biological systems. To show the general applicability of the proposed procedure, we also present results where the correction scheme was used to correct independent free energies obtained from simulations employing a cutoff scheme or LS electrostatics. In this work, we give practical guidelines on how to apply the appropriate corrections easily.


Asunto(s)
Electricidad Estática , Artefactos , Sitios de Unión , ADN/química , Distamicinas/química , Ligandos , Simulación de Dinámica Molecular , Netropsina/química , Solventes/química , Termodinámica , Inhibidores de Tripsina/química
7.
Angew Chem Int Ed Engl ; 59(35): 14861-14868, 2020 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-32421895

RESUMEN

While CH-π interactions with target proteins are crucial determinants for the affinity of arguably every drug molecule, no method exists to directly measure the strength of individual CH-π interactions in drug-protein complexes. Herein, we present a fast and reliable methodology called PI (π interactions) by NMR, which can differentiate the strength of protein-ligand CH-π interactions in solution. By combining selective amino-acid side-chain labeling with 1 H-13 C NMR, we are able to identify specific protein protons of side-chains engaged in CH-π interactions with aromatic ring systems of a ligand, based solely on 1 H chemical-shift values of the interacting protein aromatic ring protons. The information encoded in the chemical shifts induced by such interactions serves as a proxy for the strength of each individual CH-π interaction. PI by NMR changes the paradigm by which chemists can optimize the potency of drug candidates: direct determination of individual π interactions rather than averaged measures of all interactions.


Asunto(s)
Espectroscopía de Resonancia Magnética/métodos , Proteínas/química , Humanos , Modelos Moleculares
8.
Hum Mol Genet ; 26(18): 3531-3544, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28911204

RESUMEN

Human proteins are vulnerable towards disease-associated single amino acid replacements affecting protein stability and function. Interestingly, a few studies have shown that consensus amino acids from mammals or vertebrates can enhance protein stability when incorporated into human proteins. Here, we investigate yet unexplored relationships between the high vulnerability of human proteins towards disease-associated inactivation and recent evolutionary site-specific divergence of stabilizing amino acids. Using phylogenetic, structural and experimental analyses, we show that divergence from the consensus amino acids at several sites during mammalian evolution has caused local protein destabilization in two human proteins linked to disease: cancer-associated NQO1 and alanine:glyoxylate aminotransferase, mutated in primary hyperoxaluria type I. We demonstrate that a single consensus mutation (H80R) acts as a disease suppressor on the most common cancer-associated polymorphism in NQO1 (P187S). The H80R mutation reactivates P187S by enhancing FAD binding affinity through local and dynamic stabilization of its binding site. Furthermore, we show how a second suppressor mutation (E247Q) cooperates with H80R in protecting the P187S polymorphism towards inactivation through long-range allosteric communication within the structural ensemble of the protein. Our results support that recent divergence of consensus amino acids may have occurred with neutral effects on many functional and regulatory traits of wild-type human proteins. However, divergence at certain sites may have increased the propensity of some human proteins towards inactivation due to disease-associated mutations and polymorphisms. Consensus mutations also emerge as a potential strategy to identify structural hot-spots in proteins as targets for pharmacological rescue in loss-of-function genetic diseases.


Asunto(s)
Angiotensinógeno/genética , Proteínas/genética , Alanina/genética , Alanina Transaminasa/genética , Alanina Transaminasa/metabolismo , Aminoácidos/genética , Angiotensinógeno/metabolismo , Animales , Sitios de Unión , Secuencia de Consenso/genética , Evolución Molecular , Humanos , Mutación , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Filogenia , Polimorfismo Genético , Unión Proteica , Estabilidad Proteica , Proteínas/metabolismo , Transaminasas/genética , Transaminasas/metabolismo
9.
J Chem Inf Model ; 59(1): 137-148, 2019 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-30532974

RESUMEN

The protozoan cysteine proteases cruzain in Trypanosoma cruzi and rhodesain in Trypanosoma brucei are therapeutic targets for Chagas disease and Human African Trypanosomiasis (HAT), respectively. A benzimidazole series was previously characterized as potent noncovalent competitive cruzain and rhodesain inhibitors with activity against trypanosomes. Common structure-activity relationships (SAR) trends and structural modifications leading to selectivity against each enzyme were described. However, some of these trends could not be understood based on the reported binding mode of lead compound 1. Therefore, we employed microsecond molecular dynamics simulations and free energy calculations to understand qualitative SAR trends and to quantitatively recapitulate them. Simulations revealed the most stable protein-ligand interactions and provided insights concerning enzyme selectivity. Calculated relative binding free energies of compound 1 analogs exhibited deviations of 1.1 and 2.2 kcal/mol from the experimental values for cruzain and rhodesain, respectively. These data encourage prospective thermodynamic integration (TI) studies to optimize this series and facilitate the prioritization of compounds for synthesis.


Asunto(s)
Bencimidazoles/química , Inhibidores de Cisteína Proteinasa/química , Simulación del Acoplamiento Molecular , Trypanosoma brucei brucei/enzimología , Animales , Cisteína Endopeptidasas/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Relación Estructura-Actividad
10.
Proc Natl Acad Sci U S A ; 113(16): 4326-31, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27051065

RESUMEN

Nuclear factor kappa B (NF-κB) is an inducible transcription factor that plays critical roles in immune and stress responses and is often implicated in pathologies, including chronic inflammation and cancer. Although much has been learned about NF-κB-activating pathways, the specific repression of NF-κB is far less well understood. Here we identified the type I protein arginine methyltransferase 1 (PRMT1) as a restrictive factor controlling TNFα-induced activation of NF-κB. PRMT1 forms a cellular complex with NF-κB through direct interaction with the Rel homology domain of RelA. We demonstrate that PRMT1 methylates RelA at evolutionary conserved R30, located in the DNA-binding L1 loop, which is a critical residue required for DNA binding. Asymmetric R30 dimethylation inhibits the binding of RelA to DNA and represses NF-κB target genes in response to TNFα. Molecular dynamics simulations of the DNA-bound RelA:p50 predicted structural changes in RelA caused by R30 methylation or a mutation that interferes with the stability of the DNA-NF-κB complex. Our findings provide evidence for the asymmetric arginine dimethylation of RelA and unveil a unique mechanism controlling TNFα/NF-κB signaling.


Asunto(s)
Arginina/análogos & derivados , Transducción de Señal/fisiología , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Arginina/genética , Arginina/metabolismo , Línea Celular , Humanos , Metilación , Ratones , Ratones Noqueados , Simulación de Dinámica Molecular , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factor de Transcripción ReIA/genética , Factor de Necrosis Tumoral alfa/genética
11.
Chem Biodivers ; 16(5): e1800435, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30702795

RESUMEN

The small molecules that bind to DNA minor groove are considered as potential therapeutic agents to fight against many human diseases. They induce cell death by interfering with transcription, replication and progression of cell cycle. Herein, we report the synthesis of imidazopyridine-3-amines using sulfated ceria catalyst by employing Groebkee-Blackburne-Bienayme reaction. We evaluated the possible antiproliferative and antimycobacterial activity against A549 cells and Mycobacterium tuberculosis, respectively. Among the tested compounds, N-tert-butyl-2-(2-butyl-4-chloro-1H-imidazol-5-yl)-5,7-dimethylimidazo[1,2-a]pyridin-3-amine (4g) was identified as cytotoxic heterocycle and antimycobacterial agent. Molecular docking studies of the imidazopyridine derivatives revealed the consistent positioning in the minor groove with a tight shape fit between receptor and ligands. Therefore, we speculate that new imidazopyridines induce their pharmacological effect by targeting the minor groove of DNA.


