Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Oncogene ; 36(32): 4629-4640, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28368417

ABSTRACT

Accumulating studies have demonstrated the importance of long noncoding RNAs (lncRNAs) during oncogenic transformation. However, because most lncRNAs are currently uncharacterized, the identification of novel oncogenic lncRNAs is difficult. Given that intergenic lncRNA have substantially less sequence conservation patterns than protein-coding genes across species, evolutionary conserved intergenic lncRNAs are likely to be functional. The current study identified a novel intergenic lncRNA, LINC00461 (ECONEXIN) using a combined approach consisting of searching lncRNAs by evolutionary conservation and validating their expression in a glioma mouse model. ECONEXIN was the most highly conserved intergenic lncRNA containing 83.0% homology with the mouse ortholog (C130071C03Rik) for a region over 2500 bp in length within its exon 3. Expressions of ECONEXIN and C130071C03Rik were significantly upregulated in both human and mouse glioma tissues. Moreover, the expression of C130071C03Rik was upregulated even in precancerous conditions and markedly increased during glioma progression. Functional analysis of ECONEXIN in glioma cell lines, U87 and U251, showed it was dominantly located in the cytoplasm and interacted with miR-411-5p via two binding sites within ECONEXIN. Inhibition of ECONEXIN upregulated miR-411-5p together with the downregulation of its target, Topoisomerase 2 alpha (TOP2A), in glioma cell lines, resulting in decreased cell proliferation. Our data demonstrated that ECONEXIN is a potential oncogene that regulates TOP2A by sponging miR-411-5p in glioma. In addition, our investigative approaches to identify conserved lncRNA and their molecular characterization by validation in mouse tumor models may be useful to functionally annotate novel lncRNAs, especially cancer-associated lncRNAs.


Subject(s)
Antigens, Neoplasm/metabolism , Brain Neoplasms/metabolism , Carcinogenesis/genetics , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Glioma/metabolism , MicroRNAs/metabolism , Oncogenes , RNA, Long Noncoding/metabolism , Animals , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Conserved Sequence , Disease Progression , Down-Regulation , Glioma/genetics , Humans , Mice , Poly-ADP-Ribose Binding Proteins , Precancerous Conditions/genetics , RNA, Long Noncoding/genetics , RNA-Induced Silencing Complex/metabolism , Up-Regulation
2.
Oncogene ; 36(26): 3796, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28218901

ABSTRACT

This corrects the article DOI: 10.1038/onc.2011.466.

3.
Cancer Gene Ther ; 22(10): 487-95, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26450624

ABSTRACT

The epidermal growth factor receptor variant III (EGFRvIII) is exclusively expressed on the cell surface in ~50% of glioblastoma multiforme (GBM). This variant strongly and persistently activates the phosphatidylinositol 3-kinase-Akt signaling pathway in a ligand-independent manner resulting in enhanced tumorigenicity, cellular motility and resistance to chemoradiotherapy. Our group generated a recombinant single-chain variable fragment (scFv) antibody specific to the EGFRvIII, referred to as 3C10-scFv. In the current study, we constructed a lentiviral vector transducing the chimeric antigen receptor (CAR) that consisted of 3C10-scFv, CD3ζ, CD28 and 4-1BB (3C10-CAR). The 3C10-CAR-transduced peripheral blood mononuclear cells (PBMCs) and CD3(+) T cells specifically lysed the glioma cells that express EGFRvIII. Moreover, we demonstrated that CAR CD3(+) T cells migrated to the intracranial xenograft of GBM in the mice treated with 3C10-CAR PBMCs. An important and novel finding of our study was that a thalidomide derivative lenalidomide induced 3C10-CAR PBMC proliferation and enhanced the persistent antitumor effect of the cells in vivo. Lenalidomide also exhibited enhanced immunological synapses between the effector cells and the target cells as determined by CD11a and F-actin polymerization. Collectively, lentiviral-mediated transduction of CAR effectors targeting the EGFRvIII showed specific efficacy, and lenalidomide even intensified CAR cell therapy by enhanced formation of immunological synapses.


Subject(s)
ErbB Receptors/immunology , Glioma/immunology , Immunological Synapses/drug effects , Recombinant Fusion Proteins/immunology , T-Lymphocytes/immunology , Thalidomide/analogs & derivatives , Animals , Cell Line, Tumor , Combined Modality Therapy , ErbB Receptors/metabolism , Glioma/metabolism , Glioma/therapy , Humans , Immunologic Factors/pharmacology , Immunological Synapses/immunology , Immunotherapy, Adoptive/methods , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Lenalidomide , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Single-Chain Antibodies/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Thalidomide/pharmacology , Treatment Outcome , Xenograft Model Antitumor Assays
4.
Cancer Gene Ther ; 19(11): 796-801, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23018623

ABSTRACT

In our previous works, we demonstrated that human neural stem cells (NSCs) transduced with the cytosine deaminase (CD) gene showed remarkable 'bystander killer effect' on glioma and medulloblastoma cells after administration of the prodrug 5-fluorocytosine (5-FC). In addition, herpes simplex virus thymidine kinase (TK) is a widely studied enzyme used for suicide gene strategies, for which the prodrug is ganciclovir (GCV). To apply this strategy to brain metastasis treatment, we established here a human NSC line (F3.CD-TK) expressing the dual suicide genes CD and TK. We examined whether F3.CD-TK cells intensified the antitumor effect on lung cancer brain metastases. In vitro studies showed that F3.CD-TK cells exerted a marked bystander effect on human lung cancer cells after treatment with 5-FC and GCV. In a novel experimental brain metastases model, intravenously administered F3 cells migrated near lung cancer metastatic lesions, which were induced by the injection of lung cancer cells via the intracarotid artery. More importantly, F3.CD-TK cells in the presence of prodrugs 5-FC and GCV decreased tumor size and considerably prolonged animal survival. The results of the present study indicate that the dual suicide gene-engineered, NSC-based treatment strategy might offer a new promising therapeutic modality for brain metastases.


Subject(s)
Brain Neoplasms/secondary , Gene Transfer Techniques , Genes, Transgenic, Suicide , Lung Neoplasms/pathology , Neural Stem Cells/metabolism , Administration, Intravenous , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Brain Neoplasms/therapy , Bystander Effect , Carotid Arteries/metabolism , Cell Movement , Cell Survival , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , Flucytosine/administration & dosage , Flucytosine/pharmacology , Ganciclovir/administration & dosage , Ganciclovir/pharmacology , Genetic Engineering/methods , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Kaplan-Meier Estimate , Mice , Mice, Nude , Neoplasms, Experimental/therapy , Prodrugs/administration & dosage , Prodrugs/pharmacology , Simplexvirus/enzymology , Simplexvirus/genetics , Thymidine Kinase/genetics , Thymidine Kinase/metabolism
5.
Oncogene ; 31(22): 2715-24, 2012 May 31.
Article in English | MEDLINE | ID: mdl-22020337

ABSTRACT

Glioblastomas (GBMs) are the most common and aggressive type of brain tumor. GBMs usually show hyperactivation of the PI3K-Akt pathway, a pro-tumorigenic signaling cascade that contributes to pathogenesis. Girdin, an actin-binding protein identified as a novel substrate of Akt, regulates the sprouting of axons and the migration of neural progenitor cells during early postnatal-stage neurogenesis in the hippocampus. Here, we show that Girdin is highly expressed in human glioblastoma (GBM). Stable Girdin knockdown in isolated GBM stem cells resulted in decreased expression of stem cell markers, including CD133, induced multilineage neural differentiation, and inhibited in vitro cell motility, ex vivo invasion, sphere-forming capacity and in vivo tumor formation. Furthermore, exogenous expression of the Akt-binding domain of Girdin, which competitively inhibits its Akt-mediated phosphorylation, diminished the expression of stem cell markers, SOX2 and nestin, and migration on the brain slice and induced the expression of neural differentiation markers glial fibrillary acidic protein/ßIII Tubulin. Our results reveal that Girdin is required for GBM-initiating stem cells to sustain the stemness and invasive properties.


Subject(s)
Brain Neoplasms/pathology , Cell Differentiation , Cell Movement , Glioblastoma/pathology , Microfilament Proteins/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Vesicular Transport Proteins/metabolism , AC133 Antigen , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Biomarkers/metabolism , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Immunoenzyme Techniques , Immunoprecipitation , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Microfilament Proteins/antagonists & inhibitors , Microfilament Proteins/genetics , Neoplasm Grading , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Peptides/genetics , Peptides/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Cells, Cultured , Vesicular Transport Proteins/antagonists & inhibitors , Vesicular Transport Proteins/genetics
6.
Gene Ther ; 17(11): 1363-71, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20520650

ABSTRACT

Glioblastoma multiforme (GBM) is one of the most formidable brain tumors with a mean survival period of approximately 12 months. To date, a combination of radiotherapy and chemotherapy with an oral alkylating agent, temozolomide (TMZ), has been used as first-line therapy for glioma. However, the efficacy of chemotherapy for treating GBM is very limited; this is partly because of the high activity levels of the DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT) in tumor cells, which creates a resistant phenotype by blunting the therapeutic effect of alkylating agents. Thus, MGMT may be an important determinant of treatment failure and should be considered as a suitable target for intervention, in an effort to improve the therapeutic efficacy of TMZ. In this study, we showed that small-interfering RNA (siRNA)-based downregulation of MGMT could enhance the chemosensitivity of malignant gliomas against TMZ. Notably, TMZ-resistant glioma-initiating cells with increased DNA repair and drug efflux capabilities could be efficiently transduced with MGMT-siRNA by using a novel liposome, LipoTrust. Accordingly, such transduced glioma-initiating cells could be sensitized to TMZ in both in vitro and in vivo tumor models. Taken together, this study provides an experimental basis for the clinical use of such therapeutic combinations.


Subject(s)
Antineoplastic Agents, Alkylating/toxicity , Brain Neoplasms/therapy , Dacarbazine/analogs & derivatives , Glioblastoma/therapy , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , RNA, Small Interfering/administration & dosage , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Brain Neoplasms/drug therapy , Cell Line, Tumor , Combined Modality Therapy , DNA Repair , Dacarbazine/administration & dosage , Dacarbazine/toxicity , Drug Delivery Systems , Glioblastoma/drug therapy , Liposomes , Mice , Mice, Inbred NOD , Mice, SCID , O(6)-Methylguanine-DNA Methyltransferase/genetics , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Temozolomide
7.
Cancer Gene Ther ; 17(5): 299-306, 2010 May.
Article in English | MEDLINE | ID: mdl-19893595

ABSTRACT

Previously, we have shown that the genetically modified human neural stem cells (NSCs) show remarkable migratory and tumor-tropic capability to track down brain tumor cells and deliver therapeutic agents with significant therapeutic benefit. Human NSCs that were retrovirally transduced with cytosine deaminase (CD) gene showed remarkable 'bystander killer effect' on the glioma cells after application of the prodrug, 5-fluorocytosine (5-FC). Interferon-beta (IFN-beta) is known for its antiproliferative effects in a variety of cancers. In our pilot clinical trial in glioma, the IFN-beta gene has shown potent antitumor activity in patients with malignant glioma. In the present study, we sought to examine whether human NSCs genetically modified to express both CD and IFN-beta genes intensified antitumor effect on experimental glioma. In vitro studies showed that CD/IFN-beta-expressing NSCs exerted a remarkable bystander effect on human glioma cells after the application of 5-FC, as compared with parental NSCs and CD-expressing NSCs. In animal models with human glioma orthotopic xenograft, intravenously infused CD/IFN-beta-expressing NSCs produced striking antitumor effect after administration of the prodrug 5-FC. Furthermore, the same gene therapy regimen prolonged survival periods significantly in the experimental animals. The results of the present study indicate that the multimodal NSC-based treatment strategy might have therapeutic potential against gliomas.


Subject(s)
Cytosine Deaminase/physiology , Genetic Therapy/methods , Glioma/drug therapy , Glioma/therapy , Interferon-beta/physiology , Animals , Bystander Effect , Cell Line, Tumor , Cytosine Deaminase/genetics , Disease Models, Animal , Female , Flucytosine/therapeutic use , Humans , Interferon-beta/genetics , Mice , Mice, Nude
8.
Gene Ther ; 14(15): 1132-42, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17508009

ABSTRACT

Medulloblastomas are highly malignant neuroectodermal cerebellar tumors of children. One of the reasons for the difficulty for the treatment of medulloblastomas is their inherent tendency to metastasize through the cerebrospinal fluid (CSF) pathway leading to leptomeningeal dissemination. Recently, genetically modified neural stem cells (NSCs) were shown to have the capability of selectively migrating into glioma mass and delivering therapeutic agents with significant therapeutic benefits. In the present study, we applied the NSC strategy to target medulloblastomas, particularly their leptomeningeal dissemination. We used NSCs that were retrovirally transduced with the cytosine deaminase gene (CD-NSCs). In vitro studies demonstrated that CD-NSCs had sufficient migratory activity toward medulloblastoma cells and exerted a remarkable bystander effect on these cells following the application of 5-fluorocytosine (5-FC). It is noteworthy that neutralization of the hepatocyte growth factor blocked their migration In animal studies using our leptomeningeal dissemination model, CD-NSCs implanted directly into CSF space were shown to distribute diffusely within the disseminated tumor cells and could provide remarkable antitumor effect after intraperitoneal administration of 5-FC. Furthermore, CD-NSC treatment followed by 5-FC administration prolonged survival periods significantly in experimental animals. Our data suggest that the CD-NSC strategy can also be applied to target leptomeningeal dissemination of medulloblastomas.


Subject(s)
Cytosine Deaminase/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Medulloblastoma/therapy , Meningeal Neoplasms/therapy , Stem Cell Transplantation/methods , Animals , Antimetabolites/therapeutic use , Bystander Effect , Cell Line , Cell Line, Tumor , Cell Movement , Cisterna Magna , Female , Flucytosine/therapeutic use , Glioma/therapy , Humans , In Situ Nick-End Labeling , Injections , Meninges/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Neurons/enzymology , Neurons/transplantation
9.
Cancer Gene Ther ; 13(4): 385-92, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16244591

ABSTRACT

Glioblastomas are the most common primary brain tumors in adults. These tumors exhibit a high degree of vascularization, and malignant progression from astrocytoma to glioblastoma is often accompanied by increased angiogenesis and the upregulation of vascular endothelial growth factor and its receptors. In this study, we investigated the in vivo antiangiogenic and antitumor effects of brain-specific angiogenesis inhibitor 1 (BAI1) using human glioblastoma cell lines. Glioblastoma cells were transduced with an adenoviral vector encoding BAI1 (AdBAI1), and Northern and Western blot analyses, respectively, demonstrated BAI1 mRNA and protein expression in the transduced tumor cells. Using an in vivo neovascularization assay, we found that angiogenesis surrounding AdBAI1-transduced glioblastoma cells transplanted into transparent skinfold chambers of SCID mice was significantly impaired compared to control treated cells. Additionally, in vivo inoculation with AdBAI1 of established subcutaneous or intracerebral transplanted tumors significantly impaired tumor growth and promoted increased mouse survival. Morphologically, the tumors exhibited signs of impaired angiogenesis, such as extensive necrosis and reduced intratumoral vascular density. Taken together, these data strongly indicate that BAI1 may be an excellent gene therapy candidate for the treatment of brain tumors, especially human glioblastomas.


Subject(s)
Angiogenic Proteins/biosynthesis , Brain Neoplasms/blood supply , Glioblastoma/blood supply , Neovascularization, Pathologic/therapy , Adenoviridae/genetics , Angiogenic Proteins/genetics , Animals , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Line, Tumor , Genetic Therapy , Genetic Vectors , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Mice , Mice, SCID , Neoplasm Transplantation , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled , Transduction, Genetic
10.
Apoptosis ; 7(1): 77-86, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11773708

ABSTRACT

Apoptosis is originally defined by unique morphological changes of dying cells, and the biochemical hallmark associated with apoptosis is internucleosomal DNA fragmentation. However, few report has shown the precise time course of the apoptotic events. The present study was designed to try to clarify apoptotic processes using a video-enhanced contrast-differential interference contrast (VEC-DIC) microscopy. The morphological changes of murine fibroblast Ltk-cells treated with TNF-alpha were divided into four stages: (i) pre-apoptotic, (ii) cytoplasmic shrinkage, (iii) membrane blebbing, and (iv) ballooning. Almost of the cells underwent cytoplasmic shrinkage and membrane blebbing within 6 hours after TNF-alpha exposure, and at about 9 hours, they were in the ballooning stage. Based on these data, we investigated the relationship between morphological changes and other biochemical features. The earliest event was exposure of phosphatidyl-serine at the cytoplasmic membrane, which was already observed in the pre-apoptotic stage. Loss of mitochondrial membrane potential was observed in the cytoplasmic shrinkage stage. Caspase-8/-3 activities already started increasing in the pre-apoptotic stage, and reached their peak at 6 hours after TNF-alpha exposure. DNA fragmentation occurred in the late phase of the membrane blebbing.


Subject(s)
Apoptosis/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Annexin A5/metabolism , Caspases/metabolism , Cell Line , Cell Membrane/drug effects , Cell Size/drug effects , Endodeoxyribonucleases/metabolism , Enzyme Activation/drug effects , Fibroblasts , Mice , Microscopy, Fluorescence , Microscopy, Interference , Microscopy, Video , Mitochondria/drug effects , Time Factors
11.
Biotechnol Bioeng ; 74(5): 416-23, 2001 Sep 05.
Article in English | MEDLINE | ID: mdl-11427943

ABSTRACT

Cytokines and growth factors are indispensable for the propagation and maintenance of factor-dependent mammalian cells. However, cytokines are often so expensive that the use of factor-dependent cells for industrial applications such as protein production is often not practical. Based on our previous design of a binary hen egg lysozyme (HEL)-specific receptor composed of portions of the anti-HEL antibody and the erythropoietin receptor, a new pair of chimeric receptors having the intracellular domain of gp130 were made and transfected to an interleukin-6 (IL-6)-dependent hybridoma, 7TD1. The clone expressing the two new receptors showed clear HEL dose-dependent cell growth and monoclonal antibody production in both serum-based and serum-free media without IL-6. These results establish the feasibility of applying receptor design to tailor cells for the inexpensive induction of cell growth for the purpose of producing therapeutic products.


Subject(s)
DNA-Binding Proteins/metabolism , Hybridomas/metabolism , Interleukin-6/metabolism , Muramidase/metabolism , Recombinant Fusion Proteins/metabolism , Trans-Activators/metabolism , Animals , Antibodies, Monoclonal/metabolism , Cell Division/physiology , Cell Line/cytology , Cell Line/metabolism , Culture Media, Serum-Free/metabolism , Efficiency , Hybridomas/cytology , Mice , Phosphorylation , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Recombinant Fusion Proteins/genetics , STAT3 Transcription Factor , Transfection
12.
Exp Neurol ; 169(2): 231-8, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11358438

ABSTRACT

Previous studies have demonstrated that either the neurotrophin glial-derived neurotrophic factor (GDNF) or the antiapoptotic peptide Bcl-2 delivered into striatum by a viral vector protects dopaminergic neurons of the substantia nigra in vivo from degeneration induced by the administration of the neurotoxin 6-hydroxydopamine (6-OHDA). In this study we used recombinant, replication-incompetent, genomic herpes simplex virus-based vectors to deliver the genes coding for Bcl-2 and GDNF into rat substantia nigra (SN) 1 week prior to 6-OHDA injection into the striatum. Vector-mediated expression of either Bcl-2 or GDNF alone each resulted in a doubling in cell survival as measured by retrograde labeling with fluorogold (FG) and a 50% increase in tyrosine hydroxylase-immunoreactive (TH-IR) neurons in the lesioned SN compared to the unlesioned side. Gene transfer of Bcl-2 and GDNF were equivalent in this effect. Coadministration of the Bcl-2-expressing vector with the GDNF-expressing vector improved the survival of lesioned SN neurons as measured by FG labeling by 33% and by the expression of TH-IR by 15%. These results suggest that the two factors delivered together act in an additive fashion to improve DA cell survival in the face of 6-OHDA toxicity.


Subject(s)
Corpus Striatum/physiology , Dopamine/physiology , Gene Transfer Techniques , Genes, bcl-2 , Nerve Growth Factors , Nerve Tissue Proteins/physiology , Neurons/physiology , Oxidopamine/toxicity , Proto-Oncogene Proteins c-bcl-2/metabolism , Substantia Nigra/physiology , Tyrosine 3-Monooxygenase/analysis , Animals , Corpus Striatum/drug effects , Corpus Striatum/pathology , Dextroamphetamine/pharmacology , Female , Functional Laterality , Genes, Reporter , Genetic Therapy/methods , Genetic Vectors , Glial Cell Line-Derived Neurotrophic Factor , Humans , Motor Activity/drug effects , Nerve Degeneration/genetics , Nerve Degeneration/prevention & control , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Rats, Sprague-Dawley , Rotation , Simplexvirus , beta-Galactosidase/analysis , beta-Galactosidase/genetics
13.
Biochim Biophys Acta ; 1526(1): 37-43, 2001 Apr 03.
Article in English | MEDLINE | ID: mdl-11287120

ABSTRACT

Hybrid glycoprotein and neoproteoglycan probes were prepared by coupling various glycoproteins or polysaccharides to peroxidase or biotinyl bovine serum albumin, respectively. Lectins recognizable by the neoglycoconjugate probes were extracted from 16 cultivable mushrooms. Dot-blot assay revealed five extracts to be reactive with only hybrid glycoprotein probes, but others also reacted with neoproteoglycan probes. According to the reactivity pattern with probe screening, the one lectin from Oudemansiella platyphylla extract (OPL) bound best with asialotransferrin-- and asialoagalactotransferrin--peroxidase probes and was isolated using an asialotransferrin column, but it did not bind with other hybrid glycoprotein or neoproteoglycan probes. OPL, consisting of two polypeptides with high homology in the N-terminal amino acid sequences, exhibited weak hemagglutinating activity. Purified OPL specifically bound the beta-GlcNAc probe among various biotinylated polymeric sugar probes, while it exhibited essentially the same binding specificity toward neoglycoconjugate probes as that of the crude extract, showing a preference for the asialobiantennary complex type of N-linked glycans. These results indicate that the neoglycoconjugate probes are valuable in lectin screening.


Subject(s)
Acetylglucosamine/chemistry , Agaricales/chemistry , Lectins/chemistry , Acetylglucosamine/genetics , Carbohydrate Sequence , Chromatography, Affinity , Glycoproteins/genetics , Lectins/classification , Lectins/genetics , Molecular Sequence Data , Plant Extracts/chemistry , Proteoglycans/genetics
14.
J Biol Chem ; 276(25): 22032-40, 2001 Jun 22.
Article in English | MEDLINE | ID: mdl-11283017

ABSTRACT

A new member of the UDP-N-acetylglucosamine:beta-galactose beta1,3-N-acetylglucosaminyltransferase (beta3Gn-T) family having the beta3Gn-T motifs was cloned from rat and human cDNA libraries and named beta3Gn-T5 based on its position in a phylogenetic tree. We concluded that beta3Gn-T5 is the most feasible candidate for lactotriaosylceramide (Lc(3)Cer) synthase, an important enzyme which plays a key role in the synthesis of lacto- or neolacto-series carbohydrate chains on glycolipids. beta3Gn-T5 exhibited strong activity to transfer GlcNAc to glycolipid substrates, such as lactosylceramide (LacCer) and neolactotetraosylceramide (nLc(4)Cer; paragloboside), resulting in the synthesis of Lc(3)Cer and neolactopentaosylceramide (nLc(5)Cer), respectively. A marked decrease in LacCer and increase in nLc(4)Cer was detected in Namalwa cells stably expressing beta3Gn-T5. This indicated that beta3Gn-T5 exerted activity to synthesize Lc(3)Cer and decrease LacCer, followed by conversion to nLc(4)Cer via endogenous galactosylation. The following four findings further supported that beta3Gn-T5 is Lc(3)Cer synthase. 1) The beta3Gn-T5 transcript levels in various cells were consistent with the activity levels of Lc(3)Cer synthase in those cells. 2) The beta3Gn-T5 transcript was presented in various tissues and cultured cells. 3) The beta3Gn-T5 expression was up-regulated by stimulation with retinoic acid and down-regulated with 12-O-tetradecanoylphorbol-13-acetate in HL-60 cells. 4) The changes in beta3Gn-T5 transcript levels during the rat brain development were determined. Points 2, 3, and 4 were consistent with the Lc(3)Cer synthase activity reported previously.


Subject(s)
Epitopes/chemistry , Glycolipids/chemistry , Lewis X Antigen/chemistry , N-Acetylglucosaminyltransferases/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary , Humans , Molecular Sequence Data , N-Acetylglucosaminyltransferases/chemistry , N-Acetylglucosaminyltransferases/metabolism , Phylogeny , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Tumor Cells, Cultured
15.
Exp Neurol ; 168(2): 225-30, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11259110

ABSTRACT

Proximal axotomy in adult animals results in delayed death of motor neurons. Features characteristic of both necrosis and apoptosis have been described in motor neurons of the spinal cord following proximal avulsion of the ventral roots. We have previously demonstrated that a genomic herpes simplex virus (HSV)-based vector expressing the anti-apoptotic peptide Bcl-2 protects dopaminergic neurons of the substantia nigra from neurotoxin-induced apoptotic cell death and preserves the neurotransmitter phenotype of those cells. In this study we examined whether the same vector could protect adult rat lumbar motor neurons from cell death following proximal ventral root avulsion. Injection of the Bcl-2-expressing vector 1 week prior to root avulsion increased the survival of lesioned motor neurons, determined by retrograde Fluorogold labeling, by 50%. The Bcl-2-expressing vector did not preserve choline acetyltransferase neurotransmitter phenotype of the lesioned cells. These results shed light on the mechanism of cell death following axonal injury, and have implications for developing an effective treatment for the clinical problem of proximal root avulsion.


Subject(s)
Anterior Horn Cells/metabolism , Genetic Vectors/therapeutic use , Motor Neurons/metabolism , Nerve Degeneration/prevention & control , Proto-Oncogene Proteins c-bcl-2/metabolism , Radiculopathy/metabolism , Animals , Anterior Horn Cells/drug effects , Cell Death/drug effects , Cell Death/physiology , Choline O-Acetyltransferase/metabolism , Genetic Vectors/metabolism , Humans , Male , Motor Neurons/drug effects , Motor Neurons/pathology , Radiculopathy/complications , Radiculopathy/pathology , Rats , Rats, Sprague-Dawley , Simplexvirus
16.
J Biol Chem ; 276(5): 3498-507, 2001 Feb 02.
Article in English | MEDLINE | ID: mdl-11042166

ABSTRACT

We have isolated three types of cDNAs encoding novel beta1,3-N-acetylglucosaminyltransferases (designated beta3Gn-T2, -T3, and -T4) from human gastric mucosa and the neuroblastoma cell line SK-N-MC. These enzymes are predicted to be type 2 transmembrane proteins of 397, 372, and 378 amino acids, respectively. They share motifs conserved among members of the beta1,3-galactosyltransferase family and a beta1,3-N-acetylglucosaminyltransferase (designated beta3Gn-T1), but show no structural similarity to another type of beta1,3-N-acetylglucosaminyltransferase (iGnT). Each of the enzymes expressed by insect cells as a secreted protein fused to the FLAG peptide showed beta1,3-N-acetylglucosaminyltransferase activity for type 2 oligosaccharides but not beta1,3-galactosyltransferase activity. These enzymes exhibited different substrate specificity. Transfection of Namalwa KJM-1 cells with beta3Gn-T2, -T3, or -T4 cDNA led to an increase in poly-N-acetyllactosamines recognized by an anti-i-antigen antibody or specific lectins. The expression profiles of these beta3Gn-Ts were different among 35 human tissues. beta3Gn-T2 was ubiquitously expressed, whereas expression of beta3Gn-T3 and -T4 was relatively restricted. beta3Gn-T3 was expressed in colon, jejunum, stomach, esophagus, placenta, and trachea. beta3Gn-T4 was mainly expressed in brain. These results have revealed that several beta1,3-N-acetylglucosaminyltransferases form a family with structural similarity to the beta1,3-galactosyltransferase family. Considering the differences in substrate specificity and distribution, each beta1,3-N-acetylglucosaminyltransferase may play different roles.


Subject(s)
Galactosyltransferases/chemistry , N-Acetylglucosaminyltransferases/isolation & purification , Polysaccharides/metabolism , Amino Acid Sequence , Animals , Cell Line, Transformed , Cells, Cultured , Humans , Insecta , Molecular Sequence Data , N-Acetylglucosaminyltransferases/classification , N-Acetylglucosaminyltransferases/genetics , Phylogeny , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Tissue Distribution
17.
Cancer Immunol Immunother ; 50(9): 463-8, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11761440

ABSTRACT

Dendritic cells (DCs) are professional antigen-presenting cells (APCs) that locate in peripheral organs. It has been thought that a systemic immune response does not play a role in regression of central nervous system (CNS) tumors, because the CNS is an immunologically privileged site. However, recent advances in immunology have led to the possibility of immunotherapy using peripheral DCs against CNS tumors. Here, we investigated whether DCs pulsed with tumor extract could induce an antitumor effect against malignant glioma. Furthermore, we also investigated whether the antitumor effect become higher by pulsation with tumor extract-liposome complex, compared to pulsation with tumor extract alone. As a liposome, we used cationic small unilamellar vesicles composed of N-(alpha-trimethylammonioacetyl)-didodecyl-D-glutamate chloride (TMAG), dilauroylphosphatidylcholine (DLPC), and dioleoylphosphatidylethanolamine (DOPE) in a molar ratio of 1:2:2. After intracerebral inoculation of mouse malignant glioma GL261 cells into syngeneic C57BL/6 mice, DCs pulsed with extract from the glioma cells by sonication were administered intraperitoneally thrice weekly on days 7, 14 and 21. Tumor growth inhibition was evaluated by measuring the tumor size 1 month after the tumor inoculation. The group treated with DCs pulsed by tumor extract was inhibited in tumor progression compared with the control non-pulsed DCs group, and the group treated with DCs pulsed by tumor extract and liposomes showed substantial tumor volume reductions in all the mice. Among the mice, there were several with no visible masses in their brains. Immunohistochemical study showed that the CD8-positive cytotoxic T cells (CTLs) were strongly recognized among the almost disappearing tumor cells of pulsed DCs groups. The CTLs showed a specific antitumor activity for GL261 mouse glioma cells. These findings indicated that DCs pulsed with tumor extract and liposomes might play an important role in the activation of an immune response in malignant glioma.


Subject(s)
Brain Neoplasms/immunology , Dendritic Cells/immunology , Glioma/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Brain Neoplasms/prevention & control , Cytotoxicity, Immunologic , Dendritic Cells/drug effects , Female , Glioma/prevention & control , Immunoenzyme Techniques , Immunotherapy , Liposomes , Mice , Mice, Inbred C57BL , Vaccination
18.
Gan To Kagaku Ryoho ; 27(9): 1403-9, 2000 Aug.
Article in Japanese | MEDLINE | ID: mdl-10969596

ABSTRACT

The expression of topoisomerase II alpha (Topo II alpha) was investigated in six human glioma cell lines as a function of doubling time, cell cycle distribution, and sensitivity to an antineoplastic agent, etoposide (ETP). The Topo II alpha level was determined by immunohistochemical and flow cytometric studies using monoclonal antibody, 8D2, specific for Topo II alpha. In all cell lines, the Topo II alpha level correlated directly with sensitivity to ETP, but not with doubling time or percentage of cells in any phase of the cell cycle. These findings suggest that the cytocidal activity of ETP may be mediated by qualitative and quantitative changes in Topo II alpha in human glioma cells. We speculate that better knowledge of Topo II alpha expression in surgical specimens may lead to more individualized use of ETP in patients with malignant glioma.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Brain Neoplasms/enzymology , DNA Topoisomerases, Type II/metabolism , Etoposide/pharmacology , Glioma/enzymology , Isoenzymes/metabolism , Antigens, Neoplasm , Brain Neoplasms/pathology , Cell Cycle , Cell Division , DNA-Binding Proteins , Drug Screening Assays, Antitumor , Flow Cytometry , Glioma/pathology , Humans , Immunohistochemistry , Tumor Cells, Cultured/drug effects
19.
J Neurooncol ; 47(2): 117-24, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10982152

ABSTRACT

We previously demonstrated that intratumoral administration of liposomes containing the murine interferon beta (IFN-beta) gene [lip(pSV2muIFN-beta)] resulted in stronger growth-inhibitory effect on GL261 (H-2b) mouse glioma inoculated in brains of syngeneic C57BL/6 mice than conventional exogenous IFN-beta administration, and histologic evaluation revealed the massive infiltration of T lymphocytes (CD8 > CD4) within the residual tumor. The present study was aimed at determining whether such tumor-infiltrating lymphocytes (TIL) have any tumor-specific cytotoxic effects. Intratumoral administration of lip(pSV2muIFN-beta) resulted in prolonged survival time and a 50% tumor-free incidence in the mice treated. The surviving animals were subsequently re-challenged with either subcutaneous or intracranial injection of GL261 cells, and no tumors were found to develop over a 50-day period. In vivo depletion of CD8, but not CD4 cells decreased the efficacy of lip(pSV2muIFN-beta). Specific cytotoxic T lymphocytes (CTL) against GL261 cells were generated from both TIL and spleen cells of the mice treated. The results of flow cytometric analysis and antibody blocking test revealed that the bulk CTL lines thus prepared were T cell receptor (TCR) alphabeta, CD8 T lymphocytes with H-2b restriction. These findings suggest that, in addition to direct growth-inhibitory effects by the IFN-beta gene on the tumor cells, activation of systemic cellular immunity may participate in antitumor effects in vivo, despite the fact that central nervous system is generally regarded as an immunologically privileged site.


Subject(s)
Brain Neoplasms/therapy , Genetic Therapy , Glioma/therapy , Interferon-beta/genetics , Animals , Brain Neoplasms/immunology , CD8-Positive T-Lymphocytes , Disease Models, Animal , Female , Flow Cytometry , Glioma/immunology , Humans , Interferon-beta/administration & dosage , Interferon-beta/therapeutic use , Liposomes , Mice , Plasmids , Survival Analysis , Tumor Cells, Cultured
20.
Jpn J Cancer Res ; 91(4): 363-7, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10804282

ABSTRACT

Adenoviral (Ad) vectors are commonly used in gene therapy trials because of their efficiency in gene transfer. However, their use is limited by immune responses that reduce transgene expression and decrease the efficacy of repeated vector administration. In this study, we demonstrated that conjugation of Ad vector with our novel cationic liposomes could reduce viral antigenicity in vivo. Mice subcutaneously injected with liposome-conjugated Ad vector showed a 6.5-fold reduction of anti-Ad antibodies with neutralizing activity, compared to those with unconjugated Ad vector. Interestingly, we also found that the conjugated vector is less susceptible to inactivation by neutralizing antibodies in vitro and in vivo. Our results suggest that liposome conjugation reduces viral antigenicity, shields vectors from neutralizing antibody, and may allow repeated Ad vector administration.


Subject(s)
Adenoviridae/immunology , Genetic Vectors , Liposomes/administration & dosage , Adenoviridae/genetics , Animals , Antibodies, Viral/blood , Genetic Therapy , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...