Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters











Publication year range
1.
J Virol ; 75(22): 11079-87, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11602747

ABSTRACT

Foreign glycoproteins expressed in recombinant vesicular stomatitis virus (VSV) can elicit specific and protective immunity in the mouse model. We have previously demonstrated the expression of respiratory syncytial virus (RSV) G (attachment) and F (fusion) glycoprotein genes in recombinant VSV. In this study, we demonstrate the expression of RSV F and G glycoproteins in attenuated, nonpropagating VSVs which lack the VSV G gene (VSVDeltaG) and the incorporation of these RSV proteins into recombinant virions. We also show that intranasal vaccination of mice with nondefective VSV recombinants expressing RSV G (VSV-RSV G) or RSV F (VSV-RSV F) elicited RSV-specific antibodies in serum (by enzyme-linked immunosorbent assay [ELISA]) as well as neutralizing antibodies to RSV and afford complete protection against RSV challenge. In contrast, VSVDeltaG-RSV F induced detectable serum antibodies to RSV by ELISA, but no detectable neutralizing antibodies, yet it still protected from RSV challenge. VSVDeltaG-RSV G failed to induce any detectable serum (by ELISA) or neutralizing antibodies and failed to protect from RSV challenge. The attenuated, nonpropagating VSVDeltaG-RSV F is a particularly attractive candidate for a live attenuated recombinant RSV vaccine.


Subject(s)
Antigens, Viral/immunology , Respiratory Syncytial Virus Infections/prevention & control , Vaccines, Synthetic/immunology , Vesicular stomatitis Indiana virus/genetics , Viral Proteins/immunology , Viral Vaccines/immunology , Virus Replication , Administration, Intranasal , Animals , Base Sequence , Cell Line , Cricetinae , Female , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Respiratory Syncytial Virus Infections/pathology , Vaccines, Attenuated/immunology
2.
Virology ; 287(2): 427-35, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11531419

ABSTRACT

We investigated the stability and mechanisms of loss of foreign gene expression in two recombinant vesicular stomatitis viruses (VSVs). A recombinant expressing the cellular CD4 protein exhibited remarkable stability of foreign gene expression. However, after 26 sequential passages, a mutant no longer expressing CD4 was recovered from the virus stock. Sequencing of the CD4 coding region in this mutant revealed a single nucleotide deletion causing a frameshift and termination of protein synthesis. A second VSV recombinant expressing the measles virus F protein grew poorly and exhibited extreme instability of expression of the F protein. Expression of F protein was lost rapidly through mutations of the upstream transcription termination site from (3')AUAC(5') to (3')AUAU(5'), as well as lengthening of the subsequent U(7) tract that is the template for poly(A) addition to VSV G mRNA. Such mutations resulted in fusion of the F mRNA to the 3' end of the G mRNA, making the F protein translation initiation codon inaccessible. We suggest that the VSV polymerase is error prone during replication of the U(7) tract, providing a rapid means for complete elimination of expression of proteins that are toxic to the virus life cycle.


Subject(s)
Gene Expression , Vesicular stomatitis Indiana virus/genetics , Animals , Blotting, Northern , CD4 Antigens/biosynthesis , Cells, Cultured , Cricetinae , Measles virus/genetics , RNA, Messenger/biosynthesis , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction
3.
Cell ; 106(5): 539-49, 2001 Sep 07.
Article in English | MEDLINE | ID: mdl-11551502

ABSTRACT

We developed an AIDS vaccine based on attenuated VSV vectors expressing env and gag genes and tested it in rhesus monkeys. Boosting was accomplished using vectors with glycoproteins from different VSV serotypes. Animals were challenged with a pathogenic AIDS virus (SHIV89.6P). Control monkeys showed a severe loss of CD4+ T cells and high viral loads, and 7/8 progressed to AIDS with an average time of 148 days. All seven vaccinees were initially infected with SHIV89.6P but have remained healthy for up to 14 months after challenge with low or undetectable viral loads. Protection from AIDS was highly significant (p = 0.001). VSV vectors are promising candidates for human AIDS vaccine trials because they propagate to high titers and can be delivered without injection.


Subject(s)
AIDS Vaccines/immunology , Acquired Immunodeficiency Syndrome/immunology , Vesicular stomatitis Indiana virus/genetics , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Acquired Immunodeficiency Syndrome/prevention & control , Acquired Immunodeficiency Syndrome/virology , Animals , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , Enzyme-Linked Immunosorbent Assay , Gene Products, env/genetics , Gene Products, env/immunology , Gene Products, gag/genetics , Gene Products, gag/immunology , HIV/immunology , HIV/physiology , HIV Antibodies/biosynthesis , Humans , Immunization, Secondary , Macaca mulatta , Mice , Neutralization Tests , Pilot Projects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , SAIDS Vaccines/genetics , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/physiology , T-Lymphocytes, Cytotoxic/immunology , Time Factors , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/immunology , Vesicular stomatitis Indiana virus/immunology , Viral Load , Virus Shedding
4.
J Virol ; 74(23): 10903-10, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11069984

ABSTRACT

Live recombinant vesicular stomatitis viruses (VSVs) expressing foreign antigens are highly effective vaccine vectors. However, these vectors induce high-titer neutralizing antibody directed at the single VSV glycoprotein (G), and this antibody alone can prevent reinfection and boosting with the same vector. To determine if efficient boosting could be achieved by changing the G protein of the vector, we have developed two new recombinant VSV vectors based on the VSV Indiana serotype but with the G protein gene replaced with G genes from two other VSV serotypes, New Jersey and Chandipura. These G protein exchange vectors grew to titers equivalent to wild-type VSV and induced similar neutralizing titers to themselves but no cross-neutralizing antibodies to the other two serotypes. The effectiveness of these recombinant VSV vectors was illustrated in experiments in which sequential boosting of mice with the three vectors, all encoding the same primary human immunodeficiency virus (HIV) envelope protein, gave a fourfold increase in antibody titer to an oligomeric HIV envelope compared with the response in animals receiving the same vector three times. In addition, only the animals boosted with the exchange vectors produced antibodies neutralizing the autologous HIV primary isolate. These VSV envelope exchange vectors have potential as vaccines in immunizations when boosting of immune responses may be essential.


Subject(s)
AIDS Vaccines/immunology , HIV Antibodies/biosynthesis , HIV-1/immunology , Membrane Glycoproteins , Vaccines, Synthetic/immunology , Vesicular stomatitis Indiana virus/genetics , Vesiculovirus , Viral Envelope Proteins/immunology , Animals , Female , Gene Products, env/immunology , Genetic Vectors , Immunization, Secondary , Mice , Mice, Inbred BALB C , Vaccinia virus/genetics , Vesicular stomatitis Indiana virus/pathogenicity
5.
J Infect Dis ; 182(4): 1228-33, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10979923

ABSTRACT

Respiratory syncytial virus (RSV) encodes a short (64 or 65 amino acids) hydrophobic (SH) protein whose function in viral replication and pathogenesis is not understood. We carried out molecular epidemiological studies of the SH gene during the 1998-1999 seasonal epidemic in New Haven, Connecticut. Strains circulating during the epidemic were related to viruses identified worldwide. The SH gene transcriptional control signals were conserved in 70 (98.6%) of 71 isolates that we sequenced. The deduced amino acid sequence of the SH protein was nearly identical to subgroup A and subgroup B reference strains that were isolated in 1961 and 1962, respectively. Twenty-six (96.3%) of 27 subgroup A strains contained 0 or 1 amino acid substitution, compared with that of the reference A2 strain. Most subgroup B isolates (38 [86.4%] of 44 strains) contained 0, 1, or 2 amino acid substitutions, compared with that of the reference B18537 strain.


Subject(s)
Respiratory Syncytial Viruses/genetics , Viral Proteins/genetics , Amino Acid Sequence , Base Sequence , Connecticut/epidemiology , DNA Primers , Genes, Viral , Humans , Molecular Sequence Data , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Viral Proteins/chemistry , Viral Structural Proteins/genetics
6.
Virus Res ; 69(1): 3-15, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10989181

ABSTRACT

We generated a recombinant vesicular stomatitis virus (VSV-E2) encoding the bovine viral diarrhea virus (BVDV) E2 glycoprotein with the VSV-G protein signal peptide. Infection of BHK21 cells with VSV-E2 induced the synthesis of a recombinant E2 (rE2) that comigrated with authentic BVDV-E2 in PAGE-SDS gels. Non-reducing immunoblots showed that rE2 is a disulfide bond-linked homodimer with at least 10-fold higher avidity for conformation-dependent anti-BVDV-E2 antibodies than its reduced monomeric counterpart. Immunofluorescence microscopy also showed that rE2 was transported to the plasma membrane of infected cells and analysis of purified particles demonstrated that dimeric rE2 was incorporated into VSV-E2 virions in approximately 1:10 ratio with respect to the G glycoprotein. BALB/c mice inoculated intranasally with VSV-E2 doses of up to 10(7) plaque forming units (pfu) showed no symptoms of viral-induced disease and developed a specific BVDV neutralizing response that lasted for at least 180 days post inoculation.


Subject(s)
Antibodies, Viral/biosynthesis , Diarrhea Viruses, Bovine Viral/genetics , Diarrhea Viruses, Bovine Viral/immunology , Membrane Glycoproteins , Vesicular stomatitis Indiana virus/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Viral/blood , Base Sequence , Cattle , Cell Line , Chimera/genetics , Chimera/immunology , Cricetinae , DNA, Recombinant/genetics , Female , Mice , Mice, Inbred BALB C , Neutralization Tests , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Vaccines, Synthetic/genetics , Viral Vaccines/genetics
7.
J Virol ; 74(10): 4652-7, 2000 May.
Article in English | MEDLINE | ID: mdl-10775601

ABSTRACT

In humans, maternal antibodies inhibit successful immunization against measles, because they interfere with vaccine-induced seroconversion. We have investigated this problem using the cotton rat model (Sigmodon hispidus). As in humans, passively transferred antibodies inhibit the induction of measles virus (MV)-neutralizing antibodies and protection after immunization with MV. In contrast, a recombinant vesicular stomatitis virus (VSV) expressing the MV hemagglutinin (VSV-H) induces high titers of neutralizing antibodies to MV in the presence of MV-specific antibodies. The induction of neutralizing antibodies increased with increasing virus dose, and all doses gave good protection from subsequent challenge with MV. Induction of antibodies by VSV-H was observed in the presence of passively transferred human or cotton rat antibodies, which were used as the models of maternal antibodies. Because MV hemagglutinin is not a functional part of the VSV-H envelope, MV-specific antibodies only slightly inhibit VSV-H replication in vitro. This dissociation of function and antigenicity is probably key to the induction of a neutralizing antibody in the presence of a maternal antibody.


Subject(s)
Antibodies, Viral/biosynthesis , Immunity, Maternally-Acquired , Measles Vaccine/immunology , Measles virus/immunology , Measles/prevention & control , Animals , Antibodies, Viral/immunology , Antibody Specificity , Disease Models, Animal , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/metabolism , Humans , Immunization, Passive , Measles/immunology , Measles Vaccine/administration & dosage , Measles Vaccine/genetics , Neutralization Tests , Rats , Sigmodontinae , Vaccination , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/immunology
8.
J Virol ; 73(5): 3723-32, 1999 May.
Article in English | MEDLINE | ID: mdl-10196265

ABSTRACT

We showed previously that a single intranasal vaccination of mice with a recombinant vesicular stomatitis virus (VSV) expressing an influenza virus hemagglutinin (HA) protein provided complete protection from lethal challenge with influenza virus (A. Roberts, E. Kretzschmar, A. S. Perkins, J. Forman, R. Price, L. Buonocore, Y. Kawaoka, and J. K. Rose, J. Virol. 72:4704-4711, 1998). Because some pathogenesis was associated with the vector itself, in the present study we generated new VSV vectors expressing HA which are completely attenuated for pathogenesis in the mouse model. The first vector has a truncation of the cytoplasmic domain of the VSV G protein and expresses influenza virus HA (CT1-HA). This nonpathogenic vector provides complete protection from lethal influenza virus challenge after intranasal administration. A second vector with VSV G deleted and expressing HA (DeltaG-HA) is also protective and nonpathogenic and has the advantage of not inducing neutralizing antibodies to the vector itself.


Subject(s)
Genetic Vectors , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/immunology , Membrane Glycoproteins , Vaccines, Synthetic/immunology , Vesicular stomatitis Indiana virus , Animals , Cattle , Cell Line , Cricetinae , Female , Gene Expression , Genetic Vectors/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza Vaccines/genetics , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/prevention & control , Recombination, Genetic , Time Factors , Vaccines, Attenuated/immunology , Vesicular stomatitis Indiana virus/immunology , Viral Envelope Proteins/genetics , Virion/metabolism
9.
Virology ; 254(1): 81-91, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-9927576

ABSTRACT

The genes encoding the respiratory syncytial virus (RSV) attachment (G) and fusion (F) envelope glycoproteins were expressed separately as additional genes in recombinant vesicular stomatitis viruses (VSV). Cells infected with the VSV-RSV F recombinant formed large syncytia illustrating the fusion activity of F in absence of other RSV proteins. Both F and G glycoproteins were expressed at the cell surface and incorporated into virions. Incorporation of these proteins did not require cytoplasmic tail sequences of VSV G. Using a compound, ammonium chloride, that raises the endosomal pH, we showed that presence of the RSV F glycoprotein in the envelope of recombinant VSV allowed for infectivity through a low-pH-independent pathway. Recombinant VSV expressing RSV glycoproteins could be useful as an RSV vaccine.


Subject(s)
Genetic Vectors , Glycoproteins/metabolism , HN Protein , Respiratory Syncytial Virus, Human/metabolism , Vesicular stomatitis Indiana virus , Viral Envelope Proteins/metabolism , Viral Fusion Proteins/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Cricetinae , Cytoplasm/metabolism , Gene Expression , Glycoproteins/genetics , Humans , Membrane Fusion , Molecular Sequence Data , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Respiratory Syncytial Virus, Human/genetics , Tumor Cells, Cultured , Viral Envelope Proteins/genetics , Viral Fusion Proteins/genetics , Viral Proteins/genetics , Virion/metabolism
10.
J Virol ; 72(6): 4704-11, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9573234

ABSTRACT

Since the development of a system for generating vesicular stomatitis virus (VSV) from plasmid DNAs, our laboratory has reported the expression of several different glycoproteins from recombinant VSVs. In one of these studies, high-level expression of an influenza virus hemagglutinin (HA) from a recombinant VSV-HA and efficient incorporation of the HA protein into the virions was reported (E. Kretzschmar, L. Buonocore, M. J. Schnell, and J. K. Rose, J. Virol. 71:5982-5989, 1997). We report here that VSV-HA is an effective intranasal vaccine vector that raises high levels of neutralizing antibody to influenza virus and completely protects mice from bronchial pneumonia caused by challenge with a lethal dose of influenza A virus. Additionally, these recombinant VSVs are less pathogenic than wild-type VSV (serotype Indiana). This vector-associated pathogenicity was subsequently eliminated through introduction of specific attenuating deletions. These live attenuated recombinant VSVs have great potential as vaccine vectors.


Subject(s)
Hemagglutinins/immunology , Influenza A virus/immunology , Orthomyxoviridae Infections/immunology , Vaccines, Synthetic/immunology , Vesicular stomatitis Indiana virus/immunology , Viral Vaccines/immunology , Administration, Inhalation , Animals , DNA, Recombinant/genetics , Female , Hemagglutinins/genetics , Immunity , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/prevention & control , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vesicular stomatitis Indiana virus/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
11.
EMBO J ; 17(5): 1289-96, 1998 Aug 10.
Article in English | MEDLINE | ID: mdl-9482726

ABSTRACT

The cytoplasmic domains of viral glycoproteins are often involved in specific interactions with internal viral components. These interactions can concentrate glycoproteins at virus budding sites and drive efficient virus budding, or can determine virion morphology. To investigate the role of the vesicular stomatitis virus (VSV) glycoprotein (G) cytoplasmic and transmembrane domains in budding, we recovered recombinant VSVs expressing chimeric G proteins with the transmembrane and cytoplasmic domains derived from the human CD4 protein. These unrelated foreign sequences were capable of supporting efficient VSV budding. Further analysis of G protein cytoplasmic domain deletion mutants showed that a cytoplasmic domain of only 1 amino acid did not drive efficient budding, whereas 9 amino acids did. Additional studies in agreement with the CD4-chimera experiments indicated the requirement for a short cytoplasmic domain on VSV G without the requirement for a specific sequence in that domain. We propose a model for VSV budding in which a relatively non-specific interaction of a cytoplasmic domain with a pocket or groove in the viral nucleocapsid or matrix proteins generates a glycoprotein array that promotes viral budding.


Subject(s)
Membrane Glycoproteins , Vesicular stomatitis Indiana virus/growth & development , Viral Envelope Proteins/chemistry , Amino Acid Sequence , Animals , CD4 Antigens/genetics , Cell Line , Cell Membrane/virology , Cricetinae , Cytopathogenic Effect, Viral , Cytoplasm , Humans , Molecular Sequence Data , Mutation , Recombinant Fusion Proteins , Sequence Deletion , Serial Passage , Viral Envelope Proteins/biosynthesis , Viral Envelope Proteins/genetics , Viral Proteins/biosynthesis , Virion/ultrastructure
12.
Cell ; 90(5): 849-57, 1997 Sep 05.
Article in English | MEDLINE | ID: mdl-9298897

ABSTRACT

We describe a recombinant vesicular stomatitis virus lacking its glycoprotein gene and expressing instead the HIV-1 receptor CD4 and a coreceptor, CXCR4. This virus was unable to infect normal cells but did infect, propagate on, and kill cells that were first infected with HIV-1 and therefore had the HIV membrane fusion protein on their surface. Killing of HIV-1-infected cells controlled HIV infection in a T cell line and reduced titers of infectious HIV-1 in the culture by as much as 10(4)-fold. Such a targeted virus could have therapeutic value in reducing HIV viral load. Our results also demonstrate a general strategy of targeting one virus to the envelope protein of another virus to control infection.


Subject(s)
HIV Infections/therapy , HIV Infections/virology , HIV-1/physiology , Membrane Glycoproteins , Vesicular stomatitis Indiana virus/physiology , Animals , CD4 Antigens/genetics , Cricetinae , GTP-Binding Proteins/genetics , Gene Deletion , Gene Expression Regulation, Viral/physiology , Glycoproteins/genetics , HIV-1/growth & development , Humans , Jurkat Cells/virology , Kidney/cytology , Membrane Proteins/genetics , Microscopy, Immunoelectron , Mutagenesis/physiology , Receptors, CXCR4 , Receptors, HIV/genetics , Recombinant Fusion Proteins/physiology , Vesicular stomatitis Indiana virus/growth & development , Vesicular stomatitis Indiana virus/ultrastructure , Viral Envelope Proteins/genetics , Viral Envelope Proteins/physiology , Virus Replication
13.
J Virol ; 71(8): 5982-9, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9223488

ABSTRACT

We derived recombinant vesicular stomatitis virus (VSV) expressing either influenza virus hemagglutinin (HA) or neuraminidase (NA) glycoproteins from extra genes inserted in the viral genome. The HA protein was expressed from a site downstream of the VSV glycoprotein (G) gene, while NA protein was expressed from a site upstream of the VSV G gene. The HA protein was expressed at lower levels than the VSV G protein, while the NA protein was expressed at higher levels, as expected from the gradient of VSV transcription that follows the gene order. The HA and NA proteins were transported to the cell surface and were functional as demonstrated by hemadsorption, hemolysis, and NA assays. Biochemical analysis showed that both HA and NA proteins were incorporated into VSV particles at high levels, although there was a preference for incorporation of the VSV G protein over either of the influenza virus proteins. Immunoelectron microscopy of the recombinants showed that the particles derived from the recombinants were mosaics carrying both the VSV G protein and the influenza virus membrane glycoproteins. These results extend earlier studies showing incorporation of the cellular glycoprotein CD4 and two other viral glycoproteins into VSV particles. Our results indicate that there is significant space in the VSV membrane that can accommodate foreign membrane proteins and that the foreign protein can represent as much as 35% of the total protein in the viral envelope. Incorporation of foreign proteins into VSV virions can, in many cases, occur passively in the absence of specific incorporation signals.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/biosynthesis , Neuraminidase/biosynthesis , Recombinant Proteins/biosynthesis , Vesicular stomatitis Indiana virus/genetics , Animals , Base Sequence , Cricetinae , Molecular Sequence Data , Vaccines, Synthetic/biosynthesis , Virion/genetics
14.
J Virol ; 71(7): 5060-8, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9188571

ABSTRACT

We generated replication-competent, recombinant vesicular stomatitis viruses (VSVs) expressing the human immunodeficiency virus (HIV) envelope protein or an HIV-VSV chimeric envelope protein in which the cytoplasmic domain of the HIV envelope protein was replaced with that from the VSV glycoprotein (G). These recombinants were generated with HIV type 1 (HIV-1) envelopes from both laboratory and primary isolates of HIV-1. The replication-competent recombinant viruses were stable and expressed the foreign proteins at high levels from extra transcription units in VSV. The foreign proteins were processed appropriately and transported to the cell surface. The incorporation of HIV gp120 into VSV particles was demonstrated biochemically only for the construct expressing the chimeric envelopes containing the VSV G cytoplasmic domain. The incorporation of the chimeric HIV envelope protein into the membrane of the recombinant VSV was also demonstrated by electron microscopy with gold-conjugated antibodies. To determine whether specific infection of CD4-positive cells could be demonstrated for these recombinants, we neutralized VSV infectivity due to VSV glycoprotein with anti-VSV serum. The neutralized recombinants expressing the chimeric envelope were able to infect only HeLa cells expressing CD4, and this CD4-specific infectivity was neutralized with anti-HIV serum. This assay also detected a 100-fold-lower titer of CD4-specific infectivity for the VSV recombinant expressing the wild-type HIV envelope. Our results illustrate that it is possible to express functional HIV envelopes from the VSV genome and target the recombinant virus to an alternative receptor. The recombinants may also prove useful as HIV vaccines.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Genetic Vectors , HIV Envelope Protein gp160/genetics , Membrane Glycoproteins , Vesicular stomatitis Indiana virus/metabolism , Viral Envelope Proteins/genetics , Animals , Cell Line , Cricetinae , Gene Expression , HIV Envelope Protein gp160/metabolism , HIV-1/genetics , HeLa Cells , Humans , Protein Processing, Post-Translational , Rabbits , Recombinant Fusion Proteins/genetics , Recombination, Genetic , Vesicular stomatitis Indiana virus/growth & development , Vesicular stomatitis Indiana virus/ultrastructure , Virion/metabolism , Virion/ultrastructure
15.
Proc Natl Acad Sci U S A ; 93(21): 11359-65, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8876140

ABSTRACT

In a previous study we demonstrated that vesicular stomatitis virus (VSV) can be used as a vector to express a soluble protein in mammalian cells. Here we have generated VSV recombinants that express four different membrane proteins: the cellular CD4 protein, a CD4-G hybrid protein containing the ectodomain of CD4 and the transmembrane and cytoplasmic tail of the VSV glycoprotein (G), the measles virus hemagglutinin, or the measles virus fusion protein. The proteins were expressed at levels ranging from 23-62% that of VSV G protein and all were transported to the cell surface. In addition we found that all four proteins were incorporated into the membrane envelope of VSV along with the VSV G protein. The levels of incorporation of these proteins varied from 6-31% of that observed for VSV G. These results suggest that many different membrane proteins may be co-incorporated quite efficiently with VSV G protein into budding VSV virus particles and that specific signals are not required for this co-incorporation process. In fact, the CD4-G protein was incorporated with the same efficiency as wild type CD4. Electron microscopy of virions containing CD4 revealed that the CD4 molecules were dispersed throughout the virion envelope among the trimeric viral spike glycoproteins. The recombinant VSV-CD4 virus particles were about 18% longer than wild type virions, reflecting the additional length of the helical nucleocapsid containing the extra gene. Recombinant VSVs carrying foreign antigens on the surface of the virus particle may be useful for viral targeting, membrane protein purification, and for generation of immune responses.


Subject(s)
Genetic Vectors , Glycoproteins/biosynthesis , Membrane Glycoproteins , Transfection/methods , Vesicular stomatitis Indiana virus/genetics , Viral Envelope Proteins/biosynthesis , Animals , Antigens, CD/biosynthesis , CD4 Antigens/biosynthesis , Cell Line , Cricetinae , Hemagglutinins, Viral/biosynthesis , Humans , Kidney , Measles virus/genetics , Measles virus/metabolism , Microscopy, Immunoelectron , Recombinant Proteins/biosynthesis , Vesicular stomatitis Indiana virus/metabolism , Viral Fusion Proteins/biosynthesis
16.
J Virol ; 70(4): 2318-23, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8642658

ABSTRACT

A new transcription unit was generated in the 3' noncoding region of the vesicular stomatitis virus (VSV) glycoprotein gene by introducing the smallest conserved sequence found at each VSV gene junction. This sequence was introduced into a DNA copy of the VSV genome from which infectious VSV can be derived. It contained an 11-nucleotide putative transcription stop/polyadenylation signal for the glycoprotein mRNA, an intergenic dinucleotide, and a 10-nucleotide putative transcription start sequence preceding a downstream foreign gene encoding the bacterial enzyme chloramphenicol acetyltransferase. Infectious recombinant VSV was recovered from this construct and was found to express high levels of functional chloramphenicol acetyltransferase mRNA and protein. The recombinant virus grew to wild-type titers of 5 x 10(9)/ml, and expression of the foreign gene was completely stable for at least 15 passages involving 10(6)-fold expansion at each passage. These results define functionally the transcription stop/polyadenylation and start sequences for VSV and also illustrate the utility of VSV as a stable vector that should have wide application in cell biology and vaccine development.


Subject(s)
Cloning, Molecular/methods , Gene Expression Regulation, Viral , Membrane Glycoproteins , Regulatory Sequences, Nucleic Acid , Vesicular stomatitis Indiana virus/genetics , Base Sequence , Chloramphenicol O-Acetyltransferase/genetics , Conserved Sequence , Molecular Sequence Data , RNA, Viral/genetics , Serial Passage , Transcription, Genetic , Viral Envelope Proteins/genetics , Viral Plaque Assay
17.
Virology ; 204(1): 482-6, 1994 Oct.
Article in English | MEDLINE | ID: mdl-8091684

ABSTRACT

The small membrane protein Vpu of human immunodeficiency virus type 1 stimulates rapid degradation of CD4 molecules that are retained in the endoplasmic reticulum. To analyze the domain(s) of CD4 involved in Vpu-stimulated degradation, we examined degradation of hybrid proteins made between the vesicular stomatitis virus glycoprotein (VSV G) and CD4. Vpu expression stimulated rapid degradation of a hybrid consisting of the extracellular domain of VSV G linked to the transmembrane and cytoplasmic domains of CD4. Analysis of additional hybrids showed that both the cytoplasmic and transmembrane domains of CD4 were required for this Vpu-stimulated degradation. This conclusion is in apparent conflict with a recent study showing that the cytoplasmic domain of CD4 alone is sufficient to cause Vpu-stimulated degradation of a CD8-CD4 hybrid protein. The apparent conflict may be explained by the presence of related sequences or structures in the transmembrane domains of CD4 and CD8 that are involved in binding Vpu directly or that interact with the Vpu-stimulated degradation system.


Subject(s)
CD4 Antigens/metabolism , HIV-1/metabolism , Membrane Glycoproteins , Viral Regulatory and Accessory Proteins/metabolism , Amino Acid Sequence , Base Sequence , CD4 Antigens/chemistry , CD4 Antigens/genetics , Glycoproteins/genetics , HeLa Cells , Human Immunodeficiency Virus Proteins , Humans , Molecular Sequence Data , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Viral Envelope Proteins/genetics
18.
Proc Natl Acad Sci U S A ; 90(7): 2695-9, 1993 Apr 01.
Article in English | MEDLINE | ID: mdl-8464877

ABSTRACT

A retroviral vector was constructed in which a gene encoding a mutated soluble CD4 protein that is retained in the endoplasmic reticulum (sCD4-KDEL) is expressed under control of human immunodeficiency virus type 1 (HIV-1) regulatory elements. HIV-1 infection of a human T-cell line transduced with this vector led to induction of sCD4-KDEL synthesis and a block in transport of the HIV envelope protein to the cell surface. There was a complete block to maturation of infectious HIV-1 in the transduced cells, no viral spread, and little or no syncytium formation. Infected cells gradually disappeared from the culture over a period of 2 months. This intracellular trap for HIV has potential application in gene therapy for AIDS.


Subject(s)
CD4 Antigens/physiology , HIV Long Terminal Repeat , HIV-1/physiology , T-Lymphocyte Subsets/physiology , Virus Replication , Acquired Immunodeficiency Syndrome/therapy , CD4 Antigens/genetics , Cell Line , Fluorescent Antibody Technique , Genetic Therapy , Genetic Vectors , HIV Core Protein p24/analysis , HIV Core Protein p24/biosynthesis , HIV-1/genetics , HIV-1/immunology , Humans , Restriction Mapping , T-Lymphocyte Subsets/microbiology
19.
Virology ; 185(2): 681-8, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1660200

ABSTRACT

The spike glycoprotein (G protein) of rabies virus (CVS strain) expressed in HeLa cells from cloned cDNA mediated membrane fusion after exposure to pHs of 6.1 or below. Chemical crosslinking showed that the rabies G protein, like the vesicular stomatitis virus (VSV) G protein, could be crosslinked to dimers and trimers, indicating that rabies G protein is a trimer. However, unlike the VSV G protein, rabies G protein trimers were not stable to sedimentation in sucrose gradients, even at a mildly acidic pH which stabilizes the VSV G protein trimers. In addition, we report that the expressed rabies virus G protein was functional because it could assemble into VSV particles (tsO45) lacking VSV G protein and rescue infectivity. These VSV (rabies) pseudotypes were neutralized only by an antibody to the rabies G protein. We also examined the properties of a hybrid protein containing the extracellular domain of the rabies virus glycoprotein and the transmembrane and cytoplasmic domains of the VSV G protein. This protein was transported to the cell surface and could be crosslinked to form dimers and trimers, but had little or no detectable membrane fusion activity. The lack of fusion activity was paradoxical because the hybrid protein could rescue VSV infectivity, although the titers were lower than those obtained with the wild-type rabies G protein.


Subject(s)
Antigens, Viral , Glycoproteins/metabolism , Membrane Fusion , Membrane Glycoproteins , Rabies virus/metabolism , Vesicular stomatitis Indiana virus/metabolism , Viral Envelope Proteins/metabolism , Viral Fusion Proteins/metabolism , Animals , Base Sequence , Cell Line/microbiology , Centrifugation, Density Gradient , Cricetinae , Flow Cytometry , Genetic Complementation Test , Glycoproteins/chemistry , Glycoproteins/genetics , Humans , Hydrogen-Ion Concentration , Kinetics , Macromolecular Substances , Mice , Molecular Sequence Data , Plasmids/genetics , Rabies virus/genetics , Recombinant Fusion Proteins/metabolism , Vesicular stomatitis Indiana virus/genetics , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Fusion Proteins/chemistry
20.
Biotechniques ; 10(4): 520-5, 1991 Apr.
Article in English | MEDLINE | ID: mdl-1867862

ABSTRACT

One of the most efficient systems for the expression of genes in the cytoplasm of animal cells utilizes a recombinant vaccinia virus encoding the bacteriophage T7 RNA polymerase. Cells infected with this virus are transfected with plasmid DNAs containing the gene to be expressed under T7 promoter control. The major limitation of this system is the efficiency with which DNA is introduced into the cell. Recently, a cationic liposome-mediated transfection reagent has yielded transfection frequencies of greater than 80%. To determine if commercially available cationic lipids could form liposomes that would yield similar transfection efficiencies, we tested liposomes prepared with five different cationic lipids. When used at appropriate concentrations in liposomes that also contained a neutral lipid, four of the five cationic lipids were effective in the transfection of HeLa cells. However, liposomes formed with the neutral lipid and one of the cationic lipids, dimethyldioctadecylammonium bromide (DDAB), gave transfection frequencies of greater than 95% and had a broad spectrum of effectiveness on a variety of cell lines. Liposomes containing DDAB are an inexpensive, highly efficient and reproducible alternative for the transfection of animal cells and are well suited for use with the vaccinia virus/T7 expression system.


Subject(s)
Liposomes , Transfection , Animals , Cations , Cell Line , DNA , Dogs , Flow Cytometry , HeLa Cells , Humans , Microscopy, Fluorescence , Quaternary Ammonium Compounds
SELECTION OF CITATIONS
SEARCH DETAIL