Asunto(s)
Antituberculosos/síntesis química , Cerio/química , ADN/química , Imidazoles/química , Piridinas/química , Células A549 , Antituberculosos/química , Antituberculosos/farmacología , Sitios de Unión , Catálisis , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Ciclización , ADN/metabolismo , Humanos , Imidazoles/síntesis química , Imidazoles/farmacología , Simulación del Acoplamiento Molecular , Mycobacterium tuberculosis/efectos de los fármacos , Netropsina/química , Netropsina/metabolismo , Conformación de Ácido Nucleico , Piridinas/síntesis química , Piridinas/farmacología , Relación Estructura-Actividad , Sulfatos/química
12.
J Mol Recognit ; 31(10): e2727, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29785722

RESUMEN

Serine proteases of the Chymotrypsin family are structurally very similar but have very different substrate preferences. This study investigates a set of 9 different proteases of this family comprising proteases that prefer substrates containing positively charged amino acids, negatively charged amino acids, and uncharged amino acids with varying degree of specificity. Here, we show that differences in electrostatic substrate preferences can be predicted reliably by electrostatic molecular interaction fields employing customized GRID probes. Thus, we are able to directly link protease structures to their electrostatic substrate preferences. Additionally, we present a new metric that measures similarities in substrate preferences focusing only on electrostatics. It efficiently compares these electrostatic substrate preferences between different proteases. This new metric can be interpreted as the electrostatic part of our previously developed substrate similarity metric. Consequently, we suggest, that substrate recognition in terms of electrostatics and shape complementarity are rather orthogonal aspects of substrate recognition. This is in line with a 2-step mechanism of protein-protein recognition suggested in the literature.


Asunto(s)
Serina Proteasas/metabolismo , Sitios de Unión , Unión Proteica , Serina Proteasas/química , Electricidad Estática , Especificidad por Sustrato
13.
J Chem Inf Model ; 58(5): 982-992, 2018 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-29652495

RESUMEN

Macrocycles are of considerable interest as highly specific drug candidates, yet they challenge standard conformer generators with their large number of rotatable bonds and conformational restrictions. Here, we present a molecular dynamics-based routine that bypasses current limitations in conformational sampling and extensively profiles the free energy landscape of peptidic macrocycles in solution. We perform accelerated molecular dynamics simulations to capture a diverse conformational ensemble. By applying an energetic cutoff, followed by geometric clustering, we demonstrate the striking robustness and efficiency of the approach in identifying highly populated conformational states of cyclic peptides. The resulting structural and thermodynamic information is benchmarked against interproton distances from NMR experiments and conformational states identified by X-ray crystallography. Using three different model systems of varying size and flexibility, we show that the method reliably reproduces experimentally determined structural ensembles and is capable of identifying key conformational states that include the bioactive conformation. Thus, the described approach is a robust method to generate conformations of peptidic macrocycles and holds promise for structure-based drug design.


Asunto(s)
Compuestos Macrocíclicos/química , Simulación de Dinámica Molecular , Péptidos/química , Conformación Proteica , Termodinámica
14.
Mol Cell Proteomics ; 15(7): 2515-24, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27122596

RESUMEN

We present protease specificity profiling based on quantitative proteomics in combination with proteome-derived peptide libraries. Peptide libraries are generated by endoproteolytic digestion of proteomes without chemical modification of primary amines before exposure to a protease under investigation. After incubation with a test protease, treated and control libraries are differentially isotope-labeled using cost-effective reductive dimethylation. Upon analysis by liquid chromatography-tandem mass spectrometry, cleavage products of the test protease appear as semi-specific peptides that are enriched for the corresponding isotope label. We validate our workflow with two proteases with well-characterized specificity profiles: trypsin and caspase-3. We provide the first specificity profile of a protease encoded by a human endogenous retrovirus and for chlamydial protease-like activity factor (CPAF). For CPAF, we also highlight the structural basis of negative subsite cooperativity between subsites S1 and S2'. For A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) -4, -5, and -15, we show a canonical preference profile, including glutamate in P1 and glycine in P3'. In total, we report nearly 4000 cleavage sites for seven proteases. Our protocol is fast, avoids enrichment or synthesis steps, and enables probing for lysine selectivity as well as subsite cooperativity. Due to its simplicity, we anticipate usability by most proteomic laboratories.


