Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 509
Filtrar
1.
Molecules ; 26(16)2021 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-34443484

RESUMO

The COVID-19 outbreak has rapidly spread on a global scale, affecting the economy and public health systems throughout the world. In recent years, peptide-based therapeutics have been widely studied and developed to treat infectious diseases, including viral infections. Herein, the antiviral effects of the lysine linked dimer des-Cys11, Lys12,Lys13-(pBthTX-I)2K ((pBthTX-I)2K)) and derivatives against SARS-CoV-2 are reported. The lead peptide (pBthTX-I)2K and derivatives showed attractive inhibitory activities against SARS-CoV-2 (EC50 = 28-65 µM) and mostly low cytotoxic effect (CC50 > 100 µM). To shed light on the mechanism of action underlying the peptides' antiviral activity, the Main Protease (Mpro) and Papain-Like protease (PLpro) inhibitory activities of the peptides were assessed. The synthetic peptides showed PLpro inhibition potencies (IC50s = 1.0-3.5 µM) and binding affinities (Kd = 0.9-7 µM) at the low micromolar range but poor inhibitory activity against Mpro (IC50 > 10 µM). The modeled binding mode of a representative peptide of the series indicated that the compound blocked the entry of the PLpro substrate toward the protease catalytic cleft. Our findings indicated that non-toxic dimeric peptides derived from the Bothropstoxin-I have attractive cellular and enzymatic inhibitory activities, thereby suggesting that they are promising prototypes for the discovery and development of new drugs against SARS-CoV-2 infection.


Assuntos
Venenos de Crotalídeos/química , Dimerização , Papaína/antagonistas & inibidores , Peptídeos/química , Peptídeos/farmacologia , SARS-CoV-2/enzimologia , Antivirais/química , Antivirais/metabolismo , Antivirais/farmacologia , Simulação de Acoplamento Molecular , Papaína/química , Papaína/metabolismo , Peptídeos/metabolismo , Inibidores de Proteases/química , Inibidores de Proteases/metabolismo , Inibidores de Proteases/farmacologia , Conformação Proteica , SARS-CoV-2/efeitos dos fármacos
2.
Biochem Biophys Res Commun ; 546: 192-199, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33618285

RESUMO

Environmental allergen sources such as house dust mites contain proteases, which are frequently allergens themselves. Inhalation with the exogenous proteases, such as a model of protease allergen, papain, to airways evokes release and activation of IL-33, which promotes innate and adaptive allergic airway inflammation and Th2 sensitization in mice. Here, we examine whether epicutaneous (e.c.) vaccination with antigens with and without protease activity shows prophylactic effect on the Th airway sensitization and Th2-medated airway inflammation, which are driven by exogenous or endogenous IL-33. E.c. vaccination with ovalbumin restrained ovalbumin-specific Th2 airway sensitization and/or airway inflammation on subsequent inhalation with ovalbumin plus papain or ovalbumin plus recombinant IL-33. E.c. vaccination with papain or protease inhibitor-treated papain restrained papain-specific Th2 and Th9 airway sensitization, eosinophilia, and infiltration of IL-33-responsive Th2 and group 2 innate lymphoid cells on subsequent inhalation with papain. However, e.c. vaccination with papain but not protease inhibitor-treated papain induced Th17 response in bronchial draining lymph node cells. In conclusions, we demonstrated that e.c. allergen vaccination via intact skin in mice restrained even protease allergen-activated IL-33-driven airway Th2 sensitization to attenuate allergic airway inflammation and that e.c. vaccination with protease allergen attenuated the airway inflammation similar to its derivative lacking the protease activity, although the former but not the latter promoted Th17 development. In addition, the present study suggests that modified allergens, of which Th17-inducing e.c. adjuvant activity such as the protease activity was eliminated, might be preferable for safer clinical applications of the e.c. allergen administration.


Assuntos
Inflamação/imunologia , Ovalbumina/imunologia , Papaína/antagonistas & inibidores , Papaína/imunologia , Células Th17 , Células Th2/imunologia , Vacinação/métodos , Administração por Inalação , Animais , Feminino , Imunoglobulina E/imunologia , Inflamação/prevenção & controle , Mediadores da Inflamação/imunologia , Interleucina-33/administração & dosagem , Interleucina-33/imunologia , Camundongos , Ovalbumina/administração & dosagem , Ovalbumina/sangue , Papaína/administração & dosagem , Células Th17/imunologia
3.
Chem Pharm Bull (Tokyo) ; 68(11): 1074-1081, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33132374

RESUMO

Fragment-based approach combined with electrophilic reactive compounds is a powerful strategy to discover novel covalent ligands for protein target. However, the promiscuous reactivity often interferes with identification of the fragments possessing specific binding affinity to the targeted protein. In our study, we report the fragment-based covalent drug discovery using the chemically tuned weak reactivity of chlorofluoroacetamide (CFA). We constructed a small fragment library composed of 30 CFA-appended compounds and applied it to the covalent ligand screening for cysteine protease papain as a model protein target. Using the fluorescence enzymatic assay, we identified CFA-benzothiazole 30 as a papain inhibitor, which was found to irreversibly inactivate papain upon enzyme kinetic analysis. The formation of the covalent papain-30 adduct was confirmed using electrospray ionization mass spectrometry analysis. The activity-based protein profiling (ABPP) experiment using an alkynylated analog of 30 (i.e., 30-yne) revealed that 30-yne covalently labeled papain with high selectivity. These data demonstrate potential utility of the CFA-fragment library for de novo discovery of target selective covalent inhibitors.


Assuntos
Acetamidas/química , Cisteína Proteases/química , Inibidores de Cisteína Proteinase/química , Acetamidas/metabolismo , Cromatografia Líquida de Alta Pressão , Cisteína Proteases/metabolismo , Inibidores de Cisteína Proteinase/metabolismo , Descoberta de Drogas , Corantes Fluorescentes/química , Cinética , Ligantes , Papaína/antagonistas & inibidores , Papaína/metabolismo , Espectrometria de Massas por Ionização por Electrospray
4.
J Nat Prod ; 83(10): 2983-2995, 2020 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-32998509

RESUMO

Streptomyces mobaraensis produces the papain inhibitor SPI consisting of a 12 kDa protein and small active compounds (SPIac). Purification of the papain inhibitory compounds resulted in four diverse chymostatin derivatives that were characterized by NMR and MS analysis. Chymostatins are hydrophobic tetrapeptide aldehydes from streptomycetes, e.g., S. lavendulae and S. hygroscopicus, that reverse chymosin-mediated angiotensin activation and inhibit other serine and cysteine proteases. Chymotrypsin and papain were both inhibited by the SPIac compounds in the low nanomolar range. SPIac differs from the characterized chymostatins by the exchange of phenylalanine for tyrosine. The crystal structure of one of these chymostatin variants confirmed its molecular structure and revealed a S-configured hemithioacetal bond with the catalytic Cys25 thiolate as well as close interactions with hydrophobic S1 and S2 subsite amino acids. A model for chymostatin biosynthesis is provided based on the discovery of clustered genes encoding several putative nonribosomal peptide synthetases; among them, there is the unusual CstF enzyme that accommodates two canonical amino acid activation domains as well as three peptide carrier protein domains.


Assuntos
Inibidores Enzimáticos/farmacologia , Oligopeptídeos , Papaína/antagonistas & inibidores , Streptomyces , Aldeídos , Sequência de Aminoácidos , Vias Biossintéticas , Hidroxilação , Modelos Moleculares , Estrutura Molecular , Peptídeo Sintases , Especificidade por Substrato
5.
J Chem Phys ; 153(11): 115101, 2020 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-32962355

RESUMO

Broad-spectrum antiviral drugs are urgently needed to stop the Coronavirus Disease 2019 pandemic and prevent future ones. The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is related to the SARS-CoV and Middle East respiratory syndrome coronavirus (MERS-CoV), which have caused the previous outbreaks. The papain-like protease (PLpro) is an attractive drug target due to its essential roles in the viral life cycle. As a cysteine protease, PLpro is rich in cysteines and histidines, and their protonation/deprotonation modulates catalysis and conformational plasticity. Here, we report the pKa calculations and assessment of the proton-coupled conformational dynamics of SARS-CoV-2 in comparison to SARS-CoV and MERS-CoV PLpros using the recently developed graphical processing unit (GPU)-accelerated implicit-solvent continuous constant pH molecular dynamics method with a new asynchronous replica-exchange scheme, which allows computation on a single GPU card. The calculated pKa's support the catalytic roles of the Cys-His-Asp triad. We also found that several residues can switch protonation states at physiological pH among which is C270/271 located on the flexible blocking loop 2 (BL2) of SARS-CoV-2/CoV PLpro. Simulations revealed that the BL2 can open and close depending on the protonation state of C271/270, consistent with the most recent crystal structure evidence. Interestingly, despite the lack of an analogous cysteine, BL2 in MERS-CoV PLpro is also very flexible, challenging a current hypothesis. These findings are supported by the all-atom fixed-charge simulations and provide a starting point for more detailed studies to assist the structure-based design of broad-spectrum inhibitors against CoV PLpros.


Assuntos
Antivirais/farmacologia , Betacoronavirus/enzimologia , Desenho de Fármacos , Coronavírus da Síndrome Respiratória do Oriente Médio/enzimologia , Simulação de Dinâmica Molecular , Papaína/química , Papaína/metabolismo , Prótons , Sequência de Aminoácidos , Histidina , Concentração de Íons de Hidrogênio , Papaína/antagonistas & inibidores , Domínios Proteicos , SARS-CoV-2
6.
Bioessays ; 42(11): e2000094, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32815593

