Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(43): 21514-21520, 2019 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-31591233

RESUMO

The bat-borne paramyxovirus, Sosuga virus (SosV), is one of many paramyxoviruses recently identified and classified within the newly established genus Pararubulavirus, family Paramyxoviridae The envelope surface of SosV presents a receptor-binding protein (RBP), SosV-RBP, which facilitates host-cell attachment and entry. Unlike closely related hemagglutinin neuraminidase RBPs from other genera of the Paramyxoviridae, SosV-RBP and other pararubulavirus RBPs lack many of the stringently conserved residues required for sialic acid recognition and hydrolysis. We determined the crystal structure of the globular head region of SosV-RBP, revealing that while the glycoprotein presents a classical paramyxoviral six-bladed ß-propeller fold and structurally classifies in close proximity to paramyxoviral RBPs with hemagglutinin-neuraminidase (HN) functionality, it presents a receptor-binding face incongruent with sialic acid recognition. Hemadsorption and neuraminidase activity analysis confirms the limited capacity of SosV-RBP to interact with sialic acid in vitro and indicates that SosV-RBP undergoes a nonclassical route of host-cell entry. The close overall structural conservation of SosV-RBP with other classical HN RBPs supports a model by which pararubulaviruses only recently diverged from sialic acid binding functionality.


Assuntos
Proteína HN/química , Infecções por Paramyxoviridae/virologia , Paramyxoviridae/fisiologia , Proteínas Virais/química , Internalização do Vírus , Proteína HN/genética , Proteína HN/metabolismo , Humanos , Ácido N-Acetilneuramínico/metabolismo , Paramyxoviridae/química , Paramyxoviridae/genética , Infecções por Paramyxoviridae/metabolismo , Receptores Virais/genética , Receptores Virais/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Ligação Viral
2.
J Virol ; 95(2)2020 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-33087464

RESUMO

Engagement of cell surface receptors by viruses is a critical determinant of viral tropism and disease. The reovirus attachment protein σ1 binds sialylated glycans and proteinaceous receptors to mediate infection, but the specific requirements for different cell types are not entirely known. To identify host factors required for reovirus-induced cell death, we conducted a CRISPR-knockout screen targeting over 20,000 genes in murine microglial BV2 cells. Candidate genes required for reovirus to cause cell death were highly enriched for sialic acid synthesis and transport. Two of the top candidates identified, CMP N-acetylneuraminic acid synthetase (Cmas) and solute carrier family 35 member A1 (Slc35a1), promote sialic acid expression on the cell surface. Two reovirus strains that differ in the capacity to bind sialic acid, T3SA+ and T3SA-, were used to evaluate Cmas and Slc35a1 as potential host genes required for reovirus infection. Following CRISPR-Cas9 disruption of either gene, cell surface expression of sialic acid was diminished. These results correlated with decreased binding of strain T3SA+, which is capable of engaging sialic acid. Disruption of either gene did not alter the low-level binding of T3SA-, which does not engage sialic acid. Furthermore, infectivity of T3SA+ was diminished to levels similar to those of T3SA- in cells lacking Cmas and Slc35a1 by CRISPR ablation. However, exogenous expression of Cmas and Slc35a1 into the respective null cells restored sialic acid expression and T3SA+ binding and infectivity. These results demonstrate that Cmas and Slc35a1, which mediate cell surface expression of sialic acid, are required in murine microglial cells for efficient reovirus binding and infection.IMPORTANCE Attachment factors and receptors are important determinants of dissemination and tropism during reovirus-induced disease. In a CRISPR cell survival screen, we discovered two genes, Cmas and Slc35a1, which encode proteins required for sialic acid expression on the cell surface and mediate reovirus infection of microglial cells. This work elucidates host genes that render microglial cells susceptible to reovirus infection and expands current understanding of the receptors on microglial cells that are engaged by reovirus. Such knowledge may lead to new strategies to selectively target microglial cells for oncolytic applications.


Assuntos
N-Acilneuraminato Citidililtransferase/metabolismo , Proteínas de Transporte de Nucleotídeos/metabolismo , Infecções por Reoviridae/virologia , Reoviridae/fisiologia , Animais , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Sobrevivência Celular , Camundongos , Ácido N-Acetilneuramínico/metabolismo , N-Acilneuraminato Citidililtransferase/genética , Proteínas de Transporte de Nucleotídeos/genética , Receptores Virais/metabolismo , Reoviridae/genética , Reoviridae/metabolismo , Infecções por Reoviridae/metabolismo , Ligação Viral , Replicação Viral
3.
Virol J ; 18(1): 136, 2021 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-34217298

RESUMO

Flaviviruses are enveloped viruses that infect multiple hosts. Envelope proteins are the outermost proteins in the structure of flaviviruses and mediate viral infection. Studies indicate that flaviviruses mainly use envelope proteins to bind to cell attachment receptors and endocytic receptors for the entry step. Here, we present current findings regarding key envelope protein amino acids that participate in the flavivirus early infection process. Among these sites, most are located in special positions of the protein structure, such as the α-helix in the stem region and the hinge region between domains I and II, motifs that potentially affect the interaction between different domains. Some of these sites are located in positions involved in conformational changes in envelope proteins. In summary, we summarize and discuss the key envelope protein residues that affect the entry process of flaviviruses, including the process of their discovery and the mechanisms that affect early infection.


Assuntos
Infecções por Flavivirus , Flavivirus , Proteínas do Envelope Viral , Internalização do Vírus , Aminoácidos , Humanos
4.
J Virol ; 93(12)2019 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-30918079

RESUMO

In previous research, a 27.8-kDa protein in flounder Paralichthys olivaceus gill (FG) cells was identified as a putative cellular receptor (27.8R), which mediated lymphocystis disease virus (LCDV) infection via interaction with a 32-kDa viral attachment protein (VAP) of LCDV, and monoclonal antibodies (MAbs) against 27.8R and 32-kDa VAP were developed. In this study, the 27.8R was identified as voltage-dependent anion channel protein 2 (VDAC2) and receptor of activated protein C kinase 1 (RACK1) of flounder. Recombinant VDAC2 (rVDAC2) and RACK1 (rRACK1) were obtained by prokaryotic expression, and rabbit anti-VDAC2/RACK1 polyclonal antibodies were prepared. The rVDAC2, rRACK1, and 27.8-kDa proteins in FG cells were recognized by anti-27.8R MAbs and anti-VDAC2/RACK1 polyclonal antibodies simultaneously. Preincubation of FG cells with anti-VDAC2/RACK1 polyclonal antibodies significantly decreased the percentages of LCDV-infected cells and LCDV copy numbers, blocked virus infection, and delayed the development of cytopathic effect. The mRNA expressions of VDAC2 and RACK1 in FG cells were upregulated to maximum levels 12 h and 48 h after LCDV infection, respectively. VDAC2/RACK1 knockdown through short interfering RNA (siRNA) significantly reduced VDAC2/RACK1 expression and LCDV copy numbers in FG cells compared with negative controls, while VDAC2/RACK1 expression on LCDV-nonpermissive epithelial papillosum cells (EPCs) conferred susceptibility to LCDV infection, indicating the VDAC2 and RACK1 were sufficient to allow LCDV entry and infection. All these results collectively showed that VDAC2 and RACK1 function as receptors for LCDV entry and infection.IMPORTANCE Lymphocystis disease virus (LCDV) is the causative agent of lymphocystis disease in fish, which has caused huge economic losses to the aquaculture industry worldwide, but the molecular mechanism underlying the LCDV-host interaction remains unclear. Here, the 27.8-kDa putative cellular receptor for LCDV was identified as voltage-dependent anion channel protein 2 (VDAC2) and receptor of activated protein C kinase 1 (RACK1), and our results revealed that VDAC2 and RACK1 expression was sufficient to allow LCDV entry and that they are functional receptors that initiate LCDV infection for the first time, which leads to a better understanding of the molecular mechanism underlying LCDV infection and virus-host interactions.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Iridoviridae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Canal de Ânion 2 Dependente de Voltagem/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Anticorpos Monoclonais/imunologia , Infecções por Vírus de DNA/virologia , Doenças dos Peixes/virologia , Linguado , Proteínas de Ligação ao GTP/fisiologia , Brânquias/metabolismo , Iridoviridae/patogenicidade , Receptores Virais/metabolismo , Receptores Virais/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia , Proteínas Virais/genética , Replicação Viral/fisiologia , Canal de Ânion 2 Dependente de Voltagem/fisiologia
5.
Appl Environ Microbiol ; 86(3)2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31704685

RESUMO

Viruses influence microbial community structure and biogeochemical cycles in marine environments. Viral attachment to nonhost surfaces could influence host viral infection rates; however, the prevalence of such viral attachment is not investigated quantitatively. We used coastal seawater viral assemblages and, as models, marine vibriophage (SIO-2) and enterobacteriophages (T2 and T4) to investigate their attachment to probable nonhost marine bacteria. We also studied viral attachment to colloids and other abiotic surfaces in seawater. Centrifugation experiments with bacterium-virus mixtures showed substantial viral loss in the supernatant presumably due to the viral attachment to bacteria. This attachment (0.04 to 24 viruses µm-2 [bacterial surface area]) varied with bacterium-virus combinations. Surprisingly, filtering seawater on 0.2-µm Anodisc or polycarbonate filters retained ∼12 to 84% of viruses presumably attached to ≥0.2-µm-sized particles and/or the filter surface. Enzymatic digestion followed by epifluorescence and atomic force microscopy suggested that 7 to 25% of the total viruses were attached via ß-glycosidic linkages. Furthermore, a substantial proportion (7 to 48%) of viruses became attached to model abiotic surfaces (polycarbonate, polypropylene, and glass), and this has significance for laboratory protocols as well as studies of virus ecology in particle-rich marine environments. Substantial attachment of viruses to nonhost surfaces could influence virus-driven biogeochemical cycles and microbial community structure.IMPORTANCE Viruses play important roles in altering microbial community structure and biogeochemical cycles in marine environments. Viral attachment to nonhost surfaces can influence host viral infection rates; however, the prevalence of viral attachment to nonhost surfaces and the ratio of attached viruses to total viruses are little known. We used coastal seawater viral assemblages and used marine vibriophage (SIO-2) and enterobacteriophages (T2 and T4) as models to investigate their attachment to abiotic and biotic surfaces in seawater. Viral attachment was observed on several surfaces, such as nonhost bacteria, polymers, filters, cover glasses, and tube surfaces. This study cautions against commonly used protocols that require viral incubation and seawater fractionation. More importantly, these results could influence virus-driven biogeochemical cycles and microbial community structure in the ocean.


Assuntos
Bacteriófagos/isolamento & purificação , Microbiota , Água do Mar/virologia , Ligação Viral , Coloides
6.
Proc Natl Acad Sci U S A ; 114(14): E2929-E2936, 2017 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-28320973

RESUMO

Orthomyxoviruses are an important family of RNA viruses, which include the various influenza viruses. Despite global efforts to eradicate orthomyxoviral pathogens, these infections remain pervasive. One such orthomyxovirus, infectious salmon anemia virus (ISAV), spreads easily throughout farmed and wild salmonids, constituting a significant economic burden. ISAV entry requires the interplay of the virion-attached hemagglutinin-esterase and fusion glycoproteins. Preventing infections will rely on improved understanding of ISAV entry. Here, we present the crystal structures of ISAV hemagglutinin-esterase unbound and complexed with receptor. Several distinctive features observed in ISAV HE are not seen in any other viral glycoprotein. The structures reveal a unique mode of receptor binding that is dependent on the oligomeric assembly of hemagglutinin-esterase. Importantly, ISAV hemagglutinin-esterase receptor engagement does not initiate conformational rearrangements, suggesting a distinct viral entry mechanism. This work improves our understanding of ISAV pathogenesis and expands our knowledge on the overall diversity of viral glycoprotein-mediated entry mechanisms. Finally, it provides an atomic-resolution model of the primary neutralizing antigen critical for vaccine development.


Assuntos
Hemaglutininas Virais/química , Hemaglutininas Virais/metabolismo , Isavirus/patogenicidade , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Animais , Sítios de Ligação , Cristalografia por Raios X , Hemaglutininas Virais/genética , Interações Hospedeiro-Patógeno , Conformação Proteica , Domínios Proteicos , Receptores Virais/química , Receptores Virais/metabolismo , Espalhamento a Baixo Ângulo , Proteínas Virais de Fusão/genética , Ligação Viral , Difração de Raios X
7.
J Biol Chem ; 293(30): 11709-11726, 2018 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-29887526

RESUMO

Coronavirus tropism is predominantly determined by the interaction between coronavirus spikes and the host receptors. In this regard, coronaviruses have evolved a complicated receptor-recognition system through their spike proteins. Spikes from highly related coronaviruses can recognize distinct receptors, whereas spikes of distant coronaviruses can employ the same cell-surface molecule for entry. Moreover, coronavirus spikes can recognize a broad range of cell-surface molecules in addition to the receptors and thereby can augment coronavirus attachment or entry. The receptor of Middle East respiratory syndrome coronavirus (MERS-CoV) is dipeptidyl peptidase 4 (DPP4). In this study, we identified membrane-associated 78-kDa glucose-regulated protein (GRP78) as an additional binding target of the MERS-CoV spike. Further analyses indicated that GRP78 could not independently render nonpermissive cells susceptible to MERS-CoV infection but could facilitate MERS-CoV entry into permissive cells by augmenting virus attachment. More importantly, by exploring potential interactions between GRP78 and spikes of other coronaviruses, we discovered that the highly conserved human GRP78 could interact with the spike protein of bat coronavirus HKU9 (bCoV-HKU9) and facilitate its attachment to the host cell surface. Taken together, our study has identified GRP78 as a host factor that can interact with the spike proteins of two Betacoronaviruses, the lineage C MERS-CoV and the lineage D bCoV-HKU9. The capacity of GRP78 to facilitate surface attachment of both a human coronavirus and a phylogenetically related bat coronavirus exemplifies the need for continuous surveillance of the evolution of animal coronaviruses to monitor their potential for human adaptations.


Assuntos
Betacoronavirus/fisiologia , Infecções por Coronavirus/metabolismo , Coronavirus/fisiologia , Proteínas de Choque Térmico/metabolismo , Coronavírus da Síndrome Respiratória do Oriente Médio/fisiologia , Ligação Viral , Animais , Linhagem Celular , Chlorocebus aethiops , Dipeptidil Peptidase 4/metabolismo , Chaperona BiP do Retículo Endoplasmático , Interações Hospedeiro-Patógeno , Humanos , Mapas de Interação de Proteínas , Receptores Virais/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Células Vero
8.
Glycobiology ; 29(5): 419-430, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30834446

RESUMO

Galectins, highly conserved ß-galactoside-binding lectins, have diverse regulatory roles in development and immune homeostasis and can mediate protective functions during microbial infection. In recent years, the role of galectins in viral infection has generated considerable interest. Studies on highly pathogenic viruses have provided invaluable insight into the participation of galectins in various stages of viral infection, including attachment and entry. Detailed mechanistic and structural aspects of these processes remain undetermined. To address some of these gaps in knowledge, we used Zebrafish as a model system to examine the role of galectins in infection by infectious hematopoietic necrosis virus (IHNV), a rhabdovirus that is responsible for significant losses in both farmed and wild salmonid fish. Like other rhabdoviruses, IHNV is characterized by an envelope consisting of trimers of a glycoprotein that display multiple N-linked oligosaccharides and play an integral role in viral infection by mediating the virus attachment and fusion. Zebrafish's proto-typical galectin Drgal1-L2 and the chimeric-type galectin Drgal3-L1 interact directly with the glycosylated envelope of IHNV, and significantly reduce viral attachment. In this study, we report the structure of the complex of Drgal1-L2 with N-acetyl-d-lactosamine at 2.0 Å resolution. To gain structural insight into the inhibitory effect of these galectins on IHNV attachment to the zebrafish epithelial cells, we modeled Drgal3-L1 based on human galectin-3, as well as, the ectodomain of the IHNV glycoprotein. These models suggest mechanisms for which the binding of these galectins to the IHNV glycoprotein hinders with different potencies the viral attachment required for infection.


Assuntos
Galectinas/química , Galectinas/metabolismo , Glicoproteínas/química , Vírus da Necrose Hematopoética Infecciosa/química , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/metabolismo , Sequência de Aminoácidos , Animais , Glicoproteínas/metabolismo , Vírus da Necrose Hematopoética Infecciosa/metabolismo , Modelos Moleculares , Alinhamento de Sequência , Peixe-Zebra
9.
Proc Natl Acad Sci U S A ; 112(17): E2156-65, 2015 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-25825759

RESUMO

The discovery of African henipaviruses (HNVs) related to pathogenic Hendra virus (HeV) and Nipah virus (NiV) from Southeast Asia and Australia presents an open-ended health risk. Cell receptor use by emerging African HNVs at the stage of host-cell entry is a key parameter when considering the potential for spillover and infection of human populations. The attachment glycoprotein from a Ghanaian bat isolate (GhV-G) exhibits <30% sequence identity with Asiatic NiV-G/HeV-G. Here, through functional and structural analysis of GhV-G, we show how this African HNV targets the same human cell-surface receptor (ephrinB2) as the Asiatic HNVs. We first characterized this virus-receptor interaction crystallographically. Compared with extant HNV-G-ephrinB2 structures, there was significant structural variation in the six-bladed ß-propeller scaffold of the GhV-G receptor-binding domain, but not the Greek key fold of the bound ephrinB2. Analysis revealed a surprisingly conserved mode of ephrinB2 interaction that reflects an ongoing evolutionary constraint among geographically distal and phylogenetically divergent HNVs to maintain the functionality of ephrinB2 recognition during virus-host entry. Interestingly, unlike NiV-G/HeV-G, we could not detect binding of GhV-G to ephrinB3. Comparative structure-function analysis further revealed several distinguishing features of HNV-G function: a secondary ephrinB2 interaction site that contributes to more efficient ephrinB2-mediated entry in NiV-G relative to GhV-G and cognate residues at the very C terminus of GhV-G (absent in Asiatic HNV-Gs) that are vital for efficient receptor-induced fusion, but not receptor binding per se. These data provide molecular-level details for evaluating the likelihood of African HNVs to spill over into human populations.


Assuntos
Efrina-B2 , Infecções por Henipavirus/metabolismo , Henipavirus , Proteínas Virais , Internalização do Vírus , Efrina-B2/química , Efrina-B2/genética , Efrina-B2/metabolismo , Efrina-B3/química , Efrina-B3/genética , Efrina-B3/metabolismo , Células HEK293 , Henipavirus/química , Henipavirus/fisiologia , Infecções por Henipavirus/genética , Humanos , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/metabolismo
10.
Int J Mol Sci ; 19(9)2018 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-30150566

RESUMO

In previous research, a 32 kDa protein in lymphocystis disease virus (LCDV) was identified as viral attachment protein (VAP) that specifically interacted with the 27.8 kDa cellular receptor from flounder Paralichthys olivaceus gill (FG) cells, and the recombinant VAP (rVAP) was expressed in Escherichia coli strain BL21 (DE3). In this study, monoclonal antibodies (MAbs) against 32 kDa VAP are produced by immunization of BALB/c mice with the rVAP. Seven hybridoma secreting MAbs were screened by enzyme-linked immunosorbent assay, five of which designated as 1C6, 1C8, 3B5, 3D11 and 3H10 are cloned by the limiting dilution method, depending on the strongly positive results of ELISA. Western blotting analysis shows that the five MAbs can specifically react with the 32 kDa protein of LCDV and the purified 50 kDa rVAP, and the subtype of the MAbs is identified as IgG. Immunofluorescence results demonstrate that the specific fluorescence signals for LCDV appear in the cytoplasm of FG cells at 24 h post LCDV infection. Neutralization assay results indicate that pre-incubations of LCDV with the five MAbs can significantly decrease the LCDV copy numbers and delay the development of the cytopathic effect in FG cells, revealing that the five MAbs can neutralize the LCDV particles and block viral infection in vitro. The neutralizing MAbs against 32 kDa VAP would be useful for the study on the LCDV⁻host interaction and might be promising inhibitors of LCDV infection in fish.


Assuntos
Anticorpos Monoclonais/imunologia , Iridoviridae/imunologia , Proteínas Recombinantes/imunologia , Proteínas Virais/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Linhagem Celular , Citoplasma/imunologia , Citoplasma/virologia , Brânquias/citologia , Brânquias/imunologia , Brânquias/virologia , Hibridomas/imunologia , Hibridomas/metabolismo , Imunização , Iridoviridae/genética , Camundongos Endogâmicos BALB C , Peso Molecular , Testes de Neutralização , Proteínas Virais/química , Proteínas Virais/genética
11.
Appl Microbiol Biotechnol ; 101(23-24): 8331-8344, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29063173

RESUMO

Glycoprotein D (gD) of bovine herpesvirus-1 (BoHV-1) is essential for attachment and penetration of cells during infection and is a major target for neutralizing antibodies during an adaptive immune response. Currently there are no recombinant antibodies capable of binding gD epitopes for use in treating BoHV-1 infection. In this study, a bovine scFv gene derived from a hybridoma secreting monoclonal antibodies (McAbs) against the amino acid motif MEESKGYEPP of gD was expressed in E. coli. Molecular modeling, western blot and ELISA analysis showed that this scFv had a high affinity for BoHV-1 gD, with a Kd of 161.2 ± 37.58 nM and for whole BoHV-1 virus, with a Kd of 67.44 ± 16.99 nM. In addition, this scFv displayed a high affinity for BoHV-1 antigen in an ELISA and competed with BoHV-1 anti-serum in a competitive ELISA. Immunofluorescence assay (IFA) and laser confocal microscopy showed that this scFv could efficiently bind to and be internalized by BoHV-1 infected Madin-Darby bovine kidney (MDBK) cells. Importantly, this scFv was shown to inhibit BoHV-1 infectivity and to reduce the number of viral plaques by blocking viral attachment to MDBK cells. Our study suggests that this bovine single-chain antibody could be developed for use as a diagnostic and therapeutic agent against BoHV-1 infection in cattle.


Assuntos
Anticorpos Antivirais/imunologia , Herpesvirus Bovino 1/imunologia , Anticorpos de Cadeia Única/imunologia , Proteínas Virais/imunologia , Animais , Anticorpos Antivirais/química , Western Blotting , Bovinos , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Simulação de Acoplamento Molecular , Testes de Neutralização , Ligação Proteica , Anticorpos de Cadeia Única/química , Ensaio de Placa Viral , Proteínas Virais/química
12.
Carbohydr Polym ; 335: 122101, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38616079

RESUMO

In this study, we purified a partially acetylated heteropolysaccharide (Ts1-1A) from the fruit bodies of Trametes sanguinea Lloyd through cold water extraction and serial chromatographic separation. The purified polysaccharide Ts1-1A (12.8 kDa) was characterized as a branched mannogalactofucan with a backbone of alternately connected 1,3-linked α-Fucp and 1,6-linked α-Galp, which was partially substituted by non-reducing end units of ß-Manp at O-2 and O-3 positions of 1,6-linked α-Galp. Ts1-1A showed pronounced anti-human cytomegalovirus activity at the concentration of 200 and 500 µg/mL in systematical assessments including morphological changes, western blotting, qPCR, indirect immunofluorescence and tissue culture infective dose assays. Moreover, Ts1-1A exerted its antiviral activity at two distinct stages of viral proliferation manifesting as significantly inhibiting viral protein (IE1/2 and p52) expression and reducing viral gene (UL123, UL44 and UL32) replication in the HCMV-infected WI-38 cells. At viral attachment stage, Ts1-1A interacted with HCMV and prevented HCMV from attaching to its host cells. While at early phase of viral replication stage, Ts1-1A suppressed HCMV replication by downregulating NQO1 and HO-1 proteins related to oxidative stress as an antioxidant. To sum up, Ts1-1A is a promising anti-HCMV agent which could be developed for HCMV infection prevention and therapy.


Assuntos
Citomegalovirus , Polyporaceae , Humanos , Trametes , Antivirais/farmacologia
13.
mBio ; 14(5): e0139123, 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37737607

RESUMO

IMPORTANCE: Genetically diverse paramyxoviruses are united in their presentation of a receptor-binding protein (RBP), which works in concert with the fusion protein to facilitate host-cell entry. The C-terminal head region of the paramyxoviral RBP, a primary determinant of host-cell tropism and inter-species transmission potential, forms structurally distinct classes dependent upon protein and glycan receptor specificity. Here, we reveal the architecture of the C-terminal head region of the RBPs from Nariva virus (NarV) and Mossman virus (MosV), two archetypal rodent-borne paramyxoviruses within the recently established genus Narmovirus, family Paramyxoviridae. Our analysis reveals that while narmoviruses retain the general architectural features associated with paramyxoviral RBPs, namely, a six-bladed ß-propeller fold, they lack the structural motifs associated with known receptor-mediated host-cell entry pathways. This investigation indicates that the RBPs of narmoviruses exhibit pathobiological features that are distinct from those of other paramyxoviruses.


Assuntos
Proteínas de Transporte , Paramyxovirinae , Proteínas de Transporte/metabolismo , Paramyxoviridae , Proteínas Virais de Fusão/metabolismo , Ligação Proteica , Internalização do Vírus
14.
Antiviral Res ; 211: 105547, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36682463

RESUMO

Human respiratory syncytial virus (RSV) is the leading cause of severe lower respiratory tract infections in infants, the elderly, and the immunocompromised, yet no licensed vaccine and only limited therapeutic options for prevention and treatment are available, which poses a global health challenge and emphasizes the urgent medical need for novel antiviral agents. In the current study, a novel potent small molecule inhibitor of RSV was identified by performing a screening and structure optimization campaign, wherein a naturally occurring dicaffeoylquinic acid (DCQA) compound served as a chemical starting point. The reported benzamide derivative inhibitor, designated as 2f, was selected for its improved stability and potent antiviral activity from a series of investigated structurally related compounds. 2f was well tolerated by cells and able to inhibit RSV infection with a half maximal inhibitory concentration (IC50) of 35 nM and a favorable selectivity index (SI) of 3742. Although the exact molecular target for 2f is not known, in vitro mechanism of action investigations revealed that the compound inhibits the early stage of infection by interacting with RSV virion and interferes primarily with the attachment and potentially with the virus-cell fusion step. Moreover, intranasal administration of 2f to mice simultaneously or prior to intranasal infection with RSV significantly decreased viral load in the lungs, pointing to the in vivo potential of the compound. Our results suggest that 2f is a viable candidate for further preclinical development and evaluation as an antiviral agent against RSV infections.


Assuntos
Infecções por Vírus Respiratório Sincicial , Vírus Sincicial Respiratório Humano , Infecções Respiratórias , Lactente , Camundongos , Humanos , Animais , Idoso , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Pulmão , Linhagem Celular , Infecções Respiratórias/tratamento farmacológico , Antivirais/farmacologia
15.
Front Vet Sci ; 10: 1314624, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38089700

RESUMO

Pseudorabies virus (PRV) belongs to the Alphaherpesvirinae subfamily and serves as an exceptional animal model for investigating the infection mechanism of Herpes simplex virus type 1. Notably, PRV has the capability to infect a wide range of mammals, including humans, highlighting its potential as an overlooked zoonotic pathogen. The attachment and entry steps of PRV into host cells are crucial to accomplish its life cycle, which involve numerous cellular factors. In this mini review, we offer a comprehensive summary of current researches pertaining to the role of cellular factors in PRV attachment and entry stages, with the overarching goal of advancing the development of novel antiviral agents against this pathogen.

16.
J Adv Vet Anim Res ; 10(3): 563-569, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37969804

RESUMO

Objective: The research aimed to isolate, adapt to cell culture, and characterize the lumpy skin disease virus (LSDV) from clinically infected cattle in Bangladesh. Materials and Methods: From September 2019 to June 2020, 37 skin nodules and skin swabs were aseptically collected from afflicted cattle in the outbreak regions of Jhenaidah and Kishoreganj in Bangladesh. The LSDV was isolated from embryonated specific pathogen-free (SPF) chicken eggs along the chorioallantoic membrane (CAM) route and the Vero cell line after several blind passages. The viral attachment protein was targeted for molecular detection using polymerase chain reactions (PCR). For phylogenetic analysis, PCR-positive products were partially sequenced. Results: The virus was evident in the cell line, showed cytopathic effects after the 13 blind passage, and on the CAM of SPF chicken eggs, exhibited thickening of the CAM with pock-like lesions. A total of 12 samples (32.43%) tested positive for LSDV by PCR. Phylogenetic analysis of the present isolates (accession numbers MN792649 and MN792650) revealed 100% similarity with strains from India (MN295064), Kenya (AF325528, MN072619, KX683219), Greece (KY829023), Serbia (KY702007), and Kazakhstan (MN642592); moreover, 99.43% to 100% similarity to the sheep pox virus. Conclusion: Partially sequenced LSDV was developed as a vaccine seed and was first isolated in Bangladesh and characterized at the molecular level.

17.
J Colloid Interface Sci ; 622: 481-493, 2022 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-35525149

RESUMO

Inflenza A viruses (IAVs) are highly transmissible and pathogenic Orthomyxoviruses, which have led to worldwide outbreaks and seasonal pandemics of acute respiratory diseases, causing serious threats to public health. Currently used anti-influenza drugs may cause neurological side effects, and they are increasingly less effective against mutant strains. To help prevent the spread of IAVs, in this work, we have developed quercetin-derived carbonized nanogels (CNGsQur) that display potent viral inhibitory, antioxidative, and anti-inflammatory activities. The antiviral CNGsQur were synthesized by mild carbonization of quercetin (Qur), which successfully preserved their antioxidative and anti-inflammatory properties while also contributed enhanced properties, such as water solubility, viral binding, and biocompatibility. Antiviral assays of co-treatment, pre-treatment, and post-treatment indicate that CNGsQur interacts with the virion, revealing that the major antiviral mechanism resulting in the inhibition of the virus is by their attachment on the cell surface. Among them, the selectivity index (SI) of CNGsQur270 (>857.1) clearly indicated its great potential for clinical application in IAVs inhibition, which was much higher than that of pristine quercetin (63.7) and other clinical drugs (4-81). Compared with quercetin at the same dose, the combined effects of viral inhibition, antioxidative and anti-inflammatory activities impart the superior therapeutic effects of CNGsQur270 aerosol inhalation in the treatment of IAVs infection, as evidenced by a mouse model. These CNGsQur effectively prevent the spread of IAVs and suppress virus-induced inflammation while also exhibiting good in vivo biocompatibility. CNGsQur shows much promise as a clinical therapeutic agent against infection by IVAs.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Vírus da Influenza A , Animais , Anti-Inflamatórios/farmacologia , Antivirais/farmacologia , Vírus da Influenza A Subtipo H1N1/fisiologia , Camundongos , Quercetina/farmacologia
18.
Front Vet Sci ; 9: 844058, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35372537

RESUMO

African swine fever (ASF) is a highly contagious disease and provokes severe economic losses and health threats. At present no effective vaccine or treatment is available to prevent or cure ASF. Consequently, there is an urgent need to develop effective drugs against ASF virus (ASFV). Chlorine dioxide (ClO2), an ideal biocide, has broad-spectrum antibacterial activity and no drug resistance. Here, we found that ClO2 strongly inhibited ASFV replication in porcine alveolar macrophages (PAMs). The inhibitory effect of ClO2 occurred during viral attachment rather than entry, indicating that ClO2 suppressed the early stage of virus life cycle. ClO2 showed a potent anti-ASFV effect when added either before, simultaneously with, or after virus infection. Furthermore, ClO2 could destroy viral nucleic acids and proteins, which may contribute to its capacity of inactivating ASFV virions. The minimum concentration of degradation of ASFV nucleic acids by ClO2 is 1.2 µg/mL, and the degradation is a temperature-dependent manner. These have guiding significance for ClO2 prevention and control of ASFV infection in pig farms. In addition, ClO2 decreased the expression of ASFV-induced inflammatory cytokines. Overall, our findings suggest that ClO2 may be an ideal candidate for the development of novel anti-ASFV prophylactic and therapeutic drugs in swine industry.

19.
Front Vet Sci ; 9: 937653, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35754529

RESUMO

[This corrects the article DOI: 10.3389/fvets.2022.844058.].

20.
Front Pharmacol ; 13: 899181, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35847047

RESUMO

Broccoli (Brassica oleracea L. var. Italica) leaves are a byproduct of broccoli and could be used as a food source. The study aimed to evaluate the effect of broccoli leaves on influenza A virus (IAV) infection. We investigated the effect of ethanol extract of Broccoli leaves (EBL) on IAV infection using green fluorescent protein (GFP)-tagged Influenza A/PR/8/34 virus (PR8-GFP IAV). When EBL and PR8-GFP IAV were cotreated to RAW 264.7 cells, the fluorescence microscopy and fluorescence-activated cell sorting (FACS) analysis showed that EBL significantly reduced the levels of GFP expression by influenza viral infection dose-dependently. Immunofluorescence (IF) analysis confirmed that EBL decreased the expression of IAV proteins. EBL exhibited a strong inhibitory effect of IAV binding on the cells and moderate virucidal impact. Consistently, EBL potently suppressed the hemagglutination by IAV infection. These results indicate that EBL prevents IAV attachment via the inhibition of HA upon viral infection. Finally, EBL as an HA inhibitor of IAV could be used as the natural antiviral source to protect against influenza viral infection.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa