Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Front Cell Infect Microbiol ; 14: 1391758, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38716194

RESUMEN

Campylobacter jejuni, a Gram-negative bacterium, is one of the most common causes of foodborne illness worldwide. Its adhesion mechanism is mediated by several bacterial factors, including flagellum, protein adhesins, lipooligosaccharides, proteases, and host factors, such as surface glycans on epithelial cells and mucins. Fungal lectins, specialized carbohydrate-binding proteins, can bind to specific glycans on host and bacterial cells and thus influence pathogenesis. In this study, we investigated the effects of fungal lectins and protease inhibitors on the adhesion of C. jejuni to model biotic surfaces (mucin, fibronectin, and collagen) and Caco-2 cells as well as the invasion of Caco-2 cells. The lectins Marasmius oreades agglutinin (MOA) and Laccaria bicolor tectonin 2 (Tec2) showed remarkable efficacy in all experiments. In addition, different pre-incubations of lectins with C. jejuni or Caco-2 cells significantly inhibited the ability of C. jejuni to adhere to and invade Caco-2 cells, but to varying degrees. Pre-incubation of Caco-2 cells with selected lectins reduced the number of invasive C. jejuni cells the most, while simultaneous incubation showed the greatest reduction in adherent C. jejuni cells. These results suggest that fungal lectins are a promising tool for the prevention and treatment of C. jejuni infections. Furthermore, this study highlights the potential of fungi as a rich reservoir for novel anti-adhesive agents.


Asunto(s)
Adhesión Bacteriana , Campylobacter jejuni , Lectinas , Inhibidores de Proteasas , Campylobacter jejuni/efectos de los fármacos , Campylobacter jejuni/fisiología , Campylobacter jejuni/metabolismo , Humanos , Células CACO-2 , Adhesión Bacteriana/efectos de los fármacos , Lectinas/metabolismo , Lectinas/farmacología , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/metabolismo , Hongos/efectos de los fármacos , Mucinas/metabolismo , Células Epiteliales/microbiología , Fibronectinas/metabolismo
2.
J Med Chem ; 67(9): 7048-7067, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38630165

RESUMEN

Emerging RNA viruses, including SARS-CoV-2, continue to be a major threat. Cell entry of SARS-CoV-2 particles via the endosomal pathway involves cysteine cathepsins. Due to ubiquitous expression, cathepsin L (CatL) is considered a promising drug target in the context of different viral and lysosome-related diseases. We characterized the anti-SARS-CoV-2 activity of a set of carbonyl- and succinyl epoxide-based inhibitors, which were previously identified as inhibitors of cathepsins or related cysteine proteases. Calpain inhibitor XII, MG-101, and CatL inhibitor IV possess antiviral activity in the very low nanomolar EC50 range in Vero E6 cells and inhibit CatL in the picomolar Ki range. We show a relevant off-target effect of CatL inhibition by the coronavirus main protease α-ketoamide inhibitor 13b. Crystal structures of CatL in complex with 14 compounds at resolutions better than 2 Å present a solid basis for structure-guided understanding and optimization of CatL inhibitors toward protease drug development.


Asunto(s)
Antivirales , Catepsina L , SARS-CoV-2 , Catepsina L/antagonistas & inhibidores , Catepsina L/metabolismo , Antivirales/farmacología , Antivirales/química , Antivirales/síntesis química , Animales , Chlorocebus aethiops , Células Vero , SARS-CoV-2/efectos de los fármacos , Humanos , Relación Estructura-Actividad , Inhibidores de Cisteína Proteinasa/farmacología , Inhibidores de Cisteína Proteinasa/química , Inhibidores de Cisteína Proteinasa/síntesis química , Cristalografía por Rayos X , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/química , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/metabolismo , Modelos Moleculares
3.
Science ; 382(6674): 1015-1020, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38033070

RESUMEN

Photolyase is an enzyme that uses light to catalyze DNA repair. To capture the reaction intermediates involved in the enzyme's catalytic cycle, we conducted a time-resolved crystallography experiment. We found that photolyase traps the excited state of the active cofactor, flavin adenine dinucleotide (FAD), in a highly bent geometry. This excited state performs electron transfer to damaged DNA, inducing repair. We show that the repair reaction, which involves the lysis of two covalent bonds, occurs through a single-bond intermediate. The transformation of the substrate into product crowds the active site and disrupts hydrogen bonds with the enzyme, resulting in stepwise product release, with the 3' thymine ejected first, followed by the 5' base.


Asunto(s)
Desoxirribodipirimidina Fotoliasa , Cristalografía , Desoxirribodipirimidina Fotoliasa/química , Desoxirribodipirimidina Fotoliasa/metabolismo , Reparación del ADN , Daño del ADN , Transporte de Electrón
4.
Commun Biol ; 6(1): 1058, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37853179

RESUMEN

Several drug screening campaigns identified Calpeptin as a drug candidate against SARS-CoV-2. Initially reported to target the viral main protease (Mpro), its moderate activity in Mpro inhibition assays hints at a second target. Indeed, we show that Calpeptin is an extremely potent cysteine cathepsin inhibitor, a finding additionally supported by X-ray crystallography. Cell infection assays proved Calpeptin's efficacy against SARS-CoV-2. Treatment of SARS-CoV-2-infected Golden Syrian hamsters with sulfonated Calpeptin at a dose of 1 mg/kg body weight reduces the viral load in the trachea. Despite a higher risk of side effects, an intrinsic advantage in targeting host proteins is their mutational stability in contrast to highly mutable viral targets. Here we show that the inhibition of cathepsins, a protein family of the host organism, by calpeptin is a promising approach for the treatment of SARS-CoV-2 and potentially other viral infections.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2/metabolismo , Catepsinas , Antivirales/farmacología , Antivirales/uso terapéutico , Antivirales/química , Inhibidores de Proteasas/farmacología , Cisteína Endopeptidasas/metabolismo
5.
Commun Biol ; 6(1): 450, 2023 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-37095140

RESUMEN

Addressing the elusive specificity of cysteine cathepsins, which in contrast to caspases and trypsin-like proteases lack strict specificity determining P1 pocket, calls for innovative approaches. Proteomic analysis of cell lysates with human cathepsins K, V, B, L, S, and F identified 30,000 cleavage sites, which we analyzed by software platform SAPS-ESI (Statistical Approach to Peptidyl Substrate-Enzyme Specific Interactions). SAPS-ESI is used to generate clusters and training sets for support vector machine learning. Cleavage site predictions on the SARS-CoV-2 S protein, confirmed experimentally, expose the most probable first cut under physiological conditions and suggested furin-like behavior of cathepsins. Crystal structure analysis of representative peptides in complex with cathepsin V reveals rigid and flexible sites consistent with analysis of proteomics data by SAPS-ESI that correspond to positions with heterogeneous and homogeneous distribution of residues. Thereby support for design of selective cleavable linkers of drug conjugates and drug discovery studies is provided.


Asunto(s)
COVID-19 , Cisteína , Humanos , Proteómica , SARS-CoV-2
6.
Plant Physiol ; 191(3): 1934-1952, 2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36517238

RESUMEN

TGA (TGACG-binding) transcription factors, which bind their target DNA through a conserved basic region leucine zipper (bZIP) domain, are vital regulators of gene expression in salicylic acid (SA)-mediated plant immunity. Here, we investigated the role of StTGA2.1, a potato (Solanum tuberosum) TGA lacking the full bZIP, which we named a mini-TGA. Such truncated proteins have been widely assigned as loss-of-function mutants. We, however, confirmed that StTGA2.1 overexpression compensates for SA-deficiency, indicating a distinct mechanism of action compared with model plant species. To understand the underlying mechanisms, we showed that StTGA2.1 can physically interact with StTGA2.2 and StTGA2.3, while its interaction with DNA was not detected. We investigated the changes in transcriptional regulation due to StTGA2.1 overexpression, identifying direct and indirect target genes. Using in planta transactivation assays, we confirmed that StTGA2.1 interacts with StTGA2.3 to activate StPRX07, a member of class III peroxidases (StPRX), which are known to play role in immune response. Finally, via structural modeling and molecular dynamics simulations, we hypothesized that the compact molecular architecture of StTGA2.1 distorts DNA conformation upon heterodimer binding to enable transcriptional activation. This study demonstrates how protein truncation can lead to distinct functions and that such events should be studied carefully in other protein families.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , Factores de Transcripción , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional , Expresión Génica , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Regulación de la Expresión Génica de las Plantas
7.
Commun Biol ; 5(1): 805, 2022 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-35953531

RESUMEN

SARS-CoV-2 papain-like protease (PLpro) covers multiple functions. Beside the cysteine-protease activity, facilitating cleavage of the viral polypeptide chain, PLpro has the additional and vital function of removing ubiquitin and ISG15 (Interferon-stimulated gene 15) from host-cell proteins to support coronaviruses in evading the host's innate immune responses. We identified three phenolic compounds bound to PLpro, preventing essential molecular interactions to ISG15 by screening a natural compound library. The compounds identified by X-ray screening and complexed to PLpro demonstrate clear inhibition of PLpro in a deISGylation activity assay. Two compounds exhibit distinct antiviral activity in Vero cell line assays and one inhibited a cytopathic effect in non-cytotoxic concentration ranges. In the context of increasing PLpro mutations in the evolving new variants of SARS-CoV-2, the natural compounds we identified may also reinstate the antiviral immune response processes of the host that are down-regulated in COVID-19 infections.


Asunto(s)
Antivirales , Tratamiento Farmacológico de COVID-19 , Sitio Alostérico , Antivirales/farmacología , Proteasas Similares a la Papaína de Coronavirus , Humanos , Papaína/metabolismo , Péptido Hidrolasas/metabolismo , SARS-CoV-2
8.
Int J Mol Sci ; 23(9)2022 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-35563308

RESUMEN

We introduce a new family of fungal protease inhibitors with ß-trefoil fold from the mushroom Coprinopsis cinerea, named cocaprins, which inhibit both cysteine and aspartic proteases. Two cocaprin-encoding genes are differentially expressed in fungal tissues. One is highly transcribed in vegetative mycelium and the other in the stipes of mature fruiting bodies. Cocaprins are small proteins (15 kDa) with acidic isoelectric points that form dimers. The three-dimensional structure of cocaprin 1 showed similarity to fungal ß-trefoil lectins. Cocaprins inhibit plant C1 family cysteine proteases with Ki in the micromolar range, but do not inhibit the C13 family protease legumain, which distinguishes them from mycocypins. Cocaprins also inhibit the aspartic protease pepsin with Ki in the low micromolar range. Mutagenesis revealed that the ß2-ß3 loop is involved in the inhibition of cysteine proteases and that the inhibitory reactive sites for aspartic and cysteine proteases are located at different positions on the protein. Their biological function is thought to be the regulation of endogenous proteolytic activities or in defense against fungal antagonists. Cocaprins are the first characterized aspartic protease inhibitors with ß-trefoil fold from fungi, and demonstrate the incredible plasticity of loop functionalization in fungal proteins with ß-trefoil fold.


Asunto(s)
Agaricales , Proteasas de Ácido Aspártico , Proteasas de Cisteína , Lotus , Agaricales/química , Ácido Aspártico Endopeptidasas , Proteasas de Ácido Aspártico/genética , Cisteína , Proteasas de Cisteína/genética , Lotus/metabolismo , Péptido Hidrolasas/metabolismo , Inhibidores de Proteasas/química
10.
Int J Mol Sci ; 22(3)2021 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-33498210

RESUMEN

Protein inhibitors of proteases are an important tool of nature to regulate and control proteolysis in living organisms under physiological and pathological conditions. In this review, we analyzed the mechanisms of inhibition of cysteine proteases on the basis of structural information and compiled kinetic data. The gathered structural data indicate that the protein fold is not a major obstacle for the evolution of a protease inhibitor. It appears that nature can convert almost any starting fold into an inhibitor of a protease. In addition, there appears to be no general rule governing the inhibitory mechanism. The structural data make it clear that the "lock and key" mechanism is a historical concept with limited validity. However, the analysis suggests that the shape of the active site cleft of proteases imposes some restraints. When the S1 binding site is shaped as a pocket buried in the structure of protease, inhibitors can apply substrate-like binding mechanisms. In contrast, when the S1 binding site is in part exposed to solvent, the substrate-like inhibition cannot be employed. It appears that all proteases, with the exception of papain-like proteases, belong to the first group of proteases. Finally, we show a number of examples and provide hints on how to engineer protein inhibitors.


Asunto(s)
Proteasas de Cisteína/química , Inhibidores de Cisteína Proteinasa/química , Animales , Cistatinas/química , Cistatinas/metabolismo , Cistatinas/farmacología , Proteasas de Cisteína/metabolismo , Inhibidores de Cisteína Proteinasa/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Humanos , Unión Proteica , Securina/química , Securina/metabolismo , Securina/farmacología , Proteína Inhibidora de la Apoptosis Ligada a X/química , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Proteína Inhibidora de la Apoptosis Ligada a X/farmacología
11.
Biochim Biophys Acta Proteins Proteom ; 1869(2): 140567, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33227497

RESUMEN

Human cathepsin X belongs to the cathepsin family of 11 lysosomal cysteine proteases. We expressed recombinant procathepsin X in Pichia pastoris in vitro and cleaved it into its active mature form using aspartic cathepsin E. We found, using size exclusion chromatography, X-ray crystallography, and small-angle X-ray scattering, that cathepsin X is a biologically active homodimer with a molecular weight of ~53 kDa. The novel finding that cathepsin X is a dimeric protein opens new horizons in the understanding of its function and the underlying pathophysiological mechanisms of various diseases including neurodegenerative disorders in humans.


Asunto(s)
Catepsina K/genética , Catepsina Z/genética , Proteínas Recombinantes/química , Secuencia de Aminoácidos/genética , Catepsina K/ultraestructura , Catepsina Z/ultraestructura , Cristalografía por Rayos X , Humanos , Pichia/química , Pichia/genética , Proteínas Recombinantes/genética , Saccharomycetales/química , Saccharomycetales/genética
12.
Int J Mol Sci ; 21(17)2020 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-32858965

RESUMEN

SecA protein is a major component of the general bacterial secretory system. It is an ATPase that couples nucleotide hydrolysis to protein translocation. In some Gram-positive pathogens, a second paralogue, SecA2, exports a different set of substrates, usually virulence factors. To identify SecA2 features different from SecA(1)s, we determined the crystal structure of SecA2 from Clostridioides difficile, an important nosocomial pathogen, in apo and ATP-γ-S-bound form. The structure reveals a closed monomer lacking the C-terminal tail (CTT) with an otherwise similar multidomain organization to its SecA(1) homologues and conserved binding of ATP-γ-S. The average in vitro ATPase activity rate of C. difficile SecA2 was 2.6 ± 0.1 µmolPi/min/µmol. Template-based modeling combined with evolutionary conservation analysis supports a model where C. difficile SecA2 in open conformation binds the target protein, ensures its movement through the SecY channel, and enables dimerization through PPXD/HWD cross-interaction of monomers during the process. Both approaches exposed regions with differences between SecA(1) and SecA2 homologues, which are in agreement with the unique adaptation of SecA2 proteins for a specific type of substrate, a role that can be addressed in further studies.


Asunto(s)
Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Clostridioides difficile/enzimología , Adenosina Trifosfatasas/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Clostridioides difficile/química , Clostridioides difficile/genética , Secuencia Conservada , Cristalografía por Rayos X , Evolución Molecular , Modelos Moleculares , Conformación Proteica
13.
Appl Environ Microbiol ; 86(17)2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32561587

RESUMEN

Pernisine is a subtilisin-like protease that was originally identified in the hyperthermophilic archaeon Aeropyrum pernix, which lives in extreme marine environments. Pernisine shows exceptional stability and activity due to the high-temperature conditions experienced by A. pernix Pernisine is of interest for industrial purposes, as it is one of the few proteases that has demonstrated prion-degrading activity. Like other extracellular subtilisins, pernisine is synthesized in its inactive pro-form (pro-pernisine), which needs to undergo maturation to become proteolytically active. The maturation processes of mesophilic subtilisins have been investigated in detail; however, less is known about the maturation of their thermophilic homologs, such as pernisine. Here, we show that the structure of pro-pernisine is disordered in the absence of Ca2+ ions. In contrast to the mesophilic subtilisins, pro-pernisine requires Ca2+ ions to adopt the conformation suitable for its subsequent maturation. In addition to several Ca2+-binding sites that have been conserved from the thermostable Tk-subtilisin, pernisine has an additional insertion sequence with a Ca2+-binding motif. We demonstrate the importance of this insertion for efficient folding and stabilization of pernisine during its maturation. Moreover, analysis of the pernisine propeptide explains the high-temperature requirement for pro-pernisine maturation. Of note, the propeptide inhibits the pernisine catalytic domain more potently at high temperatures. After dissociation, the propeptide is destabilized at high temperatures only, which leads to its degradation and finally to pernisine activation. Our data provide new insights into and understanding of the thermostable subtilisin autoactivation mechanism.IMPORTANCE Enzymes from thermophilic organisms are of particular importance for use in industrial applications, due to their exceptional stability and activity. Pernisine, from the hyperthermophilic archaeon Aeropyrum pernix, is a proteolytic enzyme that can degrade infective prion proteins and thus has a potential use for disinfection of prion-contaminated surfaces. Like other subtilisin-like proteases, pernisine needs to mature through an autocatalytic process to become an active protease. In the present study, we address the maturation of pernisine and show that the process is regulated specifically at high temperatures by the propeptide. Furthermore, we demonstrate the importance of a unique Ca2+-binding insertion for stabilization of mature pernisine. Our results provide a novel understanding of thermostable subtilisin autoactivation, which might advance the development of these enzymes for commercial use.


Asunto(s)
Aeropyrum/genética , Proteínas Arqueales/genética , Péptido Hidrolasas/genética , Aeropyrum/enzimología , Secuencia de Aminoácidos , Proteínas Arqueales/química , Proteínas Arqueales/metabolismo , Sitios de Unión , Conformación Molecular , Péptido Hidrolasas/química , Péptido Hidrolasas/metabolismo
14.
Commun Biol ; 3(1): 178, 2020 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-32313083

RESUMEN

To achieve productive binding, enzymes and substrates must align their geometries to complement each other along an entire substrate binding site, which may require enzyme flexibility. In pursuit of novel drug targets for the human pathogen S. aureus, we studied peptidoglycan N-acetylglucosaminidases, whose structures are composed of two domains forming a V-shaped active site cleft. Combined insights from crystal structures supported by site-directed mutagenesis, modeling, and molecular dynamics enabled us to elucidate the substrate binding mechanism of SagB and AtlA-gl. This mechanism requires domain sliding from the open form observed in their crystal structures, leading to polysaccharide substrate binding in the closed form, which can enzymatically process the bound substrate. We suggest that these two hydrolases must exhibit unusual extents of flexibility to cleave the rigid structure of a bacterial cell wall.


Asunto(s)
Acetilglucosaminidasa/metabolismo , Proteínas Bacterianas/metabolismo , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Peptidoglicano/metabolismo , Staphylococcus aureus/enzimología , Acetilglucosaminidasa/química , Acetilglucosaminidasa/genética , Regulación Alostérica , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Sitios de Unión , Catálisis , Dominio Catalítico , Hidrólisis , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Mutación , N-Acetil Muramoil-L-Alanina Amidasa/química , N-Acetil Muramoil-L-Alanina Amidasa/genética , Dominios Proteicos , Staphylococcus aureus/genética , Relación Estructura-Actividad , Especificidad por Sustrato
15.
Nature ; 578(7796): 627-630, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32025030

RESUMEN

Thyroglobulin (TG) is the protein precursor of thyroid hormones, which are essential for growth, development and the control of metabolism in vertebrates1,2. Hormone synthesis from TG occurs in the thyroid gland via the iodination and coupling of pairs of tyrosines, and is completed by TG proteolysis3. Tyrosine proximity within TG is thought to enable the coupling reaction but hormonogenic tyrosines have not been clearly identified, and the lack of a three-dimensional structure of TG has prevented mechanistic understanding4. Here we present the structure of full-length human thyroglobulin at a resolution of approximately 3.5 Å, determined by cryo-electron microscopy. We identified all of the hormonogenic tyrosine pairs in the structure, and verified them using site-directed mutagenesis and in vitro hormone-production assays using human TG expressed in HEK293T cells. Our analysis revealed that the proximity, flexibility and solvent exposure of the tyrosines are the key characteristics of hormonogenic sites. We transferred the reaction sites from TG to an engineered tyrosine donor-acceptor pair in the unrelated bacterial maltose-binding protein (MBP), which yielded hormone production with an efficiency comparable to that of TG. Our study provides a framework to further understand the production and regulation of thyroid hormones.


Asunto(s)
Microscopía por Crioelectrón , Tiroglobulina/química , Tiroglobulina/ultraestructura , Proteínas Bacterianas/química , Células HEK293 , Humanos , Proteínas de Unión a Maltosa/química , Modelos Moleculares , Mutación , Reproducibilidad de los Resultados , Solventes/química , Tiroglobulina/genética , Hormonas Tiroideas/biosíntesis , Hormonas Tiroideas/metabolismo , Tirosina/química , Tirosina/genética , Tirosina/metabolismo
16.
Chem Sci ; 10(36): 8461-8477, 2019 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-31803426

RESUMEN

Human cysteine cathepsins constitute an 11-membered family of proteases responsible for degradation of proteins in cellular endosomal-lysosomal compartments as such, they play important roles in antigen processing, cellular stress signaling, autophagy, and senescence. Moreover, for many years these enzymes were also linked to tumor growth, invasion, angiogenesis and metastasis when upregulated. Individual biological roles of each cathepsin are difficult to establish, because of their redundancy and similar substrate specificities. Selective chemical tools that enable imaging of individual cathepsin activities in living cells, tumors, and the tumor microenvironment may provide a better insight into their functions. In this work, we used HyCoSuL technology to profile the substrate specificity of human cathepsin B. The use of unnatural amino acids in the substrate library enabled us to uncover the broad cathepsin B preferences that we utilized to design highly-selective substrates and fluorescent activity-based probes (ABPs). We further demonstrated that Cy5-labeled MP-CB-2 probe can selectively label cathepsin B in eighteen cancer cell lines tested, making this ABP highly suitable for other biological setups. Moreover, using Cy5-labelled MP-CB-2 we were able to demonstrate by fluorescence microscopy that in cancer cells cathepsins B and L share overlapping, but not identical subcellular localization.

17.
Sci Rep ; 9(1): 1678, 2019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30737447

RESUMEN

Validation of three-dimensional structures is at the core of structural determination methods. The local validation criteria, such as deviations from ideal bond length and bonding angles, Ramachandran plot outliers and clashing contacts, are a standard part of structure analysis before structure deposition, whereas the global and regional packing may not yet have been addressed. In the last two decades, three-dimensional models of macromolecules such as proteins have been successfully described by a network of nodes and edges. Amino acid residues as nodes and close contact between the residues as edges have been used to explore basic network properties, to study protein folding and stability and to predict catalytic sites. Using complex network analysis, we introduced common network parameters to distinguish between correct and incorrect three-dimensional protein structures. The analysis showed that correct structures have a higher average node degree, higher graph energy, and lower shortest path length than their incorrect counterparts. Thus, correct protein models are more densely intra-connected, and in turn, the transfer of information between nodes/amino acids is more efficient. Moreover, protein graph spectra were used to investigate model bias in protein structure.

18.
J Am Chem Soc ; 141(8): 3507-3514, 2019 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-30689386

RESUMEN

Irreversible covalent inhibitors can have a beneficial pharmacokinetic/pharmacodynamics profile but are still often avoided due to the risk of indiscriminate covalent reactivity and the resulting adverse effects. To overcome this potential liability, we introduced an alkyne moiety as a latent electrophile into small molecule inhibitors of cathepsin K (CatK). Alkyne-based inhibitors do not show indiscriminate thiol reactivity but potently inhibit CatK protease activity by formation of an irreversible covalent bond with the catalytic cysteine residue, confirmed by crystal structure analysis. The rate of covalent bond formation ( kinact) does not correlate with electrophilicity of the alkyne moiety, indicative of a proximity-driven reactivity. Inhibition of CatK-mediated bone resorption is validated in human osteoclasts. Together, this work illustrates the potential of alkynes as latent electrophiles in small molecule inhibitors, enabling the development of irreversible covalent inhibitors with an improved safety profile.


Asunto(s)
Alquinos/farmacología , Catepsina K/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Alquinos/química , Catepsina K/metabolismo , Inhibidores de Cisteína Proteinasa/síntesis química , Inhibidores de Cisteína Proteinasa/química , Humanos , Modelos Moleculares , Estructura Molecular , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química
19.
Acta Chim Slov ; 66(1): 5-17, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33855484

RESUMEN

The majority of lysosomal cysteine cathepsins are ubiquitously expressed enzymes. However, some of them differ in their specific cell or tissue distribution and substrate specificity, suggesting their involvement in determining normal cellular processes, as well as pathologies. Their proteolytic activities are potentially harmful if uncontrolled. Therefore, living organisms have developed several regulatory mechanisms such as endogenous protein inhibitors of the cystatin family, including the group of small cytosolic proteins, the stefins. The main focus of this review is stefins of various origins and their properties, structure, and mechanism of interaction with their target enzymes. Furthermore, oligomerization and fibrillogenesis in stefins and/or cystatins provide insights into conformational diseases. The present status of the knowledge in this field and current trends might contribute to identifying novel therapeutic targets and approaches to treat various diseases.

20.
Acta Chim Slov ; 66(1): 50-57, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33855486

RESUMEN

Legumain or asparagine endopeptidase is a unique cysteine endopeptidase with a distinctive specificity for the hydrolysis of peptide bonds after asparagine and to a lesser extent after aspartate. It is ubiquitously expressed in various tissues and besides its involvement in immune response and other physiological processes, it was also shown to play a role in pathological states such as inflammation and cancer. In order to improve our understanding of legumain substrate recognition we have performed proteomic profiling of legumain specificity on native proteins derived from MDA-MB-231 cells using two different N-terminal labelling methodologies (FPPS and ISIL). Our data revealed narrow cleavage specificity for P1 position combined with clear cleavage preference for unstructured secondary regions in the substrate proteins. No extended cleavage specificity on native proteins was observed. Moreover, a limited number of identified cleavages on individual substrates suggest its primary role in precision proteolysis and regulatory proteolytic events.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA