Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters

Publication year range
1.
J Infect Dis ; 214(3): 369-78, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27001960

ABSTRACT

BACKGROUND: Vaccination and passive antibody therapies are critical for controlling infectious diseases. Passive antibody administration has limitations, including the necessity for purification and multiple injections for efficacy. Vaccination is associated with a lag phase before generation of immunity. Novel approaches reported here utilize the benefits of both methods for the rapid generation of effective immunity. METHODS: A novel antibody-based prophylaxis/therapy entailing the electroporation-mediated delivery of synthetic DNA plasmids encoding biologically active anti-chikungunya virus (CHIKV) envelope monoclonal antibody (dMAb) was designed and evaluated for antiviral efficacy, as well as for the ability to overcome shortcomings inherent with conventional active vaccination and passive immunotherapy. RESULTS: One intramuscular injection of dMAb produced antibodies in vivo more rapidly than active vaccination with an anti-CHIKV DNA vaccine. This dMAb neutralized diverse CHIKV clinical isolates and protected mice from viral challenge. Combination of dMAb and the CHIKV DNA vaccine afforded rapid and long-lived protection. CONCLUSIONS: A DNA-based dMAb strategy induced rapid protection against an emerging viral infection. This method can be combined with DNA vaccination as a novel strategy to provide both short- and long-term protection against this emerging infectious disease. These studies have implications for pathogen treatment and control strategies.


Subject(s)
Antibodies, Viral/immunology , Chemoprevention/methods , Chikungunya Fever/prevention & control , Vaccines, DNA/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/administration & dosage , Disease Models, Animal , Electroporation , Injections, Intramuscular , Mice, Inbred BALB C , Time Factors , Treatment Outcome , Vaccines, DNA/administration & dosage , Viral Vaccines/administration & dosage
2.
Immunology ; 142(4): 594-602, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24527796

ABSTRACT

VGX-1027 [(S,R)-3-phenyl-4,5-dihydro-5-isoxasole acetic acid] is a small molecule compound with immunomodulatory properties, which favourably influences the development of immuno-inflammatory and autoimmune diseases in different animal models such as type 1 diabetes mellitus, pleurisy, rheumatoid arthritis and inflammatory bowel disease. However, the precise mechanism of action of VGX-1027 remains to be ascertained. With this aim, we have studied the immunomodulatory effects of VGX-1027 in vitro, using a genome-wide oligonucleotide microarray approach, and in vivo, using the NZB/NZW F1 model of systemic lupus erythematosus. Microarray data revealed that the administration of VGX-1027 profoundly affected the immune response to exogenous antigens, by modulating the expression of genes that are primarily involved in antigen processing and presentation as well as genes that regulate immune activation. When administered in vivo VGX-1027 ameliorated the course of the disease in the NZB/NZW F1 mice, which correlated with higher per cent survival and improved clinical and histopathological signs. The data presented herein support the theory that VGX-1027 modulates immunity, probably by inhibiting inflammatory antigen presentation and so limiting immune cell expansion.


Subject(s)
Acetates/pharmacology , Immunologic Factors/pharmacology , Lipopolysaccharides/toxicity , Lupus Erythematosus, Systemic/drug therapy , Oxazoles/pharmacology , Toll-Like Receptor 4/immunology , Animals , Disease Models, Animal , Female , Humans , Lupus Erythematosus, Systemic/chemically induced , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Male , Mice
3.
J Vasc Surg ; 59(3): 786-93, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23850058

ABSTRACT

OBJECTIVE: Hypoxia-inducible factor-1 alpha (HIF-1α) is a transcription factor that stimulates angiogenesis during tissue ischemia. In vivo electroporation (EP) enhances tissue DNA transfection. We hypothesized that in vivo EP of plasmid DNA encoding a constitutively expressed HIF-1α gene enhances neovascularization compared with intramuscular (IM) injection alone. METHODS: Left femoral artery ligation was performed in mice assigned to three groups: (1) HIF-EP (n = 13); (2) HIF-IM (n = 14); and (3) empty plasmid (pVAX)-EP (n = 12). A single dose of HIF-1α or pVAX DNA (20 µL of 5 µg/µL each) was injected into the ischemic adductor muscle followed by EP (groups one and three). Mice in group two received IM injection of HIF-1α plasmid DNA alone. From preligation to days 0, 3, 7, 14, and 21 postligation, limb perfusion recovery quantified by laser Doppler perfusion imager, limb function, and limb necrosis were measured. On day 21, the surviving mice (4-5 per group) were sacrificed and adductor muscle tissues stained for necrosis using hematoxylin and eosin, capillary density (anti-CD31 antibodies), and collateral vessels via anti-α-smooth muscle actin antibodies. RESULTS: In vivo EP of HIF-1α DNA significantly improved limb perfusion (HIF-EP: 1.03 ± 0.15 vs HIF-IM: 0.78 ± 0.064; P < .05, vs pVAX-EP: 0.41 ± 0.019; P < .001), limb functional recovery (HIF-EP: 3.5 ± 0.58 vs HIF-IM, 2.4 ± 1.14; P < .05, vs pVAX-EP: 2.4 ± 1.14; P < .001), and limb autoamputation on day 21 (HIF-EP: 77% ± 12% vs HIF-IM: 43% ± 14%; P < .05 vs pVAX-EP: 17% ± 11%; P < .01). Adductor muscle tissue necrosis decreased (HIF-EP: 20.7% ± 1.75% vs HIF-IM: 44% ± 3.73; P < .001, vs pVAX-EP: 60.05% ± 2.17%; P < .0001), capillary density increased (HIF-EP: 96.83 ± 5.72 vessels/high-powered field [hpf] vs HIF-IM: 62.87 ± 2.0 vessels/hpf; P < .001, vs pVAX-EP: 39.37 ± 2.76 vessels/hpf; P < .0001), collateral vessel formation increased (HI-EP: 76.33 ± 1.94 vessels/hpf vs HIF-IM: 37.5 ± 1.56 vessels/hpf; P < .0001, vs pVAX-EP: 18.5 ± 1.34 vessels/hpf; P < .00001), and the vessels were larger (HIF-EP: 15,521.67 ± 1298.16 µm(2) vs HIF-IM: 7788.87 ± 392.04 µm(2); P < .001 vs pVAX-EP: 4640.25 ± 614.01 µm(2); P < .0001). CONCLUSIONS: In vivo EP-mediated delivery of HIF-1α plasmid DNA improves neovascularization in a mouse model of limb ischemia and is a potentially suitable nonviral, noninvasive intervention to facilitate therapeutic angiogenesis in critical limb ischemia.


Subject(s)
Electroporation , Gene Transfer Techniques , Genetic Therapy/methods , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Ischemia/therapy , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Animals , Blood Flow Velocity , Collateral Circulation , Disease Models, Animal , Hindlimb , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Injections, Intramuscular , Ischemia/genetics , Ischemia/metabolism , Ischemia/pathology , Ischemia/physiopathology , Laser-Doppler Flowmetry , Mice , Mice, Inbred BALB C , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Necrosis , Recovery of Function , Regional Blood Flow , Time Factors
4.
Immunology ; 140(2): 202-10, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23710940

ABSTRACT

A sudden upsurge of fever cases with joint pain was observed in the outpatient department, Community Health Centre, Rangat during July-August 2010 in Rangat Middle Andaman, India. The aetiological agent responsible for the outbreak was identified as chikungunya virus (CHIKV), by using RT-PCR and IgM ELISA. The study investigated the association of polymorphisms in the human leucocyte antigen class II genes with susceptibility or protection against CHIKV. One hundred and one patients with clinical features suggestive of CHIKV infection and 104 healthy subjects were included in the study. DNA was extracted and typed for HLA-DRB1 and DQB1 alleles. Based on the amino acid sequences of HLA-DQB1 retrieved from the IMGT/HLA database, critical amino acid differences in the specific peptide-binding pockets of HLA-DQB1 molecules were investigated. The frequencies of HLA-DRB1 alleles were not significantly different, whereas lower frequency of HLA-DQB1*03:03 was observed in CHIKV patients compared with the control population [P = 0·001, corrected P = 0·024; odds ratio (OR)  = 0, 95% confidence interval (95% CI) 0·0-0·331; Peto's OR = 0·1317, 95% CI 0·0428-0·405). Significantly lower frequency of glutamic acid at position 86 of peptide-binding pocket 1 coding HLA-DQB1 genotypes was observed in CHIKV patients compared with healthy controls (P = 0·004, OR = 0·307, 95% CI 0·125-0·707). Computational binding predictions of CD4 epitopes of CHIKV by NetMHCII revealed that HLA-DQ molecules are known to bind more CHIKV peptides than HLA-DRB1 molecules. The results suggest that HLA-DQB1 alleles and critical amino acid differences in the peptide-binding pockets of HLA-DQB1 alleles might have role in influencing infection and pathogenesis of CHIKV.


Subject(s)
Alphavirus Infections/genetics , Genetic Predisposition to Disease/genetics , Histocompatibility Antigens Class II/genetics , Polymorphism, Genetic/genetics , Adult , Alleles , Alphavirus Infections/epidemiology , Binding Sites , Chikungunya Fever , Disease Outbreaks , Enzyme-Linked Immunosorbent Assay , Female , Genotype , Humans , India , Male , Reverse Transcriptase Polymerase Chain Reaction
5.
Nat Cell Biol ; 8(2): 170-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16429131

ABSTRACT

The Vpr protein of HIV-1 functions as a vital accessory gene by regulating various cellular functions, including cell differentiation, apoptosis, nuclear factor of kappaB (NF-kappaB) suppression and cell-cycle arrest of the host cell. Several reports have indicated that Vpr complexes with the glucocorticoid receptor (GR), but it remains unclear whether the GR pathway is required for Vpr to function. Here, we report that Vpr uses the GR pathway as a recruitment vehicle for the NF-kappaB co-activating protein, poly(ADP-ribose) polymerase-1 (PARP-1). The GR interaction with Vpr is both necessary and sufficient to facilitate this interaction by potentiating the formation of a Vpr-GR-PARP-1 complex. The recruitment of PARP-1 by the Vpr-GR complex prevents its nuclear localization, which is necessary for Vpr to suppress NF-kappaB. The association of GR with PARP-1 is not observed with steroid (glucocorticoid) treatment, indicating that the GR association with PARP-1 is a gain of function that is solely attributed to HIV-1 Vpr. These data provide important insights into Vpr biology and its role in HIV pathogenesis.


Subject(s)
Cell Nucleus/metabolism , Gene Products, vpr/physiology , Poly(ADP-ribose) Polymerases/metabolism , Receptors, Glucocorticoid/metabolism , Active Transport, Cell Nucleus , Animals , Antigens, Bacterial/pharmacology , Cell Line , Chlorocebus aethiops , Enterotoxins/pharmacology , Female , Gene Expression/drug effects , Gene Expression/genetics , Gene Products, vpr/metabolism , Gene Products, vpr/pharmacology , HIV Infections/metabolism , HIV Infections/physiopathology , HeLa Cells , Humans , I-kappa B Kinase/metabolism , I-kappa B Proteins/metabolism , Interleukin-1/blood , Interleukin-12/blood , Jurkat Cells , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Mifepristone/pharmacology , Mutation/genetics , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Protein Binding/drug effects , Protein Interaction Mapping , RNA, Small Interfering/genetics , Receptors, Glucocorticoid/genetics , Transcription Factor RelA/metabolism , Transfection , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , U937 Cells , vpr Gene Products, Human Immunodeficiency Virus
6.
J Immunol ; 187(6): 2932-43, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21856939

ABSTRACT

Recent evidence demonstrates that HIV-1 infection leads to the attenuation of cellular immune responses, which has been correlated with the increased expression of programmed death (PD)-1 on virus-specific CD8(+) T cells. PD-1 is induced upon T cell activation, and its prolonged expression facilitates CD8(+) T cell inhibitory signals when bound to its B7 family ligands, PD-ligand (L)1/2, which are expressed on APCs. Importantly, early reports demonstrated that blockade of the PD-1/PD-L interaction by Abs may help to counter the development of immune exhaustion driven by HIV viral persistence. To better understand the regulation of the PD-1 pathway during HIV infection, we examined the ability of the virus to induce PD-L expression on macrophages and dendritic cells. We found a direct relationship between the infection of APCs and the expression of PD-L1 in which virus-mediated upregulation induced a state of nonresponsiveness in uninfected HIV-specific T cells. Furthermore, this exhaustion phenotype was revitalized by the blockade of PD-L1, after which T cells regained their capacity for proliferation and the secretion of proinflammatory cytokines IFN-γ, IL-2, and IL-12 upon restimulation. In addition, we identify a critical role for the PI3K/serine-threonine kinase signaling pathway in PD-L1 upregulation of APCs by HIV, because inhibition of these intracellular signal transducer enzymes significantly reduced PD-L1 induction by infection. These data identify a novel mechanism by which HIV exploits the immunosuppressive PD-1 pathway and suggest a new role for virus-infected cells in the local corruption of immune responses required for viral suppression.


Subject(s)
Antigen-Presenting Cells/immunology , CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , Lymphocyte Activation/immunology , Signal Transduction/immunology , Antigen-Presenting Cells/metabolism , Antigens, CD/biosynthesis , Antigens, CD/immunology , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/immunology , Blotting, Western , CD8-Positive T-Lymphocytes/metabolism , Cell Separation , Enzyme Activation/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , HIV Infections/metabolism , HIV-1/immunology , Humans , Ligands , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Programmed Cell Death 1 Receptor , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
7.
Cytometry A ; 77(3): 275-84, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20104580

ABSTRACT

The capacity for robust proliferation upon re-infection is a hallmark of adaptive immunity and the basis of vaccination. A widely used animal model for the study of human disease is the rhesus macaque (RM), where capacity for proliferation can be assessed ex vivo using carboxyfluorescein succinimidyl ester (CFSE)-based dilution assays. However, we show over the course of the standard ex vivo proliferation assay that CFSE-labeling at commonly used dye concentrations induces significant cell death, but that this phenomenon is dose-dependent. Here, we describe an alternative semiquantitative method for estimating T cell proliferative responses that avoids the putative biases associated with chemical modification. RM peripheral blood mononuclear cells were stimulated ex vivo with cognate peptides for 5 days, immunostained for intracellular Ki-67, and then analyzed by flow cytometry. We describe a gating strategy using Ki-67 and side light scatter, also a marker of blastogenesis, which correlates strongly with data from CFSE dilution. We show that this method is a valid tool for measuring RM antigen-specific cellular proliferation ex vivo and can be used as an alternative to CFSE dilution assays.


Subject(s)
Flow Cytometry/methods , Ki-67 Antigen/biosynthesis , T-Lymphocytes/cytology , Animals , Antigens/chemistry , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Coloring Agents/pharmacology , Dose-Response Relationship, Drug , Fluoresceins/chemistry , Gene Deletion , Leukocytes, Mononuclear/cytology , Macaca mulatta , Succinimides/chemistry , T-Lymphocytes/microbiology
8.
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
9.
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
10.
Mol Ther ; 16(2): 419-26, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18087262

ABSTRACT

Non-homeostatic tissue apoptosis in vivo has been shown to induce inflammatory responses and facilitate the cross-presentation of proteins within apoptotic bodies. We hypothesize that in the presence of foreign antigens, the apoptotic-inflammatory process improves immune priming; further, molecules that trigger apoptosis may be adapted for use as immune adjuvants. One very attractive molecule in this context is the tumor necrosis factor receptor (TNFR) family molecule DR5/TRAIL-receptor 2. We show a significant improvement in CD8(+) T-cell mediated vaccine immunity with the use of death receptor-5 (DR5) as an immune adjuvant, a property that is correlated with the activation of caspases-8 (casp8) and dependent on its ability to induce apoptosis in vivo.


Subject(s)
Caspase 8/metabolism , Dendritic Cells/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Dendritic Cells/metabolism , Female , Flow Cytometry , Immunization/methods , In Situ Nick-End Labeling , Influenza A virus/immunology , Mice , Mice, Inbred C57BL , Mutation , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics
11.
Apoptosis ; 13(9): 1088-99, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18622704

ABSTRACT

The mitochondrion is an organelle that regulates various cellular functions including the production of energy and programmed cell death. Aberrant mitochondrial function is often concomitant with various cytopathies and medical disorders. The mitochondrial membrane plays a key role in the induction of cellular apoptosis, and its destabilization, as triggered by both intracellular and extracellular stimuli, results in the release of proapoptotic factors into the cytosol. Not surprisingly, proteins from the human immunodeficiency virus type 1 (HIV) have been implicated in exploiting this organelle to promote the targeted depletion of key immune cells, which assists in viral evasion of the immune system and contributes to the characteristic global immunodeficiency observed during progression of disease. Here we review the mechanisms by which HIV affects the mitochondrion, and suggest that various viral-associated genes may directly regulate apoptotic cell death.


Subject(s)
Apoptosis , Genes, Viral , HIV-1/genetics , Mitochondria/pathology , Mitochondria/virology , HIV Infections/pathology , HIV Infections/virology , Humans
12.
DNA Cell Biol ; 27(5): 267-77, 2008 May.
Article in English | MEDLINE | ID: mdl-18462066

ABSTRACT

Prior work has implicated viral protein R (Vpr) in the arrest of human immunodeficiency virus type 1 (HIV-1)-infected cells in the G2 phase of the cell cycle, associated with increased viral replication and host cell apoptosis. We and others have recently shown that virion infectivity factor (Vif ) also plays a role in the G2 arrest of HIV-1-infected cells. Here, we demonstrate that, paradoxically, at early time points postinfection, Vif expression blocks Vpr-mediated G2 arrest, while deletion of Vif from the HIV-1 genome leads to a marked increase in G2 arrest of infected CD4 T-cells. Consistent with this increased G2 arrest, T-cells infected with Vif-deleted HIV-1 express higher levels of Vpr protein than cells infected with wild-type virus. Further, expression of exogenous Vif inhibits the expression of Vpr, associated with a decrease in G2 arrest of both infected and transfected cells. Treatment with the proteasome inhibitor MG132 increases Vpr protein expression and G2 arrest in wild-type, but not Vif-deleted, NL4-3-infected cells, and in cells cotransfected with Vif and Vpr. In addition, Vpr coimmunoprecipitates with Vif in cotransfected cells in the presence of MG132. This suggests that inhibition of Vpr by Vif is mediated at least in part by proteasomal degradation, similar to Vif-induced degradation of APOBEC3G. Together, these data show that Vif mediates the degradation of Vpr and modulates Vpr-induced G2 arrest in HIV-1-infected T-cells.


Subject(s)
G2 Phase , Gene Expression Regulation, Viral , HIV Infections/metabolism , HIV-1/metabolism , vif Gene Products, Human Immunodeficiency Virus/metabolism , vpr Gene Products, Human Immunodeficiency Virus/metabolism , Apoptosis/physiology , Blotting, Western , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Proliferation , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , HIV Infections/pathology , HIV Infections/virology , HIV-1/growth & development , Humans , Leupeptins/pharmacology , Transfection , Virus Replication , vif Gene Products, Human Immunodeficiency Virus/genetics , vpr Gene Products, Human Immunodeficiency Virus/genetics
13.
Vaccine ; 35(1): 61-70, 2017 01 03.
Article in English | MEDLINE | ID: mdl-27894716

ABSTRACT

The skin is an ideal target tissue for vaccine delivery for a number of reasons. It is highly accessible, and most importantly, enriched in professional antigen presenting cells. Possessing strong similarities to human skin physiology and displaying a defined epidermis, the guinea pig is an appropriate model to study epidermal delivery of vaccine. However, whilst we have characterized the humoral responses in the guinea pig associated with skin vaccine protocols we have yet to investigate the T cell responses. In response to this inadequacy, we developed an IFN-γ ELISpot assay to characterize the cellular immune response in the peripheral blood of guinea pigs. Using a nucleoprotein (NP) influenza pDNA vaccination regimen, we characterized host T cell responses. After delivery of the DNA vaccine to the guinea pig epidermis we detected robust and rapid T cell responses. The levels of IFN-γ spot-forming units averaged approximately 5000 per million cells after two immunizations. These responses were broad in that multiple regions across the NP antigen elicited a T cell response. Interestingly, we identified a number of NP immunodominant T cell epitopes to be conserved across an outbred guinea pig population, a phenomenon which was also observed after immunization with a RSV DNA vaccine. We believe this data enhances our understanding of the cellular immune response elicited to a vaccine in guinea pigs, and globally, will advance the use of this model for vaccine development, especially those targeting skin as a delivery site.


Subject(s)
Electroporation , Influenza Vaccines/immunology , RNA-Binding Proteins/immunology , Skin/immunology , T-Lymphocytes/immunology , Vaccines, DNA/immunology , Viral Core Proteins/immunology , Animals , Enzyme-Linked Immunospot Assay , Female , Guinea Pigs , Influenza Vaccines/administration & dosage , Interferon-gamma/metabolism , Nucleocapsid Proteins , Vaccines, DNA/administration & dosage
14.
Vaccine ; 35(21): 2840-2847, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28413132

ABSTRACT

Respiratory syncytial virus (RSV) is a massive medical burden in infants, children and the elderly worldwide, and an effective, safe RSV vaccine remains an unmet need. Here we assess a novel vaccination strategy based on the intradermal delivery of a SynCon® DNA-based vaccine encoding engineered RSV-F antigen using a surface electroporation device (SEP) to target epidermal cells, in clinically relevant experimental models. We demonstrate the ability of this strategy to elicit robust immune responses. Importantly we demonstrate complete resistance to pulmonary infection at a single low dose of vaccine in the cotton rat RSV/A challenge model. In contrast to the formalin-inactivated RSV (FI-RSV) vaccine, there was no enhanced lung inflammation upon virus challenge after DNA vaccination. In summary the data presented outline the pre-clinical development of a highly efficacious, tolerable and safe non-replicating vaccine delivery strategy.


Subject(s)
Electroporation/instrumentation , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Animals , Disease Models, Animal , Female , Lung/pathology , Sigmodontinae , Treatment Outcome
15.
DNA Cell Biol ; 25(7): 383-92, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16848679

ABSTRACT

DNA vaccines expressing the envelope (env) of the human immunodeficiency virus type 1 (HIV-1) have been relatively ineffective at generating strong immune responses. In this study, we described the development of a recombinant plasmid DNA (pEK2P-B) expressing an engineered codon-optimized envelope gp140 gene of primary (nonrecombinant) HIV-1 subtype B isolate 6101. Codon usage and RNA optimization of HIV-1 structural genes has been shown to increase protein expression in vitro as well as in the context of DNA vaccines in vivo. To further increase the expression, a synthetic IgE leader with kozak sequences were fused into the env gene. The cytoplasmic tail of the gene was also truncated to prevent recycling. The expression of env by the recombinant pEK2P-B was evaluated using T7 coupled transcription/translation. The construct demonstrated high expression of the HIV-1 env gene in eukaryotic cells as demonstrated in transfected 293-T and RD cells. Immunogenicity of pEK2P-B was evaluated in mice using IFN-gamma ELISpot assay, and the construct was found to be highly immunogenic and crossreactive with HIV-1 clade C env peptides. Three immunodominant peptides were also mapped out. Furthermore, by performing a CFSE flow cytometry-based proliferation assay, 2.4 and 1.5% proliferation was observed in CD4+, CD8+, and CCR+ memory T cells, respectively. Therefore, this engineered synthetic optimized env DNA vaccine may be useful in DNA vaccine and other studies of HIV-1 immunogenicity.


Subject(s)
AIDS Vaccines/genetics , AIDS Vaccines/immunology , Gene Products, env/genetics , Gene Products, env/immunology , HIV-1/genetics , HIV-1/immunology , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cross Reactions , DNA, Viral/genetics , Epitope Mapping , Female , Gene Expression , Genes, env , Genetic Engineering , HIV Antigens/genetics , HIV Antigens/immunology , Humans , Immunodominant Epitopes/genetics , In Vitro Techniques , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Molecular Sequence Data , T-Lymphocyte Subsets/immunology , Transfection , Vaccines, DNA/genetics , Vaccines, DNA/immunology , env Gene Products, Human Immunodeficiency Virus
16.
Clin Pharmacol Drug Dev ; 5(2): 91-101, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27138022

ABSTRACT

VGX-1027, a novel oral immune modulator, is under development for the treatment of rheumatoid arthritis. The safety, tolerability, and pharmacokinetics of single (1-800 mg) and multiple (40-400 mg) oral doses were evaluated in 2 clinical studies. The doses were well tolerated up to 800 mg in a single dose and 200 mg twice daily in multiple doses. Adverse events were mild to moderate in severity with no identifiable dose-related pattern. There were no clinically significant physical or laboratory findings. The pharmacokinetic data indicated that increases in Cmax and AUC0-inf were dose-proportional, and AUC0- τ was approximately dose-proportional. For the single-dose study, median Tmax ranged from 0.5 to 2 hours and mean t1/2 ranged from 4.9 to 8.7 hours. For the multiple-dose study, median Tmax ranged from 0.5 to 2.0 hours and mean t1/2 ranged from 7.05 to 10.05 hours. No accumulation of the drug was observed after day 1, indicating that steady-state concentrations were attained with single and multiple dosing for 5 days. Approximately 90% of the administered dose was excreted in urine as unchanged drug.


Subject(s)
Acetates/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Immunologic Factors/administration & dosage , Oxazoles/administration & dosage , Acetates/adverse effects , Acetates/pharmacokinetics , Administration, Oral , Adult , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/pharmacokinetics , Area Under Curve , Biological Availability , Dose-Response Relationship, Drug , Double-Blind Method , Female , Half-Life , Humans , Immunologic Factors/adverse effects , Immunologic Factors/pharmacokinetics , Male , Oxazoles/adverse effects , Oxazoles/pharmacokinetics
17.
NPJ Vaccines ; 1: 16021, 2016.
Article in English | MEDLINE | ID: mdl-29263859

ABSTRACT

Significant concerns have been raised owing to the rapid global spread of infection and disease caused by the mosquito-borne Zika virus (ZIKV). Recent studies suggest that ZIKV can also be transmitted sexually, further increasing the exposure risk for this virus. Associated with this spread is a dramatic increase in cases of microcephaly and additional congenital abnormalities in infants of ZIKV-infected mothers, as well as a rise in the occurrence of Guillain Barre' syndrome in infected adults. Importantly, there are no licensed therapies or vaccines against ZIKV infection. In this study, we generate and evaluate the in vivo efficacy of a novel, synthetic, DNA vaccine targeting the pre-membrane+envelope proteins (prME) of ZIKV. Following initial in vitro development and evaluation studies of the plasmid construct, mice and non-human primates were immunised with this prME DNA-based immunogen through electroporation-mediated enhanced DNA delivery. Vaccinated animals were found to generate antigen-specific cellular and humoral immunity and neutralisation activity. In mice lacking receptors for interferon (IFN)-α/ß (designated IFNAR-/-) immunisation with this DNA vaccine induced, following in vivo viral challenge, 100% protection against infection-associated weight loss or death in addition to preventing viral pathology in brain tissue. In addition, passive transfer of non-human primate anti-ZIKV immune serum protected IFNAR-/- mice against subsequent viral challenge. This study in NHP and in a pathogenic mouse model supports the importance of immune responses targeting prME in ZIKV infection and suggests that additional research on this vaccine approach may have relevance for ZIKV control and disease prevention in humans.

18.
Oncogene ; 21(30): 4613-25, 2002 Jul 11.
Article in English | MEDLINE | ID: mdl-12096338

ABSTRACT

The targeted delivery of genes whose products arrest the cell cycle and/or induce apoptosis represent an important tool for the understanding and controlling forms of unregulated cell growth. The vpr gene product of HIV-1 has been reported to interfere with cell growth and induce apoptosis, but the mechanism of its action is not clearly understood. In order to study these important properties of Vpr, we created a recombinant adenovirus H5.010CMV-vpr (adCMV-vpr) as a tool to deliver the vpr gene to various cell lines to examine its biology. Vpr protein expression was confirmed by Western blot analysis in adCMV-vpr infected cells. We tested the effects of adCMV-vpr on cell growth of several tumor cell lines. Infection of both p53 positive and p53 deficient tumor cell lines with adCMV-vpr resulted in dramatic induction of cell death in short-term assays. We observed that apoptosis was induced through the mitochondrial pathway as we observed changes in the cytochrome c content accompanied by caspase 9 activation. As Bcl-2 is reported to interfere with apoptosis through the mitochondrial pathway, we examined the effect of adCMV-vpr in Bcl-2 over expressing cell lines. We observed that Bcl-2 overexpression does not inhibit adCMV-vpr induced apoptosis. The properties of adCMV-vpr inducing apoptosis through caspase 9 in a p53 pathway independent manner suggest that this is an important reagent. Such a vector may give insight into approaches designed to limit the growth of pathogenic human cells.


Subject(s)
Apoptosis , Caspases/metabolism , Gene Products, vpr/metabolism , Tumor Suppressor Protein p53/metabolism , Adenoviridae/genetics , Apoptosis/drug effects , Blotting, Western , Caspase 9 , Cell Cycle , Cell Size , Cell Survival , Enzyme Activation , Flow Cytometry , Gene Deletion , Gene Expression , Gene Products, vpr/genetics , Genetic Vectors/genetics , HeLa Cells , Humans , Polymerase Chain Reaction , Proto-Oncogene Proteins c-bcl-2/metabolism , Transduction, Genetic , Transgenes/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
19.
PLoS One ; 10(8): e0136323, 2015.
Article in English | MEDLINE | ID: mdl-26301899

ABSTRACT

REDD1 is a highly conserved stress response protein that is upregulated following many types of cellular stress, including hypoxia, DNA damage, energy stress, ER stress, and nutrient deprivation. Recently, REDD1 was shown to be involved in dexamethasone induced autophagy in murine thymocytes. However, we know little of REDD1's function in mature T cells. Here we show for the first time that REDD1 is upregulated following T cell stimulation with PHA or CD3/CD28 beads. REDD1 knockout T cells exhibit a defect in proliferation and cell survival, although markers of activation appear normal. These findings demonstrate a previously unappreciated role for REDD1 in T cell function.


Subject(s)
Autophagy/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Transcription Factors/genetics , Animals , Autophagy/drug effects , Cell Proliferation/drug effects , DNA Damage/genetics , Dexamethasone/administration & dosage , Mice , Mice, Knockout , T-Lymphocytes/metabolism , Thymocytes/metabolism , Thymocytes/pathology , Transcription Factors/metabolism , Transcriptional Activation/drug effects
20.
Sci Transl Med ; 7(301): 301ra132, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-26290414

ABSTRACT

First identified in 2012, Middle East respiratory syndrome (MERS) is caused by an emerging human coronavirus, which is distinct from the severe acute respiratory syndrome coronavirus (SARS-CoV), and represents a novel member of the lineage C betacoronoviruses. Since its identification, MERS coronavirus (MERS-CoV) has been linked to more than 1372 infections manifesting with severe morbidity and, often, mortality (about 495 deaths) in the Arabian Peninsula, Europe, and, most recently, the United States. Human-to-human transmission has been documented, with nosocomial transmission appearing to be an important route of infection. The recent increase in cases of MERS in the Middle East coupled with the lack of approved antiviral therapies or vaccines to treat or prevent this infection are causes for concern. We report on the development of a synthetic DNA vaccine against MERS-CoV. An optimized DNA vaccine encoding the MERS spike protein induced potent cellular immunity and antigen-specific neutralizing antibodies in mice, macaques, and camels. Vaccinated rhesus macaques seroconverted rapidly and exhibited high levels of virus-neutralizing activity. Upon MERS viral challenge, all of the monkeys in the control-vaccinated group developed characteristic disease, including pneumonia. Vaccinated macaques were protected and failed to demonstrate any clinical or radiographic signs of pneumonia. These studies demonstrate that a consensus MERS spike protein synthetic DNA vaccine can induce protective responses against viral challenge, indicating that this strategy may have value as a possible vaccine modality against this emerging pathogen.


Subject(s)
Middle East Respiratory Syndrome Coronavirus/immunology , Vaccines, DNA/therapeutic use , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Camelus , Macaca mulatta , Mice
SELECTION OF CITATIONS
SEARCH DETAIL