Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Biochem J ; 353(Pt 2): 369-75, 2001 01 15.
Article in English | MEDLINE | ID: mdl-11139402

ABSTRACT

Using differential mRNA expression analysis, a previously uncharacterized gene was found to be up-regulated 2-fold in brown adipose tissue (BAT) of mice exposed to cold (4 degrees C) for 48 h. Contig and homology analysis revealed that the gene represents the murine orthologue to a sequence from a public database encoding a putative human protein (CGI-69). The presence of mitochondrial carrier domains in the human protein, its transmembrane topology and cold-induction of the mouse CGI-69 gene in BAT prompted an analysis of the idea that CGI-69 may represent a new uncoupling protein (UCP) functional homologue. However, transfection of human CGI-69 isoforms in HEK-293 cells yielded no change in mitochondrial membrane potential (Deltapsi(m)), despite localization of FLAG-tagged CGI-69 to mitochondria of MCF7 cells. Surprisingly, overexpression of the human 2-oxoglutarate carrier (OGC) protein (originally designed as a negative control) sparked a significant drop in Deltapsi(m), possibly signalling a previously unappreciated uncoupling activity for the OGC.


Subject(s)
Adipose Tissue, Brown/metabolism , Carrier Proteins/biosynthesis , Intracellular Membranes/metabolism , Membrane Proteins/biosynthesis , Membrane Transport Proteins , Mitochondria/metabolism , Mitochondrial Proteins , Recombinant Fusion Proteins , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Cell Line , Cold Temperature , Male , Membrane Potentials , Mitochondrial Membrane Transport Proteins , Molecular Sequence Data , Rats , Sequence Homology, Amino Acid
2.
FASEB J ; 14(11): 1611-8, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10928996

ABSTRACT

Mitochondrial uncoupling proteins have been implicated in the maintenance of metabolic rate and adaptational thermoregulation. We recently reported the identification of a brain-specific mitochondrial uncoupling protein homologue, UCP4. Here we characterized another newly described member of the uncoupling protein family, termed UCP5 (also called BMCP1). UCP5 transcripts are present in multiple human and mouse tissues, with an especially high abundance in the brain and testis. Expression of UCP5 in mammalian cells reduces the mitochondrial membrane potential. Multiple isoforms of UCP5 were identified and exhibited tissue-specific distribution and different potency in reduction of membrane potential. Furthermore, the mRNA abundance of both UCP4 and UCP5 is modulated by nutritional status or temperature in a tissue-specific manner in mice. Brain UCP4 and UCP5 mRNA transcripts rose by 1.5- and 1.7-fold, respectively, and liver UCP5 expression increased by 1.8-fold in response to acute cold exposure. A high-fat diet increased UCP5 mRNA in liver by 1.6-fold selectively in the obesity-resistant A/J but not in the obesity-prone C57BL/6J mouse strain. Liver UCP5 expression decreased significantly with a 24 h fast and was restored to the normal level after refeeding. In contrast, brain transcripts for both genes were not significantly altered by fasting or high-fat diet. These findings are consistent with the notion that UCP4 and UCP5 may be involved in tissue-specific thermoregulation and metabolic changes associated with nutritional status.


Subject(s)
Carrier Proteins/genetics , Cold Temperature , Dietary Fats/pharmacology , Fasting/physiology , Gene Expression Regulation/drug effects , Membrane Transport Proteins , Nerve Tissue Proteins/genetics , Amino Acid Sequence , Animals , Brain/drug effects , Brain/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Line , Cloning, Molecular , Dietary Fats/administration & dosage , Down-Regulation/drug effects , Gene Expression Profiling , Humans , Liver/drug effects , Liver/metabolism , Male , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Uncoupling Proteins , Molecular Sequence Data , Multigene Family/genetics , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Organ Specificity , Protein Isoforms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection , Up-Regulation/drug effects
3.
J Pharmacol Exp Ther ; 291(3): 1100-12, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10565830

ABSTRACT

Mechanisms of immunosuppressive action of mycophenolic acid (MPA) on rat lymphocytes and correlations among MPA plasma concentrations (pharmacokinetics) and its suppression of immune functions (pharmacodynamics) were studied in vitro and in vivo. In vitro, MPA inhibited concanavalin A-stimulated lymphocyte proliferation in blood [tritium-labeled thymidine ([(3)H]TdR) incorporation, percentage of lymphocytes positive for proliferating cell nuclear antigen, and in S-G(2)M by flow cytometry] and activation (percentage of lymphocytes expressing CD25 or CD134). Maximum percent inhibitions (I(max)) of lymphocyte functions and concentrations of MPA (mg/l in blood) inhibiting 50% of I(max) (IC(50)) were 99%/0.14 mg/l for [(3)H]TdR, 93%/0.28 mg/l for S-G(2)M, 74%/0.29 mg/l for CD25, and 83%/0.24 mg/l for CD134. Blood sampled at different times after single or multiple oral MPA administrations at four dose levels was assayed for lymphocyte functions and MPA plasma concentrations. I(max) (%) and IC(50) (mg/l in plasma by HPLC) were 98 to 99%/0.18 to 0.19 mg/l for [(3)H]TdR, 88 to 98%/0.70 to 0.83 mg/l for S-G(2)M, 60 to 63%/0.65 to 0.81 mg/l for CD25, and 72 to 77%/0.61 to 0.74 mg/l for CD134. IC(50) values for S-G(2)M, CD25, and CD134 were higher after multiple daily treatments than after a single dose. There were clear and direct relationships among MPA dose levels, kinetics of MPA plasma concentrations, and dynamics of lymphocyte functions. MPA treatment in vitro and in vivo inhibits not only mitogen-stimulated lymphocyte proliferation in whole blood but also lymphocyte expression of cell surface cytokine receptors. These two different mechanisms of action may contribute to the therapeutic efficacy of MPA in vivo.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/drug effects , Lymphocytes/drug effects , Mycophenolic Acid/pharmacology , Receptors, Tumor Necrosis Factor , Animals , Antibiotics, Antineoplastic/blood , Antibiotics, Antineoplastic/pharmacokinetics , Cell Division/drug effects , DNA/biosynthesis , Flow Cytometry , Hematocrit , Immunosuppressive Agents/blood , Immunosuppressive Agents/pharmacokinetics , Leukocyte Count , Lymphocyte Count/drug effects , Lymphocytes/immunology , Male , Mycophenolic Acid/blood , Mycophenolic Acid/pharmacokinetics , Rats , Rats, Inbred Lew , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/immunology , Receptors, Interleukin-2/biosynthesis , Receptors, Interleukin-2/immunology , Receptors, OX40 , Tumor Necrosis Factor Receptor Superfamily, Member 7/biosynthesis , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
4.
Immunol Lett ; 67(3): 179-83, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10369124

ABSTRACT

Malononitrilamides (MNAs) are a new class of immunomodulatory drug highly effective in in vivo models of allo- and xenotransplantation. Knowledge of their effects on immune cells, however, is limited and has been derived solely from investigations using isolated mononuclear cells. This use of purified cells to investigate drug activity is not ideal, so we have combined the analytical power of flow cytometry with our mitogen-driven, whole blood lymphocyte activation and proliferation assays to investigate the in vitro mechanism of action of MNAs. We first show that MNAs (A77 1726, HMR1279, and HMR1715), as well as brequinar (BQR) and cyclosporine (CsA), effectively inhibit cell activation antigen expression and lymphocyte proliferation. We next show that the inhibitory effects of MNAs and BQR, but not CsA, are reversed by the addition of uridine to the culture. These results suggest that inhibition of pyrimidine biosynthesis may be a mechanism by which MNAs suppress both lymphocyte activation and proliferation since these effects were reversed when uridine nucleotide pools were replenished. This novel finding of suppression of activation antigen expression by MNAs in whole blood expands our understanding of the effects of this new class of drug.


Subject(s)
Aniline Compounds/pharmacology , Hydroxybutyrates/pharmacology , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/drug effects , Lymphocytes/drug effects , Receptors, Tumor Necrosis Factor , Acrylamides/pharmacology , Alkynes , Animals , Biphenyl Compounds/pharmacology , Caproates/pharmacology , Crotonates , Cyclosporine/pharmacology , Dose-Response Relationship, Drug , Flow Cytometry , Isoxazoles/metabolism , Leflunomide , Lymphocytes/immunology , Male , Nitriles , Rats , Rats, Inbred Lew , Receptors, Interleukin-2/metabolism , Receptors, OX40 , Toluidines , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
5.
J Immunol Methods ; 224(1-2): 1-9, 1999 Apr 22.
Article in English | MEDLINE | ID: mdl-10357200

ABSTRACT

An assay based on two-color flow cytometry has been developed to measure CTL and NK cell-mediated cytotoxicity. After effector/target cells are incubated together, CTL or NK populations are stained with an effector cell specific PE-conjugated mAb. Subsequently, annexin V-FITC binds to cells expressing phosphatidylserine (an early marker of apoptosis) on the cell surface. Target cells are gated upon as PE-negative and quantified with respect to their annexin V positivity. The shift from annexin Vneg to annexin Vhi is a discrete event such that all target cells fall within discernible populations with respect to annexin V. There is a strong correlation between cytotoxicity measured with our assay and a standard 51Cr release assay (r2 = 0.989). The PE/annexin V assay shows increased sensitivity at early timepoints after target/effector cell mixing. In addition, this method allows for analysis of target cells at the single cell level. Therefore, we have described a promising new technique to measure in vitro cell-mediated cytotoxicity. It avoids the potential difficulties of working with radioactive isotopes, and offers increased sensitivity and versatility.


Subject(s)
Annexin A5/analysis , Cytotoxicity Tests, Immunologic/methods , Flow Cytometry/methods , Killer Cells, Natural/immunology , T-Lymphocytes, Cytotoxic/immunology , Biomarkers , CD8 Antigens , Cell Line , Chromium , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Humans , Jurkat Cells , Phycoerythrin , Staining and Labeling/methods
6.
FEBS Lett ; 443(3): 326-30, 1999 Jan 29.
Article in English | MEDLINE | ID: mdl-10025957

ABSTRACT

Uncoupling proteins (UCPs) are a family of mitochondrial transporter proteins that have been implicated in thermoregulatory heat production and maintenance of the basal metabolic rate. We have identified and partially characterized a novel member of the human uncoupling protein family, termed uncoupling protein-4 (UCP4). Protein sequence analyses showed that UCP4 is most related to UCP3 and possesses features characteristic of mitochondrial transporter proteins. Unlike other known UCPs, UCP4 transcripts are exclusively expressed in both fetal and adult brain tissues. UCP4 maps to human chromosome 6p11.2-q12. Consistent with its potential role as an uncoupling protein, UCP4 is localized to the mitochondria and its ectopic expression in mammalian cells reduces mitochondrial membrane potential. These findings suggest that UCP4 may be involved in thermoregulatory heat production and metabolism in the brain.


Subject(s)
Brain/metabolism , Carrier Proteins/physiology , Membrane Transport Proteins , Mitochondria/physiology , Amino Acid Sequence , Animals , Brain/embryology , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Carrier Proteins/analysis , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , Cell Survival , Chromosome Mapping , Chromosomes, Human, Pair 6/genetics , Cloning, Molecular , Expressed Sequence Tags , Genes, Plant/genetics , Humans , Ion Channels , Membrane Potentials/drug effects , Mitochondria/metabolism , Mitochondrial Proteins , Mitochondrial Uncoupling Proteins , Molecular Sequence Data , Organ Specificity , RNA, Messenger/analysis , Sequence Homology, Amino Acid , Transfection , Tumor Cells, Cultured , Uncoupling Protein 3
7.
Invest Ophthalmol Vis Sci ; 40(1): 197-202, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9888444

ABSTRACT

PURPOSE: To investigate the relation of senescence-related beta-galactosidase activity and telomere shortening to replicative senescence in cultured human retinal pigment epithelial (RPE) cells. METHODS: A human RPE cell line was serially passaged until 80% of cells were nondividing in a 72-hour 5-bromo-2'-deoxyuridine (BrdU) labeling study. Early- and late-passage cells were double-stained for BrdU and senescence-related beta-galactosidase activity (pH 6). The average chromosomal telomere length at several population doublings was estimated by Southern blot analysis after double digestion of DNA with RsaI and HinfI and using a telomere-specific probe. RESULTS: BrdU-beta-galactosidase double-staining revealed an inverse correlation between the number of BrdU-labeled nuclei and beta-galactosidase-labeled cells as a function of population doubling level (PDL). At PDL 58, only 20% of all cells labeled for BrdU, whereas 57% stained for beta-galactosidase. The mean terminal restriction fragment length (TRF) was reduced from 10 kb in early (PDL 12) cultures to 4 kb in late (PDL 57) cultures. CONCLUSIONS: Senescence-related beta-galactosidase activity and mean TRF length may prove useful in studying the senescence of RPE cells in vitro. These techniques may be valuable in determining senescence of the retinal pigment epithelium in vivo, where senescent RPE cells could be involved in the development of age-related maculopathy and age-related macular degeneration.


Subject(s)
Cellular Senescence , Pigment Epithelium of Eye/enzymology , Telomere/metabolism , beta-Galactosidase/metabolism , Blotting, Southern , Bromodeoxyuridine/metabolism , Cell Division , Cell Line , Cells, Cultured , Cellular Senescence/physiology , DNA/analysis , DNA Replication , Histocytochemistry , Humans , Infant , Pigment Epithelium of Eye/cytology
8.
Nature ; 396(6712): 699-703, 1998 Dec 17.
Article in English | MEDLINE | ID: mdl-9872321

ABSTRACT

Fas ligand (FasL) is produced by activated T cells and natural killer cells and it induces apoptosis (programmed cell death) in target cells through the death receptor Fas/Apol/CD95. One important role of FasL and Fas is to mediate immune-cytotoxic killing of cells that are potentially harmful to the organism, such as virus-infected or tumour cells. Here we report the discovery of a soluble decoy receptor, termed decoy receptor 3 (DcR3), that binds to FasL and inhibits FasL-induced apoptosis. The DcR3 gene was amplified in about half of 35 primary lung and colon tumours studied, and DcR3 messenger RNA was expressed in malignant tissue. Thus, certain tumours may escape FasL-dependent immune-cytotoxic attack by expressing a decoy receptor that blocks FasL.


Subject(s)
Colonic Neoplasms/genetics , Lung Neoplasms/genetics , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Adult , Amino Acid Sequence , Apoptosis , Colonic Neoplasms/immunology , Cytotoxicity, Immunologic , DNA, Complementary , Expressed Sequence Tags , Fas Ligand Protein , Gene Amplification , Humans , Jurkat Cells , Killer Cells, Natural/immunology , Ligands , Lung Neoplasms/immunology , Membrane Glycoproteins/antagonists & inhibitors , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/isolation & purification , Receptors, Tumor Necrosis Factor, Member 6b , Sequence Homology, Amino Acid , Tissue Distribution , Tumor Cells, Cultured , fas Receptor
10.
Cancer Surv ; 29: 263-84, 1997.
Article in English | MEDLINE | ID: mdl-9338104

ABSTRACT

Telomere dynamics and changes in telomerase activity are consistent elements of cellular alterations associated with changes in proliferative state. In particular, the highly specific correlations and early causal relationships between telomere loss in the absence of telomerase activity and replicative senescence or crisis, on the one hand, and telomerase reactivation and cell immortality, on the other, point to a new and important paradigm in the complementary fields of ageing and cancer. Although the signalling pathways between telomeres and transcriptional and cell cycle machinery remain undefined, recently described homologies between telomeric proteins and lipid/protein kinase activities important in chromosome stability provide evidence for the existence of pathways transducing signals originating in chromosome structure to cell cycle regulatory processes. Similarities between cell cycle arrest at senescence and the response of mortal cells to DNA/oxidative damage suggest overlap in the signal transduction mechanisms culminating in irreversible and stable cell cycle arrest. The feasibility of targeting telomeres/telomerase as a strategy for antiproliferative therapeutics has been shown in studies in yeast, in which mutations in specific telomere associated genes result in delayed cell death. Similarly, antisense oligonucleotide inhibition of telomerase activity in human tumour cells (HeLa) results in delayed cell death. The mechanism of cell death and possible escape from this fate require further study. In human cells, however, it would seem reasonable to predict that in these circumstances, apoptosis is induced in the vast majority of cells either directly in response to a DNA damage signal arising from critically shortened telomeres or as a secondary consequence of genetic instability.


Subject(s)
Aging/genetics , Cellular Senescence/genetics , Neoplasms/genetics , Telomerase/metabolism , Telomere/genetics , Aging/physiology , Apoptosis , Cell Cycle/physiology , Cellular Senescence/physiology , Cyclin-Dependent Kinases/metabolism , DNA Damage/genetics , Genes, p53 , Humans , Neoplasms/etiology , Neoplasms/physiopathology , Signal Transduction , Telomere/physiology
11.
Exp Cell Res ; 219(2): 461-70, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7641798

ABSTRACT

Cells treated with compactin, an inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, the enzyme which catalyzes the rate-limiting step of the mevalonate pathway, are arrested prior to the DNA synthesis (S) phase of the cell cycle. Identification of a specific pathway product or products with a role in DNA replication, however, has remained elusive. In this report we demonstrate that farnesyl acetate, a derivative of the key isoprenoid pathway intermediate farnesyl pyrophosphate, inhibits DNA replication in both Chinese hamster ovary cells and human (HeLa) cells. This effect is revealed by measurement of DNA content using fluorescence-activated cell sorter analysis and by measurement of [3H]thymidine incorporation. We show that cells treated with farnesyl acetate retain protein synthesis capacity as DNA replication is inhibited and remain intact as viewed with the vital stain propidium iodide. The inhibition of DNA replication by farnesyl acetate occurs in cells treated with high levels of compactin and in cells lacking HMG-CoA reductase. These results indicate that farnesyl acetate action is not dependent on metabolism through the isoprenoid pathway and is not the result of the loss of a metabolite required for replication nor the accumulation of a metabolite which is inhibitory. In addition, cells treated with farnesyl acetate for over 6 h are irreversibly blocked from progressing through S phase, a phenomenon which differs sharply from the results with compactin, removal of which results in synchronous progression through S phase. Farnesyl acetate also blocks protein prenylation in cells, to a degree comparable to a known farnesylation inhibitor, BZA-5B. We propose that farnesyl acetate is acting in a manner quite different from the metabolic block caused by compactin, causing a rapid and irreversible block of DNA replication.


Subject(s)
DNA Replication/drug effects , DNA/metabolism , Farnesol/analogs & derivatives , Mevalonic Acid/metabolism , Animals , CHO Cells , Cells, Cultured , Cricetinae , Farnesol/pharmacology , Flow Cytometry , HeLa Cells , Humans , S Phase/drug effects
12.
Exp Cell Res ; 219(1): 159-68, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7628532

ABSTRACT

We have studied the effect of the cell synchronization agents compactin, ciclopirox olamine, mimosine, aphidicolin, ALLN, and colcemid on several parameters of cell cycle progression in mitotically synchronized HeLa S3 cells. Using cell size and cyclin A and B levels as markers of cytoplasmic progression and DNA content as a measure of nuclear cell cycle position, we have examined coordination of cytoplasmic and nuclear events during induction synchrony. Each synchronizing agent was unique in its effect on the coordination of the cytoplasmic and nuclear cycle. Mimosine, aphidicolin, ALLN, and colcemid disrupted cell cycle integration while compactin and ciclopirox olamine did not. Continued net cell growth during cell cycle arrest was the most dramatic in aphidicolin-treated cells, which averaged a 60% increase in size. Mimosine, ALLN, and colcemid produced an increase in cell size of approximately 25%, and ciclopyrox olamine and compactin exerted a negligible effect. Cyclin A and B were found at mitotic (high) or G1 (low) levels, or in combination of high and low concentrations not correlated with DNA content in drug-treated cells. For example, treatment with mimosine, which arrests cells in G1 with 2C DNA, resulted in cyclin A accumulating to mitotic levels, whereas cyclin B remained at a low concentration, the first time this phenomenon has been observed. These results demonstrate that populations of synchronized cells obtained by different drug treatments are blocked at biochemically distinct cell cycle points not apparent by cytometric measurement of DNA content. Our results provide conclusive evidence that induced synchrony methods differ with respect to their impact on cell cycle organization and from the pattern seen with nonperturbing cell selection methods.


Subject(s)
Cell Cycle/drug effects , Cell Nucleus/drug effects , Aphidicolin/pharmacology , Cell Cycle/physiology , Cell Nucleus/physiology , Cell Nucleus/ultrastructure , Ciclopirox , Cyclins/metabolism , Cytoplasm/drug effects , Cytoplasm/physiology , Cytoplasm/ultrastructure , DNA, Neoplasm/analysis , DNA, Neoplasm/metabolism , Demecolcine/pharmacology , HeLa Cells , Humans , Kinetics , Leupeptins/pharmacology , Mimosine/pharmacology , Protease Inhibitors/pharmacology , Pyridones/pharmacology , Time Factors
14.
Exp Cell Res ; 215(2): 373-9, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7982475

ABSTRACT

We have studied the relationship between apoptosis and drug-induced cell cycle perturbation in HeLa S3 cells when treated with the anti-tubulin drug colcemid. We found that at least two distinct mechanisms contributed to colcemid cytotoxicity and apoptosis. Continuous exposure to concentrations of colcemid sufficient to block cells at the mitotic checkpoint led to the appearance of apoptotic cells approximately one cell cycle after their initial accumulation in mitosis. Continuous exposure to concentrations sufficient to delay mitotic progression but insufficient to cause mitotic arrest, or pulse exposure to concentrations of colcemid sufficient to induce mitotic block, led to the generation of multipolar mitoses and genetically deficient hypodiploid daughter cells which underwent apoptosis while in interphase. The fact that aberrant spindle function delayed but did not block cells at the mitotic checkpoint indicates that the mitotic checkpoint senses the presence or absence of the spindle but not spindle abnormalities. In both mitotic and interphase cells, colcemid-induced apoptosis occurred after a period of cell cycle stasis during which cells failed to complete an initiated cell cycle. These results are discussed with reference to understanding the relationship between apoptosis and the regulation of cell cycle progression.


Subject(s)
Apoptosis/drug effects , Demecolcine/pharmacology , Apoptosis/physiology , Cell Cycle , Cyclins/metabolism , DNA/metabolism , HeLa Cells , Histones/metabolism , Humans , Interphase , Mitosis , Spindle Apparatus/physiology , Tubulin/metabolism , Tubulin Modulators
15.
Cancer Res ; 54(19): 5064-70, 1994 Oct 01.
Article in English | MEDLINE | ID: mdl-7923118

ABSTRACT

Chinese hamster ovary (and many rodent cell lines) transiently delay mitosis and progress into a second cell cycle without undergoing cytokinesis when treated with Colcemid, whereas HeLaS3 (and most human cell lines) arrest permanently in mitosis. We have discussed these differences and their consequences for cell survival under cell cycle-perturbing conditions within the context of mitotic checkpoint control (Schimke et al., Cold Spring Harbor Symp. Quant. Biol., 56: 417-425, 1991). Here, we report studies with mouse BALB/3T3 cell populations which, by the criterion of response to Colcemid, constitute a heterogeneous population with respect to mitotic checkpoint control. Clonal and subclonal populations retain population heterogeneity but with a bias for enrichment of cell populations that respond as do HeLaS3 cells. We have analyzed clones for their propensity for gene amplification as assessed by a stepwise increment selection protocol in methotrexate and report that there are significant differences in amplification propensities that correlate with differences in mitotic checkpoint control properties.


Subject(s)
Gene Amplification , Mitosis , 3T3 Cells , Animals , Cell Line, Transformed , Drug Resistance , Methotrexate/pharmacology , Mice , Tetrahydrofolate Dehydrogenase/genetics
16.
Exp Cell Res ; 211(2): 275-81, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8143773

ABSTRACT

Using a procedure to stain cells simultaneously for cyclin B1 protein and DNA, we have examined cyclin B1 expression by flow cytometry in human cells under a variety of perturbing and nonperturbing conditions. The method described is useful for measuring relative differences in cyclin B level (immunochemically detectable epitope) as a function of cell cycle position on an individual cell basis and thus to examine cell cycle-related changes in cyclin B expression without prior cell synchronization. We show that in HeLaS3 cells, cyclin B1 accumulates in cells only after they become 4C and have resided in G2 for a short period of time. During colcemid-induced mitotic arrest cyclin B1 continues to accumulate in HeLa S3 cells, and under specific conditions of aphidicolin-induced unbalanced cell growth induced, cyclin B accumulates to supranormal levels prior to mitosis. Flow cytometric analysis of cyclin B expression and DNA content permits detailed examination of the effects of cell cycle perturbations on cyclin B expression under a variety of conditions.


Subject(s)
Cyclins/genetics , Cyclins/metabolism , Aphidicolin/pharmacology , Cell Cycle/genetics , Cell Cycle/physiology , DNA/genetics , DNA/metabolism , Demecolcine/pharmacology , Flow Cytometry , Gene Expression , HeLa Cells , Humans
17.
Exp Cell Res ; 209(2): 238-47, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8262141

ABSTRACT

Using fluorescence-activated cell sorting combined with fluorescence microscopy the mechanism of embryonic germ cell death in the mouse has been shown to be apoptosis. Primordial germ cells (PGCs) from embryos at specific developmental stages have been analyzed, and cells with apoptotic morphology have been isolated by cell sorting. In the female, apoptotic oogonia at Day 13 and apoptotic oocytes at Days 15 and 17 were found. In the male, apoptotic cells were seen on Day 13 through Day 17. Apoptotic germ cells were not detected at Day 12 (combined male and female PGCs). Examination of sorted cells by fluorescence microscopy and by light microscopic analysis after alkaline phosphatase staining confirmed that the cells are apoptotic germ cells. Electron microscopy further confirmed that cells showing the morphological characteristics of apoptosis are present.


Subject(s)
Apoptosis , Mice, Inbred BALB C/embryology , Oocytes/cytology , Oogonia/cytology , Spermatogonia/cytology , Animals , Cell Cycle , Female , Flow Cytometry , Gestational Age , Light , Male , Mice , Scattering, Radiation
18.
J Biol Chem ; 268(31): 23072-80, 1993 Nov 05.
Article in English | MEDLINE | ID: mdl-8226824

ABSTRACT

We have shown previously that there are significant differences between mammalian cell lines in response to disruption of the assembly of the mitotic spindle apparatus (Kung, A. L., Sherwood, S. W., and Schimke, R. T. (1990) Proc. Natl. Acad. Sci. U. S. A. 87, 9553-9557). In this paper we report that there are also significant differences between mammalian cell lines in response to the inhibition of DNA synthesis. In HeLa S3 cells protein synthesis is down-regulated, and cellular growth is arrested in response to the inhibition of DNA synthesis. Upon release from inhibition and resumption of normal growth, cellular viability is maintained near untreated control levels. In contrast, Chinese hamster ovary cells continue to accumulate protein and continue to undergo cellular growth during the period of DNA synthesis inhibition. Cyclin B levels accumulate throughout the period of inhibition and rapidly exceed normal levels at mitosis. The degree of aberrant growth during the period of transient DNA synthesis inhibition is directly related to the degree of subsequent cytotoxicity. If protein accumulation and cellular growth are limited with partially inhibitory levels of cycloheximide during the period of DNA synthesis inhibition, the cytotoxic effects are abolished. These results support the concept that aberrant growth and accumulation of proteins during a transient period of DNA synthesis inhibition are primary determinants of subsequent cell killing.


Subject(s)
Aphidicolin/pharmacology , CHO Cells/drug effects , Cell Cycle/drug effects , Cyclins/metabolism , DNA Replication/drug effects , HeLa Cells/drug effects , Protein Biosynthesis , Animals , Cell Division/drug effects , Cell Survival/drug effects , Cricetinae , Cycloheximide/pharmacology , DNA/biosynthesis , Humans , In Vitro Techniques , Species Specificity , Time Factors
19.
Br J Cancer ; 67(6): 1229-35, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8512808

ABSTRACT

In this study we have investigated the interaction of caffeine, a prototypic methylxanthine, and TNF on the induction of cell death in mouse and human cell lines during progression from G1 to successive phases of the cell cycle. Exposure of cells to TNF (0.1-100 ng ml-1) as single agent for 48 h caused low or no lethality. The rates of cell death increased significantly when cells cultured with TNF for 24 h were exposed to caffeine (2.5-20 mM). The magnitude of the enhancement by caffeine was TNF and caffeine dose-dependent. The most effective response to this combination was observed in the mouse cell lines, WEHI and L929, followed by the human cell lines, HeLa, A375 and MCF-7, respectively. In L929 cells, TNF treatment did not inhibit DNA synthesis during the first S phase of the cell cycle (20-24 h), but it did block the progress toward a second S phase, indicating the cells were arrested at G2 phase or mitosis. Caffeine had great enhancer effect on L929 cells exposed to TNF for 24 h, but the effect was reduced in cells with either less than 24 h or greater than 28 h of exposure. L929 cells stimulated with TNF died via apoptosis, as judged by both morphological criteria and the occurrence of internucleosomal DNA cleavage. Exposure of TNF-treated cells to caffeine caused a greater increase in the proportion of apoptotic cells as well as the extent of internucleosomal DNA fragmentation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Caffeine/pharmacology , Neoplasms, Experimental/drug therapy , Tumor Necrosis Factor-alpha/pharmacology , Animals , Apoptosis/drug effects , Caffeine/adverse effects , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Death/drug effects , Cell Death/physiology , Drug Synergism , Humans , Mice , Neoplasms, Experimental/pathology , Tumor Cells, Cultured/drug effects , Tumor Necrosis Factor-alpha/administration & dosage
20.
Proc Natl Acad Sci U S A ; 90(8): 3353-7, 1993 Apr 15.
Article in English | MEDLINE | ID: mdl-8386372

ABSTRACT

The cytotoxic neutral cysteine protease inhibitor N-acetylleucylleucylnorleucinal (ALLN) inhibits cell-cycle progression in CHO cells, affecting the G1/S and metaphase-anaphase transition points, as well as S phase. Mitotic arrest induced by ALLN is associated with the inhibition of cyclin B degradation. At mitosis-inhibiting concentrations of ALLN, cells undergo nuclear-envelope breakdown, spindle formation, chromosome condensation, and congression to the metaphase plate. However, normal anaphase events do not occur, and cells arrest in a metaphase configuration for a prolonged period. Steady-state levels of cyclin B increase to greater than normal mitotic levels, and cyclin B is not degraded for an extended period. Histone H1 kinase activity remains elevated during mitotic arrest. Duration of mitotic arrest depends on ALLN concentration; high concentrations (> 50 micrograms/ml) produce a prolonged mitotic arrest, whereas at lower concentrations, cells are transiently delayed through mitosis (up to 4-12 hr), after which they undergo aberrant cell division resulting in randomly sized daughter cells containing variable amounts of DNA. Cyclin B degradation fails to occur, and histone H1 kinase remains activated for the duration of mitotic arrest at all ALLN concentrations.


Subject(s)
Cell Cycle/drug effects , Cyclins/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Leupeptins/pharmacology , Animals , CHO Cells , Cricetinae , Flow Cytometry , Fluorescent Antibody Technique , Kinetics , Mitosis/drug effects , Protamine Kinase/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...