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










Database
Language
Publication year range
2.
Biomaterials ; 108: 1-12, 2016 11.
Article in English | MEDLINE | ID: mdl-27614817

ABSTRACT

Targeting antigen combined with adjuvants to hepatic antigen-presenting cells (APCs) is essential for the induction of intrahepatic T cellular immunity controlling and resolving viral infections of the liver. Intravenous injection of antigen-loaded nanoparticles is a promising approach for the delivery of antigens to liver APCs. Accordingly, polymeric nanocapsules (NCs) synthesized exclusively of hepatitis C virus non-structural protein 5A (NS5A) and the adjuvant monophosphoryl lipid A (MPLA) adsorbed to the nanocapsule surface were developed. Aim of the present study was the evaluation of the in vitro and in vivo behavior of MPLA-functionalized NS5A-NCs regarding the interaction with liver dendritic cells (DCs) and the potential to induce intrahepatic immune responses in a mouse model. Maturation of DCs was significantly increased by application of NS5A+MPLA-NCs compared to non-functionalized NS5A-NCs promoting a vigorous expression of CD40, CD80, CD86 and a strong secretion of the Th1-related cytokine IL-12. NS5A-NCs were preferentially deposited in DCs and Kupffer cells residing in the liver after intravenous administration. Immunization with NS5A-NCs induced intrahepatic antigen-specific CD4(+) T cellular immune responses determined by the secretion of IFNγ and IL-2. Furthermore, supplementation with MPLA induced significant levels of NS5A-specific antibodies. The application of polymeric nanocapsules synthesized exclusively out of antigen avoids the risk of unintended side effects caused by additional carrier substances. Functionalization with adjuvants like MPLA and the efficient targeting to liver-resident APCs inherits the potential for application of antigen nanocapsules in further vaccination approaches against pathogens affecting the liver.


Subject(s)
Hepatitis C/immunology , Immunity, Innate/immunology , Lipid A/analogs & derivatives , Liver/immunology , Nanocapsules/administration & dosage , Viral Nonstructural Proteins/administration & dosage , Viral Nonstructural Proteins/immunology , Animals , Cytokines/immunology , Female , Histocompatibility Antigens Class II/immunology , Immunity, Innate/drug effects , Immunization/methods , Lipid A/administration & dosage , Lipid A/immunology , Liver/drug effects , Mice , Mice, Inbred C57BL , Nanocapsules/chemistry , Nanocapsules/ultrastructure , Particle Size , Polymers
3.
Nanomedicine ; 12(8): 2383-2394, 2016 11.
Article in English | MEDLINE | ID: mdl-27516081

ABSTRACT

Chronic hepatitis B virus (HBV) infection is the most prevalent serious liver infection in the world. A frequent route of infection represents mother-to-child transmission. Efficient control of HBV replication depends on antigen-specific cellular immune response mediated by dendritic cells (DCs). Aim of the present study was to evaluate optimized adjuvant combinations, efficiently maturing monocyte-derived neonatal and adult dendritic cells (moDCs). In addition, the potential of polymeric HBsAg-nanocapsules (HBsAg-NCs) was investigated regarding up-take by moDCs and the subsequent induction of specific T cell responses in a human co-culture model. Simultaneous stimulation of moDCs with MPLA and IFNγ induced up-regulation of CD80 and HLA-DR along with vigorous secretion of IL-12p70. MPLA-coating of HBsAg-NCs promoted NCs-uptake by moDCs. Finally, MPLA-HBsAg-NCs-pulsed moDCs with IFNγ increased T cell proliferation and induced antigen-specific IFNγ release by T cells. The herein presented vaccine approach provides a rational for neonatal and therapeutic immunization strategies against HBV.


Subject(s)
Fetal Blood , Hepatitis B Surface Antigens , Hepatitis B Vaccines , Nanocapsules , T-Lymphocytes , Antigens, Surface , Dendritic Cells , Hepatitis B/prevention & control , Hepatitis B virus , Humans
4.
Vaccine ; 33(7): 838-46, 2015 Feb 11.
Article in English | MEDLINE | ID: mdl-25573037

ABSTRACT

Enhancing delivery of antigens to dendritic cells (DCs) is essential for the induction of vigorous antigen-specific cellular immune responses. Aim of the present study was to evaluate the properties of hydroxyethyl starch nanocapsules (HES-NCs) functionalized with anti-CD40, anti-DEC205, interferon-γ (IFNγ) and/or monophosphoryl lipid A (MPLA) with respect to the overall uptake, the released cytokine profile, and the influence on phenotypic maturation of human monocyte-derived DCs using flow cytometry, confocal microscopy and enzyme-linked immunosorbent assays. NC uptake by DCs was significantly enhanced by functionalizing NCs with anti-CD40 or MPLA. With respect to the cytokine profile and the maturation status, coating with MPLA evoked a strong Th1-type cytokine response and significantly increased CD80 and CD83 expression on DCs, contrasting the moderate effects of MPLA in solution. Notably, an at least 20 fold higher amount of MPLA in solution was needed compared to the dosage of MPLA attached to HES-NCs in order to induce comparable effects, evidencing the intense dose-sparing potential of particle-bound MPLA. Reducing the amount of the vaccine adjuvant MPLA, while maintaining or even surpassing the effects on human DCs, reveals the potential of HES-NCs as a promising carrier system for the simultaneous delivery of antigen along with compounds promoting a Th1-prone cellular immune response.


Subject(s)
Dendritic Cells/metabolism , Hydroxyethyl Starch Derivatives/chemistry , Lipid A/analogs & derivatives , Nanocapsules/chemistry , Nanomedicine/methods , Adjuvants, Immunologic , Cells, Cultured , Humans , Interleukin-12/metabolism , Lipid A/chemistry , Microscopy, Confocal , Nanocapsules/administration & dosage , Th1 Cells/metabolism , Toll-Like Receptor 4/metabolism
5.
Biomacromolecules ; 15(7): 2378-88, 2014 Jul 14.
Article in English | MEDLINE | ID: mdl-24901387

ABSTRACT

A broad spectrum of infectious liver diseases emphasizes the need of microparticles for targeted delivery of immunomodulatory substances to the liver. Microcapsules (MCs) are particularly attractive for innovative drug and vaccine formulations, enabling the combination of antigen, drugs, and adjuvants. The present study aimed to develop microcapsules characterized by an enhanced liver deposition and accelerated uptake by nonparenchymal liver cells (NPCs). Initially, two formulations of biodegradable microcapsules were synthesized from either hydroxyethyl starch (HES) or mannose. Notably, HES-MCs accumulated primarily in the liver, while mannose particles displayed a lung preference. Functionalization of HES-MCs with anti-CD40, anti-DEC205, and/or monophosphoryl lipid A (MPLA) enhanced uptake of MCs by nonparenchymal liver cells in vitro. In contrast, only MPLA-coated HES-MCs promoted significantly the in vivo uptake by NPCs. Finally, HES-MCs equipped with MPLA, anti-CD40, and anti-DEC205 induced the secretion of TNF-α, IL-6 by Kupffer cells (KCs), and IFN-γ and IL-12p70 by liver dendritic cells (DCs). The enhanced uptake and activation of KCs by MPLA-HES-MCs is a promising approach to prevent or treat infection, since KCs are exploited as an entry gate in various infectious diseases, such as malaria. In parallel, loading and activating liver DCs, usually prone to tolerance, bears the potential to induce antigen specific, intrahepatic immune responses necessary to prevent and treat infections affecting the liver.


Subject(s)
Drug Delivery Systems/methods , Lipid A/analogs & derivatives , Liver/drug effects , Animals , Antigens, CD/metabolism , CD40 Antigens/metabolism , Capsules/chemistry , Capsules/pharmacokinetics , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Female , Interferon-gamma/metabolism , Interleukin-6/metabolism , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Lectins, C-Type/metabolism , Lipid A/chemistry , Lipid A/pharmacology , Liver/cytology , Liver Diseases/drug therapy , Mice , Mice, Inbred C57BL , Minor Histocompatibility Antigens , Nanoparticles/chemistry , Phagocytosis/drug effects , Receptors, Cell Surface/metabolism , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
6.
Nanomedicine ; 9(8): 1223-34, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23727100

ABSTRACT

Dexamethasone (DXM) is a synthetic glucocorticoid with anti-inflammatory properties. Targeted delivery of dexamethasone to inflammatory cells, e.g. macrophages and Kupffer cells represents a promising approach to minimize side effects. The aim of the present study was to induce a targeted transport of novel DXM-based biodegradable nanocapsules to phagocytic cells. Nanocapsules (NCs) consisting of a hydroxyethylated glucose polymer (hydroxyethyl starch, HES) shell with encapsulated DXM and NCs synthesized exclusively in inverse miniemulsion out of DXM were investigated. Non-parenchymal murine liver cells served as target cells. HES-DXM NCs were predominantly incorporated by Kupffer cells (KCs). In contrast, DXM NCs were phagocytized by KCs and endothelial cells. The release of the NC-content was confirmed by incorporation of CellTracker™ into the NCs. Uptake of DXM NCs by Kupffer cells reduced significantly the release of inflammatory cytokines in response to LPS stimulation. Importantly, the DXM NCs consisting exclusively out of a dexamethasone shell offer the potential to serve as carriers for additional therapeutics. FROM THE CLINICAL EDITOR: In this paper, nanocapsule-based targeted delivery of dexamethasone to inflammatory cells is presented as a promising approach to minimize side effects and increase efficacy of this anti-inflammatory clinically used corticosteroid.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Dexamethasone/administration & dosage , Drug Delivery Systems , Hydroxyethyl Starch Derivatives/chemistry , Kupffer Cells/drug effects , Nanocapsules/chemistry , Animals , Anti-Inflammatory Agents/pharmacology , Cells, Cultured , Cytokines/analysis , Cytokines/immunology , Dexamethasone/pharmacology , Female , Kupffer Cells/immunology , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...