Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 142
Filter
3.
PLoS One ; 13(1): e0191789, 2018.
Article in English | MEDLINE | ID: mdl-29377913

ABSTRACT

Human coronavirus (HCoV) is one of the most common causes of respiratory tract infection throughout the world. To investigate the epidemiological and genetic variation of HCoV in Guangzhou, south China, we collected totally 13048 throat and nasal swab specimens from adults and children with fever and acute upper respiratory infection symptoms in Gunazhou, south China between July 2010 and June 2015, and the epidemiological features of HCoV and its species were studied. Specimens were screened for HCoV by real-time RT-PCR, and 7 other common respiratory viruses were tested simultaneously by PCR or real-time PCR. HCoV was detected in 294 cases (2.25%) of the 13048 samples, with most of them inpatients (251 cases, 85.4% of HCoV positive cases) and young children not in nursery (53.06%, 156 out of 294 HCoV positive cases). Four HCoVs, as OC43, 229E, NL63 and HKU1 were detected prevalent during 2010-2015 in Guangzhou, and among the HCoV positive cases, 60.20% were OC43, 16.67% were 229E, 14.97% were NL63 and 7.82% were HKU1. The month distribution showed that totally HCoV was prevalent in winter, but differences existed in different species. The 5 year distribution of HCoV showed a peak-valley distribution trend, with the detection rate higher in 2011 and 2013 whereas lower in 2010, 2012 and 2014. The age distribution revealed that children (especially those <3 years old) and old people (>50 years) were both high risk groups to be infected by HCoV. Of the 294 HCoV positive patients, 34.69% (101 cases) were co-infected by other common respiratory viruses, and influenza virus was the most common co-infecting virus (30/101, 29.70%). Fifteen HCoV-OC43 positive samples of 2013-2014 were selected for S gene sequencing and phylogenetic analysis, and the results showed that the 15 strains could be divided into 2 clusters in the phylogenetic tree, 12 strains of which formed a separate cluster that was closer to genotype G found in Malaysia. It was revealed for the first time that genotype B and genotype G of HCoV-OC43 co-circulated and the newly defined genotype G was epidemic as a dominant genotype during 2013-2014 in Guanzhou, south China.


Subject(s)
Coronavirus/isolation & purification , Phylogeny , Respiratory Tract Infections/epidemiology , China/epidemiology , Coronavirus/classification , Coronavirus/pathogenicity , Humans
4.
Front Immunol ; 8: 317, 2017.
Article in English | MEDLINE | ID: mdl-28377769

ABSTRACT

Engaging the antibody-dependent cell-mediated cytotoxicity (ADCC) for killing of virus-infected cells and secretion of antiviral cytokines and chemokines was incorporated as one of the important features in the design of universal influenza vaccines. However, investigation of the ADCC epitopes on the highly immunogenic influenza hemagglutinin (HA) head region has been rarely reported. In this study, we determined the ADCC and antiviral activities of two putative ADCC epitopes, designated E1 and E2, on the HA head of a pandemic H1N1 influenza virus in vitro and in a lethal mouse model. Our data demonstrated that sera from the E1-vaccinated mice could induce high ADCC activities. Importantly, the induction of ADCC response modestly decreased viral load in the lungs of H1N1-infected mice. However, the elevated ADCC significantly increased mouse alveolar damage and mortality than that of the PBS-vaccinated group (P < 0.0001). The phenotype was potentially due to an exaggerated inflammatory cell infiltration triggered by ADCC, as an upregulated release of cytotoxic granules (perforin) was observed in the lung tissue of E1-vaccinated mice after H1N1 influenza virus challenge. Overall, our data suggested that ADCC elicited by certain domains of HA head region might have a detrimental rather than protective effect during influenza virus infection. Thus, future design of universal influenza vaccine shall strike a balance between the induction of protective immunity and potential side effects of ADCC.

