Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Adv Sci (Weinh) ; 10(33): e2303615, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37867242

RESUMEN

Biomimetic viral mineralization improves viral vaccine stability and immunogenicity using inorganic metals such as Ca, Al, or Fe. Mn is a metal found in high concentrations in mammalian tissues; however, under natural or laboratory conditions, Mn mineralization by medical viruses has yet to be established. Herein, a single IAV particle is successfully encapsulated with manganese phosphate (MnP) under specific conditions using the human influenza A virus (IAV). MnP-mineralized IAVs (IAV@Mn) exhibited physiochemical and in vitro properties similar to Ca-mineralized IAVs. In animal models, IAV@Mn shows limited replication in immune-competent cells and a significant attenuation compared to naïve cells. Moreover, a single-dose vaccination with IAV@Mn induced robust humoral and cellular immune responses and conferred significant protection against a wild-type IAV challenge in mice. Thus, Mn mineralization in pathogenic viruses provides a rapid and universal strategy for generating an emergency vaccine in response to emerging viruses.


Asunto(s)
Virus de la Influenza A , Infecciones por Orthomyxoviridae , Vacunas , Ratones , Animales , Humanos , Manganeso , Mamíferos
2.
World J Pediatr ; 19(10): 1009-1016, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36811744

RESUMEN

BACKGROUND: Previous serological studies of human bocavirus (HBoV) 1 could not exclude cross-reactivity with the other three HBoVs, particularly HBoV2. METHODS: To search for genotype-specific antibodies against HBoV1 and HBoV2, the divergent regions (DRs) located on the major capsid protein VP3 were defined through viral amino acid alignment and structure prediction. DR-deduced peptides were used as antigens to harvest corresponding anti-DR rabbit sera. To determine their genotype specificities for HBoV1 and HBoV2, these sera samples were used as antibodies against the antigens VP3 of HBoV1 and HBoV2 (expressed in Escherichia coli) in western blotting (WB), enzyme-linked immunosorbent assay (ELISA), and bio-layer interferometry (BLI) assays. Subsequently, the antibodies were evaluated with clinical specimens from pediatric patients with acute respiratory tract infection by indirect immunofluorescence assay (IFA). RESULTS: There were four DRs (DR1-4) located on VP3 with different secondary and tertiary structures between HBoV1 and HBoV2. Regarding the reactivity with VP3 of HBoV1 or HBoV2 in WB and ELISA, high intra-genotype cross-reactivity of anti-HBoV1 or HBoV2 DR1, DR3, and DR4, but not anti-DR2, was observed. Genotype-specific binding capacity of anti-DR2 sera was confirmed by BLI and IFA, in which only anti-HBoV1 DR2 antibody reacted with HBoV1-positive respiratory specimens. CONCLUSION: Antibodies against DR2, located on VP3 of HBoV1 or HBoV2, were genotype specific for HBoV1 and HBoV2, respectively.


Asunto(s)
Bocavirus Humano , Infecciones por Parvoviridae , Infecciones del Sistema Respiratorio , Animales , Niño , Humanos , Conejos , Bocavirus Humano/genética , Infecciones por Parvoviridae/diagnóstico , Ensayo de Inmunoadsorción Enzimática , Genotipo , Infecciones del Sistema Respiratorio/diagnóstico , Escherichia coli
3.
J Med Virol ; 95(1): e28411, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36524893

RESUMEN

A series of nonpharmaceutical interventions (NPIs) was launched in Beijing, China, on January 24, 2020, to control coronavirus disease 2019. To reveal the roles of NPIs on the respiratory syncytial virus (RSV), respiratory specimens collected from children with acute respiratory tract infection between July 2017 and Dec 2021 in Beijing were screened by capillary electrophoresis-based multiplex PCR (CEMP) assay. Specimens positive for RSV were subjected to a polymerase chain reaction (PCR) and genotyped by G gene sequencing and phylogenetic analysis using iqtree v1.6.12. The parallel and fixed (paraFix) mutations were analyzed with the R package sitePath. Clinical data were compared using SPSS 22.0 software. Before NPIs launched, each RSV endemic season started from October/November to February/March of the next year in Beijing. After that, the RSV positive rate abruptly dropped from 31.93% in January to 4.39% in February 2020; then, a dormant state with RSV positive rates ≤1% from March to September, a nearly dormant state in October (2.85%) and November (2.98%) and a delayed endemic season in 2020, and abnormal RSV positive rates remaining at approximately 10% in summer until September 2021 were detected. Finally, an endemic RSV season returned in October 2021. There was a game between Subtypes A and B, and RSV-A replaced RSV-B in July 2021 to become the dominant subtype. Six RSV-A and eight RSV-B paraFix mutations were identified on G. The percentage of severe pneumonia patients decreased to 40.51% after NPIs launched. NPIs launched in Beijing seriously interfered with the endemic season of RSV.


Asunto(s)
COVID-19 , Infecciones por Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano , Infecciones del Sistema Respiratorio , Niño , Humanos , Lactante , Infecciones por Virus Sincitial Respiratorio/epidemiología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Beijing/epidemiología , Filogenia , COVID-19/epidemiología , COVID-19/prevención & control , Virus Sincitial Respiratorio Humano/genética , Reacción en Cadena de la Polimerasa Multiplex
4.
J Virol ; 96(23): e0087922, 2022 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-36377874

RESUMEN

The glycan loop of Zika virus (ZIKV) envelope protein (E) contains the glycosylation site and has been well documented to be important for viral pathogenesis and transmission. In the present study, we report that deletions in the E glycan loop, which were recorded in African ZIKV strains previously, have re-emerged in their contemporary Asian lineages. Here, we generated recombinant ZIKV containing specific deletions in the E glycan loop by reverse genetics. Extensive in vitro and in vivo characterization of these deletion mutants demonstrated an attenuated phenotype in an adult A129 mouse model and reduced oral infections in mosquitoes. Surprisingly, these glycan loop deletion mutants exhibited an enhanced neurovirulence phenotype, and resulted in a more severe microcephalic brain in neonatal mouse models. Crystal structures of the ZIKV E protein and a deletion mutant at 2.5 and 2.6 Å, respectively, revealed that deletion of the glycan loop induces encephalitic flavivirus-like conformational alterations, including the appearance of perforations on the surface and a clear change in the topology of the loops. Overall, our results demonstrate that the E glycan loop deletions represent neonatal mouse neurovirulence markers of ZIKV. IMPORTANCE Zika virus (ZIKV) has been identified as a cause of microcephaly and acquired evolutionary mutations since its discovery. Previously deletions in the E glycan loop were recorded in African ZIKV strains, which have re-emerged in the contemporary Asian lineages recently. The glycan loop deletion mutants are not glycosylated, which are attenuated in adult A129 mouse model and reduced oral infections in mosquitoes. More importantly, the glycan loop deletion mutants induce an encephalitic flavivirus-like conformational alteration in the E homodimer, resulting in a significant enhancement of neonatal mouse neurovirulence. This study underscores the critical role of glycan loop deletion mutants in ZIKV pathogenesis, highlighting a need for global virological surveillance for such ZIKV variants.


Asunto(s)
Proteínas del Envoltorio Viral , Infección por el Virus Zika , Virus Zika , Animales , Ratones , Modelos Animales de Enfermedad , Polisacáridos/química , Proteínas del Envoltorio Viral/genética , Virulencia , Replicación Viral/genética , Virus Zika/genética , Virus Zika/patogenicidad , Infección por el Virus Zika/virología
5.
J Med Virol ; 94(9): 4309-4318, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35587571

RESUMEN

Zika virus (ZIKV) suddenly evolved from a neglected arthropod-borne flavivirus into a pandemic pathogen during 2015-2016. A panel of amino acid mutations has been shown to be responsible for the enhanced neurovirulence and transmissibility of ZIKV. Recent studies have demonstrated that ZIKV genomic RNA is modified by host N6-methyladenosine (m6 A) machinery during viral replication in host cells, and the m6 A profiles vary among different isolates and different host cells. In the present study, using a contemporary Asian ZIKV strain isolated in 2019 (SZ1901) as a model, we profiled m6 A modifications on both the viral genome RNA and cellular transcripts from the ZIKV-infected human hepatocarcinoma cell line Huh7. Methylated RNA immunoprecipitation sequencing (MeRIP-seq) identified a unique m6 A map in the genome of ZIKV strain SZ1901 that is different from all previous isolates. Meanwhile, ZIKV infection induced m6 A upregulation in the CDS regions but downregulation in the 3' untranslated region of host RNA transcripts. The m6 A peak intensity in the majority of host genes was downregulated, including ISG-related genes. Overall, our study describes unique viral and host m6 A profiles in contemporary ZIKV-infected Huh7 cells, highlighting the complexity and importance of m6 A modification during viral infection.


Asunto(s)
Infección por el Virus Zika , Virus Zika , Regiones no Traducidas 3' , Genoma Viral , Humanos , ARN Viral/genética , ARN Viral/metabolismo , Replicación Viral/fisiología , Virus Zika/fisiología
6.
Innovation (Camb) ; 3(2): 100221, 2022 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-35252935

RESUMEN

The highly pathogenic and readily transmissible SARS-CoV-2 has caused a global coronavirus pandemic, urgently requiring effective countermeasures against its rapid expansion. All available vaccine platforms are being used to generate safe and effective COVID-19 vaccines. Here, we generated a live-attenuated candidate vaccine strain by serial passaging of a SARS-CoV-2 clinical isolate in Vero cells. Deep sequencing revealed the dynamic adaptation of SARS-CoV-2 in Vero cells, resulting in a stable clone with a deletion of seven amino acids (N679SPRRAR685) at the S1/S2 junction of the S protein (named VAS5). VAS5 showed significant attenuation of replication in multiple human cell lines, human airway epithelium organoids, and hACE2 mice. Viral fitness competition assays demonstrated that VAS5 showed specific tropism to Vero cells but decreased fitness in human cells compared with the parental virus. More importantly, a single intranasal injection of VAS5 elicited a high level of neutralizing antibodies and prevented SARS-CoV-2 infection in mice as well as close-contact transmission in golden Syrian hamsters. Structural and biochemical analysis revealed a stable and locked prefusion conformation of the S trimer of VAS5, which most resembles SARS-CoV-2-3Q-2P, an advanced vaccine immunogen (NVAX-CoV2373). Further systematic antigenic profiling and immunogenicity validation confirmed that the VAS5 S trimer presents an enhanced antigenic mimic of the wild-type S trimer. Our results not only provide a potent live-attenuated vaccine candidate against COVID-19 but also clarify the molecular and structural basis for the highly attenuated and super immunogenic phenotype of VAS5.

8.
Cell Res ; 31(4): 404-414, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33510385

RESUMEN

The newly identified Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has resulted in a global health emergency because of its rapid spread and high mortality. The molecular mechanism of interaction between host and viral genomic RNA is yet unclear. We demonstrate herein that SARS-CoV-2 genomic RNA, as well as the negative-sense RNA, is dynamically N6-methyladenosine (m6A)-modified in human and monkey cells. Combined RIP-seq and miCLIP analyses identified a total of 8 m6A sites at single-base resolution in the genome. Especially, epidemic strains with mutations at these identified m6A sites have emerged worldwide, and formed a unique cluster in the US as indicated by phylogenetic analysis. Further functional experiments showed that m6A methylation negatively regulates SARS-CoV-2 infection. SARS-CoV-2 infection also triggered a global increase in host m6A methylome, exhibiting altered localization and motifs of m6A methylation in mRNAs. Altogether, our results identify m6A as a dynamic epitranscriptomic mark mediating the virus-host interaction.


Asunto(s)
Adenosina/análogos & derivados , Genoma Viral , SARS-CoV-2/genética , Adenosina/metabolismo , Animales , COVID-19/patología , COVID-19/virología , Línea Celular , Chlorocebus aethiops , Metilación de ADN , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Mutagénesis Sitio-Dirigida , Filogenia , ARN Mensajero/genética , SARS-CoV-2/clasificación , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/fisiología , Células Vero , Replicación Viral
10.
Cell Res ; 31(1): 25-36, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33262452

RESUMEN

Structural principles underlying the composition and synergistic mechanisms of protective monoclonal antibody cocktails are poorly defined. Here, we exploited antibody cooperativity to develop a therapeutic antibody cocktail against SARS-CoV-2. On the basis of our previously identified humanized cross-neutralizing antibody H014, we systematically analyzed a fully human naive antibody library and rationally identified a potent neutralizing antibody partner, P17, which confers effective protection in animal model. Cryo-EM studies dissected the nature of the P17 epitope, which is SARS-CoV-2 specific and distinctly different from that of H014. High-resolution structure of the SARS-CoV-2 spike in complex with H014 and P17, together with functional investigations revealed that in a two-antibody cocktail, synergistic neutralization was achieved by S1 shielding and conformational locking, thereby blocking receptor attachment and viral membrane fusion, conferring high potency as well as robustness against viral mutation escape. Furthermore, cluster analysis identified a hypothetical 3rd antibody partner for further reinforcing the cocktail as pan-SARS-CoVs therapeutics.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , COVID-19 , Epítopos/inmunología , SARS-CoV-2/inmunología , Anticuerpos de Cadena Única/inmunología , Animales , Anticuerpos Neutralizantes/farmacología , Anticuerpos Antivirales/farmacología , COVID-19/inmunología , COVID-19/prevención & control , Chlorocebus aethiops , Modelos Animales de Enfermedad , Humanos , Anticuerpos de Cadena Única/farmacología , Células Vero
11.
Small ; 16(41): e2002435, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32954651

RESUMEN

Infection with live-attenuated vaccines always inevitably induces side effects that reduce their safety. This study suggests a concept of magnetic virus produced by genetically modifying viral surfaces with Fe3 O4 nanoparticles (NPs) to control their tropisms. An iron-affinity peptide is designed to be displayed on the viral surface protein (VP1) of human enterovirus type 71 (EV71), a typical nonenveloped picornavirus, as the model. The modified EV71 can self-bind with Fe3 O4 NPs under physiological conditions, resulting in novel EV71-Fe3 O4 hybrid materials. This rationally engineered EV71 with Fe3 O4 retains its original biological infectivity, but its tropism can be precisely controlled by magnetism. Both in vitro and in vivo experiments demonstrate that EV71-Fe3 O4 can infect only a desired area within the limit of the applied magnetic field, which effectively reduces its pathological damage. More importantly, this characteristic of EV71 can be inherited due to the gene-induced coassembly of viruses and NPs. This achievement provides a proof of concept in virus vaccine improvement by a combination of gene modification and material incorporation, leading to great potential for biomedical developments.


Asunto(s)
Enterovirus , Nanopartículas , Humanos , Fenómenos Magnéticos
12.
Sci Adv ; 6(6): eaax7989, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32076641

RESUMEN

Mosquito-borne flaviviruses infect both mammals and mosquitoes. RNA interference (RNAi) has been demonstrated as an anti-flavivirus mechanism in mosquitoes; however, whether and how flaviviruses induce and antagonize RNAi-mediated antiviral immunity in mammals remains unknown. We show that the nonstructural protein NS2A of dengue virus-2 (DENV2) act as a viral suppressor of RNAi (VSR). When NS2A-mediated RNAi suppression was disabled, the resulting mutant DENV2 induced Dicer-dependent production of abundant DENV2-derived siRNAs in differentiated mammalian cells. VSR-disabled DENV2 showed severe replication defects in mosquito and mammalian cells and in mice that were rescued by RNAi deficiency. Moreover, NS2As of multiple flaviviruses act as VSRs in vitro and during viral infection in both organisms. Overall, our findings demonstrate that antiviral RNAi can be induced by flavivirus, while flavivirus uses NS2A as a bona fide VSR to evade RNAi in mammals and mosquitoes, highlighting the importance of RNAi in flaviviral vector-host life cycles.


