Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters










Publication year range
1.
Oncogene ; 25(59): 7714-22, 2006 Dec 14.
Article in English | MEDLINE | ID: mdl-16799643

ABSTRACT

The continuous production of the CXC ligand 1 (CXCL1) chemokine by melanoma cells is a major effector of tumor growth. We have previously shown that the constitutive expression of this chemokine is dependent upon transcription factors nuclear factor-kappa B (NF-kappaB), stimulating protein-1 (SP1), high-mobility group-I/Y (HMGI/Y), CAAT displacement protein (CDP) and poly(ADP-ribose) polymerase-1 (PARP-1). In this study, we demonstrate for the first time the mechanism of transcriptional regulation of CXCL1 through PARP-1 in melanoma cells. In its inactive state, PARP-1 binds to the CXCL1 promoter in a sequence-specific manner and prevents binding of NF-kappaB (p65/p50) to its element. However, activation of the PARP-1 enzymatic activity enhances CXCL1 expression, owing to the loss of PARP-1 binding to the CXCL1 promoter, accompanied by enhanced binding of p65 to the promoter. The delineation of the role of NF-kappaB-interacting factors in the putative CXCL1 enhanceosome will provide key information in developing strategies to block constitutive expression of this and other chemokines in cancer and to develop targeted therapy.


Subject(s)
Chemokines, CXC/genetics , Gene Expression Regulation, Neoplastic , Melanoma/genetics , Base Sequence , Cell Line, Tumor , Chemokine CXCL1 , Chemokines, CXC/analysis , Humans , Melanoma/pathology , Molecular Sequence Data , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/physiology , Promoter Regions, Genetic , Transcription Factor RelA/physiology , Transcription, Genetic
2.
Neoplasia ; 3(3): 179-88, 2001.
Article in English | MEDLINE | ID: mdl-11494111

ABSTRACT

The tumor-suppressor p53 undergoes extensive poly(ADP-ribosyl)ation early during apoptosis in human osteosarcoma cells, and degradation of poly(ADP-ribose) (PAR) attached to p53 coincides with poly(ADP-ribose)polymerase-1, (PARP-1) cleavage, and expression of p53 target genes. The mechanism by which poly(ADP-ribosyl)ation may regulate p53 function has now been investigated. Purified wild-type PARP-1 catalyzed the poly(ADP-ribosyl) of full-length p53 in vitro. In gel supershift assays, poly(ADP-ribosyl)ation suppressed p53 binding to its DNA consensus sequence; however, when p53 remained unmodified in the presence of inactive mutant PARP-1, it retained sequence-specific DNA binding activity. Poly(ADP-ribosyl)ation of p53 by PARP-1 during early apoptosis in osteosarcoma cells also inhibited p53 interaction with its DNA consensus sequence; thus, poly(ADP-ribosyl)ation may represent a novel means for regulating transcriptional activation by p53 in vivo.


Subject(s)
Bone Neoplasms/metabolism , Consensus Sequence , DNA-Binding Proteins/metabolism , Osteosarcoma/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Tumor Suppressor Protein p53/physiology , Apoptosis/physiology , Base Sequence , Bone Neoplasms/pathology , DNA/chemistry , DNA-Binding Proteins/genetics , Gene Targeting , Humans , Immunoblotting , Osteosarcoma/pathology , Poly(ADP-ribose) Polymerases/metabolism , Tumor Cells, Cultured
3.
J Biol Chem ; 276(41): 38185-92, 2001 Oct 12.
Article in English | MEDLINE | ID: mdl-11461900

ABSTRACT

During apoptosis, endonucleases cleave DNA into 50-300-kb fragments and subsequently into internucleosomal fragments. DNA fragmentation factor (DFF) is implicated in apoptotic DNA cleavage; this factor comprises DFF45 and DFF40 subunits, the former of which acts as a chaperone and inhibitor of the catalytic subunit and whose cleavage by caspase-3 results in DFF activation. Disruption of the DFF45 gene blocks internucleosomal DNA fragmentation and confers resistance to apoptosis in primary thymocytes. The role of DFF-mediated DNA fragmentation in apoptosis was investigated in primary fibroblasts from DFF45(-/-) and control (DFF45(+/+)) mice. DFF45 deficiency rendered fibroblasts resistant to apoptosis induced by tumor necrosis factor (TNF). TNF induced rapid cleavage of DNA into approximately 50-kb fragments in DFF45(+/+) fibroblasts but not in DFF45(-/-) cells, indicating that DFF mediates this initial step in DNA processing. The TNF-induced activation of poly(ADP-ribose) polymerase (PARP), which requires PARP binding to DNA strand breaks, and the consequent depletion of the PARP substrate NAD were markedly delayed in DFF45(-/-) cells, suggesting a role for DFF in PARP activation. The activation of caspase-3 and mitochondrial events important in apoptotic signaling, including the loss of mitochondrial membrane potential and the release of cytochrome c, induced by TNF were similarly delayed in DFF45(-/-) fibroblasts. DFF45(-/-) and DFF45(+/+) cells were equally sensitive to the DNA-damaging agent and PARP activator N-methyl-N'-nitro-N-nitrosoguanidine. Inhibition of PARP by 3-aminobenzamide partially protected DFF45(+/+) cells against TNF-induced death and inhibited the associated release of cytochrome c and activation of caspase-3. These results suggest that the generation of 50-kb DNA fragments by DFF, together with the activation of PARP, mitochondrial dysfunction, and caspase-3 activation, contributes to an amplification loop in the death process.


Subject(s)
Apoptosis/physiology , Poly(ADP-ribose) Polymerases/physiology , Proteins/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis Regulatory Proteins , Caspase 3 , Caspases/metabolism , Caspases/physiology , Cells, Cultured , Cytochrome c Group/metabolism , Enzyme Activation , Homozygote , Methylnitronitrosoguanidine/pharmacology , Mice , Mitochondria/physiology , Proteins/genetics
4.
Nucleic Acids Res ; 29(3): 841-9, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11160908

ABSTRACT

Poly(ADP-ribose) polymerase (PARP) knockout mice are resistant to murine models of human diseases such as cerebral and myocardial ischemia, traumatic brain injury, diabetes, Parkinsonism, endotoxic shock and arthritis, implicating PARP in the pathogenesis of these diseases. Potent selective PARP inhibitors are therefore being evaluated as novel therapeutic agents in the treatment of these diseases. Inhibition or depletion of PARP, however, increases genomic instability in cells exposed to genotoxic agents. We recently demonstrated the presence of a genomically unstable tetraploid population in PARP(-/-) fibroblasts and its loss after stable transfection with PARP cDNA. To elucidate whether the genomic instability is attributable to PARP deficiency or lack of PARP activity, we investigated the effects of PARP inhibition on development of tetraploidy. Immortalized wild-type and PARP(-/-) fibroblasts were exposed for 3 weeks to 20 microM GPI 6150 (1,11b-dihydro-[2H:]benzopyrano[4,3,2-de]isoquinolin-3-one), a novel small molecule specific competitive inhibitor of PARP (K(i) = 60 nM) and one of the most potent PARP inhibitors to date (IC(50) = 0.15 microM). Although GPI 6150 initially decreased cell growth in wild-type cells, there was no effect on cell growth or viability after 24 h. GPI 6150 inhibited endogenous PARP activity in wild-type cells by approximately 91%, to about the residual levels in PARP(-/-) cells. Flow cytometric analysis of unsynchronized wild-type cells exposed for 3 weeks to GPI 6150 did not induce the development of tetraploidy, suggesting that, aside from its catalytic function, PARP may play other essential roles in the maintenance of genomic stability.


Subject(s)
Benzopyrans/pharmacology , Enzyme Inhibitors/pharmacology , Isoquinolines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Polyploidy , Animals , Cell Division/drug effects , Cell Division/genetics , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , DNA/drug effects , DNA/genetics , DNA/metabolism , Dose-Response Relationship, Drug , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Flow Cytometry/methods , Genotype , Inhibitory Concentration 50 , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Mutant Strains , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism
5.
Toxicol Appl Pharmacol ; 171(2): 94-106, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11222085

ABSTRACT

JP-8 is a kerosene-based fuel widely used by the U.S. military. Various models of human occupational and animal exposure to JP-8 have demonstrated the potential for local and systemic toxicity but the mechanisms involved are unknown. The purpose of our investigation was to study the molecular mechanisms of JP-8 toxicity by using an in vitro model. JP-8 exposure in a rat lung alveolar type II epithelial cell line (RLE-6TN) induces biochemical and morphological markers of apoptotic cell death: caspase-3 activation, poly(ADP-ribose) polymerase (PARP) cleavage, chromatin condensation, membrane blebbing, cytochrome c release from mitochondria, and genomic DNA cleavage into both oligonucleosomal (DNA ladder) and high-molecular-weight (HMW) fragments. The human histiocytic lymphoma cell line (U937) also responds to JP-8 with caspase-3 activation, cleavage of caspase substrates, including PARP, DNA-PK, and lamin B1, and degradation of genomic DNA with the production of HMW fragments. Caspase-3 activation and PARP cleavage also occur in the acute T-cell leukemia cell line (Jurkat) following treatment with JP-8. Furthermore, Jurkat cells stably transfected with a plasmid encoding the antiapoptotic protein Bcl-x(L) or pretreated with the pan-caspase inhibitor Boc-d-fmk, are relatively resistant to the cytotoxic effects of JP-8 compared to control cells. Finally, we demonstrate that PARP cleavage occurs in primary mouse thymocytes exposed to JP-8. In conclusion, our data support the hypothesis that apoptotic cell death is responsible at least partially for the cytotoxic effects of JP-8 and suggest that inhibition of the apoptotic cascade might reduce JP-8 toxicity.


Subject(s)
Apoptosis/drug effects , Hydrocarbons/toxicity , Lung/pathology , Animals , Caspase 3 , Caspase Inhibitors , Caspases/metabolism , Cell Line , Cell Membrane/pathology , Chromatin/ultrastructure , Cytochrome c Group/metabolism , DNA Fragmentation , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Epithelial Cells/pathology , Humans , Hydrocarbons/administration & dosage , Jurkat Cells , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Monocytes/pathology , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/physiology , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Rats , Transfection , Tumor Cells, Cultured , bcl-X Protein
6.
Toxicol Appl Pharmacol ; 171(2): 107-16, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11222086

ABSTRACT

JP-8 induces apoptosis in rat lung epithelial cells, primary mouse T lymphocytes, Jurkat T lymphoma cells, and U937 monocytic cells (Stoica et al., 2001). Here, we have observed a different mechanism of cytotoxicity in human keratinocytes grown in culture as well as when grafted onto nude mice. At lower levels of JP-8 (80 microg/ml; 1 x 10(-4) dilution), sufficient to induce apoptosis in other cell types, including lung epithelial cells (Stoica et al., 2001), no apoptosis was observed. At higher levels (>200 microg/ml; 2.5 x 10(-4) dilution), JP-8 is cytotoxic to both primary and immortalized human keratinocytes, as evidenced by the metabolism of calcein, as well as by morphological changes such as cell rounding and cell detachment. There was no evidence of activation of caspases-3, -7, or -8 either by enzyme activity or immunoblot analysis, and the stable expression of a dominant-negative inhibitor of apoptosis (FADD-DN) did not increase the survival of keratinocytes to JP-8. The pattern of poly(ADP-ribose) polymerase (PARP) cleavage was also characteristic of necrosis. PARP has been also been implicated in necrosis via its ability to lower levels of ATP in damaged cells. However, fibroblasts derived from PARP-/- mice underwent necrotic cell death similar to those derived from PARP+/+ mice, indicating that the effects of JP-8 are independent of PARP. Immunoblot analysis further revealed that exposure of keratinocytes to the toxic higher levels of JP-8 markedly downregulates the expression of the prosurvival members of the Bcl-2 family, Bcl-2 and Bcl-x(L), and upregulates the expression of antisurvival members of this family, including Bad and Bak. Bcl-2 and Bcl-x(L) have been shown to preserve mitochondrial integrity and suppress cell death. In contrast, Bak and Bad both promote cell death by alteration of the mitochondrial membrane potential, in part by heterodimerization with and inactivation of Bcl-2 and Bcl-x(L), and either inducing necrosis or activating a downstream caspase program. High intrinsic levels of Bcl-2 and Bcl-x(L) may prevent apoptotic death of keratinocytes at lower levels of JP-8, while perturbation of the balance between pro- and antiapoptotic Bcl-2 family members at higher levels may ultimately play a role in necrotic cell death in human keratinocytes. Finally, when human keratinocytes were grafted to form a human epidermis on nude mice, treatment of these grafts with JP-8 revealed cytotoxicity and altered histology in vivo.


Subject(s)
Apoptosis/drug effects , Fibroblasts/pathology , Hydrocarbons/toxicity , Keratinocytes/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Skin/pathology , Animals , Caspase 3 , Caspases/metabolism , Cell Line , Cells, Cultured , Enzyme Activation/drug effects , Epithelial Cells/pathology , Gene Expression/drug effects , Humans , Lung/pathology , Male , Mice , Mice, Nude , Necrosis , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Skin Transplantation
7.
J Invest Dermatol ; 117(6): 1566-73, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11886524

ABSTRACT

Sulfur mustard is cytotoxic to dermal fibroblasts as well as epidermal keratinocytes. We demonstrated that poly(ADP-ribose) polymerase (PARP) modulates Fas-mediated apoptosis, and other groups and we have shown that PARP plays a role in the modulation of other types of apoptotic and necrotic cell death. We have now utilized primary dermal fibroblasts, immortalized fibroblasts, and keratinocytes derived from PARP(-/-) mice and their wildtype littermates (PARP(+/+)) to determine the contribution of PARP to sulfur mustard toxicity. Following sulfur mustard exposure, primary skin fibroblasts from PARP-deficient mice demonstrated increased internucleosomal DNA cleavage, caspase-3 processing and activity, and annexin V positivity, compared to those derived from PARP(+/+) animals. Conversely, propidium iodide staining, PARP cleavage patterns, and random DNA fragmentation revealed a dose-dependent increase in necrosis in PARP(+/+) but not PARP(-/-) cells. Using immortalized PARP(-/-) fibroblasts stably transfected with the human PARP cDNA or with empty vector alone, we show that PARP inhibits markers of apoptosis in these cells as well. Finally, primary keratinocytes were derived from newborn PARP(+/+) and PARP(-/-) mice and immortalized with the E6 and E7 genes of human papilloma virus. In contrast to fibroblasts, keratinocytes from both PARP(-/-) and PARP(+/+) mice express markers of apoptosis in response to sulfur mustard exposure. The effects of PARP on the mode of cell death in different skin cell types may determine the severity of vesication in vivo, and thus have implications for the design of PARP inhibitors to reduce sulfur mustard pathology.


Subject(s)
Apoptosis/drug effects , Dermatologic Agents/toxicity , Fibroblasts/cytology , Keratinocytes/cytology , Mustard Gas/toxicity , Poly(ADP-ribose) Polymerases/metabolism , Apoptosis/physiology , Caspase 3 , Caspases/metabolism , Epidermis/pathology , Fibroblasts/drug effects , Fibroblasts/enzymology , Humans , Keratinocytes/drug effects , Keratinocytes/enzymology , Necrosis , Wound Healing/physiology
8.
Diabetes ; 49(11): 1849-55, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11078451

ABSTRACT

This study was designed to test the hypothesis that poly(ADP-ribose) polymerase (PARP) plays a role in the repair of damage to mitochondrial DNA (mtDNA). A rat insulinoma cell line was transfected with a PARP antisense vector that was under the control of a dexamethasone promoter. Transfected cells were selected for stable integration of the antisense vector. Several cell lines containing the antisense vector were isolated. For these studies, one of these lines (clone 5) was chosen for further evaluation. When cells were treated with dexamethasone for 72 h, PARP activity was diminished by 60%. Western blot analysis revealed a concomitant reduction in PARP protein. When clone 5 cells were exposed to the simple methylating agent methylnitrosourea (MNU) without previous treatment with dexamethasone, repair of lesions in mtDNA was found to be similar to that seen in wild-type cells or in wild-type cells treated with dexamethasone. However, when clone 5 cells were pretreated with dexamethasone for 72 h, repair of MNU-induced damage was significantly inhibited. To ascertain whether the PARP activity that was inhibited by the antisense treatment was the same as that found in the nucleus, repair studies were performed on fibroblasts derived from PARP knockout mice and their normal wild-type controls. Attenuated repair was also seen in the cells in which the gene for PARP was inactivated. These are the first studies to demonstrate that PARP can facilitate the repair of simple alkylation damage to mtDNA.


Subject(s)
DNA Repair , DNA, Mitochondrial/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Purines/metabolism , Alkylation , Animals , Apoptosis , Dexamethasone/pharmacology , Genetic Vectors , Glucocorticoids/pharmacology , Insulinoma/enzymology , Methylation , Methylnitrosourea/pharmacology , Mice , Mice, Knockout , Poly(ADP-ribose) Polymerases/genetics , Polymerase Chain Reaction , RNA, Antisense/genetics , Rats , Transfection , Tumor Cells, Cultured
9.
Proc Natl Acad Sci U S A ; 97(21): 11274-9, 2000 Oct 10.
Article in English | MEDLINE | ID: mdl-11016956

ABSTRACT

Poly(ADP-ribose) polymerase (PARP) is implicated in the maintenance of genomic integrity, given that inhibition or depletion of this enzyme increases genomic instability in cells exposed to genotoxic agents. We previously showed that immortalized fibroblasts derived from PARP(-/-) mice exhibit an unstable tetraploid population, and partial chromosomal gains and losses in PARP(-/-) mice and immortalized fibroblasts are accompanied by changes in the expression of p53, Rb, and c-Jun, as well as other proteins. A tetraploid population has also now been detected in primary fibroblasts derived from PARP(-/-) mice. Oligonucleotide microarray analysis was applied to characterize more comprehensively the differences in gene expression between asynchronously dividing primary fibroblasts derived from PARP(-/-) mice and their wild-type littermates. Of the 11,000 genes monitored, 91 differentially expressed genes were identified. The loss of PARP results in down-regulation of the expression of several genes involved in regulation of cell cycle progression or mitosis, DNA replication, or chromosomal processing or assembly. PARP deficiency also up-regulates genes that encode extracellular matrix or cytoskeletal proteins that are implicated in cancer initiation or progression or in normal or premature aging. These results provide insight into the mechanism by which PARP deficiency impairs mitotic function, thereby resulting in the genomic alterations and chromosomal abnormalities as well as in altered expression of genes that may contribute to genomic instability, cancer, and aging.


Subject(s)
Gene Expression Regulation/physiology , Poly(ADP-ribose) Polymerases/physiology , Animals , Cellular Senescence/genetics , DNA Repair , DNA Replication , Female , Fibroblasts/enzymology , Fibroblasts/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/pathology , Poly(ADP-ribose) Polymerases/genetics
11.
J Biol Chem ; 275(28): 21302-8, 2000 Jul 14.
Article in English | MEDLINE | ID: mdl-10807908

ABSTRACT

Apoptosis is characterized by various cell morphological and biochemical features, one of which is the internucleosomal degradation of genomic DNA. The role of the human chromatin-bound Ca(2+)- and Mg(2+)-dependent endonuclease (CME) DNAS1L3 and its inhibition by poly(ADP-ribosyl)ation in the DNA degradation that accompanies apoptosis was investigated. The nuclear localization of this endonuclease is the unique feature that distinguishes it from other suggested apoptotic nucleases. Purified recombinant DNAS1L3 was shown to cleave nuclear DNA into both high molecular weight and oligonucleosomal fragments in vitro. Furthermore, exposure of mouse skin fibroblasts expressing DNAS1L3 to inducers of apoptosis resulted in oligonucleosomal DNA fragmentation, an effect not observed in cells not expressing this CME, as well as in a decrease in cell viability greater than that apparent in the control cells. Recombinant DNAS1L3 was modified by recombinant human poly(ADP-ribose) polymerase (PARP) in vitro, resulting in a loss of nuclease activity. The DNAS1L3 protein also underwent poly(ADP-ribosyl)ation in transfected mouse skin fibroblasts in response to inducers of apoptosis. The cleavage and inactivation of PARP by a caspase-3-like enzyme late in apoptosis were associated with a decrease in the extent of DNAS1L3 poly(ADP-ribosyl)ation, which likely releases DNAS1L3 from inhibition and allows it to catalyze the degradation of genomic DNA.


Subject(s)
Apoptosis/physiology , DNA Fragmentation , Endodeoxyribonucleases/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Animals , Calcium Chloride/pharmacology , Caspase 3 , Caspases/metabolism , Cell Line , Cell Survival , Endodeoxyribonucleases/antagonists & inhibitors , Fibroblasts/cytology , Fibroblasts/enzymology , Fibroblasts/physiology , Humans , Kinetics , Magnesium Chloride/pharmacology , Mice , Recombinant Proteins/metabolism , Skin/enzymology
12.
J Appl Toxicol ; 20 Suppl 1: S13-7, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11428624

ABSTRACT

We previously reported that, in normal human epidermal keratinocytes (NHEK) cultures exposed to the alkylating compound sulfur mustard (bis-(2-chloroethyl) sulfide, HD, 0.3-1 mM), there is a rapid (< or =1 h) activation (100% above unexposed control) of the DNA repair enzyme DNA ligase I (130 kD) followed by a first-order decay (1-5 h). The DNA ligase activation is accompanied by a time-dependent (0.5-4 h) and significant DNA repair. Inhibition of another putative DNA repair enzyme, poly(ADP-ribose) polymerase (PARP), by using 3-amino benzamide does not affect DNA ligase activation following HD exposure, but increases the half-life of the activated enzyme threefold. To examine the role of PARP in HD-induced DNA ligase activation and subsequent DNA repair, we conducted studies using cultured keratinocytes in which the level of PARP had been selectively lowered (> or =85%) by the use of induced expression of antisense RNA. In these cells, there was no stimulation of DNA ligase up to 3 h, and a small stimulation (ca. 30% above unexposed control at 5-6 h after HD exposure. A time-course (0.5-6 h) study of DNA repair in HD-exposed PARP-deficient keratinocytes revealed a much slower rate of repair compared with HD-exposed NHEK. The results suggest an active role of PARP in DNA ligase activation and DNA repair in mammalian cells, and also indicate that modulation of PARP-mediated mechanisms may provide a useful approach in preventing HD toxicity.


Subject(s)
DNA Repair , Dermatologic Agents/adverse effects , Mustard Gas/adverse effects , Poly(ADP-ribose) Polymerases/metabolism , Cell Culture Techniques , DNA Ligase ATP , DNA Ligases/biosynthesis , DNA Ligases/metabolism , Gene Expression Regulation , Humans , Keratinocytes/drug effects , Keratinocytes/pathology
13.
J Appl Toxicol ; 20 Suppl 1: S43-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11428642

ABSTRACT

We describe two pathways by which the vesicating agent sulfur mustard (HD) may cause basal cell death and detachment: induction of terminal differentiation and apoptosis. Following treatment of normal human epidermal keratinocytes (NHEK) with 10 or 100 microM HD, the differentiation-specific keratin pair K1/K10 was induced and the cornified envelope precursor protein, involucrin, was cross-linked by epidermal transglutaminase. Fibronectin levels were reduced in a time- and dose-dependent manner. The rapid increase in p53 and decrease in Bcl-2 levels was consistent not only with epidermal differentiation but with apoptosis as well. Further examination of biochemical markers of apoptosis following treatment of either NHEK or human papillomavirus (HPV)-immortalized keratinocytes revealed a burst of poly(ADP-ribose) synthesis, specific cleavage of poly(ADP-ribose)polymerase (PARP) in vivo and in vitro into characteristic 89 and 24 kDa fragments, processing of caspase-3 into its active form and the formation of DNA ladders. The intracellular calcium chelator BAPTA suppressed the differentiation markers, whereas antisense oligonucleotides and chemical inhibitors specific for calmodulin blocked both markers of differentiation and apoptosis. Modulation of p53 levels utilizing retroviral constructs expressing the E6, E7 or E6 + E7 genes of HPV-16 revealed that HD-induced apoptosis was partially p53-dependent. Finally, immortalized fibroblasts derived from PARP -/- 'knockout mice' were exquisitely sensitive to HD-induced apoptosis. These cells became HD resistant when wild-type PARP was stably expressed in these cells. These results indicate that HD exerts its effects via calmodulin, 3 and PARP-sensitive pathways.


Subject(s)
Apoptosis/drug effects , Calmodulin/pharmacology , Cell Differentiation/drug effects , Dermatologic Agents/pharmacology , Mustard Gas/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Apoptosis/physiology , Calmodulin/metabolism , Cell Culture Techniques , Epithelial Cells/drug effects , Genes, p53/drug effects , Humans , Keratinocytes , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Tumor Suppressor Protein p53/biosynthesis
14.
Proc Natl Acad Sci U S A ; 96(23): 13191-6, 1999 Nov 09.
Article in English | MEDLINE | ID: mdl-10557296

ABSTRACT

Depletion of poly(ADP-ribose) polymerase (PARP) increases the frequency of recombination, gene amplification, sister chromatid exchanges, and micronuclei formation in cells exposed to genotoxic agents, implicating PARP in the maintenance of genomic stability. Flow cytometric analysis now has revealed an unstable tetraploid population in immortalized fibroblasts derived from PARP(-/-) mice. Comparative genomic hybridization detected partial chromosomal gains in 4C5-ter, 5F-ter, and 14A1-C1 in PARP(-/-)mice and immortalized PARP(-/-)fibroblasts. Neither the chromosomal gains nor the tetraploid population were apparent in PARP(-/-) cells stably transfected with PARP cDNA [PARP(-/-)(+PARP)], indicating negative selection of cells with these genetic aberrations after reintroduction of PARP cDNA. Although the tumor suppressor p53 was not detectable in PARP(-/-) cells, p53 expression was partially restored in PARP(-/-) (+PARP) cells. Loss of 14D3-ter that encompasses the tumor suppressor gene Rb-1 in PARP(-/-) mice was associated with a reduction in retinoblastoma(Rb) expression; increased expression of the oncogene Jun was correlated with a gain in 4C5-ter that harbors this oncogene. These results further implicate PARP in the maintenance of genomic stability and suggest that altered expression of p53, Rb, and Jun, as well as undoubtedly many other proteins may be a result of genomic instability associated with PARP deficiency.


Subject(s)
Chromosome Aberrations , Poly(ADP-ribose) Polymerases/genetics , Transfection , Animals , Cell Line, Transformed , DNA, Complementary , Female , Gene Expression Regulation, Enzymologic/genetics , Genes, Retinoblastoma , Genes, jun , Genes, p53 , Genome , Mice , Mice, Inbred C57BL , Mice, Knockout , Nucleic Acid Hybridization
15.
Oncogene ; 18(36): 5015-23, 1999 Sep 09.
Article in English | MEDLINE | ID: mdl-10490838

ABSTRACT

E2F-1, a transcription factor implicated in the activation of genes required for S phase such as DNA pol alpha, is regulated by interactions with Rb and by cell-cycle dependent alterations in E2F-1 abundance. We have shown that depletion of poly(ADP-ribose) polymerase (PARP) by antisense RNA expression downregulates pol alpha and E2F-1 expression during early S phase. To examine the role of PARP in the regulation of pol alpha and E2F-1 gene expression, we utilized immortalized mouse fibroblasts derived from wild-type and PARP knockout (PARP-/-) mice as well as PARP-/- cells stably transfected with PARP cDNA [PARP-/-(+PARP)]. After release from serum deprivation, wild-type and PARP-/-(+PARP) cells, but not PARP-/- cells, exhibited a peak of cells in S phase by 16 h and had progressed through the cell cycle by 22 h. Whereas [3H]thymidine incorporation remained negligible in PARP-/- cells, in vivo DNA replication maximized after 18 h in wild-type and PARP-/-(+PARP) cells. To investigate the effect of PARP on E2F-1 promoter activity, a construct containing the E2F-1 gene promoter fused to a luciferase reporter gene was transiently transfected into these cells. E2F-1 promoter activity in control and PARP-/-(+PARP) cells increased eightfold after 9 h, but not in PARP-/- cells. PARP-/- cells did not show the marked induction of E2F-1 expression during early S phase apparent in control and PARP-/-(+PARP) cells. RT - PCR analysis and pol alpha activity assays revealed the presence of pol alpha transcripts and a sixfold increase in activity in both wild-type and PARP-/-(+PARP) cells after 20 h, but not in PARP-/- cells. These results suggest that PARP plays a role in the induction of E2F-1 promoter activity, which then positively regulates both E2F-1 and pol alpha expression, when quiescent cells reenter the cell cycle upon recovery from aphidicolin exposure or removal of serum.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , DNA Polymerase I/genetics , DNA-Binding Proteins , Poly(ADP-ribose) Polymerases/metabolism , Promoter Regions, Genetic , S Phase , Transcription Factors/genetics , Up-Regulation , Animals , Base Sequence , Culture Media, Serum-Free , DNA Polymerase I/metabolism , DNA Primers , E2F Transcription Factors , E2F1 Transcription Factor , Mice , Poly(ADP-ribose) Polymerases/genetics , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1
16.
Mol Cell Biochem ; 193(1-2): 137-48, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10331650

ABSTRACT

We have focused on the roles of PARP and poly(ADP-ribosyl)ation early in apoptosis, as well as during the early stages of differentiation-linked DNA replication. In both nuclear processes, a transient burst of PAR synthesis and PARP expression occurs early, prior to internucleosomal DNA cleavage before commitment to apoptosis as well as at the round of DNA replication prior to the onset of terminal differentiation. In intact human osteosarcoma cells undergoing spontaneous apoptosis, both PARP and PAR decreased after this early peak, concomitant with the inactivation and cleavage of PARP by caspase-3 and the onset of substantial DNA and nuclear fragmentation. Whereas 3T3-L1, osteosarcoma cells, and immortalized PARP +/+ fibroblasts exhibited this early burst of PAR synthesis during Fas-mediated apoptosis, neither PARP-depleted 3T3-L1 PARP-antisense cells nor PARP -/- fibroblasts showed this response. Consequently, whereas control cells progressed into apoptosis, as indicated by induction of caspase-3-like PARP-cleavage activity, PARP-antisense cells and PARP -/- fibroblasts did not, indicating a requirement for PARP and poly(ADP-ribosyl)ation of nuclear proteins at an early reversible stage of apoptosis. In parallel experiments, a transient increase in PARP expression and activity were also noted in 3T3-L1 preadipocytes 24 h after induction of differentiation, a stage at which approximately 95% of the cells were in S-phase, but not in PARP-depleted antisense cells, which were consequently unable to complete the round of DNA replication required for differentiation. PARP, a component of the multiprotein DNA replication complex (MRC) that catalyzes viral DNA replication in vitro, poly(ADP-ribosyl)ates 15 of approximately 40 MRC proteins, including DNA pol alpha, DNA topo I, and PCNA. Depletion of endogenous PARP by antisense RNA expression in 3T3-L1 cells results in MRCs devoid of any DNA pol alpha and DNA pol delta activities. Surprisingly, there was no new expression of PCNA and DNA pol alpha, as well as the transcription factor E2F-1 in PARP-antisense cells during entry into S-phase, suggesting that PARP may play a role in the expression of these proteins, perhaps by interacting with a site in the promoters for these genes.


Subject(s)
Apoptosis , DNA Replication/physiology , Poly(ADP-ribose) Polymerases/physiology , Proteins/physiology , 3T3 Cells , Animals , Caspase 3 , Caspases/metabolism , HL-60 Cells , Humans , Jurkat Cells , Mice , Osteosarcoma/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Proteins/metabolism , RNA, Antisense/metabolism , S Phase , Time Factors , Tumor Cells, Cultured
17.
Proc Natl Acad Sci U S A ; 96(10): 5774-9, 1999 May 11.
Article in English | MEDLINE | ID: mdl-10318960

ABSTRACT

1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is a neurotoxin that causes parkinsonism in humans and nonhuman animals, and its use has led to greater understanding of the pathogenesis of Parkinson's disease. However, its molecular targets have not been defined. We show that mice lacking the gene for poly(ADP-ribose) polymerase (PARP), which catalyzes the attachment of ADP ribose units from NAD to nuclear proteins after DNA damage, are dramatically spared from MPTP neurotoxicity. MPTP potently activates PARP exclusively in vulnerable dopamine containing neurons of the substantia nigra. MPTP elicits a novel pattern of poly(ADP-ribosyl)ation of nuclear proteins that completely depends on neuronally derived nitric oxide. Thus, NO, DNA damage, and PARP activation play a critical role in MPTP-induced parkinsonism and suggest that inhibitors of PARP may have protective benefit in the treatment of Parkinson's disease.


Subject(s)
Parkinson Disease, Secondary/metabolism , Proteins/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , 1-Methyl-4-phenylpyridinium , Animals , DNA Damage , Enzyme Activation/drug effects , Immunohistochemistry , Mice , Mice, Knockout , Monoamine Oxidase/metabolism , Nitric Oxide Synthase/metabolism , Nuclear Proteins/metabolism , Parkinson Disease, Secondary/chemically induced , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases , Substantia Nigra/drug effects
18.
Cancer Res ; 59(9): 2190-4, 1999 May 01.
Article in English | MEDLINE | ID: mdl-10232607

ABSTRACT

Spontaneous apoptosis in human osteosarcoma cells was observed to be associated with a marked increase in the intracellular abundance of p53. Immunoprecipitation and immunoblot analysis revealed that, together with a variety of other nuclear proteins, p53 undergoes extensive poly(ADP-ribosyl)ation early during the apoptotic program in these cells. Subsequent degradation of poly(ADP-ribose) (PAR), attached to p53 presumably by PAR glycohydrolase, the only reported enzyme to degrade PAR, was apparent concomitant with the onset of proteolytic processing and activation of caspase-3, caspase-3-mediated cleavage of poly(ADP-ribose) polymerase (PARP), and internucleosomal DNA fragmentation during the later stages of cell death. The decrease in PAR covalently bound to p53 also coincided with the marked induction of expression of the p53-responsive genes bax and Fas. These results suggest that poly(ADP-ribosyl)ation may play a role in the regulation of p53 function and implies a regulatory role for PARP and/or PAR early in apoptosis.


Subject(s)
Apoptosis/physiology , Bone Neoplasms/pathology , Neoplasm Proteins/metabolism , Osteosarcoma/pathology , Poly Adenosine Diphosphate Ribose/metabolism , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-bcl-2 , Tumor Suppressor Protein p53/metabolism , Bone Neoplasms/metabolism , Caspase 3 , Caspases/metabolism , DNA Fragmentation , Enzyme Activation , Gene Expression Regulation, Neoplastic , Glycoside Hydrolases/metabolism , Humans , Osteosarcoma/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Tumor Cells, Cultured , bcl-2-Associated X Protein , fas Receptor/biosynthesis , fas Receptor/genetics
19.
Nucleic Acids Res ; 27(9): 1999-2005, 1999 May 01.
Article in English | MEDLINE | ID: mdl-10198433

ABSTRACT

Ca2+- and Mg2+-dependent endonucleases have been implicated in DNA fragmentation during apoptosis. We have demonstrated that particular nucleases of this type are inhibited by poly(ADP-ribosyl)ation and suggested that subsequent cleavage of PARP by caspase-3 might release these nucleases from poly(ADP-ribosyl)ation-induced inhibition. Hence, we purified and partially sequenced such a nuclease isolated from bovine seminal plasma and identified human, rat and mouse homologs of this enzyme. The extent of sequence homology among these nucleases indicates that these four proteins are orthologous members of the family of DNase I-related enzymes. We demonstrate that the activation of the human homolog previously specified as DNAS1L3 can induce Ca2+- and Mg2+-dependent DNA fragmentation in vitro and in vivo. RT-PCR analysis failed to detect DNAS1L3 mRNA in HeLa cells and nuclei isolated from these cells did not exhibit internucleosomal DNA fragmentation when incubated in the presence of Ca2+and Mg2+. However, nuclei isolated from HeLa cells that had been stably transfected with DNAS1L3 cDNA underwent such DNA fragmentation in the presence of both ions. The Ca2+ionophore ionomycin also induced internucleosomal DNA degradation in transfected but not in control HeLa cells. Transverse alternating field electrophoresis revealed that in nuclei from transfected HeLa cells, but not in those from control cells, DNA was cleaved into fragments of >1000 kb in the presence of Mg2+; addition of Ca2+in the presence of Mg2+resulted in processing of the >1000 kb fragments into 50 kb and oligonucleosomal fragments. These results demonstrate that DNAS1L3 is necessary for Ca2+- and Mg2+-dependent cleavage of DNA into both oligonucleosomal and high molecular mass fragments in specific cell types.


Subject(s)
Calcium/metabolism , DNA/metabolism , Endodeoxyribonucleases/metabolism , Magnesium/metabolism , Nucleosomes/metabolism , Amino Acid Sequence , Animals , Base Sequence , DNA/chemistry , DNA Fragmentation , Endodeoxyribonucleases/chemistry , HeLa Cells , Humans , Hydrolysis , Molecular Sequence Data , Molecular Weight , Sequence Homology, Amino Acid
20.
Cancer Res ; 58(20): 4510-4, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9788591

ABSTRACT

The antiangiogenic, tubulin-binding drug combretastatin A-4 exhibits a selective toxicity for proliferating endothelial cells in vitro and induces vascular shutdown in tumor models in vivo. The mechanism of combretastatin A-4 cytotoxicity has now been investigated with cultured proliferating human umbilical vein endothelial cells by examining various markers of apoptosis. Incubation of cells with 0.1 mM combretastatin A-4 induced the conversion (first detected after 6 h) of the CPP32 proenzyme to active caspase-3, a cysteine protease that plays an important role in apoptosis in many cell types; the drug also increased caspase-3 activity. Another early event observed was the binding of annexin V to 50% of the cells 8 h after drug treatment. Internucleosomal DNA fragmentation, another hallmark of apoptosis, was detected in cells incubated with 0.1 mM combretastatin A-4 for 24 h. Staining with Hoechst 33258 revealed that about 75% of cells exhibited a nuclear morphology characteristic of apoptosis after incubation with drug for 24 h. Incubation of cells for up to 8 h with combretastatin A-4 did not induce the release of lactate dehydrogenase or increase the uptake of propidium iodide, both indicators of membrane integrity. These results indicate that the selective cytotoxic effect of combretastatin A-4 is mediated by the induction of apoptosis rather than by necrosis and may provide an enhanced clinical strategy in cancer chemotherapy with this new agent.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Endothelium, Vascular/drug effects , Stilbenes/pharmacology , Animals , Caspase 3 , Caspases/metabolism , Cell Membrane/drug effects , Cell Nucleus/drug effects , DNA Fragmentation/drug effects , Humans , L-Lactate Dehydrogenase/metabolism , Mice
SELECTION OF CITATIONS
SEARCH DETAIL