5.
J Gen Virol ; 98(3): 364-373, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28113045

ABSTRACT

Substitutions in the PA N-terminus (PAN) of influenza A viruses are associated with viral pathogenicity. During our previous study, which identified PAN-V63I and -A37S/I61T/V63I/V100A substitutions as virulence determinants, we observed a severe decrease in virus growth and transcription/replication capacity posed by PAN-A37S/V100A substitution. To further delineate the significance of substitutions at these positions, we generated mutant H7N7 viruses bearing the substitutions PAN-A37S, -A37S/I61T, -A37S/V63I, -V100A, -I61T/V100A and -V63I/V100A by reverse genetics. Our results showed that all mutant viruses except PAN-V100A showed a significantly reduced growth capability in infected cells. At the same time, the PAN-A37S, -A37S/I61T and -A37S/V63I mutant viruses displayed decreased viral transcription and replication by diminishing virus RNA synthesis activity. Biochemical assays indicated that the substitutions PAN-A37S, -A37S/I61T and -A37S/V63I suppressed the polymerase and endonuclease activities when compared with those of the wild-type. Together, our results demonstrated that the PAN-A37S, -A37S/I61T and -A37S/V63I substitutions contributed to a decreased pathogenicity of avian H7N7 influenza A virus.


Subject(s)
Influenza A Virus, H7N7 Subtype/pathogenicity , Influenza in Birds/virology , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , Virulence Factors/metabolism , Amino Acid Substitution , Animals , Birds , Dogs , Influenza A Virus, H7N7 Subtype/genetics , Influenza A Virus, H7N7 Subtype/growth & development , Madin Darby Canine Kidney Cells , Protein Domains , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/genetics , Viral Proteins/genetics , Virulence/genetics , Virulence Factors/genetics , Virus Replication/genetics
6.
Antiviral Res ; 137: 58-66, 2017 01.
Article in English | MEDLINE | ID: mdl-27840201

ABSTRACT

The PB1 C-terminal domain and PB2 N-terminal domain interaction of the influenza A polymerase, which modulates the assembly of PB1 and PB2 subunits, may serve as a valuable target for the development of novel anti-influenza therapeutics. In this study, we performed a systematic screening of a chemical library, followed by the antiviral evaluation of primary hits and their analogues. Eventually, a novel small-molecule compound PP7 that abrogated the PB1-PB2 association and impaired viral polymerase activity was identified. PP7 exhibited antiviral activities against influenza virus subtypes A (H1N1)pdm09, A(H7N9) and A(H9N2) in cell cultures and partially protected mice against lethal challenge of mouse-adapted influenza A (H1N1)pdm09 virus. Surprisingly, a panel of other subtypes of influenza virus, including A(H5N1) and A(H7N7), showed various degrees of resistance to the compound. Biochemical studies revealed a similar pattern of resistance on the impairment of polymerase activity. Molecular docking analyses suggested a PP7-binding site that appeared to be completely conserved among the subtypes of the virus mentioned above. Thus, we propose that alternative/additional binding site (s) may exist for the regulation of PB1-PB2 subunits assembly of influenza A virus.


Subject(s)
Antiviral Agents/pharmacology , Influenza A virus/chemistry , Influenza A virus/drug effects , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/isolation & purification , Antiviral Agents/therapeutic use , Binding Sites , Cell Line , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H7N7 Subtype/drug effects , Influenza A Virus, H7N9 Subtype/drug effects , Influenza A Virus, H9N2 Subtype/drug effects , Influenza A virus/enzymology , Mice , Molecular Docking Simulation , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Small Molecule Libraries , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Replication/drug effects
7.
Virology ; 501: 92-101, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27889648

ABSTRACT

PB2 is one of the subunits of the influenza A virus (IAV) polymerase complex. By bioinformatics analysis we identified PB2 substitutions at positions 389 and 598 among IAV isolates from humans, which might associate with viral pathogenicity. To evaluate the biological significance of these substitutions, PB2-K389R and -V598T/I mutant viruses of avian H7N9 IAVs were generated by reverse genetics. Compared to the wild type, the mutant viruses displayed an enhanced growth capacity in human and mammalian cells. Meanwhile, they presented increased transcription and replication by producing higher levels of viral mRNA, cRNA and vRNA. Minireplicon assays indicated that the polymerase activity was elevated by these substitutions. Notably, the PB2-V598T/I substitutions substantially increased virus replication and virulence in mice. Together, we demonstrated that the substitutions PB2-V598T/I contributed to higher IAV replication and virulence in mammals, which added to the knowledge of IAV virulence determinants and benefited the surveillance of IAVs.


Subject(s)
Amino Acid Substitution , Influenza A Virus, H7N9 Subtype/enzymology , Influenza A Virus, H7N9 Subtype/pathogenicity , Influenza, Human/virology , RNA-Dependent RNA Polymerase/genetics , Viral Proteins/genetics , Animals , Birds , Female , Humans , Influenza A Virus, H7N9 Subtype/genetics , Influenza A Virus, H7N9 Subtype/physiology , Influenza in Birds/virology , Mice , Mice, Inbred BALB C , Molecular Docking Simulation , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Virulence , Virus Replication
8.
Infect Genet Evol ; 47: 68-76, 2017 01.
Article in English | MEDLINE | ID: mdl-27871815

ABSTRACT

Despite recent discoveries of novel animal bocaparvoviruses, current understandings on the diversity and evolution of bocaparvoviruses are still limited. We report the identification and genome characterization of a novel bocaparvovirus, rat bocaparvovirus (RBoV), in brown rats (Rattus norvegicus) in China. RBoV was detected in 11.5%, 2.4%, 16.2% and 0.3% of alimentary, respiratory, spleen and kidney samples respectively, of 636 brown rats by PCR, but not in samples of other rodent species, suggesting that brown rats are the primary reservoir of RBoV. Six RBoV genomes sequenced from three brown rats revealed the presence of three ORFs, characteristic of bocaparvoviruses. Phylogenetic analysis showed that RBoV was distantly related to other bocaparvoviruses, forming a distinct cluster within the genus, with ≤55.5% nucleotide identities to the genome of ungulate bocaparvovirus 3, supporting its classification as a novel bocaparvovirus species. RBoV possessed a putative second exon encoding the C-terminal region of NS1 and conserved RNA splicing signals, similar to human bocaparvoviruses and canine bocaparvovirus. In contrast to human, feline and canine bocaparvoviruses which demonstrates inter/intra-host viral diversity, partial VP1/VP2 sequences of 49 RBoV strains demonstrated little inter-host genetic diversity, suggesting a single genetic group. Although the pathogenicity of RBoV remains to be determined, its presence in different host tissues suggests wide tissue tropism. RBoV represents the first bocaparvovirus in rodents with genome sequenced, which extends our knowledge on the host range of bocaparvoviruses. Further studies are required to better understand the epidemiology, genetic diversity and pathogenicity of bocaparvoviruses in different rodent populations.


Subject(s)
Bocavirus/genetics , Genome, Viral/genetics , Parvoviridae Infections/veterinary , Parvoviridae Infections/virology , Rats/virology , Animals , China , DNA, Viral/genetics , RNA Splice Sites/genetics
9.
J Gen Virol ; 97(12): 3345-3358, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27902362

ABSTRACT

We report the discovery of a novel bocaparvovirus, bat bocaparvovirus (BtBoV), in one spleen, four respiratory and 61 alimentary samples from bats of six different species belonging to three families, Hipposideridae, Rhinolophidae and Vespertilionidae. BtBoV showed a higher detection rate in alimentary samples of Rhinolophus sinicus (5.7 %) than those of other bat species (0.43-1.59 %), supporting R. sinicus as the primary reservoir and virus spillover to accidental bat species. BtBoV peaked during the lactating season of R. sinicus, and it was more frequently detected among female than male adult bats (P<0.05), and among lactating than non-lactating female bats (P<0.0001). Positive BtBoV detection was associated with lower body weight in lactating bats (P<0.05). Ten nearly complete BtBoV genomes from three bat species revealed a unique large ORF1 spanning NS1 and NP1 in eight genomes and conserved splicing signals leading to multiple proteins, as well as a unique substitution in the conserved replication initiator motif within NS1. BtBoV was phylogenetically distantly related to known bocaparvoviruses with ≤57.3 % genome identities, supporting BtBoV as a novel species. Ms-BtBoV from Miniopterus schreibersii and Hp-BtBoV from Hipposideros pomona demonstrated 97.2-99.9 % genome identities with Rs-BtBoVs from R. sinicus, supporting infection of different bat species by a single BtBoV species. Rs-BtBoV_str15 represents the first bat parvovirus genome with non-coding regions sequenced, which suggested the presence of head-to-tail genomic concatamers or episomal forms of the genome. This study represents the first to describe interspecies transmission in BoVs. The high detection rates in lactating female and juvenile bats suggest possible vertical transmission of BtBoV.


Subject(s)
Bocavirus/isolation & purification , Chiroptera/virology , Parvoviridae Infections/veterinary , Animals , Base Sequence , Bocavirus/classification , Bocavirus/genetics , China , Chiroptera/classification , Female , Genome, Viral , Male , Molecular Sequence Data , Open Reading Frames , Parvoviridae Infections/transmission , Parvoviridae Infections/virology , Phylogeny , Seasons , Viral Proteins/genetics , Viral Proteins/metabolism
10.
Sci Rep ; 6: 37800, 2016 11 25.
Article in English | MEDLINE | ID: mdl-27886255

ABSTRACT

The PA N-terminal domain (PA-Nter) is essential for viral transcription and replication. Here we identified PA-Nter substitutions A37S, I61T, V63I and V100A in recently emerged avian influenza A viruses (IAVs) with potential effect on virus pathogenicity and/or host adaptation. We introduced the identified PA-Nter substitutions into avian H7N7 IAV by reverse genetics. Our results showed that single substitution V63I and combined substitutions, I61T/V63I and A37S/I61T/V63I/V100A (Mfour), significantly increased virus growth capacity in mammalian cells. Meanwhile, these substitutions conferred higher virus transcription/replication capacity by producing more mRNA, cRNA and vRNA. Consistently, the polymerase activity and the endonuclease activity were enhanced by these PA-Nter substitutions. Notably, substitutions V63I and Mfour strongly increased virus replication and virulence in mice. Collectively, our findings demonstrated that the PA-Nter substitutions V63I and Mfour enhanced IAV pathogenicity through modification of the polymerase activity and the endonuclease activity, which added to the evolving knowledge of IAV virulence determinants.


Subject(s)
Amino Acid Substitution , Influenza A Virus, H7N7 Subtype/pathogenicity , Influenza in Birds/virology , Viral Proteins/chemistry , Animals , Birds , Cells, Cultured , Humans , Influenza A Virus, H7N7 Subtype/genetics , Influenza A Virus, H7N7 Subtype/growth & development , Mice , RNA, Messenger/genetics , RNA, Viral/genetics , Virulence
11.
ACS Infect Dis ; 2(7): 471-7, 2016 07 08.
Article in English | MEDLINE | ID: mdl-27626099

ABSTRACT

Influenza viruses are obligate parasites that hijack the host cellular system. Previous results have shown that the influenza virus PB2 subunit confers a dependence of host eukaryotic translation initiation factor 4-γ 1 (eIF4G1) for viral mRNA translation. Here, we demonstrated that peptide-mediated interference of the PB2-eIF4G1 interaction inhibited virus replication in vitro and in vivo. Remarkably, intranasal administration of the peptide provided 100% protection against lethal challenges of influenza A viruses in BALB/c mice, including H1N1, H5N1, and H7N9 influenza virus subtypes. Mapping of the PB2 protein indicated that the eIF4G1 binding sites resided within the PB2 cap-binding domain. Virtual docking analysis suggested that the inhibitory peptide associated with the conserved amino acid residues that were essential to PB2 cap-binding activity. Overall, our results identified the PB2-eIF4G1 interactive site as a druggable target for influenza therapeutics.


Subject(s)
Antiviral Agents/pharmacology , Eukaryotic Initiation Factor-4G/metabolism , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H5N1 Subtype/metabolism , Influenza A Virus, H7N9 Subtype/metabolism , Influenza, Human/metabolism , Peptides/pharmacology , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , Animals , Antiviral Agents/chemistry , Cell Line , Eukaryotic Initiation Factor-4G/chemistry , Eukaryotic Initiation Factor-4G/genetics , Female , Humans , Influenza A Virus, H1N1 Subtype/chemistry , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/chemistry , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H7N9 Subtype/chemistry , Influenza A Virus, H7N9 Subtype/genetics , Influenza, Human/drug therapy , Influenza, Human/genetics , Influenza, Human/virology , Mice , Mice, Inbred BALB C , Peptides/chemistry , Protein Binding , Protein Domains , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , Viral Proteins/chemistry , Viral Proteins/genetics
12.
Nat Microbiol ; 1: 16004, 2016 Feb 22.
Article in English | MEDLINE | ID: mdl-27572168

