Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
ACS Appl Mater Interfaces ; 14(5): 6358-6369, 2022 Feb 09.
Article in English | MEDLINE | ID: mdl-35099925

ABSTRACT

Ulcerative colitis (UC) is a chronic inflammatory bowel disease (IBD) of unknown etiology affecting the colon and rectum. Previous studies have found that reactive oxygen species (ROS) overproduction and transmembrane glycoprotein CD98 (encoded by SLC3A2) upregulation played important roles in the initiation and progression of UC. On the basis of this, a biomimetic pH-responsive metal organic framework (MOF) carrier was constructed to deliver carbon nanodot-SOD nanozyme and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) (CRISPR/Cas9) system for site-specific treatment of UC. In this system, carbon nanodots (C-dots) and CD98 CRISPR/Cas9 plasmid were successfully encapsulated into MOF carrier (ZIF-8 nanoparticles) by a one-pot approach (formed as CCZ), and then camouflaged with macrophage membrane (formed as CCZM). It was worth noting that the C-dot nanozyme showed excellent superoxide dismutase (SOD) enzymatic activity, which could scavenge ROS effectively. As expected, this biomimetic system exhibited pH-responsive, immune escape, and inflammation targeting capability simultaneously. In vitro experiments showed that ROS was significantly eliminated, and CD98 was downregulated by CCZM. In the dextran sulfate sodium salt (DSS)-induced UC model, administration of CCZM significantly ameliorated the inflammation symptoms of mice, including the colon length and pathological parameters such as epithelium integrity and inflammation infiltration. In addition, both in vitro and in vivo results demonstrated that biomimetic nanoparticles effectively reduced the expression of pro-inflammatory cytokines. Overall, this study would provide a promising approach for the precise treatment of UC.


Subject(s)
Biomimetic Materials/chemistry , CRISPR-Cas Systems/genetics , Imidazoles/chemistry , Metal-Organic Frameworks/chemistry , Nanoparticles/chemistry , Quantum Dots/chemistry , Superoxide Dismutase/chemistry , Animals , Carbon/chemistry , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colon/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Drug Carriers/chemistry , Female , Fusion Regulatory Protein-1/genetics , Fusion Regulatory Protein-1/metabolism , Humans , Mice , Mice, Inbred C57BL , Plasmids/genetics , Plasmids/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/therapeutic use
2.
Sci Rep ; 8(1): 16220, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30385787

ABSTRACT

CD98 has been implicated in the experimental model of inflammatory bowel disease. We have previously shown that IEC-specific overexpression of CD98 mediates intestinal inflammation and intestinal epithelial barrier dysfunction. Mice overexpressing CD98 exhibited severe colitis and a greater susceptibility to CAC. Here we demonstrated CD98 overexpression to dysregulate homeostatic gradient profile of miRNA and protein expression along the ileal villus-crypt axis. Using miRNA-target gene prediction module, we observed differentially expressed miRNAs to target proteins of villus and crypt profoundly affected by CD98 overexpression. We have utilized online bioinformatics as methods to further scrutinize the biological meanings of miRNA-target data. We identified significant interactions among the differentially regulated proteins targeted by altered miRNAs in Tg mice. The biological processes affected by the predicted targets of miRNAs deviate from the homeostatic functions of the miRNA-gene-protein axis of the wildtype mice. Our results emphasize a dynamic perturbation of miRNA and protein expression in villus-crypt axis contributing to potential biological consequences of altering CD98 expression. Our findings also suggest the need for a consideration of arrays of interacting biological entities (i.e. miRNAs-mRNAs, protein-protein interaction) or a combination comparison for a better understanding of the disease pathology which is necessary for an effective therapeutic target development.


Subject(s)
Fusion Regulatory Protein-1/genetics , Gene Expression Regulation , Gene Expression , Ileum/metabolism , Intestinal Mucosa/metabolism , MicroRNAs/genetics , RNA Interference , Animals , Computational Biology/methods , Female , Fusion Regulatory Protein-1/metabolism , Gene Expression Profiling , Gene Regulatory Networks , Intestinal Mucosa/ultrastructure , Mice
3.
ACS Nano ; 12(6): 5253-5265, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29860836

ABSTRACT

Colon cancer is among the most widely occurring cancer types, leading to considerably high mortality rate. The current chemotherapy achieves only limited success, and more effective therapeutic strategies are urgently needed. Human colonic biopsy specimens indicate increased expression of CD98 in patients with colon cancer, suggesting that CD98 might be a potential therapeutic target and/or a receptor for targeted drug delivery in colon cancer treatment. Herein, we coloaded CD98 siRNA (siCD98) and camptothecin (CPT) into CD98 Fab'-functionalized nanoparticles (NPs). The resultant Fab'-siCD98/CPT-NPs showed good monodispersity with an average diameter of approximately 270 nm and a ζ-potential of around -24 mV. These NPs mediated efficient drug delivery to the target cancer cells and tumor tissues, producing much better anticancer and antimigration effects compared to other relevant NPs. Mouse models with orthotopic colon tumors were treated with NP-embedded hydrogel, which revealed that Fab'-siCD98/CPT-NPs exhibited a therapeutic efficacy significantly better than that of single drug-loaded NPs or nonfunctionalized siCD98/CPT-NPs. This study indicates that the Fab'-siCD98/CPT-NP/hydrogel system is able to realize specific release of NPs in the colonic lumen and enable drugs (siCD98 and CPT) to be internalized into target cells, demonstrating a notable potential for clinical applications in colon-cancer-targeted combination therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/pharmacology , Colonic Neoplasms/drug therapy , Fusion Regulatory Protein-1/deficiency , Intestines/drug effects , Nanoparticles/administration & dosage , RNA, Small Interfering/administration & dosage , Administration, Oral , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Models, Animal , Drug Delivery Systems , Drug Screening Assays, Antitumor , Female , Fusion Regulatory Protein-1/biosynthesis , Fusion Regulatory Protein-1/genetics , Humans , Intestines/pathology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology
4.
Nanomedicine (Lond) ; 12(16): 1927-1943, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28665164

ABSTRACT

AIM: To develop novel siRNA delivery system overcoming the limitations of synthetic nanoparticles, such as potential side effects, nonspecificity and economic production for ulcerative colitis therapy. MATERIALS & METHODS: Nanoparticles composed of edible ginger-derived lipid, termed ginger-derived lipid vehicles (GDLVs) were generated from ginger lipids through hydration of a lipid film, a commonly used method for a liposome fabrication. The morphology, biocompatibility and transfection efficiency of GDLVs loaded with siRNA-CD98 (siRNA-CD98/GDLVs) were characterized by standard methods. RESULTS: Orally administered siRNA-CD98/GDLVs were effectively targeted specifically to colon tissues, resulting in reduced expression of CD98. CONCLUSION: These GDLVs have great promise as efficient siRNA-delivery vehicles while potentially obviating issues related to the traditional synthetic nanoparticles. As such, they help shift the current paradigm of siRNA delivery away from artificially synthesized nanoparticles toward the use of naturally derived nanovehicles from edible plants.


Subject(s)
Colitis, Ulcerative/therapy , Fusion Regulatory Protein-1/metabolism , Lipids/chemistry , Nanoparticles/chemistry , RNA, Small Interfering/administration & dosage , Zingiber officinale/chemistry , Administration, Oral , Animals , Apoptosis , Cell Line , Colon/drug effects , Colon/metabolism , Drug Carriers , Fusion Regulatory Protein-1/genetics , Genetic Therapy , Humans , Liposomes/chemistry , Mice , Particle Size , Surface Properties , Transfection
5.
Oncotarget ; 8(6): 10050-10063, 2017 Feb 07.
Article in English | MEDLINE | ID: mdl-28052023

ABSTRACT

The male germinal lineage, which is defined as unipotent, produces sperm through spermatogenesis. However, embryonic primordial germ cells and postnatal spermatogonial stem cells (SSCs) can change their fate and convert to pluripotency in culture when they are not controlled by the testicular microenvironment. The mechanisms underlying these reprogramming processes are poorly understood. Testicular germ cell tumors, including teratoma, share some molecular characteristics with pluripotent cells, suggesting that cancer could result from an abnormal differentiation of primordial germ cells or from an abnormal conversion of SCCs to pluripotency in the testis. Here, we investigated whether the somatic reprogramming factors Oct3/4, Sox2, Klf4 and c-Myc (OSKM) could play a role in SSCs reprogramming and induce pluripotency using a doxycycline-inducible transgenic Col1a1-4F2A-OSKM mouse model. We showed that, in contrast to somatic cells, SSCs from adult mice are resistant to this reprogramming strategy, even in combination with small molecules, hypoxia, or p53 deficiency, which were previously described to favour the conversion of somatic cells to pluripotency. This finding suggests that adult SSCs have developed specific mechanisms to repress reprogramming by OSKM factors, contributing to circumvent testicular cancer initiation events.


Subject(s)
Adult Germline Stem Cells/metabolism , Cellular Reprogramming Techniques , Cellular Reprogramming , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Transcription Factors/metabolism , Mouse Embryonic Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins c-myc/metabolism , SOXB1 Transcription Factors/metabolism , Adult Germline Stem Cells/drug effects , Animals , Cell Hypoxia , Cell Lineage , Cells, Cultured , Cellular Reprogramming/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Fusion Regulatory Protein-1/genetics , Fusion Regulatory Protein-1/metabolism , Gene Expression Regulation, Developmental , Genotype , Induced Pluripotent Stem Cells/pathology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mouse Embryonic Stem Cells/drug effects , Octamer Transcription Factor-3/genetics , Phenotype , Proto-Oncogene Proteins c-myc/genetics , SOXB1 Transcription Factors/genetics , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
Neoplasma ; 64(2): 283-288, 2017.
Article in English | MEDLINE | ID: mdl-28052681

ABSTRACT

L-type amino acid transporter 1 (LAT1) and CD98 are frequently expressed in various human cancers, and closely correlated with tumor aggressiveness and survival. However, little is known about the expression of LAT1 and CD98 in cutaneous angiosarcoma. The aim of this study is to investigate the clinicopathological significance of these markers in the dismal disease. A total of 52 patients with cutaneous angiosarcoma were retrospectively reviewed. Immunohistochemical staining of tumor specimens were evaluated using anti-LAT1, CD98 and Ki-67 antibodies. The rates of high expression for LAT1 and CD98 were 56% (29/52) and 79% (41/52), respectively. The frequency of high expression for CD98 was significantly higher than that for LAT1 (p=0.021). The low expression of CD98 was significantly associated with distant metastasis (p=0.044) and was identified as a significant prognostic predictor by multivariate analysis. The expression level of LAT1 was not significantly correlated with prognosis. The low expression of CD98 is a novel biomarker for predicting poor prognosis in patients with cutaneous angiosarcoma.


Subject(s)
Fusion Regulatory Protein-1/genetics , Hemangiosarcoma/genetics , Large Neutral Amino Acid-Transporter 1/genetics , Biomarkers, Tumor/genetics , Hemangiosarcoma/diagnosis , Humans , Prognosis , Retrospective Studies
7.
Dig Liver Dis ; 49(2): 188-196, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27939923

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is characterized by excessive lipid hepatic accumulation. Here, we investigated whether a reduction of CD98 expression mediated by CD98 siRNA-loaded nanoparticles (NPs) could attenuate liver disease markers in a mouse model of NAFLD. NPs were generated using a double emulsion/solvent evaporation technique. Mice fed a high fat diet for 8 weeks to induce fatty liver were treated with vein tail injections of CD98 siRNA-loaded NPs. In vitro, HepG2 treated with CD98 siRNA-loaded NPs showed significant downregulation of CD98 leading to a significant decrease of major pro-inflammatory cytokines and markers. In vivo, CD98 siRNA-loaded NPs strongly decreased all markers of NAFLD, including the blood levels of ALT and lipids accumulation, fibrosis evidence and pro-inflammatory cytokines. In conclusion, our results indicate that CD98 appears to function as a key actor/inducer in NAFLD, and that our NPs approach may offer a new targeted therapeutic for this disease.


Subject(s)
Fusion Regulatory Protein-1/genetics , Gene Silencing , Non-alcoholic Fatty Liver Disease/therapy , RNA, Small Interfering/genetics , Animals , Biomarkers/blood , Diet, High-Fat , Disease Models, Animal , Down-Regulation , Female , Humans , Mice , Nanoparticles/chemistry
8.
Parasitol Int ; 66(4): 471-478, 2017 Aug.
Article in English | MEDLINE | ID: mdl-26657242

ABSTRACT

L-type amino acid transporter 1 (LAT1) is highly expressed in various human cancers, including cholangiocarcinoma (CCA), the most common cancer in Northeast Thailand. Chronic inflammation and oxidative stress induced by liver fluke, Opisthorchis viverrini, infection has been recognized as the major cause of CCA in this area. We show here that an increased expression of LAT1 and its co-functional protein CD98 are found during carcinogenesis induced by Ov in hamster CCA tissues. We also demonstrate that oxidative stress induced by H2O2 is time-dependent and dramatically activates LAT1 and CD98 expression in immortal cholangiocytes (MMNK1). In addition, H2O2 treatment increased LAT1 and CD98 expression, as well as an activated form of AKT and mTOR in MMNK1 and CCA cell lines (KKU-M055 and KKU-M213). We also show that suppression of PI3K/AKT pathway activity with a dual PI3K/mTOR inhibitor, BEZ235, causes a reduction in LAT1 and CD98 expression in KKU-M055 and KKU-M213 in parallel with a reduction of activated AKT and mTOR. Interestingly, high expression of LAT1 in human CCA tissues is a significant prognostic factor for shorter survival. Taken together, our data show that LAT1 expression is significantly associated with CCA progression and cholangiocarcinogenesis induced by oxidative stress. Moreover, the expression of LAT1 and CD98 in CCA is possibly regulated by the PI3K/AKT signaling pathway.


Subject(s)
Bile Duct Neoplasms/metabolism , Cholangiocarcinoma/metabolism , Fusion Regulatory Protein-1/genetics , Large Neutral Amino Acid-Transporter 1/genetics , Opisthorchiasis/metabolism , Opisthorchis/physiology , Adult , Aged , Animals , Bile Duct Neoplasms/diagnosis , Cell Line , Cholangiocarcinoma/diagnosis , Cricetinae , Female , Fusion Regulatory Protein-1/metabolism , Humans , Large Neutral Amino Acid-Transporter 1/metabolism , Male , Mesocricetus , Middle Aged , Opisthorchiasis/complications , Opisthorchiasis/diagnosis , Oxidative Stress , Prognosis , Tissue Array Analysis , Up-Regulation
9.
Cancer Cell ; 30(5): 792-805, 2016 Nov 14.
Article in English | MEDLINE | ID: mdl-27908736

ABSTRACT

Acute myelogenous leukemia (AML) is an aggressive disease associated with drug resistance and relapse. To improve therapeutic strategies, it is critical to better understand the mechanisms that underlie AML progression. Here we show that the integrin binding glycoprotein CD98 plays a central role in AML. CD98 promotes AML propagation and lethality by driving engagement of leukemia cells with their microenvironment and maintaining leukemic stem cells. Further, delivery of a humanized anti-CD98 antibody blocks growth of patient-derived AML, highlighting the importance of this pathway in human disease. These findings indicate that microenvironmental interactions are key regulators of AML and that disrupting these signals with targeted inhibitors such as CD98 antibodies may be a valuable therapeutic approach for adults and children with this disease.


Subject(s)
Antibodies/administration & dosage , Fusion Regulatory Protein-1/genetics , Leukemia, Myeloid, Acute/drug therapy , Neoplastic Stem Cells/pathology , Animals , Antibodies/pharmacology , Cell Adhesion , Cell Line, Tumor , Cell Proliferation/drug effects , Fusion Regulatory Protein-1/antagonists & inhibitors , Gene Knockout Techniques , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Neoplasm Transplantation
10.
Int J Mol Sci ; 17(11)2016 Nov 10.
Article in English | MEDLINE | ID: mdl-27834933

ABSTRACT

Hepatocellular carcinoma (HCC) is currently the third most common cause of cancer-related death in the Asia-Pacific region. Our previous work showed that knockdown of CD98 significantly inhibits malignant HCC cell phenotypes in vitro and in vivo. The level of CD98 in the membrane is tightly regulated to mediate complex processes associated with cell-cell communication and intracellular signaling. In addition, the intracellular domain of CD98 (CD98-ICD) seems to be of vital importance for recycling CD98 to the membrane after it is endocytosed. The intracellular and transmembrane domains of CD98 associate with ß-integrins (primarily ß1 but also ß3), and this association is essential for CD98 mediation of integrin-like signaling and complements dominant suppression of ß1-integrin. We speculated that isolated CD98-ICD would similarly suppress ß1-integrin activation and inhibit the malignant behaviors of cancer cells. In particular, the exact role of CD98-ICD has not been studied independently in HCC. In this study, we found that ectopic expression of CD98-ICD inhibited the malignant phenotypes of HCC cells, and the mechanism possibly involves ß1-integrin suppression. Moreover, the expression levels of CD98, ß1-integrin-A (the activated form of ß1-integrin) and Ki-67 were significantly increased in HCC tissues relative to those of normal liver tissues. Therefore, our preliminary study indicates that ectopic CD98-ICD has an inhibitory role in the malignant development of HCC, and shows that CD98-ICD acts as a dominant negative mutant of CD98 that attenuates ß1-integrin activation. CD98-ICD may emerge as a promising candidate for antitumor treatment.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Fusion Regulatory Protein-1/metabolism , Integrin beta1/metabolism , Liver Neoplasms/metabolism , Animals , Binding Sites/genetics , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Disease Progression , Flow Cytometry , Fusion Regulatory Protein-1/genetics , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , Microscopy, Confocal , Transfection , Transplantation, Heterologous , Tumor Burden
11.
Cancer Sci ; 107(10): 1499-1505, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27465934

ABSTRACT

System l amino acid transporter 1 (LAT1) is highly expressed in various types of human cancer, and contributes to cancer growth and survival. Recently, we have shown that LAT1 expression is closely related to the growth and aggressiveness of esophageal cancer, and is an independent marker of poor prognosis. However, it remains unclear whether LAT1 inhibition could suppress esophageal cancer growth. In this study, we investigated the tumor-suppressive effects of the inhibition of LAT1. Both LAT1 and CD98, which covalently associates to LAT1 on the membrane, were expressed in human esophageal cancer cell lines KYSE30 and KYSE150. Quantitative PCR analysis showed that the expression of LAT1 was much higher than other subtypes of LAT. A selective inhibitor of LAT, 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid (BCH), suppressed cellular uptake of l-14 C-leucine and cell proliferation in a dose-dependent manner. It also suppressed phosphorylation of mammalian target of rapamycin, 4E-BP1, and p70S6K protein, and induced cell cycle arrest at G1 phase. These results suggest that suppression of both mammalian target of rapamycin signaling and cell cycle progression is involved in BCH-induced growth inhibition. In tumor-bearing mice, daily treatment with BCH significantly delayed tumor growth and decreased glucose metabolism, indicating that LAT1 inhibition potentially suppresses esophageal cancer growth in vivo. Thus, our results suggest that LAT1 inhibition could be a promising molecular target for the esophageal cancer therapy.


Subject(s)
Amino Acid Transport System L/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Amino Acid Transport System L/genetics , Amino Acid Transport System L/metabolism , Amino Acids/metabolism , Animals , Antineoplastic Agents/administration & dosage , Biological Transport/drug effects , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Fusion Regulatory Protein-1/genetics , Fusion Regulatory Protein-1/metabolism , Gene Expression Profiling , Humans , Lactate Dehydrogenases/metabolism , Male , Mice , Molecular Targeted Therapy , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Transcriptome , Xenograft Model Antitumor Assays
12.
Colloids Surf B Biointerfaces ; 143: 186-193, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27011348

ABSTRACT

CD98 plays an important role in the development and progression of inflammation. Here, CD98 siRNA (siCD98) was complexed with urocanic acid-modified chitosan (UAC) to form nanoparticles (NPs), which were transfected into Raw 264.7 macrophages in an effort to convey anti-inflammatory effects. Characterization showed that the generated NPs had a desirable particle size (156.0-247.1nm), a slightly positive zeta potential (15.8-17.5mV), and no apparent cytotoxicity against Raw 264.7 macrophages and colon-26 cells compared to control NPs fabricated by Oligofectamine (OF) and siRNA. Cellular uptake experiments demonstrated that macrophages exhibited a time-dependent accumulation profile of UAC/siRNA NPs. Further in vitro gene silencing experiments revealed that UAC/siCD98 NPs with a weight ratio of 60:1 yielded the most efficient knockdowns of CD98 and the pro-inflammatory cytokine, TNF-α. Indeed, the RNAi efficiency obtained with our NPs was even higher than that of the positive control OF/siCD98 NPs. These results suggest that UAC/siCD98 NPs might be a safe, efficient and promising candidate for the treatment of inflammatory disease.


Subject(s)
Chitosan/chemistry , Fusion Regulatory Protein-1/genetics , Macrophages/metabolism , Nanoparticles/chemistry , RNA, Small Interfering/genetics , Urocanic Acid/chemistry , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Inflammation/genetics , Inflammation/metabolism , Inflammation/prevention & control , Inflammation Mediators/metabolism , Macrophages/cytology , Macrophages/drug effects , Mice , Microscopy, Electron, Transmission , Molecular Structure , Nanoparticles/administration & dosage , Nanoparticles/ultrastructure , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , X-Ray Diffraction
13.
J Exp Clin Cancer Res ; 34: 110, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26437640

ABSTRACT

BACKGROUND: Dysregulated endocytosis of membrane proteins contributes significantly to several hallmarks of cancer. Basigin can enhance cancer progression, but its precise mechanism remains unclear. CD98 promotes cell spreading and tumorigenicity by triggering integrin clustering and enhancing cell adhesion to the extracellular matrix. The endocytosis and recyle of basigin and CD98 might play critical roles in cancer. METHODS: The role of CD98 was confirmed in liver cancer cells by cell spreading in vitro and tumorigenicity by nude mice xenograft tumor assay in vivo; membrane expression of basigin and CD98 in SMMC-7721 was measured by FCAS; pull down and SPR analysis were uses to reveal the direct association between basigin and CD98; DsRed1 tagged CD98 was blocked in the cytoplasm in K7721 (whose basigin was knockn out) and had a well colocalization with ER and Rab5a positive recycling endosomes under co-focal; finally, by FRET imaging and FCAS we observed the internalization of basigin and CD98 was flotillin-1-regulated, and their recycle at early steps was Arf6-mediated. RESULTS: Basigin and CD98 were highly expressed and co-localized on the human hepatocellular carcinoma (HCC) cell membrane; basigin can directly bind to CD98, mediating CD98 redistribution on the HCC cell membrane and activating the downstream integrin signaling pathway. Internalization of basigin and CD98 was flotillin-1 regulated the and their recycling was mediated by Arf6. This recycling process for basigin and CD98 promotes cell spreading and tumor growth in liver cancer xenografts. CONCLUSION: Basigin, as a redistribution chaperone of CD98, plays a critical role in promoting cell spreading and the progression of hepatocellular carcinoma.


Subject(s)
Basigin/genetics , Carcinoma, Hepatocellular/pathology , Cell Movement/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Fusion Regulatory Protein-1/genetics , Liver Neoplasms/pathology , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/metabolism , Animals , Cell Adhesion/physiology , Cell Line, Tumor , Cell Membrane/metabolism , Endocytosis/physiology , Extracellular Matrix/metabolism , Female , Humans , Integrins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction/physiology , Surface Plasmon Resonance , Xenograft Model Antitumor Assays/methods
14.
Chem Biol ; 22(2): 217-28, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25641165

ABSTRACT

Human ß-defensins play a pivotal role in the innate immune response. Although expressed by and acting at epithelial surfaces, little is known about their specific interaction with epithelial structures. Here, we identify the transmembrane protein CD98 as a cell surface receptor involved in the internalization of human ß-defensin 3 (hBD3) in human epithelial A549 cells. CD98 and hBD3 extensively colocalize on the basolateral domain of A549. While verifying their direct binding by fluorescence resonance energy transfer and surface plasmon resonance, we mapped the interaction to CD98 residues 304-414, i.e. to the region known to interact with the proteins of intestinal bacteria during colonic invasion. Treatment of A549 cells with hBD3 dramatically reduces CD98 expression and conversely, knockdown of CD98 expression impairs hBD3 cell surface binding and internalization. Competition for bacterial binding to CD98 and downregulation of CD98 expression may represent novel mechanisms for the antibacterial activity of hBD3.


Subject(s)
Fusion Regulatory Protein-1/metabolism , beta-Defensins/metabolism , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Biotin/chemistry , Cell Line, Tumor , Epithelial Cells/cytology , Epithelial Cells/metabolism , Escherichia coli/drug effects , Fluorescence Resonance Energy Transfer , Fusion Regulatory Protein-1/antagonists & inhibitors , Fusion Regulatory Protein-1/genetics , Gene Expression Regulation/drug effects , Humans , Microscopy, Confocal , Molecular Dynamics Simulation , Protein Binding , Protein Structure, Tertiary , Protein Transport , Proteomics , RNA Interference , RNA, Small Interfering/metabolism , Surface Plasmon Resonance , beta-Defensins/chemistry , beta-Defensins/pharmacology
15.
Sci Rep ; 4: 6941, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25373594

ABSTRACT

Although it is recognized that the abnormal accumulation of amino acid is a cause of the symptoms in metabolic disease such as phenylketonuria (PKU), the relationship between disease severity and serum amino acid levels is not well understood due to the lack of experimental model. Here, we present a novel in vitro cellular model using K562-D cells that proliferate slowly in the presence of excessive amount of phenylalanine within the clinically observed range, but not phenylpyruvate. The increased expression of the L-type amino acid transporter (LAT2) and its adapter protein 4F2 heavy chain appeared to be responsible for the higher sensitivity to phenylalanine in K562-D cells. Supplementation with valine over phenylalanine effectively restored cell proliferation, although other amino acids did not improve K562-D cell proliferation over phenylalanine. Biochemical analysis revealed mammalian target of rapamycin complex (mTORC) as a terminal target of phenylalanine in K562-D cell proliferation, and supplementation of valine restored mTORC1 activity. Our results show that K562-D cell can be a potent tool for the investigation of PKU at the molecular level and to explore new therapeutic approaches to the disease.


Subject(s)
Models, Biological , Phenylalanine/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Biological Transport , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Fusion Regulatory Protein-1/genetics , Fusion Regulatory Protein-1/metabolism , Gene Expression Regulation , Hemin/pharmacology , Humans , K562 Cells , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Phenylalanine/pharmacology , Phenylketonurias/genetics , Phenylketonurias/metabolism , Phenylketonurias/pathology , Phenylpyruvic Acids/metabolism , Phenylpyruvic Acids/pharmacology , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Valine/metabolism , Valine/pharmacology
16.
Ann Surg Oncol ; 21(13): 4359-68, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25084765

ABSTRACT

BACKGROUND: It has been reported that CD147 and CD98 heavy chain (CD98hc) form a complex on the cell plasma membrane of several cancers; however, whether this complex exists in non-small cell lung cancer (NSCLC) cells and affects the prognosis of patients remains to be elucidated. METHODS: The expression of CD147 and CD98hc was assessed in tissue samples from 241 NSCLC patients and NSCLC cell lines. The correlation between CD147 and CD98hc expression and their association with the prognosis of NSCLC patients were analyzed. We also evaluated the impact of CD147 and CD98hc on the growth of NSCLC cells as well as Akt phosphorylation. RESULTS: Both CD147 and CD98hc were significantly upregulated in NSCLC cells, and their expression levels were significantly correlated (p < 0.001). Immunoflurenece staining and co-immunoprecipitation demonstrated that CD147 and CD98hc could form a complex on NSCLC cells. Compared with NSCLC patients with CD147-/CD98hc-, those with CD147+/CD98hc+ exhibited a significantly poor overall survival (OS) with a hazard ratio (HR) of 1.92 (p = 0.010), and a significantly increased risk of recurrence with a HR of 1.97 (p = 0.004). Also, we demonstrated that the proliferation of lung cancer cell lines was significantly affected by knockdown and force-expression of the CD147-CD98hc complex. Western blot analysis indicated that the phosphorylation of Akt in NSCLC cells was significantly affected by knockdown and overexpression of either or both CD147 and CD98hc. CONCLUSIONS: Our findings indicate that the CD147-CD98hc complex significantly contributes to poor prognosis of NSCLC patients through promoting cell proliferation via the PI3K/Akt pathway.


Subject(s)
Basigin/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation , Fusion Regulatory Protein-1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Basigin/chemistry , Basigin/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Cell Membrane/metabolism , Female , Fluorescent Antibody Technique , Follow-Up Studies , Fusion Regulatory Protein 1, Heavy Chain , Fusion Regulatory Protein-1/antagonists & inhibitors , Fusion Regulatory Protein-1/genetics , Humans , Immunoprecipitation , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Prognosis , RNA, Small Interfering/genetics , Signal Transduction , Survival Rate
17.
Cancer Med ; 3(5): 1246-55, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24890221

ABSTRACT

Oncocytic L-type amino acid transporter (LAT) 1 may be a prognostic indicator and target of new molecular therapeutic agents against malignancies. To investigate whether LAT1 expression influence the outcomes of patients with bile duct cancer, the expression of LAT1, LAT2, CD98, and Ki-67 was investigated immunohistochemically in 134 surgically resected bile duct adenocarcinomas, including 84 distal extrahepatic bile duct adenocarcinomas, 21 hilar cholangiocarcinomas, 15 intrahepatic cholangiocarcinomas, and 14 ampullary adenocarcinomas. LAT1 expression was weakly correlated with CD98 expression and Ki-67 labeling index (LI). Kaplan-Meier analysis showed a significant difference in prognosis between patients with bile duct adenocarcinomas having LAT1-high and -low scores, whereas LAT2 and CD98 expression and Ki-67 LI were not predictive of poor prognosis. Prognosis tended to be worse in patients having tumors with LAT1-high/LAT2-low than LAT1-low/LAT2-high scores (P = 0.0686). Multivariable analyses revealed that LAT1 expression, surgical margin, pT stage were independent prognostic factors. In conclusion, aberrant overexpression of LAT1 in bile duct adenocarcinoma predicts poor prognosis, suggesting that LAT1 may be a potential target of anticancer therapy.


Subject(s)
Adenocarcinoma/metabolism , Amino Acid Transport Systems/metabolism , Bile Duct Neoplasms/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Adult , Aged , Aged, 80 and over , Amino Acid Transport Systems/genetics , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/mortality , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/surgery , Female , Fusion Regulatory Protein-1/genetics , Fusion Regulatory Protein-1/metabolism , Gene Expression , Humans , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Odds Ratio , Prognosis
18.
Cancer Sci ; 105(4): 396-401, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24484217

ABSTRACT

A mimotope is an antibody-epitope-mimicking peptide retrieved from a phage display random peptide library. Immunization with antitumor antibody-derived mimotopes is promising for inducing antitumor immunity in hosts. In this study, we isolated linear and constrained mimotopes from HBJ127, a tumor-suppressing anti-CD98 heavy chain mAb, and determined their abilities for induction of antitumor activity equal to that of the parent antibody. We detected elevated levels of antipeptide responses, but failed to detect reactivity against native CD98-expressing HeLa cells in sera of immunized mice. Phage display panning and selection of mimotope-immunized mouse spleen-derived antibody Fab library showed that HeLa cell-reactive Fabs were successfully retrieved from the library. This finding indicates that native antigen-reactive Fab clones represented an undetectable minor population in mimotope-induced antibody repertoire. Functional and structural analysis of retrieved Fab clones revealed that they were almost identical to the parent antibody. From these results, we confirmed that mimotope immunization was promising for retrieving antitumor antibodies equivalent to the parent antibody, although the co-administration of adjuvant compounds such as T-cell epitope peptides and Toll-like receptor 4 agonist peptides is likely to be necessary for inducing stronger antitumor immunity than mimotope injection alone.


Subject(s)
Antibodies/immunology , Fusion Regulatory Protein-1/immunology , Neoplasms/immunology , Animals , Antibodies/isolation & purification , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Epitopes/genetics , Epitopes/immunology , Fusion Regulatory Protein-1/genetics , HeLa Cells , Humans , Immunization , Mice , Neoplasms/genetics , Neoplasms/therapy , Peptide Library , Peptides/administration & dosage , Peptides/chemistry , Peptides/immunology , Toll-Like Receptor 4/metabolism
19.
Gastroenterology ; 146(5): 1289-300.e1-19, 2014 May.
Article in English | MEDLINE | ID: mdl-24503126

ABSTRACT

BACKGROUND & AIMS: Nanoparticles have been explored as carriers of small interfering RNAs (siRNAs) and might be developed to treat patients with inflammatory bowel disease (IBD). Overexpression of CD98 on the surface of colonic epithelial cells and macrophages promotes the development and progression of IBD. We developed an orally delivered hydrogel that releases nanoparticles with single-chain CD98 antibodies on their surface (scCD98 functionalized) and loaded with CD98 siRNA (siCD98). We tested the ability of the nanoparticles to reduce levels of CD98 in the colons of mice with colitis. METHODS: scCD98-functionalized siCD98-loaded nanoparticles were fabricated using a complex coacervation technique. We investigated the cellular uptake and lysosome escape profiles of the nanoparticles in Colon-26 cells and RAW 264.7 macrophages using fluorescence microscopy. Colitis was induced by transfer of CD4(+)CD45RB(high) T cells to Rag(-/-) mice or administration of dextran sodium sulfate to C57BL/6 mice. Mice were then given hydrogel (chitosan and alginate) containing scCD98-functionalized nanoparticles loaded with siCD98 or scrambled siRNA (control) via gavage. RESULTS: The scCD98-functionalized nanoparticles were approximately 200 nm in size and had high affinity for CD98-overexpressing cells. The scCD98-functionalized siCD98-loaded nanoparticles significantly reduced levels of CD98 in Colon-26 cells and RAW 264.7 macrophages, along with production of inflammatory cytokines (tumor necrosis factor α, interleukin-6, and interleukin-12). In mice with colitis, administration of the scCD98-functionalized siCD98-loaded nanoparticles reduced colon expression of CD98. Importantly, the severity of colitis was also reduced compared with controls (based on loss of body weight, myeloperoxidase activity, inflammatory cytokine production, and histological analysis). Approximately 24.1% of colonic macrophages (CD11b(+)CD11c(-)F4/80(+)) in the mice had taken up fluorescently labeled siRNA-loaded nanoparticles within 12 hours of administration. CONCLUSIONS: Nanoparticles containing surface CD98 antibody and loaded with siCD98 reduce expression of this protein by colonic epithelial cells and macrophages, and oral administration decreases the severity of colitis in mice. This nanoparticle in hydrogel (chitosan/alginate) formulation might be developed to treat patients with IBD.


Subject(s)
Colitis/prevention & control , Colon/metabolism , Fusion Regulatory Protein-1/genetics , Fusion Regulatory Protein-1/immunology , Genetic Therapy/methods , Nanomedicine/methods , Nanoparticles , RNA Interference , RNA, Small Interfering/administration & dosage , Single-Chain Antibodies/administration & dosage , Administration, Oral , Alginates/chemistry , Animals , Cell Line , Chitosan/chemistry , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Colitis/metabolism , Colitis/pathology , Colon/immunology , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Hydrogels , Interleukin-12/metabolism , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Small Interfering/metabolism , Severity of Illness Index , Time Factors , Tumor Necrosis Factor-alpha/metabolism
20.
Mol Ther ; 22(1): 69-80, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24025751

ABSTRACT

Intestinal CD98 expression plays a crucial role in controlling homeostatic and innate immune responses in the gut. Modulation of CD98 expression in intestinal cells therefore represents a promising therapeutic strategy for the treatment and prevention of inflammatory intestinal diseases, such as inflammatory bowel disease. Here, the advantages of nanoparticles (NPs) are used, including their ability to easily pass through physiological barriers and evade phagocytosis, high loading concentration, rapid kinetics of mixing and resistance to degradation. Using physical chemistry characterizations techniques, CD98 siRNA/polyethyleneimine (PEI)-loaded NPs was characterized (diameter of ~480 nm and a zeta potential of -5.26 mV). Interestingly, CD98 siRNA can be electrostatically complexed by PEI and thus protected from RNase. In addition, CD98 siRNA/PEI-loaded NPs are nontoxic and biocompatible with intestinal cells. Oral administration of CD98/PEI-loaded NPs encapsulated in a hydrogel reduced CD98 expression in mouse colonic tissues and decreased dextran sodium sulfate-induced colitis in a mouse model. Finally, flow cytometry showed that CD98 was effectively downregulated in the intestinal epithelial cells and intestinal macrophages of treated mice. Finally, the results collectively demonstrated the therapeutic effect of "hierarchical nano-micro particles" with colon-homing capabilities and the ability to directly release "molecularly specific" CD98 siRNA in colonic cells, thereby decreasing colitis.


Subject(s)
Fusion Regulatory Protein-1/chemistry , Inflammation/metabolism , Intestinal Mucosa/metabolism , Nanoparticles/chemistry , Polyethyleneimine/chemistry , RNA, Small Interfering/chemistry , Animals , Cell Line , Cell Survival , Colon/drug effects , Colon/metabolism , Colon/pathology , Dendritic Cells/metabolism , Dextran Sulfate/adverse effects , Fusion Regulatory Protein-1/genetics , Gene Transfer Techniques , Inflammation/chemically induced , Inflammation/genetics , Inflammation/pathology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestines/drug effects , Intestines/pathology , Macrophages/metabolism , Mice , Nanoparticles/administration & dosage , Nanoparticles/ultrastructure , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...