Asunto(s)
Culicidae/virología , Infecciones por Flavivirus/genética , Infecciones por Flavivirus/virología , Flavivirus/fisiología , Interacciones Huésped-Patógeno/genética , Interferencia de ARN , Animales , Infecciones por Flavivirus/transmisión , Regulación de la Expresión Génica , Mosquitos Vectores/virología , Ribonucleasa III/metabolismo , Proteínas no Estructurales Virales/metabolismo
13.
J Virol ; 94(6)2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-31896596

RESUMEN

Mosquito-borne flaviviruses consist of a positive-sense genome RNA flanked by the untranslated regions (UTRs). There is a panel of highly complex RNA structures in the UTRs with critical functions. For instance, Xrn1-resistant RNAs (xrRNAs) halt Xrn1 digestion, leading to the production of subgenomic flaviviral RNA (sfRNA). Conserved short direct repeats (DRs), also known as conserved sequences (CS) and repeated conserved sequences (RCS), have been identified as being among the RNA elements locating downstream of xrRNAs, but their biological function remains unknown. In this study, we revealed that the specific DRs are involved in the production of specific sfRNAs in both mammalian and mosquito cells. Biochemical assays and structural remodeling demonstrate that the base pairings in the stem of these DRs control sfRNA formation by maintaining the binding affinity of the corresponding xrRNAs to Xrn1. On the basis of these findings, we propose that DRs functions like a bracket holding the Xrn1-xrRNA complex for sfRNA formation.IMPORTANCE Flaviviruses include many important human pathogens. The production of subgenomic flaviviral RNAs (sfRNAs) is important for viral pathogenicity as a common feature of flaviviruses. sfRNAs are formed through the incomplete degradation of viral genomic RNA by the cytoplasmic 5'-3' exoribonuclease Xrn1 halted at the Xrn1-resistant RNA (xrRNA) structures within the 3'-UTR. The 3'-UTRs of the flavivirus genome also contain distinct short direct repeats (DRs), such as RCS3, CS3, RCS2, and CS2. However, the biological functions of these ancient primary DR sequences remain largely unknown. Here, we found that DR sequences are involved in sfRNA formation and viral virulence and provide novel targets for the rational design of live attenuated flavivirus vaccine.


Asunto(s)
Regiones no Traducidas 3'/fisiología , Flavivirus/metabolismo , Genoma Viral/fisiología , Conformación de Ácido Nucleico , ARN Viral/biosíntesis , Secuencias Repetidas en Tándem/fisiología , Células A549 , Animales , Chlorocebus aethiops , Cricetinae , Culicidae/metabolismo , Culicidae/virología , Flavivirus/genética , Humanos , ARN Viral/genética , Células Vero
14.
Cell Res ; 29(4): 265-273, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30814679

RESUMEN

The re-emergence of Zika virus (ZIKV) in the Western Hemisphere has resulted in global public health crisis since 2015. ZIKV preferentially infects and targets human neural progenitor cells (hNPCs) and causes fetal microcephaly upon maternal infection. hNPCs not only play critical roles during fetal brain development, but also persist in adult brain throughout life. Yet the mechanism of innate antiviral immunity in hNPCs remains largely unknown. Here, we show that ZIKV infection triggers the abundant production of virus-derived small interfering RNAs in hNPCs, but not in the more differentiated progenies or somatic cells. Ablation of key RNAi machinery components significantly enhances ZIKV replication in hNPCs. Furthermore, enoxacin, a broad-spectrum antibiotic that is known as an RNAi enhancer, exerts potent anti-ZIKV activity in hNPCs and other RNAi-competent cells. Strikingly, enoxacin treatment completely prevents ZIKV infection and circumvents ZIKV-induced microcephalic phenotypes in brain organoid models that recapitulate human fetal brain development. Our findings highlight the physiological importance of RNAi-mediated antiviral immunity during the early stage of human brain development, uncovering a novel strategy to combat human congenital viral infections through enhancing RNAi.


Asunto(s)
Encéfalo/inmunología , Células-Madre Neurales/inmunología , Organoides/inmunología , ARN Viral/inmunología , Infección por el Virus Zika/inmunología , Virus Zika/genética , Animales , Antivirales/farmacología , Encéfalo/patología , Línea Celular , Enoxacino/farmacología , Humanos , Inmunidad Innata , Células-Madre Neurales/patología , Organoides/patología , Interferencia de ARN , Replicación Viral , Virus Zika/inmunología , Virus Zika/fisiología
15.
Emerg Microbes Infect ; 7(1): 169, 2018 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-30333476

RESUMEN

Zika virus (ZIKV) has elicited global concern due to its unique biological features, unusual transmission routes, and unexpected clinical outcomes. Although ZIKV transmission through anal intercourse has been reported in humans, it remains unclear if ZIKV is detectable in the stool, if it can infect the host through the anal canal mucosa, and what the pathogenesis of such a route of infection might be in the mouse model. Herein, we demonstrate that ZIKV RNA can be recovered from stools in multiple mouse models, as well as from the stool of a ZIKV patient. Remarkably, intra-anal (i.a.) inoculation with ZIKV leads to efficient infection in both Ifnar1-/- and immunocompetent mice, characterized by extensive viral replication in the blood and multiple organs, including the brain, small intestine, testes, and rectum, as well as robust humoral and innate immune responses. Moreover, i.a. inoculation of ZIKV in pregnant mice resulted in transplacental infection and delayed fetal development. Overall, our results identify the anorectal mucosa as a potential site of ZIKV infection in mice, reveal the associated pathogenesis of i.a. infection, and highlight the complexity of ZIKV transmission through anal intercourse.


Asunto(s)
Heces/virología , Mucosa Intestinal/virología , Recto/virología , Esparcimiento de Virus , Infección por el Virus Zika/virología , Virus Zika/fisiología , Animales , Femenino , Humanos , Inmunidad Innata , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Complicaciones del Embarazo/genética , Complicaciones del Embarazo/inmunología , Complicaciones del Embarazo/virología , Replicación Viral , Virus Zika/genética , Virus Zika/aislamiento & purificación , Infección por el Virus Zika/genética , Infección por el Virus Zika/inmunología
16.
Science ; 358(6365): 933-936, 2017 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-28971967

RESUMEN

Zika virus (ZIKV) has evolved into a global health threat because of its unexpected causal link to microcephaly. Phylogenetic analysis reveals that contemporary epidemic strains have accumulated multiple substitutions from their Asian ancestor. Here we show that a single serine-to-asparagine substitution [Ser139→Asn139 (S139N)] in the viral polyprotein substantially increased ZIKV infectivity in both human and mouse neural progenitor cells (NPCs) and led to more severe microcephaly in the mouse fetus, as well as higher mortality rates in neonatal mice. Evolutionary analysis indicates that the S139N substitution arose before the 2013 outbreak in French Polynesia and has been stably maintained during subsequent spread to the Americas. This functional adaption makes ZIKV more virulent to human NPCs, thus contributing to the increased incidence of microcephaly in recent ZIKV epidemics.


Asunto(s)
Microcefalia/virología , Proteínas del Envoltorio Viral/genética , Infección por el Virus Zika/virología , Virus Zika/genética , Virus Zika/patogenicidad , Américas/epidemiología , Sustitución de Aminoácidos , Animales , Asparagina/genética , Línea Celular Tumoral , Cricetinae , Brotes de Enfermedades , Humanos , Incidencia , Ratones , Microcefalia/epidemiología , Mutación , Células-Madre Neurales/virología , Polinesia/epidemiología , Serina/genética , Infección por el Virus Zika/complicaciones , Infección por el Virus Zika/epidemiología
17.
J Asian Nat Prod Res ; 19(4): 358-387, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28276768

RESUMEN

Some novel josamycin derivatives bearing an arylalkyl-type side chain were designed and synthesized. By HWE or Wittig reaction, 16-aldehyde group of josamycin analogs were converted into unsaturated carbonyl compounds. They were evaluated for their in vitro antibacterial activities against a panel of respiratory pathogens. 8b and 8e exhibited comparable activities against a panel of respiratory pathogens, especially to resistant ones in the series of desmycarosyl josamycin analogs. Among of all the target molecules, 21 showed the best antibacterial activities.


Asunto(s)
Antibacterianos , Josamicina , Cetonas , Antibacterianos/síntesis química , Antibacterianos/química , Antibacterianos/farmacología , Josamicina/análogos & derivados , Josamicina/síntesis química , Josamicina/química , Josamicina/farmacología , Cetonas/síntesis química , Cetonas/química , Cetonas/farmacología , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Staphylococcus aureus/efectos de los fármacos
18.
J Virol ; 90(14): 6538-6548, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27147750

RESUMEN

UNLABELLED: Flaviviruses pose a significant threat to both animals and humans. Recently, a novel flavivirus, duck Tembusu virus (DTMUV), was identified to be the causative agent of a serious duck viral disease in Asia. Its rapid spread, expanding host range, and uncertain transmission routes have raised substantial concerns regarding its potential threats to nonavian hosts, including humans. Here, we demonstrate that DTMUV is not pathogenic for nonhuman primates and is highly sensitive to mammal type I interferon (IFN) signaling. In vitro assays demonstrated that DTMUV infected and replicated efficiently in various mammalian cell lines. Further tests in mice demonstrated high neurovirulence and the age-dependent neuroinvasiveness of the virus. In particular, the inoculation of DTMUV into rhesus monkeys did not result in either viremia or apparent clinical symptoms, although DTMUV-specific humoral immune responses were detected. Furthermore, we revealed that although avian IFN failed to inhibit DTMUV in avian cells, DTMUV was more sensitive to the antiviral effects of type I interferon than other known human-pathogenic flaviviruses. Knockout of the type I IFN receptor in mice caused apparent viremia, viscerotropic disease, and mortality, indicating a vital role of IFN signaling in protection against DTMUV infection. Collectively, we provide direct experimental evidence that this novel avian-origin DTMUV possesses a limited capability to establish infection in immunocompetent primates due to its decreased antagonistic activity in the mammal IFN system. Furthermore, our findings highlight the potential risk of DTMUV infection in immunocompromised individuals and warrant studies on the cross-species transmission and pathogenesis of this novel flavivirus. IMPORTANCE: Mosquito-borne flaviviruses comprise a large group of pathogenic and nonpathogenic members. The pathogenic flaviviruses include dengue, West Nile, and Japanese encephalitis viruses, and the nonpathogenic flaviviruses normally persist in a natural cycle and rarely cause disease in humans. A novel flavivirus, DTMUV (also known as duck egg drop syndrome flavivirus [DEDSV]) was identified in 2012 in ducks and then rapidly spread to several Asian countries. This new flavivirus was then shown to infect multiple avian species, resulting in neurological symptoms with unknown routes of transmission. There is public concern regarding its potential transmission from birds to humans and other nonavian hosts. Our present study shows that the mammalian IFN system can efficiently eliminate DTMUV infection and that the emergence of severe DTMUV-associated disease in mammals, especially humans, is unlikely. Currently, DTMUV infection mostly affects avian species.


Asunto(s)
Antivirales/farmacología , Patos/virología , Infecciones por Flavivirus/tratamiento farmacológico , Flavivirus/patogenicidad , Interferón Tipo I/farmacología , Enfermedades de las Aves de Corral/tratamiento farmacológico , Receptores de Interferón/fisiología , Células A549 , Animales , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Femenino , Infecciones por Flavivirus/inmunología , Infecciones por Flavivirus/virología , Células HeLa , Células Hep G2 , Humanos , Macaca mulatta , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Células Vero
19.
J Gen Virol ; 96(8): 2165-2175, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25908779

RESUMEN

Japanese encephalitis remains the leading cause of viral encephalitis in children in Asia and is expanding its geographical range to larger areas in Asia and Australasia. Five genotypes of Japanese encephalitis virus (JEV) co-circulate in the geographically affected areas. In particular, the emergence of genotype I (GI) JEV has displaced genotype III (GIII) as the dominant circulating genotype in many Asian regions. However, all approved vaccine products are derived from GIII strains. In the present study, bioinformatic analysis revealed that GI and GIII JEV strains shared two distinct amino acid residues within the envelope (E) protein (E222 and E327). By using reverse genetics approaches, A222S and S327T mutations were demonstrated to decrease live-attenuated vaccine (LAV) SA14-14-2-induced neutralizing antibodies in humans, without altering viral replication. A222S or S327T mutations were then rationally engineered into the infectious clone of SA14-14-2, and the resulting mutant strains retained the same genetic stability and attenuation characteristics as the parent strain. More importantly, immunization of mice with LAV-A222S or LAV-S327T elicited increased neutralizing antibodies against GI strains. Together, these results demonstrated that E222 and E327 are potential genotype-related neutralization determinants and are critical in determining the protective efficacy of live Japanese encephalitis vaccine SA14-14-2 against circulating GI strains. Our findings will aid in the rational design of the next generation of Japanese encephalitis LAVs capable of providing broad protection against all JEV strains belonging to different genotypes.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/genética , Encefalitis Japonesa/virología , Vacunas contra la Encefalitis Japonesa/genética , Proteínas del Envoltorio Viral/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Virus de la Encefalitis Japonesa (Especie)/química , Virus de la Encefalitis Japonesa (Especie)/clasificación , Virus de la Encefalitis Japonesa (Especie)/inmunología , Encefalitis Japonesa/inmunología , Femenino , Genotipo , Humanos , Vacunas contra la Encefalitis Japonesa/química , Vacunas contra la Encefalitis Japonesa/inmunología , Masculino , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Filogenia , Alineación de Secuencia , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética
20.
J Basic Microbiol ; 55(1): 22-32, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24652702

RESUMEN

This study investigated the influence of corn straw application on soil microbial communities and the relationship between such communities and soil properties in black soil. The crop used in this study was maize (Zea mays L.). The five treatments consisted of applying a gradient (50, 100, 150, and 200%) of shattered corn straw residue to the soil. Soil samples were taken from May through September during the 2012 maize growing season. The microbial community structure was determined using phospholipid fatty acid (PLFA) analysis. Our results revealed that the application of corn straw influenced the soil properties and increased the soil organic carbon and total nitrogen. Applying corn straw to fields also influenced the variation in soil microbial biomass and community composition, which is consistent with the variations found in soil total nitrogen (TN) and soil respiration (SR). However, the soil carbon-to-nitrogen ratio had no effect on soil microbial communities. The abundance of PLFAs, TN, and SR was higher in C1.5 than those in other treatments, suggesting that the soil properties and soil microbial community composition were affected positively by the application of corn straw to black soil. A Principal Component Analysis indicated that soil microbial communities were different in the straw decomposition processes. Moreover, the soil microbial communities from C1.5 were significantly different from those of CK (p < 0.05). We also found a high ratio of fungal-to-bacterial PLFAs in black soil and significant variations in the ratio of monounsaturated-to-branched fatty acids with different straw treatments that correlated with SR (p < 0.05). These results indicated that the application of corn straw positively influences soil properties and soil microbial communities and that these properties affect these communities. The individual PLFA signatures were sensitive indicators that reflected the changes in the soil environment condition.


Asunto(s)
Consorcios Microbianos , Microbiología del Suelo , Suelo/química , Zea mays , Biomasa , Carbono/análisis , China , Ecosistema , Ácidos Grasos/análisis , Nitrógeno/análisis , Fosfolípidos/análisis , Análisis de Componente Principal , Estaciones del Año , Zea mays/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...