78 results on '"SFV"'
Search Results
2. Análisis y Fundamentos de Sistemas Fotovoltaicos Interconectados a la Red de Distribución.
- Author
-
Arizpe Islas, Jorge Luis, Porras González, Victor Alejandro, Aguiñaga Guerrero, José Fernando, Leal Beltrán, David Enrique, and Galindo Martínez, Jesús Alejandro
- Abstract
Copyright of Congreso Internacional de Investigacion Academia Journals is the property of PDHTech, LLC and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2019
3. Semliki Forest virus infection of mosquito cells : novel insights into host responses and antiviral immunity
- Author
-
Rodriguez, Julio, Kohl, Alain, and Fazakerley, John
- Subjects
616.9 ,Semliki Forest virus ,SFV ,mosquito ,arboviruses ,innate immunity - Abstract
Arboviruses are transmitted between vertebrate hosts by arthropod vectors, such as mosquitoes or ticks. In vertebrates arboviruses cause cytopathic effects and disease, however, arbovirus infection of arthropods usually results in persistence. Control of arboviral infection is mediated by the arthropod’s immune system. Pathways such as RNAi, JAK/STAT, Toll and IMD have previously been implicated in controlling arbovirus infections. In contrast, the antiviral role of other pathways in mosquitoes, such as melanisation, is unknown. Using high through output 454 sequencing the transcriptome of U4.4 cells infected with the model arbovirus Semliki Forest virus (SFV)(Togaviridae, Alphavirus) was generated. This experiment revealed intriguing patterns of differential transcript abundance that suggest a broad impact of SFV infection in U4.4 cells, such as in metabolism, cell structure and nucleic acid processing. SFV infection induces differential expression of genes in pathways such as apoptosis, stress response and cell cycle. Most interestingly, this study indicated that melanisation might have an antiviral role in mosquitoes. In arthropods, melanisation is a process involved in wound healing and antimicrobial defences. Phenoloxidase (PO), a key enzyme involved in melanisation, is cytotoxic and therefore kept in its inactive form, prophenoloxidase (PPO), until activation is triggered. The PPO activation process is tightly regulated by serine protease inhibitors (serpins) which inhibit the proteolytic activation reaction. In this thesis I demonstrate that the supernatant of cultured Aedes albopictus-derived U4.4 cells contains a functional proPO-activating system, which is activated by infection with bacteria and virions of SFV. Activation of this pathway reduces the spread and infectivity of SFV in vitro and in vivo. In order to further characterise the PO cascade and its antiviral role the serpins in Ae. albopictus were also investigated. Using the transcriptome sequencing and bioinformatics we identified and classified 11 serpins. We silenced each of the serpins and monitored PPO levels and antiviral activity showing that homologues to drosophila’s serpin- 27a plays a role in melanisation against SFV in vitro. Collectively, these results characterise the mosquito PO cascade as a novel immune defence against arbovirus infection in mosquitoes.
- Published
- 2013
4. Lack of expression of hepatitis C virus core protein in human monocyte-erived dendritic cells using recombinant semliki forest virus
- Author
-
Maria-Cristina Navas, Françoise Stoll-Keller, and Jovan Pavlovic
- Subjects
Dendritic cells ,HCV ,SFV ,viral vector ,Biology (General) ,QH301-705.5 - Abstract
Hepatitis C Virus belongs to the Flaviviridae family. One proposed mechanism of HCV persistence in the ability to infect hematopoietic cells, including Dendritic cells (DCs). HCV infection of DCs could impair their functions that represent one of the mechanisms, thus hampering viral clearance by the host immune system. Among HCV-encoded proteins, the highly conserved Core protein has been suggested to be responsible for the immunomodulatory properties of this Hepacivirus. Recombinant viral vectors expressing the HCV Core protein and allowing its transduction and therefore the expression of the protein into DCs could be useful tools for the analysis of the properties of the Core protein. Vaccinia Virus and retrovirus have been used to transduce human DCs. Likewise, gene transfer into DCs using Semliki Forest Virus has been reported. This study aimed to express the HCV Core protein in human monocyte-derived DCs using an SFV vector, in which the subgenomic RNA encoding the structural proteins was replaced by the HCV Core sequence and then analyze the effects of its expression on DCs functions.
- Published
- 2019
- Full Text
- View/download PDF
5. An optimization study for expression of the rabies virus glycoprotein (RVGP) in mammalian cell lines using the Semliki Forest virus (SFV).
- Author
-
de Rezende, Alexandre Gonçalves, Fernández Núñez, Eutimio Gustavo, Astray, Renato Mancini, Puglia, Ana Lia Pradella, Pereira, Carlos Augusto, and Jorge, Soraia Attie Calil
- Subjects
- *
SEMLIKI Forest virus , *CELL lines , *RABIES virus , *ENZYME-linked immunosorbent assay , *PROTEIN expression - Abstract
• A production approach for viral particles as a delivery system of RNA encoding the rabies virus Glycoprotein was set up. • The vectored particles were able to enter different mammalian cell lines and promote the expression of the target protein. • The vectored particles were able to RNA delivery to 5 mammalian cell lines (BHK-21, HuH-7, Vero, L929, and HEK-293T). The Semliki Forest virus (SFV) viral vector has been widely used for transient protein expression. This study aimed to analyze comprehensively the capacity of SFV vector to express rabies lyssavirus glycoprotein (RVGP) in mammalian cells. The assessed parameters were transfection strategy, multiplicity of infection (MOI), harvest time and mammalian cell host. Two transfection approaches, electroporation and lipofection were evaluated to obtain the recombinant SFV, and the electroporation was found to be the most effective. Viral quantification by RT-qPCR was performed to elucidate the relation between the amount of recombinant virus utilized in the infection process and the production levels of the heterologous protein. Four different multiplicities of infection (MOIs = 1; 10; 15; 50) were evaluated using five mammalian cell lines: BHK-21, HuH-7, Vero, L929, and HEK-293T. Protein expression was assessed at two harvest times after infection (24 and 48 h). The recombinant protein generated was characterized by western blot, dot blot, and indirect immunofluorescence (IIF), while its concentration was determined by enzyme-linked immunosorbent assay (ELISA). Similar expression patterns were observed in cell lines BHK-21, HEK-293T, L929, and Vero, with higher RVGP production in the first 24 h. The BHK-21 cells showed yields of up to 4.3 μg per 106 cells when lower MOIs (1 and 10) were used. The HEK-293 T cells also showed similar production (4.3 μg per 106 cells) with MOI of 1, while the L929 and Vero cell lines showed lower expression rates of 2.82 and 1.26 μg per 106 cells, respectively. These cell lines showed lower expression levels at 48 h after infection compared to 24 h. Controversially, in the case of the HuH-7 cell line, RVGP production was higher at 48 h after infection (4.0 μg per 106 cells) and using MOIs of 15 and 50. This work may contribute to optimize the RVGP production using SFV system in mammalian cells. This study can also substantiate for example, the development of approaches that use of SFV for applications for other protein expressions and suggests values for relevant parameters and cell lines of this biotechnique. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
6. LACK OF EXPRESSION OF HEPATITIS C VIRUS CORE PROTEIN IN HUMAN MONOCYTE-DERIVED DENDRITIC CELLS USING RECOMBINANT SEMLIKI FOREST VIRUS.
- Author
-
NAVAS, Maria-Cristina, STOLL-KELLER, Françoise, and PAVLOVIC, Jovan
- Subjects
- *
SEMLIKI Forest virus , *HEPATITIS C virus , *VIRAL proteins , *CYTOSKELETAL proteins , *DENDRITIC cells , *VACCINIA - Abstract
Hepatitis C Virus belongs to the Flaviviridae family. One proposed mechanism of HCV persistence in the ability to infect hematopoietic cells, including Dendritic cells (DCs). HCV infection of DCs could impair their functions that represent one of the mechanisms, thus hampering viral clearance by the host immune system. Among HCV-encoded proteins, the highly conserved Core protein has been suggested to be responsible for the immunomodulatory properties of this Hepacivirus. Recombinant viral vectors expressing the HCV Core protein and allowing its transduction and therefore the expression of the protein into DCs could be useful tools for the analysis of the properties of the Core protein. Vaccinia Virus and retrovirus have been used to transduce human DCs. Likewise, gene transfer into DCs using Semliki Forest Virus has been reported. This study aimed to express the HCV Core protein in human monocyte-derived DCs using an SFV vector, in which the subgenomic RNA encoding the structural proteins was replaced by the HCV Core sequence and then analyze the effects of its expression on DCs functions. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
7. Organelle dynamics and viral infections: at cross roads.
- Author
-
Glingston, R. Sahaya, Deb, Rachayeeta, Kumar, Sachin, and Nagotu, Shirisha
- Subjects
- *
ORGANELLES , *INTRACELLULAR pathogens , *VIRUS diseases , *HOST-parasite relationships , *VIRUSES - Abstract
Abstract Viruses are obligate intracellular parasites of the host cells. A commonly accepted view is the requirement of internal membranous structures for various aspects of viral life cycle. Organelles enable favourable intracellular environment for several viruses. However, studies reporting organelle dynamics upon viral infections are scant. In this review, we aim to summarize and highlight modulations caused to various organelles upon viral infection or expression of its proteins. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
8. Bayesian inference reveals ancient origin of simian foamy virus in orangutans.
- Author
-
Reid, Michael J.C., Switzer, William M., Schillaci, Michael A., Klegarth, Amy R., Campbell, Ellsworth, Ragonnet-Cronin, Manon, Joanisse, Isabelle, Caminiti, Kyna, Lowenberger, Carl A., Galdikas, Birute Mary F., Hollocher, Hope, Sandstrom, Paul A., and Brooks, James I
- Subjects
- *
SIMIAN foamy virus , *ORANGUTANS , *HOSTS (Biology) , *BIOLOGICAL evolution , *BAYESIAN analysis , *DISEASES - Abstract
Simian foamy viruses (SFVs) infect most nonhuman primate species and appears to co-evolve with its hosts. This co-evolutionary signal is particularly strong among great apes, including orangutans (genus Pongo ). Previous studies have identified three distinct orangutan SFV clades. The first of these three clades is composed of SFV from P . abelii from Sumatra, the second consists of SFV from P . pygmaeus from Borneo, while the third clade is mixed, comprising an SFV strain found in both species of orangutan. The existence of the mixed clade has been attributed to an expansion of P . pygmaeus into Sumatra following the Mount Toba super-volcanic eruption about 73,000 years ago. Divergence dating, however, has yet to be performed to establish a temporal association with the Toba eruption. Here, we use a Bayesian framework and a relaxed molecular clock model with fossil calibrations to test the Toba hypothesis and to gain a more complete understanding of the evolutionary history of orangutan SFV. As with previous studies, our results show a similar three-clade orangutan SFV phylogeny, along with strong statistical support for SFV-host co-evolution in orangutans. Using Bayesian inference, we date the origin of orangutan SFV to > 4.7 million years ago (mya), while the mixed species clade dates to approximately 1.7 mya, > 1.6 million years older than the Toba super-eruption. These results, combined with fossil and paleogeographic evidence, suggest that the origin of SFV in Sumatran and Bornean orangutans, including the mixed species clade, likely occurred on the mainland of Indo-China during the Late Pliocene and Calabrian stage of the Pleistocene, respectively. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
9. Structure of Semliki Forest virus in complex with its receptor VLDLR.
- Author
-
Cao, Duanfang, Ma, Bingting, Cao, Ziyi, Zhang, Xinzheng, and Xiang, Ye
- Subjects
- *
SEMLIKI Forest virus , *LOW density lipoprotein receptors , *ALPHAVIRUSES , *LIPOPROTEIN receptors , *INSECT hosts - Abstract
Semliki Forest virus (SFV) is an alphavirus that uses the very-low-density lipoprotein receptor (VLDLR) as a receptor during infection of its vertebrate hosts and insect vectors. Herein, we used cryoelectron microscopy to study the structure of SFV in complex with VLDLR. We found that VLDLR binds multiple E1-DIII sites of SFV through its membrane-distal LDLR class A (LA) repeats. Among the LA repeats of the VLDLR, LA3 has the best binding affinity to SFV. The high-resolution structure shows that LA3 binds SFV E1-DIII through a small surface area of 378 Å2, with the main interactions at the interface involving salt bridges. Compared with the binding of single LA3s, consecutive LA repeats around LA3 promote synergistic binding to SFV, during which the LAs undergo a rotation, allowing simultaneous key interactions at multiple E1-DIII sites on the virion and enabling the binding of VLDLRs from divergent host species to SFV. [Display omitted] • The receptor VLDLR binds multiple E1-DIII sites of SFV through its LA repeats • Single LA binds E1-DIII with a small contact surface and a low binding affinity • Multiple LAs promote synergistic binding and a high binding affinity to SFV Semliki Forest virus (SFV) enters host cells by binding to repeat regions of the host cell's very-low-density lipoprotein receptors. The repeat regions enable synergic, multimodal binding with SFV, which is highly tolerant of mutations, allowing SFV to bind efficiently to the receptors of highly divergent species. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
10. Local delivery of optimized nanobodies targeting the PD-1/PD-L1 axis with a self-amplifying RNA viral vector induces potent antitumor responses.
- Author
-
Silva-Pilipich, Noelia, Blanco, Ester, Lozano, Teresa, Martisova, Eva, Igea, Ana, Herrador-Cañete, Guillermo, Ballesteros-Briones, María Cristina, Gorraiz, Marta, Sarrión, Patricia, González-Sapienza, Gualberto, Lasarte, Juan José, Vanrell, Lucía, and Smerdou, Cristian
- Subjects
- *
IMMUNOGLOBULINS , *GENETIC vectors , *PROGRAMMED cell death 1 receptors , *PROGRAMMED death-ligand 1 , *IMMUNE checkpoint proteins - Abstract
Despite the success of immune checkpoint blockade for cancer therapy, many patients do not respond adequately. We aimed to improve this therapy by optimizing both the antibodies and their delivery route, using small monodomain antibodies (nanobodies) delivered locally with a self-amplifying RNA (saRNA) vector based on Semliki Forest virus (SFV). We generated nanobodies against PD-1 and PD-L1 able to inhibit both human and mouse interactions. Incorporation of a dimerization domain reduced PD-1/PD-L1 IC50 by 8- and 40-fold for anti-PD-L1 and anti-PD-1 nanobodies, respectively. SFV viral particles expressing dimeric nanobodies showed a potent antitumor response in the MC38 model, resulting in >50% complete regressions, and showed better therapeutic efficacy compared to vectors expressing conventional antibodies. These effects were also observed in the B16 melanoma model. Although a short-term expression of nanobodies was observed due to the cytopathic nature of the saRNA vector, it was enough to generate a strong proinflammatory response in tumors, increasing infiltration of NK and CD8+ T cells. Delivery of the SFV vector expressing dimeric nanobodies by local plasmid electroporation, which could be more easily translated to the clinic, also showed a potent antitumor effect. • Nanobodies able to block human and mouse PD-1/PD-L1 interactions have been developed. • Dimerization of nanobodies considerably decreased IC50 for PD-1/PD-L1 interactions. • SFV viral particles expressing dimeric nanobodies led to potent antitumor effects. • Delivery of SFV saRNA by plasmid electroporation showed a potent antitumor effect. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
11. Semliki forest virus as a vector: pros and cons for its use in biopharmaceuticals production
- Author
-
Eutimio Gustavo Fernández Núñez, Soraia Attie Calil Jorge, Renato Mancini Astray, Alexandre Gonçalves de Rezende, Bruno Labate Vale da Costa, Daniella Cristina Ventini Monteiro, Carlos Augusto Pereira, and Aldo Tonso
- Subjects
BHK-21 ,Large-scale bioprocesses ,Mammalian cells ,Recombinant proteins ,SFV ,Transient expression ,Biotechnology ,TP248.13-248.65 - Abstract
The number of biopharmaceuticals for medical and veterinarian use produced in mammalian cells is increasing year after year. All of them are obtained by stable recombinant cell lines. However, it is recognized that transient gene expression produces high level expression in a short time. In that sense, viral vectors have been extensively used for producing recombinant proteins on lab-scale. Among them, Semliki Forest virus is commonly employed for this purpose. This review discusses the main aspects related to the use of Semliki Forest virus technology as well as its advantages and drawbacks which limit currently its utilization in biopharmaceutical industry on large-scale.
- Published
- 2013
- Full Text
- View/download PDF
12. Simian Foamy Virus in Non-Human Primates and Cross-Species Transmission to Humans in Gabon: An Emerging Zoonotic Disease in Central Africa?
- Author
-
Augustin Mouinga-Ondémé and Mirdad Kazanji
- Subjects
SFV ,mandrills ,wild-born non-human primates ,interspecies transmission ,Gabon ,central Africa ,Microbiology ,QR1-502 - Abstract
It is now known that all human retroviruses have a non-human primate counterpart. It has been reported that the presence of these retroviruses in humans is the result of interspecies transmission. Several authors have described the passage of a simian retrovirus, simian foamy virus (SFV), from primates to humans. To better understand this retroviral “zoonosis” in natural settings, we evaluated the presence of SFV in both captive and wild non-human primates and in humans at high risk, such as hunters and people bitten by a non-human primate, in Gabon, central Africa. A high prevalence of SFV was found in blood samples from non-human primates and in bush meat collected across the country. Mandrills were found to be highly infected with two distinct strains of SFV, depending on their geographical location. Furthermore, samples collected from hunters and non-human primate laboratory workers showed clear, extensive cross-species transmission of SFV. People who had been bitten by mandrills, gorillas and chimpanzees had persistent SFV infection with low genetic drift. Thus, SFV is presumed to be transmitted from non-human primates mainly through severe bites, involving contact between infected saliva and blood. In this review, we summarize and discuss our five-year observations on the prevalence and dissemination of SFV in humans and non-human primates in Gabon.
- Published
- 2013
- Full Text
- View/download PDF
13. Local delivery of optimized nanobodies targeting the PD-1/PD-L1 axis with a self-amplifying RNA viral vector induces potent antitumor responses
- Author
-
Silva-Pilipich, N.R. (Noelia Romina)
- Subjects
- Nanobody, SFV, Alphavirus, PD-1/PD-L1, Cancer immunotherapy
- Abstract
Despite the success of immune checkpoint blockade for cancer therapy, many patients do not respond adequately. We aimed to improve this therapy by optimizing both the antibodies and their delivery route, using small monodomain antibodies (nanobodies) delivered locally with a self-amplifying RNA (saRNA) vector based on Semliki Forest virus (SFV). We generated nanobodies against PD-1 and PD-L1 able to inhibit both human and mouse interactions. Incorporation of a dimerization domain reduced PD-1/PD-L1 IC50 by 8- and 40-fold for antiPD-L1 and anti-PD-1 nanobodies, respectively. SFV viral particles expressing dimeric nanobodies showed a potent antitumor response in the MC38 model, resulting in >50% complete regressions, and showed better therapeutic efficacy compared to vectors expressing conventional antibodies. These effects were also observed in the B16 melanoma model. Although a short-term expression of nanobodies was observed due to the cytopathic nature of the saRNA vector, it was enough to generate a strong proinflammatory response in tumors, increasing infiltration of NK and CD8+ T cells. Delivery of the SFV vector expressing dimeric nanobodies by local plasmid electroporation, which could be more easily translated to the clinic, also showed a potent antitumor effect.
- Published
- 2023
14. Alphavirus Restriction by IFITM Proteins.
- Author
-
Weston, Stuart, Czieso, Stephanie, White, Ian J., Smith, Sarah E., Wash, Rachael S., Diaz‐Soria, Carmen, Kellam, Paul, and Marsh, Mark
- Subjects
- *
ALPHAVIRUS diseases , *THERAPEUTIC use of interferons , *MEMBRANE proteins , *CELL membranes , *ANTIVIRAL agents , *THERAPEUTICS - Abstract
Interferon inducible transmembrane proteins ( IFITMs) are broad-spectrum antiviral factors. In cell culture the entry of many enveloped viruses, including orthomyxo-, flavi-, and filoviruses, is inhibited by IFITMs, though the mechanism(s) involved remain unclear and may vary between viruses. We demonstrate that Sindbis and Semliki Forest virus ( SFV), which both use endocytosis and acid-induced membrane fusion in early endosomes to infect cells, are restricted by the early endosomal IFITM3. The late endosomal IFITM2 is less restrictive and the plasma membrane IFITM1 does not inhibit normal infection by either virus. IFITM3 inhibits release of the SFV capsid into the cytosol, without inhibiting binding, internalization, trafficking to endosomes or low pH-induced conformational changes in the envelope glycoprotein. Infection by SFV fusion at the cell surface was inhibited by IFITM1, but was equally inhibited by IFITM3. Furthermore, an IFITM3 mutant ( Y20A) that is localized to the plasma membrane inhibited infection by cell surface fusion more potently than IFITM1. Together, these results indicate that IFITMs, in particular IFITM3, can restrict alphavirus infection by inhibiting viral fusion with cellular membranes. That IFITM3 can restrict SFV infection by fusion at the cell surface equivalently to IFITM1 suggests that IFITM3 has greater antiviral potency against SFV. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
15. Long noncoding RNA EGOT negatively affects the antiviral response and favors HCV replication.
- Author
-
Carnero, Elena, Barriocanal, Marina, Prior, Celia, Pablo Unfried, Juan, Segura, Victor, Guruceaga, Elizabeth, Enguita, Mónica, Smerdou, Cristian, Gastaminza, Pablo, and Fortes, Puri
- Abstract
The role of long noncoding RNAs (lncRNAs) in viral infection is poorly studied. We have identified hepatitis C virus (HCV)-Stimulated lncRNAs (CSRs) by transcriptome analysis. Interestingly, two of these CSRs (PVT1 and UCA1) play relevant roles in tumorigenesis, providing a novel link between HCV infection and development of liver tumors. Expression of some CSRs seems induced directly by HCV, while others are upregulated by the antiviral response against the virus. In fact, activation of pathogen sensors induces the expression of CSR32/EGOT. RIG-I and the RNA-activated kinase PKR sense HCV RNA, activate NF-κB and upregulate EGOT. EGOT is increased in the liver of patients infected with HCV and after infection with influenza or Semliki Forest virus (SFV). Genome-wide guilt-by-association studies predict that EGOT may function as a negative regulator of the antiviral pathway. Accordingly, EGOT depletion increases the expression of several interferon-stimulated genes and leads to decreased replication of HCV and SFV. Our results suggest that EGOT is a lncRNA induced after infection that increases viral replication by antagonizing the antiviral response. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
16. Organelle dynamics and viral infections: at cross roads
- Author
-
Sachin Kumar, R. Sahaya Glingston, Shirisha Nagotu, and Rachayeeta Deb
- Subjects
0301 basic medicine ,Human cytomegalovirus ,Adeno Virus ,Severe Acute Respiratory Syndrome Corona Virus ,Mitochondria Permeability Transition Pore ,viruses ,UPR ,PV ,Dengue virus ,medicine.disease_cause ,SV40 ,HBV ,Cucumber Mosaic Virus ,MMP ,HSV ,CMV ,SARS-CoV ,ROS ,Vaccinia Virus ,Human Foreskin Fibroblasts ,Virus ,Mitochondria ,Cell biology ,Promyelocytic Leukemia Nuclear Bodies ,Infectious Diseases ,Virus Diseases ,Host-Pathogen Interactions ,Viruses ,Human Cytomegalovirus ,Polio Virus ,Hepatitis B Virus ,Intracellular ,030106 microbiology ,Immunology ,HFFs ,Ad ,Peroxisome ,Biology ,Microbiology ,Article ,Nucleus ,ESCRT ,Human Hepatoma cell lines ,Viral Proteins ,03 medical and health sciences ,Simian Virus ,Viral life cycle ,Huh ,Organelle ,medicine ,Human Immunodeficiency virus ,VACV ,HCMV ,MPTP ,SFV ,Organelles ,DENV ,Endosomal Sorting Complexes Required for Transport ,Mitochondrial Membrane Potential ,Semliki Forest Virus ,Intracellular parasite ,HIV ,Dengue Virus ,ERAD ,medicine.disease ,Herpes Simplex Virus ,Endoplasmic Reticulum Associated Degradation ,030104 developmental biology ,ER ,Gene Expression Regulation ,PML NB ,Unfolded Protein Response ,HCV, Hepatitis C Virus ,Reactive Oxygen Species ,Virus Physiological Phenomena - Abstract
Viruses are obligate intracellular parasites of the host cells. A commonly accepted view is the requirement of internal membranous structures for various aspects of viral life cycle. Organelles enable favourable intracellular environment for several viruses. However, studies reporting organelle dynamics upon viral infections are scant. In this review, we aim to summarize and highlight modulations caused to various organelles upon viral infection or expression of its proteins.
- Published
- 2019
17. Impact des multiples infections naturelles par des rétrovirus simiens sur la charge provirale et la clonalité rétrovirale in vivo
- Author
-
Jegado, Brice, Centre International de Recherche en Infectiologie - UMR (CIRI), École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Université de Lyon, Renaud Mahieux, David Fouchet, and STAR, ABES
- Subjects
Modèle animal ,Retrovirus ,viruses ,STLV-1 ,LM-PCR ,virus diseases ,biochemical phenomena, metabolism, and nutrition ,Clonalité ,Interspecies transmission ,ATL ,[SDV.MHEP.MI]Life Sciences [q-bio]/Human health and pathology/Infectious diseases ,Transmission inter-espèces ,[SDV.MHEP.MI] Life Sciences [q-bio]/Human health and pathology/Infectious diseases ,Animal model ,SFV ,Clonality ,Rétrovirus - Abstract
Non-human primates (NHPs) are natural hosts for exogenous retroviruses such as STLV-1 (Simian T-cell Leukemia Virus type 1) and SFV (Simian Foamy Virus), and natural cases of co-infections have been reported (SFV/STLV-1). As described for HTLV-1 in humans, STLV-1 is the etiologic agent of ATL (Adult T cell Leukemia) in NHPs, but the majority of infected animals remain asymptomatic. Furthermore, as for HTLV-1, which is able to replicate through clonal expansion of infected cells, the quantification of the proviral load together with the identification of proviral integration sites in STLV-1-infected NHPs have highlighted the clonal expansion of STLV-1 in vivo (Turpin et al., 2017). On the other hand, SFV is considered as non-pathogenic, but whether co-infections might regulate SFV replication is unknown. The aim of the project is to evaluate the influence of STLV-1 on the proviral load and the retroviral clonality of SFV in naturally infected baboons. First, we have reported that the SFV proviral load increases in the blood of baboons naturally co-infected with SFV and STLV-1 (Alais et al., 2018). We next demonstrate for the first time that SFV is able to replicate by clonal expansion of infected cells in vivo, as described for HTLV-1 and STLV-1. However, STLV-1 does not seem to promote clonal expansion of SFV in vivo. Lastly, in order to analyze if one unique cell can be co-infected by both SFV and STLV-1 in vivo, we develop a DNA-FISH (fluorescence in situ hybridization) approach. Altogether, this work sheds light on the replication of SFV in naturally infected NHPs, and emphasizes the fact that co-infection can significantly impact the replication of exogenous viruses in vivo, Les primates non-humains (PNH) sont les hôtes naturels de plusieurs rétrovirus exogènes tels que STLV-1 (virus de la leucémie simienne de type 1) et SFV (virus foamy simien), et certains cas de co-infections naturelles ont été répertoriés (SFV/STLV-1). Comme HTLV-1 chez l’Homme, STLV-1 est l’agent étiologique de l’ATL (leucémie à cellule T de l’adulte), mais la majorité des PNH infectés restent asymptomatiques. HTLV-1 promeut l’expansion clonale des cellules infectées et l’expansion clonale de STLV-1 a également été démontrée in vivo chez les PNH (Turpin et al., 2017). SFV, lui, provoque une infection asymptomatique, mais aucune donnée n'est disponible quant à la régulation de la réplication de SFV en cas de co-infection. L’objectif du projet consiste à évaluer l’influence de STLV-1 sur la charge provirale (PVL) et la clonalité rétrovirale de SFV chez des babouins naturellement infectés par SFV et STLV-1. Nous avons tout d'abord montré que la PVL de SFV augmente dans le sang des babouins naturellement co-infectés (Alais et al., 2018), par rapport aux animaux infectés uniquement par SFV. Nous avons ensuite démontré que SFV est capable de se répliquer par expansion clonale des cellules infectées in vivo et nos premiers résultats suggèrent que l’expansion clonale de SFV n’est pas favorisée par STLV-1. Enfin, une approche de DNA-FISH (Fluorescence in situ hybridization), nous permettra de déterminer si, in vivo, une même cellule peut être co-infectée par les deux rétrovirus SFV et STLV-1. Ce projet permet d’apporter des informations sur la réplication du virus SFV et met en avant l’influence des co-infections rétrovirales sur la réplication des rétrovirus exogènes in vivo
- Published
- 2020
18. Nonhuman primate retroviruses from Cambodia: High simian foamy virus prevalence, identification of divergent STLV-1 strains and no evidence of SIV infection.
- Author
-
Ayouba, Ahidjo, Duval, Linda, Liégeois, Florian, Ngin, Sopheak, Ahuka-Mundeke, Steve, Switzer, William M., Delaporte, Eric, Ariey, Frédéric, Peeters, Martine, and Nerrienet, Eric
- Subjects
- *
RETROVIRUSES , *VIRUS identification , *SIMIAN immunodeficiency virus , *SIMIAN viruses , *FOAMY viruses - Abstract
Highlights: [•] We tested 118 samples from 6 different primates species from Cambodia for retroviruses. [•] We showed no evidence of SIV infection. [•] We observed high prevalence of SFV infection complying with host/virus co-divergence. [•] We identified a well-supported clade of Cambodian STLV-1 from 3 different species. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
19. Simian Foamy Virus in Non-Human Primates and Cross-Species Transmission to Humans in Gabon: An Emerging Zoonotic Disease in Central Africa?
- Author
-
Mouinga-Ondémé, Augustin and Kazanji, Mirdad
- Subjects
- *
RETROVIRUSES , *SIMIAN foamy virus , *ZOONOSES , *PRIMATES , *RNA viruses , *ONCOGENIC viruses - Abstract
It is now known that all human retroviruses have a non-human primate counterpart. It has been reported that the presence of these retroviruses in humans is the result of interspecies transmission. Several authors have described the passage of a simian retrovirus, simian foamy virus (SFV), from primates to humans. To better understand this retroviral "zoonosis" in natural settings, we evaluated the presence of SFV in both captive and wild non-human primates and in humans at high risk, such as hunters and people bitten by a non-human primate, in Gabon, central Africa. A high prevalence of SFV was found in blood samples from non-human primates and in bush meat collected across the country. Mandrills were found to be highly infected with two distinct strains of SFV, depending on their geographical location. Furthermore, samples collected from hunters and non-human primate laboratory workers showed clear, extensive cross-species transmission of SFV. People who had been bitten by mandrills, gorillas and chimpanzees had persistent SFV infection with low genetic drift. Thus, SFV is presumed to be transmitted from non-human primates mainly through severe bites, involving contact between infected saliva and blood. In this review, we summarize and discuss our five-year observations on the prevalence and dissemination of SFV in humans and non-human primates in Gabon. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
20. Simian retroviruses in African apes.
- Author
-
Peeters, M. and Delaporte, E.
- Subjects
- *
RETROVIRUSES , *SIMIAN immunodeficiency virus , *SIMIAN foamy virus , *HIV infections , *APES - Abstract
Clin Microbiol Infect 2012; 18: 514-520 Abstract It is now well established that simian immunodeficiency viruses (SIVs) from chimpanzees (SIVcpz) and gorillas (SIVgor) from west Central Africa are at the origin of HIV-1/AIDS. Apes are also infected with other retroviruses, notably simian T-cell lymphotropic viruses (STLVs) and simian foamy viruses (SFVs), that can be transmitted to humans. We discuss the actual knowledge on SIV, STLV and SFV infections in chimpanzees, gorillas, and bonobos. We especially elaborate on how the recent development of non-invasive methods has allowed us to identify the reservoirs of the HIV-1 ancestors in chimpanzees and gorillas, and increased our knowledge of the natural history of SIV infections in chimpanzees. Multiple cross-species events with retroviruses from apes to humans have occurred, but only one transmission of SIVcpz from chimpanzees in south-eastern Cameroon spread worldwide, and is responsible for the actual HIV pandemic. Frequent SFV transmissions have been recently reported, but no human-to-human transmission has been documented yet. Because humans are still in contact with apes, identification of pathogens in wild ape populations can signal which pathogens may be cause risk for humans, and allow the development of serological and molecular assays with which to detect transmissions to humans. Finally, non-invasive sampling also allows the study of the impact of retroviruses and other pathogens on the health and survival of endangered species such as chimpanzees, gorillas, and bonobos. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
21. The infection of mammalian and insect cells with SFV bearing nsP1 palmitoylation mutations
- Author
-
Karo-Astover, Liis, Šarova, Oksana, Merits, Andres, and Žusinaite, Eva
- Subjects
- *
VIRUS diseases , *SEMLIKI Forest virus , *VIRAL replication , *VERTEBRATES , *CYTOPLASM , *CELL membranes ,MAMMAL cytology - Abstract
Abstract: Semliki Forest virus (SFV), an alphavirus, replicates in vertebrate host and mosquito vector cells. The virus-specific part of the replicase complex constitutes nonstructural proteins 1–4 (nsP1–nsP4) and is bound to cytoplasmic membranes by an amphipathic helix inside of nsP1 and through the palmitoylation of cysteine residues in nsP1. In mammalian cells, defects in these viral functions result in a nonviable phenotype or the emergence of second-site compensatory mutations that have a positive impact on SFV infection. In most cases, these second-site compensatory mutations were found to compensate for the defect caused by the absence of palmitoylation in mosquito cells (C6/36). In C6/36 cells, however, all palmitoylation-defective viruses had severely reduced synthesis of subgenomic RNA; at the same time, several of them had very efficient formation of defective interfering genomes. Analysis of C6/36 cells that individually expressed either wild type (wt) or palmitoylation-deficient nsP1 forms revealed that similar to mammalian cells, the wt nsP1 localized predominantly to the plasma membrane, whereas its mutant forms localized to the cytoplasm. In contrast to transfected mammalian cells, all forms of nsP1 induced the formation of filopodia-like structures on some, but not all, transfected mosquito cells. These findings indicate that the plasma membrane and associated host factors may have different roles in alphavirus replicase complex formation in mammalian and mosquito cells. In general, the lack of nsP1 palmitoylation had a less severe effect on the function of the replication complex in mammalian cells when compared with that in mosquito cells. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
22. Multiprotein genetic vaccine in the SIV- Macaca animal model: a promising approach to generate sterilizing immunity to HIV infection.
- Author
-
Maggiorella, Maria Teresa, Sernicola, Leonardo, Crostarosa, Federica, Belli, Roberto, Pavone-Cossut, Maria Rosaria, Macchia, Iole, Farcomeni, Stefania, Tenner-Racz, Klara, Racz, Paul, Ensoli, Barbara, and Titti, Fausto
- Subjects
- *
DNA vaccines , *VACCINATION , *AIDS vaccines , *IMMUNIZATION , *VIRAL replication , *SIMIAN viruses , *LENTIVIRUS diseases , *KRA - Abstract
Background Vaccine combining structural and regulatory proteins is an emerging approach to develop an HIV/AIDS vaccine and therefore, the immunogenicity and efficacy of two regimens of immunization combining structural (Gag/Pol, Env) and regulatory (Rev, Tat, Nef) Simian immunodeficiency virus (SIV) proteins were compared in cynomolgus monkeys. Methods Monkeys were immunized with Modified Vaccine Ankara vector (MVA-J5) (protocol 1) or with DNA, Semliki forest virus and MVA vectors (DNA/SFV/MVA) (protocol 2). At week 32, all monkeys were challenge intravenously (protocol 1) or intrarectally (protocol 2) with 50 MID50 of SIVmac251. Humoral, proliferative responses and in particular in protocol 2, the frequency of IFN-γ producing cells, were measured in all monkeys before and after the challenge. Results Both vaccine regimens elicited humoral and proliferative responses but failed to induce neutralizing antibodies. Upon intravenous challenge, two out of three MVA-J5 vaccinated monkeys exhibited a long-term control of the viral replication whereas DNA/SFV/MVA vaccine abrogated the virus replication up to undetectable level in three out of four vaccinated monkeys. A major contribution to this vaccine effect appeared to be the IFN-γ/ELISPOT responses to vaccine antigens (Gag, Rev Tat and Nef). Conclusions These results, indicate that multiprotein heterologous prime-boost vaccination can induce a robust vaccine-induced immunity able to abrogate virus replication. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
23. Recombinant alphaviruses as vectors for anti-tumour and anti-microbial immunotherapy
- Author
-
Riezebos-Brilman, Annelies, Mare, Arjan de, Bungener, Laura, Huckriede, Anke, Wilschut, Jan, and Daemen, Toos
- Subjects
- *
CANCER treatment , *IMMUNOTHERAPY , *IMMUNOLOGY , *IMMUNIZATION , *PAPILLOMAVIRUSES , *VIRUSES , *GENETIC transformation - Abstract
Abstract: Background: Vectors derived from alphaviruses are gaining interest for their high transfection potency and strong immunogenicity. Objectives: After a brief introduction on alphaviruses and their vectors, an overview is given on current preclinical immunotherapy studies using vector systems based on alphaviruses. The efficacy of alphavirus vectors in inducing immune responses will be illustrated by a more detailed description of immunization studies using recombinant Semliki Forest virus for the treatment of human papilloma virus-induced cervical cancer. Results: Immunization with recombinant alphavirus results in the induction of humoral and cellular immune responses against microbes, infected cells and cancer cells. Preclinical studies demonstrate that infectious diseases and cancer can be treated prophylactically as well as therapeutically. Conclusions: Alphavirus-based genetic immunization strategies are highly effective in animal model systems, comparing quite favourably with any other approach. Therefore, we hope and expect to see an efficient induction of tumour-or microbial immunity and a positive outcome in future clinical efficacy studies. [Copyright &y& Elsevier]
- Published
- 2006
- Full Text
- View/download PDF
24. MONITOREO DEL DESEMPEÑO DEL INVERSOR QUE CONFORMA EL PRIMER SISTEMA FOTOVOLTAICO INTERCONECTADO DE COLOMBIA, USANDO INSTRUMENTACION VIRTUAL.
- Author
-
Aristizábal, A. J. and Gordillo, G.
- Subjects
- *
ELECTRIC inverters , *ELECTRIC machinery monitoring , *PHOTOVOLTAIC power systems , *ELECTRIC generators - Abstract
This article presents results obtained from the performance monitoring of the inverter, part of the first Colombian interconnected photovoltaic system (PVS), which was installed and in operation since June 2003, at the Physics Department of the Universidad National de Colombia, Bogotá Campus; the PVS is constituted of a 980 Wp photovoltaic generator and a 1000 VA inverter. Data acquisition and analysis equipment was developed from virtual instrumentation capable of permanent monitoring parameters yielding information on the performance of the inverter (current and d.c voltage generated by the photovoltaic array, current and a.c voltage at inverter output, inverter efficiency, irradiance and ambient temperature measurements) and on the PVS efficiency. This equipment was developed by using hardware made up of Field Point modules and a NI-6024E data acquisition board, along with LabVIEW 7.1 graphic programming software. The results obtained, from a three-month monitoring, indicate the viability of the installation and operation of the interconnected to the local electric grid photovoltaic systems, as embedded systems capable of supporting the power supplied by the distribution grid. [ABSTRACT FROM AUTHOR]
- Published
- 2006
25. Induction of human papilloma virus E6/E7-specific cytotoxic T-lymphocyte activity in immune-tolerant, E6/E7-transgenic mice.
- Author
-
Riezebos-Brilman, A., Regts, J., Freyschmidt, E.-J., Dontje, B., Wilschut, J., and Daemen, T.
- Subjects
- *
PAPILLOMAVIRUSES , *TRANSGENIC mice , *ANIMAL models of cancer , *CANCER treatment - Abstract
Despite promising preclinical results of various therapeutic anticancer immunization strategies, these approaches may not be effective enough to eradicate tumors in cancer patients. While most animal models are based on fast-growing transplantable tumors, malignancies in, for example, cervical cancer patients in general develop much more slowly, which may lead to immune suppression and/or immune tolerance. As a consequence, the immunomodulating signal of any therapeutic immunization regimen should be sufficiently potent to overcome this immunocompromised condition. In previous studies, we demonstrated that an experimental vaccine against human papillomavirus (HPV)-induced cervical cancer, based on Semliki Forest virus (SFV), induces robust HPV-specific cellular immune responses in mice. Now we studied whether this strategy is potent enough to also prime a cellular immune response in immune-tolerant HPV transgenic mice, in which CTL activity cannot be induced using protein or DNA vaccines. We demonstrate that, depending on the route of immunization, SFV-expressing HPV16 E6 and E7 indeed has the capacity to induce HPV16 E7-specific cytotoxic T cells in HPV-transgenic mice.Gene Therapy (2005) 12, 1410–1414. doi:10.1038/sj.gt.3302536; published online 21 April 2005 [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
26. Codon optimization and mRNA amplification effectively enhances the immunogenicity of the hepatitis C virus nonstructural 3/4A gene.
- Author
-
Frelin, L, Ahlén, G, Alheim, M, Weiland, O, Barnfield, C, and Liljeström, P
- Subjects
- *
HEPATITIS C virus , *DNA vaccines , *GENES , *IMMUNITY , *TUMORS , *RNA editing - Abstract
We have recently shown that the NS3-based genetic immunogens should contain also hepatitis C virus (HCV) nonstructural (NS) 4A to utilize fully the immunogenicity of NS3. The next step was to try to enhance immunogenicity by modifying translation or mRNA synthesis. To enhance translation efficiency, a synthetic NS3/4A-based DNA (coNS3/4A-DNA) vaccine was generated in which the codon usage was optimized (co) for human cells. In a second approach, expression of the wild-type (wt) NS3/4A gene was enhanced by mRNA amplification using the Semliki forest virus (SFV) replicon (wtNS3/4A-SFV). Transient tranfections of human HepG2 cells showed that the coNS3/4A gene gave 11-fold higher levels of NS3 as compared to the wtNS3/4A gene when using the CMV promoter. We have previously shown that the presence of NS4A enhances the expression by SFV. Both codon optimization and mRNA amplification resulted in an improved immunogenicity as evidenced by higher levels of NS3-specific antibodies. This improved immunogenicity also resulted in a more rapid priming of cytotoxic T lymphocytes (CTLs). Since HCV is a noncytolytic virus, the functionality of the primed CTL responses was evaluated by an in vivo challenge with NS3/4A-expressing syngeneic tumor cells. The priming of a tumor protective immunity required an endogenous production of the immunogen and CD8+ CTLs, but was independent of B and CD4+ T cells. This model confirmed the more rapid in vivo activation of an NS3/4A-specific tumor-inhibiting immunity by codon optimization and mRNA amplification. Finally, therapeutic vaccination with the coNS3/4A gene using gene gun 6-12 days after injection of tumors significantly reduced the tumor growth in vivo. Codon optimization and mRNA amplification effectively enhances the overall immunogenicity of NS3/4A. Thus, either, or both, of these approaches should be utilized in an NS3/4A-based HCV genetic vaccine.Gene Therapy (2004) 11, 522-533. doi:10.1038/sj.gt.3302184 [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
27. A single-chain Fv intrabody provides functional protection against the effects of mutant protein in an organotypic slice culture model of Huntington's disease
- Author
-
Murphy, Robert C. and Messer, Anne
- Subjects
- *
HUNTINGTON disease , *NEURODEGENERATION , *PROTEINS , *CELL death - Abstract
Huntington''s disease (HD) is a progressive, hereditary, neurodegenerative disorder caused by an expanded polyglutamine tract in huntingtin protein, leading to misfolding and abnormal protein–protein interactions. Reducing the initial misfolding should lead to decreased pathogenesis. We show that malonate stress increases the number of dead or dying cells when organotypic slice cultures are transduced to express pathological-length huntingtin fragments. Co-transfected anti-HD single-chain Fv (sFv) intrabodies can reverse this HD-specific increase in malonate-induced morbidity. [Copyright &y& Elsevier]
- Published
- 2004
- Full Text
- View/download PDF
28. Differential Requirements of Rab5 and Rab7 for Endocytosis of Influenza and Other Enveloped Viruses.
- Author
-
Sieczkarski, Sara B. and Whittaker, Gary R.
- Subjects
- *
INFLUENZA viruses , *HELA cells - Abstract
Enveloped viruses often enter cells via endocytosis; however, specific endocytic trafficking pathway(s) for many viruses have not been determined. Here we demonstrate, through the use of dominant-negative Rab5 and Rab7, that influenza virus (Influenza A/WSN/33 (H1N1) and A/X-31 (H3N2)) requires both early and late endosomes for entry and subsequent infection in HeLa cells. Time-course experiments, monitoring viral ribonucleoprotein colocalization with endosomal markers, indicated that influenza exhibits a conventional endocytic uptake pattern – reaching early endosomes after approximately 10 min, and late endosomes after 40 min. Detection with conformation-specific hemagglutinin antibodies indicated that hemagglutinin did not reach a fusion-competent form until the virus had trafficked beyond early endosomes. We also examined two other enveloped viruses that are also pH-dependent for entry – Semliki Forest virus and vesicular stomatitis virus. In contrast to influenza virus, infection with both Semliki Forest virus and vesicular stomatitis virus was inhibited only by the expression of dominant negative Rab5 and not by dominant negative Rab7, indicating an independence of late endosome function for infection by these viruses. As a whole, these data provide a definitive characterization of influenza virus endocytic trafficking and show differential requirements for endocytic trafficking between pH-dependent enveloped viruses . [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
29. Pathogenesis of Semliki Forest virus encephalitis.
- Author
-
Fazakerley, John K
- Abstract
This article provides a review of the pathogenesis of Semliki Forest virus (SFV) encephalitis. In mice, outcome of infection varies according to age of the mouse and strain of the virus and can include acute encephalitis, subacute demyelinating meningoencephalomyelitis, and persistent subclinical central nervous system (CNS) infection. All strains of virus are virulent in mice infected < 12 days of age. The L10 strain is also virulent in mice > 14 days age, whereas the A7(74) strain is avirulent. The genetic difference between these strains maps to the nsp3 gene. For A7(74) virus, age-related virulence correlates with ability of CNS neurons to replicate virus and undergo apoptotic cell death. Immature developing neurons support complete virus replication but as neuronal populations and circuits mature in the postnatal brain, virus infection becomes progressively restricted and nonproductive. This restricted replication can be overcome by gold I compounds, which may function by inducing neuronal dedifferentiation to a state permissive for virus replication. Biochemical pathways associated with membrane biogenesis may be an important determinant of this effect. Infection of some developing neuronal populations results in apoptosis, whereas infection of mature neurons results in persistent infection. An active type-I interferon system prevents virus spread in extraneural tissues. An initial high-titer plasma viremia is controlled by immunoglobulin M (IgM) antibodies. Virus enters the brain across cerebral endothelial cells and initiates scattered foci of perivascular infection. The blood-brain barrier is disrupted. Neurons and oligodendrocytes are the cell types most frequently infected. Infectivity in the brain can be eliminated by IgG antibodies, though an active T-cell response is required for virus elimination. Lesions of inflammatory demyelination require the presence of CD8[sup +] T lymphocytes and probably result from destruction by these cells of virally infected oligodendrocytes. [ABSTRACT FROM AUTHOR]
- Published
- 2002
- Full Text
- View/download PDF
30. Designing immunotoxins for cancer therapy.
- Author
-
Pennell, Christopher and Erickson, Heidi
- Abstract
Immunotoxins are the rapeutic agents with a high degree of specificity and unique mechanism of action. An immunotoxin is achimeric protein consisting of a targeting moiety linked to a toxin. The targeting moiety selectively binds to a tumor cell and targets it for death via the attached toxin. Generally, immunotoxins are specifically potent against cancer cells in vitro and in animal models of human malignancies. However, immunotoxins can be limited clinically by immunogenicity, toxicity, and instability. In this review, weofferwaysto overcome these limitations to create “ideal immunotoxins” for cancer therapy. These include producing single chain targeting/toxin fusion proteins of fully human origin that are extracellularly stable but once internalized, can be cleaved by intracellular proteases to free the toxin and facilitate its translocation to the cytosol. [ABSTRACT FROM AUTHOR]
- Published
- 2002
- Full Text
- View/download PDF
31. Lack of expression of hepatitis C virus core protein in human monocyte-erived dendritic cells using recombinant semliki forest virus
- Author
-
Françoise Stoll-Keller, Maria-Cristina Navas Navas, and Jovan Pavlovic
- Subjects
QH301-705.5 ,Hepatitis C virus ,Hepacivirus ,viruses ,medicine.disease_cause ,Semliki Forest virus ,Dendritic cells ,Virus ,Viral vector ,03 medical and health sciences ,Transduction (genetics) ,chemistry.chemical_compound ,0302 clinical medicine ,Retrovirus ,medicine ,Biology (General) ,SFV ,030304 developmental biology ,0303 health sciences ,biology ,viral vector ,virus diseases ,biology.organism_classification ,Virology ,chemistry ,HCV ,030211 gastroenterology & hepatology ,Vaccinia ,General Agricultural and Biological Sciences - Abstract
Hepatitis C Virus belongs to the Flaviviridae family. One proposed mechanism of HCV persistence in the ability to infect hematopoietic cells, including Dendritic cells (DCs). HCV infection of DCs could impair their functions that represent one of the mechanisms, thus hampering viral clearance by the host immune system. Among HCV-encoded proteins, the highly conserved Core protein has been suggested to be responsible for the immunomodulatory properties of this Hepacivirus. Recombinant viral vectors expressing the HCV Core protein and allowing its transduction and therefore the expression of the protein into DCs could be useful tools for the analysis of the properties of the Core protein. Vaccinia Virus and retrovirus have been used to transduce human DCs. Likewise, gene transfer into DCs using Semliki Forest Virus has been reported. This study aimed to express the HCV Core protein in human monocyte-derived DCs using an SFV vector, in which the subgenomic RNA encoding the structural proteins was replaced by the HCV Core sequence and then analyze the effects of its expression on DCs functions.
- Published
- 2019
32. Development and characterization of immunological tools directed against membrane proteins of therapeutic interest
- Author
-
Hartmann, Lucie and STAR, ABES
- Subjects
[SDV.IMM] Life Sciences [q-bio]/Immunology ,Anticorps ,Nanodiscs ,Récepteur de l’adénosine A2A ,RCPG ,Adenosine A2A receptor ,Nanodisques ,GPCRs ,Melatonine MT1 receptor ,Antibodies ,Polyclonal serum ,Viral particles ,Particules virales ,[SDV.BIO] Life Sciences [q-bio]/Biotechnology ,Récepteur de la mélatonine MT1 ,Nanobodies ,Sérum polyclonal ,SFV - Abstract
G Protein Coupled Receptors (GPCRs) constitute the largest membrane protein family represented in the human genome. Their involvement in a wide number of biological processes fully supports their study. GPCR-targeting antibodies are versatile and valuable tools, which remain scarcely available, chiefly because their generation is a challenging process. This thesis presents an alternative and innovative strategy in which recombinant Semliki Forest Virus (SFV) particles coding for the receptor of interest are used as immunogens. When applied to the human version of the Adenosine A2A receptor, this method enables to cause the receptor’s overexpression at the surface of the infected animal cells, which generates an immune response. This strategy enables to raise receptor-specific mouse polyclonal serum. It opens a new path towards the generation of monoclonal mouse antibodies. Additionally, it seems to also be a promising approach to develop nanobodies., Les Récepteurs Couplés aux Protéines G (RCPG) constituent la plus grande famille de protéines membranaires chez l’Homme, et leur implication dans un grand nombre de processus physiologiques justifie pleinement l’intérêt de leur étude. Les anticorps spécifiques de ces récepteurs sont des outils polyvalents à haute valeur ajoutée, qui restent toutefois encore trop rarement disponibles, notamment en raison des difficultés techniques posées par leur génération. Ce manuscrit présente la mise au point d’une méthode d’immunisation alternative et innovante, mettant en jeu des particules virales recombinantes dérivées du Virus de la Forêt de Semliki (SFV) codant pour le récepteur d’intérêt. Appliquée au récepteur de l’adénosine A2A humain, l’immunisation permet d’engendrer la surexpression de celui-ci à la surface des cellules de l’animal infecté, et de provoquer l’apparition d’une réponse immunitaire. Cette approche permet d’une part de générer un sérum polyclonal de souris spécifique au récepteur, et ouvre donc une nouvelle voie pour l’obtention d’anticorps monoclonaux murins. Elle semble d’autre part prometteuse pour la génération de nanobodies.
- Published
- 2019
33. Bacterial aspects associated with the expression of a single-chain antibody fragment in Escherichia coli.
- Author
-
Somerville, J., Goshorn, S., Fell, H., and Darveau, R.
- Abstract
The bacterial expression of a single-chain antibody fragment, designated L6 sFv, was examined. Periplasmic targeting resulted in the production of a correctly folded protein that bound tumor antigen. However, immediately after induction at either 30°C or 37°C there was a significant loss in bacterial viability, which was followed by a loss in absorbance. The loss in absorbance correlated with cell lysis and release of the L6 sFv into the culture supernatant. The kinetics of appearance of L6 sFv in the supernatant paralleled that of periplasmic \-lactamase and confirmed an initial loss of cell-wall integrity prior to cell lysis. Bacteria incubated at 30°C produced approximately threefold more correctly folded antibody fragment because of an increase in the number of cells/ A at the lower incubation temperature. More than 95% of the L6 sFv, made at either incubation temperature, was incorrectly folded. Osmotic-shock procedures did not release L6 sFv. However, in situ subtilisin susceptibility experiments with bacterial spheroplasts confirmed a periplasmic location. French press disruption resulted in the release of correctly but not incorrectly folded material. Membrane fractionation revealed that the incorrectly folded L6 sFv remained associated with both the inner and outer membrane. These results demonstrate that, in this system, antibody fragment expression resulted initially in cell death, which was followed by release of protein into the culture supernatant and eventually cell lysis. It is also suggested that membrane association in the periplasmic space may impede proper folding. [ABSTRACT FROM AUTHOR]
- Published
- 1994
- Full Text
- View/download PDF
34. Insertion of the Type-I IFN Decoy Receptor B18R in a miRNA-Tagged Semliki Forest Virus Improves Oncolytic Capacity but Results in Neurotoxicity
- Author
-
Sarén, Tina, Ramachandran, Mohanraj, Martikainen, Miika, and Yu, Di
- Subjects
Medicin och hälsovetenskap ,Semliki Forest virus ,B18R ,viruses ,glioblastoma ,type I interferon ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,Medical and Health Sciences ,Article ,SFV - Abstract
Oncolytic Semliki Forest virus (SFV) has been suggested as a potential candidate for the treatment of glioblastoma and neuroblastoma. However, the oncolytic capacity of SFV is restricted by the anti-viral type-I interferon (IFN) response. The aim of this study was to increase the oncolytic capacity of a microRNA target tagged SFV against glioblastoma by arming it with the Vaccinia-virus-encoded type-I IFN decoy receptor B18R (SFV4B18RmiRT) to neutralize type-I IFN response. Expression of B18R by SFV4B18RmiRT aided neutralization of IFN-β, which was shown by reduced STAT-1 phosphorylation and improved virus spread in plaque assays. B18R expression by SFV4 increased its oncolytic capacity in vitro against murine glioblastoma (CT-2A), regardless of the presence of exogenous IFN-β. Both SFV4B18RmiRT and SFV4miRT treatments controlled tumor growth in mice with syngeneic orthotopic gliomablastoma (CT-2A). However, treatment with SFV4B18RmiRT induced severe neurological symptoms in some mice because of virus replication in the healthy brain. Neither neurotoxicity nor virus replication in the brain was observed when SFV4miRT was administered. In summary, our results indicate that the oncolytic capacity of SFV4 was improved in vitro and in vivo by incorporation of B18R, but neurotoxicity of the virus was increased, possibly due to loss of microRNA targets. Keywords: Semliki Forest virus, SFV, glioblastoma, type I interferon, B18R
- Published
- 2017
35. Contemporary Distribution, Estimated Age, and Prehistoric Migrations of Old World Monkey Retroviruses.
- Author
-
van der Kuyl AC
- Abstract
Old World monkeys (OWM), simians inhabiting Africa and Asia, are currently affected by at least four infectious retroviruses, namely, simian foamy virus (SFV), simian immunodeficiency virus (SIV), simian T-lymphotropic virus (STLV), and simian type D retrovirus (SRV). OWM also show chromosomal evidence of having been infected in the past with four more retroviral species, baboon endogenous virus (BaEV), Papio cynocephalus endogenous virus (PcEV), simian endogenous retrovirus (SERV), and Rhesus endogenous retrovirus-K (RhERV-K/SERV-K1). For some of the viruses, transmission to other primates still occurs, resulting, for instance, in the HIV pandemic. Retroviruses are intimately connected with their host as they are normally spread by close contact. In this review, an attempt to reconstruct the distribution and history of OWM retroviruses will be made. A literature overview of the species infected by any of the eight retroviruses as well as an age estimation of the pathogens will be given. In addition, primate genomes from databases have been re-analyzed for the presence of endogenous retrovirus integrations. Results suggest that some of the oldest retroviruses, SERV and PcEV, have travelled with their hosts to Asia during the Miocene, when a higher global temperature allowed simian expansions. In contrast, younger viruses, such as SIV and SRV, probably due to the lack of a primate continuum between the continents in later times, have been restricted to Africa and Asia, respectively.
- Published
- 2021
- Full Text
- View/download PDF
36. Simian Foamy Virus in Non-Human Primates and Cross-Species Transmission to Humans in Gabon: An Emerging Zoonotic Disease in Central Africa?
- Author
-
Mirdad Kazanji and Augustin Mouinga-Ondémé
- Subjects
Primates ,viruses ,lcsh:QR1-502 ,Cross-species transmission ,Simian foamy virus ,interspecies transmission ,Review ,central Africa ,lcsh:Microbiology ,Simian retrovirus ,Zoonoses ,Virology ,biology.animal ,Disease Transmission, Infectious ,medicine ,Animals ,Humans ,Primate ,Bites and Stings ,Gabon ,mandrills ,SFV ,wild-born non-human primates ,biology ,Transmission (medicine) ,Zoonosis ,Primate Diseases ,virus diseases ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,medicine.disease ,Infectious Diseases ,Non-human ,Retroviridae Infections - Abstract
It is now known that all human retroviruses have a non-human primate counterpart. It has been reported that the presence of these retroviruses in humans is the result of interspecies transmission. Several authors have described the passage of a simian retrovirus, simian foamy virus (SFV), from primates to humans. To better understand this retroviral "zoonosis" in natural settings, we evaluated the presence of SFV in both captive and wild non-human primates and in humans at high risk, such as hunters and people bitten by a non-human primate, in Gabon, central Africa. A high prevalence of SFV was found in blood samples from non-human primates and in bush meat collected across the country. Mandrills were found to be highly infected with two distinct strains of SFV, depending on their geographical location. Furthermore, samples collected from hunters and non-human primate laboratory workers showed clear, extensive cross-species transmission of SFV. People who had been bitten by mandrills, gorillas and chimpanzees had persistent SFV infection with low genetic drift. Thus, SFV is presumed to be transmitted from non-human primates mainly through severe bites, involving contact between infected saliva and blood. In this review, we summarize and discuss our five-year observations on the prevalence and dissemination of SFV in humans and non-human primates in Gabon.
- Published
- 2013
37. Alphavirus restriction by IFITM proteins
- Author
-
Weston, S, Czieso, S, White, IJ, Smith, SE, Wash, RS, Diaz-Soria, C, Kellam, P, and Marsh, M
- Subjects
Semliki Forest virus ,viruses ,SEMLIKI-FOREST-VIRUS ,restriction factor ,VIRAL ENTRY ,Endosomes ,virus entry ,0601 Biochemistry and Cell Biology ,TRANSMEMBRANE PROTEIN-3 ,IFITM ,FUSION ,interferon inducible transmembrane protein ,BHK-21-CELLS ,alphavirus ,Humans ,DENGUE VIRUS ,SFV ,Science & Technology ,virus-host interaction ,Alphavirus Infections ,Membrane Proteins ,RNA-Binding Proteins ,LOCALIZATION ,Cell Biology ,biochemical phenomena, metabolism, and nutrition ,Virus Internalization ,Antigens, Differentiation ,Endocytosis ,WEST NILE VIRUS ,A549 Cells ,CELLS ,REPLICATION ,Sindbis Virus ,Life Sciences & Biomedicine ,Developmental Biology - Abstract
Interferon inducible transmembrane proteins (IFITMs) are broad‐spectrum antiviral factors. In cell culture the entry of many enveloped viruses, including orthomyxo‐, flavi‐, and filoviruses, is inhibited by IFITMs, though the mechanism(s) involved remain unclear and may vary between viruses. We demonstrate that Sindbis and Semliki Forest virus (SFV), which both use endocytosis and acid‐induced membrane fusion in early endosomes to infect cells, are restricted by the early endosomal IFITM3. The late endosomal IFITM2 is less restrictive and the plasma membrane IFITM1 does not inhibit normal infection by either virus. IFITM3 inhibits release of the SFV capsid into the cytosol, without inhibiting binding, internalization, trafficking to endosomes or low pH‐induced conformational changes in the envelope glycoprotein. Infection by SFV fusion at the cell surface was inhibited by IFITM1, but was equally inhibited by IFITM3. Furthermore, an IFITM3 mutant (Y20A) that is localized to the plasma membrane inhibited infection by cell surface fusion more potently than IFITM1. Together, these results indicate that IFITMs, in particular IFITM3, can restrict alphavirus infection by inhibiting viral fusion with cellular membranes. That IFITM3 can restrict SFV infection by fusion at the cell surface equivalently to IFITM1 suggests that IFITM3 has greater antiviral potency against SFV.
- Published
- 2016
38. Alphavirus Restriction by IFITM Proteins
- Author
-
Stuart, Weston, Stephanie, Czieso, Ian J, White, Sarah E, Smith, Rachael S, Wash, Carmen, Diaz-Soria, Paul, Kellam, and Mark, Marsh
- Subjects
Semliki Forest virus ,Alphavirus Infections ,viruses ,Membrane Proteins ,RNA-Binding Proteins ,virus–host interaction ,Endosomes ,Original Articles ,restriction factor ,biochemical phenomena, metabolism, and nutrition ,Virus Internalization ,virus entry ,Antigens, Differentiation ,Endocytosis ,IFITM ,A549 Cells ,interferon inducible transmembrane protein ,Humans ,alphavirus ,Original Article ,Sindbis Virus ,SFV - Abstract
Interferon inducible transmembrane proteins (IFITMs) are broad‐spectrum antiviral factors. In cell culture the entry of many enveloped viruses, including orthomyxo‐, flavi‐, and filoviruses, is inhibited by IFITMs, though the mechanism(s) involved remain unclear and may vary between viruses. We demonstrate that Sindbis and Semliki Forest virus (SFV), which both use endocytosis and acid‐induced membrane fusion in early endosomes to infect cells, are restricted by the early endosomal IFITM3. The late endosomal IFITM2 is less restrictive and the plasma membrane IFITM1 does not inhibit normal infection by either virus. IFITM3 inhibits release of the SFV capsid into the cytosol, without inhibiting binding, internalization, trafficking to endosomes or low pH‐induced conformational changes in the envelope glycoprotein. Infection by SFV fusion at the cell surface was inhibited by IFITM1, but was equally inhibited by IFITM3. Furthermore, an IFITM3 mutant (Y20A) that is localized to the plasma membrane inhibited infection by cell surface fusion more potently than IFITM1. Together, these results indicate that IFITMs, in particular IFITM3, can restrict alphavirus infection by inhibiting viral fusion with cellular membranes. That IFITM3 can restrict SFV infection by fusion at the cell surface equivalently to IFITM1 suggests that IFITM3 has greater antiviral potency against SFV.
- Published
- 2016
39. Recombinant alphaviruses as vectors for anti-tumour and anti-microbial immunotherapy
- Author
-
Jan Wilschut, Toos Daemen, Arjan de Mare, Annelies Riezebos-Brilman, Laura Bungener, and Anke Huckriede
- Subjects
HPV ,animal structures ,medicine.medical_treatment ,viruses ,Genetic Vectors ,SEMLIKI-FOREST-VIRUS ,RESPIRATORY SYNCYTIAL VIRUS ,Alphavirus ,Infections ,Semliki Forest virus ,Cancer Vaccines ,Immune system ,Neoplasms ,Virology ,medicine ,Humans ,alphavirus ,Vector (molecular biology) ,SFV ,Infection Control ,Vaccines, Synthetic ,T-LYMPHOCYTE ACTIVITY ,biology ,IMMUNE-RESPONSES ,Immunogenicity ,REPLICON PARTICLES ,virus diseases ,VEE ,Immunotherapy ,biochemical phenomena, metabolism, and nutrition ,SIN ,biology.organism_classification ,ENCEPHALITIS-VIRUS ,SIMIAN IMMUNODEFICIENCY VIRUS ,Infectious Diseases ,Immunization ,Immunology ,Togaviridae ,immunotherapy ,MESSENGER-RNA ,SINDBIS-VIRUS ,ENHANCED IMMUNOGENICITY - Abstract
Background: Vectors derived from alphaviruses are gaining interest for their high transfection potency and strong immunogenicity. Objectives: After a brief introduction on alphaviruses and their vectors, an overview is given on current preclinical immunotherapy studies using vector systems based on alphaviruses. The efficacy of alphavirus vectors in inducing immune responses will be illustrated by a more detailed description of immunization studies using recombinant Semliki Forest virus for the treatment of human papilloma virus-induced cervical cancer. Results: Immunization with recombinant alphavirus results in the induction of humoral and cellular immune responses against microbes, infected cells and cancer cells. Preclinical studies demonstrate that infectious diseases and cancer can be treated prophylactically as well as therapeutically. Conclusions: Alphavirus-based genetic immunization stratagies are highly effective in animal model systems, comparing quite favourably with any other approach. Therefore, we hope and expect to see an efficient induction Of tumour-or microbial immunity and a positive outcome in future clinical efficacy studies. (c) 2005 Elsevier B.V. All rights reserved.
- Published
- 2006
40. Recombinant alphaviruses as vectors for anti-tumour and anti-microbial immunotherapy
- Subjects
HPV ,T-LYMPHOCYTE ACTIVITY ,IMMUNE-RESPONSES ,REPLICON PARTICLES ,SEMLIKI-FOREST-VIRUS ,RESPIRATORY SYNCYTIAL VIRUS ,VEE ,SIN ,ENCEPHALITIS-VIRUS ,SIMIAN IMMUNODEFICIENCY VIRUS ,alphavirus ,immunotherapy ,MESSENGER-RNA ,SINDBIS-VIRUS ,ENHANCED IMMUNOGENICITY ,SFV - Abstract
Background: Vectors derived from alphaviruses are gaining interest for their high transfection potency and strong immunogenicity.Objectives: After a brief introduction on alphaviruses and their vectors, an overview is given on current preclinical immunotherapy studies using vector systems based on alphaviruses. The efficacy of alphavirus vectors in inducing immune responses will be illustrated by a more detailed description of immunization studies using recombinant Semliki Forest virus for the treatment of human papilloma virus-induced cervical cancer.Results: Immunization with recombinant alphavirus results in the induction of humoral and cellular immune responses against microbes, infected cells and cancer cells. Preclinical studies demonstrate that infectious diseases and cancer can be treated prophylactically as well as therapeutically.Conclusions: Alphavirus-based genetic immunization stratagies are highly effective in animal model systems, comparing quite favourably with any other approach. Therefore, we hope and expect to see an efficient induction Of tumour-or microbial immunity and a positive outcome in future clinical efficacy studies. (c) 2005 Elsevier B.V. All rights reserved.
- Published
- 2006
41. Replizierende foamyvirale Vektoren zur Stimulation einer effektiven und permanenten Immunantwort gegen HIV und tumorassoziierte Antigene
- Author
-
Blochmann, Rico
- Subjects
foamy virus ,tumorassoziierte Antigene ,mice ,animal model ,610 Medizin ,HIV ,gene gun ,replicating vector ,hamster ,HSRV2 ,PFV ,vaccine ,CTL ,ddc:610 ,SFV - Abstract
Hinweis: Im Zeitraum vom 20.08.2015 bis zum 15.01.2016 war dem Datensatz eine veraltete Version der Dissertation beigefügt. Für wissenschaftliche Zwecke ist ausschließlich die nun beigefügte Version der Dissertation zu nutzen. Die alte Version muss gelöscht werden und darf nicht weiterverbreitet werden. Auf Basis der bislang zur Verfügung stehenden Methoden konnte weder ein effektiver HIV- Impfstoff entwickelt noch der durchschlagende Erfolg bei der Induktion einer anti-Tumorantwort erzielt werden. Aus diesem Grund ist es Aufgabe der Grundlagenforschung, innovative Lösungsansätze bereit zu stellen, um die Effektivität der Vakzinierung zu verbessern. Diesbezüglich zeigte der Einsatz von replikationskompetenten Vektorsystemen erste vielversprechende Ergebnisse im SIV-Primatenmodell. Hinsichtlich dieses Ansatzes war es Ziel dieser Dissertation, ein neues replika- tionskompetentes Impf-Vektor-System basierend auf simianen Foamyviren zu entwickeln, um eine effektive und permanente Immunantwort gegen definierte Antigene stimulieren zu können. Auf der Basis des foamyviralen Vektors pHSRV2/13 (PFV/HFV) erfolgte die Konstruktion von replizierenden PFV-ΔBet-Basisvektoren. In einer ersten Tierversuchsphase konnte durch ballis- tische Applikation der PFV-ΔBet-Deletionsmutanten via Gene Gun eine permanente PFV- gerichtete Antikörperantwort in Hamstern und Mäusen induziert werden. Die Entwicklung eines effektiven viralen Impf-Vektor-Systems bedingte maßgeblich die stabile Expression definierter Antigene unter Kontrolle der viralen Replikation. Durch die gezielte genetische Modifikation der PFV-ΔBet-Basisvektoren konnte eine Palette von Hybridviren mit variablen Replikations- und Antigen-Expressionseigenschaften generiert werden. Dabei war es möglich, eine stabile Expression von GFP und HIV-CTL-Epitopen über einen Zeitraum von 68 Tagen in vitro nachzuweisen. Im Anschluss erfolgte die Immunisierung von Hamstern und Mäusen mit den generierten PFV- HIV-Epitop-Hybridviren. Durch diese Hybridviren konnte eine permanente, PFV-gerichtete Anti- körperantwort über einen Zeitraum von knapp 400 Tagen induziert werden. Darüber hinaus ist es gelungen, die in vivo Replikation der generierten Impf-Vektoren über Re-Isolation des applizierten Impfvirus aus dem Gewebe eines infizierten Hamsters eindeutig nachzuwiesen. Einen endgültigen Beweis für die Funktionalität des generierten Impf-Vektor-Systems lieferte der geführte Nachweis einer induzierten HIV-Epitop-spezifischen T-Zellpopulation im Milzgewebe von immunisierten Mäusen knapp 400 Tage nach Applikation der Hybrid-Impfviren. Dabei wurde eine variable Immunantwort in Abhängigkeit zu den unterschiedlichen Eigenschaften der konstruierten Impf- Vektoren stimuliert. Dadurch konnte bewiesen werden, dass durch die Applikation von replizierenden foamyviralen Impf-Vektoren die Induktion einer starken und permanenten Immunabwehr prinzipiell möglich ist., Attempts to develop a successful HIV vaccine or induce an effective anti-tumor response using the methods available have so far been unsuccessful. The evaluation of innovative strategies aimed at improving the efficacy of vaccination is therefore an important aspect of basic research. Recent studies using replication vector systems in the SIV-primate model have, in this respect, been promising. The aim of this thesis was therefore to develop a novel replication-competent vaccine vector system based on simian foamy viruses able to induce an effective and enduring immune response against defined antigens. Replicating PFV-ΔBet vectors were constructed using the foamy viral vector pHSRV2/13 (PFV/HFV) as a basis. An initial animal experiment demonstrated the induction of a long-lasting PFV-specific antibody response in hamsters and mice by gene gun ballistic application of the PFV-ΔBet deletion mutants. In order to be effective, a vaccine viral vector of this kind requires the stable expression of defined antigens under the control of viral replication. A variety of hybrid viruses with differing replication and antigen expression characteristics were generated by specific genetic modification of the PFV-ΔBet basis vectors. Virus replication and the stable expression of GFP and an HIV-CTL epitope string could be demonstrated for up to 68 days in vitro. Hamsters and mice were therefore inoculated with the PFV-HIV epitope hybrid viruses and a persistent PFV-specific antibody response was observed over a period of nearly 400 days. Furthermore, in vivo replication of the vaccine vectors was demonstrated by re-isolation of the inoculated virus from the tissues of an infected hamster. The functionality of the vaccine vector system was clearly demonstrated by the presence of HIV epitope-specific T-cells in the spleens of mice immunized almost 400 days previously with the hybrid viruses. The strength of the response appeared to depend on the particular characteristics of the vaccine vectors. These data demonstrate that is in principle possible to induce a strong and persisting immunity by application of replicating foamy virus vaccine vectors.
- Published
- 2014
42. Clinical Applications of Phage-Derived sFvs and sFv Fusion Proteins
- Author
-
J Bhatia, R. H. J. Begent, A. Huhalov, SP Cooke, GM Boxer, RB Pedley, Surinder K. Sharma, A Mayer, Lisa Robson, KA Chester, AA Flynn, and Dir Spencer
- Subjects
Recombinant Fusion Proteins ,antibody targeting ,Clinical Biochemistry ,Immunoglobulin Variable Region ,Review ,Carcinoembryonic antigen ,CEA ,Peptide Library ,Carboxypeptidase-G2 ,Genetics ,fusion protein ,Humans ,cancer ,Radioimmunoguided surgery ,Molecular Biology ,Immunoglobulin Fragments ,sFv ,chemistry.chemical_classification ,Clinical Trials as Topic ,lcsh:R5-920 ,biology ,Genes, Immunoglobulin ,Biochemistry (medical) ,Adept ,General Medicine ,biology.organism_classification ,Virology ,Fusion protein ,chemistry ,Filamentous bacteriophage ,biology.protein ,Antibody ,Glycoprotein ,lcsh:Medicine (General) ,ADEPT - Abstract
Single chain Fv antibodies (sFvs) have been produced from filamentous bacteriophage libraries obtained from immunised mice. MFE-23, the most characterised of these sFvs, is reactive with carcinoembryonic antigen (CEA), a glycoprotein that is highly expressed in colorectal adenocarcinomas. MFE-23 has been expressed in bacteria and purified in our laboratory for two clinical trials; a gamma camera imaging trial using123I-MFE-23 and a radioimmunoguided surgery trial using125I-MFE-23, where tumour deposits are detected by a hand-held probe during surgery. Both these trials show MFE-23 is safe and effective in localising tumour deposits in patients with cancer. We are now developing fusion proteins which use MFE-23 to deliver a therapeutic moiety; MFE-23::CPG2 targets the enzyme carboxypeptidase G2 (CPG2) for use in the ADEPT (antibody directed enzyme prodrug therapy) system and MFE::TNFα aims to reduce sequestration and increase tumor concentrations of systemically administered TNFα.
- Published
- 2000
43. Inhibition of HIV-1 infection by down-regulation of the CXCR4 co-receptor using an intracellular single chain variable fragment against CXCR4
- Author
-
BouHamdan, M, Strayer, DS, Wei, D, Mukhtar, M, Duan, L-X, Hoxie, J, and Pomerantz, RJ
- Published
- 2001
- Full Text
- View/download PDF
44. Short-Term Local Expression of a PD-L1 Blocking Antibody from a Self-Replicating RNA Vector Induces Potent Antitumor Responses.
- Author
-
Ballesteros-Briones MC, Martisova E, Casales E, Silva-Pilipich N, Buñuales M, Galindo J, Mancheño U, Gorraiz M, Lasarte JJ, Kochan G, Escors D, Sanchez-Paulete AR, Melero I, Prieto J, Hernandez-Alcoceba R, Hervas-Stubbs S, and Smerdou C
- Subjects
- Animals, B7-H1 Antigen genetics, B7-H1 Antigen metabolism, Cell Line, Dependovirus genetics, Disease Models, Animal, Female, Genetic Therapy methods, Genetic Vectors administration & dosage, Humans, Immunomodulation drug effects, Immunophenotyping, Injections, Intralesional, Mice, Neoplasms pathology, Neoplasms therapy, Recombinant Fusion Proteins genetics, Semliki forest virus genetics, Survival Rate, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, Tumor Burden, Antibodies, Monoclonal genetics, Antibodies, Monoclonal pharmacology, B7-H1 Antigen antagonists & inhibitors, Gene Expression, Genetic Vectors genetics, Neoplasms genetics, Neoplasms immunology, RNA Viruses genetics
- Abstract
Immune checkpoint blockade has shown anti-cancer efficacy, but requires systemic administration of monoclonal antibodies (mAbs), often leading to adverse effects. To avoid toxicity, mAbs could be expressed locally in tumors. We developed adeno-associated virus (AAV) and Semliki Forest virus (SFV) vectors expressing anti-programmed death ligand 1 (aPDL1) mAb. When injected intratumorally in MC38 tumors, both viral vectors led to similar local mAb expression at 24 h, diminishing quickly in SFV-aPDL1-treated tumors. However, SFV-aPDL1 induced >40% complete regressions and was superior to AAV-aPDL1, as well as to aPDL1 mAb given systemically or locally. SFV-aPDL1 induced abscopal effects and was also efficacious against B16-ovalbumin (OVA). The higher SFV-aPDL1 antitumor activity could be related to local upregulation of interferon-stimulated genes because of SFV RNA replication. This was confirmed by combining local SFV-LacZ administration and systemic aPDL1 mAb, which provided higher antitumor effects than each separated agent. SFV-aPDL1 promoted tumor-specific CD8 T cells infiltration in both tumor models. In MC38, SFV-aPDL1 upregulated co-stimulatory markers (CD137/OX40) in tumor CD8 T cells, and its combination with anti-CD137 mAb showed more pronounced antitumor effects than each single agent. These results indicate that local transient expression of immunomodulatory mAbs using non-propagative RNA vectors inducing type I interferon (IFN-I) responses represents a potent and safe approach for cancer treatment., (Copyright © 2019 The American Society of Gene and Cell Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
45. Characterization of New Simian Foamy Viruses from African Nonhuman Primates
- Author
-
Jonathan S. Allan, Karen L. Leighton, Anthony G. Comuzzie, Suzanne R. Broussard, M. Michelle Leland, and Evelyn M. Whitehead
- Subjects
Primates ,Hamlyn's guenon ,viruses ,Molecular Sequence Data ,Gene Products, gag ,Simian foamy virus ,Simian ,phylogeny ,Virus ,Patas monkey ,biology.animal ,Virology ,Animals ,Spumavirus ,SFV ,Base Sequence ,baboon ,biology ,patas ,spumavirus ,biology.organism_classification ,Guenon ,African green monkey ,Africa ,African Green Monkey ,Sequence Alignment ,Baboon - Abstract
Simian foamy viruses (SFV) are exogenous retroviruses present in most if not all nonhuman primate species. Baboons and other African monkey species are known to harbor SFVs, yet there is presently no data in regard to their genetic relationship. Here we studied SFVs from baboons as compared to other SFVs isolated from a Hamlyn's guenon, a patas monkey, and a vervet. By Western blot analysis, the gag precursor proteins (p74/p70) were detected from all SFVs. In addition, the envelope glycoproteins from a vervet isolate (SFV-Agm2) were comparable in size to the env precursor gp130, the exterior glycoprotein (gp70), and the transmembrane protein (gp48) as detected by lentil lectin binding and radioimmunoprecipitation (RIPA). Molecular comparison of PCR amplified products from pol and LTR regions of each SFV demonstrated a close relationship among baboon SFVs while SFVs from patas, Hamlyn's guenon, and vervet clustered together. The baboon viruses only varied by 4% among each other in the LTR region; however, as much as 26% variation was noted when compared to the other African monkey SFVs. To determine the prevalence rate of SFV-Bab in our baboon colony, we employed both Western blotting and PCR analysis. Antibodies to SFV gag precursor proteins were seen in 7 of 10 infants; however, none were positive by PCR, suggesting that these infants were virus negative and that their antibodies were maternal in origin. Only one juvenile (1/10) and all adults (38/38) were infected with SFV. Taken together these results suggest that SFVs have arisen and diverged along with the evolution of their natural hosts. Furthermore, the high prevalence rates to SFV seen in adult baboons strongly suggest a sexual or oral routes of transmission.
- Published
- 1997
- Full Text
- View/download PDF
46. Semliki forest virus as a vector: pros and cons for its use in biopharmaceuticals production
- Author
-
Renato Mancini Astray, Eutimio Gustavo Fernández Núñez, Aldo Tonso, Alexandre Gonçalves de Rezende, Soraia Attie Calil Jorge, Bruno Labate Vale da Costa, Carlos Augusto Pereira, Daniella Cristina Ventini Monteiro, Universidade de São Paulo (USP), Universidade Estadual Paulista (UNESP), Instituto Butantan, Instituto de Pesquisas Tecnológicas do Estado de São Paulo, Universidade Estadual Paulista (Unesp), and Inst Pesquisas Tecnol Estado Sao Paulo SA
- Subjects
BHK-21 ,Recombinant proteins ,Multidisciplinary ,lcsh:Biotechnology ,viruses ,Large-scale bioprocesses ,Mammalian cells ,Transient expression ,Biology ,Semliki Forest virus ,biology.organism_classification ,Virology ,Viral vector ,Biopharmaceutical industry ,lcsh:TP248.13-248.65 ,Vector (molecular biology) ,MAMÍFEROS ,High level expression ,SFV - Abstract
Made available in DSpace on 2014-12-03T13:11:37Z (GMT). No. of bitstreams: 0 Previous issue date: 2013-09-01Bitstream added on 2014-12-03T13:22:33Z : No. of bitstreams: 1 S1516-89132013000500018.pdf: 107013 bytes, checksum: 0a4b1e7c2d2c2bac93af970e5ee6fbc7 (MD5) Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP) Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) The number of biopharmaceuticals for medical and veterinarian use produced in mammalian cells is increasing year after year. All of them are obtained by stable recombinant cell lines. However, it is recognized that transient gene expression produces high level expression in a short time. In that sense, viral vectors have been extensively used for producing recombinant proteins on lab-scale. Among them, Semliki Forest virus is commonly employed for this purpose. This review discusses the main aspects related to the use of Semliki Forest virus technology as well as its advantages and drawbacks which limit currently its utilization in biopharmaceutical industry on large-scale. Univ Sao Paulo, Escola Politecn, Dept Engn Quim, Lab Celulas Anim, Sao Paulo, Brazil Univ Estadual Paulista Julho de Mesquita Filho, Fac Ciencia & Letras, Dept Ciencias Biol, Assis, SP, Brazil Inst Butantan, Lab Imunol Viral, Sao Paulo, Brazil Inst Pesquisas Tecnol Estado Sao Paulo SA, Nucleo Bionanomanufatura, Lab Biotecnol Ind, Sao Paulo, Brazil Univ Estadual Paulista Julho de Mesquita Filho, Fac Ciencia & Letras, Dept Ciencias Biol, Assis, SP, Brazil FAPESP: 10/52521-6
- Published
- 2013
47. Nonhuman primate retroviruses from Cambodia: high simian foamy virus prevalence, identification of divergent STLV-1 strains and no evidence of SIV infection
- Author
-
Linda Duval, Ahidjo Ayouba, Sopheak Ngin, Eric Nerrienet, Frédéric Ariey, Florian Liegeois, Eric Delaporte, Martine Peeters, William M. Switzer, and Steve Ahuka-Mundeke
- Subjects
Microbiology (medical) ,Sequence analysis ,viruses ,Molecular Sequence Data ,Simian foamy virus ,Simian ,Antibodies, Viral ,STLV ,Microbiology ,Macaque ,Serology ,Simian retrovirus ,biology.animal ,Hylobates ,Retroviruses ,Catarrhini ,Genetics ,Prevalence ,Animals ,Molecular Biology ,Nonhuman primates ,SFV ,Ecology, Evolution, Behavior and Systematics ,Phylogeny ,Phylogenetic tree ,biology ,Primate Diseases ,biology.organism_classification ,Virology ,Lorisidae ,Retroviruses, Simian ,Tumor Virus Infections ,Infectious Diseases ,SIV ,DNA, Viral ,Cambodia ,Retroviridae Infections - Abstract
Nonhuman primates (NHPs) carry retroviruses such as simian immunodeficiency viruses (SIV), simian T-cell lymphotropic viruses (STLV) and simian foamy viruses (SFV). Here, we revisited NHPs from Cambodia to assess the prevalence and diversity of these retroviruses using updated viral detection tools. We screened blood from 118 NHPs consisting of six species (Macaca fascicularis (n = 91), Macaca leonine (n = 8), Presbytis cristata (n = 3), Nycticebus coucang (n = 1), Hylobates pileatus (n = 14), and Pongo pygmaeus) (n = 1) by using a Luminex-based multiplex serology assay that allows the detection of all known SIV/HIV and SFV lineages. We also used highly sensitive PCR assays to detect each simian retrovirus group. Positive PCR products were sequenced and phylogenetically analyzed to infer evolutionary histories. Fifty-three of 118 (44.9%) NHPs tested positive for SFV by serology and 8/52 (15.4%), all from M. fascicularis, were PCR-confirmed. The 8 novel SFV sequences formed a highly supported distinct lineage within a clade composed of other macaque SFV. We observed no serological or molecular evidence of SIV infection among the 118 NHP samples tested. Four of 118 (3.3%) NHPs were PCR-positive for STLV, including one M. fascicularis, one P. cristata, and two H. pileatus. Phylogenetic analyses revealed that the four novel STLV belonged to the PTLV-1 lineage, outside the African radiation of PTLV-1, like all Asian PTLV identified so far. Sequence analysis of the whole STLV-1 genome from a H. pileatus (C578_Hp) revealed a genetic structure characteristic of PTLV. Similarity analysis comparing the STLV-1 (C578_Hp) sequence with prototype PTLVs showed that C578_Hp is closer to PTLV-1s than to all other types across the entire genome. In conclusion, we showed a high frequency of SFV infection but found no evidence of SIV infection in NHPs from Cambodia. We identified for the first time STLV-1 in a P. cristata and in two H. pileatus.
- Published
- 2013
48. Simian retroviruses in African apes
- Author
-
Eric Delaporte and Martine Peeters
- Subjects
Microbiology (medical) ,Disease reservoir ,Pan troglodytes ,viruses ,bonobo ,Gorilla ,Simian ,STLV ,Acquired immunodeficiency syndrome (AIDS) ,chimpanzee ,Zoonoses ,biology.animal ,Pandemic ,Disease Transmission, Infectious ,medicine ,Animals ,Humans ,SFV ,Disease Reservoirs ,Molecular Epidemiology ,Gorilla gorilla ,biology ,Molecular epidemiology ,Transmission (medicine) ,Bonobo ,Primate Diseases ,General Medicine ,Pan paniscus ,biology.organism_classification ,medicine.disease ,gorilla ,Virology ,Retroviruses, Simian ,Infectious Diseases ,SIV ,Africa ,Retroviridae Infections - Abstract
It is now well established that simian immunodeficiency viruses (SIVs) from chimpanzees (SIVcpz) and gorillas (SIVgor) from west Central Africa are at the origin of HIV-1/AIDS. Apes are also infected with other retroviruses, notably simian T-cell lymphotropic viruses (STLVs) and simian foamy viruses (SFVs), that can be transmitted to humans. We discuss the actual knowledge on SIV, STLV and SFV infections in chimpanzees, gorillas, and bonobos. We especially elaborate on how the recent development of non-invasive methods has allowed us to identify the reservoirs of the HIV-1 ancestors in chimpanzees and gorillas, and increased our knowledge of the natural history of SIV infections in chimpanzees. Multiple cross-species events with retroviruses from apes to humans have occurred, but only one transmission of SIVcpz from chimpanzees in south-eastern Cameroon spread worldwide, and is responsible for the actual HIV pandemic. Frequent SFV transmissions have been recently reported, but no human-to-human transmission has been documented yet. Because humans are still in contact with apes, identification of pathogens in wild ape populations can signal which pathogens may be cause risk for humans, and allow the development of serological and molecular assays with which to detect transmissions to humans. Finally, non-invasive sampling also allows the study of the impact of retroviruses and other pathogens on the health and survival of endangered species such as chimpanzees, gorillas, and bonobos.
- Published
- 2012
49. Induction of membrane proliferation in mouse CNS by gold sodium thiomalate with reference to increased virulence of the avirulent Semliki Forest virus
- Author
-
Mehta, S., Pathak, S., and Webb, H. E.
- Published
- 1990
- Full Text
- View/download PDF
50. Induction of human papilloma virus E6/E7-specific cytotoxic T-lymphocyte activity in immune-tolerant, E6/E7-transgenic mic
- Author
-
Annelies Riezebos-Brilman, Joke Regts, EJ Freyschmidt, Toos Daemen, Bert Dontje, Jan Wilschut, and Targeted Gynaecologic Oncology (TARGON)
- Subjects
immune tolerance ,FUSION PROTEIN ,Papillomavirus E7 Proteins ,viruses ,Lymphocyte Activation ,Immune tolerance ,Mice ,Cytotoxic T cell ,E7 ,E6 ,TRANSGENIC MICE ,virus diseases ,LANGERHANS CELLS ,HPV-transgenic mice ,IMMUNIZATION ,Injections, Intravenous ,Molecular Medicine ,VACCINATION ,Female ,HPV ,CERVICAL-CANCER ,ANTIGEN ,Mice, Transgenic ,chemical and pharmacologic phenomena ,INNATE ,Biology ,Semliki Forest virus ,Cancer Vaccines ,Injections, Intramuscular ,DNA vaccination ,Immune system ,Antigen ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,SFV ,Papillomavirus Infections ,Cancer ,Genetic Therapy ,Oncogene Proteins, Viral ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,medicine.disease ,Semliki forest virus ,Repressor Proteins ,Tumor Virus Infections ,Immunization ,Immunology ,CTL ,bacteria ,T-Lymphocytes, Cytotoxic - Abstract
Despite promising preclinical results of various therapeutic anticancer immunization strategies, these approaches may not be effective enough to eradicate tumors in cancer patients. While most animal models are based on fast-growing transplantable tumors, malignancies in, for example, cervical cancer patients in general develop much more slowly, which may lead to immune suppression and/or immune tolerance. As a consequence, the immunomodulating signal of any therapeutic immunization regimen should be sufficiently potent to overcome this immunocompromised condition. In previous studies, we demonstrated that an experimental vaccine against human papillomavirus (HPV)-induced cervical cancer, based on Semliki Forest virus (SFV), induces robust HPV-specific cellular immune responses in mice. Now we studied whether this strategy is potent enough to also prime a cellular immune response in immune-tolerant HPV transgenic mice, in which CTL activity cannot be induced using protein or DNA vaccines. We demonstrate that, depending on the route of immunization, SFV-expressing HPV16 E6 and E7 indeed has the capacity to induce HPV16 E7-specific cytotoxic T cells in HPV-transgenic mice.
- Published
- 2005
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.