Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 275(9): 6337-45, 2000 Mar 03.
Article in English | MEDLINE | ID: mdl-10692433

ABSTRACT

The transcription factor E2F plays an important role in G(1) to S phase transition in the higher eukaryotic cell cycle. Although a number of E2F-inducible genes have been identified, the biochemical cascades from E2F to the S phase entry remain to be investigated. In this study, we generated stably transfected mouse NIH3T3 cells that express exogenous human E2F-1 under the control of a heavy metal-inducible metallothionein promoter and analyzed the molecular mechanism of the E2F-1-mediated initiation of chromosomal DNA replication. Ectopic E2F-1 expression in cells arrested in G(0)/G(1) by serum deprivation enabled them to progress through G(1) and to enter S phase. During the G(1) progression, mouse cyclin E, but little of cyclin D1, was induced to express, which subsequently activated Cdk2. Experiments using the Cdk inhibitory proteins p27, p18, and p19 proved that the activity of Cdk2, but not of Cdk4, was required for S phase entry mediated by E2F-1. Minichromosome maintenance proteins (MCM) 4 and 7, the components of the DNA-replication initiation complex (RC), were constitutively expressed during the cell cycle, although the MCM genes are well known E2F-inducible genes. However, tight association of these two proteins with chromatin depended upon ectopic E2F-1 expression. In contrast, the Cdc45 protein, another RC component, which turned out to be a transcriptional target of E2Fs, was induced to express and subsequently bound to chromatin in response to E2F-1. Experiments utilizing a chemical Cdk-specific inhibitor, butyrolactone I, revealed that Cdk2 activity was required only for chromatin binding of the Cdc45 proteins, and not for the expression of Cdc45 or chromatin binding of MCM4 and -7. These results indicate that at least two separate pathways function downstream of E2F to initiate S phase; one depends upon the activity of Cdk2 and the other does not.


Subject(s)
CDC2-CDC28 Kinases , Carrier Proteins , Cyclin-Dependent Kinases/metabolism , Cyclins/pharmacology , Protein Serine-Threonine Kinases/metabolism , S Phase , Transcription Factors/metabolism , 3T3 Cells , 4-Butyrolactone/analogs & derivatives , 4-Butyrolactone/pharmacology , Animals , Base Sequence , Cell Cycle Proteins/metabolism , Chromatin/metabolism , Cyclin-Dependent Kinase 2 , DNA Replication/genetics , DNA-Binding Proteins/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , Humans , Interphase , Mice , Minichromosome Maintenance Complex Component 4 , Minichromosome Maintenance Complex Component 7 , Molecular Sequence Data , Nuclear Proteins/metabolism , Protein Binding , RNA, Messenger/metabolism , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcriptional Activation , Transfection
2.
J Biol Chem ; 273(26): 16494-500, 1998 Jun 26.
Article in English | MEDLINE | ID: mdl-9632717

ABSTRACT

Although no chromosomal DNA replication actually occurs during Xenopus oocyte maturation, the capability develops during the late meiosis I (MI) phase in response to progesterone. This ability, however, is suppressed by Mos proteins and maturation/mitosis promoting factor during the second meiosis phase (meiosis II; MII) until fertilization. Inhibition of RNA synthesis by actinomycin D during early MI prevented induction of the replication ability, but did not interfere with initiation of the meiotic cell cycle progression characterized by oscillation of the maturation/mitosis promoting factor activity and germinal vesicle breakdown. Microinjection of recombinant proteins such as dominant-negative E2F or universal Cdk inhibitors, p21 and p27, but not wild type human E2F-1 or Cdk4-specific inhibitor, p19, into maturing oocytes during MI abolished induction of the DNA replication ability. Co-injection of human E2F-1 and cyclin E proteins into immature oocytes allowed them to initiate DNA replication even in the absence of progesterone treatment. Injection of cyclin E alone, which was sufficient to activate endogenous Cdk2 kinase, failed to induce DNA replication. Moreover, the activation of Cdk2 was not affected under the conditions where DNA replication was blocked by actinomycin D. Thus, like somatic cells, both activities of E2F and cyclin E-Cdk2 complex are required for induction of the DNA replication ability in maturing Xenopus oocytes, and enhancement of both activities enables oocytes to override DNA-replication inhibitory mechanisms that specifically lie in maturing oocytes.


Subject(s)
CDC2-CDC28 Kinases , Carrier Proteins , Cell Cycle Proteins , Cyclin E/metabolism , Cyclin-Dependent Kinases/metabolism , DNA Replication , DNA-Binding Proteins , Oocytes/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , Chromosomes/metabolism , Cyclin-Dependent Kinase 2 , Dactinomycin/pharmacology , E2F Transcription Factors , E2F1 Transcription Factor , Enzyme Activation , Humans , Macromolecular Substances , Microinjections , Oncogene Proteins v-mos/metabolism , Protein Synthesis Inhibitors/pharmacology , RNA/biosynthesis , Retinoblastoma-Binding Protein 1 , S Phase/genetics , Transcription Factor DP1 , Xenopus , Xenopus Proteins
3.
Leukemia ; 11 Suppl 3: 347-51, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9209386

ABSTRACT

Mammalian D-type cyclins are differentially expressed during the first gap phase (G1) of the cell cycle in various cell types, and function as regulatory subunits of cyclin-dependent kinases (cdks), cdk4 and cdk6, to form holoenzymes whose activities are both necessary and rate limiting for G1 progression. Mitogenic signals induce the expression of cyclin D and cdk4 proteins, and facilitate their assembly into holoenzymes and their post-translational modification, while anti-proliferative stimuli extinguish the activity of cyclin D-dependent kinases by inducing cdk inhibitors which directly interfere with their catalytic functions and/or inhibit the post-translational activation of cyclin-bound cdks. Therefore, a variety of extracellular signals target and regulate the cyclin D/cdk4 serine/threonine kinases, which execute their critical functions during middle to late G1 phase by phosphorylating key substrates, including the retinoblastoma tumor suppressor gene products (pRb). Although overexpression of cyclin D, or inactivation of Rb or cdk inhibitor gene alone is not sufficient for cell transformation, high frequency of alterations of these genes in cancers suggests that inactivation of this particular pathway is involved in tumor development.


Subject(s)
Cell Cycle/physiology , Cyclins/physiology , Proto-Oncogene Proteins , Animals , Cell Division , Cell Transformation, Neoplastic , Cyclin D , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/metabolism , G1 Phase , Gene Expression , Humans , Mammals , Retinoblastoma Protein/metabolism , Signal Transduction
4.
Mol Cell Biol ; 15(6): 3247-55, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7760820

ABSTRACT

The E2A-HLF fusion gene, formed by the t(17;19)(q22;p13) translocation in childhood acute pro-B-cell leukemia, encodes a hybrid protein that contains the paired trans-activation domains of E2A (E12/E47) linked to the basic region/leucine zipper DNA-binding and dimerization domain of hepatic leukemia factor (HLF). To assess the transforming potential of this novel gene, we introduced it into NIH 3T3 murine fibroblasts by using an expression vector that also contained the neomycin resistance gene. Cells selected for resistance to the neomycin analog G418 formed aberrant colonies in monolayer cultures, marked by increased cell density and altered morphology. Transfected cells also grew readily in soft agar, producing colonies whose sizes correlated with E2A-HLF expression levels. Subclones expanded from colonies with high levels of the protein reproducibly formed tumors in nude mice and grew to higher plateau-phase cell densities in reduced-serum conditions than did parental NIH 3T3 cells. By contrast, NIH 3T3 cells expressing mutant E2A-HLF proteins that lacked either of the bipartite E2A trans-activation domains or the HLF leucine zipper domain failed to show oncogenic properties, including anchorage-independent cell growth. Thus, both of the E2A trans-activation motifs and the HLF leucine zipper dimerization domain are essential for the transforming potential of the chimeric E2A-HLF protein, suggesting a model in which aberrant regulation of the expression pattern of downstream target genes contributes to leukemogenesis.


Subject(s)
Adenovirus E2 Proteins/genetics , Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/genetics , Leucine Zippers/genetics , Oncogene Proteins, Fusion/genetics , 3T3 Cells , Adenovirus E2 Proteins/pharmacology , Animals , Base Sequence , Basic-Leucine Zipper Transcription Factors , DNA-Binding Proteins/pharmacology , Mice , Molecular Sequence Data , Oncogene Proteins, Fusion/pharmacology , Plasmids , Protein Folding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
5.
Mol Cell Biol ; 15(5): 2672-81, 1995 May.
Article in English | MEDLINE | ID: mdl-7739547

ABSTRACT

Cyclin D-dependent kinases act as mitogen-responsive, rate-limiting controllers of G1 phase progression in mammalian cells. Two novel members of the mouse INK4 gene family, p19 and p18, that specifically inhibit the kinase activities of CDK4 and CDK6, but do not affect those of cyclin E-CDK2, cyclin A-CDK2, or cyclin B-CDC2, were isolated. Like the previously described human INK4 polypeptides, p16INK4a/MTS1 and p15INK4b/MTS2, mouse p19 and p18 are primarily composed of tandemly repeated ankyrin motifs, each ca. 32 amino acids in length, p19 and p18 bind directly to CDK4 and CDK6, whether untethered or in complexes with D cyclins, and can inhibit the activity of cyclin D-bound cyclin-dependent kinases (CDKs). Although neither protein interacts with D cyclins or displaces them from preassembled cyclin D-CDK complexes in vitro, both form complexes with CDKs at the expense of cyclins in vivo, suggesting that they may also interfere with cyclin-CDK assembly. In proliferating macrophages, p19 mRNA and protein are periodically expressed with a nadir in G1 phase and maximal synthesis during S phase, consistent with the possibility that INK4 proteins limit the activities of CDKs once cells exit G1 phase. However, introduction of a vector encoding p19 into mouse NIH 3T3 cells leads to constitutive p19 synthesis, inhibits cyclin D1-CDK4 activity in vivo, and induces G1 phase arrest.


Subject(s)
Carrier Proteins/pharmacology , Cell Cycle Proteins , Cyclin-Dependent Kinases , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins , Tumor Suppressor Proteins , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p16 , DNA Primers/genetics , Humans , Macrophages/cytology , Macrophages/metabolism , Mice , Molecular Sequence Data , Protein Serine-Threonine Kinases/metabolism , Sequence Homology, Amino Acid , Tissue Distribution
6.
Cell ; 79(3): 487-96, 1994 Nov 04.
Article in English | MEDLINE | ID: mdl-7954814

ABSTRACT

Cyclic AMP (cAMP) blocks the mitogenic effects of colony-stimulating factor 1 (CSF-1) in macrophages, inducing cell cycle arrest in mid-G1 phase. Complexes between cyclin D1 and cyclin-dependent kinase 4 (cdk4) assemble in growth arrested cells, but cdk4 is not phosphorylated in vivo by the cdk-activating kinase (CAK) and remains inactive. Although undetectable in lysates of cAMP-treated cells, active CAK is recovered after antibody precipitation, indicating that it is not the direct target of inhibition. Levels of the cdk inhibitor p27Klp1 increase in cAMP-treated cells, and its immunodepletion from inhibitory lysates restores CAK-mediated cdk4 activation. Kip1 does not bind to CAK, but its association with cyclin D-cdk4 prevents CAK from phosphorylating and activating the holoenzyme.


Subject(s)
Cell Cycle Proteins , Cyclic AMP/pharmacology , Cyclin-Dependent Kinases , Fungal Proteins/metabolism , G1 Phase/physiology , Macrophages/physiology , Microtubule-Associated Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins , Tumor Suppressor Proteins , Animals , Antigen-Antibody Complex , Cells, Cultured , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p27 , Enzyme Activation , G1 Phase/drug effects , Macrophages/drug effects , Mice , Peptide Mapping , Phosphorylation , Polyenes/pharmacology , Precipitin Tests , Protein Serine-Threonine Kinases/antagonists & inhibitors , Sirolimus , Time Factors , Cyclin-Dependent Kinase-Activating Kinase
7.
Mol Cell Biol ; 14(11): 7265-75, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7935441

ABSTRACT

The assembly of functional holoenzymes composed of regulatory D-type cyclins and cyclin-dependent kinases (cdks) is rate limiting for progression through the G1 phase of the mammalian somatic cell cycle. Complexes between D-type cyclins and their major catalytic subunit, cdk4, are catalytically inactive until cyclin-bound cdk4 undergoes phosphorylation on a single threonyl residue (Thr-172). This step is catalyzed by a cdk-activating kinase (CAK) functionally analogous to the enzyme which phosphorylates cdc2 and cdk2 at Thr-161/160. Here, we demonstrate that the catalytic subunit of mouse cdc2/cdk2 CAK (a 39-kDa protein designated p39MO15) can assemble with a regulatory protein present in either insect or mammalian cells to generate a CAK activity capable of phosphorylating and enzymatically activating both cdk2 and cdk4 in complexes with their respective cyclin partners. A newly identified 37-kDa cyclin-like protein (cyclin H [R. P. Fisher and D. O. Morgan, Cell 78:713-724, 1994]) can assemble with p39MO15 to activate both cyclin A-cdk2 and cyclin D-cdk4 in vitro, implying that CAK is structurally reminiscent of cyclin-cdk complexes themselves. Antisera produced to the p39MO15 subunit can completely deplete mammalian cell lysates of CAK activity for both cyclin A-cdk2 and cyclin D-cdk4, with recovery of activity in the resulting immune complexes. By using an immune complex CAK assay, CAK activity for cyclin A-cdk2 and cyclin D-cdk4 was detected both in quiescent cells and invariantly throughout the cell cycle. Therefore, although it is essential for the enzymatic activation of cyclin-cdk complexes, CAK appears to be neither rate limiting for the emergence of cells from quiescence nor subject to upstream regulatory control by stimulatory mitogens.


Subject(s)
CDC2-CDC28 Kinases , Cyclin-Dependent Kinases , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins , Amino Acid Sequence , Animals , Base Sequence , Cell Cycle , Cloning, Molecular , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , DNA Probes/genetics , DNA, Complementary/genetics , Enzyme Activation , Gene Expression , Humans , Mice , Molecular Sequence Data , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Recombinant Fusion Proteins/metabolism , Xenopus , Xenopus Proteins , Cyclin-Dependent Kinase-Activating Kinase
8.
Mol Cell Biol ; 14(4): 2713-21, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8139570

ABSTRACT

The accumulation of assembled holoenzymes composed of regulatory D-type cyclins and their catalytic partner, cyclin-dependent kinase 4 (cdk4), is rate limiting for progression through the G1 phase of the cell cycle in mammalian fibroblasts. Both the synthesis and assembly of D-type cyclins and cdk4 depend upon serum stimulation, but even when both subunits are ectopically overproduced, they do not assemble into complexes in serum-deprived cells. When coexpressed from baculoviral vectors in intact Sf9 insect cells, cdk4 assembles with D-type cyclins to form active protein kinases. In contrast, recombinant D-type cyclin and cdk4 subunits produced in insect cells or in bacteria do not assemble as efficiently into functional holoenzymes when combined in vitro but can be activated in the presence of lysates obtained from proliferating mammalian cells. Assembly of cyclin D-cdk4 complexes in coinfected Sf9 cells facilitates phosphorylation of cdk4 on threonine 172 by a cdk-activating kinase (CAK). Assembly can proceed in the absence of this modification, but cdk4 mutants which cannot be phosphorylated by CAK remain catalytically inactive. Therefore, formation of the cyclin D-cdk4 complex and phosphorylation of the bound catalytic subunit are independently regulated, and in addition to the requirement for CAK activity, serum stimulation is required to promote assembly of the complexes in mammalian cells.


Subject(s)
Cell Cycle/physiology , Cyclin-Dependent Kinases , Cyclins/biosynthesis , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cyclin-Dependent Kinase 4 , DNA Primers , Fibroblasts/cytology , Fibroblasts/enzymology , G1 Phase , Homeostasis , Mammals , Molecular Sequence Data , Moths , Mutagenesis, Site-Directed , Peptide Fragments/analysis , Phosphopeptides/analysis , Point Mutation , Polymerase Chain Reaction , Protein Kinases/biosynthesis , Transfection
9.
Mol Cell Biol ; 14(3): 2066-76, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8114738

ABSTRACT

D-type cyclin-dependent kinase activities have not so far been detected in mammalian cells. Lysis of rodent fibroblasts, mouse macrophages, or myeloid cells with Tween 20 followed by precipitation with antibodies to cyclins D1, D2, and D3 or to their major catalytic partner, cyclin-dependent kinase 4 (cdk4), yielded kinase activities in immune complexes which readily phosphorylated the retinoblastoma protein (pRb) but not histone H1 or casein. Virtually all cyclin D1-dependent kinase activity in proliferating macrophages and fibroblasts could be attributed to cdk4. When quiescent cells were stimulated by growth factors to enter the cell cycle, cyclin D1-dependent kinase activity was first detected in mid G1, reached a maximum near the G1/S transition, and remained elevated in proliferating cells. The rate of appearance of kinase activity during G1 phase lagged significantly behind cyclin induction and correlated with the more delayed accumulation of cdk4 and formation of cyclin D1-cdk4 complexes. Thus, cyclin D1-associated kinase activity was not detected during the G0-to-G1 transition, which occurs within the first few hours following growth factor stimulation. Rodent fibroblasts engineered to constitutively overexpress either cyclin D1 alone or cyclin D3 together with cdk4 exhibited greatly elevated cyclin D-dependent kinase activity, which remained absent in quiescent cells but rose to supraphysiologic levels as cells progressed through G1. Therefore, despite continued enforced overproduction of cyclins and cdk4, the assembly of cyclin D-cdk4 complexes and the appearance of their kinase activities remained dependent upon serum stimulation, indicating that upstream regulators must govern formation of the active enzymes.


Subject(s)
Cell Cycle , Cyclin-Dependent Kinases , Cyclins/physiology , Oncogene Proteins/physiology , Protein Kinases/physiology , Proto-Oncogene Proteins , 3T3 Cells , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Cell Line , Cyclin D1 , Cyclin D3 , Cyclin-Dependent Kinase 4 , Cyclins/metabolism , Fibroblasts/enzymology , Macrophages/enzymology , Mice , Molecular Sequence Data , Recombinant Proteins , Retinoblastoma Protein/metabolism
10.
Hybridoma ; 13(1): 37-44, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8200657

ABSTRACT

D-type cyclins are necessary and rate-limiting for G1 progression during the mammalian cell cycle. Cyclins D1, D2, and D3 are encoded by distinct genes and are expressed in proliferating cells in a lineage-specific manner. Monoclonal antibodies (mAbs) generated to bacterially produced recombinant D-type cyclins were able to react with the native proteins expressed in mammalian cells. One mouse and three rat mAbs immunoprecipitated cyclin D1 from mouse macrophages. Only rat mAbs reacted with human cyclin D1 and cross-reacted with cyclin D2 expressed in proliferating T lymphocytes and human tumor cell lines. A single rat mAb to cyclin D2 exhibited a pattern of reactivity reciprocal to that of rat mAbs to D1. Three rat mAbs reacted specifically with mouse or human cyclin D3, but did not cross-react with cyclins D1 or D2 from either species. Representative mAbs were useful for immunoblotting and detected D-type cyclins coprecipitating in complexes recovered with antiserum to cyclin-dependent kinase-4 (CDK4). Because these mAbs detect D-type cyclins in the nuclei of fixed permeabilized cells, they should prove useful in documenting cyclin overexpression in those human tumors in which the genes are amplified or are targets of specific chromosomal rearrangements.


Subject(s)
Antibodies, Monoclonal/immunology , Cyclins/immunology , G1 Phase/immunology , Animals , Blotting, Western , Cross Reactions , Cyclin D1 , Cyclin D2 , Cyclin D3 , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Mice , Oncogene Proteins/immunology , Precipitin Tests , Rats
11.
Genes Dev ; 8(1): 9-22, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8288131

ABSTRACT

Cell-cell contact and TGF-beta can arrest the cell cycle in G1. Mv1Lu mink epithelial cells arrested by either mechanism are incapable of assembling active complexes containing the G1 cyclin, cyclin E, and its catalytic subunit, Cdk2. These growth inhibitory signals block Cdk2 activation by raising the threshold level of cyclin E necessary to activate Cdk2. In arrested cells the threshold is set higher than physiological cyclin E levels and is determined by an inhibitor that binds to cyclin E-Cdk2 complexes. A 27-kD protein that binds to and prevents the activation of cyclin E-Cdk2 complexes can be purified from arrested cells but not from proliferating cells, using cyclin E-Cdk2 affinity chromatography. p27 is present in proliferating cells, but it is sequestered and unavailable to interact with cyclin E-Cdk2 complexes. Cyclin D2-Cdk4 complexes bind competitively to and down-regulate the activity of p27 and may thereby act in a pathway that reverses Cdk2 inhibition and enables G1 progression.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle , Cyclin-Dependent Kinases , Cyclins/metabolism , Protein Kinases/metabolism , Proteins/metabolism , Transforming Growth Factor beta/physiology , Animals , Cell Communication , Cell Line , Cyclin-Dependent Kinase 2 , Mink , Protein Kinase Inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Cyclin-Dependent Kinase-Activating Kinase
12.
Proc Natl Acad Sci U S A ; 90(24): 11513-7, 1993 Dec 15.
Article in English | MEDLINE | ID: mdl-7505440

ABSTRACT

Growth factor-induced signals govern the expression of three D-type cyclins, which, in turn, function as regulatory subunits of cyclin-dependent kinases (cdks) to control cell cycle transitions during the late G1 interval. 32D myeloid cells, which self-renew as uncommitted precursors in interleukin 3 (IL-3), express cyclins D2 and D3 (but not D1) in complexes with cdk4 and cdk2. When transferred to granulocyte colony-stimulating factor (G-CSF), 32D cells stop dividing and terminally differentiate to mature neutrophils. Cyclin D and cdk4 expression ceased as cells underwent growth arrest in G-CSF, but cdk2 levels were sustained. 32D cells engineered to ectopically express D-type cyclins exhibited contracted G1 intervals with a compensatory lengthening of S phase but remained IL-3 dependent for cell growth; those overexpressing cyclins D2 and D3 (but not D1) were unable to differentiate and died in G-CSF. Cyclin D2 mutants, which cannot efficiently bind to, or functionally interact with, the retinoblastoma protein (pRb) or its relatives (p107) did not block differentiation. Conversely, the introduction of a catalytically inactive cdk4 mutant into cells overexpressing cyclin D2 restored their G-CSF response. The persistence of cdk2 and its predilection to functionally interact with cyclins D2 and D3 rather than D1 might explain the specificity of the differentiation blockade.


Subject(s)
Cell Cycle , Cell Differentiation/physiology , Cyclins/physiology , Granulocyte Colony-Stimulating Factor/physiology , Granulocytes/cytology , Neutrophils/physiology , Oncogene Proteins/physiology , Amino Acid Sequence , Animals , Cell Line , Clone Cells , Cyclin D1 , Cyclin D2 , Cyclin D3 , Cyclins/biosynthesis , G1 Phase , Granulocytes/physiology , Mammals , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Neutrophils/cytology , Oncogene Proteins/biosynthesis , Protein Kinases/metabolism , Recombinant Proteins/biosynthesis , Retinoblastoma Protein/physiology , Transfection
13.
Genes Dev ; 7(8): 1559-71, 1993 Aug.
Article in English | MEDLINE | ID: mdl-8339933

ABSTRACT

Mammalian D-type cyclins are growth factor-regulated, delayed early response genes that are presumed to control progression through the G1 phase of the cell cycle by governing the activity of cyclin-dependent kinases (cdks). Overexpression of mouse cyclin D1 in serum-stimulated mouse NIH-3T3 and rat-2 fibroblasts increased their rates of G0 to S- and G1- to S-phase transit by several hours, leading to an equivalent contraction of their mean cell generation times. Although such cells remained contact inhibited and anchorage dependent, they manifested a reduced serum requirement for growth and were smaller in size than their normal counterparts. Ectopic expression of cyclin D2 in rodent fibroblasts, either alone or together with exogenous cdk4, shortened their G0- to S-phase interval and reduced their serum dependency, but cyclin D2 alone did not alter cell size significantly. When cells were microinjected during the G1 interval with a monoclonal antibody specifically reactive to cyclin D1, parental rodent fibroblasts and derivatives overexpressing this cyclin were inhibited from entering S phase, but cells injected near the G1/S phase transition were refractory to antibody-induced growth suppression. Thus, cyclin D1, and most likely D2, are rate limiting for G1 progression.


Subject(s)
Cyclin-Dependent Kinases , Cyclins/physiology , G1 Phase , Oncogene Proteins/physiology , Proto-Oncogene Proteins , 3T3 Cells , Animals , Cell Size , Cyclin D1 , Cyclin D2 , Cyclin-Dependent Kinase 4 , Cyclins/biosynthesis , Cyclins/genetics , DNA Replication , Fibroblasts/drug effects , Fibroblasts/metabolism , G1 Phase/drug effects , Gene Expression , Mice , Oncogene Proteins/biosynthesis , Oncogene Proteins/genetics , Protein Kinases/metabolism , Rats
14.
Cell ; 71(2): 323-34, 1992 Oct 16.
Article in English | MEDLINE | ID: mdl-1423597

ABSTRACT

Murine D type cyclins associate with a catalytic subunit (p34PSK-J3) with properties distinct from known cyclin-dependent kinases (cdks). Mouse p34PSK-J3 shows less than 50% amino acid identity to p34cdc2, p33cdk2, and p36cdk3, lacks a PSTAIRE motif, and does not bind to p13suc1. Cyclin D1-p34PSK-J3 complexes accumulate in macrophages during G1 and decline in S phase, whereas complexes involving cyclins D2 and D3 form in proliferating T cells. Although histone H1 kinase activity is not detected in cyclin D or PSK-J3 immunoprecipitates, cyclin D-p34PSK-J3 complexes assembled in vitro stably bind and phosphorylate the retinoblastoma gene product (pRb) and an Rb-like protein (p107) but do not interact with pRb mutants that are functionally inactive. Thus, p34PSK-J3 is a cyclin D-regulated catalytic subunit that acts as an Rb (but not H1) kinase.


Subject(s)
Cyclin-Dependent Kinases , Cyclins/chemistry , Oncogene Proteins/chemistry , Protein Kinases/chemistry , Proto-Oncogene Proteins , Amino Acid Sequence , Animals , Base Sequence , CDC2 Protein Kinase , Cells, Cultured , Cyclin D1 , Cyclin-Dependent Kinase 4 , G1 Phase , Humans , Insecta , Macrophages , Mice , Molecular Sequence Data , Protein Kinases/genetics , Protein Kinases/isolation & purification , Sequence Homology, Amino Acid
15.
Mol Cell Biol ; 10(5): 2407-12, 1990 May.
Article in English | MEDLINE | ID: mdl-1691441

ABSTRACT

A chimeric receptor composed of the extracellular domain of the human T-cell antigen CD2 (T11) joined to the membrane-spanning segment and the intracellular tyrosine kinase domain of the human colony-stimulating factor 1 receptor (CSF-1R) was expressed in murine NIH 3T3 fibroblasts. Stimulation of these cells with monoclonal antibodies to CD2 induced phosphorylation of the chimeric glycoprotein on tyrosine, receptor downmodulation, and mitogenesis. In contrast, neither human CSF-1R nor the chimeric receptor was able to function in interleukin-2-dependent murine T cells. In fibroblasts, then, CSF-1 per se is not required for activation of the receptor kinase or for a biological response, whereas in T cells, CSF-1R may be unable to engage the downstream signal transduction machinery.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/physiology , Cell Division , Proto-Oncogene Proteins/physiology , Receptors, Cell Surface/physiology , Receptors, Immunologic/physiology , Amino Acid Sequence , Antibodies, Monoclonal/immunology , Base Sequence , CD2 Antigens , Down-Regulation , Epitopes , Molecular Sequence Data , Phosphotyrosine , Protein-Tyrosine Kinases/physiology , Receptor, Macrophage Colony-Stimulating Factor , Recombinant Fusion Proteins , Restriction Mapping , Signal Transduction , Tyrosine/analogs & derivatives , Tyrosine/metabolism
16.
Ciba Found Symp ; 148: 96-104; discussion 104-9, 1990.
Article in English | MEDLINE | ID: mdl-2156660

ABSTRACT

Colony-stimulating factor-1 (CSF-1 or M-CSF) supports the proliferation and survival of mononuclear phagocytes by binding to a receptor (CSF-1R) encoded by the c-fms proto-oncogene. Whereas the CSF-1R kinase is normally regulated by ligand, receptors bearing 'activating mutations' act constitutively as enzymes and can transform fibroblasts and haemopoietic cells of different lineages. Introduction of human CSF-1R enables mouse NIH-3T3 cells to form colonies in agar in response to human CSF-1 and to proliferate in serum-free medium supplemented with CSF-1, albumin, transferrin and insulin. Similarly, expression of human CSF-1R in interleukin 3-dependent mouse FDC-P1 myeloid cells enables them to grow in CSF-1. High levels of CSF-1R expression in FDC-P1 cells can induce factor-independent growth which is abrogated by a 'neutralizing' monoclonal antibody to the receptor. Therefore, critical mutations in the c-fms gene or overexpression of CSF-1R in immature myeloid precursors might each contribute to leukaemia.


Subject(s)
Proto-Oncogene Proteins/physiology , Receptors, Cell Surface/genetics , Signal Transduction/drug effects , Animals , Cell Line, Transformed , Cell Transformation, Neoplastic/drug effects , Humans , Mice , Mutation , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Receptor, Macrophage Colony-Stimulating Factor , Receptors, Colony-Stimulating Factor , Signal Transduction/physiology
17.
Mol Cell Biol ; 9(9): 4069-73, 1989 Sep.
Article in English | MEDLINE | ID: mdl-2528688

ABSTRACT

A retroviral vector encoding the receptor for human colony-stimulating factor-1 (CSF-1) was introduced into murine myeloid FDC-P1 cells which require interleukin-3 (IL-3) for their proliferation and survival in culture. Cells expressing the CSF-1 receptor (CSF-1R), selected by fluorescence-activated cell sorting in the continued presence of murine IL-3, formed colonies in semisolid medium and were able to proliferate continuously in liquid cultures containing human recombinant CSF-1. Thus, although they do not synthesize endogenous murine CSF-1R, FDC-P1 cells express the downstream components of the CSF-1 mitogenic pathway necessary for its signal-response coupling. After receptor transduction, slowly proliferating factor-independent variants that produced neither CSF-1 nor growth factors able to support the proliferation of parental FDC-P1 cells also arose. When the human CSF-1R was expressed in FDC-P1 cells under the control of an inducible metallothionein promoter, the frequencies of both CSF-1-responsive and factor-independent variants increased after heavy-metal treatment. In addition, a monoclonal antibody to human CSF-1R arrested colony formation by both the CSF-1-dependent and factor-independent cells but did not affect their growth in response to IL-3. Therefore, the induction of both the CSF-1-dependent and factor-independent phenotypes depended on expression of the transduced human CSF-1R.


Subject(s)
Colony-Stimulating Factors/pharmacology , Proto-Oncogene Proteins/genetics , Animals , Cell Division/drug effects , Cells, Cultured , Genetic Vectors , Humans , Interleukin-3/pharmacology , Mice , Receptor, Macrophage Colony-Stimulating Factor , Transduction, Genetic
18.
Mol Cell Biol ; 6(12): 4155-60, 1986 Dec.
Article in English | MEDLINE | ID: mdl-2432397

ABSTRACT

We have previously shown that Rous sarcoma virus variants that carry the cellular homolog (c-src) of the viral src gene (v-src) do not transform chicken embryo fibroblasts. We also have shown that replacement of sequences upstream or downstream from the BglI site of the cellular src gene with the corresponding regions of v-src restored transforming activity to the hybrid genes. Since there are only six amino acid changes between p60c-src and p60v-src within the sequences upstream from BglI, we constructed chimeric molecules involving v-src and c-src to determine the effect of each amino acid substitution on the biological activities of the gene product. We found that the change from Thr to Ile at position 338 or the replacement of a fragment of c-src containing Gly-63, Arg-95, and Thr-96 with a corresponding fragment of v-src containing Asp-63, Trp-95, and Ile-96 converted p60c-src into a transforming protein by the criteria of focus formation, anchorage-independent growth, and tumor formation in newborn chickens. These mutations also resulted in elevation of the protein kinase activity of p60c-src.


Subject(s)
Avian Sarcoma Viruses/genetics , Cell Transformation, Neoplastic , Genes, Viral , Genes , Genetic Variation , Proto-Oncogene Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Chick Embryo , Fibroblasts/metabolism , Plasmids , Protein Kinases/genetics , Protein Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins pp60(c-src)
SELECTION OF CITATIONS
SEARCH DETAIL
...