Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Neuroscience ; 141(2): 863-874, 2006 Aug 25.
Article in English | MEDLINE | ID: mdl-16750894

ABSTRACT

Corticocortical disconnection in Alzheimer's disease occurs by the progressive impairment and eventual loss of a small subset of pyramidal neurons in layers III and V of association areas of the neocortex. These neurons exhibit large somatic size, extensive dendritic arborization and high levels of nonphosphorylated neurofilaments of medium and high molecular weight that can be identified using a monoclonal SMI-32 antibody. It is thought that the accumulation of amyloid Abeta and neurofibrillary tangles may provoke metabolic disturbances that result in the loss of these SMI-32 immunoreactive neurons. The recent detection of increased levels of caspase-3 cleaved fodrin in frontal, temporal and parietal association areas in Alzheimer's disease brains suggests that programmed cell death may contribute to the destruction of SMI-32 positive neurons. In the present study, we utilized an antibody that selectively recognizes the 120 kDa breakdown product of alphaIIspectrin (fodrin) generated by caspase-3 to determine whether this protease is activated in vulnerable pyramidal neurons located in layers III and V of Alzheimer's disease brains. Neurons immunoreactive for caspase-3 cleaved alphaIIspectrin were located predominantly in layers III and V of the inferior frontal and superior temporal cortices of patients with Alzheimer's disease but not age-matched controls. Pyramidal neurons immunoreactive for caspase-3 cleaved alphaIIspectrin invariably displayed SMI-32 immunoreactivity suggesting that caspase-3 activation is a pathological event that may be responsible for the loss of a subset of pyramidal neurons that comprise corticocortical projections.


Subject(s)
Alzheimer Disease/metabolism , Caspases/metabolism , Neurofilament Proteins/metabolism , Neurons/metabolism , Spectrin/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Analysis of Variance , Animals , Blotting, Western/methods , Brain/metabolism , Brain/pathology , Caspase 3 , Female , Humans , Immunohistochemistry/methods , Immunoprecipitation/methods , Male , Rats
2.
Neuroscience ; 126(4): 927-40, 2004.
Article in English | MEDLINE | ID: mdl-15207327

ABSTRACT

The ability of fimbria-fornix bilateral axotomy to elicit calpain and caspase-3 activation in the rat septohippocampal pathway was determined using antibodies that selectively recognize either calpain- or caspase-cleaved products of the cytoskeletal protein alphaII-spectrin. Radioenzymatically determined choline acetyl transferase (ChAT) activity was elevated in the septum at day 5, but reduced in the dorsal hippocampus at days 3, 5 and 7, after axotomy. Prominent accumulation of calpain-, but not caspase-3-, cleaved spectrin proteolytic fragments was observed in both the septum and dorsal hippocampus 1-7 days after axotomy. ChAT-positive neuronal cell bodies in the septum also displayed calpain-cleaved spectrin indicating that calpain activation occurred in cholinergic septal neurons as a consequence of transection of the septohippocampal pathway. Calpain-cleaved alphaII-spectrin immunoreactivity was observed in cholinergic fibers coursing through the fimbria-fornix, but not in pyramidal neurons of the dorsal hippocampus, suggesting that degenerating cholinergic nerve terminals were the source of calpain activity in the dorsal hippocampus following axotomy. Accumulation of calpain-cleaved spectrin proteolytic fragments in the dorsal hippocampus and septum at day 5 after axotomy was reduced by i.c.v. administration of two calpain inhibitors. Calpain inhibition partially reduced the elevation of ChAT activity in the septum produced by transection but failed to decrease the loss of ChAT activity in the dorsal hippocampus following axotomy. These findings suggest that calpain activation contributes to the cholinergic cell body response and hippocampal axonal cytoskeletal degradation produced by transection of the septohippocampal pathway.


