Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 205
Filtrar
1.
J Antimicrob Chemother ; 77(6): 1625-1634, 2022 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-35245364

RESUMEN

BACKGROUND: The macrophage infectivity potentiator (Mip) protein, which belongs to the immunophilin superfamily, is a peptidyl-prolyl cis/trans isomerase (PPIase) enzyme. Mip has been shown to be important for virulence in a wide range of pathogenic microorganisms. It has previously been demonstrated that small-molecule compounds designed to target Mip from the Gram-negative bacterium Burkholderia pseudomallei bind at the site of enzymatic activity of the protein, inhibiting the in vitro activity of Mip. OBJECTIVES: In this study, co-crystallography experiments with recombinant B. pseudomallei Mip (BpMip) protein and Mip inhibitors, biochemical analysis and computational modelling were used to predict the efficacy of lead compounds for broad-spectrum activity against other pathogens. METHODS: Binding activity of three lead compounds targeting BpMip was verified using surface plasmon resonance spectroscopy. The determination of crystal structures of BpMip in complex with these compounds, together with molecular modelling and in vitro assays, was used to determine whether the compounds have broad-spectrum antimicrobial activity against pathogens. RESULTS: Of the three lead small-molecule compounds, two were effective in inhibiting the PPIase activity of Mip proteins from Neisseria meningitidis, Klebsiella pneumoniae and Leishmania major. The compounds also reduced the intracellular burden of these pathogens using in vitro cell infection assays. CONCLUSIONS: These results indicate that Mip is a novel antivirulence target that can be inhibited using small-molecule compounds that prove to be promising broad-spectrum drug candidates in vitro. Further optimization of compounds is required for in vivo evaluation and future clinical applications.


Asunto(s)
Proteínas Bacterianas , Bacterias Gramnegativas , Leishmania major , Isomerasa de Peptidilprolil , Proteínas Protozoarias , Proteínas Bacterianas/antagonistas & inhibidores , Bacterias Gramnegativas/efectos de los fármacos , Leishmania major/efectos de los fármacos , Macrófagos/metabolismo , Neisseria meningitidis , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Recombinantes
2.
Cell Rep ; 36(11): 109694, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34525372

RESUMEN

Chromatin organization plays a crucial role in tissue homeostasis. Heterochromatin relaxation and consequent unscheduled mobilization of transposable elements (TEs) are emerging as key contributors of aging and aging-related pathologies, including Alzheimer's disease (AD) and cancer. However, the mechanisms governing heterochromatin maintenance or its relaxation in pathological conditions remain poorly understood. Here we show that PIN1, the only phosphorylation-specific cis/trans prolyl isomerase, whose loss is associated with premature aging and AD, is essential to preserve heterochromatin. We demonstrate that this PIN1 function is conserved from Drosophila to humans and prevents TE mobilization-dependent neurodegeneration and cognitive defects. Mechanistically, PIN1 maintains nuclear type-B Lamin structure and anchoring function for heterochromatin protein 1α (HP1α). This mechanism prevents nuclear envelope alterations and heterochromatin relaxation under mechanical stress, which is a key contributor to aging-related pathologies.


Asunto(s)
Proteínas de Drosophila/metabolismo , Heterocromatina/metabolismo , Lamina Tipo B/metabolismo , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Isomerasa de Peptidilprolil/metabolismo , Estrés Mecánico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Células Cultivadas , Homólogo de la Proteína Chromobox 5/genética , Homólogo de la Proteína Chromobox 5/metabolismo , Elementos Transponibles de ADN/genética , Drosophila/metabolismo , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Humanos , Lamina Tipo B/química , Ratones , Ratones Endogámicos C57BL , Peptidilprolil Isomerasa de Interacción con NIMA/antagonistas & inhibidores , Peptidilprolil Isomerasa de Interacción con NIMA/genética , Neocórtex/citología , Neocórtex/metabolismo , Neuronas/citología , Neuronas/metabolismo , Membrana Nuclear/química , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Isomerasa de Peptidilprolil/genética , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
3.
Comput Biol Chem ; 89: 107398, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33059132

RESUMEN

Theileria annulata secretes peptidyl prolyl isomerase enzyme (TaPIN1) to manipulate the host cell oncogenic signaling pathway by disrupting the tumor suppressor F-box and WD repeat domain-containing 7 (FBW7) protein level leading to an increased level of c-Jun proto-oncogene. Buparvaquone is a hydroxynaphthoquinone anti-theilerial drug and has been used to treat theileriosis. However, TaPIN1 contains the A53 P mutation that causes drug resistance. In this study, potential TaPIN1 inhibitors were investigated using a library of naphthoquinone derivatives. Comparative models of mutant (m) and wild type (wt) TaPIN1 were predicted and energy minimization was followed by structure validation. A naphthoquinone (hydroxynaphthalene-1,2-dione, hydroxynaphthalene-1,4-dione) and hydroxynaphthalene-2,3-dione library was screened by Schrödinger Glide HTVS, SP and XP docking methodologies and the docked compounds were ranked by the Glide XP scoring function. The two highest ranked docked compounds Compound 1 (4-hydroxy-3-[3,4,5-trihydroxy-6-(hydroxymethyl)oxan-2-yl]oxynaphthalene-1,2-dione) and Compound 2 (6-acetyl-1,4,5,7,8-pentahydroxynaphthalene-2,3-dione) were used for further molecular dynamics (MD) simulation studies. The MD results showed that ligand Compound 1 was located in the active site of both mTaPIN1 and wtTaPIN1 and could be proposed as a potential inhibitor by acting as a substrate antagonist. However, ligand Compound 2 was displaced away from the binding pocket of wtTaPIN1 but was located near the active site binding pocket of mTaPIN1 suggesting that could be selectively evaluated as a potential inhibitor against the mTaPIN1. Compound 1 and Compound 2 ligands are potential inhibitors but Compound 2 is suggested as a better inhibitor for mTaPIN1. These ligands could also further evaluated as potential inhibitors against human peptidyl prolyl isomerase which causes cancer in humans by using the same mechanism as TaPIN1.


Asunto(s)
Inhibidores Enzimáticos/química , Naftoquinonas/química , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Proteínas Protozoarias/antagonistas & inhibidores , Theileria annulata/enzimología , Dominio Catalítico , Bases de Datos de Compuestos Químicos/estadística & datos numéricos , Inhibidores Enzimáticos/metabolismo , Ensayos Analíticos de Alto Rendimiento , Ligandos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Mutación , Naftoquinonas/metabolismo , Isomerasa de Peptidilprolil/química , Isomerasa de Peptidilprolil/genética , Isomerasa de Peptidilprolil/metabolismo , Unión Proteica , Proto-Oncogenes Mas , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
4.
Cells ; 9(7)2020 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-32708451

RESUMEN

The secretome is an important mediator in the permanent process of reciprocity between cells and their environment. Components of secretome are involved in a large number of physiological mechanisms including differentiation, migration, and extracellular matrix modulation. Alteration in secretome composition may therefore trigger cell transformation, inflammation, and diseases. In the kidney, aberrant protein secretion plays a central role in cell activation and transition and in promoting renal fibrosis onset and progression. Using comparative proteomic analyses, we investigated in the present study the impact of cell transition on renal fibroblast cells secretome. Human renal cell lines were stimulated with profibrotic hormones and cytokines, and alterations in secretome were investigated using proteomic approaches. We identified protein signatures specific for the fibrotic phenotype and investigated the impact of modeling secretome proteins on extra cellular matrix accumulation. The secretion of peptidyl-prolyl cis-trans isomerase A (PPIA) was demonstrated to be associated with fibrosis phenotype. We showed that the in-vitro inhibition of PPIA with ciclosporin A (CsA) resulted in downregulation of PPIA and fibronectin (FN1) expression and significantly reduced their secretion. Knockdown studies of PPIA in a three-dimensional (3D) cell culture model significantly impaired the secretion and accumulation of the extracellular matrix (ECM), suggesting a positive therapeutic effect on renal fibrosis progression.


Asunto(s)
Fibroblastos/enzimología , Fibroblastos/patología , Riñón/patología , Isomerasa de Peptidilprolil/metabolismo , Proteoma/metabolismo , Línea Celular Transformada , Supervivencia Celular/efectos de los fármacos , Ciclosporina/farmacología , Progresión de la Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibroblastos/efectos de los fármacos , Fibronectinas/metabolismo , Fibrosis , Humanos , Inflamación/patología , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Fenotipo , ARN Interferente Pequeño/metabolismo , Tacrolimus/farmacología
5.
Bioorg Med Chem Lett ; 28(22): 3540-3548, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30301675

RESUMEN

SurA is a gram-negative, periplasmic chaperone protein involved in the proper folding of outer membrane porins (OMPs), which protect bacteria against toxins in the extracellular environment by selectively regulating the passage of nutrients into the cell. Previous studies demonstrated that deletion of SurA renders bacteria more sensitive to toxins that compromise the integrity of the outer membrane. Inhibitors of SurA will perturb the folding of OMPs, leading to disruption of the outer membrane barrier and making the cell more vulnerable to toxic insults. The discovery of novel SurA inhibitors is therefore of great importance for developing alternative strategies to overcome antibiotic resistance. Our laboratory has screened over 10,000,000 compoundsin silicoby computationally docking these compounds onto the crystal structure of SurA. Through this screen and a screen of fragment compounds (molecular weight less than 250 g/mol), we found twelve commercially readily available candidate compounds that bind to the putative client binding site of SurA. We confirmed binding to SurA by developing and employing a competitive fluorescence anisotropy-based binding assay. Our results show that one of these compounds, Fmoc-ß-(2-quinolyl)-d-alanine, binds the client binding site with high micromolar affinity. Using this compound as a lead, we also discovered that Fmoc-l-tryptophan and Fmoc-l-phenylalanine, but not Fmoc-l-tyrosine, bind SurA with similar micromolar affinity. To our knowledge, this is the first report of a competitive fluorescence anisotropy assay developed for the identification of inhibitors of the chaperone SurA, and the identification of three small molecules that bind SurA at its client binding site.


Asunto(s)
Proteínas Portadoras/antagonistas & inhibidores , Proteínas de Escherichia coli/antagonistas & inhibidores , Escherichia coli/metabolismo , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Alanina/análogos & derivados , Alanina/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Proteínas Portadoras/metabolismo , Proteínas de Escherichia coli/metabolismo , Polarización de Fluorescencia , Simulación del Acoplamiento Molecular , Péptidos/química , Péptidos/metabolismo , Isomerasa de Peptidilprolil/metabolismo , Estructura Terciaria de Proteína
6.
Cell Signal ; 52: 147-154, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30213686

RESUMEN

Extracellular signal-regulated kinase (ERK), also known as classical mitogen-activated protein kinase, plays critical roles in cell regulation. ERK is activated through phosphorylation by a cascade of protein kinases including MEK. Various ligands activate the MEK/ERK pathway through receptor-dependent cell signaling. In cultured cells, many ligands such as growth factors, hormones, cytokines and vasoactive peptides elicit transient activation of MEK/ERK, often peaking at ~10 min after the cell treatment. Here, we describe a novel biological event, in which ligand-mediated cell signaling results in the dephosphorylation of MEK/ERK. Neuromedin N and neurotensin, peptides derived from the same precursor polypeptide, elicit cell signaling through the neurotensin receptors. In cultured human pulmonary artery smooth muscle cells (PASMCs), but not in human pulmonary artery endothelial cells (PAECs), we found that both neuromedin N and neurotensin promoted the dephosphorylation of ERK and MEK. Human PASMCs were found to express neurotensin receptor (NTR)-1, -2 and -3, while human PAECs only express NTR3. Neuromedin N-mediated dephosphorylation was suppressed by small chemical inhibitors of protein phosphatase 1/2A and peptidyl-prolyl isomerase. Transmission electron microscopy showed the formation of endocytic vesicles in response to neuromedin N treatment, and dephosphorylation did not occur when sorting nexin 9, a critical regulator of the endocytic vesicle formation, was knocked down. We conclude that neuromedin N and neurotensin elicit a unique dephosphorylation signaling in the MEK/ERK pathway that is regulated by endocytosis. Considering the pathophysiological importance of the MEK/ERK pathway, this discovery of the dephosphorylation mechanism should advance the field of cell signaling.


Asunto(s)
Células Endoteliales/metabolismo , Miocitos del Músculo Liso/metabolismo , Neurotensina/fisiología , Fragmentos de Péptidos/fisiología , Arteria Pulmonar/metabolismo , Células Endoteliales/citología , Humanos , Péptidos y Proteínas de Señalización Intracelular/farmacología , Sistema de Señalización de MAP Quinasas , Miocitos del Músculo Liso/citología , Proteínas Nucleares , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Nexinas de Proteasas/metabolismo , Arteria Pulmonar/citología , Proteínas de Unión al ARN , Receptores de Neurotensina/metabolismo
7.
Virology ; 522: 46-55, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30014857

RESUMEN

Cyclophilins (Cyps) belong to the family of peptidyl-prolyl isomerases (PPIases). The PPIase activity of most Cyps is inhibited by the immunosuppressive drug cyclosporin A and several of its non-immunosuppressive analogs, which can also block the replication of nidoviruses (arteriviruses and coronaviruses). Cyclophilins have been reported to play an essential role in the replication of several other RNA viruses, including human immunodeficiency virus-1, hepatitis C virus, and influenza A virus. Likewise, the replication of various nidoviruses was reported to depend on Cyps or other PPIases. This review summarizes our current understanding of this class of nidovirus-host interactions, including the potential function of in particular CypA and the inhibitory effect of Cyp inhibitors. Also the involvement of the FK-506-binding proteins and parvulins is discussed. The nidovirus data are placed in a broader perspective by summarizing the most relevant data on Cyp interactions and Cyp inhibitors for other RNA viruses.


Asunto(s)
Ciclofilinas/antagonistas & inhibidores , Ciclofilinas/metabolismo , Interacciones Huésped-Patógeno , Nidovirales/fisiología , Replicación Viral , Animales , Humanos , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Isomerasa de Peptidilprolil/metabolismo
8.
Oxid Med Cell Longev ; 2018: 1714896, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29849865

RESUMEN

BACKGROUND AND AIM: Upregulation of prolyl isomerase-1 (Pin1) protein expression and activity was associated with the pathogenesis of diabetic vasculopathy through induction of endothelial oxidative stress and inflammation. Moreover, VDR agonist protects against high glucose-induced endothelial apoptosis through the inhibition of oxidative stress. We aimed to explore the effects of the VDR agonist on diabetes-associated endothelial dysfunction and the role of Pin1 in this process. METHODS: Streptozocin-induced diabetic mice were randomly treated with vehicle, VDR agonist (10 µg/kg/d, i.g., twice a week), or Pin1 inhibitor, Juglone (1 mg/kg/d, i.p., every other day), for eight weeks. In parallel, human umbilical vein endothelial cells (HUVECs) exposed to high-glucose condition were treated with 1,25-dihydroxyvitamin D3 and Juglone or vehicle for 72 hours. Organ chamber experiments were performed to assess endothelium-dependent relaxation to acetylcholine. Circulatory levels of Pin1, SOD, MDA, IL-1ß, IL-6, and NO in diabetic mice, Pin1 protein expression and activity, subcellular distribution of p66Shc, and NF-κB p65 in high glucose-cultured HUVECs were determined. RESULTS: Both VDR agonist and Juglone significantly improved diabetes-associated endothelial dysfunction and reduced high glucose-induced endothelial apoptosis. Mechanistically, the circulatory levels of SOD and NO were increased compared with those of vehicle-treated diabetic mice. Additionally, Pin1 protein expression and activity, p66Shc mitochondrial translocation, and NF-κB p65 in high glucose-cultured HUVECs were also inhibited by VDR agonist and Juglone. Knockdown of VDR abolished the inhibitory effects of VDR agonist on high glucose-induced upregulation of Pin1 protein expression and activity. CONCLUSIONS: VDR agonist prevents diabetic endothelial dysfunction through inhibition of Pin1-mediated mitochondrial oxidative stress and inflammation.


Asunto(s)
Endotelio Vascular/patología , Inflamación/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Receptores de Calcitriol/uso terapéutico , Animales , Diabetes Mellitus Experimental/patología , Modelos Animales de Enfermedad , Humanos , Inflamación/patología , Masculino , Ratones , Transfección
9.
Protein J ; 37(3): 270-279, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29761378

RESUMEN

SIB1 FKBP22 is a peptidyl prolyl cis-trans isomerase (PPIase) member from a psychrotrophic bacterium, Shewanella sp. SIB1, consisting of N- and C-domains responsible for dimerization and catalytic PPIase activity, respectively. This protein was assumed to be involved in cold adaptation of SIB1 cells through its dual activity of PPIase activity and chaperone like-function. Nevertheless, the catalytic inhibition by FK506 and its substrate specificity remain unknown. Besides, ability of SIB1 FKBP22 to inhibit phosphatase activity of calcinuerin is also interesting to be studied since it may reflect wider cellular functions of SIB1 FKBP22. In this study, we found that wild type (WT) SIB1 FKBP22 bound to FK506 with IC50 of 77.55 nM. This value is comparable to that of monomeric mutants (NNC-FKBP22, C-domain+ and V37R/L41R mutants), yet significantly higher than that of active site mutant (R142A). In addition, WT SIB1 FKBP22 and monomeric variants were found to prefer hydrophobic residues preceding proline. Meanwhile, R142A mutant has wider preferences on bulkier hydrophobic residues due to increasing hydrophobicity and binding pocket space. Surprisingly, in the absence of FK506, SIB1 FKBP22 and its variants inhibited, with the exception of N-domain, calcineurin phosphatase activity, albeit low. The inhibition of SIB1 FKBP22 by FK506 is dramatically increased in the presence of FK506. Altogether, we proposed that local structure at substrate binding pocket of C-domain plays crucial role for the binding of FK506 and peptide substrate preferences. In addition, C-domain is essential for inhibition, while dimerization state is important for optimum inhibition through efficient binding to calcineurin.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/síntesis química , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Isomerasa de Peptidilprolil/química , Shewanella/enzimología , Tacrolimus/farmacología , Dominios Proteicos , Especificidad por Sustrato
10.
Eur Rev Med Pharmacol Sci ; 22(1): 79-86, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29364473

RESUMEN

OBJECTIVE: MicroRNAs have been reported to contribute to the development of osteosarcoma via negatively regulating the target genes. Nevertheless, the undiscovered function of miR-361 on osteosarcoma development remains uncertain. PATIENTS AND METHODS: MiR-361 and FKBP14 (FK506-binding protein 14) expression in osteosarcoma samples were detected by Real-time polymerase chain reaction (PCR). Cells invasive ability was examined via the transwell invasion assay. The luciferase reporter assay was used to examine the regulation mechanism. The protein level of FKBP14 was detected by Western blot. Cell Counting Kit-8 (CCK-8) was used to detect cell lines proliferation. RESULTS: MiR-361 was reduced both in osteosarcoma samples and cell lines. Up-regulation of miR-361 significantly inhibited cells invasive and proliferative abilities, while down-regulation of miR-361 promoted cell lines invasion and proliferation. miR-361 could negatively regulate FKBP14 in osteosarcoma. Suppression of FKBP14 could reverse the function of miR-361 inhibitor. CONCLUSIONS: MiR-361 inhibits osteosarcoma cell lines invasion and proliferation by targeting FKBP14.


Asunto(s)
Neoplasias Óseas/patología , MicroARNs/metabolismo , Osteosarcoma/patología , Isomerasa de Peptidilprolil/metabolismo , Regiones no Traducidas 3' , Antagomirs/metabolismo , Neoplasias Óseas/genética , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Femenino , Humanos , Masculino , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Persona de Mediana Edad , Osteosarcoma/genética , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Isomerasa de Peptidilprolil/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Regulación hacia Arriba
11.
Protein Expr Purif ; 137: 64-76, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28668496

RESUMEN

The gene for a protein domain, derived from a tumor marker, fused to His tag codons and under control of a T7 promotor was expressed in E. coli strain BL21 (DE3). The recombinant protein was purified from cell lysates through immobilized metal affinity chromatography and size-exclusion chromatography. A contaminating bacterial protein was consistently co-purified, even using stringent washing solutions containing 50 or 100 mM imidazole. Immunization of a dromedary with this contaminated protein preparation, and the subsequent generation and panning of the immune Nanobody library yielded several Nanobodies of which 2/3 were directed against the bacterial contaminant, reflecting the immunodominance of this protein to steer the dromedary immune response. Affinity adsorption of this contaminant using one of our specific Nanobodies followed by mass spectrometry identified the bacterial contaminant as FKBP-type peptidyl-prolyl cis-trans isomerase (SlyD) from E. coli. This SlyD protein contains in its C-terminal region 14 histidines in a stretch of 31 amino acids, which explains its co-purification on Ni-NTA resin. This protein is most likely present to varying extents in all recombinant protein preparations after immobilized metal affinity chromatography. Using our SlyD-specific Nb 5 we generated an immune-complex that could be removed either by immunocapturing or by size exclusion chromatography. Both methods allow us to prepare a recombinant protein sample where the SlyD contaminant was quantitatively eliminated.


Asunto(s)
Proteínas de Escherichia coli/química , Escherichia coli , Isomerasa de Peptidilprolil/química , Anticuerpos de Dominio Único , Animales , Camelus , Escherichia coli/química , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/antagonistas & inhibidores , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/aislamiento & purificación
12.
Sci Rep ; 7: 42141, 2017 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-28176839

RESUMEN

Molecular chaperones are essential molecules for cell growth, whereby they maintain protein homeostasis. Because of their central cellular function, bacterial chaperones might be potential candidates for drug targets. Antimicrobial resistance is currently one of the greatest threats to human health, with gram-negative bacteria being of major concern. We found that a Cu2+ complex readily crosses the bacterial cell wall and inhibits SlyD, which is a molecular chaperone, cis/trans peptidyl prolyl isomerise (PPIase) and involved in various other metabolic pathways. The Cu2+ complex binds to the active sites of SlyD, which suppresses its PPIase and chaperone activities. Significant cell growth retardation could be observed for pathogenic bacteria (e.g., Staphylococcus aureus and Pseudomonas aeruginosa). We anticipate that rational development of drugs targeting molecular chaperones might help in future control of pathogenic bacterial growth, in an era of rapidly increasing antibiotic resistance.


Asunto(s)
Antracenos/química , Antibacterianos/farmacología , Complejos de Coordinación/farmacología , Cobre/química , Proteínas de Escherichia coli/antagonistas & inhibidores , Escherichia coli/efectos de los fármacos , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Piridinas/química , Antibacterianos/síntesis química , Sitios de Unión , Complejos de Coordinación/síntesis química , Sistemas de Liberación de Medicamentos , Farmacorresistencia Bacteriana/efectos de los fármacos , Escherichia coli/enzimología , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Expresión Génica , Pruebas de Sensibilidad Microbiana , Isomerasa de Peptidilprolil/química , Isomerasa de Peptidilprolil/genética , Isomerasa de Peptidilprolil/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/enzimología , Pseudomonas aeruginosa/genética , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/enzimología , Staphylococcus aureus/genética
13.
PLoS One ; 11(12): e0168830, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28036348

RESUMEN

BACKGROUND: While screening for an inhibitor of the peptidyl prolyl cis/trans isomerase, Pin1, we came across a brown algae polyphenol that blocks the differentiation of fibroblasts into adipocytes. However, its effectiveness on the accumulation of fat in the body has never been studied. METHODOLOGY/PRINCIPAL FINDINGS: Oral administration of brown algae polyphenol to mice fed with a high fat diet, suppressed the increase in fat volume to a level observed in mice fed with a normal diet. We speculate that Pin1 might be required for the differentiation of stem cell to adipocytes. We established wild type (WT) and Pin1-/- (Pin1-KO) adipose-derived mesenchymal stem cell (ASC) lines and found that WT ASCs differentiate to adipocytes but Pin1-KO ASCs do not. CONCLUSION AND SIGNIFICANCE: Oral administration of brown algae polyphenol, a Pin1 inhibitor, reduced fat buildup in mice. We showed that Pin1 is required for the differentiation of stem cells into adipocytes. We propose that oral intake of brown algae polyphenol is useful for the treatment of obesity.


Asunto(s)
Adipocitos/efectos de los fármacos , Peptidilprolil Isomerasa de Interacción con NIMA/antagonistas & inhibidores , Obesidad/tratamiento farmacológico , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Phaeophyceae/química , Polifenoles/farmacología , Células Madre/efectos de los fármacos , Adiposidad/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Fibroblastos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL
14.
Bioorg Med Chem ; 24(21): 5134-5147, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27591009

RESUMEN

The bacteria Burkholderia pseudomallei and Legionella pneumophila cause severe diseases like melioidosis and Legionnaire's disease with high mortality rates despite antibiotic treatment. Due to increasing antibiotic resistances against these and other Gram-negative bacteria, alternative therapeutical strategies are in urgent demand. As a virulence factor, the macrophage infectivity potentiator (Mip) protein constitutes an attractive target. The Mip proteins of B. pseudomallei and L. pneumophila exhibit peptidyl-prolyl cis/trans isomerase (PPIase) activity and belong to the PPIase superfamily. In previous studies, the pipecolic acid moiety proved to be a valuable scaffold for inhibiting this PPIase activity. Thus, a library of pipecolic acid derivatives was established guided by structural information and computational analyses of the binding site and possible binding modes. Stability and toxicity considerations were taken into account in iterative extensions of the library. Synthesis and evaluation of the compounds in PPIase assays resulted in highly active inhibitors. The activities can be interpreted in terms of a common binding mode obtained by docking calculations.


Asunto(s)
Burkholderia pseudomallei/enzimología , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Legionella pneumophila/enzimología , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Simulación del Acoplamiento Molecular , Estructura Molecular , Isomerasa de Peptidilprolil/metabolismo , Relación Estructura-Actividad
15.
J Med Chem ; 59(21): 9622-9644, 2016 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-27409354

RESUMEN

Peptidyl-proline isomerases (PPIases) are a chaperone superfamily comprising the FK506-binding proteins (FKBPs), cyclophilins, and parvulins. PPIases catalyze the cis/trans isomerization of proline, acting as a regulatory switch during folding, activation, and/or degradation of many proteins. These "clients" include proteins with key roles in cancer, neurodegeneration, and psychiatric disorders, suggesting that PPIase inhibitors could be important therapeutics. However, the active site of PPIases is shallow, solvent-exposed, and well conserved between family members, making selective inhibitor design challenging. Despite these hurdles, macrocyclic natural products, including FK506, rapamycin, and cyclosporin, bind PPIases with nanomolar or better affinity. De novo attempts to derive new classes of inhibitors have been somewhat less successful, often showcasing the "undruggable" features of PPIases. Interestingly, the most potent of these next-generation molecules tend to integrate features of the natural products, including macrocyclization or proline mimicry strategies. Here, we review recent developments and ongoing challenges in the inhibition of PPIases, with a focus on how natural products might inform the creation of potent and selective inhibitors.


Asunto(s)
Productos Biológicos/farmacología , Ciclosporina/farmacología , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Sirolimus/farmacología , Tacrolimus/farmacología , Productos Biológicos/química , Ciclosporina/química , Humanos , Modelos Moleculares , Conformación Molecular , Isomerasa de Peptidilprolil/metabolismo , Pliegue de Proteína/efectos de los fármacos , Sirolimus/química , Relación Estructura-Actividad , Tacrolimus/química
16.
J Biomol Screen ; 21(7): 701-12, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27185744

RESUMEN

In this work, a sensitive and convenient protease-based fluorimetric high-throughput screening (HTS) assay for determining peptidyl-prolyl cis-trans isomerase activity was developed. The assay was based on a new intramolecularly quenched substrate, whose fluorescence and structural properties were examined together with kinetic constants and the effects of solvents on its isomerization process. Pilot screens performed using the Library of Pharmacologically Active Compounds (LOPAC) and cyclophilin A (CypA), as isomerase model enzyme, indicated that the assay was robust for HTS, and that comparable results were obtained with a CypA inhibitor tested both manually and automatically. Moreover, a new compound that inhibits CypA activity with an IC50 in the low micromolar range was identified. Molecular docking studies revealed that the molecule shows a notable shape complementarity with the catalytic pocket confirming the experimental observations. Due to its simplicity and precision in the determination of extent of inhibition and reaction rates required for kinetic analysis, this assay offers many advantages over other commonly used assays.


Asunto(s)
Ciclofilina A/antagonistas & inhibidores , Inhibidores Enzimáticos/aislamiento & purificación , Ensayos Analíticos de Alto Rendimiento/métodos , Isomerasa de Peptidilprolil/química , Dominio Catalítico , Ciclofilina A/química , Inhibidores Enzimáticos/química , Transferencia Resonante de Energía de Fluorescencia/métodos , Humanos , Cinética , Espectroscopía de Resonancia Magnética/métodos , Simulación del Acoplamiento Molecular/métodos , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Isomerasa de Peptidilprolil/farmacología , Solventes/química , Especificidad por Sustrato
17.
Int J Cardiol ; 203: 702-7, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26583846

RESUMEN

BACKGROUND: Atherosclerosis is a major cause of mortality in patients with diabetes. However, novel breakthrough therapies have yet to be approved in this setting. Prolyl-isomerase-1 (Pin1) is emerging as a key molecule implicated in vascular oxidative stress and inflammation. In the present study, we investigate whether pharmacological inhibition of Pin1 may protect against diabetes-induced oxidative stress, endothelial dysfunction and vascular inflammation. METHODS AND RESULTS: Experiments were performed in human aortic endothelial cells (HAECs) exposed to normal (5 mmol/L) or high glucose (25 mmol/L) concentrations, in the presence of Pin1 inhibitor Juglone (10 µM) or vehicle (<1% ethanol). In parallel, streptozotocin-induced diabetic mice were treated with Juglone i.p. every other day for 30 days (1mg/Kg). Organ chamber experiments were performed in aortic rings to assess endothelium-dependent relaxations to acetylcholine (Ach 10(-9) to 10(-6)mol/L). Mitochondrial oxidative stress, organelle integrity as well as NF-kB-dependent inflammatory signatures were determined both in HAECs and aortas from diabetic mice. In HAECs, ambient hyperglycemia increased mitochondrial superoxide anion generation while treatment with Juglone prevented this phenomenon. Pharmacological inhibition of Pin1 also preserved mitochondrial integrity, nitric oxide availability and endothelial expression of adhesion molecules. Interestingly enough, endothelial dysfunction, oxidative stress and NF-kB-driven inflammation were significantly attenuated in diabetic mice chronically treated with Juglone as compared to vehicle-treated animals. CONCLUSION: Pharmacological inhibition of Pin1 by Juglone prevents hyperglycemia-induced vascular dysfunction. Taken together, our findings may set the stage for novel therapeutic approaches to prevent vascular complications in patients with diabetes.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Angiopatías Diabéticas/prevención & control , Endotelio Vascular/fisiopatología , Naftoquinonas/farmacología , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Vasodilatación/efectos de los fármacos , Animales , Células Cultivadas , ADN/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Angiopatías Diabéticas/etiología , Angiopatías Diabéticas/genética , Endotelio Vascular/patología , Inhibidores Enzimáticos/farmacología , Humanos , Inmunohistoquímica , Masculino , Ratones , Mitocondrias Musculares/genética , Mitocondrias Musculares/metabolismo , Peptidilprolil Isomerasa de Interacción con NIMA , Estrés Oxidativo/genética , Isomerasa de Peptidilprolil/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
18.
Oxid Med Cell Longev ; 2016: 4528906, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26697133

RESUMEN

Long-term heavy alcohol consumption is considered to be one of the main causes of left ventricular dysfunction in alcoholic cardiomyopathy (ACM). As previously suggested, high-dose alcohol induces oxidation stress and apoptosis of cardiomyocytes. However, the underlying mechanisms are yet to be elucidated. In this study, we found that high-dose alcohol treatment stimulated expression and activity of Pin1 in mouse primary cardiomyocytes. While siRNA-mediated knockdown of Pin1 suppressed alcohol-induced mouse cardiomyocyte apoptosis, overexpression of Pin1 further upregulated the numbers of apoptotic mouse cardiomyocytes. We further demonstrated that Pin1 promotes mitochondria oxidative stress and loss of mitochondrial membrane potential but suppresses endothelial nitric oxide synthase (eNOS) expression in the presence of alcohol. Taken together, our results revealed a pivotal role of Pin1 in regulation of alcohol-induced mouse cardiomyocytes apoptosis by promoting reactive oxygen species (ROS) accumulation and repressing eNOS expression, which could be potential therapeutic targets for ACM.


Asunto(s)
Apoptosis/efectos de los fármacos , Etanol/toxicidad , Miocitos Cardíacos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Isomerasa de Peptidilprolil/antagonistas & inhibidores , ARN Interferente Pequeño/farmacología , Animales , Apoptosis/genética , Ratones , Miocitos Cardíacos/patología , Peptidilprolil Isomerasa de Interacción con NIMA , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo/genética , Estrés Oxidativo/inmunología , Isomerasa de Peptidilprolil/genética , Isomerasa de Peptidilprolil/metabolismo , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo
19.
J Biomol Struct Dyn ; 34(2): 399-413, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26046477

RESUMEN

Cyclophilins (CyPs) are enzymes involved in protein folding. In Trypanosoma cruzi (T. cruzi), the most abundantly expressed CyP is the isoform TcCyP19. It has been shown that TcCyP19 is inhibited by the immunosuppressive drug cyclosporin A (CsA) and analogs, which also proved to have potent trypanosomicidal activity in vitro. In this work, we continue and expand a previous study on the molecular interactions of CsA, and a set of analogs modeled in complexes with TcCyP19. The modeled complexes were used to evaluate binding free energies by molecular dynamics (MD), applying the Linear Interaction Energy (LIE) method. In addition, putative binding sites were identified by molecular docking. In our analysis, the binding free energy calculations did not correlate with experimental data. The heterogeneity of the non-bonded energies and the variation in the pattern of hydrogen bonds suggest that the systems may not be suitable for the application of the LIE method. Further, the docking calculations identified two other putative binding sites with comparable scoring energies to the active site, a fact that may also explain the lack of correlation found. Kinetic experiments are needed to confirm or reject the multiple binding sites hypothesis. In the meantime, MD simulations at the alternative sites, employing other methods to compute binding free energies, might be successful at finding good correlations with the experimental data.


Asunto(s)
Ciclofilinas/antagonistas & inhibidores , Ciclosporina/farmacología , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Trypanosoma cruzi/metabolismo , Aminoácidos/química , Sitios de Unión , Intervalos de Confianza , Cristalografía por Rayos X , Ciclosporina/química , Enlace de Hidrógeno , Ligandos , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Isomerasa de Peptidilprolil/metabolismo , Solventes , Electricidad Estática , Termodinámica , Trypanosoma cruzi/efectos de los fármacos
20.
Bioorg Med Chem Lett ; 25(23): 5619-24, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26508545

RESUMEN

Pin1 is a peptidyl prolyl isomerase that specifically catalyzes cis-trans isomerization of phosphorylated Thr/Ser-Pro peptide bonds in substrate proteins and peptides. Pin1 is involved in many important cellular processes, including cancer progression, so it is a potential target of cancer therapy. We designed and synthesized a novel series of Pin1 inhibitors based on a glutamic acid or aspartic acid scaffold bearing an aromatic moiety to provide a hydrophobic surface and a cyclic aliphatic amine moiety with affinity for the proline-binding site of Pin1. Glutamic acid derivatives bearing cycloalkylamino and phenylthiazole groups showed potent Pin1-inhibitory activity comparable with that of known inhibitor VER-1. The results indicate that steric interaction of the cyclic alkyl amine moiety with binding site residues plays a key role in enhancing Pin1-inhibitory activity.


Asunto(s)
Aminas/síntesis química , Ácido D-Aspártico/síntesis química , Ácido Glutámico/síntesis química , Isomerasa de Peptidilprolil/antagonistas & inhibidores , Aminas/química , Aminas/farmacología , Dominio Catalítico , Cristalografía por Rayos X , Ácido D-Aspártico/química , Ácido D-Aspártico/farmacología , Ácido Glutámico/química , Ácido Glutámico/farmacología , Hidrocarburos Cíclicos , Concentración 50 Inhibidora , Simulación del Acoplamiento Molecular , Estructura Molecular , Peptidilprolil Isomerasa de Interacción con NIMA
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...