Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Mol Cell Biol ; 20(1): 372-8, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10594039

ABSTRACT

The INK4 family of cyclin-dependent kinase (CDK) inhibitors includes four 15- to 19-kDa polypeptides (p16(INK4a), p15(INK4b), p18(INK4c), and p19(INK4d)) that bind to CDK4 and CDK6. By disrupting cyclin D-dependent holoenzymes, INK4 proteins prevent phosphorylation of the retinoblastoma protein and block entry into the DNA-synthetic phase of the cell division cycle. The founding family member, p16(INK4a), is a potent tumor suppressor in humans, whereas involvement, if any, of other INK4 proteins in tumor surveillance is less well documented. INK4c and INK4d are expressed during mouse embryogenesis in stereotypic tissue-specific patterns and are also detected, together with INK4b, in tissues of young mice. INK4a is expressed neither before birth nor at readily appreciable levels in young animals, but its increased expression later in life suggests that it plays some checkpoint function in response to cell stress, genotoxic damage, or aging per se. We used targeted gene disruption to generate mice lacking INK4d. These animals developed into adulthood, had a normal life span, and did not spontaneously develop tumors. Tumors did not arise at increased frequency in animals neonatally exposed to ionizing radiation or the carcinogen dimethylbenzanthrene. Mouse embryo fibroblasts, bone marrow-derived macrophages, and lymphoid T and B cells isolated from these animals proliferated normally and displayed typical lineage-specific differentiation markers. Males exhibited marked testicular atrophy associated with increased apoptosis of germ cells, although they remained fertile. The absence of tumors in INK4d-deficient animals demonstrates that, unlike INK4a, INK4d is not a tumor suppressor but is instead involved in spermatogenesis.


Subject(s)
Carrier Proteins/genetics , Cell Cycle Proteins , Cyclin-Dependent Kinase Inhibitor p16 , Fertility/genetics , Gene Expression Regulation/physiology , Testis/pathology , Animals , Cyclin-Dependent Kinase Inhibitor p19 , Cyclin-Dependent Kinases/antagonists & inhibitors , Male , Mice , Mice, Knockout , Testis/physiology
2.
Mol Cell Biol ; 11(2): 1171-5, 1991 Feb.
Article in English | MEDLINE | ID: mdl-1846666

ABSTRACT

Inhibition of okadaic acid-sensitive phosphatases released the cyclin degradation pathway from its inhibited state in extracts prepared from unfertilized Xenopus eggs arrested at the second meiotic metaphase. It also switched on cyclin protease activity in a permanent fashion in interphase extracts prepared from activated eggs. Even after cdc2 kinase inactivation, microinjection of okadaic acid-treated interphase extracts pushed G2-arrested recipient oocytes into the M phase, suggesting that the phosphatase inhibitor stabilizes the activity of an unidentified factor which shares in common with cdc2 kinase the maturation-promoting factor activity.


Subject(s)
Cyclins/metabolism , Ethers, Cyclic/pharmacology , Oocytes/metabolism , Phosphoprotein Phosphatases/metabolism , Animals , CDC2 Protein Kinase/metabolism , Cell Cycle/drug effects , Female , Homeostasis , Ionophores/pharmacology , Kinetics , Meiosis/drug effects , Metaphase , Models, Biological , Okadaic Acid , Oocytes/cytology , Oocytes/drug effects , Xenopus
3.
Mol Cell Biol ; 21(9): 3244-55, 2001 May.
Article in English | MEDLINE | ID: mdl-11287627

ABSTRACT

Male mice lacking both the Ink4c and Ink4d genes, which encode two inhibitors of D-type cyclin-dependent kinases (Cdks), are infertile, whereas female fecundity is unaffected. Both p18(Ink4c) and p19(Ink4d) are expressed in the seminiferous tubules of postnatal wild-type mice, being largely confined to postmitotic spermatocytes undergoing meiosis. Their combined loss is associated with the delayed exit of spermatogonia from the mitotic cell cycle, leading to the retarded appearance of meiotic cells that do not properly differentiate and instead undergo apoptosis at an increased frequency. As a result, mice lacking both Ink4c and Ink4d produce few mature sperm, and the residual spermatozoa have reduced motility and decreased viability. Whether or not Ink4d is present, animals lacking Ink4c develop hyperplasia of interstitial testicular Leydig cells, which produce reduced levels of testosterone. The anterior pituitary of fertile mice lacking Ink4c or infertile mice doubly deficient for Ink4c and Ink4d produces normal levels of luteinizing hormone (LH). Therefore, the failure of Leydig cells to produce testosterone is not secondary to defects in LH production, and reduced testosterone levels do not account for infertility in the doubly deficient strain. By contrast, Ink4d-null or double-null mice produce elevated levels of follicle-stimulating hormone (FSH). Because Ink4d-null mice are fertile, increased FSH production by the anterior pituitary is also unlikely to contribute to the sterility observed in Ink4c/Ink4d double-null males. Our data indicate that p18(Ink4c) and p19(Ink4d) are essential for male fertility. These two Cdk inhibitors collaborate in regulating spermatogenesis, helping to ensure mitotic exit and the normal meiotic maturation of spermatocytes.


Subject(s)
Carrier Proteins/physiology , Cell Cycle Proteins , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/metabolism , Enzyme Inhibitors , Proto-Oncogene Proteins , Spermatogenesis/physiology , Tumor Suppressor Proteins , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cyclin D , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , Cyclin-Dependent Kinase Inhibitor p18 , Cyclin-Dependent Kinase Inhibitor p19 , Cyclin-Dependent Kinases/biosynthesis , Female , Follicle Stimulating Hormone/metabolism , Infertility, Male , Luteinizing Hormone/metabolism , Male , Meiosis/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Protein Serine-Threonine Kinases/biosynthesis , Testis/metabolism , Testis/pathology
4.
Oncogene ; 36(37): 5231-5242, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28504719

ABSTRACT

Group3 medulloblastoma (MBG3) that predominantly occur in young children are usually associated with MYC amplification and/or overexpression, frequent metastasis and a dismal prognosis. Physiologically relevant MBG3 models are currently lacking, making inferences related to their cellular origin thus far limited. Using in utero electroporation, we here report that MBG3 mouse models can be developed in situ from different multipotent embryonic cerebellar progenitor cells via conditional expression of Myc and loss of Trp53 function in several Cre driver mouse lines. The Blbp-Cre driver that targets embryonic neural progenitors induced tumors exhibiting a large-cell/anaplastic histopathology adjacent to the fourth ventricle, recapitulating human MBG3. Enforced co-expression of luciferase together with Myc and a dominant-negative form of Trp53 revealed that GABAergic neuronal progenitors as well as cerebellar granule cells give rise to MBG3 with their distinct growth kinetics. Cross-species gene expression analysis revealed that these novel MBG3 models shared molecular characteristics with human MBG3, irrespective of their cellular origin. We here developed MBG3 mouse models in their physiological environment and we show that oncogenic insults drive this MB subgroup in different cerebellar lineages rather than in a specific cell of origin.


Subject(s)
Cerebellar Neoplasms/genetics , Cerebellum/embryology , Cerebellum/pathology , Medulloblastoma/genetics , Proto-Oncogene Proteins c-myc/genetics , Animals , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , Cerebellum/cytology , Cerebellum/metabolism , Disease Models, Animal , Female , Humans , Male , Medulloblastoma/metabolism , Medulloblastoma/pathology , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-myc/metabolism , Transfection
5.
Cancer Res ; 53(2): 235-8, 1993 Jan 15.
Article in English | MEDLINE | ID: mdl-8417814

ABSTRACT

Several in vitro studies have shown that the cyclin A gene is expressed and plays an important role in both the S and G2-M phases of the cell cycle. We analyzed cells from the blood and bone marrow of patients with various, mostly neoplastic, hematological disorders to determine whether (a) the cyclin A protein level correlated with that of cyclin A RNA and (b) cell distribution among the different phases of the cell cycle correlated with cyclin A RNA expression. Thirty-eight patients were studied by means of dot blot and Western blot techniques for cyclin A RNA and protein accumulation, and 21 were also studied for cell cycle distribution by using flow cytometric analysis. Semiquantitative studies were based on densitometric computerized evaluation of dot and Western blots. There was a very strong positive correlation between cyclin A RNA and protein expression (r = 0.99; P < 0.00005), indicating that cyclin A accumulation is regulated in these cells at a transcriptional level. There was also a highly significant positive correlation between cyclin A RNA expression and the cumulative percentage of cells in S plus G2-M phase (r = 0.98; P < 0.00005). Therefore, this in vivo study shows that the expression of cyclin A RNA and protein in human hematological malignancies correlates with the percentage of cells in S plus G2-M phase and identifies cyclin A as a new potential cell proliferation index in oncology.


