Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Exp Hematol ; 26(13): 1202-8, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9845375

ABSTRACT

In this study we describe the expression and function of Fas in mouse bone marrow (BM) stromal cells (SCs) and cell lines derived from long-term BM cultures. Flow cytometry analysis showed that Fas was expressed on adherent cells from freshly isolated BM and on all cloned SC lines tested. The SC line ME-25 was Fas+ but negative for FasL as detected by reverse transcriptase-polymerase chain reaction. Furthermore, ME-25 was CD44+, VCAM-1+, Mac-3-, Gr-1-, and type IV collagen-. ME-25 treatment with interferon-gamma or tumor necrosis factor-alpha significantly induced upregulation of Fas expression as detected by both flow cytometry and Western blot immunoassay. The same treatment with interleukin (IL)-1, IL-2, or IL-13 had no effect. Functional studies demonstrated that Fas induced a strong increase in apoptosis when engaged with an anti-Fas monoclonal antibody (MoAb). Activated BM T cells induced Fas-dependent cytotoxicity of ME-25 insofar as blocking anti-FasL MoAb inhibited the killing of ME-25 induced by activated BM T cells. These data suggest a possible involvement of Fas-expressing SCs in negative regulatory functions in the BM and provide a starting point for further studies on the role of Fas+ SCs.


Subject(s)
Bone Marrow Cells/metabolism , Gene Expression Regulation , Interferon-gamma/pharmacology , Stromal Cells/metabolism , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor/genetics , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis , Cell Line , Cytotoxicity, Immunologic , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , fas Receptor/immunology
2.
Cancer Lett ; 114(1-2): 97-9, 1997 Mar 19.
Article in English | MEDLINE | ID: mdl-9103263

ABSTRACT

Short chain fatty acids (propionate and butyrate) and deoxycholic acid (DCA) are able to induce apoptosis in HT-29 colonic tumor cell line, but DCA induces a much higher level of apoptosis than butyrate and propionate. Mixtures of DCA with butyrate or propionate enhance the effect of the single components. Apoptosis is not affected by the PKC, PTK or de novo mRNA and protein synthesis inhibitors, so that the involvement of these enzymes and processes is ruled out. In contrast, DCA-induced apoptosis is directly related to [Ca2+]i concentration as demonstrated by the apoptosis inhibition caused by [Ca2+]i chelator BAPTA/AM.


Subject(s)
Apoptosis/drug effects , Butyrates/pharmacology , Colonic Neoplasms/pathology , Deoxycholic Acid/pharmacology , Propionates/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Butyric Acid , Calcium/analysis , Chelating Agents/pharmacology , Colonic Neoplasms/chemistry , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Dose-Response Relationship, Drug , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Genistein , Humans , Isoflavones/pharmacology , Protein Synthesis Inhibitors/pharmacology , Time Factors , Tumor Cells, Cultured
3.
Leuk Res ; 18(6): 431-9, 1994 Jun.
Article in English | MEDLINE | ID: mdl-8207961

ABSTRACT

Genistein, a natural isoflavonoid phytoestrogen, is a strong inhibitor of protein tyrosine kinases. We analyzed the effects of genistein on in vitro growth, cell-cycle progression and chromatin structure of Jurkat cells, a T-cell leukemia line with a constitutively increased tyrosine phosphorylation pattern. Exposure of in vitro cultured Jurkat cells to genistein resulted in a dose-dependent, growth inhibition. Cell-cycle analysis of genistein-treated cells revealed a G2/M arrest at low genistein concentrations (5-10 micrograms/ml), while at higher doses (20-30 micrograms/ml) there was also a perturbation in S-phase progression. The derangements in cell-cycle control were followed by apoptotic death of genistein-treated cells. Immunocytochemical analysis of cells stained with a FITC-conjugated anti-phosphotyrosine monoclonal antibody showed that 30 micrograms/ml genistein effectively inhibit tyrosine kinase activity in cultured Jurkat cells. Our results indicate that the natural isoflavone genistein antagonizes tumor cell growth through both cell-cycle arrest and induction of apoptosis and suggest that it could be a promising new agent in cancer therapy.


Subject(s)
Antineoplastic Agents/toxicity , Isoflavones/toxicity , Protein-Tyrosine Kinases/antagonists & inhibitors , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line , DNA, Neoplasm/analysis , Dose-Response Relationship, Drug , Genistein , Humans , Kinetics , Leukemia, T-Cell , Time Factors , Tumor Cells, Cultured
4.
Am J Surg ; 176(4): 311-4, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9817245

ABSTRACT

BACKGROUND: Gastrostomies provide reliable long-term enteral access in patients with traumatic brain injuries. The impact of technique of gastrostomy on total hospital cost is not known. METHODS: A retrospective analysis of patients who sustained head trauma and required gastrostomies for long-term enteral access between 1 July 1990 and 1 July 1996 was performed. RESULTS: The patients who received percutaneous endoscopic gastrostomies (PEG) were similar to patients who received Stamm gastrostomies (OPEN) with respect to age, injury severity score, mechanism of injury, associated injuries, complication rates, and deaths. Total hospital costs ($ x 10(3)) were lower for patients who had PEGs placed in the intensive care unit (78.2 +/- 37.4) or endoscopy suite (71.9 +/- 37.7) compared with PEGs placed in the operating room (122.4 +/- 75.7) or OPEN gastrostomies (119.8 +/- 65.1). CONCLUSIONS: In head-injured patients, PEGs are a reliable method of obtaining long-term enteral access with a complication rate equivalent to Stamm gastrostomies. If performed in either the intensive care unit or the endoscopy suite, PEGs are associated with significantly reduced total hospital costs.


Subject(s)
Craniocerebral Trauma/complications , Endoscopy/economics , Gastrostomy/economics , Hospital Costs , Adult , Aged , Cost Control , Cost-Benefit Analysis , Craniocerebral Trauma/therapy , Endoscopy/adverse effects , Endoscopy/methods , Enteral Nutrition/economics , Enteral Nutrition/methods , Female , Gastrostomy/adverse effects , Gastrostomy/methods , Humans , Male , Middle Aged , Retrospective Studies , Treatment Outcome
5.
Haemophilia ; 8(1): 28-32, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11886462

ABSTRACT

Acquired inhibitors to FVIII (anti-FVIII) are uncommon in children. An acquired anti-FVIII developed in a previously healthy 4-year-old boy treated with penicillin for streptococcal pharyngitis. Aspirin prophylaxis begun for suspected rheumatic fever led to compartment syndromes of all four extremities, which resolved with high-dose FVIII and surgical decompression. Anti-FVIII in this patient, and the five additional cases identified in a survey of 160 haemophilia treatment centres, occurred at a median age of 8 years, with median initial and peak titres of 4.6 and 6.9 Bethesda Units (BU), respectively. All six presented with bleeding, including haematomas (three intramuscular, one intracranial), and ecchymoses in three. The median baseline FVIII was 0.05 U mL(-1), and the median baseline activated partial thromboplastin time (APTT) was 79.8 s. The inhibitor resolved completely in five patients (83%) within a median 5 months, after treatment with FVIII concentrate, steroids, cytoxan, methotrexate, and no treatment. The inhibitor persisted in the patient with Goodpasture's disease, despite steroids, cytoxan, cyclosporin, and intravenous gamma globulin. Aspirin therapy, in two, worsened ongoing bleeding. The association of penicillin-like drugs in this and three other cases in the literature suggest that to avoid potential catastrophic bleeding, it is prudent to obtain an APTT prior to initiating aspirin for suspected rheumatic fever. In conclusion, acquired anti-FVIII inhibitors in children may cause severe bleeding, and remit in the majority after FVIII and/or immunosuppressive therapy.


Subject(s)
Autoantibodies/blood , Factor VIII/immunology , Hemophilia A/chemically induced , Adolescent , Aspirin/administration & dosage , Aspirin/adverse effects , Autoimmune Diseases/chemically induced , Autoimmune Diseases/etiology , Autoimmune Diseases/immunology , Child , Child, Preschool , Compartment Syndromes/chemically induced , Compartment Syndromes/complications , Compartment Syndromes/drug therapy , Contraindications , Factor VIII/administration & dosage , Female , Hemophilia A/etiology , Hemophilia A/immunology , Hemorrhage/chemically induced , Hemorrhage/etiology , Hemorrhage/immunology , Humans , Male , Partial Thromboplastin Time , Penicillins/administration & dosage , Penicillins/adverse effects , Rheumatic Fever/drug therapy
6.
Cell Immunol ; 141(2): 323-31, 1992 May.
Article in English | MEDLINE | ID: mdl-1533568

ABSTRACT

We have previously shown that interleukin-2 (IL-2) is able to induce the generation of natural killer (NK) activity in bone marrow (BM) cell cultures from mice pretreated with 5-fluorouracil (5-FU). Cell fractionation experiments to analyze the nature of BM precursors indicate that MAC-1-, NK1-1- noncytotoxic precursors are induced by IL-2 to proliferate and generate cytolytic NK cells. These data demonstrate that the phenotype and functional characteristics of the IL-2-responsive cells in the FUBM are different from those of mature NK cells in that they are MAC-1+, NK1.1+, CD3- and susceptible to boosting by IFN-alpha.


Subject(s)
Bone Marrow/drug effects , Hematopoietic Stem Cells/immunology , Interleukin-2/pharmacology , Killer Cells, Natural/immunology , Animals , Antigens/analysis , Antigens, Differentiation, T-Lymphocyte/analysis , Antigens, Ly , Antigens, Surface , Bone Marrow Cells , CD3 Complex , Cell Differentiation , Female , Lectins, C-Type , Macrophage-1 Antigen/analysis , Male , Mice , NK Cell Lectin-Like Receptor Subfamily B , Proteins/analysis , Receptors, Antigen, T-Cell/analysis , Tumor Cells, Cultured
7.
Cell Immunol ; 178(2): 124-31, 1997 Jun 15.
Article in English | MEDLINE | ID: mdl-9225003

ABSTRACT

Glucocorticoids (GCH) are highly effective agents in controlling inflammation and immune response. We studied the effect of the synthetic GCH dexamethasone (DEX) on the expression of TCR zeta gene splicings that code for some chains belonging to the T-cell receptor (TCR)/CD3 complex. In the DEX-treated hybridoma T-cell line 3DO, TCR zeta gene splicings increase within the first 24 hr (about fourfold increase), as demonstrated by reverse transcriptase-polymerase chain reaction and RNase protection assay. This increase is due to the stimulation of TCR zeta gene locus transcription, as demonstrated by the "run-on" assay. A similar upregulation was observed in murine thymocytes following in vivo DEX treatment. As a consequence of TCR zeta gene locus modulation, the expression of the spliced mRNAs coding for TCR zeta and TCR eta subunits is increased, whereas their relative ratio is only slightly changed. Indeed, the amount of TCR zeta protein in 24-hr DEX-treated cells is fivefold more than that in the untreated cells. A similar effect was seen in 3DO cells treated with hydrocortisone but not in those treated with testosterone. TCR zeta protein increase was confined to the cytoplasm and therefore TCR/CD3 complex expression did not increase. This newly described effect of DEX may constitute an additional molecular mechanism that contributes to its immunomodulating activity.


Subject(s)
CD3 Complex/genetics , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Hydrocortisone/pharmacology , Membrane Proteins/genetics , Receptors, Antigen, T-Cell/genetics , Animals , Cell Compartmentation/drug effects , Cell Membrane/metabolism , Gene Expression Regulation/drug effects , Mice , Mice, Inbred C3H , RNA Splicing , RNA, Messenger/genetics , Testosterone/pharmacology , Transcription, Genetic/drug effects , Up-Regulation/drug effects
8.
Mol Cell Biochem ; 167(1-2): 135-44, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9059990

ABSTRACT

Glucocorticoid hormones (GCH) are anti-inflammatory and immunosuppressive agents that inhibit T-cell growth and activation. Since the T-cell receptor (TCR)/CD3 complex mediates T-lymphocyte activation, we studied the effect of in vitro dexamethasone (DEX), a synthetic GCH, on TCR/CD3 expression. DEX-treatment of a hybridoma T-cell line and normal un-transformed T-cell clones induced a decrease of the TCR/ CD3 membrane expression after 4 days. After 4 weeks, TCR/CD3 was undetectable. However, the amount of mRNAs coding TCR/CD3 chains, including TCR alpha, TCR beta, CD3 gamma, CD3 theta and CD3 epsilon, as well as the amount of CD3 epsilon protein, a major component of the complex, were unaltered. By contrast, a decrease of the mRNAs deriving from the TCR zeta gene locus, as well as of the TCR zeta protein which is responsible for the membrane expression of the TCR/CD3 complex, was induced. These data suggest that the down-modulation of TCR expression is due to the diminution of TCR zeta gene products in DEX-treated cells.


Subject(s)
CD3 Complex/drug effects , CD3 Complex/metabolism , Dexamethasone/pharmacology , Animals , Blotting, Western , CD3 Complex/genetics , Down-Regulation , Glucocorticoids/pharmacology , Membrane Proteins/drug effects , Membrane Proteins/genetics , Mice , RNA Splicing , RNA, Messenger/biosynthesis , Receptors, Antigen, T-Cell/drug effects , Receptors, Antigen, T-Cell/genetics , Transcription, Genetic
9.
Int J Immunopharmacol ; 18(12): 677-84, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9172010

ABSTRACT

Glucocorticoid hormones (GCs) are able to modulate leukocyte activity. We studied the effect of dexamethasone (DEX) on the expression of CD2, an adhesion molecule involved in T-lymphocyte homing and activation. Results of flow cytometry analysis and immunoprecipitation with anti-CD2 monoclonal antibodies (mAbs) indicated that in vitro treatment with DEX augments CD2 expression in transformed T-cell lines. This effect correlated with a rapid increase in the mRNA and was inhibited by actinomycin-D (AD). The DEX-induced CD2 augmentation was transient, peaked at days 1-2 and returned to the levels of untreated controls at days 3-4. It was a dose-dependent phenomenon, mediated by the GC receptor (GCR), because it was inhibited by the GCR antagonist RU486, and was not induced by other steroids such as testosterone and progesterone. This CD2 modulation could presumably contribute to GC-induced effects on T-cell activity.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , CD2 Antigens/biosynthesis , Dexamethasone/pharmacology , Animals , CD2 Antigens/drug effects , CD2 Antigens/genetics , Dactinomycin/pharmacology , Hormone Antagonists/pharmacology , Mifepristone/pharmacology , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/metabolism , T-Lymphocytes/metabolism , Tumor Cells, Cultured
10.
Blood ; 89(10): 3717-26, 1997 May 15.
Article in English | MEDLINE | ID: mdl-9160677

ABSTRACT

Anti-CD3 monoclonal antibodies (MoAbs) and glucocorticoid hormones induce apoptosis in immature thymocytes and peripheral T lymphocytes. This process is inhibited by a number of growth factors, including interleukin-2 (IL-2), IL-3, and IL-4, as well as by triggering of the adhesion molecule CD44, which would indicate that signals generated by membrane receptors can modulate the survival of lymphoid cells. To investigate whether triggering of CD2 may also affect apoptosis in lymphoid cells, we analyzed the effect of stimulation with anti-CD2 MoAbs on T-cell apoptosis induced by two stimuli, anti-CD3 MoAbs and dexamethasone (DEX), using a hybridoma T-cell line and a T-helper cell clone. The results show that CD2 engagement decreased anti-CD3 MoAb-induced apoptosis, but did not influence DEX-induced cell death. Furthermore, the decrease appeared to be related to the expression of Fas/APO-1 (CD95) and Fas-ligand (Fas-L). In fact, we show that CD2 stimulation inhibits apoptosis by preventing the CD3-induced upregulation of Fas and Fas-L in a Fas-dependent experimental system. These data suggest that a costimulatory molecule may control a deletion pathway and may therefore contribute to the regulation of peripheral tolerance.


Subject(s)
Apoptosis , CD2 Antigens/physiology , Membrane Glycoproteins/physiology , Receptor-CD3 Complex, Antigen, T-Cell/physiology , T-Lymphocytes/cytology , fas Receptor/physiology , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , CD2 Antigens/immunology , Cytotoxicity, Immunologic , Dexamethasone/pharmacology , Fas Ligand Protein , Hybridomas/cytology , Hybridomas/drug effects , Mice , Mice, Inbred C3H , Muromonab-CD3/pharmacology , Receptor-CD3 Complex, Antigen, T-Cell/antagonists & inhibitors , T-Lymphocytes/drug effects , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/drug effects
11.
Int J Clin Lab Res ; 21(4): 300-3, 1992.
Article in English | MEDLINE | ID: mdl-1591384

ABSTRACT

Glucocorticoid hormones, calcium ionophores and anti-CD3 monoclonal antibodies induce apoptosis in mouse thymocytes. This type of cell death, which is characterized by an extensive DNA fragmentation into oligonucleosomal subunits, occurs in the intrathymic process of negative selection, and is involved in the deletion of autoreactive T-cells during thymic maturation. A number of cytokines are able to modulate apoptosis, and interleukins, including interleukin-1, interleukin-2, and interleukin-4, play a crucial role in thymic maturation and T-cell development. We tested the effects of several cytokines on the glucocorticoid hormone-induced apoptosis of mouse thymocytes in vitro, and demonstrated that interleukin-1 alpha, interleukin-2, and interleukin-4 inhibit the apoptosis induced by dexamethasone, but that interleukin-3 and interleukin-6 exert no noteworthy effect. Dose-response experiments indicated that interleukin-4 is more potent than interleukin-1 alpha and interleukin-2 in inhibiting dexamethasone-induced apoptosis. Furthermore, interleukin-4 fully inhibited the DNA fragmentation induced by the protein kinase-C activator 12-O-tetradecanoylphorbol-13-acetate, but was ineffective against apoptosis induced by the calcium ionophore A23187. These results suggest that interleukins regulate the thymic selection process by acting as modulators of the negative selection process.


Subject(s)
Dexamethasone/pharmacology , Interleukins/physiology , T-Lymphocytes/drug effects , Thymus Gland/cytology , Animals , Calcimycin/pharmacology , Cell Death/drug effects , Cell Differentiation/drug effects , Cells, Cultured , DNA Replication , Interleukins/pharmacology , Mice , Mice, Inbred C3H , T-Lymphocytes/cytology , Tetradecanoylphorbol Acetate/pharmacology
12.
Immunology ; 86(3): 379-84, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8550074

ABSTRACT

Activation of immature thymocytes or transformed T lymphocytes via T-cell receptor (TCR)/CD3 signalling can induce programmed cell death (apoptosis). Recent data indicate that anti-CD3/TCR monoclonal antibodies (mAb) also trigger apoptosis in activated (but not resting) mature peripheral blood T lymphocytes. Here we report that triggering of resting CD4-CD8-TCR alpha beta+ and/or TCR gamma delta+ via the alternative CD2-dependent activation pathway is able to induce programmed cell death. A pair of mitogenic anti-CD2 mAb provoked a dramatic rise in [Ca2+]i that was almost entirely sustained by extracellular fluxes, and the inhibition of membrane [Ca2+/Mg2+] ATPase. The resulting endonuclease activation was able to induce DNA fragmentation, as revealed by propidium iodide staining and gel electrophoresis. Induction of apoptosis was prevented by the presence of interleukin-4 (IL-4) as well as by endonuclease inactivation with 100 microM ZnCl2, but enhanced by the contemporary block of protein kinase C. Thus it seems that in resting T lymphocytes the strong calcium signal delivered by the alternative CD2 activation pathway may act as a negative apoptotic signal in both alpha beta and gamma delta T cells with low (non-major histocompatibility complex restricted) antigenic affinity, so limiting the extension of polyclonal T-cell growth.


Subject(s)
CD2 Antigens/metabolism , Interleukin-4/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Signal Transduction/physiology , T-Lymphocytes/metabolism , Antibodies, Monoclonal , Apoptosis/immunology , CD4 Antigens , CD8 Antigens , Calcium/metabolism , Calcium-Transporting ATPases/metabolism , Cells, Cultured , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Lymphocyte Activation , Lymphocyte Count , T-Lymphocyte Subsets/immunology
13.
Nutr Cancer ; 28(1): 74-80, 1997.
Article in English | MEDLINE | ID: mdl-9200153

ABSTRACT

Apoptosis of tumor cells is an important growth-regulating event in tumor masses. In this study we have confirmed that deoxycholic acid (DCA) and the short-chain fatty acids (SCFA) butyrate and propionate induce a time- and concentration-dependent apoptosis in two human colon tumor cell lines: HT-29 and CaCO2. DCA is more potent, inducing effects at low concentration (50 microM) and after 24 hours of incubation, whereas SCFA (4 mM) requires 72-96 hours of treatment. Combining low concentrations of DCA (12.5-25 microM) with butyrate and propionate (4 mM) produces an additive effect on the percentage of apoptotic cells, as demonstrated by flow cytometry and DNA fragmentation. Protein kinase C, protein tyrosine kinase, and gene transcription/translation inhibitors do not significantly modify the rate of apoptosis, whereas the intracellular Ca2+ chelator 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid acetoxymethyl ester (BAPTA-AM) completely abolishes the DCA-induced effect without affecting the SCFA-induced apoptosis. Measurement of intracellular Ca2+ by inverted fluorescence microscopy reveals that DCA induces a rapid increase of cytosolic Ca2+ that is abolished when the cells are preincubated with BAPTA-AM, whereas ethyleneglycolbis(beta-aminoethyl ether)-N,N,N',N'-tetraacetic acid has a minimal effect. In contrast, SCFA does not modify the intracellular Ca2+ concentration. Thus the DCA-induced apoptosis is a Ca(2+)-dependent process, whereas the intracellular signals responsible for the SCFA-induced effect remain unknown. The ionophore activity of DCA could be responsible for the increased intracellular Ca2+, but other mechanisms, such as activation of phospholipase C and phosphoinositide hydrolysis, have to be considered.


Subject(s)
Apoptosis/drug effects , Colonic Neoplasms/pathology , Deoxycholic Acid/pharmacology , Fatty Acids/pharmacology , Butyrates/pharmacology , Butyric Acid , Calcium/metabolism , Chelating Agents/pharmacology , Colonic Neoplasms/metabolism , DNA Fragmentation , Deoxycholic Acid/administration & dosage , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Fatty Acids/administration & dosage , Flow Cytometry , Humans , Propionates/pharmacology , Protein Biosynthesis , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , Transcription, Genetic , Tumor Cells, Cultured
14.
Proc Natl Acad Sci U S A ; 94(12): 6216-21, 1997 Jun 10.
Article in English | MEDLINE | ID: mdl-9177197

ABSTRACT

By comparing untreated and dexamethasone-treated murine T cell hybridoma (3DO) cells by the differential display technique, we have cloned a new gene, GITR (glucocorticoid-induced tumor necrosis factor receptor family-related gene) encoding a new member of the tumor necrosis factor/nerve growth factor receptor family. GITR is a 228-amino acids type I transmembrane protein characterized by three cysteine pseudorepeats in the extracellular domain and similar to CD27 and 4-1BB in the intracellular domain. GITR resulted to be expressed in normal T lymphocytes from thymus, spleen, and lymph nodes, although no expression was detected in other nonlymphoid tissues, including brain, kidney, and liver. Furthermore, GITR expression was induced in T lymphocytes upon activation by anti-CD3 mAb, Con A, or phorbol 12-myristate 13-acetate plus Ca-ionophore treatment. The constitutive expression of a transfected GITR gene induced resistance to anti-CD3 mAb-induced apoptosis, whereas antisense GITR mRNA expression lead to increased sensitivity. The protection toward T cell receptor-induced apoptosis was specific, because other apoptotic signals (Fas triggering, dexamethasone treatment, or UV irradiation) were not modulated by GITR transfection. Thus, GITR is a new member of tumor necrosis factor/nerve growth factor receptor family involved in the regulation of T cell receptor-mediated cell death.


Subject(s)
Apoptosis , Receptors, Nerve Growth Factor/chemistry , Receptors, Nerve Growth Factor/physiology , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Tumor Necrosis Factor/physiology , T-Lymphocytes/physiology , Amino Acid Sequence , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Clone Cells , Cloning, Molecular , Dexamethasone/pharmacology , Gene Library , Glucocorticoid-Induced TNFR-Related Protein , Hybridomas , Lymph Nodes/immunology , Mice , Mice, Inbred C3H , Molecular Sequence Data , Protein Biosynthesis , Protein Sorting Signals/chemistry , Receptors, Nerve Growth Factor/biosynthesis , Receptors, Tumor Necrosis Factor/biosynthesis , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Spleen/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymus Gland/immunology , Transcription, Genetic , Transfection , Ultraviolet Rays
15.
Scand J Immunol ; 41(5): 504-8, 1995 May.
Article in English | MEDLINE | ID: mdl-7725070

ABSTRACT

The mechanisms by which glucocorticoids suppress immune responses have not yet been clearly defined. In steroid-sensitive pathological conditions, an increase in gamma delta T cells can occur in certain untreated systemic autoimmune disorders and seems to be a peristent feature in most cases of systemic lupus erythematosus (SLE). Our previously published data demonstrated that immunosuppressive therapy normalized this expanded SLE T cell subset in parallel with clinical remission of the symptoms. To establish how corticosteroid treatment determines the disappearance of peripheral blood gamma delta T lymphocytes, circulating alpha beta and gamma delta T lymphocytes from seven SLE subjects with active disease and seven healthy individuals were cultured in the presence or absence of 10(-7) M Dexamethasone (DEX). Cell suspensions were then analysed for DNA fragmentation, characteristic of apoptotic cell death, by a new cytofluorimetric method. Conventional agarose-gel electrophoresis on the same T cell populations was carried out for comparison. Regular follow-ups for 6 months revealed in vivo steroid treatment determined a dramatic fall in SLE blood gamma delta T cells, and in vitro experiments seem to indicate that DEX-triggered apoptotic signals are confined to the double negative (CD4-CD8-) gamma delta T cell subpopulation which disappears after in vivo immunosuppressive therapy. Clinical and pathological remission of some autoimmune diseases is often obtained by corticosteroids. Our results offer new insights on the mechanisms through these hormones exert their potent inhibitory activities on immune system cells postulated to play a role in the generation of autoimmune responses.


Subject(s)
Apoptosis/drug effects , Dexamethasone/pharmacology , Receptors, Antigen, T-Cell, gamma-delta/analysis , T-Lymphocytes/physiology , Adult , Cells, Cultured , Child , DNA/analysis , Female , Humans , Lupus Erythematosus, Systemic/immunology , Middle Aged
16.
Immunity ; 7(6): 803-12, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9430225

ABSTRACT

By comparing mRNA species expressed in dexamethasone (DEX)-treated and untreated murine thymocytes, we have identified a gene, glucocorticoid-induced leucine zipper (GILZ), encoding a new member of the leucine zipper family. GILZ was found expressed in normal lymphocytes from thymus, spleen, and lymph nodes, whereas low or no expression was detected in other nonlymphoid tissues, including brain, kidney, and liver. In thymocytes and peripheral T cells, GILZ gene expression is induced by DEX. Furthermore, GILZ expression selectively protects T cells from apoptosis induced by treatment with anti-CD3 monoclonal antibody but not by treatment with other apoptotic stimuli. This antiapoptotic effect correlates with inhibition of Fas and Fas ligand expression. Thus, GILZ is a candidate transcription factor involved in the regulation of apoptosis of T cells.


Subject(s)
Apoptosis , Dexamethasone/pharmacology , Leucine Zippers/immunology , Receptor-CD3 Complex, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , Base Sequence , Cell Line , DNA, Complementary , Fas Ligand Protein , Gene Expression , Leucine Zippers/genetics , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C3H , Molecular Sequence Data , Subcellular Fractions , T-Lymphocytes/drug effects , T-Lymphocytes/radiation effects , Tissue Distribution , Transfection , Ultraviolet Rays , fas Receptor/biosynthesis , fas Receptor/immunology
17.
Blood ; 94(10): 3456-67, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10552956

ABSTRACT

The Fas/FasL system mediates apoptosis in several different cell types, including T lymphocytes. Fas ligand (FasL), a 40-kD type II membrane protein also expressed in activated T cells, belongs to the tumor necrosis factor ligand family. We describe a new alternative splicing of mouse FasL, named FasL short (FasLs), cloned by reverse transcriptase-polymerase chain reaction. FasLs is encoded by part of exon 1 and part of exon 4 of FasL gene. The protein encoded by FasLs mRNA has a putative initiation code at position 756 and preserves the same reading frame as FasL, resulting in a short molecule lacking the intracellular, the transmembrane, and part of the extracellular domains. RNase protection and immunoprecipitation analysis showed that FasLs is expressed in nonactivated normal spleen cells and in hybridoma T cells and that it is upregulated upon activation by anti-CD3 monoclonal antibody (MoAb). Moreover, FasLs-transfected cells expressed soluble FasLs in the supernatant and became resistant to apoptosis induced by agonist anti-Fas MoAb. Thus, FasLs, a new alternative splicing of FasL, is involved in the regulation of Fas/FasL-mediated cell death.


Subject(s)
Membrane Glycoproteins/genetics , fas Receptor/metabolism , Alternative Splicing , Amino Acid Sequence , Animals , Apoptosis , Base Sequence , CD3 Complex/immunology , Cells, Cultured , Cloning, Molecular , DNA, Complementary/metabolism , Fas Ligand Protein , Hybridomas , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/physiology , Mice , Molecular Sequence Data , RNA, Messenger/metabolism , Spleen/cytology , Spleen/metabolism , T-Lymphocytes/pathology , T-Lymphocytes/physiology , Transfection , Up-Regulation , fas Receptor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL