Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Int J Mol Sci ; 21(16)2020 Aug 17.
Article in English | MEDLINE | ID: mdl-32824620

ABSTRACT

Claudin-1 (CLDN1), a tight junctional protein, is highly expressed in lung cancer cells and may contribute to chemoresistance. A drug which decreases CLDN1 expression could be a chemosensitizer for enhancing the efficacy of anticancer drugs, but there is no such drug known. We found that PMTPV, a short peptide, which mimics the structure of second extracellular loop (ECL2) of CLDN1, can reduce the protein level of CLDN1 without affecting the mRNA level in A549 cells derived from human lung adenocarcinoma. The PMTPV-induced decrease in CLDN1 expression was inhibited by monodansylcadaverine, a clathrin-mediated endocytosis inhibitor, and chloroquine, a lysosome inhibitor. Quartz crystal microbalance assay showed that PMTPV can directly bind to the ECL2 of CLDN1. In transwell assay, PMTPV increased fluxes of Lucifer yellow (LY), a paracellular flux marker, and doxorubicin (DXR), an anthracycline anticancer drug, without affecting transepithelial electrical resistance. In three-dimensional spheroid culture, the size and cell viability were unchanged by short peptides, but the fluorescence intensity of hypoxia probe LOX-1 was decreased by PMTPV. PMTPV elevated the accumulation and cytotoxicity of DXR in the spheroids. Similar results were observed by knockdown of CLDN1. Furthermore, the sensitivities to cisplatin (CDDP), docetaxel, and gefitinib were enhanced by PMTPV. The level of CLDN1 expression in CDDP-resistant cells was higher than that in parental A549 cells, which was reduced by PMTPV. PMTPV restored the toxicity to DXR in the CDDP-resistant cells. Our data suggest that PMTPV may become a novel chemosensitizer for lung adenocarcinoma.


Subject(s)
Antineoplastic Agents/toxicity , Claudin-1/metabolism , Oligopeptides/pharmacology , A549 Cells , Binding Sites , Cell Survival/drug effects , Claudin-1/antagonists & inhibitors , Claudin-1/chemistry , Humans , Ligands , Protein Binding
2.
Yakugaku Zasshi ; 139(1): 89-95, 2019.
Article in Japanese | MEDLINE | ID: mdl-30606936

ABSTRACT

Hepatitis C virus (HCV) infection is a major leading cause of chronic severe liver diseases such as cirrhosis and hepatocellular carcinoma. The recent direct-acting antivirals (DAAs) for the treatment of HCV infection offer very high cure rates, but DAAs are vulnerable to drug resistance because HCV is an RNA virus, which generally has very high mutation rates. DAA resistance-associated variants of HCV could reduce the effectiveness of DAAs in the future. Thus, the continuous development of new anti-HCV drugs against different target molecules is needed. We have been studying the host factors involved in HCV entry into cells. From those studies, we obtained novel candidates for host-targeting anti-HCV entry inhibitors, such as monoclonal antibodies against HCV receptors, which can be used together with DAAs. In this symposium review, we present and discuss our recent work on anti-HCV strategies targeting HCV entry steps.


Subject(s)
Antibodies, Monoclonal , Antiviral Agents , Drug Discovery , Hepacivirus/drug effects , Hepacivirus/pathogenicity , Hepatitis C/drug therapy , Hepatitis C/virology , Molecular Targeted Therapy , Receptors, Virus/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Claudin-1/antagonists & inhibitors , Drug Resistance, Viral , Humans , Mice , Occludin/antagonists & inhibitors
3.
Cell Commun Signal ; 16(1): 14, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29636110

ABSTRACT

BACKGROUND: In addition to physiological events such as fertilisation, placentation, osteoclastogenesis, or tissue regeneration/wound healing, cell fusion is involved in pathophysiological conditions such as cancer. Cell fusion, which applies to both the proteins and conditions that induce the merging of two or more cells, is not a fully understood process. Inflammation/pro-inflammatory cytokines might be a positive trigger for cell fusion. Using a Cre-LoxP-based cell fusion assay we demonstrated that the fusion between human M13SV1-Cre breast epithelial cells and human MDA-MB-435-pFDR1 cancer cells was induced by the pro-inflammatory cytokine tumour necrosis factor-α (TNF-α). METHODS: The gene expression profile of the cells in the presence of TNF-α and under normoxic and hypoxic conditions was analysed by cDNA microarray analysis. cDNA microarray data were verified by qPCR, PCR, Western blot and zymography. Quantification of cell fusion events was determined by flow cytometry. Proteins of interest were either blocked or knocked-down using a specific inhibitor, siRNA or a blocking antibody. RESULTS: The data showed an up-regulation of various genes, including claudin-1 (CLDN1), ICAM1, CCL2 and MMP9 in M13SV1-Cre and/or MDA-MB-435-pFDR1 cells. Inhibition of these proteins using a blocking ICAM1 antibody, CLDN1 siRNA or an MMP9 inhibitor showed that only the blockage of MMP9 was correlated with a decreased fusion rate of the cells. Likewise, the tetracycline-based antibiotic minocycline, which exhibits anti-inflammatory properties, was also effective in both inhibiting the TNF-α-induced MMP9 expression in M13SV1-Cre cells and blocking the TNF-α-induced fusion frequency of human M13SV1-Cre breast epithelial cells and human MDA-MB-435-pFDR1 cancer cells. CONCLUSIONS: The matrix metalloproteinase-9 (MMP9) is most likely involved in the TNF-α-mediated fusion of human M13SV1-Cre breast epithelial cells and human MDA-MB-435-pFDR1 cancer cells. Likewise, our data indicate that the tetracycline-based antibiotic minocycline might exhibit anti-fusogenic properties because it inhibits a cell fusion-related mechanism.


Subject(s)
Cell Fusion , Epithelial Cells/drug effects , Matrix Metalloproteinase 9/metabolism , Transcriptome/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Breast/cytology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Hypoxia , Cell Line, Tumor , Claudin-1/antagonists & inhibitors , Claudin-1/genetics , Claudin-1/metabolism , Down-Regulation/drug effects , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Integrases/genetics , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Matrix Metalloproteinase 9/chemistry , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase Inhibitors/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Up-Regulation/drug effects
4.
Gut ; 67(4): 736-745, 2018 04.
Article in English | MEDLINE | ID: mdl-28360099

ABSTRACT

OBJECTIVE: HCV infection is a leading cause of chronic liver disease and a major indication for liver transplantation. Although direct-acting antivirals (DAAs) have much improved the treatment of chronic HCV infection, alternative strategies are needed for patients with treatment failure. As an essential HCV entry factor, the tight junction protein claudin-1 (CLDN1) is a promising antiviral target. However, genotype-dependent escape via CLDN6 and CLDN9 has been described in some cell lines as a possible limitation facing CLDN1-targeted therapies. Here, we evaluated the clinical potential of therapeutic strategies targeting CLDN1. DESIGN: We generated a humanised anti-CLDN1 monoclonal antibody (mAb) (H3L3) suitable for clinical development and characterised its anti-HCV activity using cell culture models, a large panel of primary human hepatocytes (PHH) from 12 different donors, and human liver chimeric mice. RESULTS: H3L3 pan-genotypically inhibited HCV pseudoparticle entry into PHH, irrespective of donor. Escape was likely precluded by low surface expression of CLDN6 and CLDN9 on PHH. Co-treatment of a panel of PHH with a CLDN6-specific mAb did not enhance the antiviral effect of H3L3, confirming that CLDN6 does not function as an entry factor in PHH from multiple donors. H3L3 also inhibited DAA-resistant strains of HCV and synergised with current DAAs. Finally, H3L3 cured persistent HCV infection in human-liver chimeric uPA-SCID mice in monotherapy. CONCLUSIONS: Overall, these findings underscore the clinical potential of CLDN1-targeted therapies and describe the functional characterisation of a humanised anti-CLDN1 antibody suitable for further clinical development to complement existing therapeutic strategies for HCV.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antiviral Agents/pharmacology , Claudin-1/antagonists & inhibitors , Hepacivirus/drug effects , Hepatitis C/prevention & control , Hepatocytes/drug effects , Immunologic Factors/pharmacology , Animals , Claudin-1/immunology , Hepatitis C/immunology , Hepatocytes/immunology , Hepatocytes/virology , Humans , Mice , Mice, SCID , Treatment Outcome
5.
Cell Physiol Biochem ; 42(4): 1493-1513, 2017.
Article in English | MEDLINE | ID: mdl-28719888

ABSTRACT

BACKGROUND/AIMS: Hallmark of diabetic macular edema is the enhanced permeability of retinal endothelial cells (REC) induced by vascular endothelial growth factor (VEGF-A165), which acts through activating specific receptors. To improve the predictability of inhibitors' potentials to block harmful effects of VEGF-A165, we investigated if its signaling pathways triggered in REC are redundant. METHODS: Immortalized bovine REC monolayers were treated with inhibitors specific for various protein kinases in combination with VEGF-A165. Permeability was monitored continuously by measurements of the cell index (CI) to reveal even subtle and transient changes. Expression of tight junction (TJ) proteins was determined as additional indicator of barrier stability. RESULTS: After a sharp but transient CI drop caused by VEGF-A165 early after its addition, further exposure resulted in a continuous CI decline over several days associated with loss of TJ protein claudin-1. Both phases were blocked by inhibition of VEGF receptor 2. Tested inhibitors of intracellular kinases had a limited or no effect, or were efficient only in certain phases of exposure to VEGF-A165, e.g. inhibiting protein kinase C only prevented the early response. High concentrations of some inhibitors even resulted in VEGF-independent barrier destabilization. CONCLUSIONS: Specific kinase inhibitors differently affect VEGF-A165-triggered processes in distinct phases of its action. VEGF-A165-initiated signaling is redundant and blocking of key proteins of single pathways is not sufficient to suppress REC barrier breakdown.


Subject(s)
Antibodies/pharmacology , Endothelial Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Retina/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Animals , Cattle , Cell Line, Transformed , Cell Membrane Permeability/drug effects , Claudin-1/antagonists & inhibitors , Claudin-1/genetics , Claudin-1/metabolism , Claudin-5/antagonists & inhibitors , Claudin-5/genetics , Claudin-5/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation , Humans , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase C/metabolism , Quinazolines/pharmacology , Recombinant Proteins/pharmacology , Retina/cytology , Retina/metabolism , Signal Transduction , Tight Junctions/drug effects , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
6.
J Exp Clin Cancer Res ; 36(1): 89, 2017 06 28.
Article in English | MEDLINE | ID: mdl-28659146

ABSTRACT

BACKGROUND: Metastatic colorectal cancer (mCRC) is one of the major causes of cancer-related death. Despite the substantial progress in mCRC management, it remains important to identify new therapeutic options and biological markers for personalized medicine. Here, we investigated the expression of claudin-1 (CLDN1), a major tight junction transmembrane protein, in the different colorectal cancer (CRC) molecular subtypes and then assessed the anti-tumor effect of a new anti-CLDN1 monoclonal antibody (mAb). METHODS: Gene expression profiling and immunochemistry analysis of normal and tumor tissue samples from patients with stage IV CRC were used to determine CLDN1 gene expression. Then, the 6F6 mAb against CLDN1 extracellular part was generated. Its effect on CRC cell cycle, proliferation, survival and migration was assessed in vitro, using a 3D cell culture system, flow cytometry, clonogenic and migration assays. In vivo, 6 F6 mAb efficacy was evaluated in nude mice after subcutaneous xenografts or intrasplenic injection of CRC cells. RESULTS: Compared with normal mucosa where it was almost exclusively cytoplasmic, in CRC samples CLDN1 was overexpressed (p < 0.001) and mainly localized at the membrane. Moreover, it was differentially expressed in the various CRC molecular subtypes. The strongest expressions were found in the consensus molecular subtype CMS2 (p < 0.001), the transit-ampliflying (p < 0.001) and the C5 subtypes (p < 0.001). Lower CLDN1 expression predicted a better outcome in the molecular subtypes C3 and C5 (p = 0.012 and p = 0.004, respectively). CLDN1 targeting with the 6 F6 mAb led to reduction of survival, growth and migration of CLDN1-positive cells. In preclinical mouse models, the 6F6 mAb decreased tumor growth and liver metastasis formation. CONCLUSION: Our data indicate that CLDN1 targeting with an anti-CLDN1 mAb results in decreased growth and survival of CRC cells. This suggests that CLDN1 could be a new potential therapeutic target.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Claudin-1/antagonists & inhibitors , Colorectal Neoplasms/metabolism , Animals , Biomarkers, Tumor , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Molecular Targeted Therapy , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Retrospective Studies , Single Photon Emission Computed Tomography Computed Tomography/methods , Xenograft Model Antitumor Assays
7.
Biochem Biophys Res Commun ; 486(3): 732-737, 2017 05 06.
Article in English | MEDLINE | ID: mdl-28342862

ABSTRACT

CLDN1 (claudin1) is essential for intercellular junctions and has been reported to be involving in cell migration and metastasis, making it as an oncogene in various cancer types. However, the biological function roles and regulatory mechanisms of CLDN1 in hepatocellular carcinoma (HCC) are still not clarified. In this study, we found down-regulation of miR-29a and up-regulation of CLDN1 in HCC tissues and cell lines. Further found an inverse relation between the expressions of miR-29a and CLDN1 in HCC. Dual-luciferase reporter assay indicated that miR-29a regulated the expression of CLDN1 by binding to its 3' untranslated region (3'UTR). Knockdown of CLDN1 led to decrease in tumor cell growth and migration capacities in vitro and in vivo. While overexpression of miR-29a suppressed tumor growth and migration, these effects could be reversed by re-expressing CLDN1. Taken together, out data suggested that miR-29a may regulate tumor growth and migration by targeting CLDN1, providing promising therapeutic targets for HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Claudin-1/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , MicroRNAs/genetics , Animals , Base Sequence , Binding Sites , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Claudin-1/antagonists & inhibitors , Claudin-1/metabolism , Female , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Neoplasm Invasiveness , Neoplasm Transplantation , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Survival Analysis , Transfection , Tumor Burden
8.
Nutrients ; 8(11)2016 Oct 28.
Article in English | MEDLINE | ID: mdl-27801835

ABSTRACT

The composition of the gut microbiome reflects the overall health status of the host. In this study, stool samples representing the gut microbiomes from 6 gluten-sensitive (GS) captive juvenile rhesus macaques were compared with those from 6 healthy, age- and diet-matched peers. A total of 48 samples representing both groups were studied using V4 16S rRNA gene DNA analysis. Samples from GS macaques were further characterized based on type of diet administered: conventional monkey chow, i.e., wheat gluten-containing diet (GD), gluten-free diet (GFD), barley gluten-derived diet (BOMI) and reduced gluten barley-derived diet (RGB). It was hypothesized that the GD diet would lower the gut microbial diversity in GS macaques. This is the first report illustrating the reduction of gut microbial alpha-diversity (p < 0.05) following the consumption of dietary gluten in GS macaques. Selected bacterial families (e.g., Streptococcaceae and Lactobacillaceae) were enriched in GS macaques while Coriobacteriaceae was enriched in healthy animals. Within several weeks after the replacement of the GD by the GFD diet, the composition (beta-diversity) of gut microbiome in GS macaques started to change (p = 0.011) towards that of a normal macaque. Significance for alpha-diversity however, was not reached by the day 70 when the feeding experiment ended. Several inflammation-associated microRNAs (miR-203, -204, -23a, -23b and -29b) were upregulated (p < 0.05) in jejunum of 4 biopsied GS macaques fed GD with predicted binding sites on 16S ribosomal RNA of Lactobacillus reuteri (accession number: NR_025911), Prevotella stercorea (NR_041364) and Streptococcus luteciae (AJ297218) that were overrepresented in feces. Additionally, claudin-1, a validated tight junction protein target of miR-29b was significantly downregulated in jejunal epithelium of GS macaques. Taken together, we predict that with the introduction of effective treatments in future studies the diversity of gut microbiomes in GS macaques will approach those of healthy individuals. Further studies are needed to elucidate the regulatory pathways of inflammatory miRNAs in intestinal mucosa of GS macaques and to correlate their expression with gut dysbiosis.


Subject(s)
Celiac Disease/metabolism , Disease Models, Animal , Dysbiosis/metabolism , Glutens/adverse effects , Intestinal Mucosa/metabolism , MicroRNAs/metabolism , Plant Proteins, Dietary/adverse effects , Animals , Biomarkers/metabolism , Celiac Disease/immunology , Celiac Disease/microbiology , Celiac Disease/pathology , Claudin-1/antagonists & inhibitors , Claudin-1/genetics , Claudin-1/metabolism , Dysbiosis/immunology , Dysbiosis/microbiology , Dysbiosis/pathology , Feces/chemistry , Feces/microbiology , Female , Gastrointestinal Microbiome/immunology , Gene Expression Regulation , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Jejunum/immunology , Jejunum/metabolism , Jejunum/microbiology , Jejunum/pathology , Macaca mulatta , Male , MicroRNAs/chemistry , Nucleotide Motifs , RNA, Bacterial/metabolism , RNA, Ribosomal, 16S/metabolism , Specific Pathogen-Free Organisms , Tight Junctions/immunology , Tight Junctions/metabolism , Tight Junctions/pathology
10.
J Gen Virol ; 97(1): 82-94, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26519290

ABSTRACT

Hepatitis C virus (HCV) is a major cause of chronic hepatitis and liver carcinoma and new therapies based on novel targets are needed. The tight junction protein claudin 1 (CLDN-1) is essential for HCV cell entry and spread, and anti-CLDN-1 rat and mouse mAbs are safe and effective in preventing and treating HCV infection in a human liver chimeric mouse model. To accelerate translation of these observations into a novel approach to treat HCV infection and disease in humans, we screened a phage display library of human single-chain antibody fragments by using a panel of CLDN-1-positive and -negative cell lines and identified phage specifically binding to CLDN-1. The 12 clones showing the highest levels of binding were converted into human IgG4. Some of these mAbs displayed low-nanomolar affinity, and inhibited infection of human hepatoma Huh7.5 cells by different HCV isolates in a dose-dependent manner. Cross-competition experiments identified six inhibitory mAbs that recognized distinct epitopes. Combination of the human anti-SRB1 mAb C-1671 with these anti-CLDN-1 mAbs could either increase or reduce inhibition of cell culture-derived HCV infection in vitro. These novel human anti-CLDN-1 mAbs are potentially useful to develop a new strategy for anti-HCV therapy and lend support to the combined use of antibodies targeting the HCV receptors CLDN-1 and SRB1, but indicate that care must be taken in selecting the proper combination.


Subject(s)
Antibodies, Monoclonal/metabolism , Antiviral Agents/metabolism , Claudin-1/antagonists & inhibitors , Hepacivirus/physiology , Scavenger Receptors, Class B/antagonists & inhibitors , Single-Chain Antibodies/metabolism , Virus Internalization/drug effects , Antibodies, Monoclonal/isolation & purification , Antiviral Agents/isolation & purification , Cell Line , Claudin-1/immunology , Hepatocytes/virology , Humans , Models, Theoretical , Peptide Library , Scavenger Receptors, Class B/immunology , Single-Chain Antibodies/isolation & purification , Viral Load , Virus Cultivation
12.
Viruses ; 7(6): 2965-79, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-26067567

ABSTRACT

Breast cancer has a high incidence and mortality rate worldwide. Several viral vectors including lentiviral, adenoviral and adeno-associated viral vectors have been used in gene therapy for various forms of human cancer, and have shown promising effects in controlling tumor development. Claudin1 (CLDN1) is a member of the tetraspan transmembrane protein family that plays a major role in tight junctions and is associated with tumor metastasis. However, the role of CLDN1 in breast cancer is largely unexplored. In this study, we tested the therapeutic potential of silencing CLDN1 expression in two breast cancer (MDA-MB-231 and MCF7) cell lines using lentiviral vector mediated RNA interference. We found that a CLDN1 short hairpin (shRNA) construct efficiently silenced CLDN1 expression in both breast cancer cell lines, and CLDN1 knockdown resulted in reduced cell proliferation, survival, migration and invasion. Furthermore, silencing CLDN1 inhibited epithelial to mesenchymal transition (EMT) by upregulating the epithelial cell marker, E-cadherin, and downregulating mesenchymal markers, smooth muscle cell alpha-actin (SMA) and Snai2. Our data demonstrated that lentiviral vector mediated CLDN1 RNA interference has great potential in breast cancer gene therapy by inhibiting EMT and controlling tumor cell growth.


Subject(s)
Antineoplastic Agents/metabolism , Claudin-1/antagonists & inhibitors , Epithelial Cells/physiology , Epithelial-Mesenchymal Transition/drug effects , Gene Silencing , RNA, Small Interfering/metabolism , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Genetic Vectors , Humans , Lentivirus/genetics , Models, Biological , RNA Interference , RNA, Small Interfering/genetics , Transformation, Genetic
13.
Asian Pac J Cancer Prev ; 16(6): 2473-81, 2015.
Article in English | MEDLINE | ID: mdl-25824783

ABSTRACT

Colon cancer is one of the leading causes of cancer-associated death worldwide. The prognosis for advanced colorectal cancers remains dismal, mainly due to the propensity for metastatic progression. Accordingly, there is a need for effective anti-metastasis therapeutic agents. Since a great body of research has indicated anticancer effects for curcumin, we investigated the effects of dendrosomal curcumin (DNC) on cellular migration and adhesion of human SW480 cells and possible molecular mechanisms involved. Different methods were applied in this study including MTT, Scratch and adhesion assays as well as real-time PCR and transwell chamber assays. Based on the results obtained, DNC inhibits metastasis by decreasing Hef 1, Zeb 1 and Claudin 1 mRNA levels and can reduce SW480 cell proliferation with IC50values of 15.9, 11.6 and 7.64 µM at 24, 48 and 72 h post-treatment. Thus it might be considered as a safe formulation for therapeutic purpose in colorectal cancer cases.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Cell Movement/drug effects , Claudin-1/antagonists & inhibitors , Colonic Neoplasms/drug therapy , Curcumin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Homeodomain Proteins/antagonists & inhibitors , Phosphoproteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Claudin-1/genetics , Claudin-1/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/secondary , Curcumin/chemistry , Drug Carriers , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunoenzyme Techniques , Nanocapsules/administration & dosage , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured , Zinc Finger E-box-Binding Homeobox 1
14.
Biochem Biophys Res Commun ; 459(3): 379-86, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25727011

ABSTRACT

Interleukin-18 (IL-18) was recently reported to have a pro-tumor effect in various cancers. Increased IL-18 levels in the serum of cancer patients correlated with malignancy, and IL-18 acts a crucial factor for cell migration in gastric cancer and melanoma. Claudins, which are the most important tight junction proteins, are also linked with cancer progression and metastasis. However, the relationship between claudins and IL-18 is not well-understood. Here, we show that the migratory ability of MCF-7 cells was reduced when endogenous IL-18 expression was inhibited with IL-18 siRNA. Moreover, exogenous IL-18 enhanced breast cancer cell migration and suppressed the expression of the tight junction proteins claudin-1, claudin-3, claudin-4, and claudin-12 in MCF-7 cells. Knockdown of claudin-3, claudin-4, and claudin-12, but not claudin-1, increased breast cancer migration with maximal effects observed in claudin-12 siRNA-transfected cells. To investigate whether the mitogen-activated protein kinase (MAPK) signaling pathway is involved in IL-18-induced cell migration and claudin-12 expression, cells were pretreated with SB203580 (an inhibitor of p38 MAPK) or PD98059 (an inhibitor of ERK1/2) prior to the addition of IL-18. Although pretreatment of MCF-7 cells with SB203580 blocked both the enhanced cell migration and the decreased claudin-12 expression, PD98059 only blocked cell migration and did not affect claudin-12 expression. In addition, exogenous IL-18 induced rapid phosphorylation of p38 MAPK. These results suggest that IL-18 is an important factor inducing breast cancer cell migration through down-regulation of claudin-12 and activation of the p38 MAPK pathway.


Subject(s)
Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Claudins/physiology , Interleukin-18/physiology , MAP Kinase Signaling System , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Movement/physiology , Claudin-1/antagonists & inhibitors , Claudin-1/genetics , Claudin-1/physiology , Claudin-3/antagonists & inhibitors , Claudin-3/genetics , Claudin-3/physiology , Claudin-4/antagonists & inhibitors , Claudin-4/genetics , Claudin-4/physiology , Claudins/antagonists & inhibitors , Claudins/genetics , Down-Regulation/drug effects , Female , Flavonoids/pharmacology , Gene Knockdown Techniques , Humans , Imidazoles/pharmacology , Interleukin-18/antagonists & inhibitors , Interleukin-18/genetics , MAP Kinase Signaling System/drug effects , MCF-7 Cells , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , RNA, Small Interfering/genetics , Recombinant Proteins/pharmacology , Tight Junctions/drug effects , Tight Junctions/physiology
15.
J Virol ; 89(9): 4866-79, 2015 May.
Article in English | MEDLINE | ID: mdl-25673725

ABSTRACT

UNLABELLED: Hepatitis C virus (HCV) entry into host cells is a complex process requiring multiple host factors, including claudin-1 (CLDN1). Safe and effective therapeutic entry inhibitors need to be developed. We isolated a human hepatic Huh7.5.1-derived cell mutant that is nonpermissive to HCV, and comparative microarray analysis showed that the mutant was CLDN1 defective. Four hybridomas were obtained, which produced monoclonal antibodies (MAbs) that interacted with the parental Huh7.5.1 cell but not with the CLDN1-defective mutant. All MAbs produced by these hybridomas specifically bound to human CLDN1 with a very high affinity and prevented HCV infection of Huh7.5.1 cells in a dose-dependent manner, without apparent cytotoxicity. Two selected MAbs also inhibited HCV infection of human liver-chimeric mice without significant adverse effects. CLDN1 may be a potential target to prevent HCV infection in vivo. Anti-CLDN1 MAbs may hence be promising candidates as novel anti-HCV agents. IMPORTANCE: Safe and effective therapeutic entry inhibitors against hepatitis C virus (HCV) are very useful for combination therapies with other anti-HCV drugs, such as direct-acting antivirals. In this study, we first showed an effective strategy for developing functional monoclonal antibodies (MAbs) against extracellular domains of a multimembrane-spanning target protein, claudin-1 (CLDN1), by using parental cells expressing the intact target membrane protein and target-defective cells. The established MAbs against CLDN1, which had a very high affinity for intact CLDN1, efficiently inhibited in vitro and in vivo HCV infections. These anti-CLDN1 MAbs are promising leads for novel entry inhibitors against HCV.


Subject(s)
Antibodies, Monoclonal/immunology , Claudin-1/antagonists & inhibitors , Hepacivirus/physiology , Hepatitis C/prevention & control , Receptors, Virus/antagonists & inhibitors , Virus Internalization/drug effects , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/therapeutic use , Cell Line , Claudin-1/immunology , Disease Models, Animal , Drug-Related Side Effects and Adverse Reactions , Hepacivirus/drug effects , Hepatocytes/virology , Humans , Male , Mice , Receptors, Virus/immunology , Treatment Outcome
16.
PLoS One ; 9(9): e107970, 2014.
Article in English | MEDLINE | ID: mdl-25268969

ABSTRACT

As is widely recognized, human parvovirus B19 (B19) and human bocavirus (HBoV) are important human pathogens. Obviously, both VP1 unique region (VP1u) of B19 and HBoV exhibit the secreted phospholipase A2 (sPLA2)-like enzymatic activity and are recognized to participate in the pathogenesis of lower respiratory tract illnesses. However, exactly how, both VP1u from B19 and HBoV affect tight junction has seldom been addressed. Therefore, this study investigates how B19-VP1u and HBoV-VP1u may affect the tight junction of the airway epithelial A549 cells by examining phospholipase A2 activity and transepithelial electrical resistance (TEER) as well as performing immunoblotting analyses. Experimental results indicate that TEER is more significantly decreased in A549 cells by treatment with TNF-α (10 ng), two dosages of B19-VP1u and BoV-VP1u (400 ng and 4000 ng) or bee venom PLA2 (10 ng) than that of the control. Accordingly, more significantly increased claudin-1 and decreased occludin are detected in A549 cells by treatment with TNF-α or both dosages of HBoV-VP1u than that of the control. Additionally, more significantly decreased Na+/K+ ATPase is observed in A549 cells by treatment with TNF-α, high dosage of B19-VP1u or both dosages of BoV-VP1u than that of the control. Above findings suggest that HBoV-VP1u rather than B19 VP1u likely plays more important roles in the disruption of tight junction in the airway tract. Meanwhile, this discrepancy appears not to be associated with the secreted phospholipase A2 (sPLA2)-like enzymatic activity.


Subject(s)
Epithelial Cells/drug effects , Human bocavirus/chemistry , Parvovirus B19, Human/chemistry , Tight Junctions/drug effects , Viral Core Proteins/pharmacology , Bee Venoms/chemistry , Bee Venoms/enzymology , Cell Line , Claudin-1/antagonists & inhibitors , Claudin-1/genetics , Claudin-1/metabolism , Electric Impedance , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression , Humans , Phospholipases A2/pharmacology , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Respiratory Mucosa/cytology , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Viral Core Proteins/biosynthesis , Viral Core Proteins/isolation & purification
17.
Neuro Oncol ; 16(2): 256-64, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24305709

ABSTRACT

BACKGROUND: Claudins are tight junction proteins expressed in epithelial tissues that play important roles in cell polarity and adhesion. Altered distribution of claudin-1(CLDN1) affects cell mobility and tumor invasiveness. Craniopharyngiomas (CPs) represent epithelial tumors of the sellar region, consisting of adamantinomatous (adaCP) and papillary (papCP) variants. Their tendency to infiltrate surrounding brain structures complicates successful surgery. Reliable markers are required to predict tumor behavior and to establish individualized treatment protocols. METHODS: We describe the distribution pattern of CLDN1 in a large cohort of 66 adaCPs, 21 papCPs, and 24 Rathke`s cleft cyst (RCC) cases using immunohistochemistry. CLDN1 mRNA levels were analyzed with qRT-PCR in 33 CP samples. The impact on the migration potential was studied in primary adaCP cell cultures (n = 11) treated with small interfering RNA (siRNA) for CLDN1. Furthermore, CLDN1 distribution patterns and expression levels were compared between invasive (n = 16) and noninvasive (n = 17) tumor groups. RESULTS: PapCPs and RCCs exhibited a distinct homogenous and membranous expression pattern, whereas CLDN1 immunoreactivity appeared weaker and more heterogeneous in adaCPs. In the latter cases, whirl-like cell clusters showed complete absence of CLDN1. mRNA analysis confirmed reduced CLDN1 levels in adaCPs versus papCPs. Interestingly, invasive tumors exhibited significantly lower CLDN1 expression compared with noninvasive counterparts regardless of CP subtype. Accordingly, siRNA treatment for CLDN1 altered tumor cell migration in vitro. CONCLUSION: CLDN1 represents a novel marker in the differential diagnosis of CP variants and RCCs. Low CLDN1 expression levels correlate with an invasive CP growth pattern and may serve as a prognostic marker.


Subject(s)
Carcinoma, Papillary/pathology , Claudin-1/metabolism , Craniopharyngioma/pathology , Pituitary Neoplasms/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Apoptosis , Blotting, Western , Carcinoma, Papillary/genetics , Carcinoma, Papillary/metabolism , Cell Movement , Cell Proliferation , Child , Child, Preschool , Claudin-1/antagonists & inhibitors , Claudin-1/genetics , Cohort Studies , Craniopharyngioma/genetics , Craniopharyngioma/metabolism , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Pituitary Neoplasms/genetics , Pituitary Neoplasms/metabolism , Prognosis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Young Adult
18.
Cancer Prev Res (Phila) ; 6(7): 666-74, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23682078

ABSTRACT

The length of time required for preinvasive adenoma to progress to carcinoma, the immunogenicity of colorectal cancer (CRC), and the identification of high-risk populations make development and testing of a prophylactic vaccine for the prevention of CRC possible. We hypothesized that genes upregulated in adenoma relative to normal tissue, which maintained increased expression in CRC, would encode proteins suitable as putative targets for immunoprevention. We evaluated existing adenoma and CRC microarray datasets and identified 160 genes that were ≥2-fold upregulated in both adenoma and CRC relative to normal colon tissue. We further identified 23 genes that showed protein overexpression in colon adenoma and CRC based on literature review. Silencing the most highly upregulated genes, CDH3, CLDN1, KRT23, and MMP7, in adenoma and CRC cell lines resulted in a significant decrease in viability (P < 0.0001) and proliferation (P < 0.0001) as compared to controls and an increase in cellular apoptosis (P < 0.05 for CDH3, KRT23). Results were duplicated across cell lines representing microsatellite instability, CpG island methylator, and chromosomal instability phenotypes, suggesting immunologic elimination of cells expressing these proteins could impact the progression of all CRC phenotypes. To determine whether these proteins were immunogens, we interrogated sera from early stage CRC patients and controls and found significantly elevated CDH3 (P = 0.006), KRT23 (P = 0.0007), and MMP7 (P < 0.0001) serum immunoglobulin G in cases as compared to controls. These data show a high throughput approach to the identification of biologically relevant putative immunologic targets for CRC and identified three candidates suitable for vaccine development.


Subject(s)
Adenoma/diagnosis , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/diagnosis , Gene Expression Regulation, Neoplastic , Precancerous Conditions/diagnosis , Adenoma/metabolism , Adenoma/prevention & control , Adult , Aged , Aged, 80 and over , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Blotting, Western , Cadherins/antagonists & inhibitors , Cadherins/genetics , Cadherins/metabolism , Case-Control Studies , Cell Proliferation , Claudin-1/antagonists & inhibitors , Claudin-1/genetics , Claudin-1/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/prevention & control , DNA Methylation , Enzyme-Linked Immunosorbent Assay , Female , Humans , Keratins, Type I/antagonists & inhibitors , Keratins, Type I/genetics , Keratins, Type I/metabolism , Male , Matrix Metalloproteinase 7/chemistry , Matrix Metalloproteinase 7/genetics , Matrix Metalloproteinase 7/metabolism , Microsatellite Instability , Middle Aged , Neoplasm Staging , Precancerous Conditions/metabolism , Precancerous Conditions/prevention & control , Prognosis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Young Adult
19.
Biochem Pharmacol ; 85(12): 1770-82, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23603294

ABSTRACT

Infliximab, a monoclonal antibody directed against human tumor necrosis factor-alpha (TNF-α), effectively treats anterior uveitis, which can accompany Behçet's disease. Here, we investigated the underlying mechanism of this action. We examined human, non-pigmented ciliary epithelial cells (HNPCECs), which make up the blood-aqueous barrier (BAB) in the uvea. We measured the expression levels of matrix metalloproteinases (MMPs) and tissue inhibitors of MMPs in the presence or absence of TNF-α using quantitative, real-time polymerase chain reaction and enzyme-linked immunosorbent assays. The expression of MMP-1, MMP-3, and MMP-9 increased in the presence of TNF-α, and the addition of infliximab reversed the increase. The TNF-α effects were more attenuated when infliximab was added before than when it was added after TNF-α exposure. Gelatin zymography demonstrated that the protease activity of these MMPs was also increased in the presence of TNF-α and attenuated with infliximab. Immunostaining showed that MMP-1, MMP-3, and MMP-9 degraded claudin-1 and occludin in HNPCECs and in non-pigmented ciliary epithelial cells of the swine ciliary body. In a monolayer of HNPCECs, we found that permeability was significantly increased with MMP treatment. Thus, TNF-α increased levels of MMPs in cells that form the BAB, and MMPs degraded components of the tight junctions in the BAB, which increased permeability through the cellular barrier. Furthermore, infliximab effectively attenuated the TNF-α-induced increases in MMP expression in cells that make up the BAB. These findings might suggest a basis for the clinical prevention of anterior uveitis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Ciliary Body/metabolism , Claudin-1/antagonists & inhibitors , Matrix Metalloproteinases/metabolism , Occludin/antagonists & inhibitors , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/toxicity , Animals , Cells, Cultured , Ciliary Body/drug effects , Claudin-1/metabolism , Down-Regulation/drug effects , Down-Regulation/immunology , Enzyme Induction/drug effects , Enzyme Induction/physiology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Infliximab , Matrix Metalloproteinase Inhibitors/metabolism , Matrix Metalloproteinase Inhibitors/toxicity , Matrix Metalloproteinases/biosynthesis , Occludin/metabolism , Swine
20.
Tumour Biol ; 33(6): 2307-15, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22941467

ABSTRACT

Accumulating evidence reveals that aberrant expression of claudins manifests in various tumors; however, their biological functions are poorly understood. Here, we report on the elevated expression of claudin-1 in human breast cancer MCF-7 cells under tumor necrosis factor (TNF)-α treatment. Interestingly, the increased expression of claudin-1 contributes to an anti-apoptotic role in TNF-α-induced apoptosis. In line with this, upon TNF-α stimulus, downregulation of claudin-1 by siRNA knockdown results in a significant increase in cleavage of caspase-8 and poly (ADP-ribose) polymerase, a decrease of cyclinD1 expression, and DNA fragmentation. Consistently, TdT-mediated dUTP nick end labeling assay also shows that loss of claudin-1 increases the susceptibility of MCF-7 cells to TNF-α-induced apoptosis. However, there is no obvious effect on the expression of Bax and p53 after the treatment aforementioned. In addition, TNF-α increases the amount of claudin-1 and the cytoplasmic accumulation of ß-catenin, while claudin-1 siRNA increases the amount of ß-catenin in the cell membrane as well as the amount of E-cadherin in the cytoplasm. In conclusion, our data reveal a novel role of claudin-1 in regulating apoptosis in MCF-7 cells.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Claudin-1/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Blotting, Western , Breast Neoplasms/metabolism , Cadherins/genetics , Cadherins/metabolism , Caspases/genetics , Caspases/metabolism , Cell Membrane , Cell Proliferation/drug effects , Claudin-1/antagonists & inhibitors , Claudin-1/genetics , Cytoplasm , Female , Fluorescent Antibody Technique , Humans , Protein Transport , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Subcellular Fractions , Tumor Cells, Cultured , beta Catenin/genetics , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...