ABSTRACT

Middle East respiratory syndrome coronavirus (MERS-CoV) causes sporadic zoonotic disease and healthcare-associated outbreaks in human. MERS is often complicated by acute respiratory distress syndrome (ARDS) and multi-organ failure(1,2). The high incidence of renal failure in MERS is a unique clinical feature not often found in other human coronavirus infections(3,4). Whether MERS-CoV infects the kidney and how it triggers renal failure are not understood(5,6). Here, we demonstrated renal infection and apoptotic induction by MERS-CoV in human ex vivo organ culture and a nonhuman primate model. High-throughput analysis revealed that the cellular genes most significantly perturbed by MERS-CoV have previously been implicated in renal diseases. Furthermore, MERS-CoV induced apoptosis through upregulation of Smad7 and fibroblast growth factor 2 (FGF2) expression in both kidney and lung cells. Conversely, knockdown of Smad7 effectively inhibited MERS-CoV replication and protected cells from virus-induced cytopathic effects. We further demonstrated that hyperexpression of Smad7 or FGF2 induced a strong apoptotic response in kidney cells. Common marmosets infected by MERS-CoV developed ARDS and disseminated infection in kidneys and other organs. Smad7 and FGF2 expression were elevated in the lungs and kidneys of the infected animals. Our results provide insights into the pathogenesis of MERS-CoV and host targets for treatment.


Subject(s)
Apoptosis , Coronavirus Infections/pathology , Fibroblast Growth Factor 2/metabolism , Kidney/pathology , Lung/pathology , Middle East Respiratory Syndrome Coronavirus/pathogenicity , Smad7 Protein/metabolism , Animals , Callithrix , Cytopathogenic Effect, Viral , Disease Models, Animal , Host-Pathogen Interactions , Humans , Organ Culture Techniques
13.
Virology ; 498: 1-8, 2016 11.
Article in English | MEDLINE | ID: mdl-27525812

ABSTRACT

To evaluate the pathogenicity, a highly pathogenic avian influenza H7N7 virus (A/Netherlands/219/03) isolated from human was passaged in mice. A mutant virus (mH7N7) with attenuated virulence was isolated from mouse lung, which had a 3-log higher MLD50 than the wild-type virus (wH7N7). Sequence analysis and reverse genetics study revealed that mutations in PA account for the compromised viral replication in mammalian cells and mice. A mini-genome assay demonstrated that PA mutations P103H and S659L can cooperatively decrease polymerase activity. Actually, PA with double mutation P103H-S659L cannot sustain the generation of live virus by reverse genetics. Interestingly, the prior infection of mH7N7 virus provided mice with cross-protection against lethal challenge of other subtypes of influenza A virus including H1N1, H5N1 and H7N9. In conclusion, we demonstrated that PA mutations P103H and S659L can cooperatively reduce polymerase activity and viral replication in mammalian cells and attenuate pathogenicity in mice.


Subject(s)
Amino Acid Substitution , Host-Pathogen Interactions , Influenza A Virus, H7N7 Subtype/genetics , Influenza A Virus, H7N7 Subtype/pathogenicity , RNA-Dependent RNA Polymerase/genetics , Viral Proteins/genetics , Animals , Cell Line , Cells, Cultured , Female , Humans , Mice , Mutation , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Polymorphism, Genetic , Viral Load , Virulence/genetics , Virulence Factors/genetics , Virus Replication
14.
J Antimicrob Chemother ; 71(9): 2489-97, 2016 09.
Article in English | MEDLINE | ID: mdl-27272726

ABSTRACT

OBJECTIVES: The conserved residues 318-483 in the PB2 subunit of influenza A polymerase is an independently folded cap-binding domain (PB2cap) that exhibits a distinct binding mode from other host cap-binding proteins, which suggests that PB2cap might be an ideal drug target. This study aimed to identify a new class of anti-influenza inhibitors that specifically disrupts the interaction between PB2cap and host cap structures. METHODS: An innovative fluorescence polarization assay was established for primary screening, followed by cap-binding inhibitory activity, antiviral efficacy and cytotoxicity evaluations of the selected compounds. The best compound was characterized by multi-cycle virus growth assay, cross-protection test, synergism evaluation, mini-replicon assay, binding affinity analysis, docking simulation and mouse study. RESULTS: Several PB2 cap-binding inhibitors were discovered. The compound 7-(4-hydroxy-2-oxo-2H-chromen-3-yl)-6H,7H,8H-chromeno[3',4':5,6]pyrano[3,2-c]chromene-6,8-dione, designated PB2-39, was identified as a potent inhibitor of replication of multiple subtypes of influenza A virus, including H1N1, H3N2, H5N1, H7N7, H7N9 and H9N2 in vitro and H1N1, H5N1 and H7N9 in vivo. Combinational treatment with the influenza virus release inhibitor zanamivir and PB2-39 exerted a synergistic anti-influenza effect. Mechanistic experiments supported that PB2-39 suppressed viral polymerase activity. Docking and binding affinity analyses demonstrated that PB2-39 interacted with the PB2 cap-binding pocket, suggesting its role as a cap-binding competitor. CONCLUSIONS: Our study provides new insights for the strategic development of novel cap-binding inhibitors of influenza A viruses.


Subject(s)
Antiviral Agents/isolation & purification , Antiviral Agents/pharmacology , Influenza A virus/drug effects , Influenza A virus/physiology , RNA Cap-Binding Proteins/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Virus Replication/drug effects , Animals , Antiviral Agents/toxicity , Cell Line , Cell Survival/drug effects , Disease Models, Animal , Drug Synergism , Female , Fluorescence Polarization , Humans , Mice, Inbred BALB C , Orthomyxoviridae Infections/drug therapy , Treatment Outcome
15.
AIDS Res Hum Retroviruses ; 32(9): 909-17, 2016 09.
Article in English | MEDLINE | ID: mdl-27067022

ABSTRACT

The emergence of drug resistance mutations is increasing after the implementation of highly active antiretroviral therapy. To characterize two novel mutations L228I and Y232H in the primer grip of reverse transcriptase (RT) of HIV-1 circulating recombination form 08_BC (CRF08_BC) subtype, both mutant clones were constructed to determine their impacts on viral phenotypic susceptibility and replication capacity (RC). Results showed that the novel mutation, L228I, conferred a low-level resistance to etravirine by itself. L228I in combination with Y188C displayed a high level of cross-resistance to both nevirapine (NVP) and efavirenz (EFV). The copresence of A139V and Y232H induced a moderate level of resistance to NVP and EFV. Mutations Y188C/L228I, A139V, Y232H, and A139V/Y232H reduced more than 55% of viral RC compared with that of the wild-type (WT) reference virus. Modeling study suggested that the copresence of Y188C/L228I or A139V/Y232H might induce conformational changes to RT, which might result in reduced drug susceptibility and viral RC due to abolished hydrogen bonding or complex interaction with vicinal residues. Our results demonstrated that L228I and Y232H were novel accessory nonnucleoside reverse transcriptase inhibitor resistance-related mutations and provided valuable information for clinicians to design more effective treatment to patients infected with HIV-1 subtype CRF08_BC.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Resistance, Viral , HIV Reverse Transcriptase/genetics , HIV-1/drug effects , Mutant Proteins/genetics , Mutation, Missense , Reverse Transcriptase Inhibitors/pharmacology , Genotype , HIV-1/enzymology , HIV-1/genetics , HIV-1/physiology , Humans , Microbial Sensitivity Tests , Mutation , Virus Replication
16.
Sci Rep ; 6: 22880, 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-26956222

ABSTRACT

The RNA-dependent RNA polymerase of influenza A virus comprises conserved and independently-folded subdomains with defined functionalities. The N-terminal domain of the PA subunit (PA(N)) harbors the endonuclease function so that it can serve as a desired target for drug discovery. To identify a class of anti-influenza inhibitors that impedes PA(N) endonuclease activity, a screening approach that integrated the fluorescence resonance energy transfer based endonuclease inhibitory assay with the DNA gel-based endonuclease inhibitory assay was conducted, followed by the evaluation of antiviral efficacies and potential cytotoxicity of the primary hits in vitro and in vivo. A small-molecule compound ANA-0 was identified as a potent inhibitor against the replication of multiple subtypes of influenza A virus, including H1N1, H3N2, H5N1, H7N7, H7N9 and H9N2, in cell cultures. Combinational treatment of zanamivir and ANA-0 exerted synergistic anti-influenza effect in vitro. Intranasal administration of ANA-0 protected mice from lethal challenge and reduced lung viral loads in H1N1 virus infected BALB/c mice. In summary, ANA-0 shows potential to be developed to novel anti-influenza agents.


Subject(s)
Antiviral Agents/pharmacology , Influenza A virus/drug effects , Influenza A virus/enzymology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Virus Replication/drug effects , Administration, Intranasal , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/isolation & purification , Drug Evaluation, Preclinical , Drug Synergism , Female , Influenza A virus/physiology , Lung/virology , Mice, Inbred BALB C , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Survival Analysis , Treatment Outcome , Viral Load
17.
Sci Rep ; 6: 22008, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26911565

ABSTRACT

A safe, potent and broad-spectrum antiviral is urgently needed to combat emerging respiratory viruses. In light of the broad antiviral activity of ß-defensins, we tested the antiviral activity of 11 peptides derived from mouse ß-defensin-4 and found that a short peptide, P9, exhibited potent and broad-spectrum antiviral effects against multiple respiratory viruses in vitro and in vivo, including influenza A virus H1N1, H3N2, H5N1, H7N7, H7N9, SARS-CoV and MERS-CoV. The antiviral activity of P9 was attributed to its high-affinity binding to viral glycoproteins, as well as the abundance of basic amino acids in its composition. After binding viral particles through viral surface glycoproteins, P9 entered into cells together with the viruses via endocytosis and prevented endosomal acidification, which blocked membrane fusion and subsequent viral RNA release. This study has paved the avenue for developing new prophylactic and therapeutic agents with broad-spectrum antiviral activities.


Subject(s)
Antiviral Agents/pharmacology , Peptides/pharmacology , Animals , Endosomes/drug effects , Endosomes/metabolism , Endosomes/virology , Influenza A virus/drug effects , Influenza A virus/physiology , Inhibitory Concentration 50 , Mice , Microbial Sensitivity Tests , Middle East Respiratory Syndrome Coronavirus/drug effects , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Peptide Fragments/pharmacology , Peptides/chemistry , Peptides/metabolism , Protein Binding , Severe acute respiratory syndrome-related coronavirus/drug effects , Viral Envelope Proteins/metabolism , Viral Load , beta-Defensins/chemistry
18.
PLoS Pathog ; 12(1): e1005367, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26735852

ABSTRACT

B-1 cells play a critical role in early protection during influenza infections by producing natural IgM antibodies. However, the underlying mechanisms involved in regulating this process are largely unknown. Here we found that during influenza infection pleural cavity B-1a cells rapidly infiltrated lungs, where they underwent plasmacytic differentiation with enhanced IgM production. This process was promoted by IL-17A signaling via induction of Blimp-1 expression and NF-κB activation in B-1a cells. Deficiency of IL-17A led to severely impaired B-1a-derived antibody production in the respiratory tract, resulting in a deficiency in viral clearance. Transfer of B-1a-derived natural antibodies rescued Il17a-/- mice from otherwise lethal infections. Together, we identify a critical function of IL-17A in promoting the plasmacytic differentiation of B-1a cells. Our findings provide new insights into the mechanisms underlying the regulation of pulmonary B-1a cell response against influenza infection.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation , Interleukin-17/immunology , Orthomyxoviridae Infections/immunology , Transcription Factors/biosynthesis , Animals , B-Lymphocytes/cytology , Cell Differentiation/immunology , Chromatin Immunoprecipitation , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Gene Expression Regulation/immunology , Immunity, Humoral/immunology , Influenza A Virus, H1N1 Subtype/immunology , Interleukin-17/deficiency , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1 , Real-Time Polymerase Chain Reaction , Respiratory Tract Infections/immunology , Transcription Factors/immunology
19.
J Control Release ; 222: 130-40, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26682504

ABSTRACT

Progress in bacterial therapy for cancer and infectious diseases is hampered by the absence of safe and efficient vectors. Sustained delivery and high gene expression levels are critical for the therapeutic efficacy. Here we developed a Salmonella typhimrium strain to maintain and safely deliver a plasmid vector to target tissues. This vector is designed to allow dual transcription of therapeutic factors, such as cytotoxic proteins, short hairpin RNAs or combinations, in the nucleus or cytoplasm of eukaryotic cells, with this expression sustained by an autocatalytic positive-feedback loop. Mechanisms to prime the system and maintain the plasmid in the bacterium are also provided. Synergistic effects of attenuated Salmonella and our inter-kingdom system allow the precise expression of Diphtheria toxin A chain (DTA) gene in tumor microenvironment and eradicate large established tumors in immunocompetent animals. In the experiments reported here, 26% of mice (n=5/19) with aggressive tumors were cured and the others all survived until the end of the experiment. We also demonstrated that ST4 packaged with shRNA-encoding plasmids has sustained knockdown effects in nude mice bearing human MDA-MB-231 xenografts. Three weeks after injection of 5×10(6) ST4/pIKT-shPlk, PLK1 transcript levels in tumors were 62.5±18.6% lower than the vector control group (P=0.015). The presence of PLK1 5' RACE-PCR cleavage products confirmed a sustained RNAi-mediated mechanism of action. This innovative technology provides an effective and versatile vehicle for efficient inter-kingdom gene delivery that can be applied to cancer therapy and other purposes.


Subject(s)
Genetic Therapy/methods , Salmonella typhimurium/genetics , Animals , Cell Cycle Proteins/genetics , Cell Line, Tumor , Diphtheria Toxin/genetics , Feedback, Physiological , Female , Genetic Vectors , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/therapy , Peptide Fragments/genetics , Plasmids , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , RNA Interference , RNA, Small Interfering/genetics , Polo-Like Kinase 1
20.
Antiviral Res ; 125: 34-42, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26593979

ABSTRACT

Assembly of the heterotrimeric influenza virus polymerase complex from the individual subunits PB1, PA, and PB2 is a prerequisite for viral replication, in which the interaction between the C terminal of PA (PAC) and the N-terminal of PB1 (PB1N) may be a desired target for antiviral development. In this study, we compared the feasibility of high throughput screening by enzyme-linked immunosorbent assay (ELISA) and fluorescence polarization assay. Among the two, ELISA was demonstrated to own broader dynamic range so that it was used for screening inhibitors that blocked PAC and PB1N interaction. Several binding inhibitors of PAC-PB1N were identified and subsequently tested for the antiviral efficacy. Apparently, 3-(2-chlorophenyl)-6-ethyl-7-methyl[1,2,4]triazolo[4,3-a]pyrimidin-5-ol, designated ANA-1, was found to be a strong inhibitor of viral polymerase activity and act as a potent antiviral agent against the infections of multiple subtypes of influenza A virus, including H1N1, H3N2, H5N1, H7N7, H7N9 and H9N2 subtypes, in cell cultures. Intranasal administration of ANA-1 protected mice from lethal challenge and reduced lung viral loads in H1N1 virus infected BALB/c mice. Docking analyses predicted that ANA-1 bound to an allosteric site of PAC, which might cause conformational changes thereby disrupting the PAC-PB1N interaction. Overall, our study has identified a novel compound with potential to be developed as an anti-influenza drug.


Subject(s)
Antiviral Agents/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Orthomyxoviridae/drug effects , Orthomyxoviridae/enzymology , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , Virus Replication/drug effects , Allosteric Site/drug effects , Animals , Cell Line , Dogs , Enzyme-Linked Immunosorbent Assay , Female , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H1N1 Subtype/physiology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Molecular Docking Simulation , Orthomyxoviridae/classification , Orthomyxoviridae/physiology , Protein Binding/drug effects , Protein Conformation , Protein Structure, Tertiary , Small Molecule Libraries/pharmacology , Virus Replication/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...