Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 420
Filtrar
1.
Front Immunol ; 14: 1163367, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37469515

RESUMO

Background: Salivary glands from blood-feeding arthropods secrete several molecules that inhibit mammalian hemostasis and facilitate blood feeding and pathogen transmission. The salivary functions from Simulium guianense, the main vector of Onchocerciasis in South America, remain largely understudied. Here, we have characterized a salivary protease inhibitor (Guianensin) from the blackfly Simulium guianense. Materials and methods: A combination of bioinformatic and biophysical analyses, recombinant protein production, in vitro and in vivo experiments were utilized to characterize the molecula mechanism of action of Guianensin. Kinetics of Guianensin interaction with proteases involved in vertebrate inflammation and coagulation were carried out by surface plasmon resonance and isothermal titration calorimetry. Plasma recalcification and coagulometry and tail bleeding assays were performed to understand the role of Guianensin in coagulation. Results: Guianensin was identified in the sialotranscriptome of adult S. guianense flies and belongs to the Kunitz domain of protease inhibitors. It targets various serine proteases involved in hemostasis and inflammation. Binding to these enzymes is highly specific to the catalytic site and is not detectable for their zymogens, the catalytic site-blocked human coagulation factor Xa (FXa), or thrombin. Accordingly, Guianensin significantly increased both PT (Prothrombin time) and aPTT (Activated partial thromboplastin time) in human plasma and consequently increased blood clotting time ex vivo. Guianensin also inhibited prothrombinase activity on endothelial cells. We show that Guianensin acts as a potent anti-inflammatory molecule on FXa-induced paw edema formation in mice. Conclusion: The information generated by this work highlights the biological functionality of Guianensin as an antithrombotic and anti-inflammatory protein that may play significant roles in blood feeding and pathogen transmission.


Assuntos
Hemostáticos , Simuliidae , Camundongos , Humanos , Animais , Células Endoteliais , Hemostasia , Anti-Inflamatórios/farmacologia , Inflamação , Proteínas e Peptídeos Salivares/farmacologia , Mamíferos
2.
World J Microbiol Biotechnol ; 39(8): 215, 2023 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-37269390

RESUMO

Blast disease caused by Magnaporthe oryzae is a major contributor to decreased crop yield and rice production globally. The use of chemical fungicides to combat crop pathogens is not only unsafe but also promotes the emergence of pathogenic variants, leading to recurrent host infections. To address plant diseases, antimicrobial peptides have emerged as a promising alternative as they are effective, safe, and biodegradable antifungal agents. This study examines the antifungal activity and mechanism of action of the human salivary peptide histatin 5 (Hst5) on M. oryzae. Hst5 causes morphogenetic defects in the fungus, including non-uniform chitin distribution on the fungal cell wall and septa, deformed hyphal branching, and cell lysis. Importantly, a pore-forming mechanism of Hst5 in M. oryzae was ruled out. Furthermore, the interaction of Hst5 with the M. oryzae genomic DNA suggests that the peptide may also influence gene expression in the blast fungus. In addition to its effects on morphogenetic defects and cell lysis, Hst5 also inhibits conidial germination, appressorium formation, and the appearance of blast lesions on rice leaves. The elucidated multi-target antifungal mechanism of Hst5 in M. oryzae provides an environmentally friendly alternative to combating blast infections in rice by preventing fungal pathogenicity. The promising antifungal characteristics of the AMP peptide may also be explored for other crop pathogens, making it a potential biofungicide for the future.


Assuntos
Magnaporthe , Oryza , Humanos , Antifúngicos/farmacologia , Antifúngicos/metabolismo , Histatinas/farmacologia , Histatinas/metabolismo , Peptídeos Antimicrobianos , Oryza/microbiologia , Proteínas e Peptídeos Salivares/metabolismo , Proteínas e Peptídeos Salivares/farmacologia , Doenças das Plantas/prevenção & controle , Doenças das Plantas/microbiologia , Proteínas Fúngicas/genética
3.
Caries Res ; 57(1): 52-58, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36630932

RESUMO

The effect of solutions containing a statherin-derived peptide (Stn15pSpS) on the protection against enamel erosion in vitro was evaluated. Bovine enamel specimens were divided into 4 groups (n = 15/group): (1) deionized water (negative control), (2) Elmex Erosion Protection™ (positive control), (3) 1.88 × 10-5 M Stn15pSpS, and (4) 3.76 × 10-5 M Stn15pSpS. The solutions were applied on the specimens for 1 min. Stimulated saliva was collected from 3 donors and used to form a 2-h acquired pellicle on the specimens. Then, the specimens were submitted to an erosive pH-cycling protocol 4 times/day, for 7 days (0.01 M HCl pH 2.0/45 s, artificial saliva/2 h, and artificial saliva overnight). The solutions were applied again during pH-cycling, 2 times/day for 1 min after the first and last erosive challenges. Enamel loss (µm) was assessed by contact profilometry. Data were analyzed by Kruskal-Wallis and Dunn's test (p < 0.05). The best protection against erosion was conferred by Elmex Erosion Protection that significantly differed from all the other treatments, followed by the solutions containing Stn15pSpS, regardless of the concentration. However, 3.76 × 10-5 M Stn15pSpS did not differ from the negative control. The solution containing the lower concentration of Stn15pSpS protected against erosion in vitro, which should be confirmed using protocols that more closely resemble the clinical condition.


Assuntos
Erosão Dentária , Animais , Bovinos , Humanos , Esmalte Dentário , Fluoretos/farmacologia , Saliva Artificial/farmacologia , Erosão Dentária/prevenção & controle , Proteínas e Peptídeos Salivares/farmacologia
4.
Biochimie ; 208: 13-19, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36580989

RESUMO

Triabin, a lipocalin-like thrombin inhibitor from the saliva of the blood-sucking triatomine bug Triatoma pallidipennis, exhibits effective inhibition comparable to hirudin despite binding exclusively at exosite I. Interestingly, it was reported that higher triabin doses would not inhibit thrombin completely, which makes it a promising antithrombotic candidate agent with a larger therapeutic window. However, few structural and functional studies about triabin have been reported in the past three decades, mostly due to the lack of a reliable and practicable recombinant expression technology for this seemingly small protein. In this work, we have adopted the SUMO fusion technology for the expression of triabin in E. coli cells-with facile refolding and purification procedures-and the bioactive triabin was produced in ∼12 mg/L culture medium. Subsequently, the structure-function studies through extensive site-directed mutagenesis reveal that triabin's Phe-106 involved in the hydrophobic contacts plays a surprisingly important role in the thrombin inhibition, in contrast to the negatively charged residues Asp-135 or Glu-128 involved in the salt-bridge interaction. As such, this study complements our understanding of the interaction mechanism of natural thrombin inhibitors, which should facilitate the development of anticoagulant drugs with a novel mode of action against thrombin.


Assuntos
Escherichia coli , Trombina , Trombina/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas e Peptídeos Salivares/farmacologia , Proteínas de Insetos/metabolismo , Sítios de Ligação
5.
J Biochem ; 172(4): 205-216, 2022 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-35792074

RESUMO

Saliva contributes to the innate immune system, which suggests that it can prevent SARS-CoV-2 entry. We studied the ability of healthy salivary proteins to bind to angiotensin-converting enzyme 2 (ACE2) using biolayer interferometry and pull-down assays. Their effects on binding between the receptor-binding domain of the SARS-CoV-2 spike protein S1 (S1) and ACE2 were determined using an enzyme-linked immunosorbent assay. Saliva bound to ACE2 and disrupted the binding of S1 to ACE2 and four ACE2-binding salivary proteins were identified, including cationic histone H2A and neutrophil elastase, which inhibited the S1-ACE2 interaction. Calf thymus histone (ct-histone) also inhibited binding as effectively as histone H2A. The results of a cell-based infection assay indicated that ct-histone suppressed SARS-CoV-2 pseudoviral invasion into ACE2-expressing host cells. Manufactured polypeptides, such as ε-poly-L-lysine, also disrupted S1-ACE2 binding, indicating the importance of the cationic properties of salivary proteins in ACE2 binding. Overall, we demonstrated that positively charged salivary proteins are a barrier against SARS-CoV-2 entry by cloaking the negatively charged surface of ACE2 and provided a view that the cationic polypeptides represent a preventative and therapeutic treatment against COVID-19.


Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , Histonas/metabolismo , Humanos , Elastase de Leucócito/metabolismo , Peptidil Dipeptidase A/metabolismo , Polilisina/metabolismo , Ligação Proteica , SARS-CoV-2 , Proteínas e Peptídeos Salivares/metabolismo , Proteínas e Peptídeos Salivares/farmacologia , Glicoproteína da Espícula de Coronavírus
6.
Insect Biochem Mol Biol ; 143: 103739, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35149206

RESUMO

Triatoma infestans is one of the most important vectors of Trypanosoma cruzi in the Americas. While feeding, they release large amounts of saliva that will counteract the host's responses triggered at the bite site. Despite the various activities described on T. infestans saliva, little is known about its effect on the modulation of the host's immune system. This work aimed to describe the effects of T. infestans saliva on cells of the mouse immune system and access the role in hematophagy. The effect of saliva or salivary gland extract (SGE) was evaluated in vivo and in vitro by direct T. infestans feeding on mice or using different biological assays. Mice that were submitted to four bites by three specimens of T. infestans had their anti-saliva IgG serum levels approximately 2.4 times higher than controls, but no change in serum IL-2, IL-4, IL-6, IL-10, IL-17A, IFN-γ, and TNF-α levels was observed. No macroscopic alterations were seen at the bite site, but an accumulation of mononuclear and polymorphonuclear cells shortly after the bite and 24 h later were observed in histological cuts. At low concentrations (up to ∼5 µg/well), SGE induced TNF-α production by macrophages and spleen cells, IFN-γ and IL-10 by spleen cells and NO by macrophages. However, at higher concentrations (10 and 20 µg/well), viability of macrophages and spleen cells was reduced by SGE, reducing the production of NO and cytokines (except TNF-α). The salivary trialysin was the main inducer of cell death as macrophage viability and NO production was restored in assays carried out with SGE from trialysin knockdown insects. The reduction of the salivary trialysin by RNAi affected the total ingestion rate, the weight gain, and retarded the molt from second to the fifth instar of T. infestans nymphs fed on mice. The results show that T. infestans saliva modulates the activity of cells of the host immune system and trialysin is an important salivary molecule that reduces host cells viability and impacts the feeding performance of T. infestans feeding on live hosts.


Assuntos
Triatoma , Trypanosoma cruzi , Animais , Sistema Imunitário , Camundongos , Saliva , Proteínas e Peptídeos Salivares/farmacologia
7.
Histochem Cell Biol ; 157(4): 443-457, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35037129

RESUMO

Stress stimulates both the sympathetic-adrenomedullary and hypothalamus-pituitary-adrenal axes. Activation of these axes results in the release of catecholamines, which in turn affects salivary secretion. Thus, repetitive stimulation of the α1-adrenergic receptor could be useful for studying the effects of chronic stress on the salivary gland. Salivary protein concentration and kallikrein activity were significantly lower in mice following chronic phenylephrine (PHE) administration. Chronic PHE administration led to significantly increased expression of the 78-kDa glucose-regulated protein, activating transcription factor 4, and activating transcription factor 6. Histological analyses revealed a decrease in the size of the serous cell and apical cytoplasm. These results suggest that repetitive pharmacological stimulation of the sympathetic nervous system elicits ER stress and translational suppression. In addition, PHE-treated mice exhibited a decrease in intracellular Ca2+ influx elicited by carbachol, a muscarine receptor agonist in the submandibular gland. The present findings suggest that chronic psychological, social, and physical stress could adversely affect Ca2+ regulation.


Assuntos
Estresse do Retículo Endoplasmático , Glândula Submandibular , Agonistas Adrenérgicos/metabolismo , Agonistas Adrenérgicos/farmacologia , Animais , Catecolaminas , Camundongos , Proteínas e Peptídeos Salivares/metabolismo , Proteínas e Peptídeos Salivares/farmacologia , Glândula Submandibular/metabolismo
8.
Front Immunol ; 12: 626200, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33732248

RESUMO

Tick saliva is a rich source of pharmacologically and immunologically active molecules. These salivary components are indispensable for successful blood feeding on vertebrate hosts and are believed to facilitate the transmission of tick-borne pathogens. Here we present the functional and structural characterization of Iripin-3, a protein expressed in the salivary glands of the tick Ixodes ricinus, a European vector of tick-borne encephalitis and Lyme disease. Belonging to the serpin superfamily of protease inhibitors, Iripin-3 strongly inhibited the proteolytic activity of serine proteases kallikrein and matriptase. In an in vitro setup, Iripin-3 was capable of modulating the adaptive immune response as evidenced by reduced survival of mouse splenocytes, impaired proliferation of CD4+ T lymphocytes, suppression of the T helper type 1 immune response, and induction of regulatory T cell differentiation. Apart from altering acquired immunity, Iripin-3 also inhibited the extrinsic blood coagulation pathway and reduced the production of pro-inflammatory cytokine interleukin-6 by lipopolysaccharide-stimulated bone marrow-derived macrophages. In addition to its functional characterization, we present the crystal structure of cleaved Iripin-3 at 1.95 Å resolution. Iripin-3 proved to be a pluripotent salivary serpin with immunomodulatory and anti-hemostatic properties that could facilitate tick feeding via the suppression of host anti-tick defenses. Physiological relevance of Iripin-3 activities observed in vitro needs to be supported by appropriate in vivo experiments.


Assuntos
Imunidade Adaptativa/efeitos dos fármacos , Anticoagulantes/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Proteínas de Insetos/farmacologia , Ixodes/metabolismo , Saliva/metabolismo , Proteínas e Peptídeos Salivares/farmacologia , Animais , Anticoagulantes/isolamento & purificação , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citocinas/metabolismo , Cobaias , Humanos , Fatores Imunológicos/isolamento & purificação , Proteínas de Insetos/isolamento & purificação , Ativação Linfocitária/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibidores de Proteases/isolamento & purificação , Inibidores de Proteases/farmacologia , Coelhos , Proteínas e Peptídeos Salivares/isolamento & purificação , Baço/efeitos dos fármacos , Baço/imunologia , Baço/metabolismo
9.
Int J Mol Sci ; 22(2)2021 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-33477282

RESUMO

The saliva of blood-sucking leeches contains a plethora of anticoagulant substances. One of these compounds derived from Haementeria ghilianii, the 66mer three-disulfide-bonded peptide tridegin, specifically inhibits the blood coagulation factor FXIIIa. Tridegin represents a potential tool for antithrombotic and thrombolytic therapy. We recently synthesized two-disulfide-bonded tridegin variants, which retained their inhibitory potential. For further lead optimization, however, structure information is required. We thus analyzed the structure of a two-disulfide-bonded tridegin isomer by solution 2D NMR spectroscopy in a combinatory approach with subsequent MD simulations. The isomer was studied using two fragments, i.e., the disulfide-bonded N-terminal (Lys1-Cys37) and the flexible C-terminal part (Arg38-Glu66), which allowed for a simplified, label-free NMR-structure elucidation of the 66mer peptide. The structural information was subsequently used in molecular modeling and docking studies to provide insights into the structure-activity relationships. The present study will prospectively support the development of anticoagulant-therapy-relevant compounds targeting FXIIIa.


Assuntos
Fator XIIIa/antagonistas & inibidores , Espectroscopia de Ressonância Magnética/métodos , Proteínas e Peptídeos Salivares/farmacologia , Sequência de Aminoácidos , Animais , Dissulfetos/química , Fator XIIIa/metabolismo , Fibrinolíticos/farmacologia , Humanos , Isomerismo , Sanguessugas/metabolismo , Imageamento por Ressonância Magnética/métodos , Modelos Moleculares , Simulação de Dinâmica Molecular , Proteínas e Peptídeos Salivares/química , Proteínas e Peptídeos Salivares/metabolismo , Relação Estrutura-Atividade
10.
Brain Behav Immun ; 93: 288-298, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33401017

RESUMO

Recently it was shown that the hematophagous salivary gland protein agaphelin exhibits multiple antithrombotic effects without promoting the risk of bleeding. Agaphelin inhibits neutrophil elastase and thereby reduces cathepsin G-induced platelet aggregation. However, it is still unclear, whether pharmacological treatment with agaphelin in brain ischemia is protective and, regarding its bleeding risk, safe. To elucidate this issue, male C57BL/6 mice were subjected to 60 min of transient middle cerebral artery occlusion (tMCAO) and treated with 0.25 mg/kg agaphelin intravenously immediately after tMCAO. On day 1 and 7, infarct volume and functional neurological outcome were assessed by behavioural tests, histochemistry and magnetic resonance imaging. Thrombus formation, intracerebral bleeding risk, blood-brain barrier damage and the local inflammatory response were determined on day 1. This study shows for the first time a protective effect of agaphelin characterized by smaller infarct volume, reduced neurological deficits and reduced animal mortality. This protective effect was associated with reduced local thrombus formation, increased blood-brain barrier integrity and reduced brain inflammatory response. It is essential to mention that the protective effect of agaphelin was not linked to an increased risk of intracerebral bleeding. The promotion of brain tissue survival and inhibition of thromboinflammation identifies agaphelin as a promising treatment option in ischemic stroke, which considering the lack of bleeding risk should potentially be safe.


Assuntos
Isquemia Encefálica , Proteínas de Insetos/farmacologia , AVC Isquêmico , Elastase Pancreática/antagonistas & inibidores , Proteínas e Peptídeos Salivares/farmacologia , Trombose , Animais , Barreira Hematoencefálica , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/tratamento farmacológico , Inflamação/tratamento farmacológico , AVC Isquêmico/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL
11.
Int J Mol Sci ; 23(1)2021 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-35008844

RESUMO

Mucin 7 (encoded byMUC7) is a human salivary protein that has a role in the natural immune system. Fragments of mucin 7 exhibit antimicrobial activity against bacteria and yeast. Although the antimicrobial properties of peptides have been known and studied for decades, the exact mechanism of action of antimicrobial peptides (AMPs) is still unclear. It is known that some AMPs require divalent metal ions to activate their activity. Herein, we investigated three 15-mer MUC7 peptides, one of which (mother peptide, sequence, L3) is a synthetic analog of a fragment naturally excised from MUC7 (with His3, His8, and His 14) and its two structural analogs, containing only two histidine residues, His3, His13 and His8, His13 (L2 and L1, respectively). Since there is a correlation between lipophilicity, the presence of metal ions (such as Cu(II) and Zn(II)) and antimicrobial activity of AMP, antimicrobial properties of the studied peptides, as well as their complexes with Cu(II) and Zn(II) ions, were tested for activity against Gram-positive (Enterococcus faecalis, Staphylococcus epidermidis) and Gram-negative (Escherichia coli, Pseudomonas aeruginosa) bacteria and fungi (Candida albicans). The results were correlated with their lipophilicity. Coordination and thermodynamic studies (potentiometry, UV-Vis, CD) revealed the formation of mainly mononuclear complexes in solution for all studied systems with different stability in the physiological pH range.


Assuntos
Peptídeos Antimicrobianos/farmacologia , Complexos de Coordenação/farmacologia , Mucinas/farmacologia , Proteínas e Peptídeos Salivares/farmacologia , Sequência de Aminoácidos , Peptídeos Antimicrobianos/química , Humanos , Testes de Sensibilidade Microbiana , Mucinas/química , Proteínas e Peptídeos Salivares/química , Termodinâmica
12.
Virology ; 553: 1-8, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33190061

RESUMO

MUC5B and MUC7 salivary mucins are reported to inhibit HIV-1 entry into target cells in vitro; however, their relative inhibitory potencies have not been quantitively compared. There is also conflicting evidence regarding whether HIV-1 infection diminishes mucins' inhibitory efficacy. We explored the effect of donor HIV-1 status upon the anti-HIV-1 potency of purified MUC5B and MUC7 while comparing their relative inhibitory potential using a pseudovirus-based neutralization assay. HIV status of sample donors had no detectable effect on HIV-1 inhibition by salivary mucins. MUC5B (median IC50 50 µg/ml, IQR 10-116 µg/ml) exhibited significantly more potent HIV-1 inhibition than MUC7 (median IC50 458 µg/ml, IQR 192->2000 µg/ml; Mann-Whitney U p < 0.0001). We suggest that larger size, gel-forming properties and extensive glycosylation of MUC5B allow more effective binding and aggregation of viral particles. MUC5B is also more abundant in the saliva and is therefore likely to make a substantially greater contribution to it's anti-HIV-1 properties.


Assuntos
HIV-1/fisiologia , Mucina-5B/fisiologia , Mucinas/fisiologia , Saliva/química , Proteínas e Peptídeos Salivares/fisiologia , Adulto , Fármacos Anti-HIV , Linhagem Celular , Sobrevivência Celular , Glicosilação , Infecções por HIV/metabolismo , HIV-1/efeitos dos fármacos , Humanos , Pessoa de Meia-Idade , Mucina-5B/química , Mucina-5B/isolamento & purificação , Mucina-5B/farmacologia , Mucinas/química , Mucinas/isolamento & purificação , Mucinas/farmacologia , Saliva/fisiologia , Proteínas e Peptídeos Salivares/química , Proteínas e Peptídeos Salivares/isolamento & purificação , Proteínas e Peptídeos Salivares/farmacologia , Pseudotipagem Viral , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Adulto Jovem
13.
Biomolecules ; 10(10)2020 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-32992542

RESUMO

Mosquitoes inject saliva into the host skin to facilitate blood meal acquisition through active compounds that prevent hemostasis. D7 proteins are among the most abundant components of the mosquito saliva and act as scavengers of biogenic amines and eicosanoids. Several members of the D7 family have been characterized at the biochemical level; however, none have been studied thus far in Aedes albopictus, a permissive vector for several arboviruses that causes extensive human morbidity and mortality. Here, we report the binding capabilities of a D7 long form protein from Ae. albopictus (AlboD7L1) by isothermal titration calorimetry and compared its model structure with previously solved D7 structures. The physiological function of AlboD7L1 was demonstrated by ex vivo platelet aggregation and in vivo leukocyte recruitment experiments. AlboD7L1 binds host hemostasis agonists, including biogenic amines, leukotrienes, and the thromboxane A2 analog U-46619. AlboD7L1 protein model predicts binding of biolipids through its N-terminal domain, while the C-terminal domain binds biogenic amines. We demonstrated the biological function of AlboD7L1 as an inhibitor of both platelet aggregation and cell recruitment of neutrophils and eosinophils. Altogether, this study reinforces the physiological relevance of the D7 salivary proteins as anti-hemostatic and anti-inflammatory molecules that help blood feeding in mosquitoes.


Assuntos
Aedes/química , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Inflamação/genética , Proteínas de Insetos/química , Animais , Hemostasia/efeitos dos fármacos , Humanos , Inflamação/prevenção & controle , Proteínas de Insetos/genética , Proteínas de Insetos/farmacologia , Leucócitos/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Saliva/química , Proteínas e Peptídeos Salivares/química , Proteínas e Peptídeos Salivares/farmacologia
14.
Front Immunol ; 11: 1643, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32849563

RESUMO

Introduction: Proteinuria contributes to progression of renal damage, partly by complement activation on proximal tubular epithelial cells. By pattern recognition, properdin has shown to bind to heparan sulfate proteoglycans on tubular epithelium and can initiate the alternative complement pathway (AP). Properdin however, also binds to C3b(Bb) and properdin binding to tubular cells might be influenced by the presence of C3b(Bb) on tubular cells and/or by variability in properdin proteins in vitro. In this study we carefully evaluated the specificity of the properdin - heparan sulfate interaction and whether this interaction could be exploited in order to block alternative complement activation. Methods: Binding of various properdin preparations to proximal tubular epithelial cells (PTEC) and subsequent AP activation was determined in the presence or absence of C3 inhibitor Compstatin and properdin inhibitor Salp20. Heparan sulfate proteoglycan dependency of the pattern recognition of properdin was evaluated on PTEC knocked down for syndecan-1 by shRNA technology. Solid phase binding assays were used to evaluate the effectivity of heparin(oids) and recombinant Salp20 to block the pattern recognition of properdin. Results: Binding of serum-derived and recombinant properdin preparations to PTECs could be dose-dependently inhibited (P < 0.01) and competed off (P < 0.01) by recombinant Salp20 (IC50: ~125 ng/ml) but not by Compstatin. Subsequent properdin-mediated AP activation on PTECs could be inhibited by Compstatin (P < 0.01) and blocked by recombinant Salp20 (P < 0.05). Syndecan-1 deficiency in PTECs resulted in a ~75% reduction of properdin binding (P = 0.057). In solid-phase binding assays, properdin binding to C3b could be dose-dependently inhibited by recombinant Salp20> heparin(oid) > C3b. Discussion: In this study we showed that all properdin preparations recognize heparan sulfate/syndecan-1 on PTECs with and without Compstatin C3 blocking conditions. In contrast to Compstatin, recombinant Salp20 prevents heparan sulfate pattern recognition by properdin on PTECs. Both complement inhibitors prevented properdin-mediated C3 activation. Binding of properdin to C3b could also be blocked by heparin(oids) and recombinant Salp20. This work indicates that properdin serves as a docking station for AP activation on PTECs and a Salp20 analog or heparinoids may be viable inhibitors in properdin mediated AP activation.


Assuntos
Complemento C3b/metabolismo , Inativadores do Complemento/farmacologia , Células Epiteliais/efeitos dos fármacos , Heparitina Sulfato/metabolismo , Proteínas de Insetos/farmacologia , Túbulos Renais Proximais/efeitos dos fármacos , Properdina/metabolismo , Receptores de Reconhecimento de Padrão/metabolismo , Proteínas e Peptídeos Salivares/farmacologia , Sindecana-1/metabolismo , Animais , Linhagem Celular , Ativação do Complemento/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Ixodes , Túbulos Renais Proximais/metabolismo , Peptídeos Cíclicos/farmacologia , Ligação Proteica , Transdução de Sinais , Sindecana-1/genética
15.
Chem Biol Interact ; 327: 109179, 2020 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-32534990

RESUMO

Excessive osteoclast leads to the imbalance in bone reconstruction and results in osteolytic diseases, such as osteoporosis and rheumatic arthritis. Integrin αvß3 abundantly expresses on osteoclast and plays a critical role in the formation and function of osteoclast, therefore, blockage of αvß3 has become an attractive therapeutic option for osteolytic diseases. In this study, we find that Tablysin-15, a RGD motif containing disintegrin, concentration-dependently suppresses RANKL-induced osteoclastogenesis, F-actin ring formation and bone resorption without affecting the cell viabilities. Tablysin-15 binds to integrin αvß3 and inhibits the activation of FAK-associated signaling pathways. Tablysin-15 also suppresses the activation of NF-кB, MAPK, and Akt-NFATc1 signaling pathways, which are crucial transcription factors during osteoclast differentiation. Moreover, Tablysin-15 decreases the osteoclastogenesis marker gene expression, including MMP-9, TRAP, CTSK, and c-Src. Finally, Tablysin-15 significantly inhibits LPS-induced bone loss in a mouse model. Taken together, our results indicate that Tablysin-15 significantly suppresses osteoclastogenesis in vitro and in vivo, thus it might be a excellent candidate for treating osteolytic-related diseases.


Assuntos
Conservadores da Densidade Óssea/farmacologia , Reabsorção Óssea/prevenção & controle , Proteínas de Insetos/farmacologia , Osteogênese/efeitos dos fármacos , Proteínas e Peptídeos Salivares/farmacologia , Animais , Conservadores da Densidade Óssea/toxicidade , Reabsorção Óssea/induzido quimicamente , Fêmur/efeitos dos fármacos , Fêmur/patologia , Proteínas de Insetos/toxicidade , Integrina alfaVbeta3/metabolismo , Lipopolissacarídeos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/efeitos dos fármacos , Ligante RANK/metabolismo , Células RAW 264.7 , Proteínas e Peptídeos Salivares/toxicidade , Fator de Transcrição RelA/metabolismo , Regulação para Cima/efeitos dos fármacos
16.
Clin Exp Metastasis ; 37(4): 489-497, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32394234

RESUMO

Tissue factor (TF), a blood coagulation protein, plays an important role in tumor growth, invasion, and metastasis. Ixolaris, a tick-derived non-immunogenic molecule that binds to TF, has demonstrated in vivo inhibitory effect on murine models of melanoma, including primary growth and metastasis. This work aimed to: I) develop an efficient and stable labeling technique of ixolaris with Iodine-131(131I); II) compare the biodistribution of 131I and 131I-ixolaris in tumor-free and melanoma-bearing mice; III) evaluate whether 131I-ixolaris could serve as an antimetastatic agent. Ixolaris radioiodination was performed using iodogen, followed by liquid paper chromatography. Labeling stability and anticoagulant activity were measured. Imaging studies were performed after intravenous administration of free 131I or 131I-ixolaris in a murine melanoma model employing the B16-F10 cell line. Animals were divided in three experimental groups: the first experimental group, D0, received a single-dose of 9.25 MBq of 131I-ixolaris at the same day the animals were inoculated with melanoma cells. In the second group, D15, a single-dose of 9.25 MBq of 131I-ixolaris or free 131I was applied into mice on the fifteenth day after the tumor induction. The third group, D1-D15, received two therapeutic doses of 9.25 MBq of 131I-ixolaris or 131I. In vitro studies demonstrated that 131I-ixolaris is stable for up to 24 h and retains its inhibitory activity on blood coagulation. Biodistribution analysis and metastasis assays showed that all treatment regimens with 131I-ixolaris were effective, being the double-treatment (D1/D15) the most effective one. Remarkably, treatment with free 131I showed no anti-metastatic effect. 131I-ixolaris is a promising theranostic agent for metastatic melanoma.


Assuntos
Anticoagulantes/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Melanoma Experimental/tratamento farmacológico , Medicina de Precisão/métodos , Proteínas e Peptídeos Salivares/farmacologia , Tromboplastina/metabolismo , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Radioisótopos do Iodo/farmacologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas e Peptídeos Salivares/metabolismo , Proteínas e Peptídeos Salivares/farmacocinética
17.
Immunobiology ; 225(3): 151937, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32201094

RESUMO

Sand fly saliva presents molecules with potential to development of compounds for treatment of inflammatory diseases. Agaphelin, isolated from the saliva of the mosquito Anopheles gambiae, demonstrates anti-inflammatory properties such as neutrophils chemotaxis inhibition. Here, we extend these results and evaluated the role of agaphelin (0.1-100 nM) in an in vitro model consisting in the activation of human bronchial epithelial cells (BEAS-2B) by IL-4 (50 ng/mL) or lipopolysaccharide (LPS; 10 ng/mL). Agaphelin is non-cytotoxic for BEAS-2B cells. Notably, agaphelin markedly reduces CCL2 and IL-8 production induced by IL-4 or LPS, without altering the IL-10 production. The TLR4 expression and STAT1 phosphorylation induced by LPS were inhibited by agaphlin. In addition, agaphelin decreased the phosphorylation of STAT6 induce by IL-4, whose effect was independent of IL-4-binding activity. Taken together, these findings identify agaphelin as a potential anti-inflammatory therapeutic agent for airway inflammations.


Assuntos
Anti-Inflamatórios/farmacologia , Proteínas de Insetos/farmacologia , Interleucina-4/metabolismo , Lipopolissacarídeos/imunologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Proteínas e Peptídeos Salivares/farmacologia , Biomarcadores , Citocinas/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Ligação Proteica , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo
18.
Curr Cancer Drug Targets ; 20(4): 306-315, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31893992

RESUMO

BACKGROUND: Integrins are crucial anti-cancer therapy targets. We previously showed that tablysin-15 is an integrin antagonist with its Arg-Gly-Asp motif in a novel structural context. OBJECTIVE: Here we investigated the anti-cancer effects and mechanisms of action of tablysin-15 in melanoma cells. METHODS: Cell adhesion, competitive binding, cell viability, and ATP chemiluminescence assays were used to analyze the binding of tablysin-15 to αvß3 integrin and its phenotypic effects. Wound healing, transwells, and zymography were performed to detect motility and matrix metalloproteinase- 2/-9 activities. PARP and caspase-3 cleavage were used as apoptosis assays, while LDH release and flow cytometry were used for necrosis and cell cycle analysis. The expression of mRNAs and proteins of target molecules was measured by qRT-PCR and western blotting, respectively. RESULTS: Tablysin-15 dose-dependently inhibited the proliferation, migration, and invasion of M21 cells through integrin αvß3. The proliferation inhibition caused by tablysin-15 was attributable to G0/G1 phase arrest rather than apoptosis or necrosis. Furthermore, tablysin-15 suppressed MMP-2/- 9 activities and the mRNA expression of MMP-2/-9 and COX-2 but was upregulated TIMP-1 in M21 cells. Meanwhile, tablysin-15 suppressed the expression of cyclin D1/E and CDK 2/6, the phosphorylation of FAK, Akt, and ERK, and nuclear translocation of NF-κB, while increasing the expression of the CDK inhibitor p21waf1/C1. Taken together, tablysin-15 might inhibit melanoma cell metastasis and proliferation by competing with αvß3 integrin, thereby blocking FAK-associated signaling pathways and nuclear translocation of NF-κB. CONCLUSION: Tablysin-15 has reliable anti-cancer effects against M21 melanoma cells, suggesting tablysin-15 is a promising anti-tumor drug.


Assuntos
Proteínas de Insetos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Melanoma/tratamento farmacológico , NF-kappa B/metabolismo , Proteínas e Peptídeos Salivares/farmacologia , Neoplasias Cutâneas/tratamento farmacológico , Apoptose/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Desintegrinas/química , Desintegrinas/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinase 1 de Adesão Focal/antagonistas & inibidores , Quinase 1 de Adesão Focal/metabolismo , Humanos , Integrina alfaVbeta3/antagonistas & inibidores , Integrina alfaVbeta3/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Oligopeptídeos/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia
19.
Cell Adh Migr ; 14(1): 129-138, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-30238848

RESUMO

In a tumor microenvironment, endothelial cell migration and angiogenesis allow cancer to spread to other organs causing metastasis.  Indeed, a number of molecules that are involved in cytoskeleton re-organization and intracellular signaling have been investigated for their effects on tumor cell growth and metastasis. Alongside that, Amblyomin-X, a recombinant Kunitz-type protein, has been shown to reduce metastasis and tumor growth in in vivo experiments. In the present report, we provide a mechanistic insight to these antitumor effects, this is,  Amblyomin-X modulates Rho-GTPases and uPAR signaling, and reduces the release of MMPs, leading to disruption of the actin cytoskeleton and decreased cell migration of tumor cell lines. Altogether, our data support a role for Amblyomin-X as a novel potential antitumor drug. ABBREVIATIONS: Amb-X: Amblyomin-X; ECGF: endotelial cell growth factor; ECM: extracellular matrix; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; HUVEC: human umbilical vein endothelial cell; LRP1: low-density lipoprotein receptor-related protein; MMP: matrix metalloproteinase; HPI-4: hedgehog pathway inhibitor 4; PAI-1: plasminogen activator inhibitor 1; PMA: phorbol 12-myristate-13-acetate; TFPI: tissue factor pathway inhibitor; uPA: urokinase plasminogen activator; uPAR: uPA receptor.


Assuntos
Aprotinina/farmacologia , Proteínas de Artrópodes/farmacologia , Movimento Celular/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Proteínas e Peptídeos Salivares/farmacologia , Adesão Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Humanos , Metaloproteinases da Matriz/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
20.
Behav Brain Res ; 378: 112263, 2020 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-31568834

RESUMO

BACKGROUND: Antidepressants are the first-choice for pharmacological treatment of panic disorder. However, they present disadvantages, such as delayed therapeutic effect, many side effects and a considerable rate of non-responders. These shortcomings prompt the development of new therapeutic strategies. Among these are the adjunctive use of enkephalinase inhibitors, such as opiorphin, which supposedly acts by increasing the availability of brain enkephalins and other endogenous opioids. AIMS: We here evaluated whether opiorphin in the dorsal periaqueductal grey matter (dPAG), a key panic-related area, accelerates and/or facilitates the antipanic-like effect of fluoxetine or imipramine. We also verified whether the panicolytic effect of imipramine depends on activation of µ-opioid receptors (MORs). METHODS: Male Wistar rats were submitted to the escape task of the elevated T-maze, an index of panic attack, after treatment with imipramine (3, 7 or 21 days) or fluoxetine (3, 7, 14 or 21 days), combined with an intra-dPAG injection of opiorphin. RESULTS: Opiorphin facilitated and accelerated the panicolytic-like effect caused by imipramine, but not with fluoxetine. The antipanic-like effect caused by chronic imipramine did not depend on MOR activation in the dPAG. CONCLUSION: Combined treatment of antidepressant drugs with opiorphin for hastening or potentiating the effects of the former compounds may not be generally effective, with the results varying depending on the type/class of these panicolytic drugs.


Assuntos
Antidepressivos/farmacologia , Fluoxetina/farmacologia , Imipramina/farmacologia , Neprilisina/antagonistas & inibidores , Oligopeptídeos/farmacologia , Transtorno de Pânico/tratamento farmacológico , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Inibidores de Proteases/farmacologia , Proteínas e Peptídeos Salivares/farmacologia , Animais , Antidepressivos/administração & dosagem , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Interações Medicamentosas , Quimioterapia Combinada , Fluoxetina/administração & dosagem , Imipramina/administração & dosagem , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Oligopeptídeos/administração & dosagem , Inibidores de Proteases/administração & dosagem , Ratos , Ratos Wistar , Proteínas e Peptídeos Salivares/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...