Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
Dokl Biochem Biophys ; 490(1): 43-46, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32342312

ABSTRACT

CDK8-mediated transcriptional reprogramming is essential for an extensive gene expression. Constitutive knockouts of the cdk8 gene are lethal at the morula stage. For modeling transcriptional reprogramming in an adult organism, we investigated the possibility to attenuate the CDK8 kinase activity with a F97G mutation in exon 3 of the cdk8 gene. According to preliminary experimental data, this mutation should lead to a decrease in CDK8 kinase activity. To edit the genome of laboratory mice, the CRISPR/Cas9 technology was used, in which the introduction of a double-stranded gap occurred at a distance of 128 nucleotide pairs from the planned site of the introduced mutation. To introduce the mutation, a matrix for homologous repair was used as part of plasmid DNA, with homologous arms 903 and 484 bp in the 5'-3' region from the point of double-stranded rupture, respectively. As a result, mice with site-specific target mutations in exon 3 of the cdk8 gene were obtained. We for the first time demonstrated a high efficacy of the mutation 128 bp apart from the site of double-strand break. Viable animals with the F97G mutation in the catalytic domain of CDK8 kinase were obtained for the first time. The resulting cdk8 mutant mice will be used in subsequent studies to simulate the processes involving transcription reprogramming.


Subject(s)
Cyclin-Dependent Kinase 8/metabolism , Gene Editing/methods , Genome , RNA, Guide, Kinetoplastida , Transcription, Genetic , Animals , CRISPR-Cas Systems , Catalytic Domain , Exons , Heterozygote , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mutation , Oligonucleotides/genetics
2.
Br J Cancer ; 101(11): 1900-8, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19935801

ABSTRACT

BACKGROUND: The mitotic arrest deficiency protein 2 (MAD2) is a key component of the mitotic spindle assembly checkpoint, monitoring accurate chromosomal alignment at the metaphase plate before mitosis. MAD2 also has a function in cellular senescence and in a cell's response to microtubule inhibitory (MI) chemotherapy exemplified by paclitaxel. METHODS: Using an siRNA approach, the impact of MAD2 down-regulation on cellular senescence and paclitaxel responsiveness was investigated. The endpoints of senescence, cell viability, migration, cytokine expression, cell cycle analysis and anaphase bridge scoring were carried out using standard approaches. RESULTS: We show that MAD2 down-regulation induces premature senescence in the MCF7 breast epithelial cancer cell line. These MAD2-depleted (MAD2) cells are also significantly replicative incompetent but retain viability. Moreover, they show significantly higher levels of anaphase bridges and polyploidy compared to controls. In addition, these cells secrete higher levels of IL-6 and IL-8 representing key components of the senescence-associated secretory phenotype (SASP) with the ability to impact on neighbouring cells. In support of this, MAD2 cells show enhanced migratory ability. At 72 h after paclitaxel, MAD2 cells show a significant further induction of senescence compared with paclitaxel naive controls. In addition, there are significantly more viable cells in the MAD2 MCF7 cell line after paclitaxel reflecting the observed increase in senescence. CONCLUSION: Considering that paclitaxel targets actively dividing cells, these senescent cells will evade cytotoxic kill. In conclusion, compromised MAD2 levels induce a population of senescent cells resistant to paclitaxel.


Subject(s)
Breast Neoplasms/drug therapy , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Cellular Senescence/drug effects , Paclitaxel/pharmacology , Repressor Proteins/metabolism , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Cellular Senescence/genetics , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Female , Humans , Mad2 Proteins , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Spindle Apparatus/drug effects , Spindle Apparatus/genetics , Spindle Apparatus/metabolism , Transfection
3.
Oncogene ; 26(48): 6954-8, 2007 Oct 18.
Article in English | MEDLINE | ID: mdl-17486059

ABSTRACT

Damage-induced G1 checkpoint in mammalian cells involves upregulation of p53, which activates transcription of p21(Waf1) (CDKN1A). Inhibition of cyclin-dependent kinase (CDK)2 and CDK4/6 by p21 leads to dephosphorylation and activation of Rb. We now show that ectopic p21 expression in human HT1080 fibrosarcoma cells causes not only dephosphorylation but also depletion of Rb; this effect was p53-independent and susceptible to a proteasome inhibitor. CDK inhibitor p27 (CDKN1B) also caused Rb dephosphorylation and depletion, but another CDK inhibitor p16 (CDKN2A) induced only dephosphorylation but not depletion of Rb. Rb depletion was observed in both HT1080 and HCT116 colon carcinoma cells, where p21 was induced by DNA-damaging agents. Rb depletion after DNA damage did not occur in the absence of p21, and it was reduced when p21 induction was inhibited by p21-targeting short hairpin RNA or by a transdominant inhibitor of p53. These results indicate that p21 both activates Rb through dephosphorylation and inactivates it through degradation, suggesting negative feedback regulation of damage-induced cell-cycle checkpoint arrest.


Subject(s)
Colonic Neoplasms/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Fibrosarcoma/metabolism , Retinoblastoma Protein/metabolism , Antibiotics, Antineoplastic/pharmacology , Colonic Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Damage/drug effects , Doxorubicin/pharmacology , Fibrosarcoma/pathology , Humans , Immunoblotting , Phosphorylation/drug effects , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Suppressor Protein p53/metabolism
4.
Cancer Res ; 61(21): 7754-62, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11691789

ABSTRACT

The cell cycle regulatory events that interface with polyamine requirements for cell growth have not yet been clearly identified. Here we use specific inhibitors of polyamine biosynthetic enzymes to investigate the effect of polyamine pool depletion on cell cycle regulation. Treatment of MALME-3M cells with either the ornithine decarboxylase inhibitor alpha-difluoromethylornithine or the S-adenosylmethionine decarboxylase inhibitor MDL-73811 lowered specific polyamine pools and slowed cell growth but did not induce cell cycle arrest. By contrast, treatment with the combination of inhibitors halted cell growth and caused a distinct G1 arrest. The latter was associated with marked reduction of all three polyamine pools, a strong increase in p21(WAF1/CIP1/SDI1) (p21), and hypophosphorylation of retinoblastoma protein. All effects were fully prevented by exogenous polyamines. p21 induction preceded p53 stabilization in MALME-3M cells and also occurred in a polyamine-depleted, p53-nonfunctional melanoma cell line, indicating that p21 is induced at least in part through p53-independent mechanisms. Conditional overexpression of p21 in a fibrosarcoma cell line was shown previously to inhibit the expression of multiple proliferation-associated genes and to induce the expression of genes associated with various aspects of cell senescence and organism aging. Polyamine depletion in MALME-3M cells was associated with inhibition of seven of seven tested p21-inhibited genes and with induction of 13 of 14 tested p21-induced genes. p21 expression is also known to induce a senescence-like phenotype, and phenotypic features of senescence were observed in polyamine-depleted MALME-3M cells. Cells increased in size, appeared more granular, and expressed senescence-associated beta-galactosidase. Cells released from the polyamine inhibition lost the ability to form colonies, failed to replicate their DNA, and approximately 25% became bi- or multinucleated. These events parallel the outcome of prolonged p21 induction in fibrosarcoma cells. The results of this study indicate that polyamine pool depletion achieved by specific biosynthetic enzyme inhibitors causes p21-mediated G1 cell cycle arrest followed by p21-mediated changes in gene expression, development of a senescence-like phenotype, and loss of cellular proliferative capacity.


Subject(s)
Biogenic Polyamines/physiology , Cyclins/physiology , G1 Phase/physiology , Gene Expression Regulation, Neoplastic , Melanoma/metabolism , Melanoma/pathology , Adenosylmethionine Decarboxylase/antagonists & inhibitors , Biogenic Polyamines/biosynthesis , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Division/drug effects , Cell Division/physiology , Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , Deoxyadenosines/pharmacology , Eflornithine/pharmacology , Enzyme Inhibitors/pharmacology , G1 Phase/drug effects , Humans , Melanoma/genetics , Ornithine Decarboxylase Inhibitors , Phosphorylation , Retinoblastoma Protein/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
5.
Biochemistry ; 40(14): 4323-31, 2001 Apr 10.
Article in English | MEDLINE | ID: mdl-11284688

ABSTRACT

The multidrug transporter P-glycoprotein (Pgp) is an ATPase efflux pump for multiple cytotoxic agents, including vinblastine and colchicine. We have found that resistance to vinblastine but not to colchicine in cell lines derived from different types of tissues and expressing the wild-type human Pgp correlates with the Pgp density. Vinblastine induces a conformational change in Pgp, evidenced by increased reactivity with a conformation-sensitive monoclonal antibody UIC2, in all the tested cell lines. In contrast, colchicine increases the UIC2 reactivity in only some of the cell lines. In those lines where colchicine alone did not affect UIC2 reactivity, this drug was, however, able to reverse the vinblastine-induced increase in UIC2 reactivity. The magnitude of the increase in UIC2 reactivity in the presence of saturating concentrations of colchicine correlates with the relative ability of Pgp to confer colchicine resistance in different cell lines, suggesting the existence of some cell-specific factors that have a coordinate effect on the ability of colchicine to induce conformational transitions and to be transported by Pgp. Colchicine, like vinblastine, reverses the decrease in UIC2 reactivity produced by nonhydrolyzable nucleotides, but unlike vinblastine, it does not reverse the effect of ATP at a high concentration. Colchicine, however, decreases the Hill number for the effect of ATP on the UIC2 reactivity from 2 to 1. Colchicine increases the UIC2 reactivity and reverses the effect of ATP in ATPase-deficient Pgp mutants, but not in the wild-type Pgp expressed in the same cellular background, suggesting that ATP hydrolysis counteracts the effects of colchicine on the Pgp conformation.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Colchicine/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , 3T3 Cells , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/immunology , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Antibodies, Monoclonal/metabolism , Antigen-Antibody Reactions/drug effects , Binding Sites/genetics , Carrier Proteins/genetics , Cell Communication/drug effects , Cell Line , Dose-Response Relationship, Drug , Drug Resistance, Multiple/immunology , Drug Resistance, Neoplasm/immunology , Humans , Intracellular Signaling Peptides and Proteins , K562 Cells , Mice , Protein Conformation/drug effects , Transfection , Tumor Cells, Cultured
6.
Biochemistry ; 40(14): 4312-22, 2001 Apr 10.
Article in English | MEDLINE | ID: mdl-11284687

ABSTRACT

The reactivity of the ATP-dependent multidrug transporter P-glycoprotein (Pgp) with the conformation-sensitive monoclonal antibody UIC2 is increased in the presence of Pgp transport substrates, ATP-depleting agents, or mutations that reduce the level of nucleotide binding by Pgp. We have investigated the effects of nucleotides and vinblastine, a Pgp transport substrate, on the UIC2 reactivity of Pgp in cells permeabilized by Staphylococcus aureus alpha-toxin. ATP, ADP, and nonhydrolyzable ATP analogues decreased the UIC2 reactivity; this effect was potentiated by vanadate, a nucleotide-trapping agent. The Hill number for the nucleotide-induced conformational transition was 2 for ATP and ADP but 1 for nonhydrolyzable ATP analogues. The Hill numbers for ATP and ADP were decreased to 1 by mutations in one of the two nucleotide binding sites of Pgp, whereas mutation of both sites greatly diminished the overall effect of nucleotides. Vinblastine reversed the decrease in the UIC2 reactivity brought about by all the nucleotides, including nonhydrolyzable analogues; this effect of vinblastine was blocked by vanadate. These data indicate that UIC2-detectable conformational changes of Pgp are driven by binding and debinding of nucleotides, that nucleotide hydrolysis affects the Hill number for its Pgp interactions, and that Pgp transport substrates promote nucleotide dissociation from Pgp. These findings are consistent with a conventional E1/E2 model that explains conformational transitions of a transporter protein through a series of linked equilibria.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antibodies, Monoclonal/metabolism , Binding Sites, Antibody , Cell Membrane Permeability , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/immunology , Adenine Nucleotides/metabolism , Adenine Nucleotides/pharmacology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Bacterial Toxins/pharmacology , Binding Sites, Antibody/drug effects , Binding Sites, Antibody/genetics , Cell Line , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/genetics , Cell Membrane Permeability/immunology , Hemolysin Proteins/pharmacology , Humans , K562 Cells , Mice , Protein Conformation/drug effects , Staphylococcus aureus , Vinblastine/pharmacology
7.
Biochemistry ; 40(14): 4332-9, 2001 Apr 10.
Article in English | MEDLINE | ID: mdl-11284689

ABSTRACT

The MDR1 P-glycoprotein (Pgp), responsible for a clinically important form of multidrug resistance in cancer, is an ATPase efflux pump for multiple lipophilic drugs. The G185V mutation near transmembrane domain 3 of human Pgp increases its relative ability to transport several drugs, including etoposide, but decreases the transport of other substrates. MDR1 cDNA with the G185V substitution was used in a function-based selection to identify mutations that would further increase Pgp-mediated resistance to etoposide. This selection yielded the I186N substitution, adjacent to G185V. Pgps with G185V, I186N, or both mutations were compared to the wild-type Pgp for their ability to confer resistance to different drugs in NIH 3T3 cells. In contrast to the differential effects of G185V, I186N mutation increased resistance to all the tested drugs and augmented the effect of G185V on etoposide resistance. The effects of the mutations on conformational transitions of Pgp induced by different drugs were investigated using a conformation-sensitive antibody UIC2. Ligand-binding analysis of the drug-induced increase in UIC2 reactivity was used to determine the K(m) value that reflects the apparent affinity of drugs for Pgp, and the Hill number reflecting the apparent number of drug-binding sites. Both mutations altered the magnitude of drug-induced increases in UIC2 immunoreactivity, the K(m) values, and the Hill numbers for individual drugs. Mutation-induced changes in the magnitude of UIC2 reactivity shift did not correlate with the effects of the mutations on resistance to the corresponding drugs. In contrast, an increase or a decrease in drug resistance relative to that of the wild type was accompanied by a corresponding increase or decrease in the K(m) or in both the K(m) and the Hill number. These results suggest that mutations that alter the ability of Pgp to transport individual drugs change the apparent affinity and the apparent number of drug-binding sites in Pgp.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Drug Resistance, Multiple/genetics , Mutagenesis, Site-Directed , 3T3 Cells , ATP Binding Cassette Transporter, Subfamily B, Member 1/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Amino Acid Substitution/genetics , Animals , Antibodies, Monoclonal/metabolism , Antigen-Antibody Reactions/drug effects , Asparagine/genetics , Cell Line , Colchicine/metabolism , Colchicine/pharmacology , Etoposide/metabolism , Etoposide/pharmacology , Genetic Vectors/biosynthesis , Genetic Vectors/chemistry , Genetic Vectors/physiology , Glycine/genetics , Humans , Isoleucine/genetics , Mice , Protein Conformation/drug effects , Transduction, Genetic , Transfection , Valine/genetics , Vinblastine/metabolism , Vinblastine/pharmacology
8.
Drug Resist Updat ; 4(5): 303-13, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11991684

ABSTRACT

Inhibition of the program of apoptosis has been reported to have little or no effect on clonogenic survival after treatment with drugs or radiation in several tumor cell lines. A decrease in apoptosis is compensated in such cell lines by an increase in the fractions of cells that undergo permanent growth arrest with phenotypic features of cell senescence, or die through the process of mitotic catastrophe. Most of the tested tumor cell lines have retained the capacity of normal cells to undergo accelerated senescence after treatment with DNA-interactive drugs, ionizing radiation, or cytostatic agents. p53 and p21(Waf1/Cip1/Sdi1) act as positive regulators of treatment-induced senescence, but they are not required for this response in tumor cells. The senescent phenotype distinguishes tumor cells that survived drug exposure but lost the ability to form colonies from those that recover and proliferate after treatment. Although senescent cells do not proliferate, they are metabolically active and may produce secreted proteins with potential tumor-promoting activities. The expression of such proteins is mediated at least in part by the induction of p21(Waf1/Cip1/Sdi1). The other anti-proliferative response of tumor cells is mitotic catastrophe, a form of cell death that results from abnormal mitosis and leads to the formation of interphase cells with multiple micronuclei. Mitotic catastrophe is induced by different classes of cytotoxic agents, but the pathways of abnormal mitosis differ depending on the nature of the inducer and the status of cell-cycle checkpoints. Mitotic catastrophe can also develop as a consequence of aberrant reentry of tumor cells into cell cycle after prolonged growth arrest. Elucidation of the factors that regulate different aspects of treatment-induced senescence and mitotic catastrophe should assist in improving the efficacy and decreasing side effects of cancer therapy.


Subject(s)
Apoptosis/drug effects , Growth Inhibitors/toxicity , Mitosis/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Animals , Apoptosis/physiology , Apoptosis/radiation effects , Cell Division/drug effects , Cellular Senescence/drug effects , Cellular Senescence/physiology , Cellular Senescence/radiation effects , Humans , Mitosis/physiology , Mitosis/radiation effects , Tumor Cells, Cultured
9.
Cancer Res ; 60(18): 5027-30, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-11016623

ABSTRACT

A combination of four genetic suppressor elements (GSEs), two of which are derived from putative transcriptional regulators, was previously found to increase resistance to drugs inhibiting DNA replication in HT1080 fibrosarcoma cells. In the present study, two GSE-transduced cell lines, isolated with and without cytotoxic selection, were found to be resistant to a diverse group of DNA-interactive agents, including aphidicolin, hydroxyurea, cytarabine, etoposide, doxorubicin, and mafosfamide. Changes in gene expression associated with GSE-induced drug resistance were analyzed by cDNA array hybridization and reverse transcription-PCR. Twenty genes were found to be up-regulated in both of the resistant cell lines. These include genes involved in DNA replication and repair (e.g., PCNA, XRCC1, B-MYB, and GADD45), transcriptional regulators associated with stress response, and cell cycle checkpoint control (e.g., YB-1, DBPA, and ATF4), and genes for signal transduction proteins (e.g., protein tyrosine phosphatase 1B and regulatory subunits alpha and beta of cAMP-dependent protein kinase). The observed changes in gene expression may play a role in pleiotropic resistance to different classes of DNA-targeting drugs.


Subject(s)
DNA Repair/genetics , DNA Replication/genetics , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/genetics , Genes, Suppressor/genetics , Antineoplastic Agents/pharmacology , Aphidicolin/pharmacology , Cell Cycle/genetics , DNA Repair/drug effects , DNA Replication/drug effects , DNA, Neoplasm/drug effects , DNA, Neoplasm/genetics , Fibrosarcoma/drug therapy , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Heat-Shock Response/genetics , Humans , Proliferating Cell Nuclear Antigen/biosynthesis , Proliferating Cell Nuclear Antigen/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transduction, Genetic , Tumor Cells, Cultured
10.
Int J Oncol ; 17(3): 579-86, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10938401

ABSTRACT

P-glycoprotein (P-gp) is a transmembrane glycoprotein that confers multidrug resistance (MDR). It has been demonstrated that the Gly185 residue within the cytoplasmic loop between predicted transmembrane portions 2 and 3 plays an important role in substrate specificity of human P-gp. Derivatives of cyclosporin interact with and reverse the ability of P-gp to act as a drug efflux pump. To determine if the Gly185 residue of human P-gp is also important for the interaction of P-gp with closely related cyclosporin derivatives, we examined the effect of PSC-833 and CsA on P-gp in KB3-1 cells transfected with human wild-type P-gp (GSV-2) or with the mutant P-gp (VSV-1) that habored the Gly185-->Val substitution. While the ability of CsA to sensitize VSV-1 cells to anticancer agents was enhanced, no changes in the potency of PSC-833 against cells transfected with either the wild-type or mutant P-gp were observed. In addition, VSV-1 transfected cells were more sensitive to CsA inhibition of verapamil-stimulated ATPase activity than cells transfected with wild-type P-gp. Furthermore, the intracellular accumulation of CsA was low in GSV-2 P-gp-expressing cells, compared with its accumulation in VSV-1 cells and it was found to be as high as in non-P-gp expressing KB3-1 cells. These results indicated an enhanced sensitivity of Val185-P-gp expressing cells to CsA that correlated with increased intracellular accumulation in these cells. In contrast, no significant difference in the accumulation of PSC-833 was observed among the parental, wild-type or resistant cells. Since PSC-833 was found to be more potent than CsA, these studies provided insight into the effects of the structure of MDR modulators in mediating sensitivity to anticancer drugs.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Amino Acid Substitution , Antineoplastic Agents/metabolism , Cyclosporine/pharmacology , Cyclosporins/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Neoplasm Proteins/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Adenosine Triphosphatases/metabolism , Biological Transport, Active/drug effects , Calcium Channel Blockers/pharmacology , Carcinoma, Squamous Cell/pathology , Colchicine/metabolism , Daunorubicin/metabolism , Humans , Neoplasm Proteins/chemistry , Neoplasm Proteins/physiology , Paclitaxel/metabolism , Point Mutation , Structure-Activity Relationship , Transfection , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Verapamil/pharmacology , Vinblastine/metabolism
11.
Cancer Res ; 60(10): 2576-8, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10825123

ABSTRACT

Ionizing radiation induces apoptosis, mitotic catastrophe, and senescence-like terminal proliferation arrest in tumor cells. We investigated the effect of the MDR1 P-glycoprotein (Pgp), recently shown to inhibit caspase-mediated apoptosis, on cellular responses to radiation. Pgp strongly inhibited radiation-induced apoptosis in a HeLa-derived cell line with inducible MDR1 expression and in NIH 3T3 cells transduced with a MDR1-expressing retroviral vector. The inhibition of apoptosis by Pgp was associated, however, with increases in radiation-induced mitotic catastrophe and senescence and produced only a marginal change in the survival of irradiated cells. Pgp had no effect on radiation responses in apoptosis-resistant HT1080 cells. These results indicate that Pgp inhibits radiation-induced apoptosis, but this effect of Pgp provides no substantial increase in radiation resistance of the tested cell lines because apoptosis-resistant cells die from mitotic catastrophe or undergo senescence-like terminal proliferation arrest.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Cellular Senescence/drug effects , Cellular Senescence/radiation effects , Mitosis/drug effects , Mitosis/radiation effects , 3T3 Cells , Animals , Cell Survival/drug effects , Cell Survival/radiation effects , HeLa Cells , Humans , Mice , Radiation, Ionizing
12.
Oncogene ; 19(17): 2165-70, 2000 Apr 20.
Article in English | MEDLINE | ID: mdl-10815808

ABSTRACT

Induction of a cyclin-dependent kinase inhibitor p21Waf1/ Cip1/Sdi1 is an integral part of cell growth arrest associated with senescence and damage response. p21 overexpression from an inducible promoter resulted in senescence-like growth arrest in a human fibrosarcoma cell line. After release from p21-induced growth arrest, cells re-entered the cell cycle but displayed growth retardation, cell death and decreased clonogenicity. The failure to form colonies was associated with abnormal mitosis and endoreduplication in the recovering cells and was correlated with the induced level of p21 and the duration of p21 induction. p21 induction was found to inhibit the expression of multiple proteins involved in the execution and control of mitosis. p21-induced depletion of the cellular pools of mitosis-control proteins was followed by asynchronous resynthesis of such proteins after release from p21, which explains the observed mitotic abnormalities. Genetic destabilization in cells recovering from p21-induced growth arrest may conceivably play a role in carcinogenesis and tumor progression.


Subject(s)
Autoantigens , Carrier Proteins , Chromosomal Proteins, Non-Histone/metabolism , Cyclin B/metabolism , Cyclins/metabolism , Mitosis , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins , Cell Division , Centromere Protein A , Cyclin B/genetics , Cyclin B1 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/drug effects , Cyclins/genetics , DNA/drug effects , Dose-Response Relationship, Drug , Fibrosarcoma/drug therapy , Fibrosarcoma/pathology , Fluorescent Dyes/analysis , Fluorescent Dyes/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Humans , Isopropyl Thiogalactoside/pharmacology , Microfilament Proteins , Nuclear Proteins , Organic Chemicals , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins , Tumor Cells, Cultured , Polo-Like Kinase 1
13.
Proc Natl Acad Sci U S A ; 97(8): 4291-6, 2000 Apr 11.
Article in English | MEDLINE | ID: mdl-10760295

ABSTRACT

Induction of cyclin-dependent kinase inhibitor p21(Waf1/Cip1/Sdi1) triggers cell growth arrest associated with senescence and damage response. Overexpression of p21 from an inducible promoter in a human cell line induces growth arrest and phenotypic features of senescence. cDNA array hybridization showed that p21 expression selectively inhibits a set of genes involved in mitosis, DNA replication, segregation, and repair. The kinetics of inhibition of these genes on p21 induction parallels the onset of growth arrest, and their reexpression on release from p21 precedes the reentry of cells into cell cycle, indicating that inhibition of cell-cycle progression genes is a mechanism of p21-induced growth arrest. p21 also up-regulates multiple genes that have been associated with senescence or implicated in age-related diseases, including atherosclerosis, Alzheimer's disease, amyloidosis, and arthritis. Most of the tested p21-induced genes were not activated in cells that had been growth arrested by serum starvation, but some genes were induced in both forms of growth arrest. Several p21-induced genes encode secreted proteins with paracrine effects on cell growth and apoptosis. In agreement with the overexpression of such proteins, conditioned media from p21-induced cells were found to have antiapoptotic and mitogenic activity. These results suggest that the effects of p21 induction on gene expression in senescent cells may contribute to the pathogenesis of cancer and age-related diseases.


Subject(s)
Aging/genetics , Cell Transformation, Neoplastic/genetics , Cyclins/physiology , Gene Expression Regulation/physiology , Cell Cycle/genetics , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , DNA Repair/genetics , DNA, Complementary , Humans , Nucleic Acid Hybridization
14.
Oncogene ; 18(34): 4808-18, 1999 Aug 26.
Article in English | MEDLINE | ID: mdl-10490814

ABSTRACT

Exposure of human tumor cell lines to moderate doses of anticancer agents induces terminal proliferation arrest accompanied by morphologic and enzymatic changes that resemble senescence of normal cells. We have investigated the role of p53 and p21waf1/cip1 in the induction of this response in drug-treated tumor cells. Doxorubicin treatment induced the senescence-like phenotype (SLP) and its associated terminal growth arrest in wild-type HCT116 colon carcinoma cells; this response was strongly decreased but not abolished in HCT116 lines with homozygous knockout of p53 or p21. Transduction of HT1080 fibrosarcoma cells with a genetic inhibitor of p53 also decreased the induction of SLP and increased drug-induced mitotic cell death. To determine if drug-stimulated p21 expression was responsible for senescence-like growth arrest, we have expressed different levels of p21 from an inducible promoter. While high-level overexpression of p21 was sufficient to induce SLP in HT1080 cells, the levels of p21 expressed in doxorubicin-treated cells could account for only a fraction of doxorubicin-induced SLP. Our results indicate that p53 and p21 act as positive regulators of senescence-like terminal proliferation arrest, but their function is neither sufficient nor absolutely required for this treatment response in tumor cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cellular Senescence/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Cyclins/genetics , Tumor Suppressor Protein p53/genetics , Carcinoma/drug therapy , Carcinoma/pathology , Cell Death/drug effects , Cell Death/genetics , Cell Division/drug effects , Cell Division/genetics , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/drug effects , Cyclins/metabolism , Doxorubicin/pharmacology , Fibrosarcoma/drug therapy , Fibrosarcoma/pathology , Fluorescent Dyes/metabolism , Gene Expression Regulation, Neoplastic , Humans , Organic Chemicals , Regulatory Sequences, Nucleic Acid , Tumor Cells, Cultured , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism , beta-Galactosidase/drug effects , beta-Galactosidase/metabolism
15.
Cancer Res ; 59(15): 3761-7, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10446993

ABSTRACT

Exposure of human tumor cell lines to different chemotherapeutic drugs, ionizing radiation, and differentiating agents induced morphological, enzymatic, and ploidy changes resembling replicative senescence of normal cells. Moderate doses of doxorubicin induced this senescence-like phenotype (SLP) in 11 of 14 tested cell lines derived from different types of human solid tumors, including all of the lines with wild-type p53 and half of p53-mutated cell lines. SLP induction seemed to be independent from mitotic cell death, the other major effect of drug treatment. Among cells that survived drug exposure, SLP markers distinguished those cells that became terminally growth-arrested within a small number of cell divisions from the cells that recovered and resumed proliferation. SLP induction in breast carcinoma cells treated with retinoids in vitro or in vivo was found to correlate with permanent growth inhibition under the conditions of minimal cytotoxicity, suggesting that this response may be particularly important for the antiproliferative effect of differentiating agents. The senescence-like program of terminal proliferation arrest may provide an important determinant of treatment outcome and a target for augmentation in cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cellular Senescence/drug effects , Neoplastic Stem Cells/drug effects , Adenocarcinoma/pathology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/pathology , Doxorubicin/pharmacology , Female , Fibrosarcoma/pathology , Gamma Rays , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , Phenotype , Ploidies , Tretinoin/pharmacology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/pathology , Tumor Cells, Cultured/radiation effects
16.
Gene Ther ; 6(1): 130-7, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10341885

ABSTRACT

The development of general approaches for the isolation of efficient antivirals and the identification and validation of targets for drug screening are becoming increasingly important, due to the emergence of previously unrecognized viral diseases. The genetic suppressor element (GSE) technology is an approach based on the functional expression selection of efficient genetic inhibitors from random fragment libraries derived from a gene or genome of interest. We have applied this technology to isolate potent genetic inhibitors against HIV-1. Two strategies were used to select for GSEs that interfere with latent virus induction and productive HIV-1 infection based on the expression of intracellular and surface antigens. The selected GSEs clustered in seven narrowly defined regions of the HIV-1 genome and were found to be functionally active. These elements are potential candidates for the gene therapy of AIDS. The developed approaches can be applied to other viral pathogens, as well as for the identification of cellular genes supporting the HIV-1 life cycle.


Subject(s)
Acquired Immunodeficiency Syndrome/therapy , Antiviral Agents , Genes, Suppressor , Genetic Therapy/methods , HIV-1/genetics , Flow Cytometry , Gene Library , Genome, Viral , Humans
17.
J Dermatol ; 26(4): 203-9, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10343463

ABSTRACT

Protein kinase C (PKC) has been implicated in regulation of hair growth. In this study, the role of PKC alpha in induced mouse hair growth was studied. Hair growth in C57BL6 mice, a well known model for hair growth research, was induced by plucking the telogen hair. PKC alpha protein levels during the induced hair growth cycle were analyzed by Western immunoblot and mRNA levels were measured by RT-PCR. At 1 day and 4 days postdepilation, when the induced hair cycle was in early and midanagen, the PKC alpha protein level was decreased. At 10 days after depilation, when the induced hair cycle was in mature anagen, the PKC alpha protein level was increased. At 17 days after plucking the hair, when the induced hair cycle was in early catagen, PKC alpha protein returned to the control level. PKC alpha mRNA was relatively unchanged at 1 day and 4 days after plucking the hair but significantly elevated at 10 days postdepilation. At 17 days after hair growth induction, PKC alpha mRNA reverted to the control level. These results suggest that: 1) in early and mid anagen of the induced hair growth cycle, PKC alpha was downregulated posttranscriptionally. This downregulation may play a role in the induction of hair growth; 2) in mature anagen of induced hair growth cycle, PKC alpha was overexpressed, and this overexpression may play a part in maintaining the hair growth. Since the expression of PKC alpha was roughly correlated with mouse skin pigmentation, we hypothesize that PKC alpha may regulate hair growth partially through modulation of skin melanogenesis.


Subject(s)
Hair/growth & development , Isoenzymes/metabolism , Protein Kinase C/metabolism , RNA, Messenger/analysis , Skin/enzymology , Animals , Blotting, Western , Down-Regulation , Female , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Protein Kinase C-alpha , Reference Values , Sensitivity and Specificity
18.
Oncogene ; 18(2): 377-84, 1999 Jan 14.
Article in English | MEDLINE | ID: mdl-9927194

ABSTRACT

Drug design targeted at microtubules has led to the advent of some potent anti-cancer drugs. In the present study, we demonstrated that microtubule-binding agents (MBAs) taxol and colchicine induced immediate early gene (c-jun and ATF3) expression, cell cycle arrest, and apoptosis in the human breast cancer cell line MCF-7. To elucidate the signal transduction pathways that mediate such biological activities of MBAs, we studied the involvement of mitogen-activated protein (MAP) kinases. Treatment with taxol, colchicine, or other MBAs (vincristine, podophyllotoxin, nocodazole) stimulated the activity of c-jun N-terminal kinase 1 (JNK1) in MCF-7 cells. In contrast, p38 was activated only by taxol and none of the MBAs changed the activity of extracellular signal-regulated protein kinase 2 (ERK2). Activation of JNK1 or p38 by MBAs occurred subsequent to the morphological changes in the microtubule cytoskeleton induced by these compounds. Furthermore, baccatine III and beta-lumicolchicine, inactive analogs of taxol and colchicine, respectively, did not activate JNKI or p38. These results suggest that interactions between microtubules and MBAs are essential for the activation of these kinases. Pretreatment with the antioxidants N-acetyl-L-cysteine (NAC), ascorbic acid or vitamin E, blocked H2O2- or doxorubicin-induced JNKI activity, but had no effect on JNKI activation by MBAs, excluding a role for oxidative stress. However, BAPTA/AM, a specific intracellular Ca2+ chelator, attenuated JNK1 activation by taxol but not by colchicine, and had no effect on microtubule changes induced by taxol. Thus, stabilization or depolymerization of microtubules may regulate JNK1 activity via distinct downstream signaling pathways. The differential activation of MAP kinases opens up a new avenue for addressing the mechanism of action of antimicrotubule drugs.


Subject(s)
Breast Neoplasms/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Microtubules/drug effects , Apoptosis/drug effects , Breast Neoplasms/pathology , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Cycle/drug effects , Colchicine/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Activation , Humans , Microtubules/metabolism , Nocodazole/pharmacology , Paclitaxel/pharmacology , Podophyllotoxin/pharmacology , Tumor Cells, Cultured , Vincristine/pharmacology
19.
Somat Cell Mol Genet ; 25(1): 9-26, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10925700

ABSTRACT

Many anticancer drugs inhibit DNA replication. To investigate the mechanism of permanent growth inhibition after transient arrest of DNA replication, we selected genetic suppressor elements (GSEs) conferring resistance to replication inhibitor Aphidicolin. Starting from a retroviral expression library carrying normalized fragments of human cell cDNA, we isolated four GSEs which, when introduced as a combination, produced resistance to Aphidicolin, doxorubicin and hydroxyurea in HT1080 fibrosarcoma cells. The four GSEs were derived from ORFX bromodomain protein gene, WIZ zinc finger protein gene, the gene for subunit 3 of cytochrome c oxidase, and the gene corresponding to an EST with no known function. A cell line carrying all four GSEs showed a weaker induction of the senescence-like phenotype after treatment with Aphidicolin or doxorubicin; the resistance of this cell line was not associated with decreased doxorubicin accumulation. These results indicate that combined effects of GSEs derived from these four genes increase cellular resistance to replication-inhibiting drugs, possibly by inhibiting drug-induced senescence.


Subject(s)
Aphidicolin/pharmacology , DNA Replication/drug effects , Genes, Suppressor , Antineoplastic Agents/pharmacology , Base Sequence , DNA, Antisense/genetics , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Doxorubicin/pharmacology , Drug Resistance/genetics , Gene Library , HeLa Cells , Humans , Hydroxyurea/pharmacology , Molecular Sequence Data , Tumor Cells, Cultured
20.
Oncol Res ; 10(4): 185-92, 1998.
Article in English | MEDLINE | ID: mdl-9778689

ABSTRACT

Treatment-induced secondary drug resistance of tumor cells is a major cause of relapsed disease and therapeutic failure in cancer patients. It has been shown that the expression of the multidrug resistance MDR1/P-glycoprotein gene could be induced by short-term in vitro exposure of cells to protein kinase C (PKC) agonists or different chemotherapeutic drugs. We studied whether other genes involved in drug resistance are regulated by similar signaling pathways. Transient (up to 24 h) treatment of HL-60 or K562 leukemia cells with phorbol 12-myristate 13-acetate (TPA) resulted in increased steady-state level of LRP (lung resistance-related protein) mRNA and protein. Among conventional chemotherapeutic drugs tested, only cytarabine (Ara C) induced the LRP mRNA expression though no increase in LRP protein was detected. LRP gene activation was not detectable in either H9 T-cell leukemia or in solid carcinoma cell lines (BT-20, ZR-75-1, and SW 1573). None of the agents influenced the levels of MRP (multidrug resistance-associated protein) mRNA in any cell line tested. In HL-60 cells, the LRP activation by TPA or Ara C was sustained for at least 23 days after withdrawal of inducing agents. bis-Indolylmaleimide I, a potent PKC inhibitor, attenuated TPA-induced LRP activation. In contrast, the inhibitor had no effect on the LRP induction by Ara C. These data indicate that the LRP gene can be activated by different mechanisms, some of which involve PKC.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Carcinogens/pharmacology , Cytarabine/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Leukemia, Experimental/genetics , Leukemia, Experimental/metabolism , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Tetradecanoylphorbol Acetate/pharmacology , Vault Ribonucleoprotein Particles , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Genes, MDR/drug effects , HL-60 Cells/drug effects , HL-60 Cells/metabolism , Humans , K562 Cells/drug effects , K562 Cells/metabolism , Leukemia, T-Cell/genetics , Leukemia, T-Cell/metabolism , Protein Kinase C/physiology , RNA, Messenger/metabolism , Transcriptional Activation , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...