Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 628: 25-31, 2022 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-36063599

RESUMO

α-1-antichymotrypsin (ACT) is a serine proteinase inhibitor that controls the activity of proteases like chymotrypsin, cathepsin G and mast cell chymase. Familial variants of ACT results in liver and lung diseases, but it is also reported to be associated with several other disease conditions. ACT is mainly synthesized in the liver using four coding exons, namely E1, E2, E3 and E4 encoding a 423 amino acid protein that also includes a 23 amino acid signal peptide. It is found to be associated with amyloid plaques and is elevated during inflammatory response and modulates cytokine based signal transduction pathways, independent of its anti-protease activity. Therefore, the multispecificity of ACT and its non-inhibitory roles in diseased conditions warrants an assessment of possible existence of the other isoforms. Consequently, scanning of introns, 5' and 3' region of the ACT gene using computational tools like FGENESH and FEX did indicate the presence of coding regions. Using a combined approach of bioinformatics and molecular biology, we have found one novel exon located in the intronic region between exons E1 and E2, that splices with exon E2 and replaces N-terminal exon E1, generating an ACT isoform with a novel 151 base pair N-terminus. This isoform was found to lack the signal sequence and is smaller in size but its reactive centre loop remains intact. A truncated transcript was also confirmed with an extension of the E3 by a 12 nucleotide intronic region including a stop codon. Modelling studies show that due to removal of E4 this isoform lacks the RCL. Novel isoform ACT-N lacks E1 but has a conserved RCL. However, due to loss of strands of ß-sheet A, it may also be inactive, but with ability to bind the target proteases. The novel truncated ACT-T isoform lacks the RCL and may have a non-inhibitory role. These hypothesis will need further work for functional validation.


Assuntos
Inibidores de Serina Proteinase , Processamento Alternativo , Sequência de Aminoácidos , Aminoácidos/metabolismo , Catepsina G/metabolismo , Quimases/metabolismo , Quimotripsina/metabolismo , Códon de Terminação , Citocinas/metabolismo , Humanos , Nucleotídeos/metabolismo , Isoformas de Proteínas/metabolismo , Sinais Direcionadores de Proteínas , Inibidores de Serina Proteinase/genética , Serpinas
2.
IUBMB Life ; 73(7): 941-952, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33893722

RESUMO

Neuroserpin is a serine protease inhibitor expressed mainly in the brain and at low levels in other tissues like the kidney, testis, heart, and spinal cord. It is involved in the inhibition of tissue plasminogen activator (tPA), plasmin, and to a lesser extent, urokinase-type plasminogen (uPA). Neuroserpin has also been shown to plays noninhibitory roles in the regulation of N-cadherin-mediated cell adhesion. It is involved in neuroprotection from seizure and stroke through tPA-mediated inhibition and also through its other protease targets. Mutations in critical domains of neuroserpin lead to its polymerization and neuronal death. In this study, a novel truncated isoform of human neuroserpin was identified in the brain and liver, which was confirmed by reverse transcriptase-PCR and DNA sequencing using exon-specific primers. Structural characterization of novel isoform using MD simulations studies indicated that it lacks the reactive center loop (RCL) but largely maintains its secondary structure fold. The novel truncated variant was cloned, expressed, and purified. A comparative intrinsic fluorescence and 4,4'-bis-1-anilino naphthalene 8-sulfonate studies revealed a decrease in fluorescence emission intensity and a more exposed hydrophobic surface as compared to the reported isoform. However, the novel isoform has lost its ability for tPA inhibition and complex formation. The absence of RCL indicates a noninhibitory role for the truncated isoform, prompting a detailed search and identification of two smaller isoforms in the human brain. With indications of the noninhibitory role of neuroserpin, identifying novel isoforms that appear to be without the tPA recognition domain is significant.


Assuntos
Neuropeptídeos/química , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Serpinas/química , Serpinas/genética , Serpinas/metabolismo , Processamento Alternativo , Encéfalo/metabolismo , Fluorescência , Expressão Gênica , Humanos , Interações Hidrofóbicas e Hidrofílicas , Fígado/metabolismo , Simulação de Dinâmica Molecular , Isoformas de Proteínas , Reprodutibilidade dos Testes , Ativador de Plasminogênio Tecidual/metabolismo , Neuroserpina
3.
IUBMB Life ; 72(10): 2180-2193, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32827448

RESUMO

Heparin cofactor II (HCII) is predominantly expressed in the liver and inhibits thrombin in blood plasma to influence the blood coagulation cascade. Its deficiency is associated with arterial thrombosis. Its cleavage by neutrophil elastase produces fragment that helps in neutrophil chemotaxis in the acute inflammatory response in human. In the present study, we have identified a novel alternatively spliced transcript of the HCII gene in human liver. This novel transcript includes an additional novel region in continuation with exon 3 called exon 3b. Exon 3b acts like an alternate last exon, and hence its inclusion in the transcript due to alternative splicing removes exon 4 and encodes for a different C-terminal region to give a novel protein, HCII-N. MD simulations of HCII-N and three-dimensional structure showed a unique 51 amino acid sequence at the C-terminal having unique RCL-like structure. The HCII-N protein purified from bacterial culture showed a protein migrating at lower molecular weight (MW 55 kDa) as compared to native HCII (MW 66 kDa). A fluorescence-based analysis revealed a more compact structure of HCII-N that was in a more hydrophilic environment. The HCII-N protein, however, showed no inhibitory activity against thrombin. Due to large conformational variation observed in comparison with native HCII, HCII-N may have alternate protease specificity or a non-inhibitory role. Western blot of HCII purified from large plasma volume showed the presence of a low MW 59 kDa band with no thrombin activity. This study provides the first evidence of alternatively spliced novel isoform of the HCII gene.


Assuntos
Cofator II da Heparina/química , Cofator II da Heparina/genética , Cofator II da Heparina/metabolismo , Fígado/metabolismo , Processamento Alternativo , Fator Xa/metabolismo , Humanos , Modelos Moleculares , Simulação de Dinâmica Molecular , Isoformas de Proteínas , Espectrometria de Fluorescência , Trombina/metabolismo , Ativador de Plasminogênio Tecidual/antagonistas & inibidores , Ativador de Plasminogênio Tecidual/metabolismo
4.
Arch Biochem Biophys ; 685: 108332, 2020 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-32194043

RESUMO

Increased tendency of cancer patients to develop venous thromboembolism (VTE) is associated with high rates of mortality. Elevation of procoagulant proteins and down regulation of naturally occurring coagulation inhibitors appears to form the basis of high risk of VTE in malignancy. A reduced level of anticoagulant protein like antithrombin (AT) will influence both coagulation and angiogenesis, as its cleaved and latent conformations show potent antiangiogenic activity. We show a concentration dependent perturbation in the secondary and tertiary structures of AT conformers exposed to hypochlorous acid (HOCl). Modulated under a very narrow concentration range of HOCl, native AT undergoes oligomerization, aggregation and fragmentation based on spectroscopic, SDS and native-PAGE studies. Factor Xa inhibition assay demonstrated a progressive decrease in inhibition activity of AT on modification by HOCl. Bis-ANS result showed that hydrophobic patches were more exposed in the case of HOCl-modified AT when assessed fluorometrically. Dosage of HOCl-modified AT in experimental animals induced high titer antibodies showing more specificity towards modified forms in comparison to unmodified forms. Auto-antibodies isolated from cancer patients also showed enhanced binding with HOCl-modified AT in comparison to native counterpart. Compared to normal AT, structurally and functionally altered conformation of HOCl-modified AT showed increased immunogenic sensitivity. HOCl modified AT can contribute to prothrombotic and angiogenic environment during cancer progression/development.


Assuntos
Antitrombinas/imunologia , Epitopos/imunologia , Ácido Hipocloroso/química , Adolescente , Adulto , Idoso , Animais , Anticorpos Antineoplásicos/imunologia , Anticorpos Antineoplásicos/isolamento & purificação , Antitrombinas/química , Autoanticorpos/imunologia , Autoanticorpos/isolamento & purificação , Feminino , Humanos , Imunoglobulina G/imunologia , Imunoglobulina G/isolamento & purificação , Pessoa de Meia-Idade , Coelhos , Adulto Jovem
5.
Clin Sci (Lond) ; 134(17): 2235-2241, 2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32869854

RESUMO

Human serine protease inhibitors (serpins) are the main inhibitors of serine proteases, but some of them also have the capability to effectively inhibit cysteine proteases. Severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) main protease (Mpro) is a chymotrypsin-type cysteine protease that is needed to produce functional proteins essential for virus replication and transcription. Serpin traps its target proteases by presenting a reactive center loop (RCL) as protease-specific cleavage site, resulting in protease inactivation. Mpro target sites with its active site serine and other flanking residues can possibly interact with serpins. Alternatively, RCL cleavage site of serpins with known evidence of inhibition of cysteine proteases can be replaced by Mpro target site to make chimeric proteins. Purified chimeric serpin can possibly inhibit Mpro that can be assessed indirectly by observing the decrease in ability of Mpro to cleave its chromogenic substrate. Chimeric serpins with best interaction and active site binding and with ability to form 1:1 serpin-Mpro complex in human plasma can be assessed by using SDS/PAGE and Western blot analysis with serpin antibody. Trapping SARS-CoV-2 Mpro cysteine protease using cross-class serpin cysteine protease inhibition activity is a novel idea with significant therapeutic potential.


Assuntos
Antivirais/farmacologia , Betacoronavirus/efeitos dos fármacos , Infecções por Coronavirus/tratamento farmacológico , Pneumonia Viral/tratamento farmacológico , Serpinas/farmacologia , Proteínas não Estruturais Virais/antagonistas & inibidores , Antivirais/uso terapêutico , Betacoronavirus/enzimologia , Western Blotting , COVID-19 , Proteases 3C de Coronavírus , Infecções por Coronavirus/virologia , Cisteína Endopeptidases/química , Eletroforese em Gel de Poliacrilamida , Humanos , Pandemias , Pneumonia Viral/virologia , SARS-CoV-2 , Serpinas/uso terapêutico , Proteínas não Estruturais Virais/química
6.
J Thromb Thrombolysis ; 50(3): 678-685, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32020514

RESUMO

Antithrombin (AT) deficiency is a rare but strong risk factor for the thrombosis development. Mutations in gene encoding AT (SERPINC1) have provided a detailed understanding of AT deficiency and subsequent development of thrombotic complications. In the present study, we describe a case of thrombotic patient with reduced AT activity and normal AT antigen levels. AT deficiency in the patient was explained by the presence of heterozygous mutation g.13397A>G (Ala427Thr) in exon 6 of SERPINC1. Reduced APTT and TT with normal PT were observed. The mutation was found to be absent in healthy controls (n = 62). In vitro purification and characterization of variant AT showed significant decrease in fluorescence emission intensity, decreased bis-ANS fluorescence emission, changes in secondary structure and presence of polymerized AT in patient's plasma as assessed by fluorescence, circular dichroism and transmission electron microscopy respectively. Furthermore, molecular dynamics simulation studies showed altered conformation due to Ala427Thr substitution. Our study shows that genetic screening should be carried out in AT deficient patients in addition to the routinely used functional assays to understand the molecular basis of disease development.


Assuntos
Antitrombina III/genética , Trombofilia/genética , Adulto , Antitrombina III/química , Coagulação Sanguínea , Feminino , Humanos , Simulação de Dinâmica Molecular , Mutação Puntual , Conformação Proteica , Trombofilia/sangue
7.
Biochem Biophys Res Commun ; 517(3): 421-426, 2019 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-31378371

RESUMO

Antithrombin (AT3) is one of the most important inhibitors of blood coagulation proteases that belong to the serpin family of protease inhibitors. In this study, a novel alternatively spliced isoform of AT3 was identified, both at transcript and protein level. This novel transcript contains an additional region in the continuation of exon 3b that was included in the transcript due to use of an alternate 5' splice site. The existence of the novel transcript was confirmed in human brain and liver through RT-PCR. An analysis of the complete transcript indicated that the native reactive centre loop (RCL) of AT3 is maintained; however the novel amino acid sequence projects out as an additional loop as evident from MD simulation studies. A unique amino acid sequence present in the novel isoform was used for the development of polyclonal antibody. The expression of novel isoform was confirmed in human brain and liver tissue using Western blot analysis. Interestingly an alignment of RCL like domain with other inhibitory serpins showed significant similarity with the neuroserpin RCL. To the best of our knowledge, this is the first evidence of alternatively spliced AT3 sequence containing an additional loop and could have physiological relevance.


Assuntos
Processamento Alternativo , Antitrombina III/química , Heparina/química , Neuropeptídeos/química , Serpinas/química , Sequência de Aminoácidos , Animais , Anticorpos/química , Anticorpos/isolamento & purificação , Antitrombina III/genética , Antitrombina III/metabolismo , Sequência de Bases , Sítios de Ligação , Encéfalo/metabolismo , Expressão Gênica , Heparina/metabolismo , Humanos , Fígado/metabolismo , Simulação de Dinâmica Molecular , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Coelhos , Serpinas/genética , Serpinas/metabolismo , Neuroserpina
8.
J Ind Microbiol Biotechnol ; 46(12): 1669-1683, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31531745

RESUMO

Due to the potential toxicity of mercury, there is an immediate need to understand its uptake, transport and flux within living cells. Conventional techniques used to analyze Hg2+ are invasive, involve high cost and are less sensitive. In the present study, a highly efficient genetically encoded mercury FRET sensor (MerFS) was developed to measure the cellular dynamics of Hg2+ at trace level in real time. To construct MerFS, the periplasmic mercury-binding protein MerP was sandwiched between enhanced cyan fluorescent protein (ECFP) and venus. MerFS is pH stable, offers a measurable fluorescent signal and binds to Hg2+ with high sensitivity and selectivity. Mutant MerFS-51 binds with an apparent affinity (Kd) of 5.09 × 10-7 M, thus providing a detection range for Hg2+ quantification between 0.210 µM and 1.196 µM. Furthermore, MerFS-51 was targeted to Escherichia coli (E. coli), yeast and human embryonic kidney (HEK)-293T cells that allowed dynamic measurement of intracellular Hg2+ concentration with a highly responsive saturation curve, proving its potential application in cellular systems.


Assuntos
Transferência Ressonante de Energia de Fluorescência , Mercúrio/análise , Transporte Biológico , Sobrevivência Celular , Escherichia coli/química , Células HEK293 , Humanos , Espaço Intracelular/química , Saccharomyces cerevisiae/química
9.
Arch Biochem Biophys ; 604: 128-42, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27372899

RESUMO

Pro-coagulant, anti-coagulant and fibrinolytic pathways are responsible for maintaining hemostatic balance under physiological conditions. Any deviation from these pathways would result in hypercoagulability leading to life threatening diseases like myocardial infarction, stroke, portal vein thrombosis, deep vein thrombosis (DVT) and pulmonary embolism (PE). Angiogenesis is the process of sprouting of new blood vessels from pre-existing ones and plays a critical role in vascular repair, diabetic retinopathy, chronic inflammation and cancer progression. Serpins; a superfamily of protease inhibitors, play a key role in regulating both angiogenesis and coagulation. They are characterized by the presence of highly conserved secondary structure comprising of 3 ß-sheets and 7-9 α-helices. Inhibitory role of serpins is modulated by binding to cofactors, specially heparin and heparan sulfate proteoglycans (HSPGs) present on cell surfaces and extracellular matrix. Heparin and HSPGs are the mainstay of anti-coagulant therapy and also have therapeutic potential as anti-angiogenic inhibitors. Many of the heparin binding serpins that regulate coagulation cascade are also potent inhibitors of angiogenesis. Understanding the molecular mechanism of the switch between their specific anti-coagulant and anti-angiogenic role during inflammation, stress and regular hemostasis is important. In this review, we have tried to integrate the role of different serpins, their interaction with cofactors and their interplay in regulating coagulation and angiogenesis.


Assuntos
Coagulação Sanguínea , Heparina/química , Neovascularização Patológica , Serpinas/fisiologia , Angiotensinogênio/química , Animais , Anticoagulantes/química , Antitrombinas/química , Proteínas Sanguíneas/química , Vasos Sanguíneos/fisiopatologia , Progressão da Doença , Matriz Extracelular/química , Proteínas do Olho/química , Fibrinólise , Proteoglicanas de Heparan Sulfato/química , Heparina de Baixo Peso Molecular/química , Homeostase , Humanos , Inflamação , Conformação Molecular , Fatores de Crescimento Neural/química , Inibidor 1 de Ativador de Plasminogênio/química , Inibidor da Proteína C/química , Serpina E2/química , Serpinas/química , Transdução de Sinais
10.
Org Biomol Chem ; 14(45): 10599-10619, 2016 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-27735963

RESUMO

The increasing incidence of human candidiasis and the tendency of Candida species to become resistant to existing chemotherapies are well-recognized health problems. The present study demonstrates the successful synthesis of novel triazole-amino acid hybrids with potent in vitro and in vivo inhibitory activity against Candida species. Particularly, compounds 68 and 70 showed potent in vitro activity against fluconazole (FLC) resistant as well as sensitive clinical isolates of Candida albicans. Time kill curve analysis of lead inhibitors 68 and 70 showed their fungistatic nature. Secretion of hydrolytic enzymes, mainly proteinases and phospholipases, decreased considerably in the presence of 68 and 70 indicating their interference in fungal virulence. TEM analysis of Candida cells exposed to compounds 68 and 70 clearly showed morphological changes and intracellular damage as their possible mode of action. A preliminary mechanistic study carried out on the two most effective inhibitors (68 and 70) revealed the inhibition of ergosterol biosynthesis thereby causing the cells to lose their integrity and viability. The selected compounds did not show significant cytotoxicity up to a concentration of 200 µg mL-1 in the HEK293 cell line. An in silico analysis of 68 and 70 binding to a modeled C. albicans CYP51 showed critical H-bonding as well as hydrophobic interactions with the important active site residues indicating the basis of their anti-Candida role. Studies on the larvae of Galleria mellonella showed that the selected inhibitors (68 and 70) were non-toxic, did not provoke an immune response and significantly reduced Candida proliferation in vivo.


Assuntos
Aminoácidos/química , Aminoácidos/farmacologia , Antifúngicos/química , Antifúngicos/farmacologia , Candida/efeitos dos fármacos , Triazóis/química , Triazóis/farmacologia , Candida/crescimento & desenvolvimento , Candida/metabolismo , Candida/patogenicidade , Candidíase/tratamento farmacológico , Fluconazol/farmacologia , Células HEK293 , Humanos , Testes de Sensibilidade Microbiana , Simulação de Acoplamento Molecular , Esterol 14-Desmetilase/metabolismo , Virulência/efeitos dos fármacos
11.
Eur Biophys J ; 44(3): 139-48, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25652544

RESUMO

The importance of domain II in the molecular interaction of bovine serum albumin (BSA) with curcumin was investigated by fluorescence spectroscopy and molecular docking. At pH 7.4 BSA is in its native state. Domain III of BSA unfolds at pH 4.0, and domains I and III unfold in the presence of 5 M urea. Curcumin has a high quenching constant (K SV ~ 10(4) M (-1)) and moderate binding affinity (n ~ 0.5). The standard free energy change (∆G° ~ -25 kJ mol(-1)) indicates that binding is spontaneous. No significant change in ∆G° observed after unfolding of domain I or domain III. The standard change in enthalpy (∆H°) and entropy (∆S°) show that ionic and hydrophobic interactions are important in the binding. Computational studies revealed that the inter-domain helix h10DOMI-h1DOMII of BSA is the region of binding of curcumin, and residues Arg198 and Arg208 are important in binding. The binding site is located between sub-domains IB and IIA, and overlaps drug binding site-1.


Assuntos
Curcumina/farmacologia , Simulação de Acoplamento Molecular , Soroalbumina Bovina/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Bovinos , Curcumina/química , Dados de Sequência Molecular , Ligação Proteica , Soroalbumina Bovina/metabolismo
12.
J Thromb Thrombolysis ; 39(4): 481-8, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25771983

RESUMO

Recurrent pregnancy loss (RPL) can be caused due to diverse factors with thrombophilia being one of them. The association of various thrombophilic risk factors with RPL is inconsistent in different studies and the frequency of these risk factors in Indian population is obscure. Five hundred and eighty patients with either recurrent early miscarriage or a history of at least one late miscarriage were screened for deficiency of protein C (PC), protein S (PS), antithrombin III (AT), APC resistance and prothrombin 20210G > A mutation. APC resistance positive patients were typed for the factor V Leiden, factor V Hong Kong/Cambridge mutations, and HR2 haplotype. PstI and rs2227589 AT mutations were detected by direct sequencing. APC resistance (13.4 %) was detected to be most common in Indian RPL patients followed by PS (10.6 %), PC (9.8 %) and AT deficiency (4.31 %.). FV Leiden was shown to be associated with APC resistance while HR2 haplotype was not associated with APC resistance (p values: 0.0001 and 0.327 respectively) and the increased risk of RPL. PstI and rs2227589 polymorphisms were similar in patients and controls and not associated with AT deficiency in RPL. Our study emphasizes the presence of other contributory factors towards APC resistance rather than FV Leiden alone. This is the first Indian study where HR2 haplotype and rs2227589 are observed to be present in RPL population. Although not significant, occurrence of rs2227589 and FV HR2 in homozygous condition necessitates the study of these polymorphisms in a larger sample size.


Assuntos
Aborto Habitual/genética , Antitrombina III/genética , Fator V/genética , Mutação , Polimorfismo Genético , Resistência à Proteína C Ativada/genética , Adulto , Estudos de Casos e Controles , Feminino , Haplótipos , Homozigoto , Humanos , Índia , Gravidez
13.
Int J Biol Macromol ; 266(Pt 2): 131065, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38521329

RESUMO

Protein C inhibitor (PCI) maintains hemostasis by inhibiting both procoagulant and anticoagulant serine proteases, and plays important roles in coagulation, fibrinolysis, reproduction, and anti-angiogenesis. The reactive site loop of PCI traps and irreversibly inhibits the proteases like APC (activating protein C), thrombin (FIIa) and factor Xa (FXa). Previous studies on antithrombin (ATIII) had identified Tyr253 and Glu255 as functional exosites that interact and aid in the inhibition of factor IXa and FXa. Presence of exosite in PCI is not known, however a sequence comparison with the PCI from different vertebrate species and ATIII identified Glu239 to be absolutely conserved. PCI residues analogous to ATIII exosite residues were mutated to R238A and E239A. Purified variant PCI in the presence of heparin (10 µg/ml) showed a 2-4 fold decrease in the rate of inhibition of the proteases. However, the stoichiometry of inhibition of FIIa, APC, and FXa by native PCI, R238A and E239A variants were found to be close to 1.0, which also indicated the formation of stable complexes based on SDS-PAGE and western blot analysis with thrombin and APC. Our findings revealed the possible presence of an exosite in PCI that influences the protease inhibition rates.


Assuntos
Heparina , Inibidor da Proteína C , Serina Proteases , Inibidor da Proteína C/química , Inibidor da Proteína C/metabolismo , Heparina/química , Heparina/farmacologia , Humanos , Serina Proteases/metabolismo , Serina Proteases/química , Trombina/metabolismo , Proteína C/metabolismo , Proteína C/química , Fator Xa/metabolismo , Fator Xa/química , Sequência de Aminoácidos , Ativação Enzimática/efeitos dos fármacos
14.
Biosci Rep ; 44(5)2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38660763

RESUMO

During inflammation and situations of cellular stress protein disulfide isomerase (PDI) is released in the blood plasma from the platelet and endothelial cells to influence thrombosis. The addition of exogenous PDI makes the environment pro-thrombotic by inducing disulfide bond formation in specific plasma protein targets like vitronectin, factor V, and factor XI. However, the mechanistic details of PDI interaction with its target remain largely unknown. A decrease in the coagulation time was detected in activated partial thromboplastin time (APTT), prothrombin time (PT), and thrombin time (TT) on addition of the purified recombinant PDI (175 nM). The coagulation time can be controlled using an activator (quercetin penta sulfate, QPS) or an inhibitor (quercetin 3-rutinoside, Q3R) of PDI activity. Likewise, the PDI variants that increase the PDI activity (H399R) decrease, and the variant with low activity (C53A) increases the blood coagulation time. An SDS-PAGE and Western blot analysis showed that the PDI does not form a stable complex with either thrombin or antithrombin (ATIII) but it uses the ATIII-thrombin complex as a template to bind and maintain its activity. A complete inhibition of thrombin activity on the formation of ATIII-thrombin-PDI complex, and the complex-bound PDI-catalyzed disulfide bond formation of the target proteins may control the pro- and anti-thrombotic role of PDI.


Assuntos
Coagulação Sanguínea , Isomerases de Dissulfetos de Proteínas , Trombina , Humanos , Isomerases de Dissulfetos de Proteínas/metabolismo , Trombina/metabolismo , Antitrombina III/metabolismo , Ligação Proteica , Antitrombinas/metabolismo , Antitrombinas/química , Quercetina/farmacologia , Quercetina/análogos & derivados
15.
Int J Biol Macromol ; 252: 126241, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37567521

RESUMO

Currently used antithrombotic drugs are beset with several drawbacks which necessitates the need for new and cheaper alternatives. Protein disulfide isomerase (PDI) is secreted in the blood plasma in cellular stress conditions and initiates the thrombus formation. A screening of library of natural compounds revealed that naringin had a high binding affinity for the PDI (-8.2 kcal/mol). Recombinant PDI was purified using the affinity chromatography. Incubation of purified PDI (3 µM) with naringin (0-100 µM, pH 7.4, 25 °C) partially modulated its conformation. Consequently, the fluorescence emission spectra of the PDI binding to naringin were assessed using the Stern-Volmer equation, which indicated an association constant of 2.78 × 104 M-1 suggesting an appreciable affinity for the naringin, with a unique binding site. An insulin turbidity assay showed that PDI activity is decreased in the presence of naringin indicating inhibition. Molecular dynamic simulation studies showed the changes in the PDI structure on binding to the naringin. Incubation of naringin (80 µM) in fresh human plasma along with exogenous PDI (175 nM) showed a significant delay in the intrinsic and extrinsic coagulation pathways. We show that naringin is able to modulate the PDI conformation and activity resulting in altered blood coagulation rates.


Assuntos
Flavanonas , Trombose , Humanos , Isomerases de Dissulfetos de Proteínas/metabolismo , Coagulação Sanguínea , Trombose/metabolismo , Flavanonas/farmacologia
16.
J Biomol Struct Dyn ; 41(9): 3717-3727, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-35343865

RESUMO

Thromboembolic diseases are a major cause of mortality in human and the currently available anticoagulants are associated with various drawbacks, therefore the search for anticoagulants that have better safety profile is highly desirable. Compounds that are part of the dietary routine can be modified to possibly increase their anticoagulant potential. We show mannose 2,3,4,5,6-O-pentasulfate (MPS) as a synthetically modified form of mannose that has appreciable anticoagulation properties. An in silico study identified that mannose in sulfated form can bind effectively to the heparin-binding site of antithrombin (ATIII) and heparin cofactor II (HCII). Mannose was sulfated using a simple sulfation strategy-involving triethylamine-sulfur trioxide adduct. HCII and ATIII were purified from human plasma and the binding analysis using fluorometer and isothermal calorimetry showed that MPS binds at a unique site. A thrombin inhibition analysis using the chromogenic substrate showed that MPS partially enhances the activity of HCII. Further an assessment of in vitro blood coagulation assays using human plasma showed that the activated partial thromboplastin time (APTT) and prothrombin time (PT) were prolonged in the presence of MPS. A molecular dynamics simulation analysis of the HCII-MPS complex showed fluctuations in a N-terminal loop and the cofactor binding site of HCII. The results indicate that MPS is a promising lead due to its effect on the in vitro coagulation rate.Communicated by Ramaswamy H. Sarma.


Assuntos
Cofator II da Heparina , Manose , Humanos , Cofator II da Heparina/química , Cofator II da Heparina/metabolismo , Manose/farmacologia , Coagulação Sanguínea , Anticoagulantes/farmacologia , Anticoagulantes/química , Heparina/farmacologia , Antitrombina III/farmacologia , Antitrombina III/fisiologia , Antitrombinas/farmacologia , Trombina/química
17.
J Thromb Thrombolysis ; 34(2): 251-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22453684

RESUMO

An elevated prothrombotic state is a major risk factor for venous thromboembolism, atrial fibrillation and cardiac strokes. The regulation of various coagulation cascade proteases plays an important role in determining a prothrombotic state. Clinically used anticoagulants are inhibitor of enzymes that are involved in the coagulation pathway, primarily thrombin and factor Xa. The conformational activation of antithrombin by heparin is a critical step in the inhibition of factor Xa by antithrombin. Despite heparin being the most potent physiological activator which enhances the otherwise very lethargic antithrombin inhibition of factor Xa by approximately 1,000-fold, the conventional heparin therapy poses serious complications because of heparin's polyanionic nature and its cross-reactivity. A number of attempts have been carried out in designing alternative non-heparin based conformational activators of antithrombin for factor Xa inhibition. Studies have demonstrated appreciable activation of antithrombin by small organic molecules, but not much is known about the specificity and effects of these molecules on structure and stability. It is assumed that these activators of antithrombin perform their function by binding to heparin binding site. A recently identified cavity which links the heparin binding site to the strand 2A for antithrombin activation also seems to be an ideal target apart the heparin binding site of antithrombin. There are opportunities in discovering more activators from naturally available organic scaffolds and also for modifying such scaffolds for designing better conformational activators with minimum associated complications. This review summarizes the current literature on the mainstay anticoagulants and non-heparin based antithrombin conformation modulators.


Assuntos
Anticoagulantes/farmacologia , Antitrombina III/metabolismo , Heparina/farmacologia , Fibrilação Atrial/tratamento farmacológico , Fibrilação Atrial/metabolismo , Fator Xa/metabolismo , Inibidores do Fator Xa , Humanos , Conformação Proteica/efeitos dos fármacos , Trombose Venosa/tratamento farmacológico , Trombose Venosa/metabolismo
18.
Biosci Rep ; 42(12)2022 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-36408789

RESUMO

Neuroserpin (NS) is predominantly expressed in the brain and is the primary inhibitor of tissue plasminogen activator (tPA). NS variants are associated with the neurogenerative disease termed familial encephalopathy with neuroserpin inclusion bodies (FENIB). The disease is characterized by variable age of onset and severity. The reactive center loop (RCL) insertion-based inhibitory mechanism of NS requires a coordinated conformational change leading to a shift in the strands of the ß-sheet A and movement of helix F. Strand 1A is connected to the helix F at its C terminal end and with the strand 2A at its N terminal, both these domain move for accommodating the inserting loop; therefore, a variant that influences their movement may alter the inhibition rates. A molecular dynamic simulation analysis of a H138C NS variant from strand 1A showed a large decrease in conformational fluctuations as compared with wild-type NS. H138 was mutated, expressed, purified and a native-PAGE and transmission electron microscopy (TEM) analysis showed that this variant forms large molecular weight aggregates on a slight increase in temperature. However, a circular dichroism analysis showed its secondary structure to be largely conserved. Surprisingly, its tPA inhibition activity and complex formation remain unhindered even after the site-specific labeling of H138C with Alexa fluor C5 maleimide. Further, a helix F-strand 1A (W154C-H138C) double variant still shows appreciable inhibitory activity. Increasingly, it appears that aggregation and not loss of inhibition is the more likely cause of shutter region-based variants phenotypes, indicating that hindering polymer formation using small molecules may retain inhibitory activity in pathological variants of NS.


