Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 463
Filtrar
1.
Viruses ; 16(4)2024 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-38675866

RESUMO

Gu-Sui-Bu, the dried rhizome of Davallia mariesii, is a traditional Chinese herbal remedy with a significant history of treating osteoporosis and inflammatory conditions. However, its potential as an anti-influenza agent and its underlying mechanisms of action remain unexplored. To obtain a more potent extract from D. mariesii and gain insights into its mechanism of action against influenza A virus (IAV), we utilized a partitioning process involving organic solvents and water, resulting in the isolation of butanolic subfractions of the D. mariesii extract (DMBE). DMBE exhibited a broad anti-viral spectrum, effectively inhibiting IAV, with an EC50 of 24.32 ± 6.19 µg/mL and a selectivity index of 6.05. We subsequently conducted a series of in vitro assays to evaluate the antiviral effects of DMBE and to uncover its mechanisms of action. DMBE was found to inhibit IAV during the early stages of infection by hindering the attachment of the virus onto and its penetration into host cells. Importantly, DMBE was observed to hinder IAV-mediated cell-cell fusion. It also inhibited neuraminidase activity, plaque size, and the expression levels of phospho-AKT. In summary, this study provides evidence for the effectiveness of D. mariesii as a complementary and alternative herbal remedy against IAV. Specifically, our data highlight DMBE's capabilities in inhibiting viral entry and the release of virions.


Assuntos
Antivirais , Vírus da Influenza A , Extratos Vegetais , Antivirais/farmacologia , Antivirais/química , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/fisiologia , Humanos , Extratos Vegetais/farmacologia , Extratos Vegetais/química , Animais , Células Madin Darby de Rim Canino , Cães , Internalização do Vírus/efeitos dos fármacos , Sapindaceae/química , Replicação Viral/efeitos dos fármacos , Ligação Viral/efeitos dos fármacos , Influenza Humana/tratamento farmacológico , Influenza Humana/virologia , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/química , Neuraminidase/metabolismo , Células A549 , Linhagem Celular
2.
Microb Pathog ; 190: 106628, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38508422

RESUMO

Rotavirus infections in suckling and weaning piglets cause severe dehydration and death, resulting in significant economic losses in the pig breeding industry. With the continuous emergence of porcine rotavirus (PoRV) variants and poor vaccine cross-protection among various genotypes, there is an urgent need to develop alternative strategies such as seeking effective antiviral products from nature, microbial metabolites and virus-host protein interaction. Sialidases play a crucial role in various physiopathological processes and offer a promising target for developing antivirus drugs. However, the effect of bacterial-derived sialidases on the infection of PoRVs remains largely unknown. Herein, we investigated the impact of bacterial-derived sialidases (sialidase Cp and Vc) on PoRV strain OSU(Group A) infection, using differentiated epithelial monkey kidney cells (MA104) as a model. Our results indicated that the pretreatment of MA104 with exogenous sialidases effectively suppressed PoRV OSU in a concentration-dependent manner. Notably, even at a concentration of 0.01 µU/mL, sialidases significantly inhibited the virus (MOI = 0.01). Meanwhile, we found that sialidase Vc pretreatment sharply reduced the binding rate of PoRV OSU. Last, we demonstrated that PoRV OSU might recognize α-2,3-linked sialic acid as the primary attachment factor in MA104. Our findings provide new insights into the underlying mechanism of PoRV OSU infections, shedding lights on the development of alternative antivirus approaches based on bacteria-virus interaction.


Assuntos
Neuraminidase , Infecções por Rotavirus , Rotavirus , Replicação Viral , Animais , Neuraminidase/metabolismo , Neuraminidase/genética , Rotavirus/efeitos dos fármacos , Rotavirus/fisiologia , Suínos , Replicação Viral/efeitos dos fármacos , Linhagem Celular , Células Epiteliais/virologia , Células Epiteliais/microbiologia , Ligação Viral/efeitos dos fármacos , Ácido N-Acetilneuramínico/metabolismo , Ácido N-Acetilneuramínico/farmacologia , Antivirais/farmacologia , Haplorrinos , Doenças dos Suínos/virologia , Doenças dos Suínos/microbiologia
3.
J Virol ; 97(3): e0146322, 2023 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-36779754

RESUMO

Entry of influenza A viruses (IAVs) into host cells is initiated by binding to sialic acids (Sias), their primary host cell receptor, followed by endocytosis and membrane fusion to release the viral genome into the cytoplasm of the host cell. Host tropism is affected by these entry processes, with a primary factor being receptor specificity. Sias exist in several different chemical forms, including the hydroxylated N-glycolylneuraminic acid (Neu5Gc), which is found in many hosts; however, it has not been clear how modified Sias affect viral binding and entry. Neu5Gc is commonly found in many natural influenza hosts, including pigs and horses, but not in humans or ferrets. Here, we engineered HEK293 cells to express the hydoxylase gene (CMAH) that converts Neu5Ac to Neu5Gc, or knocked out the Sia-CMP transport gene (SLC35A1), resulting in cells that express 95% Neu5Gc or minimal level of Sias, respectively. H3N2 (X-31) showed significantly reduced infectivity in Neu5Gc-rich cells compared to wild-type HEK293 (>95% Neu5Ac). To determine the effects on binding and fusion, we generated supported lipid bilayers (SLBs) derived from the plasma membranes of these cells and carried out single particle microscopy. H3N2 (X-31) exhibited decreased binding to Neu5Gc-containing SLBs, but no significant difference in H3N2 (X-31)'s fusion kinetics to either SLB type, suggesting that reduced receptor binding does not affect subsequent membrane fusion. This finding suggests that for this virus to adapt to host cells rich in Neu5Gc, only receptor affinity changes are required without further adaptation of virus fusion machinery. IMPORTANCE Influenza A virus (IAV) infections continue to threaten human health, causing over 300,000 deaths yearly. IAV infection is initiated by the binding of influenza glycoprotein hemagglutinin (HA) to host cell sialic acids (Sias) and the subsequent viral-host membrane fusion. Generally, human IAVs preferentially bind to the Sia N-acetylneuraminic acid (Neu5Ac). Yet, other mammalian hosts, including pigs, express diverse nonhuman Sias, including N-glycolylneuraminic acid (Neu5Gc). The role of Neu5Gc in human IAV infections in those hosts is not well-understood, and the variant form may play a role in incidents of cross-species transmission and emergence of new epidemic variants. Therefore, it is important to investigate how human IAVs interact with Neu5Ac and Neu5Gc. Here, we use membrane platforms that mimic the host cell surface to examine receptor binding and membrane fusion events of human IAV H3N2. Our findings improve the understanding of viral entry mechanisms that can affect host tropism and virus evolution.


Assuntos
Interações entre Hospedeiro e Microrganismos , Vírus da Influenza A Subtipo H3N2 , Ácidos Siálicos , Internalização do Vírus , Animais , Humanos , Células HEK293 , Vírus da Influenza A Subtipo H3N2/genética , Vírus da Influenza A Subtipo H3N2/metabolismo , Fusão de Membrana , Proteínas de Transporte de Nucleotídeos/genética , Proteínas de Transporte de Nucleotídeos/metabolismo , Ácidos Siálicos/química , Ácidos Siálicos/farmacologia , Imagem Individual de Molécula , Ligação Viral/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Interações entre Hospedeiro e Microrganismos/genética , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia
4.
Thromb Haemost ; 122(6): 984-997, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35322395

RESUMO

Two years since the outbreak of the novel coronavirus SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) pandemic, there remain few clinically effective drugs to complement vaccines. One is the anticoagulant, heparin, which in 2004 was found able to inhibit invasion of SARS-CoV (CoV-1) and which has been employed during the current pandemic to prevent thromboembolic complications and moderate potentially damaging inflammation. Heparin has also been shown experimentally to inhibit SARS-CoV-2 attachment and infection in susceptible cells. At high therapeutic doses however, heparin increases the risk of bleeding and prolonged use can cause heparin-induced thrombocytopenia, a serious side effect. One alternative, with structural similarities to heparin, is the plant-derived, semi-synthetic polysaccharide, pentosan polysulfate (PPS). PPS is an established drug for the oral treatment of interstitial cystitis, is well-tolerated, and exhibits weaker anticoagulant effects than heparin. In an established Vero cell model, PPS and its fractions of varying molecular weights inhibited invasion by SARS-CoV-2. Intact PPS and its size-defined fractions were characterized by molecular weight distribution and chemical structure using nuclear magnetic resonance spectroscopy and liquid chromatography-mass spectrometry, then employed to explore the structural basis of interactions with SARS-CoV-2 spike protein receptor-binding domain (S1 RBD) and the inhibition of Vero cell invasion. PPS was as effective as unfractionated heparin, but more effective in inhibiting cell infection than low-molecular-weight heparin (on a weight/volume basis). Isothermal titration calorimetry and viral plaque-forming assays demonstrated size-dependent binding to S1 RBD and inhibition of Vero cell invasion, suggesting the potential application of PPS as a novel inhibitor of SARS-CoV-2 infection.


Assuntos
Poliéster Sulfúrico de Pentosana , SARS-CoV-2 , Ligação Viral , Animais , Anticoagulantes/farmacologia , Chlorocebus aethiops , Heparina/uso terapêutico , Poliéster Sulfúrico de Pentosana/farmacologia , Ligação Proteica , SARS-CoV-2/efeitos dos fármacos , Glicoproteína da Espícula de Coronavírus , Células Vero , Ligação Viral/efeitos dos fármacos
5.
Molecules ; 27(4)2022 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-35209042

RESUMO

Porcine pseudorabies (PR) is an important infectious disease caused by pseudorabies virus (PRV), which poses a major threat to food safety and security. Vaccine immunization has become the main means to prevent and control the disease. However, since 2011, a new PRV variant has caused huge economic losses to the Chinese pig industry. Panax notoginseng polysaccharides have immunomodulatory activity and other functions, but the antiviral effect has not been reported. We studied the anti-PRV activity of Panax notoginseng polysaccharides in vitro. A less cytopathic effect was observed by increasing the concentration of Panax notoginseng polysaccharides. Western blot, TCID50, plaque assay, and IFA revealed that Panax notoginseng polysaccharides could significantly inhibit the infectivity of PRV XJ5 on PK15 cells. In addition, we also found that Panax notoginseng polysaccharides blocked the adsorption and replication of PRV to PK15 cells in a dose-dependent manner. These results show that Panax notoginseng polysaccharides play an antiviral effect mainly by inhibiting virus adsorption and replication in vitro. Therefore, Panax notoginseng polysaccharides may be a potential anti-PRV agent.


Assuntos
Herpesvirus Suídeo 1/fisiologia , Fatores Imunológicos/farmacologia , Panax notoginseng/química , Polissacarídeos/farmacologia , Pseudorraiva/metabolismo , Doenças dos Suínos/metabolismo , Ligação Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Fatores Imunológicos/química , Polissacarídeos/química , Pseudorraiva/tratamento farmacológico , Pseudorraiva/patologia , Suínos , Doenças dos Suínos/patologia , Doenças dos Suínos/virologia
6.
PLoS Pathog ; 18(2): e1010343, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35176124

RESUMO

The continuous emergence of severe acute respiratory coronavirus 2 (SARS-CoV-2) variants and the increasing number of breakthrough infection cases among vaccinated people support the urgent need for research and development of antiviral drugs. Viral entry is an intriguing target for antiviral drug development. We found that diltiazem, a blocker of the L-type calcium channel Cav1.2 pore-forming subunit (Cav1.2 α1c) and an FDA-approved drug, inhibits the binding and internalization of SARS-CoV-2, and decreases SARS-CoV-2 infection in cells and mouse lung. Cav1.2 α1c interacts with SARS-CoV-2 spike protein and ACE2, and affects the attachment and internalization of SARS-CoV-2. Our finding suggests that diltiazem has potential as a drug against SARS-CoV-2 infection and that Cav1.2 α1c is a promising target for antiviral drug development for COVID-19.


Assuntos
Tratamento Farmacológico da COVID-19 , COVID-19 , Diltiazem/farmacologia , Pulmão/efeitos dos fármacos , SARS-CoV-2/efeitos dos fármacos , Células A549 , Animais , COVID-19/patologia , COVID-19/virologia , Células Cultivadas , Chlorocebus aethiops , Diltiazem/uso terapêutico , Modelos Animais de Doenças , Feminino , Células HEK293 , Células HeLa , Humanos , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , SARS-CoV-2/fisiologia , Células Vero , Ligação Viral/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos
7.
Int J Mol Sci ; 23(3)2022 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-35163532

RESUMO

Since the beginning of the HIV epidemic, lasting more than 30 years, the main goal of scientists was to develop effective methods for the prevention and treatment of HIV infection. Modern medicines have reduced the death rate from AIDS by 80%. However, they still have side effects and are very expensive, dictating the need to search for new drugs. Earlier, it was shown that phospholipases A2 (PLA2s) from bee and snake venoms block HIV replication, the effect being independent on catalytic PLA2 activity. However, the antiviral activity of human PLA2s against Lentiviruses depended on catalytic function and was mediated through the destruction of the viral membrane. To clarify the role of phospholipolytic activity in antiviral effects, we analyzed the anti-HIV activity of several snake PLA2s and found that the mechanisms of their antiviral activity were similar to that of mammalian PLA2. Our results indicate that snake PLA2s are capable of inhibiting syncytium formation between chronically HIV-infected cells and healthy CD4-positive cells and block HIV binding to cells. However, only dimeric PLA2s had pronounced virucidal and anti-HIV activity, which depended on their catalytic activity. The ability of snake PLA2s to inactivate the virus may provide an additional barrier to HIV infection. Thus, snake PLA2s might be considered as candidates for lead molecules in anti-HIV drug development.


Assuntos
Fármacos Anti-HIV/farmacologia , Linfócitos T CD4-Positivos/citologia , Células Gigantes/citologia , HIV-1/fisiologia , Fosfolipases A2/farmacologia , Venenos de Serpentes/enzimologia , Serpentes/metabolismo , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Células Cultivadas , Células Gigantes/efeitos dos fármacos , Células Gigantes/virologia , HIV-1/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Proteínas de Répteis/farmacologia , Serpentes/classificação , Ativação Viral/efeitos dos fármacos , Ligação Viral/efeitos dos fármacos
8.
Viruses ; 14(2)2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-35216006

RESUMO

Coronaviruses (CoVs) are common among humans and many animals, causing respiratory or gastrointestinal diseases. Currently, only a few antiviral drugs against CoVs are available. Especially for SARS-CoV-2, new compounds for treatment of COVID-19 are urgently needed. In this study, we characterize the antiviral effects of two high-sulfated glycosaminoglycan (GAG) derivatives against SARS-CoV-2 and bovine coronaviruses (BCoV), which are both members of the Betacoronavirus genus. The investigated compounds are based on hyaluronan (HA) and chondroitin sulfate (CS) and exhibit a strong inhibitory effect against both CoVs. Yield assays were performed using BCoV-infected PT cells in the presence and absence of the compounds. While the high-sulfated HA (sHA3) led to an inhibition of viral growth early after infection, high-sulfated CS (sCS3) had a slightly smaller effect. Time of addition assays, where sHA3 and sCS3 were added to PT cells before, during or after infection, demonstrated an inhibitory effect during all phases of infection, whereas sHA3 showed a stronger effect even after virus absorbance. Furthermore, attachment analyses with prechilled PT cells revealed that virus attachment is not blocked. In addition, sHA3 and sCS3 inactivated BCoV by stable binding. Analysis by quantitative real-time RT PCR underlines the high potency of the inhibitors against BCoV, as well as B.1-lineage, Alpha and Beta SARS-CoV-2 viruses. Taken together, these results demonstrated that the two high-sulfated GAG derivatives exhibit low cytotoxicity and represent promising candidates for an anti-CoV therapy.


Assuntos
Antivirais/farmacologia , Infecções por Coronavirus/veterinária , Coronavirus Bovino/efeitos dos fármacos , Glicosaminoglicanos/farmacologia , SARS-CoV-2/efeitos dos fármacos , Animais , Bovinos , Linhagem Celular , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacologia , Infecções por Coronavirus/tratamento farmacológico , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Humanos , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Sulfatos/química , Sulfatos/farmacologia , Ligação Viral/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
9.
Biomed Res Int ; 2022: 1558860, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35039793

RESUMO

Increasing outbreaks of new pathogenic viruses have promoted the exploration of novel alternatives to time-consuming vaccines. Thus, it is necessary to develop a universal approach to halt the spread of new and unknown viruses as they are discovered. One such promising approach is to target lipid membranes, which are common to all viruses and bacteria. The ongoing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has reaffirmed the importance of interactions between the virus envelope and the host cell plasma membrane as a critical mechanism of infection. Metadichol®, a nanolipid emulsion of long-chain alcohols, has been demonstrated as a strong candidate that inhibits the proliferation of SARS-CoV-2. Naturally derived substances, such as long-chain saturated lipid alcohols, reduce viral infectivity, including that of coronaviruses (such as SARS-CoV-2) by modifying their lipid-dependent attachment mechanism to human host cells. The receptor ACE2 mediates the entry of SARS-CoV-2 into the host cells, whereas the serine protease TMPRSS2 primes the viral S protein. In this study, Metadichol® was found to be 270 times more potent an inhibitor of TMPRSS2 (EC50 = 96 ng/mL) than camostat mesylate (EC50 = 26000 ng/mL). Additionally, it inhibits ACE with an EC50 of 71 ng/mL, but it is a very weak inhibitor of ACE2 at an EC50 of 31 µg/mL. Furthermore, the live viral assay performed in Caco-2 cells revealed that Metadichol® inhibits SARS-CoV-2 replication at an EC90 of 0.16 µg/mL. Moreover, Metadichol® had an EC90 of 0.00037 µM, making it 2081 and 3371 times more potent than remdesivir (EC50 = 0.77 µM) and chloroquine (EC50 = 1.14 µM), respectively.


Assuntos
Álcoois Graxos/farmacologia , Sistemas de Liberação de Fármacos por Nanopartículas/farmacologia , SARS-CoV-2/efeitos dos fármacos , Vírus/efeitos dos fármacos , Animais , Antivirais/farmacologia , Linhagem Celular , Chlorocebus aethiops , Ésteres/farmacologia , Guanidinas/farmacologia , Humanos , Metabolismo dos Lipídeos/fisiologia , Lipídeos/química , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidade , Serina Endopeptidases/efeitos dos fármacos , Serina Endopeptidases/metabolismo , Serina Proteases/metabolismo , Inibidores de Serina Proteinase/farmacologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Células Vero , Ligação Viral/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
10.
SLAS Discov ; 27(1): 8-19, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35058179

RESUMO

The severe acute respiratory syndrome coronavirus 2 responsible for COVID-19 remains a persistent threat to mankind, especially for the immunocompromised and elderly for which the vaccine may have limited effectiveness. Entry of SARS-CoV-2 requires a high affinity interaction of the viral spike protein with the cellular receptor angiotensin-converting enzyme 2. Novel mutations on the spike protein correlate with the high transmissibility of new variants of SARS-CoV-2, highlighting the need for small molecule inhibitors of virus entry into target cells. We report the identification of such inhibitors through a robust high-throughput screen testing 15,000 small molecules from unique libraries. Several leads were validated in a suite of mechanistic assays, including whole cell SARS-CoV-2 infectivity assays. The main lead compound, calpeptin, was further characterized using SARS-CoV-1 and the novel SARS-CoV-2 variant entry assays, SARS-CoV-2 protease assays and molecular docking. This study reveals calpeptin as a potent and specific inhibitor of SARS-CoV-2 and some variants.


Assuntos
Antivirais/farmacologia , Tratamento Farmacológico da COVID-19 , Inibidores de Cisteína Proteinase/farmacologia , Dipeptídeos/farmacologia , Ligação Viral/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Enzima de Conversão de Angiotensina 2/metabolismo , Animais , Catepsina L/antagonistas & inibidores , Linhagem Celular , Chlorocebus aethiops , Avaliação Pré-Clínica de Medicamentos , Reposicionamento de Medicamentos , Células HEK293 , Humanos , Simulação de Acoplamento Molecular , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/crescimento & desenvolvimento , Serina Endopeptidases/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Células Vero
11.
J Mol Biol ; 434(2): 167395, 2022 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-34896364

RESUMO

GSK3732394 is a multi-specific biologic inhibitor of HIV entry currently under clinical evaluation. A key component of this molecule is an adnectin (6940_B01) that binds to CD4 and inhibits downstream actions of gp160. Studies were performed to determine the binding site of the adnectin on CD4 and to understand the mechanism of inhibition. Using hydrogen-deuterium exchange with mass spectrometry (HDX), CD4 peptides showed differential rates of deuteration (either enhanced or slowed) in the presence of the adnectin that mapped predominantly to the interface of domains 2 and 3 (D2-D3). In addition, an X-ray crystal structure of an ibalizumab Fab/CD4(D1-D4)/adnectin complex revealed an extensive interface between the adnectin and residues on CD4 domains D2-D4 that stabilize a novel T-shaped CD4 conformation. A cryo-EM map of the gp140/CD4/GSK3732394 complex clearly shows the bent conformation for CD4 while bound to gp140. Mutagenic analyses on CD4 confirmed that amino acid F202 forms a key interaction with the adnectin. In addition, amino acid L151 was shown to be a critical indirect determinant of the specificity for binding to the human CD4 protein over related primate CD4 molecules, as it appears to modulate CD4's flexibility to adopt the adnectin-bound conformation. The significant conformational change of CD4 upon adnectin binding brings the D1 domain of CD4 in proximity to the host cell membrane surface, thereby re-orienting the gp120 binding site in a direction that is inaccessible to incoming virus due to a steric clash between gp160 trimers on the virus surface and the target cell membrane.


Assuntos
Fármacos Anti-HIV/farmacologia , Antígenos CD4/química , Antígenos CD4/metabolismo , HIV-1/metabolismo , Ligação Viral/efeitos dos fármacos , Animais , Anticorpos Monoclonais , Sítios de Ligação , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Internalização do Vírus/efeitos dos fármacos
12.
Biomed Pharmacother ; 146: 112581, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34965505

RESUMO

Epimedium koreanum Nakai (EKN) is a popular plant in Korean and Chinese medicine for treating a variety of ailments. The aqueous extract of EKN has a significant inhibitory impact on influenza A virus (IAV) infection by directly blocking viral attachment and having a virucidal effect, according to this study. Using fluorescent microscopy and fluorescence-activated cell sorting (FACS) with a green fluorescent protein (GFP)-tagged Influenza A/PR/8/34 virus, we examined the effect of EKN on viral infection. By viral infection, EKN strongly suppresses GFP expression, and at a dosage of 100 µg/mL, EKN decreased GFP expression by up to 90% of the untreated infected control. Immunofluorescence and Western blot analyses against influenza viral proteins revealed that EKN decreased influenza viral protein expression in a dose-dependent manner. EKN inhibited the H1N1 influenza virus's hemagglutinin (HA) and neuraminidase (NA), preventing viral attachment to cells. Furthermore, EKN had a virucidal impact and inhibited the cytopathic effects of H1N1, H3N2 and influenza B virus infection. Finally, our findings show that EKN has the potential to be developed as a natural viral inhibitor against influenza virus infection.


Assuntos
Alphainfluenzavirus/efeitos dos fármacos , Antivirais/farmacologia , Epimedium , Extratos Vegetais/farmacologia , Animais , Hemaglutininas/efeitos dos fármacos , Humanos , Camundongos , Neuraminidase/efeitos dos fármacos , Proteínas Virais/efeitos dos fármacos , Ligação Viral/efeitos dos fármacos
13.
Eur Rev Med Pharmacol Sci ; 25(23): 7565-7584, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34919258

RESUMO

OBJECTIVE: With the recent direction in drug repurposing, many approved drugs have been evaluated to assess their effect on the coronavirus or SARS-CoV-2 infection (COVID-19). Driving this path, chloroquine (CQ) has been used in the treatment of malaria and hydroxychloroquine (HCQ) in immunomodulatory and anti-thrombotic action, playing a leading role in initial management of the viral infection. MATERIALS AND METHODS: Literature search was done using Google Scholar, PubMed and Scopus database using keywords "chloroquine" "SARS-CoV-2" "COVID-19" "mechanism of action" and articles of interest were selected providing evidence of the possible role of CQ in viral infection. RESULTS: In a bid to understand how and if CQ and HCQ would exert their anti-viral property, mechanistic exegesis was done to review various proposed mechanisms of action. This revealed the inhibition of viral attachment and entry, inhibition of enveloped glycoprotein, inhibition of the development and proliferation of new viral particles as the way they perform their action. There is an interplay between iron metabolism and homeostasis with COVID-19 infection and viral reproduction. CONCLUSIONS: This study aims to show the functional role of CQ and HCQ, as well as to provide possible mechanistic insight on the role of iron on viral infection, iron starvation and its downstream cellular pathways involving hepcidin and proinflammatory cytokines. The overall aim of providing possible mode of action of CQ and HCQ in the management of COVID-19 infection is exhibited via its anti-viral, anti-inflammatory and anti-thrombotic activities.


Assuntos
Tratamento Farmacológico da COVID-19 , Cloroquina/farmacologia , Hidroxicloroquina/farmacologia , Ferro/metabolismo , COVID-19/metabolismo , Cloroquina/uso terapêutico , Reposicionamento de Medicamentos , Homeostase , Humanos , Hidroxicloroquina/uso terapêutico , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/fisiologia , Ligação Viral/efeitos dos fármacos
14.
Viruses ; 13(12)2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34960796

RESUMO

Porcine epidemic diarrhea virus (PEDV), an enteric coronavirus, causes neonatal pig acute gastrointestinal infection with a characterization of severe diarrhea, vomiting, high morbidity, and high mortality, resulting in tremendous damages to the swine industry. Neither specific antiviral drugs nor effective vaccines are available, posing a high priority to screen antiviral drugs. The aim of this study is to investigate anti-PEDV effects of carbazole alkaloid derivatives. Eighteen carbazole derivatives (No.1 to No.18) were synthesized, and No.5, No.7, and No.18 were identified to markedly reduce the replication of enhanced green fluorescent protein (EGFP) inserted-PEDV, and the mRNA level of PEDV N. Flow cytometry assay, coupled with CCK8 assay, confirmed No.7 and No.18 carbazole derivatives displayed high inhibition effects with low cell toxicity. Furthermore, time course analysis indicated No.7 and No.18 carbazole derivatives exerted inhibition at the early stage of the viral life cycle. Collectively, the analysis underlines the benefit of carbazole derivatives as potential inhibitors of PEDV, and provides candidates for the development of novel therapeutic agents.


Assuntos
Antivirais/farmacologia , Carbazóis/farmacologia , Vírus da Diarreia Epidêmica Suína/efeitos dos fármacos , Animais , Antivirais/química , Carbazóis/química , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Estrutura Molecular , Células Vero , Ligação Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
15.
Int J Mol Sci ; 22(24)2021 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-34948390

RESUMO

Since the start of the COVID-19 outbreak, pharmaceutical companies and research groups have focused on the development of vaccines and antiviral drugs against SARS-CoV-2. Here, we apply a drug repurposing strategy to identify drug candidates that are able to block the entrance of the virus into human cells. By combining virtual screening with in vitro pseudovirus assays and antiviral assays in Human Lung Tissue (HLT) cells, we identify entrectinib as a potential antiviral drug.


Assuntos
Benzamidas/farmacologia , Tratamento Farmacológico da COVID-19 , Indazóis/farmacologia , SARS-CoV-2/efeitos dos fármacos , Animais , Antivirais/farmacologia , Benzamidas/metabolismo , COVID-19/metabolismo , Linhagem Celular , Chlorocebus aethiops , Avaliação Pré-Clínica de Medicamentos , Reposicionamento de Medicamentos/métodos , Humanos , Indazóis/metabolismo , Pulmão/patologia , Pulmão/virologia , Simulação de Acoplamento Molecular , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidade , Células Vero , Ligação Viral/efeitos dos fármacos
16.
Viruses ; 13(11)2021 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-34835112

RESUMO

Anti-viral small molecules are currently lacking for treating coronavirus infection. The long development timescales for such drugs are a major problem, but could be shortened by repurposing existing drugs. We therefore screened a small library of FDA-approved compounds for potential severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) antivirals using a pseudovirus system that allows a sensitive read-out of infectivity. A group of structurally-related compounds, showing moderate inhibitory activity with IC50 values in the 2-5 µM range, were identified. Further studies demonstrated that these "kite-shaped" molecules were surprisingly specific for SARS-CoV-1 and SARS-CoV-2 and that they acted early in the entry steps of the viral infectious cycle, but did not affect virus attachment to the cells. Moreover, the compounds were able to prevent infection in both kidney- and lung-derived human cell lines. The structural homology of the hits allowed the production of a well-defined pharmacophore that was found to be highly accurate in predicting the anti-viral activity of the compounds in the screen. We discuss the prospects of repurposing these existing drugs for treating current and future coronavirus outbreaks.


Assuntos
Antivirais/farmacologia , Tratamento Farmacológico da COVID-19 , COVID-19/virologia , Vírus da Leucemia Murina/efeitos dos fármacos , SARS-CoV-2 , Internalização do Vírus/efeitos dos fármacos , Animais , Linhagem Celular , Chlorocebus aethiops , Descoberta de Drogas/métodos , Reposicionamento de Medicamentos , Sinergismo Farmacológico , Humanos , Vírus da Leucemia Murina/metabolismo , Camundongos , Simulação de Acoplamento Molecular , Glicoproteína da Espícula de Coronavírus/metabolismo , Células Vero , Ligação Viral/efeitos dos fármacos
17.
Antiviral Res ; 195: 105188, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34648875

RESUMO

Zika virus (ZIKV) has been the cause of some epidemics since 2007. The correlations of microcephaly and Guillain-Barré syndrome with ZIKV have been noticed. Unfortunately, researchers have yet to develop an effective vaccine or drug approved for ZIKV infection. Anidulafungin is a member of echinocandins that is used to treat candida infections. This study assessed the antiviral capability of anidulafungin against ZIKV. Anidulafungin was shown to significantly decrease viral RNA levels, protein expression levels, viral yields, and the rate of infection. In time of addition assays, anidulafungin exhibited inhibitory activities in the early stages of ZIKV infection. In binding and entry assays, administering anidulafungin did not lead to a corresponding decrease in quantity of viral RNA, but a significant decrease in ZIKV infectivity was observed in virucidal assays. This indicated that anidulafungin interferes directly with virions. T-1105 is a viral polymerase inhibitor, which functions in the late stage of ZIKV infection. When anidulafungin was administered in combination with T-1105, an obvious synergistic effect was observed, resulting in a combination index (CI) value of 0.85 ± 0.13. Finally, we evaluated the effects of echinocandins in terms of half-maximal inhibitory concentration (IC50), calculation of cytotoxicity concentration 50% (CC50), selectivity index (SI), and Patchdock score. Among the tests, anidulafungin bears the lowest IC50 and highest Patchdock score. Although anidulafungin is classified as a pregnancy category C agent; however, combination therapy of anidulafungin with a viral RNA replication inhibitor could expand treatment options for ZIKV infection.


Assuntos
Anidulafungina/farmacologia , Antivirais/síntese química , Pirazinas/farmacologia , RNA Viral/efeitos dos fármacos , Zika virus/efeitos dos fármacos , Animais , Antivirais/farmacologia , Chlorocebus aethiops , Sinergismo Farmacológico , Humanos , Concentração Inibidora 50 , Células Vero , Vírion/efeitos dos fármacos , Ligação Viral/efeitos dos fármacos , Infecção por Zika virus/tratamento farmacológico , Infecção por Zika virus/virologia
18.
J Virol ; 95(24): e0093821, 2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34613794

RESUMO

Sodium taurocholate cotransporting polypeptide (NTCP) is a receptor that is essential for hepatitis B virus (HBV) entry into the host cell. A number of HBV entry inhibitors targeting NTCP have been reported to date; these inhibitors have facilitated a mechanistic analysis of the viral entry process. However, the mechanism of HBV internalization into host cells after interaction of virus with NTCP remains largely unknown. Recently, we reported that troglitazone, a thiazolidinedione derivative, specifically inhibits both HBV internalization and NTCP oligomerization, resulting in inhibition of HBV infection. Here, using troglitazone as a chemical probe to investigate entry process, the contribution of NTCP oligomerization to HBV internalization was evaluated. Using surface plasmon resonance and transporter kinetics, we found that troglitazone directly interacts with NTCP and noncompetitively interferes with NTCP-mediated bile acid uptake, suggesting that troglitazone allosterically binds to NTCP, rather than to the bile acid-binding pocket. Additionally, alanine scanning mutagenesis showed that a mutation at phenylalanine 274 of NTCP (F274A) caused a loss of HBV susceptibility and disrupted both the oligomerization of NTCP and HBV internalization without affecting viral attachment to the cell surface. An inhibitor of the interaction between NTCP and epidermal growth factor receptor (EGFR), another host cofactor essential for HBV internalization, impeded NTCP oligomerization. Meanwhile, coimmunoprecipitation analysis revealed that neither troglitazone nor the F274A mutation in NTCP affects the NTCP-EGFR interaction. These findings suggest that NTCP oligomerization is initiated downstream of the NTCP-EGFR interaction and then triggers HBV internalization. This study provides significant insight into the HBV entry mechanisms. IMPORTANCE Hepatitis B virus (HBV) infection is mediated by a specific interaction with sodium taurocholate cotransporting polypeptide (NTCP), a viral entry receptor. Although the virus-receptor interactions are believed to trigger viral internalization into host cells, the exact molecular mechanisms of HBV internalization are not understood. In this study, we revealed the mode of action whereby troglitazone, a specific inhibitor of HBV internalization, impedes NTCP oligomerization and identified NTCP phenylalanine 274 as a residue essential for this oligomerization. We further analyzed the association between NTCP oligomerization and HBV internalization, a process that is mediated by epidermal growth factor receptor (EGFR), another essential host cofactor for HBV internalization. Our study provides critical information on the mechanism of HBV entry and suggests that oligomerization of the viral receptor serves as an attractive target for drug discovery.


Assuntos
Vírus da Hepatite B/fisiologia , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Multimerização Proteica , Receptores Virais/metabolismo , Simportadores/metabolismo , Internalização do Vírus/efeitos dos fármacos , Transporte Biológico , Receptores ErbB/genética , Receptores ErbB/metabolismo , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/virologia , Humanos , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Simportadores/genética , Troglitazona/farmacologia , Ligação Viral/efeitos dos fármacos
19.
Int J Biol Sci ; 17(14): 3786-3794, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34671199

RESUMO

COVID-19, caused by a novel coronavirus, SARS-CoV-2, poses a serious global threat. It was first reported in 2019 in China and has now dramatically spread across the world. It is crucial to develop therapeutics to mitigate severe disease and viral spread. The receptor-binding domains (RBDs) in the spike protein of SARS-CoV and MERS-CoV have shown anti-viral activity in previous reports suggesting that this domain has high potential for development as therapeutics. To evaluate the potential antiviral activity of recombinant SARS-CoV-2 RBD proteins, we determined the RBD residues of SARS-CoV-2 using a homology search with RBD of SARS-CoV. For efficient expression and purification, the signal peptide of spike protein was identified and used to generate constructs expressing recombinant RBD proteins. Highly purified RBD protein fused with the Fc domain of human IgG showed potent anti-viral efficacy, which was better than that of a protein fused with a histidine tag. Intranasally pre-administrated RBD protein also inhibited the attachment of SARS-COV-2 to mouse lungs. These findings indicate that RBD protein could be used for the prevention and treatment of SARS-CoV-2 infection.


Assuntos
Tratamento Farmacológico da COVID-19 , SARS-CoV-2/efeitos dos fármacos , Glicoproteína da Espícula de Coronavírus/uso terapêutico , Ligação Viral/efeitos dos fármacos , Administração Intranasal , Sequência de Aminoácidos , Animais , Sítios de Ligação , Chlorocebus aethiops , Feminino , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Domínios Proteicos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Glicoproteína da Espícula de Coronavírus/biossíntese , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/farmacologia , Células Vero
20.
Antiviral Res ; 195: 105192, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34687821

RESUMO

Due to the lack of relevant therapies for infectious haematopoietic necrosis virus (IHNV) infection, the viral outbreak invariably causes serious economic losses in salmonid species. In this study, we evaluated the anti-IHNV effects of 7-(6-benzimidazole) coumarin (C10) and 4-phenyl-2-thioxo-1,2,3,4-tetrahydro-5H-chromeno[4,3-d]pyrimidin-5-one (S5) in vitro and in vivo. The results revealed that C10 at 12.5 mg/L and S5 at 25 mg/L significantly inhibited IHNV replication in epithelioma papulosum cyprini (EPC) cells with a maximum inhibitory rate >90%, showing that IHNV-induced cytopathic effect (CPE) was alleviated by C10 and S5. There are two complementary effects on antiviral mechanism: 1. C10 completely inhibited IHNV infectivity when the virus was preincubated with C10 at 12.5 mg/L, determining that C10 may have a negative impact on IHNV binding to the cell; 2. C10 also up-regulated the gene expression of extracellular proto type galectin-1 (Gal1-L2) and a chimera galectin-3 (Gal3-L1) of EPC cells to inhibit IHNV adhesion. For the in vivo study, injection and immersion of the coumarins enhanced the survival rate of rainbow trout (Oncorhynchus mykiss) juveniles by 25% (at least) at 12 dpi. IHNV loads in the kidney and spleen were also obviously decreased at 96 h, and thus we considered that they had a delaying effect on IHNV replication in vivo. Meanwhile, C10 with a high stability in aquacultural water in immersion suppressed IHNV horizontal transmission by decreasing the viral loads in recipient fish. Overall, our data suggest that there is a positive effect of C10 and S5 against IHNV infection in aquaculture, and C10 had the potential to be a broad-spectrum antiviral against fish rhabdoviruses.


Assuntos
Antivirais/farmacologia , Cumarínicos/farmacologia , Vírus da Necrose Hematopoética Infecciosa/efeitos dos fármacos , Ligação Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Animais , Aquicultura , Linhagem Celular , Doenças dos Peixes/tratamento farmacológico , Doenças dos Peixes/mortalidade , Doenças dos Peixes/patologia , Oncorhynchus mykiss/virologia , Infecções por Rhabdoviridae/tratamento farmacológico , Infecções por Rhabdoviridae/mortalidade , Infecções por Rhabdoviridae/patologia , Taxa de Sobrevida , Carga Viral/efeitos dos fármacos , Proteínas Virais/genética , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...