Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 7934, 2024 04 04.
Article in English | MEDLINE | ID: mdl-38575614

ABSTRACT

Biodistribution tests are crucial for evaluating the safety of cell therapy (CT) products in order to prevent unwanted organ homing of these products in patients. Quantitative polymerase chain reaction (qPCR) using intronic Alu is a popular method for biodistribution testing owing to its ability to detect donor cells without modifying CT products and low detection limit. However, Alu-qPCR may generate inaccurate information owing to background signals caused by the mixing of human genomic DNA with that of experimental animals. The aim of this study was to develop a test method that is more specific and sensitive than Alu-qPCR, targeting the mitochondrial DNA (mtDNA) sequence that varies substantially between humans and experimental animals. We designed primers for 12S, 16S, and cytochrome B in mtDNA regions, assessed their specificity and sensitivity, and selected primers and probes for the 12S region. Human adipose-derived stem cells, used as CT products, were injected into the tail vein of athymic NCr-nu/nu mice and detected, 7 d after administration, in their lungs at an average concentration of 2.22 ± 0.69 pg/µg mouse DNA, whereas Alu was not detected. Therefore, mtDNA is more specific and sensitive than Alu and is a useful target for evaluating CT product biodistribution.


Subject(s)
DNA, Mitochondrial , Mitochondria , Humans , Mice , Animals , DNA, Mitochondrial/genetics , Tissue Distribution , DNA Primers , Mitochondria/genetics
3.
EMBO Mol Med ; 16(1): 40-63, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38177528

ABSTRACT

Despite intensive clinical and scientific efforts, the mortality rate of sepsis remains high due to the lack of precise biomarkers for patient stratification and therapeutic guidance. Secreted human tryptophanyl-tRNA synthetase 1 (WARS1), an endogenous ligand for Toll-like receptor (TLR) 2 and TLR4 against infection, activates the genes that signify the hyperinflammatory sepsis phenotype. High plasma WARS1 levels stratified the early death of critically ill patients with sepsis, along with elevated levels of cytokines, chemokines, and lactate, as well as increased numbers of absolute neutrophils and monocytes, and higher Sequential Organ Failure Assessment (SOFA) scores. These symptoms were recapitulated in severely ill septic mice with hypercytokinemia. Further, injection of WARS1 into mildly septic mice worsened morbidity and mortality. We created an anti-human WARS1-neutralizing antibody that suppresses proinflammatory cytokine expression in marmosets with endotoxemia. Administration of this antibody into severe septic mice attenuated cytokine storm, organ failure, and early mortality. With antibiotics, the antibody almost completely prevented fatalities. These data imply that blood-circulating WARS1-guided anti-WARS1 therapy may provide a novel theranostic strategy for life-threatening systemic hyperinflammatory sepsis.


Subject(s)
Sepsis , Tryptophan-tRNA Ligase , Humans , Animals , Mice , Tryptophan-tRNA Ligase/genetics , Precision Medicine , Cytokines/metabolism , Chemokines
4.
Nat Nanotechnol ; 18(4): 390-402, 2023 04.
Article in English | MEDLINE | ID: mdl-36635335

ABSTRACT

Although conventional innate immune stimuli contribute to immune activation, they induce exhausted immune cells, resulting in suboptimal cancer immunotherapy. Here we suggest a kinetically activating nanoadjuvant (K-nanoadjuvant) that can dynamically integrate two waves of innate immune stimuli, resulting in effective antitumour immunity without immune cell exhaustion. The combinatorial code of K-nanoadjuvant is optimized in terms of the order, duration and time window between spatiotemporally activating Toll-like receptor 7/8 agonist and other Toll-like receptor agonists. K-nanoadjuvant induces effector/non-exhausted dendritic cells that programme the magnitude and persistence of interleukin-12 secretion, generate effector/non-exhausted CD8+ T cells, and activate natural killer cells. Treatment with K-nanoadjuvant as a monotherapy or in combination therapy with anti-PD-L1 or liposomes (doxorubicin) results in strong antitumour immunity in murine models, with minimal systemic toxicity, providing a strategy for synchronous and dynamic tailoring of innate immunity for enhanced cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Animals , Mice , Immunotherapy/methods , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/therapeutic use , Immunity, Innate , Neoplasms/therapy
5.
Int J Mol Sci ; 23(16)2022 Aug 16.
Article in English | MEDLINE | ID: mdl-36012471

ABSTRACT

Alveolar organoids (AOs), derived from human pluripotent stem cells (hPSCs) exhibit lung-specific functions. Therefore, the application of AOs in pulmonary disease modeling is a promising tool for understanding disease pathogenesis. However, the lack of immune cells in organoids limits the use of human AOs as models of inflammatory diseases. In this study, we generated AOs containing a functional macrophage derived from hPSCs based on human fetal lung development using biomimetic strategies. We optimized culture conditions to maintain the iMACs (induced hPSC-derived macrophages) AOs for up to 14 days. In lipopolysaccharide (LPS)-induced inflammatory conditions, IL-1ß, MCP-1 and TNF-α levels were significantly increased in iMAC-AOs, which were not detected in AOs. In addition, chemotactic factor IL-8, which is produced by mononuclear phagocytic cells, was induced by LPS treatment in iMACs-AOs. iMACs-AOs can be used to understand pulmonary infectious diseases and is a useful tool in identifying the mechanism of action of therapeutic drugs in humans. Our study highlights the importance of immune cell presentation in AOs for modeling inflammatory pulmonary diseases.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Cell Differentiation , Humans , Lipopolysaccharides/pharmacology , Lung , Macrophages , Organoids
6.
Clin Transl Sci ; 14(6): 2161-2170, 2021 11.
Article in English | MEDLINE | ID: mdl-34080313

ABSTRACT

KMRC011 is a novel Toll-like receptor 5 agonist under development as a treatment for acute radiation syndrome (ARS). The aim of this first-in-human study was to investigate the tolerability, pharmacokinetics, and pharmacodynamics of a single intramuscular dose of KMRC011 in healthy subjects. A randomized, single-blind, placebo-controlled, single dose-escalation study was conducted with the starting dose of 5 µg. Eight (4 only for 5 µg cohort) subjects per cohort were randomly assigned to KMRC011 or placebo in a 3:1 ratio. Dose-limiting toxicity (DLT) was assessed throughout the study. Serum concentrations of KMRC011, granulocyte colony-stimulating factor (G-CSF), and interleukin-6 (IL-6) were measured up to 48 h postdose. Based on safety review, the dose of KMRC011 escalated up to 20 µg, and consequently, a total of 4 dose levels (5, 10, 15, and 20 µg) were explored. The most common adverse event was injection site reaction, showing no dose-related trend. Three DLTs (2 cases of hepatic enzyme increased and 1 of pyrexia) were observed; 1 in the 15 µg cohort and 2 in the 20 µg cohort. A developed method could not detect any KMRC011 in serum. KMRC011 15 µg and 20 µg showed significant increases of G-CSF, IL-6, and absolute neutrophil counts, compared with the placebo. A single intramuscular administration of KMRC011 ranging from 5 to 15 µg was tolerated in healthy subjects. Doses of KMRC011 equal to or greater than 15 µg exerted TLR5 agonist-like activities by increasing serum G-CSF and IL-6. It suggests that KMRC011 has the potential for a treatment for ARS.


Subject(s)
Acute Radiation Syndrome/drug therapy , Dose-Response Relationship, Drug , Peptide Fragments/pharmacology , Peptide Fragments/pharmacokinetics , Adult , Humans , Male , Middle Aged , Peptide Fragments/administration & dosage , Peptide Fragments/therapeutic use , Young Adult
7.
Tissue Eng Regen Med ; 17(3): 335-350, 2020 06.
Article in English | MEDLINE | ID: mdl-32451775

ABSTRACT

BACKGROUND: Human adipose tissue-derived stem cells (ADSCs) are attractive multipotent stem cell sources with therapeutic potential in various fields requiring repair and regeneration, such as acute and chronically damaged tissues. ADSC is suitable for cell-based therapy, but its use has been hampered due to poor survival after administration. Potential therapeutic use of ADSC requires mass production of cells through in vitro expansion. Many studies have consistently observed the tendency of senescence by mesenchymal stem cell (MSC) proliferation upon expansion. Hypoxia has been reported to improve stem cell proliferation and survival. METHODS: We investigated the effects of hypoxia pretreatment on ADCS proliferation, migration capacity, differentiation potential and cytokine production. We also analyzed the effects of vascular endothelial growth factor (VEGF) on osteogenic and chondrogenic differentiation of ADSCs by hypoxia pretreatment. RESULTS: Hypoxia pretreatment increased the proliferation of ADSCs by increasing VEGF levels. Interestingly, hypoxia pretreatment significantly increased chondrogenic differentiation but decreased osteogenic differentiation compared to normoxia. The osteogenic differentiation of ADSC was decreased by the addition of VEGF but increased by the depletion of VEGF. We have shown that hypoxia pretreatment increases the chondrogenic differentiation of ADSCs while reducing osteogenic differentiation in a VEGF-dependent manner. CONCLUSION: These results show that hypoxia pretreatment can provide useful information for studies that require selective inhibition of osteogenic differentiation, such as cartilage regeneration.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Chondrocytes/metabolism , Hypoxia/metabolism , Hypoxia/therapy , Stem Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adipose Tissue/cytology , Cell Differentiation/drug effects , Cell Movement , Cell Proliferation/drug effects , Chondrogenesis/drug effects , Cytokines/metabolism , Gene Expression , Gene Expression Profiling , Humans , Multipotent Stem Cells/metabolism , Osteogenesis , Stem Cells/cytology
8.
ACS Nano ; 13(11): 12671-12686, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31589013

ABSTRACT

The low therapeutic efficacy of current cancer immunotherapy is related to nonimmunogenic and immunosuppressive tumor microenvironments (TMEs). To overcome these limitations, both the immune priming of antitumoral lymphocytes and the reprogramming of immunosuppressive factors in TMEs are essential. Here, we suggest a nanoemulsion (NE)-based immunotherapeutic platform that can not only modulate tumor-induced suppression but also induce an effective cell-mediated immune response for T cell proliferation. Multifunctional NEs can be fabricated by integrating the efficacy of NEs as delivery systems and the multifaceted immunomodulation characteristics (i.e., immunostimulation and reprogramming of immunosuppression) of small molecule-based Toll-like receptor 7/8 agonists. Local in situ vaccination of melanoma and cervical tumor models with tumor antigens (protein and peptide) adjuvanted with NE loaded with TLR7/8 agonists [NE (TLR7/8a)] induced the recruitment and activation of innate immune cells, infiltration of lymphocytes, and polarization of tumor-associated M2 macrophages, which resulted in inhibition of tumor growth and prolonged survival in both primary and rechallenged tumor models. Antibody-depletion experiments also suggested that macrophages, type I IFN (IFN-α and IFN-ß), CD8+ T cells, and NK1.1+ cells contributed to the antitumor effect of NE (TLR7/8a). The combination of antitumoral lymphocytes and reprogramming of immunosuppressive TMEs induced by NE (TLR7/8a) treatment evoked a synergistic antitumor immune response with immune checkpoint blockade therapy (anti-PD-1 and anti-PD-L1).


Subject(s)
Cancer Vaccines , Immunotherapy/methods , Membrane Glycoproteins/agonists , Nanostructures/chemistry , Toll-Like Receptor 7/agonists , Tumor Microenvironment/immunology , Animals , Antigens, Neoplasm/immunology , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Emulsions/chemistry , Emulsions/pharmacology , Female , Immunosuppression Therapy , Mice , Mice, Inbred C57BL , Toll-Like Receptor 8/agonists
9.
Cell Cycle ; 17(9): 1138-1145, 2018.
Article in English | MEDLINE | ID: mdl-29895199

ABSTRACT

Double-strand breaks (DSBs) are repaired through two major pathways, homology-directed recombination (HDR) and non-homologous end joining (NHEJ). The choice between these two pathways is largely influenced by cell cycle phases. HDR can occur only in S/G2 when sister chromatid can provide homologous templates, whereas NHEJ can take place in all phases of the cell cycle except mitosis. Central to NHEJ repair is the Ku70/80 heterodimer which forms a ring structure that binds DSB ends and serves as a platform to recruit factors involved in NHEJ. Upon completion of NHEJ repair, DNA double strand-encircling Ku dimers have to be removed. The removal depends on ubiquitylation and proteasomal degradation of Ku80 by the ubiquitin E3 ligases RNF8. Here we report that RNF8 is a substrate of APCCdh1 and the latter keeps RNF8 level in check at DSBs to prevent premature turnover of Ku80.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , DNA Damage/physiology , DNA End-Joining Repair/physiology , Ku Autoantigen/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Cell Cycle Checkpoints , DNA Breaks, Double-Stranded , DNA-Binding Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
10.
Biomaterials ; 130: 56-66, 2017 06.
Article in English | MEDLINE | ID: mdl-28364631

ABSTRACT

In this study, synthetic vaccine nanoparticles (SVNPs) that efficiently targeted lymph nodes, where immune responses against foreign antigens are primed, were developed to enhance antitumor immunity. The size (20-70 nm) and surface character (amination) of poly(γ-glutamic acid)-based SVNPs were selected for effective loading and delivery (i.e., migration and retention) of model tumor antigen (OVA) and toll-like receptor 3 agonist (poly (I:C)) to immune cells in lymph nodes. Antigen-presenting cells treated with SVNP-OVA and SVNP-IC showed higher uptake of OVA and poly (I:C) and higher secretion of inflammatory cytokines (TNF-α, IL-6) and type I interferon (IFN-α, IFN-ß) than those treated with OVA and poly (I:C) alone. In vivo analysis revealed higher levels of activation markers, inflammatory cytokines, and type I IFNs in the lymph nodes of mice immunized with SVNP-IC compared to those of mice in other groups. SVNP-IC-treated mice showed significantly greater in vivo natural killer cell expansion/activation (NK1.1+ cells) and CD8+ T cell response (CD8+ INF-γ+ cells) in innate and adaptive immunity, respectively. Both preventive and therapeutic vaccination of EG7-OVA tumor-bearing mice using the simultaneous injection of both SVNP-OVA and SVNP-IC induced higher antitumor immunity and inhibited tumor growth.


Subject(s)
Adaptive Immunity , Cancer Vaccines/immunology , Immunity, Innate , Lymph Nodes/metabolism , Nanoparticles/chemistry , Neoplasms/immunology , Vaccines, Synthetic/immunology , Animals , Chickens , Immunotherapy, Adoptive , Mice, Inbred C57BL , Nanoparticles/ultrastructure , Ovalbumin/immunology , Poly I-C/immunology
11.
Carbohydr Polym ; 157: 476-483, 2017 Feb 10.
Article in English | MEDLINE | ID: mdl-27987951

ABSTRACT

In this study, the multifaceted properties of hyaluronic acid (HA) were used for the targeted therapy of cancer by photodynamic therapy (PDT) guided by molecular imaging. Near-infrared (NIR) photosensitizers (Chlorin e6; Ce6) were encapsulated into poly(lactic-co-glycolic acid) nanoparticles (PLGA NPs) coated with HA that can act as CD44 targeting ligand. The abundant carboxylate groups of HA also enabled the chelation of gadolinium ions (Gd3+), T1-weighted MRI contrast agents, on the surface of PLGA NPs. Through both in vitro and in vivo fluorescence and MRI signal analysis, we confirmed that the HA-Gd-Ce6-PLGA NPs (HAGCP-NPs) could efficiently target CD44-overexpressing A549 cancer cells. When an NIR laser was illuminated to irradiate A549 tumor-bearing mice, the groups treated with HAGCP-NPs showed remarkable delays in tumor growth or tumor regression. Taken together, the HAGCP-NPs are expected to be used as a theranostic platform for the dual modal (MR/NIR) imaging and PDT of cancer.


Subject(s)
Hyaluronic Acid , Nanoparticles , Neoplasms, Experimental/drug therapy , Photochemotherapy , Photosensitizing Agents/pharmacology , A549 Cells , Animals , Humans , Magnetic Resonance Imaging , Mice
12.
Carbohydr Polym ; 155: 1-10, 2017 Jan 02.
Article in English | MEDLINE | ID: mdl-27702491

ABSTRACT

A novel powder-form combination adjuvant system containing two immunostimulatory compounds was firstly developed and evaluated as a therapeutic intervention for cancer immunotherapy. With the help of hyaluronic acid (HA), water insoluble monophosphoryl lipid A (MPL), QS21 and imiquimod (R837), could be easily dispersed in aqueous solution and lyophilized as powder-form, which have an advantage in room-temperature storage stability compared with those conventional liquid formulation that requires cold storage. Two kinds of HA-based combination vaccine adjuvants (HA/MPL/QS21, HMQ and HA/MPL/R837, HMR) contributed to the increase of both humoral and cellular immunity, which is very important for efficient cancer immunotherapy. Through the challenge experiments in EG7-OVA (mouse lymphoma-expressing OVA) tumor-bearing mice model, we found out that the immunostimulatory effects of HMQ and HMR were successful in the inhibition of tumor proliferation. Taken together, both HA-based powder-form combination adjuvant systems are expected to be used as potent prophylactic and therapeutic cancer vaccine.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Cancer Vaccines/therapeutic use , Hyaluronic Acid/pharmacology , Lymphoma/therapy , Adjuvants, Immunologic/chemistry , Aminoquinolines/chemistry , Aminoquinolines/immunology , Aminoquinolines/therapeutic use , Animals , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Drug Carriers , Female , Hyaluronic Acid/chemistry , Hydrogen Bonding , Imiquimod , Immunotherapy , Lipid A/analogs & derivatives , Lipid A/chemistry , Lipid A/immunology , Lipid A/therapeutic use , Lymphoma/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Saponins/chemistry , Saponins/immunology , Saponins/therapeutic use , Solubility
13.
Int J Nanomedicine ; 11: 5621-5632, 2016.
Article in English | MEDLINE | ID: mdl-27822040

ABSTRACT

In this research, we synthesized bioderived poly(amino acid) hydrogel particles that showed pH-dependent membrane-disrupting properties and controlled cytosolic delivery of antitumor drugs. Poly(γ-glutamic acid) (γ-PGA) that has been produced extensively using bacteria, especially those of Bacillus subtilis species, was modified with cholesterol (γ-PGA/Chol), and the γ-PGA/Chol conjugates were used to form polymeric nanoparticles the size of 21.0±1.1 nm in aqueous solution. When the polymeric nanoparticles were mixed with doxorubicin (Dox), raspberry-like hydrogel particles (RBHPs) were formed by the electrostatic interaction between the cationically charged Dox and the anionically charged nanoparticles. The average size and surface charge of the RBHPs in aqueous solution were 444.9±122.5 nm and -56.44 mV, respectively. The loaded amount of Dox was approximately 63.9 µg/mg of RBHPs. The RBHPs showed controlled drug release behavior in both in vitro and ex vivo cell-based experiments. Through fluorescence microscopy and fluorescence-activated cell sorting, the cellular uptake of RBHPs into human cervical cancer cells (HeLa) was analyzed. The cytotoxic effect, evaluated by the methyl tetrazolium salt assay, was dependent on both the concentration of RBHPs and the treatment time. The pH-dependent membrane-disrupting properties of the RBHPs and the subsequent cytosolic delivery of Dox were evaluated using a standard hemolysis assay. Upon an increase in hydrophobicity at the lysosomal acidic pH, RBHPs could easily interact, penetrate cell membranes, and destabilize them. Taken together, the data suggested that RBHPs could be used as drug delivery carriers after loading with other therapeutic drugs, such as proteins or small interfering RNA for cancer therapy.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Cell Membrane/drug effects , Doxorubicin/pharmacology , Drug Delivery Systems , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Nanoparticles/administration & dosage , Polyglutamic Acid/analogs & derivatives , Uterine Cervical Neoplasms/drug therapy , Animals , Antibiotics, Antineoplastic/chemistry , Cytosol/metabolism , Doxorubicin/chemistry , Drug Carriers/chemistry , Erythrocytes/drug effects , Female , Flow Cytometry , Hemolysis/drug effects , Humans , Hydrogen-Ion Concentration , Nanoparticles/chemistry , Polyglutamic Acid/chemistry , Rubus , Sheep , Tumor Cells, Cultured , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
14.
Small ; 12(45): 6279-6288, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27671946

ABSTRACT

Seasonal emerging infectious diseases such as influenza A impose substantial risk and need new translational strategies to achieve active immunomodulation. Here, a novel injectable pathogen-mimicking hydrogel (iPMH) that can enhance both cellular and humoral immune responses is suggested. By the help of poly(γ-glutamic acid) that has abundant carboxylate groups and dispersion helper function, hydrophobic immunostimulatory 3-O-desacyl-4'-monophosphoryl lipid A (MPLA) molecules and viral antigens (PR8, W150) can be successfully combined as pathogen-mimicking adjuvants. Polyelectrolyte complex between the poly(γ-glutamic acid)-based adjuvants and collagens generate in situ gel-forming hydrogel at physiological temperature. When the iPMH are immunized, they act as a pathogen-mimicking (MPLA, H1N1, H5N1) immune priming center and a depot for continuous stimulation of immune system, resulting in the induction of high levels (8.5 times higher) of antigen-specific IgG titers in the sera of mice and the increased number of IFN-γ-producing cells (7.3 times higher) compared with those in the groups immunized with antigen plus clinically used aluminum gels. Following the intranasal infection of the mouse adapted virus (emerging infectious 2009 H1N1 and highly pathogenic 2006 H5N1) at 50 times the 50% lethal dose, the mice immunized with viral antigens plus iPMH exhibit 100% protective immunity against lethal virus challenge.


Subject(s)
Hydrogels/chemistry , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Adjuvants, Immunologic , Animals , Cells, Cultured , Female , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Lipid A/analogs & derivatives , Lipid A/chemistry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Polyglutamic Acid/analogs & derivatives , Polyglutamic Acid/chemistry
15.
Int J Nanomedicine ; 11: 3753-64, 2016.
Article in English | MEDLINE | ID: mdl-27540289

ABSTRACT

Effective induction of an antigen-specific cytotoxic T lymphocyte (CTL) immune response is one of the key goals of cancer immunotherapy. We report the design and fabrication of polyethylenimine (PEI)-coated polymer nanoparticles (NPs) as efficient antigen-delivery carriers that can induce antigen cross-presentation and a strong CTL response. After synthesis of poly(d,l-lactide-co-glycolide) (PLGA) NPs containing ovalbumin (OVA) by the double-emulsion solvent-evaporation method, cationic-charged PLGA NPs were generated by coating them with PEI. In a methyl tetrazolium salt assay, no discernible cytotoxic effect of PEI-coated PLGA (OVA) NPs was observed. The capacity and mechanism of PEI-coated PLGA (OVA) NPs for antigen delivery and cross-presentation on dendritic cells (DCs) were determined by fluorescence microscopy and flow cytometry. PEI-coated PLGA (OVA) NPs were internalized efficiently via phagocytosis or macropinocytosis in DCs and induced efficient cross-presentation of the antigen on MHC class I molecules via both endosome escape and a lysosomal processing mechanism. The DCs treated with PEI-coated PLGA (OVA) NPs induced a release of IL-2 cytokine from OVA-specific CD8-OVA1.3 T cells more efficiently than DCs treated with PLGA (OVA) NPs. Therefore, the PEI-coated PLGA (OVA) NPs can induce antigen cross-presentation and are expected to be used for induction of a strong CTL immune response and for efficient anticancer immunotherapy.


Subject(s)
Antigens/immunology , Cross-Priming , Lactic Acid/chemistry , Nanoparticles/chemistry , Ovalbumin/immunology , Polyethyleneimine/chemistry , Polyglycolic Acid/chemistry , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens/administration & dosage , Cell Differentiation , Cell Survival , Dendritic Cells/cytology , Dendritic Cells/immunology , Endocytosis , Female , Intracellular Space/metabolism , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Nanoparticles/ultrastructure , Polylactic Acid-Polyglycolic Acid Copolymer
16.
Molecules ; 21(5)2016 Apr 29.
Article in English | MEDLINE | ID: mdl-27136523

ABSTRACT

In this study, we developed electrostatically self-assembled ternary nanocomplexes as a safe and effective non-viral vector for the delivery of plasmid DNA (pDNA) into human adipose-derived stem cells (hASCs). Although polyethylenimine (PEI) polymers initially showed excellent performance as gene delivery carriers, their broad use has been limited by cytotoxicity resulting from their strong positive charge. To reduce the cytotoxicity, we utilized anionic hyaluronic acid (HA) as a corona layer material for pDNA/PEI binary nanocomplexes. HA was also introduced to increase the targeting efficiency of pDNA/PEI nanocomplexes because HA has can bind CD44 that is highly expressed on the surface of hASCs. We confirmed that the addition of HA changed the surface charge of pDNA/PEI nanocomplexes from positive to negative. The pDNA/PEI/HA ternary nanocomplexes showed high transfection efficiency and low cytotoxicity compared with commercially available products. When hASCs were pretreated with HA to passivate CD44, the transfection efficiency of pDNA/PEI/HA nanocomplexes was significantly reduced. These results suggest that HA that can act as a targeting ligand to CD44 contributed to the improved transfection of pDNA into hASCs. Our novel pDNA/PEI/HA nanocomplexes may be used as an effective non-viral pDNA delivery system for hASCs.


Subject(s)
DNA/metabolism , Genetic Vectors , Mesenchymal Stem Cells , Nanoparticles , Plasmids/genetics , Transfection , Adipose Tissue/cytology , Cells, Cultured , Humans , Hyaluronan Receptors/metabolism , Polyethyleneimine , Static Electricity
17.
Vaccine ; 33(38): 4762-9, 2015 Sep 11.
Article in English | MEDLINE | ID: mdl-26271830

ABSTRACT

Here, monophosphoryl lipid A (MPLA) and aluminum salt (Alum) were introduced into a hyaluronic acid (HA)-based combination vaccine adjuvant for hepatitis B vaccine (HBV). Although Alum is a well-known hepatitis B vaccine adjuvant that induces an enhanced humoral immune response, it cannot induce the cellular immune responses. On the other hand, MPLA has been generally reported to promote IFN-γ production via antigen-specific CD4(+) T cells, but it is not water soluble as a result of its long hydrophobic alkyl chains. To this end, water insoluble MPLA could be solubilized in an aqueous solution with the help of HA, which contains many carboxyl and hydroxyl groups that can be used to attach to the hydroxyl head groups of MPLA via hydrogen bonds. Three groups of mice were treated with either hepatitis B surface antigen (HBsAg) alone, HBsAg_Alum complex, or HBsAg_Alum_MPLA/HA complex. The group immunized with the HBsAg_Alum_MPLA/HA complex exhibited a high increase in cellular immune response as well as in humoral immune response relative to the other two groups. The antibody, cytokine and T cell levels were most elevated in the group of mice immunized with HBsAg_Alum_MPLA/HA complex, even at a 1µg/mice dose, and the magnitude was still maintained even after 8 weeks. Specifically, the antibody value was 120 times larger in mice vaccinated with HBsAg_Alum_MPLA/HA complex than in mice vaccinated with HBsAg_Alum complex designed similar to commercially available hepatitis B vaccine, Engerix B. The cytokine and T cell proliferation levels were 2 times and 6 times larger in mice adjuvanted with HBsAg_Alum_MPLA/HA complex than in those vaccinated with HBsAg_Alum. The results therefore indicate that incorporating MPLA and Alum with HA can be a potent strategy to increase both the magnitude and the persistence of HBsAg-specific immune responses to protect hosts against hepatitis B virus infection.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Aluminum/administration & dosage , Hepatitis B Antibodies/blood , Hepatitis B Vaccines/immunology , Hyaluronic Acid/administration & dosage , Lipid A/analogs & derivatives , T-Lymphocytes/immunology , Animals , Cell Proliferation , Cytokines/metabolism , Female , Hepatitis B Vaccines/administration & dosage , Lipid A/administration & dosage , Mice, Inbred C57BL , Treatment Outcome
18.
Gastric Cancer ; 18(1): 55-64, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24481855

ABSTRACT

BACKGROUND: This study aimed to examine tracers designed to overcome the disadvantages of indocyanine green (ICG), which disperses quickly to multiple lymph nodes, using a near-infrared (NIR) imaging system in animal models. METHODS: Diluted ICG, ICG/poly-γ-glutamic acid (PGA) complex, and IRDye900-conjugated pullulan-cholesterol nanoprobe "near-infrared polynagogel" (NIR-PNG) were injected into the stomachs of dogs and pigs, and the patterns of dispersion were observed using an NIR imaging system. To compare retention times, fluorescence signals were evaluated in the stomach and small bowel of animals 1 week after injection. RESULTS: A diluted concentration (~0.1 mg/ml) of ICG was optimal for NIR imaging compared with the conventional concentration (5 mg/ml) for visual inspection. When injected into the stomach, the signals of ICG and ICG/PGA complex were relatively large at the injection site, and signals were detected at multiple sentinel nodes and lymph nodes beyond them. The NIR-PNG signal intensity was relatively small at the injection site and limited to only one sentinel node with no additional node. When evaluated 1 week after injection, only the NIR-PNG signal was detected in the canine stomach, and the signal intensity at the lymph nodes of the porcine small bowel was the highest with NIR-PNG, followed by ICG/PGA complex and finally ICG. CONCLUSION: NIR-PNG showed the best characteristics of less dispersion and longer retention in the sentinel nodes, and ICG/PGA complex remained longer than diluted ICG. These tracers could potentially be used as optimal tracers for sentinel node navigation surgery in gastric cancer.


Subject(s)
Diagnostic Imaging/methods , Glucans , Indocyanine Green , Lymph Nodes/pathology , Sentinel Lymph Node Biopsy/methods , Animals , Disease Models, Animal , Dogs , Female , Fluorescent Dyes , Indocyanine Green/pharmacokinetics , Intestines/drug effects , Lymphatic Metastasis/diagnosis , Mice, Inbred BALB C , Nanostructures , Polyglutamic Acid , Quantum Dots , Sus scrofa
19.
Nat Commun ; 5: 3846, 2014 May 14.
Article in English | MEDLINE | ID: mdl-24828763

ABSTRACT

The ability to manipulate small fluid droplets, colloidal particles and single cells with the precision and parallelization of modern-day computer hardware has profound applications for biochemical detection, gene sequencing, chemical synthesis and highly parallel analysis of single cells. Drawing inspiration from general circuit theory and magnetic bubble technology, here we demonstrate a class of integrated circuits for executing sequential and parallel, timed operations on an ensemble of single particles and cells. The integrated circuits are constructed from lithographically defined, overlaid patterns of magnetic film and current lines. The magnetic patterns passively control particles similar to electrical conductors, diodes and capacitors. The current lines actively switch particles between different tracks similar to gated electrical transistors. When combined into arrays and driven by a rotating magnetic field clock, these integrated circuits have general multiplexing properties and enable the precise control of magnetizable objects.


Subject(s)
Magnets , Nanoparticles , Single-Cell Analysis/methods , Colloids , Computers , Hydrodynamics , Single-Cell Analysis/instrumentation
20.
Magn Reson Med ; 71(3): 1054-63, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23640738

ABSTRACT

PURPOSE: We determined whether poly(lactic-co-glycolic acid) nanoparticles would be a useful reagent for the successful monitoring of isolated islets by magnetic resonance imaging and optical imaging systems, without clinically relevant toxicity in vitro or in vivo. METHODS: We used iron oxide for MR imaging and a cyanide dye approved by the Food and Drug Administration (indocyanine green) for optical imaging and estimated the in vivo detection of transplanted pancreatic islets. RESULTS: The poly(lactic-co-glycolic acid) nanoparticles were associated with the islets in vitro and were successfully detected by 4.7 T (MR) and optical imaging, without other toxic effects. When labeled islets were transplanted under the mouse kidney capsule, in vivo T2/ T2*-weighted scans with 4.7 T MR detected as few as 300 labeled islets by 4 weeks. Optical in vivo imaging revealed indocyanine green fluorescence by 2 and 4 days after transplantation of islets containing 250 and 500 µg/mL poly(lactic-co-glycolic acid) nanoparticles, respectively. These results were further supported by the immunohistochemical results for insulin and iron in the recipient mouse kidney and pancreas. CONCLUSIONS: Taken together, these data indicate that poly(lactic-co-glycolic acid) nanoparticles may be used to label transplanted islets and may be imaged with in vivo MR and optical imaging systems.


Subject(s)
Indocyanine Green , Islets of Langerhans Transplantation/methods , Islets of Langerhans/cytology , Lactic Acid/chemistry , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles , Polyglycolic Acid/chemistry , Animals , Cell Tracking/methods , Cells, Cultured , Diffusion , Image Enhancement/methods , Magnetite Nanoparticles/ultrastructure , Mice , Mice, Inbred C57BL , Microscopy/methods , Nanocapsules/chemistry , Nanocapsules/ultrastructure , Polylactic Acid-Polyglycolic Acid Copolymer , Reproducibility of Results , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...