Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Signal ; 21(6): 926-35, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19385066

ABSTRACT

A phosphoinositide signalling cycle is present in the nucleus, independent of that which occurs at the plasma membrane. The key enzyme involved in this cycle is phospholipase (PLC) beta1. This nuclear cycle has been shown to be involved in both cell proliferation and differentiation. Here, we report that nuclear PLCbeta1 activity is upregulated during differentiation of 3T3-L1 adipocytes. During differentiation there are two phases of PLCbeta1 activity; the first occurs within 5 min of treatment with differentiation media, does not require new PLCbeta1 to enter the nucleus and is regulated by pERK and PKC alpha while the second phase occurs from day 2 of differentiation, requires new PLCbeta1 protein to enter the nucleus and is independent of regulation by pERK and PKC alpha. Over-expression with the PLC mutants, Deltamk (which lacks the ERK phosphorylation site) and M2B (which lacks the nuclear localisation sequence), revealed that both phases of PLCbeta1 activity are required for terminal differentiation to occur. Inhibition of PLCbeta1 activity prevents the upregulation of cyclinD3 and cdk4 protein, suggesting that PLCbeta1 plays a role in the control of the cell cycle during differentiation. These results indicate nuclear PLCbeta1 as a key regulator of adipocyte differentiation.


Subject(s)
Adipocytes/cytology , Adipocytes/enzymology , Cell Differentiation , Cell Nucleus/enzymology , Cyclin-Dependent Kinase 4/metabolism , Cyclins/metabolism , Phospholipase C beta/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Animals , Cell Differentiation/drug effects , Cell Nucleus/drug effects , Cyclin D3 , Down-Regulation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Isoenzymes/metabolism , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mutant Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Kinase C-alpha/metabolism , Protein Kinase Inhibitors/pharmacology , Up-Regulation/drug effects
2.
Endocrinology ; 148(3): 1108-17, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17122077

ABSTRACT

Our main goal in this study was to investigate the role of phospholipase C (PLC) beta(1) and PLCgamma(1) in skeletal muscle differentiation and the existence of potential downstream targets of their signaling activity. To examine whether PLC signaling can modulate the expression of cyclin D3, a target of PLCbeta(1) in erythroleukemia cells, we transfected C2C12 cells with expression vectors containing PLCbeta(1) or PLCgamma(1) cDNA and with small interfering RNAs from regions of the PLCbeta(1) or PLCgamma(1) gene and followed myogenic differentiation in this well-established cell system. Intriguingly, overexpressed PLCbeta(1) and PLCgamma(1) were able to mimic insulin induction of both cyclin D3 and muscle differentiation. By knocking down PLCbeta(1) or PLCgamma(1) expression, C2C12 cells almost completely lost the increase in cyclin D3, and the differentiation program was down-regulated. To explore the induction of the cyclin D3 gene promoter during this process, we used a series of 5'-deletions of the 1.68-kb promoter linked to a reporter gene and noted a 5-fold augmentation of promoter activity upon insulin stimulation. These constructs were also cotransfected with PLCbeta(1) or PLCgamma(1) cDNAs and small interfering RNAs, respectively. Our data indicate that PLCbeta(1) or PLCgamma(1) signaling is capable of acting like insulin in regard to both the myogenic differentiation program and cyclin D3 up-regulation. Taken together, this is the first study that hints at cyclin D3 as a target of PLCbeta(1) and PLCgamma(1) during myogenic differentiation in vitro and implies that up-regulation of these enzymes is sufficient to mimic the actions of insulin in this process.


Subject(s)
Cyclins/genetics , Insulin/physiology , Isoenzymes/physiology , Muscle Development , Muscle, Skeletal/enzymology , Type C Phospholipases/physiology , Animals , Cells, Cultured , Cyclin D3 , Cyclins/metabolism , Cyclins/physiology , Immunohistochemistry , Isoenzymes/metabolism , Mice , Muscle Development/drug effects , Myogenin/physiology , Phospholipase C beta , Phospholipase C gamma/metabolism , Phospholipase C gamma/physiology , Promoter Regions, Genetic , RNA, Small Interfering/pharmacology , Signal Transduction , Type C Phospholipases/metabolism
3.
Biochim Biophys Acta ; 1761(5-6): 509-21, 2006.
Article in English | MEDLINE | ID: mdl-16624616

ABSTRACT

Over the last years, evidence has suggested that phosphoinositides, which are involved in the regulation of a large variety of cellular processes both in the cytoplasm and in the plasma membrane, are present also within the nucleus. A number of advances has resulted in the discovery that phosphoinositide-specific phospholipase C signalling in the nucleus is involved in cell growth and differentiation. Remarkably, the nuclear inositide metabolism is regulated independently from that present elsewhere in the cell. Even though nuclear inositol lipids hydrolysis generates second messengers such as diacylglycerol and inositol 1,4,5-trisphosphate, it is becoming increasingly clear that in the nucleus polyphosphoinositides may act by themselves to influence pre-mRNA splicing and chromatin structure. Among phosphoinositide-specific phospholipase C, the beta(1) isoform appears to be one of the key players of the nuclear lipid signaling. This review aims at highlighting the most significant and up-dated findings about phosphoinositide-specific phospholipase C beta(1) in the nucleus.


Subject(s)
Cell Nucleus/metabolism , Cell Physiological Phenomena , Lipid Bilayers/metabolism , Nuclear Envelope/metabolism , Phosphatidylinositol Diacylglycerol-Lyase/metabolism , Phosphatidylinositols/metabolism , Signal Transduction/physiology , Animals , Humans , Phosphoinositide Phospholipase C
4.
Mol Pharmacol ; 68(1): 169-78, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15821150

ABSTRACT

15-Deoxy delta(12,14)-prostaglandin J(2) (15d-PGJ(2)), an activator of peroxisome proliferator-activated receptor (PPAR)-gamma and -delta, is a prostanoid metabolite with anti-inflammatory actions. In intrauterine tissues, proinflammatory cytokines and prostaglandins have been identified as playing key roles in the maintenance of pregnancy and the onset of labor. We investigated and compared the early (<3 h) effects of 15d-PGJ(2) with rosiglitazone (PPAR-gamma ligand) and 2-methyl-4-((4-methyl-2-(4-trifluoromethylphenyl)-1,3-thiazol-5-yl)-methylsulfanyl)phenoxy-acetic acid (GW501516) (PPAR-delta ligand) on interleukin (IL)-1beta-induced prostaglandin and cytokine production by amnion-derived WISH cells. We show that 15d-PGJ(2) exerts differential effects depending on concentration. At low concentrations (<0.1 microM), 15d-PGJ(2) inhibited IL-1beta-stimulated prostaglandin E(2) (PGE(2)) but not cytokine (IL-6/IL-8) production or cyclooxygenase-2 (COX-2) expression. This effect was attenuated by a PPAR-gamma inhibitor [2-chloro-5-nitro-N-phenyl-benzamide (GW9662)], by transfection with a dominant-negative PPAR construct, and was reproduced by the PPAR-gamma ligand rosiglitazone. At higher concentrations (1-10 microM), 15d-PGJ(2) inhibited IL-1beta-stimulated PGE(2) and cytokine production and COX-2 expression, and this effect was not blocked by GW9662. Rosiglitazone at high concentrations (1-10 microM) stimulated PGE(2) production in the absence or presence of the dominant-negative PPAR. The PPAR-delta ligand GW501516 also inhibited IL-1beta-stimulated PGE(2) production but only at high concentrations (1 microM). IL-1beta-induced nuclear factor-kappaB (NF-kappaB) DNA binding activity was significantly inhibited by 15d-PGJ(2) (10 microM) and GW501516 (1 microM) but increased with 10 microM rosiglitazone. We conclude that 1) at low concentrations, 15d-PGJ(2) acts through a PPAR-gamma signaling pathway; b) at higher concentrations, its actions are mediated most likely through other pathways such as activation of PPAR-delta and/or inhibition of NF-kappaB; and 3) rosiglitazone exerts PPAR-independent effects at high concentrations (>1 microM).


Subject(s)
Amnion/drug effects , Epithelial Cells/drug effects , NF-kappa B/physiology , PPAR delta/physiology , PPAR gamma/physiology , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/administration & dosage , Amnion/cytology , Amnion/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Epithelial Cells/cytology , Epithelial Cells/physiology , Humans , Nanotechnology/methods
5.
Biochim Biophys Acta ; 1676(2): 127-37, 2004 Jan 20.
Article in English | MEDLINE | ID: mdl-14746907

ABSTRACT

Many studies have been undertaken to investigate the mechanisms of skin differentiation. In particular, growth factors and hormones are believed to play important roles in skin proliferation, differentiation and survival. Insulin-like growth factor-1 (IGF-1) has been identified as a survival factor in many tissues including the skin, but the molecular mechanism of IGF-1 in epidermal differentiation is not completely understood. Neonatal mouse skin is useful for studying changes in gene expression, as the mitotic activity of skin cells changes shortly after birth. Using RNA differential display (DD), a 357-nt message that is specifically expressed in the epidermal keratinocytes of IGF-1-injected newborn mice but not in controls, has been identified. Confirmation of expression of this gene by ribonuclease protection assay (RPA) showed that its mRNA expression in the epidermal keratinocytes is induced by IGF-1. Using RNA ligase-mediated rapid amplification of 5' cDNA ends (RLM-5'-RACE), we have successfully isolated a 3473-bp full-length gene, c98, that has 97% sequence homology to a bcl-2-like gene, bcl-w. The latter has been identified as a proto-oncogene in several murine myeloid cell lines. Amino acid sequence analysis of the c98 showed that it has 97% sequence identity to the bcl-w protein and possesses bcl-2 homology domains (BH) 1, 2 and 3. Immunoblotting data revealed similar increases of c98 protein expression to its mRNA expression in the keratinocytes of IGF-1-injected animals. Weak expression of other bcl-2 family member proteins, bax, bcl-2 and bcl-xL, were also found in the immunoblots. Additionally, IGF-1 was found to be able to protect epidermal keratinocytes from dexamethasone (DEX)-induced apoptosis, based on the findings that after the cells were treated with DEX, DNA laddering was present in the control mice but not in those injected with IGF-1. Further, using a photometric enzyme-linked immunoassay to quantitate keratinocyte death, we found that after addition of DEX, the amounts of cytoplasmic histone-associated DNA fragments were not significantly (P>0.05) different in IGF-1-treated cells compared with untreated control cells during the high mitotic stage of skin epidermis. To assess the role of c98 in these anti-apoptotic processes, we have generated a recombinant plasmid that contains an expression vector and c98 and transfected this plasmid into the keratinocytes from mice without IGF-1-treatment. Expression of the c98 protein was found to completely (P>0.05) block DEX-induced apoptosis after cell transfection. Taken together, our current data demonstrated that IGF-1 plays an anti-apoptotic role in the DEX-induced apoptosis in epidermal keratinocytes and this, at least in part, may be mediated through expression of c98.


Subject(s)
Apoptosis/drug effects , DNA, Complementary/biosynthesis , Genes, bcl-2 , Insulin-Like Growth Factor I/pharmacology , Keratinocytes/drug effects , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/chemistry , Dexamethasone/antagonists & inhibitors , Dexamethasone/pharmacology , Keratinocytes/metabolism , Mice , Molecular Sequence Data , RNA/analysis , RNA/isolation & purification , Sequence Homology, Nucleic Acid , Skin/growth & development , Skin/metabolism
6.
Mol Pharmacol ; 64(6): 1586-90, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14645690

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are a family of nuclear receptors that are involved in lipid metabolism, differentiation, proliferation, cell death, and inflammation. Three subtypes have been identified: PPAR-alpha, -delta, and -gamma. We have previously shown presence of PPAR-gamma mRNA in the amnion, choriodecidua, and placenta, and its level of expression was unchanged with labor. To evaluate whether PPAR-alpha and -delta subtypes are present in intrauterine tissues, placentae were obtained from women at term after spontaneous vaginal delivery (TSL; n = 15) and elective caesarean section before labor (TNL; n = 15). Northern blot analyses were used to evaluate the mRNA for PPARs. Activities of PPARs were assessed using JEG3 choriocarcinoma cells transfected with a PPAR-response element reporter construct (pTK-PPREx3-luc) and treated with PPAR ligands. The PPAR-gamma-specific ligand rosiglitazone induced PPAR response element (PPRE)-mediated activity in a concentration-dependent manner, whereas the PPAR-gamma-specific irreversible inhibitor GW9662 fully inhibited this induction. However, GW9662 only partially inhibited 15-deoxy-Delta12,14-prostaglandin J2 (15d-PGJ2)-induced luciferase activity, suggesting that 15d-PGJ2 may also activate either of the other isoforms. PPAR-alpha and -delta are expressed in the amnion, choriodecidua, and placental villous tissues. In the amnion, although for PPAR-alpha no significant difference in expression was observed with labor, PPAR-delta expression increased significantly (p < 0.001). In the choriodecidua, expression of PPAR-alpha declined with labor (p < 0.01), whereas, as in the amnion, PPAR-delta expression increased (p < 0.05). In the placenta, both PPAR-alpha and -delta expression increased with labor (p < 0.005). The changes observed with labor suggest that regulation of PPAR expression and function may have roles to the mechanisms that maintain pregnancy or initiate labor.


Subject(s)
Amnion/metabolism , Labor, Obstetric/metabolism , Placenta/metabolism , Receptors, Cytoplasmic and Nuclear/biosynthesis , Transcription Factors/biosynthesis , Cell Line, Tumor , Female , Humans , Pregnancy , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics
8.
J Cell Physiol ; 195(3): 446-52, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12704654

ABSTRACT

Phospholipase C beta(1) (PLCbeta(1)) signaling in both cell proliferation and differentiation has been largely investigated, but its role in myoblast differentiation is still unclear. The C2C12 myogenic cell line has been used in this study in order to find out the role of the two subtypes of PLCbeta(1), i.e., a and b in this process. C2C12 myoblast proliferate in response to mitogens and upon mitogen withdrawal differentiates into multinucleated myotubes. We found that differentiation of C2C12 skeletal muscle cells is characterized by a marked increase in the amount of nuclear PLCbeta(1)a and PLCbeta(1)b. Indeed, treatment with insulin induces a dramatic rise of both PLCbeta(1) subtypes expression and activity, as determined by immunochemical and enzymatic assays. Immunofluorescence experiments with anti-PLCbeta(1) specific monoclonal antibody showed a low level of cytoplasmatic and nuclear staining during the initial 12 h of differentiation whilst a massive nuclear staining is appreciable in differentiating cells. The time course of PLCbeta(1) expression versus Troponin T expression clearly indicates that the increase in the amount of PLCbeta(1) takes place 24 h earlier than that of Troponin T. Moreover, the overexpression of the PLCbeta(1)M2b mutant, lacking the nuclear localization signal and entirely located in the cytoplasm, represses the formation of mature multinucleated myotube. Taken together these results suggest that nuclear PLCbeta(1) is a key player in myoblast differentiation, functioning as a positive regulator of this process.


Subject(s)
Cell Nucleus/enzymology , Isoenzymes/metabolism , Muscle, Skeletal/enzymology , Type C Phospholipases/metabolism , Up-Regulation , Animals , Cell Differentiation , Cell Line , Enzyme Activation , Isoenzymes/chemistry , Isoenzymes/genetics , Muscle, Skeletal/cytology , Mutation , Nuclear Localization Signals , Phospholipase C beta , RNA, Messenger/biosynthesis , Rats , Transcription, Genetic , Type C Phospholipases/chemistry , Type C Phospholipases/genetics
9.
Biochim Biophys Acta ; 1589(3): 305-10, 2002 May 08.
Article in English | MEDLINE | ID: mdl-12031797

ABSTRACT

It is well established that phospholipase C (PLC) beta(1) plays a role in the nuclear compartment and is involved in the signalling pathway that controls the switching of the erythroleukemia cells programming from an undifferentiated to a differentiated state. Constitutive overexpression of nuclear PLCbeta(1) has been previously shown to inhibit Friend cells differentiation. For further characterization, we investigated the localization of PLCbeta(1)a and PLCbeta(1)b in Friend cells by fusing their cDNA to enhanced green fluorescent protein (GFP). To investigate the potential target of nuclear PLCbeta(1) in Friend differentiation, we studied the expression of p45/NF-E2 transcription factor, which is an enhancer binding protein for expression of the beta-globin gene and the expression of GATA proteins that are important for the survival and differentiation of erythroid cells. Our data suggest that the overexpression of PLCbeta(1) (both 1a and 1b) only in the nuclear compartment significantly reduces the expression of p45/NF-E2. The effect observed is attributable to the specific action of nuclear PLCbeta(1) signalling given that GATA-1 and GATA-3 are not affected at all. Here we show the existence of a unique target, i.e. the transcription factor p45/NF-E2, whose expression is specifically inhibited by the nuclear signalling evoked by PLCbeta(1) forms.


Subject(s)
DNA-Binding Proteins/metabolism , Isoenzymes/metabolism , Leukemia, Erythroblastic, Acute/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Type C Phospholipases/metabolism , Animals , Cell Differentiation , Cell Division , Dimethyl Sulfoxide , Erythroid-Specific DNA-Binding Factors , Green Fluorescent Proteins , Luminescent Proteins , NF-E2 Transcription Factor, p45 Subunit , Phospholipase C beta , Signal Transduction , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...