Asunto(s)
Péptido Hidrolasas/metabolismo , Proteoma/análisis , Proteómica/métodos , Cromatografía Liquida , Humanos , Marcaje Isotópico , Biblioteca de Péptidos , Especificidad por Sustrato , Espectrometría de Masas en Tándem
15.
Biophys J ; 113(7): 1485-1495, 2017 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-28978442

RESUMEN

Mutations of positively charged amino acids in the S4 transmembrane segment of a voltage-gated ion channel form ion-conducting pathways through the voltage-sensing domain, named ω-current. Here, we used structure modeling and MD simulations to predict pathogenic ω-currents in CaV1.1 and CaV1.3 Ca2+ channels bearing several S4 charge mutations. Our modeling predicts that mutations of CaV1.1-R1 (R528H/G, R897S) or CaV1.1-R2 (R900S, R1239H) linked to hypokalemic periodic paralysis type 1 and of CaV1.3-R3 (R990H) identified in aldosterone-producing adenomas conducts ω-currents in resting state, but not during voltage-sensing domain activation. The mechanism responsible for the ω-current and its amplitude depend on the number of charges in S4, the position of the mutated S4 charge and countercharges, and the nature of the replacing amino acid. Functional characterization validates the modeling prediction showing that CaV1.3-R990H channels conduct ω-currents at hyperpolarizing potentials, but not upon membrane depolarization compared with wild-type channels.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Adenoma/genética , Adenoma/metabolismo , Animales , Canales de Calcio Tipo L/química , Canales de Calcio Tipo L/genética , Simulación por Computador , Células HEK293 , Humanos , Parálisis Periódica Hipopotasémica/genética , Parálisis Periódica Hipopotasémica/metabolismo , Potenciales de la Membrana/fisiología , Modelos Moleculares , Mutación , Dominios Proteicos , Estructura Secundaria de Proteína , Conejos , Homología Estructural de Proteína , Agua/química , Agua/metabolismo
16.
Biochim Biophys Acta ; 1858(11): 2846-2857, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27526680

RESUMEN

Cholesterol-phospholipid bilayers continue to be the current state of the art in membrane models and serve as representative systems for studying the effect of cholesterol on the cell membrane. As the mixing of different lipid species requires long spatio-temporal scales, coarse-grained models have gained increasing popularity in modeling such membrane systems. In this paper, a systematic study of the MARTINI coarse-grained model for the DPPC-cholesterol binary system has been performed. We construct the phase diagram of DPPC lipid bilayers in the presence of different cholesterol concentrations and at different temperatures using coarse-grained Molecular Dynamics (MD) simulations with the MARTINI force field. The phase diagram based on the condensation effect is directly comparable to available experimental data and demonstrates qualitative agreement over all cholesterol concentrations. Self-assembled bilayers quantitatively reproduce experimental observables, such as lateral diffusion of lipids, electron density, area per lipid and lipid order parameters. The phase diagram of the DPPC-cholesterol binary system also reveals the profound effect of cholesterol on the physical properties of phospholipid bilayers such lipid order, diffusion, and fluidity. Cholesterol induces the liquid-ordered phase, which increases the fluidity of the phospholipid hydrocarbon chains above the gel to liquid-crystalline phase transition temperature and decreases it below the phase transition. The present study suggests that the MARTINI force field can be successfully used to obtain molecular level insights into cholesterol-DPPC model membranes.


Asunto(s)
1,2-Dipalmitoilfosfatidilcolina/química , Colesterol/química , Membrana Dobles de Lípidos/química , Difusión , Cinética , Simulación de Dinámica Molecular , Transición de Fase , Temperatura , Termodinámica
17.
BMC Cancer ; 17(1): 235, 2017 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-28359266

RESUMEN

BACKGROUND: Expression and activity of heparanase, an endoglycosidase that cleaves heparan sulfate (HS) side chains of proteoglycans, is associated with progression and poor prognosis of many cancers which makes it an attractive drug target in cancer therapeutics. METHODS: In the present work, we report the in vitro screening of a library of 150 small molecules with the scaffold bearing quinolones, oxazines, benzoxazines, isoxazoli(di)nes, pyrimidinones, quinolines, benzoxazines, and 4-thiazolidinones, thiadiazolo[3,2-a]pyrimidin-5-one, 1,2,4-triazolo-1,3,4-thiadiazoles, and azaspiranes against the enzymatic activity of human heparanase. The identified lead compounds were evaluated for their heparanase-inhibiting activity using sulfate [35S] labeled extracellular matrix (ECM) deposited by cultured endothelial cells. Further, anti-invasive efficacy of lead compound was evaluated against hepatocellular carcinoma (HepG2) and Lewis lung carcinoma (LLC) cells. RESULTS: Among the 150 compounds screened, we identified 1,2,4-triazolo-1,3,4-thiadiazoles bearing compounds to possess human heparanase inhibitory activity. Further analysis revealed 2,4-Diiodo-6-(3-phenyl-[1, 2, 4]triazolo[3,4-b][1, 3, 4]thiadiazol-6yl)phenol (DTP) as the most potent inhibitor of heparanase enzymatic activity among the tested compounds. The inhibitory efficacy was demonstrated by a colorimetric assay and further validated by measuring the release of radioactive heparan sulfate degradation fragments from [35S] labeled extracellular matrix. Additionally, lead compound significantly suppressed migration and invasion of LLC and HepG2 cells with IC50 value of ~5 µM. Furthermore, molecular docking analysis revealed a favourable interaction of triazolo-thiadiazole backbone with Asn-224 and Asp-62 of the enzyme. CONCLUSIONS: Overall, we identified biologically active heparanase inhibitor which could serve as a lead structure in developing compounds that target heparanase in cancer.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Glucuronidasa/antagonistas & inhibidores , Neoplasias/enzimología , Neoplasias/patología , Tiadiazoles/química , Triazoles/química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/química , Humanos , Neoplasias/tratamiento farmacológico , Células Tumorales Cultivadas
18.
J Chem Inf Model ; 57(2): 345-354, 2017 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-28079371

RESUMEN

The anomalous binding modes of five highly similar fragments of TIE2 inhibitors, showing three distinct binding poses, are investigated. We report a quantitative rationalization for the changes in binding pose based on molecular dynamics simulations. We investigated five fragments in complex with the transforming growth factor ß receptor type 1 kinase domain. Analyses of these simulations using Grid Inhomogeneous Solvation Theory (GIST), pKA calculations, and a tool to investigate enthalpic differences upon binding unraveled the various thermodynamic contributions to the different binding modes. While one binding mode flip can be rationalized by steric repulsion, the second binding pose flip revealed a different protonation state for one of the ligands, leading to different enthalpic and entropic contributions to the binding free energy. One binding pose is stabilized by the displacement of entropically unfavored water molecules (binding pose determined by solvation entropy), ligands in the other binding pose are stabilized by strong enthalpic interactions, overcompensating the unfavorable water entropy in this pose (binding pose determined by enthalpic interactions). This analysis elucidates unprecedented details determining the flipping of the binding modes, which can elegantly explain the experimental findings for this system.


Asunto(s)
Entropía , Receptor TIE-2/metabolismo , Descubrimiento de Drogas , Simulación de Dinámica Molecular , Unión Proteica , Dominios Proteicos , Receptor TIE-2/antagonistas & inhibidores , Solventes/química , Agua/química
19.
J Allergy Clin Immunol ; 137(5): 1525-34, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26559323

RESUMEN

BACKGROUND: The search for intrinsic factors, which account for a protein's capability to act as an allergen, is ongoing. Fold stability has been identified as a molecular feature that affects processing and presentation, thereby influencing an antigen's immunologic properties. OBJECTIVE: We assessed how changes in fold stability modulate the immunogenicity and sensitization capacity of the major birch pollen allergen Bet v 1. METHODS: By exploiting an exhaustive virtual mutation screening, we generated mutants of the prototype allergen Bet v 1 with enhanced thermal and chemical stability and rigidity. Structural changes were analyzed by means of x-ray crystallography, nuclear magnetic resonance, and molecular dynamics simulations. Stability was monitored by using differential scanning calorimetry, circular dichroism, and Fourier transform infrared spectroscopy. Endolysosomal degradation was simulated in vitro by using the microsomal fraction of JAWS II cells, followed by liquid chromatography coupled to mass spectrometry. Immunologic properties were characterized in vitro by using a human T-cell line specific for the immunodominant epitope of Bet v 1 and in vivo in an adjuvant-free BALB/c mouse model. RESULTS: Fold stabilization of Bet v 1 was pH dependent and resulted in resistance to endosomal degradation at a pH of 5 or greater, affecting presentation of the immunodominant T-cell epitope in vitro. These properties translated in vivo into a strong allergy-promoting TH2-type immune response. Efficient TH2 cell activation required both an increased stability at the pH of the early endosome and efficient degradation at lower pH in the late endosomal/lysosomal compartment. CONCLUSIONS: Our data indicate that differential pH-dependent fold stability along endosomal maturation is an essential protein-inherent determinant of allergenicity.


Asunto(s)
Alérgenos/química , Antígenos de Plantas/química , Alérgenos/genética , Alérgenos/inmunología , Animales , Antígenos de Plantas/genética , Antígenos de Plantas/inmunología , Endosomas , Femenino , Concentración de Iones de Hidrógeno , Inmunoglobulina E/inmunología , Inmunoglobulina G/inmunología , Ratones Endogámicos BALB C , Mutación , Polen/inmunología , Pliegue de Proteína , Estabilidad Proteica
20.
Int J Mol Sci ; 18(6)2017 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-28587205

RESUMEN

The protein Bet v 1 represents the main cause for allergic reactions to birch pollen in Europe and North America. Structurally homologous isoforms of Bet v 1 can have different properties regarding allergic sensitization and Th2 polarization, most likely due to differential susceptibility to proteolytic cleavage. Using NMR relaxation experiments and molecular dynamics simulations, we demonstrate that the initial proteolytic cleavage sites in two naturally occurring Bet v 1 isoforms, Bet v 1.0101 (Bet v 1a) and Bet v 1.0102 (Bet v 1d), are conformationally flexible. Inaccessible cleavage sites in helices and strands are highly flexible on the microsecond-millisecond time scale, whereas those located in loops display faster nanosecond-microsecond flexibility. The data consistently show that Bet v 1.0102 is more flexible and conformationally heterogeneous than Bet v 1.0101. Moreover, NMR hydrogen-deuterium exchange measurements reveal that the backbone amides in Bet v 1.0102 are significantly more solvent exposed, in agreement with this isoform's higher susceptibility to proteolytic cleavage. The differential conformational flexibility of Bet v 1 isoforms, along with the transient exposure of inaccessible sites to the protein surface, may be linked to proteolytic susceptibility, representing a potential structure-based rationale for the observed differences in Th2 polarization and allergic sensitization.


Asunto(s)
Antígenos de Plantas/química , Modelos Moleculares , Proteínas de Plantas/química , Conformación Proteica , Simulación de Dinámica Molecular , Resonancia Magnética Nuclear Biomolecular , Isoformas de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA