Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 365
Filtrar
1.
Molecules ; 25(7)2020 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-32260270

RESUMEN

Different parts of Nuphar lutea L. (yellow water lily) have been used to treat several inflammatory and pathogen-related diseases. It has shown that Nuphar lutea extracts (NUP) are active against various pathogens including bacteria, fungi, and leishmanial parasites. In an effort to detect novel therapeutic agents against negative-stranded RNA (- RNA) viruses, we have tested the effect of a partially-purified alkaloid mixture of Nuphar lutea leaves on the measles virus (MV). The MV vaccine's Edmonston strain was used to acutely or persistently infect cells. The levels of several MV proteins were detected by a Western blot and immunocytochemistry. Viral RNAs were quantitated by qRT-PCR. Virus infectivity was monitored by infecting African green monkey kidney VERO cells' monolayers. We showed that NUP protected cells from acute infection. Decreases in the MV P-, N-, and V-proteins were observed in persistently infected cells and the amount of infective virus released was reduced as compared to untreated cells. By examining viral RNAs, we suggest that NUP acts at the post-transcriptional level. We conclude, as a proof of concept, that NUP has anti-viral therapeutic activity against the MV. Future studies will determine the mechanism of action and the effect of NUP on other related viruses.


Asunto(s)
Alcaloides/farmacología , Antivirales/farmacología , Virus del Sarampión/crecimiento & desarrollo , Nuphar/química , Alcaloides/química , Animales , Antivirales/química , Chlorocebus aethiops , Regulación Viral de la Expresión Génica/efectos de los fármacos , Virus del Sarampión/efectos de los fármacos , Virus del Sarampión/genética , Extractos Vegetales/química , Prueba de Estudio Conceptual , ARN Viral/efectos de los fármacos , Células Vero , Proteínas Virales/efectos de los fármacos , Proteínas Virales/metabolismo
2.
J Gen Virol ; 101(4): 399-409, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32053093

RESUMEN

Oncolytic virotherapy is an emerging treatment option for numerous cancers, with several virus families currently being evaluated in clinical trials. More specifically, vaccine-strain measles virus has arisen as a promising candidate for the treatment of different tumour types in several early clinical trials. Replicating viruses, and especially RNA viruses without proofreading polymerases, can rapidly adapt to varying environments by selecting quasispecies with advantageous genetic mutations. Subsequently, these genetic alterations could potentially weaken the safety profile of virotherapy. In this study, we demonstrate that, following an extended period of virus replication in producer or cancer cell lines, the quasispecies consensus sequence of vaccine strain-derived measles virus accrues a remarkably small number of mutations throughout the nonsegmented negative-stranded RNA genome. Interestingly, we detected a nonrandom distribution of genetic alterations within the genome, with an overall decreasing frequency of mutations from the 3' genome start to its 5' end. Comparing the serially passaged viruses to the parental virus on producer cells, we found that the acquired consensus mutations did not drastically change viral replication kinetics or cytolytic potency. Collectively, our data corroborate the genomic stability and excellent safety profile of oncolytic measles virus, thus supporting its continued development and clinical translation as a promising viro-immunotherapeutic.


Asunto(s)
Inestabilidad Genómica , Virus del Sarampión/genética , Cuasiespecies/genética , Animales , Línea Celular Tumoral , Supervivencia Celular , Chlorocebus aethiops , Humanos , Virus del Sarampión/crecimiento & desarrollo , Mutación , Viroterapia Oncolítica , Pase Seriado , Células Vero , Virulencia/genética
3.
Methods Mol Biol ; 2095: 335-364, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31858478

RESUMEN

The production of biopharmaceuticals in cell culture involves stringent controls to ensure product safety and quality. To meet these requirements, quality by design principles must be applied during the development of cell culture processes so that quality is built into the product by understanding the manufacturing process. One key aspect is process analytical technology, in which comprehensive online monitoring is used to identify and control critical process parameters that affect critical quality attributes such as the product titer and purity. The application of industry-ready technologies such as turbidimetry and dielectric spectroscopy provides a deeper understanding of biological processes within the bioreactor and allows the physiological status of the cells to be monitored on a continuous basis. This in turn enables selective and targeted process controls to respond in an appropriate manner to process disturbances. This chapter outlines the principles of online dielectric spectroscopy and turbidimetry for the measurement of optical density as applied to mammalian and insect cells cultivated in stirred-tank bioreactors either in suspension or as adherent cells on microcarriers.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Espectroscopía Dieléctrica/métodos , Nefelometría y Turbidimetría/métodos , Animales , Reactores Biológicos , Técnicas de Cultivo de Célula/instrumentación , Chlorocebus aethiops , Espectroscopía Dieléctrica/instrumentación , Drosophila melanogaster , Virus del Sarampión/crecimiento & desarrollo , Virus del Sarampión/aislamiento & purificación , Nefelometría y Turbidimetría/instrumentación , Proteínas Recombinantes/metabolismo , Células Vero
4.
Arch Virol ; 164(2): 439-446, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30390151

RESUMEN

Measles virus (MeV) first infects immune cells in the respiratory tract of a human host, spreads to lymphoid organs throughout the body, and finally enters and grows in respiratory epithelial cells before being released and transmitted to the next host. Thus, efficient growth in respiratory epithelial cells is important for the person-to-person transmission of MeV. Upon viral entry, host cells detect viral nucleic acids and produce interferons (IFNs) to control viral growth. Type I (IFN-α/ß) and type III (IFN-λ) IFNs have largely common induction and signaling mechanisms and stimulate expression of similar target genes but utilize distinct receptors. To determine the relative contributions of type I and type III IFNs to the control of MeV growth in epithelial cells, we examined the growth of MeV and that of its mutants lacking either type I or type III IFN receptor in the human lung epithelial cell line H358. Our results revealed that both type I and type III IFNs are required to restrict MeV growth in H358 cells and that the induction of type III as well as type I IFNs was increased in the absence of the MeV nonstructural V protein.


Asunto(s)
Células Epiteliales/inmunología , Interferón Tipo I/inmunología , Interferones/inmunología , Virus del Sarampión/crecimiento & desarrollo , Sarampión/inmunología , Línea Celular , Células Epiteliales/virología , Humanos , Interferón Tipo I/genética , Interferones/genética , Pulmón/citología , Pulmón/inmunología , Pulmón/virología , Sarampión/genética , Sarampión/virología , Virus del Sarampión/genética , Virus del Sarampión/fisiología , Interferón lambda
5.
Biol Chem ; 399(10): 1115-1123, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-29975662

RESUMEN

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/inmunología
6.
Biotechnol Bioeng ; 115(5): 1186-1194, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29288575

RESUMEN

Oncolytic viruses offer new hope to millions of patients with incurable cancer. One promising class of oncolytic viruses is Measles virus, but its broad administration to cancer patients is currently hampered by the inability to produce the large amounts of virus needed for treatment (1010 -1012 virus particles per dose). Measles virus is unstable, leading to very low virus titers during production. The time of infection and time of harvest are therefore critical parameters in a Measles virus production process, and their optimization requires an accurate online monitoring system. We integrated a probe based on dielectric spectroscopy (DS) into a stirred tank reactor to characterize the Measles virus production process in adherent growing Vero cells. We found that DS could be used to monitor cell adhesion on the microcarrier and that the optimal virus harvest time correlated with the global maximum permittivity signal. In 16 independent bioreactor runs, the maximum Measles virus titer was achieved approximately 40 hr after the permittivity maximum. Compared to an uncontrolled Measles virus production process, the integration of DS increased the maximum virus concentration by more than three orders of magnitude. This was sufficient to achieve an active Measles virus concentration of > 1010 TCID50 ml-1 .


Asunto(s)
Espectroscopía Dieléctrica/métodos , Virus del Sarampión/crecimiento & desarrollo , Virus Oncolíticos/crecimiento & desarrollo , Tecnología Farmacéutica/métodos , Cultivo de Virus/métodos , Animales , Chlorocebus aethiops , Células Vero
7.
Biotechnol Prog ; 33(4): 989-997, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28054473

RESUMEN

Measles virus (MV) has a natural affinity for cancer cells and oncolytic MV preparations have therefore been investigated in several clinical trials as a potential treatment for cancer. The main bottleneck in the administration of oncolytic MV to cancer patients is the production process, because very large doses of virus particles are required for each treatment. Here, we investigated the productivity of different host cells and found that a high infection efficiency did not necessarily result in high virus yields because virus release is also dependent on the host cell. As well as producing large numbers of active MV particles, host cells must perform well in dynamic cultivation systems. In screening experiments, the highest productivity was achieved by Vero and BJAB cells, but only the Vero cells maintained their high virus productivity when transferred to a stirred tank reactor. We used dielectric spectroscopy as an online monitoring system to control the infection and harvest times, which are known to be critical process parameters. The precise control of these parameters allowed us to achieve higher virus titers with Vero cells in a stirred tank reactor than in a static cultivation system based on T-flasks, with maximum titers of up to 1011 TCID50 ml-1 . © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 33:989-997, 2017.


Asunto(s)
Reactores Biológicos , Técnicas de Cultivo de Célula , Virus del Sarampión/crecimiento & desarrollo , Virus del Sarampión/aislamiento & purificación , Animales , Línea Celular , Chlorocebus aethiops , Espectroscopía Dieléctrica , Humanos , Células Vero
8.
J Virol ; 90(15): 6808-6817, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27194761

RESUMEN

UNLABELLED: Measles is a highly contagious, acute viral illness. Immune cells within the airways are likely first targets of infection, and these cells traffic measles virus (MeV) to lymph nodes for amplification and subsequent systemic dissemination. Infected immune cells are thought to return MeV to the airways; however, the mechanisms responsible for virus transfer to pulmonary epithelial cells are poorly understood. To investigate this process, we collected blood from human donors and generated primary myeloid cells, specifically, monocyte-derived macrophages (MDMs) and dendritic cells (DCs). MDMs and DCs were infected with MeV and then applied to primary cultures of well-differentiated airway epithelial cells from human donors (HAE). Consistent with previous results obtained with free virus, infected MDMs or DCs were incapable of transferring MeV to HAE when applied to the apical surface. Likewise, infected MDMs or DCs applied to the basolateral surface of HAE grown on small-pore (0.4-µm) support membranes did not transfer virus. In contrast, infected MDMs and DCs applied to the basolateral surface of HAE grown on large-pore (3.0-µm) membranes successfully transferred MeV. Confocal microscopy demonstrated that MDMs and DCs are capable of penetrating large-pore membranes but not small-pore membranes. Further, by using a nectin-4 blocking antibody or recombinant MeV unable to enter cells through nectin-4, we demonstrated formally that transfer from immune cells to HAE occurs in a nectin-4-dependent manner. Thus, both infected MDMs and DCs rely on cell-to-cell contacts and nectin-4 to efficiently deliver MeV to the basolateral surface of HAE. IMPORTANCE: Measles virus spreads rapidly and efficiently in human airway epithelial cells. This rapid spread is based on cell-to-cell contact rather than on particle release and reentry. Here we posit that MeV transfer from infected immune cells to epithelial cells also occurs by cell-to-cell contact rather than through cell-free particles. In addition, we sought to determine which immune cells transfer MeV infectivity to the human airway epithelium. Our studies are based on two types of human primary cells: (i) myeloid cells generated from donated blood and (ii) well-differentiated airway epithelial cells derived from donor lungs. We show that different types of myeloid cells, i.e., monocyte-derived macrophages and dendritic cells, transfer infection to airway epithelial cells. Furthermore, cell-to-cell contact is an important component of successful MeV transfer. Our studies elucidate a mechanism by which the most contagious human respiratory virus is delivered to the airway epithelium.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Células Epiteliales/virología , Macrófagos/virología , Virus del Sarampión/crecimiento & desarrollo , Sarampión/virología , Células Mieloides/virología , Sistema Respiratorio/virología , Fusión Celular , Células Cultivadas , Células Dendríticas/virología , Humanos , Sarampión/metabolismo , Nectinas , Receptores Virales/metabolismo , Internalización del Virus
9.
Viral Immunol ; 29(5): 296-306, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27035543

RESUMEN

Changes in interferon (IFN)-stimulated gene (ISG) expression in cells infected with measles virus (MeV), four wild strains (belonging to different genotypes), and the laboratory strain Edmonston were examined. ISGs [MxA, 2'-5'-oligoadenylate synthetase, and interferon regulatory factor-1] were upregulated in an MeV-infection-induced manner and in an IFN-induced manner. In MeV-infected SiHa cell lines, the MeV infection-induced expression levels were in the order of A>H1>D8>D5>D3. On the other hand, all infected cell lines abolished type I and III IFN-induced ISG expression. However, partial type II IFN-mediated induction was observed in the MeV-infected cells. The wild strain of genotype D3 was the most potent inhibitor of MeV infection-induced and IFN-induced ISG expression and generated the highest titer of infectious viral particles. Edmonston triggered the highest levels of MeV infection-induced ISG expression in SiHa cells and produced the lowest titer of infectious particles. Expression of the viral C protein was associated with suppression of MeV infection-induced and type II IFN-induced ISG expression.


Asunto(s)
Células Epiteliales/virología , Genoma Viral , Interacciones Huésped-Patógeno , Virus del Sarampión/efectos de los fármacos , Proteínas no Estructurales Virales/inmunología , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Cuello del Útero/efectos de los fármacos , Cuello del Útero/inmunología , Cuello del Útero/virología , Chlorocebus aethiops , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Femenino , Regulación de la Expresión Génica , Genotipo , Humanos , Interferón-alfa/farmacología , Interferón gamma/farmacología , Interferones/farmacología , Virus del Sarampión/genética , Virus del Sarampión/crecimiento & desarrollo , Virus del Sarampión/inmunología , Nitrilos , Proteínas de la Nucleocápside/genética , Proteínas de la Nucleocápside/inmunología , Plásmidos/química , Plásmidos/inmunología , Pirazoles/farmacología , Pirimidinas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Células Vero , Carga Viral/efectos de los fármacos , Proteínas no Estructurales Virales/genética
10.
Emerg Infect Dis ; 22(4): 687-90, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26926035
11.
Oncotarget ; 6(42): 44892-904, 2015 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-26539644

RESUMEN

Attenuated measles virus (MV) is currently being evaluated as an oncolytic virus in clinical trials and could represent a new therapeutic approach for malignant pleural mesothelioma (MPM). Herein, we screened the sensitivity to MV infection and replication of twenty-two human MPM cell lines and some healthy primary cells. We show that MV replicates in fifteen of the twenty-two MPM cell lines. Despite overexpression of CD46 by a majority of MPM cell lines compared to healthy cells, we found that the sensitivity to MV replication did not correlate with this overexpression. We then evaluated the antiviral type I interferon (IFN) responses of MPM cell lines and healthy cells. We found that healthy cells and the seven insensitive MPM cell lines developed a type I IFN response in presence of the virus, thereby inhibiting replication. In contrast, eleven of the fifteen sensitive MPM cell lines were unable to develop a complete type I IFN response in presence of MV. Finally, we show that addition of type I IFN onto MV sensitive tumor cell lines inhibits replication. These results demonstrate that defects in type I IFN response are frequent in MPM and that MV takes advantage of these defects to exert oncolytic activity.


Asunto(s)
Interferón Tipo I/metabolismo , Virus del Sarampión/crecimiento & desarrollo , Mesotelioma/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/crecimiento & desarrollo , Neoplasias Pleurales/terapia , Replicación Viral , Antígenos CD/metabolismo , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Interacciones Huésped-Patógeno , Humanos , Interferón Tipo I/inmunología , Virus del Sarampión/inmunología , Virus del Sarampión/metabolismo , Proteína Cofactora de Membrana/metabolismo , Mesotelioma/inmunología , Mesotelioma/metabolismo , Mesotelioma/virología , Virus Oncolíticos/inmunología , Virus Oncolíticos/metabolismo , Neoplasias Pleurales/inmunología , Neoplasias Pleurales/metabolismo , Neoplasias Pleurales/virología , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Factores de Tiempo
12.
Vaccine ; 33(36): 4540-7, 2015 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-26192359

RESUMEN

Disposable-syringe jet injectors (DSJIs) with single-use, auto disable, needle-free syringes offer the opportunity to avoid hazards associated with injection using a needle and syringe. Clinical studies have evaluated DSJIs for vaccine delivery, but most studies have focused on inactivated, subunit, or DNA vaccines. Questions have been raised about possible damage to live attenuated viral vaccines by forces generated during the jet injection process. This study examines the effect of jet injection on the integrity of measles, mumps, and rubella vaccine (MMR), measured by viral RNA content and infectivity. Three models of DSJIs were evaluated, each generating a different ejection force. Following jet injection, the RNA content for each of the vaccine components was measured using RT-qPCR immediately after injection and following passage in Vero cells. Jet injection was performed with and without pig skin as a simulation of human skin. There was little to no reduction of RNA content immediately following jet injection with any of the three DSJIs. Samples passaged in Vero cells showed no loss in infectivity of the measles vaccine following jet injection. Mumps vaccine consistently showed increased replication following jet injection. Rubella vaccine showed no loss after jet injection alone but some infectivity loss following injection through pig skin with two of the devices. Overall, these data demonstrated that forces exerted on a live attenuated MMR vaccine did not compromise vaccine infectivity. The bench model and protocol used in this study can be applied to evaluate the impact of jet injection on other live virus vaccines.


Asunto(s)
Equipos Desechables , Inyecciones a Chorro/métodos , Vacuna contra el Sarampión-Parotiditis-Rubéola/química , Vacuna contra el Sarampión-Parotiditis-Rubéola/inmunología , Animales , Chlorocebus aethiops , Virus del Sarampión/crecimiento & desarrollo , Viabilidad Microbiana , Virus de la Parotiditis/crecimiento & desarrollo , ARN Viral/análisis , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virus de la Rubéola/crecimiento & desarrollo , Células Vero , Cultivo de Virus
13.
J Virol ; 89(14): 7089-96, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25926640

RESUMEN

UNLABELLED: The discovery that measles virus (MV) uses the adherens junction protein nectin-4 as its epithelial receptor provides a new vantage point from which to characterize its rapid spread in the airway epithelium. We show here that in well-differentiated primary cultures of airway epithelial cells from human donors (HAE), MV infectious centers form rapidly and become larger than those of other respiratory pathogens: human respiratory syncytial virus, parainfluenza virus 5, and Sendai virus. While visible syncytia do not form after MV infection of HAE, the cytoplasm of an infected cell suddenly flows into an adjacent cell, as visualized through wild-type MV-expressed cytoplasmic green fluorescent protein (GFP). High-resolution video microscopy documents that GFP flows through openings that form on the lateral surfaces between columnar epithelial cells. To assess the relevance of the protein afadin, which connects nectin-4 to the actin cytoskeleton, we knocked down its mRNA. This resulted in more-limited infectious-center formation. We also generated a nectin-4 mutant without the afadin-binding site in its cytoplasmic tail. This mutant was less effective than wild-type human nectin-4 at promoting MV infection in primary cultures of porcine airway epithelia. Thus, in airway epithelial cells, MV spread requires the nectin-4/afadin complex and is based on cytoplasm transfer between columnar cells. Since the viral membrane fusion apparatus may open the passages that allow cytoplasm transfer, we refer to them as intercellular membrane pores. Virus-induced intercellular pores may contribute to extremely efficient measles contagion by promoting the rapid spread of the virus through the upper respiratory epithelium. IMPORTANCE: Measles virus (MV), while targeted for eradication, still causes about 120,000 deaths per year worldwide. The recent reemergence of measles in insufficiently vaccinated populations in Europe and North America reminds us that measles is extremely contagious, but the processes favoring its spread in the respiratory epithelium remain poorly defined. Here we characterize wild-type MV spread in well-differentiated primary cultures of human airway epithelial cells. We observed that viral infection promotes the flow of cytoplasmic contents from infected to proximal uninfected columnar epithelial cells. Cytoplasm flows through openings that form on the lateral surfaces. Infectious-center growth is facilitated by afadin, a protein connecting the adherens junction and the actin cytoskeleton. The viral fusion apparatus may open intercellular pores, and the cytoskeleton may stabilize them. Rapid homogenization of cytoplasmic contents in epithelial infectious centers may favor rapid spread and contribute to the extremely contagious nature of measles.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Células Epiteliales/virología , Interacciones Huésped-Patógeno , Virus del Sarampión/crecimiento & desarrollo , Proteínas de Microfilamentos/metabolismo , Animales , Células Cultivadas , Humanos , Microscopía por Video , Virus de la Parainfluenza 5/crecimiento & desarrollo , Virus Sincitial Respiratorio Humano/crecimiento & desarrollo , Virus Sendai/crecimiento & desarrollo , Porcinos , Internalización del Virus
14.
Virus Res ; 189: 206-13, 2014 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-24911240

RESUMEN

Adult T cell leukemia/lymphoma (ATL) is a highly aggressive CD4+/CD25+ T-cell malignancy caused by human T cell lymphotropic virus type 1 (HTLV-1). Previous studies in the MET-1 cell/NOD/SCID mouse model of ATL demonstrated that MET-1 cells are very susceptible to measles virus (MV) oncolytic therapy. To further evaluate the potential of MV therapy in ATL, the susceptibility of several HTLV-1 transformed CD4+ T cell lines (MT-1, MT-2, MT-4 and C8166-45) as well as HTLV-1 negative CD4+ T cell lines (Jurkat and CCRF-CEM) to infection with MV was tested in vitro. All cell lines were permissive to MV infection and subsequent cell death, except MT-1 and CCRF-CEM cells which were susceptible and permissive to MV infection, but resistant to cell death. The resistance to MV-mediated cell death was associated with IFNß produced by MT-1 and CCRF-CEM cells. Inhibition of IFNß rendered MT-1 and CCRF-CEM cells susceptible to MV-mediated cell death. Cells susceptible to MV-induced cell death did not produce nor were responsive to IFNß. Upon infection with Newcastle Disease Virus (NDV), MT-1 and CCRF-CEM but not the susceptible cell lines up-regulated pSTAT-2. In vivo, treatment of tumors induced by MT-1 cell lines which produce IFNß demonstrated only small increases in mean survival time, while only two treatments prolonged mean survival time in mice with MET-1 tumors deficient in type I interferon production. These results indicate that type I interferon production is closely linked with the inability of tumor cells to respond to type I interferon. Screening of tumor cells for type I interferon could be a useful strategy to select candidate patients for MV virotherapy.


Asunto(s)
Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Leucemia-Linfoma de Células T del Adulto/inmunología , Leucemia-Linfoma de Células T del Adulto/terapia , Virus del Sarampión/crecimiento & desarrollo , Viroterapia Oncolítica/métodos , Animales , Línea Celular , Supervivencia Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones SCID , Análisis de Supervivencia
15.
J Virol ; 88(15): 8332-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24829351

RESUMEN

UNLABELLED: VSV-FH is a hybrid vesicular stomatitis virus (VSV) with a deletion of its G glycoprotein and encoding the measles virus (MV) fusion (F) and hemagglutinin (H) envelope glycoproteins. VSV-FH infects cells expressing MV receptors and is fusogenic and effective against myeloma xenografts in mice. We evaluated the fusogenic activities of MV and VSV-FH in relationship to the density of receptor on the target cell surface and the kinetics of F and H expression in infected cells. Using a panel of cells expressing increasing numbers of the MV receptor CD46, we evaluated syncytium size in MV- or VSV-FH-infected cells. VSV-FH is not fusogenic at low CD46 density but requires less CD46 for syncytium formation than MV. The size of each syncytium is larger in VSV-FH-infected cells at a specific CD46 density. While syncytium size reached a plateau and did not increase further in MV-infected CHO cells expressing ≥4,620 CD46 copies/cell, there was a corresponding increase in syncytium size with increases in CD46 levels in VSV-FH-infected CD46-expressing CHO (CHO-CD46) cells. Further analysis in VSV-FH-infected cell lines shows earlier and higher expression of F and H mRNAs and protein. However, VSV-FH cytotoxic activity was reduced by pretreatment of the cells with type I interferon. In contrast, the cytopathic effects are not affected in MV-infected cells. In summary, VSV-FH has significant advantages over MV as an oncolytic virus due to its higher viral yield, faster replication kinetics, and larger fusogenic capabilities but should be used in cancer types with defective interferon signaling pathways. IMPORTANCE: We studied the cytotoxic activity of a vesicular stomatitis/measles hybrid virus (VSV-FH), which is superior to that of measles virus (MV), in different cancer cell lines. We determined that viral RNA and protein were produced faster and in higher quantities in VSV-FH-infected cells. This resulted in the formation of larger syncytia, higher production of infectious particles, and a more potent cytopathic effect in permissive cells. Importantly, VSV-FH, similar to MV, can discriminate between low- and high-expressing CD46 cells, a phenotype important for cancer therapy as the virus will be able to preferentially infect cancer cells that overexpress CD46 over low-CD46-expressing normal cells.


Asunto(s)
Glicoproteínas/metabolismo , Hemaglutininas/metabolismo , Virus del Sarampión/fisiología , Recombinación Genética , Vesiculovirus/fisiología , Proteínas Virales de Fusión/metabolismo , Internalización del Virus , Animales , Células CHO , Cricetinae , Cricetulus , Células Gigantes/virología , Glicoproteínas/genética , Hemaglutininas/genética , Virus del Sarampión/genética , Virus del Sarampión/crecimiento & desarrollo , Proteína Cofactora de Membrana/metabolismo , Receptores Virales/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Vesiculovirus/genética , Vesiculovirus/crecimiento & desarrollo , Proteínas Virales de Fusión/genética
16.
J Math Biol ; 68(3): 533-48, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23143337

RESUMEN

For a certain class of multi-type branching processes in a continuous-time periodic environment, we show that the extinction probability is equal to (resp. less than) 1 if the basic reproduction number R(0) is less than (resp. bigger than) 1. The proof uses results concerning the asymptotic behavior of cooperative systems of differential equations. In epidemiology the extinction probability may be used as a time-periodic measure of the epidemic risk. As an example we consider a linearized SEIR epidemic model and data from the recent measles epidemic in France. Discrete-time models with potential applications in conservation biology are also discussed.


Asunto(s)
Número Básico de Reproducción , Epidemias , Extinción Biológica , Modelos Biológicos , Humanos , Sarampión/epidemiología , Virus del Sarampión/crecimiento & desarrollo
17.
J Med Chem ; 56(3): 1311-22, 2013 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-23320521

RESUMEN

Dideoxy bicyclic pyrimidine nucleoside analogues (ddBCNAs) with d-chirality have previously been described by us to inhibit replication of human cytomegalovirus. We herein report for the first time that activity against vaccinia virus (VACV) was achieved using novel l-analogues. A structure-activity relationship was established: Antiviral activity versus VACV was highest with an ether side chain with an optimum of n-C(9)H(18)-O-n-C(5)H(11). This gave an IC(50) of 190 nM, a 60-fold enhancement over the FDA-approved antiviral cidofovir. Interestingly, l-ddBCNAs also inhibit wild type measles virus syncytia formation with a TCID(50) of 7.5 µM for the lead compound. We propose that l-ddBCNAs represent significant innovative antiviral candidates versus measles and poxviruses, and we suggest a mechanism of action versus one or more cellular targets that are essential for viral replication.


Asunto(s)
Antivirales/farmacología , Virus del Sarampión/efectos de los fármacos , Virus Vaccinia/efectos de los fármacos , Antivirales/química , Células HeLa , Humanos , Técnicas In Vitro , Espectroscopía de Resonancia Magnética , Virus del Sarampión/crecimiento & desarrollo , Pruebas de Sensibilidad Microbiana , Espectrometría de Masa por Ionización de Electrospray , Virus Vaccinia/crecimiento & desarrollo , Ensayo de Placa Viral
18.
J Virol ; 87(6): 3484-501, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23302892

RESUMEN

The oncolytic potential of measles vaccine virus (MeV) has been demonstrated in several tumor entities. Here, we investigated the susceptibility of eight sarcoma cell lines to MeV-mediated oncolysis and found five to be susceptible, whereas three proved to be resistant. In the MeV-resistant cell lines, we often observed an inhibition of viral replication along with a strong upregulation of the intracellular virus-sensing molecule RIG-I and of the interferon (IFN)-stimulated gene IFIT1. Not only expression of IFIT1 but also phosphorylation of IFN-stimulated Stat1 took place rapidly and were found to be persistent over time. In contrast, susceptible cell lines showed a much weaker, delayed, or completely missing expression of IFIT1 as well as a delayed or only transient phosphorylation of Stat1, whereas exogenic stimulation with beta interferon (IFN-ß) resulted in a comparable profound activation of Stat1 and expression of IFIT1 in all cell lines. Pretreatment with IFN-ß rendered three of the susceptible cell lines more resistant to MeV-mediated oncolysis. These data suggest that differences in the innate immune defense often account for different degrees of susceptibility of sarcoma cell lines to MeV-mediated oncolysis. From a therapeutic perspective, we were able to overcome resistance to MeV by increasing the multiplicity of infection (MOI) and by addition of the prodrug 5-fluorocytosine (FC), thereby exploiting the suicide gene function of virotherapeutic vector MeV-SCD armed with the SCD fusion protein, which consists of yeast cytosine deaminase and yeast uracil phosphoribosyltransferase.


Asunto(s)
Virus del Sarampión/crecimiento & desarrollo , Virus del Sarampión/inmunología , Virus Oncolíticos/crecimiento & desarrollo , Virus Oncolíticos/inmunología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Humanos , Interferón beta/inmunología , Virus del Sarampión/fisiología , Virus Oncolíticos/fisiología , Proteínas de Unión al ARN , Factor de Transcripción STAT1/metabolismo , Replicación Viral
19.
J Gen Virol ; 93(Pt 3): 565-576, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22170635

RESUMEN

The cytidine deaminase APOBEC3G (apolipoprotein B mRNA-editing enzyme-catalytic polypeptide 3G; A3G) exerts antiviral activity against retroviruses, hepatitis B virus, adeno-associated virus and transposable elements. We assessed whether the negative-strand RNA viruses measles, mumps and respiratory syncytial might be affected by A3G, and found that their infectivity was reduced by 1-2 logs (90-99 %) in A3G overexpressing Vero cells, and in T-cell lines expressing A3G at physiological levels. Viral RNA was co-precipitated with HA-tagged A3G and could be amplified by RT-PCR. Interestingly, A3G reduced viral transcription and protein expression in infected cells by 50-70 %, and caused an increased mutation frequency of 0.95 mutations per 1000 nt in comparison to the background level of 0.22/1000. The observed mutations were not specific for A3G [cytidine to uridine (C→U) or guanine to adenine (G→A) hypermutations], nor specific for ADAR (adenosine deaminase acting on RNA, A→G and U→C transitions, with preference for next neighbour-nucleotides U = A>C>G). In addition, A3G mutants with inactivated catalytic deaminase (H257R and E259Q) were inhibitory, indicating that the deaminase activity is not required for the observed antiviral activity. In combination, impaired transcription and increased mutation frequencies are sufficient to cause the observed reduction in viral infectivity and eliminate virus replication within a few passages in A3G-expressing cells.


Asunto(s)
Citidina Desaminasa/metabolismo , Virus del Sarampión/patogenicidad , Virus de la Parotiditis/patogenicidad , Virus Sincitiales Respiratorios/patogenicidad , Replicación Viral , Desaminasa APOBEC-3G , Animales , Antivirales/metabolismo , Línea Celular , Citidina Desaminasa/inmunología , Humanos , Virus del Sarampión/crecimiento & desarrollo , Virus del Sarampión/inmunología , Virus de la Parotiditis/crecimiento & desarrollo , Virus de la Parotiditis/inmunología , Mutación Puntual , ARN Viral/genética , Virus Sincitiales Respiratorios/crecimiento & desarrollo , Virus Sincitiales Respiratorios/inmunología
20.
Appl Microbiol Biotechnol ; 93(3): 1031-40, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21935589

RESUMEN

Measles vaccination remains the most efficient way to control the spread of the virus. This work focuses on the production of a measles vaccine using stirred conditions as an advanced option for process scale up. Non-porous Cytodex 1 microcarriers were used to support MRC-5 cell growth in suspension cultures. Virus replication was first optimized in spinner flasks, and the effects of various operational parameters were investigated. Cell infection with AIK-C measles strain at an MOI (multiplicity of infection) of 0.005, without glucose regulation and in M199 medium, resulted in a virus titer of 106·²5 TCID50 (median tissue culture infective dose)/ml. To optimize the production process in a 7-l bioreactor, we carried out various perfused cultures using minimum essential medium (MEM) + 5% FCS diluted with phosphate-buffered saline (PBS). We achieved a high cell density level (4.1 × 106 cells/ml) with an efficient use of the medium when MEM + 5% FCS diluted with PBS at 25% was used during the cell amplification step. Optimization of measles production in MRC-5 cells grown on Cytodex 1 beads in a 7-l bioreactor showed that perfusion was the most efficient when compared to repeated-batch culture. Perfusion at a rate of 0.25 V (reactor volume)/day showed the highest specific productivity (1.6 IVP [infectious virus particle] cell⁻¹ day⁻¹). Testing of several stabilizers containing pharmaceutically improved components such as sugars, amino acids, and charged ions showed that the formulation composed of sucrose and MgCl2, led to the maintenance of the infectivity of the AIK-C measles virus strain to a significant level, when stored at +28 °C, +4 °C and -60 °C.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Dextranos , Fibroblastos/virología , Vacuna Antisarampión , Virus del Sarampión/crecimiento & desarrollo , Cultivo de Virus/métodos , Reactores Biológicos , Biotecnología/métodos , Técnicas de Cultivo de Célula/instrumentación , Línea Celular , Medios de Cultivo , Humanos , Microesferas , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...