Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Viral Immunol ; 26(1): 75-83, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23409931

ABSTRACT

Chikungunya virus (CHIKV) is an important emerging mosquito-borne alphavirus, indigenous to tropical Africa and Asia. It can cause epidemic fever and acute illness characterized by fever and arthralgias. The epidemic cycle of this infection is similar to dengue and urban yellow fever viral infections. The generation of an efficient vaccine against CHIKV is necessary to prevent and/or control the disease manifestations of the infection. In this report, we studied immune response against a CHIKV-envelope DNA vaccine (pEnv) and the role of the CHIKV nonstructural gene 2 (nsP2) as an adjuvant for the induction of protective immune responses in a relevant mouse challenge model. When injected with the CHIKV pEnv alone, 70% of the immunized mice survived CHIKV challenge, whereas when co-injected with pEnv+pnsP2, 90% of the mice survived viral challenge. Mice also exhibited a delayed onset signs of illness, and a marked decrease in morbidity, suggesting a nsP2 mediated adjuvant effect. Co-injection of the pnsP2 adjuvant with pEnv also qualitatively and quantitatively increased antigen specific neutralizing antibody responses compared to vaccination with pEnv alone. In sum, these novel data imply that the addition of nsP2 to the pEnv vaccine enhances anti-CHIKV-Env immune responses and maybe useful to include in future CHIKV clinical vaccination strategies.


Subject(s)
Adjuvants, Immunologic/metabolism , Alphavirus Infections/prevention & control , Chikungunya virus/immunology , Vaccines, DNA/immunology , Viral Envelope Proteins/immunology , Viral Nonstructural Proteins/metabolism , Adjuvants, Immunologic/genetics , Alphavirus Infections/immunology , Alphavirus Infections/pathology , Animals , Chikungunya Fever , Chikungunya virus/genetics , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Severity of Illness Index , Survival Analysis , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Viral Envelope Proteins/genetics , Viral Nonstructural Proteins/genetics
2.
Viral Immunol ; 24(4): 265-71, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21830898

ABSTRACT

Chikungunya virus (CHIKV) has caused large outbreaks worldwide in recent years. Acute-phase CHIKV infection has been reported to cause mild to severe febrile illness, and in some patients, this may be followed by long-lasting polyarthritis. The mainstay of treatment includes nonsteroidal anti-inflammatory drugs and other disease-modifying agents, the use of which is based on the assumption of an immunological interference mechanism in the pathogenesis. The present study has been designed to generate preliminary evidence to test this hypothesis. The levels of 30 cytokines were estimated in serum samples of acute CHIKV-infected patients, fully-recovered patients, patients with chronic CHIKV arthritis, and controls, using a quantitative multiplex bead ELISA. The levels of the proinflammatory cytokines IL-1 and IL-6 were elevated in acute patients, but IFN-γ/ß and TNF-α levels remained stable. IL-10, which might have an anti-inflammatory effect, was also elevated, indicating a predominantly anti-inflammatory response in the acute phase of infection. Elevation of MCP-1, IL-6, IL-8, MIP-1α, and MIP-1ß was most prominent in the chronic phase. These cytokines and chemokines have been shown to play important roles in other arthritides, including epidemic polyarthritis (EPA) caused by Ross River virus (RRV) and rheumatoid arthritis (RA).The immunopathogenesis of chronic CHIKV arthritis might have similarities to these arthritides. The novel intervention strategies being developed for EPA and RA, such as IL-6 and IL-8 signaling blockade, may also be considered for chronic CHIKV arthritis.


Subject(s)
Alphavirus Infections/immunology , Alphavirus Infections/pathology , Arthritis, Infectious/immunology , Arthritis, Infectious/pathology , Chikungunya virus/pathogenicity , Cytokines/immunology , Adult , Alphavirus Infections/complications , Chronic Disease , Cytokines/blood , Enzyme-Linked Immunosorbent Assay , Humans , Middle Aged
3.
PLoS Negl Trop Dis ; 5(1): e928, 2011 Jan 11.
Article in English | MEDLINE | ID: mdl-21264351

ABSTRACT

Chikungunya virus (CHIKV) is an emerging mosquito-borne alphavirus indigenous to tropical Africa and Asia. Acute illness is characterized by fever, arthralgias, conjunctivitis, rash, and sometimes arthritis. Relatively little is known about the antigenic targets for immunity, and no licensed vaccines or therapeutics are currently available for the pathogen. While the Aedes aegypti mosquito is its primary vector, recent evidence suggests that other carriers can transmit CHIKV thus raising concerns about its spread outside of natural endemic areas to new countries including the U.S. and Europe. Considering the potential for pandemic spread, understanding the development of immunity is paramount to the development of effective counter measures against CHIKV. In this study, we isolated a new CHIKV virus from an acutely infected human patient and developed a defined viral challenge stock in mice that allowed us to study viral pathogenesis and develop a viral neutralization assay. We then constructed a synthetic DNA vaccine delivered by in vivo electroporation (EP) that expresses a component of the CHIKV envelope glycoprotein and used this model to evaluate its efficacy. Vaccination induced robust antigen-specific cellular and humoral immune responses, which individually were capable of providing protection against CHIKV challenge in mice. Furthermore, vaccine studies in rhesus macaques demonstrated induction of nAb responses, which mimicked those induced in convalescent human patient sera. These data suggest a protective role for nAb against CHIKV disease and support further study of envelope-based CHIKV DNA vaccines.


Subject(s)
Alphavirus Infections/prevention & control , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Chikungunya virus/immunology , Vaccines, DNA/immunology , Alphavirus Infections/virology , Animals , Chikungunya virus/genetics , Chikungunya virus/isolation & purification , Disease Models, Animal , Electroporation , Female , Humans , Macaca mulatta , Mice , Mice, Inbred BALB C , Neutralization Tests/methods , Vaccination/methods , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Virology/methods
4.
Immunology ; 128(1 Suppl): e612-20, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19740322

ABSTRACT

DNA vaccination is a novel immunization strategy that has great potential for the development of vaccines and immune therapeutics. This strategy has been highly effective in mice, but is less immunogenic in non-human primates and in humans. Enhancing DNA vaccine potency remains a challenge. It is likely that antigen-presenting cells (APCs), and especially dendritic cells (DCs), play a significant role in the presentation of the vaccine antigen to the immune system. A new study reports the synergistic recruitment, expansion and activation of DCs in vivo by high-mobility group box 1 (HMGB1) protein. Such combinational strategies for delivering vaccine in a single, simple platform will hypothetically bolster the cellular immunity in vivo. Here, we combined plasmid encoding human immunodeficiency virus-1 (HIV-1) Gag and Env with an HMGB1 plasmid as a DNA adjuvant in BALB/c mice (by intramuscular immunization via electroporation), and humoral and cellular responses were measured. Co-administration of this potent immunostimulatory adjuvant strongly enhanced the cellular interferon-gamma (IFN-gamma) and humoral immune response compared with that obtained in mice immunized with vaccine only. Our results show that co-immunization with HMGB1 can have a strong adjuvant activity, driving strong cellular and humoral immunity that may be an effective immunological adjuvant in DNA vaccination against HIV-1.


Subject(s)
AIDS Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , HIV-1/immunology , HMGB1 Protein/immunology , Vaccines, DNA/immunology , AIDS Vaccines/genetics , Adjuvants, Immunologic/metabolism , Amino Acid Sequence , Animals , Antibodies/blood , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Female , Genetic Vectors/immunology , Genetic Vectors/metabolism , HMGB1 Protein/genetics , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Vaccines, DNA/genetics , env Gene Products, Human Immunodeficiency Virus/immunology , env Gene Products, Human Immunodeficiency Virus/metabolism , gag Gene Products, Human Immunodeficiency Virus/immunology , gag Gene Products, Human Immunodeficiency Virus/metabolism
5.
J Virol ; 82(23): 11536-44, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18799583

ABSTRACT

Chronic viral infection is characterized by the functional impairment of virus-specific T-cell responses. Recent evidence has suggested that the inhibitory receptor programmed death 1 (PD-1) is specifically upregulated on antigen-specific T cells during various chronic viral infections. Indeed, it has been reported that human immunodeficiency virus (HIV)-specific T cells express elevated levels of PD-1 and that this expression correlates with the viral load and inversely with CD4(+) T-cell counts. More importantly, antibody blockade of the PD-1/PD-L1 pathway was sufficient to both increase and stimulate virus-specific T-cell proliferation and cytokine production. However, the mechanisms that mediate HIV-induced PD-1 upregulation are not known. Here, we provide evidence that the HIV type 1 (HIV-1) accessory protein Nef can transcriptionally induce the expression of PD-1 during infection in vitro. Nef-induced PD-1 upregulation requires its proline-rich motif and the activation of the downstream kinase p38. Further, inhibition of Nef activity by p38 MAPK inhibitor effectively blocked PD-1 upregulation, suggesting that p38 MAPK activation is an important initiating event in Nef-mediated PD-1 expression in HIV-1-infected cells. These data demonstrate an important signaling event of Nef in HIV-1 pathogenesis.


Subject(s)
Antigens, CD/genetics , Apoptosis Regulatory Proteins/genetics , nef Gene Products, Human Immunodeficiency Virus/physiology , p38 Mitogen-Activated Protein Kinases/physiology , Acquired Immunodeficiency Syndrome/metabolism , Antigens, CD/biosynthesis , Apoptosis Regulatory Proteins/biosynthesis , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Gene Expression Regulation , Humans , Programmed Cell Death 1 Receptor , Up-Regulation
6.
Vaccine ; 26(40): 5128-34, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18471943

ABSTRACT

Chikungunya virus (CHIKV) is an emerging arbovirus and is an important human pathogen. Infection of humans by CHIKV can cause a syndrome characterized by fever, headache, rash, nausea, vomiting, myalgia, arthralgia and occasionally neurological manifestations such as acute limb weakness. It is also associated with a fatal haemorrhagic condition. CHIKV is geographically distributed from Africa through Southeast Asia and South America, and its transmission to humans is mainly through the Aedes aegypti species mosquitoes. The frequency of recent epidemics in the Indian Ocean and La Reunion islands suggests that a new vector perhaps is carrying the virus, as A. aegypti are not found there. In fact, a relative the Asian tiger mosquito, Aedes albopictus, may be the culprit which has raised concerns in the world health community regarding the potential for a CHIK virus pandemic. Accordingly steps should be taken to develop methods for the control of CHIKV. Unfortunately, currently there is no specific treatment for Chikungunya virus and there is no vaccine currently available. Here we present data of a novel consensus-based approach to vaccine design for CHIKV, employing a DNA vaccine strategy. The vaccine cassette was designed based on CHIKV capsid- and envelope-specific consensus sequences with several modifications, including codon optimization, RNA optimization, the addition of a Kozak sequence, and a substituted immunoglobulin E leader sequence. The expression of capsid, envelope E1 and E1 was evaluated using T7-coupled transcription/translation and immunoblot analysis. A recently developed, adaptive constant-current electroporation technique was used to immunize C57BL/6 mice with an intramuscular injection of plasmid coding for the CHIK-Capsid, E1 and E2. Analysis of cellular immune responses, including epitope mapping, demonstrates that electroporation of these constructs induces both potent and broad cellular immunity. In addition, antibody ELISAs demonstrate that these synthetic immunogens are capable of inducing high titer antibodies capable of recognizing native antigen. Taken together, these data support further study of the use of consensus CHIK antigens in a potential vaccine cocktail.


Subject(s)
Alphavirus Infections/immunology , Capsid Proteins/immunology , Chikungunya virus/immunology , Genes, env/immunology , Vaccines, DNA/immunology , Viral Vaccines/immunology , Alphavirus Infections/prevention & control , Alphavirus Infections/virology , Animals , Antibodies, Viral/blood , Capsid/immunology , Capsid Proteins/genetics , Chikungunya virus/genetics , Cricetinae , Electroporation , Female , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology , Vaccination , Vaccines, DNA/administration & dosage , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...