Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Proc Natl Acad Sci U S A ; 119(7)2022 02 15.
Article in English | MEDLINE | ID: mdl-35140181

ABSTRACT

Foxp3-expressing CD25+CD4+ regulatory T cells (Tregs) are abundant in tumor tissues. Here, hypothesizing that tumor Tregs would clonally expand after they are activated by tumor-associated antigens to suppress antitumor immune responses, we performed single-cell analysis on tumor Tregs to characterize them by T cell receptor clonotype and gene-expression profiles. We found that multiclonal Tregs present in tumor tissues predominantly expressed the chemokine receptor CCR8. In mice and humans, CCR8+ Tregs constituted 30 to 80% of tumor Tregs in various cancers and less than 10% of Tregs in other tissues, whereas most tumor-infiltrating conventional T cells (Tconvs) were CCR8- CCR8+ tumor Tregs were highly differentiated and functionally stable. Administration of cell-depleting anti-CCR8 monoclonal antibodies (mAbs) indeed selectively eliminated multiclonal tumor Tregs, leading to cure of established tumors in mice. The treatment resulted in the expansion of CD8+ effector Tconvs, including tumor antigen-specific ones, that were more activated and less exhausted than those induced by PD-1 immune checkpoint blockade. Anti-CCR8 mAb treatment also evoked strong secondary immune responses against the same tumor cell line inoculated several months after tumor eradication, indicating that elimination of tumor-reactive multiclonal Tregs was sufficient to induce memory-type tumor-specific effector Tconvs. Despite induction of such potent tumor immunity, anti-CCR8 mAb treatment elicited minimal autoimmunity in mice, contrasting with systemic Treg depletion, which eradicated tumors but induced severe autoimmune disease. Thus, specific removal of clonally expanding Tregs in tumor tissues for a limited period by cell-depleting anti-CCR8 mAb treatment can generate potent tumor immunity with long-lasting memory and without deleterious autoimmunity.


Subject(s)
Immunologic Memory , Neoplasms/metabolism , Receptors, CCR8/metabolism , Animals , Antibodies, Monoclonal , Biomarkers, Tumor , Cell Differentiation , Cell- and Tissue-Based Therapy , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Deletion , Gene Expression Regulation, Neoplastic , Humans , Mice , Receptors, CCR8/genetics , T-Lymphocytes, Regulatory
2.
Blood ; 121(14): 2804-13, 2013 Apr 04.
Article in English | MEDLINE | ID: mdl-23365463

ABSTRACT

Monoclonal antibodies (mAbs) that are internalized into cells are a current focus in the development of antibody-drug conjugates (ADCs). We describe a phage display-based high-throughput screening system to rapidly isolate cell-internalizing mAbs. We simultaneously examined the cell-internalizing activities of several hundred independent mAbs and successfully isolated cell-internalizing mAbs against the tumor endothelial markers Roundabout homolog 4 (Robo4) and vascular endothelial growth factor receptor 2 (VEGFR2). Tumor accumulation of mAbs with high cell-internalizing activity was significantly higher than that of mAbs with low cell-internalizing activity. Furthermore, the antitumor effects of ADCs of mAbs with high cell-internalizing activity were significantly stronger than those of mAbs with low cell-internalizing activity. Although anti-VEGFR2 therapy caused a significant loss of body weight, anti-Robo4 therapy did not. These findings indicate that cell-internalizing activity plays an important role in the biodistribution and therapeutic effects of ADCs. Further, Robo4 can be an effective marker for tumor vascular targeting.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Immunoconjugates/pharmacokinetics , Melanoma/therapy , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Skin Neoplasms/therapy , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Biological Transport/immunology , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Cell Line, Transformed , Drug Design , Female , High-Throughput Screening Assays , Humans , Immunoconjugates/immunology , Melanoma/blood supply , Melanoma/immunology , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/therapy , Peptide Library , Receptors, Cell Surface , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Skin Neoplasms/blood supply , Skin Neoplasms/immunology , Tissue Distribution , Vascular Endothelial Growth Factor Receptor-2/immunology , Vascular Endothelial Growth Factor Receptor-2/metabolism
3.
Bioorg Med Chem ; 22(21): 6039-46, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25281270

ABSTRACT

NAD(P)H: quinone oxidoreductase 1 (NQO1) is an obligate two-electron reductase and is highly expressed in many human solid cancers. Because NQO1 can be induced immediately after exposure to ionizing radiation, we aimed to develop an NQO1-targeted radiolabeled agent to establish a novel internal radiation therapy that amplifies the therapeutic effects when combined with external radiation therapy. We designed three NQO1-targeted radioiodinated compounds including two ether linkage compounds ([(125)I]1 and [(125)I]2) and a sulfide linkage compound ([(125)I]3) based on the selective binding of indolequinone analogs to the active site of NQO1 by the stacking effect. These compounds were successfully prepared using an oxidative iododestannylation reaction with high radiochemical yields and purity. In NQO1-expressing tumor cells, [(125)I]1 and [(125)I]2 were readily metabolized to p-[(125)I]iodophenol or m-[(125)I]iodophenol and [(125)I]I(-), whereas over 85% of the initial radioactivity of [(125)I]3 was observed as an intact form at 1h after incubation. The cellular uptake of [(125)I]3 was significantly higher than those of [(125)I]1 and [(125)I]2. The uptake of [(125)I]3 was specific and was dependent on the expression of NQO1. These data suggest that the novel NQO1-targeted radioiodinated compound [(125)I]3 could be used as a novel internal radiation agent for the treatment of cancer.


Subject(s)
Indolequinones/chemistry , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neoplasms/radiotherapy , Radiopharmaceuticals/chemistry , Cell Line, Tumor , Halogenation , Humans , Indolequinones/chemical synthesis , Indolequinones/metabolism , Indolequinones/pharmacokinetics , Iodine Radioisotopes/chemistry , Iodine Radioisotopes/metabolism , Iodine Radioisotopes/pharmacokinetics , Neoplasms/metabolism , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/metabolism , Radiopharmaceuticals/pharmacokinetics , Radiotherapy/methods
4.
J Immunol ; 187(12): 6249-55, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22095716

ABSTRACT

We previously reported that poly (γ-glutamic acid)-based nanoparticles (γ-PGA NPs) are excellent vaccine carriers for inducing efficient cross-presentation in dendritic cells, thereby producing strong antitumor immunity in vivo. Analyzing the mechanism of cross-presentation induced by γ-PGA NPs will be useful toward designing novel vaccine carriers. In this study, we show an intracellular mechanism of efficient cross-presentation induced by OVA-loaded γ-PGA NPs. Cross-presentation induced by γ-PGA NPs depended on cytoplasmic proteasomes and TAP, similar to the classical MHC class I presentation pathway for endogenous Ags. Intracellular behavior analyzed by confocal laser scanning microscopy revealed that encapsulated OVA and γ-PGA accumulated in both the endoplasmic reticulum (ER) and endosome compartments within 2 h. At the same time, electron microscopy analysis clearly showed that intracellular γ-PGA NPs and encapsulated Au NPs were enveloped in endosome-like vesicles, not in the ER. These findings strongly suggest that γ-PGA NPs enhance ER-endosome fusion for cross-presentation. Moreover, inhibition of ER translocon sec61 significantly decreased the γ-PGA NP/OVA-mediated cross-presentation efficiency, indicating that sec61 is important for transporting Ags from the fused ER-endosome to the cytoplasm. These findings imply that the ER-endosome complex is key for the efficient cross-presentation of Ags encapsulated in γ-PGA NPs.


Subject(s)
Cancer Vaccines/immunology , Cross-Priming/immunology , Endoplasmic Reticulum/immunology , Endosomes/immunology , H-2 Antigens/immunology , Nanoparticles , Phenylalanine/analogs & derivatives , Polyglutamic Acid/pharmacology , Vaccines, DNA/immunology , Animals , Cancer Vaccines/chemical synthesis , Cancer Vaccines/genetics , Cells, Cultured , Cross-Priming/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endosomes/genetics , Endosomes/metabolism , Female , H-2 Antigens/genetics , H-2 Antigens/metabolism , Immunity, Cellular/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Phenylalanine/chemical synthesis , Phenylalanine/genetics , Phenylalanine/pharmacology , Polyglutamic Acid/chemical synthesis , Polyglutamic Acid/genetics , Vaccines, DNA/chemical synthesis , Vaccines, DNA/genetics
5.
Mol Cancer Ther ; 22(9): 1063-1072, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37420296

ABSTRACT

Although regulatory T cells (Treg) are inhibitory immune cells that are essential for maintaining immune homeostasis, Tregs that infiltrate tumor tissue promote tumor growth by suppressing antitumor immunity. Selective reduction of tumor-infiltrating Tregs is, therefore, expected to activate antitumor immunity without affecting immune homeostasis. We previously reported that selective Treg depletion targeted by a C-C motif chemokine receptor 8 (CCR8) resulted in induction of strong antitumor immunity without any obvious autoimmunity in mouse models. Thus, herein, we developed a novel humanized anti-CCR8 monoclonal antibody, S-531011, aimed as a cancer immunotherapy strategy for patients with cancer. S-531011 exclusively recognized human CCR8 among all chemokine receptors and showed potent antibody-dependent cell-mediated cytotoxicity activity toward CCR8+ cells and neutralization activity against CCR8-mediated signaling. We observed that S-531011 reduced tumor-infiltrating CCR8+ Tregs and induced potent antitumor activity in a tumor-bearing human-CCR8 knock-in mouse model. Moreover, combination therapy with S-531011 and anti-mouse programmed cell death 1 (PD-1) antibody strongly suppressed tumor growth compared with anti-PD-1 antibody alone with no observable adverse effects. S-531011 also depleted human tumor-infiltrating Tregs, but not Tregs derived from human peripheral blood mononuclear cells. These results suggest that S-531011 is a promising drug for inducing antitumor immunity without severe side effects in the clinical setting.


Subject(s)
Neoplasms , Receptors, Chemokine , Humans , Receptors, Chemokine/metabolism , T-Lymphocytes, Regulatory , Neoplasms/drug therapy , Immunity , Lymphocytes, Tumor-Infiltrating
6.
Sci Adv ; 9(24): eadf0661, 2023 06 16.
Article in English | MEDLINE | ID: mdl-37315144

ABSTRACT

Severe acute respiratory syndrome coronavirus 2-neutralizing antibodies primarily target the spike receptor binding domain (RBD). However, B cell antigen receptors (BCRs) on RBD-binding memory B (Bmem) cells have variation in the neutralizing activities. Here, by combining single Bmem cell profiling with antibody functional assessment, we dissected the phenotype of Bmem cell harboring the potently neutralizing antibodies in coronavirus disease 2019 (COVID-19)-convalescent individuals. The neutralizing subset was marked by an elevated CD62L expression and characterized by distinct epitope preference and usage of convergent VH (variable region of immunoglobulin heavy chain) genes, accounting for the neutralizing activities. Concordantly, the correlation was observed between neutralizing antibody titers in blood and CD62L+ subset, despite the equivalent RBD binding of CD62L+ and CD62L- subset. Furthermore, the kinetics of CD62L+ subset differed between the patients who recovered from different COVID-19 severities. Our Bmem cell profiling reveals the unique phenotype of Bmem cell subset that harbors potently neutralizing BCRs, advancing our understanding of humoral protection.


Subject(s)
B-Lymphocyte Subsets , COVID-19 , L-Selectin , Humans , Antibodies, Neutralizing , Antibodies, Viral , Epitopes , SARS-CoV-2
7.
Proc Biol Sci ; 279(1730): 959-66, 2012 Mar 07.
Article in English | MEDLINE | ID: mdl-21881138

ABSTRACT

How and why diverse sexual systems evolve are fascinating evolutionary questions, but few empirical studies have dealt with these questions in animals. Pedunculate (gooseneck) barnacles show such diversity, including simultaneous hermaphroditism, coexistence of dwarf males and hermaphrodites (androdioecy), and coexistence of dwarf males and females (dioecy). Here, we report the first phylogenetically controlled test of the hypothesis that the ultimate cause of the diverse sexual systems and presence of dwarf males in this group is limited mating opportunities for non-dwarf individuals, owing to mating in small groups. Within the pedunculate barnacle phylogeny, dwarf males and females have evolved repeatedly. Females are more likely to evolve in androdioecious than hermaphroditic populations, suggesting that evolution of dwarf males has preceded that of females in pedunculates. Both dwarf males and females are associated with a higher proportion of solitary individuals in the population, corroborating the hypothesis that limited mating opportunities have favoured evolution of these diverse sexual systems, which have puzzled biologists since Darwin.


Subject(s)
Adaptation, Physiological , Biological Evolution , Hermaphroditic Organisms/physiology , Thoracica/physiology , Animals , Bayes Theorem , Female , Likelihood Functions , Male , Phylogeny , Reproduction , Sexual Behavior, Animal , Sexual Maturation , Thoracica/genetics
8.
Bioconjug Chem ; 21(6): 1026-31, 2010 Jun 16.
Article in English | MEDLINE | ID: mdl-20446679

ABSTRACT

Gold/iron oxide magnetic nanoparticles are hybrid nanoparticles containing a core of magnetic iron oxide and surface colloidal gold, which allows for various biomaterials to be immobilized on the surface of the iron oxide nanoparticles via colloidal gold. Here, we developed a novel magnetic resonance (MR) imaging agent to broaden the MR tumor-imaging spectrum of superparamagnetic iron oxide nanoparticles (SPIO), e.g., Feridex(), a clinical MR imaging agent for diagnosing liver cancer. Au/Feridex was synthesized by electron beam irradiation, and thiol-modified poly(ethylene glycol) (PEG-SH) was easily conjugated to its surface via an Au-S bond without the need for any chemical reactions. PEG conjugation of Au/Feridex enhanced its accumulation in Meth-A tumor tissue and decreased its accumulation in normal liver tissue. In addition, MRI using PEG-Au/Feridex, in contrast to MRI using unmodified Au/Feridex and Feridex, detected B16BL6 and Meth-A tumor tissues in vivo. This finding indicates that PEG-Au/Feridex is useful for diagnosing various types of tumors. In addition, because the synthesis of PEG-Au/Feridex is simple and high yields are easily produced, PEG-modified SPIO for tumor diagnosis can be prepared on an industrial scale with low cost.


Subject(s)
Ferrosoferric Oxide/chemistry , Gold/chemistry , Magnetic Resonance Imaging/methods , Neoplasms/pathology , Polyethylene Glycols/chemistry , Sulfur/chemistry , Animals , Cell Line, Tumor , Dextrans , Ferric Compounds/chemistry , Ferrosoferric Oxide/pharmacokinetics , Gold/pharmacokinetics , Liver Neoplasms/pathology , Magnetite Nanoparticles , Melanoma/pathology , Metal Nanoparticles/chemistry , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Polyethylene Glycols/pharmacokinetics
9.
Protein Expr Purif ; 61(1): 78-82, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18571431

ABSTRACT

Magic roundabout (Robo4) is the fourth recently identified member of the roundabout receptor family. Robo4 is predominantly expressed in embryonic or tumor vascular endothelium and is considered important for vascular development and as a candidate tumor endothelial marker. Much remains unknown about the Robo4 molecule, however, such as its ligands, structure, and the details of its function. Thus, we aimed to establish an expression and purification method for obtaining soluble recombinant human Robo4 (shRobo4) and mouse Robo4 (smRobo4) for use in Robo4 characterization studies. In this work, we expressed the extracellular domain of hRobo4 and mRobo4 in mammalian 293F cells and purified them by two-step chromatography. Based on gel-filtration chromatography and Blue Native polyacrylamide gel electrophoresis, these purified proteins exist as multimers. The shRobo4 and smRobo4 we obtained will be useful in advanced studies to determine the importance of multimerization, identify the ligands, and elucidate the ligand-receptor interactions and Robo4-mediated signaling. The results of these studies will help to elucidate the role of Robo4 in angiogenesis and perhaps eventually contribute to the development of novel vessel-targeting therapies.


Subject(s)
Biomarkers, Tumor/metabolism , Receptors, Cell Surface/metabolism , Base Sequence , Blotting, Western , Cell Line , Chromatography, Gel , DNA Primers , Electrophoresis, Polyacrylamide Gel , Humans , Ligands , Neovascularization, Pathologic , Receptors, Cell Surface/isolation & purification , Solubility
10.
Bioorg Med Chem ; 16(11): 6027-33, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18479926

ABSTRACT

Malaria is a leading cause of death in developing countries, and the emergence of strains resistant to the main therapeutic agent, chloroquine, has become a serious problem. We have developed cationic-dimer type antimalarials, MAP-610 and PMAP-H10, which are structurally different from chloroquine. In this study, we introduced several substituents on the terminal phenyl rings of PMAP-H10. The electronic and hydrophobic properties of the substituents were correlated with the antimalarial activity and cytotoxicity of the compounds, respectively. Studies with synchronized cultures of malarial plasmodia showed that our cationic-dimers act selectively between the schizont stage and the ring stage of the parasitic cycle, unlike chloroquine, which has a stage-independent action. Thus, the mechanism of action of our antimalarials appears to be different from that of chloroquine, and our compounds may be effective against chloroquine-resistant strains.


Subject(s)
Antimalarials/chemistry , Antimalarials/toxicity , Chloroquine/toxicity , Plasmodium falciparum/drug effects , Plasmodium falciparum/growth & development , Pyridinium Compounds/chemistry , Pyridinium Compounds/toxicity , Animals , Cations , Cell Line, Tumor , Chloroquine/chemistry , Dimerization , Drug Resistance , Erythrocytes/drug effects , Erythrocytes/parasitology , Humans , Inhibitory Concentration 50 , Mice , Parasitemia/drug therapy , Parasitemia/parasitology , Structure-Activity Relationship
11.
Org Lett ; 20(6): 1589-1592, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29488386

ABSTRACT

A novel cascade reaction of indole-2,3-epoxide equivalents with γ-carbolines by utilizing a double "open and shut" transformation to access multiheterocyclic compounds containing both isotryptamines and pyrimido[1,6- a]indoles has been developed. This strategy utilizes the in situ formation of a bulky quaternary ammonium salt via ammonium exchange, which undergoes Hofmann elimination/vinylogous Mannich/retro-Mannich/cyclization cascade sequences.

12.
Clin Case Rep ; 6(9): 1829-1833, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30214773

ABSTRACT

Sjögren's syndrome concurrent with protein-losing gastroenteropathy can develop into secondary systemic capillary leak syndrome. Thus, it is important to diagnose the condition as soon as possible and simultaneously administer treatment for Sjögren's syndrome, protein-losing gastroenteropathy, and systemic capillary leak syndrome.

13.
J Biosci Bioeng ; 111(5): 597-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21277253

ABSTRACT

Immune phage antibody libraries are an attractive technology for isolating antigen-specific monoclonal antibodies (mAbs). Here we show that the immunization schedule affects the immune phage antibody library properties. We subcutaneously (s.c.) administered HIV-1 Nef and Vif antigens with different schedules (25 µg × 2 s.c. and 10 µg × 3 s.c.). The variety of isolated mAbs in 25 µg × 2 s.c. groups (Nef: 11 clones, Vif: 9 clones) was superior to that in the 10 µg × 3 s.c. groups (Nef: 2 clones, Vif: 1 clone). This finding suggests that it is important to optimize the immunization schedule for isolating a wide variety of mAbs.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Immunization Schedule , Peptide Library , nef Gene Products, Human Immunodeficiency Virus/immunology , vif Gene Products, Human Immunodeficiency Virus/immunology , Animals , Antibodies, Monoclonal/immunology , Cloning, Molecular , Female , HIV Antibodies/biosynthesis , HIV Antibodies/blood , HIV Antibodies/immunology , HIV-1/immunology , Mice , Mice, Inbred BALB C , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/immunology
14.
Sci Signal ; 3(148): ra83, 2010 Nov 16.
Article in English | MEDLINE | ID: mdl-21081755

ABSTRACT

Tumor necrosis factor (TNF) is an inflammatory cytokine that has important roles in various immune responses, which are mediated through its two receptors, TNF receptor 1 (TNFR1) and TNFR2. Antibody-based therapy against TNF is used clinically to treat several chronic autoimmune diseases; however, such treatment sometimes results in serious side effects, which are thought to be caused by the blocking of signals from both TNFRs. Therefore, knowledge of the structural basis for the recognition of TNF by each receptor would be invaluable in designing TNFR-selective drugs. Here, we solved the 3.0 angstrom resolution structure of the TNF-TNFR2 complex, which provided insight into the molecular recognition of TNF by TNFR2. Comparison to the known TNFR1 structure highlighted several differences between the ligand-binding interfaces of the two receptors. Additionally, we also demonstrated that TNF-TNFR2 formed aggregates on the surface of cells, which may be required for signal initiation. These results may contribute to the design of therapeutics for autoimmune diseases.


Subject(s)
Receptors, Tumor Necrosis Factor, Type II/chemistry , Tumor Necrosis Factors/chemistry , Drug Design , Models, Molecular , Molecular Structure , Solutions
15.
Article in Japanese | MEDLINE | ID: mdl-20601834

ABSTRACT

A 30-year-old pregnant woman experienced mild dyspnea in April 2009. She complained of mild myalgia and was subsequently admitted to our hospital in June 2009 because of worsening dyspnea. Physical examination revealed fine crackles in the lower lung field, but no eruptions externally. Laboratory findings revealed elevated serum levels of myogenic enzymes (aldolase, 17.6 IU/l and myoglobin, 247.2 ng/ml) and positive titers for the anti-Jo-1 antibody and hypoxia (PaO(2), 79.4 Torr). The chest radiograph revealed a ground-glass opacity. The patient was diagnosed as interstitial pneumonia (IP) associated with polymyositis (PM) at 20 weeks of gestation. On July 9, we commenced the initial treatment-steroid pulse therapy with 60 mg/day of prednisolone and 3 mg/day of tacrolimus. We also induced abortion. The treatment of corticosteroids and tacrolimus was, however, ineffective even after increasing the tacrolimus dose to 6 mg/day. On July 30, she suddenly experienced chest pain along with severe dyspnea. Computed tomography revealed the presence of pneumomediastinum and deterioration of the IP. We added cyclophosphamide pulse therapy to the existing regimen ; this improved the disease course, reduced hypoxia, and improved radiographic findings. We believe that this is a rare case of IP with PM during pregnancy.


Subject(s)
Cyclophosphamide/administration & dosage , Immunosuppressive Agents/administration & dosage , Lung Diseases, Interstitial/drug therapy , Mediastinal Emphysema/drug therapy , Polymyositis/complications , Pregnancy Complications/drug therapy , Tacrolimus/administration & dosage , Adult , Drug Therapy, Combination , Female , Humans , Pregnancy , Pulse Therapy, Drug
16.
Biomaterials ; 30(9): 1809-14, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19136151

ABSTRACT

Gold/iron-oxide MAgnetic Nanoparticles (GoldMAN) imparts useful magnetic properties to various biomolecules. Gold nanoparticles immobilized on the surface of magnetic nanoparticles allow for the conjugation of biomolecules via an Au-S bond. Here, we present a practical application by utilizing GoldMAN and a magnetic field to induce intracellular transduction. This method has great potential for application of the adenovirus gene delivery vector (Ad), widely used for in vitro/in vivo gene transfer, to Ad-resistant cells. We demonstrated that Ad was easily immobilized on GoldMAN and the Ad/GoldMAN complex was introduced into the cell by the magnetic field, which increased gene expression over 1000 times that of Ad alone. The GoldMAN penetrated the plasma membrane directly, independent of the cell-surface virus receptors and endocytosis pathway. This mechanism will contribute to improve the gene expression efficiency of Ad. This technology is a useful tool for extending Ad tropism and enhancing transduction efficiency. GoldMAN also makes possible the effective use of various biomolecules within the cell because of its interesting cell-entry mechanism.


Subject(s)
Adenoviridae/genetics , Ferric Compounds/metabolism , Gene Transfer Techniques , Gold/metabolism , Magnetics , Nanoparticles/chemistry , Animals , Cell Line, Tumor , Humans , Intracellular Space/metabolism , Mice , Microscopy, Confocal , Nanoparticles/ultrastructure , Transduction, Genetic
17.
ChemMedChem ; 2(10): 1527-32, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17607684

ABSTRACT

Malaria is one of the three major serious infectious diseases in the world. As the area affected by malaria includes a large proportion of developing countries, there is a need for new antimalarials that can be synthesized and supplied inexpensively. To generate low-cost antimalarials, the MAP series 6-10, bis-cation dimers, synthesized by amidating the carboxyl group of isonicotinic acid (11) with various amines and by cationizing the nitrogen atoms of the pyridine ring with the corresponding alkyl bromides, were designed. This design enabled expansion of the structural variations of bis-cation-type antimalarial compounds. The compounds bearing alkyl or phenyl groups in the amide moieties showed remarkable antimalarial activities in vitro. Moreover, 1,1'-(1,12-dodecanediyl)bis[4-[(buthylamino)carbonyl]pyridinium bromide], MAP-412 (6 d), exhibited a potent antimalarial activity (ED(50)=8.2 mg kg(-1)). Being prepared at low cost, our bis-cation-type antimalarial compounds may be useful as lead compounds for inexpensive antimalarials.


Subject(s)
Antimalarials/chemical synthesis , Isonicotinic Acids/chemistry , Antimalarials/chemistry , Cations , Dimerization , Isonicotinic Acids/economics
SELECTION OF CITATIONS
SEARCH DETAIL