Subject(s)
Calpain/metabolism , Choline O-Acetyltransferase/metabolism , Fornix, Brain/physiology , Hippocampus/enzymology , Neural Pathways/enzymology , Septum Pellucidum/enzymology , Animals , Axotomy/methods , Blotting, Western/methods , Caspase 3 , Caspases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Fornix, Brain/injuries , Fornix, Brain/surgery , Hippocampus/drug effects , Immunohistochemistry/methods , Leupeptins/pharmacology , Male , Neural Pathways/drug effects , Protease Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Septum Pellucidum/drug effects , Spectrin/metabolism , Time Factors
3.
Cell Death Differ ; 11(4): 439-47, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14713960

ABSTRACT

The activation of caspase-3 represents a critical step in the pathways leading to the biochemical and morphological changes that underlie apoptosis. Upon induction of apoptosis, the large (p17) and small (p12) subunits, comprising active caspase-3, are generated via proteolytic processing of a latent proenzyme dimer. Two copies of each individual subunit are generated to form an active heterotetramer. The tetrameric form of caspase-3 cleaves specific protein substrates within the cell, thereby producing the apoptotic phenotype. In contrast to the proenzyme, once activated in HeLa cells, caspase-3 is difficult to detect due to its rapid degradation. Interestingly, however, enzyme stability and therefore detection of active caspase-3 by immunoblot analysis can be restored by treatment of cells with a peptide-based caspase-3 selective inhibitor, suggesting that the active form can be stabilized through protein-inhibitor interaction. The heteromeric active enzyme complex is necessary for its stabilization by inhibitors, as expression of the large subunit alone is not stabilized by the presence of inhibitors. Our results show for the first time, that synthetic caspase inhibitors not only block caspase activity, but may also increase the stability of otherwise rapidly degraded mature caspase complexes. Consistent with these findings, experiments with a catalytically inactive mutant of caspase-3 show that rapid turnover is dependent on the activity of the mature enzyme. Furthermore, turnover of otherwise stable active site mutants of capase-3 is rescued by the presence of the active enzyme suggesting that turnover can be mediated in trans.


Subject(s)
Caspase Inhibitors , Caspases/metabolism , Enzyme Inhibitors/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Caspase 3 , Catalysis , Cell Line, Tumor , Enzyme Stability/drug effects , Gene Expression Regulation, Enzymologic/drug effects , HeLa Cells , Humans , Macromolecular Substances , Molecular Structure , Tumor Cells, Cultured
4.
Neuroscience ; 115(1): 125-36, 2002.
Article in English | MEDLINE | ID: mdl-12401327

ABSTRACT

In the present study, we evaluated the time-course of caspase-3 activation, and the evolution of cell death following focal cerebral ischemia produced by transient middle cerebral artery occlusion in rats. Ischemia-induced active caspase-3 immunoreactivity in the striatum but not the cortex at 3 and 6 h time points post-reperfusion. Furthermore, using a novel approach to visualize enzymatic activity, deltaC-APP, a C-terminal cleavage product of APP generated by caspase-3, was found to immunolocalize to the same areas as active caspase-3. Double-labeling studies demonstrated co-localization of these two proteins at the cellular level. Further double-labeling experiments revealed that active caspase-3 was confined to neuronal cells which were still viable and thus immunoreactive for NeuN. DNA fragmentation, assessed histologically by terminal dUTP nick-end labeling (TUNEL), was observed in a small number of cells in the striatum as early as 3 h, but only began to appear in the cortex by 6 h. DNA fragmentation was progressive, and by 24 h post-reperfusion, large portions of both the striatum and cortex showed TUNEL positive cells. However, double-labeling of active caspase-3 with TUNEL showed only minimal co-localization at all time-points. Thus, caspase-3 activation is an event that appears to occur prior to DNA fragmentation. As a confirmation of the histological TUNEL data, 24 h ischemia also induced the generation of nucleosome fragments, evidenced by cell death enzyme-linked immunosorbent assay. Using a novel ischemia-induced substrate cleavage biochemical approach, spectrin P120 fragment, a caspase-specific cleavage product of alpha II spectrin, a cytoskeletal protein, was shown to be elevated by western blotting. Brain concentrations of both nucleosomes and spectrin P120 correlate with the degree of injury previously assessed by triphenyltetrazolium chloride staining and infarct volume calculation. Together, our findings suggest a possible association between caspase-3 activation and ischemic cell death following middle cerebral artery occlusion brain injury.


Subject(s)
Caspases/analysis , Caspases/metabolism , DNA Fragmentation , Ischemic Attack, Transient/enzymology , Animals , Caspase 3 , Cell Death/genetics , DNA Fragmentation/genetics , Enzyme Activation/genetics , Immunohistochemistry , In Situ Nick-End Labeling , Ischemic Attack, Transient/genetics , Male , Rats , Rats, Wistar
5.
Cell Death Differ ; 9(8): 818-31, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12107825

ABSTRACT

Neuronal death, which follows ischemic injury or is triggered by excitotoxins, can occur by both apoptosis and necrosis. Caspases, which are not directly required for necrotic cell death, are central mediators of the apoptotic program. Here we demonstrate that caspases cleave and inactivate the plasma membrane Ca(2+) pump (PMCA) in neurons and non-neuronal cells undergoing apoptosis. PMCA cleavage impairs intracellular Ca(2+) handling, which results in Ca(2+) overload. Expression of non-cleavable PMCA mutants prevents the disturbance in Ca(2+) handling, slows down the kinetics of apoptosis, and markedly delays secondary cell lysis (necrosis). These findings suggest that caspase-mediated cleavage and inactivation of PMCAs can lead to necrosis, an event that is reduced by caspase inhibitors in brain ischemia.


Subject(s)
Apoptosis/physiology , Calcium-Transporting ATPases/metabolism , Caspases/metabolism , Cell Membrane/enzymology , Hypoxia-Ischemia, Brain/enzymology , Necrosis , Neurons/enzymology , Animals , Animals, Newborn , Apoptosis/drug effects , Binding Sites/drug effects , Binding Sites/physiology , CHO Cells , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Calcium-Transporting ATPases/drug effects , Caspase 3 , Caspases/drug effects , Caspases/genetics , Cation Transport Proteins , Cell Membrane/drug effects , Clone Cells/cytology , Clone Cells/drug effects , Clone Cells/metabolism , Coloring Agents , Cricetinae , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/physiopathology , Immunohistochemistry , Intracellular Fluid/metabolism , Mice , Mutation/drug effects , Mutation/genetics , Neurons/drug effects , Neurons/pathology , Neurotoxins/pharmacology , Plasma Membrane Calcium-Transporting ATPases , Rats
6.
Cell Death Differ ; 8(1): 30-7, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11313700

ABSTRACT

Caspase activation and proteolytic cleavage of specific target proteins represents an integral step in the pathway leading to the apoptotic death of cells. Analysis of caspase activity in intact cells, however, has been generally limited to the measurement of end-point biochemical and morphological markers of apoptosis. In an effort to develop a strategy with which to monitor caspase activity, early in the cell death cascade and in real-time, we have generated cell lines that overexpress recombinant GFP-based caspase substrates that display a quantifiable change in their spectral properties when cleaved by group II caspases. Specifically, tandem GFP substrates linked by a caspase-sensitive cleavage site show diminished fluorescence resonance energy transfer (FRET), as a consequence of cleavage, due to physical separation of the GFP moieties in apoptotic cells. We have evaluated the influence of different caspase-sensitive linkers on both FRET efficiency and cleavage by caspase-3. We also demonstrate that caspase activity as well as inhibition by pharmacological agents can be monitored, with minimal manipulation, in intact adherent cells seeded in a 96-well cell culture dish. Finally, we have adapted this technology to a high throughput screening platform to identify novel small molecule and cell permeable inhibitors of apoptosis. Based on a biochemical analysis of the compounds identified it is clear that this assay can be used to detect drugs which inhibit caspases directly as well as those which target upstream components of the caspase cascade.


Subject(s)
Apoptosis , Biological Assay/methods , Caspases/metabolism , Luminescent Proteins/genetics , Recombinant Fusion Proteins/metabolism , Apoptosis/drug effects , Caspase 3 , Caspase Inhibitors , Dose-Response Relationship, Drug , Energy Transfer/physiology , Enzyme Activation/drug effects , Enzyme Inhibitors/analysis , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Genes, Reporter , Green Fluorescent Proteins , HeLa Cells , Humans , Hydrolysis/drug effects , Peptide Fragments/analysis , Poly(ADP-ribose) Polymerases/genetics , Recombinant Fusion Proteins/genetics , Spectrometry, Fluorescence , Substrate Specificity/physiology
8.
Nat Immunol ; 1(6): 496-501, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11101871

ABSTRACT

Sepsis induces lymphocyte apoptosis and prevention of lymphocyte death may improve the chances of surviving this disorder. We compared the efficacy of a selective caspase-3 inhibitor to a polycaspase inhibitor and to caspase-3-/- mice. Both inhibitors prevented lymphocyte apoptosis and improved survival. Caspase-3-/- mice shared a decreased, but not total, block of apoptosis. The polycaspase inhibitor caused a very substantial decrease in bacteremia. Caspase inhibitors did not benefit RAG-1-/- mice, which had a > tenfold increase in bacteremia compared to controls. Adoptive transfer of T cells that overexpressed the anti-apoptotic protein Bcl-2 increased survival. T cells stimulated with anti-CD3 and anti-CD28 produced increased interleukin 2 and interferon gamma by 6 h. Thus, caspase inhibitors enhance immunity by preventing lymphocyte apoptosis and lymphocytes act rapidly, within 24 h, to control infection.


Subject(s)
Apoptosis/drug effects , Caspase Inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Lymphocytes/drug effects , Sepsis/drug therapy , Adoptive Transfer , Animals , Bacteremia/drug therapy , Bacteremia/microbiology , Bacteremia/pathology , Caspase 3 , Caspases/genetics , Colony Count, Microbial , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , In Vitro Techniques , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Lymphocytes/pathology , Mice , Mice, Knockout , Sepsis/pathology
9.
Brain Res Mol Brain Res ; 70(1): 159-63, 1999 Jun 18.
Article in English | MEDLINE | ID: mdl-10381555

ABSTRACT

In this study, we examined the levels of activated caspase-3 in the kainic acid (KA) model of hippocampal degeneration in both sensitive (FVB/N) and resistant (129/SvEMS) strains of mice. At 30 h, 2 and 4 days following KA administration, animals were sacrificed and brains examined for pyknosis, TUNEL labeling, and activated caspase-3 immunoreactivity. Catalytically active caspase-3 was first detected 30 h following KA treatment in the sensitive, FVB/N strain. This was 18 h before the appearance of pyknosis or TUNEL labeling. The expression of activated caspase-3 continues up to 4 days post-injection. No activated caspase-3 immunoreactivity was detected in the resistant, 129/SvEMS strain, neither was there evidence of pyknosis or TUNEL staining. This suggests that activation of caspase-3 is a necessary component of KA-induced cell death.


Subject(s)
Apoptosis/drug effects , Caspases/physiology , Excitatory Amino Acid Agonists/toxicity , Hippocampus/drug effects , Kainic Acid/toxicity , Nerve Tissue Proteins/physiology , Neurons/pathology , Animals , Caspase 3 , Drug Resistance , Enzyme Activation/drug effects , Excitatory Amino Acid Agonists/pharmacology , Gene Expression Regulation/drug effects , Hippocampus/pathology , In Situ Nick-End Labeling , Kainic Acid/pharmacology , Male , Mice , Mice, Inbred Strains , Seizures/chemically induced , Seizures/enzymology , Seizures/pathology
10.
Cell ; 97(3): 395-406, 1999 Apr 30.
Article in English | MEDLINE | ID: mdl-10319819

ABSTRACT

The amyloid-beta precursor protein (APP) is directly and efficiently cleaved by caspases during apoptosis, resulting in elevated amyloid-beta (A beta) peptide formation. The predominant site of caspase-mediated proteolysis is within the cytoplasmic tail of APP, and cleavage at this site occurs in hippocampal neurons in vivo following acute excitotoxic or ischemic brain injury. Caspase-3 is the predominant caspase involved in APP cleavage, consistent with its marked elevation in dying neurons of Alzheimer's disease brains and colocalization of its APP cleavage product with A beta in senile plaques. Caspases thus appear to play a dual role in proteolytic processing of APP and the resulting propensity for A beta peptide formation, as well as in the ultimate apoptotic death of neurons in Alzheimer's disease.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloidosis/enzymology , Caspases/metabolism , Acute Disease , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Amyloidosis/genetics , Amyloidosis/pathology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Aspartic Acid , Aspartic Acid Endopeptidases , Brain Diseases/chemically induced , Brain Diseases/enzymology , Brain Diseases/pathology , Camptothecin/pharmacology , Caspase 3 , Caspases/analysis , Cysteine Proteinase Inhibitors/pharmacology , Endopeptidases/genetics , Enzyme Inhibitors/pharmacology , Enzyme Precursors/analysis , Enzyme Precursors/metabolism , Excitatory Amino Acid Agonists , Hippocampus/cytology , Humans , In Situ Nick-End Labeling , Kainic Acid , Leukemia, Erythroblastic, Acute , Male , Mice , Mice, Inbred C57BL , Mutation/physiology , Neurons/chemistry , Neurons/cytology , Neurons/enzymology , Oligopeptides/pharmacology , Rabbits , Rats , Rats, Wistar , Sweden , Tumor Cells, Cultured
11.
EMBO J ; 18(8): 2049-56, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10205159

ABSTRACT

The activation of caspases represents a critical step in the pathways leading to the biochemical and morphological changes that underlie apoptosis. Multiple pathways leading to caspase activation appear to exist and vary depending on the death-inducing stimulus. We demonstrate that the activation of caspase-3, in Jurkat cells stimulated to undergo apoptosis by a Fas-independent pathway, is catalyzed by caspase-6. Caspase-6 was found to co-purify with caspase-3 as part of a multiprotein activation complex from extracts of camptothecin-treated Jurkat cells. A biochemical analysis of the protein constituents of the activation complex showed that Hsp60 was also present. Furthermore, an interaction between Hsp60 and caspase-3 could be demonstrated by co-immunoprecipitation experiments using HeLa as well as Jurkat cell extracts. Using a reconstituted in vitro system, Hsp60 was able to substantially accelerate the maturation of procaspase-3 by different upstream activator caspases and this effect was dependent on ATP hydrolysis. We propose that the ATP-dependent 'foldase' activity of Hsp60 improves the vulnerability of pro-caspase-3 to proteolytic maturation by upstream caspases and that this represents an important regulatory event in apoptotic cell death.


Subject(s)
Apoptosis , Caspases/metabolism , Chaperonin 60/metabolism , Enzyme Precursors/metabolism , Amino Acid Sequence , Caspase 3 , Caspases/isolation & purification , Chromatography, Ion Exchange , Enzyme Activation , Enzyme Precursors/isolation & purification , HeLa Cells , Humans , Jurkat Cells , Molecular Sequence Data , Protein Processing, Post-Translational , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Spectrophotometry, Ultraviolet
12.
Cell Death Differ ; 5(4): 271-88, 1998 Apr.
Article in English | MEDLINE | ID: mdl-10200473

ABSTRACT

Apoptotic cell suicide initiated by ligation of CD95 (Fas/APO-1) occurs through recruitment, oligomerization and autocatalytic activation of the cysteine protease, caspase-8 (MACH, FLICE, Mch5). An endogenous mammalian regulator of this process, named Usurpin, has been identified (aliases for Usurpin include CASH, Casper, CLARP, FLAME-1, FLIP, I-FLICE and MRIT). This protein is ubiquitously expressed and exists as at least three isoforms arising by alternative mRNA splicing. The Usurpin gene is comprised of 13 exons and is clustered within approximately 200 Kb with the caspase-8 and -10 genes on human chromosome 2q33-34. The Usurpin polypeptide has features in common with pro-caspase-8 and -10, including tandem 'death effector domains' on the N-terminus of a large subunit/small subunit caspase-like domain, but it lacks key residues that are necessary for caspase proteolytic activity, including the His and Cys which form the catalytic substrates diad, and residues that stabilize the P1 aspartic acid in substrates. Retro-mutation of these residues to functional caspase counterparts failed to restore proteolytic activity, indicating that other determinants also ensure the absence of catalytic potential. Usurpin heterodimerized with pro-caspase-8 in vitro and precluded pro-caspase-8 recruitment by the FADD/MORT1 adapter protein. Cell death induced by CD95 (Fas/APO-1) ligation was attenuated in cells transfected with Usurpin. In vivo, a Usurpin deficit was found in cardiac infarcts where TUNEL-positive myocytes and active caspase-3 expression were prominent following ischemia/reperfusion injury. In contrast, abundant Usurpin expression (and a caspase-3 deficit) occurred in surrounding unaffected cardiac tissue, suggesting reciprocal regulation of these pro- and anti-apoptotic molecules in vivo. Usurpin thus appears to be an endogenous modulator of apoptosis sensitivity in mammalian cells, including the susceptibility of cardiac myocytes to apoptotic death following ischemia/ reperfusion injury.


Subject(s)
Apoptosis/physiology , Caspases/physiology , Intracellular Signaling Peptides and Proteins , fas Receptor/physiology , Alternative Splicing , Amino Acid Sequence , Animals , Apoptosis/genetics , Apoptosis/immunology , Base Sequence , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/genetics , Carrier Proteins/physiology , Caspase 8 , Caspase 9 , Caspases/genetics , Chromosome Mapping , Chromosomes, Human, Pair 2/genetics , Cloning, Molecular , DNA Primers/genetics , Enzyme Activation , Female , HeLa Cells , Humans , Jurkat Cells , Male , Models, Biological , Molecular Sequence Data , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Sequence Homology, Amino Acid , Tissue Distribution
13.
Genes Dev ; 11(5): 558-70, 1997 Mar 01.
Article in English | MEDLINE | ID: mdl-9119221

ABSTRACT

p53 can be isolated from cells in a form that is inert for binding to DNA but that can be stimulated dramatically by phosphorylation, antibody binding, or short single strands of DNA. This suggests that upon genotoxic stress, cells can convert latent p53 to one that is active for DNA binding. Surprisingly, we observed that latent p53 is as effective in activating transcription in vitro as is active p53. We found that HeLa nuclear extracts can stimulate DNA binding by latent p53 and have purified from them a p53-stimulating protein that we have determined to be the product of the Ref-1 gene. Interestingly, Ref-1 is a dual function protein that can both regulate the redox state of a number of proteins and function as a DNA repair (A/P) endonuclease. We observed that oxidized forms of full-length and carboxy-terminally truncated p53 (p53 delta30), which are inactive for DNA binding, are both stimulated by the Ref-1 protein. However, in the presence of reducing agent, Ref-1 is an extremely potent stimulator of full-length p53 but not p53 delta30. These and additional data indicate that Ref-1 protein stimulates p53 by both redox-dependent and -independent means and imply a key role for it in p53 regulation. Importantly, we have also determined that Ref-1 can stimulate p53 transactivation in vivo. This is the first example of a noncovalent protein modifier of p53 function identified in cells.


Subject(s)
Carbon-Oxygen Lyases , DNA-(Apurinic or Apyrimidinic Site) Lyase , Nuclear Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Cell Extracts , DNA/metabolism , HeLa Cells/chemistry , Humans , Mutation , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Oxidation-Reduction , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcription, Genetic
14.
Proc Natl Acad Sci U S A ; 93(17): 8919-23, 1996 Aug 20.
Article in English | MEDLINE | ID: mdl-8799128

ABSTRACT

The DNA-binding activity of AP-1 proteins is modulated, in vitro, by a posttranslational mechanism involving reduction oxidation. This mode of regulation has been proposed to control both the transcriptional activity and the oncogenic potential of Fos and Jun. Previous studies revealed that reduction of oxidized Fos and Jun by a cellular protein, Ref-1, stimulates sequence-specific AP-1 DNA-binding activity. Ref-1, a bifunctional protein, is also capable of initiating the repair of apurinic/apyrymidinic sites in damaged DNA. The relationship between the redox and DNA repair activities of Ref-1 is intriguing; both activities have been suggested to play an important role in the cellular response to oxidative stress. To investigate the physiological function of Ref-1, we used a gene targeting strategy to generate mice lacking a functional ref-1 gene. We report here that heterozygous mutant mice develop into adulthood without any apparent abnormalities. In contrast, homozygous mutant mice, lacking a functional ref-1 gene, die during embryonic development. Detailed analysis indicates that death occurs following blastocyst formation, shortly after the time of implantation. Degeneration of the mutant embryos is clearly evident at embryonic day 5.5. These findings demonstrate that Ref-1 is essential for early embryonic development.


Subject(s)
Carbon-Oxygen Lyases , DNA-(Apurinic or Apyrimidinic Site) Lyase , Genes, Lethal , Mice, Mutant Strains/embryology , Nuclear Proteins/genetics , Animals , Base Sequence , Blastocyst/pathology , DNA Repair , Gene Targeting , Heterozygote , Homozygote , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Oxidation-Reduction
15.
Science ; 272(5263): 892-5, 1996 May 10.
Article in English | MEDLINE | ID: mdl-8629027

ABSTRACT

Transcription factors of the NFAT family are thought to play a major role in regulating the expression of cytokine genes and other inducible genes during the immune response. The role of NFAT1 was investigated by targeted disruption of the NFAT1 gene. Unexpectedly, cells from NFAT1 -/- mice showed increased primary responses to Leishmania major and mounted increased secondary responses to ovalbumin in vitro. In an in vivo model of allergic inflammation, the accumulation of eosinophils and levels of serum immunoglobulin E were increased in NFAT1 -/- mice. These results suggest that NFAT1 exerts a negative regulatory influence on the immune response.


Subject(s)
DNA-Binding Proteins/physiology , Hypersensitivity/immunology , Immunity , Lymphocyte Activation , Nuclear Proteins , Transcription Factors/physiology , Amino Acid Sequence , Animals , Antigens, Protozoan/immunology , Cell Line , Cytokines/biosynthesis , DNA-Binding Proteins/genetics , Eosinophils/immunology , Gene Targeting , Immunoglobulin E/biosynthesis , Immunologic Memory , Leishmania major/immunology , Mice , Molecular Sequence Data , NFATC Transcription Factors , Ovalbumin/immunology , T-Lymphocytes/immunology , Transcription Factors/genetics
17.
J Biol Chem ; 269(45): 27855-62, 1994 Nov 11.
Article in English | MEDLINE | ID: mdl-7961715

ABSTRACT

A rise in extracellular calcium (Ca2+e) suppresses not only secretion of parathyroid hormone (PTH) but also expression of the PTH gene to ensure constant plasma Ca2+ level. A nuclear protein(s) in a wide variety of cells bound to the specific DNA elements (negative Ca2+ responsive elements, nCaREs) in the human PTH gene in sequence-specific and Ca2+e concentration-dependent manners. Our Southwestern cloning revealed that a redox factor protein (ref1), which was known to activate several transcription factors via alterations of their redox state, belonged to an nCaRE binding protein. The level of ref1 mRNA as well as of its protein was elevated by an increase in Ca2+e concentration. In gel shift assay, anti-ref1 antibody eliminated formation of the nCaRE-protein complex. We also found that there was another protein(s) interacting with nCaREs and ref1. Further, experiments with an antisense-ref cDNA expression vector introduced into cultured cells suggested that DNA (nCaRE)-ref1 interaction led to Ca2+e-mediated transcriptional suppression. Thus, it is concluded that ref1 possesses transcription repressor activity in addition to its function as a transcriptional auxiliary protein.


Subject(s)
Calcium/pharmacology , Carbon-Oxygen Lyases , Cell Nucleus/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase , Gene Expression Regulation/drug effects , Nuclear Proteins/metabolism , Parathyroid Hormone/biosynthesis , Animals , Base Sequence , Cell Line , Chloramphenicol O-Acetyltransferase/biosynthesis , Chlorocebus aethiops , Cloning, Molecular , Cricetinae , DNA-Binding Proteins/isolation & purification , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Immunoblotting , Kidney , Molecular Sequence Data , Oligodeoxyribonucleotides , Plasmids , Promoter Regions, Genetic , Suppression, Genetic , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Transfection
18.
Mol Cell Biol ; 14(9): 5997-6003, 1994 Sep.
Article in English | MEDLINE | ID: mdl-8065332

ABSTRACT

Many solid tumors contain substantial fractions of hypoxic cells which are relatively resistant to both radiation therapy and certain cytotoxic drugs. We have previously shown that exposure of human HT29 cells to hypoxic conditions results in the overexpression of certain enzymes involved in the detoxication of xenobiotics, including NAD(P)H:(quinone acceptor) oxidoreductase (DT)-diaphorase, and gamma-glutamylcysteine synthetase, the rate-limiting enzyme in glutathione synthesis. This hypoxic effect on DT-diaphorase was shown to involve both transcriptional induction and altered message stability. We have investigated the effects of hypoxia on elements in the promoter region of DT-diaphorase. Electrophoretic mobility shift assays demonstrate the induction of a binding activity to the AP-1 response element of DT-diaphorase. Supershift assays suggest that this binding is due to AP-1 nuclear factors and that members of the jun family are induced to a greater degree than fos by hypoxia. Analysis of the kinetics of transcription factor expression indicates that the expression of c-jun and junD is induced during hypoxic exposure; mRNA levels fall during reoxygenation. Induction of fos on the other hand is not as florid during hypoxia (5-fold) and is most pronounced (17-fold) 24 h after the restoration of an oxic environment. Thus, the hypoxic response of DT-diaphorase expression is mediated in part through AP-1, initially by a jun-related mechanism and then by the involvement of fos. The affinity of transcription factors for the AP-1 binding site depends on the redox state of a cysteine residue located close to the DNA-binding region of both Fos and Jun. A nuclear protein, Ref-1, maintains the reduced state of Fos and Jun and promotes binding to AP-1. Nuclear extracts of HT29 cells exposed to hypoxia show markedly increased Ref-1 protein content. Elevation of ref-1 steady-state mRNA levels occurs as an early event following induction of hypoxia and persists when cells are restored to a normally oxygenated environment. Nuclear run-on analysis demonstrates that induction of transcription is the mechanism of ref-1 mRNA elevation. Electrophoretic mobility shift assays and immunodepletion assays were used to further define the interaction of Ref-1 with specific AP-1-binding proteins under hypoxic conditions. These data demonstrate that the induction of detoxicating enzyme expression in HT29 cells exposed to hypoxia results from the induction of both transactivating factors that bind to the AP-1 element and of redox proteins that enhance their affinity for this element.


Subject(s)
Carbon-Oxygen Lyases , DNA-(Apurinic or Apyrimidinic Site) Lyase , Hypoxia/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Gene Expression Regulation , Humans , In Vitro Techniques , Oxidation-Reduction , RNA, Messenger/genetics , Time Factors , Tumor Cells, Cultured
20.
Proc Natl Acad Sci U S A ; 91(1): 23-7, 1994 Jan 04.
Article in English | MEDLINE | ID: mdl-7506414

ABSTRACT

The DNA binding activity of transcription factor AP-1 is regulated in vitro by a posttranslational mechanism involving reduction/oxidation (redox). Redox regulation is mediated by a conserved cysteine residue in the DNA-binding domain of Fos and Jun. Previously, we demonstrated that a DNA repair protein, Ref-1, could stimulate the DNA binding activity of Fos-Jun dimers by reducing this cysteine residue. To examine the relationship between the redox and repair functions of Ref-1, we generated a series of deletion mutants. Analysis of the truncated proteins in vitro revealed that the redox and repair activities are encoded by distinct regions of Ref-1. Sequences in the N-terminal domain of Ref-1 that are not present in functionally related proteins from other organisms are required for the redox activity, whereas the DNA repair activity requires conserved C-terminal sequences. Chemical alkylation or oxidation of cysteine sulfhydryls inhibits the redox activity of Ref-1 without affecting its DNA repair activity. Crosslinking studies suggest that a direct cysteine-mediated interaction occurs between Ref-1 and Jun.


Subject(s)
Carbon-Oxygen Lyases , DNA Repair , Nuclear Proteins/chemistry , Cysteine/chemistry , DNA Mutational Analysis , DNA-(Apurinic or Apyrimidinic Site) Lyase , Deoxyribonuclease IV (Phage T4-Induced) , Endodeoxyribonucleases/chemistry , Humans , Macromolecular Substances , Oxidation-Reduction , Protein Binding , Proto-Oncogene Proteins c-jun/chemistry , Proto-Oncogene Proteins c-jun/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...