Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters










Publication year range
1.
Curr Top Microbiol Immunol ; 329: 129-62, 2009.
Article in English | MEDLINE | ID: mdl-19198565

ABSTRACT

An overview is given on the development of technologies to allow reverse genetics of RNA viruses, i.e., the rescue of viruses from cDNA, with emphasis on nonsegmented negative-strand RNA viruses (Mononegavirales), as exemplified for measles virus (MV). Primarily, these technologies allowed site-directed mutagenesis, enabling important insights into a variety of aspects of the biology of these viruses. Concomitantly, foreign coding sequences were inserted to (a) allow localization of virus replication in vivo through marker gene expression, (b) develop candidate multivalent vaccines against measles and other pathogens, and (c) create candidate oncolytic viruses. The vector use of these viruses was experimentally encouraged by the pronounced genetic stability of the recombinants unexpected for RNA viruses, and by the high load of insertable genetic material, in excess of 6 kb. The known assets, such as the small genome size of the vector in comparison to DNA viruses proposed as vectors, the extensive clinical experience of attenuated MV as vaccine with a proven record of high safety and efficacy, and the low production cost per vaccination dose are thus favorably complemented.


Subject(s)
Genetic Engineering , Measles Vaccine/immunology , Measles virus/genetics , Measles/immunology , Animals , Genome, Viral , Humans , Measles/prevention & control , Measles/virology , Measles Vaccine/administration & dosage , Measles Vaccine/genetics , Measles virus/immunology , Mutagenesis, Site-Directed , Vaccines, Combined/administration & dosage , Vaccines, Combined/genetics , Vaccines, Combined/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
2.
Vaccine ; 19(17-19): 2329-36, 2001 Mar 21.
Article in English | MEDLINE | ID: mdl-11257357

ABSTRACT

We have genetically engineered a panel of recombinant measles viruses (rMVs) that express from various positions within the MV genome either the HN or F surface glycoproteins of mumps virus (MuV) or the env, gag or pol proteins from simian immunodeficiency virus (SIV). All rMVs were rescued from the respective antigenomic plasmid constructs; progeny viruses replicated comparably to the progenitor Edmonston B MV, but showed slight propagation retardation, which was dependent on the size and nature of the expressed proteins and on the genomic position of the inserts. All transgenes except that encoding mumps F glycoprotein were faithfully maintained and expressed even after virus amplification by 10(20). Our results suggest possible applications of rMVs as live-attenuated, multivalent vaccines against retroviruses such as SIV and HIV as well as other pathogens more distantly related to MV than MuV.


Subject(s)
Antigens, Viral/genetics , Measles virus/genetics , Measles virus/immunology , Mumps virus/genetics , Mumps virus/immunology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Animals , Base Sequence , Chlorocebus aethiops , DNA Primers/genetics , Gene Expression , Genes, Viral , Genetic Engineering , Genetic Vectors , Measles virus/growth & development , Plasmids/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombination, Genetic , Vaccines, Attenuated/genetics , Vaccines, Attenuated/isolation & purification , Vaccines, Synthetic/genetics , Vaccines, Synthetic/isolation & purification , Vero Cells , Viral Proteins/genetics , Viral Proteins/immunology
3.
J Virol ; 75(7): 3343-51, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11238860

ABSTRACT

Knowledge of the mechanisms of virus dissemination in acute measles is cursory, but cells of the monocyte/macrophage (MM) lineage appear to be early targets. We characterized the dissemination of the Edmonston B vaccine strain of measles virus (MV-Ed) in peripheral blood mononuclear cells (PBMC) of two mouse strains expressing the human MV-Ed receptor CD46 with human-like tissue specificity and efficiency. In one strain the alpha/beta interferon receptor is defective, allowing for efficient MV-Ed systemic spread. In both mouse strains the PBMC most efficiently infected were F4/80-positive MMs, regardless of the inoculation route used. Circulating B lymphocytes and CD4-positive T lymphocytes were infected at lower levels, but no infected CD8-positive T lymphocytes were detected. To elucidate the roles of MMs in infection, we depleted these cells by clodronate liposome treatment in vivo. MV-Ed infection of splenic MM-depleted mice caused strong activation and infection of splenic dendritic cells (DC), followed by enhanced virus replication in the spleen. Similarly, depletion of lung macrophages resulted in strong activation and infection of lung DC. Thus, in MV infections of genetically modified mice, blood monocytes and tissue macrophages provide functions beneficial for both the virus and the host: they support virus replication early after infection, but they also contribute to protecting other immune cells from infection. Human MM may have similar roles in acute measles.


Subject(s)
Antigens, CD/physiology , Macrophages/physiology , Measles/immunology , Membrane Glycoproteins/physiology , Receptors, Interferon/physiology , Animals , Dendritic Cells/virology , Humans , Macrophages/virology , Measles virus/physiology , Membrane Cofactor Protein , Mice , Monocytes/physiology , Monocytes/virology , Receptor, Interferon alpha-beta , Virus Replication
4.
Virology ; 291(2): 215-25, 2001 Dec 20.
Article in English | MEDLINE | ID: mdl-11878891

ABSTRACT

Subacute sclerosing panencephalitis (SSPE) is a progressive degenerative disease of the brain uniformly leading to death. Although caused by measles virus (MV), the virus recovered from patients with SSPE differs from wild-type MV; biologically SSPE virus is defective and its genome displays a variety of mutations among which biased replacements of many uridine by cytidine resides primarily in the matrix (M) gene. To address the question of whether the SSPE MVs with M mutations are passive in that they are not infectious, cannot spread within the CNS, and basically represent an end-stage result of a progressive infection or alternatively SSPE viruses are infectious, and their mutations enable them to persist and thereby cause a prolonged neurodegenerative disease, we utilized reverse genetics to generate an infectious virus in which the M gene of MV was replaced with the M gene of Biken strain SSPE MV and inoculated the recombinant virus into transgenic mice bearing the MV receptor. Our results indicate that despite biased hypermutations in the M gene, the virus is infectious in vivo and produces a protracted progressive infection with death occurring as long as 30 to 50 days after that caused by MV. In primary neuron cultures, the mutated M protein is not essential for MV replication, prevents colocalization of the viral N with membrane glycoproteins, and is associated with accumulation of nucleocapsids in cells' cytoplasm and nucleus.


Subject(s)
SSPE Virus/metabolism , Subacute Sclerosing Panencephalitis/virology , Viral Matrix Proteins/physiology , Animals , Cells, Cultured , Chlorocebus aethiops , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurons/cytology , Neurons/virology , Nucleocapsid Proteins/metabolism , SSPE Virus/genetics , SSPE Virus/growth & development , SSPE Virus/physiology , Subacute Sclerosing Panencephalitis/mortality , Subcellular Fractions , Vero Cells , Viral Fusion Proteins/metabolism , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism
5.
EMBO J ; 19(14): 3576-85, 2000 Jul 17.
Article in English | MEDLINE | ID: mdl-10899112

ABSTRACT

In polarized epithelial cells measles virus (MV) is predominantly released at the apical cell surface, irrespective of the sorting of its two envelope glycoproteins F and H. It has been reported previously that the viral matrix (M) protein modulates the fusogenic capacity of the viral envelope glycoproteins. Here, extant MV mutants and chimeras were used to determine the role of M protein in the transport of viral glycoproteins and release of progeny virions in polarized epithelial CaCo2 cells. In the absence of M, envelope glycoproteins are sorted to the basolateral surface, suggesting that they possess intrinsic basolateral sorting signals. However, interactions of M with the glycoprotein cytoplasmic tails allow M-glycoprotein co-segregation to the apical surface, suggesting a vectorial function of M to retarget the glycoproteins for apical virion release. Whereas this may allow virus airway shedding, the intrinsic sorting of the glycoproteins to the basolateral surface may account for systemic host infection by allowing efficient cell-cell fusion.


Subject(s)
Cell Polarity , Epithelial Cells/virology , Glycoproteins/metabolism , Measles virus/physiology , Viral Matrix Proteins/metabolism , Amino Acid Sequence , Animals , Caco-2 Cells , Cell Line , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fluorescent Antibody Technique , Glycoproteins/chemistry , Glycoproteins/genetics , Humans , Measles virus/genetics , Measles virus/metabolism , Membrane Fusion , Molecular Sequence Data , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Viral Matrix Proteins/genetics , Virus Replication
6.
J Virol ; 74(3): 1364-72, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10627547

ABSTRACT

The dissemination of the Edmonston measles virus (Ed-MV) vaccine strain was studied with genetically modified mice defective for the alpha/beta interferon receptor and expressing human CD46 with human-like tissue specificity and efficiency. A few days after intranasal infection, macrophages expressing Ed-MV RNA were detected in the lungs, in draining lymph nodes, and in the thymus. In lymph nodes, large syncytia which stained positive for viral RNA and for macrophage surface marker proteins were found and apoptotic cell death was monitored. In the thymus, smaller syncytia which stained positive for macrophage and dendritic cell markers were detected. Thus, macrophages appear to be the main vectors for dissemination of MV infection in these mice; human macrophages may have a similar function in the natural host. We then compared the pathogenicities of two recombinant viruses lacking the C or V nonstructural proteins to that of the parental strain, Ed-MV. These viruses were less effective in spreading through the lymphatic system and, unlike Ed-MV, were not detected in the liver. After intracerebral inoculation the recombinant viruses caused lethal disease less often than Ed-MV and induced distinctive patterns of gliosis and inflammation. Ed-MV was reisolated from brain tissue, but its derivatives were not. C- and V-defective viruses should be considered as more-attenuated MV vaccine candidates.


Subject(s)
Defective Viruses/physiology , Lymphatic System/virology , Measles virus/genetics , Measles virus/physiology , Measles/pathology , Animals , Antigens, CD/analysis , Antigens, CD/genetics , Brain/pathology , Defective Viruses/genetics , Giant Cells , Humans , Immunohistochemistry , In Situ Hybridization , Liver/pathology , Liver/virology , Lung/pathology , Lung/virology , Lymphatic System/pathology , Macrophages/virology , Measles/virology , Measles virus/classification , Measles virus/isolation & purification , Membrane Cofactor Protein , Membrane Glycoproteins/analysis , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Receptors, Interferon/analysis , Receptors, Interferon/genetics , Virus Replication
7.
J Gen Virol ; 81(Pt 2): 441-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10644843

ABSTRACT

Proteolytic cleavage of the fusion protein (F) is an important control mechanism of the biological activity of paramyxoviruses. The sequence R-R-H-K-R(112) at the cleavage site of the F protein of measles virus (MV) was altered by site-directed mutagenesis to R-N-H-N-R(112), which is not recognized by the ubiquitous cellular protease furin. When transiently expressed in cell cultures standard F protein was cleaved, whereas the mutant remained in the uncleaved form. Syncytium formation by the mutant that was analysed after coexpression with haemagglutinin protein depended on the presence of trypsin. Recombinant MV containing the mutation required trypsin activation for fusion and infectivity in cell culture. Intranasal infection of transgenic mice susceptible to MV infection (Ifnar(tm)-CD46Ge) resulted in a moderately productive infection and inflammation of the lung. In contrast to parental virus, intracerebral inoculation did not induce neural disease. The possible effects of the change in cleavage activation on tissue tropism and pathogenicity are discussed.


Subject(s)
Measles virus/genetics , Measles virus/pathogenicity , Trypsin/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Binding Sites/genetics , Cell Fusion/drug effects , Cell Line , Chlorocebus aethiops , DNA Primers/genetics , Humans , Measles/etiology , Measles/pathology , Measles/virology , Measles virus/drug effects , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Recombination, Genetic , Vero Cells , Viral Fusion Proteins/genetics , Viral Fusion Proteins/physiology , Virulence/drug effects , Virulence/genetics , Virulence/physiology
8.
Virology ; 267(1): 80-9, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10648185

ABSTRACT

The measles virus (MV) P gene encodes three proteins: the P protein and two nonstructural proteins, C and V. Because the functions of both the C and V protein are unknown, we used MV C (C-) and V (V-) deletion recombinants generated by the MV reverse genetics system (F. Radecke, P. Spielhofer, H. Schnieder, K. Kaelin, M. Huber, C. Dotsch, G. Christiansen, and M. A. Billeter 1995. EMBO J. 14, 5773-5784). Compared to parental vaccine strain, Edmonston (Ed) MV, both had normal growth and cytopathic effects in Vero cells and showed similar growth kinetics in human neuroblastoma SK-N-MC cells and in primary mouse neurons expressing the MV receptor, CD46. However, in vivo, using YAC-CD46 transgenic mice as a model for MV induced CNS disease (M. B. A. Oldstone, H. Lewicki, D. Thomas, A. Tishon, S. Dales, J. Patterson, M. Manchester, D. Homann, D. Naniche, and A. Holz 1999. Cell 98, 629-640), C- and V- viruses differed markedly from wt Ed(V(+)C(+)) virus. Newborn mice inoculated with as little as 10(3) PFU of Ed strain became ill and died after 10-15 days. In contrast, those inoculated with 10(3) or 10(4) PFU of MV C- or MV V- showed significantly fewer and milder clinical symptoms and had a lower mortality. A total of 10(5) PFU V- virus were required to kill most YAC-CD46 mice, and less than half (44%) were killed with a corresponding dose of MV C-. Immunohistochemical staining for MV antigens showed similar extents of spread for MV C- and MV Ed but restricted spread for MV V- throughout the brain. Viral load and transcription were markedly reduced for V- but not for C-. Multiple cytokines and chemokines were equivalently upregulated for all three viruses. Therefore, MV C and V proteins encode virulence functions in vivo and likely operate via separate mechanisms.


Subject(s)
Measles virus/pathogenicity , Phosphoproteins/physiology , Viral Nonstructural Proteins/physiology , Viral Proteins/physiology , Animals , Base Sequence , Brain/virology , Cell Line , Chlorocebus aethiops , DNA Primers/genetics , Gene Deletion , Genes, Viral , Humans , Measles/virology , Measles virus/genetics , Measles virus/physiology , Mice , Mice, Transgenic , Neurons/virology , Phosphoproteins/genetics , RNA, Viral/genetics , Recombination, Genetic , Vero Cells , Viral Nonstructural Proteins/genetics , Viral Proteins/genetics , Virulence/genetics , Virus Replication
9.
J Virol ; 73(11): 9568-75, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10516065

ABSTRACT

A recombinant measles virus (MV) which expresses enhanced green fluorescent protein (EGFP) has been rescued. This virus, MVeGFP, expresses the reporter gene from an additional transcription unit which is located prior to the gene encoding the measles virus nucleocapsid protein. The recombinant virus was used to infect human astrocytoma cells (GCCM). Immunocytochemistry (ICC) together with EGFP autofluorescence showed that EGFP is both an early and very sensitive indicator of cell infection. Cells that were EGFP-positive and ICC-negative were frequently observed. Confocal microscopy was used to indirectly visualize MV infection of GCCM cells and to subsequently follow cell-to-cell spread in real time. These astrocytoma cells have extended processes, which in many cases are intimately associated. The processes appear to have an important role in cell-to-cell spread, and MVeGFP was observed to utilize them in the infection of surrounding cells. Heterogeneity was seen in cell-to-cell spread in what was expected to be a homogeneous monolayer. In tissue culture, physical constraints govern the integrity of the syncytia which are formed upon extensive cell fusion. When around 50 cells were fused, the syncytia rapidly disintegrated and many of the infected cells detached. Residual adherent EGFP-positive cells were seen to either continue to be involved in the infection of surrounding cells or to remain EGFP positive but no longer participate in the transmission of MV infection to neighboring cells.


Subject(s)
Astrocytes/virology , Measles virus/genetics , Measles virus/physiology , Animals , Astrocytoma , Cell Line , Fluorescent Antibody Technique , Genes, Reporter , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Recombinant Proteins/metabolism , Tumor Cells, Cultured
10.
J Virol ; 73(8): 6916-22, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10400789

ABSTRACT

Molecular determinants of neuropathogenesis have been shown to be present in the hemagglutinin (H) protein of measles virus (MV). An H gene insertion vector has been generated from the Edmonston B vaccine full-length infectious clone of MV. Using this vector, it is possible to insert complete H open reading frames into the parental (Edtag) background. The H gene from a rodent brain-adapted MV strain (CAM/RB) was inserted into this vector, and a recombinant virus (EdtagCAMH) was rescued by using a modified vaccinia virus which expresses T7 RNA polymerase (MVA-T7). The recombinant virus grew at an equivalent rate and to similar titers as the CAM/RB and Edtag parental viruses. Neurovirulence was assayed in a mouse model for MV encephalitis. Viruses were injected intracerebrally into the right cortex of C57/BL/6 suckling mice. After infection mice inoculated with the CAM/RB strain developed hind limb paralysis and ataxia. Clinical symptoms were never observed with an equivalent dose of Edtag virus or in sham infections. Immunohistochemistry (IHC) was used to detect viral antigen in formalin-fixed brain sections. Measles antigen was observed in neurons and neuronal processes of the hippocampus, frontal, temporal, and olfactory cortices and neostriatum on both sides of symmetrical structures. Viral antigen was not detected in mice infected with Edtag virus. Mice infected with the recombinant virus, EdtagCAMH, became clinically ill, and virus was detected by IHC in regions of the brain similar to those in which it was detected in animals infected with CAM/RB. The EdtagCAMH infection had, however, progressed much less than the CAM/RB virus at 4 days postinfection. It therefore appears that additional determinants are encoded in other regions of the MV genome which are required for full neurovirulence equivalent to CAM/RB. Nevertheless, replacement of the H gene alone is sufficient to cause neuropathology.


Subject(s)
Brain/virology , Hemagglutinins, Viral/physiology , Measles virus/pathogenicity , Adaptation, Biological , Animals , Chlorocebus aethiops , Disease Models, Animal , Genes, Viral , Genetic Vectors , HeLa Cells , Hemagglutinins, Viral/genetics , Humans , Measles/pathology , Measles/virology , Measles Vaccine , Measles virus/genetics , Measles virus/growth & development , Measles virus/immunology , Mice , Mice, Inbred C57BL , Mutagenesis, Insertional , Rodentia , Vero Cells , Virulence
11.
J Virol ; 73(6): 4823-8, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10233943

ABSTRACT

It has been shown previously that measles virus (MV) can be successfully used to express foreign proteins (M. Singh and M. A. Billeter, J. Gen. Virol. 80:101-106, 1998). To develop an inexpensive MV-based vaccine, we generated recombinant MVs that produce structural proteins of hepatitis B virus (HBV). A recombinant virus that expressed the HBV small surface antigen (HBsAg) was analyzed in terms of its replication characteristics, its genetic stability in cell culture, and its immunogenic potential in genetically modified mice. Although this virus showed a progression of replication slightly slower than that of the parental MV, it appeared to stably maintain the added genetic information; it uniformly expressed the appropriately glycosylated HBsAg after 10 serial passages. Genetically modified mice inoculated with this recombinant MV produced humoral immune responses against both HBsAg and MV proteins.


Subject(s)
Hepatitis B Surface Antigens/immunology , Hepatitis B Vaccines/immunology , Measles virus/genetics , Vaccines, Synthetic/immunology , Animals , Antibodies, Viral/blood , Chlorocebus aethiops , Hepatitis B Core Antigens/genetics , Hepatitis B Core Antigens/immunology , Hepatitis B Surface Antigens/genetics , Immunization , Measles virus/immunology , Mice , Recombinant Proteins/immunology , Vero Cells , Virus Replication
12.
J Virol Methods ; 79(1): 65-74, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10328536

ABSTRACT

Measles virus encodes three proteins required for the encapsidation, transcription and replication of viral genomes. The genes for these proteins have been inserted into the vaccinia virus genome together with the gene for the bacteriophage T7 RNA polymerase. Cells infected with this recombinant virus were able to encapsidate, transcribe and replicate a CAT gene positioned in the negative polarity behind a T7 promoter and flanked by measles virus genomic termini. Inhibition of the accumulation of the nucleocapsid proteins by actinomycin D led to an increase in CAT expression. Thus the measles virus polymerase activity, encoded by the vaccinia genome, was regulated by the level of measles proteins just as the authentic polymerase. The recombinant vaccinia described in this study could be useful for the production of measles virus-like particles encoding foreign genes and employed in vaccination or gene therapy strategies.


Subject(s)
Measles virus/genetics , Measles virus/physiology , Transcription, Genetic/genetics , Vaccinia virus/genetics , Virus Replication , Bacteriophage T7/enzymology , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Genetic Vectors , Genome, Viral , Nucleocapsid/genetics , Nucleocapsid/metabolism , Recombination, Genetic , Restriction Mapping , Reverse Transcriptase Polymerase Chain Reaction
13.
J Gen Virol ; 80 ( Pt 1): 101-106, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9934691

ABSTRACT

Suppression of cell-mediated immunity (CMI) is well-documented during and after measles. This immunosuppression is suggested to result from decreased production of interleukin-12 (IL-12), a key interleukin for CMI. In an attempt to clearly discern the role of IL-12 in measles-induced immunosuppression, a measles virus (MV) that expresses biologically active human IL-12 was generated. This was achieved by inserting the coding sequences of the two subunits (p35 and p40) of human IL-12 separated by an internal ribosome entry site in an additional transcription unit between the H and the L genes of MV. Although the IL-12-expressing MV grew slightly slower than the normal MV, it stably maintained the inserted sequences (3.2 kb) and uniformly expressed the foreign genes after 10 passages in cell culture. These findings suggest that MV is a well-suited vector for delivery of proteins of immunogenic and therapeutic importance.


Subject(s)
Genetic Vectors/immunology , Interleukin-12/immunology , Measles virus/immunology , Animals , Base Sequence , Chlorocebus aethiops , Genetic Vectors/physiology , Humans , Interferon-gamma/metabolism , Interleukin-12/genetics , Measles virus/physiology , Molecular Sequence Data , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Vero Cells
14.
J Virol ; 72(10): 7754-61, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9733811

ABSTRACT

An understanding of the determinants of measles virus (MV) virulence has been hampered by the lack of an experimental model of infection. We have previously demonstrated that virulence phenotypes in human infections are faithfully reproduced by infection of human thymus/liver (thy/liv) implants engrafted into SCID mice, where the virus grows primarily in stromal cells but induces thymocyte apoptosis (P. G. Auwaerter et al., J. Virol. 70:3734-3740, 1996). To begin to elucidate the roles of the C protein, V protein, and the 5' untranslated region of the F gene (F 5'UTR) in MV infection in vivo, the replication of strains bearing mutations of these genes was compared to that of the parent sequence-tagged Edmonston strain (EdTag). Growth curves show that mutants fall into two phenotypic classes. One class of mutants demonstrated kinetics of growth similar to that of EdTag, with decreased peak titers. The second class of mutants manifested peak titers similar to that of EdTag but had different replication kinetics. Abrogation of V expression led to delayed and markedly prolonged replication. Additionally, thymocyte survival was prolonged and implant architecture was preserved throughout the course of infection. In contrast, massive bystander thymocyte death occurred after infection with EdTag and all other mutants. A mutant which overexpressed V in Vero cells (V+) had the opposite phenotype of the A mutant not expressing V (V-). V+ grew more rapidly than EdTag with 100-fold-greater levels of virus production 3 days after infection. These results suggest that C, V, and the F 5'UTR are accessory factors required for efficient virus replication in vivo. In addition, thymocyte survival after V- infection suggests this protein may play multiple roles in pathogenesis of MV infection of thymus. Since these recombinant mutant viruses grew identically to the parent virus in Vero cells, the data show that thy/liv implants are an excellent model for investigating the determinants of MV virulence.


Subject(s)
Measles virus/growth & development , Mutation , Recombination, Genetic , Viral Proteins/genetics , Animals , Base Sequence , DNA Primers , Humans , Liver/virology , Male , Measles virus/genetics , Measles virus/physiology , Mice , Mice, SCID , Phenotype , Thymus Gland/virology , Virus Replication/genetics
15.
J Virol ; 72(10): 8124-32, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9733853

ABSTRACT

Nonstructural proteins encoded by measles virus (MV) include the V protein which is translated from an edited P mRNA. V protein is not associated with intracellular or released viral particles and has recently been found to be dispensable for MV propagation in cell culture (H. Schneider, K. Kaelin, and M. A. Billeter, Virology 227:314-322, 1997). Using recombinant MVs (strain Edmonston [ED]) genetically engineered to overexpress V protein (ED-V+) or to be deficient for V protein (ED-V-), we found that in the absence of V both MV-specific proteins and RNAs accumulated to levels higher than those in the parental MV molecular clone (ED-tag), whereas MV-specific gene expression was strongly attenuated in human U-87 glioblastomas cells after infection with ED-V+. The titers of virus released from these cells 48 h after infection with either V mutant virus were lower than those from cells infected with ED-tag. Similarly, significantly reduced titers of infectious virus were reisolated from lung tissue of cotton rats (Sigmodon hispidus) after intranasal infection with both editing mutants compared to titers isolated from ED-tag-infected animals. In cell culture, expression of V protein led to a redistribution of MV N protein in doubly transfected Cos-7 cells, indicating that these proteins form heterologous complexes. This interaction was further confirmed by using a two-hybrid approach with both proteins expressed as Gal4 or VP16 fusion products. Moreover, V protein efficiently competed complexes formed between MV N and P proteins. These findings indicate that V protein acts to balance accumulation of viral gene products in cell culture, and this may be dependent on its interaction with MV N protein. Furthermore, expression of V protein may contribute to viral pathogenicity in vivo.


Subject(s)
Measles virus/pathogenicity , Phosphoproteins/genetics , RNA, Viral/biosynthesis , Viral Proteins/genetics , Animals , Humans , Measles virus/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , Rats , Rats, Inbred Strains , Recombination, Genetic , Tumor Cells, Cultured
16.
Nat Med ; 4(8): 945-8, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9701248

ABSTRACT

B-cell activation depends on the intensity of B-cell receptor cross-linking. Studies of haptenated antigens and vesicular stomatitis virus (VSV) have demonstrated a correlation between antigen repetitiveness and the degree to which B-cell activation is independent of T cells. Here, we compare neutralizing antibody responses to inactivated VSV with those to two inactivated human pathogenic viruses: highly cytopathic poliovirus (PV) and poorly cytopathic measles virus (MV). The rigidly structured PV efficiently induced neutralizing IgM antibodies independent of T cells. In contrast, neutralizing antibodies to the pleomorphic MV were dependent on helper T cells. To test whether this resulted from the differences in virus structure or the capacity of MV to induce cell fusion and/or immunosuppression, we analyzed antibody responses to chimeric MV expressing VSV glycoprotein instead of MV fusion protein and hemagglutinin. IgM antibodies were independent of T cells; in addition, we found IgG responses dependent on T-cell help that were enduring and protective against lethal VSV infection. Because chimeric MV viruses look like MV ultrastructurally, we conclude that not only structural differences in the envelope but also the ability of MV to induce immunosuppression may limit its capacity to directly activate B cells. These findings are relevant for our understanding of B-cell activation by two prototypic human pathogenic viruses and for the design of new recombinant vaccines.


Subject(s)
Immunoglobulin G/biosynthesis , Immunoglobulin M/biosynthesis , Measles virus/genetics , Measles virus/immunology , Poliovirus/immunology , T-Lymphocytes/immunology , Animals , Antibody Formation , CD4-Positive T-Lymphocytes/immunology , Chimera , Female , Humans , Lymphocyte Depletion , Measles virus/radiation effects , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Neutralization Tests , Poliovirus/genetics , RNA Viruses/genetics , RNA Viruses/immunology , T-Lymphocytes, Helper-Inducer/immunology , Ultraviolet Rays , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/immunology
17.
EMBO J ; 17(14): 3899-908, 1998 Jul 15.
Article in English | MEDLINE | ID: mdl-9670007

ABSTRACT

Measles viruses (MV) can be isolated from the brains of deceased subacute sclerosing panencephalitis patients only in a cell-associated form. These viruses are often defective in the matrix (M) protein and always seem to have an altered fusion protein cytoplasmic tail. We reconstituted a cell-free, infectious M-less MV (MV-DeltaM) from cDNA. In comparison with standard MV, MV-DeltaM was considerably more efficient at inducing cell-to-cell fusion but virus titres were reduced approximately 250-fold. In MV-DeltaM-induced syncytia the ribonucleocapsids and glycoproteins largely lost co-localization, confirming the role of M protein as the virus assembly organizer. Genetically modified mice were inoculated with MV-DeltaM or with another highly fusogenic virus bearing glycoproteins with shortened cytoplasmic tails (MV-Delta(tails)). MV-DeltaM and MV-Delta(tails) lost acute pathogenicity but penetrated more deeply into the brain parenchyma than standard MV. We suggest that enhanced cell fusion may also favour the propagation of mutated, assembly-defective MV in human brains.


Subject(s)
Brain/virology , Cell Fusion/physiology , Measles virus/pathogenicity , Subacute Sclerosing Panencephalitis/virology , Viral Matrix Proteins/physiology , Amino Acid Sequence , Animals , Antigens, CD/physiology , Brain/pathology , Chlorocebus aethiops , Genome, Viral , Giant Cells , Measles virus/physiology , Membrane Cofactor Protein , Membrane Glycoproteins/physiology , Mice , Molecular Sequence Data , Receptors, Virus/physiology , Subacute Sclerosing Panencephalitis/pathology , Vero Cells , Viral Structural Proteins/analysis , Virus Replication
18.
J Virol ; 72(3): 2150-9, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9499071

ABSTRACT

Measles virus (MV) and vesicular stomatitis virus (VSV) are both members of the Mononegavirales but are only distantly related. We generated two genetically stable chimeric viruses. In MGV, the reading frames of the MV envelope glycoproteins H and F were substituted by a single reading frame encoding the VSV G glycoprotein; MG/FV is similar but encodes a G/F hybrid in which the VSV G cytoplasmic tail was replaced by that of MV F. In contrast to MG/FV, MGV virions do not contain the MV matrix (M) protein. This demonstrates that virus assembly is possible in the absence of M; conversely, the cytoplasmic domain of F allows incorporation of M and enhances assembly. The formation of chimeric viruses was substantially delayed and the titers obtained were reduced about 50-fold in comparison to standard MV. In the novel chimeras, transcription and replication are mediated by the MV ribonucleoproteins but the envelope glycoproteins dictate the host range. Mice immunized with the chimeric viruses were protected against lethal doses of wild-type VSV. These findings suggest that it is feasible to construct MV variants bearing a variety of different envelopes for use as vaccines or for gene therapeutic purposes.


Subject(s)
Hemagglutinins, Viral/immunology , Measles virus/immunology , Membrane Glycoproteins , Rhabdoviridae Infections/prevention & control , Vesicular stomatitis Indiana virus/immunology , Viral Envelope Proteins/immunology , Viral Fusion Proteins/immunology , Amino Acid Sequence , Animals , Base Sequence , Biological Transport , Cell Line , Cell Line, Transformed , Chlorocebus aethiops , Cloning, Molecular , Cricetinae , Cytopathogenic Effect, Viral , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/metabolism , Humans , Measles virus/genetics , Measles virus/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Immunoelectron , Molecular Sequence Data , Plasmids , Rhabdoviridae Infections/immunology , Vero Cells , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Viral Proteins/biosynthesis , Virion/ultrastructure , Virus Assembly
19.
J Virol Methods ; 64(1): 57-64, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9029530

ABSTRACT

A system which allows the reconstitution of measles virus (MV) from cloned cDNA is described. The severely host cell restricted vaccinia vector MVA-T7 expressing bacteriophage T7 RNA polymerase was used to generate full-length antigenomic MV RNA and simultaneously the mRNAs encoding the viral N, P and L proteins in order to produce replicationally and transcriptionally active nucleocapsids. The functionality of the N, P and L proteins was demonstrated first by their ability to rescue MV specific subgenomic RNAs. Assembly and budding of reconstituted MV was shown by syncytia formation and subsequently by virus isolation. The inability of MVA-T7 to produce progeny virus in most mammalian cells circumvents the necessity to separate the reconstituted MV from the MVA-T7 helper virus. Since all components are expressed transiently, this system is especially suitable for studying the functions of N, P and L. Furthermore, it is useful for investigating later steps in the MV life cycle.


Subject(s)
Defective Viruses/genetics , Genetic Vectors , Measles virus/isolation & purification , Vaccinia virus/genetics , Animals , Bacteriophage T7/enzymology , Base Sequence , Cell Line , Chlorocebus aethiops , DNA, Viral/analysis , DNA-Directed RNA Polymerases/genetics , Genome, Viral , HeLa Cells , Humans , Measles virus/genetics , Measles virus/physiology , Molecular Sequence Data , Vero Cells , Viral Proteins , Virus Replication
20.
Virology ; 227(2): 314-22, 1997 Jan 20.
Article in English | MEDLINE | ID: mdl-9018130

ABSTRACT

The measles virus (MV) phosphoprotein (P) gene encodes three proteins, P, C, and V. The V protein is synthesized by pseudo-templated transcription, also designated as RNA editing: during P gene transcription one G residue is inserted at a defined position in about 50% of the mRNAs. To study the importance of sequence elements for the nontemplated G insertion, we generated recombinant MVs in which six different mutations were introduced within the region where editing occurs (3' UUUUUCCC, template strand). These viruses were then analyzed for their ability to edit their P mRNA and to produce V protein. Single U to C changes within the U stretch abolished editing. Extending the template by three C residues at the site of G insertion resulted in a less precise editing phenotype and overproduction of V. None of these mutants were impaired in their multiplication behavior when analyzed in cultured cells. However, the syncytia of a recombinant MV overproducing V protein were in general smaller and lysed 1 to 2 days later than usual.


Subject(s)
Defective Viruses/physiology , Measles virus/physiology , Phosphoproteins/genetics , RNA Editing , Viral Proteins/genetics , Virus Replication , Animals , Base Sequence , Cell Line , Chlorocebus aethiops , DNA Primers , Defective Viruses/genetics , HeLa Cells , Humans , Kidney , Kinetics , Measles virus/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphoproteins/biosynthesis , Polymerase Chain Reaction , Recombination, Genetic , Transcription, Genetic , Vero Cells , Viral Proteins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...