Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Vet Res ; 55(1): 32, 2024 Mar 16.
Article in English | MEDLINE | ID: mdl-38493182

ABSTRACT

Outbreaks of West Nile virus (WNV) occur periodically, affecting both human and equine populations. There are no vaccines for humans, and those commercialised for horses do not have sufficient coverage. Specific antiviral treatments do not exist. Many drug discovery studies have been conducted, but since rodent or primate cell lines are normally used, results cannot always be transposed to horses. There is thus a need to develop relevant equine cellular models. Here, we used induced pluripotent stem cells to develop a new in vitro model of WNV-infected equine brain cells suitable for microplate assay, and assessed the cytotoxicity and antiviral activity of forty-one chemical compounds. We found that one nucleoside analog, 2'C-methylcytidine, blocked WNV infection in equine brain cells, whereas other compounds were either toxic or ineffective, despite some displaying anti-viral activity in human cell lines. We also revealed an unexpected proviral effect of statins in WNV-infected equine brain cells. Our results thus identify a potential lead for future drug development and underscore the importance of using a tissue- and species-relevant cellular model for assessing the activity of antiviral compounds.


Subject(s)
Horse Diseases , Induced Pluripotent Stem Cells , West Nile Fever , West Nile virus , Animals , Horses , Humans , West Nile Fever/veterinary , West Nile Fever/epidemiology , Brain , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Horse Diseases/drug therapy
2.
Int J Mol Sci ; 25(3)2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38338659

ABSTRACT

Vaccinia virus (Orthopoxvirus) F17 protein is a major virion structural phosphoprotein having a molecular weight of 11 kDa. Recently, it was shown that F17 synthesised in infected cells interacts with mTOR subunits to evade cell immunity and stimulate late viral protein synthesis. Several years back, we purified an 11 kDa protein that inhibited protein synthesis in reticulocyte lysate from virions, and that possesses all physico-chemical properties of F17 protein. To investigate this discrepancy, we used defective vaccinia virus particles devoid of the F17 protein (designated iF17- particles) to assess their ability to inhibit protein synthesis. To this aim, we purified iF17- particles from cells infected with a vaccinia virus mutant which expresses F17 only in the presence of IPTG. The SDS-PAGE protein profiles of iF17- particles or derived particles, obtained by solubilisation of the viral membrane, were similar to that of infectious iF17 particles. As expected, the profiles of full iF17- particles and those lacking the viral membrane were missing the 11 kDa F17 band. The iF17- particles did attach to cells and injected their viral DNA into the cytoplasm. Co-infection of the non-permissive BSC40 cells with a modified vaccinia Ankara (MVA) virus, expressing an mCherry protein, and iF17- particles, induced a strong mCherry fluorescence. Altogether, these experiments confirmed that the iF17- particles can inject their content into cells. We measured the rate of protein synthesis as a function of the multiplicity of infection (MOI), in the presence of puromycin as a label. We showed that iF17- particles did not inhibit protein synthesis at high MOI, by contrast to the infectious iF17 mutant. Furthermore, the measured efficiency to inhibit protein synthesis by the iF17 mutant virus generated in the presence of IPTG, was threefold to eightfold lower than that of the wild-type WR virus. The iF17 mutant contained about threefold less F17 protein than wild-type WR. Altogether these results strongly suggest that virion-associated F17 protein is essential to mediate a stoichiometric inhibition of protein synthesis, in contrast to the late synthesised F17. It is possible that this discrepancy is due to different phosphorylation states of the free and virion-associated F17 protein.


Subject(s)
Vaccinia virus , Vaccinia , Humans , Vaccinia virus/genetics , Vaccinia/genetics , Isopropyl Thiogalactoside , Cell Line , Phosphoproteins , Virion/genetics
3.
Viruses ; 13(11)2021 11 10.
Article in English | MEDLINE | ID: mdl-34835061

ABSTRACT

Tick-borne encephalitis virus (TBEV), a member of the Flaviviridae family, Flavivirus genus, is responsible for neurological symptoms that may cause permanent disability or death. With an incidence on the rise, it is the major arbovirus affecting humans in Central/Northern Europe and North-Eastern Asia. Neuronal death is a critical feature of TBEV infection, yet little is known about the type of death and the molecular mechanisms involved. In this study, we used a recently established pathological model of TBEV infection based on human neuronal/glial cells differentiated from fetal neural progenitors and transcriptomic approaches to tackle this question. We confirmed the occurrence of apoptotic death in these cultures and further showed that genes involved in pyroptotic death were up-regulated, suggesting that this type of death also occurs in TBEV-infected human brain cells. On the contrary, no up-regulation of major autophagic genes was found. Furthermore, we demonstrated an up-regulation of a cluster of genes belonging to the extrinsic apoptotic pathway and revealed the cellular types expressing them. Our results suggest that neuronal death occurs by multiple mechanisms in TBEV-infected human neuronal/glial cells, thus providing a first insight into the molecular pathways that may be involved in neuronal death when the human brain is infected by TBEV.


Subject(s)
Apoptosis , Encephalitis Viruses, Tick-Borne/pathogenicity , Neuroglia/virology , Neurons/virology , Pyroptosis , Apoptosis/genetics , Astrocytes/metabolism , Humans , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Pyroptosis/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , Transcriptome
4.
Vet Res ; 52(1): 65, 2021 May 03.
Article in English | MEDLINE | ID: mdl-33941270

ABSTRACT

The brain is a complex organ and any model for studying it in its normal and pathological aspects becomes a tool of choice for neuroscientists. The mastering and dissemination of protocols allowing brain organoids development have paved the way for a whole range of new studies in the field of brain development, modeling of neurodegenerative or neurodevelopmental diseases, understanding tumors as well as infectious diseases that affect the brain. While studies are so far limited to the use of human cerebral organoids, there is a growing interest in having similar models in other species. This review presents what is currently developed in this field, with a particular focus on the potential of cerebral organoids for studying neuro-infectious diseases in human and domestic animals.


Subject(s)
Animals, Domestic , Brain Diseases , Brain , Organoids , Animals , Brain/pathology , Brain/physiology , Brain/physiopathology , Brain Diseases/pathology , Brain Diseases/physiopathology , Humans , Organoids/pathology , Organoids/physiology , Organoids/physiopathology
5.
Clin Infect Dis ; 72(10): 1701-1708, 2021 05 18.
Article in English | MEDLINE | ID: mdl-32516409

ABSTRACT

BACKGROUND: Human encephalitis represents a medical challenge from a diagnostic and therapeutic point of view. We investigated the cause of 2 fatal cases of encephalitis of unknown origin in immunocompromised patients. METHODS: Untargeted metatranscriptomics was applied on the brain tissue of 2 patients to search for pathogens (viruses, bacteria, fungi, or protozoans) without a prior hypothesis. RESULTS: Umbre arbovirus, an orthobunyavirus never previously identified in humans, was found in 2 patients. In situ hybridization and reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR) showed that Umbre virus infected neurons and replicated at high titers. The virus was not detected in cerebrospinal fluid by RT-qPCR. Viral sequences related to Koongol virus, another orthobunyavirus close to Umbre virus, were found in Culex pipiens mosquitoes captured in the south of France where the patients had spent some time before the onset of symptoms, demonstrating the presence of the same clade of arboviruses in Europe and their potential public health impact. A serological survey conducted in the same area did not identify individuals positive for Umbre virus. The absence of seropositivity in the population may not reflect the actual risk of disease transmission in immunocompromised individuals. CONCLUSIONS: Umbre arbovirus can cause encephalitis in immunocompromised humans and is present in Europe.


Subject(s)
Agammaglobulinemia , Encephalitis , Orthobunyavirus , Viruses , Animals , Europe , France/epidemiology , Humans , Orthobunyavirus/genetics
6.
J Neuroinflammation ; 17(1): 76, 2020 Mar 03.
Article in English | MEDLINE | ID: mdl-32127025

ABSTRACT

BACKGROUND: Tick-borne encephalitis virus (TBEV) is a member of the Flaviviridae family, Flavivirus genus, which includes several important human pathogens. It is responsible for neurological symptoms that may cause permanent disability or death, and, from a medical point of view, is the major arbovirus in Central/Northern Europe and North-Eastern Asia. TBEV tropism is critical for neuropathogenesis, yet little is known about the molecular mechanisms that govern the susceptibility of human brain cells to the virus. In this study, we sought to establish and characterize a new in vitro model of TBEV infection in the human brain and to decipher cell type-specific innate immunity and its relation to TBEV tropism and neuropathogenesis. METHOD: Human neuronal/glial cells were differentiated from neural progenitor cells and infected with the TBEV-Hypr strain. Kinetics of infection, cellular tropism, and cellular responses, including innate immune responses, were characterized by measuring viral genome and viral titer, performing immunofluorescence, enumerating the different cellular types, and determining their rate of infection and by performing PCR array and qRT-PCR. The specific response of neurons and astrocytes was analyzed using the same approaches after enrichment of the neuronal/glial cultures for each cellular subtype. RESULTS: We showed that infection of human neuronal/glial cells mimicked three major hallmarks of TBEV infection in the human brain, namely, preferential neuronal tropism, neuronal death, and astrogliosis. We further showed that these cells conserved their capacity to mount an antiviral response against TBEV. TBEV-infected neuronal/glial cells, therefore, represented a highly relevant pathological model. By enriching the cultures for either neurons or astrocytes, we further demonstrated qualitative and quantitative differential innate immune responses in the two cell types that correlated with their particular susceptibility to TBEV. CONCLUSION: Our results thus reveal that cell type-specific innate immunity is likely to contribute to shaping TBEV tropism for human brain cells. They describe a new in vitro model for in-depth study of TBEV-induced neuropathogenesis and improve our understanding of the mechanisms by which neurotropic viruses target and damage human brain cells.


Subject(s)
Astrocytes/immunology , Astrocytes/virology , Encephalitis, Tick-Borne/immunology , Encephalitis, Tick-Borne/virology , Neurons/immunology , Neurons/virology , Cell Culture Techniques/methods , Cells, Cultured , Disease Susceptibility , Encephalitis Viruses, Tick-Borne/physiology , Humans , Immunity, Innate , Viral Tropism
7.
Viruses ; 12(1)2019 12 24.
Article in English | MEDLINE | ID: mdl-31878129

ABSTRACT

Neurological disorders represent an important sanitary and economic threat for the equine industry worldwide. Among nervous diseases, viral encephalitis is of growing concern, due to the emergence of arboviruses and to the high contagiosity of herpesvirus-infected horses. The nature, severity and duration of the clinical signs could be different depending on the etiological agent and its virulence. However, definite diagnosis generally requires the implementation of combinations of direct and/or indirect screening assays in specialized laboratories. The equine practitioner, involved in a mission of prevention and surveillance, plays an important role in the clinical diagnosis of viral encephalitis. The general management of the horse is essentially supportive, focused on controlling pain and inflammation within the central nervous system, preventing injuries and providing supportive care. Despite its high medical relevance and economic impact in the equine industry, vaccines are not always available and there is no specific antiviral therapy. In this review, the major virological, clinical and epidemiological features of the main neuropathogenic viruses inducing encephalitis in equids in Europe, including rabies virus (Rhabdoviridae), Equid herpesviruses (Herpesviridae), Borna disease virus (Bornaviridae) and West Nile virus (Flaviviridae), as well as exotic viruses, will be presented.


Subject(s)
Encephalomyelitis, Equine/veterinary , Horse Diseases/epidemiology , Horse Diseases/virology , Viruses/pathogenicity , Animals , Arboviruses/pathogenicity , Bornaviridae/pathogenicity , Encephalomyelitis, Equine/complications , Encephalomyelitis, Equine/epidemiology , Europe/epidemiology , Herpesviridae/pathogenicity , Horses , Humans , Rhabdoviridae/pathogenicity , Viruses/classification , West Nile virus/pathogenicity
8.
Emerg Microbes Infect ; 8(1): 1003-1016, 2019.
Article in English | MEDLINE | ID: mdl-31282298

ABSTRACT

Zika virus (ZIKV) is a mosquito-borne Flavivirus that causes Zika disease with particular neurological complications, including Guillain-Barré Syndrome and congenital microcephaly. Although ZIKV has been shown to directly infect human neural progenitor cells (hNPCs), thereby decreasing their viability and growth, it is as yet unknown which of the cellular pathways involved in the disruption of neurogenesis are affected following ZIKV infection. By comparing the effect of two ZIKV strains in vitro on hNPCs, the differentiation process of the latter cells was found to lead to a decreased susceptibility to infection and cell death induced by each of the ZIKV strains, which was associated with an earlier and stronger antiviral innate immune response in infected, differentiated hNPCs, as compared to undifferentiated cells. Moreover, ZIKV modulated, both in hNPCs and in vivo in fetal brain in an experimental mouse model, the expression of the Notch pathway which is involved in cellular proliferation, apoptosis and differentiation during neurogenesis. These results show that the differentiation state of hNPCs is a significant factor contributing to the outcome of ZIKV infection and furthermore suggest that ZIKV infection might initiate early activation of the Notch pathway resulting in an abnormal differentiation process, implicated in ZIKV-induced brain injury.


Subject(s)
Neural Stem Cells/virology , Neurogenesis , Receptor, Notch1/metabolism , Zika Virus Infection/virology , Zika Virus/physiology , Animals , Apoptosis , Female , Humans , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Receptor, Notch1/genetics , Signal Transduction , Zika Virus/genetics , Zika Virus Infection/genetics , Zika Virus Infection/metabolism , Zika Virus Infection/physiopathology
9.
mSphere ; 4(3)2019 06 05.
Article in English | MEDLINE | ID: mdl-31167947

ABSTRACT

The use of high-throughput sequencing (HTS) to identify viruses in biologicals differs from current molecular approaches, since its use enables an unbiased approach to detection without the need to design specific primers to preamplify target sequences. Its broad range of detection and analytical sensitivity make it an important tool to ensure that biologicals are free from adventitious viruses. Similar to other molecular methods, however, identification of viral sequences in cells by HTS does not prove viral infection, since this could reflect carryover of inert viral sequences from reagents or other sources or the presence of transcriptionally inactive cellular sequences. Due to the broad range of detection associated with HTS, the above can potentially be perceived as a drawback for the testing of pharmaceutical biological products using this method. In order to avoid the identification of inert viral sequences, we present a methodology based on metabolic RNA labeling and sequencing, which enables the specific identification of newly synthesized viral RNAs in infected cells, resulting in the ability to unambiguously distinguish active infection by DNA or RNA viruses from inert nucleic acids. In the present study, we report the ability to differentiate Vero cells acutely infected by a single-stranded positive-sense RNA virus (tick-borne encephalitis virus) from cells which have been in contact with nonreplicating virus particles. Additionally, we also found a laboratory contamination by the squirrel monkey retrovirus of our Vero cell line, which was derived from an Old World (African green) monkey, a type of contamination which until now has been identified only in cells derived from primates from the New World.IMPORTANCE The use of high-throughput sequencing (HTS) to identify viral contamination of biological products is extremely sensitive and provides a broad range of detection. Nevertheless, viral sequences identified can also be inert. Examples include contamination resulting from reagents or the presence of inactivated viruses in animal-derived components of the cell culture medium. We therefore developed a method that relies on the sequencing of newly synthesized RNAs, an unequivocal sign of the presence of a transcriptionally active virus. This improvement in the specificity of viral testing increases the acceptability of HTS as a standard test for cells used in manufacturing biologicals and in biotherapies.


Subject(s)
DNA Contamination , DNA, Viral/analysis , RNA, Viral/analysis , Viruses/genetics , Animals , Cell Differentiation , Chlorocebus aethiops , Computational Biology , High-Throughput Nucleotide Sequencing , Vero Cells , Viruses/isolation & purification
10.
Article in English | MEDLINE | ID: mdl-29423380

ABSTRACT

To define the bottlenecks that restrict antigen expression after oral administration of viral-vectored vaccines, we tracked vectors derived from the human adenovirus type 5 at whole body, tissue, and cellular scales throughout the digestive tract in a murine model of oral delivery. After intragastric administration of vectors encoding firefly luciferase or a model antigen, detectable levels of transgene-encoded protein or mRNA were confined to the intestine, and restricted to delimited anatomical zones. Expression of luciferase in the form of multiple small bioluminescent foci in the distal ileum, cecum, and proximal colon suggested multiple crossing points. Many foci were unassociated with visible Peyer's patches, implying that transduced cells lay in proximity to villous rather than follicle-associated epithelium, as supported by detection of transgene-encoded antigen in villous epithelial cells. Transgene-encoded mRNA but not protein was readily detected in Peyer's patches, suggesting that post-transcriptional regulation of viral gene expression might limit expression of transgene-encoded antigen in this tissue. To characterize the pathways by which the vector crossed the intestinal epithelium and encountered sentinel cells, a fluorescent-labeled vector was administered to mice by the intragastric route or inoculated into ligated intestinal loops comprising a Peyer's patch. The vector adhered selectively to microfold cells in the follicle-associated epithelium, and, after translocation to the subepithelial dome region, was captured by phagocytes that expressed CD11c and lysozyme. In conclusion, although a large number of crossing events took place throughout the intestine within and without Peyer's patches, multiple firewalls prevented systemic dissemination of vector and suppressed production of transgene-encoded protein in Peyer's patches.


Subject(s)
Adenoviruses, Human/genetics , Adenoviruses, Human/immunology , Genetic Vectors/genetics , Genetic Vectors/immunology , Transgenes/genetics , Transgenes/immunology , Administration, Oral , Animals , Female , Gene Expression , Genes, Reporter , Genetic Vectors/administration & dosage , Humans , Immunization , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice , Organ Specificity , Peyer's Patches/immunology , Peyer's Patches/metabolism , Phagocytes/metabolism , Protein Transport , Vaccination
11.
Cell Rep ; 18(2): 324-333, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28076778

ABSTRACT

ZIKA virus (ZIKV) is an emerging pathogen responsible for neurological disorders and congenital microcephaly. However, the molecular basis for ZIKV neurotropism remains poorly understood. Here, we show that Axl is expressed in human microglia and astrocytes in the developing brain and that it mediates ZIKV infection of glial cells. Axl-mediated ZIKV entry requires the Axl ligand Gas6, which bridges ZIKV particles to glial cells. Following binding, ZIKV is internalized through clathrin-mediated endocytosis and traffics to Rab5+ endosomes to establish productive infection. During entry, the ZIKV/Gas6 complex activates Axl kinase activity, which downmodulates interferon signaling and facilitates infection. ZIKV infection of human glial cells is inhibited by MYD1, an engineered Axl decoy receptor, and by the Axl kinase inhibitor R428. Our results highlight the dual role of Axl during ZIKV infection of glial cells: promoting viral entry and modulating innate immune responses. Therefore, inhibiting Axl function may represent a potential target for future antiviral therapies.


Subject(s)
Immunity, Innate , Neuroglia/metabolism , Neuroglia/virology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Virus Internalization , Zika Virus/physiology , Brain/embryology , Brain/metabolism , Clathrin/metabolism , Endocytosis , Endosomes/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Interferon Type I/metabolism , Neuroglia/pathology , Signal Transduction , Zika Virus Infection/pathology , Zika Virus Infection/virology , Axl Receptor Tyrosine Kinase
12.
J Virol ; 91(1)2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27795408

ABSTRACT

Schmallenberg virus (SBV) was discovered in Germany in late 2011 and then spread rapidly to many European countries. SBV is an orthobunyavirus that causes abortion and congenital abnormalities in ruminants. A virus-encoded nonstructural protein, termed NSs, is a major virulence factor of SBV, and it is known to promote the degradation of Rpb1, a subunit of the RNA polymerase II (Pol II) complex, and therefore hampers global cellular transcription. In this study, we found that NSs is mainly localized in the nucleus of infected cells and specifically appears to target the nucleolus through a nucleolar localization signal (NoLS) localized between residues 33 and 51 of the protein. NSs colocalizes with nucleolar markers such as B23 (nucleophosmin) and fibrillarin. We observed that in SBV-infected cells, B23 undergoes a nucleolus-to-nucleoplasm redistribution, evocative of virus-induced nucleolar disruption. In contrast, the nucleolar pattern of B23 was unchanged upon infection with an SBV recombinant mutant with NSs lacking the NoLS motif (SBVΔNoLS). Interestingly, unlike wild-type SBV, the inhibitory activity of SBVΔNoLS toward RNA Pol II transcription is impaired. Overall, our results suggest that a putative link exists between NSs-induced nucleolar disruption and its inhibitory function on cellular transcription, which consequently precludes the cellular antiviral response and/or induces cell death. IMPORTANCE: Schmallenberg virus (SBV) is an emerging arbovirus of ruminants that spread in Europe between 2011 and 2013. SBV induces fetal abnormalities during gestation, with the central nervous system being one of the most affected organs. The virus-encoded NSs protein acts as a virulence factor by impairing host cell transcription. Here, we show that NSs contains a nucleolar localization signal (NoLS) and induces disorganization of the nucleolus. The NoLS motif in the SBV NSs is absolutely necessary for virus-induced inhibition of cellular transcription. To our knowledge, this is the first report of nucleolar functions for NSs within the Bunyaviridae family.


Subject(s)
Cell Nucleolus/virology , Ependymoglial Cells/virology , Host-Pathogen Interactions , Orthobunyavirus/pathogenicity , RNA Polymerase II/chemistry , Viral Nonstructural Proteins/chemistry , Animals , Cell Line, Transformed , Cell Nucleolus/metabolism , Cell Nucleolus/ultrastructure , Choroid Plexus/cytology , Choroid Plexus/metabolism , Choroid Plexus/virology , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Ependymoglial Cells/metabolism , Ependymoglial Cells/ultrastructure , Gene Expression Regulation , HeLa Cells , Humans , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleophosmin , Orthobunyavirus/genetics , Orthobunyavirus/metabolism , Protein Sorting Signals , Protein Transport , Proteolysis , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Sheep , Signal Transduction , Transcription, Genetic , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
14.
PLoS Pathog ; 11(4): e1004859, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25923687

ABSTRACT

It is well established that persistent viral infection may impair cellular function of specialized cells without overt damage. This concept, when applied to neurotropic viruses, may help to understand certain neurologic and neuropsychiatric diseases. Borna disease virus (BDV) is an excellent example of a persistent virus that targets the brain, impairs neural functions without cell lysis, and ultimately results in neurobehavioral disturbances. Recently, we have shown that BDV infects human neural progenitor cells (hNPCs) and impairs neurogenesis, revealing a new mechanism by which BDV may interfere with brain function. Here, we sought to identify the viral proteins and molecular pathways that are involved. Using lentiviral vectors for expression of the bdv-p and bdv-x viral genes, we demonstrate that the phosphoprotein P, but not the X protein, diminishes human neurogenesis and, more particularly, GABAergic neurogenesis. We further reveal a decrease in pro-neuronal factors known to be involved in neuronal differentiation (ApoE, Noggin, TH and Scg10/Stathmin2), demonstrating that cellular dysfunction is associated with impairment of specific components of the molecular program that controls neurogenesis. Our findings thus provide the first evidence that a viral protein impairs GABAergic human neurogenesis, a process that is dysregulated in several neuropsychiatric disorders. They improve our understanding of the mechanisms by which a persistent virus may interfere with brain development and function in the adult.


Subject(s)
Borna disease virus/physiology , Down-Regulation , GABAergic Neurons/metabolism , Host-Pathogen Interactions , Neurogenesis , Phosphoproteins/metabolism , Viral Structural Proteins/metabolism , Active Transport, Cell Nucleus , Apolipoproteins E/antagonists & inhibitors , Apolipoproteins E/metabolism , Biomarkers/chemistry , Biomarkers/metabolism , Borna Disease/metabolism , Borna Disease/pathology , Borna Disease/virology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Proliferation , Cells, Cultured , France , GABAergic Neurons/cytology , GABAergic Neurons/pathology , GABAergic Neurons/virology , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/pathology , Human Embryonic Stem Cells/virology , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Phosphoproteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/toxicity , Stathmin , Tyrosine 3-Monooxygenase/antagonists & inhibitors , Tyrosine 3-Monooxygenase/metabolism , Viral Structural Proteins/genetics
15.
Vaccine ; 33(1): 141-8, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25444801

ABSTRACT

Contagious bovine pleuropneumonia (CBPP), caused by Mycoplasma mycoides subsp. mycoides small colony type (MmmSC), is a devastating respiratory disease of cattle. In sub-Saharan Africa, where CBPP is enzootic, live attenuated vaccines are deployed but afford only short-lived protection. In cattle, recovery from experimental MmmSC infection has been associated with the presence of CD4(+) T lymphocytes that secrete interferon gamma in response to MmmSC, and in particular to the lipoprotein A (LppA) antigen. In an effort to develop a better vaccine against CBPP, a viral vector (Ad5-LppA) that expressed LppA was generated from human adenovirus type 5. The LppA-specific immune responses elicited by the Ad5-LppA vector were evaluated in mice, and compared to those elicited by recombinant LppA formulated with a potent adjuvant. Notably, a single administration of Ad5-LppA, but not recombinant protein, sufficed to elicit a robust LppA-specific humoral response. After a booster administration, both vector and recombinant protein elicited strong LppA-specific humoral and cell-mediated responses. Ex vivo stimulation of splenocytes induced extensive proliferation of CD4(+) T cells for mice immunized with vector or protein, and secretion of T helper 1-associated and proinflammatory cytokines for mice immunized with Ad5-LppA. Our study - by demonstrating the potential of a viral-vectored prototypic vaccine to elicit prompt and robust immune responses against a major antigen of MmmSC - represents a first step in developing a recombinant vaccine against CBPP.


Subject(s)
Adenoviruses, Human/genetics , Bacterial Vaccines/immunology , Drug Carriers , Genetic Vectors , Lipoprotein(a)/immunology , Mycoplasma mycoides/immunology , Pleuropneumonia, Contagious/prevention & control , Animals , Antibodies, Bacterial/blood , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Cytokines/metabolism , Female , Humans , Lipoprotein(a)/biosynthesis , Lipoprotein(a)/genetics , Mice, Inbred BALB C , Mycoplasma mycoides/genetics
16.
Biologicals ; 42(4): 218-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24930452

ABSTRACT

Specific Pathogen Free (SPF) embryonated eggs are used for the production of many veterinary and human vaccines. We have used High Throughput Sequencing to screen allantoic fluids and embryos for the presence of encapsidated viral genomes and viral transcripts, respectively. SPF eggs from two different producers were tested. We evidenced sequences corresponding to known endogenous retroviruses and sequences of Avian Leukosis Virus, but no sequence that might suggest a productive infection of eggs with a virus even distant from known viruses. Our results strongly suggest that SPF eggs such as those used for this study represent a safe substrate for the production of vaccines.


Subject(s)
Eggs/analysis , Eggs/virology , High-Throughput Nucleotide Sequencing/methods , Specific Pathogen-Free Organisms , Animals , Avian Leukosis Virus/genetics , Chick Embryo , Chickens/virology , DNA, Viral/analysis , Endogenous Retroviruses/genetics , RNA, Viral/analysis , Vaccines/biosynthesis
17.
PLoS One ; 8(12): e84473, 2013.
Article in English | MEDLINE | ID: mdl-24367664

ABSTRACT

Some strains of West Nile virus (WNV) are neuroinvasive and may induce fatal encephalitis/meningitis in a variety of animal species including humans. Whether, however, there is a strain-specific signature in the brain is as yet unknown. Here we investigated the neuropathogenesis induced by two phylogenetically distant WNV strains of lineage 1, WNV(IS98) and WNV(KUN35 911). While four-week old C57Bl/6J mice were susceptible to both strains and succumbed rapidly after intraperitoneal inoculation, differences were observed in virulence and clinical disease. WNV(KUN35 911), the less virulent strain as judged by determination of LD50, induced typical signs of encephalitis. Such signs were not observed in WNV(IS98)-infected mice, although they died more rapidly. Histological examination of brain sections also revealed differences, as the level of apoptosis and inflammation was higher in WNV(KUN35 911)- than WNV(IS98)-infected mice. Moreover, staining for cleaved caspase 3 showed that the two WNV strains induced apoptotic death through different molecular mechanisms in one particular brain area. Finally, the two strains showed similar tropism in cortex, striatum, brainstem, and cerebellum but a different one in hippocampus. In summary, our data show that, upon peripheral administration, WNV(IS98) and WNV(KUN35 911) strains induce partially distinct lesions and tissue tropism in the brain. They suggest that the virulence of a WNV strain is not necessarily correlated with the severity of apoptotic and inflammatory lesions in the brain.


Subject(s)
Brain/pathology , Brain/virology , West Nile virus/pathogenicity , Animals , Apoptosis , Inflammation/virology , Mice , Mice, Inbred C57BL , Species Specificity , West Nile virus/physiology
18.
Viruses ; 5(11): 2856-80, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24284878

ABSTRACT

West Nile virus (WNV) is a neurotropic flavivirus that cycles between mosquitoes and birds but that can also infect humans, horses, and other vertebrate animals. In most humans, WNV infection remains subclinical. However, 20%-40% of those infected may develop WNV disease, with symptoms ranging from fever to meningoencephalitis. A large variety of WNV strains have been described worldwide. Based on their genetic differences, they have been classified into eight lineages; the pathogenic strains belong to lineages 1 and 2. Ten years ago, Beasley et al. (2002) found that dramatic differences exist in the virulence and neuroinvasion properties of lineage 1 and lineage 2 WNV strains. Further insights on how WNV interacts with its hosts have recently been gained; the virus acts either at the periphery or on the central nervous system (CNS), and these observed differences could help explain the differential virulence and neurovirulence of WNV strains. This review aims to summarize the current state of knowledge on factors that trigger WNV dissemination and CNS invasion as well as on the inflammatory response and CNS damage induced by WNV. Moreover, we will discuss how WNV strains differentially interact with the innate immune system and CNS cells, thus influencing WNV pathogenesis.


Subject(s)
Central Nervous System Diseases/virology , West Nile Fever/virology , West Nile virus/pathogenicity , Animals , Central Nervous System Diseases/immunology , Humans , Virulence , West Nile Fever/immunology , West Nile virus/classification , West Nile virus/genetics , West Nile virus/isolation & purification
19.
J Virol ; 86(5): 2512-22, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22190725

ABSTRACT

Understanding the complex mechanisms by which infectious agents can disrupt behavior represents a major challenge. The Borna disease virus (BDV), a potential human pathogen, provides a unique model to study such mechanisms. Because BDV induces neurodegeneration in brain areas that are still undergoing maturation at the time of infection, we tested the hypothesis that BDV interferes with neurogenesis. We showed that human neural stem/progenitor cells are highly permissive to BDV, although infection does not alter their survival or undifferentiated phenotype. In contrast, upon the induction of differentiation, BDV is capable of severely impairing neurogenesis by interfering with the survival of newly generated neurons. Such impairment was specific to neurogenesis, since astrogliogenesis was unaltered. In conclusion, we demonstrate a new mechanism by which BDV might impair neural function and brain plasticity in infected individuals. These results may contribute to a better understanding of behavioral disorders associated with BDV infection.


Subject(s)
Borna Disease/physiopathology , Borna disease virus/physiology , Neurogenesis , Neurons/cytology , Stem Cells/cytology , Borna Disease/virology , Brain/cytology , Brain/virology , Cells, Cultured , Humans
20.
Neurobiol Dis ; 23(3): 603-11, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16854590

ABSTRACT

Neurodegeneration is a common neuropathological feature of prion diseases. Although evidence of apoptosis was found in natural and experimental prion diseases, the precise mechanisms by which neurons die are poorly understood. The pro-apoptotic BAX protein, a key factor of the mitochondrial pathway, plays a central role in the regulation of neuronal apoptosis. Recently, BAX was implicated in neuronal death in a transgenic model of inherited prion disease. Nevertheless, whether neurodegeneration occurs by similar mechanisms in other prion diseases remains unknown. Here, using mice knocked out for the Bax gene, we investigated BAX implication in neuronal death induced by a prion disease of infectious origin. A mouse-adapted prion strain of bovine spongiform encephalopathy (BSE) was inoculated intracerebrally into Bax-/- mice and their wild-type littermates. We found that Bax inactivation did not alter the development of the disease. Clinical illness was not prevented. PrP(res) deposition and astrogliosis occurred to the usual extent. Neuronal integrity was not maintained, and neurons in hippocampus and thalamus were not protected. These results demonstrated that BAX is not necessary for neuron death induced by the BSE strain. They suggest the existence of multiple molecular death pathways in prion diseases.


Subject(s)
Apoptosis/genetics , Brain/metabolism , Encephalopathy, Bovine Spongiform/genetics , Encephalopathy, Bovine Spongiform/therapy , Neurons/metabolism , bcl-2-Associated X Protein/genetics , Animals , Brain/pathology , Brain/physiopathology , Cattle , Disease Models, Animal , Encephalopathy, Bovine Spongiform/physiopathology , Female , Genetic Predisposition to Disease/genetics , Genetic Therapy , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/pathology , PrPSc Proteins/metabolism , Prions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...