RESUMO

More than 15 million people have been affected by coronavirus disease 2019 (COVID-19) and it has caused 640 016 deaths as of July 26, 2020. Currently, no effective treatment option is available for COVID-19 patients. Though many drugs have been proposed, none of them has shown particular efficacy in clinical trials. In this article, the relationship between the Adrenergic system and the renin-angiotensin-aldosterone system (RAAS) is focused in COVID-19 and a vicious circle consisting of the Adrenergic system-RAAS-Angiotensin converting enzyme 2 (ACE2)-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (which is referred to as the "ARAS loop") is proposed. Hyperactivation of the ARAS loop may be the underlying pathophysiological mechanism in COVID-19, and beta-adrenergic blockers are proposed as a potential treatment option. Beta-adrenergic blockers may decrease the SARS-CoV-2 cellular entry by decreasing ACE2 receptors expression and cluster of differentiation 147 (CD147) in various cells in the body. Beta-adrenergic blockers may decrease the morbidity and mortality in COVID-19 patients by preventing or reducing acute respiratory distress syndrome (ARDS) and other complications. Retrospective and prospective clinical trials should be conducted to check the validity of the hypothesis. Also see the video abstract here https://youtu.be/uLoy7do5ROo.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/uso terapêutico , Infecções por Coronavirus/tratamento farmacológico , Pneumonia Viral/tratamento farmacológico , Enzima de Conversão de Angiotensina 2 , Betacoronavirus/efeitos dos fármacos , Betacoronavirus/fisiologia , COVID-19 , Carvedilol/farmacologia , Carvedilol/uso terapêutico , Infecções por Coronavirus/epidemiologia , Proteases Semelhantes à Papaína de Coronavírus , Reposicionamento de Medicamentos/métodos , Humanos , Inflamassomos/efeitos dos fármacos , Inflamassomos/metabolismo , Mediadores da Inflamação/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Pandemias , Papaína/antagonistas & inibidores , Papaína/metabolismo , Peptidil Dipeptidase A/efeitos dos fármacos , Peptidil Dipeptidase A/metabolismo , Pneumonia Viral/epidemiologia , Inibidores de Proteases/farmacologia , Inibidores de Proteases/uso terapêutico , Embolia Pulmonar/prevenção & controle , Sistema Renina-Angiotensina/efeitos dos fármacos , Sistema Renina-Angiotensina/fisiologia , Insuficiência Respiratória/prevenção & controle , SARS-CoV-2 , Choque Séptico/prevenção & controle , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/metabolismo , Internalização do Vírus/efeitos dos fármacos
7.
Planta ; 252(2): 16, 2020 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-32661769

RESUMO

MAIN CONCLUSION: A new Piper nigrum cysteine proteinase inhibitor, PnCPI, belonging to group I of phytocystatins, with inhibitory activity against papain and growth of Fusarium solani f. sp. piperis, was isolated and characterized. Previous studies (de Souza et al. 2011) have identified a partial cDNA sequence of putative cysteine proteinase inhibitor differentially expressed in roots of black pepper (P. nigrum L.) infected by F. solani f. sp. piperis. Here, we aimed to isolate the full-length cDNA and genomic sequences of the P. nigrum cysteine proteinase inhibitor gene, named PnCPI. Sequence analyses showed that the PnCPI gene encodes a deduced protein of 108 amino acid residues with a predicted molecular mass of 12.3 kDa and isoelectric point of 6.51. Besides the LARFAV-like sequence, common to all phytocystatins, PnCPI contains three conserved motifs of the superfamily cystatin: a glycine residue at the N-terminal region, the QxVxG reactive site more centrally positioned, and one tryptophan in the C-terminal region. PnCPI, belonging to group I of phytocystatins, showed high identity with cystatins isolated from several plant species. Sequence analyses also revealed no putative signal peptide at the N-terminal of PnCPI, as well as no introns within the genomic sequence corresponding to the PnCPI coding region. Molecular modeling showed the ability of PnCPI to interact with papain, while its inhibitory activity against this protease was confirmed after heterologous expression in Escherichia coli. The effects of heat treatments on the inhibitory activity of recombinant PnCPI, rPnCPI, were evaluated. In addition, rPnCPI exhibited in vitro activity against F. solani f. sp. piperis, revealing a new cystatin with the potential antifungal application. The identification of PnCPI as a functional cystatin able to inhibit the in vitro growth of F. solani f. sp. piperis indicates other factors contributing to in vivo susceptibility of black pepper to root rot disease.


Assuntos
Antifúngicos/farmacologia , Cistatinas/farmacologia , Inibidores de Cisteína Proteinase/farmacologia , Fusarium/efeitos dos fármacos , Papaína/antagonistas & inibidores , Piper nigrum/genética , Doenças das Plantas/prevenção & controle , Antifúngicos/isolamento & purificação , Clonagem Molecular , Inibidores de Cisteína Proteinase/isolamento & purificação , DNA Complementar/genética , Fusarium/enzimologia , Piper nigrum/química , Doenças das Plantas/microbiologia
8.
Parasit Vectors ; 13(1): 41, 2020 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-31996242

RESUMO

BACKGROUND: Naegleria fowleri is a free-living amoeba that causes an opportunistic fatal infection known as primary amoebic meningoencephalitis (PAM) in humans. Cysteine proteases produced by the amoeba may play critical roles in the pathogenesis of infection. In this study, a novel cysteine protease inhibitor of N. fowleri (fowlerstefin) was characterized to elucidate its biological function as an endogenous cysteine protease inhibitor of the parasite as well as a pathogenic molecule that induces immune responses in microglial cells. METHODS: Recombinant fowlerstefin was expressed in Escherichia coli. The inhibitory activity of fowlerstefin against several cysteine proteases, including human cathepsins B and L, papain and NfCPB-L, was analyzed. Fowlerstefin-induced pro-inflammatory response in BV-2 microglial cells was anayzed by cytokine array assay, reverse transcription polymerase chain reaction, and enzyme-linked immunosorbent assay. RESULTS: Fowlerstefin is a cysteine protease inhibitor with a monomeric structure, and belongs to the stefin family. Recombinant fowlerstefin effectively inhibited diverse cysteine proteases including cathepsin B-like cysteine proteases of N. fowleri (NfCPB-L), human cathepsins B and L, and papain. Expression of fowlerstefin in the amoeba was optimal during the trophozoite stage and gradually decreased in cysts. Fowlerstefin induced an inflammatory response in BV-2 microglial cells. Fowlerstefin induced the expression of several pro-inflammatory cytokines and chemokines including IL-6 and TNF in BV-2 microglial cells. Fowlerstefin-induced expression of IL-6 and TNF in BV-2 microglial cells was regulated by mitogen-activated protein kinase (MAPKs). The inflammatory response induced by fowlerstefin in BV-2 microglial cells was downregulated via inhibition of NF-κB and AP-1. CONCLUSIONS: Fowlerstefin is a pathogenic molecule that stimulates BV-2 microglial cells to produce pro-inflammatory cytokines through NF-κB- and AP-1-dependent MAPK signaling pathways. Fowlerstefin-induced inflammatory cytokines exacerbate the inflammatory response in N. fowleri-infected areas and contribute to the pathogenesis of PAM.


Assuntos
Infecções Protozoárias do Sistema Nervoso Central/parasitologia , Cistatinas/farmacologia , Inibidores de Cisteína Proteinase/farmacologia , Microglia/efeitos dos fármacos , Naegleria fowleri/metabolismo , Análise de Variância , Animais , Anticorpos Antiprotozoários/biossíntese , Anticorpos Antiprotozoários/imunologia , Anticorpos Antiprotozoários/isolamento & purificação , Especificidade de Anticorpos , Catepsina B/antagonistas & inibidores , Catepsina L/antagonistas & inibidores , Linhagem Celular , Cistatinas/química , Inibidores de Cisteína Proteinase/química , Inibidores de Cisteína Proteinase/imunologia , Citocinas/metabolismo , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Humanos , Concentração de Íons de Hidrogênio , Imunoglobulina G/biossíntese , Imunoglobulina G/imunologia , Imunoglobulina G/isolamento & purificação , Camundongos , Camundongos Endogâmicos BALB C , Microglia/imunologia , Microglia/patologia , Naegleria fowleri/classificação , Naegleria fowleri/genética , Papaína/antagonistas & inibidores , Filogenia , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia
9.
Biochim Biophys Acta Proteins Proteom ; 1867(9): 854-865, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31247344

RESUMO

Falcipain-2(FP2), a cysteine protease from Plasmodium falciparum, cleaves host erythrocyte hemoglobin and specific membrane skeleton components during the parasite life cycle. Therefore its inhibition has been considered as an attractive approach to combat the disease. SerpinB3 (SPB3) belongs to the ovalbumin-serpin family and is a potent cross-class inhibitor of cysteine cathepsins L, K, S and papain. This study explored the possibility of inhibition of FP2 by SPB3. It turned out that general proteolytic activities as well as specific hemoglobinolytic activity of FP2 have been inhibited by SPB3. Furthermore, studies have been designed to investigate and characterize the mechanism of inhibition in comparison with proteases Cathepsin L (CTSL) and papain. The Ki value of inhibition for FP2, measured against its specific substrate (VLK-pNA), is 338.11 nM and stoichiometry (I/E ratio) of inhibition is 1. These values are comparable to CTSL and papain. Analytical gel filtration profile and CD spectroscopy data confirm FP2-SPB3 complex formation. Our studies revealed that interaction of SPB3 with FP2 is non-covalent type like that of CTSL and papain but unlike other serine protease-inhibiting serpins. An in-silico docking and simulation study have been performed with FP2 as well as CTSL and results suggest different binding mode for FP2 and CTSL, though both the complexes are stable with significant contribution from electrostatic energy of interaction. We further showed a disease state mutant SPB3-Gly351Ala performed better anti-protease activity against FP2. This study, for the first time, has shown a serpin family inhibitor from human could efficiently inhibit activity of FP2.


Assuntos
Antígenos de Neoplasias/química , Cisteína Endopeptidases/química , Simulação de Acoplamento Molecular , Plasmodium falciparum/enzimologia , Serpinas/química , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Catepsina L/antagonistas & inibidores , Catepsina L/química , Catepsina L/genética , Catepsina L/metabolismo , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Humanos , Papaína/antagonistas & inibidores , Papaína/química , Papaína/genética , Papaína/metabolismo , Plasmodium falciparum/genética , Serpinas/genética , Serpinas/metabolismo
10.
FEBS Lett ; 593(12): 1313-1325, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31077354

RESUMO

Cystatins are important regulators of papain-like cysteine proteases. In the protozoan parasite Giardia intestinalis, papain-like cysteine proteases play an essential role in the parasite's biology and pathogenicity. Here, we characterized a cysteine protease inhibitor of G. intestinalis that belongs to type-I-cystatins. The parasite cystatin is shown to be a strong inhibitor of papain (Ki  ≈ 0.3 nm) and three parasite cysteine proteases (CP14019, CP16160 and CP16779, Ki  ≈ 0.9-5.8 nm), but a weaker inhibitor of human cathepsin B (Ki  ≈ 79.9 nm). The protein localizes mainly in the cytoplasm. Together, these data suggest that cystatin of G. intestinalis plays a role in the regulation of cysteine protease activities in the parasite and, possibly, in the interaction with the host.


Assuntos
Catepsina B/antagonistas & inibidores , Cistatinas/farmacologia , Inibidores de Cisteína Proteinase/farmacologia , Giardia lamblia/metabolismo , Papaína/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Cistatinas/química , Giardia lamblia/enzimologia , Interações Hospedeiro-Parasita , Humanos , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Homologia de Sequência de Aminoácidos
11.
Protein Expr Purif ; 158: 36-43, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30807851

RESUMO

Insect-derived serine protease inhibitors (serpins) exhibit multiple inhibitory activities. Todate some functional roles for serpins in Hyphantria cunea Drury have been identified. Here, new functional features of the H. cunea serine protease inhibitor (dHC-serpin) were characterized. In this study, the cDNA encoding serpin was amplified from H. cunea (dHC) pupa fat body total RNA using RT-PCR. The full-length dHC-serpin cDNA encoded a protein of 440 amino acids with a predicted 19-amino acid signal peptide and a 421-amino acid functional domain. The functional domain was cloned into a pSUMO vector and transformed into Escherichia coli, resulting in the production of a pSUMO-dHC-serpin fusion protein. The soluble form of this protein was then purified by Ni-IDA chromatography. The SUMO-dHC-serpin fusion protein was then cleaved using a SUMO protease and purified again by Ni-IDA chromatography. dHC-serpin did not inhibit trypsin, elastase, proteinase K or cathepsin B, but strongly inhibited papain. The inhibitor retained its inhibitory activity over a broad range of pH (pH 2-12), temperature (20-50 °C), and DTT concentration (up to 100 mM). A complete loss of inhibitory activity was observed at pH 13 and 70 °C. Serpins generally serve as inhibitors that use a mobile reactive center loop (RCL) as bait to trap protease targets. dHC-serpin, like others serpins, binds papain using the RCL structure.


Assuntos
Proteínas de Insetos , Mariposas , Papaína , Proteínas Recombinantes de Fusão , Proteína SUMO-1 , Serpinas , Animais , Proteínas de Insetos/biossíntese , Proteínas de Insetos/química , Proteínas de Insetos/isolamento & purificação , Mariposas/química , Mariposas/genética , Papaína/antagonistas & inibidores , Papaína/química , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteína SUMO-1/biossíntese , Proteína SUMO-1/química , Proteína SUMO-1/genética , Proteína SUMO-1/isolamento & purificação , Serpinas/biossíntese , Serpinas/química , Serpinas/genética , Serpinas/isolamento & purificação
12.
Mar Drugs ; 16(12)2018 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-30501116

RESUMO

Cystatins are a family of cysteine protease inhibitors which are associated with a variety of physiological and pathological processes in vivo. In the present study, the cDNA sequence of a cystatin F homologue called Lm-cystatin F was cloned from the buccal glands of Lampetra morii. Although Lm-cystatin F shares a lower homology with cystatin superfamily members, it is also composed of a signal peptide and three highly conserved motifs, including the G in the N-terminal, QXVXG, as well as the PW in the C-terminal of the sequence. After sequence optimization and recombination, the recombinant protein was expressed as a soluble protein in Escherichia coli with a molecular weight of 19.85 kDa. Through affinity chromatography and mass spectrometry analysis, the purified protein was identified as a recombinant Lm-cystatin F (rLm-cystatin F). Additionally, rLm-cystatin F could inhibit the activity of papain. Based on MTT assay, rLm-cystatin F inhibited the proliferation of human umbilical vein endothelial cells (HUVECs) dose dependently with an IC50 of 5 µM. In vitro studies show that rLm-cystatin F suppressed the adhesion, migration, invasion, and tube formation of HUVECs, suggesting that rLm-cystatin F possesses anti-angiogenic activity, which provides information on the feeding mechanisms of Lampetra morii and insights into the application of rLm-cystatin F as a potential drug in the future.


Assuntos
Inibidores da Angiogênese/farmacologia , Cistatinas/farmacologia , Lampreias , Neovascularização Fisiológica/efeitos dos fármacos , Papaína/antagonistas & inibidores , Motivos de Aminoácidos , Sequência de Aminoácidos , Inibidores da Angiogênese/química , Inibidores da Angiogênese/genética , Inibidores da Angiogênese/isolamento & purificação , Animais , Proliferação de Células/efeitos dos fármacos , Cistatinas/química , Cistatinas/genética , Cistatinas/isolamento & purificação , DNA Complementar , Células Endoteliais da Veia Umbilical Humana , Humanos , Concentração Inibidora 50 , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/farmacologia , Homologia de Sequência de Aminoácidos
13.
Antiviral Res ; 158: 199-205, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30138642

RESUMO

Porcine epidemic diarrhea virus (PEDV) is a coronavirus (CoV) discovered in the 1970s that infects the intestinal tract of pigs, resulting in diarrhea and vomiting. It can cause extreme dehydration and death in neonatal piglets. In Asia, modified live attenuated vaccines have been used to control PEDV infection in recent years. However, a new strain of PEDV that belongs to genogroup 2a appeared in the USA in 2013 and then quickly spread to Canada and Mexico as well as Asian and European countries. Due to the less effective protective immunity provided by the vaccines against this new strain, it has caused considerable agricultural and economic loss worldwide. The emergence of this new strain increases the importance of understanding PEDV as well as strategies for inhibiting it. Coronaviral proteases, including main proteases and papain-like proteases, are ideal antiviral targets because of their essential roles in viral maturation. Here we provide a first description of the expression, purification and structural characteristics of recombinant PEDV papain-like protease 2, moreover present our finding that 6-thioguanine, a chemotherapeutic drug, in contrast to its competitive inhibition on SARS- and MERS-CoV papain-like proteases, is a noncompetitive inhibitor of PEDV papain-like protease 2.


Assuntos
Antivirais/farmacologia , Papaína/antagonistas & inibidores , Vírus da Diarreia Epidêmica Suína/efeitos dos fármacos , Tioguanina/farmacologia , Sítios de Ligação/efeitos dos fármacos , Coronavirus/efeitos dos fármacos , Infecções por Coronavirus , Proteases Semelhantes à Papaína de Coronavírus , Cinética , Simulação de Acoplamento Molecular , Papaína/química , Papaína/genética , Papaína/isolamento & purificação , Vírus da Diarreia Epidêmica Suína/genética , Conformação Proteica/efeitos dos fármacos , Proteínas Recombinantes
14.
J Virol ; 92(19)2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30021905

RESUMO

Middle East respiratory syndrome coronavirus (MERS-CoV) utilizes host cellular proteases to enter cells. A previous report shows that furin, which is distributed mainly in the Golgi apparatus and cycled to the cell surface and endosomes, proteolytically activates the MERS-CoV spike (S) protein following receptor binding to mediate fusion between the viral and cellular membranes. In this study, we reexamined furin usage by MERS-CoV using a real-time PCR-based virus cell entry assay after inhibition of cellular proteases. We found that the furin inhibitor dec-RVKR-CMK blocked entry of MERS-CoV harboring an S protein lacking furin cleavage sites; it even blocked entry into furin-deficient LoVo cells. In addition, dec-RVKR-CMK inhibited not only the enzymatic activity of furin but also those of cathepsin L, cathepsin B, trypsin, papain, and TMPRSS2. Furthermore, a virus cell entry assay and a cell-cell fusion assay provided no evidence that the S protein was activated by exogenous furin. Therefore, we conclude that furin does not play a role in entry of MERS-CoV into cells and that the inhibitory effect of dec-RVKR-CMK is specific for TMPRSS2 and cathepsin L rather than furin.IMPORTANCE Previous studies using the furin inhibitor dec-RVKR-CMK suggest that MERS-CoV utilizes a cellular protease, furin, to activate viral glycoproteins during cell entry. However, we found that dec-RVKR-CMK inhibits not only furin but also other proteases. Furthermore, we found no evidence that MERS-CoV uses furin. These findings suggest that previous studies in the virology field based on dec-RVKR-CMK should be reexamined carefully. Here we describe appropriate experiments that can be used to assess the effect of protease inhibitors on virus cell entry.


Assuntos
Furina/metabolismo , Coronavírus da Síndrome Respiratória do Oriente Médio/metabolismo , Proteólise , Glicoproteína da Espícula de Coronavírus/metabolismo , Internalização do Vírus , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Catepsina B/antagonistas & inibidores , Catepsina B/genética , Catepsina B/metabolismo , Catepsina L/antagonistas & inibidores , Catepsina L/genética , Catepsina L/metabolismo , Chlorocebus aethiops , Furina/antagonistas & inibidores , Furina/genética , Humanos , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Papaína/antagonistas & inibidores , Papaína/genética , Papaína/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Glicoproteína da Espícula de Coronavírus/genética , Células Vero
15.
J Biol Chem ; 293(34): 13151-13165, 2018 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-29967063

RESUMO

Protein activity is often regulated by altering the oligomerization state. One mechanism of multimerization involves domain swapping, wherein proteins exchange parts of their structures and thereby form long-lived dimers or multimers. Domain swapping has been specifically observed in amyloidogenic proteins, for example the cystatin superfamily of cysteine protease inhibitors. Cystatins are twin-headed inhibitors, simultaneously targeting the lysosomal cathepsins and legumain, with important roles in cancer progression and Alzheimer's disease. Although cystatin E is the most potent legumain inhibitor identified so far, nothing is known about its propensity to oligomerize. In this study, we show that conformational destabilization of cystatin E leads to the formation of a domain-swapped dimer with increased conformational stability. This dimer was active as a legumain inhibitor by forming a trimeric complex. By contrast, the binding sites toward papain-like proteases were buried within the cystatin E dimer. We also showed that the dimers could further convert to amyloid fibrils. Unexpectedly, cystatin E amyloid fibrils contained functional protein, which inhibited both legumain and papain-like enzymes. Fibril formation was further regulated by glycosylation. We speculate that cystatin amyloid fibrils might serve as a binding platform to stabilize the pH-sensitive legumain and cathepsins in the extracellular environment, contributing to their physiological and pathological functions.


Assuntos
Amiloide/química , Cistatina M/química , Cistatina M/metabolismo , Papaína/antagonistas & inibidores , Multimerização Proteica , Sítios de Ligação , Cristalografia por Raios X , Humanos , Modelos Moleculares , Conformação Proteica , Relação Estrutura-Atividade
16.
Food Chem ; 265: 1-8, 2018 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-29884359

RESUMO

The autolysis of pretreated bovine skin (PBS) (treated with 0.1 M NaOH and 1% HCl), its endogenous proteases, inhibitors and their effects on quality attributes of gelatin were examined. PBS was subjected to different temperatures (20-90 °C) and pH (2-9) and treated with different protease inhibitors. Maximum autolytic activity of PBS was observed at 40 °C and pH 5. Ethylene-bis (oxyethylenenitrilo) tetraacetic acid (EGTA) was the most effective in impeding the degradation of γ-, ß- and α- chains of PBS protein indicating that metallocollagenases were the predominant endogenous proteases in bovine skin. Gelatin was extracted in the absence (GAE) and presence (GPE) of EGTA, and EGTA with papain enzyme (GPEP). GPEP had a higher yield and lower gel strength than GEA and GPE. Metallocollagenases partook in the degradation of gelatin thereby affecting its functional properties. Pretreating PBS with or without EGTA, and papain influenced the quality attributes of gelatin.


Assuntos
Gelatina/metabolismo , Papaína/antagonistas & inibidores , Papaína/metabolismo , Proteólise , Animais , Bovinos , Ácido Egtázico/farmacologia , Gelatina/química , Gelatina/isolamento & purificação , Proteólise/efeitos dos fármacos
17.
Angew Chem Int Ed Engl ; 57(27): 8022-8026, 2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29469231

RESUMO

Fluorobenzene probes for protein profiling through selective cysteine labeling have been developed by rational reactivity tuning. Tuning was achieved by selecting an electron-withdrawing para substituent in combination with variation of the number of fluorine substituents. Optimized probes chemoselectively arylated cysteine residues in proteins under aqueous conditions. Probes linked to azide, biotin, or a fluorophore were applicable to labeling of eGFP and albumin. Selective inhibition of cysteine proteases was also demonstrated with the probes. Additionally, probes were tuned for site-selective labeling of cysteine residues and for activity-based protein profiling in cell lysates.


Assuntos
Cisteína/química , Fluorbenzenos/química , Proteínas de Fluorescência Verde/química , Soroalbumina Bovina/química , Cisteína/metabolismo , Endopeptidases/química , Endopeptidases/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Papaína/antagonistas & inibidores , Papaína/metabolismo , Soroalbumina Bovina/metabolismo
18.
Pestic Biochem Physiol ; 145: 66-75, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29482733

RESUMO

Carbendazim is a broad spectrum benzimidazole fungicide which is used to ensure plants' protection from pest and pathogens' invasion. The present work describes the impact of carbendazim (CAR) on garlic phytocystatin (GPC) which is a crucial plant regulatory protein. Interaction of carbendazim with GPC has been investigated through various biophysical techniques viz. UV absorption, fluorescence spectroscopy, isothermal titration calorimetry, far-UV circular dichroism and FTIR spectroscopy which showed binding between them with consequent modulatory effects. Functional activity of GPC was monitored by the anti-papain inhibitory assay which suggests that incubation of GPC with the higher concentration of CAR disrupts the inhibitory function of GPC. UV spectroscopy confirmed the formation of GPC-CAR complex. Intrinsic fluorescence suggests binding of CAR to GPC which reflects the changes in microenvironment around tryptophan residues of GPC. Isothermal titration calorimetry suggests that interaction of CAR to GPC is an exothermic reaction. Secondary structure analysis was also performed which confirmed that binding of CAR decreases the alpha-helical content of GPC. Collectively, these results demonstrated that GPC exhibited significant structural and functional alteration upon interaction with carbendazim. Since GPC is involved in various regulatory processes, therefore, its structural or functional alteration may lead to disruption of physiological and biological balance within the plant. Hence, our study signifies that exposure of carbendazim to plant exerts physicochemical alteration within the plant.


Assuntos
Benzimidazóis/farmacologia , Carbamatos/farmacologia , Fungicidas Industriais/farmacologia , Alho/metabolismo , Proteínas de Plantas/metabolismo , Análise Espectral/métodos , Benzimidazóis/metabolismo , Sítios de Ligação , Calorimetria , Carbamatos/metabolismo , Fungicidas Industriais/metabolismo , Papaína/antagonistas & inibidores , Plantas/microbiologia , Relação Estrutura-Atividade
19.
Microbiol Res ; 207: 211-225, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29458857

RESUMO

Cysteine proteases of the papain family, including mammalian cathepsins, play important physiological roles, however, their excessive activity may contribute to the development of various pathologies. Therefore, cysteine cathepsin inhibitors are being considered as promising drugs to treat cathepsin-driven diseases. Diverse saprophytic and parasitic microbes produce such inhibitors, which target the host's proteases playing pivotal roles in immune responses, thus leading to the survival of microbes within their host. Yersinia enterocolitica is a Gram-negative zoopathogenic coccobacillus, which has developed several mechanisms to evade the host's immune system. Nevertheless, the bacterium has not yet been shown to produce any cysteine protease inhibitors. Here we demonstrate that Y. enterocolitica strains of different bioserotypes and genotypes synthesize papain and human cathepsin L inhibitors, but not bovine cathepsin B inhibitors. By employing fluorimetry and zymography, the cell-surface inhibitors were shown to associate peripherally with the outer membrane, while the inhibitors present in cell-free extracts proved to: interact reversibly with their target enzymes, exhibit thermolability and stability in a range of pH values (5-9), and have high molecular weights. Batch affinity chromatography on papain-agarose resin was then undertaken to isolate putative inhibitors of cysteine proteases from the bacterial extract. The isolated 18 kDa protein was identified by LC-MS/MS as the periplasmic chaperone Skp. The Skp-containing eluate inhibited the activity of cysteine cathepsins produced by human dermal fibroblasts. The homologous Skp protein was also isolated from the extract of Escherichia coli. Our results point to a possible new biological role of the bacterial chaperone Skp.


Assuntos
Catepsinas/metabolismo , Extratos Celulares/química , Inibidores de Cisteína Proteinase/metabolismo , Papaína/antagonistas & inibidores , Yersinia enterocolitica/metabolismo , Animais , Bovinos , Cisteína Proteases/metabolismo , Proteínas de Ligação a DNA , Escherichia coli/metabolismo , Proteínas de Escherichia coli , Humanos , Chaperonas Moleculares , Papaína/metabolismo
20.
Int J Biol Macromol ; 94(Pt B): 819-826, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26751400

RESUMO

In the present study a thiol proteinase inhibitor was isolated from buffalo kidney making use of ammonium sulphate precipitation and gel filtration chromatography on Sephacryl S-100HR column. Purified inhibitor is homogeneous as it displayed a single band in gel electrophoresis both under reducing and non-reducing environment and is of 65KDa as revealed by gel filtration and SDS PAGE. Kinetic studies revealed the presence of reversible accompanied with competitive mode of inhibition; showing maximum efficacy against papain (Ki=2.90×10-4). It was maximally active at pH 8.0 and was stable for a period of 30, 60 and 90 days at 37, 4 and -20°C respectively. Immunological studies confirmed its purity of epitopes as a single precipitin line is obtained in immunodiffusion. N-terminal analysis revealed that it shared a good homology with mouse kidney cystatin as well as with Human Cys C and Cys E thereby advocating its use as a model for various human oriented studies which targets how the kidney cystatin level varies in accordance with various drugs that are currently being used as a target for variety of diseases.


Assuntos
Cistatinas/química , Rim/química , Papaína/química , Inibidores de Proteases/química , Compostos de Sulfidrila/química , Sequência de Aminoácidos , Animais , Bromelaínas/antagonistas & inibidores , Bromelaínas/química , Búfalos , Cistatinas/imunologia , Cistatinas/isolamento & purificação , Ficina/antagonistas & inibidores , Ficina/química , Humanos , Concentração de Íons de Hidrogênio , Rim/imunologia , Cinética , Camundongos , Peso Molecular , Papaína/antagonistas & inibidores , Inibidores de Proteases/imunologia , Inibidores de Proteases/isolamento & purificação , Estabilidade Proteica , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...