Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 184
Filter
Add more filters

Publication year range
1.
Minerva Endocrinol ; 35(2): 109-25, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20595939

ABSTRACT

Modifications of the hypothalamo-pituitary-adrenal axis and associated changes in circulating levels of glucocorticoids form a key component of the response of an organism to stressful challenges. Increased levels of glucocorticoids promote gluconeogenesis, mobilization of amino acids, and stimulation of fat breakdown to maintain circulating levels of glucose necessary to mount a stress response. In addition to profound changes in the physiology and function of multiple tissues, stress and elevated glucocorticoids can also inhibit reproduction, a logical effect for the survival of self. Precise levels of glucocorticoids are required for proper gonadal function; where the balance is disrupted, so is fertility. Glucocorticoids affect gonadal function at multiple levels in hypothalamo-pituitary-gonadal axis: 1) the hypothalamus (to decrease the synthesis and release of gonadotropin-releasing hormone [GnRH]); 2) the pituitary gland (to inhibit the synthesis and release of luteinizing hormone [LH] and follicle stimulating hormone [FSH]); 3) the testis/ovary (to modulate steroidogenesis and/or gametogenesis directly). Furthermore, maternal exposure to prenatal stress or exogenous glucocorticoids can lead to permanent modification of hypothalamo-pituitary-adrenal function and stress-related behaviors in offspring. Glucocorticoids are vital to many aspects of normal brain development, but fetal exposure to superabundant glucocorticoids can result in life-long effects on neuroendocrine function. This review focuses on the molecular mechanisms believed to mediate glucocorticoid inhibition of reproductive functions and the anatomical sites at which these effects take place.


Subject(s)
Fertility , Glucocorticoids/metabolism , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Stress, Physiological , Stress, Psychological/metabolism , Female , Follicle Stimulating Hormone/biosynthesis , Glucocorticoids/adverse effects , Gonadotropin-Releasing Hormone/biosynthesis , Humans , Male , Ovary/metabolism , Pregnancy , Testis/metabolism
2.
Trends Cell Biol ; 5(1): 21-6, 1995 Jan.
Article in English | MEDLINE | ID: mdl-14731429

ABSTRACT

The formation of distinct DNA fragments of oligonucleosomal size (180-200 bp lengths) is a biochemical hallmark of apoptosis in many cells. Recent observations also suggest large DNA fragments and even single-strand cleavage events occur during cell death. These observations have raised many questions. What are the types of DNA cleavage observed during apoptosis? What are the nucleases involved? And what is the role of these nucleolytic events in apoptosis?

3.
Cell Death Differ ; 14(4): 840-50, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17170751

ABSTRACT

Our laboratory has shown that glucocorticoids can inhibit apoptosis in rat hepatoma cells; however, the mechanisms are incompletely understood. To address this issue we sought to determine if glucocorticoid inhibition is effective when death is induced by stimuli that more selectively activate either the intrinsic (UV-C) or extrinsic (FasL) apoptotic pathways. Using flow cytometric analysis, we show that pretreatment of HTC cells with dexamethasone (Dex) inhibits UV-C- but not FasL-induced apoptosis. This inhibition requires Dex pretreatment and can be abrogated by the glucocorticoid antagonist RU486 indicating glucocorticoid receptor-mediated action. Dex increases anti-apoptotic Bcl-x(L) at both mRNA and protein levels. The Bcl-x(L) protein level remains elevated even after apoptosis induction with either UV-C or FasL although only UV-C-induced cell death is inhibited. Repression of Bcl-x(L) protein with siRNA abrogates the anti-apoptotic effect of glucocorticoids. Together these data provide direct evidence that Bcl-x(L) mediates glucocorticoid inhibition of UV-C induced apoptosis.


Subject(s)
Apoptosis/drug effects , Carcinoma, Hepatocellular/metabolism , Dexamethasone/pharmacology , Fas Ligand Protein/metabolism , Glucocorticoids/pharmacology , bcl-X Protein/metabolism , Animals , Carcinoma, Hepatocellular/drug therapy , Fas Ligand Protein/drug effects , Immunologic Factors , RNA, Small Interfering , Rats , Tumor Cells, Cultured , Ultraviolet Rays , fas Receptor/metabolism
5.
Mol Cell Biol ; 15(2): 943-53, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7823959

ABSTRACT

Glucocorticoids are potent immunosuppressants which work in part by inhibiting cytokine gene transcription. We show here that NF-kappa B, an important regulator of numerous cytokine genes, is functionally inhibited by the synthetic glucocorticoid dexamethasone (DEX). In transfection experiments, DEX treatment in the presence of cotransfected glucocorticoid receptor (GR) inhibits NF-kappa B p65-mediated gene expression and p65 inhibits GR activation of a glucocorticoid response element. Evidence is presented for a direct interaction between GR and the NF-kappa B subunits p65 and p50. In addition, we demonstrate that the ability of p65, p50, and c-rel subunits to bind DNA is inhibited by DEX and GR. In HeLa cells, DEX activation of endogenous GR is sufficient to block tumor necrosis factor alpha or interleukin 1 activation of NF-kappa B at the levels of both DNA binding and transcriptional activation. DEX treatment of HeLa cells also results in a significant loss of nuclear p65 and a slight increase in cytoplasmic p65. These data reveal a second mechanism by which NF-kappa B activity may be regulated by DEX. We also report that RU486 treatment of wild-type GR and DEX treatment of a transactivation mutant of GR each can significantly inhibit p65 activity. In addition, we found that the zinc finger domain of GR is necessary for the inhibition of p65. This domain is also required for GR repression of AP-1. Surprisingly, while both AP-1 and NF-kappa B can be inhibited by activated GR, synergistic NF-kappa B/AP-1 activity is largely unaffected. These data suggest that NF-kappa B, AP-1, and GR interact in a complex regulatory network to modulate gene expression and that cross-coupling of NF-kappa B and GR plays an important role in glucocorticoid-mediated repression of cytokine transcription.


Subject(s)
Dexamethasone/pharmacology , Gene Expression , NF-kappa B/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Cell Line , Chloramphenicol O-Acetyltransferase/metabolism , Chlorocebus aethiops , Drug Synergism , Gene Expression/drug effects , Glutathione Transferase/metabolism , HeLa Cells , Humans , Interleukin-1/pharmacology , Kinetics , Macromolecular Substances , Mifepristone/pharmacology , NF-kappa B/antagonists & inhibitors , NF-kappa B/drug effects , NF-kappa B p50 Subunit , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Transcription Factor AP-1/metabolism , Transcription Factor RelA , Transfection , Tumor Necrosis Factor-alpha/pharmacology
6.
Cancer Res ; 41(7): 2687-91, 1981 Jul.
Article in English | MEDLINE | ID: mdl-7248937

ABSTRACT

Growth of HeLa S3 cells in monolayer cultures of Joklik's minimum essential medium, 5% fetal calf serum, and 2 mM glutamine in the presence of 10(-6) M dexamethasone results in an approximately 70% inhibition of exogenously added [3H]thymidine incorporation into DNA. In marked contrast, dexamethasone does not alter HeLa S3 cell growth rate under these conditions. The half-maximal inhibitory concentration of dexamethasone is approximately 10(-9) M which correlates well with the dissociation constant of the nuclear glucocorticoid receptor at 37 degrees. Only active glucocorticoids, e.g., dexamethasone and cortisol, inhibit [3H]thymidine incorporation into DNA. 17 beta-Estradiol, 5 alpha-dihydrotestosterone, progesterone, and cortisone are ineffective. A measurable effect of dexamethasone at 10(-8) M occurs within 3 to 4 hr after hormone administration. The presence of transcriptional and translational inhibitors during exposure of the HeLa S3 cells to glucocorticoids blocks the accumulation of the hormone effect. Dexamethasone has little or no effect on uridine and leucine incorporation into RNA and protein, respectively, under these conditions. These results demonstrate that the incorporation of a DNA precursor is regulated by glucocorticoid hormones in HeLa S3 cells. This effect is most likely mediated via an alteration in the thymidine precursor pool specific activity.


Subject(s)
DNA/biosynthesis , Glucocorticoids/pharmacology , Thymidine/metabolism , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Depression, Chemical , Dexamethasone/antagonists & inhibitors , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , HeLa Cells , Humans , Hydrocortisone/pharmacology , Time Factors
7.
Cell Death Differ ; 23(5): 889-902, 2016 May.
Article in English | MEDLINE | ID: mdl-26658018

ABSTRACT

We developed a model system to investigate apoptotic resistance in T cells using osmotic stress (OS) to drive selection of death-resistant cells. Exposure of S49 (Neo) T cells to multiple rounds of OS followed by recovery of surviving cells resulted in the selection of a population of T cells (S49 (OS 4-25)) that failed to die in response to a variety of intrinsic apoptotic stimuli including acute OS, but remained sensitive to extrinsic apoptotic initiators. Genome-wide microarray analysis comparing the S49 (OS 4-25) with the parent S49 (Neo) cells revealed over 8500 differentially regulated genes, with almost 90% of those identified being repressed. Surprisingly, our data revealed that apoptotic resistance is not associated with expected changes in pro- or antiapoptotic Bcl-2 family member genes. Rather, these cells lack several characteristics associated with the initial signaling or activation of the intrinsic apoptosis pathway, including failure to increase mitochondrial-derived reactive oxygen species, failure to increase intracellular calcium, failure to deplete glutathione, failure to release cytochrome c from the mitochondria, along with a lack of induced caspase activity. The S49 (OS 4-25) cells exhibit metabolic characteristics indicative of the Warburg effect, and, despite numerous changes in mitochondria gene expression, the mitochondria have a normal metabolic capacity. Interestingly, the S49 (OS 4-25) cells have developed a complete dependence on glucose for survival, and glucose withdrawal results in cell death with many of the essential characteristics of apoptosis. Furthermore, we show that other dietary sugars such as galactose support the viability of the S49 (OS 4-25) cells in the absence of glucose; however, this carbon source sensitizes these cells to die. Our findings suggest that carbon substrate reprogramming for energy production in the S49 (OS 4-25) cells results in stimulus-specific recognition defects in the activation of intrinsic apoptotic pathways.


Subject(s)
Apoptosis , Carbon/metabolism , T-Lymphocytes/pathology , Animals , Apoptosis/genetics , Cell Survival , Mice , T-Lymphocytes/metabolism , Tumor Cells, Cultured
8.
Oncogene ; 35(48): 6189-6202, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27157613

ABSTRACT

ErbB-2 amplification/overexpression accounts for an aggressive breast cancer (BC) subtype (ErbB-2-positive). Enhanced ErbB-2 expression was also found in gastric cancer (GC) and has been correlated with poor clinical outcome. The ErbB-2-targeted therapies trastuzumab (TZ), a monoclonal antibody, and lapatinib, a tyrosine kinase inhibitor, have proved highly beneficial. However, resistance to such therapies remains a major clinical challenge. We here revealed a novel mechanism underlying the antiproliferative effects of both agents in ErbB-2-positive BC and GC. TZ and lapatinib ability to block extracellular signal-regulated kinases 1/2 and phosphatidylinositol-3 kinase (PI3K)/AKT in sensitive cells inhibits c-Myc activation, which results in upregulation of miR-16. Forced expression of miR-16 inhibited in vitro proliferation in BC and GC cells, both sensitive and resistant to TZ and lapatinib, as well as in a preclinical BC model resistant to these agents. This reveals miR-16 role as tumor suppressor in ErbB-2-positive BC and GC. Using genome-wide expression studies and miRNA target prediction algorithms, we identified cyclin J and far upstream element-binding protein 1 (FUBP1) as novel miR-16 targets, which mediate miR-16 antiproliferative effects. Supporting the clinical relevance of our results, we found that high levels of miR-16 and low or null FUBP1 expression correlate with TZ response in ErbB-2-positive primary BCs. These findings highlight a potential role of miR-16 and FUBP1 as biomarkers of sensitivity to TZ therapy. Furthermore, we revealed miR-16 as an innovative therapeutic agent for TZ- and lapatinib-resistant ErbB-2-positive BC and GC.


Subject(s)
Breast Neoplasms/genetics , Cyclins/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , MicroRNAs/genetics , Quinazolines/pharmacology , Stomach Neoplasms/genetics , Trastuzumab/pharmacology , 3' Untranslated Regions , Animals , Antineoplastic Agents/pharmacology , Binding Sites , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Female , Genes, Tumor Suppressor , Humans , Lapatinib , Male , Mice , Models, Biological , Promoter Regions, Genetic , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , RNA-Binding Proteins , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism
9.
Biochim Biophys Acta ; 800(3): 258-68, 1984 Aug 21.
Article in English | MEDLINE | ID: mdl-6466704

ABSTRACT

Previous studies have demonstrated that the vitamin pyridoxal phosphate can alter the physicochemical properties of glucocorticoid receptors. We now report the localization of a pyridoxal phosphate binding site within the mero-receptor domain of this glucocorticoid receptor. Mero-glucocorticoid receptors that are generated by trypsin (10 micrograms/ml) or chymotrypsin (100 micrograms/ml) digestion of intact receptors sediment as 2.6 S species on 5-20% sucrose gradients in the presence or absence of pyridoxal phosphate. Mero-glucocorticoid receptors prepared by exogenous proteinases are hydrophobic and show no affinity for DEAE Bio-Gel A. Treating either trypsin-generated or chymotrypsin-generated mero-receptors with pyridoxal phosphate rapidly converts the proteins (60 and 35%, respectively) into forms that bind to DEAE Bio-Gel A. Induction of DEAE binding is specific to pyridoxal phosphate, for treating mero-receptors with pyridoxal, pyridoxamine or pyridoxine phosphate is ineffective. Furthermore, DEAE binding cannot be induced by adding other pyridoxal phosphate-treated cytosols to untreated mero-receptors. High-resolution polyacrylamide gel isoelectric focussing studies indicated that treating mero-receptor generated by either proteinase with pyridoxal phosphate shifted the isoelectric points of both to lower pH values. The conversion of the mero-receptor to a more acidic form also occurred when the intact glucocorticoid receptor was treated with the vitamin prior to proteolysis. These studies localize at least one pyridoxal phosphate binding site on the mero-receptor domain of the rat thymocyte glucocorticoid receptor.


Subject(s)
Pyridoxal Phosphate/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Steroid/metabolism , Animals , Binding Sites , Chymotrypsin , Isoelectric Point , Male , Peptide Fragments , Protein Conformation , Rats , Solubility , Thymus Gland , Trypsin
10.
Biochim Biophys Acta ; 543(4): 545-55, 1978 Nov 01.
Article in English | MEDLINE | ID: mdl-718988

ABSTRACT

Binding of dexamethasone . receptors with isolated nuclei, DNA-cellulose and cellulose has been compared with respect to dependence on salt concentration and resistance to KCl extraction and DNAse I digestion. A solution of cytoplasmic dexamethasone . receptor complexes was prepared by the incubation of rat thymus cells with steroid at 3 degrees C and breaking the cells by hypotonic lysis. Activation of the complexes was accomplished by warming the solution at 25 degrees C for 15 min. Activation significantly increased the ability of dexamethasone . receptors to bind to nuclei and DNA-cellulose but not to cellulose. Dexamethasone-receptor complexes bound to nuclei at 3 degrees C are completely resistant to extraction with 0.1 M KCl, 76% resistant to 0.2 M KCl and 20% resistant to 0.4 M KCl. Dexamethasone . receptors bound to DNA-cellulose are 45% resistant to extraction with 0.1 M and 0.2 M KCl and 29% resistant to 0.4 M KCl extraction. Cellulose-bound dexamethasone . receptors are not resistant to any of these extractions. DNAase I treatment releases 60% of the dexamethasone . receptors bound to DNA-cellulose but only 13% of those bound to nuclei, though at least 60% of the nuclear DNA is solubilized. The presence of 0.15 M KCl decreases binding of activated dexamethasone . receptors to nuclei by 73% but to DNA-cellulose by only 17%. Pretreatment of nuclei with 0.1--0.4 M KCl reduces their capacity to bind activated dexamethasone . receptors by 90% whereas similar treatment reduces the capacity of DNA-cellulose to bind dexamethasone . receptors by only 29%. Nuclei extracted with 0.1 M KCl appear to have a limited capacity to accept dexamethasone . receptors. These studies demonstrate that binding of dexamethasone . receptors to nuclei and DNA-cellulose differs by (a) the higher resistance of nuclear complexes to KCl and DNAase I treatment; (b) the much greater sensitivity of nuclei to KCl treatment.


Subject(s)
Cell Nucleus/metabolism , DNA/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Steroid/metabolism , Cell-Free System , Cellulose , Deoxyribonucleases/metabolism , Dexamethasone/metabolism , Kinetics , Protein Binding , Salts
11.
Biochim Biophys Acta ; 630(3): 375-85, 1980 Jul 03.
Article in English | MEDLINE | ID: mdl-6249382

ABSTRACT

This study analyzes the sensitivity of nuclear bound glucocorticoid receptors to solubilization from nuclei by DNAase I and DNAase II. Thymocytes were incubated with 10(-8) M [3H]dexamethasone, [3H]cortisol or [3H]triamcinolone acetonide, without or with 10(-6) M unlabelled dexamethasone, for 30 min at 37 degrees C and nuclei from these cells were digested with either DNAase I and DNAase II. DNAase I for 2 h at 3 degrees C leads to solubilization of 60% of the nuclear DNA and release of 10--20% triamcinolone acetonide-receptor, 30--40% dexamethasone-receptor and 85--90% cortisol-receptor. DNAase II at the same enzymatic concentration solubilizes only 10--20% of the nuclear DNA, but releases 40--50% triamcinolone-receptor, 60--70% dexamethasone-receptor and 100% cortisol-receptor. Release of nuclear bound dexamethasone-receptor by DNAase I parallels the solubilization of DNA, reaching maximum values by 2 h at 3 degrees C, whereas maximal release by DNAase II is obtained within 45 min when DNA solubilization is not complete. When nuclei initially extracted with DNAase I are re-extracted with DNAase II, greater than 65% of the DNAase I residual dexamethasone-receptors are solubilized, whereas DNAase I is ineffective in solubilizing DNAase II residual dexamethasone-receptors. DNAase I solubilizes only 30% of the 0.4 M KCl residual dexamethasone-receptor whereas DNAase II digests over 90% of this fraction. DNAase I extracts of nuclear dexamethasone-receptor chromatograph on G-100 Sephadex as a single radioactive peak just after the void volume, whereas DNAase II extracts of nuclear dexamethasone-receptor chromatograph as two peaks of radioactivity, one which is similar to the DNAase I solubilized receptor and a second broad peak of macromolecular bound radioactivity which is smaller in size.


Subject(s)
Deoxyribonucleases/metabolism , Endodeoxyribonucleases , Endonucleases/metabolism , Receptors, Glucocorticoid/physiology , Receptors, Steroid/physiology , Animals , Cell Nucleus/enzymology , Deoxyribonuclease I , Dexamethasone/metabolism , Hydrocortisone/metabolism , Male , Rats , Solubility , Thymus Gland/cytology , Triamcinolone Acetonide/metabolism
12.
Biochim Biophys Acta ; 678(1): 18-26, 1981 Nov 18.
Article in English | MEDLINE | ID: mdl-7030406

ABSTRACT

A quantitative assay employing binding of [3H] diisopropylfluorophosphate ([3H]DFP) and SDS-polyacrylamide gel electrophoresis was used to measure serine hydrolases in cell-free extracts from rat splenic lymphocytes. After labeling with [3H]DFP at pH7, six major serine hydrolases are detected on 10% gels, having molecular weights of 78, 55, 34, 30, 28, and 17 (.10(-3)). When labeled at pH4, only four activities are measured, with Mr of 79, 55, 33 and 17(.10(-3)). Incubation of splenic lymphocytes for 8 h in vitro with 1 microM dexamethasone followed by [3H]-DFP labeling at pH 7 produces a 91% increase in the 17000 [3H]DFP. Hormone treatment for 8 h with subsequent labeling at pH 4 results in a 15% increase in the largest (78000) species, as well as 73% increase in the 17000 enzyme, compared with lysates from cells incubated without steroid. These effects are not observed after only 4 h of glucocorticoid exposure. Dexamethasone treatment for 8 h does not produce a decrease in any of these serine hydrolases, nor is there an apparent induction of new enzymes (i.e., having a molecular weight different from the preexisting species). Studies examining the effect of protease inhibitors on the [3H]DFP capacity of these proteins, show that the 17000 enzyme is sensitive to the protease inhibitor, pepstatin A, as well as the sulfhydryl reagents dithiothreitol and N-ethylmaleimide. These results suggest that this dexamethasone-responsive enzyme is a protease which requires a free thiol group for optimal activity. These findings are discussed with regard to the mechanism of glucocorticoid action in lymphocytes.


Subject(s)
Dexamethasone/pharmacology , Endopeptidases/blood , Lymphocytes/enzymology , Animals , Electrophoresis, Polyacrylamide Gel , Isoflurophate/metabolism , Male , Molecular Weight , Protease Inhibitors/pharmacology , Protein Binding , Rats , Rats, Inbred Strains , Spleen , Substrate Specificity
13.
Biochim Biophys Acta ; 717(2): 236-47, 1982 Aug 06.
Article in English | MEDLINE | ID: mdl-7115767

ABSTRACT

In the presence of tracer concentrations of extracellular leucine (5 microM), treatment of rat splenic lymphocyte suspensions in vitro with 1 microM dexamethasone for 2.5-4 h caused a 30-35% inhibition of [3H]leucine incorporation into protein. As the extracellular leucine concentration was raised to 5mM, this inhibition was progressively reduced to 0-12%. This phenomenon correlated with a marked dependence on extracellular leucine concentration of the dexamethasone-dependent enlargement of free intracellular leucine pools in splenic lymphocytes: a 123% increase in pool size with tracer extracellular leucine; a 10% increase with 5 mM leucine. Varying extracellular leucine had no effect on: (1) nuclear [3H]dexamethasone binding by the cells; (2) the concentration of dexamethasone needed for half-maximal inhibiton of [3H]leucine incorporation; (3) the time course of onset and maximal expression of the hormonal inhibition of [3H]leucine incorporation; or (4) the magnitude of dexamethasone-dependent inhibition of [3H]uridine incorporation into RNA by these cells. There was no detectable effect of dexamethasone on uptake and retention of [3H]leucine by the cells regardless of the extracellular leucine concentration. Treatment of splenic lymphocytes for 4 h in vitro with 1 microM dexamethasone caused a small shift of ribosomes from larger aggregate polysomes to smaller forms. Thus, glucocorticoid-induced inhibition of amino aicd incorporation in splenic lymphocytes is a multicomponent response, of which an actual decrease in protein synthesis is only a small part. Enlargement of free intracellular amino acid pools, probably resulting from increased protein degradation, is the major contributing factor to the hormonal inhibition of amino acid incorporation.


Subject(s)
Amino Acids/metabolism , Dexamethasone/pharmacology , Lymphocytes/metabolism , Protein Biosynthesis , Animals , Biological Transport , Cell Nucleus/metabolism , Dexamethasone/metabolism , Kinetics , Leucine/metabolism , Lymphocytes/drug effects , Polyribosomes/drug effects , Polyribosomes/metabolism , Proteins/genetics , Rats , Receptors, Glucocorticoid/metabolism , Spleen/metabolism
14.
Cell Death Differ ; 10(7): 791-7, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12815462

ABSTRACT

The Fas-associated death domain (FADD) adaptor protein FADD/Mort-1 is recruited by several members of the tumor necrosis factor receptor (TNFR) superfamily during cell death activated via death receptors. Since most studies have focused on the interaction of FADD with plasma membrane proteins, FADD's subcellular location is thought to be confined to the cytoplasm. In this report, we show for the first time that FADD is present in both the cytoplasm and the nucleus of cells, and that its nuclear localization relies on strong nuclear localization and nuclear export signals (NLS and NES, respectively) that reside in the death-effector domain (DED) of the protein. Specifically, we found that a conserved basic KRK35 sequence of the human protein is necessary for FADD's nuclear localization, since disruption of this motif leads to the confinement of FADD in the cytoplasm. Furthermore, we show that the leucine-rich motif LTELKFLCL28 in the DED is necessary for FADD's nuclear export. Functionally, mutation of the NES of FADD and its seclusion in the nucleus reduces the cell death-inducing efficacy of FADD reconstituted in FADD-deficient T cells.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis/physiology , Carrier Proteins/metabolism , Cell Compartmentation/genetics , Cell Nucleus/metabolism , Active Transport, Cell Nucleus/physiology , Amino Acid Sequence/physiology , Carrier Proteins/genetics , Cytoplasm/metabolism , Fas-Associated Death Domain Protein , HeLa Cells , Humans , Jurkat Cells , Mutation/genetics , Protein Structure, Tertiary/physiology , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , T-Lymphocytes/metabolism
15.
Cell Death Differ ; 1(1): 11-7, 1994 Jul.
Article in English | MEDLINE | ID: mdl-17180002

ABSTRACT

Apoptosis is characterized by multiple morphological and biochemical changes. One biochemical change that has been primarily associated with apoptosis is the cleavage of chromatin in the internucleosomal regions. We have taken two independent approaches to investigating the enzyme(s) responsible for such cleavage. First, using SDS-PAGE gels with (32)P-labelled DNA incorporated into the matrix, we identified a nuclease activity (termed NUC18) from apoptotic thymocytes. This enzyme has been purified to homogeneity and the activity of the pure protein is dependent on Ca(2+) and Mg(2+) while inhibited by Zn(2+) and aurintricarboxylic acid. This protein is found in the nucleus of apoptotic and nonapoptotic cells but is maintained in nondying cells in a large-molecular-weight inactive complex. NUC18 has a denatured molecular weight of 18 Kd but elutes from gel filtration columns with a native molecular weight of approximately 25 Kd. Although an exhaustive search has not been performed, NUC18 has been identified in several cell lines and tissues. Our second approach is designed specifically to detect internucleosomal cleavage of DNA, an obvious requirement for an apoptotic nuclease. By examining the degradation of HeLa chromatin, we have identified a low-molecular-weight of approximately 23 Kd native molecular weight) internucleosomal cleavage enzyme active in nuclear extracts from glucocorticoid-treated thymocytes. This activity is also dependent upon Ca(2+)and Mg(2+) and is inhibited by Zn(2+) as well as aurintricarboxylic acid. It is present in a variety of cell lines and tissues and is maintained in control cells in a latent state prior to apoptosis. In addition to similarities in physical properties, the two enzymes appear to be immunologically related to one another by virtue of their ability to interact with the same antibody. Overall, using independent approaches, we have identified two nucleases with similar biochemical properties whose activity correlates with apoptosis. The current work suggests that these are novel and perhaps closely related enzymes.

16.
Cell Death Differ ; 4(3): 200-8, 1997 Apr.
Article in English | MEDLINE | ID: mdl-16465229

ABSTRACT

Apoptosis is commonly associated with the catabolism of the genome in the dying cell. The chromatin degradation occurs in essentially two forms: (1) internucleosomal DNA cleavage to generate oligonucleosomal-length fragments (180-200 bp and multiples thereof), and (2) cleavage of higher order chromatin structures to generate approximately 30-50 Kb fragments. To investigate this component of apoptosis and identify the nuclease(s) responsible, we have developed and utilized an in vitro assay that recapitulates the genomic destruction seen during apoptosis in vivo and allows the simultaneous analysis of both forms of DNA degradation from the same sample. Using this assay we evaluated the digestion patterns of several candidate apoptotic nucleases: DNase I, DNase II, and cyclophilin (NUC18) as well as the bacterial enzyme micrococcal nuclease (not thought to be involved in apoptosis). Chromatin degraded by DNase I formed a smear of DNA on conventional static-field agarose gels and approximately amp;30 - 50 Kb DNA fragments on pulsed field gels. In contrast, DNase II, at a physiologically relevant pH, had no effect on the integrity of HeLa chromatin in either analysis. Similar to DNase I, cyclophilin C produced only approximately 30-50 Kb DNA fragments but did not generate internucleosomal fragments. In contrast, micrococcal nuclease generated both oligonucleosomal and approximately 30-50 Kb DNA fragments. Nuclear extracts from glucocorticoid-treated apoptotic thymocytes generated oligonucleosomal DNA fragments and the larger approximately 30-50 Kb DNA fragments, fully recapitulating both types of apoptotic DNA degradation. Previously, differential sensitivity of nucleases to inhibition by Zn2+ was used to argue that two distinct enzymes mediate approximately 30-50 Kb DNA cleavage and internucleosomal DNA degradation. While, the nuclease activity present in thymocyte nuclear extracts was differentially sensitive to inhibition by Zn2+ during short term incubations it was not during prolonged digestions, suggesting that differences in DNA detection are likely to account for previous results. Together our studies show that none of the nucleases commonly associated with apoptosis could fully recapitulate the DNA degradation seen in vivo.

17.
Cell Death Differ ; 5(12): 1017-27, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9894608

ABSTRACT

Chromatin degradation into oligonucleosomal and approximately 30-50 Kb fragments is a hallmark of apoptosis. Crude nuclear extract from apoptotic rat thymocytes is able to recapitulate both types of DNA fragmentation in an assay using HeLa cell nuclei as an exogenous substrate. Using size exclusion chromatography we have identified a novel activity (approximately 260 Kd) that produces only approximately 30-50 Kb DNA fragments, and a 25 Kd activity that generates both approximately 30-50 Kb and oligonucleosomal fragments. Both activities produced DNA fragments with 3'-OH termini, are dependent on Ca2+ and Mg2+ and are inhibited by N-ethyl-maleimide, sodium tetrathionate, aurintricarboxylic acid and sodium chloride, similar to other nucleases implicated in apoptosis. These activities were inhibited by the serine protease inhibitors N-tosyl-L-phenylalanine chloromethyl ketone and N alpha-p-tosyl-L-lysine chloromethyl ketone, but not by the serine protease inhibitor diisopropyl fluorophosphate, or by calpain inhibitors I or II, or the capsase inhibitors Ac-Asp-Glu-Val-Asp-aldehyde, Ac-Tyr-Val-Ala-Asp-aldehyde, or Z-Val-Ala-Asp-fluoromethyl ketone. Both activities were insensitive to protease inhibitors when extracts were incubated with naked linear DNA, indicating the presence of both nuclease and protease activities in the preparation. Together, these observations suggest the involvement of non-caspase proteases in apoptosis which perhaps function by altering chromatin substructure and exposing it to nucleolytic attack.


Subject(s)
Apoptosis/physiology , Chromatin/metabolism , DNA Fragmentation/physiology , Serine Endopeptidases/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/drug effects , Calcium/metabolism , Calpain/antagonists & inhibitors , Calpain/metabolism , Caspase 1/metabolism , Caspase 3 , Caspase Inhibitors , Caspases/metabolism , Cell Nucleolus/chemistry , Cell Nucleolus/enzymology , Cysteine Proteinase Inhibitors/pharmacology , Electrophoresis, Gel, Pulsed-Field , Endonucleases/metabolism , HeLa Cells , Humans , Isoflurophate/pharmacology , Magnesium/metabolism , Male , Oligopeptides/pharmacology , Peptide Fragments/analysis , Peptide Fragments/metabolism , Protease Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Serine Proteinase Inhibitors/pharmacology , Thymus Gland/cytology , Tosyllysine Chloromethyl Ketone/pharmacology , Tosylphenylalanyl Chloromethyl Ketone/pharmacology
18.
Cell Death Differ ; 7(10): 994-1001, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11279546

ABSTRACT

Apoptosis, a physiological form of cell death, is characterized by the activation of a program that kills cells and recycles their constituents. We have used thymoma cell lines to examine the role of Bcl-2 and caspases in ribosomal destruction during apoptosis. Glucocorticoid- and calcium ionophore (A23187)-induced apoptosis of S49 Neo cells resulted in both 28S rRNA and DNA degradation. Interestingly, anisomycin, a potent protein synthesis inhibitor, also induced 28S rRNA and DNA fragmentation suggesting that the responsible nucleases are present in the viable cells and become activated during apoptosis. The anti-apoptotic protein, Bcl-2, inhibited both glucocorticoid- and anisomycin-induced DNA and 28S rRNA degradation but could not protect against A23187-induced nucleic acid degradation. We next examined the role of caspase activation in the generation of 28S rRNA degradation through the use of ZVAD, a general caspase inhibitor. Under conditions where ZVAD substantially decreased 28S rRNA degradation induced by glucocorticoid or anisomycin, no decrease was observed when A23187 was used to induce apoptosis. Surprisingly, RNA degradation, like DNA degradation, occurs exclusively in shrunken lymphocytes but not those with normal cell volume despite equivalent exposure of the cells to the apoptotic signals. Together, these findings indicate the ribosome is a specific target for death effectors during apoptosis and that a caspase/Bcl-2-independent pathway exists to activate its destruction.


Subject(s)
Apoptosis/immunology , Caspases/metabolism , Lymphocytes/cytology , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Ribosomal, 28S/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Anisomycin/pharmacology , Apoptosis/drug effects , Blotting, Northern , Cysteine Proteinase Inhibitors/pharmacology , DNA, Neoplasm/metabolism , Lymphocytes/metabolism , Mice , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/analysis , Thymoma , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/enzymology
19.
Cell Death Differ ; 5(11): 973-83, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9846184

ABSTRACT

Retinoids play an important role in the control of lymphocyte function and homeostasis in the thymus. In this study, we show that the induction of growth arrest and apoptosis in a variety of T-cell lymphoma cell lines, including Jurkat and Molt-4 cells, is highly specific for the synthetic retinoid 6-[3-(1-adamantyl)-4-hydroxyphenyl]-2-naphthalene carboxylic acid (AHPN) since all-trans retinoic acid (RA), the RAR-selective retinoid TTAB, the RXR-selective retinoid SR11217 and the retinoid SR11302 exhibiting selective anti-AP1 activity, do not induce apoptosis or cause growth arrest. These findings support the concept that the effects of AHPN on proliferation and induction of apoptosis are mediated by a novel signaling pathway. AHPN-induced apoptosis is associated with an induction of internucleosomal DNA-fragmentation, increased annexin V binding and a 30-fold stimulation of caspase-3-like activity. Overexpression of Bcl-2 in Molt-4 cells greatly inhibits the induction of apoptosis by AHPN as indicated by the inhibition of DNA-fragmentation, annexin V binding and caspase-3-like activity. However, Bcl-2 overexpression does not interfere with the ability of AHPN to cause growth arrest or accumulation of cells in the early S-phase of the cell cycle, indicating that the effects of AHPN on growth arrest can be uncoupled from the effects on apoptosis. The caspase inhibitor Z-VAD-FMK, at concentrations that totally block caspase activity, delays but does not prevent cell death and does not affect the accumulation of cells in the S-phase of the cell cycle. Our results show that induction of caspase-3-like activity plays an important role in the execution of AHPN-induced apoptosis but cells can undergo cell death in the absence of this activity suggesting that AHPN-induced cell death involves caspase-dependent and -independent mechanisms.


Subject(s)
Apoptosis/drug effects , Apoptosis/physiology , Caspases/physiology , Lymphoma, T-Cell/drug therapy , Lymphoma, T-Cell/enzymology , Retinoids/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Annexin A5/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/genetics , Caspase 3 , Caspase Inhibitors , Caspases/biosynthesis , Cell Division/drug effects , Cysteine Proteinase Inhibitors/pharmacology , DNA Fragmentation/drug effects , Enzyme Induction/drug effects , Genes, bcl-2 , Humans , Jurkat Cells , Lymphoma, T-Cell/pathology , Nucleosomes/drug effects , Nucleosomes/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Signal Transduction , Tumor Cells, Cultured
20.
Cell Death Differ ; 5(1): 38-49, 1998 Jan.
Article in English | MEDLINE | ID: mdl-10200444

ABSTRACT

To continue elucidation of the biochemical and molecular pathways involved in the induction of apoptosis in granulosa cells (GC) of ovarian follicles destined for atresia, we characterized the occurrence and protease modulation of high and low molecular weight (MW) DNA fragmentation during rat GC death. Atresia of ovarian follicles, occurring either spontaneously in vivo or induced in vitro, was associated with both high MW and internucleosomal (low MW) DNA cleavage. Incubation of follicles in the presence of a putative irreversible and non-competitive inhibitor of caspase-1 (interleukin-1beta-converting enzyme or ICE), sodium aurothiomalate (SAM), completely prevented internucleosomal, but not high MW, DNA cleavage. As reported previously, morphological features of apoptosis (pyknosis, cellular condensation) and atresia (granulosa cell disorganization, oocyte pseudomaturation) remained detectable in SAM-treated follicles. The potential involvement of proteases in endonuclease activation was further analyzed in cell-free assays using nuclei from both GC (which autodigest their DNA) and HeLa cells (HC, which do not autodigest their DNA unless incubated with extracts prepared from other cell types). Crude cytoplasmic extracts prepared from GC induced both high MW and internucleosomal DNA cleavage in HC nuclei. The induction of low, but not high, MW DNA cleavage in HC nuclei by GC extracts was suppressed by pretreatment of the extracts with SAM or with any one of the serine protease inhibitors, dichloroisocoumarin (DCI), N-tosyl-L-leucylchloromethylketone (TLCK) or N-tosyl-L-phenylchloromethylketone (TPCK). Interestingly, SAM and DCI also prevented cation-induced low MW DNA fragmentation in GC nuclei; however, TLCK and TPCK were without effect. Our results support a role for cytoplasmic and nuclear serine proteases in the activation of the endonuclease(s) responsible for internucleosomal DNA cleavage during apoptosis.


Subject(s)
Apoptosis/physiology , Cell Nucleus/enzymology , Deoxyribonucleases/metabolism , Endopeptidases/metabolism , Follicular Atresia/metabolism , Ovarian Follicle/cytology , Animals , Antirheumatic Agents/pharmacology , Apoptosis/drug effects , Caspase 1/metabolism , Caspase Inhibitors , Cell Nucleus/chemistry , Cell-Free System , Cysteine Proteinase Inhibitors/pharmacology , DNA/chemistry , DNA/metabolism , Deoxyribonucleases/antagonists & inhibitors , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Female , Gold Sodium Thiomalate/pharmacology , HeLa Cells , Humans , Molecular Weight , Oligopeptides/pharmacology , Ovarian Follicle/metabolism , Rats , Rats, Sprague-Dawley , Serine Proteinase Inhibitors/pharmacology , Tosyllysine Chloromethyl Ketone/pharmacology , Tosylphenylalanyl Chloromethyl Ketone/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL