Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 260
Filtrar
1.
J Org Chem ; 89(17): 12432-12438, 2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39178422

RESUMEN

Substituted 5-hydroxy γ-pyrones have shown promise as covalent inhibitor leads against cysteine proteases and transcription factors, but their hydrolytic instability has hindered optimization efforts. Previous mechanistic proposals have suggested that these molecules function as Michael acceptor prodrugs, releasing a leaving group to generate an o-quinone methide-like structure. Addition to this electrophile of either water or an active site cysteine was purported to lead to inhibitor hydrolysis or enzyme inhibition, respectively. Through the use of kinetic nuclear magnetic resonance experiments, Hammett analysis, kinetic isotope effect studies, and density functional theory calculations, our findings suggest that enzyme inhibition and hydrolysis proceed by distinct pathways and are differentially influenced by substituent electronics. This mechanistic revision helps enable a more rational optimization for this class of promising compounds.


Asunto(s)
Profármacos , Pironas , Profármacos/química , Profármacos/farmacología , Pironas/química , Pironas/farmacología , Estructura Molecular , Teoría Funcional de la Densidad , Cinética , Hidrólisis , Proteasas de Cisteína/química , Proteasas de Cisteína/metabolismo
2.
PLoS One ; 19(8): e0307512, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39093838

RESUMEN

The multifunctional autoprocessing repeat-in-toxin (MARTX) toxin is the primary virulence factor of Vibrio vulnificus displaying cytotoxic and hemolytic properties. The cysteine protease domain (CPD) is responsible for activating the MARTX toxin by cleaving the toxin precursor and releasing the mature toxin fragments. To investigate the structural determinants for inositol hexakisphosphate (InsP6)-mediated activation of the CPD, we determined the crystal structures of unprocessed and ß-flap truncated MARTX CPDs of Vibrio vulnificus strain MO6-24/O in complex with InsP6 at 1.3 and 2.2Å resolution, respectively. The CPD displays a conserved domain with a central seven-stranded ß-sheet flanked by three α-helices. The scissile bond Leu3587-Ala3588 is bound in the catalytic site of the InsP6-loaded form of the Cys3727Ala mutant. InsP6 interacts with the conserved basic cleft and the ß-flap inducing the active conformation of catalytic residues. The ß-flap of the post-CPD is flexible in the InsP6-unbound state. The structure of the CPD Δß-flap showed an inactive conformation of the catalytic residues due to the absence of interaction between the active site and the ß-flap. This study confirms the InsP6-mediated activation of the MARTX CPDs in which InsP6-binding induces conformational changes of the catalytic residues and the ß-flap that holds the N terminus of the CPD in the active site, facilitating hydrolysis of the scissile bond.


Asunto(s)
Ácido Fítico , Vibrio vulnificus , Vibrio vulnificus/enzimología , Vibrio vulnificus/genética , Vibrio vulnificus/metabolismo , Ácido Fítico/metabolismo , Dominio Catalítico , Proteasas de Cisteína/metabolismo , Proteasas de Cisteína/química , Proteasas de Cisteína/genética , Cristalografía por Rayos X , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/genética , Dominios Proteicos , Modelos Moleculares , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Secuencia de Aminoácidos
3.
Molecules ; 29(11)2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38893535

RESUMEN

The aim of this study was to investigate the transition from non-covalent reversible over covalent reversible to covalent irreversible inhibition of cysteine proteases by making delicate structural changes to the warhead scaffold. To this end, dipeptidic rhodesain inhibitors with different N-terminal electrophilic arenes as warheads relying on the SNAr mechanism were synthesized and investigated. Strong structure-activity relationships of the inhibition potency, the degree of covalency, and the reversibility of binding on the arene substitution pattern were found. The studies were complemented and substantiated by molecular docking and quantum-mechanical calculations of model systems. Furthermore, the improvement in the membrane permeability of peptide esters in comparison to their corresponding carboxylic acids was exemplified.


Asunto(s)
Proteasas de Cisteína , Inhibidores de Cisteína Proteinasa , Simulación del Acoplamiento Molecular , Inhibidores de Cisteína Proteinasa/química , Inhibidores de Cisteína Proteinasa/farmacología , Inhibidores de Cisteína Proteinasa/metabolismo , Relación Estructura-Actividad , Proteasas de Cisteína/metabolismo , Proteasas de Cisteína/química , Cisteína Endopeptidasas/metabolismo , Cisteína Endopeptidasas/química , Estructura Molecular
4.
Cells ; 13(11)2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38891048

RESUMEN

Cysteine cathepsins F and W are members of the papain-like cysteine protease family, which have distinct structural features and functional roles in various physiological and pathological processes. This review provides a comprehensive overview of the current understanding of the structure, biological functions, and pathological implications of cathepsins F and W. Beginning with an introduction to these proteases, we delve into their structural characteristics and elucidate their unique features that dictate their enzymatic activities and substrate specificity. We also explore the intricate involvement of cathepsins F and W in malignancies, highlighting their role as potential biomarkers and therapeutic targets in cancer progression. Furthermore, we discuss the emerging roles of these enzymes in immune response modulation and neurological disorders, shedding light on their implications in autoimmune and neurodegenerative diseases. Finally, we review the landscape of inhibitors targeting these proteases, highlighting their therapeutic potential and challenges in clinical translation. This review brings together the diverse facets of cysteine cathepsins F and W, providing insights into their roles in health and disease and guiding future investigations for therapeutic advances.


Asunto(s)
Catepsina F , Humanos , Animales , Catepsina F/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/patología , Proteasas de Cisteína/metabolismo , Proteasas de Cisteína/química , Catepsinas/metabolismo , Catepsinas/química , Especificidad por Sustrato
6.
Int J Biol Macromol ; 271(Pt 1): 132505, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38768911

RESUMEN

Proteases, essential regulators of plant stress responses, remain enigmatic in their precise functional roles. By employing activity-based probes for real-time monitoring, this study aimed to delve into protease activities in Chlamydomonas reinhardtii exposed to oxidative stress induced by hydrogen peroxide. However, our work revealed that the activity-based probes strongly labelled three non-proteolytic proteins-PsbO, PsbP, and PsbQ-integral components of photosystem II's oxygen-evolving complex. Subsequent biochemical assays and mass spectrometry experiments revealed the involvement of CrCEP1, a previously uncharacterized papain-like cysteine protease, as the catalyst of this labelling reaction. Further experiments with recombinant CrCEP1 and PsbO proteins replicated the reaction in vitro. Our data unveiled that endopeptidase CrCEP1 also has transpeptidase activity, ligating probes and peptides to the N-termini of Psb proteins, thereby expanding the repertoire of its enzymatic activities. The hitherto unknown transpeptidase activity of CrCEP1, working in conjunction with its proteolytic activity, unveils putative complex and versatile roles for proteases in cellular processes during stress responses.


Asunto(s)
Chlamydomonas reinhardtii , Proteasas de Cisteína , Proteasas de Cisteína/metabolismo , Proteasas de Cisteína/química , Chlamydomonas reinhardtii/enzimología , Estrés Oxidativo , Complejo de Proteína del Fotosistema II/metabolismo , Complejo de Proteína del Fotosistema II/química , Proteínas de Plantas/metabolismo , Proteínas de Plantas/química , Peróxido de Hidrógeno/metabolismo , Cisteína Endopeptidasas/metabolismo , Cisteína Endopeptidasas/química
7.
J Biol Chem ; 299(6): 104801, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37164157

RESUMEN

Papain-like cysteine peptidases form a big and highly diverse superfamily of proteins involved in many important biological functions, such as protein turnover, deubiquitination, tissue remodeling, blood clotting, virulence, defense, and cell wall remodeling. High sequence and structure diversity observed within these proteins hinders their comprehensive classification as well as the identification of new representatives. Moreover, in general protein databases, many families already classified as papain like lack details regarding their mechanism of action or biological function. Here, we use transitive remote homology searches and 3D modeling to newly classify 21 families to the papain-like cysteine peptidase superfamily. We attempt to predict their biological function and provide structural characterization of 89 protein clusters defined based on sequence similarity altogether spanning 106 papain-like families. Moreover, we systematically discuss observed diversity in sequences, structures, and catalytic sites. Eventually, we expand the list of human papain-related proteins by seven representatives, including dopamine receptor-interacting protein 1 as potential deubiquitinase, and centriole duplication regulating CEP76 as retaining catalytically active peptidase-like domain. The presented results not only provide structure-based rationales to already existing peptidase databases but also may inspire further experimental research focused on peptidase-related biological processes.


Asunto(s)
Proteasas de Cisteína , Papaína , Humanos , Dominio Catalítico , Centriolos/metabolismo , Proteasas de Cisteína/química , Proteasas de Cisteína/clasificación , Proteasas de Cisteína/metabolismo , Enzimas Desubicuitinizantes/metabolismo , Modelos Moleculares , Papaína/química , Papaína/clasificación , Bases de Datos de Proteínas
8.
J Mol Biol ; 435(6): 168012, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36792007

RESUMEN

The Venezuelan equine encephalitis virus (VEEV) belongs to the Togaviridae family and is pathogenic to both humans and equines. The VEEV non-structural protein 2 (nsP2) is a cysteine protease (nsP2pro) that processes the polyprotein and thus it is a drug target for inhibitor discovery. The atomic structure of the VEEV nsP2 catalytic domain was previously characterized by both X-ray crystallography and computational studies. A modified nsP2pro harboring a N475A mutation in the N terminus was observed to exhibit an unexpected conformation: the N-terminal residues bind to the active site, mimicking binding of a substrate. The large conformational change of the N terminus was assumed to be induced by the N475A mutation, as N475 has an important role in stabilization of the N terminus and the active site. This conformation was first observed in the N475A mutant, but we also found it while determining a crystal structure of the catalytically active nsP2pro containing the wild-type N475 active site residue and K741A/K767A surface entropy reduction mutations. This suggests that the N475A mutation is not a prerequisite for self-inhibition. Here, we describe a high resolution (1.46 Å) crystal structure of a truncated nsP2pro (residues 463-785, K741A/K767A) and analyze the structure further by molecular dynamics to study the active and self-inhibited conformations of nsP2pro and its N475A mutant. A comparison of the different conformations of the N-terminal residues sheds a light on the interactions that play an important role in the stabilization of the enzyme.


Asunto(s)
Dominio Catalítico , Proteasas de Cisteína , Virus de la Encefalitis Equina Venezolana , Animales , Humanos , Cristalografía por Rayos X , Proteasas de Cisteína/química , Proteasas de Cisteína/genética , Virus de la Encefalitis Equina Venezolana/enzimología , Caballos , Simulación de Dinámica Molecular
9.
J Chem Inf Model ; 63(3): 950-958, 2023 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-36648276

RESUMEN

Alzheimer's disease represents one of the most ambitious challenges for biomedical sciences due to the growing number of cases worldwide in the elderly population and the lack of efficient treatments. One of the recent attempts to develop a treatment points to the cysteine protease RgpB as a promising drug target. In this attempt, several small-molecule covalent inhibitors of this enzyme have been proposed. Here, we report a computational study at the atomic level of the inhibition mechanism of the most promising reported compounds. Molecular dynamics simulations were performed on six of them, and their binding energies in the active site of the protein were computed. Contact maps and interaction energies were decomposed by residues to disclose those key interactions with the enzyme. Finally, quantum mechanics/molecular mechanics (QM/MM) molecular dynamics (MD) simulations were performed to evaluate the reaction mechanism by which these drug candidates lead to covalently bound complexes, inhibiting the RgpB protease. The results provide a guide for future re-design of prospective and efficient inhibitors for the treatment of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer , Cisteína-Endopeptidasas Gingipaínas , Anciano , Humanos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Proteasas de Cisteína/química , Cisteína-Endopeptidasas Gingipaínas/efectos adversos , Cisteína-Endopeptidasas Gingipaínas/antagonistas & inhibidores , Cisteína-Endopeptidasas Gingipaínas/metabolismo , Simulación de Dinámica Molecular
10.
Acta Crystallogr D Struct Biol ; 77(Pt 12): 1535-1542, 2021 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-34866609

RESUMEN

Legionella pneumophila is a human pathogen that causes Legionnaires' disease, a severe form of pneumonia. It can be found in various aquatic environments ranging from cooling towers to ponds. In addition to causing disease in humans, it can also infect free-living amoebae commonly found in various aquatic environments. Once inside a human lung macrophage, it creates a niche called the Legionella-containing vacuole where it can evade phagolysosomal degradation and replicate. During infection, normal cellular functions are hijacked by proteins that are secreted by the pathogen, called bacterial effectors. Here, the structural characterization of the effector LegA15/AnkD is reported. The protein contains an ankyrin-repeat domain followed by a cysteine protease-like (CPL) domain with a putative catalytic triad consisting of His268-Asn290-Cys361. The CPL domain shows similarity to the CE clan in the MEROPS database, which contains ubiquitin-like hydrolases. The C-terminal segment of LegA15, including the CPL domain, shows structural similarity to another effector, LegA3/AnkH, while they share only 12% sequence identity. When expressed in mammalian cells, LegA15 is localized within the cytoplasm, in contrast to LegA3, which localizes to the nucleus.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteasas de Cisteína/metabolismo , Legionella/metabolismo , Proteínas Bacterianas/química , Proteasas de Cisteína/química , Interacciones Huésped-Patógeno , Legionella/patogenicidad , Conformación Proteica , Dominios Proteicos
11.
Int J Mol Sci ; 22(21)2021 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-34768970

RESUMEN

The papain-like cysteine proteases (PLCPs), the most important group of cysteine proteases, have been reported to participate in the regulation of growth, senescence, and abiotic stresses in plants. However, the functions of PLCPs and their roles in stress response in microalgae was rarely reported. The responses to different abiotic stresses in Haematococcus pluvialis were often observed, including growth regulation and astaxanthin accumulation. In this study, the cDNA of HpXBCP3 containing 1515 bp open reading frame (ORF) was firstly cloned from H. pluvialis by RT-PCR. The analysis of protein domains and molecular evolution showed that HpXBCP3 was closely related to AtXBCP3 from Arabidopsis. The expression pattern analysis revealed that it significantly responds to NaCl stress in H. pluvialis. Subsequently, transformants expressing HpXBCP3 in Chlamydomonas reinhardtii were obtained and subjected to transcriptomic analysis. Results showed that HpXBCP3 might affect the cell cycle regulation and DNA replication in transgenic Chlamydomonas, resulting in abnormal growth of transformants. Moreover, the expression of HpXBCP3 might increase the sensitivity to NaCl stress by regulating ubiquitin and the expression of WD40 proteins in microalgae. Furthermore, the expression of HpXBCP3 might improve chlorophyll content by up-regulating the expression of NADH-dependent glutamate synthases in C. reinhardtii. This study indicated for the first time that HpXBCP3 was involved in the regulation of cell growth, salt stress response, and chlorophyll synthesis in microalgae. Results in this study might enrich the understanding of PLCPs in microalgae and provide a novel perspective for studying the mechanism of environmental stress responses in H. pluvialis.


Asunto(s)
Proteínas Algáceas/metabolismo , Chlorophyceae/enzimología , Proteasas de Cisteína/metabolismo , Microalgas/crecimiento & desarrollo , Microalgas/fisiología , Proteínas Algáceas/química , Proteínas Algáceas/genética , Chlamydomonas reinhardtii/genética , Chlamydomonas reinhardtii/crecimiento & desarrollo , Chlamydomonas reinhardtii/fisiología , Chlorophyceae/genética , Clorofila/biosíntesis , Proteasas de Cisteína/química , Proteasas de Cisteína/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ontología de Genes , Redes y Vías Metabólicas/genética , Redes y Vías Metabólicas/fisiología , Microalgas/genética , Filogenia , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tolerancia a la Sal/genética , Tolerancia a la Sal/fisiología , Estrés Fisiológico/genética , Estrés Fisiológico/fisiología , Transformación Genética
12.
Molecules ; 26(16)2021 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-34443335

RESUMEN

The specificity of inhibition by 6,6'-dihydroxythiobinupharidine (DTBN) on cysteine proteases was demonstrated in this work. There were differences in the extent of inhibition, reflecting active site structural-steric and biochemical differences. Cathepsin S (IC50 = 3.2 µM) was most sensitive to inhibition by DTBN compared to Cathepsin B, L and papain (IC50 = 1359.4, 13.2 and 70.4 µM respectively). DTBN is inactive for the inhibition of Mpro of SARS-CoV-2. Docking simulations suggested a mechanism of interaction that was further supported by the biochemical results. In the docking results, it was shown that the cysteine sulphur of Cathepsin S, L and B was in close proximity to the DTBN thiaspirane ring, potentially forming the necessary conditions for a nucleophilic attack to form a disulfide bond. Covalent docking and molecular dynamic simulations were performed to validate disulfide bond formation and to determine the stability of Cathepsins-DTBN complexes, respectively. The lack of reactivity of DTBN against SARS-CoV-2 Mpro was attributed to a mismatch of the binding conformation of DTBN to the catalytic binding site of Mpro. Thus, gradations in reactivity among the tested Cathepsins may be conducive for a mechanism-based search for derivatives of nupharidine against COVID-19. This could be an alternative strategy to the large-scale screening of electrophilic inhibitors.


Asunto(s)
Alcaloides/farmacología , Proteasas de Cisteína/metabolismo , Alcaloides/química , Animales , Antivirales/farmacología , Sitios de Unión , COVID-19/metabolismo , Dominio Catalítico , Catepsinas/farmacología , Línea Celular Tumoral , Proteasas de Cisteína/química , Inhibidores de Cisteína Proteinasa/química , Inhibidores de Cisteína Proteinasa/farmacología , Humanos , Ratones , Simulación del Acoplamiento Molecular/métodos , Nuphar/química , Papaína/farmacología , Extractos Vegetales/farmacología , Unión Proteica , SARS-CoV-2/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
13.
Vet Res ; 52(1): 113, 2021 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-34446106

RESUMEN

The aim of this study was to investigate the biological properties of a novel gut-specific cysteine protease in Trichinella spiralis (TsGSCP) and its role in larval intrusion, development and fecundity. TsGSCP has a functional C1 peptidase domain; C1 peptidase belongs to cathepsin B family. The TsGSCP gene cloned and expressed in Escherichia coli BL21 showed intensive immunogenicity. qPCR and Western blotting revealed that TsGSCP mRNA and protein were expressed at various T. spiralis stages, but their expression levels in intestinal infectious larvae (IIL) were clearly higher than those in muscle larvae (ML), adult worms (AWs) and new-born larvae (NBL). Indirect immunofluorescence (IIF) analysis showed that TsGSCP was primarily located at the outer cuticle and the intrauterine embryos of this parasite. rTsGSCP showed the ability to specifically bind with IECs, and the binding site is within the IEC cytoplasm. rTsGSCP accelerated larval intrusion into host intestinal epithelial cells (IECs), whereas anti-rTsGSCP antibodies suppressed larval intrusion; the acceleration and suppression was induced by rTsGSCP and anti-rTsGSCP antibodies, respectively, in a dose-dependent manner. When ML were transfected with TsGSCP-specific dsRNA, TsGSCP expression and enzymatic activity were reduced by 46.82 and 37.39%, respectively, and the capacity of the larvae to intrude into IECs was also obviously impeded. Intestinal AW burden and adult female length and fecundity were significantly decreased in the group of mice infected with dsRNA-transfected ML compared to the control dsRNA and PBS groups. The results showed that TsGSCP plays a principal role in gut intrusion, worm development and fecundity in the T. spiralis lifecycle and might be a candidate target for vaccine development against Trichinella intrusion and infection.


Asunto(s)
Proteasas de Cisteína/genética , Proteínas del Helminto/genética , Trichinella spiralis/fisiología , Secuencia de Aminoácidos , Animales , Proteasas de Cisteína/química , Proteasas de Cisteína/metabolismo , Femenino , Fertilidad , Proteínas del Helminto/química , Proteínas del Helminto/metabolismo , Larva/genética , Larva/crecimiento & desarrollo , Larva/metabolismo , Larva/fisiología , Ratones , Filogenia , Alineación de Secuencia/veterinaria , Trichinella spiralis/genética , Trichinella spiralis/crecimiento & desarrollo , Trichinella spiralis/metabolismo , Triquinelosis/veterinaria
14.
Front Immunol ; 12: 680279, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34335582

RESUMEN

Cysteine cathepsins are primarily involved in the degradation and recycling of proteins in endo-lysosomal compartments but are also gaining recognition as pivotal proteolytic contributors to various immune functions. Through their extracellular proteolytic activities within the hematopoietic stem cell niche, they are involved in progenitor cell mobilization and differentiation. Cysteine cathepsins, such as cathepsins L and S contribute to antigen-induced adaptive immunity through major histocompatibility complex class II antigen presentation whereas cathepsin X regulates T-cell migration. By regulating toll-like receptor signaling and cytokine secretion cysteine cathepsins activate innate immune cells and affect their functional differentiation. Cathepsins C and H are expressed in cytotoxic T lymphocytes and natural killer cells and are involved in processing of pro-granzymes into proteolytically active forms. Cytoplasmic activities of cathepsins B and L contribute to the maintenance of homeostasis of the adaptive immune response by regulating cell death of T and B lymphocytes. The expression pattern, localization, and activity of cysteine cathepsins is tightly connected to their function in immune cells. Furthermore, cysteine cathepsins together with their endogenous inhibitors, serve as mediators in the interplay between cancer and immune cells that results in immune cell anergy. The aim of the present article is to review the mechanisms of dysregulation of cysteine cathepsins and their inhibitors in relation to immune dysfunction to address new possibilities for regulation of their function.


Asunto(s)
Diferenciación Celular/inmunología , Proteasas de Cisteína/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Inmunomodulación , Animales , Diferenciación Celular/genética , Anergia Clonal/inmunología , Proteasas de Cisteína/química , Proteasas de Cisteína/genética , Inhibidores de Cisteína Proteinasa/farmacología , Humanos , Tolerancia Inmunológica , Inmunomodulación/efectos de los fármacos , Inmunosenescencia/efectos de los fármacos , Familia de Multigenes , Organogénesis/genética , Organogénesis/inmunología , Relación Estructura-Actividad , Linfocitos T/inmunología , Linfocitos T/metabolismo
15.
Acta Crystallogr D Struct Biol ; 77(Pt 8): 1040-1049, 2021 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-34342277

RESUMEN

The ß-link is a composite protein motif consisting of a G1ß ß-bulge and a type II ß-turn, and is generally found at the end of two adjacent strands of antiparallel ß-sheet. The 1,2-positions of the ß-bulge are also the 3,4-positions of the ß-turn, with the result that the N-terminal portion of the polypeptide chain is orientated at right angles to the ß-sheet. Here, it is reported that the ß-link is frequently found in certain protein folds of the SCOPe structural classification at specific locations where it connects a ß-sheet to another area of a protein. It is found at locations where it connects one ß-sheet to another in the ß-sandwich and related structures, and in small (four-, five- or six-stranded) ß-barrels, where it connects two ß-strands through the polypeptide chain that crosses an open end of the barrel. It is not found in larger (eight-stranded or more) ß-barrels that are straightforward ß-meanders. In some cases it initiates a connection between a single ß-sheet and an α-helix. The ß-link also provides a framework for catalysis in serine proteases, where the catalytic serine is part of a conserved ß-link, and in cysteine proteases, including Mpro of human SARS-CoV-2, in which two residues of the active site are located in a conserved ß-link.


Asunto(s)
Estructura Secundaria de Proteína , Serina Proteasas/química , Secuencias de Aminoácidos , Animales , Dominio Catalítico , Proteasas 3C de Coronavirus/química , Proteasas 3C de Coronavirus/metabolismo , Proteasas de Cisteína/química , Proteasas de Cisteína/metabolismo , Bases de Datos de Proteínas , Humanos , Enlace de Hidrógeno , Modelos Moleculares , SARS-CoV-2/química , SARS-CoV-2/enzimología , Serina Proteasas/metabolismo , Homología Estructural de Proteína
16.
Int J Mol Sci ; 22(6)2021 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-33810118

RESUMEN

Experimental evidence for enzymatic mechanisms is often scarce, and in many cases inadvertently biased by the employed methods. Thus, apparently contradictory model mechanisms can result in decade long discussions about the correct interpretation of data and the true theory behind it. However, often such opposing views turn out to be special cases of a more comprehensive and superior concept. Molecular dynamics (MD) and the more advanced molecular mechanical and quantum mechanical approach (QM/MM) provide a relatively consistent framework to treat enzymatic mechanisms, in particular, the activity of proteolytic enzymes. In line with this, computational chemistry based on experimental structures came up with studies on all major protease classes in recent years; examples of aspartic, metallo-, cysteine, serine, and threonine protease mechanisms are well founded on corresponding standards. In addition, experimental evidence from enzyme kinetics, structural research, and various other methods supports the described calculated mechanisms. One step beyond is the application of this information to the design of new and powerful inhibitors of disease-related enzymes, such as the HIV protease. In this overview, a few examples demonstrate the high potential of the QM/MM approach for sophisticated pharmaceutical compound design and supporting functions in the analysis of biomolecular structures.


Asunto(s)
Simulación de Dinámica Molecular , Péptido Hidrolasas/química , Inhibidores de Proteasas/química , Teoría Cuántica , Algoritmos , Proteasas de Cisteína/química , Proteasas de Cisteína/metabolismo , Metaloproteasas/metabolismo , Estructura Molecular , Péptido Hidrolasas/metabolismo , Inhibidores de Proteasas/farmacología , Conformación Proteica , Serina Proteasas/química , Serina Proteasas/metabolismo , Termodinámica
17.
J Am Chem Soc ; 143(17): 6423-6433, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33885283

RESUMEN

Terminal unactivated alkynes are nowadays considered the golden standard for cysteine-reactive warheads in activity-based probes (ABPs) targeting cysteine deubiquitinating enzymes (DUBs). In this work, we study the versatility of the thiol-alkyne addition reaction in more depth. Contrary to previous findings with UCHL3, we now show that covalent adduct formation can progress with substituents on the terminal or internal alkyne position. Strikingly, acceptance of alkyne substituents is strictly DUB-specific as this is not conserved among members of the same subfamily. Covalent adduct formation with the catalytic cysteine residue was validated by gel analysis and mass spectrometry of intact ABP-treated USP16CDWT and catalytically inactive mutant USP16CDC205A. Bottom-up mass spectrometric analysis of the covalent adduct with a deuterated propargyl ABP provides mechanistic understanding of the in situ thiol-alkyne reaction, identifying the alkyne rather than an allenic intermediate as the reactive species. Furthermore, kinetic analysis revealed that introduction of (bulky/electron-donating) methyl substituents on the propargyl moiety decreases the rate of covalent adduct formation, thus providing a rational explanation for the commonly lower level of observed covalent adduct compared to unmodified alkynes. Altogether, our work extends the scope of possible propargyl derivatives in cysteine targeting ABPs from unmodified terminal alkynes to internal and substituted alkynes, which we anticipate will have great value in the development of ABPs with improved selectivity profiles.


Asunto(s)
Alquinos/química , Proteasas de Cisteína/química , Pargilina/análogos & derivados , Compuestos de Sulfhidrilo/química , Enzimas Desubicuitinizantes/química , Células HEK293 , Humanos , Pargilina/química , Propilaminas/química , Ubiquitina Tiolesterasa/química
18.
Int J Biol Macromol ; 179: 601-609, 2021 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-33713772

RESUMEN

Proteinases with the (chymo)trypsin-like serine/cysteine fold comprise a large superfamily performing their function through the Acid - Base - Nucleophile catalytic triad. In our previous work (Denesyuk AI, Johnson MS, Salo-Ahen OMH, Uversky VN, Denessiouk K. Int J Biol Macromol. 2020;153:399-411), we described a universal three-dimensional (3D) structural motif, NBCZone, that contains eleven amino acids: dipeptide 42 T-43 T, pentapeptide 54 T-55 T-56 T-57 T(base)-58 T, tripeptide 195 T(nucleophile)-196 T-197 T and residue 213 T (T - numeration of amino acids in trypsin). The comparison of the NBCZones among the members of the (chymo)trypsin-like protease family suggested the existence of 15 distinct groups. Within each group, the NBCZones incorporate an identical set of conserved interactions and bonds. In the present work, the structural environment of the catalytic acid at the position 102 T and the fourth member of the "catalytic tetrad" at the position 214 T was analyzed in 169 3D structures of proteinases with the (chymo)trypsin-like serine/cysteine fold. We have identified a complete Structural Catalytic Core (SCC) consisting of two classes and four groups. The proteinases belonging to different classes and groups differ from each other by the nature of the interaction between their N- and C-terminal ß-barrels. Comparative analysis of the 3CLpro(s) from SARS-CoV-2 and SARS-CoV, used as an example, showed that the amino acids at positions 103 T and 179 T affect the nature of the interaction of the "catalytic acid" core (102 T-Core, N-terminal ß-barrel) with the "supplementary" core (S-Core, C-terminal ß-barrel), which ultimately results in the modulation of the enzymatic activity. The reported analysis represents an important standalone contribution to the analysis and systematization of the 3D structures of (chymo)trypsin-like serine/cysteine fold proteinases. The use of the developed approach for the comparison of 3D structures will allow, in the event of the appearance of new representatives of a given fold in the PDB, to quickly determine their structural homologues with the identification of possible differences.


Asunto(s)
Proteasas de Cisteína/química , Serina Proteasas/química , Secuencia de Aminoácidos , Sitios de Unión , COVID-19/metabolismo , Catálisis , Dominio Catalítico , Proteasas de Cisteína/metabolismo , Humanos , Modelos Moleculares , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/química , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo , SARS-CoV-2/química , SARS-CoV-2/metabolismo , Serina Endopeptidasas/química , Serina Endopeptidasas/metabolismo , Serina Proteasas/metabolismo , Tripsina/metabolismo
19.
J Chem Inf Model ; 61(2): 1020-1032, 2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33538596

RESUMEN

Currently the entire human population is in the midst of a global pandemic caused by SARS-CoV-2 (Severe Acute Respiratory Syndrome CoronaVirus 2). This highly pathogenic virus has to date caused >71 million infections and >1.6 million deaths in >180 countries. Several vaccines and drugs are being studied as possible treatments or prophylactics of this viral infection. M3CLpro (coronavirus main cysteine protease) is a promising drug target as it has a significant role in viral replication. Here we use the X-ray crystal structure of M3CLpro in complex with boceprevir to study the dynamic changes of the protease upon ligand binding. The binding free energy was calculated for water molecules at different locations of the binding site, and molecular dynamics (MD) simulations were carried out for the M3CLpro/boceprevir complex, to thoroughly understand the chemical environment of the binding site. Several HCV NS3/4a protease inhibitors were tested in vitro against M3CLpro. Specifically, asunaprevir, narlaprevir, paritaprevir, simeprevir, and telaprevir all showed inhibitory effects on M3CLpro. Molecular docking and MD simulations were then performed to investigate the effects of these ligands on M3CLpro and to provide insights into the chemical environment of the ligand binding site. Our findings and observations are offered to help guide the design of possible potent protease inhibitors and aid in coping with the COVID-19 pandemic.


Asunto(s)
Antivirales/farmacología , Proteasas de Cisteína/química , SARS-CoV-2/efectos de los fármacos , Inhibidores de Serina Proteinasa/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Simulación por Computador , Cristalografía por Rayos X , Proteasas de Cisteína/efectos de los fármacos , Humanos , Técnicas In Vitro , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Conformación Proteica , SARS-CoV-2/enzimología , Serina Proteasas
20.
Sci Rep ; 11(1): 4573, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33633359

RESUMEN

Thermostability and substrate specificity of proteases are major factors in their industrial applications. rEla is a novel recombinant cysteine protease obtained from a thermophilic bacterium, Cohnella sp.A01 (PTCC No: 1921). Herein, we were interested in recombinant production and characterization of the enzyme and finding the novel features in comparison with other well-studied cysteine proteases. The bioinformatics analysis showed that rEla is allosteric cysteine protease from DJ-1/ThiJ/PfpI superfamily. The enzyme was heterologously expressed and characterized and the recombinant enzyme molecular mass was 19.38 kD which seems to be smaller than most of the cysteine proteases. rEla exhibited acceptable activity in broad pH and temperature ranges. The optimum activity was observed at 50℃ and pH 8 and the enzyme showed remarkable stability by keeping 50% of residual activity after 100 days storage at room temperature. The enzyme Km and Vmax values were 21.93 mM, 8 U/ml, respectively. To the best of our knowledge, in comparison with the other characterized cysteine proteases, rEla is the only reported cysteine protease with collagen specificity. The enzymes activity increases up to 1.4 times in the presence of calcium ion (2 mM) suggesting it as the enzyme's co-factor. When exposed to surfactants including Tween20, Tween80, Triton X-100 and SDS (1% and 4% v/v) the enzyme activity surprisingly increased up to 5 times.


Asunto(s)
Bacillales/enzimología , Proteasas de Cisteína/metabolismo , Secuencia de Aminoácidos , Bacillales/efectos de los fármacos , Bacillales/genética , Sitios de Unión , Proteasas de Cisteína/química , Proteasas de Cisteína/genética , Activación Enzimática/efectos de los fármacos , Estabilidad de Enzimas/efectos de los fármacos , Concentración de Iones de Hidrógeno , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Filogenia , Unión Proteica , Conformación Proteica , Análisis de Secuencia de ADN , Relación Estructura-Actividad , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...