Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Diabetologia ; 53(8): 1795-806, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20407745

ABSTRACT

AIMS/HYPOTHESIS: Inflammation contributes to both insulin resistance and pancreatic beta cell failure in human type 2 diabetes. Toll-like receptors (TLRs) are highly conserved pattern recognition receptors that coordinate the innate inflammatory response to numerous substances, including NEFAs. Here we investigated a potential contribution of TLR2 to the metabolic dysregulation induced by high-fat diet (HFD) feeding in mice. METHODS: Male and female littermate Tlr2(+/+) and Tlr2(-/-) mice were analysed with respect to glucose tolerance, insulin sensitivity, insulin secretion and energy metabolism on chow and HFD. Adipose, liver, muscle and islet pathology and inflammation were examined using molecular approaches. Macrophages and dendritic immune cells, in addition to pancreatic islets were investigated in vitro with respect to NEFA-induced cytokine production. RESULTS: While not showing any differences in glucose homeostasis on chow diet, both male and female Tlr2(-/-) mice were protected from the adverse effects of HFD compared with Tlr2(+/+) littermate controls. Female Tlr2(-/-) mice showed pronounced improvements in glucose tolerance, insulin sensitivity, and insulin secretion following 20 weeks of HFD feeding. These effects were associated with an increased capacity of Tlr2(-/-) mice to preferentially burn fat, combined with reduced tissue inflammation. Bone-marrow-derived dendritic cells and pancreatic islets from Tlr2(-/-) mice did not increase IL-1beta expression in response to a NEFA mixture, whereas Tlr2(+/+) control tissues did. CONCLUSION/INTERPRETATION: These data suggest that TLR2 is a molecular link between increased dietary lipid intake and the regulation of glucose homeostasis, via regulation of energy substrate utilisation and tissue inflammation.


Subject(s)
Dietary Fats/metabolism , Insulin Resistance/genetics , Insulin-Secreting Cells/metabolism , Toll-Like Receptor 2/metabolism , Analysis of Variance , Animals , Blood Glucose/metabolism , Calorimetry, Indirect , Cells, Cultured , Female , Inflammation/genetics , Inflammation/metabolism , Insulin/metabolism , Insulin-Secreting Cells/cytology , Male , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 2/genetics
2.
Transpl Int ; 13 Suppl 1: S324-8, 2000.
Article in English | MEDLINE | ID: mdl-11112024

ABSTRACT

The interaction between Fas and its ligand (FasL) induces apoptosis in the Fas-expressing cell. We hypothesized that liposome-mediated FasL gene transduction to the lung allograft, in addition to low-dose immunosuppression, might reduce acute rejection. Orthotopic left lung allotransplantation was performed in male rats (Brown Norway to Fischer F344). FasL gene transfer was performed by use of the plasmid pBCMGSNeo carrying the gene coding for murine FasL and the cationic liposome GL#67:DOPE. Six hundred and sixty micrograms of DNA in 250 microl H2O and 0.5 micromol GL#67 in 250 microl H2O were diluted to 5 ml with saline solution. This emulsion (20 degrees C) was instilled retrogradely through the left pulmonary vein after flushing with LPD solution (20 ml, at 4 degrees C). Subsequently, the graft was stored at 10 degrees C for 3 h. A single dose of cyclosporine A (CsA; 2.5 mg/kg i.m.) was given to all groups 48 h after the transplantation. In group 1 (n = 6), FasL/GL#67 was instilled as described. In group 2 (n = 5), GL#67 was given without DNA. Group 3 (n = 5) animals received CsA only. Five days after transplantation, gas exchange was assessed after exclusion of the contralateral native lung (FiO2 = 1.0). Grafts were flushed with saline solution and fixed in formaldehyde for histological evaluation. No statistical difference in gas exchange (PaO2) between the two control groups 2 (6.4 +/- 0.4 kPa) and 3 (7.4 +/- 0.4 kPa) could be detected 5 days postoperatively (P = 0.9). In contrast, grafts transduced with FasL (group 1) had significantly better gas exchange on postoperative day 5 (PaO2: group 1 37.0 +/- 10.6 kPa vs group 2 6.4 +/- 0.41 kPa; P = 0.002). Two animals in group 1 revealed no or only minimal improvement in gas exchange. Histologically, all lung specimen of all groups showed signs of acute rejection (A2). Leukocyte infiltrates, rated by two independent observers, were less severe in all group 1 animals. Liposome-mediated FasL gene transfer at the time of harvest in combination with low-dose CsA reduces acute rejection in four out of six animals in this model of rat lung allotransplantation.


Subject(s)
Cyclosporine/therapeutic use , Graft Rejection/prevention & control , Lung Transplantation/immunology , Membrane Glycoproteins/physiology , Animals , Dose-Response Relationship, Drug , Fas Ligand Protein , Gene Transfer Techniques , Immunosuppression Therapy/methods , Liposomes , Lung Transplantation/physiology , Male , Membrane Glycoproteins/genetics , Oxygen/blood , Partial Pressure , Plasmids , Pulmonary Veins , Rats , Rats, Inbred BN , Rats, Inbred F344 , Transplantation, Homologous , fas Receptor/physiology
3.
Eur J Immunol ; 30(3): 794-802, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10741394

ABSTRACT

In the normal central nervous system (CNS) expression of MHC class II is minimal, but has been found to be highly up-regulated on microglia cells in experimental autoimmune encephalitis (EAE). Here we used the EAE model to examine the regulation of expression of the class II transactivator (CIITA), which is required for activation of MHC class II genes. EAE was induced in C57BL/6 mice by immunization with myelin oligodendrocyte glycoprotein peptide 35-55. CIITA mRNA form I (specific for dendritic cells) and form IV (IFN-gamma inducible) but not form III (B cell specific) were detected in brain and spinal cord of mice with acute EAE. In unimmunized or mock-immunized mice, none of the three CIITA forms was found to be induced. Dendritic cells (DC) were identified by immunostainings for CD11c in perivascular and meningeal cell infiltrates in EAE spinal cord and brain. Time-course analysis showed (1) the appearance of DC in the CNS shortly before onset of disease, (2) the recruitment of CD11b cells occuring much earlier and (3) the absence of CIITA and MHC class II expression in these CD11b+ cells at preclinical stages.


Subject(s)
Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Genes, MHC Class II , Animals , Astrocytes/immunology , CD11 Antigens/metabolism , Central Nervous System/immunology , Central Nervous System/metabolism , Central Nervous System/pathology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Immunohistochemistry , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Microglia/immunology , Pregnancy , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spinal Cord/immunology , Spinal Cord/metabolism , Spinal Cord/pathology , Transcriptional Activation
4.
Eur J Immunol ; 30(12): 3680-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11169411

ABSTRACT

During inflammatory reactions in the central nervous system (CNS), resident macrophages, the microglia, are exposed to Th1 cell-derived cytokines and pro-apoptotic Fas ligand (FasL). Despite the presence of TNF-alpha and IFN-gamma, both being capable of sensitizing microglia to FasL, apoptosis of microglia is not a hallmark of inflammatory diseases of the CNS. In the present study, TGF-beta is found to counteract the effect of TNF-alpha and IFN-gamma to sensitize microglia to FasL-mediated apoptosis. Resistance to Fas-mediated apoptosis by TGF-beta does not correlate with a down-regulation of Fas expression. As a key inhibitor of Fas-mediated apoptosis, we found expression of the cellular FLICE-inhibitory protein (c-FLIP) to be induced by TGF-beta in resting as well as in activated microglia. Induction of FLIP was found to depend on a mitogen-activated protein kinase kinase (MKK)-dependent pathway as shown by the use of the specific MKK-inhibitor PD98059. The presence of FLIP strongly interfered with FasL-induced activation of caspase-8 and caspase-3 preventing subsequent cell death. The presented data provide the first evidence for a TGF-beta-mediated FLIP in macrophage-like cells and suggest a mode of action for the anti-apoptotic role of TGF-beta in the CNS.


Subject(s)
Apoptosis/drug effects , Caspase Inhibitors , Microglia/physiology , Transforming Growth Factor beta/pharmacology , fas Receptor/physiology , Animals , Caspase 3 , Caspase 8 , Caspase 9 , Fas Ligand Protein , Membrane Glycoproteins/pharmacology , Mice , Mice, Inbred C57BL , Microglia/drug effects
5.
Eur J Immunol ; 29(10): 3053-62, 1999 10.
Article in English | MEDLINE | ID: mdl-10540316

ABSTRACT

The mechanisms underlying neurotoxicity mediated by cytotoxic T lymphocytes (CTL) and their secretory granule proteins perforin and granzymes remain unclear. We evaluated the possible role of the neurotransmitter glutamate in cell death observed in differentiated neurons exposed to CTL-derived granules. Excitotoxicity induced by excessive concentrations of extracellular glutamate is associated with a rise in intracellular calcium and can lead to generation of NO through the activation of glutamatergic N-methyl-D-aspartate (NMDA) receptors. Consistent with an involvement of glutamate, we found that cell death in mature cerebral granule cells was inhibited by 65-80% by two NMDA receptor blockers (MK-801 and D-2-amino-5-phosphonovaleric acid) or a NO synthase blocker (N(G)-nitro-L-arginine methylester). Furthermore, neurons treated with secretory granules responded with a biphasic rise in the intracellular calcium concentration ([Ca2+]i). Whereas MK-801 did not interfere with the immediate rise of [Ca2+]i, the second wave of calcium accumulation starting at 40 min was delayed by 20 min and reduced in amplitude in the presence of MK-801. In immature, NMDA receptor-negative neurons, MK-801 prevented neither the cytotoxicity nor the calcium influx observed 5 min after addition of cytotoxic granules. The demonstration that NMDA receptors and NO are involved in granule-mediated killing of mature neurons opens new avenues in the treatment of neuronal cell death in CTL-mediated diseases such as viral encephalitis.


Subject(s)
Cytoplasmic Granules/immunology , Neurons/immunology , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , T-Lymphocytes, Cytotoxic/immunology , 2-Amino-5-phosphonovalerate/pharmacology , Animals , Calcium/immunology , Calcium/metabolism , Cell Death/immunology , Cell Differentiation , Cells, Cultured , Cerebellum/cytology , Cerebellum/immunology , Cytoplasmic Granules/metabolism , Dizocilpine Maleate/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Mice , Mice, Inbred C57BL , NG-Nitroarginine Methyl Ester/pharmacology , Neurons/cytology , Nitric Oxide/immunology , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
6.
Hum Gene Ther ; 10(10): 1641-8, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10428209

ABSTRACT

Fas ligand (FasL) is a cytokine, produced by activated T cells and NK cells, that triggers apoptosis of Fas-positive target cells including human glioma cells. As shown here, in vitro infection of rat F98 and human LN18 glioma cell lines with recombinant adenovirus (rAd) expressing FasL cDNA under control of the cytomegalovirus promoter (rAd-CMV-FasL) induced striking cytotoxicity in Fas-positive glioma cell lines but not in the Fas-negative F98 glioma subline F98/ZH. The extent of FasL-mediated cytotoxic effects outranged the expectations based on expression of beta-galactosidase (beta-Gal) by F98 cells infected with a control virus expressing the lacZ gene (rAd-CMV-lacZ). The detection of FasL bioactivity in supernatants of infected cells provides evidence of a bystander mechanism involving the cytotoxic action of FasL on uninfected cells. In F98 tumor-bearing rats, infection with rAd-CMV-FasL increased the mean survival time by 50% compared with infection with rAd-CMV-lacZ or untreated controls. These data suggest that viral vector transduction of the FasL gene could be part of a successful glioma gene therapy.


Subject(s)
Adenoviruses, Human , Genetic Therapy , Genetic Vectors , Glioma/therapy , Membrane Glycoproteins/genetics , Animals , Drug Resistance , Fas Ligand Protein , Gene Expression , Genetic Therapy/methods , Humans , Male , Membrane Glycoproteins/biosynthesis , Mice , Neoplasms, Experimental/therapy , Rats , Rats, Inbred F344 , Tumor Cells, Cultured
7.
J Neurooncol ; 39(1): 19-32, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9760067

ABSTRACT

Loss of wild-type p53 activity is one of the most common molecular abnormalities in human cancers including malignant gliomas. The p53 status is also thought to modulate sensitivity to irradiation and chemotherapy. Here, we studied the effect of a p53 gene transfer on the chemosensitivity of three human glioma cell lines with different endogenous p53 status (LN-229, wild-type; LN-18, mutant; LN-308, deleted), using the murine temperature-sensitive p53 val135 mutant. Expression of mutant p53 enhanced proliferation of LN-308 cells but reduced proliferation in the other cell lines. Expression of wild-type p53 caused reversible growth arrest of all cell lines but failed to induce apoptosis. Growth arrest induced by wild-type p53 was associated with strong induction of p21 expression. Strong induction of BAX expression and loss of BCL-2 expression, which are associated with p53-dependent apoptosis rather than growth arrest, were not observed. Wild-type p53 failed to sensitize glioma cells to cytotoxic drugs including BCNU, cytarabine, doxorubicin, teniposide and vincristine. The combined effects of wild-type p53 gene transfer and drug treatment were less than additive rather than synergistic, suggesting that the intracellular cascades activated by p53 and chemotherapy are redundant. Unexpectedly, forced expression of mutant p53 modulated drug sensitivity in that it enhanced the toxicity of some drugs but attenuated the effects of others. These effects may represent a dominant negative effect of mutant p53 in LN-229 cells which have wild-type p53 activity but must be considered a gain of function-type effect in the other two cell lines which have no wild-type p53 activity. Importantly, no clear-cut pattern emerged among the three cell lines studied. We conclude that somatic gene therapy based on the reintroduction of p53 will limit the proliferation of human malignant glioma cells but is unlikely to induce clinically relevant sensitization to chemotherapy in these tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Genes, p53 , Glioma/pathology , Tumor Suppressor Protein p53/physiology , Amino Acid Substitution , Animals , Apoptosis , Carmustine/pharmacology , Cell Division , Cytarabine/pharmacology , Doxorubicin/pharmacology , Gene Deletion , Gene Expression Regulation, Neoplastic/genetics , Genes, bcl-2 , Humans , Mice , Point Mutation , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Recombinant Fusion Proteins/physiology , Temperature , Teniposide/pharmacology , Transfection , Tumor Cells, Cultured/drug effects , Vincristine/pharmacology , bcl-2-Associated X Protein
8.
Leukemia ; 11(11): 1842-9, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9369416

ABSTRACT

A gene encoding the p53 val135 mutant, which assumes mutant conformation at 38.5 degrees C and wild-type conformation at 32.5 degrees C, was introduced into p53-deficient K562 myeloid leukemia cells. Forced expression of wild-type, but not mutant, p53 resulted in growth arrest, accumulation of p21 and Bax proteins, and delayed cell death. Wild-type p53 enhanced the cytotoxic effects of some drugs and attenuated those of others. Compared with wild-type p53, mutant p53 induced much stronger sensitization to drug cytotoxicity. This occurred in the absence of effects on cell cycle progression or activation of several p53 target genes. Although both mutant and wild-type p53 induced changes of immunophenotype, no specific pattern of differentiation was associated with enhanced chemosensitivity. Thus, (1) induction of growth arrest and activation of p53 target genes such as p21 and bax are linked to the wild-type conformation of p53; (2) p53 induces immunophenotypic changes of myeloid leukemia cells suggestive of multidirectional differentiation in a conformation-dependent manner; and (3) (so-called) mutant p53 induces chemosensitization in the absence of effects on cell cycle progression, activation of bax, p21, gadd45 and mdm-2, or a specific pattern of differentiation; and (4) chemosensitization mediated by wild-type p53 may be masked by transcription-dependent induction of growth arrest.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, Myeloid/metabolism , Tumor Suppressor Protein p53/physiology , Animals , Cell Cycle/drug effects , Cell Division/drug effects , Drug Resistance, Neoplasm , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Leukemia, Myeloid/pathology , Leukemia, Myeloid/physiopathology , Mice , Microscopy, Electron , Mutation , Temperature , Transfection , Tumor Cells, Cultured/ultrastructure , Tumor Suppressor Protein p53/genetics
9.
J Neurochem ; 68(3): 889-97, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9048733

ABSTRACT

The puromycin-sensitive aminopeptidase (PSA) is thought to contribute to the degradation of enkephalins. Besides being the most abundant aminopeptidase in the brain, PSA is expressed in other organs as well. From a human fetal brain cDNA library, we have isolated a cDNA encoding the human PSA (huPSA) protein. The isolated cDNA gave rise to a protein with a molecular mass of 99 kDa. Compared with mouse PSA, homology at the amino acid and cDNA level was 98 and 93%, respectively. Translation of the huPSA was found to be initiated at the second of two possible start codons, as shown by studies with antibodies directed against peptide sequences of both potential N-terminal regions. Northern blot analysis with RNA isolated from different human organs demonstrated that the huPSA transcript is strongest but not exclusively expressed in the brain. Vesicular stomatitis virus epitope-tagged huPSA protein was expressed in HeLa cells and found to be localized in the cytoplasm, especially in the perinuclear region. By in situ hybridization, huPSA transcript could be identified in cortical and cerebellar neurons, whereas glial cells and blood vessels remained negative.


Subject(s)
Aminopeptidases/genetics , Aminopeptidases/metabolism , Cloning, Molecular , Neurons/metabolism , Amino Acid Sequence , Animals , Blotting, Northern , HeLa Cells , Humans , In Situ Hybridization , Intracellular Membranes/metabolism , Mice , Molecular Sequence Data , Protein Biosynthesis
10.
Eur J Immunol ; 27(12): 3151-60, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9464800

ABSTRACT

The expression and action of Fas/Fas ligand (FasL) in multiple sclerosis has been postulated as a major pathway leading to inflammatory demyelination. To formally test this hypothesis, C57BL/6-lpr and -gld mice, which due to gene mutation express Fas and FasL in an inactive form, were immunized with myelin oligodendrocyte glycoprotein peptide(35-55). Whereas in wild-type C57BL/6 mice, experimental autoimmune encephalomyelitis (EAE), was chronic/relapsing, EAE in lpr and gld mice was characterized by a lower incidence of disease and a monophasic course. This contrasts with C57BL/6 perforin knockout mice, which showed the most severe form of EAE of all mouse strains tested, the course being chronic relapsing. The difference noted cannot be attributed to an involvement of FasL in oligodendrocyte damage since oligodendrocytes are insensitive to FasL-mediated cytotoxicity in vitro, and since in the acute phase of EAE gld mice also show CD4+ T cell infiltrates with associated demyelination in brain and spinal cord. Unlike oligodendrocytes, astrocytes were killed by FasL in vitro. It remains to be established whether this latter finding explains the different disease course of lpr and gld mice compared to wild-type and perforin knockout mice.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Membrane Glycoproteins/immunology , Myelin-Associated Glycoprotein/immunology , fas Receptor/immunology , Animals , Chronic Disease , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Fas Ligand Protein , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Myelin Proteins , Myelin-Associated Glycoprotein/administration & dosage , Myelin-Oligodendrocyte Glycoprotein , Oligodendroglia/immunology , Oligodendroglia/pathology , Perforin , Pore Forming Cytotoxic Proteins , Recurrence , fas Receptor/genetics
12.
Eur J Immunol ; 26(9): 2271-4, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8814277

ABSTRACT

Cytotoxic T lymphocytes (CTL) kill target cells by perforin-mediated pore formation, induction of apoptosis by the Fas ligand, or both. It has been demonstrated that depolarized neurons can be induced to express major histocompatibility complex (MHC) class I antigens by interferon-gamma. Evidence for antigen-dependent CTL-mediated killing was obtained by transfecting neurons with MHC class I cDNA. The present study was designed to investigate the mechanisms of killing of cerebellar granule neurons depolarized by high K+ concentrations and thereby inducible for MHC class I antigen expression. We found that neurons express only low levels of Fas (APO-1/CD95) and are resistant to Fas ligand-mediated killing even when pretreated with cytokines. However, granules extracted from CTL as well as purified perforin induce almost complete lysis of neurons. These data suggest that CTL-mediated elimination of neurons involves the perforin, but not the Fas pathway of target cell killing.


Subject(s)
Histocompatibility Antigens Class I/physiology , Membrane Glycoproteins/physiology , Neurons/immunology , T-Lymphocytes, Cytotoxic/physiology , fas Receptor/physiology , Animals , Cytotoxicity, Immunologic , Interferon-alpha/pharmacology , Interferon-gamma/pharmacology , Mice , Mice, Inbred C57BL , Perforin , Pore Forming Cytotoxic Proteins
13.
Exp Cell Res ; 221(2): 395-403, 1995 Dec.
Article in English | MEDLINE | ID: mdl-7493639

ABSTRACT

The phosphatase inhibitors okadaic acid and calyculin A were used to examine the role of phosphorylation processes in T cell apoptosis induced by interleukin-2 (IL-2) deprivation or transforming growth factor-beta 2 (TGF-beta 2). Okadaic acid and calyculin A inhibited IL-2-driven T cell proliferation and induced apoptosis at concentrations known to inhibit protein phosphatase 1. High concentrations of both agents caused toxic changes of prominent cellular swelling and dilatation of rough endoplasmic reticular profiles. When the T cells were induced to undergo apoptosis by IL-2 deprivation, okadaic acid and calyculin A delayed loss of membrane integrity, nucleosomal size DNA fragmentation, and loss of bcl-2 mRNA. However, T cells deprived of IL-2 in the presence of okadaic acid or calyculin A revealed DNA breaks by in situ DNA end labeling and apoptotic morphology by electron microscopy and failed to show enhanced survival after reexposure to IL-2. Although TGF-beta-mediated signaling is thought to involve the dephosphorylation of specific substrates, okadaic acid and calyculin A not only failed to inhibit, but actually augmented, TGF-beta 2-induced inhibition of T cell proliferation and induction of apoptosis. Exposure to either TGF-beta 2 or the phosphatase inhibitors prevented phosphorylation of the retinoblastoma protein RB. In summary, okadaic acid and calyculin A: (i) induce T cell apoptosis in the presence of IL-2, (ii) allow us to distinguish essential from epiphenomenal features of T cell apoptosis after IL-2 deprivation, and (iii) cooperate with TGF-beta 2 in inducing growth arrest and apoptosis of murine T cells via intracellular cascades that converge in the prevention of RB phosphorylation.


Subject(s)
Apoptosis , Interleukin-2/physiology , Phosphoprotein Phosphatases/antagonists & inhibitors , T-Lymphocytes/cytology , Transforming Growth Factor beta/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Division/drug effects , Cell Line , Cell Membrane/drug effects , DNA Damage , Endoplasmic Reticulum, Rough/drug effects , Enzyme Inhibitors/pharmacology , Ethers, Cyclic/pharmacology , Gene Expression Regulation/drug effects , Interleukin-2/pharmacology , Marine Toxins , Mice , Nucleosomes/drug effects , Okadaic Acid , Oxazoles/pharmacology , Phosphorylation , Protein Phosphatase 1 , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2 , RNA, Messenger/metabolism , Retinoblastoma Protein/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/enzymology
14.
Cancer Res ; 55(13): 2936-44, 1995 Jul 01.
Article in English | MEDLINE | ID: mdl-7540953

ABSTRACT

Human malignant glioma cells are susceptible to apoptosis induced by antibodies to Fas/APO-1, a cytokine receptor protein of the nerve growth factor/tumor necrosis factor receptor superfamily. Here we show that a critical level of cell surface expression of Fas/APO-1 is a prerequisite for induction of glioma cell apoptosis via Fas/APO-1. Although Fas/APO-1 mRNA was expressed in three Fas/APO-1 antibody-resistant glioma cell lines, these cells expressed either little Fas/APO-1 protein (LN-319 and LN-405) or an abnormal Fas/APO-1 protein that was not translocated to the cell membrane and therefore functionally inactive (LN-308). Although all glioma cell lines expressed mRNA for Fas/APO-1-delta TM, a soluble form of Fas/APO-1 lacking the transmembrane domain, none of the cell lines released detectable amounts of soluble Fas/APO-1, a potential endogenous antagonist of Fas/APO-1-mediated glioma cell apoptosis. Stable transfection of three resistant glioma cell lines with a human Fas/APO-1 cDNA expression vector dramatically enhanced cell surface expression of Fas/APO-1 and induced susceptibility to Fas/APO-1 antibody-mediated apoptosis. These data indicate that malignant glioma cells, unlike other tumor cells, uniformly harbor the intracellular cascade required for Fas/APO-1-mediated apoptosis. Low level of Fas/APO-1 expression results from inefficient transcription and translation of the Fas/APO-1 gene or the synthesis of mutant Fas/APO-1 proteins. gamma-Interferon, tumor necrosis factor-alpha, and interleukin 1 beta augmented Fas/APO-1-mediated apoptosis of Fas/APO-1-transfected glioma cells by acting on the subcellular suicidal cascade triggered by Fas/APO-1 activation. Dexamethasone attenuated Fas/APO-1 antibody-induced apoptosis, not only of constitutively Fas/APO-1-positive glioma cells, but also of Fas/APO-1-transfected glioma cells. The antiapoptotic effect of dexamethasone could be overcome by preexposure of the glioma cells to gamma-interferon or by coexposure to Fas/APO-1 antibodies and cycloheximide. Thus, Fas/APO-1 gene transfer and combined immunotherapy using Fas/APO-1 antibodies and cytokines may overcome Fas/APO-1 antibody resistance of Fas/APO-1-negative human malignant glioma cells, which may represent subpopulations within single gliomas or form a separate subgroup of human malignant gliomas.


Subject(s)
Antigens, Surface/administration & dosage , Glioma/therapy , Antibodies/administration & dosage , Antigens, Surface/genetics , Apoptosis , Base Sequence , Cycloheximide/pharmacology , DNA Primers/chemistry , Dexamethasone/pharmacology , Gene Transfer Techniques , Genetic Therapy , Glioma/genetics , Humans , In Vitro Techniques , Interferon-gamma/pharmacology , Membrane Glycoproteins/metabolism , Molecular Sequence Data , Tumor Cells, Cultured , fas Receptor
15.
J Clin Invest ; 95(6): 2633-43, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7539458

ABSTRACT

The majority of human malignant glioma cells express Fas/APO-1 and are susceptible to Fas/APO-1 antibody-mediated apoptosis in vitro. The sensitivity of Fas/APO-1-positive glioma cell lines to Fas/APO-1 antibody-mediated killing correlates inversely with the constitutive expression of the antiapoptotic protooncogene bcl-2. Here we report that BCL-2 protein expression of human glial tumors in vivo correlates with malignant transformation in that BCL-2 immunoreactive glioma cells were more abundant in WHO grade III/IV gliomas than in grade I/II gliomas. Fas/APO-1 antibody-sensitive human glioma cell lines stably transfected with a murine bcl-2 cDNA acquired resistance to Fas/APO-1 antibody-mediated apoptosis. Forced expression of bcl-2 also attenuated TNF alpha-mediated cytotoxicity of glioma cell lines in the presence of actinomycin D and cycloheximide and conferred partial protection from irradiation and the cancer chemotherapy drugs, cisplatin and BCNU. Preexposure of the glioma cell lines to the cytokines, IFN gamma and TNF alpha, which sensitize for Fas/APO-1-dependent killing, partially overcame bcl-2-mediated rescue from apoptosis, suggesting that multimodality immunotherapy involving cytokines and Fas/APO-1 targeting might eventually provide a promising approach to the treatment of human malignant gliomas.


Subject(s)
Antigens, Surface/physiology , Apoptosis , Glioma/pathology , Proto-Oncogene Proteins/genetics , Animals , Cytokines/pharmacology , DNA Damage , DNA, Neoplasm/chemistry , Dexamethasone/pharmacology , Drug Resistance , Gene Expression Regulation, Neoplastic , Gene Transfer Techniques , Glioma/drug therapy , Glioma/radiotherapy , Humans , In Vitro Techniques , Mice , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogenes , RNA, Messenger/genetics , Transfection , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor
16.
Genomics ; 26(2): 272-80, 1995 Mar 20.
Article in English | MEDLINE | ID: mdl-7601453

ABSTRACT

N-Oct 3 is a human POU domain transcription factor that binds to the octamer sequence ATGCAAAT. The protein is expressed in the central nervous system during development and in adult brain. We have isolated and characterized genomic clones encoding the human N-Oct 3 gene (HGMW-approved symbol POUF3). Comparison of the structure of these clones with the N-Oct 3 cDNA revealed that POUF3 is an intronless gene. Sequencing of 650 bp of the promoter region showed 84% sequence identity of POUF3 with its murine homologue, the brain-2 (designated brn-2) gene. Whereas both POUF3 and brn-2 lack a TATA box, consensus sequences for AP-2, GCF, and SP1 transcription factors were identified within the highly conserved 5'-flanking region. These sequences may play a crucial role for the tissue-specific transcription activation of the POUF3 gene. Southern blotting and in situ hybridization localized the human POUF3 gene to chromosome 6q16.


Subject(s)
Chromosomes, Human, Pair 6 , Genes , Genome, Human , Nerve Tissue Proteins/genetics , Transcription Factors/genetics , Animals , Base Sequence , Chromosome Mapping , Homeodomain Proteins , Humans , In Situ Hybridization, Fluorescence , Mice , Molecular Sequence Data , POU Domain Factors , Sequence Alignment , Sequence Homology, Nucleic Acid , Species Specificity , Transcription, Genetic
17.
Dev Biol ; 165(2): 602-14, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7958425

ABSTRACT

AP-2 is a cell type-specific DNA-binding transcription factor that regulates selected target genes in vertebrate organisms. Here we investigated cell type-specific expression and regulation of AP-2 in neuroectodermal cell lineages. During retinoic acid (RA)-mediated differentiation of P19 embryonal carcinoma cells into neuroectodermal cell types that include immunohistochemically defined neurons and astrocytes, we observed a strong induction of AP-2 transcripts and protein. In contrast, AP-2 mRNA was not induced in P19 cells which undergo mesoendodermal differentiation in response to 1% dimethylsulfoxide or low concentrations of RA, respectively. The potential of both neurons and astrocytes to express AP-2 was ascertained by using cerebellar neurons and astrocytes derived from newborn mice. Unlike these types of cells, microglial cells do not express AP-2. Dibutyryl cyclic AMP further enhanced levels of AP-2 transcripts in both P19 astrocytes and primary astrocytes which also respond to agents elevating intracellular cAMP (noradrenaline, isoproterenol, forskolin). The cAMP-dependent induction of AP-2 could be blocked by inhibitors of protein kinase A. In contrast to its action in P19 cells, RA had no effect on AP-2 mRNA levels in primary astrocytes. Our results indicate that AP-2 may play a role as a retinoic acid-sensitive regulator during differentiation of neurons and glia from an embryonic neural precursor. Furthermore, AP-2 may be involved in gene transcription in both mature neurons and astrocytes.


Subject(s)
Astrocytes/metabolism , Cerebellum/metabolism , DNA-Binding Proteins/metabolism , Ectoderm/metabolism , Microglia/metabolism , Neurons/metabolism , Transcription Factors/metabolism , Animals , Animals, Newborn , Base Sequence , Bucladesine/pharmacology , Cell Differentiation , Cyclic AMP/physiology , DNA Primers/chemistry , Gene Expression/drug effects , Mice , Molecular Sequence Data , Protein Kinase Inhibitors , RNA, Messenger/genetics , Transcription Factor AP-2 , Tretinoin/pharmacology
18.
Eur J Immunol ; 24(6): 1293-300, 1994 Jun.
Article in English | MEDLINE | ID: mdl-8206089

ABSTRACT

Transforming growth factor-beta (TGF beta) is a potent immunosuppressive cytokine which inhibits the antigen (Ag)-dependent expansion of T cells both in vitro and in vivo by mechanisms not well defined yet. Here we report that exposure of interleukin (IL)-2-dependent T cell lines to TGF beta 2 results in apoptosis defined by morphology, nucleosomal size DNA fragmentation and in situ DNA end labeling. TGF beta 2-induced T cell apoptosis showed the following characteristics: (1) in contrast to the rapid evolution of apoptosis following IL-2 deprivation, apoptosis of T cells triggered by TGF beta 2 was delayed; (2) cycloheximide prevented TGF beta 2-induced apoptosis of CTLL-2 but not of OVA-7 T helper cells; (3) in contrast to apoptosis following IL-2 deprivation, TGF beta 2-mediated T cell apoptosis was not associated with decreased expression of the proto-oncogenes, bcl-2 or c-myc; (4) TGF beta 2-induced apoptosis was not restricted to IL-2-dependent T cell lines since the IL-4-dependent T cell line, CT.4S, as well as EL4 lymphoma cells, which grow independently of exogenous IL-2, were also susceptible to TGF beta 2-mediated apoptosis. Taken together, these data may present a novel mechanism of TGF beta 2-mediated suppression of T cell expansion in response to Ag and IL-2, the activation of the endogenous death program of apoptosis, which appears to operate independently of direct interactions of TGF beta 2 with the IL-2/IL-2 receptor system.


Subject(s)
Apoptosis/physiology , Proto-Oncogene Proteins/biosynthesis , RNA, Messenger/biosynthesis , T-Lymphocytes/physiology , Transforming Growth Factor beta/physiology , Animals , Apoptosis/genetics , Cells, Cultured , Down-Regulation/physiology , Genes, myc/genetics , Interleukin-2/physiology , Mice , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogenes
20.
Nucleic Acids Res ; 21(2): 253-8, 1993 Jan 25.
Article in English | MEDLINE | ID: mdl-8441633

ABSTRACT

Octamer transcription factors (Oct or OTF) are a subset of the POU family of transcription factors which regulate transcription of cellular and viral genes by binding to the octamer sequence motif ATGCAAAT. Neurons and astroglial cells harbour, in addition to the ubiquitous Oct 1 factor, at least four specific factors termed N-Oct 2,3,4 and 5. Here we report the cloning of a human brain-derived cDNA that encodes the N-Oct 3 protein (443 aa) which is the human counterpart of the murine brain-2 gene product. Extracts from mammalian cells transfected with an N-Oct 3 expression vector yield three octamer DNA binding complexes in the electrophoretic mobility shift assay (EMSA): N-Oct 3 and two smaller complexes comigrating with the N-Oct 5A and 5B proteins of brain extracts. We present data suggesting that the N-Oct 5A and 5B proteins are generated by alternative translation initiation at internal AUG residues which are located before the POU domain. In contrast to the putative N-Oct 5 proteins, which are transcriptionally inert, the N-Oct 3 protein activates transcription from a reporter gene promoter with an octamer sequence, when transiently expressed in HeLa cells.


Subject(s)
Brain/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Blotting, Northern , Cloning, Molecular , DNA/metabolism , Gene Expression Regulation , HeLa Cells , Homeodomain Proteins , Humans , Mice , Molecular Sequence Data , Organ Specificity/genetics , POU Domain Factors , Promoter Regions, Genetic , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...