Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 129
Filter
Add more filters










Publication year range
2.
Emerg Med J ; 24(5): e26, 2007 May.
Article in English | MEDLINE | ID: mdl-17452686

ABSTRACT

A man presented with recurrent syncope, weakness and fatigue. His ECG showed marked QRS widening and he had gross hyponatraemia and hypokalaemia. His medications included bendroflumethiazide (long term) and flecainide (started 2 months previously). This presentation was consistent with flacainide cardiotoxicity exacerbated by electrolyte disturbance. The syncopal episodes probably represented life-threatening arrhythmias. The ECG and symptoms resolved completely once the electrolytes were corrected. Increased cardiotoxicity with hypokalaemia is documented, but not widely recognised. Hyponatraemia-induced flecainide cardiotoxicity has not been documented. The clinical effects of flecainide are due to use-dependent block of sodium channels. There are reports that support the use of hypertonic sodium salts to reverse flecainide toxicity via antagonism at the receptor. By this rationale, hyponatraemia would lead to Flecainide toxicity. Flecainide has been shown to reduce salt absorption in animal bowel. It is possible that in combination with bendroflumethiazide it acted synergistically to produce profound electrolyte disturbance. Flecainide cardiotoxicity has a significant mortality and can present non-specifically. Thus, early recognition is essential. This case demonstrates the importance of strict electrolyte control in patients who are on flecainide. We would discourage concomitant use of flecainide and bendroflumethiazide.


Subject(s)
Anti-Arrhythmia Agents/adverse effects , Arrhythmias, Cardiac/chemically induced , Bendroflumethiazide/adverse effects , Diuretics/adverse effects , Flecainide/adverse effects , Hypokalemia/chemically induced , Hyponatremia/chemically induced , Arrhythmias, Cardiac/diagnosis , Arrhythmias, Cardiac/drug therapy , Drug Interactions , Electrocardiography , Humans , Hypertension/complications , Hypertension/drug therapy , Hypokalemia/blood , Hypokalemia/diagnosis , Hyponatremia/blood , Hyponatremia/diagnosis , Male , Middle Aged , Potassium/blood , Sodium/blood
4.
Apoptosis ; 11(6): 1049-59, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16544100

ABSTRACT

The ubiquitin Carboxyl-terminal Hydrolase-L1 gene (UCHL1) is a key enzyme in the protein degradation pathway; however, its precise role in protecting cells under stress conditions is unclear. In the present study we investigated the activity of this gene in human NT2/D1 embryonal carcinoma cells subjected to oxygen-glucose deprivation (OGD) and reoxygenation. OGD/reoxygenation cause global metabolic changes due to energy withdrawal and the subsequent generation of reactive oxygen species which initiates either a stress-adaptation-survival response or cell death, depending on the severity of the insult. A bi-phasic change in UCHL1 expression was observed by Q-PCR, Western blotting and flow cytometry. Down regulation of UCHL1 was detected immediately after OGD treatment and its expression was subsequently restored and increased 6 h after OGD treatment as well as during reoxygenation. Furthermore, flow cytometry analysis detected a lower level of UCHL1 only in apoptotic cells that had severe loss of mitochondrial membrane potential. Accordingly, down-regulation of endogenous UCHL1 by antisense cDNA in mouse N2a neuroblastoma cells increased the cell's sensitivity to OGD treatment. This down-regulation of endogenous UCHL1 led to the accumulation of p27, suggesting that UCHL1 is an essential gene to maintain cell homeostasis under normal growth and oxidative stress conditions.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Oxidative Stress/physiology , Ubiquitin Thiolesterase/genetics , Cell Line, Tumor , Cell Survival/physiology , Humans , Ubiquitin Thiolesterase/biosynthesis
5.
Cell Death Differ ; 12(11): 1368-77, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15920534

ABSTRACT

DNA fragmentation in apoptosis, especially in lymphocytic cells, is initiated at scaffold/matrix attachment regions (S/MARs) and is preceded by the degradation of nuclear proteins. The present study was performed to establish whether the same mechanism occurred in human NT2 cells subjected to oxygen and glucose deprivation (OGD). We analyzed the integrity of c-myc S/MAR containing a base-unpairing region (BUR)-like element, which we established to be a binding site of the transcription factor Sox2. An accumulation of DNA breaks in close proximity to this element and a degradation of Sox2 were observed early in the OGD-induced apoptotic response. Identification of Sox2 as a novel c-myc BUR-binding protein was achieved through yeast one-hybrid screening and the Sox2/DNA interaction was confirmed by electrophoretic mobility shift assay and immunoprecipitation with Sox2 antibody. Our data support the notion that early proteolysis of unique BUR-binding proteins might represent a universal mechanism that renders these DNA sites vulnerable to endonucleolysis.


Subject(s)
Apoptosis/physiology , DNA, Neoplasm/metabolism , DNA-Binding Proteins/metabolism , Genes, myc/physiology , HMGB Proteins/metabolism , Matrix Attachment Regions/physiology , Neurons/cytology , Transcription Factors/metabolism , Apoptosis/genetics , Base Sequence , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/metabolism , Cell Line, Tumor , DNA Damage , DNA, Neoplasm/genetics , DNA-Binding Proteins/genetics , Genes, myc/genetics , HMGB Proteins/genetics , Humans , Jurkat Cells , Matrix Attachment Regions/genetics , Neurons/metabolism , Polymerase Chain Reaction , SOXB1 Transcription Factors , Transcription Factors/genetics
6.
Cell Death Differ ; 11(6): 645-54, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15002038

ABSTRACT

DNaseY, a Ca(2+)- and Mg(2+)-dependent endonuclease, has been implicated in apoptotic DNA degradation; however, the molecular mechanisms controlling its involvement in this process have not been fully elucidated. We have obtained evidence from yeast two-hybrid screening and coimmunoprecipitation experiments that DNaseY interacted physically with actinin-alpha4 and this interaction significantly enhanced its endonuclease activity. Accordingly, simultaneous overexpression of both proteins in PC12 cells dramatically increased the rate of apoptosis in response to teniposide' VM26. However, overexpression of DNaseY alone neither triggered apoptosis nor facilitated cell death in response to VM26 or serum deprivation. Instead, the overexpression of DNaseY increased the production of single-strand DNA breaks and evoked a profound upregulation of DNA repair pathways. Taken together, our results point to a novel regulatory mechanism of DNaseY activity and offer an explanation for why cells must first cleave key DNA repair and replication proteins before the successful execution of apoptosis.


Subject(s)
Actinin/metabolism , Apoptosis/physiology , Deoxyribonuclease I/metabolism , Microfilament Proteins/metabolism , Actinin/immunology , Animals , DNA Fragmentation/physiology , Deoxyribonuclease I/immunology , Electrophoresis, Gel, Pulsed-Field , Fluorescent Antibody Technique , In Situ Nick-End Labeling , Microfilament Proteins/immunology , Rats , Two-Hybrid System Techniques
7.
Cell Death Differ ; 10(3): 278-89, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12700628

ABSTRACT

Apoptotic DNA degradation could be initiated by the accumulation of single-strand (ss) breaks in vulnerable chromatin regions, such as base unpairing regions (BURs), which might be preferentially targeted for degradation by both proteases and nucleases. We tested this hypothesis in anti-Fas-treated apoptotic Jurkat cells. Several nuclear proteins known for their association with both MARs and the nuclear matrix, that is, PARP, NuMA, lamin B and SATB1, were degraded, but the morphological rearrangement of the BUR-binding SATB1 protein was one of the earliest detected changes. Subsequently, we have identified several genes containing sequences homologous to the 25 bp BUR element of the IgH gene, a known SATB1-binding site, and examined the integrity of genomic DNA in their vicinity. Multiple ss breaks were found in close proximity to these sites relative to adjacent regions of DNA. Consistent with our prediction, the results indicated that the initiation of DNA cleavage in anti-Fas-treated Jurkat cells occurred within the BUR sites, which likely became accessible to endonucleases due to the degradation of BUR-binding proteins.


Subject(s)
DNA Damage , Polymerase Chain Reaction/methods , Base Sequence , Binding Sites , Blotting, Western , Cell Nucleus/metabolism , Cell Survival , Chromatin/metabolism , DNA Fragmentation , DNA, Single-Stranded , Electrophoresis, Gel, Two-Dimensional , Humans , In Situ Nick-End Labeling , Jurkat Cells , Kinetics , Matrix Attachment Region Binding Proteins/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Models, Genetic , Molecular Sequence Data , Nuclear Proteins/metabolism , Time Factors
10.
Curr Opin Immunol ; 13(2): 121-8, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11228401

ABSTRACT

A selection of interesting papers that were published in the two months before our press date in major journals most likely to report significant results in immunology.


Subject(s)
Allergy and Immunology , Animals , Autoimmunity , Humans , Immunogenetics , Immunotherapy , Neoplasms/immunology , Transplantation Immunology
11.
Cancer Res ; 61(5): 2047-54, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11280765

ABSTRACT

HLA-A2+ melanoma patients develop naturally a strong CD8+ T cell response to a self-peptide derived from Melan-A. Here, we have used HLA-A2/peptide tetramers to isolate Melan-A-specific T cells from tumor-infiltrated lymph nodes of two HLA-A2+ melanoma patients and analyzed their TCR beta chain V segment and complementarity determining region 3 length and sequence. We found a broad diversity in Melan-A-specific immune T-cell receptor (TCR) repertoires in terms of both TCR beta chain variable gene segment usage and clonal composition. In addition, immune TCR repertoires selected in the patients were not overlapping. In contrast to previously characterized CD8+ T-cell responses to viral infections, this study provides evidence against usage of highly restricted TCR repertoire in the natural response to a self-differentiation tumor antigen.


Subject(s)
Antigens, Neoplasm/immunology , Melanoma/immunology , Neoplasm Proteins/immunology , Peptide Fragments/immunology , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Base Sequence , Complementarity Determining Regions/immunology , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/immunology , Humans , Immunodominant Epitopes/immunology , Immunoglobulin Variable Region/immunology , Lymph Nodes/immunology , Lymph Nodes/pathology , MART-1 Antigen , Molecular Sequence Data , Receptors, Antigen, T-Cell, alpha-beta/immunology
12.
J Immunol ; 165(6): 3128-35, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10975826

ABSTRACT

Effective antitumor immune responses against cerebral malignancies have been demonstrated in several models, but precise cellular function of specific effector cells is poorly understood. We have explored this topic by analyzing the MHC class I-restricted T cell response elicited after implantation of HLA-CW3-transfected P815 mastocytoma cells (P815-CW3) in syngeneic mice. In this model, tumor-specific CTLs use a distinctive repertoire of TCRs that allows ex vivo assessment of the response by immunophenotyping and TCR spectratyping. Thus, for the first time in a brain tumor model, we are able to directly visualize ex vivo CTLs specific for a tumor-expressed Ag. Tumor-specific CTLs are detected in the CNS after intracerebral implantation of P815-CW3, together with other inflammatory cells. Moreover, despite observations in other models suggesting that CTLs infiltrating the brain may be functionally compromised and highly dependent upon CD4 T cells, in this syngeneic P815-CW3 model, intracerebral tumors were efficiently rejected, whether or not CD4 T cells were present. This observation correlated with potent ex vivo cytotoxicity of brain-infiltrating CTLs, specific for the immunodominant epitope CW3170-179 expressed on P815-CW3 tumor cells.


Subject(s)
Brain Neoplasms/immunology , CD4-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Graft Rejection/immunology , Lymphopenia/immunology , Sarcoma, Experimental/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Brain Neoplasms/pathology , Brain Neoplasms/prevention & control , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Movement/immunology , Cell Separation , Clone Cells , Cytotoxicity, Immunologic/genetics , Female , Graft Rejection/genetics , Graft Rejection/pathology , HLA-C Antigens/genetics , HLA-C Antigens/immunology , Humans , Injections, Intraventricular , Lymphocyte Activation/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Inbred DBA , Neoplasm Transplantation , Receptors, Antigen, T-Cell/biosynthesis , Sarcoma, Experimental/pathology , Sarcoma, Experimental/prevention & control , Transfection , Tumor Cells, Cultured/transplantation , Weight Loss/immunology
13.
Glia ; 32(1): 102-7, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10975915

ABSTRACT

Astrocytes exert many active roles in brain homeostasis, potentially including the regulation of immune reactions. They possess a substantial aptitude for plasticity and, indeed, functional and phenotypic changes are frequently encountered in reactive gliosis observed in brain injuries. The significance of reactive astrocytes is still poorly defined, but it is clear that these cells are an important source of cytokines in inflamed brain. How tumor necrosis factor (TNF) and TNF-receptor family members contribute to this reaction is an interesting issue that is currently being explored. It was previously shown that reactive astrocytes express high levels of Fas (CD95) and respond to Fas ligand (CD95L) by apoptosis or IL-8 production. TWEAK (Apo-3 ligand) is a recently identified member of the TNF family that is produced mainly by leukocytes that can infiltrate the inflamed brain and thus influence astrocyte behavior. Here we show that human astrocytes derived from different regions of the brain specifically bind TWEAK and are totally resistant to TWEAK mediated apoptosis. In addition, high amounts of IL-8 and IL-6 were secreted by astrocytes after TWEAK exposure. Finally, expression of cell surface molecules involved in the propagation and/or maintenance of brain inflammation was determined. TWEAK significantly increased ICAM-1 expression on astrocytes, whereas no modification was detected in the expression of Fas, TNFRI, B7-1, or MHC molecules. In conclusion, the proinflammatory effects induced by TWEAK on astrocytes in culture recapitulate many characteristics of reactive astrocytes observed in vivo, suggesting that TWEAK could play a significant role in brain inflammation.


Subject(s)
Astrocytes/drug effects , Astrocytes/metabolism , Brain/physiopathology , Carrier Proteins/metabolism , Carrier Proteins/pharmacology , Encephalitis/metabolism , Encephalitis/physiopathology , Apoptosis Regulatory Proteins , Astrocytes/cytology , Brain/drug effects , Brain/pathology , Cytokine TWEAK , Encephalitis/pathology , Fetus , Gliosis/metabolism , Gliosis/pathology , Gliosis/physiopathology , Humans , Intercellular Adhesion Molecule-1/drug effects , Intercellular Adhesion Molecule-1/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Rhombencephalon/cytology , Rhombencephalon/drug effects , Rhombencephalon/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factors
14.
Cancer Lett ; 153(1-2): 13-23, 2000 May 29.
Article in English | MEDLINE | ID: mdl-10779625

ABSTRACT

This study examined changes in protein kinase C (PKC) isoforms in rodent fibroblasts (rat F111 and mouse NIH3T3), transformed by the polyoma virus middle T antigen (mT) and undergoing apoptosis in response to teniposide (VM26). The mT-transformed cells up-regulated PKC delta and down-regulated both PKC epsilon and PKC lambda expression, and were more sensitive to the drug than their non-transformed counterparts. The drug treatment further lowered the expression of PKC epsilon, triggered nuclear translocation of PKC delta and its site-specific proteolysis, consistent with the notion that changes in specific PKC isoforms play a role not only in the neoplastic transformation of fibroblasts, but also in their apoptotic response.


Subject(s)
Cell Transformation, Neoplastic , Isoenzymes/metabolism , Protein Kinase C/metabolism , Teniposide/pharmacology , 3T3 Cells , Animals , Antineoplastic Agents/pharmacology , Cells, Cultured , Gene Expression , Mice , Protein Kinase C-delta , Protein Kinase C-epsilon , Rats
15.
Cell Immunol ; 206(1): 51-8, 2000 Nov 25.
Article in English | MEDLINE | ID: mdl-11161437

ABSTRACT

Elimination of clonally expanded peripheral CD8 T cells was thought to involve apoptosis induction mediated principally by TNF, but recently Fas (CD95/APO-1) has been shown to play a role in certain responses. Here we study Fas expression and sensitivity to its ligation on murine CD8 cells specific for the CW3 antigen expressed by transfected P815 cells. Fas was progressively downregulated after successive in vitro restimulations of antigen-specific CD8 cells, until clones became Fas negative and totally resistant to the effects of recombinant Fas ligand. In contrast, Fas expression by in vivo restimulated antigen-specific cells did not diminish. Loss of Fas expression in vitro was not totally irreversible, since it could be reinduced by inhibition of DNA methylation. Understanding how Fas may be differentially regulated in vivo and in vitro is an important issue for the optimal manipulation of T cells for adoptive immunotherapy protocols.


Subject(s)
Azacitidine/analogs & derivatives , DNA Methylation/drug effects , Gene Expression Regulation/physiology , T-Lymphocytes, Cytotoxic/drug effects , fas Receptor/physiology , Animals , Apoptosis/drug effects , Azacitidine/pharmacology , Clone Cells/drug effects , Clone Cells/immunology , Clone Cells/metabolism , Decitabine , Enzyme Inhibitors/pharmacology , Fas Ligand Protein , Female , Flow Cytometry , HLA-C Antigens/genetics , HLA-C Antigens/immunology , Immunotherapy, Adoptive , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , Mice, Inbred DBA , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Transfection , Tumor Cells, Cultured , fas Receptor/biosynthesis , fas Receptor/genetics
16.
Apoptosis ; 5(3): 265-75, 2000 Jun.
Article in English | MEDLINE | ID: mdl-11225848

ABSTRACT

The objective of this study was to establish whether apoptosis in 5123tc rat hepatoma cells required the caspase-3 dependent pathway. Apoptosis was induced by either growth factor deprivation or treatment with a topoisomerase II inhibitor, VM26, in the absence or presence of caspase inhibitors (DEVD-fmk, z-VAD-fmk and BAF). The results indicated that, although these inhibitors at 10 microM concentration completely blocked caspase-3 activity, they had no effect on either the rate of cell death or on any other apoptotic features, e.g., chromatin condensation, DNA fragmentation, protein cleavage, suggesting that caspase-3 was not required to mediate nuclear destruction in these hepatoma cells. At higher concentrations, up to 100 microM, z-VAD-fmk and BAF, but not DEVD-fmk, did block apoptosis, however, they also caused cell swelling and membrane permeabilization, which are the hallmarks of necrotic cell death. Clearly, high concentrations of these inhibitors must have interfered non-specifically with other metabolic pathways, e.g., z-VAD-fmk at a high concentration blocked protein phosphorylation, and caused cell death by a different mechanism.


Subject(s)
Apoptosis , Caspases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Carcinoma, Hepatocellular , Caspase 3 , Caspase Inhibitors , Cell Nucleus/metabolism , Culture Media, Serum-Free , DNA Fragmentation , Electrophoresis, Gel, Pulsed-Field , Electrophoresis, Gel, Two-Dimensional , Enzyme Inhibitors/pharmacology , Humans , Lamins , Microscopy, Fluorescence , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases , Proteins/metabolism , Rats , Teniposide/pharmacology , Tumor Cells, Cultured
17.
Mol Immunol ; 36(11-12): 745-53, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10593513

ABSTRACT

The development of T cell effector and memory responses against foreign antigens (Ags) involves the activation, differentiation and proliferation of naive T cells expressing distinct Ag-specific TCRs. Understanding the complexity of Ag-selected TCR repertoires in individual responders in terms of the sequences selected and their relative frequencies may provide indications about how a repertoire is established and suggest ways to influence the outcome of an immune response. Most methods of repertoire analysis are unsuitable for calculating the relative in vivo frequencies of Ag-specific clones (expressing distinct TCRs) selected during an immune response, whereas sequence data obtained by single-cell PCR analysis directly reflect cell frequencies if a sufficiently large number of cells is sampled. Using a CD8 T cell response in normal mice in which Ag-selected cells are identified by cell surface phenotype and rearranged TCRBV sequences are determined by PCR amplification of genomic DNA directly from single cells, we have analyzed a large number (>200 per animal) of structurally-related Ag-specific TCRs to calculate the frequencies of distinct TCRs selected by individual mice. We found that each responder selects a unique Ag-specific TCR repertoire in which the various TCRBV sequences are present in a wide range of frequencies. However, the overall distribution of sequences is quite similar for different responder animals. Moreover, an individual's selected TCR repertoire is uniformly represented among Ag-specific CD8 cells circulating in the blood or localized in the spleen or liver. Relatively few sequences make up the bulk of the repertoire and account for the oligoclonality observed in earlier studies. We discuss various models that could account for this skewed distribution of an Ag-selected TCR repertoire.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/genetics , Animals , Cell Line , Female , Gene Rearrangement, T-Lymphocyte , HLA-C Antigens/genetics , HLA-C Antigens/immunology , Humans , Immunization , Liver/cytology , Liver/immunology , Mice , Mice, Inbred DBA , Polymerase Chain Reaction , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Spleen/cytology , Spleen/immunology , Transfection
18.
Int Immunol ; 11(8): 1337-50, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10421791

ABSTRACT

Anaplastic astrocytoma and glioblastoma are frequent and malignant brain tumors that are infiltrated by T lymphocytes. Whether these cells result from non-specific inflammation following blood-brain barrier disruption or an antigen-driven specific immune response is unknown. In this study, an in-depth characterization of TCR diversity in tumor and blood RNA biopsies was performed in a series of 16 patients with malignant astrocytoma. Whilst there was no obvious restriction of the AV and BV gene segment usage, complementarity-determining region 3 size analysis and sequencing of amplified TCR transcripts revealed multiple T cell oligoclonal expansions in all astrocytomas analyzed. Unique T cell clones were present in different adjacent areas of a given tumor, but never detected in the blood. Quantification of the number of TCR clonal transcripts per microg of tumor RNA indicated that certain T cell clonal expansions may represent at least 300 cells/10(6) tumor cells. Furthermore, we demonstrated that the in vivo expanded clones were almost exclusively confined to the CD8(+) subset. Overall, these data suggest that spontaneous antigen-driven immune responses may be elicited against human astrocytoma despite the immunosuppressive microenvironment generated by the brain and the tumor itself. However, the ultimate failure of the immune system to control tumor growth could be the consequence of a deficient CD4 T(h) component of the response. This observation could have important consequences for the development of immunotherapies for astrocytoma patients.


Subject(s)
Astrocytoma/immunology , Brain Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Complementarity Determining Regions , Lymphocytes, Tumor-Infiltrating/immunology , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , Clone Cells , Female , Genes, T-Cell Receptor , Humans , Immunoglobulin Variable Region/genetics , Lymphocytes, Tumor-Infiltrating/cytology , Male , Middle Aged , Molecular Sequence Data , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
19.
Methods ; 17(4): 329-38, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10196104

ABSTRACT

DNA degradation during apoptosis is endonuclease mediated and proceeds through an ordered series of stages commencing with the production of large DNA pieces of 300 kb which are then degraded to fragments of 50 kb. The 50-kb fragments are further degraded, in some but not all cells, to smaller pieces (10-40 kb) releasing the small oligonucleosome fragments that are detected as a characteristic DNA ladder on conventional agarose gels. Methodology is presented for the detection of both DNA ladders and the initial stages of DNA fragmentation using pulsed-field gel electrophoresis. We have developed electrophoresis conditions that resolve large fragments of DNA and also retain the smaller fragments on the same gel. Methods for the detection of endonuclease activities responsible for the cleavage of DNA during apoptosis are also presented.


Subject(s)
Apoptosis , DNA Fragmentation/genetics , Endonucleases/analysis , Animals , DNA/analysis , Electrophoresis, Agar Gel/methods , Electrophoresis, Gel, Pulsed-Field/methods , Humans , Image Processing, Computer-Assisted , Mice , Protease Inhibitors/pharmacology , Tumor Cells, Cultured
20.
J Immunol ; 162(4): 2326-33, 1999 Feb 15.
Article in English | MEDLINE | ID: mdl-9973511

ABSTRACT

Astrocytes are a major cellular component of the brain that are capable of intense proliferation and metabolic activity during diverse inflammatory brain diseases (such as multiple sclerosis, Alzheimer's dementia, tumor, HIV encephalitis, or prion disease). In this biological process, called reactive gliosis, astrocyte apoptosis is frequently observed and could be an important mechanism of regulation. However, the factors responsible for apoptosis in human astrocytes are poorly defined. Here, we report that short term cultured astrocytes derived from different brain regions express significant levels of CD95 at their surface. Only late passage astrocytes are sensitive to CD95 ligation using either CD95 mAb or recombinant CD95 ligand. Blocking experiments using caspase inhibitors with different specificities (DEVD-CHO, z-VAD-fmk, and YVAD-cmk), an enzymatic activity assay, and immunoblotting show that CPP32/caspase-3 play a prominent role in CD95-induced astrocyte death. In contrast, early passage astrocytes are totally resistant to death, but a significant increase in astrocytic IL-8 secretion (p < 0.001, by Wilcoxon's test for paired samples) is observed after CD95 triggering. Production of IL-8 contributes to the resistance of astrocytes to CD95 ligation. Furthermore, in the presence of IFN-gamma, resistant astrocytes became sensitive to CD95-mediated death. These data suggest that microenvironmental factors can influence the consequences of CD95 ligation on astrocytes. Therefore, we propose that CD95 expressed by human astrocytes plays a pivotal role in the regulation of astrocyte life and death and may be a key factor in inflammatory processes in the brain, such as reactive gliosis.


Subject(s)
Apoptosis/immunology , Astrocytes/immunology , Astrocytes/pathology , Encephalitis/immunology , Receptors, Tumor Necrosis Factor/physiology , fas Receptor/metabolism , Adjuvants, Immunologic/pharmacology , Astrocytes/enzymology , Astrocytes/metabolism , Caspase 3 , Caspases/metabolism , Cell Death/immunology , Cells, Cultured , Encephalitis/etiology , Encephalitis/pathology , Enzyme Activation/immunology , Humans , Immunity, Innate , Interferon-gamma/pharmacology , Interleukin-8/metabolism , Ligands , Tumor Cells, Cultured , fas Receptor/biosynthesis , fas Receptor/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...