Subject(s)
Cell Division , Cyclins/genetics , Leukemia/pathology , Lymphoma/pathology , Cell Cycle , Cyclins/metabolism , Gene Expression , Humans , Leukemia/diagnosis , Lymphoma/diagnosis , RNA, Messenger/genetics , RNA, Neoplasm/genetics
6.
Cancer Res ; 59(10): 2464-9, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10344759

ABSTRACT

The alternative reading frame product (p19ARF) of the mouse INK4a/ARF locus is induced by oncoproteins such as Myc and E1A as part of a checkpoint response that limits cell cycle progression in response to hyperproliferative signals. ARF binds directly to Mdm2 to prevent down-regulation of p53 and thereby promotes p53-dependent transcription and cell cycle arrest. However, ARF is not required for p53 induction in response to ionizing radiation or other forms of DNA damage. Animals lacking a functional ataxia telangiectasia (Atm) gene are exquisitely sensitive to ionizing radiation; Atm-null mouse embryo fibroblasts (MEFs) undergo premature replicative arrest, which is relieved by the loss of p53. Here we show that the loss of ARF expands the life expectancy of Atm-null MEFs, but alters neither the sensitivity of Atm-null mice to ionizing radiation nor their propensity to develop lymphomas early in life. Therefore, whereas ARF and Atm signal to p53 through distinct pathways, the loss of ARF can modify p53-dependent features of the Atm-null phenotype.


Subject(s)
Nuclear Proteins , Protein Serine-Threonine Kinases , Proteins/genetics , Proteins/physiology , Radiation Tolerance/genetics , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle/genetics , Cell Cycle Proteins , Cells, Cultured , DNA Damage , DNA Repair/genetics , DNA-Binding Proteins , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/radiation effects , Lymphoma/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Tumor Suppressor Protein p14ARF , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins
7.
Oncogene ; 7(8): 1653-6, 1992 Aug.
Article in English | MEDLINE | ID: mdl-1321406

ABSTRACT

We have previously reported the identification of a hepatitis B virus (HBV) DNA integration in an intron of the cyclin A gene in an early hepatocellular carcinoma (HCC) and the isolation of human cyclin A cDNA. We have now constructed a cDNA library from the tumor and isolated several hybrid HBV-cyclin A cDNAs from it. The hybrid cDNAs encode an HBV-cyclin A fusion protein. In the chimeric protein, the N-terminus of cyclin A, including the signals for cyclin degradation, is deleted and replaced by viral PreS2/S sequences, transcription being initiated from the viral PreS2/S promoter. This chimeric protein is undegradable in an in vitro cyclin degradation assay. Northern blot analyses showed strong expression of the hybrid transcripts in the tumor, while cyclin A- or HBV-specific transcripts were not detected in the non-tumorous liver of the same patient. Thus, HBV DNA integration in the cyclin A gene resulted in a strong expression of hybrid HBV-cyclin A transcripts encoding a stabilized cyclin A. This chimeric protein may play an important role in the development of the tumor.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cell Transformation, Neoplastic/genetics , Cyclins/genetics , Hepatitis B virus/genetics , Liver Neoplasms/genetics , Virus Integration , Base Sequence , Blotting, Northern , DNA, Viral/genetics , Humans , Introns/genetics , Molecular Sequence Data , Mutagenesis, Insertional/genetics , Proviruses/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
8.
Oncogene ; 15(2): 203-11, 1997 Jul 10.
Article in English | MEDLINE | ID: mdl-9244355

ABSTRACT

Four INK4 proteins can prevent cell proliferation by specifically inhibiting cyclin D-dependent kinases. Both p18INK4c and p19INK4d were widely expressed during mouse embryogenesis, but p16INK4a and p15INK4b were not readily detected prenatally. Although p15INK4b, p18INK4c and p19INK4d were demonstrated in many tissues by 4 weeks after birth, p16INK4a protein expression was restricted to the lung and spleen of older mice, with increased, more widespread mRNA expression during aging. Transcripts encoding the INK4a alternative reading frame product p19ARF were not detected before birth but were ubiquitous postnatally. Expression of p16INK4a and p15INK4b was induced when mouse embryos were disrupted and cultured as mouse embryo 'fibroblasts' (MEFs). The levels of p16INK4a and p18INK4c, but not p15INK4b or p19INK4d, further increased as MEFs approached senescence. Following crisis and establishment, three of four independently-derived cell lines became polyploid and expressed higher levels of functional p16INK4a. In contrast, one MEF line that sustained bi-allelic deletions of INK4a initially remained diploid. Therefore, loss of p16INK4a and other events predisposing to polyploidy may represent alternative processes contributing to cell immortalization. Whereas p18INK4c and p19INK4d may regulate pre- and postnatal development, p16INK4a more likely plays a checkpoint function during cell senescence that underscores its selective role as a tumor suppressor.


Subject(s)
Aging/metabolism , Carrier Proteins/genetics , Cell Cycle Proteins , Embryonic and Fetal Development , Enzyme Inhibitors , RNA, Messenger/analysis , Tumor Suppressor Proteins , Amino Acid Sequence , Animals , Carrier Proteins/analysis , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinase Inhibitor p18 , Gene Expression Regulation , Mice , Molecular Sequence Data
9.
Oncogene ; 10(8): 1501-9, 1995 Apr 20.
Article in English | MEDLINE | ID: mdl-7731704

ABSTRACT

Cyclin A is a cell cycle regulatory protein that functions in mitotic and S-phase control in mammalian cells. Using a genomic construction corresponding to the human cyclin A gene under the control of its own promoter, we have established stable transfectants overexpressing cyclin A protein. Experiments assisted by laser scanning image cytometry showed that this overexpression begins from late G1 phase onwards and is therefore cell cycle-regulated in this model. We demonstrated that this overexpression advances entry into S phase, leading to a contraction of the overall cell generation time. These results provide evidence that cyclin A can be a rate-limiting factor with respect to the control of the transition to S phase in mammalian cells.


Subject(s)
Cyclins/biosynthesis , S Phase , Cyclins/genetics , DNA/biosynthesis , G1 Phase , HeLa Cells , Humans , Transfection
10.
Article in English | MEDLINE | ID: mdl-16869746

ABSTRACT

The Ink4a-Arf locus encodes two closely wedded tumor suppressor proteins (p16(Ink4a) and p19(Arf)) that inhibit cell proliferation by activating Rb and p53, respectively. With few exceptions, the Arf gene is repressed during mouse embryonic development, thereby helping to limit p53 expression during organogenesis. However, in adult mice, sustained hyperproliferative signals conveyed by somatically activated oncogenes can induce Arf gene expression and trigger a p53 response that eliminates incipient cancer cells. Disruption of this tumor surveillance pathway predisposes to cancer, and inactivation of INK4a- ARF by deletion, silencing, or mutation has been frequently observed in many forms of human cancer. Although it is accepted that much of Arf's tumor-suppressive activity is mediated by p53, more recent genetic evidence has pointed to additional p53- independent functions of Arf, including its ability to inhibit gene expression by a number of other transcription factors. Surprisingly, the enforced expression of Arf in mammalian cells promotes the sumoylation of several Arf-interacting proteins, implying that Arf has an associated catalytic activity. We speculate that transcriptional down-regulation in response to Arf-induced sumoylation may account for Arf's p53-independent functions.


Subject(s)
Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Genes, p53 , Humans , Mice , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Sequence Homology, Amino Acid , Small Ubiquitin-Related Modifier Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Tumor Suppressor Protein p14ARF/chemistry , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
11.
Genes Dev ; 11(8): 957-72, 1997 Apr 15.
Article in English | MEDLINE | ID: mdl-9136925

ABSTRACT

The expression of D-type G1 cyclins and their assembly with their catalytic partners, the cyclin-dependent kinases 4 and 6 (CDK4 and CDK6), into active holoenzyme complexes are regulated by growth factor-induced signals. In turn, the ability of cyclin D-dependent kinases to trigger phosphorylation of the retinoblastoma (Rb) protein in the mid- to late G1 phase of the cell cycle makes the inactivation of Rb's growth suppressive function a mitogen-dependent step. The ability of D-type cyclins to act as growth factor sensors depends not only on their rapid induction by mitogens but also on their inherent instability, which ensures their precipitous degradation in cells deprived of growth factors. However, the mechanisms governing the turnover of D-type cyclins have not yet been elucidated. We now show that cyclin D1 turnover is governed by ubiquitination and proteasomal degradation, which are positively regulated by cyclin D1 phosphorylation on threonine-286. Although "free" or CDK4-bound cyclin D1 molecules are intrinsically unstable (t1/2 < 30 min), a cyclin D1 mutant (T286A) containing an alanine for threonine-286 substitution fails to undergo efficient polyubiquitination in an in vitro system or in vivo, and it is markedly stabilized (t1/2 approximately 3.5 hr) when inducibly expressed in either quiescent or proliferating mouse fibroblasts. Phosphorylation of cyclin D1 on threonine-286 also occurs in insect Sf9 cells, and although the process is enhanced significantly by the binding of cyclin D1 to CDK4, it does not depend on CDK4 catalytic activity. This implies that another kinase can phosphorylate cyclin D1 to accelerate its destruction and points to yet another means by which cyclin D-dependent kinase activity may be exogenously regulated.


Subject(s)
Cyclins/metabolism , Cysteine Endopeptidases/metabolism , Multienzyme Complexes/metabolism , Oncogene Proteins/metabolism , Proto-Oncogene Proteins , Threonine/metabolism , Ubiquitins/metabolism , 3T3 Cells , Animals , Cell Line , Cyclin D1 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/metabolism , Mice , Mutation , Phosphopeptides/analysis , Phosphorylation , Proteasome Endopeptidase Complex , Recombinant Fusion Proteins , Retinoblastoma Protein/metabolism , Spodoptera
12.
Cell ; 83(6): 993-1000, 1995 Dec 15.
Article in English | MEDLINE | ID: mdl-8521522

ABSTRACT

The INK4a (MTS1, CDKN2) gene encodes an inhibitor (p16INK4a) of the cyclin D-dependent kinases CDK4 and CDK6 that blocks them from phosphorylating the retinoblastoma protein (pRB) and prevents exit from the G1 phase of the cell cycle. Deletions and mutations involving INK4a occur frequently in cancers, implying that p16INK4a, like pRB, suppresses tumor formation. An unrelated protein (p19ARF) arises in major part from an alternative reading frame of the mouse INK4a gene, and its ectopic expression in the nucleus of rodent fibroblasts induces G1 and G2 phase arrest. Economical reutilization of coding sequences in this manner is practically without precedent in mammalian genomes, and the unitary inheritance of p16INK4a and p19ARF may underlie their dual requirement in cell cycle control.


Subject(s)
Carrier Proteins/genetics , Cell Cycle/genetics , Genes, Tumor Suppressor/genetics , Proteins/physiology , Reading Frames/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Cell Nucleus/chemistry , Cloning, Molecular , Cyclin-Dependent Kinase Inhibitor p16 , Exons/genetics , Humans , Mice , Molecular Sequence Data , Mutation/genetics , Organ Specificity , Protein Biosynthesis , Proteins/analysis , Proteins/genetics , RNA, Messenger/analysis , Sequence Homology, Amino Acid , Tumor Suppressor Protein p14ARF
13.
Exp Cell Res ; 206(1): 43-8, 1993 May.
Article in English | MEDLINE | ID: mdl-8097727

ABSTRACT

Cyclin A is a nuclear protein which is part of a kinase complex with either p34cdc2 or p33cdk2. Cyclin A is required in higher eukaryotic cells at the G1/S and the G2/M transitions. To examine the relationship between cyclin A and DNA replication, we simultaneously labeled exponentially growing HeLa cells for the distribution of cyclin A and proliferating cell nuclear antigen (PCNA). We have now demonstrated, by means of immunoelectron microscopy, that cyclin A is located at the sites of DNA replication visualized by both BrdU and PCNA labeling. Thus cyclin A may play a significant role in the phosphorylation of proteins at or near the sites of DNA replication.


Subject(s)
Cyclins/analysis , DNA Replication , Bromodeoxyuridine/metabolism , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cyclins/physiology , DNA/metabolism , HeLa Cells , Humans , Microscopy, Immunoelectron , Nuclear Proteins/analysis , Phosphorylation , Proliferating Cell Nuclear Antigen
14.
Hepatology ; 13(5): 936-40, 1991 May.
Article in English | MEDLINE | ID: mdl-1851495

ABSTRACT

Insulin-like growth factor II is believed to play an important role in fetal growth and development. The insulin-like growth factor II gene expression is tissue specific and developmentally regulated. We have previously shown an enhanced level of insulin-like growth factor II messenger RNA and protein in human hepatocellular carcinomas. This led to the suggestion that hepatocytes might be involved in insulin-like growth factor II expression. However, previous studies based on in situ hybridization only showed insulin-like growth factor II messenger RNA in liver sinusoidal cells. This paper reports on the analysis of the expression of insulin-like growth factor II and insulin-like growth factor II, insulin-like growth factor I and insulin receptor messenger RNAs in vivo in isolated rat hepatocytes at various stages of development and in vitro in adult rat hepatocytes primary culture. Our study indicates that isolated rat hepatocytes synthesize insulin-like growth factor II messenger RNA with a switch between fetal and adult messenger RNA profiles occurring 21 days after birth. In addition, adult rat hepatocytes in culture expressed insulin-like growth factor II messenger RNA and protein. Insulin-like growth factor II, insulin-like growth factor I and insulin receptor messenger RNAs were also detected. Therefore these results are consistent with the hypothesis that insulin-like growth factor II acts as an autocrine growth factor for hepatocytes in addition to having a paracrine effect. They also indicate that primary culture of hepatocytes is a good model for further studies on insulin-like growth factor II gene regulation.


Subject(s)
Gene Expression Regulation , Insulin-Like Growth Factor II/genetics , Liver/metabolism , Receptor, Insulin/genetics , Receptors, Cell Surface/genetics , Animals , Animals, Newborn , Cells, Cultured , Insulin-Like Growth Factor II/biosynthesis , Liver/cytology , Liver/embryology , Male , RNA, Messenger/biosynthesis , Rats , Receptor, Insulin/biosynthesis , Receptors, Cell Surface/biosynthesis , Receptors, Somatomedin
15.
J Hepatol ; 14(1): 30-4, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1310705

ABSTRACT

The insulin-like growth factor II (IGF-II) is involved in embryonic growth. Modifications of its expression might play a role in the development of primary liver cancer in humans and woodchucks. In the liver, little information is available on the cell types involved in its synthesis. We have investigated the expression of IGF-II as well as IGF-II, IGF-I and insulin receptor mRNAs in non parenchymal liver cell preparations in rats of various ages. The results indicate that Kupffer cells, endothelial cells and fat-storing cells express both IGF-II and the three different receptor mRNAs. Furthermore, a switch from a fetal to an adult IGF-II mRNA profile was obtained in the different cell preparations. Therefore, our results indicate that regulation of IGF-II gene expression can be analyzed through these isolated liver cell preparations. These results might also be important in investigating the potential role of IGF-II in liver carcinogenesis.


Subject(s)
Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor I/genetics , Liver/chemistry , Liver/cytology , RNA, Messenger/genetics , Receptor, Insulin/genetics , Receptors, Cell Surface/genetics , Animals , Blotting, Northern , Cells, Cultured , Gene Expression , Immunohistochemistry , Liver/ultrastructure , Male , Plasmids , Rats , Rats, Inbred Strains , Receptors, Somatomedin
16.
Proc Natl Acad Sci U S A ; 98(17): 9654-9, 2001 Aug 14.
Article in English | MEDLINE | ID: mdl-11481442

ABSTRACT

Establishment of cell lines from primary mouse embryo fibroblasts depends on loss of either the Arf tumor suppressor or its downstream target, the p53 transcription factor. Mouse p19(Arf) is encoded by the Ink4a-Arf locus, which also specifies a second tumor suppressor protein, the cyclin D-dependent kinase inhibitor p16(Ink4a). We surveyed bone marrow-derived cells from wild-type, Ink4a-Arf-null, or Arf-null mice for their ability to bypass senescence during continuous passage in culture. Unlike preB cells from wild-type mice, those from mice lacking Arf alone could be propagated indefinitely when placed onto stromal feeder layers engineered to produce IL-7. The preB cell lines remained diploid and IL-7-dependent and continued to express elevated levels of p16(Ink4a). By contrast, Arf-null bone marrow-derived macrophages that depend on colony-stimulating factor-1 for proliferation and survival in culture initially grew at a slow rate but gave rise to rapidly and continuously growing, but still growth factor-dependent, variants that ceased to express p16(Ink4a). Wild-type bone marrow-derived macrophages initially expressed both p16(Ink4a) and p19(Arf) but exhibited an extended life span when p16(Ink4a) expression was extinguished. In all cases, gene silencing was accompanied by methylation of the Ink4a promoter. Therefore, whereas Arf loss alone appears to be the major determinant of establishment of murine fibroblast and preB cell lines in culture, p16(Ink4a) provides an effective barrier to immortalization of bone marrow-derived macrophages.


Subject(s)
B-Lymphocytes/cytology , Bone Marrow Cells/cytology , Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p16/physiology , Genes, p16 , Hematopoietic Stem Cells/cytology , Macrophages/cytology , Proteins/physiology , 3T3 Cells , Animals , Apoptosis/radiation effects , B-Lymphocytes/metabolism , B-Lymphocytes/radiation effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/radiation effects , Cell Line, Transformed , Cell Lineage , Cell Transformation, Viral , Coculture Techniques , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Methylation , DNA, Complementary/genetics , Exons/genetics , Gene Expression Regulation , Gene Silencing , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/radiation effects , Macrophages/metabolism , Mice , Promoter Regions, Genetic , Proteins/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Simian virus 40/physiology , Transcription, Genetic , Tumor Suppressor Protein p14ARF
17.
Genes Dev ; 15(22): 2934-9, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11711428

ABSTRACT

Loss of Dmp1, an Arf transcriptional activator, leads to spontaneous tumorigenesis in mice, causing death from various forms of cancer by two years of age. Retention and expression of the wild-type Dmp1 allele in tumors arising in Dmp1(+/-) mice demonstrate that Dmp1 can be haplo-insufficient for tumor suppression. The mean latency of E(mu)-Myc-induced B-cell lymphomas is halved on a Dmp1(-/-) or Dmp1(+/-) genetic background. Although p53 mutations or Arf deletion normally occur in approximately 50% of E(mu)-Myc-induced lymphomas, Dmp1 loss obviates selection for such mutations, indicating that Dmp1 is a potent genetic modifier of the Arf-p53 pathway in vivo.


Subject(s)
Genes, p53/genetics , Mutation , Proto-Oncogene Proteins c-myc/genetics , Transcription Factors/genetics , Transcription Factors/physiology , Tumor Suppressor Protein p14ARF/genetics , Age Factors , Alleles , Animals , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA, Complementary/metabolism , Disease-Free Survival , Gene Deletion , Genotype , Immunoblotting , In Situ Hybridization , Lymphoma/chemically induced , Lymphoma/genetics , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplasms, Experimental , RNA/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Transcriptional Activation
18.
J Biol Chem ; 276(32): 29994-30001, 2001 Aug 10.
Article in English | MEDLINE | ID: mdl-11402027

ABSTRACT

Myc overexpression is a hallmark of human cancer and promotes transformation by facilitating immortalization. This function has been linked to the ability of c-Myc to induce the expression of the catalytic subunit of telomerase, telomerase reverse transcriptase (TERT), as ectopic expression of TERT immortalizes some primary human cell types. c-Myc up-regulates telomerase activity in primary mouse embryonic fibroblasts (MEFs) and myeloid cells. Paradoxically, Myc overexpression also triggers the ARF-p53 apoptotic program, which is activated when MEFs undergo replicative crises following culture ex vivo. The rare immortal variants that arise from these cultures generally suffer mutations in p53 or delete Ink4a/ARF, and Myc greatly increases the frequency of these events. Alternative reading frame (ARF)- and p53-null MEFs have increased telomerase activity, as do variant immortal clones that bypass replicative crisis. Similarly, immortal murine NIH-3T3 fibroblasts and myeloid 32D.3 and FDC-P1.2 cells do not express ARF and have robust telomerase activity. However, Myc overexpression in these immortal cells results in remarkably discordant regulation of TERT and telomerase activity. Furthermore, in MEFs and 32D.3 cells TERT expression and telomerase activity are regulated independently of endogenous c-Myc. Thus, the regulation of TERT and telomerase activity is complex and is also regulated by factors other than Myc, ARF, or p53.


Subject(s)
Proto-Oncogene Proteins c-myc/metabolism , Telomerase/metabolism , Transcription, Genetic , 3T3 Cells , Animals , Apoptosis , Blotting, Western , Cell Line , Cells, Cultured , Down-Regulation , Fibroblasts/metabolism , Humans , Mice , Mutation , Reading Frames , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Up-Regulation
19.
Biochem Biophys Res Commun ; 182(3): 1144-54, 1992 Feb 14.
Article in English | MEDLINE | ID: mdl-1531751

ABSTRACT

We have investigated cyclin A expression in a primary culture of normal rat hepatocytes and during rat liver regeneration after partial hepatectomy. In both cases, cyclin A mRNA and protein accumulate as the cells enter S phase. To investigate the potential implication of cyclin A accumulation at S phase, we microinjected anti-sense DNA constructs for cyclin A, resulting in effective inhibition of S phase entry. These effects were specific for cyclin A since anti-sense cyclin B construct had no similar effects. These results therefore, obtained in normal epithelial cells, indicate that cyclin A is involved in S phase and thus should not be only considered as a mitotic cyclin.


Subject(s)
Cyclins/physiology , Liver/physiology , S Phase , Animals , Autoradiography , Blotting, Northern , Cells, Cultured , Culture Media, Serum-Free , Cyclins/genetics , DNA Replication/drug effects , DNA, Antisense/pharmacology , Dexamethasone/pharmacology , Epithelial Cells , Epithelium/physiology , Hepatectomy , Immunohistochemistry , Insulin/pharmacology , Liver/cytology , Liver/drug effects , Male , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Rats, Inbred Strains , S Phase/drug effects , Thymidine/metabolism , Tritium
20.
Cell Growth Differ ; 8(11): 1139-50, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9372237

ABSTRACT

In situ hybridization of mouse embryo sections demonstrated expression of mRNAs encoding two polypeptide inhibitors (p18INK4c and p19INK4d) of cyclin D-dependent kinase (CDK) 4 and CDK6 in the central nervous system. No expression of two other INK4 members, p16INK4a and p15INK4b, was observed. The p19INK4d and p18INK4c proteins formed complexes with either CDK4 or CDK6 in a temporal pattern consistent with the results of in situ hybridization. Expression of INK4c was observed at embryonic day 13.5 in neuroepithelial zones of the developing brain, being restricted to dividing neuroblasts but absent from differentiating postmitotic neurons. In the neocortex, p18INK4c was expressed precisely at those developmental stages when neuroblasts switch from a symmetric to an asymmetric pattern of cell division with concomitant increases in their G1 interval. INK4d RNA was detected from embryonic day 11.5 onward, at higher levels than INK4c and with a distinctly different spatial and temporal pattern. Marked INK4d expression was seen in dorsal root ganglia, spinal cord, and focally throughout the brain, but primarily in postmitotic neurons. Neural expression of INK4d continued postnatally into adulthood in postmitotic cells of the dentate gyrus, the pyramidal layer of the hippocampus, and in discrete regions of the cerebral cortex, cerebellum, thalamus, and brainstem. Downregulation of p19INK4d in the dentate gyrus after kainic acid-induced seizures indicated that its expression could also be modified in nondividing cells by excitotoxic stress. Therefore, p19INK4d may contribute to maintaining the quiescent state, acting as a buffer to prevent reactivation of cyclin D-dependent kinases in terminally differentiated cells.


Subject(s)
Brain/embryology , Cell Cycle Proteins , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Inhibitors/metabolism , Proto-Oncogene Proteins , Tumor Suppressor Proteins , Animals , Brain/drug effects , Carrier Proteins/biosynthesis , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p18 , Cyclin-Dependent Kinase Inhibitor p19 , Dentate Gyrus/embryology , Excitatory Amino Acid Agonists/pharmacology , Kainic Acid/pharmacology , Mice , Neocortex/embryology
SELECTION OF CITATIONS
SEARCH DETAIL