RESUMEN
Macrophages are important target cells for diverse viruses and thus represent a valuable system for studying virus biology. Isolation of primary human macrophages is done by culture of dissociated tissues or from differentiated blood monocytes, but these methods are both time consuming and result in low numbers of recovered macrophages. Here, we explore whether macrophages derived from human induced pluripotent stem cells (iPSCs)-which proliferate indefinitely and potentially provide unlimited starting material-could serve as a faithful model system for studying virus biology. Human iPSC-derived monocytes were differentiated into macrophages and then infected with HIV-1, dengue virus, or influenza virus as model human viruses. We show that iPSC-derived macrophages support the replication of these viruses with kinetics and phenotypes similar to human blood monocyte-derived macrophages. These iPSC-derived macrophages were virtually indistinguishable from human blood monocyte-derived macrophages based on surface marker expression (flow cytometry), transcriptomics (RNA sequencing), and chromatin accessibility profiling. iPSC lines were additionally generated from non-human primate (chimpanzee) fibroblasts. When challenged with dengue virus, human and chimpanzee iPSC-derived macrophages show differential susceptibility to infection, thus providing a valuable resource for studying the species-tropism of viruses. We also show that blood- and iPSC-derived macrophages both restrict influenza virus at a late stage of the virus lifecycle. Collectively, our results substantiate iPSC-derived macrophages as an alternative to blood monocyte-derived macrophages for the study of virus biology. IMPORTANCE: Macrophages have complex relationships with viruses: while macrophages aid in the removal of pathogenic viruses from the body, macrophages are also manipulated by some viruses to serve as vessels for viral replication, dissemination, and long-term persistence. Here, we show that iPSC-derived macrophages are an excellent model that can be exploited in virology.
Asunto(s)
Virus del Dengue , VIH-1 , Células Madre Pluripotentes Inducidas , Macrófagos , Modelos Biológicos , Orthomyxoviridae , Virología , Animales , Humanos , Diferenciación Celular/genética , VIH-1/crecimiento & desarrollo , VIH-1/fisiología , Células Madre Pluripotentes Inducidas/citología , Macrófagos/citología , Macrófagos/metabolismo , Macrófagos/virología , Orthomyxoviridae/crecimiento & desarrollo , Orthomyxoviridae/fisiología , Pan troglodytes , Virus del Dengue/crecimiento & desarrollo , Virus del Dengue/fisiología , Fibroblastos/citología , Monocitos/citología , Replicación Viral , Citometría de Flujo , Perfilación de la Expresión Génica , Ensamble y Desensamble de Cromatina , Tropismo Viral , Virología/métodos , Biomarcadores/análisis , Biomarcadores/metabolismoRESUMEN
The heterotrimeric influenza polymerase (FluPol), comprising subunits PA, PB1 and PB2, binds to the conserved 5' and 3' termini (the 'promoter') of each of the eight single-stranded viral RNA (vRNA) genome segments and performs both transcription and replication of vRNA in the infected cell nucleus. To transcribe viral mRNAs, FluPol associates with cellular RNA polymerase II (Pol II), which enables it to take 5'-capped primers from nascent Pol II transcripts. Here we present a co-crystal structure of bat influenza A polymerase bound to a Pol II C-terminal domain (CTD) peptide mimic, which shows two distinct phosphoserine-5 (SeP5)-binding sites in the polymerase PA subunit, accommodating four CTD heptad repeats overall. Mutagenesis of the SeP5-contacting basic residues (PA K289, R454, K635 and R638) weakens CTD repeat binding in vitro without affecting the intrinsic cap-primed (transcription) or unprimed (replication) RNA synthesis activity of recombinant polymerase, whereas in cell-based minigenome assays the same mutations substantially reduce overall polymerase activity. Only recombinant viruses with a single mutation in one of the SeP5-binding sites can be rescued, but these viruses are severely attenuated and genetically unstable. Several previously described mutants that modulate virulence can be rationalized by our results, including a second site mutation (PA(C453R)) that enables the highly attenuated mutant virus (PA(R638A)) to revert to near wild-type infectivity. We conclude that direct binding of FluPol to the SeP5 Pol II CTD is fine-tuned to allow efficient viral transcription and propose that the CTD-binding site on FluPol could be targeted for antiviral drug development.
Asunto(s)
Quirópteros/virología , Orthomyxoviridae/enzimología , ARN Polimerasa II/química , ARN Polimerasa II/metabolismo , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/metabolismo , Secuencia de Aminoácidos , Animales , Antivirales/farmacología , Sitios de Unión/efectos de los fármacos , Sitios de Unión/genética , Cristalografía por Rayos X , Virus de la Influenza A/enzimología , Virus de la Influenza B/enzimología , Modelos Moleculares , Terapia Molecular Dirigida , Mutación , Orthomyxoviridae/genética , Orthomyxoviridae/crecimiento & desarrollo , Orthomyxoviridae/patogenicidad , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/enzimología , Infecciones por Orthomyxoviridae/virología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fosfoserina/metabolismo , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Subunidades de Proteína , ARN Polimerasa Dependiente del ARN/genética , Virulencia/genética , Replicación ViralRESUMEN
Based on our previous studies, we show that the M gene is critical for the replication and pathogenicity of the chimeric H17 bat influenza virus (Bat09:mH1mN1) by replacing the bat M gene with those from human and swine influenza A viruses. However, the key amino acids of the M1 and/or M2 proteins that are responsible for virus replication and pathogenicity remain unknown. In this study, replacement of the PR8 M gene with the Eurasian avian-like M gene from the A/California/04/2009 pandemic H1N1 virus significantly decreased viral replication in both mammalian and avian cells in the background of the chimeric H17 bat influenza virus. Further studies revealed that M1 was more crucial for viral growth and pathogenicity than M2 and that the amino acid residues M1-41V and M2-27A were responsible for these characteristics in cells and in mice. These key residues of the M1 and M2 proteins identified in this study might be important for influenza virus surveillance and could be used to produce live attenuated vaccines in the future. IMPORTANCE The M1 and M2 proteins influence the morphology, replication, virulence, and transmissibility of influenza viruses. Although a few key residues in the M1 and M2 proteins have been identified, whether other residues of the M1 and M2 proteins are involved in viral replication and pathogenicity remains to be discovered. In the background of the chimeric H17 bat influenza virus, the Eurasian avian-like M gene from the A/California/04/2009 virus significantly decreased viral growth in mammalian and avian cells. Further study showed that M1 was implicated more than M2 in viral growth and pathogenicity in vitro and in vivo and that the key amino acid residues M1-41V and M2-27A were responsible for these characteristics in cells and in mice. These key residues of the M1 and M2 proteins could be used for influenza virus surveillance and live attenuated vaccine applications in the future. These findings provide important contributions to knowledge of the genetic basis of the virulence of influenza viruses.
Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/genética , Infecciones por Orthomyxoviridae/virología , Orthomyxoviridae/crecimiento & desarrollo , Orthomyxoviridae/patogenicidad , Proteínas de la Matriz Viral/metabolismo , Aminoácidos/metabolismo , Animales , Línea Celular , Quirópteros , Genes Virales , Humanos , Pulmón/virología , Ratones , Orthomyxoviridae/genética , Virus Reordenados/genética , Virus Reordenados/crecimiento & desarrollo , Virus Reordenados/patogenicidad , Cornetes Nasales/virología , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/genética , Virulencia , Replicación ViralRESUMEN
To characterize bat influenza H18N11 virus, we propagated a reverse genetics-generated H18N11 virus in Madin-Darby canine kidney subclone II cells and detected two mammal-adapting mutations in the neuraminidase (NA)-like protein (NA-F144C and NA-T342A, N2 numbering) that increased the virus titers in three mammalian cell lines (i.e., Madin-Darby canine kidney, Madin-Darby canine kidney subclone II, and human lung adenocarcinoma [Calu-3] cells). In mice, wild-type H18N11 virus replicated only in the lungs of the infected animals, whereas the NA-T342A and NA-F144C/T342A mutant viruses were detected in the nasal turbinates, in addition to the lungs. Bat influenza viruses have not been tested for their virulence or organ tropism in ferrets. We detected wild-type and single mutant viruses each possessing NA-F144C or NA-T342A in the nasal turbinates of one or several infected ferrets, respectively. A mutant virus possessing both the NA-F144C and NA-T342A mutations was isolated from both the lung and the trachea, suggesting that it has a broader organ tropism than the wild-type virus. However, none of the H18N11 viruses caused symptoms in mice or ferrets. The NA-F144C/T342A double mutation did not substantially affect virion morphology or the release of virions from cells. Collectively, our data demonstrate that the propagation of bat influenza H18N11 virus in mammalian cells can result in mammal-adapting mutations that may increase the replicative ability and/or organ tropism of the virus; overall, however, these viruses did not replicate to high titers throughout the respiratory tract of mice and ferrets.IMPORTANCE Bats are reservoirs for several severe zoonotic pathogens. The genomes of influenza A viruses of the H17N10 and H18N11 subtypes have been identified in bats, but no live virus has been isolated. The characterization of artificially generated bat influenza H18N11 virus in mammalian cell lines and animal models revealed that this virus can acquire mammal-adapting mutations that may increase its zoonotic potential; however, the wild-type and mutant viruses did not replicate to high titers in all infected animals.
Asunto(s)
Quirópteros/virología , Mutación , Neuraminidasa/genética , Neuraminidasa/metabolismo , Orthomyxoviridae/enzimología , Orthomyxoviridae/genética , Replicación Viral/fisiología , Animales , Línea Celular , Modelos Animales de Enfermedad , Femenino , Hurones/virología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Neuraminidasa/química , Orthomyxoviridae/crecimiento & desarrollo , Infecciones por Orthomyxoviridae/veterinaria , Infecciones por Orthomyxoviridae/virología , Tráquea/virología , Zoonosis/virologíaRESUMEN
The correlation of viral growth capability (n = 156) with the viral load in nasopharyngeal swabs (n = 76) was assessed. Epidemic influenza A/H1N1, A/H3N2, and B viruses showed a wide range of growth capability (104-1011 copies/mL) in Madin-Darby canine kidney cells. The growth was correlated with the nasopharyngeal viral load (r = 0.53). Six selected strains showed growth-dependent cell death (r = 0.96) in a growth kinetics assay. Epidemic influenza viruses exhibit a wide range of growth capability. Growth capability should be considered one of the key factors in disease prognosis.
Asunto(s)
Epidemias , Gripe Humana/epidemiología , Gripe Humana/virología , Orthomyxoviridae/crecimiento & desarrollo , Células A549 , Animales , Supervivencia Celular , Perros , Humanos , Japón/epidemiología , Células de Riñón Canino Madin Darby , Nasofaringe/virología , Orthomyxoviridae/clasificación , Orthomyxoviridae/genética , Orthomyxoviridae/aislamiento & purificación , Pronóstico , ARN Viral/análisis , Carga ViralRESUMEN
Carbohydrates play important and diverse roles in the fundamental processes of life. We have established a method for accurately and a large-scale synthesis of functional carbohydrates with diverse properties using a unique enzymatic method. Furthermore, various artificial glycan-conjugated molecules have been developed by adding these synthetic carbohydrates to macromolecules and to middle- and low-molecular-weight molecules with different properties. These glycan-conjugated molecules have biological activities comparable to or higher than those of natural compounds and present unique functions. In this review, several synthetic glycan-conjugated molecules are taken as examples to show design, synthesis, and function.
Asunto(s)
Antivirales/síntesis química , Factores Biológicos/síntesis química , Glicoconjugados/síntesis química , Polisacáridos/química , Sialoglicoproteínas/síntesis química , Animales , Antivirales/farmacología , Factores Biológicos/farmacología , Conformación de Carbohidratos , Quimioinformática/métodos , Reactivos de Enlaces Cruzados/química , Diseño de Fármacos , Glicoconjugados/farmacología , Glicósidos/química , Glicosilación , Humanos , Nanopartículas/química , Orthomyxoviridae/efectos de los fármacos , Orthomyxoviridae/crecimiento & desarrollo , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/virología , Sialoglicoproteínas/farmacologíaRESUMEN
The establishment of long-lived pathogen-specific T cells is a fundamental property of the adaptive immune response. However, the mechanisms underlying long-term persistence of antigen-specific CD4+ T cells are not well-defined. Here we identify a subset of memory CD4+ T cells capable of effluxing cellular toxins, including rhodamine (Rho), through the multidrug efflux protein MDR1 (also known as P-glycoprotein and ABCB1). Drug-effluxing CD4+ T cells were characterized as CD161+CD95+CD45RA-CD127hiCD28+CD25int cells with a distinct chemokine profile and a Th1-polarized pro-inflammatory phenotype. CD4+CD161+Rho-effluxing T cells proliferated vigorously in response to stimulation with anti-CD3/CD28 beads and gave rise to CD161- progeny in vitro. These cells were also capable of self-renewal and maintained their phenotypic and functional characteristics when cultured with homeostatic cytokines. Multidrug-effluxing CD4+CD161+ T cells were enriched within the viral-specific Th1 repertoire of healthy donors and patients with acute myeloid leukemia (AML) and survived exposure to daunorubicin chemotherapy in vitro. Multidrug-effluxing CD4+CD161+ T cells also resisted chemotherapy-induced cytotoxicity in vivo and underwent significant expansion in AML patients rendered lymphopenic after chemotherapy, contributing to the repopulation of anti-CMV immunity. Finally, after influenza vaccination, the proportion of influenza-specific CD4+ T cells coexpressing CD161 was significantly higher after 2 years compared with 4 weeks after immunization, suggesting CD161 is a marker for long-lived antigen-specific memory T cells. These findings suggest that CD4+CD161+ T cells with rapid efflux capacity contribute to the maintenance of viral-specific memory T cells. These data provide novel insights into mechanisms that preserve antiviral immunity in patients undergoing chemotherapy and have implications for the development of novel immunotherapeutic approaches.
Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Regulación Leucémica de la Expresión Génica , Memoria Inmunológica , Gripe Humana/prevención & control , Leucemia Mieloide Aguda/inmunología , Subfamilia B de Receptores Similares a Lectina de Células NK/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/inmunología , Antibióticos Antineoplásicos/farmacología , Anticuerpos/farmacología , Transporte Biológico , Antígenos CD4/genética , Antígenos CD4/inmunología , Linfocitos T CD4-Positivos/clasificación , Linfocitos T CD4-Positivos/patología , Linfocitos T CD4-Positivos/virología , Citomegalovirus/efectos de los fármacos , Citomegalovirus/crecimiento & desarrollo , Citomegalovirus/inmunología , Daunorrubicina/farmacología , Resistencia a Antineoplásicos/genética , Humanos , Inmunofenotipificación , Vacunas contra la Influenza/administración & dosificación , Gripe Humana/inmunología , Gripe Humana/virología , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/virología , Subfamilia B de Receptores Similares a Lectina de Células NK/inmunología , Orthomyxoviridae/efectos de los fármacos , Orthomyxoviridae/crecimiento & desarrollo , Orthomyxoviridae/inmunología , Rodaminas/metabolismo , Rodaminas/farmacología , Transducción de Señal , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células TH1/patologíaRESUMEN
At every step of their replication cycle influenza viruses depend heavily on their host cells. The multifaceted interactions that occur between the virus and its host cell determine the outcome of the infection, including efficiency of progeny virus production, tropism, and pathogenicity. In order to understand viral disease and develop therapies for influenza it is therefore pertinent to study the intricate interplay between influenza viruses and their required host factors. Here, we review the current knowledge on host cell factors required by influenza virus at the different stages of the viral replication cycle. We also discuss the roles of host factors in zoonotic transmission of influenza viruses and their potential for developing novel antivirals.
Asunto(s)
Interacciones Huésped-Patógeno/genética , Gripe Humana/genética , Gripe Humana/virología , Orthomyxoviridae/crecimiento & desarrollo , Replicación Viral , Humanos , Gripe Humana/tratamiento farmacológico , Orthomyxoviridae/patogenicidad , VirulenciaRESUMEN
RNA secondary structures play a key role in splicing, gene expression, microRNA biogenesis, RNA editing, and other biological processes. The importance of RNA structures has been demonstrated in the life cycle of RNA-containing viruses, including the influenza virus. At least two regions of conserved secondary structure in NS segment (+) RNA are predicted to vary among influenza virus strains with respect to thermodynamic stability; both fall in the NS1 open reading frame. The NS1 protein is involved in multiple virus-host interaction processes, and its main function is to inhibit the cellular immune response to viral infection. Using a reverse genetics approach, four influenza virus strains were constructed featuring mutations that have different effects on RNA secondary structure. Growth curve experiments and ELISA data show that, at least in the first viral replication cycle, mutations G123A and A132G affecting RNA structure in the (82-148) NS RNA region influence NS1 protein expression.
Asunto(s)
Expresión Génica , Conformación de Ácido Nucleico , Orthomyxoviridae/crecimiento & desarrollo , ARN Viral/química , Proteínas no Estructurales Virales/biosíntesis , Animales , Perros , Células de Riñón Canino Madin Darby , Mutagénesis Sitio-Dirigida , Orthomyxoviridae/genética , Mutación Puntual , ARN Viral/metabolismo , Genética Inversa , Replicación ViralRESUMEN
Influenza infection is common worldwide with many individuals affected each year during epidemics and occasionally pandemics. Previous studies in animal models and a few human cases have established an important role of innate type I and III interferon (IFN) for viral elimination and mounting of antiviral responses. However, genetic and immunological determinants of very severe disseminated influenza virus infection in humans remain incompletely understood. Here, we describe an adult patient with severe influenza virus A (IAV) infection, in whom we identified a rare variant E331V in IFN regulatory factor (IRF)7 by whole-exome sequencing. Examination of patient cells demonstrated a cellular phenotype suggesting functional IRF7 impairment, since priming with IFN was almost abolished and IFN responses to IAV were significantly impaired in patient cells. Moreover, IAV replication was significantly higher in patient cells than in controls. Finally, expression of IRF7 E331V in HEK293 cells demonstrated significantly reduced activation of both IFNA7 and IFNB promoters in a luciferase reporter gene expression assay compared to IRF7 wild type. These findings provide further support for the essential role of IRF7 in amplifying antiviral IFN responses to ensure potent and sustained IFN responses during influenza virus infection in humans.
Asunto(s)
Inmunidad Innata , Factores Inmunológicos/metabolismo , Gripe Humana/inmunología , Gripe Humana/patología , Factor 7 Regulador del Interferón/genética , Interferones/metabolismo , Mutación Missense , Adulto , Células HEK293 , Humanos , Factor 7 Regulador del Interferón/metabolismo , Interferón-alfa/biosíntesis , Masculino , Persona de Mediana Edad , Orthomyxoviridae/crecimiento & desarrollo , Replicación Viral , Secuenciación Completa del GenomaRESUMEN
In this review, we summarize the mechanisms by which sphingolipids modulate virus multiplication and the host innate immune response, using a number of host-virus systems as illustrative models. Sphingolipids exert diverse functions, both at the level of the viral life cycle and in the regulation of antiviral immune responses. Sphingolipids may influence viral replication in three ways: by serving as (co)receptors during viral entry, by modulating virus replication, and by shaping the antiviral immune response. Several studies have demonstrated that sphingosine kinases (SphK) and their product, sphingosine-1-phosphate (S1P), enhance the replication of influenza, measles, and hepatitis B virus (HBV). In contrast, ceramides, particularly S1P and SphK1, influence the expression of type I interferon (IFN-I) by modulating upstream antiviral signaling and enhancing dendritic cell maturation, differentiation, and positioning in tissue. The synthetic molecule α-galactosylceramide has also been shown to stimulate natural killer cell activation and interferon (IFN)-γ secretion. However, to date, clinical trials have failed to demonstrate any clinical benefit for sphingolipids in the treatment of cancer or HBV infection. Taken together, these findings show that sphingolipids play an important and underappreciated role in the control of virus replication and the innate immune response.
Asunto(s)
Inmunidad Innata/inmunología , Esfingolípidos/inmunología , Esfingolípidos/metabolismo , Replicación Viral , Animales , Virus de la Hepatitis B/crecimiento & desarrollo , Virus de la Hepatitis B/inmunología , Humanos , Virus del Sarampión/crecimiento & desarrollo , Virus del Sarampión/inmunología , Orthomyxoviridae/crecimiento & desarrollo , Orthomyxoviridae/inmunologíaRESUMEN
The infectious salmon anemia virus (ISAV) is an aquatic pathogen that is a member of the Orthomyxoviridae family with lethal hemorrhagic potential. Although it affects other species of salmonid fish, ISAV only causes disease in Atlantic salmon (Salmo salar) specimens in sea water. In spite of the fact that the virus has been described as enveloped with icosahedral symmetry, viral like particles with anomalous morphology have been observed in field samples, this we have not been able to recover then in adequate quantities for full demonstration. We report a procedure to concentrate and recover these novel forms of the virus, comparing two cell lines from different origins, demonstrating that these forms were preferentially expressed in cells of epithelial origin.
Asunto(s)
Células Epiteliales/virología , Isavirus/clasificación , Isavirus/aislamiento & purificación , Infecciones por Orthomyxoviridae/veterinaria , Infecciones por Orthomyxoviridae/virología , Salmo salar/virología , Animales , Línea Celular , Enfermedades de los Peces/virología , Isavirus/crecimiento & desarrollo , Isavirus/patogenicidad , Microscopía Electrónica , Orthomyxoviridae/clasificación , Orthomyxoviridae/crecimiento & desarrollo , Orthomyxoviridae/aislamiento & purificación , Orthomyxoviridae/patogenicidad , Infecciones por Orthomyxoviridae/patología , ARN Viral/análisis , ARN Viral/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/veterinaria , Agua de Mar , Cultivo de VirusRESUMEN
Large scale continuous cell-line cultures promise greater reproducibility and efficacy for the production of influenza vaccines, and adenovirus for gene therapy. This paper seeks to use an existing validated ultra scale-down tool, which is designed to mimic the commercial scale process environment using only milliliters of material, to provide some initial insight into the performance of the harvest step for these processes. The performance of industrial scale centrifugation and subsequent downstream process units is significantly affected by shear. The properties of these cells, in particular their shear sensitivity, may be changed considerably by production of a viral product, but literature on this is limited to date. In addition, the scale-down tool used here has not previously been applied to the clarification of virus production processes. The results indicate that virus infected cells do not actually show any increase in sensitivity to shear, and may indeed become less shear sensitive, in a similar manner to that previously observed in old or dead cell cultures. Clarification may be most significantly dependent on the virus release mechanism, with the budding influenza virus producing a much greater decrease in clarification than the lytic, non-enveloped adenovirus. A good match was also demonstrated to the industrial scale performance in terms of clarification, protein release, and impurity profile.
Asunto(s)
Centrifugación/métodos , Tecnología Farmacéutica/métodos , Vacunas Virales/aislamiento & purificación , Cultivo de Virus/métodos , Adenoviridae/crecimiento & desarrollo , Supervivencia Celular , Orthomyxoviridae/crecimiento & desarrolloRESUMEN
Despite all the advantages that cell-cultured influenza vaccines have over egg-based influenza vaccines, the inferior productivity of cell-culture systems is a major drawback that must be addressed. BST-2 (tetherin) is a host restriction factor which inhibits budding-out of various enveloped viruses from infected host cells. We developed BST-2-deficient MDCK and Vero cell lines to increase influenza virus release in cell culture. BST-2 gene knock-out resulted in increased release of viral particles into the culture medium, by at least 2-fold and up to 50-fold compared to release from wild-type counterpart cells depending on cell line and virus type. The effect was not influenza virus/MDCK/Vero-specific, but was also present in a broad range of host cells and virus families; we observed similar results in murine, human, canine, and monkey cell lines with viruses including MHV-68 (Herpesviridae), influenza A virus (Orthomyxoviridae), porcine epidemic diarrhea virus (Coronaviridae), and vaccinia virus (Poxviridae). Our results suggest that the elimination of BST-2 expression in virus-producing cell lines can enhance the production of viral vaccines. Biotechnol. Bioeng.2017;114: 2289-2297. © 2017 Wiley Periodicals, Inc.
Asunto(s)
Antígenos CD/genética , Mejoramiento Genético/métodos , Vacunas contra la Influenza/biosíntesis , Orthomyxoviridae/crecimiento & desarrollo , Orthomyxoviridae/aislamiento & purificación , Virión/aislamiento & purificación , Virión/metabolismo , Animales , Chlorocebus aethiops , Perros , Proteínas Ligadas a GPI/genética , Técnicas de Silenciamiento del Gen , Vacunas contra la Influenza/aislamiento & purificación , Células de Riñón Canino Madin Darby , Ingeniería Metabólica/métodos , Orthomyxoviridae/genética , Células Vero , Virión/genéticaRESUMEN
The yields of egg-grown influenza vaccines are maximized by the production of a seed strain using a reassortment of the seasonal influenza virus isolate with a highly egg-adapted strain. The seed virus is selected based on high yields of viral hemagglutinin (HA) and expression of the surface antigens from the seasonal isolate. The remaining proteins are usually derived from the high-growth parent. However, a retrospective analysis of vaccine seeds revealed that the seasonal PB1 gene was selected in more than 50% of reassortment events. Using the model seasonal H3N2 virus A/Udorn/307/72 (Udorn) virus and the high-growth A/Puerto Rico/8/34 (PR8) virus, we assessed the influence of the source of the PB1 gene on virus growth and vaccine yield. Classical reassortment of these two strains led to the selection of viruses that predominantly had the Udorn PB1 gene. The presence of Udorn PB1 in the seed virus, however, did not result in higher yields of virus or HA compared to the yields in the corresponding seed virus with PR8 PB1. The 8-fold-fewer virions produced with the seed virus containing the Udorn PB1 were somewhat compensated for by a 4-fold increase in HA per virion. A higher HA/nucleoprotein (NP) ratio was found in past vaccine preparations when the seasonal PB1 was present, also indicative of a higher HA density in these vaccine viruses. As the HA viral RNA (vRNA) and mRNA levels in infected cells were similar, we propose that PB1 selectively alters the translation of viral mRNA. This study helps to explain the variability of vaccine seeds with respect to HA yield.
Asunto(s)
Hemaglutininas/análisis , Vacunas contra la Influenza/química , Orthomyxoviridae/química , Virus Reordenados/química , Proteínas Virales/metabolismo , Animales , Hemaglutininas/inmunología , Humanos , Vacunas contra la Influenza/inmunología , Orthomyxoviridae/crecimiento & desarrollo , Orthomyxoviridae/inmunología , Virus Reordenados/crecimiento & desarrollo , Virus Reordenados/inmunología , Tecnología Farmacéutica/métodosRESUMEN
Study of effectiveness of CaCo-2 cell line for influenza virus isolation was carried out. It was shown that the properties of this cell line strongly depended on the source of its origin and cultivation conditions. The infectious activity of the influenza viruses on CaCo-2 cell line was virtually the same as in the MDCK cell line. The rate of the viral isolation was virtually identical for both cell lines tested, but viruses from post-mortem materials were isolated only in CaCo-2 cell line. In general, the CaCo-2 line is believed to be a valuable cell line for virological research, particularly for influenza virus isolation.
Asunto(s)
Gripe Humana/virología , Orthomyxoviridae/aislamiento & purificación , Replicación Viral/genética , Animales , Células CACO-2 , Perros , Humanos , Células de Riñón Canino Madin Darby , Orthomyxoviridae/crecimiento & desarrolloRESUMEN
Forced by major drawbacks of egg-based influenza virus production, several studies focused on the establishment and optimization of cell-based production systems. Among numerous possible host cell lines from duck, monkey, canine, chicken, mouse, and human origin, only a few will meet regulatory requirements, accomplish industrial standards, and result in high virus titers. From primary virus isolation up to large-scale manufacturing of human vaccines, however, the most logical choice seems to be the use of human cell lines. For this reason, we evaluated the recently established CAP cell line derived from human amniocytes for its potential in influenza virus production in suspension culture in small scale shaker flask and stirred tank bioreactor experiments. Different human and animal influenza viruses could be adapted to produce hemagglutination (HA) titers of at least 2.0 log(10) HA units/100 µL without further process optimization. Adjusting trypsin activity as well as infection conditions (multiplicity of infection, infection medium) resulted in HA titers of up to 3.2 log(10) HA units/100 µL and maximum cell-specific virus productivities of 6,400 virions/cell (for human influenza A/PR/8/34 as a reference). Surface membrane expression of sialyloligosaccharides as well as HA N-glycosylation patterns were characterized. Overall, experimental results clearly demonstrate the potential of CAP cells for achieving high virus yields for different influenza strains and the option to introduce a highly attractive fully characterized human cell line compliant with regulatory and industrial requirements as an alternative for influenza virus vaccine production.
Asunto(s)
Vacunas contra la Influenza/aislamiento & purificación , Orthomyxoviridae/crecimiento & desarrollo , Tecnología Farmacéutica/métodos , Línea Celular , Glicosilación , Glicoproteínas Hemaglutininas del Virus de la Influenza/análisis , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Humanos , Cultivo de Virus/métodosRESUMEN
Emergence of resistance is a major concern in influenza antiviral treatment and prophylaxis. Combination antiviral therapy might overcome this problem. Here, we estimate that all possible single mutants and a sizeable fraction of double mutants are generated during an uncomplicated influenza infection. While most of them may sustain a fitness cost, some variants may confer drug resistance and be selected during therapy. We argue that a triple combination regimen would markedly reduce the risk of antiviral resistance emergence in seasonal and pandemic influenza viruses, especially in seriously ill or immunocompromised hosts.
Asunto(s)
Antivirales/administración & dosificación , Farmacorresistencia Viral , Gripe Humana/tratamiento farmacológico , Gripe Humana/virología , Mutación , Orthomyxoviridae/efectos de los fármacos , Adulto , Quimioprevención/métodos , Quimioterapia Combinada/métodos , Humanos , Modelos Teóricos , Orthomyxoviridae/genética , Orthomyxoviridae/crecimiento & desarrollo , Selección GenéticaRESUMEN
BACKGROUND: The influenza virus is still one of the most important respiratory risks affecting humans which require effective treatments. In this case, traditional medications are of interest. HESA-A is an active natural biological compound from herbal-marine origin. Previous studies have reported that the therapeutic properties of HESA-A are able to treat psoriasis vulgaris and cancers. However, no antiviral properties have been reported. METHODS: This study was designed to investigate the potential antiviral properties of HESA-A and its effects in modulating TNF-α and IL-6 cytokine levels. HESA-A was prepared in normal saline as a stock solution (0.8 mg/ml, pH = 7.4). Percentages of cell survival when exposed to different concentrations of HESA-A at different time intervals was determined by MTT assay. To study the potential antiviral activity of HESA-A, Madin-Darby Canine Kidney (MDCK) cells were treated with the effective concentration (EC50) of HESA-A (0.025 mg/ml) and 100 TCID50/0.1 ml of virus sample under different types of exposure. RESULTS: Based on the MTT method and hemagglutination assay (HA), HESA-A is capable of improving cell viability to 31% and decreasing HA titre to almost 99% in co-penetration exposures. In addition, based on quantitative real-time PCR (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA), it was found that HESA-A causes decrements in TNF-α and IL-6 cytokine expressions, which was significant for TNF-α (p ≤ 0.05) but not for IL-6. CONCLUSION: In conclusion, HESA-A was effective against influenza infection through suppressing cytokine expression.
Asunto(s)
Antivirales/farmacología , Orthomyxoviridae/efectos de los fármacos , Orthomyxoviridae/patogenicidad , Preparaciones de Plantas/farmacología , Animales , Línea Celular , Supervivencia Celular , Perros , Ensayo de Inmunoadsorción Enzimática , Perfilación de la Expresión Génica , Factores Inmunológicos/farmacología , Interleucina-6/metabolismo , Orthomyxoviridae/crecimiento & desarrollo , Reacción en Cadena en Tiempo Real de la Polimerasa , Sales de Tetrazolio/metabolismo , Tiazoles/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Carga Viral , VirulenciaRESUMEN
Trypsin is commonly used in Madin-Darby canine kidney (MDCK) cell culture-based influenza vaccine production to facilitate virus infection by proteolytic activation of viral haemagglutinin, which enables multi-cycle replication. In this study, we were able to demonstrate that trypsin also interferes with pathogen defence mechanisms of host cells. In particular, a trypsin concentration of 5 BAEE U/mL (4.5 µg/mL porcine trypsin) used in vaccine manufacturing strongly inhibited interferon (IFN) signalling by proteolytic degradation of secreted IFN. Consequently, absence of trypsin during infection resulted in a considerably stronger induction of IFN signalling and apoptosis, which significantly reduced virus yields. Under this condition, multi-cycle virus replication in MDCK cells was not prevented but clearly delayed. Therefore, incomplete infection can be ruled out as the reason for the lower virus titres. However, suppression of IFN signalling by overexpression of viral IFN antagonists (influenza virus PR8-NS1, rabies virus phosphoprotein) partially rescued virus titres in the absence of trypsin. In addition, virus yields could be almost restored by using the influenza strain A/WSN/33 in combination with fetal calf serum (FCS). For this strain, FCS enabled trypsin-independent fast propagation of virus infection, probably outrunning cellular defence mechanisms and apoptosis induction in the absence of trypsin. Overall, addition of trypsin provided optimal conditions for high yield vaccine production in MDCK cells by two means. On the one hand, proteolytic degradation of IFN keeps cellular defence at a low level. On the other hand, enhanced virus spreading enables viruses to replicate before the cellular response becomes fully activated.