Assuntos
Neuropeptídeos , Serpinas , Polimerização , Ativador de Plasminogênio Tecidual , Serpinas/genética , Neuropeptídeos/genética , Neuroserpina
19.
Int J Biol Macromol ; 176: 117-125, 2021 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-33516851

RESUMO

Neuroserpin (NS) is an inhibitory protein of serpin super family, its shutter region variants have high propensity to aggregate leading to pathological disorders like familial encephalopathy with NS inclusion bodies (FENIB). Helix F and ß-sheet A of NS participate in the tissue plasminogen activator (tPA) inhibition but the mechanism is not yet completely understood. A microsecond (µs) molecular dynamics simulation of the helix F and strand 3A variants showed predominant fluctuations in the loop connecting the strands of ß-sheet A. Therefore to understand the role of helix F and strand 3A of ß-sheet A, cysteine was incorporated at the position N182 in stand 3A (N182C) and position W154 (W154C) in the helix F using site-directed mutagenesis. Purified variants were further labeled with Alexa Fluor488 C5 maleimide dye. Temperature dependent study using non-denaturing PAGE showed the formation of large aggregates of helix F variant W154C but not the strand 3A N182C variant. Interestingly tPA inhibition was found to be decreased in the labeled N182C with decreased tPA-complex formation as compared to labeled W154C NS variant. The fluorescence emission intensity of the labeled helix F variant W154C decreased in the presence of an increasing concentration of tPA, whereas an increase in emission intensity was observed in labeled strand 3A variant N182C, indicating more exposure of strand 3A and shielding of helix F. Taken together the data shows that helix F has a predominant role in the aggregation but a minor role in the inhibition mechanism.


Assuntos
Neuropeptídeos/química , Serpinas/química , Corantes Fluorescentes , Humanos , Maleimidas , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Neuropeptídeos/antagonistas & inibidores , Neuropeptídeos/genética , Agregados Proteicos , Conformação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Serpinas/genética , Ativador de Plasminogênio Tecidual/farmacologia , Neuroserpina
20.
Biochim Biophys Acta Proteins Proteom ; 1868(4): 140363, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31954927

RESUMO

Neuroserpin (NS) is predominantly expressed in brain and inhibits tissue-type plasminogen activator (tPA) with implications in brain development and memory. Nature of conformational change in pathological variants in strand 6B and helix B of NS that cause a relatively mild to severe epilepsy (and/or dementia) remains largely elusive. MD simulation with wild type (WT) NS, strand 6B and helix B variants indicated that substitution in this region affects the conformation of the strands 5B, 5A and reactive centre loop. Therefore, we designed variants of NS in strand 6B (I46D and F48S) and helix B (A54F, L55A and L55P) to investigate their role in tPA inhibition mechanism and propensity to aggregate. An interaction analysis showed disturbance of a hydrophobic patch centered at strands 5B, 6B and helix B in I46D and F48S but not in A54F, L55A, L55P and WT NS. Purified I46D, F48S and L55P variants showed decrease in fluorescence emission intensity but have similar α-helical content, however results of A54F and L55A were comparable to WT NS. Analysis of tPA inhibition showed marginal effect on A54F and L55A variant with tPA-NS complex formation. In contrast, I46D, F48S and L55P variants showed massive decrease in tPA inhibition, with no tPA-NS complex formation. Analysis of native PAGE under under polymerization condition showed prompt conversion of I46D, F48S and L55P to latent conformation but not A54F and L55A variants. Identification of these novel conformational changes will aid in the understanding of variable clinical phenotype of shutter region NS variants and other serpins.


Assuntos
Neuropeptídeos/química , Serpinas/química , Epilepsias Mioclônicas/genética , Transtornos Heredodegenerativos do Sistema Nervoso/genética , Humanos , Interações Hidrofóbicas e Hidrofílicas , Simulação de Dinâmica Molecular , Mutação , Neuropeptídeos/genética , Neuropeptídeos/isolamento & purificação , Neuropeptídeos/metabolismo , Fenótipo , Polimerização , Agregados Proteicos , Conformação Proteica , Conformação Proteica em alfa-Hélice , Serpinas/genética , Serpinas/isolamento & purificação , Serpinas/metabolismo , Ativador de Plasminogênio Tecidual/antagonistas & inibidores , Neuroserpina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA