Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Biotechnol Adv ; 66: 108167, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37164239

RESUMO

The Asian tiger mosquito Aedes albopictus is currently spreading across Europe, facilitated by climate change and global transportation. It is a vector of arboviruses causing human diseases such as chikungunya, dengue hemorrhagic fever and Zika fever. For the majority of these diseases, no vaccines or therapeutics are available. Options for the control of Ae. albopictus are limited by European regulations introduced to protect biodiversity by restricting or phasing out the use of pesticides, genetically modified organisms (GMOs) or products of genome editing. Alternative solutions are thus urgently needed to avoid a future scenario in which Europe faces a choice between prioritizing human health or biodiversity when it comes to Aedes-vectored pathogens. To ensure regulatory compliance and public acceptance, these solutions should preferably not be based on chemicals or GMOs and must be cost-efficient and specific. The present review aims to synthesize available evidence on RNAi-based mosquito vector control and its potential for application in the European Union. The recent literature has identified some potential target sites in Ae. albopictus and formulations for delivery. However, we found little information concerning non-target effects on the environment or human health, on social aspects, regulatory frameworks, or on management perspectives. We propose optimal designs for RNAi-based vector control tools against Ae. albopictus (target product profiles), discuss their efficacy and reflect on potential risks to environmental health and the importance of societal aspects. The roadmap from design to application will provide readers with a comprehensive perspective on the application of emerging RNAi-based vector control tools for the suppression of Ae. albopictus populations with special focus on Europe.


Assuntos
Aedes , Dengue , Infecção por Zika virus , Zika virus , Animais , Humanos , Dengue/genética , Aedes/genética , Interferência de RNA , Europa (Continente) , Mosquitos Vetores/genética
2.
Toxins (Basel) ; 15(5)2023 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-37235338

RESUMO

The venoms of spiders from the RTA (retro-lateral tibia apophysis) clade contain diverse short linear peptides (SLPs) that offer a rich source of therapeutic candidates. Many of these peptides have insecticidal, antimicrobial and/or cytolytic activities, but their biological functions are unclear. Here, we explore the bioactivity of all known members of the A-family of SLPs previously identified in the venom of the Chinese wolf spider (Lycosa shansia). Our broad approach included an in silico analysis of physicochemical properties and bioactivity profiling for cytotoxic, antiviral, insecticidal and antibacterial activities. We found that most members of the A-family can form α-helices and resemble the antibacterial peptides found in frog poison. The peptides we tested showed no cytotoxic, antiviral or insecticidal activities but were able to reduce the growth of bacteria, including clinically relevant strains of Staphylococcus epidermidis and Listeria monocytogenes. The absence of insecticidal activity may suggest that these peptides have no role in prey capture, but their antibacterial activity may help to defend the venom gland against infection.


Assuntos
Inseticidas , Venenos de Aranha , Aranhas , Animais , Peçonhas , Peptídeos/farmacologia , Peptídeos/química , Inseticidas/química , Antibacterianos/farmacologia , Antibacterianos/química , Aranhas/química , Venenos de Aranha/farmacologia , Venenos de Aranha/química
3.
Front Bioeng Biotechnol ; 11: 1166601, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37207126

RESUMO

Venoms are complex chemical arsenals that have evolved independently many times in the animal kingdom. Venoms have attracted the interest of researchers because they are an important innovation that has contributed greatly to the evolutionary success of many animals, and their medical relevance offers significant potential for drug discovery. During the last decade, venom research has been revolutionized by the application of systems biology, giving rise to a novel field known as venomics. More recently, biotechnology has also made an increasing impact in this field. Its methods provide the means to disentangle and study venom systems across all levels of biological organization and, given their tremendous impact on the life sciences, these pivotal tools greatly facilitate the coherent understanding of venom system organization, development, biochemistry, and therapeutic activity. Even so, we lack a comprehensive overview of major advances achieved by applying biotechnology to venom systems. This review therefore considers the methods, insights, and potential future developments of biotechnological applications in the field of venom research. We follow the levels of biological organization and structure, starting with the methods used to study the genomic blueprint and genetic machinery of venoms, followed gene products and their functional phenotypes. We argue that biotechnology can answer some of the most urgent questions in venom research, particularly when multiple approaches are combined together, and with other venomics technologies.

4.
Toxins (Basel) ; 14(12)2022 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-36548743

RESUMO

The venoms of ants (Formicidae) are a promising source of novel bioactive molecules with potential for clinical and agricultural applications. However, despite the rich diversity of ant species, only a fraction of this vast resource has been thoroughly examined in bioprospecting programs. Previous studies focusing on the venom of Central European ants (subfamily Myrmicinae) identified a number of short linear decapeptides and nonapeptides resembling antimicrobial peptides (AMPs). Here, we describe the in silico approach and bioactivity profiling of 10 novel AMP-like peptides from the fellow Central European myrmicine ants Myrmica rubra and Myrmica ruginodis. Using the sequences of known ant venom peptides as queries, we screened the venom gland transcriptomes of both species. We found transcripts of nine novel decapeptides and one novel nonapeptide. The corresponding peptides were synthesized for bioactivity profiling in a broad panel of assays consisting of tests for cytotoxicity as well as antiviral, insecticidal, and antimicrobial activity. U-MYRTX-Mrug5a showed moderately potent antimicrobial effects against several bacteria, including clinically relevant pathogens such as Listeria monocytogenes and Staphylococcus epidermidis, but high concentrations showed negligible cytotoxicity. U-MYRTX-Mrug5a is, therefore, a probable lead for the development of novel peptide-based antibiotics.


Assuntos
Venenos de Formiga , Anti-Infecciosos , Formigas , Animais , Formigas/genética , Peptídeos/química , Transcriptoma , Peçonhas , Venenos de Formiga/toxicidade , Venenos de Formiga/química
5.
Viruses ; 14(11)2022 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-36366550

RESUMO

Influenza is a severe contagious disease caused by influenza A and B viruses. The WHO estimates that annual outbreaks lead to 3-5 million severe infections of which approximately 10% lead to the death of the patient. While vaccination is the cornerstone of prevention, antiviral drugs represent the most important treatment option of acute infections. Only two classes of drugs are currently approved for the treatment of influenza in numerous countries: M2 channel blockers and neuraminidase inhibitors. In some countries, additional compounds such as the recently developed cap-dependent endonuclease inhibitor baloxavir marboxil or the polymerase inhibitor favipiravir are available. However, many of these compounds suffer from poor efficacy, if not applied early after infection. Furthermore, many influenza strains have developed resistances and lost susceptibility to these compounds. As a result, there is an urgent need to develop new anti-influenza drugs against a broad spectrum of subtypes. Natural products have made an important contribution to the development of new lead structures, particularly in the field of infectious diseases. Therefore, this article aims to review the research on the identification of novel lead structures isolated from natural resources suitable to treat influenza infections.


Assuntos
Influenza Humana , Infecções por Orthomyxoviridae , Humanos , Antivirais/farmacologia , Antivirais/uso terapêutico , Infecções por Orthomyxoviridae/tratamento farmacológico , Influenza Humana/tratamento farmacológico , Inibidores Enzimáticos/uso terapêutico , Recursos Naturais
6.
Anal Biochem ; 655: 114836, 2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-35964735

RESUMO

Proprotein convertases (PCs) are involved in the pathogenesis of various diseases, making them promising drug targets. Most assays for PCs have been performed with few standard substrates, regardless of differences in cleavage efficiencies. Derived from studies on substrate-analogue inhibitors, 11 novel substrates were synthesized and characterized with five PCs. H-Arg-Arg-Tle-Lys-Arg-AMC is the most efficiently cleaved furin substrate based on its kcat/KM value. Due to its higher kcat value, acetyl-Arg-Arg-Tle-Arg-Arg-AMC was selected for further measurements to demonstrate the benefit of this improved substrate. Compared to our standard conditions, its use allowed a 10-fold reduction of the furin concentration, which enabled Ki value determinations of previously described tight-binding inhibitors under classical conditions. Under these circumstances, a slow-binding behavior was observed for the first time with inhibitor MI-1148. In addition to furin, four additional PCs were used to characterize these substrates. The most efficiently cleaved PC1/3 substrate was acetyl-Arg-Arg-Arg-Tle-Lys-Arg-AMC. The highest kcat/KM values for PC2 and PC7 were found for the N-terminally unprotected analogue of this substrate, although other substrates possess higher kcat values. The highest efficiency for PC5/6A was observed for the substrate acetyl-Arg-Arg-Tle-Lys-Arg-AMC. In summary, we have identified new substrates for furin, PC1/3, PC2, and PC7 suitable for improved enzyme-kinetic measurements.


Assuntos
Furina , Pró-Proteína Convertases , Sequência de Aminoácidos , Carbamatos , Corantes Fluorescentes , Oligopeptídeos , Proteínas , Subtilisinas/metabolismo
7.
Toxins (Basel) ; 14(1)2022 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-35051034

RESUMO

Linear cationic venom peptides are antimicrobial peptides (AMPs) that exert their effects by damaging cell membranes. These peptides can be highly specific, and for some, a significant therapeutic value was proposed, in particular for treatment of bacterial infections. A prolific source of novel AMPs are arthropod venoms, especially those of hitherto neglected groups such as pseudoscorpions. In this study, we describe for the first time pharmacological effects of AMPs discovered in pseudoscorpion venom. We examined the antimicrobial, cytotoxic, and insecticidal activity of full-length Checacin1, a major component of the Chelifer cancroides venom, and three truncated forms of this peptide. The antimicrobial tests revealed a potent inhibitory activity of Checacin1 against several bacteria and fungi, including methicillin resistant Staphylococcus aureus (MRSA) and even Gram-negative pathogens. All peptides reduced survival rates of aphids, with Checacin1 and the C-terminally truncated Checacin11-21 exhibiting effects comparable to Spinosad, a commercially used pesticide. Cytotoxic effects on mammalian cells were observed mainly for the full-length Checacin1. All tested peptides might be potential candidates for developing lead structures for aphid pest treatment. However, as these peptides were not yet tested on other insects, aphid specificity has not been proven. The N- and C-terminal fragments of Checacin1 are less potent against aphids but exhibit no cytotoxicity on mammalian cells at the tested concentration of 100 µM.


Assuntos
Anti-Infecciosos , Proteínas de Artrópodes , Venenos de Artrópodes , Citotoxinas , Inseticidas , Sequência de Aminoácidos , Animais , Anti-Infecciosos/química , Anti-Infecciosos/farmacologia , Anti-Infecciosos/toxicidade , Afídeos/efeitos dos fármacos , Aracnídeos , Proteínas de Artrópodes/química , Proteínas de Artrópodes/farmacologia , Proteínas de Artrópodes/toxicidade , Venenos de Artrópodes/química , Venenos de Artrópodes/farmacologia , Venenos de Artrópodes/toxicidade , Citotoxinas/química , Citotoxinas/farmacologia , Citotoxinas/toxicidade , Cães , Inseticidas/química , Inseticidas/farmacologia , Inseticidas/toxicidade , Células Madin Darby de Rim Canino , Alinhamento de Sequência
8.
Life Sci Alliance ; 3(9)2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32703818

RESUMO

The novel emerged SARS-CoV-2 has rapidly spread around the world causing acute infection of the respiratory tract (COVID-19) that can result in severe disease and lethality. For SARS-CoV-2 to enter cells, its surface glycoprotein spike (S) must be cleaved at two different sites by host cell proteases, which therefore represent potential drug targets. In the present study, we show that S can be cleaved by the proprotein convertase furin at the S1/S2 site and the transmembrane serine protease 2 (TMPRSS2) at the S2' site. We demonstrate that TMPRSS2 is essential for activation of SARS-CoV-2 S in Calu-3 human airway epithelial cells through antisense-mediated knockdown of TMPRSS2 expression. Furthermore, SARS-CoV-2 replication was also strongly inhibited by the synthetic furin inhibitor MI-1851 in human airway cells. In contrast, inhibition of endosomal cathepsins by E64d did not affect virus replication. Combining various TMPRSS2 inhibitors with furin inhibitor MI-1851 produced more potent antiviral activity against SARS-CoV-2 than an equimolar amount of any single serine protease inhibitor. Therefore, this approach has considerable therapeutic potential for treatment of COVID-19.


Assuntos
Células Epiteliais Alveolares/virologia , Betacoronavirus/fisiologia , Furina/genética , Serina Endopeptidases/genética , Glicoproteína da Espícula de Coronavírus/metabolismo , Células Epiteliais Alveolares/citologia , Animais , Sítios de Ligação , Linhagem Celular , Chlorocebus aethiops , Células HEK293 , Humanos , Proteólise , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/química , Células Vero , Internalização do Vírus , Replicação Viral
9.
J Virol ; 93(21)2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31391268

RESUMO

Cleavage of influenza virus hemagglutinin (HA) by host cell proteases is essential for virus infectivity and spread. We previously demonstrated in vitro that the transmembrane protease TMPRSS2 cleaves influenza A virus (IAV) and influenza B virus (IBV) HA possessing a monobasic cleavage site. Subsequent studies revealed that TMPRSS2 is crucial for the activation and pathogenesis of H1N1pdm and H7N9 IAV in mice. In contrast, activation of H3N2 IAV and IBV was found to be independent of TMPRSS2 expression and supported by an as-yet-undetermined protease(s). Here, we investigated the role of TMPRSS2 in proteolytic activation of IAV and IBV in three human airway cell culture systems: primary human bronchial epithelial cells (HBEC), primary type II alveolar epithelial cells (AECII), and Calu-3 cells. Knockdown of TMPRSS2 expression was performed using a previously described antisense peptide-conjugated phosphorodiamidate morpholino oligomer, T-ex5, that interferes with splicing of TMPRSS2 pre-mRNA, resulting in the expression of enzymatically inactive TMPRSS2. T-ex5 treatment produced efficient knockdown of active TMPRSS2 in all three airway cell culture models and prevented proteolytic activation and multiplication of H7N9 IAV in Calu-3 cells and H1N1pdm, H7N9, and H3N2 IAV in HBEC and AECII. T-ex5 treatment also inhibited the activation and spread of IBV in AECII but did not affect IBV activation in HBEC and Calu-3 cells. This study identifies TMPRSS2 as the major HA-activating protease of IAV in human airway cells and IBV in type II pneumocytes and as a potential target for the development of novel drugs to treat influenza infections.IMPORTANCE Influenza A viruses (IAV) and influenza B viruses (IBV) cause significant morbidity and mortality during seasonal outbreaks. Cleavage of the viral surface glycoprotein hemagglutinin (HA) by host proteases is a prerequisite for membrane fusion and essential for virus infectivity. Inhibition of relevant proteases provides a promising therapeutic approach that may avoid the development of drug resistance. HA of most influenza viruses is cleaved at a monobasic cleavage site, and a number of proteases have been shown to cleave HA in vitro This study demonstrates that the transmembrane protease TMPRSS2 is the major HA-activating protease of IAV in primary human bronchial cells and of both IAV and IBV in primary human type II pneumocytes. It further reveals that human and murine airway cells can differ in their HA-cleaving protease repertoires. Our data will help drive the development of potent and selective protease inhibitors as novel drugs for influenza treatment.


Assuntos
Vírus da Influenza A/fisiologia , Vírus da Influenza B/fisiologia , Influenza Humana/virologia , Serina Endopeptidases/metabolismo , Animais , Brônquios/citologia , Células Cultivadas , Células Epiteliais/virologia , Técnicas de Silenciamento de Genes , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Influenza Humana/enzimologia , Influenza Humana/metabolismo , Camundongos , Infecções por Orthomyxoviridae/enzimologia , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Alvéolos Pulmonares/citologia , Serina Endopeptidases/genética , Regulação para Cima , Replicação Viral
10.
FEBS J ; 286(22): 4597-4620, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31276291

RESUMO

The basic proprotein convertases (PCs) furin, PC1/3, PC2, PC5/6, PACE4, PC4, and PC7 are promising drug targets for human diseases. However, developing selective inhibitors remains challenging due to overlapping substrate recognition motifs and limited structural information. Classical drug screening approaches for basic PC inhibitors involve homogeneous biochemical assays using soluble recombinant enzymes combined with fluorogenic substrate peptides that may not accurately recapitulate the complex cellular context of the basic PC-substrate interaction. Herein we report basic PC sensor (BPCS), a novel cell-based molecular sensor that allows rapid screening of candidate inhibitors and their selectivity toward individual basic PCs within mammalian cells. BPCS consists of Gaussia luciferase linked to a sortilin-1 membrane anchor via a cleavage motif that allows efficient release of luciferase specifically if individual basic PCs are provided in the same membrane. Screening of selected candidate peptidomimetic inhibitors revealed that BPCS can readily distinguish between general and selective PC inhibitors in a high-throughput screening format. The robust and cost-effective assay format of BPCS makes it suitable to identify novel specific small-molecule inhibitors against basic PCs for therapeutic application. Its cell-based nature will allow screening for drug targets in addition to the catalytically active mature enzyme, including maturation, transport, and cellular factors that modulate the enzyme's activity. This broadened 'target range' will enhance the likelihood to identify novel small-molecule compounds that inhibit basic PCs in a direct or indirect manner and represents a conceptual advantage.


Assuntos
Técnicas Biossensoriais/métodos , Descoberta de Drogas/métodos , Inibidores Enzimáticos/farmacologia , Peptidomiméticos/farmacologia , Pró-Proteína Convertases/metabolismo , Células A549 , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Técnicas Biossensoriais/normas , Descoberta de Drogas/normas , Inibidores Enzimáticos/química , Genes Reporter , Células HEK293 , Células HeLa , Ensaios de Triagem em Larga Escala/métodos , Ensaios de Triagem em Larga Escala/normas , Humanos , Luciferases/genética , Luciferases/metabolismo , Peptidomiméticos/química , Pró-Proteína Convertases/antagonistas & inibidores , Sensibilidade e Especificidade
11.
J Med Chem ; 62(7): 3513-3523, 2019 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-30852892

RESUMO

Tridegin is a potent and specific 66mer peptide inhibitor of coagulation factor XIIIa with six cysteines involved in three disulfide bonds. Three of the 15 possible 3-disulfide-bonded isomers have been identified, which share a bridge between cysteines 19 and 25. We synthesized the three possible 2-disulfide-bonded analogues using a targeted protecting group strategy to investigate the impact of the C19-C25 bond on tridegin's folding, stability, and function. The FXIIIa inhibitory activity of the analogues was retained, which was shown by in vitro fluorogenic activity and whole blood clotting assays. Molecular dynamics simulations of wild-type tridegin and the analogues as well as molecular docking studies with FXIIIa were performed to elucidate the impact of the C19-C25 bond on conformational stability and binding mode. The strategy of selectively reducing disulfide bonds to facilitate large-scale synthesis, while retaining the functionality of disulfide-bonded peptides, has been demonstrated with our present study.


Assuntos
Dissulfetos/química , Fator XIIIa/antagonistas & inibidores , Proteínas e Peptídeos Salivares/farmacologia , Animais , Coagulação Sanguínea/efeitos dos fármacos , Humanos , Isomerismo , Sanguessugas , Simulação de Dinâmica Molecular , Dobramento de Proteína , Estabilidade Proteica , Proteínas e Peptídeos Salivares/química
12.
ChemMedChem ; 14(6): 673-685, 2019 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-30680958

RESUMO

The activation of viral glycoproteins by the host protease furin is an essential step in the replication of numerous pathogenic viruses. Thus, effective inhibitors of furin could serve as broad-spectrum antiviral drugs. A crystal structure of an inhibitory hexapeptide derivative in complex with furin served as template for the rational design of various types of new cyclic inhibitors. Most of the prepared derivatives are relatively potent furin inhibitors with inhibition constants in the low nanomolar or even sub-nanomolar range. For seven derivatives the crystal structures in complex with furin could be determined. In three complexes, electron density was found for the entire inhibitor. In the other cases the structures could be determined only for the P6/P5-P1 segments, which directly interact with furin. The cyclic derivatives together with two non-cyclic reference compounds were tested as inhibitors of the proteolytic activation and replication of respiratory syncytial virus in cells. Significant antiviral activity was found for both linear reference inhibitors, whereas a negligible efficacy was determined for the cyclic derivatives.


Assuntos
Inibidores Enzimáticos/farmacologia , Furina/antagonistas & inibidores , Compostos Macrocíclicos/farmacologia , Pró-Proteína Convertases/antagonistas & inibidores , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Humanos , Compostos Macrocíclicos/síntese química
13.
Cell Rep ; 25(2): 312-320.e7, 2018 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-30304672

RESUMO

Bats harbor a plethora of viruses with an unknown zoonotic potential. In-depth functional characterization of such viruses is often hampered by a lack of virus isolates. The genome of a virus closely related to human mumps viruses (hMuV) was detected in African fruit bats, batMuV. Efforts to characterize batMuV were based on directed expression of the batMuV glycoproteins or use of recombinant chimeric hMuVs harboring batMuV glycoprotein. Although these studies provided initial insights into the functionality of batMuV glycoproteins, the host range, replication competence, immunomodulatory functions, virulence, and zoonotic potential of batMuV remained elusive. Here, we report the successful rescue of recombinant batMuV. BatMuV infects human cells, is largely resistant to the host interferon response, blocks interferon induction and TNF-α activation, and is neurotoxic in rats. Anti-hMuV antibodies efficiently neutralize batMuV. The striking similarities between hMuV and batMuV point at the putative zoonotic potential of batMuV.


Assuntos
Quirópteros/virologia , Evasão da Resposta Imune/imunologia , Vírus da Caxumba/imunologia , Caxumba/virologia , Síndromes Neurotóxicas/etiologia , Internalização do Vírus , Replicação Viral , Animais , Feminino , Humanos , Vírus da Caxumba/patogenicidade , Síndromes Neurotóxicas/patologia , Ratos , Ratos Endogâmicos Lew
14.
Biochemistry ; 57(6): 925-934, 2018 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-29314830

RESUMO

The proprotein convertase furin is a highly specific serine protease modifying and thereby activating proteins in the secretory pathway by proteolytic cleavage. Its substrates are involved in many diseases, including cancer and infections caused by bacteria and viruses. Understanding furin's substrate specificity is crucially important for the development of pharmacologically applicable inhibitors. Using protein X-ray crystallography, we investigated the extended substrate binding site of furin in complex with three peptide-derived inhibitors at up to 1.9 Å resolution. The structure of the protease bound with a hexapeptide inhibitor revealed molecular details of its S6 pocket, which remained completely unknown so far. The arginine residue at P6 induced an unexpected turnlike conformation of the inhibitor backbone, which is stabilized by intra- and intermolecular H-bonds. In addition, we confirmed the binding of arginine to the previously proposed S5 pocket (S51). An alternative S5 site (S52) could be utilized by shorter side chains as demonstrated for a 4-aminomethyl-phenylacetyl residue, which shows steric properties similar to those of a lysine side chain. Interestingly, we also observed binding of a peptide with citrulline at P4 substituting for the highly conserved arginine. The structural data might indicate an unusual protonation state of Asp264 maintaining the interaction with uncharged citrulline. The herein identified molecular interaction sites at P5 and P6 can be utilized to improve next-generation furin inhibitors. Our data will also help to predict furin substrates more precisely on the basis of the additional specificity determinants observed for P5 and P6.


Assuntos
Furina/química , Sítios de Ligação , Cristalografia por Raios X , Furina/antagonistas & inibidores , Furina/metabolismo , Células HEK293 , Humanos , Ligação de Hidrogênio , Simulação de Acoplamento Molecular , Peptídeos/química , Peptídeos/farmacologia , Inibidores de Proteases/química , Inibidores de Proteases/farmacologia , Conformação Proteica , Especificidade por Substrato
15.
ChemMedChem ; 12(23): 1953-1968, 2017 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-29059503

RESUMO

The proprotein convertase furin is a potential target for drug design, especially for the inhibition of furin-dependent virus replication. All effective synthetic furin inhibitors identified thus far are multibasic compounds; the highest potency was found for our previously developed inhibitor 4-(guanidinomethyl)phenylacetyl-Arg-Tle-Arg-4-amidinobenzylamide (MI-1148). An initial study in mice revealed a narrow therapeutic range for this tetrabasic compound, while significantly reduced toxicity was observed for some tribasic analogues. This suggests that the toxicity depends at least to some extent on the overall multibasic character of this inhibitor. Therefore, in a first approach, the C-terminal benzamidine of MI-1148 was replaced by less basic P1 residues. Despite decreased potency, a few compounds still inhibit furin in the low nanomolar range, but display negligible efficacy in cells. In a second approach, the P2 arginine was replaced by lysine; compared to MI-1148, this furin inhibitor has slightly decreased potency, but exhibits similar antiviral activity against West Nile and Dengue virus in cell culture and decreased toxicity in mice. These results provide a promising starting point for the development of efficacious and well-tolerated furin inhibitors.


Assuntos
Antivirais/farmacologia , Inibidores Enzimáticos/farmacologia , Furina/antagonistas & inibidores , Animais , Antivirais/síntese química , Antivirais/química , Células Cultivadas , Vírus da Dengue/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Furina/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos ICR , Testes de Sensibilidade Microbiana , Estrutura Molecular , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato , Vírus do Nilo Ocidental/efeitos dos fármacos
16.
J Enzyme Inhib Med Chem ; 32(1): 712-721, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28385094

RESUMO

West Nile virus (WNV) and Dengue virus (DENV) replication depends on the viral NS2B-NS3 protease and the host enzyme furin, which emerged as potential drug targets. Modification of our previously described WNV protease inhibitors by basic phenylalanine analogs provided compounds with reduced potency against the WNV and DENV protease. In a second series, their decarboxylated P1-trans-(4-guanidino)cyclohexylamide was replaced by an arginyl-amide moiety. Compound 4-(guanidinomethyl)-phenylacetyl-Lys-Lys-Arg-NH2 inhibits the NS2B-NS3 protease of WNV with an inhibition constant of 0.11 µM. Due to the similarity in substrate specificity, we have also tested the potency of our previously described multibasic furin inhibitors. Their further modification provided chimeric inhibitors with additional potency against the WNV and DENV proteases. A strong inhibition of WNV and DENV replication in cell culture was observed for the specific furin inhibitors, which reduced virus titers up to 10,000-fold. These studies reveal that potent inhibitors of furin can block the replication of DENV and WNV.


Assuntos
Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Furina/antagonistas & inibidores , Inibidores de Proteases/farmacologia , Proteínas não Estruturais Virais/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos , Vírus do Nilo Ocidental/efeitos dos fármacos , Antivirais/síntese química , Antivirais/química , Vírus da Dengue/enzimologia , Vírus da Dengue/crescimento & desenvolvimento , Relação Dose-Resposta a Droga , Furina/metabolismo , Testes de Sensibilidade Microbiana , Estrutura Molecular , Inibidores de Proteases/síntese química , Inibidores de Proteases/química , RNA Helicases/antagonistas & inibidores , RNA Helicases/metabolismo , Serina Endopeptidases/metabolismo , Relação Estrutura-Atividade , Proteínas não Estruturais Virais/metabolismo , Vírus do Nilo Ocidental/enzimologia , Vírus do Nilo Ocidental/crescimento & desenvolvimento
17.
ChemMedChem ; 12(8): 613-620, 2017 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-28334511

RESUMO

Novel elongated and shortened derivatives of the peptidomimetic furin inhibitor phenylacetyl-Arg-Val-Arg-4-amidinobenzylamide were synthesized. The most potent compounds, such as Nα (carbamidoyl)Arg-Arg-Val-Arg-4-amidinobenzylamide (Ki =6.2 pm), contain additional basic residues at the N terminus and inhibit furin in the low-picomolar range. Furthermore, to decrease the molecular weight of this inhibitor type, compounds that lack the P5 moiety were prepared. The best inhibitors of this series, 5-(guanidino)valeroyl-Val-Arg-4-amidinobenzylamide and its P3 tert-leucine analogue displayed Ki values of 2.50 and 1.26 nm, respectively. Selected inhibitors, together with our previously described 4-amidinobenzylamide derivatives as references, were tested in cell culture for their activity against furin-dependent infectious pathogens. The propagation of the alphaviruses Semliki Forest virus and chikungunya virus was strongly inhibited in the presence of selected derivatives. Moreover, a significant protective effect of the inhibitors against diphtheria toxin was observed. These results confirm that the inhibition of furin should be a promising approach for the short-term treatment of acute infectious diseases.


Assuntos
Benzamidas/farmacologia , Furina/antagonistas & inibidores , Oligopeptídeos/farmacologia , Peptidomiméticos/farmacologia , Inibidores de Serina Proteinase/farmacologia , Animais , Antivirais/síntese química , Antivirais/farmacologia , Benzamidas/síntese química , Linhagem Celular , Vírus Chikungunya/efeitos dos fármacos , Chlorocebus aethiops , Cricetinae , Toxina Diftérica/metabolismo , Furina/metabolismo , Oligopeptídeos/síntese química , Peptidomiméticos/síntese química , Vírus da Floresta de Semliki/efeitos dos fármacos , Inibidores de Serina Proteinase/síntese química
18.
Microbiologyopen ; 5(4): 637-46, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27038342

RESUMO

GxGD-type intramembrane cleaving proteases (I-CLiPs) form a family of proteolytic enzymes that feature an aspartate-based catalytic mechanism. Yet, they structurally and functionally largely differ from the classical pepsin-like aspartic proteases. Among them are the archaeal enzyme FlaK, processing its substrate FlaB2 during the formation of flagella and γ-secretase, which is centrally involved in the etiology of the neurodegenerative Alzheimer's disease. We developed an optimized activity assay for FlaK and based on screening of a small in-house library and chemical synthesis, we identified compound 9 as the first inhibitor of this enzyme. Our results show that this intramembrane protease differs from classical pepsin-like aspartic proteases and give insights into the substrate recognition of this enzyme. By providing the needed tools to further study the enzymatic cycle of FlaK, our results also enable further studies towards a functional understanding of other GxGD-type I-CLiPs.


Assuntos
Endopeptidases/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Mathanococcus/enzimologia , Inibidores de Proteases/metabolismo , Sequência de Aminoácidos , Proteínas Arqueais/metabolismo , Flagelos/metabolismo , Flagelina/metabolismo , Proteínas de Membrana/metabolismo
19.
Eur J Cell Biol ; 94(7-9): 375-83, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26095298

RESUMO

Cellular proteases are reponsible for activation of influenza virus hemagglutinin (HA) in epithelial tissues of the respiratory tract. The trans-Golgi network (TGN) is the main subcellular compartment where HA cleavage occurs during its biosynthesis. The proteolytic HA cleavage is an indispensable prerequisite for the fusion of viral with endosomal membrane and the delivery of the virus genome into the cell. Both, the structure and accessibility of the HA cleavage site determine the responsible host protease(s) for cutting. Most influenza virus strains contain a HA sequence with a single arginine at the cleavage site suitable for processing by the trypsin-like serine proteases human airway trypsin-like protease (HAT) and transmembrane protease serine 2 (TMPRSS2), albeit a minority of viruses possesses HA cleavage site motifs that are processed by other proteases. TMPRSS2-deficient mice demonstrated the relevance of TMPRSS2 for pneumotropism and pathogenicity of H1N1 and H7N9 virus infections. In contrast, H3N2 virus infections are promoted by an additional not yet identified protease. Highly pathogenic avian H5 and H7 viruses are characterized by an enlarged cleavage site loop containing a multibasic amino acid motif, where the eukaryotic subtilases furin or PC5/6 cleave. Their ubiquitous presence in the organism allows a systemic virus infection. Peptidomimetic inhibitors derived from the HA cleavage site inhibit the HA-activating proteases and thus virus propagation.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Vírus da Influenza A Subtipo H1N1/patogenicidade , Subtipo H7N9 do Vírus da Influenza A/patogenicidade , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Serina Proteases/metabolismo , Animais , Furina/metabolismo , Glicoproteínas de Hemaglutininação de Vírus da Influenza/biossíntese , Humanos , Vírus da Influenza A Subtipo H3N2/patogenicidade , Influenza Humana/patologia , Influenza Humana/virologia , Camundongos , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Pró-Proteína Convertase 5/metabolismo , Mucosa Respiratória/virologia , Rede trans-Golgi/metabolismo
20.
Antiviral Res ; 120: 89-100, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26022200

RESUMO

Antiviral medication is used for the treatment of severe influenza infections, of which the neuraminidase inhibitors (NAIs) are the most effective drugs, approved so far. Here, we investigated the antiviral efficacy of the peptidomimetic furin inhibitor MI-701 in combination with oseltamivir carboxylate and ribavirin against the infection of highly pathogenic avian influenza viruses (HPAIV) that are activated by the host protease furin. Cell cultures infected with the strains A/Thailand/1(KAN-1)/2004 (H5N1) and A/FPV/Rostock/1934 (H7N1) were treated with each agent alone, or in double and triple combinations. MI-701 alone achieved a concentration-dependent reduction of virus propagation. Double treatment of MI-701 with oseltamivir carboxylate and triple combination with ribavirin showed synergistic inhibition and a pronounced delay of virus propagation. MI-701 resistant mutants were not observed. Emergence of NA mutation H275Y conferring high oseltamivir resistance was significantly delayed in the presence of MI-701. Our data indicate that combination with a potent furin inhibitor significantly enhances the therapeutic efficacy of conventional antivirals drugs against HPAIV infection.


Assuntos
Antivirais/metabolismo , Furina/antagonistas & inibidores , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Vírus da Influenza A Subtipo H7N1/efeitos dos fármacos , Oseltamivir/metabolismo , Peptidomiméticos/metabolismo , Ribavirina/metabolismo , Animais , Cães , Farmacorresistência Viral , Sinergismo Farmacológico , Virus da Influenza A Subtipo H5N1/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H7N1/crescimento & desenvolvimento , Células Madin Darby de Rim Canino , Testes de Sensibilidade Microbiana , Proteínas Mutantes/genética , Mutação de Sentido Incorreto , Neuraminidase/genética , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA