Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Elife ; 132024 Jan 22.
Article in English | MEDLINE | ID: mdl-38252125

ABSTRACT

Inflammasomes are filamentous signaling platforms essential for host defense against various intracellular calamities such as pathogen invasion and genotoxic stresses. However, dysregulated inflammasomes cause an array of human diseases including autoinflammatory disorders and cancer. It was recently identified that endogenous pyrin-only-proteins (POPs) regulate inflammasomes by directly inhibiting their filament assembly. Here, by combining Rosetta in silico, in vitro, and in cellulo methods, we investigate the target specificity and inhibition mechanisms of POPs. We find here that POP1 is ineffective in directly inhibiting the central inflammasome adaptor ASC. Instead, POP1 acts as a decoy and targets the assembly of upstream receptor pyrin-domain (PYD) filaments such as those of AIM2, IFI16, NLRP3, and NLRP6. Moreover, not only does POP2 directly suppress the nucleation of ASC, but it can also inhibit the elongation of receptor filaments. In addition to inhibiting the elongation of AIM2 and NLRP6 filaments, POP3 potently suppresses the nucleation of ASC. Our Rosetta analyses and biochemical experiments consistently suggest that a combination of favorable and unfavorable interactions between POPs and PYDs is necessary for effective recognition and inhibition. Together, we reveal the intrinsic target redundancy of POPs and their inhibitory mechanisms.


Subject(s)
Cytoskeleton , Inflammasomes , Humans , Pyrin , DNA Damage , Inhibition, Psychological
2.
JCI Insight ; 7(12)2022 06 22.
Article in English | MEDLINE | ID: mdl-35588060

ABSTRACT

The Aedes aegypti mosquito transmits both dengue virus (DENV) and Zika virus (ZIKV) . Individuals in endemic areas are at risk for infection with both viruses, as well as for repeated DENV infection. In the presence of anti-DENV antibodies, outcomes of secondary DENV infection range from mild to life threatening. Furthermore, the role of cross-reactive antibodies on the course of ZIKV infection remains unclear. We assessed the ability of cross-reactive DENV mAbs or polyclonal immunoglobulin isolated after DENV vaccination to upregulate type I IFN production by plasmacytoid DCs (pDCs) in response to both heterotypic DENV- and ZIKV-infected cells. We found a range in the ability of antibodies to increase pDC IFN production and a positive correlation between IFN production and the ability of an antibody to bind to the infected cell surface. Engagement of Fc receptors on the pDC and engagement of epitope on the infected cell by the Fab portion of the same antibody molecule was required to mediate increased IFN production by providing specificity to and promoting pDC sensing of DENV or ZIKV. This represents a mechanism independent of neutralization by which preexisting cross-reactive DENV antibodies could protect a subset of individuals from severe outcomes during secondary heterotypic DENV or ZIKV infection.


Subject(s)
Dengue Virus , Dengue , Zika Virus Infection , Zika Virus , Animals , Cross Reactions , Humans
5.
Nucleic Acids Res ; 48(8): 4435-4447, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32170294

ABSTRACT

Cyclic-G/AMP (cGAMP) synthase (cGAS) triggers host innate immune responses against cytosolic double-stranded (ds)DNA arising from genotoxic stress and pathogen invasion. The canonical activation mechanism of cGAS entails dsDNA-binding and dimerization. Here, we report an unexpected activation mechanism of cGAS in which Mn2+ activates monomeric cGAS without dsDNA. Importantly, the Mn2+-mediated activation positively couples with dsDNA-dependent activation in a concerted manner. Moreover, the positive coupling between Mn2+ and dsDNA length-dependent activation requires the cognate ATP/GTP substrate pair, while negative-cooperativity suppresses Mn2+ utilization by either ATP or GTP alone. Additionally, while Mn2+ accelerates the overall catalytic activity, dsDNA length-dependent dimerization specifically accelerates the cyclization of cGAMP. Together, we demonstrate how the intrinsic allostery of cGAS efficiently yet precisely tunes its activity.


Subject(s)
DNA/metabolism , Manganese , Nucleotidyltransferases/metabolism , Adenosine Triphosphate/metabolism , Allosteric Regulation , Biocatalysis , Cell Line , DNA/chemistry , Enzyme Activation , Humans , Nucleotidyltransferases/chemistry , Substrate Specificity
7.
Methods Enzymol ; 625: 269-285, 2019.
Article in English | MEDLINE | ID: mdl-31455531

ABSTRACT

The innate immune system is an evolutionarily conserved host defense system and is the first barrier to infection. The system utilizes genetically conserved receptors to identify the presence of microbial structures. Engagement of innate immune receptors by primarily by ligands that discriminate pathogens from the host activates programmed responses that limit pathogen expansion. Despite its ubiquitous nature, surprisingly DNA is a critical structure that triggers innate immune responses. Focusing on structural modifications or aberrant location of DNA, innate immune receptors identify physiologic stress. Inflammasomes and interferons are critical innate immune pathways that are activated by DNA. DNA binding proteins that tie recognition of DNA to both programmed responses have been identified, and their importance demonstrated in infection models. In this chapter, we discuss techniques to analyze AIM2 inflammasome and cGAS interferon activation by synthetic DNA and DNA viruses. We also discuss methods to measure the activity of these immune pathways.


Subject(s)
Inflammasomes/metabolism , Pathogen-Associated Molecular Pattern Molecules/metabolism , Animals , DNA Viruses/physiology , DNA-Binding Proteins/metabolism , Humans , Immunity, Innate/physiology , Interferon Type I/metabolism
8.
J Clin Invest ; 129(8): 3038-3040, 2019 06 24.
Article in English | MEDLINE | ID: mdl-31232703

ABSTRACT

Solid organ transplantation from hepatitis C virus-positive (HCV-positive) deceased donors into HCV-negative recipients is a recent approach aimed to expand the donor organ pool in the setting of severe shortage. Good short-term outcomes have been reported with this approach in combination with direct-acting antivirals. In this issue of the JCI, Zahid and colleagues have characterized early viral kinetics and the genetic landscape of donor-to-recipient HCV transmission using single-genome sequencing. In seven HCV-negative recipients of four HCV-positive donor organs, productive infection with a highly diverse viral population was seen by day three after transplantation. The degree of genetic diversity seen in recipients of HCV-positive organs was unlike the narrow genetic bottleneck typically observed with acute HCV acquisition from intravenous drug use or sexual activity. All recipients achieved HCV cure with treatment. The consequences of acute infection with a genetically diverse HCV population are unknown; however, early clinical experience with this transplantation strategy is promising.


Subject(s)
Antiviral Agents , Hepatitis C, Chronic , Hepatitis C , Organ Transplantation , Hepacivirus , Humans , Tissue Donors
9.
Ann Intern Med ; 168(8): 533-540, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29507971

ABSTRACT

Background: Given the high mortality rate for patients with end-stage kidney disease receiving dialysis and the efficacy and safety of hepatitis C virus (HCV) treatments, discarded kidneys from HCV-infected donors may be a neglected public health resource. Objective: To determine the tolerability and feasibility of using direct-acting antivirals (DAAs) as prophylaxis before and after kidney transplantation from HCV-infected donors to non-HCV-infected recipients (that is, HCV D+/R- transplantation). Design: Open-label nonrandomized trial. (ClinicalTrials.gov: NCT02781649). Setting: Single center. Participants: 10 HCV D+/R- kidney transplant candidates older than 50 years with no available living donors. Intervention: Transplantation of kidneys from deceased donors aged 13 to 50 years with positive HCV RNA and HCV antibody test results. All recipients received a dose of grazoprevir (GZR), 100 mg, and elbasvir (EBR), 50 mg, immediately before transplantation. Recipients of kidneys from donors with genotype 1 infection continued receiving GZR-EBR for 12 weeks after transplantation; those receiving organs from donors with genotype 2 or 3 infection had sofosbuvir, 400 mg, added to GZR-EBR for 12 weeks of triple therapy. Measurements: The primary safety outcome was the incidence of adverse events related to GZR-EBR treatment. The primary efficacy outcome was the proportion of recipients with an HCV RNA level below the lower limit of quantification 12 weeks after prophylaxis. Results: Among 10 HCV D+/R- transplant recipients, no treatment-related adverse events occurred, and HCV RNA was not detected in any recipient 12 weeks after treatment. Limitation: Nonrandomized study design and a small number of patients. Conclusion: Pre- and posttransplantation HCV treatment was safe and prevented chronic HCV infection in HCV D+/R- kidney transplant recipients. If confirmed in larger studies, this strategy should markedly expand organ options and reduce mortality for kidney transplant candidates without HCV infection. Primary Funding Source: Merck Sharp & Dohme.


Subject(s)
Antiviral Agents/therapeutic use , Hepatitis C/drug therapy , Hepatitis C/transmission , Kidney Transplantation , Kidney/virology , Tissue Donors , Adolescent , Adult , Amides , Benzofurans/therapeutic use , Carbamates , Cyclopropanes , Drug Therapy, Combination , Feasibility Studies , Female , Genotype , Graft Rejection , Graft Survival , Humans , Imidazoles/therapeutic use , Male , Middle Aged , Quinoxalines/therapeutic use , RNA, Viral/analysis , Sofosbuvir/therapeutic use , Sulfonamides , Treatment Outcome
10.
J Clin Invest ; 127(12): 4352-4364, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29083319

ABSTRACT

Type I IFN production is essential for innate control of acute viral infection; however, prolonged high-level IFN production is associated with chronic immune activation in HIV-infected individuals. Although plasmacytoid DCs (pDCs) are a primary source of IFN, the mechanisms that regulate IFN levels following the acute phase are unknown. We hypothesized that HIV-specific Ab responses regulate late IFN production. We evaluated the mechanism through which HIV-activated pDCs produce IFN as well as how both monoclonal HIV-specific Abs and Abs produced in natural HIV infection modulated normal pDC sensing of HIV. We found that HIV-induced IFN production required TLR7 signaling, receptor-mediated entry, fusion, and viral uncoating, but not endocytosis or HIV life cycle stages after uncoating. Abs directed against the HIV envelope that do not interfere with CD4 binding markedly enhanced the IFN response, irrespective of their ability to neutralize CD4+ T cell infection. Ab-mediated enhancement of IFN production required Fc γ receptor engagement, bypassed fusion, and initiated signaling through both TLR7 and TLR9, which was not utilized in the absence of Ab. Polyclonal Abs isolated from HIV-infected subjects also enhanced pDC production of IFN in response to HIV. Our data provide an explanation for high levels of IFN production and immune activation in chronic HIV infection.


Subject(s)
Antigen-Antibody Complex/immunology , Dendritic Cells/immunology , HIV Antibodies/immunology , HIV-1/immunology , Interferon Type I/immunology , Plasma Cells/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Dendritic Cells/pathology , Humans , Plasma Cells/pathology , Signal Transduction/immunology , Toll-Like Receptor 7/immunology , Viral Envelope Proteins/immunology
11.
Clin Infect Dis ; 64(5): 589-596, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27927859

ABSTRACT

BACKGROUND: HIV/HCV coinfection and elevated interleukin (IL)-18 levels are both associated with enhanced progression of hepatic inflammation and increased risk of diabetes, kidney disease, and cardiovascular disease. IL-18 is a proinflammatory cytokine made upon activation of the inflammasome, an innate sensing system. We assessed whether increased IL-18 could explain the increased incidence and progression of inflammatory conditions seen with HIV/HCV coinfection. METHODS: Serum from 559 subjects with HIV monoinfection, HCV monoinfection, HIV/HCV coinfection, or people who inject drugs with neither infection was tested for IL-18 by ELISA and for 16 other analytes by electrochemiluminescence immunoassay. IL-18 levels were measured in 14 additional chronically HCV infected subjects who developed incident HIV infection to determine if IL-18 increases with coinfection. RESULTS: IL-18 was significantly elevated in coinfected individuals versus both monoinfections (p<0.0001) independent of age, sex, and race. IL-18 levels were significantly higher in HIV monoinfection than in HCV monoinfection. High IL-18 levels were correlated with detectable HIV viremia and inversely with CD4 count (p<0.0001), consistent with HIV activation of the inflammasome resulting in CD4 T cell depletion. Incident HIV infection of chronically HCV infected subjects resulted in increased IL-18 (p<0.001), while HIV suppression was associated with normal IL-18 levels. Four additional analytes (IP-10, IL-12/23p40, IFNy, IL-15) were found to be elevated in HIV/HCV coinfection when compared to both monoinfections. CONCLUSIONS: HIV/HCV coinfection results in significantly elevated serum IL-18. The elevated levels of this proinflammatory cytokine may explain the increased incidence and progression of inflammatory illnesses seen in coinfected individuals.


Subject(s)
Coinfection/blood , HIV Infections/blood , HIV Infections/complications , Hepatitis C/blood , Hepatitis C/complications , Interleukin-18/blood , Adult , Aged , CD4 Lymphocyte Count , Cohort Studies , Coinfection/epidemiology , Cytokines/blood , Female , HIV Infections/epidemiology , Hepatitis C/epidemiology , Humans , Male , Middle Aged , Young Adult
12.
Cancer Discov ; 6(12): 1366-1381, 2016 12.
Article in English | MEDLINE | ID: mdl-27683557

ABSTRACT

Programmed death-1 (PD-1) is a coinhibitory receptor that downregulates the activity of tumor-infiltrating lymphocytes (TIL) in cancer and of virus-specific T cells in chronic infection. The molecular mechanisms driving high PD-1 expression on TILs have not been fully investigated. We demonstrate that TGFß1 enhances antigen-induced PD-1 expression through SMAD3-dependent, SMAD2-independent transcriptional activation in T cells in vitro and in TILs in vivo The PD-1hi subset seen in CD8+ TILs is absent in Smad3-deficient tumor-specific CD8+ TILs, resulting in enhanced cytokine production by TILs and in draining lymph nodes and antitumor activity. In addition to TGFß1's previously known effects on T-cell function, our findings suggest that TGFß1 mediates T-cell suppression via PD-1 upregulation in the tumor microenvironment (TME). They highlight bidirectional cross-talk between effector TILs and TGFß-producing cells that upregulates multiple components of the PD-1 signaling pathway to inhibit antitumor immunity. SIGNIFICANCE: Engagement of the coinhibitory receptor PD-1 or its ligand, PD-L1, dramatically inhibits the antitumor function of TILs within the TME. Our findings represent a novel immunosuppressive function of TGFß and demonstrate that TGFß1 allows tumors to evade host immune responses in part through enhanced SMAD3-mediated PD-1 expression on TILs. Cancer Discov; 6(12); 1366-81. ©2016 AACRThis article is highlighted in the In This Issue feature, p. 1293.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Neoplasms/immunology , Programmed Cell Death 1 Receptor/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Cell Line, Tumor , Cytokines/metabolism , Drug Resistance, Neoplasm , Humans , Jurkat Cells , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Neoplasms/metabolism , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction , Tumor Microenvironment
13.
PLoS Pathog ; 10(5): e1004082, 2014 May.
Article in English | MEDLINE | ID: mdl-24788318

ABSTRACT

Innate immune sensing of viral infection results in type I interferon (IFN) production and inflammasome activation. Type I IFNs, primarily IFN-α and IFN-ß, are produced by all cell types upon virus infection and promote an antiviral state in surrounding cells by inducing the expression of IFN-stimulated genes. Type I IFN production is mediated by Toll-like receptor (TLR) 3 in HCV infected hepatocytes. Type I IFNs are also produced by plasmacytoid dendritic cells (pDC) after sensing of HIV and HCV through TLR7 in the absence of productive pDC infection. Inflammasomes are multi-protein cytosolic complexes that integrate several pathogen-triggered signaling cascades ultimately leading to caspase-1 activation and generation pro-inflammatory cytokines including interleukin (IL)-18 and IL-1ß. Here, we demonstrate that HIV and HCV activate the inflammasome, but not Type I IFN production, in monocytes and macrophages in an infection-independent process that requires clathrin-mediated endocytosis and recognition of the virus by distinct endosomal TLRs. Knockdown of each endosomal TLR in primary monocytes by RNA interference reveals that inflammasome activation in these cells results from HIV sensing by TLR8 and HCV recognition by TLR7. Despite its critical role in type I IFN production by pDCs stimulated with HIV, TLR7 is not required for inflammasome activation by HIV. Similarly, HCV activation of the inflammasome in monocytes does not require TLR3 or its downstream signaling adaptor TICAM-1, while this pathway leads to type I IFN in infected hepatocytes. Monocytes and macrophages do not produce type I IFN upon TLR8 or TLR7 sensing of HIV or HCV, respectively. These findings reveal a novel infection-independent mechanism for chronic viral induction of key anti-viral programs and demonstrate distinct TLR utilization by different cell types for activation of the type I IFN vs. inflammasome pathways of inflammation.


Subject(s)
HIV-1/immunology , Hepacivirus/immunology , Inflammasomes/metabolism , Interferon Type I/metabolism , Macrophages/metabolism , Monocytes/metabolism , Toll-Like Receptors/physiology , Cells, Cultured , Cytokines/metabolism , Endocytosis/immunology , Endosomes/immunology , Endosomes/metabolism , HIV Infections/immunology , HIV Infections/metabolism , HIV-1/physiology , Hep G2 Cells , Hepacivirus/physiology , Hepatitis C/immunology , Hepatitis C/metabolism , Humans , Macrophages/immunology , Macrophages/virology , Monocytes/immunology , Monocytes/virology , Virus Internalization
14.
J Infect Dis ; 204(11): 1730-40, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21984735

ABSTRACT

BACKGROUND: Proinflammatory cytokines play a critical role in antiviral immune responses. Large-scale genome studies have found correlations between single-nucleotide polymorphisms (SNPs) in the interleukin (IL) 18 promoter and spontaneous control of hepatitis C virus (HCV), suggesting a role in clearance. METHODS: Plasma IL-18, IL-1ß, IL-6, IL-8, IL-12, interferon-γ, tumor necrosis factor-α, alanine aminotransferase (ALT), and HCV RNA levels were assessed longitudinally in subjects with known dates of HCV acquisition and analyzed according to IL-18 SNPs and outcome, either spontaneous clearance (SC) (n = 13) or persistent infection (PI) (n = 25). RESULTS: No significant change in plasma proinflammatory cytokine expression was observed with the exception of IL-18, which increased in every subject with initial detection of HCV RNA. In every SC subject, IL-18 returned to the preinfection baseline concomitant with HCV control. In PI subjects, IL-18 declined following the acute phase of infection but remained above the preinfection baseline throughout chronic infection and did not correlate with HCV RNA or ALT levels. CONCLUSIONS: Plasma IL-18 was an early and the most reliably detected host response to HCV infection measured in blood. Reduced IL-18 production with transition to chronic infection without correlation with HCV RNA or ALT levels suggests modulation of the innate response with persistent infection.


Subject(s)
Hepatitis C/immunology , Interleukin-18/blood , Acute Disease , Alanine Transaminase/blood , Biomarkers/blood , Genotype , Hepatitis C/genetics , Humans , Interleukin-18/genetics , Longitudinal Studies , Polymorphism, Single Nucleotide , RNA, Viral/blood , Statistics, Nonparametric
15.
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
16.
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
17.
J Immunol ; 175(1): 112-23, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15972637

ABSTRACT

DNA vaccines are a promising technology for the induction of Ag-specific immune responses, and much recent attention has gone into improving their immune potency. In this study we test the feasibility of delivering a plasmid encoding IL-15 as a DNA vaccine adjuvant for the induction of improved Ag-specific CD8(+) T cellular immune responses. Because native IL-15 is poorly expressed, we used PCR-based strategies to develop an optimized construct that expresses 80-fold higher than the native IL-15 construct. Using a DNA vaccination model, we determined that immunization with optimized IL-15 in combination with HIV-1gag DNA constructs resulted in a significant enhancement of Ag-specific CD8(+) T cell proliferation and IFN-gamma secretion, and strong induction of long-lived CD8(+) T cell responses. In an influenza DNA vaccine model, coimmunization with plasmid expressing influenza A PR8/34 hemagglutinin with the optimized IL-15 plasmid generated improved long term CD8(+) T cellular immunity and protected the mice against a lethal mucosal challenge with influenza virus. Because we observed that IL-15 appeared to mostly adjuvant CD8(+) T cell function, we show that in the partial, but not total, absence of CD4(+) T cell help, plasmid-delivered IL-15 could restore CD8 secondary immune responses to an antigenic DNA plasmid, supporting the idea that the effects of IL-15 on CD8(+) T cell expansion require the presence of low levels of CD4 T cells. These data suggest a role for enhanced plasmid IL-15 as a candidate adjuvant for vaccine or immunotherapeutic studies.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Interleukin-15/genetics , Interleukin-15/immunology , AIDS Vaccines/genetics , AIDS Vaccines/immunology , AIDS Vaccines/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Cell Proliferation , Cellular Senescence , Cloning, Molecular , Female , Genetic Vectors , HeLa Cells , Humans , Immunization , Immunologic Memory , In Vitro Techniques , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Influenza Vaccines/pharmacology , Interferon-gamma/biosynthesis , Lymphocyte Activation , Lymphocyte Cooperation , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Plasmids , Transfection , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, DNA/pharmacology
18.
Vaccine ; 22(13-14): 1744-50, 2004 Apr 16.
Article in English | MEDLINE | ID: mdl-15068858

ABSTRACT

Plasmid encoded exogenous IL-12 delivered as a DNA vaccine adjuvant has been shown to improve vaccine-induced immunity. In particular, pIL-12 greatly improves antigen (Ag)-specific cytotoxic tlymphocyte (CTL) activity in immunized mice. The longevity of this response has not previously been studied in detail. We have studied the effect of co-immunization with pIL-12 on HIV gp160 and Influenza A Hemeagglutinnin-specific memory immune responses. Mice co-immunized with pIL-12 and plasmid encoded antigens maintained a greater memory response than those immunized with the plasmid antigen alone which could be measured at least 6 months after vaccination. Further, this translated to an improved outcome after challenge of long term rested mice that were previously immunized. The strength of the immune response as well as the number of Ag-specific T-cells is proportional to the number of Ag-specific cells primed by the vaccination regimen.


Subject(s)
Adjuvants, Immunologic/pharmacology , Immunologic Memory , Interleukin-12/pharmacology , T-Lymphocytes/immunology , AIDS Vaccines/immunology , Animals , Cell Division/drug effects , Chromium Radioisotopes , Female , HIV Infections/virology , Immunization , Immunization Schedule , Interferon-gamma/biosynthesis , Mice , Mice, Inbred BALB C , Plasmids/immunology , Spleen/immunology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA/immunology
19.
J Immunol ; 171(8): 4311-9, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14530356

ABSTRACT

There is great interest in understanding the role of costimulatory molecules in immune activation. In both the influenza and HIV DNA immunization models, several groups have reported that coimmunization of mice with plasmids encoding immunogen and CD86, but not CD80, effectively boosts Ag-specific T cell activation. This difference in immune priming provided an opportunity to examine the functional importance of different regions of the B.7 molecules in immune activation. To examine this issue, we developed a series of chimeric CD80 and CD86 constructs as well as deletion mutants, and examined their immune activating potential in the DNA vaccine model. We demonstrate that the lack of an Ig constant-like region in the CD80 molecule is critically important to the enhanced immune activation observed. CD80 C-domain deletion mutants induce a highly inflammatory Ag-specific cellular response when administered as part of a plasmid vaccine. The data suggest that the constant-like domains, likely through intermolecular interactions, are critically important for immune regulation during costimulation and that engineered CD80/86 molecules represent more potent costimulatory molecules and may improve vaccine adjuvant efficacy.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/physiology , Antigens, CD/physiology , B7-1 Antigen/physiology , Immunoglobulin Constant Regions/physiology , Membrane Glycoproteins/physiology , Models, Immunological , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Adjuvants, Immunologic/antagonists & inhibitors , Adjuvants, Immunologic/genetics , Animals , Antigens, CD/administration & dosage , Antigens, CD/biosynthesis , Antigens, CD/genetics , B7-1 Antigen/administration & dosage , B7-1 Antigen/biosynthesis , B7-1 Antigen/genetics , B7-2 Antigen , Cell Movement/genetics , Cell Movement/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Down-Regulation/genetics , Down-Regulation/immunology , HIV-1/genetics , HIV-1/immunology , Humans , Immunoglobulin Constant Regions/administration & dosage , Immunoglobulin Constant Regions/genetics , Immunoglobulin Variable Region/administration & dosage , Immunoglobulin Variable Region/genetics , Lymphocyte Subsets/cytology , Lymphocyte Subsets/immunology , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mutagenesis, Site-Directed , Plasmids , Protein Structure, Tertiary/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Sequence Deletion/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Simian virus 40/genetics , Simian virus 40/immunology , Transfection , Up-Regulation/genetics , Up-Regulation/immunology , Vaccines, DNA/genetics
20.
Expert Opin Biol Ther ; 3(6): 887-94, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12943447

ABSTRACT

Apoptotic bodies deliver antigens (Ags) to the cross-presentation pathways of dendritic cells (DCs), where their presentation has been associated with both the maintenance of tolerance as well as the induction of protective immunity. The manner in which apoptotic bodies are generated, their abundance in relation to local DCs, and the milieu in which they are generated appear to be the major factors determining whether apoptotic bodies will induce CD8(+) T cell activation or anergy. These observations have been extended to the field of vaccination, where the engineered apoptosis of Ag-bearing/loaded cells in vivo has been used to prime strong CD8(+) T cell immunity. This review will examine Ag capture and cross-presentation by DCs, with particular emphasis on the manipulation of apoptotic bodies in vivo for the purpose of vaccination.


Subject(s)
Antigen Presentation/immunology , Apoptosis , Dendritic Cells/physiology , Immune Tolerance , T-Lymphocytes/immunology , Animals , Cytokines/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...