Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer Ther ; 18(6): 1069-1080, 2019 06.
Article in English | MEDLINE | ID: mdl-30962317

ABSTRACT

Recognition of immunoactive oligonucleotides by the immune system, such as Toll-like receptor ligand CpG, leads to increased antibody and T-cell responses. Systemic application often results in unwanted generalized nonantigen-specific activation of the immune system. Nanoparticles are ideal carriers for small and large molecules. Recently, we have demonstrated that calcium phosphate (CaP) nanoparticles functionalized with CpG, and viral antigens are able to induce specific T-cell immunity that protects mice against viral infection and efficiently reactivates the exhausted CD8+ T-cell compartment during chronic retroviral infection. Therefore, CaP nanoparticles are promising vaccine vehicles for therapeutic applications. In this study, we investigated the therapeutic potential use of these nanoparticles in a murine xenograft colorectal cancer model. Therapeutic vaccination with CaP nanoparticles functionalized with CpG and tumor model antigens increased the frequencies of cytotoxic CD8+ T cells in the tumor in a type I interferon-dependent manner. This was accompanied with significantly repressed tumor growth in contrast to the systemic administration of soluble CpG and antigens. Combination therapy of CaP nanoparticles and immune checkpoint blocker against PD-L1 further enhanced the cytotoxic CD8+ T-cell response and eradicated the tumors. Strikingly, vaccination with CaP nanoparticles functionalized with CpG and a primary tumor cell lysate was also sufficient to control the tumor growth. In conclusion, our results represent a translational approach for the use of CaP nanoparticles as a potent cancer vaccine vehicle.


Subject(s)
Adoptive Transfer/methods , Antigens, Neoplasm/chemistry , Cancer Vaccines/therapeutic use , Colonic Neoplasms/therapy , Drug Delivery Systems/methods , Nanoparticles/chemistry , Peptides/chemistry , Allografts , Animals , Antibodies, Monoclonal/pharmacology , Antigens, Viral/genetics , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Calcium Phosphates/chemistry , Cell Line, Tumor , Colonic Neoplasms/pathology , CpG Islands , Disease Models, Animal , Hemagglutinins/genetics , Interferon Type I/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Transgenic , Transfection
2.
Front Immunol ; 9: 614, 2018.
Article in English | MEDLINE | ID: mdl-29740425

ABSTRACT

T cell dysfunction and immunosuppression are characteristic for chronic viral infections and contribute to viral persistence. Overcoming these burdens is the goal of new therapeutic strategies to cure chronic infectious diseases. We recently described that therapeutic vaccination of chronic retrovirus infected mice with a calcium phosphate (CaP) nanoparticle (NP)-based vaccine carrier, functionalized with CpG and viral peptides is able to efficiently reactivate the CD8+ T cell response and improve the eradication of virus infected cells. However, the mechanisms underlying this effect were largely unclear. While type I interferons (IFNs I) are considered to drive T cell exhaustion by persistent immune activation during chronic viral infection, we here describe an indispensable role of IFN I induced by therapeutic vaccination to efficiently reinforce cytotoxic CD8+ T cells (CTL) and improve control of chronic retroviral infection. The induction of IFN I is CpG dependent and leads to significant IFN signaling indicated by upregulation of IFN stimulated genes. By vaccinating chronically retrovirus-infected mice lacking the IFN I receptor (IFNAR-/-) or by blocking IFN I signaling in vivo during therapeutic vaccination, we demonstrate that IFN I signaling is necessary to drive full reactivation of CTLs. Surprisingly, we also identified an impaired suppressive capability of regulatory T cells in the presence of IFNα, which implicates an important role for vaccine-induced IFNα in the regulation of the T cell response during chronic retroviral infection. Our data suggest that inducing IFN I signaling in conjunction with the presentation of viral antigens can reactivate immune functions and reduce viral loads in chronic infections. Therefore, we propose CaP NPs as potential therapeutic tool to treat chronic infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interferon Type I/metabolism , Nanoparticles/administration & dosage , Retroviridae Infections/immunology , Retroviridae/physiology , Viral Vaccines/immunology , Animals , Calcium Phosphates/chemistry , Cell Line , Cytotoxicity, Immunologic , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Nanoparticles/chemistry , Receptor, Interferon alpha-beta/genetics , Signal Transduction , Vaccination
3.
PLoS One ; 12(6): e0178260, 2017.
Article in English | MEDLINE | ID: mdl-28586345

ABSTRACT

Nanoparticles can be used as carriers to transport biomolecules like proteins and synthetic molecules across the cell membrane because many molecules are not able to cross the cell membrane on their own. The uptake of nanoparticles together with their cargo typically occurs via endocytosis, raising concerns about the possible degradation of the cargo in the endolysosomal system. As the tracking of a dye-labelled protein during cellular uptake and processing is not indicative of the presence of the protein itself but only for the fluorescent label, a label-free tracking was performed with the red-fluorescing model protein R-phycoerythrin (R-PE). Four different eukaryotic cell lines were investigated: HeLa, HEK293T, MG-63, and MC3T3. Alone, the protein was not taken up by any cell line; only with the help of calcium phosphate nanoparticles, an efficient uptake occurred. After the uptake into HeLa cells, the protein was found in early endosomes (shown by the marker EEA1) and lysosomes (shown by the marker Lamp1). There, it was still intact and functional (i.e. properly folded) as its red fluorescence was detected. However, a few hours after the uptake, proteolysis started as indicated by the decreasing red fluorescence intensity in the case of HeLa and MC3T3 cells. 12 h after the uptake, the protein was almost completely degraded in HeLa cells and MC3T3 cells. In HEK293T cells and MG-63 cells, no degradation of the protein was observed. In the presence of Bafilomycin A1, an inhibitor of acidification and protein degradation in lysosomes, the fluorescence of R-PE remained intact over the whole observation period in the four cell lines. These results indicate that despite an efficient nanoparticle-mediated uptake of proteins by cells, a rapid endolysosomal degradation may prevent the desired (e.g. therapeutic) effect of a protein inside a cell.


Subject(s)
Drug Delivery Systems , Nanoparticles/administration & dosage , Phycoerythrin/administration & dosage , Proteolysis/drug effects , Biological Transport/drug effects , Calcium Phosphates/administration & dosage , Calcium Phosphates/chemistry , Endocytosis , Endosomes/drug effects , HEK293 Cells , HeLa Cells , Humans , Lysosomes/drug effects , Macrolides/administration & dosage , Nanoparticles/chemistry , Phycoerythrin/chemistry
4.
Beilstein J Nanotechnol ; 8: 381-393, 2017.
Article in English | MEDLINE | ID: mdl-28326227

ABSTRACT

The efficient intracellular delivery of (bio)molecules into living cells remains a challenge in biomedicine. Many biomolecules and synthetic drugs are not able to cross the cell membrane, which is a problem if an intracellular mode of action is desired, for example, with a nuclear receptor. Calcium phosphate nanoparticles can serve as carriers for small and large biomolecules as well as for synthetic compounds. The nanoparticles were prepared and colloidally stabilized with either polyethyleneimine (PEI; cationic nanoparticles) or carboxymethyl cellulose (CMC; anionic nanoparticles) and loaded with defined amounts of the fluorescently labelled proteins HTRA1, HTRA2, and BSA. The nanoparticles were purified by ultracentrifugation and characterized by dynamic light scattering and scanning electron microscopy. Various cell types (HeLa, MG-63, THP-1, and hMSC) were incubated with fluorescently labelled proteins alone or with protein-loaded cationic and anionic nanoparticles. The cellular uptake was followed by light and fluorescence microscopy, confocal laser scanning microscopy (CLSM), and flow cytometry. All proteins were readily transported into the cells by cationic calcium phosphate nanoparticles. Notably, only HTRA1 was able to penetrate the cell membrane of MG-63 cells in dissolved form. However, the application of endocytosis inhibitors revealed that the uptake pathway was different for dissolved HTRA1 and HTRA1-loaded nanoparticles.

5.
Genes Cells ; 21(7): 682-95, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27238217

ABSTRACT

This study aimed to fabricate a growth factor-releasing biodegradable scaffold for tissue regeneration. We prepared multishell calcium phosphate (CaP) nanoparticles functionalized with DNA, polyethyleneimine (PEI), protamine and octa-arginine (R8) and compared their respective transfection activity and cell viability measures using human mesenchymal stem cells. DNA-protamine complexes improved the transfection efficiency of CaP nanoparticles with the exception of those functionalized with R8. These complexes also greatly reduced the cytotoxicity of PEI. In addition, we also fabricated DNA-protamine-functionalized CaP nanoparticle-loaded nano-hydroxyapatite-collagen scaffolds and investigated their gene transfection efficiencies. These experiments showed that the scaffolds were associated with moderate hMSC cell viability and were capable of releasing the BMP-2 protein into hMSCs following gene transfection. In particular, the scaffold loaded with protamine-containing CaP nanoparticles showed the highest cell viability and transfection efficiency in hMSCs; thus, it might be suitable to serve as an efficient growth factor-releasing scaffold.


Subject(s)
Collagen/therapeutic use , Durapatite/therapeutic use , Gene Transfer Techniques , Mesenchymal Stem Cells/metabolism , Nanoparticles/chemistry , Regeneration , Bone Marrow Cells/drug effects , Bone Morphogenetic Protein 2/genetics , Calcium Phosphates/chemistry , Cell Survival/drug effects , Collagen/chemistry , Durapatite/chemistry , Green Fluorescent Proteins/genetics , Humans , Mesenchymal Stem Cells/drug effects , Nanoparticles/therapeutic use , Plasmids/genetics , Tissue Scaffolds , Transfection
6.
Retrovirology ; 13: 24, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27076190

ABSTRACT

BACKGROUND: Regulatory T cells (Tregs) have been shown to limit anti-viral immunity during chronic retroviral infection and to restrict vaccine-induced T cell responses. The objective of the study was to assess whether a combinational therapy of nanoparticle-based therapeutic vaccination and concomitant transient ablation of Tregs augments anti-viral immunity and improves virus control in chronically retrovirus-infected mice. Therefore, chronically Friend retrovirus (FV)-infected mice were immunized with calcium phosphate (CaP) nanoparticles functionalized with TLR9 ligand CpG and CD8(+) or CD4(+) T cell epitope peptides (GagL85-93 or Env gp70123-141) of FV. In addition, Tregs were ablated during the immunization process. Reactivation of CD4(+) and CD8(+) effector T cells was analysed and the viral loads were determined. RESULTS: Therapeutic vaccination of chronically FV-infected mice with functionalized CaP nanoparticles transiently reactivated cytotoxic CD8(+) T cells and significantly reduced the viral loads. Transient ablation of Tregs during nanoparticle-based therapeutic vaccination strongly enhanced anti-viral immunity and further decreased viral burden. CONCLUSION: Our data illustrate a crucial role for CD4(+) Foxp3(+) Tregs in the suppression of anti-viral T cell responses during therapeutic vaccination against chronic retroviral infection. Thus, the combination of transient Treg ablation and therapeutic nanoparticle-based vaccination confers robust and sustained anti-viral immunity.


Subject(s)
Leukemia, Experimental/therapy , Leukocyte Reduction Procedures , Nanoparticles/administration & dosage , Retroviridae Infections/therapy , T-Lymphocytes, Regulatory/immunology , Tumor Virus Infections/therapy , Viral Vaccines/administration & dosage , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Combined Modality Therapy/methods , Friend murine leukemia virus/immunology , Leukemia, Experimental/immunology , Mice, Inbred C57BL , Retroviridae Infections/immunology , Treatment Outcome , Tumor Virus Infections/immunology
7.
Nanomedicine ; 10(8): 1787-98, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25014891

ABSTRACT

Retroviral infections e.g. HIV still represent a unique burden in the field of vaccine research. A common challenge in vaccine design is to find formulations that create appropriate immune responses to protect against and/or control the given pathogen. Nanoparticles have been considered to be ideal vaccination vehicles that mimic invading pathogens. In this study, we present biodegradable calcium phosphate (CaP) nanoparticles, functionalized with CpG and retroviral T cell epitopes of Friend virus (FV) as excellent vaccine delivery system. CaP nanoparticles strongly increased antigen delivery to antigen-presenting cells to elicit a highly efficient T cell-mediated immune response against retroviral FV infection. Moreover, single-shot immunization of chronically FV-infected mice with functionalized CaP nanoparticles efficiently reactivated effector T cells which led to a significant decrease in viral loads. Thus, our findings clearly indicate that a nanoparticle-based peptide immunization is a promising approach to improve antiretroviral vaccination. FROM THE CLINICAL EDITOR: In this study, biodegradable calcium phosphate nanoparticles were used as a vaccine delivery system after functionalization with CpG and Friend virus-derived T-cell epitopes. This vaccination strategy resulted in increased T-cell mediated immune response even in chronically infected mice, providing a promising approach to the development of clinically useful antiretroviral vaccination strategies.


Subject(s)
Immunity, Cellular/immunology , Nanoparticles/chemistry , Retroviridae Infections/immunology , Retroviridae Infections/prevention & control , Vaccines, Subunit/chemistry , Vaccines, Subunit/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Flow Cytometry , Mice
8.
J Immunol ; 190(12): 6221-9, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23667109

ABSTRACT

The ability of vaccines to induce T cell responses is crucial for preventing diseases caused by viruses or bacteria. Nanoparticles (NPs) are considered an efficient tool for inducing potent immune responses. In this study, we describe a novel vaccination approach with biodegradable calcium phosphate (CaP) NPs that serve as carrier of immunoactive TLR9 ligand (CpG) combined with a viral Ag from the influenza A virus hemagglutinin. Functionalized CaP NPs were efficiently taken up by dendritic cells in vivo and elicited a potent T cell-mediated immune response in immunized mice with high numbers of IFN-γ-producing CD4(+) and CD8(+) effector T cells. Most importantly, both i.p. and intranasal immunization with these NPs offered protection in a mouse model of influenza virus infection. This study demonstrates the great potential of CaP NPs as a novel vaccination tool that offers substantial flexibility for several infection models.


Subject(s)
Immunity, Cellular/immunology , Influenza Vaccines/immunology , Nanoparticles , Vaccination/methods , Administration, Intranasal , Animals , Calcium Phosphates/pharmacology , Flow Cytometry , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/chemistry , Injections, Intraperitoneal , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , T-Lymphocytes/immunology
9.
Colloids Surf B Biointerfaces ; 87(1): 146-50, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21640565

ABSTRACT

Exosomes from three different cell types (HEK 293T, ECFC, MSC) were characterised by scanning electron microscopy (SEM), dynamic light scattering (DLS) and nanoparticle tracking analysis (NTA). The diameter was around 110 nm for the three cell types. The stability of exosomes was examined during storage at -20°C, 4°C, and 37°C. The size of the exosomes decreased at 4°C and 37°C, indicating a structural change or degradation. Multiple freezing to -20°C and thawing did not affect the exosome size. Multiple ultracentrifugation also did not change the exosome size.


Subject(s)
Endothelial Cells/cytology , Exosomes/ultrastructure , Mesenchymal Stem Cells/cytology , Nanoparticles/ultrastructure , Nanotechnology/methods , Colloids , HEK293 Cells , Humans , Microscopy, Electron, Scanning , Particle Size , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...