Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 616: 140-144, 2022 08 06.
Article in English | MEDLINE | ID: mdl-35679696

ABSTRACT

Although modulation of claudin-1-based tight junction (TJ) in stratum granulosum is an option for transdermal absorption of drugs, granular permeation enhancers have never been developed. We previously found that homoharringtonine (HHT), a natural alkanoid, weakened intestinal epithelial barrier with changing expression and cellular localization of TJ components such as claudin-1 and claudin-4. In the present study, we investigated whether HHT is an epidermal granular permeation enhancer. Treatment of normal human epidermal keratinocytes (NHEK) cells with HHT decreased claudin-1 and claudin-4 but not zonula occludens-1 and E-cadherin. HHT lowered TJ-integrity in NHEK cells, accompanied by permeation-enhancement of dextran (4 kDa) in a dose-dependent manner. Transdermal treatment of mice with HHT weakened epidermal barrier. HHT treatment enhanced transdermal absorption of dextran with a molecular mass of up to 10 kDa. Together, HHT may be a transdermal absorption enhancer.


Subject(s)
Dextrans , Homoharringtonine , Tight Junctions , Animals , Claudin-1/metabolism , Claudin-4/metabolism , Dextrans/metabolism , Homoharringtonine/pharmacology , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Tight Junctions/metabolism
2.
J Control Release ; 283: 126-134, 2018 08 10.
Article in English | MEDLINE | ID: mdl-29753959

ABSTRACT

Within the field of RNA therapeutics, antisense oligonucleotide-based therapeutics are a potentially powerful means of treating intractable diseases. However, if these therapeutics are used for the treatment of neurological disorders, safe yet efficient methods of delivering antisense oligonucleotides across the blood-brain barrier to the central nervous system must be developed. Here, we examined the use of angubindin-1, a binder to the tricellular tight junction, to modulate paracellular transport between brain microvascular endothelial cells in the blood-brain barrier for the delivery of antisense oligonucleotides to the central nervous system. This proof-of-concept study demonstrated that intravenously injected angubindin-1 increased the permeability of the blood-brain barrier and enabled transient delivery of subsequently administered antisense oligonucleotides into the mouse brain and spinal cord, leading to silencing of a target RNA without any overt adverse effects. We also found that two bicellular tight junction modulators did not produce such a silencing effect, suggesting that the tricellular tight junction is likely a better target for the delivery of antisense oligonucleotides than the bicellular tight junction. Our delivery strategy of modulating the tricellular tight junction in the blood-brain barrier via angubindin-1 provides a novel avenue of research for the development of antisense oligonucleotide-based therapeutics for the treatment of neurological disorders.


Subject(s)
Bacterial Toxins/pharmacology , Blood-Brain Barrier/drug effects , Oligonucleotides, Antisense/metabolism , Tight Junctions/metabolism , Animals , Bacterial Toxins/administration & dosage , Blood-Brain Barrier/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enterotoxins/administration & dosage , Female , Mice, Inbred C57BL , Oligonucleotides, Antisense/administration & dosage , RNA, Long Noncoding/genetics , Rats , Receptors, Lipoprotein/metabolism
3.
Eur J Pharm Sci ; 117: 161-167, 2018 May 30.
Article in English | MEDLINE | ID: mdl-29448044

ABSTRACT

Claudin-2 (CLDN-2), a pore-forming tight junction protein with a tetra-transmembrane domain, is involved in carcinogenesis and the metastasis of some cancers. Although CLDN-2 is highly expressed in the tight junctions of the liver and kidney, whether CLDN-2 is a safe target for cancer therapy remains unknown. We recently generated a rat monoclonal antibody (mAb, clone 1A2) that recognizes the extracellular domains of human and mouse CLDN-2. Here, we investigated the safety of CLDN-2-targeted cancer therapy by using 1A2 as a model therapeutic antibody. Because most human therapeutic mAbs are IgG1 subtype that can induce antibody-dependent cellular cytotoxicity, we generated a human-rat chimeric IgG1 form of 1A2 (xi-1A2). xi-1A2 activated Fcγ receptor IIIa in the presence of CLDN-2-expressing cells, indicating that xi-1A2 likely exerts antibody-dependent cellular cytotoxicity. At 24 h after its intravenous injection, xi-1A2 was distributed into the liver, kidney, and tumor tissues of mice bearing CLDN-2-expressing fibrosarcoma cells. Treatment of the xenografted mice with xi-1A2 attenuated tumor growth without apparent adverse effects, such as changes in body weight and biochemical markers of liver and kidney injury. These results support xi-1A2 as the lead candidate mAb for safe CLDN-2-targeted cancer therapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Claudin-2/immunology , Neoplasms/drug therapy , Protein Domains/immunology , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Female , Humans , Immunoglobulin G/immunology , Jurkat Cells , Kidney/metabolism , Liver/metabolism , Mice, Inbred BALB C , Neoplasms/immunology , Neoplasms/metabolism , Rats , Receptors, IgG/genetics , Receptors, IgG/immunology
4.
J Nat Med ; 72(2): 448-455, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29275476

ABSTRACT

Lentinula edodes mycelia solid culture extract (MSCE) is used as a medical food ingredient and provides beneficial effects to patients with cancer and chronic type C hepatitis. Low molecular weight lignin (LM-lignin), which is an active component of MSCE, exhibits hepatoprotective, antitumor, antiviral, and immunomodulatory effects. In this study, we investigated the effect of LM-lignin/lignosulfonic acid on intestinal barrier function. Lignosulfonic acid enhanced transepithelial membrane electrical resistance in human intestinal Caco-2 cell monolayers. In Caco-2 cells treated with lignosulfonic acid, expression of claudin-2, which forms high conductive cation pores in tight junctions (TJs), was decreased. Lignosulfonic acid also attenuated the barrier dysfunction that is caused by tumor necrosis factor (TNF)-α and interferon (IFN)-γ in Caco-2 cells. TNF-α- and IFN-γ-induced activation of NF-κB, such as translocation of NF-κB p65 into the nucleus and induction of gene expression, was inhibited by lignosulfonic acid treatment. Furthermore, lignosulfonic acid decreased the TNF-α- and IFN-γ-induced increase in interleukin (IL)-1ß and IL-6 expression in Caco-2 cells. These results suggest that lignosulfonic acid not only enhances TJ barrier function but also restores TJ barrier integrity impaired by inflammatory cytokines. Therefore, lignosulfonic acid may be beneficial for the treatment of inflammation-induced intestinal barrier dysfunction observed in inflammatory bowel disease.


Subject(s)
Epithelial Cells/drug effects , Intestinal Mucosa/drug effects , Lignin/analogs & derivatives , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , Caco-2 Cells , Cell Line, Tumor , Humans , Lignin/pharmacology , Lignin/therapeutic use , Signal Transduction , Transfection
5.
Sci Rep ; 7(1): 14514, 2017 11 06.
Article in English | MEDLINE | ID: mdl-29109448

ABSTRACT

Claudins are key functional and structural components of tight junctions (TJs) in epithelial cell sheets. The C-terminal fragment of Clostridium perfringens enterotoxin (C-CPE) binds to claudin-4 and reversibly modulates intestinal TJ seals, thereby enhancing paracellular transport of solutes. However, the use of C-CPE as an absorption enhancer is limited by the molecule's immunogenicity and manufacturing cost. Here, we developed a high-throughput screening system based on the Time-Resolved Fluorescence Resonance Energy Transfer (TR-FRET) method to identify claudin-4 binders in a library collection of 32,560 compounds. Thiostrepton, identified from the screen, decreased transepithelial electrical resistance and increased flux of 4-kDa fluorescein isothiocyanate-labelled dextran (FD-4) in Caco-2 cell monolayers, a model of intestinal epithelium. Thiostrepton changed the expression, but not the localisation, of TJ components. Treatment of rat jejunum with thiostrepton increased the absorption of FD-4 without tissue toxicity, indicating that thiostrepton is a novel claudin-4 binder that enhances intestinal permeability. The screening system may therefore be a useful tool for identifying claudin-4 binders to enhance drug absorption in mucosa.


Subject(s)
Claudin-4/metabolism , Enterotoxins/pharmacology , Thiostrepton/pharmacology , Tight Junctions/drug effects , Tight Junctions/metabolism , Animals , Caco-2 Cells , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Electric Impedance , Fluorescence Resonance Energy Transfer , High-Throughput Screening Assays , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Jejunum/drug effects , Jejunum/metabolism , L-Lactate Dehydrogenase/metabolism , Male , Rats, Wistar , Recombinant Proteins/metabolism
6.
J Pharmacol Exp Ther ; 363(3): 444-451, 2017 12.
Article in English | MEDLINE | ID: mdl-28928120

ABSTRACT

Disruption of the gastrointestinal epithelial barrier is a hallmark of chronic inflammatory bowel diseases (IBDs). The transmembrane protein claudin 2 (CLDN2) is a component of epithelial tight junctions (TJs). In the intestines of patients with IBDs, the expression of the pore-forming TJ protein CLDN2 is upregulated. Although CLDN2 is involved in these leaky barriers, whether it can be a target to enhance TJ integrity is unknown because a CLDN2-specific inhibitor has not been developed. Here, we used DNA immunization to generate a monoclonal antibody (mAb) that recognized an extracellular loop of CLDN2. Treatment of epithelial cell monolayers with the mAb increased barrier integrity. In addition, the anti-CLDN2 mAb attenuated the decrease in TJ integrity induced by the proinflammatory cytokine tumor necrosis factor-α (TNF-α), and cotreatment of cells with anti-TNF-α mAb and anti-CLDN2 mAb showed additive attenuating effects. These findings indicate that CLDN2 may be a target for enhancing TJ integrity, and CLDN2 binder may be an enhancer of mucosal barrier integrity and a potential therapeutic option for IBDs.


Subject(s)
Claudins/metabolism , Inflammatory Bowel Diseases/metabolism , Tight Junctions/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Cell Line, Tumor , Claudins/immunology , Female , Humans , Inflammatory Bowel Diseases/therapy , Intestinal Mucosa/metabolism , Mice , Mice, Inbred BALB C , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
7.
J Pharmacol Exp Ther ; 363(2): 275-283, 2017 11.
Article in English | MEDLINE | ID: mdl-28819070

ABSTRACT

A current bottleneck in the development of central nervous system (CNS) drugs is the lack of drug delivery systems targeting the CNS. The intercellular space between endothelial cells of the blood-brain barrier (BBB) is sealed by complex protein-based structures called tight junctions (TJs). Claudin-5 (CLDN-5), a tetra-transmembrane protein is a key component of the TJ seal that prevents the paracellular diffusion of drugs into the CNS. In the present study, to investigate whether CLDN-5 binders can be used for delivery of drugs to the CNS, we generated monoclonal antibodies (mAbs) specific to the extracellular domains of CLDN-5. In an in vitro model of the BBB, the anti-CLDN-5 mAbs attenuated trans-epithelial/endothelial electrical resistance and enhanced solute permeation. These anti-CLDN-5 mAbs are potential leads for the development of novel drug delivery systems targeting the CNS.


Subject(s)
Blood-Brain Barrier/metabolism , Claudin-5/metabolism , Animals , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Claudin-5/chemistry , Claudin-5/immunology , Extracellular Space/metabolism , Female , Humans , Male , Mice , Permeability , Protein Domains , Tight Junctions/metabolism
8.
J Control Release ; 260: 1-11, 2017 08 28.
Article in English | MEDLINE | ID: mdl-28528740

ABSTRACT

A limiting barrier for mucosal absorption of drugs is the tight junction (TJ). TJs exist between two adjacent cells (bicellular TJ, bTJ) and at the sites where three cells meet (tricellular TJ, tTJ). We present a novel approach which employs a physiologically regulated pathway for the passage of large molecules through the tTJ. Main barrier-relevant tTJ proteins are tricellulin and angulin-1 to -3. We developed an angulin binder from Clostridium perfringens iota-toxin (Ib) whose receptor is angulin-1. An Ib fragment corresponding to amino acids 421-664 (Ib421-664) of iota-toxin proved to bind in cells expressing angulin-1 and -3, but not angulin-2. This binding led to removal of angulin-1 and tricellulin from the tTJ which enhanced the permeation of macromolecular solutes. Ib421-664 enhanced intestinal absorption in rats and mice. Our findings indicate that Ib421-664, which we designate angubindin-1, is a modulator of the tTJ barrier and that modulation of that barrier qualifies for a new strategy of developing a mucosal absorption enhancer.


Subject(s)
ADP Ribose Transferases/chemistry , Bacterial Toxins/chemistry , Intestinal Absorption , MARVEL Domain Containing 2 Protein/metabolism , Peptide Fragments/pharmacology , Receptors, Cell Surface/metabolism , Animals , Cell Line , Humans , Male , Mice , Rats, Wistar , Tight Junctions/metabolism
9.
Cell Physiol Biochem ; 41(5): 1924-1934, 2017.
Article in English | MEDLINE | ID: mdl-28391269

ABSTRACT

BACKGROUND/AIMS: Although proinflammatory cytokine-induced disruption of intestinal epithelial barrier integrity is associated with intestinal inflammatory disease, effective treatment for barrier dysfunction is lacking. Previously, we demonstrated that rebeccamycin alleviates epithelial barrier dysfunction induced by inflammatory cytokines in Caco-2 cell monolayers; however, the underlying mechanism remained unclear. Here, we investigated the mechanism by which rebeccamycin protects the epithelial barrier function of Caco-2 cells exposed to TNF-α. METHODS: To confirm the epithelial barrier function of Caco-2 cell monolayers, transepithelial electrical resistance (TER) and paracellular permeability were measured. Production levels and localization of tight junction (TJ) proteins were analyzed by immunoblot and immunofluorescence, respectively. Phosphorylated myosin light chain (pMLC) and MLC kinase (MLCK) mRNA expression levels were determined by immunoblot and quantitative RT-PCR, respectively. RESULTS: Rebeccamycin attenuated the TNF-α-induced reduction in TER and increase in paracellular permeability. Rebeccamycin increased claudin-5 expression, but not claudin-1, -2, -4, occludin or ZO-1 expression, and prevented the TNF-α-induced changes in ZO-1 and occludin localization. Rebeccamycin suppressed the TNF-α-induced increase in MLCK mRNA expression, thus suppressing MLC phosphorylation. The rebeccamycin-mediated reduction in MLCK production and protection of epithelial barrier function were alleviated by Chk1 inhibition. CONCLUSION: Rebeccamycin attenuates TNF-α-induced disruption of intestinal epithelial barrier integrity by inducing claudin-5 expression and suppressing MLCK production via Chk1 activation.


Subject(s)
Carbazoles/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Intestinal Mucosa/enzymology , Myosin-Light-Chain Kinase/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology , Caco-2 Cells , Checkpoint Kinase 1/metabolism , Claudin-5/biosynthesis , Enzyme Activation/drug effects , Humans , Myosin Light Chains/metabolism , Phosphorylation/drug effects , Tight Junctions/enzymology
10.
Pharmacol Res Perspect ; 4(5): e00266, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27713828

ABSTRACT

Claudin-4 (CLDN-4), a tight-junction protein, is overexpressed in various malignant tumors, including gastric, colorectal, pancreatic, and breast cancers. However, CLDN-4 is also expressed in normal tissues, including the liver, pancreas, kidney, and small intestine. Whether CLDN-4 is an effective and safe target for cancer therapy has been unclear owing to the lack of a binder with both CLDN-4 specificity and cross-reactivity to human and murine cells. In this study, we successfully generated a rat anti-CLDN-4 monoclonal antibody (5D12) that was specific to, and cross-reactive with, human and mouse CLDN-4. 5D12 recognized the second extracellular domain of human CLDN-4 in a conformation-dependent manner. A human-rat chimeric IgG1 of 5D12 (xi-5D12) activated the Fcγ IIIa receptor, indicating the activation of antibody-dependent cellular cytotoxicity in CLDN-4-expressing cells. Moreover, xi-5D12 significantly suppressed tumor growth in mice bearing human colorectal and gastric tumors without apparent adverse effects, such as weight loss or liver and kidney damage. These results suggest that CLDN-4 is a potent target for cancer therapy and that an anti-CLDN-4 antibody is a promising candidate anticancer agent.

11.
Biochem Biophys Res Commun ; 477(1): 91-95, 2016 08 12.
Article in English | MEDLINE | ID: mdl-27286708

ABSTRACT

Claudin-1 (CLDN-1), an integral transmembrane protein, is an attractive target for drug absorption, prevention of infection, and cancer therapy. Previously, we generated mouse anti-CLDN-1 monoclonal antibodies (mAbs) and found that they enhanced epidermal absorption of a drug and prevented hepatitis C virus infection in human hepatocytes. Here, we investigated anti-tumor activity of a human-mouse chimeric IgG1, xi-3A2, from one of the anti-CLDN-1 mAbs, clone 3A2. Xi-3A2 accumulated in the tumor tissues in mice bearing with human CLDN-1-expressing tumor cells. Xi-3A2 activated Fcγ receptor IIIa-expressing reporter cells in the presence of human CLDN-1-expressing cells, suggesting xi-3A2 has a potential to exhibit antibody-dependent cellular cytotoxicity against CLDN-1 expressing tumor cells. We also constructed a mutant xi-3A2 antibody with Gly, Ser, and Ile substituted with Ala, Asp, and Arg at positions 236, 239, and 332 of the Fc domain. This mutant antibody showed greater activation of Fcγ receptor IIIa and in vivo anti-tumor activity in mice bearing human CLDN-1-expressing tumors than xi-3A2 did. These findings indicate that the G236A/S239D/I332E mutant of xi-3A2 might be a promising lead for tumor therapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Claudin-1/immunology , Disease Models, Animal , Neoplasms/therapy , Animals , Antibody-Dependent Cell Cytotoxicity , Cell Line, Tumor , Chimera , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays
12.
PLoS One ; 11(1): e0145631, 2016.
Article in English | MEDLINE | ID: mdl-26727128

ABSTRACT

Several stressors are known to influence epithelial tight junction (TJ) integrity, but the association between DNA damage and TJ integrity remains unclear. Here we examined the effects of daunorubicin and rebeccamycin, two anti-tumor chemicals that induce DNA damage, on TJ integrity in human intestinal epithelial cells. Daunorubicin and rebeccamycin dose-dependently enhanced transepithelial electrical resistance (TER) and decreased flux of the 4 kDa FITC-dextran in Caco-2 cell monolayer. Daunorubicin- or rebeccamycin-induced enhancement of the TJ barrier function partly rescued attenuation of the barrier function by the inflammatory cytokines TNF-α and IFN-γ. Daunorubicin and rebeccamycin increased claudin-5 expression and the product was distributed in the actin cytoskeleton fraction, which was enriched with TJ proteins. Caffeine, which is an inhibitor of ataxia telangiectasia mutated protein (ATM) and ataxia telangiectasia mutated and Rad3-related protein (ATR), and the Chk1 inhibitor inhibited the TER increases induced by daunorubicin and rebeccamycin, whereas a Chk2 inhibitor did not. Treatment with Chk1 siRNA also significantly inhibited the TER increases. Induction of claudin-5 expression was inhibited by Chk1 inhibitor and by siRNA treatment. Our results suggest that Chk1 activation by daunorubicin and rebeccamycin induced claudin-5 expression and enhanced TJ barrier function in Caco-2 cell monolayer, which suggests a link between DNA damage and TJ integrity in the human intestine.


Subject(s)
Claudin-5/genetics , Gene Expression Regulation, Neoplastic/drug effects , Intestinal Mucosa/physiology , Protein Kinases/metabolism , Caco-2 Cells , Carbazoles/pharmacology , Checkpoint Kinase 1 , Daunorubicin/pharmacology , Enzyme Activation , Humans
13.
J Pharmacol Exp Ther ; 354(3): 440-7, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26139628

ABSTRACT

Tight junctions (TJs) are complex biochemical structures that seal the intercellular space and prevent the free movement of solutes across epithelial cell sheets. Modulating the TJ seal is a promising option for increasing the transdermal absorption of drugs. Within TJs, the binding of the claudin (CLDN) family of tetratransmembrane proteins through cis- and trans-interactions is an integral part of seal formation. Because epidermal TJs contain CLDN-1 and CLDN-4, a binder for these CLDNs may be a useful modulator of the permeability of the epidermal barrier. Here, we investigated whether m19, which can bind to CLDN-1/-4 (also CLDN-2/-5), modulates the integrity of epidermal TJs and the permeability of cell sheets to solutes. Treatment of normal human epidermal keratinocytes (NHEKs) with the CLDN binder reduced the integrity of TJs. A CLDN-1-specific binder (a monoclonal antibody, clone 7A5) also weakened the TJ seal in NHEKs. Although m19 attenuated the TJ barrier in human intestinal epithelial cells (Caco-2), 7A5 did not. Treatment of NHEKs with 7A5 enhanced permeation of a paracellular permeation marker. These findings indicate that CLDN-1 is a potential target for modulating the permeability of the epidermis, and that our CLDN-1 binder is a promising candidate molecule for development as a dermal absorption enhancer.


Subject(s)
Claudin-1/metabolism , Keratinocytes/metabolism , Protein Binding/physiology , Caco-2 Cells , Cell Line, Tumor , Claudin-4/metabolism , Epidermis/metabolism , Epithelial Cells/metabolism , Humans , Membrane Proteins/metabolism , Permeability , Tight Junctions/metabolism
14.
Biochem Biophys Res Commun ; 462(1): 52-7, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25935479

ABSTRACT

Lentinula edodes mycelia solid culture extract (MSCE) contains several bioactive molecules, including some polyphenolic compounds, which exert immunomodulatory, antitumor, and hepatoprotective effects. In this study, we examined the anti-hepatitis C virus (HCV) activity of MSCE and low-molecular-weight lignin (LM-lignin), which is the active component responsible for the hepatoprotective effect of MSCE. Both MSCE and LM-lignin inhibited the entry of two HCV pseudovirus (HCVpv) types into Huh7.5.1 cells. LM-lignin inhibited HCVpv entry at a lower concentration than MSCE and inhibited the entry of HCV particles in cell culture (HCVcc). MSCE also inhibited HCV subgenome replication. LM-lignin had no effect on HCV replication, suggesting that MSCE contains additional active substances. We demonstrate here for the first time the anti-HCV effects of plant-derived LM-lignin and MSCE. The hepatoprotective effect of LM-lignin suggests that lignin derivatives, which can be produced in abundance from existing plant resources, may be effective in the treatment of HCV-related diseases.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/drug effects , Lignin/pharmacology , Shiitake Mushrooms/chemistry , Antiviral Agents/isolation & purification , Cell Line, Tumor , Dose-Response Relationship, Drug , Hepacivirus/genetics , Hepacivirus/physiology , Humans , Lignin/chemistry , Lignin/isolation & purification , Molecular Weight , Virion/drug effects , Virion/physiology , Virus Internalization/drug effects , Virus Replication/drug effects , Virus Replication/genetics
15.
PLoS One ; 10(5): e0126352, 2015.
Article in English | MEDLINE | ID: mdl-26018248

ABSTRACT

Efficient vaccine delivery to mucosal tissues including mucosa-associated lymphoid tissues is essential for the development of mucosal vaccine. We previously reported that claudin-4 was highly expressed on the epithelium of nasopharynx-associated lymphoid tissue (NALT) and thus claudin-4-targeting using C-terminal fragment of Clostridium perfringens enterotoxin (C-CPE) effectively delivered fused antigen to NALT and consequently induced antigen-specific immune responses. In this study, we applied the C-CPE-based vaccine delivery system to develop a nasal pneumococcal vaccine. We fused C-CPE with pneumococcal surface protein A (PspA), an important antigen for the induction of protective immunity against Streptococcus pneumoniae infection, (PspA-C-CPE). PspA-C-CPE binds to claudin-4 and thus efficiently attaches to NALT epithelium, including antigen-sampling M cells. Nasal immunization with PspA-C-CPE induced PspA-specific IgG in the serum and bronchoalveolar lavage fluid (BALF) as well as IgA in the nasal wash and BALF. These immune responses were sufficient to protect against pneumococcal infection. These results suggest that C-CPE is an efficient vaccine delivery system for the development of nasal vaccines against pneumococcal infection.


Subject(s)
Bacterial Proteins/immunology , Enterotoxins/administration & dosage , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/administration & dosage , Administration, Intranasal , Animals , Bacterial Proteins/genetics , Bronchoalveolar Lavage Fluid/immunology , Claudin-4/metabolism , Drug Delivery Systems/methods , Enterotoxins/genetics , Female , Mice, Inbred BALB C , Pneumococcal Infections/immunology , Pneumococcal Vaccines/genetics , Pneumococcal Vaccines/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/pathogenicity
16.
Biotechnol Lett ; 37(6): 1177-85, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25700824

ABSTRACT

Claudins constitute a family of at least 27 proteins with four transmembrane domains, and play a pivotal role in maintaining tight-junctions seals in diverse epithelial tissues. The expression of claudin-4 often changes in intestinal tissues of inflammatory bowel disease and various human cancers. Therefore, claudin-4 is a promising target for treatment of these diseases. In our previous study, we established a reporter cell line to monitor claudin-4 expression on the basis of a functional claudin-4 promoter. Using this cell line, we have performed a cell-based screen of a library containing 2642 biologically active small-molecule compounds to identify modulators of claudin-4 expression. The screen identified 24 potential modulators of the claudin-4 promoter activity. Fourteen of these compounds (12 of them novel) induced endogenous claudin-4 expression. The identified compounds might serve as lead compounds targeting aberrant gene expression in inflammatory bowel disease.


Subject(s)
Claudin-4/biosynthesis , Cytological Techniques/methods , Drug Evaluation, Preclinical/methods , Gene Expression/drug effects , Transcriptional Activation , Cell Line , Humans , Promoter Regions, Genetic
17.
J Pharmacol Exp Ther ; 353(1): 112-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25628391

ABSTRACT

Claudin-1 (CLDN1), a known host factor for hepatitis C virus (HCV) entry and cell-to-cell transmission, is a target molecule for inhibiting HCV infection. We previously developed four clones of mouse anti-CLDN1 monoclonal antibody (mAb) that prevented HCV infection in vitro. Two of these mAbs showed the highest antiviral activity. Here, we optimized the anti-CLDN1 mAbs as candidates for therapeutics by protein engineering. Although Fab fragments of the mAbs prevented in vitro HCV infection, their inhibitory effects were much weaker than those of the whole mAbs. In contrast, human chimeric IgG1 mAbs generated by grafting the variable domains of the mouse mAb light and heavy chains inhibited in vitro HCV infection as efficiently as the parental mouse mAbs. However, the chimeric IgG1 mAbs activated Fcγ receptor, suggesting that cytotoxicity against mAb-bound CLDN1-expressing cells occurred through the induction of antibody-dependent cellular cytotoxicity (ADCC). To avoid ADCC-induced side effects, we prepared human chimeric IgG4 mAbs. The chimeric IgG4 mAbs did not activate Fcγ receptor or induce ADCC, but they prevented in vitro HCV infection as efficiently as did the parental mouse mAbs. These findings indicate that the IgG4 form of human chimeric anti-CLDN1 mAb may be a candidate molecule for clinically applicable HCV therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antiviral Agents/pharmacology , Claudin-1/immunology , Hepacivirus/drug effects , Animals , Antibodies, Monoclonal/genetics , Cell Line , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/genetics , Immunoglobulin G/pharmacology , Mice , Receptors, IgG/metabolism , Virus Internalization/drug effects
18.
Eur J Pharm Biopharm ; 89: 232-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25513955

ABSTRACT

Homoharringtonine (HHT), a natural alkaloid produced by various Cephalotaxus species, has antileukemic activity in acute and chronic myelogenous leukemia. However, HHT can also induce unanticipated effects in the gastrointestinal tract, such as diarrhea and nausea/vomiting, but the mechanism behind these adverse effects has not been clarified. In the present study, we show that HHT affects the epithelial permeability of intestinal Caco-2 cell monolayers. HHT reduced the transepithelial electrical resistance (TER) of Caco-2 cells in a dose- and time-dependent manner. The HHT effect was reversible and no cytotoxicity was observed at the concentrations used. HHT simultaneously increased the paracellular flux of the 4 kDa and 40 kDa FITC-dextrans associated with the TER reduction. Immunoblotting analysis revealed that HHT decreased the protein expression of TJ components such as claudin-3, -5, and -7. However, the transcription levels of these claudins were not repressed by HHT treatment. HHT also disturbed the cellular localization of claudin-1 and -4. These changes coincided with the reduced barrier function. Our findings suggest that HHT enhances the paracellular permeability of Caco-2 cell monolayers by modulating the protein expression and localization of claudin isoforms; these actions might be responsible for the gastrointestinal effects of HHT.


Subject(s)
Cell Membrane Permeability/physiology , Claudins/metabolism , Epithelial Cells/metabolism , Epithelial Cells/physiology , Harringtonines/metabolism , Intestinal Mucosa/metabolism , Protein Isoforms/metabolism , Caco-2 Cells , Cell Line, Tumor , Dextrans/metabolism , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/metabolism , Homoharringtonine , Humans , Intestines/physiology , Tight Junctions/metabolism , Tight Junctions/physiology , Transcription, Genetic/physiology
19.
J Pharmacol Exp Ther ; 351(1): 206-13, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25118216

ABSTRACT

Most malignant tumors are derived from epithelium, and claudin (CLDN)-3 and CLDN-4 are frequently overexpressed in such tumors. Although antibodies have potential in cancer diagnostics and therapy, development of antibodies against CLDNs has been difficult because the extracellular domains of CLDNs are too small and there is high homology among human, rat, and mouse sequences. Here, we created a monoclonal antibody that recognizes human CLDN-3 and CLDN-4 by immunizing rats with a plasmid vector encoding human CLDN-4. A hybridoma clone that produced a rat monoclonal antibody recognizing both CLDN-3 and -4 (clone 5A5) was obtained from a hybridoma screen by using CLDN-3- and -4-expressing cells; 5A5 did not bind to CLDN-1-, -2-, -5-, -6-, -7-, or -9-expressing cells. Fluorescence-conjugated 5A5 injected into xenograft mice bearing human cancer MKN74 or LoVo cells could visualize the tumor cells. The human-rat chimeric IgG1 monoclonal antibody (xi5A5) activated FcγRIIIa in the presence of CLDN-3- or -4-expressing cells, indicating that xi5A5 may exert antibody-dependent cellular cytotoxicity. Administration of xi5A5 attenuated tumor growth in xenograft mice bearing MKN74 or LoVo cells. These results suggest that 5A5 shows promise in the development of a diagnostic and therapeutic antibody for cancers.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity , Claudin-3/immunology , Claudin-4/immunology , Neoplasms, Experimental/therapy , Animals , Antibodies, Monoclonal/therapeutic use , Cell Line , Cell Line, Tumor , Female , Humans , Immunoglobulin G/immunology , Immunoglobulin G/therapeutic use , Immunotherapy , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/diagnosis , Rats , Rats, Wistar , Receptors, IgG/immunology , Xenograft Model Antitumor Assays
20.
Eur J Pharm Sci ; 52: 132-7, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24231339

ABSTRACT

We previously found that claudin (CL) is a potent target for cancer therapy using a CL-3 and -4-targeting molecule, namely the C-terminal fragment of Clostridium perfringens enterotoxin (C-CPE). Although CL-3 and -4 are expressed in various normal tissues, the safety of this CL-targeting strategy has never been investigated. Here, we evaluated the tissue distribution of C-CPE in mice. Ten minutes after intravenous injection into mice, C-CPE was distributed to the liver and kidney (24.0% and 9.5% of the injected dose, respectively). The hepatic level gradually fell to 3.2% of the injected dose by 3 h post-injection, whereas the renal C-CPE level gradually rose to 46.5% of the injected dose by 6 h post-injection and then decreased. A C-CPE mutant protein lacking the ability to bind CL accumulated in the liver to a much lesser extent (2.0% of the dose at 10 min post-injection) than did C-CPE, but its renal profile was similar to that of C-CPE. To investigate the acute toxicity of CL-targeted toxin, we intravenously administered C-CPE-fused protein synthesis inhibitory factor to mice. The CL-targeted toxin dose-dependently increased the levels of serum biomarkers of liver injury, but not of kidney injury. Histological examination confirmed that injection of CL-targeted toxin injured the liver but not the kidney. These results indicate that potential adverse hepatic effects should be considered in C-CPE-based cancer therapy.


Subject(s)
Claudin-3/metabolism , Claudin-4/metabolism , Enterotoxins/chemistry , Enterotoxins/pharmacology , Peptide Fragments/pharmacology , Protein Synthesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cells, Cultured , Clostridium perfringens , Enterotoxins/genetics , Female , Fibroblasts , Intestinal Mucosa/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred BALB C , Mutation , Peptide Fragments/chemistry , Protein Synthesis Inhibitors/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Thyroid Gland/metabolism , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...