Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 138
Filter
Add more filters

Publication year range
1.
Biomacromolecules ; 21(1): 214-229, 2020 01 13.
Article in English | MEDLINE | ID: mdl-31686502

ABSTRACT

The development of in situ-gelling hydrogels that can enable prolonged protein release is increasingly important due to the emergence of a growing number of protein-based therapeutics. Herein, we describe a high-throughput strategy to fabricate, characterize, and subsequently optimize hydrazone-cross-linked in situ-gelling hydrogels for protein delivery. Hydrogels are fabricated using an automated high-throughput robot to mix a variety of thermoresponsive, nonthermoresponsive, charged, neutral, naturally sourced, and synthetic polymers functionalized with hydrazide or aldehyde groups, generating in situ-gelling hydrogels with well-defined compositions within a 96-well plate. High-throughput characterization strategies are subsequently developed to enable on-plate analysis of hydrogel swelling, mechanics, degradation, transparency, and protein (ovalbumin) release kinetics that yield results consistent with those collected using traditional bulk hydrogel analysis techniques. Dynamic regression and latent variable modeling are then applied to fit performance statistics to the collected data set; subsequently, numerical optimization is used to identify mixtures of precursor polymers that exhibit targeted combinations of minimal burst release, maximum total protein release, minimum release rate, and maximum transparency (the latter of particular relevance for ophthalmic protein delivery applications). Given the rapid throughput of the protocols developed (i.e., 126 hydrogels can be synthesized and screened in quadruplicate within hours), this approach offers particular promise for accelerating the identification of injectable hydrogel compositions relevant for both protein delivery as well as other biomedical applications for which clearly predefined materials properties are required.


Subject(s)
Hydrogels/administration & dosage , Hydrogels/chemical synthesis , Proteins/administration & dosage , Acrylic Resins/chemistry , Chitosan/chemistry , Dextrans/chemistry , Drug Delivery Systems/methods , Hydrogels/pharmacokinetics , Injections , Kinetics , Models, Theoretical , Ovalbumin/administration & dosage , Ovalbumin/pharmacokinetics , Polyethylene Glycols/chemistry , Polymers/chemistry , Proteins/pharmacokinetics , Robotics/methods , Temperature
2.
Biomacromolecules ; 21(2): 621-629, 2020 02 10.
Article in English | MEDLINE | ID: mdl-31800235

ABSTRACT

Although current vaccine technology induces sufficient antibody responses to prophylactically ward off viral infections, an anticancer vaccine that directs the patient's immune system to directly fight extant malignant cells will require inducing Th1 and cytotoxic T lymphocyte responses in addition to antibody-mediated activities. Thus, new mechanisms are necessary to deliver antigen to cells in the lymphatic system that will induce these responses. To this end, we have developed a cholesterol-bearing pullulan (CHP) self-assembly nanogel of less than 100 nm, which we have now further modified to be anionic by carboxyl group substitution. Overall, the nanogel-protected antigens during transport to the lymphatic system and converting the vehicle to an anionic charge improved interactions with antigen-presenting cells. We further show that these modified nanogels are a more efficient system for delivering antigen to antigen-presenting cells, particularly langerin-expressing cells, and that this induced significant adaptive immunity. Therefore, we think that this technology could be used to improve anticancer immunotherapies.


Subject(s)
Adaptive Immunity/drug effects , Antigen-Presenting Cells/immunology , Vaccines/administration & dosage , Vaccines/chemistry , Animals , Antigen-Presenting Cells/drug effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Drug Delivery Systems , Epitopes , Female , Immunoglobulin G/blood , Lymph Nodes/drug effects , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Nanogels/chemistry , Ovalbumin/administration & dosage , Ovalbumin/pharmacokinetics , Polysaccharides/chemistry , RAW 264.7 Cells , Vaccines/pharmacology
3.
Mol Pharm ; 15(3): 975-984, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29359945

ABSTRACT

In immunotherapy, induction of potent cellular immunity by vaccination is essential to treat intracellular infectious diseases and tumors. In this work, we designed a new synthetic peptide carrier, Cys-Trp-Trp-Arg8-Cys-Arg8-Cys-Arg8-Cys, for vaccine delivery by integrating a redox-responsive disulfide bond cross-linking and cell-penetrating peptide arginine octamer. The carrier peptide bound to the antigen protein ovalbumin (OVA) via electrostatic self-assembly to form peptide/OVA nanocomposites. Then, the spontaneous oxidization of the thiols of the cysteine residues induced interpeptide disulfide bond cross-linking to construct denser peptide/OVA condensates. The cell-penetrating peptides incorporated in the carrier peptide could increase antigen uptake by antigen presenting cells. After being internalized by antigen presenting cells, the antigen could be rapidly released in cytoplasm along with degradation of the disulfide bonds by intracellular glutathione, which could promote potent CD8+ T cell immunity. The cross-linked peptide/OVA condensates were used for subcutaneous vaccination. The results showed that the peptide carrier mediated potent antigen-specific immune response by significantly increasing IgG titer; splenocyte proliferation; the secretion level of cytokines INF-γ, IL-12, IL-4, and IL-10; immune memory function, and the activation and maturation of dendritic cells. From the results, the low-molecular weight vaccine-condensing peptide with definite chemical composition could be developed as a novel class of vaccine delivery systems.


Subject(s)
Cell-Penetrating Peptides/chemistry , Drug Delivery Systems/methods , Ovalbumin/administration & dosage , Vaccination/methods , Vaccines, Subunit/administration & dosage , Animals , Antigens/administration & dosage , Antigens/immunology , Cell Line , Cell Membrane Permeability/drug effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Immunity, Cellular/drug effects , Immunologic Memory/drug effects , Injections, Subcutaneous , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Animal , Nanoparticles/chemistry , Ovalbumin/immunology , Ovalbumin/pharmacokinetics , Oxidation-Reduction , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacokinetics
4.
Pharm Res ; 35(2): 36, 2018 Jan 16.
Article in English | MEDLINE | ID: mdl-29368249

ABSTRACT

PURPOSE: To investigate the sustained ocular delivery of small and large drug molecules from photocrosslinked poly(ethylene glycol) diacrylate (PEGDA) implants with varying pore forming agents. METHODS: Triamcinolone acetonide and ovalbumin loaded photocrosslinked PEGDA implants, with or without pore-forming agents, were fabricated and characterised for chemical, mechanical, swelling, network parameters, as well as drug release and biocompatibility. HPLC-based analytical methods were employed for analysis of two molecules; ELISA was used to demonstrate bioactivity of ovalbumin. RESULTS: Regardless of PEGDA molecular weight or pore former composition all implants loaded with triamcinolone acetonide released significantly faster than those loaded with ovalbumin. Higher molecular weight PEGDA systems (700 Da) resulted in faster drug release of triamcinolone acetonide than their 250 Da counterpart. All ovalbumin released over the 56-day time period was found to be bioactive. Increasing PEGDA molecular weight resulted in increased system swelling, decreased crosslink density (Ve), increased polymer-water interaction parameter (χ), increased average molecular weight between crosslinks (Mc) and increased mesh size (ε). SEM studies showed the porosity of implants increased with increasing PEGDA molecular weight. Biocompatibility showed both PEGDA molecular weight implants were non-toxic when exposed to retinal epithelial cells over a 7-day period. CONCLUSION: Photocrosslinked PEGDA implant based systems are capable of controlled drug release of both small and large drug molecules through adaptations in the polymer system network. We are currently continuing evaluation of these systems as potential sustained drug delivery devices.


Subject(s)
Biological Products/pharmacokinetics , Drug Delivery Systems/methods , Drug Liberation , Glucocorticoids/pharmacokinetics , Polyethylene Glycols/chemical synthesis , Administration, Ophthalmic , Biological Products/administration & dosage , Cell Line , Diabetic Retinopathy/drug therapy , Drug Compounding/methods , Drug Implants , Glucocorticoids/administration & dosage , Humans , Macular Degeneration/drug therapy , Materials Testing , Molecular Weight , Ovalbumin/administration & dosage , Ovalbumin/pharmacokinetics , Porosity , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/drug effects , Triamcinolone Acetonide/administration & dosage , Triamcinolone Acetonide/pharmacokinetics
5.
AAPS PharmSciTech ; 18(7): 2418-2426, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28160208

ABSTRACT

The purpose of this study was to evaluate the use of different types of microneedles and doses of ovalbumin antigen for in vitro skin permeation and in vivo immunization. In vitro skin permeation experiments and confocal laser scanning microscopy revealed that hollow microneedles had a superior enhancing effect on skin permeation compared with a solid microneedle patch and untreated skin by efficiently delivering ovalbumin-fluorescein conjugate into the deep skin layers. The flux and cumulative amount of ovalbumin-fluorescein conjugate at 8 h after administering with various conditions could be ranked as follows: hollow MN; high dose > medium dose > low dose > MN patch; high dose > medium dose > low dose > untreated skin; high dose > medium dose > low dose > without ovalbumin-fluorescein conjugate. As the dose of ovalbumin-fluorescein conjugate was increased to 500 µg, the antigen accumulated in the skin to a greater extent, as evidenced by the increasing green fluorescence intensity. When the hollow microneedle was used for the delivery of ovalbumin into the skin of mice, it was capable of inducing a stronger immunoglobulin G immune response than conventional subcutaneous injection at the same antigen dose. Immunoglobulin G levels in the hollow MN group were 5.7, 11.6, and 13.3 times higher than those of the subcutaneous injection group for low, medium, and high doses, respectively. Furthermore, the mice immunized using the hollow microneedle showed no signs of skin infection or pinpoint bleeding. The results suggest that the hollow MN is an efficient device for delivering the optimal dose of antigen via the skin for successful immunization.


Subject(s)
Ovalbumin/administration & dosage , Skin/metabolism , Administration, Cutaneous , Animals , Antigens/administration & dosage , Antigens/immunology , Immunization , Immunoglobulin G/biosynthesis , Mice , Needles , Ovalbumin/immunology , Ovalbumin/pharmacokinetics
6.
AAPS PharmSciTech ; 18(4): 1070-1083, 2017 May.
Article in English | MEDLINE | ID: mdl-27975192

ABSTRACT

The control over the crystallinity of chitosan and chitosan/ovalbumin films can be achieved via an appropriate balance of the hydrophilic/hydrophobic interactions during the film formation process, which then controls the release kinetics of ovalbumin. Chitosan films were prepared by solvent casting. The presence of the anhydrous allomorph can be viewed as a probe of the hydrophobic conditions at the neutralization step. The semicrystalline structure, the swelling behavior of the films, the protein/chitosan interactions, and the release behavior of the films were impacted by the DA and the film processing parameters. At low DAs, the chitosan films neutralized in the solid state corresponded to the most hydrophobic environment, inducing the crystallization of the anhydrous allomorph with and without protein. The most hydrophilic conditions, leading to the hydrated allomorph, corresponded to non-neutralized films for the highest DAs. For the non-neutralized chitosan acetate (amorphous) films, the swelling increased when the DA decreased, whereas for the neutralized chitosan films, the swelling decreased. The in vitro release of ovalbumin (model protein) from chitosan films was controlled by their swelling behavior. For fast swelling films (DA = 45%), a burst effect was observed. On the contrary, a lag time was evidenced for DA = 2.5% with a limited release of the protein. Furthermore, by blending chitosans (DA = 2.5% and 45%), the release behavior was improved by reducing the burst effect and the lag time. The secondary structure of ovalbumin was partially maintained in the solid state, and the ovalbumin was released under its native form.


Subject(s)
Chitosan , Hydrophobic and Hydrophilic Interactions , Ovalbumin , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Chitosan/chemistry , Chitosan/pharmacology , Crystallization , Drug Delivery Systems , Ovalbumin/chemistry , Ovalbumin/pharmacokinetics
7.
Pharm Res ; 32(1): 22-36, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24970313

ABSTRACT

PURPOSE: Antigens were conjugated on the surface of N-trimethylaminoethylmethacrylate chitosan (TMC) nanoparticles to induce systemic and mucosal immune responses after nasal immunization. METHODS: TMC was synthesized by free radical polymerization and blank nanoparticles were prepared by ionic crosslinking of TMC and sodium tripolyphosphate. The model antigen (ovalbumin) was conjugated on the surface of blank nanoparticles (OVA-NP) through thioester bond formation. The cellular uptake of OVA-NP was investigated in Raw 264.7 macrophages and biodistribution of antigens was studied by the radioiodine labeling method. The immunological effects were evaluated by nasal administration of OVA-NP to Balb/C mice. The transport mechanism and nasal toxicity of OVA-NP were studied in rats. RESULTS: The cellular uptake of OVA-NP was significantly higher than that of ovalbumin-encapsulated nanoparticles (NPe) after 30 min. Nasally administered OVA-NP showed higher transport of antigens to cervical lymph nodes with higher targeting efficiency than all other groups. Compared with NPe, OVA-NP induced much higher levels of systemic and mucosal immune responses in Balb/C mice after three nasal immunizations. Ex vivo culturing of nasopharynx-associated lymphoid tissue (NALT) confirmed its participation in nasal immunization. The transport mechanism study revealed that OVA-NP can be transported across the nasal epithelium through glands and may be taken up in NALT through M cells. OVA-NP did not induce obvious toxicity to nasal mucosa or hemolysis in animals. CONCLUSION: The present study demonstrated that the conjugation of TMC nanoparticles with antigens is an effective strategy for nasal vaccination.


Subject(s)
Adjuvants, Immunologic/chemistry , Antibody Formation/immunology , Antigens/administration & dosage , Chitosan/analogs & derivatives , Drug Carriers/chemistry , Methacrylates/chemistry , Nanoparticles/chemistry , Ovalbumin/administration & dosage , Adjuvants, Immunologic/pharmacokinetics , Adjuvants, Immunologic/toxicity , Administration, Intranasal , Animals , Antigens/immunology , Cell Line , Chitosan/chemistry , Chitosan/pharmacokinetics , Chitosan/toxicity , Drug Carriers/pharmacokinetics , Drug Carriers/toxicity , Drug Compounding , Female , Immunoglobulin A/blood , Immunoglobulin G/blood , Lymph Nodes/immunology , Lymph Nodes/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Methacrylates/pharmacokinetics , Methacrylates/toxicity , Mice, Inbred BALB C , Nasal Mucosa/immunology , Nasal Mucosa/metabolism , Nasopharynx/immunology , Nasopharynx/metabolism , Ovalbumin/immunology , Ovalbumin/pharmacokinetics , Ovalbumin/toxicity , Rabbits , Rats, Sprague-Dawley , Vaccination
8.
Blood ; 119(6): 1407-17, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-22184405

ABSTRACT

Cross-presentation allows antigen-presenting cells to present exogenous antigens to CD8(+) T cells, playing an essential role in controlling infections and tumor development. IFN-α induces the rapid differentiation of human mono-cytes into dendritic cells, known as IFN-DCs, highly efficient in mediating cross-presentation, as well as the cross-priming of CD8(+) T cells. Here, we have investigated the mechanisms underlying the cross-presentation ability of IFN-DCs by studying the intracellular sorting of soluble ovalbumin and nonstructural-3 protein of hepatitis C virus. Our results demonstrate that, independently from the route and mechanism of antigen entry, IFN-DCs are extraordinarily competent in preserving internalized proteins from early degradation and in routing antigens toward the MHC class-I processing pathway, allowing long-lasting, cross-priming capacity. In IFN-DCs, both early and recycling endosomes function as key compartments for the storage of both antigens and MHC-class I molecules and for proteasome- and transporter-associated with Ag processing-dependent auxiliary cross-presentation pathways. Because IFN-DCs closely resemble human DCs naturally occurring in vivo in response to infections and other danger signals, these findings may have important implications for the design of vaccination strategies in neoplastic or chronic infectious diseases.


Subject(s)
Antigens/metabolism , Cross-Priming/drug effects , Dendritic Cells/drug effects , Endocytosis/drug effects , Interferon-alpha/pharmacology , Antigens/immunology , Blotting, Western , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cross-Priming/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endosomes/chemistry , Endosomes/metabolism , Flow Cytometry , Hepacivirus/immunology , Hepacivirus/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Hydrogen-Ion Concentration , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Microscopy, Confocal , Ovalbumin/immunology , Ovalbumin/metabolism , Ovalbumin/pharmacokinetics , Viral Nonstructural Proteins/immunology , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/pharmacokinetics
9.
Biol Pharm Bull ; 37(8): 1359-65, 2014.
Article in English | MEDLINE | ID: mdl-25087958

ABSTRACT

Ingested proteins are absorbed from the intestinal lumen via the paracellular and/or transcellular pathways, depending on their physicochemical properties. In this study, we investigated the absorption pathway(s) of ovalbumin (OVA), an egg white-allergen, as well as the mechanisms of aspirin-facilitated OVA absorption in rats. In situ intestinal re-circulating perfusion experiments showed that the absorption rate of fluorescein isothiocyanate (FITC)-labeled OVA in the distal intestine was higher than that for a marker of non-specific absorption, FITC-dextran (FD-40), and that colchicine, a general endocytosis inhibitor, suppressed OVA absorption. In the distal intestine, bafiromycin A1 and phenylarsine oxide inhibited the OVA absorption rate, whereas mehyl-ß-cyclodextrin exerted no significant effects. Thus, OVA is preferentially absorbed from the distal intestine via the paracellular and receptor- and clathrin-mediated endocytic pathways. Furthermore, aspirin increased OVA absorption in the presence or absence of colchicine, indicating that aspirin facilitated OVA absorption by inducing intestinal barrier disruption and paracellular permeability.


Subject(s)
Allergens/pharmacology , Aspirin/pharmacology , Intestine, Small/metabolism , Ovalbumin/pharmacology , Allergens/blood , Animals , Arsenicals/pharmacology , Colchicine/pharmacology , Dextrans/pharmacology , Endocytosis/drug effects , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/pharmacology , Intestinal Absorption/drug effects , Intestine, Small/drug effects , Macrolides/pharmacology , Male , Ovalbumin/blood , Ovalbumin/pharmacokinetics , Rats, Sprague-Dawley , Serum Albumin, Bovine/pharmacology , beta-Cyclodextrins/pharmacology
10.
J Am Soc Nephrol ; 24(6): 906-16, 2013 May.
Article in English | MEDLINE | ID: mdl-23539760

ABSTRACT

Podocytes are essential to the structure and function of the glomerular filtration barrier; however, they also exhibit increased expression of MHC class II molecules under inflammatory conditions, and they remove Ig and immune complexes from the glomerular basement membrane (GBM). This finding suggests that podocytes may act as antigen-presenting cells, taking up and processing antigens to initiate specific T cell responses, similar to professional hematopoietic cells such as dendritic cells or macrophages. Here, MHC-antigen complexes expressed exclusively on podocytes of transgenic mice were sufficient to activate CD8+ T cells in vivo. In addition, deleting MHC class II exclusively on podocytes prevented the induction of experimental anti-GBM nephritis. Podocytes ingested soluble and particulate antigens, activated CD4+ T cells, and crosspresented exogenous antigen on MHC class I molecules to CD8+ T cells. In conclusion, podocytes participate in the antigen-specific activation of adaptive immune responses, providing a potential target for immunotherapies of inflammatory kidney diseases and transplant rejection.


Subject(s)
Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Glomerular Filtration Barrier/immunology , Podocytes/cytology , Podocytes/immunology , Adaptive Immunity/immunology , Animals , Anti-Glomerular Basement Membrane Disease/immunology , Anti-Glomerular Basement Membrane Disease/pathology , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Female , Glomerular Basement Membrane/cytology , Glomerular Basement Membrane/immunology , Glomerular Basement Membrane/metabolism , Graft Rejection/immunology , Graft Rejection/pathology , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Immunoglobulin G/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Microspheres , Ovalbumin/immunology , Ovalbumin/pharmacokinetics , Phagocytosis/immunology , Podocytes/metabolism
11.
Med Mol Morphol ; 47(1): 14-20, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23475277

ABSTRACT

We investigated the mechanism of transplacental macromolecular transport in rats on the nineteenth day of pregnancy using tracers, transmission electron microscopy and immunohistochemistry. The blood-placental barrier of full-term rat placentas was composed of a trilaminar layer of trophoblast cells that separates the fetal capillaries from the maternal blood spaces: a layer of cytotrophoblasts lining the maternal blood space and a bilayer of syncytiotrophoblast surrounding the fetal capillaries. Horseradish peroxidase, intravenously injected into the maternal circulation, was found in the maternal blood spaces, the interspaces between the cytotrophoblasts and the syncytiotrophoblast I, many pits and small vesicles in the syncytiotrophoblast I, vesicles of the syncytiotrophoblast II, fetal connective tissue and fetal capillaries. Intravenously injected ovalbumin was detected in the maternal blood spaces, a trilaminar layer and the fetal capillaries. Neonatal Fc receptor (FcRn), a receptor for IgG, was localized at the maternal side of the blood-placental barrier. These results show that the structure of the rat blood-placental barrier is quite similar to the human blood-placental barrier, and non-specific macromolecules and food allergens may penetrate through the blood-placental barrier of the full-term placenta from the maternal to fetal circulation mediated by FcRn.


Subject(s)
Maternal-Fetal Exchange , Ovalbumin/pharmacokinetics , Placenta/metabolism , Trophoblasts/metabolism , Allergens/administration & dosage , Allergens/pharmacokinetics , Animals , Biological Transport , Female , Histocompatibility Antigens Class I/metabolism , Horseradish Peroxidase/administration & dosage , Horseradish Peroxidase/pharmacokinetics , Humans , Immunohistochemistry , Injections, Intravenous , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Ovalbumin/administration & dosage , Placenta/blood supply , Placenta/embryology , Pregnancy , Rats, Wistar , Receptors, Fc/metabolism , Trophoblasts/ultrastructure
12.
FASEB J ; 26(3): 1272-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22106367

ABSTRACT

Alum adjuvants have been in continuous clinical use for more than 80 yr. While the prevailing theory has been that depot formation and the associated slow release of antigen and/or inflammation are responsible for alum enhancement of antigen presentation and subsequent T- and B-cell responses, this has never been formally proven. To examine antigen persistence, we used the chimeric fluorescent protein EαGFP, which allows assessment of antigen presentation in situ, using the Y-Ae antibody. We demonstrate that alum and/or CpG adjuvants induced similar uptake of antigen, and in all cases, GFP signal did not persist beyond 24 h in draining lymph node antigen-presenting cells. Antigen presentation was first detectable on B cells within 6-12 h of antigen administration, followed by conventional dendritic cells (DCs) at 12-24 h, then finally plasmacytoid DCs at 48 h or later. Again, alum and/or CpG adjuvants did not have an effect on the magnitude or sequence of this response; furthermore, they induced similar antigen-specific T-cell activation in vivo. Notably, removal of the injection site and associated alum depot, as early as 2 h after administration, had no appreciable effect on antigen-specific T- and B-cell responses. This study clearly rules out a role for depot formation in alum adjuvant activity.


Subject(s)
Alum Compounds/pharmacology , Antigen Presentation/drug effects , Antigen-Presenting Cells/drug effects , Antigens/immunology , Adjuvants, Immunologic/pharmacology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens/administration & dosage , Antigens/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Flow Cytometry , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Oligodeoxyribonucleotides/pharmacology , Ovalbumin/administration & dosage , Ovalbumin/immunology , Ovalbumin/pharmacokinetics , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors
13.
J Immunol ; 187(1): 133-40, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21642544

ABSTRACT

Memory T cells are distinguished from naive T cells by their rapid production of effector cytokines, although mechanisms for this recall response remain undefined. In this study, we investigated transcriptional mechanisms for rapid IFN-γ production by Ag-specific memory CD4 T cells. In naive CD4 T cells, IFN-γ production only occurred after sustained Ag activation and was associated with high expression of the T-bet transcription factor required for Th1 differentiation and with T-bet binding to the IFN-γ promoter as assessed by chromatin immunoprecipitation analysis. By contrast, immediate IFN-γ production by Ag-stimulated memory CD4 T cells occurred in the absence of significant nuclear T-bet expression or T-bet engagement on the IFN-γ promoter. We identified rapid induction of NF-κB transcriptional activity and increased engagement of NF-κB on the IFN-γ promoter at rapid times after TCR stimulation of memory compared with naive CD4 T cells. Moreover, pharmacologic inhibition of NF-κB activity or peptide-mediated inhibition of NF-κB p50 translocation abrogated early memory T cell signaling and TCR-mediated effector function. Our results reveal a molecular mechanism for memory T cell recall through enhanced NF-κB p50 activation and promoter engagement, with important implications for memory T cell modulation in vaccines, autoimmunity, and transplantation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Immunologic Memory , Lymphocyte Activation/immunology , Transcription, Genetic/immunology , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/cytology , Cells, Cultured , Densitometry , Immunologic Memory/genetics , Interferon-gamma/biosynthesis , Interferon-gamma/metabolism , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , NF-kappa B p50 Subunit/biosynthesis , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/physiology , Ovalbumin/immunology , Ovalbumin/pharmacokinetics , Ovalbumin/physiology , Peptide Fragments/physiology , Promoter Regions, Genetic/immunology , Resting Phase, Cell Cycle/genetics , Resting Phase, Cell Cycle/immunology , T-Box Domain Proteins/biosynthesis , T-Box Domain Proteins/genetics , Up-Regulation/genetics , Up-Regulation/immunology
14.
Pharm Res ; 29(7): 1843-53, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22373666

ABSTRACT

PURPOSE: To use noninvasive fluorescence imaging to investigate the influence of molecular weight (MW) of proteins on the rate of loss from a subcutaneous (SC) injection site and subsequent uptake by the draining lymph nodes in mice. METHODS: Bevacizumab (149 kDa), bovine serum albumin (BSA, 66 kDa), ovalbumin (44.3 kDa) or VEGF-C156S (23 kDa), labeled with the near infrared dye IRDye 680, were injected SC into the front footpad of SKH-1 mice. Whole body non-invasive fluorescence imaging was performed to quantitate the fluorescence signal at the injection site and in axillary lymph nodes. RESULTS: The half-life values, describing the times for 50% loss of proteins from the injection site, were 6.81 h for bevacizumab, 2.85 h for BSA, 1.57 h for ovalbumin and 0.31 h for VEGF-C156S. The corresponding axillary lymph node exposure, represented as the area of the % dose versus time curve, was 6.27, 5.13, 4.06 and 1.54% dose ∙ h, respectively. CONCLUSIONS: Our results indicate that the rate of loss of proteins from a SC injection site is inversely related to MW of proteins, while lymph node exposure is proportionally related to the MW of proteins in a mouse model.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Lymph Nodes/metabolism , Ovalbumin/pharmacokinetics , Serum Albumin, Bovine/pharmacokinetics , Vascular Endothelial Growth Factor A/pharmacokinetics , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Bevacizumab , Cattle , Fluorescence , Fluorescent Dyes/analysis , Half-Life , Injections, Subcutaneous , Male , Mice , Molecular Weight , Ovalbumin/administration & dosage , Serum Albumin, Bovine/administration & dosage , Spectrometry, Fluorescence , Vascular Endothelial Growth Factor A/administration & dosage , Whole Body Imaging
15.
Pharm Res ; 29(2): 483-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21887598

ABSTRACT

PURPOSE: To develop a novel cancer vaccine using the targeting system of antigen protein to antigen-presenting cells (APCs) for efficient and safe cancer therapy. METHODS: The novel delivery system was constructed with antigen protein, benzalkonium chloride (BK), and γ-polyglutamic acid (γ-PGA), using ovalbumin (OVA) as a model antigen protein and evaluating its immune induction effects and utilities for cancer vaccine. RESULTS: BK and γ-PGA enabled encapsulation of OVA and formed stable anionic particles at nanoscale, OVA/BK/γ-PGA complex. Complex was taken up by dendritic cell line DC2.4 cells efficiently. We subcutaneously administered the complex to mice and examined induction of IgGs. The complex induced not only Th2-type immunoglobulins but also Th1-type immunoglobulins. OVA/BK/γ-PGA complex inhibited tumor growth of E.G7 cells expressing OVA regularly; administered OVA/BK/γ-PGA complex completely rejected tumor cells. CONCLUSION: The novel vaccine could be platform technology for a cancer vaccine.


Subject(s)
Antigen-Presenting Cells/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/therapeutic use , Drug Delivery Systems , Neoplasms/prevention & control , Ovalbumin/administration & dosage , Ovalbumin/therapeutic use , Animals , Benzalkonium Compounds/chemistry , Cancer Vaccines/immunology , Cancer Vaccines/pharmacokinetics , Cell Line , Dendritic Cells/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Male , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Ovalbumin/immunology , Ovalbumin/pharmacokinetics , Polyglutamic Acid/chemistry
16.
Bioconjug Chem ; 22(7): 1416-21, 2011 Jul 20.
Article in English | MEDLINE | ID: mdl-21688843

ABSTRACT

Oxidative stress and reduced pH are important stimuli targets for intracellular delivery and for delivery to diseased tissue. However, there is a dearth of materials able to deliver bioactive agents selectively under these conditions. We employed our recently developed dual response strategy to build a polymeric nanoparticle that degrades upon exposure to two stimuli in tandem. Our polythioether ketal based nanoparticles undergo two chemical transformations; the first is the oxidation of the thioether groups along the polymer backbone of the nanoparticles upon exposure to reactive oxygen species (ROS). This transformation switches the polymeric backbone from hydrophobic to hydrophilic and thus allows, in mildly acidic environments, the rapid acid-catalyzed degradation of the ketal groups also along the polymer backbone. Dynamic light scattering and payload release studies showed full particle degradation only in conditions that combined both oxidative stress and acidity, and these conditions led to higher release of encapsulated protein within 24 h. Nanoparticles in neutral pH and under oxidative conditions showed small molecule release and swelling of otherwise intact nanparticles. Notably, cellular studies show absence of toxicity and efficient uptake of nanoparticles by macrophages followed by cytoplasmic release of ovalbumin. Future work will apply this system to inflammatory diseases.


Subject(s)
Delayed-Action Preparations/chemistry , Nanoparticles/chemistry , Ovalbumin/administration & dosage , Sulfides/chemistry , Animals , Cell Line , Cell Survival , Delayed-Action Preparations/adverse effects , Hydrogen-Ion Concentration , Inflammation/drug therapy , Macrophages/cytology , Mice , Nanoparticles/adverse effects , Ovalbumin/pharmacokinetics , Oxidation-Reduction , Particle Size , Sulfides/chemical synthesis
17.
Biol Pharm Bull ; 34(3): 415-9, 2011.
Article in English | MEDLINE | ID: mdl-21372394

ABSTRACT

The aim of this study was to determine whether a spermine (SPM)-induced increase in gastrointestinal absorption of an allergen leads to an anaphylactic response in sensitized mice. First, we examined the enhancing effect of SPM on the gastrointestinal absorption of ovalbumin (OVA) in an in situ jejunum loop study in rats and an in vivo oral absorption study in mice. Second, we investigated whether enhancement of gastrointestinal absorption of OVA caused by SPM induces an anaphylactic response in mice sensitized to OVA. In the in situ jejunum loop study in rats, a significant amount of immune-reactive OVA was detected in the plasma after co-administration of OVA and SPM. Oral co-administration of OVA and SPM to mice in vivo also increased plasma OVA concentrations in an SPM dose-dependent manner. Furthermore, in sensitized mice, a significant increase in plasma histamine levels occurred along with the increase in plasma OVA levels after co-administration of OVA with SPM. This finding suggests that an SPM-induced increase in gastrointestinal absorption of OVA leads to an anaphylactic response. These results indicate that excess oral ingestion of SPM may have widespread health effects, including the induction of food allergies, via modulation of the function of the gastrointestinal epithelial barrier.


Subject(s)
Anaphylaxis/chemically induced , Histamine/blood , Intestinal Mucosa/metabolism , Jejunum/metabolism , Ovalbumin/pharmacokinetics , Spermine/adverse effects , Animals , Drug Synergism , Female , Intestinal Absorption , Intestinal Mucosa/immunology , Jejunum/immunology , Male , Mice , Mice, Inbred BALB C , Ovalbumin/adverse effects , Ovalbumin/immunology , Rats , Rats, Sprague-Dawley
18.
Eur J Pharm Biopharm ; 165: 306-318, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34048879

ABSTRACT

Treatment of neovascular ocular diseases involves intravitreal injections of therapeutic proteins using conventional hypodermic needles every 4-6 weeks. Due to the chronic nature of these diseases, these injections will be administrated to patients for the rest of their lives and their frequent nature can potentially pose a risk of sight-threatening complications and poor patient compliance. Therefore, we propose to develop nanoparticle (NP)-loaded bilayer dissolving microneedle (MN) arrays, to sustain delivery of protein drugs in a minimally invasive manner. In this research, a model protein, ovalbumin (OVA)-encapsulated PLGA NPs were prepared and optimised using a water-in-oil-in-water (W/O/W) double emulsion method. The impact of stabilisers and primary sonication time on the stability of encapsulated OVA was evaluated using an enzyme-linked immunosorbent assay (ELISA). Results showed that the lower primary sonication time was capable of sustaining release (77 days at 28.5% OVA loading) and improving the OVA bioactivity. The optimised NPs were then incorporated into a polymeric matrix to fabricate bilayer MNs and specifically concentrated into MN tips by high-speed centrifugation. Optimised bilayer MNs exhibited good mechanical and insertion properties and rapid dissolution kinetics (less than 3 min) in excised porcine sclera. Importantly, ex vivo transscleral distribution studies conducted using a multiphoton microscope confirmed the important function of MN arrays in the localisation of proteins and NPs in the scleral tissue. Furthermore, the polymers selected to prepare bilayer MNs and OVA NPs were determined to be biocompatible with retinal cells (ARPE-19). This delivery approach could potentially sustain the release of encapsulated proteins for more than two months and effectively bypass the scleral barrier, leading to a promising therapy for treating neovascular ocular diseases.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Drug Delivery Systems/methods , Nanoparticles/chemistry , Administration, Ophthalmic , Angiogenesis Inhibitors/pharmacokinetics , Animals , Cell Line , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/pathology , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Drug Compounding/methods , Drug Liberation , Humans , Ovalbumin/administration & dosage , Ovalbumin/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Posterior Eye Segment/blood supply , Posterior Eye Segment/pathology , Ranibizumab/administration & dosage , Ranibizumab/pharmacokinetics , Retinal Neovascularization/drug therapy , Retinal Neovascularization/pathology , Sclera/metabolism , Swine
19.
Immunol Cell Biol ; 88(6): 658-66, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20195280

ABSTRACT

Inhaled allergens are known for their immediate and ongoing effects in the respiratory tract (RT). In this report, we track inhaled antigen in normal mice for 7 days and find that while it is cleared from the airways, inhaled antigen persists in peripheral lung tissue and the draining lymph nodes (DLNs). The persistence of antigen led to ongoing presentation in the lymph nodes, but not the lungs, that decreased with time in direct proportion with the frequency of antigen-bearing RT dendritic cells (DCs). There was evidence of functional changes among the antigen-bearing DCs in the lymph nodes, as the expression of CD40, CD80 and CD86 were modulated over the course of 7 days. At the same time, there was a decrease in both CD4(+) T-cell proliferation in lymph nodes and the generation of recirculating CD4(+) T cells. However, early presentation of lower doses of inhaled antigen also resulted in a decrease in CD4(+) T-cell proliferation and recirculation. Thus, T-cell recirculation depends on the strength of stimulus in the DLNs and is produced by a combination of the dose of antigen delivered to the RT, DC migration and co-stimulatory molecule expression. These results provide an important insight into the fate of inhaled antigen in vivo and the influence of persistent antigen presentation on T-cell activation in the lymph nodes.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Dendritic Cells/metabolism , Lung/metabolism , Lymph Nodes/metabolism , Ovalbumin/pharmacokinetics , Animals , Antigen Presentation/genetics , Antigen Presentation/immunology , Antigens, CD/biosynthesis , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/pathology , Lung/immunology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Ovalbumin/administration & dosage
20.
Br J Dermatol ; 162(1): 29-41, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19863514

ABSTRACT

Background We previously have shown that nona-arginine protein transduction domain (R9-PTD) induced efficient protein-antigen (Ag) transduction of dendritic cells (DCs) in vitro, resulting in the efficient induction of strong Ag-specific immune responses mediated by CD8+ and CD4+ T cells and in superior antitumour effects in vivo in cancer-bearing mice. Objectives The Ag-specific immune responses caused by intradermal (i.d.) injections of R9-PTD-containing protein Ags without DC preparation were investigated. We also investigated the antitumour effects by intratumoral (i.t.) injections of rR9-containing protein Ags. Methods Synthesized SIINFEKL peptide, or recombinant ovalbumin fusion proteins (rOVA, rR9-OVA), were directly injected into abdominal skin in naïve C57BL/6 mice. OVA-specific cytotoxic T lymphocyte (CTL) activity, serum IgG titre and cytokine profiles were investigated. Histopathological analyses were also performed. In a cancer vaccination model, EG.7 (OVA-cDNA transfectants thymoma) cells were inoculated intradermally in C57BL/6 mice, and the antitumour effects were evaluated by i.t. injections of rR9-OVA in a treatment setting. Results i.d. injections of rR9-OVA into naïve C57BL/6 mice elicited OVA-specific CTLs and produced IgG2-dominant immunoglobulin. The i.d. injections of rR9-OVA also induced inflammatory cell infiltrates containing neutrophils, monocytes and lymphocytes, as well as production of inflammatory cytokines such as interferon (IFN)-gamma, interleukin-2 and IFN-inducible protein 10, with presenting SIINFEKL epitopes on major histocompatibility complex (MHC) class I molecules at the injection area. i.t. injections of rR9-OVA into EG.7 tumour mass significantly suppressed tumour growth, and these effects were completely abrogated by the depletion of CD8+ T cells. These antitumour effects were superior to those elicited by i.t. injections of rR9-OVA-treated DCs. Conclusions i.d. injections of rR9-containing immunogenic Ag without adjuvants simultaneously induce dual immunological effects: the induction of Tc1- and Th1-dominant immune responses, and the induction of inflammatory and CTL-mediated immune responses at the injection area by expressing Ag epitopes on MHC class I molecules as targets. This simple vaccination approach with R9-PTD-containing fusion proteins might be useful as prophylactic immunotherapy for cancer or infectious diseases.


Subject(s)
Antigen Presentation/immunology , Cancer Vaccines/administration & dosage , Oligopeptides/immunology , Skin Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cytokines/administration & dosage , Cytokines/analysis , Female , Histocompatibility Antigens/immunology , Immunoglobulin G/blood , Injections, Intradermal , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Ovalbumin/pharmacokinetics , Recombinant Fusion Proteins/immunology , Skin/immunology , Skin/pathology , Skin Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL