Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Bioconjug Chem ; 29(10): 3362-3376, 2018 10 17.
Article in English | MEDLINE | ID: mdl-30169965

ABSTRACT

The impact of influenza virus infection is felt each year on a global scale when approximately 5-10% of adults and 20-30% of children globally are infected. While vaccination is the primary strategy for influenza prevention, there are a number of likely scenarios for which vaccination is inadequate, making the development of effective antiviral agents of utmost importance. Anti-influenza treatments with innovative mechanisms of action are critical in the face of emerging viral resistance to the existing drugs. These new antiviral agents are urgently needed to address future epidemic (or pandemic) influenza and are critical for the immune-compromised cohort who cannot be vaccinated. We have previously shown that lipid tagged peptides derived from the C-terminal region of influenza hemagglutinin (HA) were effective influenza fusion inhibitors. In this study, we modified the influenza fusion inhibitors by adding a cell penetrating peptide sequence to promote intracellular targeting. These fusion-inhibiting peptides self-assemble into ∼15-30 nm nanoparticles (NPs), target relevant infectious tissues in vivo, and reduce viral infectivity upon interaction with the cell membrane. Overall, our data show that the CPP and the lipid moiety are both required for efficient biodistribution, fusion inhibition, and efficacy in vivo.


Subject(s)
Antiviral Agents/pharmacology , Cell-Penetrating Peptides/pharmacology , Influenza A virus/drug effects , Membrane Fusion/drug effects , Administration, Intranasal , Amino Acid Sequence , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Biological Availability , Cell Membrane/metabolism , Cell-Penetrating Peptides/chemistry , Endocytosis , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Immunocompromised Host , Nanoparticles/chemistry , Sigmodontinae , Viral Proteins/chemistry , tat Gene Products, Human Immunodeficiency Virus/chemistry
2.
J Virol ; 91(1)2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27733647

ABSTRACT

Measles virus (MV) infection is undergoing resurgence and remains one of the leading causes of death among young children worldwide despite the availability of an effective measles vaccine. MV infects its target cells by coordinated action of the MV hemagglutinin (H) and fusion (F) envelope glycoproteins; upon receptor engagement by H, the prefusion F undergoes a structural transition, extending and inserting into the target cell membrane and then refolding into a postfusion structure that fuses the viral and cell membranes. By interfering with this structural transition of F, peptides derived from the heptad repeat (HR) regions of F can inhibit MV infection at the entry stage. In previous work, we have generated potent MV fusion inhibitors by dimerizing the F-derived peptides and conjugating them to cholesterol. We have shown that prophylactic intranasal administration of our lead fusion inhibitor efficiently protects from MV infection in vivo We show here that peptides tagged with lipophilic moieties self-assemble into nanoparticles until they reach the target cells, where they are integrated into cell membranes. The self-assembly feature enhances biodistribution and the half-life of the peptides, while integration into the target cell membrane increases fusion inhibitor potency. These factors together modulate in vivo efficacy. The results suggest a new framework for developing effective fusion inhibitory peptides. IMPORTANCE: Measles virus (MV) infection causes an acute illness that may be associated with infection of the central nervous system (CNS) and severe neurological disease. No specific treatment is available. We have shown that fusion-inhibitory peptides delivered intranasally provide effective prophylaxis against MV infection. We show here that specific biophysical properties regulate the in vivo efficacy of MV F-derived peptides.


Subject(s)
Hemagglutinins, Viral/immunology , Measles Vaccine/administration & dosage , Measles virus/drug effects , Measles/prevention & control , Nanoparticles/administration & dosage , Peptides/immunology , Viral Fusion Proteins/immunology , Administration, Intranasal , Amino Acid Sequence , Animals , Brain/drug effects , Brain/immunology , Cholesterol/chemistry , Female , Half-Life , Hemagglutinins, Viral/chemistry , Humans , Lung/drug effects , Lung/immunology , Male , Measles/immunology , Measles/mortality , Measles/virology , Measles Vaccine/chemical synthesis , Measles virus/chemistry , Measles virus/immunology , Nanoparticles/chemistry , Peptides/chemical synthesis , Sigmodontinae , Survival Analysis , Viral Fusion Proteins/chemistry , Virus Internalization/drug effects
3.
J Virol ; 89(2): 1143-55, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25378493

ABSTRACT

UNLABELLED: Measles virus (MV) infection is undergoing resurgence and remains one of the leading causes of death among young children worldwide despite the availability of an effective measles vaccine. MV infects its target cells by coordinated action of the MV H and the fusion (F) envelope glycoprotein; upon receptor engagement by H, the prefusion F undergoes a structural transition, extending and inserting into the target cell membrane and then refolding into a postfusion structure that fuses the viral and cell membranes. By interfering with this structural transition of F, peptides derived from the heptad-repeat (HR) regions of F can potently inhibit MV infection at the entry stage. We show here that specific features of H's interaction with its receptors modulate the susceptibility of MV F to peptide fusion inhibitors. A higher concentration of inhibitory peptides is required to inhibit F-mediated fusion when H is engaged to its nectin-4 receptor than when H is engaged to its CD150 receptor. Peptide inhibition of F may be subverted by continued engagement of receptor by H, a finding that highlights the ongoing role of H-receptor interaction after F has been activated and that helps guide the design of more potent inhibitory peptides. Intranasal administration of these peptides results in peptide accumulation in the airway epithelium with minimal systemic levels of peptide and efficiently prevents MV infection in vivo in animal models. The results suggest an antiviral strategy for prophylaxis in vulnerable and/or immunocompromised hosts. IMPORTANCE: Measles virus (MV) infection causes an acute illness that may be associated with infection of the central nervous system (CNS) and severe neurological disease. No specific treatment is available. We have shown that parenterally delivered fusion-inhibitory peptides protect mice from lethal CNS MV disease. Here we show, using established small-animal models of MV infection, that fusion-inhibitory peptides delivered intranasally provide effective prophylaxis against MV infection. Since the fusion inhibitors are stable at room temperature, this intranasal strategy is feasible even outside health care settings, could be used to protect individuals and communities in case of MV outbreaks, and could complement global efforts to control measles.


Subject(s)
Antiviral Agents/administration & dosage , Measles virus/drug effects , Measles/prevention & control , Oligopeptides/administration & dosage , Viral Fusion Proteins/administration & dosage , Virus Internalization/drug effects , Administration, Intranasal , Animals , Chemoprevention/methods , Disease Models, Animal , Female , Male , Mice, Inbred C57BL , Mice, Transgenic , Sigmodontinae
4.
Vet Pathol ; 47(4): 677-89, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20442421

ABSTRACT

Human T-lymphotropic virus type-1 (HTLV-1), the first human retrovirus discovered, is the causative agent of adult T-cell leukemia/lymphoma (ATL) and a number of lymphocyte-mediated inflammatory conditions including HTLV-1-associated myelopathy/tropical spastic paraparesis. Development of animal models to study the pathogenesis of HTLV-1-associated diseases has been problematic. Mechanisms of early infection and cell-to-cell transmission can be studied in rabbits and nonhuman primates, but lesion development and reagents are limited in these species. The mouse provides a cost-effective, highly reproducible model in which to study factors related to lymphoma development and the preclinical efficacy of potential therapies against ATL. The ability to manipulate transgenic mice has provided important insight into viral genes responsible for lymphocyte transformation. Expansion of various strains of immunodeficient mice has accelerated the testing of drugs and targeted therapy against ATL. This review compares various mouse models to illustrate recent advances in the understanding of HTLV-1-associated ATL development and how improvements in these models are critical to the future development of targeted therapies against this aggressive T-cell lymphoma.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Human T-lymphotropic virus 1/immunology , Immunity, Innate/immunology , Leukemia-Lymphoma, Adult T-Cell/immunology , Paraparesis, Tropical Spastic/immunology , Animals , Disease Models, Animal , Human T-lymphotropic virus 1/genetics , Humans , Leukemia-Lymphoma, Adult T-Cell/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Paraparesis, Tropical Spastic/genetics
5.
Vet Pathol ; 46(5): 1003-14, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19429977

ABSTRACT

Infection with human T-cell leukemia virus type 1 (HTLV-1) leads sometimes to the development of adult T-cell lymphoma/leukemia (ATL), which is invariably fatal and often associated with humoral hypercalcemia of malignancy. The transformation of infected CD4 T cells and the pathogenesis of leukemia have been studied with great limitation in tissue culture and patients. To better understand the pathogenesis and perform preclinical drug studies, animal models of ATL are urgently needed. In mice, inoculation of HTLV-1 cell lines mostly leads to development of localized lymphomas. To develop an ATL animal model with leukemic spread of ATL cells, mouse strains with different well-defined immune deficiencies were inoculated intraperitoneally with different HTLV-1-infected cell lines (ACH.2, C8166, MT-2, MET-1). Inoculation of MET-1 cells into NOD/SCID mice provided the best model system for slowly developing T-cell leukemia with multiple organ involvement. In leukemic mice, an increase in serum calcium levels correlated with expression of receptor activator of nuclear factor kappa-light-chain-enhancer of activated B cells ligand on leukemic cells and secretion of parathyroid hormone-related protein and interleukin-6. In contrast to the other cell lines that did not spread systemically, MET-1 expressed both the adhesion molecules CD11a (LFA-1alpha) and CD49d (VLA-4alpha) and produced or induced expression of matrix metalloproteinases 1, 2, 3, and 9, thus underlining the importance of these molecules in the spread of adult T-cell leukemia cells. The MET-1/NOD/SCID model will be useful for developing interventions against invasion and spread of leukemic cells and subsequent humoral hypercalcemia of malignancy.


Subject(s)
Disease Models, Animal , Hypercalcemia/etiology , Leukemia-Lymphoma, Adult T-Cell/immunology , Mice, Inbred NOD/immunology , Mice, SCID/immunology , Animals , Basigin/immunology , Blotting, Western/veterinary , CD11a Antigen/immunology , Cell Line , Female , Humans , Hypercalcemia/immunology , Immunohistochemistry/veterinary , Integrin alpha4/immunology , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/immunology , Mice , Mice, Knockout , Parathyroid Hormone-Related Protein/genetics , Parathyroid Hormone-Related Protein/immunology , RANK Ligand/immunology , RNA/chemistry , RNA/genetics , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology , Reverse Transcriptase Polymerase Chain Reaction/veterinary
6.
Curr Top Microbiol Immunol ; 330: 89-110, 2009.
Article in English | MEDLINE | ID: mdl-19203106

ABSTRACT

The cotton rat (Sigmodon hispidus) model has proven to be a suitable small animal model for measles virus pathogenesis to fill the niche between tissue culture and studies in macaques. Similar to mice, inbred cotton rats are available in a microbiologically defined quality with an ever-increasing arsenal of reagents and methods available for the study of infectious diseases. Cotton rats replicate measles virus in the respiratory tract and (depending on virus strain) in lymphoid organs. They can be infected with vaccine, wild-type, and recombinant measles viruses and have been used to study viruses with genetic modifications. Other areas of study include efficacy testing of antivirals and vaccines. The cotton rat also has been an informative animal model to investigate measles virus-induced immune suppression and suppression of vaccination by maternal antibodies. In addition, the cotton rat promises to be a useful model for the study of polymicrobial disease (interaction between measles virus and secondary pathogens).


Subject(s)
Disease Models, Animal , Measles virus/pathogenicity , Measles/virology , Animals , Antibodies, Viral/immunology , Antiviral Agents/pharmacology , Humans , Measles/drug therapy , Measles/immunology , Measles Vaccine/administration & dosage , Measles Vaccine/immunology , Measles virus/drug effects , Measles virus/immunology , Measles virus/physiology , Mice , Rats , Sigmodontinae , T-Lymphocytes/immunology , T-Lymphocytes/virology , Virus Replication
7.
J Gen Virol ; 88(Pt 11): 3112-3120, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17947537

ABSTRACT

Rodent brain-adapted measles virus (MV) strains, such as CAM/RB and recombinant MVs based on the Edmonston strain containing the haemagglutinin (H) of CAM/RB, cause acute encephalitis after intracerebral infection of newborn rodents. We have demonstrated that rodent neurovirulence is modulated by two mutations at amino acid positions 195 and 200 in the H protein, one of these positions (200) being a potential glycosylation site. In order to analyse the effects of specific amino acids at these positions, we introduced a range of individual and combined mutations into the open reading frame of the H gene to generate a number of eukaryotic expression plasmids. The functionality of the mutant H proteins was assessed in transfected cells and by generating recombinant viruses. Interestingly, viruses caused acute encephalitis only if the amino acid Ser at position 200 was coupled with Gly at position 195, whereas viruses with single or combined mutations at these positions, including glycosylation at position 200, were attenuated. Neurovirulence was associated with virus spread and induction of neuronal apoptosis, whereas attenuated viruses failed to infect brain cells. Similar results were obtained by using primary brain-cell cultures. Our findings indicate that a structural alteration in the stem 2 region of the H protein at position 195 or 200 interferes with infectivity of rodent neurons, and suggest that the interaction of the viral attachment protein with cellular receptors on neurons is affected.


Subject(s)
Central Nervous System/virology , Encephalitis/virology , Measles virus/genetics , Measles virus/pathogenicity , Viral Proteins/genetics , Viral Proteins/physiology , Virulence/genetics , Amino Acid Substitution/genetics , Animals , Apoptosis , Cell Line , Cells, Cultured , Measles virus/growth & development , Models, Molecular , Mutagenesis, Site-Directed , Neurons/virology , Rats , Rodentia
8.
Clin Exp Immunol ; 148(2): 218-29, 2007 May.
Article in English | MEDLINE | ID: mdl-17335559

ABSTRACT

Epidemiological studies show an association between early exposure to respiratory syncytial virus (RSV) and the development or exacerbation of asthma. This idea is supported by studies in mice that demonstrate worsened airway hyper-reactivity (AHR) when RSV-infected animals are exposed to allergen. The effect of allergen on RSV disease, however, has not been reported. Cotton rats (Sigmodon hispidus) that have been used as a model to study RSV pathogenesis were sensitized to extracts of Aspergillus fumigatus (Af), a common household mould. The allergic response to Af included eosinophilia, formation of granulomas and induction of Th2 type cytokines. RSV infection prior to allergen challenge resulted in exacerbation of the inflammatory response as well as increased airway responsiveness to methacholine. The exacerbated response was indeed dependent on virus replication. Virus replication in turn was influenced by the allergic response, with persistence in the noses for 2 days longer in animals challenged with allergen. This diminished clearance corresponded to decreased induction of mRNA for IFN-gamma, a Th1-type cytokine that is characteristic of viral infection. Treatment of RSV-infected Af-challenged animals with recombinant IFN-gamma reduced the allergic inflammatory response as well as the relative levels of Th1 and Th2 cytokine mRNA. However, this treatment did not reduce airway reactivity, showing that these pathologic and physiologic measures of exacerbated disease are independent. We speculate that the reciprocal effect of the allergic response on viral immunity may benefit the host by limiting exacerbation of physiologic responses that are IFN-gamma-dependent.


Subject(s)
Allergens/immunology , Respiratory Hypersensitivity/virology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/physiology , Respiratory Tract Infections/virology , Animals , Antiviral Agents/therapeutic use , Aspergillus fumigatus/immunology , Cytokines/biosynthesis , Cytokines/genetics , Female , Granuloma/immunology , Granuloma/pathology , Granuloma/virology , Interferon-gamma/therapeutic use , Male , RNA, Messenger/genetics , Recombinant Proteins , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/pathology , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/immunology , Respiratory Tract Infections/pathology , Sigmodontinae , Th2 Cells/immunology , Virus Replication/immunology
9.
J Gen Virol ; 87(Pt 7): 2011-2019, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16760404

ABSTRACT

Measles virus (MV) nucleocapsids are present abundantly in brain cells of patients with subacute sclerosing panencephalitis (SSPE). This invariably lethal brain disease develops years after acute measles as result of a persistent MV infection. Various rodent models for MV infection of the central nervous system (CNS) have been described in the past, in which the detection of viral antigens is based on histological staining procedures of paraffin embedded brains. Here, the usage of a recombinant MV (MV-EGFP-CAMH) expressing the haemagglutinin (H) of the rodent-adapted MV-strain CAM/RB and the enhanced green fluorescent protein (EGFP) is described. In newborn rodents the virus infects neurons and causes an acute lethal encephalitis. From 2 weeks on, when the immune system of the genetically unmodified animal is maturating, intracerebral (i.c.) infection is overcome subclinically, however, a focal persistent infection in groups of neurons remains. The complete brain can be analysed in 50 or 100 microm slices, and infected autofluorescent cells are readily detected. Seven and 28 days post-infection (p.i.) 86 and 81% of mice are infected, respectively, and virus persists for more than 50 days p.i. Intraperitoneal immunization with MV 1 week before infection, but not after infection, protects and prevents persistence. The high percentage of persistence demonstrates that this is a reliable and useful model of a persistent CNS infection in fully immunocompetent mice, which allows the investigation of determinants of the immune system.


Subject(s)
Measles virus/genetics , Measles virus/pathogenicity , Measles/etiology , Subacute Sclerosing Panencephalitis/etiology , Age Factors , Animals , Animals, Newborn , Brain/immunology , Brain/pathology , Brain/virology , Disease Models, Animal , Green Fluorescent Proteins/genetics , Hemagglutinins, Viral/genetics , Humans , Immunization , Immunocompetence , Measles/immunology , Measles/pathology , Measles/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neurons/virology , Recombinant Proteins/genetics , Recombination, Genetic , Subacute Sclerosing Panencephalitis/immunology , Subacute Sclerosing Panencephalitis/pathology , Subacute Sclerosing Panencephalitis/virology , T-Lymphocytes/pathology
10.
J Virol ; 75(16): 7612-20, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11462033

ABSTRACT

Measles virus (MV) strain CAM/RB, which was adapted to growth in the brain of newborn rodents, is highly neurovirulent. It has been reported earlier that experimentally selected virus variants escaping from the monoclonal antibodies (MAbs) Nc32 and L77 to hemagglutinin (H) preserved their neurovirulence, whereas mutants escaping MAbs K71 and K29 were found to be strongly attenuated (U. G. Liebert et al., J. Virol. 68:1486-1493, 1994). To investigate the molecular basis of these findings, we have generated a panel of recombinant MVs expressing the H protein from CAM/RB and introduced the amino acid substitutions thought to be responsible for antibody escape and/or neurovirulence. Using these recombinant viruses, we identified the amino acid changes conferring escape from the MAbs L77 (377R-->Q and 378M-->K), Nc32 (388G-->S), K71 (492E-->K and 550S-->P), and K29 (535E-->G). When the corresponding recombinant viruses were tested in brains of newborn rodents, we found that the mutations mediating antibody escape did not confer differential neurovirulence. In contrast, however, replacement of two different amino acids, at positions 195G-->R and 200S-->N, which had been described for the escape mutant set, caused the change in neurovirulence. Thus, antibody escape and neurovirulence appear not to be associated with the same structural alterations of the MV H protein.


Subject(s)
Brain/virology , Hemagglutinins/genetics , Measles virus/genetics , Measles/virology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Antigens, Viral , Cell Line , Measles/immunology , Measles virus/immunology , Measles virus/pathogenicity , Mutation , Rats , Recombination, Genetic , Virulence/genetics
11.
Nat Med ; 7(6): 725-31, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11385511

ABSTRACT

Surface-contact-mediated signaling induced by the measles virus (MV) fusion and hemagglutinin glycoproteins is necessary and sufficient to induce T-cell unresponsiveness in vitro and in vivo. To define the intracellular pathways involved, we analyzed interleukin (IL)-2R signaling in primary human T cells and in Kit-225 cells. Unlike IL-2-dependent activation of JAK/STAT pathways, activation of Akt kinase was impaired after MV contact both in vitro and in vivo. MV interference with Akt activation was important for immunosuppression, as expression of a catalytically active Akt prevented negative signaling by the MV glycoproteins. Thus, we show here that MV exploits a novel strategy to interfere with T-cell activation during immunosuppression.


Subject(s)
Immune Tolerance , Measles virus/immunology , Measles/immunology , Milk Proteins , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Receptors, Interleukin-2/metabolism , Signal Transduction/physiology , T-Lymphocytes/immunology , Androstadienes/pharmacology , Animals , Apoptosis/drug effects , Carrier Proteins/metabolism , Cell Line , Chromones/pharmacology , DNA-Binding Proteins/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Hemagglutinins, Viral/metabolism , Humans , Interleukin-2/metabolism , Janus Kinase 1 , Janus Kinase 3 , Lymphocyte Activation , Measles/virology , Measles virus/metabolism , Measles virus/radiation effects , Mice , Mice, Transgenic , Morpholines/pharmacology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-akt , STAT3 Transcription Factor , STAT5 Transcription Factor , Sigmodontinae , Spleen/cytology , T-Lymphocytes/enzymology , T-Lymphocytes/metabolism , Trans-Activators/metabolism , Viral Fusion Proteins/metabolism , Wortmannin , bcl-Associated Death Protein
12.
Vaccine ; 19(20-22): 2764-8, 2001 Apr 06.
Article in English | MEDLINE | ID: mdl-11282186

ABSTRACT

Modified vaccinia virus Ankara (MVA) has been used as an experimental vaccine vector against respiratory infections. We have tested the safety and immunogenicity of a recombinant virus expressing the hemagglutinin of measles virus (MVA-MV-H) using the mouse model of measles virus induced encephalitis and the cotton rat model for respiratory infection. MVA-MV-H proved to induce a TH1 response, neutralizing antibodies and conferred protection against both encephalitis and lung infection. The cotton rat is very sensitive to infection with replication competent vaccinia virus. In these animals MVA-MV-H proved to be a very well tolerated vaccine. However, the efficiency in the presence of MV specific maternal antibodies was low (even using a prime-boost strategy) and therefore might have to be improved.


Subject(s)
Hemagglutinins, Viral/immunology , Measles Vaccine/immunology , Measles/prevention & control , Vaccines, Synthetic/immunology , Vaccinia virus/immunology , Animals , Antibodies, Viral/blood , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Sigmodontinae , Vaccination , Vaccinia virus/genetics
13.
Neurology ; 56(7): 938-43, 2001 Apr 10.
Article in English | MEDLINE | ID: mdl-11294933

ABSTRACT

BACKGROUND: Natural infections bear the risk of triggering MS bouts, whereas epidemiologic studies have not delineated an increased risk for disease activity after influenza virus vaccination. OBJECTIVE: To examine influenza A virus-specific and myelin protein-reactive T-cell frequencies by interferon gamma (IFNgamma)-enzyme-linked immunospot and the response of these cells by IFNgamma-reverse transcription (RT) PCR after immunization and any incidental upper respiratory tract infection (URI) in 12 patients with MS (seven with a relapsing-remitting course; five with a secondary progressive course; Kurtzke Expanded Disability Status Scale [EDSS] score from 1.0 to 6.5, without immunosuppressive treatment) and 28 healthy volunteers. RESULTS: A cellular immune response against influenza A virus was mounted in both populations at 2 weeks after vaccination. Patients with MS showed a higher relative increase (p = 0.008) than controls with respect to the number of influenza-specific T cells. Mean antibody responses against influenza A virus were increased in both populations after 2 weeks (p < 0.01). Despite these virus-specific reactions, no increase in T-cell frequencies responsive to human myelin basic protein (MBP) or recombinant human myelin oligodendrocyte protein (MOG) was observed after immunization, arguing against a general immune stimulation by influenza vaccination. In contrast, MBP-specific T-cell responses became detectable in several individuals after febrile infection. CONCLUSION: These data support the clinical observations that influenza vaccination is effective and safe in patients with MS with respect to cellular immunoreactivity against two main CNS myelin proteins.


Subject(s)
Brain/immunology , Influenza Vaccines/immunology , Multiple Sclerosis/immunology , T-Lymphocytes/immunology , Vaccination , Adult , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged
14.
Vaccine ; 19(17-19): 2250-3, 2001 Mar 21.
Article in English | MEDLINE | ID: mdl-11257342

ABSTRACT

The inhibition of vaccine-induced seroconversion after vaccination is one of the problems associated with measles virus (MV) immunization. In cotton rats, after transfer of human MV specific antibodies, vaccine-induced seroconversion is inhibited. With this model, it was shown that plasmid immunization (although successful in seronegative animals) was inhibited by maternal antibodies. In contrast, immunization via a mucosal surface with a vesicular stomatitis virus expressing the MV hemagglutinin induced seroconversion in the presence of maternal antibodies and subsequent protection.


Subject(s)
Antibodies, Viral/blood , Measles Vaccine/immunology , Measles virus/immunology , Animals , Antibody Specificity , Disease Models, Animal , Female , Genetic Vectors , Humans , Immunity, Maternally-Acquired , Measles/immunology , Measles/prevention & control , Measles Vaccine/genetics , Measles Vaccine/pharmacology , Measles virus/genetics , Pregnancy , Rats , Sigmodontinae , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, DNA/pharmacology , Vesicular stomatitis Indiana virus/genetics
15.
J Virol ; 75(7): 3059-65, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11238832

ABSTRACT

BALB/c mice are resistant to measles virus (MV)-induced encephalitis due to their strong MV-specific CD4(+) T-cell response. Resistance is broken by neutralization of gamma interferon with monoclonal antibodies, indicating an important role for this pleiotropic cytokine. Here, we demonstrate that mouse gamma interferon has no direct antiviral effect in vitro and in vivo. The breakdown of resistance is due neither to a switch in the T-helper response nor to an impaired migration of CD4(+) T cells. Neutralization of gamma interferon interferes with the major histocompatibility complex class II-dependent antigen presentation and subsequent proliferation of CD4(+) T cells in vitro and in vivo. In consequence, the reduction in numbers of CD4(+) T cells below a protective threshold leads to susceptibility to MV-induced encephalitis.


Subject(s)
Antigen Presentation , Encephalitis, Viral/immunology , Histocompatibility Antigens Class II/physiology , Interferon-gamma/physiology , Measles/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Th1 Cells/immunology , Th2 Cells/immunology
16.
Curr Mol Med ; 1(2): 163-81, 2001 May.
Article in English | MEDLINE | ID: mdl-11899069

ABSTRACT

A profound, transient suppression of immune functions during and after the acute infection is the major cause of more than one million cases of infant deaths associated with measles worldwide. Concommittant with the generation of an efficient measles virus (MV) specific immunity, immune responses towards other pathogens are strongly impaired and provide the basis for the establishment and severe course of opportunistic infections. The molecular basis for MV-induced immunosuppression has not been resolved as yet. Similar to other immunosuppressive viruses, MV is lymphotropic and viral nucleic acid and proteins are detectable in peripheral blood mononuclear cells (PBMC). It is considered central to MV-induced immunosuppression that PBMC isolated from patients largely fail to proliferate in response to antigen specific and polyclonal stimulation. The low abundancy of MV-infected PBMC suggests that MV-induced immunosuppression is not directly caused by infection-mediated cell loss or fusion, but rather by indirect mechanisms such as deregulation of cytokines or surface contact-mediated signaling which may lead to apoptosis or impair the proliferative response of uninfected PBMC. Evidence for a role of any of these mechanisms was obtained in vitro, however, much has still to be learned about the tropism of MV and its interactions with particular host cells such as dendritic cells in vivo.


Subject(s)
Immune Tolerance , Measles virus/immunology , Measles/immunology , Animals , Antigen-Presenting Cells/immunology , Antigens, CD/metabolism , Cell Death/physiology , Humans , Lymphocytes/physiology , Measles/physiopathology , Measles virus/physiology , Membrane Cofactor Protein , Membrane Glycoproteins/metabolism , Models, Immunological , Signal Transduction/physiology
17.
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
18.
Proc Natl Acad Sci U S A ; 97(8): 4251-5, 2000 Apr 11.
Article in English | MEDLINE | ID: mdl-10759546

ABSTRACT

During and after measles virus (MV) infection humans are highly susceptible to opportunistic infections because of a marked immunosuppressive effect of the virus. The mechanisms by which the virus induces this phenomenon is not well understood. In particular, detailed information is missing on the targets of suppression in relation to antigen-specific T and B cell responses. Because such studies require animal experiments, we used the cotton rat model, in which the MV causes a respiratory tract infection. Primary as well as secondary T cell responses were impaired in vivo and ex vivo by MV infection. The proliferation of T cells was greatly reduced, but their effector functions, such as cytolysis or cytokine secretion, were not. In contrast, primary and secondary B cell responses in vivo as measured by the frequency of antigen-specific plasma cells in an enzyme-linked immunospot (ELISPOT) assay were not altered by MV infection. Only the secretion of immunoglobulins was reduced slightly in animals primarily infected with MV after 2 weeks. These data demonstrate that MV-induced immunosuppression acts primarily on the T cell responses in vivo.


Subject(s)
Measles/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Division/physiology , Enzyme-Linked Immunosorbent Assay , Female , Hemocyanins/immunology , Immunoglobulins/immunology , Male , Measles virus/physiology , Rats , Sigmodontinae , Skin/immunology , Vaccinia virus/immunology
19.
J Gen Virol ; 81(Pt 5): 1313-9, 2000 May.
Article in English | MEDLINE | ID: mdl-10769074

ABSTRACT

Measles virus (MV), a single-stranded negative-sense RNA virus, is an important pathogen causing almost 1 million deaths annually. Acute MV infection induces immunity against disease throughout life. The immunological factors which are responsible for protection against measles are still poorly understood. However, T-cell-mediated immune responses seem to play a central role. The emergence of new single-cell methods for quantification of antigen-specific T-cells directly ex vivo has prompted us to measure frequencies of MV-specific memory T-cells. As an indicator for T-cell activation IFN-gamma production was measured. PBMC were analysed by intracellular staining and ELISPOT assay after stimulation with MV-infected autologous B-lymphoblastoid cell lines or dendritic cells. T-cell responses were exclusively seen with PBMC from MV-seropositive healthy adults with a history of natural measles in childhood. The median frequency of MV-specific T-cells was 0.35% for CD3(+)CD4(+) and 0.24% for the CD3(+)CD8(+) T-cell subset. These frequencies are comparable with T-cell numbers reported by other investigators for persistent virus infections such as Epstein-Barr virus, cytomegalovirus or human immunodeficiency virus. Hence, this study illustrates that MV-specific CD4(+) and CD8(+) T-cells are readily detectable long after the acute infection, and thus are probably contributing to long-term immunity. Furthermore, this new approach allows efficient analysis of T-cell responses from small samples of blood and could therefore be a useful tool to further elucidate the role of cell-mediated immunity in measles as well as in other viral infections.


Subject(s)
Immunologic Memory , Measles virus/immunology , Measles/immunology , T-Lymphocyte Subsets/immunology , Adult , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/virology , Cell Line , Enzyme-Linked Immunosorbent Assay/methods , Humans , Interferon-gamma/metabolism , Lymphocyte Activation , Major Histocompatibility Complex
20.
J Gen Virol ; 81(Pt 5): 1321-5, 2000 May.
Article in English | MEDLINE | ID: mdl-10769075

ABSTRACT

Plasmids that expressed the nucleocapsid, haemagglutinin and fusion proteins of measles virus (MV) were used to immunize cotton rats (Sigmodon hispidus) against intranasal MV infection. After immunization with all three plasmids, T cell responses and MV-specific antibodies were induced. A reduction in virus titre was observed in lung tissue from animals immunized with plasmids expressing the viral glycoproteins. Histologically, however, a moderate peribronchitis was observed after immunization with the plasmid expressing the fusion protein whereas, after immunization with plasmids expressing haemagglutinin or both glycoproteins, only mild or focal peribronchitis was seen. Immunization with the nucleocapsid did not reduce virus titres, probably because of the failure to induce neutralizing antibodies. A disadvantage of plasmid immunization was its inefficacy in the presence of MV-specific 'maternal' antibodies. This indicates that genetic immunization has to be improved to be a useful alternative vaccine against measles.


Subject(s)
Measles virus/immunology , Measles/prevention & control , Vaccines, DNA , Animals , Antibodies, Viral/blood , Enzyme-Linked Immunosorbent Assay , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/immunology , Immunization , Measles virus/genetics , Neutralization Tests , Nucleocapsid/genetics , Nucleocapsid/immunology , Plasmids/genetics , Sigmodontinae , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...