Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Oncogene ; 33(1): 108-15, 2014 Jan 02.
Article in English | MEDLINE | ID: mdl-23178491

ABSTRACT

Checkpoint kinase 2 (CHK2) kinase is a key mediator in many cellular responses to genotoxic stresses, including ionizing radiation (IR) and topoisomerase inhibitors. Upon IR, CHK2 is activated by ataxia telangiectasia mutated kinase and regulates the S-phase and G1-S checkpoints, apoptosis and DNA repair by phosphorylating downstream target proteins, such as p53 and Brca1. In addition, CHK2 is thought to be a multi-organ cancer susceptibility gene. In this study, we used a tandem affinity purification strategy to identify proteins that interact with CHK2 kinase. Cyclin-dependent kinase 11 (CDK11)(p110) kinase, implicated in pre-mRNA splicing and transcription, was identified as a CHK2-interacting protein. CHK2 kinase phosphorylated CDK11(p110) on serine 737 in vitro. Unexpectedly, CHK2 kinase constitutively phosphorylated CDK11(p110) in a DNA damage-independent manner. At a molecular level, CDK11(p110) phosphorylation was required for homodimerization without affecting its kinase activity. Overexpression of CHK2 promoted pre-mRNA splicing. Conversely, CHK2 depletion decreased endogenous splicing activity. Mutation of the phosphorylation site in CDK11(p110) to alanine abrogated its splicing-activating activity. These results provide the first evidence that CHK2 kinase promotes pre-mRNA splicing via phosphorylating CDK11(p110).


Subject(s)
Checkpoint Kinase 2/physiology , Cyclin-Dependent Kinases/metabolism , RNA Precursors/genetics , RNA, Messenger/genetics , Amino Acid Sequence , Checkpoint Kinase 2/chemistry , Cyclin-Dependent Kinases/chemistry , DNA Damage , HEK293 Cells , HT29 Cells , Humans , Phosphorylation , Protein Interaction Mapping , Protein Multimerization , Protein Processing, Post-Translational , RNA Precursors/metabolism , RNA Splicing , RNA, Messenger/metabolism
2.
Oncogene ; 25(41): 5601-11, 2006 Sep 14.
Article in English | MEDLINE | ID: mdl-16636671

ABSTRACT

Bcl-2 can both promote and attenuate tumorigenesis. Although the former function is relatively well characterized, the mechanism of the latter remains elusive. We report here that enforced Bcl-2 expression in MCF7 cells stabilizes p53, induces phosphorylation of p53 serine 15 (p53pSer15) and inhibits MCF7 cell growth. Consistent with p53 Ser15 being a target of ataxia telangiectasia mutated protein(ATM)/ATR (ATM- and rad3-related) in the DNA damage response, Bcl-2 activates ATM by inducing ATM Ser1981 phosphorylation, which is accompanied with the phosphorylaton of two additional ATM substrates, Chk2 Thr68 and H2AX Ser139. Downregulation of ATM using a specific small interference RNA fragment (ATMRNAi) abolished Bcl-2-induced p53pSer15 and Bcl-2-mediated growth inhibition of MCF7 cells. Ectopic expression of a dominant-negative p53 mutant, p53175H, partially rescued this growth inhibition. Taken together, these observations demonstrate the contribution of ATM-p53 function to Bcl-2-mediated inhibition of MCF7 cell growth, indicating an ATM-mediated surveillance system for regulating Bcl-2 overexpression. Consistent with this concept, we found that MCF7 cells express Bcl-2 heterogeneously with 34.5% of cells being Bcl-2 negative. In general, Bcl-2-positive MCF7 cells proliferate slower than those of Bcl-2 negative. Thus, we provide evidence suggesting that activation of ATM suppresses Bcl-2-induced tumorigenesis, and that attenuation of ATM function may be an important event in breast cancer progression.


Subject(s)
Cell Cycle Proteins/physiology , DNA-Binding Proteins/physiology , Gene Expression/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Suppressor Proteins/physiology , Ataxia Telangiectasia Mutated Proteins , Base Sequence , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Division/genetics , Cell Line, Tumor , DNA Primers , Fluorescent Antibody Technique , Humans , RNA, Small Interfering , Tumor Suppressor Protein p53/physiology
3.
J Mol Med (Berl) ; 79(8): 428-36, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11511973

ABSTRACT

Neuroblastomas that overexpress N-Myc due to amplification of the MYCN oncogene are aggressive tumors that become very resistant to treatment by chemotherapy and irradiation. to identify tumor suppressor genes in this group of neuroblastomas we analyzed the expression and function of both apoptosis-related cell cycle regulatory genes in cell lines and patient tumor samples. We found that in a high percentage of neuroblastoma cell lines and patient samples with amplified MYCN, caspase-8 mRNA is not expressed. The caspase-8 gene, CASP8, was deleted or silenced by methylation in the neuroblastoma cell lines while methylation of its promoter region was the predominant mechanism for its inactivation in the patient tumor samples. Reintroduction of caspase-8 into the neuroblastoma cell lines resensitized these cells to drug-induced and survival factor dependent apoptosis. Subsequently others have also shown that caspase-8 is silenced by methylation in neuroblastoma and peripheral neural ectodermal tumors, and that the caspase-9 regulator Apaf-1 is silenced by methylation in melanoma cell lines and patient samples. We conclude that caspase-8 acts as a tumor suppressor gene in neuroblastomas, that its silencing provides a permissive environment for MYCN gene amplification once the tumors are treated with chemotherapeutic drugs/irradiation, and that expression of this gene in these tumor cells may be of clinical benefit. We also discuss the possible significance of the neural crest cell progenitor cell origin and the silencing of important apoptotic regulators via methylation in both neuroblastoma and melanoma tumors.


Subject(s)
Apoptosis , Caspases/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Apoptotic Protease-Activating Factor 1 , Caspase 8 , Caspase 9 , Caspases/genetics , Child , Chromosomes, Human, Pair 1/genetics , DNA Methylation , Drug Resistance, Neoplasm , Gene Amplification/genetics , Humans , Loss of Heterozygosity/genetics , Neuroblastoma/enzymology , Proteins/metabolism , fas Receptor/metabolism
4.
Semin Cell Dev Biol ; 11(3): 191-201, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10906276

ABSTRACT

Much of the proteolysis that occurs during apoptosis is directed by caspases, a family of related cysteinyl proteases. A relatively small number of cellular proteins are targeted by caspases, yet their function is dramatically affected and apoptosis is triggered. Other proteases, such as granzymes and calpain, are also involved in the apoptotic signaling process, but in a much more cell type- and/or stimulus type-specific manner. At least three distinct caspase-signaling pathways exist; one activated through ligand-dependent death receptor oligomerization, the second through mitochondrial disruption, and the third through stress-mediated events involving the endoplasmic reticulum. These pathways also appear to interact to amplify weak apoptotic signals and shorten cellular execution time. Finally, defects in caspases contribute to autoimmune disease, cancer and certain neurological disorders.


Subject(s)
Apoptosis , Caspases/metabolism , Proteins/metabolism , Alzheimer Disease/metabolism , Calpain/metabolism , Cathepsin D/metabolism , Enzyme Activation , Granzymes , Humans , Huntington Disease/metabolism , Models, Biological , Serine Endopeptidases/metabolism , Signal Transduction
5.
Nat Med ; 6(5): 529-35, 2000 May.
Article in English | MEDLINE | ID: mdl-10802708

ABSTRACT

Caspase 8 is a cysteine protease regulated in both a death-receptor-dependent and -independent manner during apoptosis. Here, we report that the gene for caspase 8 is frequently inactivated in neuroblastoma, a childhood tumor of the peripheral nervous system. The gene is silenced through DNA methylation as well as through gene deletion. Complete inactivation of CASP8 occurred almost exclusively in neuroblastomas with amplification of the oncogene MYCN. Caspase 8-null neuroblastoma cells were resistant to death receptor- and doxorubicin-mediated apoptosis, deficits that were corrected by programmed expression of the enzyme. Thus, caspase 8 acts as a tumor suppressor in neuroblastomas with amplification of MYCN.


Subject(s)
Caspases/genetics , Gene Amplification , Gene Silencing , Genes, myc , Neuroblastoma/genetics , Antineoplastic Agents/pharmacology , Apoptosis , Caspase 8 , Caspase 9 , Caspases/biosynthesis , Child , DNA Methylation , Doxorubicin/pharmacology , Gene Deletion , Gene Expression Regulation, Neoplastic , Humans , Recombinant Proteins/biosynthesis , Retroviridae/genetics , Signal Transduction , Tumor Cells, Cultured
6.
J Biol Chem ; 275(13): 9303-7, 2000 Mar 31.
Article in English | MEDLINE | ID: mdl-10734071

ABSTRACT

There are at least two distinct classes of caspases, initiators (e.g. caspases-8, -9, and -10) and effectors (e.g. caspase-3). Furthermore, it is believed that there are two distinct primary apoptotic signaling pathways, one of which is mediated by death receptors controlled by caspases-8/10, and the other by the release of cytochrome c and activation of a caspase-9/Apaf1/cytochrome c apoptosome. However, several recent reports have demonstrated that caspase-8, and its substrate Bid, are frequently activated in response to certain apoptotic stimuli in a death receptor-independent manner. These results suggest that significant cross-talk may exist between these two distinct signaling arms, allowing each to take advantage of elements unique to the other. Here we provide evidence that activation of caspase-8, and subsequent Bid cleavage, does indeed participate in cytochrome c-mediated apoptosis, at least in certain circumstances and cell types. Furthermore, the participation of activated caspase-3 is essential for activation of caspase-8 and Bid processing to occur. Although caspase-8 activation is not required for the execution of a cytochrome c-mediated death signal, we found that it greatly shortens the execution time. Thus, caspase-8 involvement in cytochrome c-mediated cell death may help to amplify weaker death signals and ensure that apoptosis occurs within a certain time frame.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Caspases/metabolism , Staurosporine/pharmacology , Breast Neoplasms/enzymology , Caspase 3 , Caspase 8 , Caspase 9 , Cytochrome c Group/metabolism , Enzyme Activation , Humans , Hydrolysis , Tumor Cells, Cultured
7.
Nucleic Acids Res ; 28(4): 917-24, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10648783

ABSTRACT

Recently, we cloned two highly related human genes, hChlR1 ( DDX11 ) and hChlR2 ( DDX12 ), which appear to be homologs of the Saccharomyces cerevisiae CHL1 gene. Nucleotide sequence analysis suggests that these genes encode new members of the DEAH family of DNA helicases. While the enzymatic activity of CHL1 has not been characterized, the protein is required for the maintenance of high fidelity chromosome segregation in yeast. Here we report that the hChlR1 protein is a novel human DNA helicase. We have expressed and purified hChlR1 using a baculovirus system and analyzed its enzymatic activity. The recombinant hChlR1 protein possesses both ATPase and DNA helicase activities that are strictly dependent on DNA, divalent cations and ATP. These activities are abolished by a single amino acid substitution in the ATP-binding domain. The hChlR1 protein can unwind both DNA/DNA and RNA/DNA substrates. It has a preference for movement in the 5'-->3' direction on short single-stranded DNA templates. However, unlike other DNA helicases, the hChlR1 DNA helicase can translocate along single-stranded DNA in both directions when substrates have a very long single-stranded DNA region. The enzymatic activities of hChlR1 suggest that DNA helicases are required for maintaining the fidelity of chromosome segregation.


Subject(s)
DNA Helicases/metabolism , Base Sequence , Cloning, Molecular , DEAD-box RNA Helicases , DNA Helicases/genetics , DNA Helicases/isolation & purification , DNA Primers , Humans , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
8.
J Biol Chem ; 274(40): 28505-13, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10497214

ABSTRACT

The Cdc2L locus encoding the PITSLRE protein kinases maps to chromosome band 1p36 and consists of two duplicated and tandemly linked genes. The purpose of the present study was to determine whether diminution of PITSLRE kinases leads to deregulation of apoptosis. The human melanoma cell lines A375 (Cdc2L wild-type alleles) and UACC 1227 (mutant Cdc2L alleles) were tested with agonist anti-Fas monoclonal antibody. We found that exposure of these cells to anti-Fas for 24, 48, or 72 h resulted in differential sensitivity to Fas-induced apoptosis. In A375, cell death started at 24-48 h post-treatment, and it was maximal by 72 h. Conversely, UACC 1227 cells were resistant to Fas-mediated apoptosis. Induction of PITSLRE histone H1 kinase activity was observed in A375 anti-Fas treated but not in UACC 1227 cells. Also, the PITSLRE protein kinase activity in A375 anti-Fas-treated cells preceded maximal levels of apoptosis. Finally, fluorescence confocal microscopy revealed a nuclear localization of PITSLRE proteins in normal melanocytes and A375 cells but a cytoplasmic localization in UACC 1227 cells. The differences in PITSLRE protein and cellular localization between A375 and UACC 1227 cells appear to account for the differences in sensitivity of the two cells lines to anti-Fas and staurosporine. These observations suggest that alterations in PITSLRE gene expression and protein localization may result in the loss of apoptotic signaling.


Subject(s)
Apoptosis/physiology , CDC2 Protein Kinase/metabolism , Melanoma/pathology , Protein Kinases/metabolism , fas Receptor/physiology , Antibodies, Monoclonal/immunology , Apoptosis/drug effects , Caspase 3 , Caspase 8 , Caspase 9 , Caspase Inhibitors , Cell Nucleus/metabolism , Cyclin-Dependent Kinases , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Activation , Fas Ligand Protein , Humans , Melanoma/enzymology , Melanoma/metabolism , Membrane Glycoproteins/metabolism , Microinjections , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases , Staurosporine/pharmacology , Tumor Cells, Cultured , fas Receptor/immunology
9.
Methods ; 17(4): 305-12, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10196101

ABSTRACT

The avian DT40 cell system represents a novel method to generate loss of function mutations in vertebrate cells. These chicken B lymphoma cells undergo homologous recombination at very high frequencies and can thus be used to "knock out" genes believed to function in apoptotic processes. The knockout cells can then be used to determine how the cell death process is modulated after induction of apoptosis and to order components in cell death pathways. The system can be further modified, using tetracycline-responsive promoters, to allow expression of wild-type cDNAs to rescue "knockout cells" if the gene of interest is essential. Alternatively, cDNA expression constructs containing mutations or deletions in the cDNA encoding the absent protein can be used to delineate functional domains. cDNA expression libraries or known proteins believed to function downstream of the target in a signal transduction pathway could also be transfected into the knockout cell line, and the resultant cells could be assayed for complementation and/or rescue of the apoptotic alteration/defect. Finally, the system has recently been adapted to allow disruption of human genes in DT40/human hybrid cell lines thereby potentially extending this system for use in studying human genes.


Subject(s)
Apoptosis/genetics , Gene Targeting/methods , Signal Transduction/genetics , Animals , Blotting, Southern , Chickens , Cyclin D1/genetics , Dexamethasone/pharmacology , Gene Library , Humans , Hybrid Cells , Mutation , Recombination, Genetic , Transfection , Tumor Cells, Cultured
10.
J Biol Chem ; 274(11): 7245-52, 1999 Mar 12.
Article in English | MEDLINE | ID: mdl-10066786

ABSTRACT

Cycloheximide (CHX) can contribute to apoptotic processes, either in conjunction with another agent (e.g. tumor necrosis factor-alpha) or on its own. However, the basis of this CHX-induced apoptosis has not been clearly established. In this study, the molecular mechanisms of CHX-induced cell death were examined in two different human T-cell lines. In T-cells undergoing CHX-induced apoptosis (Jurkat), but not in T-cells resistant to the effects of CHX (CEM C7), caspase-8 and caspase-3 were activated. However, the Fas ligand was not expressed in Jurkat cells either before or after treatment with CHX, suggesting that the activation of these caspases does not involve the Fas receptor. To determine whether CHX-induced apoptosis was mediated by a Fas-associated death domain (FADD)-dependent mechanism, a FADD-DN protein was expressed in cells prior to CHX treatment. Its expression effectively inhibited CHX-induced cell death, suggesting that CHX-mediated apoptosis primarily involves a FADD-dependent mechanism. Since CHX treatment did not result in the induction of Fas or FasL, and neutralizing anti-Fas and anti-tumor necrosis factor receptor-1 antibodies did not block CHX-mediated apoptosis, these results may also indicate that FADD functions in a receptor-independent manner. Surprisingly, death effector filaments containing FADD and caspase-8 were observed during CHX treatment of Jurkat, Jurkat-FADD-DN, and CEM C7 cells, suggesting that their formation may be necessary, but not sufficient, for cell death.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis/drug effects , Carrier Proteins/metabolism , Cycloheximide/pharmacology , T-Lymphocytes/drug effects , Cell Line , Fas-Associated Death Domain Protein , Humans , Jurkat Cells , Subcellular Fractions/metabolism , T-Lymphocytes/cytology
11.
Cancer Genet Cytogenet ; 108(2): 91-9, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9973934

ABSTRACT

The two genes encoding the PITSLRE protein kinase isoforms, CDC2L1 and CDC2L2, are localized to human chromosome band 1p36. The PITSLRE protein kinases are a part of the p34cdc2 supergene family. Several protein products of the CDC2L locus may be effector(s) in apoptotic signaling. The larger PITSLRE p110 isoforms appear to regulate some aspect of RNA splicing/transcription during the cell cycle. One or more of these genes may function as tumor suppressor genes in melanoma. Using fluorescence in situ hybridization, one allele of the CDC2L gene complex on chromosome 1 was either deleted or translocated in 8 of 14 different melanoma cell lines. We also observed mutations in the 5' promoter region of the CDC2L1 gene in four different cell lines relative to normal melanocytes using PCR-SSCP analysis and direct DNA sequencing. Western blot analysis revealed decreased level of PITSLRE protein expression in several cell lines, as well as in four surgical malignant melanoma specimens relative to normal melanocytes. Thus, the decreased PITSLRE protein expression appears to result from deletion of the CDC2L alleles and possibly by mutations within the 5' promoter region. We propose that aberrations in the CDC2L genes may contribute to the pathogenesis or progression of melanoma.


Subject(s)
Chromosomes, Human, Pair 1 , Melanoma/genetics , Mutation , Protein Kinases/genetics , Base Sequence , Cyclin-Dependent Kinases , DNA, Neoplasm , Humans , In Situ Hybridization , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Protein Serine-Threonine Kinases , Tumor Cells, Cultured
12.
Genome Res ; 8(9): 929-39, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9750192

ABSTRACT

Cdc2L1 and Cdc2L2 span approximately 140 kb on human chromosome 1p36.3. The products of the Cdc2L genes encode almost identical protein kinases, the PITSLRE kinases, which have functions that may be relevant to the regulation of transcription/splicing and apoptotic signaling. These genes are deleted/translocated in neuroblastomas with MYCN gene amplification, a subset of malignant melanomas, and in a newly delineated deletion syndrome. Here we report that the p36.3 region of human chromosome 1 consists of two identical genomic regions, each of which contain a Cdc2L gene linked to a metalloprotease (MMP) gene in a tail-to-tail configuration. This duplicated genomic region is also linked tightly to D1Z2, a genetic marker containing a highly polymorphic VNTR (variable number tandem repeat) consisting of an unusual 40-bp reiterated sequence. Thus, these genes and the polymorphic marker D1Z2 are organized as follows: telomere-D1Z2-5'-MMP22-3'-3'-Cdc2L2-5'-5'-Cdc2L1 -3'- 3'-MMP21-5'-centromere. Remarkably, the introns and exons of Cdc2L1 and Cdc2L2, as well as their flanking regions, are essentially identical. A total of 15 amino acid differences, 12 nonconservative and 3 conservative, can be found in the 773-786 amino acids specified by the various products of the Cdc2L genes. Two separate promoter/5' untranslated (UT) regions, CpG1 and CpG2, are identical to a reported previously methylated genomic CpG sequence and are used to express >20 different Cdc2L transcripts from the two genes. The expression of CpG2 transcripts from Cdc2L1 and Cdc2L2 is tissue/cell-line specific. CpG1 transcripts are expressed ubiquitously from both genes, with perhaps some bias towards the expression of CpG1 Cdc2L1 mRNAs in certain hematopoietic cells.


Subject(s)
Chromosome Mapping , Chromosomes, Human, Pair 1/genetics , Gene Duplication , Metalloendopeptidases/genetics , Protein Kinases/genetics , 5' Untranslated Regions/genetics , Alternative Splicing , Bacteriophage P1/genetics , Cloning, Molecular , Cosmids/genetics , Cyclin-Dependent Kinases , DNA, Complementary/isolation & purification , Genes, Duplicate , Genetic Linkage , Genetic Markers/genetics , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Tumor Cells, Cultured
13.
Genomics ; 52(1): 101-6, 1998 Aug 15.
Article in English | MEDLINE | ID: mdl-9740677

ABSTRACT

The terminal end of the short arm of human chromosome 1, 1p36.3, is frequently deleted in a number of tumors and is believed to be the location of multiple tumor suppressor genes. Thus far, a bona fide tumor suppressor gene from this region has not been identified. The isolation and characterization of new 1p36 genes is, therefore, of some interest. Two novel matrix metalloproteinase genes, MMP21 and MMP22, have been identified in the Cdc2L1-2 locus, which spans approximately 120 kb on 1p36.3. These genes encode novel metalloproteinases that contain prepro, catalytic, cysteine-rich, interleukin-1 receptor-related, and proline-rich domains. Their catalytic domains are most closely related to stromelysin-3 and contain the consensus HEXXH zinc-binding region required for enzyme activation, while their cysteine-rich domains appear to be related to a number of human, mouse, and Caenorhabditis elegans metalloproteinase sequences. Of some possible interest is the absence of a highly conserved cysteine residue in the proenzyme domain, the so-called "cysteine switch," which has been shown to be involved in the autocatalytic activation of many metalloproteinases. The MMP genes are located less than 1 kb from the 3' regions of Cdc2L1 and Cdc2L2, suggesting that the MMP and Cdc2L genes are part of a larger region that has been duplicated. Finally, the MMP21/22 genes express multiple mRNAs, some of which are derived by alternative splicing, in a tissue-specific manner.


Subject(s)
Chromosomes, Human, Pair 1/genetics , Genetic Linkage , Metalloendopeptidases/chemistry , Metalloendopeptidases/genetics , Amino Acid Sequence , Blotting, Northern , Cloning, Molecular , DNA, Complementary/analysis , DNA, Complementary/isolation & purification , Humans , Male , Molecular Sequence Data , Organ Specificity , RNA/analysis , Sequence Analysis, DNA , Sequence Homology, Amino Acid
14.
Cancer Res ; 58(12): 2624-32, 1998 Jun 15.
Article in English | MEDLINE | ID: mdl-9635589

ABSTRACT

The p16INK4a (MTS1) and pl8INK4c gene products are normal, and highly expressed, in human neuroblastoma cell lines. The retinoblastoma protein (pRb) was, nonetheless, phosphorylated and functional in these cells. Such high levels of p16INK4a/p18INK4c should normally inhibit cyclin-dependent kinase (CDK) 4 and 6 activities in cells containing functional pRb, delaying cell cycle progression and growth. These neuroblastoma cell lines express both CDK4 and CDK6 mRNA and protein, but only significant CDK6 protein kinase activity was detected in this study. In addition, CDK6 was not present in p16INK4a immune complexes in cells with significant kinase activity, although p16INK4a levels were high. Others have shown that a specific mutation in the NH2-terminal region of the CDK4 gene product can disrupt p16INK4a binding, thereby bypassing its inhibitory activity. To determine whether mutation of the CDK6 gene, or some other mechanism, is responsible for the CDK6 kinase activity in these cell lines, several complementary analyses were performed. The CDK6 gene from each cell line was examined for mutations that might affect p16INK4a binding, whereas p16INKa add-back experiments were performed with CDK6 immune complexes to assess p16INK4a function. A bona fide CDK6 mutation that disrupts p16INK4a binding and prevents inhibition of CDK6 protein kinase activity was identified in 1 of 17 neuroblastoma cell lines. The mechanism(s) responsible for disruption of p16INK4a inhibitory activity in the remaining cell lines is unknown, but these results suggest that neuroblastoma cells may bypass the cell cycle block imposed by constitutive expression of wild-type p16INK4a in novel ways.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Neoplasm Proteins/metabolism , Neuroblastoma/enzymology , Protein Serine-Threonine Kinases/analysis , RNA, Messenger/metabolism , Retinoblastoma Protein/analysis , Cyclin D , Cyclin-Dependent Kinase 6 , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p19 , Humans , Mutation/genetics , Neuroblastoma/metabolism , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Protein Serine-Threonine Kinases/genetics , Retinoblastoma Protein/metabolism , Tumor Cells, Cultured
16.
J Cell Sci ; 111 ( Pt 11): 1495-506, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9580558

ABSTRACT

The PITSLRE protein kinases are members of the p34cdc2 superfamily, with >20 different isoforms expressed from two linked genes in humans. PITSLRE homologues have been identified in mouse, chicken, Drosophila, Xenopus, and possibly Plasmodium falciparum, suggesting that their function may be well conserved. A possible role for a caspase processed PITSLRE isoform has been suggested by studies of Fas- and TNF-induced cell death. However, the function of these kinases in proliferating cells is still unknown. Here we demonstrate that the 110 kDa PITSLRE isoforms (p110) are localized to both the nucleoplasm and nuclear speckles, and that these isoforms specifically interact in vitro and in vivo with the RNA-binding protein RNPS1. RNPS1 is also localized to nuclear speckles, and its over expression disrupts normal nuclear speckle organization by causing the aggregation of many nuclear speckles into approximately 6 'mega' speckles. This type of nuclear speckle aggregation closely resembles what occurs when cells are treated with several transcriptional inhibitors. These data indicate that the PITSLRE p110 isoforms interact with RNPS1 in vivo, and that these proteins may in turn influence some aspect of transcriptional and/or splicing regulation.


Subject(s)
CDC2 Protein Kinase/physiology , DNA-Binding Proteins/physiology , Protein Kinases/physiology , RNA-Binding Proteins/physiology , Ribonucleoproteins , Signal Transduction , Transcription, Genetic , Animals , Cyclin-Dependent Kinases , HeLa Cells , Humans , Mice , Protein Serine-Threonine Kinases
17.
J Biol Chem ; 272(18): 11694-7, 1997 May 02.
Article in English | MEDLINE | ID: mdl-9115219

ABSTRACT

Emerging evidence suggests that multiple aspartate-specific cysteine proteases (caspases (CASPs)) play a crucial role in programmed cell death. Many cellular proteins have been identified as their substrates and serve as markers to assay the activation of CASPs during the death process. However, no substrate has yet been unambiguously identified as an effector molecule in apoptosis. PITSLRE kinases are a superfamily of Cdc2-like kinases that have been implicated in apoptotic signaling and tumorigenesis. In this paper we report that tumor necrosis factor (TNF)-mediated apoptosis is associated with a CrmA- and Bcl-2-inhibitable cleavage of PITSLRE kinases, indicating a role for CASPs. Testing of seven murine CASPs for their ability to cleave p110 PITSLRE kinase alpha2-1 in vitro revealed that only CASP-1 (ICE (interleukin-1beta-converting enzyme)) and CASP-3 (CPP32) were able to produce the same 43-kDa cleavage product as observed in cells undergoing TNF-induced apoptosis. Mutational analysis revealed that cleavage of p110 PITSLRE kinase alpha2-1 occurred at Asp393 within the sequence YVPDS, which is similar to that involved in the CASP-1-mediated cleavage of prointerleukin-1beta. TNF-induced proteolysis of PITSLRE kinases was still observed in fibroblasts from CASP-1(0/0) mice. These data implicate CASP-3 as a potentially important CASP family protease responsible for the cleavage of PITSLRE kinases during TNF-induced apoptosis.


Subject(s)
Antigens, CD/physiology , Apoptosis/drug effects , Caspases , Cysteine Endopeptidases/metabolism , Protein Kinases/metabolism , Receptors, Tumor Necrosis Factor/physiology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antigens, CD/biosynthesis , Caspase 1 , Caspase 3 , Cloning, Molecular , Cyclin-Dependent Kinases , Cysteine Endopeptidases/biosynthesis , Enzyme Precursors/biosynthesis , Enzyme Precursors/metabolism , HeLa Cells , Humans , Mice , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Recombinant Proteins/metabolism , Substrate Specificity , T-Lymphocytes , Transfection
18.
J Biol Chem ; 272(16): 10859-69, 1997 Apr 18.
Article in English | MEDLINE | ID: mdl-9099742

ABSTRACT

DT40 lymphoma B-cells normally express cyclins D1 and D2 but not D3. When cyclin D1 expression was extinguished in these cells by gene knockout, specific alterations in their ability to transit the cell cycle were observed. These changes are exemplified by a delay of approximately 2 h in their progression through a normal 14-h cell cycle. This delay results in an increase in the number of cells in the G2/M phase population, most likely due to triggering of checkpoints in G2/M, inability to enter G1 normally, and/or alterations of crucial event(s) in early G1. The defect(s) in the cell cycle of these D1 "knockout" cells can be rescued by overexpression of any normal mouse D-type cyclin but not by a mutant mouse cyclin D1 protein that lacks the LXCXE motif at its amino terminus. These data suggest that the cell cycle alterations observed in the D1-/- cells are a direct effect of the absence of the cyclin D1 protein and support the hypothesis that the D-type cyclins have separate, but overlapping, functions. Elimination of cyclin D1 also resulted in enhanced sensitivity to radiation, resulting in a significant increase in apoptotic cells. Expression of any normal murine D-type cyclin in the D1-/- cells reversed this phenotype. Intriguingly, expression of the mutant cyclin D1 in the D1 -/- cells partially restored resistance to radiation-induced apoptosis. Thus, there may be distinct differences in cyclin D1 complexes and/or its target(s) in proliferating and apoptotic DT40 lymphoma B-cells.


Subject(s)
Cell Cycle/physiology , Cyclins/biosynthesis , Oncogene Proteins/biosynthesis , Amino Acid Sequence , Animals , Birds , Cell Division , Cell Survival , Cloning, Molecular , Cyclin D1 , Cyclin D2 , Cyclin D3 , Cyclins/chemistry , Cyclins/deficiency , G2 Phase , Gene Library , Humans , Lymphoma, B-Cell , Mice , Mice, Knockout , Mitosis , Mitotic Index , Molecular Sequence Data , Mutagenesis, Site-Directed , Oncogene Proteins/chemistry , Oncogene Proteins/deficiency , Open Reading Frames , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , T-Lymphocytes , Tumor Cells, Cultured , Vertebrates
19.
J Biol Chem ; 272(6): 3823-32, 1997 Feb 07.
Article in English | MEDLINE | ID: mdl-9013641

ABSTRACT

Helicases are components of numerous protein complexes, including those regulating transcription, translation, DNA replication and repair, splicing, and mitotic chromosome transmission. Helicases unwind double-stranded DNA and RNA homo- and hetero-duplexes. The yeast CHL1 helicase has been linked to maintenance of the high fidelity of chromosome transmission during mitosis. Mutations in this gene result in a 200-fold increase in the rate of aberrant chromosome segregation with a concomitant delay in the cell cycle at G2-M, suggesting that CHL1 is required for the maintenance of proper chromosome transmission. Two highly related human cDNA clones encoding proteins which are homologous to the yeast CHL1 gene product have been isolated. Here we show that these two distinct human CHL1-related mRNAs and proteins (hCHLR1 and hCHLR2) are expressed only in proliferating human cell lines. Quiescent normal human fibroblasts stimulated to re-enter the cell cycle by addition of serum begin to express the CHL1-related proteins as the cells enter S phase, concomitant with the expression of proliferating cell nuclear antigen. Furthermore, expression of the CHL1-related mRNAs is lost when human K562 cells cease to proliferate and terminally differentiate in response to phorbol ester treatments. Human hCHLR expression is not extinguished during hemin-induced differentiation of the same cell line, which produces erythrocyte-like cells that continue to proliferate. These experiments are consistent with the requirement of this putative helicase during either S or G2-M phase but not G1. In vitro transcribed and translated hCHLR1 protein binds to both single- and double-stranded DNA, supporting the possibility that these proteins are DNA helicases. Finally, affinity-purified hCHLR1 antisera was used to demonstrate the localization of the hCHLR proteins to the nucleolus by indirect immunofluorescence as well as by cell fractionation.


Subject(s)
Chromosomal Proteins, Non-Histone , Fungal Proteins/chemistry , Saccharomyces cerevisiae Proteins , Amino Acid Sequence , Animals , Blotting, Western , Cell Differentiation , Cloning, Molecular , DNA/metabolism , DNA, Complementary/chemistry , Drosophila melanogaster , HeLa Cells , Humans , Molecular Sequence Data , Saccharomyces cerevisiae , Sequence Homology, Amino Acid
20.
Cell Death Differ ; 4(2): 130-9, 1997 Feb.
Article in English | MEDLINE | ID: mdl-16465219

ABSTRACT

The p55 tumor necrosis factor (TNF) receptor and the Fas (CD95/APO-1) receptor share an intracellular domain necessary to induce apoptosis, suggesting they utilize common signaling pathways. To define pathways triggered by Fas and TNF-alpha we utilized human CEM-C7 T-cells. As expected, stimulation of either receptor induced apoptosis and TNF-alpha-induced signaling included the activation of NF-kappaB. Surprisingly, Fas-induced signaling also triggered the activation of NF-kappaB in T cells, yet the kinetics of NF-kappaB induction by Fas was markedly delayed. NF-kappaB activation by both pathways was persistent and due to the sequential degradation of IkappaB-alpha and IkappaB-beta. However, the kinetics of IkappaB degradation were different and there were differential effects of protease inhibitors and antioxidants on NF-kappaB activation. Signaling pathways leading to activation of apoptosis were similarly separable and were also independent of NF-kappaB activation. Thus, the Fas and TNF receptors utilize distinct signal transduction pathways in T-cells to induce NF-kappaB and apoptosis.

SELECTION OF CITATIONS
SEARCH DETAIL
...