Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
FASEB J ; 37(3): e22798, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36753401

RESUMEN

The precise physiological functions and mechanisms regulating RNase Regnase-2 (Reg-2/ZC3H12B/MCPIP2) activity remain enigmatic. We found that Reg-2 actively modulates neuroinflammation in nontransformed cells, including primary astrocytes. Downregulation of Reg-2 in these cells results in increased mRNA levels of proinflammatory cytokines IL-1ß and IL-6. In primary astrocytes, Reg-2 also regulates the mRNA level of Regnase-1 (Reg-1/ZC3H12A/MCPIP1). Reg-2 is expressed at high levels in the healthy brain, but its expression is reduced during neuroinflammation as well as glioblastoma progression. This process is associated with the upregulation of Reg-1. Conversely, overexpression of Reg-2 is accompanied by the downregulation of Reg-1 in glioma cells in a nucleolytic NYN/PIN domain-dependent manner. Interestingly, low levels of Reg-2 and high levels of Reg-1 correlate with poor-glioblastoma patients' prognoses. While Reg-2 restricts the basal levels of proinflammatory cytokines in resting astrocytes, its expression is reduced in IL-1ß-activated astrocytes. Following IL-1ß exposure, Reg-2 is phosphorylated, ubiquitinated, and degraded by proteasomes. Simultaneously, the Reg-2 transcript is destabilized by tristetraprolin (TTP) and Reg-1 through the AREs elements and conservative stem-loop structure present in its 3'UTR. Thus, the peer-control loop, of Reg-1 and Reg-2 opposing each other, exists. The involvement of TTP in Reg-2 mRNA turnover is confirmed by the observation that high TTP levels correlate with the downregulation of the Reg-2 expression in high-grade human gliomas. Additionally, obtained results reveal the importance of Reg-2 in inhibiting human and mouse glioma cell proliferation. Our current studies identify Reg-2 as a critical regulator of homeostasis in the brain.


Asunto(s)
Glioblastoma , Enfermedades Neuroinflamatorias , Animales , Humanos , Ratones , Citocinas/metabolismo , Regulación hacia Abajo , ARN Mensajero/genética , ARN Mensajero/metabolismo
2.
Cell Commun Signal ; 20(1): 10, 2022 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-35057808

RESUMEN

BACKGROUND: A universal adaptor protein, MyD88, orchestrates the innate immune response by propagating signals from toll-like receptors (TLRs) and interleukin-1 receptor (IL-1R). Receptor activation seeds MyD88 dependent formation of a signal amplifying supramolecular organizing center (SMOC)-the myddosome. Alternatively spliced variant MyD88S, lacking the intermediate domain (ID), exhibits a dominant negative effect silencing the immune response, but the mechanistic understanding is limited. METHODS: Luciferase reporter assay was used to evaluate functionality of MyD88 variants and mutants. The dimerization potential of MyD88 variants and myddosome nucleation process were monitored by co-immunoprecipitation and confocal microscopy. The ID secondary structure was characterized in silico employing I-TASSER server and in vitro using nuclear magnetic resonance (NMR) and circular dichroism (CD). RESULTS: We show that MyD88S is recruited to the nucleating SMOC and inhibits its maturation by interfering with incorporation of additional components. Biophysical analysis suggests that important functional role of ID is not supported by a well-defined secondary structure. Mutagenesis identifies Tyr116 as the only essential residue within ID required for myddosome nucleation and signal propagation (NF-κB activation). CONCLUSIONS: Our results argue that the largely unstructured ID of MyD88 is not only a linker separating toll-interleukin-1 receptor (TIR) homology domain and death domain (DD), but contributes intermolecular interactions pivotal in MyD88-dependent signaling. The dominant negative effect of MyD88S relies on quenching the myddosome nucleation and associated signal transduction. Video abstract.


Asunto(s)
Quinasas Asociadas a Receptores de Interleucina-1 , Factor 88 de Diferenciación Mieloide/metabolismo , Línea Celular , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/química , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Estructura Terciaria de Proteína , Receptores de Interleucina-1/química , Receptores de Interleucina-1/metabolismo , Receptores Toll-Like/metabolismo
3.
Int J Mol Sci ; 22(4)2021 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-33672341

RESUMEN

Accumulating evidence suggests that six proteases encoded in the spl operon of a dangerous human pathogen, Staphylococcus aureus, may play a role in virulence. Interestingly, SplA, B, D, and E have complementary substrate specificities while SplF remains to be characterized in this regard. Here, we describe the prerequisites of a heterologous expression system for active SplF protease and characterize the enzyme in terms of substrate specificity and its structural determinants. Substrate specificity of SplF is comprehensively profiled using combinatorial libraries of peptide substrates demonstrating strict preference for long aliphatic sidechains at the P1 subsite and significant selectivity for aromatic residues at P3. The crystal structure of SplF was provided at 1.7 Å resolution to define the structural basis of substrate specificity of SplF. The obtained results were compared and contrasted with the characteristics of other Spl proteases determined to date to conclude that the spl operon encodes a unique extracellular proteolytic system.


Asunto(s)
Péptido Hidrolasas/química , Péptido Hidrolasas/metabolismo , Staphylococcus aureus/enzimología , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , Escherichia coli/genética , Metionina/metabolismo , Modelos Moleculares , Péptido Hidrolasas/genética , Péptidos/química , Péptidos/metabolismo , Especificidad por Sustrato
4.
J Biol Chem ; 289(22): 15544-53, 2014 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-24713703

RESUMEN

Staphylococcal SplB protease belongs to the chymotrypsin family. Chymotrypsin zymogen is activated by proteolytic processing at the N terminus, resulting in significant structural rearrangement at the active site. Here, we demonstrate that the molecular mechanism of SplB protease activation differs significantly and we characterize the novel mechanism in detail. Using peptide and protein substrates we show that the native signal peptide, or any N-terminal extension, has an inhibitory effect on SplB. Only precise N-terminal processing releases the full proteolytic activity of the wild type analogously to chymotrypsin. However, comparison of the crystal structures of mature SplB and a zymogen mimic show no rearrangement at the active site whatsoever. Instead, only the formation of a unique hydrogen bond network, distant form the active site, by the new N-terminal glutamic acid of mature SplB is observed. The importance of this network and influence of particular hydrogen bond interactions at the N terminus on the catalytic process is demonstrated by evaluating the kinetics of a series of mutants. The results allow us to propose a consistent model where changes in the overall protein dynamics rather than structural rearrangement of the active site are involved in the activation process.


Asunto(s)
Serina Proteasas/química , Serina Proteasas/metabolismo , Staphylococcus aureus/enzimología , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Quimotripsina/química , Quimotripsina/genética , Quimotripsina/metabolismo , Cristalografía por Rayos X , Precursores Enzimáticos/metabolismo , Enlace de Hidrógeno , Modelos Moleculares , Señales de Clasificación de Proteína/fisiología , Estructura Terciaria de Proteína , Serina Proteasas/genética , Staphylococcus aureus/genética , Relación Estructura-Actividad
5.
Int J Biol Macromol ; 267(Pt 1): 131392, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38582483

RESUMEN

The main protease (Mpro) of SARS-CoV-2 is critical in the virus's replication cycle, facilitating the maturation of polyproteins into functional units. Due to its conservation across taxa, Mpro is a promising target for broad-spectrum antiviral drugs. Targeting Mpro with small molecule inhibitors, such as nirmatrelvir combined with ritonavir (Paxlovid™), which the FDA has approved for post-exposure treatment and prophylaxis, can effectively interrupt the replication process of the virus. A key aspect of Mpro's function is its ability to form a functional dimer. However, the mechanics of dimerization and its influence on proteolytic activity remain less understood. In this study, we utilized biochemical, structural, and molecular modelling approaches to explore Mpro dimerization. We evaluated critical residues, specifically Arg4 and Arg298, that are essential for dimerization. Our results show that changes in the oligomerization state of Mpro directly affect its enzymatic activity and dimerization propensity. We discovered a synergistic relationship influencing dimer formation, involving both intra- and intermolecular interactions. These findings highlight the potential for developing allosteric inhibitors targeting Mpro, offering promising new directions for therapeutic strategies.


Asunto(s)
Antivirales , Proteasas 3C de Coronavirus , Multimerización de Proteína , SARS-CoV-2 , SARS-CoV-2/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/química , Proteasas 3C de Coronavirus/metabolismo , Humanos , Antivirales/farmacología , Antivirales/química , Tratamiento Farmacológico de COVID-19 , Modelos Moleculares , COVID-19/virología , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/química
6.
ACS Chem Neurosci ; 15(17): 3181-3201, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39158934

RESUMEN

In the pathogenesis of Alzheimer's disease, the overexpression of glycogen synthase kinase-3ß (GSK-3ß) stands out due to its multifaced nature, as it contributes to the promotion of amyloid ß and tau protein accumulation, as well as neuroinflammatory processes. Therefore, in the present study, we have designed, synthesized, and evaluated a new series of GSK-3ß inhibitors based on the N-(pyridin-2-yl)cyclopropanecarboxamide scaffold. We identified compound 36, demonstrating an IC50 of 70 nM against GSK-3ß. Subsequently, through crystallography studies and quantum mechanical analysis, we elucidated its binding mode and identified the structural features crucial for interactions with the active site of GSK-3ß, thereby understanding its inhibitory potency. Compound 36 was effective in the cellular model of hyperphosphorylated tau-induced neurodegeneration, where it restored cell viability after okadaic acid treatment and showed anti-inflammatory activity in the LPS model, significantly reducing NO, IL-6, and TNF-α release. In ADME-tox in vitro studies, we confirmed the beneficial profile of 36, including high permeability in PAMPA (Pe equals 9.4) and high metabolic stability in HLMs as well as lack of significant interactions with isoforms of the CYP enzymes and lack of considerable cytotoxicity on selected cell lines (IC50 > 100 µM on HT-22 cells and 89.3 µM on BV-2 cells). Based on promising pharmacological activities and favorable ADME-tox properties, compound 36 may be considered a promising candidate for in vivo research as well as constitute a reliable starting point for further studies.


Asunto(s)
Antiinflamatorios , Glucógeno Sintasa Quinasa 3 beta , Fármacos Neuroprotectores , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/química , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/síntesis química , Antiinflamatorios/química , Humanos , Ratones , Supervivencia Celular/efectos de los fármacos , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , Proteínas tau/metabolismo
7.
Sci Rep ; 13(1): 18114, 2023 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-37872245

RESUMEN

The selective inhibition of kinases from the diabetic kinome is known to promote the regeneration of beta cells and provide an opportunity for the curative treatment of diabetes. The effect can be achieved by carefully tailoring the selectivity of inhibitor toward a particular kinase, especially DYRK1A, previously associated with Down syndrome and Alzheimer's disease. Recently DYRK1A inhibition has been shown to promote both insulin secretion and beta cells proliferation. Here, we show that commonly available flavones are effective inhibitors of DYRK1A. The observed biochemical activity of flavone compounds is confirmed by crystal structures solved at 2.06 Å and 2.32 Å resolution, deciphering the way inhibitors bind in the ATP-binding pocket of the kinase, which is driven by the arrangement of hydroxyl moieties. We also demonstrate antidiabetic properties of these biomolecules and prove that they could be further improved by therapy combined with TGF-ß inhibitors. Our data will allow future structure-based optimization of the presented scaffolds toward potent, bioavailable and selective anti-diabetic drugs.


Asunto(s)
Enfermedad de Alzheimer , Flavonas , Humanos , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Flavonas/farmacología , Flavonas/uso terapéutico , Enfermedad de Alzheimer/tratamiento farmacológico , Proliferación Celular , Inhibidores de Proteínas Quinasas/uso terapéutico
8.
J Med Chem ; 66(6): 4009-4024, 2023 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-36883902

RESUMEN

A clinical casein kinase 2 inhibitor, CX-4945 (silmitasertib), shows significant affinity toward the DYRK1A and GSK3ß kinases, involved in down syndrome phenotypes, Alzheimer's disease, circadian clock regulation, and diabetes. This off-target activity offers an opportunity for studying the effect of the DYRK1A/GSK3ß kinase system in disease biology and possible line extension. Motivated by the dual inhibition of these kinases, we solved and analyzed the crystal structures of DYRK1A and GSK3ß with CX-4945. We built a quantum-chemistry-based model to rationalize the compound affinity for CK2α, DYRK1A, and GSK3ß kinases. Our calculations identified a key element for CK2α's subnanomolar affinity to CX-4945. The methodology is expandable to other kinase selectivity modeling. We show that the inhibitor limits DYRK1A- and GSK3ß-mediated cyclin D1 phosphorylation and reduces kinase-mediated NFAT signaling in the cell. Given the CX-4945's clinical and pharmacological profile, this inhibitory activity makes it an interesting candidate with potential for application in additional disease areas.


Asunto(s)
Quinasa de la Caseína II , Naftiridinas , Glucógeno Sintasa Quinasa 3 beta , Naftiridinas/farmacología , Fenazinas , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/química
9.
Cells ; 10(9)2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34571911

RESUMEN

The rising prevalence of diabetes is threatening global health. It is known not only for the occurrence of severe complications but also for the SARS-Cov-2 pandemic, which shows that it exacerbates susceptibility to infections. Current therapies focus on artificially maintaining insulin homeostasis, and a durable cure has not yet been achieved. We demonstrate that our set of small molecule inhibitors of DYRK1A kinase potently promotes ß-cell proliferation, enhances long-term insulin secretion, and balances glucagon level in the organoid model of the human islets. Comparable activity is seen in INS-1E and MIN6 cells, in isolated mice islets, and human iPSC-derived ß-cells. Our compounds exert a significantly more pronounced effect compared to harmine, the best-documented molecule enhancing ß-cell proliferation. Using a body-like environment of the organoid, we provide a proof-of-concept that small-molecule-induced human ß-cell proliferation via DYRK1A inhibition is achievable, which lends a considerable promise for regenerative medicine in T1DM and T2DM treatment.


Asunto(s)
Homeostasis , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/enzimología , Insulina/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Genes Reporteros , Harmina/farmacología , Homeostasis/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Cinética , Masculino , Ratones , Modelos Biológicos , Factores de Transcripción NFATC/metabolismo , Organoides/efectos de los fármacos , Organoides/metabolismo , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Ratas , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo , Quinasas DyrK
10.
Biochem J ; 419(3): 555-64, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19175361

RESUMEN

Staphylococcus aureus is a dangerous human pathogen whose antibiotic resistance is steadily increasing and no efficient vaccine is as yet available. This serious threat drives extensive studies on staphylococcal physiology and pathogenicity pathways, especially virulence factors. Spl (serine protease-like) proteins encoded by an operon containing up to six genes are a good example of poorly characterized secreted proteins probably involved in virulence. In the present study, we describe an efficient heterologous expression system for SplA and detailed biochemical and structural characterization of the recombinant SplA protease. The enzyme shares a significant sequence homology to V8 protease and epidermolytic toxins which are well documented staphylococcal virulence factors. SplA has a very narrow substrate specificity apparently imposed by the precise recognition of three amino acid residues positioned N-terminal to the hydrolysed peptide bond. To explain determinants of this extended specificity we resolve the crystal structure of SplA and define the consensus model of substrate binding. Furthermore we demonstrate that artificial N-terminal elongation of mature SplA mimicking a naturally present signal peptide abolishes enzymatic activity. The probable physiological role of the process is discussed. Of interest, even though precise N-terminal trimming is a common regulatory mechanism among S1 family enzymes, the crystal structure of SplA reveals novel significantly different mechanistic details.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Serina Endopeptidasas/química , Serina Endopeptidasas/metabolismo , Staphylococcus aureus/enzimología , Secuencia de Aminoácidos , Animales , Aniones , Biocatálisis , Quimotripsina/química , Cristalografía por Rayos X , Histidina , Enlace de Hidrógeno , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Proteínas Recombinantes/biosíntesis , Especificidad por Sustrato
12.
Free Radic Biol Med ; 45(3): 321-8, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18466775

RESUMEN

Tgalphaq44 mice with targeted overexpression of activated Galphaq protein in cardiomyocytes mimic many of the phenotypic characteristics of dilated cardiomyopathy in humans. However, it is not known whether the phenotype of Tgalphaq44 mice would also involve dysfunction of cardiac mitochondria. The aim of the present work was to examine changes in EPR signals of semiquinones and iron in Fe-S clusters, as compared to classical biochemical indices of mitochondrial function in hearts from Tgalphaq44 mice in relation to the progression of heart failure. Tgalphaq44 mice at the age of 14 months displayed pulmonary congestion, increased heart/body ratio and impairment of cardiac function as measured in vivo by MRI. However, in hearts from Tgalphaq44 mice already at the age of 10 months EPR signals of semiquinones, as well as cyt c oxidase activity were decreased, suggesting alterations in mitochondrial electron flow. Furthermore, in 14-months old Tgalphaq44 mice loss of iron in Fe-S clusters, impaired citrate synthase activity, and altered mitochondrial ultrastructure were observed, supporting mitochondrial dysfunction in Tgalphaq44 mice. In conclusion, the assessment of semiquinones content and Fe(III) analysis by EPR represents a rational approach to detect dysfunction of cardiac mitochondria. Decreased contents of semiquinones detected by EPR and a parallel decrease in cyt c oxidase activity occurs before hemodynamic decompensation of heart failure in Tgalphaq44 mice suggesting that alterations in function of cardiac mitochondria contribute to the development of the overt heart failure in this model.


Asunto(s)
Cardiomiopatía Dilatada/patología , Espectroscopía de Resonancia por Spin del Electrón , Mitocondrias Cardíacas/química , Mitocondrias Cardíacas/patología , Animales , Cardiomiopatía Dilatada/metabolismo , Modelos Animales de Enfermedad , Hierro/análisis , Imagen por Resonancia Magnética , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Mitocondrias Cardíacas/metabolismo , Quinonas/análisis
13.
Int J Radiat Oncol Biol Phys ; 71(2): 542-9, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18474313

RESUMEN

PURPOSE: Tumor hypoxia has long been known to produce resistance to radiation. In this study, electron paramagnetic resonance (EPR) oxygen imaging was investigated for its power to predict the success of tumor control according to tumor oxygenation level and radiation dose. METHODS AND MATERIALS: A total of 34 EPR oxygen images were obtained from the legs of C3H mice bearing 0.5-cm(3) FSa fibrosarcomas under both normal (air breathing) and clamped tumor conditions. Under the same conditions as those during which the images were obtained, the tumors were irradiated to a variety of doses near the FSa dose at which 50% of tumors were cured. Tumor tissue was distinguished from normal tissue using co-registration of the EPR oxygen images with spin-echo magnetic resonance imaging of the tumor and/or stereotactic localization. The tumor voxel statistics in the EPR oxygen image included the mean and median partial pressure of oxygen and the fraction of tumor voxels below the specified partial pressure of oxygen values of 3, 6, and 10 mm Hg. Bivariate logistic regression analysis using the radiation dose and each of the EPR oxygen image statistics to determine which best separated treatment failure from success. RESULTS: The measurements of the dose at which 50% of tumors were cured were similar to those found in published data for this syngeneic tumor. Bivariate analysis of 34 tumors demonstrated that tumor cure correlated with dose (p = 0.004) and with a <10 mm Hg hypoxic fraction (p = 0.023). CONCLUSION: Our results have shown that, together, radiation dose and EPR image hypoxic fraction separate the population of FSa fibrosarcomas that are cured from those that fail, thus predicting curability.


Asunto(s)
Hipoxia de la Célula , Fibrosarcoma/metabolismo , Fibrosarcoma/radioterapia , Oxígeno/análisis , Tolerancia a Radiación/fisiología , Animales , Espectroscopía de Resonancia por Spin del Electrón , Femenino , Fibrosarcoma/fisiopatología , Ratones , Ratones Endogámicos C3H , Consumo de Oxígeno/fisiología , Presión Parcial , Radioterapia/métodos , Dosificación Radioterapéutica , Inducción de Remisión
14.
Cell Mol Biol Lett ; 13(2): 212-29, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18026858

RESUMEN

Proteases, also referred to as peptidases, are the enzymes that catalyse the hydrolysis of peptide bonds in polipeptides. A variety of biological functions and processes depend on their activity. Regardless of the organism's complexity, peptidases are essential at every stage of life of every individual cell, since all protein molecules produced must be proteolytically processed and eventually recycled. Protease inhibitors play a crucial role in the required strict and multilevel control of the activity of proteases involved in processes conditioning both the physiological and pathophysiological functioning of an organism, as well as in host-pathogen interactions. This review describes the regulation of activity of bacterial proteases produced by dangerous human pathogens, focusing on the Staphylococcus genus.


Asunto(s)
Péptido Hidrolasas/metabolismo , Staphylococcus/enzimología , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Péptido Hidrolasas/genética , Inhibidores de Proteasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Staphylococcus/efectos de los fármacos , Staphylococcus/genética , Transcripción Genética/efectos de los fármacos
16.
Acta Biochim Pol ; 61(1): 149-52, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24649483

RESUMEN

Staphylococcus aureus is a dangerous human pathogen characterized by growing antibiotic resistance. Virulence of S. aureus relies on a variety of secreted and cell surface associated virulence factors among which certain proteolytic enzymes play an important role. Amid staphylococcal extracellular proteases, those encoded by the spl operon remain poorly characterized, both in terms of enzymology and their physiological role. Initial data demonstrated that Spl proteases exhibit restricted substrate specificity. This study describes development of convenient protein FRET substrates for SplB protease and characterization of the substrate preference of the protease at the P1' position. Kinetic data on hydrolysis of a panel of substrates substituted at the said position is provided.


Asunto(s)
Proteínas Bacterianas/metabolismo , Péptido Hidrolasas/metabolismo , Serina Proteasas/metabolismo , Infecciones Estafilocócicas/enzimología , Staphylococcus aureus/enzimología , Secuencia de Aminoácidos , Humanos , Hidrólisis , Cinética , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/patología , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidad , Especificidad por Sustrato
17.
Protein Sci ; 23(2): 179-89, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24375505

RESUMEN

Staphylococcus aureus is responsible for a variety of human infections, including life-threatening, systemic conditions. Secreted proteome, including a range of proteases, constitutes the major virulence factor of the bacterium. However, the functions of individual enzymes, in particular SplA protease, remain poorly characterized. Here, we report development of specific inhibitors of SplA protease. The design, synthesis, and activity of a series of α-aminoalkylphosphonate diaryl esters and their peptidyl derivatives are described. Potent inhibitors of SplA are reported, which may facilitate future investigation of physiological function of the protease. The binding modes of the high-affinity compounds Cbz-Phe(P) -(OC6 H4 -4-SO2 CH3 )2 and Suc-Val-Pro-Phe(P) -(OC6 H5 )2 are revealed by high-resolution crystal structures of complexes with the protease. Surprisingly, the binding mode of both compounds deviates from previously characterized canonical interaction of α-aminoalkylphosphonate peptidyl derivatives and family S1 serine proteases.


Asunto(s)
Proteínas Bacterianas/química , Organofosfonatos/farmacología , Serina Proteasas/química , Inhibidores de Serina Proteinasa/farmacología , Staphylococcus aureus/enzimología , Cristalografía por Rayos X , Humanos , Cinética , Organofosfonatos/química , Unión Proteica/efectos de los fármacos , Inhibidores de Serina Proteinasa/química , Staphylococcus aureus/patogenicidad
18.
PLoS One ; 7(6): e37518, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22675482

RESUMEN

SJ-172550 (1) was previously discovered in a biochemical high throughput screen for inhibitors of the interaction of MDMX and p53 and characterized as a reversible inhibitor (J. Biol. Chem. 2010; 285:10786). Further study of the biochemical mode of action of 1 has shown that it acts through a complicated mechanism in which the compound forms a covalent but reversible complex with MDMX and locks MDMX into a conformation that is unable to bind p53. The relative stability of this complex is influenced by many factors including the reducing potential of the media, the presence of aggregates, and other factors that influence the conformational stability of the protein. This complex mechanism of action hinders the further development of compound 1 as a selective MDMX inhibitor.


Asunto(s)
Acetatos/farmacología , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Pirazoles/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Acetatos/química , Secuencia de Aminoácidos , Tampones (Química) , Proteínas de Ciclo Celular , Humanos , Concentración 50 Inhibidora , Modelos Biológicos , Datos de Secuencia Molecular , Proteínas Nucleares/química , Péptidos/metabolismo , Docilidad/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Conformación Proteica , Estabilidad Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas/química , Pirazoles/química , Temperatura , Proteína p53 Supresora de Tumor/química
19.
Cell Cycle ; 9(22): 4584-91, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21088494

RESUMEN

The negative regulation of p53, a major human tumor suppressor, by Mdm2 and Mdmx is crucial for the survival of a cell, whereas its aberrant function is a common feature of cancer.  Both Mdm proteins act through the spatial occlusion of the p53 transactivation (TA) domain and by the ubiquitination of p53, resulting in its degradation.  Two p53 homologues, p63 and p73, have been described in humans.  Unlike p53, these proteins regulate developmental processes rather than genome stability.  Both p63 and p73 contain TA domains homologous to that of p53, but relatively little is known about their regulation by Mdm2 or Mdmx.  Here, we present a detailed characterization of the interaction of Mdm2 and Mdmx with the TA domains of p63 and p73. Earlier reports of Mdm2 and Mdmx interactions with p73 are substantiated by the detailed quantitative characterization reported in this study. Most importantly, earlier contradictions concerning the presumed interaction of the Mdm proteins with p63 are convincingly resolved and for the first time, the affinities of these interactions are determined.  Finally, the contribution of these findings to our understanding of the physiological role of these interactions is discussed.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Ciclo Celular , Proteínas de Unión al ADN/química , Humanos , Proteínas Nucleares/química , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas c-mdm2/química , Termodinámica , Transactivadores/química , Factores de Transcripción , Triptófano/química , Proteína Tumoral p73 , Proteína p53 Supresora de Tumor/química , Proteínas Supresoras de Tumor/química
20.
J Mol Biol ; 379(2): 343-56, 2008 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-18448121

RESUMEN

Proteases are of significant importance for the virulence of Staphylococcus aureus. Nevertheless, their subset, the serine protease-like proteins, remains poorly characterized. Here presented is an investigation of SplB protease catalytic activity revealing that the enzyme possesses exquisite specificity and only cleaves efficiently after the sequence Trp-Glu-Leu-Gln. To understand the molecular basis for such selectivity, we solved the three-dimensional structure of SplB to 1.8 A. Modeling of substrate binding to the protease demonstrated that selectivity relies in part on a canonical specificity pockets-based mechanism. Significantly, the conformation of residues that ordinarily form the oxyanion hole, an essential structural element of the catalytic machinery of serine proteases, is not canonical in the SplB structure. We postulate that within SplB, the oxyanion hole is only formed upon docking of a substrate containing the consensus sequence motif. It is suggested that this unusual activation mechanism is used in parallel with classical determinants to further limit enzyme specificity. Finally, to guide future development, we attempt to point at likely physiological substrates and thus the role of SplB in staphylococcal physiology.


Asunto(s)
Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Serina Endopeptidasas/química , Serina Endopeptidasas/metabolismo , Staphylococcus aureus/enzimología , Animales , Proteínas Bacterianas/genética , Sitios de Unión , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Biblioteca de Péptidos , Péptidos/química , Péptidos/genética , Péptidos/metabolismo , Serina Endopeptidasas/genética , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA