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










Publication year range
1.
Cancers (Basel) ; 15(24)2023 Dec 08.
Article in English | MEDLINE | ID: mdl-38136298

ABSTRACT

Nexavant was reported as an alternative to the TLR3 agonist of Poly(I:C) and its derivatives. The physicochemical properties, signaling pathways, anti-cancer effects, and mechanisms of Nexavant were investigated. The distinctive characteristics of Nexavant compared to that of Poly(I:C) were demonstrated by precise quantification, enhanced thermostability, and increased resistance to RNase A. Unlike Poly(I:C), which activates TLR3, RIG-I, and MDA5, Nexavant stimulates signaling through TLR3 and RIG-I but not through MDA5. Compared to Poly(I:C), an intratumoral Nexavant treatment led to a unique immune response, immune cell infiltration, and suppression of tumor growth in various animal cancer models. Nexavant therapy outperformed anti-PD-1 antibody treatment in all the tested models and showed a synergistic effect in combinational therapy, especially in well-defined cold tumor models. The effect was similar to that of nivolumab in a humanized mouse model. Intranasal instillation of Nexavant led to the recruitment of immune cells (NK, CD4+ T, and CD8+ T) to the lungs, suppressing lung metastasis and improving animal survival. Our study highlighted Nexavant's defined nature for clinical use and unique signaling pathways and its potential as a standalone anti-cancer agent or in combination with anti-PD-1 antibodies.

2.
Cancer Lett ; 579: 216480, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37931834

ABSTRACT

Glioblastoma (GBM) is an aggressive brain tumor with a median survival of 15 months and has limited treatment options. Immunotherapy with checkpoint inhibitors has shown minimal efficacy in combating GBM, and large clinical trials have failed. New immunotherapy approaches and a deeper understanding of immune surveillance of GBM are needed to advance treatment options for this devastating disease. In this study, we used two preclinical models of GBM: orthotopically delivering either GBM stem cells or employing CRISPR-mediated tumorigenesis by adeno-associated virus, to establish immunologically proficient and non-inflamed tumors, respectively. After tumor development, the innate immune system was activated through long-term STING activation by a pharmacological agonist, which reduced tumor progression and prolonged survival. Recruitment and activation of cytotoxic T-cells were detected in the tumors, and T-cell specificity towards the cancer cells was observed. Interestingly, prolonged STING activation altered the tumor vasculature, inducing hypoxia and activation of VEGFR, as measured by a kinome array and VEGF expression. Combination treatment with anti-PD1 did not provide a synergistic effect, indicating that STING activation alone is sufficient to activate immune surveillance and hinder tumor development through vascular disruption. These results guide future studies to refine innate immune activation as a treatment approach for GBM, in combination with anti-VEGF to impede tumor progression and induce an immunological response against the tumor.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Glioblastoma/immunology , Glioblastoma/metabolism , Immunotherapy/methods , Tumor Microenvironment , Immunity, Innate
3.
Mol Cell ; 82(13): 2385-2400.e9, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35594856

ABSTRACT

Inflammation observed in SARS-CoV-2-infected patients suggests that inflammasomes, proinflammatory intracellular complexes, regulate various steps of infection. Lung epithelial cells express inflammasome-forming sensors and constitute the primary entry door of SARS-CoV-2. Here, we describe that the NLRP1 inflammasome detects SARS-CoV-2 infection in human lung epithelial cells. Specifically, human NLRP1 is cleaved at the Q333 site by multiple coronavirus 3CL proteases, which triggers inflammasome assembly and cell death and limits the production of infectious viral particles. Analysis of NLRP1-associated pathways unveils that 3CL proteases also inactivate the pyroptosis executioner Gasdermin D (GSDMD). Subsequently, caspase-3 and GSDME promote alternative cell pyroptosis. Finally, analysis of pyroptosis markers in plasma from COVID-19 patients with characterized severe pneumonia due to autoantibodies against, or inborn errors of, type I interferons (IFNs) highlights GSDME/caspase-3 as potential markers of disease severity. Overall, our findings identify NLRP1 as a sensor of SARS-CoV-2 infection in lung epithelia.


Subject(s)
COVID-19 , Coronavirus 3C Proteases , Epithelial Cells , Inflammasomes , NLR Proteins , SARS-CoV-2 , COVID-19/genetics , COVID-19/metabolism , COVID-19/virology , Caspase 3/metabolism , Coronavirus 3C Proteases/genetics , Coronavirus 3C Proteases/metabolism , Epithelial Cells/metabolism , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , Lung/metabolism , Lung/virology , NLR Proteins/genetics , NLR Proteins/metabolism , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/metabolism , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , Pyroptosis , SARS-CoV-2/enzymology , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity
4.
EBioMedicine ; 58: 102922, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32739871

ABSTRACT

BACKGROUND: PRR (Pattern Recognition Receptor) agonists have been widely tested as potent vaccine adjuvants. TLR7 (Toll-Like Receptor 7) and NOD2 (nucleotide-binding oligomerization domain 2) are key innate receptors widely expressed at mucosal levels. METHODS: Here, we evaluated the immunostimulatory properties of a novel hybrid chemical compound designed to stimulate both TLR7 and NOD2 receptors. FINDING: The combined TLR7/NOD2 agonist showed increase efficacy than TLR7L or NOD2L agonists alone or combined in different in vitro models. Dual TLR7/NOD2 agonist efficiently stimulates TLR7 and NOD2, and promotes the maturation and reprogramming of human dendritic cells, as well as the secretion of pro-inflammatory or adaptive cytokines. This molecule also strongly induces autophagy in human cells which is a major intracellular degradation system that delivers cytoplasmic constituents to lysosomes in both MHC class I and II-restricted antigen presentation. In vivo, TLR7/NOD2L agonist is a potent adjuvant after intranasal administration with NP-p24 HIV vaccine, inducing high-quality humoral and adaptive responses both in systemic and mucosal compartments. Use of TLR7/NOD2L adjuvant improves very significantly the protection of mice against an intranasal challenge with a vaccinia virus expressing the p24. INTERPRETATION: Dual TLR7/NOD2L agonist is a very potent and versatile vaccine adjuvant and promote very efficiently both systemic and mucosal immunity. FUNDING: This work was supported by Sidaction.


Subject(s)
AIDS Vaccines/administration & dosage , Adjuvants, Immunologic/administration & dosage , CD8-Positive T-Lymphocytes/metabolism , Nod2 Signaling Adaptor Protein/agonists , Toll-Like Receptor 7/agonists , AIDS Vaccines/immunology , Adaptive Immunity , Administration, Intranasal , Animals , Cell Line , Female , HeLa Cells , Humans , Immunity, Humoral , Mice , Nod2 Signaling Adaptor Protein/immunology , Toll-Like Receptor 7/immunology
5.
Oncogene ; 39(8): 1652-1664, 2020 02.
Article in English | MEDLINE | ID: mdl-31740782

ABSTRACT

Hepatocellular carcinoma (HCC) is the most common primary liver cancer, and the incidence of HCC is increasing. Recently, cancer immunotherapy has emerged as an efficient treatment against some cancers. Here we have used a mouse model of mutagen-induced HCC to explore the therapeutic usefulness of targeting the DNA-activated STING pathway in HCC. STING-deficient mice exhibited unaltered initial development of HCC, but had higher number of large tumors at late stages of disease. In the liver of STING-deficient HCC mice, we observed reduced levels of phospho-STAT1, autophagy, and cleaved caspase3. These responses were activated in the liver by treatment with a cyclic dinucleotide (CDN) STING agonist. Importantly, CDN treatment of mice after HCC development efficiently reduced tumor size. Initiation of CDN treatment at an even later stage of disease to allow HCC detection by MR scanning revealed that the majority of tumors regressed in response to CDN, but new tumors were also detected, which were unresponsive to CDN treatment. Overall, the modulation of the STING pathway affects the development of HCC, and holds promise for a use as a treatment of this disease, most likely in combination with other immunomodulatory treatments such as PD1 inhibitors or with standard of care.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Membrane Proteins/metabolism , Molecular Targeted Therapy , Nucleotidyltransferases/metabolism , Animals , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic , Humans , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Membrane Proteins/agonists , Mice , Signal Transduction/drug effects , Tumor Burden/drug effects
6.
JCI Insight ; 4(7)2019 04 04.
Article in English | MEDLINE | ID: mdl-30944257

ABSTRACT

Pathogen recognition receptor (PRR) agonists are currently being developed and tested as adjuvants in various formulations to optimize the immunogenicity and efficacy of vaccines. Using an original in vitro approach to prime naive precursors from unfractionated human peripheral blood mononuclear cells, we assessed the influence of cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), a ligand for the stimulator of interferon genes (STING), on the induction of antigen-specific CD8+ T cells. We found that 2'3'-cGAMP and 3'3'-cGAMP were especially potent adjuvants in this system, driving the expansion and maturation of functionally replete antigen-specific CD8+ T cells via the induction of type I IFNs. The biological relevance of these findings was confirmed in vivo using two mouse models, in which 2'3'-cGAMP-adjuvanted vaccination elicited protective antitumor or antiviral CD8+ T cell responses. These results identify particular isoforms of cGAMP as effective adjuvants that may find utility in the development of novel immunotherapies and vaccines.


Subject(s)
Adjuvants, Immunologic/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Membrane Proteins/agonists , Nucleotides, Cyclic/immunology , Vaccination/methods , Animals , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Line, Tumor/transplantation , Cells, Cultured , Disease Models, Animal , Female , HIV Infections/immunology , HIV Infections/prevention & control , HIV Infections/virology , HIV-1/immunology , Humans , Immunogenicity, Vaccine , Interferon Type I/immunology , Interferon Type I/metabolism , Ligands , Mice , Nucleotides, Cyclic/administration & dosage , Primary Cell Culture , Thymoma/immunology , Thymoma/pathology , Thymoma/prevention & control , Thymus Neoplasms/immunology , Thymus Neoplasms/pathology , Thymus Neoplasms/prevention & control , Viral Vaccines/administration & dosage , Viral Vaccines/immunology
7.
PLoS Pathog ; 14(4): e1006976, 2018 04.
Article in English | MEDLINE | ID: mdl-29608601

ABSTRACT

In recent years, there has been an increasing interest in immunomodulatory therapy as a means to treat various conditions, including infectious diseases. For instance, Toll-like receptor (TLR) agonists have been evaluated for treatment of genital herpes. However, although the TLR7 agonist imiquimod was shown to have antiviral activity in individual patients, no significant effects were observed in clinical trials, and the compound also exhibited significant side effects, including local inflammation. Cytosolic DNA is detected by the enzyme cyclic GMP-AMP (2'3'-cGAMP) synthase (cGAS) to stimulate antiviral pathways, mainly through induction of type I interferon (IFN)s. cGAS is activated upon DNA binding to produce the cyclic dinucleotide (CDN) 2'3'-cGAMP, which in turn binds and activates the adaptor protein Stimulator of interferon genes (STING), thus triggering type I IFN expression. In contrast to TLRs, STING is expressed broadly, including in epithelial cells. Here we report that natural and non-natural STING agonists strongly induce type I IFNs in human cells and in mice in vivo, without stimulating significant inflammatory gene expression. Systemic treatment with 2'3'-cGAMP reduced genital herpes simplex virus (HSV) 2 replication and improved the clinical outcome of infection. More importantly, local application of CDNs at the genital epithelial surface gave rise to local IFN activity, but only limited systemic responses, and this treatment conferred total protection against disease in both immunocompetent and immunocompromised mice. In direct comparison between CDNs and TLR agonists, only CDNs acted directly on epithelial cells, hence allowing a more rapid and IFN-focused immune response in the vaginal epithelium. Thus, specific activation of the STING pathway in the vagina evokes induction of the IFN system but limited inflammatory responses to allow control of HSV2 infections in vivo.


Subject(s)
Antiviral Agents/pharmacology , Herpes Genitalis/prevention & control , Herpesvirus 2, Human/drug effects , Host-Pathogen Interactions/drug effects , Membrane Proteins/agonists , Nucleotides, Cyclic/pharmacology , Animals , Cells, Cultured , Cytosol/virology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/virology , Female , Herpes Genitalis/metabolism , Herpes Genitalis/virology , Herpesvirus 2, Human/pathogenicity , Humans , Interferon Type I/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/virology , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Nucleotidyltransferases/physiology , Signal Transduction
8.
J Immunol ; 198(11): 4205-4209, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28432147

ABSTRACT

TLR agonists are currently being developed and tested as adjuvants in various formulations to optimize the immunogenicity and efficacy of vaccines. The aim of this study was to evaluate the immunostimulatory properties of a novel compound incorporating covalently linked moieties designed to stimulate both TLR2 and TLR7. This dual TLR2/TLR7 agonist induced the maturation of dendritic cells and primed substantial populations of cytolytic and highly polyfunctional effector CD8+ T cells in vitro, and safely potentiated the immunogenic properties of a nanoparticulate Ag in vivo, eliciting humoral responses with a balanced TH1/TH2 profile in mice. Collectively, these data reveal the potential utility of chimeric adjuvants with synergistic activities mediated via TLRs.


Subject(s)
Adjuvants, Immunologic , Immunity, Cellular , Immunity, Humoral , Membrane Glycoproteins/agonists , Membrane Glycoproteins/immunology , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/immunology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/immunology , Animals , Antibody Formation , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Cytokines , Dendritic Cells/immunology , Dendritic Cells/physiology , HIV Core Protein p24/administration & dosage , HIV Core Protein p24/immunology , Ligands , Mice , Mice, Inbred C57BL , Nanoparticles , Recombinant Fusion Proteins/immunology , Vaccination
9.
Proc Natl Acad Sci U S A ; 114(10): 2675-2680, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28223515

ABSTRACT

The advances in subunit vaccines development have intensified the search for potent adjuvants, particularly adjuvants inducing cell-mediated immune responses. Identification of the C-type lectin Mincle as one of the receptors underlying the remarkable immunogenicity of the mycobacterial cell wall, via recognition of trehalose-6,6'-dimycolate (TDM), has opened avenues for the rational design of such molecules. Using a combination of chemical synthesis, biological evaluation, molecular dynamics simulations, and protein mutagenesis, we gained insight into the molecular bases of glycolipid recognition by Mincle. Unexpectedly, the fine structure of the fatty acids was found to play a key role in the binding of a glycolipid to the carbohydrate recognition domain of the lectin. Glucose and mannose esterified at O-6 by a synthetic α-ramified 32-carbon fatty acid showed agonist activity similar to that of TDM, despite their much simpler structure. Moreover, they were seen to stimulate proinflammatory cytokine production in primary human and murine cells in a Mincle-dependent fashion. Finally, they were found to induce strong Th1 and Th17 immune responses in vivo in immunization experiments in mice and conferred protection in a murine model of Mycobacterium tuberculosis infection. Here we describe the rational development of new molecules with powerful adjuvant properties.


Subject(s)
Adjuvants, Immunologic/chemistry , Lectins, C-Type/immunology , Receptors, Immunologic/immunology , Tuberculosis/prevention & control , Vaccines, Subunit/immunology , Adaptive Immunity/drug effects , Adjuvants, Immunologic/therapeutic use , Animals , Cell Wall/drug effects , Cell Wall/immunology , Cord Factors/chemistry , Cord Factors/immunology , Humans , Lectins, C-Type/chemistry , Lectins, C-Type/therapeutic use , Mice , Molecular Dynamics Simulation , Mutagenesis/drug effects , Mycobacterium/immunology , Mycobacterium/pathogenicity , Receptors, Immunologic/chemistry , Tuberculosis/immunology , Tuberculosis/microbiology , Vaccines, Subunit/therapeutic use
11.
Trends Immunol ; 37(9): 573-587, 2016 09.
Article in English | MEDLINE | ID: mdl-27474233

ABSTRACT

Immune adjuvants are components that stimulate, potentiate, or modulate the immune response to an antigen. They are key elements of vaccines in both the prophylactic and therapeutic domains. In the past decade substantial progress in our understanding of innate immunity has paved the way for the design of next-generation adjuvants that stimulate a wide range of receptors. Within the framework of vaccine adjuvant design, this review outlines the interest of targeting endosomal and intracellular receptors to enhance and guide the immune response. We present and compare the molecules as well as potential combinations which are currently in the spotlight. We emphasize how targeting the appropriate receptor can direct immunity towards the appropriate response, such as a cytotoxic or mucosal response.


Subject(s)
Adjuvants, Immunologic , Endosomes/metabolism , Second Messenger Systems/immunology , Signal Transduction , Vaccines/immunology , Animals , Humans , Immunity, Innate , Molecular Targeted Therapy , Vaccination
12.
Biomaterials ; 75: 327-339, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26539801

ABSTRACT

Mucosal surfaces are a major portal of entry for many pathogens that are the cause of infectious diseases. Therefore, effective vaccines that induce a protective immune response at these sites are much needed. However, despite early success with the live attenuated oral polio vaccine over 50 years ago, only a few new mucosal vaccines have been subsequently licensed. Development of new adjuvants, comprising antigen delivery platforms and immunostimulatory molecules, are critical for the successful development of new mucosal vaccines. Among them, biodegradable nanoparticle delivery systems are promising and NOD-like receptors are considered as potential new targets for immunostimulatory molecules. In this work, different NOD1 and NOD2 ligands were encapsulated in polylactic acid (PLA) nanoparticles, coated with HIV-1 gag p24 antigen. We showed that these new formulations are able to induce proliferation of HIV-specific T cells from HIV(+) individuals as well as autophagy. In vivo, these formulations highly enhanced p24-specific systemic and mucosal immune responses in mice not only after mucosal administration but also after immunization via the parenteral route. Our results provide a rational approach for combining nanosized particulate carriers and encapsulated NOD receptor ligands as potent synergistic tools for induction of specific mucosal immunity.


Subject(s)
AIDS Vaccines/immunology , Drug Carriers/chemistry , Immunity , Mucous Membrane/immunology , Nanoparticles/chemistry , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Particle Size , AIDS Vaccines/administration & dosage , Administration, Intranasal , Administration, Oral , Animals , B-Lymphocytes/immunology , Cell Proliferation , Cytokines/metabolism , Dendritic Cells/metabolism , Endocytosis , Female , HIV Infections/immunology , Humans , Immunoglobulin A/metabolism , Immunoglobulin G/metabolism , Injections, Subcutaneous , Lactic Acid/chemistry , Ligands , Mice, Inbred BALB C , Polyesters , Polymers/chemistry , Vaccination , Vagina
13.
J Immunol ; 196(2): 607-13, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26685205

ABSTRACT

Because IgG1 allotypes might have different half-lives, their influence on infliximab (G1m17,1 allotype) pharmacokinetics was investigated in a group of spondyloarthritis patients. Infliximab was found to have a shorter half-life in patients homozygous for the G1m17,1 allotypes than in those carrying the G1m3 with no G1m1 (G1m3,-1) allotype. Because the neonatal FcR (FcRn) is involved in the pharmacokinetics of mAbs, the interaction of different IgG1 allotypes with FcRn was examined using cellular assays and surface plasmon resonance. G1m17,1 mAbs, such as infliximab and rituximab, were shown to bind more efficiently to FcRn and to be transcytosed better than the G1m3,-1 mAb cetuximab, which explains why infliximab is a better competitor for endogenous IgG1 in G1m3,-1 allotype-bearing patients. A set of four allotype variants of adalimumab (G1m17,1; G1m17,-1; G1m3,1; and G1m3,-1) was also tested for its binding to FcRn, revealing that the G1m3,1 variant, not present in commercial mAbs, binds more efficiently to FcRn and is transcytosed better than the other three variants, all of which are found in therapeutic mAbs.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Histocompatibility Antigens Class I/metabolism , Immunoglobulin G/genetics , Infliximab/pharmacokinetics , Receptors, Fc/metabolism , Spondylarthritis/drug therapy , Spondylarthritis/genetics , Female , Flow Cytometry , Humans , Immunoglobulin Allotypes/genetics , Immunoglobulin Allotypes/immunology , Immunoglobulin G/metabolism , Male , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Surface Plasmon Resonance
14.
Mol Cell Biol ; 35(2): 479-95, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25384972

ABSTRACT

Cyclic dinucleotides are important messengers for bacteria and protozoa and are well-characterized immunity alarmins for infected mammalian cells through intracellular binding to STING receptors. We sought to investigate their unknown extracellular effects by adding cyclic dinucleotides to the culture medium of freshly isolated human blood cells in vitro. Here we report that adenosine-containing cyclic dinucleotides induce the selective apoptosis of monocytes through a novel apoptotic pathway. We demonstrate that these compounds are inverse agonist ligands of A2a, a Gαs-coupled adenosine receptor selectively expressed by monocytes. Inhibition of monocyte A2a by these ligands induces apoptosis through a mechanism independent of that of the STING receptors. The blockade of basal (adenosine-free) signaling from A2a inhibits protein kinase A (PKA) activity, thereby recruiting cytosolic p53, which opens the mitochondrial permeability transition pore and impairs mitochondrial respiration, resulting in apoptosis. A2a antagonists and inverse agonist ligands induce apoptosis of human monocytes, while A2a agonists are antiapoptotic. In vivo, we used a mock developing human hematopoietic system through NSG mice transplanted with human CD34(+) cells. Treatment with cyclic di-AMP selectively depleted A2a-expressing monocytes and their precursors via apoptosis. Thus, monocyte recognition of cyclic dinucleotides unravels a novel proapoptotic pathway: the A2a Gαs protein-coupled receptor (GPCR)-driven tonic inhibitory signaling of mitochondrion-induced cell death.


Subject(s)
Adenosine/metabolism , Monocytes/metabolism , Receptor, Adenosine A2A/metabolism , Animals , Cell Death/physiology , Cyclic AMP/metabolism , Humans , Mice, Inbred C57BL , Mitochondria/metabolism , Phosphorylation/physiology , RNA, Small Interfering/genetics , Signal Transduction/physiology
15.
J Immunol ; 193(12): 5781-5, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25392526

ABSTRACT

TLR ligands are critical activators of innate immunity and are being developed as vaccine adjuvants. However, their usefulness in conjunction with NOD-like receptor agonists remains poorly studied. In this study, we evaluated a new ligand that targets both TLR2 and NOD2 receptors. We assessed its ability to enhance dendritic cell maturation in vitro in addition to improving systemic and mucosal immune responses in mice. The chimeric NOD2/TLR2 ligand induced synergistic upregulation of dendritic cell maturation markers, costimulatory molecules, and secretion of proinflammatory cytokines compared with combinations of separate ligands. Furthermore, when coadministered with biodegradable nanoparticles carrying a model Ag, the ligand was able to induce high Ag-specific IgA and IgG titers at both systemic and mucosal sites after parenteral immunizations. These findings point out the potential utility of chimeric molecules TLR/NOD as adjuvants for vaccines to induce systemic and mucosal immune responses.


Subject(s)
Adjuvants, Immunologic/metabolism , Ligands , Nod2 Signaling Adaptor Protein/metabolism , Toll-Like Receptor 2/metabolism , Vaccines/immunology , Acetylmuramyl-Alanyl-Isoglutamine/analogs & derivatives , Acetylmuramyl-Alanyl-Isoglutamine/chemistry , Acetylmuramyl-Alanyl-Isoglutamine/immunology , Adjuvants, Immunologic/chemistry , Animals , Biomarkers/metabolism , Cell Differentiation , Cytokines/biosynthesis , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , HIV Core Protein p24/immunology , Humans , Immunity, Mucosal/immunology , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Lipopeptides/genetics , Lipopeptides/immunology , Mice , Phenotype , Vaccines/genetics
16.
PLoS Biol ; 11(9): e1001658, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24068891

ABSTRACT

Intestinal microfold (M) cells possess a high transcytosis capacity and are able to transport a broad range of materials including particulate antigens, soluble macromolecules, and pathogens from the intestinal lumen to inductive sites of the mucosal immune system. M cells are also the primary pathway for delivery of secretory IgA (SIgA) to the gut-associated lymphoid tissue. However, although the consequences of SIgA uptake by M cells are now well known and described, the mechanisms whereby SIgA is selectively bound and taken up remain poorly understood. Here we first demonstrate that both the Cα1 region and glycosylation, more particularly sialic acid residues, are involved in M cell-mediated reverse transcytosis. Second, we found that SIgA is taken up by M cells via the Dectin-1 receptor, with the possible involvement of Siglec-5 acting as a co-receptor. Third, we establish that transcytosed SIgA is taken up by mucosal CX3CR1⁺ dendritic cells (DCs) via the DC-SIGN receptor. Fourth, we show that mucosal and systemic antibody responses against the HIV p24-SIgA complexes administered orally is strictly dependent on the expression of Dectin-1. Having deciphered the mechanisms leading to specific targeting of SIgA-based Ag complexes paves the way to the use of such a vehicle for mucosal vaccination against various infectious diseases.


Subject(s)
Antigen-Antibody Complex/metabolism , Immunoglobulin A, Secretory/immunology , Intestinal Mucosa/immunology , Lectins, C-Type/metabolism , Transcytosis/immunology , Animals , Antigen-Antibody Complex/immunology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , CHO Cells , CX3C Chemokine Receptor 1 , Caco-2 Cells , Cell Adhesion Molecules/metabolism , Cell Line , Cricetulus , Dendritic Cells/immunology , Dendritic Cells/metabolism , Glycosylation , HIV Core Protein p24/immunology , HeLa Cells , Humans , Immunoglobulin A/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestines/cytology , Lectins/metabolism , Lectins, C-Type/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , N-Acetylneuraminic Acid/chemistry , Receptors, Cell Surface/metabolism , Receptors, Chemokine/metabolism
17.
J Control Release ; 167(1): 60-7, 2013 Apr 10.
Article in English | MEDLINE | ID: mdl-23352911

ABSTRACT

Most successful vaccines are able to induce persistent antibody responses that can last a lifetime. Emerging evidences indicate that activation of immune cells through pattern recognition receptors (PRRs) such as Toll-like receptors (TLRs) or Nod-like receptors (NLRs) may be critical mechanisms. Among PRRs, the use of TLR ligands as adjuvants is already largely described whereas the use of NLRs ligands remains largely unexplored. As activation of intracytoplasmic NLRs is able to induce proinflammatory molecules, the added value of encapsulation of Nod1 and Nod2 receptor ligands into Poly(Lactic Acid) (PLA) biodegradable nanocarriers to modulate their immune properties on human dendritic cells (DCs) maturation has been evaluated. Their ability to induce systemic immune responses in mice was also measured and compared to free ligands and the Alum adjuvant. Nod ligands encapsulated into PLA NPs were efficiently taken up by DCs and subsequently induced a strong up-regulation of maturation markers and the enhancement of proinflammatory cytokine secretion by DCs. Furthermore, co-injection of encapsulated Nod-ligands with PLA particles carrying Gag p24 HIV-1 antigen allowed a 100 fold increase in antibody responses in comparison to Alum. These results suggest that encapsulation of Nod ligands into PLA-NPs could be an effective way to improve vaccine efficiency.


Subject(s)
AIDS Vaccines/administration & dosage , HIV Core Protein p24/immunology , Nanoparticles/administration & dosage , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Adjuvants, Immunologic , Animals , Cell Line , Cells, Cultured , Dendritic Cells/cytology , HEK293 Cells , HIV Core Protein p24/chemistry , Humans , Immunoglobulin G/blood , Lactic Acid/chemistry , Ligands , Mice , Mice, Inbred BALB C , Monocytes , NF-kappa B p50 Subunit/metabolism , Nanoparticles/chemistry , Nod1 Signaling Adaptor Protein/chemistry , Nod2 Signaling Adaptor Protein/chemistry , Polyesters , Polymers/chemistry
18.
Org Biomol Chem ; 4(18): 3489-97, 2006 Sep 21.
Article in English | MEDLINE | ID: mdl-17036144

ABSTRACT

Cellular entry of imaging probes, such as contrast agents for magnetic resonance imaging (MRI), is a key requirement for many molecular imaging studies, particularly imaging intracellular events and cell tracking. Here, we describe the successful development and in vitro analysis of MAGfect, a novel liposome formulation containing a lipidic gadolinium contrast agent for MRI, Gd-DOTA-Chol , designed to enter and label cells. Liposome formulation and cell incubation time were optimised for maximum cellular uptake of the imaging probe in a variety of cell lines. MRI analysis of cells incubated with MAGfect showed them to be highly MRI active. This formulation was examined further for cytotoxicity, cell viability and mechanism of cell labelling. One of the key advantages of using MAGfect as a labelling vehicle arises from its potential for additional functions, such as concomitant drug or gene delivery and fluorescent labelling. The gadolinium liposome was found to be an effective vehicle for transport of plasmid DNA (pDNA) into cells and expression levels were comparable to the commercial transfection agent Trojene.


Subject(s)
Cells/chemistry , Liposomes/metabolism , Magnetic Resonance Spectroscopy/methods , Staining and Labeling/methods , Cells/drug effects , DNA/metabolism , Gadolinium/metabolism , HeLa Cells , Heterocyclic Compounds/chemistry , Humans , Lipids , Organometallic Compounds/chemistry , Plasmids/metabolism , Transfection
19.
J Gene Med ; 8(6): 754-63, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16532508

ABSTRACT

BACKGROUND: To develop more efficient non-viral vectors, we have previously described a novel approach to attach a nuclear localisation signal (NLS) to plasmid DNA, by generating a fusion protein between the tetracycline repressor protein TetR and an SV40 NLS peptide (TetR-NLS). The high affinity of TetR for the DNA sequence tetO is used to bind the NLS to DNA. We have now investigated the ability of this system displaying the SV40 NLS or HIV-1 TAT peptide to enhance nuclear import of a minimised DNA construct more suitable for in vivo gene delivery: a minicircle. METHODS: We have produced a new LacZ minicircle compatible with the TetR system. After transfection of the minicircle in combination with TetR-NLS or TetR-TAT using different transfection agents, we first measured beta-galactosidase activity in vitro. We then used a special delivery technique, in which DOTAP/cholesterol liposomes and DNA/protein complexes are sequentially injected intravenously, to evaluate the activity of this system in vivo. RESULTS: In vitro results showed a 30-fold increase in transfection efficiency of the nuclear-targeted minicircle compared to normal plasmid lipofection. Results on cell cycle arrested cells seem to indicate a different mechanism between the TetR-NLS and TetR-TAT. Finally, we demonstrate a more than 6-fold increase in beta-galactosidase expression in the mouse lung using the minicircle and the TetR-TAT protein. This increase is specific for the peptide sequence and is not observed with the control protein TetR. CONCLUSIONS: Our results indicate that the combination of a minicircle DNA construct with a TetR nuclear-targeting system is able to potentiate gene expression of non-viral vectors.


Subject(s)
Cell Nucleus/metabolism , DNA, Circular/genetics , Genetic Vectors , Transfection/methods , Animals , Female , Gene Expression , Mice , NIH 3T3 Cells , Tetracycline/metabolism , Transgenes , beta-Galactosidase/metabolism
20.
J Med Chem ; 49(1): 349-57, 2006 Jan 12.
Article in English | MEDLINE | ID: mdl-16392819

ABSTRACT

A novel set of dialkynoyl analogues of the cationic, gene delivery lipid DOTAP (1) was synthesized. Structure-activity studies demonstrate that replacement of the cis-double bonds of DOTAP with triple bonds in varying positions alters both the physical properties of the resultant cationic liposome-DNA complexes and their biological functionalities, both in vitro and in vivo. Particularly, in vivo studies demonstrate that pDNA transfection of mouse lung endothelial cells with lead analogue DS(14-yne)TAP (4):cholesterol lipoplexes exhibits double the transfection level with less associated toxicity relative to the well-established DOTAP:cholesterol system. In fact, 4:cholesterol delivers up to 3 times the dose of pDNA in mice than can be tolerated by DOTAP, leading to nearly 3 times greater marker-gene expression. X-ray diffraction studies suggest that lipoplexes containing analogue 4 display increased stability at physiological temperatures. Our results thus suggest that analogue 4 is a potentially strong candidate for the gene therapy of lung tumors.


Subject(s)
DNA/genetics , DNA/pharmacology , Fatty Acids, Monounsaturated/pharmacology , Gene Transfer Techniques , Lung/cytology , Quaternary Ammonium Compounds/pharmacology , Animals , COS Cells , Cell Line, Tumor , Cell Survival/drug effects , Chlorocebus aethiops , Cholesterol/pharmacology , DNA/drug effects , Endothelial Cells/chemistry , Endothelial Cells/drug effects , Fatty Acids, Monounsaturated/chemical synthesis , Fatty Acids, Monounsaturated/chemistry , Female , Genes, Reporter , HeLa Cells , Humans , In Vitro Techniques , Liposomes , Lung/drug effects , Lung/metabolism , Mice , Mice, Inbred BALB C , Molecular Structure , Quaternary Ammonium Compounds/chemical synthesis , Quaternary Ammonium Compounds/chemistry , Structure-Activity Relationship , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL
...