Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
Int J Mol Sci ; 22(22)2021 Nov 21.
Article in English | MEDLINE | ID: mdl-34830422

ABSTRACT

Immortalized cell lines have been used in a wide range of applications in research on immune disorders and cellular metabolic regulation due to the stability and uniformity of their cellular characteristics. At present, the investigation into molecular functions and signaling pathways within bovine cells remains largely limited by the lack of immortalized model cells. Current methods for immortalizing bovine cells are mainly restricted to the ectopic expression of human telomerase reverse transcriptase (hTERT) through transient transfection or virus-mediated delivery, which have defects in efficiency and reliability. In this study, we identified bovine TERT (bTERT) as a novel potent biofactor for immortalizing bovine cells with great advantages over hTERT, and established an efficient and easily manipulated strategy for the immortalization of bovine primary cells. Through the homology-mediated end-joining-based insertion of bTERT at the ROSA26 locus, we successfully generated immortalized bovine fetal fibroblast cell lines with stable characteristics. The observed limitation of this strategy in immortalizing bovine bone marrow-derived macrophages was attributed to the post-translational modification of bTERT, causing inhibited nuclear localization and depressed activity of bTERT in this terminally differentiated cell. In summary, we constructed an innovative method to achieve the high-quality immortalization of bovine primary cells, thereby expanding the prospects for the future application of immortalized bovine model cell lines.


Subject(s)
Cell Line, Transformed/cytology , DNA End-Joining Repair/genetics , Telomerase/genetics , Animals , Cattle , Cell Line, Transformed/enzymology , Gene Expression Regulation, Enzymologic , Humans , Recombinational DNA Repair/genetics
2.
Clin Exp Immunol ; 161(1): 71-80, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20491788

ABSTRACT

Main features of rheumatoid arthritis (RA), hyperplasia of fibroblast-like synoviocytes (FLS) and joint destruction are caused by inflammatory cytokines produced in chronic autoimmune inflammation. Cell-intrinsic acquisition of tumour-like phenotypes of RA-FLS could also be responsible for the aggressive proliferation and invasion, which are supported by the fact that in some cases RA-FLS has mutations of a tumour suppressor gene TP53. However, the underlying molecular mechanism for TP53 mutations in RA-FLS has not yet been clarified. Recently it has been reported that the non-lymphoid cells in the inflammatory tissues express ectopically the activation-induced cytidine deaminase (AID) gene that induces somatic hypermutations, not only at the immunoglobulin (Ig) gene variable regions in germinal centre B lymphocytes but also at coding regions in TP53. Real-time polymerase chain reaction (PCR) analyses revealed more than half (five of nine) of the RA-FLS lines we established showed the markedly increased expression of AID. AID transcription in RA-FLS was augmented by tumour necrosis factor (TNF)-alpha and even by physiological concentration of beta-oestradiol that could not induce AID transcription in osteoarthritis-FLS. Furthermore, AID-positive RA-FLS presented a higher frequency of somatic mutations in TP53. Cytological and immunohistochemical analyses demonstrated clearly the ectopic expression of AID in the FLS at the RA synovium. These data suggested strongly a novel consequence of RA; the ectopic expression of AID in RA-FLS causes the somatic mutations and dysfunction of TP53, leading to acquisition of tumour-like properties by RA-FLS.


Subject(s)
Arthritis, Rheumatoid/pathology , Cytidine Deaminase/physiology , Genes, p53 , Mutation , Synovial Membrane/enzymology , Tumor Suppressor Protein p53/physiology , Aged , Aged, 80 and over , Arthritis, Rheumatoid/enzymology , Arthritis, Rheumatoid/genetics , Cell Line, Transformed/enzymology , Cell Line, Transformed/metabolism , Cell Line, Transformed/pathology , Cell Transformation, Neoplastic , Computer Systems , Cytidine Deaminase/biosynthesis , Enzyme Induction , Estradiol/pharmacology , Female , Gene Expression Regulation/drug effects , Humans , Hyperplasia , Male , Middle Aged , Osteoarthritis/genetics , Osteoarthritis/pathology , Reverse Transcriptase Polymerase Chain Reaction , Synovial Membrane/metabolism , Synovial Membrane/pathology , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/pharmacology
3.
Theriogenology ; 71(9): 1417-24, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19303628

ABSTRACT

Although mammary epithelial cell lines can provide a rapid and reliable indicator of gene expression efficiency of transgenic animals, their short lifespan greatly limits this application. To provide stable and long lifespan cells, goat mammary epithelial cells (GMECs) were transduced with pLNCX2-hTERT by retrovirus-mediated gene transfer. Transduced GMECs were evaluated by reverse transcriptase polymerase chain reaction (RT-PCR), proliferation assays, karyotype analysis, telomerase activity assay, western blotting, soft agar assay, and injection into nude mice. Non-transduced GMECs were used as a control. The hTERT-GMECs had higher telomerase activity and extended proliferative lifespan compared to non-transfected GMECs; even after Passage 50, hTERT-GMECs had a near diploid complement of chromosomes. Furthermore, they did not gain the anchorage-independent growth property and were not associated with a malignant phenotype in vitro or in vivo.


Subject(s)
Cell Line, Transformed/enzymology , Goats , Mammary Glands, Animal/cytology , Telomerase/genetics , Animals , Cell Division , Epithelial Cells , Female , Gene Expression , Genetic Vectors , Humans , Karyotyping , Mice , Mice, Nude , Reverse Transcriptase Polymerase Chain Reaction , Telomerase/metabolism , Transfection
4.
Oncogene ; 28(2): 279-88, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-18836480

ABSTRACT

The recepteur d'origine nantais (RON) is a receptor tyrosine kinase (RTK) in the scatter factor family, which includes the c-Met receptor. RON exhibits increased expression in a significant number of human breast cancer tissues as well as in many established breast cancer cell lines. Recent studies have indicated that in addition to ligand-dependent signaling events, RON also promotes signals in the absence of its only known ligand, MSP, when expressed in epithelial cells. In this study, we found that when expressed in MCF-10A breast epithelial cells, RON exhibits both MSP-dependent and MSP-independent signaling, which lead to distinct biological outcomes. In the absence of MSP, RON signaling promotes cell survival, increased cell spreading and enhanced migration in response to other growth factors. However, both RON-mediated proliferation and migration require the addition of MSP in MCF-10A cells. Both MSP-dependent and MSP-independent signaling by RON are mediated in part by Src family kinases. These data suggest that RON has two alternative modes of signaling that can contribute to oncogenic behavior in normal breast epithelial cells.


Subject(s)
Breast/cytology , Cell Transformation, Neoplastic , Epithelial Cells/enzymology , Hepatocyte Growth Factor/physiology , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Signal Transduction/physiology , Animals , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Line/cytology , Cell Line/drug effects , Cell Line/enzymology , Cell Line, Transformed/cytology , Cell Line, Transformed/drug effects , Cell Line, Transformed/enzymology , Cell Movement/drug effects , Cell Movement/physiology , Cell Shape/drug effects , Cell Shape/physiology , Cell Survival/drug effects , Cell Survival/physiology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Female , Humans , Mice , NIH 3T3 Cells/cytology , NIH 3T3 Cells/drug effects , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/physiology , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction/drug effects , src-Family Kinases/physiology
5.
Biotechnol Lett ; 29(11): 1623-30, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17636386

ABSTRACT

A secretory form of human alpha3-fucosyltransferase IX (sFUT9) was overexpressed in Spodoptera frugiperda (Sf9) insect cells using the stable expression vector pIB/V5-His-TOPO and the signal sequence of human interleukin 2 for efficient secretion. sFUT9 was active and its three potential N-glycosylation sites were occupied. sFUT9 efficiently fucosylated the type II acceptors Galbeta4GlcNAC-R and Fucalpha2Galbeta4GlcNAc-R (R = (CH2)3NHCO(CH2)5-NH-biotin) but not the corresponding sialylated acceptor, and only very poorly the type I (Galbeta3GlcNAc-R) related acceptors. sFUT9 showed a clear preference for glycoproteins containing type II acceptors, with values of 121, 113 and 110 microU/million cell for asialofetuin, erythropoietin and asialoerythropoietin, respectively, values approximately 11-fold higher than those obtained for the small acceptors.


Subject(s)
Cell Line, Transformed/enzymology , Fucosyltransferases/biosynthesis , Recombinant Proteins/biosynthesis , Spodoptera , Animals , Cells, Cultured , Genetic Vectors , Glycosylation , Humans , Lewis Blood Group Antigens/physiology , Spodoptera/enzymology , Spodoptera/genetics , Spodoptera/metabolism
6.
Leukemia ; 21(3): 421-6, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17252018

ABSTRACT

Imatinib mesylate is a selective inhibitor of the oncogenic tyrosine kinase, Bcr-Abl, and is widely used as a first-line treatment for chronic myeloid leukaemia (CML). Prolonged monotherapy is frequently associated with patients becoming refractory to imatinib. Therefore, there is considerable interest in small molecule inhibitors which may be used either as replacements or as adjuncts to existing imatinib therapy. For this purpose, it is most likely that drugs which do not share imatinib's mechanism of action will be most valuable. We compared two such compounds with different modes of action, adaphostin and 17-allylamino-17-demethoxygeldanamycin (17-AAG), for their cytotoxic effect and ability to induce the downregulation of cellular proteins in a murine haemopoietic cell line transformed with human p210(Bcr-Abl), and two subclones resistant to imatinib owing to an Abl-kinase domain mutation (E255K) or amplification of the BCR-ABL gene, respectively. We found that, whereas 17-AAG selectively killed Bcr-Abl-positive cells and inhibited proteins dependent on heat-shock protein 90 for their stability (p210(Bcr-Abl) and Akt), adaphostin induced the downregulation of multiple cell-signalling proteins (p210(Bcr-Abl), Akt, Bcr, Abl and STAT5a) and was cytotoxic to both Bcr-Abl-positive and -negative cells. We suggest that both compounds may prove useful in the treatment of CML but caution that undesirable side-effects may result from the inhibition of multiple cell signalling proteins.


Subject(s)
Adamantane/analogs & derivatives , Antineoplastic Agents/pharmacology , Benzoquinones/pharmacology , Hydroquinones/pharmacology , Lactams, Macrocyclic/pharmacology , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Adamantane/adverse effects , Adamantane/pharmacology , Animals , Benzamides , Benzoquinones/adverse effects , Cell Line, Transformed/drug effects , Cell Line, Transformed/enzymology , Cell Line, Tumor/drug effects , Cell Line, Tumor/enzymology , Clone Cells/drug effects , Clone Cells/enzymology , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/biosynthesis , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/physiology , Gene Expression Regulation, Leukemic/drug effects , Genes, abl , HSP90 Heat-Shock Proteins/physiology , Humans , Hydrogen Peroxide/pharmacology , Hydroquinones/adverse effects , Imatinib Mesylate , Lactams, Macrocyclic/adverse effects , Mice , Mutant Proteins/genetics , Mutant Proteins/physiology , Mutation, Missense , Oxidative Stress/drug effects , Point Mutation , Protein Kinase Inhibitors/adverse effects , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-bcr/biosynthesis , Proto-Oncogene Proteins c-bcr/genetics , Reactive Oxygen Species , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , STAT5 Transcription Factor/biosynthesis , STAT5 Transcription Factor/genetics , Signal Transduction/drug effects , Substrate Specificity , Transfection
7.
Biol Reprod ; 76(1): 29-35, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17005940

ABSTRACT

To study mechanisms governing fetoplacental vascular function, we have established a primary ovine fetoplacental artery endothelial (OFPAE) cell line. These OFPAE cells produce nitric oxide (NO), proliferate, and migrate in response to fibroblast growth factor 2 (FGF2) and vascular endothelial growth factor (VEGF). To overcome the senescence crisis that this primary OFPAE cell line will eventually enter, we attempted to establish a functional OFPAE cell line with a prolonged life span by transfecting cells with plasmids containing a neomycin resistance gene and a simian virus 40 gene (SV40) expressing large T (T) and small t (t) antigens. The OFPAE cells at passage 8 were transfected. After neomycin selection, the surviving OFPAE (designated SV40 OFPAE) cells were expanded up to passage 80. Up to passage 30, these SV40 OFPAE cells maintained a morphology similar to untransfected OFPAE cells. Expression of T and t antigens in SV40 OFPAE cells was confirmed by immunocytochemistry. These SV40 OFPAE cells exhibited positive uptake of acetylated low-density lipoprotein (Ac-LDL) and positive staining for NO synthase 3 (NOS3) and formed capillary-like tube structures on Matrigel. Up to passages 20-23, these SV40 OFPAE cells proliferated (P < 0.05) and produced (P < 0.05) NO in response to both FGF2 and VEGF. Moreover, this cell proliferation stimulated by FGF2 and VEGF was dose-dependently inhibited (P < 0.05) by PD98059 (a selective mitogen-activated protein kinase 1 and 2 [MAP2K1/2, also termed MEK1/2] inhibitor) or by LY294002 (a selective phosphoinositide 3-kinase [PI3K] inhibitor). These data indicate that SV40 OFPAE cells, at least at passage 23, retain endothelial phenotypes and functions similar to their parental, untransfected OFPAE cells. Thus, a functional OFPAE cell line with an extended life span has been successfully established, potentially providing a valuable cell model for studying fetoplacental endothelial function.


Subject(s)
Arteries/cytology , Cell Line, Transformed/cytology , Endothelial Cells/cytology , Placenta/blood supply , Animals , Arteries/enzymology , Cell Line, Transformed/drug effects , Cell Line, Transformed/enzymology , Cell Proliferation , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Fibroblast Growth Factor 2/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Plasmids/genetics , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Sheep, Domestic , Simian virus 40/genetics , Transfection , Vascular Endothelial Growth Factor A/pharmacology
8.
Biochem J ; 396(1): 23-30, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16478441

ABSTRACT

CD39/ecto-NTPDase 1 (nucleoside triphosphate diphosphohydrolase 1) is an ecto-nucleotidase that influences P2 receptor activation to regulate vascular and immune cell adhesion and signalling events pivotal in inflammation. Whether CD39 interacts with other membrane or cytoplasmic proteins has not been established to date. Using the yeast two-hybrid system, we note that the N-terminus of CD39 binds to RanBPM (Ran binding protein M; also known as RanBP9), a multi-adaptor scaffolding membrane protein originally characterized as a binding protein for the small GTPase Ran. We confirm formation of complexes between CD39 and RanBPM in transfected mammalian cells by co-immunoprecipitation studies. Endogenous CD39 and RanBPM are also found to be co-expressed and abundant in cell membranes of B-lymphocytes. NTPDase activity of recombinant CD39, but not of N-terminus-deleted-CD39 mutant, is substantially diminished by RanBPM co-expression in COS-7 cells. The conserved SPRY [repeats in splA and RyR (ryanodine receptor)] moiety of RanBPM is insufficient alone for complete physical and functional interactions with CD39. We conclude that CD39 associations with RanBPM have the potential to regulate NTPDase catalytic activity. This intermolecular interaction may have important implications for the regulation of extracellular nucleotide-mediated signalling.


Subject(s)
Antigens, CD/physiology , Apyrase/physiology , B-Lymphocytes/enzymology , Nuclear Proteins/physiology , ran GTP-Binding Protein/physiology , Adaptor Proteins, Signal Transducing , Animals , Antigens, CD/chemistry , Apyrase/chemistry , COS Cells , Cell Line, Transformed/enzymology , Cell Membrane/enzymology , Chlorocebus aethiops , Cytoskeletal Proteins , DNA, Complementary/genetics , Humans , Mice , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary , Recombinant Fusion Proteins/physiology , Structure-Activity Relationship , Transfection , Two-Hybrid System Techniques
9.
Mol Cell Biol ; 25(15): 6464-74, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16024784

ABSTRACT

Murine embryo fibroblasts are readily transformed by the introduction of specific combinations of oncogenes; however, the expression of those same oncogenes in human cells fails to convert such cells to tumorigenicity. Using normal human and murine embryonic fibroblasts, we show that the transformation of human cells requires several additional alterations beyond those required to transform comparable murine cells. The introduction of the c-Myc and H-RAS oncogenes in the setting of loss of p53 function efficiently transforms murine embryo fibroblasts but fails to transform human cells constitutively expressing hTERT, the catalytic subunit of telomerase. In contrast, transformation of multiple strains of human fibroblasts requires the constitutive expression of c-Myc, H-RAS, and hTERT, together with loss of function of the p53, RB, and PTEN tumor suppressor genes. These manipulations permit the development of transformed human fibroblasts with genetic alterations similar to those found associated with human cancers and define specific differences in the susceptibility of human and murine fibroblasts to experimental transformation.


Subject(s)
Cell Line, Transformed/pathology , Fibroblasts/pathology , Oncogene Proteins, Viral/physiology , Animals , Cell Line, Transformed/enzymology , Cell Line, Transformed/virology , Cell Transformation, Neoplastic/pathology , Cyclin-Dependent Kinase Inhibitor p16 , DNA-Binding Proteins/metabolism , Fibroblasts/virology , Humans , Mice , Oncogene Proteins, Viral/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Retinoblastoma Protein/metabolism , Signal Transduction/physiology , Telomerase/metabolism , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/metabolism
10.
Hum Mutat ; 25(3): 239-47, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15714522

ABSTRACT

The cblE type of homocystinuria is a rare autosomal recessive disorder caused by impaired reductive activation of methionine synthase. Although earlier biochemical studies proposed that the methionine synthase enzyme might be activated by two different reducing systems, mutations were reported in only the methionine synthase reductase gene (MTRR) in cblE patients. The pathogenicity of MTRR mutations, however, has not yet been tested functionally. We report on nine patients of European origin affected by the cblE type of homocystinuria. They presented between 2 weeks and 3 years of age (median age 4 weeks) with anemia, which was macrocytic in only three patients, and with neurological involvement in all but two cases. Bone marrow examination performed in seven patients showed megaloblastic changes in all but one of them. All patients exhibited moderate to severe hyperhomocysteinemia (median plasma total homocysteine [Hcy] 92 mumol/L, range 44-169), while clearly reduced methionine was observed only in four cases. Pathogenic mutations were identified in both parental alleles of the MTRR gene in all patients. Five known (c.903+469T>C, c.1361C>T, c.1459G>A, c.1557-4_1557+3del7, and c.1622_1623dupTA) and three novel mutations (c.7A>T, c.1573C>T, and c.1953-6_1953-2del5) were detected. Importantly, transfection of fibroblasts of cblE patients with a wild-type MTRR minigene expression construct resulted in a significant approximately four-fold increase of methionine synthesis, indicating correction of the enzyme defect. Our study shows a link between a milder predominantly hematological presentation and homozygosity for the c.1361C>T mutation, but no other obvious genotype-phenotype correlation. The identification of mutations in the MTRR gene, together with restoration of methionine synthesis following MTRR minigene expression in cblE cells confirms that this disease is caused by defects in the MTRR gene.


Subject(s)
5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/deficiency , Ferredoxin-NADP Reductase/deficiency , Genetic Therapy , Homocystinuria/genetics , Amino Acid Substitution , Betaine/therapeutic use , Brain/pathology , Cell Line, Transformed/enzymology , Cell Line, Transformed/pathology , Codon, Nonsense , DNA Mutational Analysis , Ferredoxin-NADP Reductase/genetics , Fibroblasts/enzymology , Fibroblasts/pathology , Folic Acid/therapeutic use , Genes, Synthetic , Genetic Complementation Test , Haplotypes/genetics , Homocysteine/blood , Homocystinuria/blood , Homocystinuria/classification , Homocystinuria/drug therapy , Homocystinuria/enzymology , Homocystinuria/pathology , Homocystinuria/therapy , Humans , Hydroxocobalamin/therapeutic use , Mutation, Missense , Point Mutation , Polymerase Chain Reaction , Polymorphism, Genetic , Polymorphism, Restriction Fragment Length , Recombinant Fusion Proteins/physiology , Sequence Deletion , Transfection , White People/genetics
11.
Expert Opin Biol Ther ; 4(9): 1423-34, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15335310

ABSTRACT

Chronic granulomatous disease (CGD) is a congenital immune deficiency that is a promising therapeutic target for gene replacement into haematopoietic stem cells (HSCs). CGD results from mutations in any one of four genes encoding subunits of the superoxide-generating NADPH oxidase of phagocytes. Life-threatening, recurrent bacterial and fungal infections, as well as inflammatory granulomas, are the hallmarks of the disease. NADPH oxidase activity can be reconstituted by retroviral- or lentiviral-mediated gene transfer to human CGD marrow in vitro and in xenograft transplant models. Gene transfer studies in knockout mouse models that resemble the human disease suggest that correction of oxidase activity in a minority of phagocytes will be of clinical benefit. Phase I clinical studies in unconditioned CGD patients showed transient expression of small numbers of gene-corrected neutrophils. Areas of research at present include efforts to enhance gene transfer rates into repopulating HSCs using vectors that transduce quiescent cells, and to increase the engraftment of genetically corrected HSCs using non-myeloablative conditioning and drug resistance genes for selection.


Subject(s)
Genetic Therapy , Granulomatous Disease, Chronic/therapy , Animals , Bone Marrow Cells/enzymology , Bone Marrow Transplantation , Cell Line, Transformed/enzymology , Cell Line, Transformed/transplantation , Cells, Cultured/enzymology , Cells, Cultured/transplantation , Clinical Trials, Phase I as Topic , Disease Susceptibility , Dosage Compensation, Genetic , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , Granulomatous Disease, Chronic/complications , Granulomatous Disease, Chronic/genetics , Granulomatous Disease, Chronic/immunology , Hematopoietic Stem Cell Transplantation , Humans , Infections/etiology , Inflammation/etiology , Mice , Mice, Knockout , NADPH Oxidases/deficiency , NADPH Oxidases/genetics , Phagocytes/enzymology , Phagocytosis , Recurrence
12.
Cytogenet Genome Res ; 104(1-4): 87-94, 2004.
Article in English | MEDLINE | ID: mdl-15162019

ABSTRACT

Ectopic expression of telomerase results in an immortal phenotype in various types of normal cells, including primary human fibroblasts. In addition to its role in telomere lengthening, telomerase has now been found to have various functions, including the control of DNA repair, chromatin modification, and the control of expression of genes involved in cell cycle regulation. The investigations on the long-term effects of telomerase expression in normal human fibroblast highlighted that these cells show low frequencies of chromosomal aberrations. In this paper, we describe the karyotypic stability of human fibroblasts immortalized by expression of hTERT. The ectopic overexpression of telomerase is associated with unusual spontaneous as well as radiation-induced chromosome stability. In addition, we found that irradiation did not enhance plasmid integration in cells expressing hTERT, as has been reported for other cell types. Long-term studies illustrated that human fibroblasts immortalized by telomerase show an unusual stability for chromosomes and for plasmid integration sites, both with and without exposure to ionizing radiation. These results confirm a role for telomerase in genome stabilisation by a telomere-independent mechanism and point to the possibility for utilizing hTERT-immortalized normal human cells for the study of gene targeting.


Subject(s)
Chromosomes, Human/radiation effects , Fibroblasts/radiation effects , Telomerase/physiology , Cell Line, Transformed/enzymology , Cell Line, Transformed/radiation effects , Cell Line, Transformed/ultrastructure , Chromosome Aberrations , Chromosomes, Human/metabolism , Clone Cells/enzymology , Clone Cells/radiation effects , Clone Cells/ultrastructure , DNA-Binding Proteins , Fibroblasts/enzymology , Fibroblasts/ultrastructure , Gene Targeting , Humans , Karyotyping , Plasmids/genetics , Radiation Tolerance , Recombinant Fusion Proteins/physiology , Telomerase/genetics , Telomere/ultrastructure , Transfection , Urinary Bladder Neoplasms/pathology
13.
Neuroreport ; 15(2): 245-9, 2004 Feb 09.
Article in English | MEDLINE | ID: mdl-15076745

ABSTRACT

It is necessary to expand human neural progenitor cells in vitro to obtain large numbers for research purposes and cell transplantation. A potential obstacle to in vitro expansion, however, is that neural progenitor cells have a limited replication life-span and gradually lose their differentiation potential. We report here that ectopic expression of the catalytic subunit of human telomerase (hTERT) gene in neural progenitor cells could induce telomerase activity, stabilize telomeres and extend their replicative life-spans. The telomerase-immortalized cells (hNPC-TERT) maintained the normal diploid karyotype, expressed the markers of human neural progenitor cells and meanwhile held the differentiation potential in vitro for up to 120 population doublings. This study provides a new approach for obtaining unlimited quantities of normal phenotypic and homogeneous human neural progenitor cells in vitro.


Subject(s)
Cell Culture Techniques/methods , Cell Line, Transformed/enzymology , Neurons/enzymology , Stem Cells/enzymology , Telomerase/genetics , Transduction, Genetic/methods , Animals , Biomarkers , Catalytic Domain/genetics , Cell Differentiation/genetics , Cell Division/genetics , Cell Line, Transformed/cytology , Cellular Senescence/genetics , Genetic Vectors/genetics , Glial Fibrillary Acidic Protein/metabolism , Humans , Intermediate Filament Proteins/metabolism , Karyotyping , Mice , Nerve Tissue Proteins/metabolism , Nestin , Neurons/cytology , Phenotype , Stem Cells/cytology , Telomere/genetics , Transgenes/genetics
14.
Biochemistry (Mosc) ; 69(3): 306-10, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15061698

ABSTRACT

We studied comparative expression and activity of cytochrome P450 family 1 (CYP1) isoforms in rat embryo cells, both primary and immortalized by Rausher leukemia virus (RLV). In RLV-infected embryonal cells compared with the initial ones the expression levels of CYP1A1 and 1B1 mRNAs and benzo[a]pyrene (BP) hydroxylase activity were higher, regardless of their treatment with the CYP1 inducer 2,3,7,8-tetrachlorodibenzo-p-dioxin. The sensitivity to BP and 7,12-dimethylbenzo[a]anthracene was higher in the cells immortalized with RLV. The expression level of mRNAs of induction-mediating proteins aryl hydrocarbon receptor and aryl hydrocarbon receptor nuclear translocator was the same in both cell cultures tested. Higher sensitivity of cells immortalized with RLV compared with the initial embryo cells to transforming effect of BP, which was described previously, is possibly associated with elevated expression of CYP1 isoforms.


Subject(s)
Aryl Hydrocarbon Hydroxylases/biosynthesis , Cell Line, Transformed/enzymology , Cell Transformation, Viral , Cytochrome P-450 CYP1A1/biosynthesis , Rauscher Virus , Animals , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Receptor Nuclear Translocator , Cell Line, Transformed/virology , Cell Transformation, Viral/drug effects , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1B1 , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/enzymology , Gene Expression Regulation, Neoplastic/drug effects , Isoenzymes/genetics , Isoenzymes/metabolism , Polychlorinated Dibenzodioxins/pharmacology , RNA, Messenger/biosynthesis , Rats , Rats, Inbred F344 , Receptors, Aryl Hydrocarbon/biosynthesis , Receptors, Aryl Hydrocarbon/genetics , Reverse Transcriptase Polymerase Chain Reaction , Teratogens/pharmacology , Transcription Factors/biosynthesis , Transcription Factors/genetics
15.
Exp Cell Res ; 294(2): 559-70, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15023542

ABSTRACT

Human Neural Stem Cells (hNSCs) are excellent candidates for in vitro and in vivo molecular, cellular, and developmental research, and also for ex-vivo gene transfer and cell therapy in the nervous system. However, hNSCs are mortal somatic cells, and thus invariably enter an irreversible growth arrest after a finite number of cell divisions in culture. It has been proposed that this is due to telomere shortening. Here, we show that long-term cultured (up to 4 years) v-myc perpetuated hNSC lines do preserve short but stable and homogeneous telomeres (TRF and Q-FISH determinations). hNSC lines (but not strains) express high levels of telomerase activity, which is activated by v-myc, as demonstrated here. Telomerase activity is not constitutive, becoming non-detectable after differentiation (in parallel to v-myc down-regulation). hNSC lines also maintain a stable cell cycle length, mitotic potential, differentiation and neuron generation capacity, and do not express senescence-associated beta-galactosidase over years, as studied here. These data, collectively, help to explain the immortal nature of v-myc-perpetuated hNSC lines, and to establish them as excellent research tools for basic and applied neurobiological and translational studies.


Subject(s)
Cell Culture Techniques/methods , Cell Line, Transformed/enzymology , Cellular Senescence/genetics , Neurons/enzymology , Pluripotent Stem Cells/enzymology , Telomerase/metabolism , Telomere/enzymology , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Line, Transformed/cytology , Cell Survival/genetics , DNA/genetics , DNA/metabolism , DNA-Binding Proteins , Down-Regulation/genetics , Gene Expression Regulation, Developmental/genetics , Genes, myc/genetics , Humans , Neurons/cytology , Pluripotent Stem Cells/cytology , RNA Stability/genetics , RNA, Messenger/metabolism , Telomerase/genetics , Telomere/genetics
16.
Toxicol Ind Health ; 20(1-5): 57-68, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15807409

ABSTRACT

An in vitro study was conducted to determine the effects of variable concentrations of trace metals on human cultured mammary cells. Monolayers of human mortal (MCF-12A) and immortal (MDA-MB231) mammary epithelial cells were incubated in the absence or presence of increasing concentrations of arsenic (As), mercury (Hg) and copper (Cu) for 24-h, 72-h, 4-d, and 7-d. The MTT assay was used to assess viability for all time periods and cell proliferation was monitored for 4-d and 7-d studies. Monolayers were also labeled with rhodamine-110 (R-6501), Sytox green, and Celltiter blue fluorescent dyes as indicators for intracellular esterase activity, nucleic acid staining, and cell reduction/viability, respectively. Total incubation time with chemical plus dyes was 24 h. For 24-h and 72-h studies, cells were seeded in 96-well plates, after which confluent monolayers were exposed to increasing concentrations of chemicals. For 4-d and 7-d studies, cells were seeded in 12-well plates at 1/3 confluent density (day 0) and exposed to increasing concentrations of metals on day 1. All cells were counted on days 4 and 7. In addition, test medium was removed from select groups of cultures on day 4, replaced with fresh medium in the absence of chemical (recovery studies), and assays were performed on day 7 as above. The data suggest that there is a consistent protective and/or stimulating effect of metals at the lowest concentrations in MCF-12A cells that is not observed in immortal MDA-MB231 cells. In fact, cell viability of MCF-12A cells is stimulated by otherwise equivalent inhibitory concentrations of As, Cu, and Hg on MDA-MB231 cells at 24-h. Whereas As and Hg suppress proliferation and viability in both cell lines after 4-d and 7-d of exposure, Cu enhances cell proliferation and viability of MCF-12A cells. MDA-MB231, however, recover better after 4-days of toxic insult. In addition, nutritional manipulation of media between the cell lines, or pretreatment with penicillamine, did not alter the hormesis effect displayed by MCF-12A. Growth of these cells however was not maintained in the alternative medium. The study demonstrates that a hormesis effect from trace metals is detectable in cultured mammary cells; fluorescent indicators, however, are not as sensitive as cell proliferation or MTT in recognizing the subtle responses. Also, sensitivity of mammary cells to lower concentrations of Cu, a biologically important trace metal, may play an important role in controlling cellular processes and proliferation. The ability to detect this in vitro phenomenon implies that similar processes, occurring in vivo, may be responsible for the development, induction, or enhancement of human cancers.


Subject(s)
Cell Line, Transformed/drug effects , Cell Proliferation/drug effects , Trace Elements/toxicity , Arsenic/toxicity , Cell Line, Transformed/enzymology , Cell Line, Transformed/pathology , Cell Survival/drug effects , Copper/toxicity , Dose-Response Relationship, Drug , Esterases/metabolism , Female , Formazans/metabolism , Humans , Mercury/toxicity , Tetrazolium Salts/metabolism
17.
Int J Oncol ; 23(2): 477-82, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12851698

ABSTRACT

Imidazolium trans-imidazole dimethyl sulfoxide tetrachlororuthenate (NAMI-A) is a new compound active against lung metastasis of solid metastasizing tumours. While its in vivo effect has been studied, the molecular insights that underlie its action are largely unknown. Among the possible pathways responsible for malignant transformation, PKC arose as one of the most promising targets for new antineoplastic drugs. We demonstrated the capability of NAMI-A of inhibiting PMA induced-PKC activity in ECV304 in a dose-dependent fashion. Furthermore, NAMI-A through modulation of PKC activity has been proved capable of reducing the phorbol ester induced expression of ornithine decarboxilase (ODC) gene and to abrogate the activation of the Raf/MEK/ERK pathway. Taken together these results suggest that many of the in vivo outcomes of NAMI-A treatment may be the result of a direct action on PKC.


Subject(s)
Cell Line, Transformed/enzymology , Dimethyl Sulfoxide/analogs & derivatives , Dimethyl Sulfoxide/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Organometallic Compounds/pharmacology , Ornithine Decarboxylase/genetics , Protein Kinase C/metabolism , Signal Transduction/physiology , Tetradecanoylphorbol Acetate/antagonists & inhibitors , Blotting, Western , Humans , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Ornithine Decarboxylase/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ruthenium Compounds , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured
18.
Cancer Res ; 63(12): 3247-56, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12810655

ABSTRACT

Demonstration of the existence of G-quadruplex structures in telomeres of Stylonychia macronuclei and in the promoter of c-myc in human cells has validated these secondary DNA structures as potential targets for drug design. The next important issue is the selectivity of G-quadruplex-interactive agents for the different types of G-quadruplex structures. In this study, we have taken an important step in associating specific biological effects of these drugs with selective interaction with either intermolecular or intramolecular G-quadruplex structures formed in telomeres. Telomestatin is a natural product isolated from Streptomyces anulatus 3533-SV4 and has been shown to be a very potent telomerase inhibitor through its G-quadruplex interaction. We have demonstrated that telomestatin interacts preferentially with intramolecular versus intermolecular G-quadruplex structures and also has a 70-fold selectivity for intramolecular G-quadruplex structures over duplex DNA. Telomestatin is able to stabilize G-quadruplex structures that are formed from duplex human telomeric DNA as well as from single-stranded DNA. Importantly, telomestatin stabilizes these G-quadruplex structures in the absence of monovalent cations, which is a unique characteristic among G-quadruplex-interactive compounds. At noncytotoxic concentrations, telomestatin suppresses the proliferation of telomerase-positive cells within several weeks. In contrast, TMPyP4, a compound that preferentially facilitates the formation of intermolecular G-quadruplex structures, suppresses the proliferation of alternative lengthening of telomeres (ALT)-positive cells as well as telomerase-positive cells. We have also demonstrated that TMPyP4 induces anaphase bridges in sea urchin embryos, whereas telomestatin did not have this effect, leading us to conclude that the selectivity of telomestatin for intramolecular G-quadruplex structures and TMPyP4 for intermolecular G-quadruplex structures is important in mediating different biological effects: stabilization of intramolecular G-quadruplex structures produces telomerase inhibition and accelerated telomere shortening, whereas facilitation of the formation of intermolecular G-quadruplex structures induces the formation of anaphase bridges.


Subject(s)
Enzyme Inhibitors/pharmacology , Nucleic Acid Conformation , Oxazoles/pharmacology , Porphyrins/pharmacology , Telomerase/antagonists & inhibitors , Telomere/drug effects , Anaphase/drug effects , Animals , Cell Line, Transformed/drug effects , Cell Line, Transformed/enzymology , DNA/drug effects , DNA/ultrastructure , Humans , Macromolecular Substances , Potassium/pharmacology , Sea Urchins/embryology , Single-Strand Specific DNA and RNA Endonucleases/metabolism , Sodium/pharmacology , Substrate Specificity , Telomere/ultrastructure
19.
Mol Cell Biol ; 23(6): 2096-108, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12612081

ABSTRACT

The human protein tyrosine phosphatase TCPTP exists as two forms: an endoplasmic reticulum-targeted 48-kDa form (TC48) and a nuclear 45-kDa form (TC45). Although targeted to the nucleus, TC45 can exit in response to specific stimuli to dephosphorylate cytoplasmic substrates. In this study, we investigated the downregulation of insulin receptor (IR) signaling by TCPTP. In response to insulin stimulation, the TC48-D182A and TC45-D182A "substrate-trapping" mutants formed stable complexes with the endogenous tyrosine-phosphorylated IR beta-subunit in 293 cells. Moreover, in response to insulin stimulation, the TC45-D182A mutant accumulated in the cytoplasm of cells overexpressing the IR and in part colocalized with the IR beta-subunit at the cell periphery. These results indicate that the IR may serve as a cellular substrate for both TC48 and TC45. In immortalized TCPTP(-/-) murine embryo fibroblasts, insulin-induced IR beta-subunit tyrosine phosphorylation and protein kinase PKB/Akt activation were enhanced relative to the values in TCPTP(+/+) cells. Importantly, the expression of TC45 or TC48 to physiological levels suppressed the enhanced insulin-induced signaling in TCPTP(-/-) cells. These results indicate that the differentially localized variants of TCPTP may dephosphorylate the IR and downregulate insulin-induced signaling in vivo.


Subject(s)
Insulin/pharmacology , Isoenzymes/physiology , Protein Serine-Threonine Kinases , Protein Tyrosine Phosphatases/physiology , Receptor, Insulin/metabolism , Signal Transduction/drug effects , Animals , CHO Cells/drug effects , CHO Cells/enzymology , Cattle , Cell Line, Transformed/drug effects , Cell Line, Transformed/enzymology , Cell Nucleus/enzymology , Cells, Cultured/drug effects , Cells, Cultured/enzymology , Cricetinae , Cricetulus , Cytoplasm/enzymology , Endoplasmic Reticulum/enzymology , Enzyme Activation , Fibroblasts/drug effects , Fibroblasts/enzymology , Genetic Complementation Test , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , MAP Kinase Signaling System , Macromolecular Substances , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Phosphorylation , Protein Interaction Mapping , Protein Processing, Post-Translational , Protein Tyrosine Phosphatase, Non-Receptor Type 2 , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptor, Insulin/drug effects , Recombinant Fusion Proteins/metabolism
20.
Oncogene ; 22(7): 1024-34, 2003 Feb 20.
Article in English | MEDLINE | ID: mdl-12592389

ABSTRACT

Increased expression of mac25/insulin-like growth factor binding-protein related protein-1 (IGFBP-rP1) in human breast and prostate epithelial cell lines results in the suppression of tumor growth. CDNA expression array analysis revealed increased manganese superoxide dismutase (SOD-2) expression in the mac25/IGFBP-rP1-transfected M12 human prostate cancer cell line compared to M12 control cells. SOD-2 has been postulated to be a tumor suppressor. SOD-2 was also increased in LNCaP cells stably transfected with mac25/IGFBP-rP1, but not in mac25/IGFBP-rP1-transfected PC-3 cells. Mac25 LNCaP cells had a marked decrease in tumor growth in nude mice compared to controls, but there was no difference in tumor growth in mac25 PC-3 cells compared to control. Phosphorylated Erk and Akt were increased in the M12 and LNCaP transfected mac25/IGFBP-rP1 cells but not PC-3 mac25. Inhibition of PI-3 kinase results in a marked decrease in viability of the M12-mac25 cells compared to M12 controls. Cells treated with H(2)O(2) result in an increase in phospho-ERK. Transfection of SOD-2 in M12 cells markedly decreased tumor growth, apoptosis, G1 delay in the cell cycle, and expression of senescence associated beta-galactosidase. These results suggest that one of the downstream mediators of the senescence-associated tumor suppression effect of mac25/IGFBP-rP1 is SOD-2.


Subject(s)
Adenocarcinoma/pathology , Carrier Proteins/physiology , Insulin-Like Growth Factor Binding Proteins , Neoplasm Proteins/physiology , Prostatic Neoplasms/pathology , Signal Transduction/physiology , Superoxide Dismutase/physiology , Adenocarcinoma/enzymology , Animals , Apoptosis/physiology , Cell Line, Transformed/enzymology , Cell Line, Transformed/transplantation , Cellular Senescence , Chromones/pharmacology , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , G1 Phase/physiology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydrogen Peroxide/pharmacology , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Morpholines/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Phenotype , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Promoter Regions, Genetic , Prostatic Neoplasms/enzymology , Protein Processing, Post-Translational , Recombinant Fusion Proteins/physiology , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics , Transfection , Tumor Cells, Cultured/enzymology , Tumor Cells, Cultured/transplantation , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...