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










Publication year range
2.
Oncogene ; 36(32): 4573-4584, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28368399

ABSTRACT

Recent evidences suggest that stearoyl-CoA-desaturase 1 (SCD1), the enzyme involved in monounsaturated fatty acids synthesis, has a role in several cancers. We previously demonstrated that SCD1 is important in lung cancer stem cells survival and propagation. In this article, we first show, using primary cell cultures from human lung adenocarcinoma, that the effectors of the Hippo pathway, Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ), are required for the generation of lung cancer three-dimensional cultures and that SCD1 knock down and pharmacological inhibition both decrease expression, nuclear localization and transcriptional activity of YAP and TAZ. Regulation of YAP/TAZ by SCD1 is at least in part dependent upon ß-catenin pathway activity, as YAP/TAZ downregulation induced by SCD1 blockade can be rescued by the addition of exogenous wnt3a ligand. In addition, SCD1 activation of nuclear YAP/TAZ requires inactivation of the ß-catenin destruction complex. In line with the in vitro findings, immunohistochemistry analysis of lung adenocarcinoma samples showed that expression levels of SCD1 co-vary with those of ß-catenin and YAP/TAZ. Mining available gene expression data sets allowed to observe that high co-expression levels of SCD1, ß-catenin, YAP/TAZ and downstream targets have a strong negative prognostic value in lung adenocarcinoma. Finally, bioinformatics analyses directed to identify which gene combinations had synergistic effects on clinical outcome in lung cancer showed that poor survival is associated with high co-expression of SCD1, ß-catenin and the YAP/TAZ downstream target birc5. In summary, our data demonstrate for the first time the involvement of SCD1 in the regulation of the Hippo pathway in lung cancer, and point to fatty acids metabolism as a key regulator of lung cancer stem cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenocarcinoma/metabolism , Cell Nucleus/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Phosphoproteins/metabolism , Stearoyl-CoA Desaturase/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Axin Signaling Complex/metabolism , Down-Regulation , Fatty Acids/metabolism , Female , HEK293 Cells , Hippo Signaling Pathway , Humans , Immunohistochemistry , Inhibitor of Apoptosis Proteins/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Neoplasm Proteins/metabolism , Primary Cell Culture , Prognosis , Protein Serine-Threonine Kinases/metabolism , Protein Stability , RNA, Messenger/metabolism , Stearoyl-CoA Desaturase/antagonists & inhibitors , Stearoyl-CoA Desaturase/genetics , Survivin , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Wnt3A Protein/metabolism , YAP-Signaling Proteins
3.
Oncogene ; 35(29): 3760-70, 2016 07 21.
Article in English | MEDLINE | ID: mdl-26996669

ABSTRACT

Downregulation of microRNAs (miRNAs) is commonly observed in cancers and promotes tumorigenesis suggesting that miRNAs may function as tumor suppressors. However, the mechanism through which miRNAs are regulated in cancer, and the connection between oncogenes and miRNA biogenesis remain poorly understood. The TP53 tumor-suppressor gene is mutated in half of human cancers resulting in an oncogene with gain-of-function activities. Here we demonstrate that mutant p53 (mutp53) oncoproteins modulate the biogenesis of a subset of miRNAs in cancer cells inhibiting their post-transcriptional maturation. Interestingly, among these miRNAs several are also downregulated in human tumors. By confocal, co-immunoprecipitation and RNA-chromatin immunoprecipitation experiments, we show that endogenous mutp53 binds and sequesters RNA helicases p72/82 from the microprocessor complex, interfering with Drosha-pri-miRNAs association. In agreement with this, the overexpression of p72 leads to an increase of mature miRNAs levels. Moreover, functional experiments demonstrate the oncosuppressive role of mutp53-dependent miRNAs (miR-517a, -519a, -218, -105). Our study highlights a previously undescribed mechanism by which mutp53 interferes with Drosha-p72/82 association leading, at least in part, to miRNA deregulation observed in cancer.


Subject(s)
MicroRNAs/genetics , Mutation , RNA Processing, Post-Transcriptional , Tumor Suppressor Protein p53/genetics , Apoptosis/genetics , Blotting, Western , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/genetics , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , Membrane Potential, Mitochondrial/genetics , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Tumor Suppressor Protein p53/metabolism
4.
Cell Death Differ ; 21(2): 321-32, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24270405

ABSTRACT

The Groucho/transducin-like Enhancer of split 1 (Gro/TLE1):Hes1 transcriptional repression complex acts in cerebral cortical neural progenitor cells to inhibit neuronal differentiation. The molecular mechanisms that regulate the anti-neurogenic function of the Gro/TLE1:Hes1 complex during cortical neurogenesis remain to be defined. Here we show that prolyl isomerase Pin1 (peptidyl-prolyl cis-trans isomerase NIMA-interacting 1) and homeodomain-interacting protein kinase 2 (HIPK2) are expressed in cortical neural progenitor cells and form a complex that interacts with the Gro/TLE1:Hes1 complex. This association depends on the enzymatic activities of both HIPK2 and Pin1, as well as on the association of Gro/TLE1 with Hes1, but is independent of the previously described Hes1-activated phosphorylation of Gro/TLE1. Interaction with the Pin1:HIPK2 complex results in Gro/TLE1 hyperphosphorylation and weakens both the transcriptional repression activity and the anti-neurogenic function of the Gro/TLE1:Hes1 complex. These results provide evidence that HIPK2 and Pin1 work together to promote cortical neurogenesis, at least in part, by suppressing Gro/TLE1:Hes1-mediated inhibition of neuronal differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carrier Proteins/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Homeodomain Proteins/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Peptidylprolyl Isomerase/metabolism , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/metabolism , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cells, Cultured , HEK293 Cells , Humans , Mice , NIMA-Interacting Peptidylprolyl Isomerase , Naphthoquinones/pharmacology , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Peptidylprolyl Isomerase/antagonists & inhibitors , Transcription Factor HES-1 , Tretinoin/pharmacology
5.
Cell Death Differ ; 20(2): 198-208, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22935610

ABSTRACT

In response to intense stress, the tumor protein p53 (p53) tumor suppressor rapidly mounts a direct mitochondrial death program that precedes transcription-mediated apoptosis. By eliminating severely damaged cells, this pathway contributes to tumor suppression as well as to cancer cell killing induced by both genotoxic drugs and non-genotoxic p53-reactivating molecules. Here we have explored the role had in this pathway by the prolyl-isomerase Pin1 (peptidylprolyl cis/trans isomerase, NIMA-interacting 1), a crucial transducer of p53's phosphorylation into conformational changes unleashing its pro-apoptotic activity. We show that Pin1 promotes stress-induced localization of p53 to mitochondria both in vitro and in vivo. In particular, we demonstrate that upon stress-induced phosphorylation of p53 on Ser46 by homeodomain interacting protein kinase 2, Pin1 stimulates its mitochondrial trafficking signal, that is, monoubiquitination. This pathway is induced also by the p53-activating molecule RITA, and we demonstrate the strong requirement of Pin1 for the induction of mitochondrial apoptosis by this compound. These findings have significant implications for treatment of p53-expressing tumors and for prospective use of p53-activating compounds in clinics.


Subject(s)
Mitochondria/metabolism , Peptidylprolyl Isomerase/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Furans/pharmacology , HCT116 Cells , Humans , Mice , Peptidylprolyl Isomerase/antagonists & inhibitors , Peptidylprolyl Isomerase/genetics , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
6.
Cell Death Differ ; 19(3): 488-500, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21921940

ABSTRACT

TRAF and TNF receptor-associated protein (TTRAP) is a multifunctional protein that can act in the nucleus as a 5'-tyrosyl DNA phosphodiesterase and in the cytoplasm as a regulator of cell signaling. In this paper we show that in response to proteasome inhibition TTRAP accumulates in nucleolar cavities in a promyelocytic leukemia protein-dependent manner. In the nucleolus, TTRAP contributes to control levels of ribosomal RNA precursor and processing intermediates, and this phenotype is independent from its 5'-tyrosyl DNA phosphodiesterase activity. Our findings suggest a previously unidentified function for TTRAP and nucleolar cavities in ribosome biogenesis under stress.


Subject(s)
Cell Nucleolus/metabolism , Nuclear Proteins/metabolism , Proteasome Inhibitors , RNA Processing, Post-Transcriptional/physiology , RNA, Ribosomal/metabolism , Ribosomes/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Cell Line, Tumor , Cell Nucleolus/genetics , DNA-Binding Proteins , HEK293 Cells , Humans , Nuclear Proteins/genetics , Phosphoric Diester Hydrolases , Promyelocytic Leukemia Protein , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , RNA, Ribosomal/genetics , Ribosomes/genetics , Stress, Physiological/physiology , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
7.
Cell Death Differ ; 17(6): 901-11, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20379196

ABSTRACT

The tumor suppressor p53 is a central hub in a molecular network controlling cell proliferation and death in response to potentially oncogenic conditions, and a wide array of covalent modifications and protein interactions modulate the nuclear and cytoplasmic activities of p53. The p53 relatives, p73 and p63, are entangled in the same regulatory network, being subject at least in part to the same modifications and interactions that convey signals on p53, and actively contributing to the resulting cellular output. The emerging picture is that of an interconnected pathway, in which all p53-family proteins are involved in the response to oncogenic stress and physiological inputs. Therefore, common and specific interactors of p53-family proteins can have a wide effect on function and dysfunction of this pathway. Many years of research have uncovered an impressive number of p53-interacting proteins, but much less is known about protein interactions of p63 and p73. Yet, many interactors may be shared by multiple p53-family proteins, with similar or different effects. In this study we review shared interactors of p53-family proteins with the aim to encourage research into this field; this knowledge promises to unveil regulatory elements that could be targeted by a new generation of molecules, and allow more efficient use of currently available drugs for cancer treatment.


Subject(s)
Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Humans , Mice , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Protein Processing, Post-Translational , Transcription Factors/physiology , Tumor Protein p73 , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/physiology
8.
Cell Death Differ ; 16(3): 428-38, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19023331

ABSTRACT

Mutations in PARK7 DJ-1 have been associated with autosomal-recessive early-onset Parkinson's disease (PD). This gene encodes for an atypical peroxiredoxin-like peroxidase that may act as a regulator of transcription and a redox-dependent chaperone. Although large gene deletions have been associated with a loss-of-function phenotype, the pathogenic mechanism of several missense mutations is less clear. By performing a yeast two-hybrid screening from a human fetal brain library, we identified TRAF and TNF receptor-associated protein (TTRAP), an ubiquitin-binding domain-containing protein, as a novel DJ-1 interactor, which was able to bind the PD-associated mutations M26I and L166P more strongly than wild type. TTRAP protected neuroblastoma cells from apoptosis induced by proteasome impairment. In these conditions, endogenous TTRAP relocalized to a detergent-insoluble fraction and formed cytoplasmic aggresome-like structures. Interestingly, both DJ-1 mutants blocked the TTRAP protective activity unmasking a c-jun N-terminal kinase (JNK)- and p38-MAPK (mitogen-activated protein kinase)-mediated apoptosis. These results suggest an active role of DJ-1 missense mutants in the control of cell death and position TTRAP as a new player in the arena of neurodegeneration.


Subject(s)
Apoptosis/physiology , Inclusion Bodies/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mutation, Missense , Nuclear Proteins/metabolism , Oncogene Proteins/genetics , Parkinson Disease , Transcription Factors/metabolism , Antineoplastic Agents/metabolism , Brain Neoplasms , Cell Line , DNA-Binding Proteins , Dopamine/metabolism , Enzyme Activation , Humans , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Leupeptins/metabolism , Neuroblastoma , Nuclear Proteins/genetics , Oncogene Proteins/metabolism , Oxidative Stress , Parkinson Disease/genetics , Parkinson Disease/metabolism , Phosphoric Diester Hydrolases , Protein Binding , Protein Deglycase DJ-1 , Substantia Nigra/cytology , Substantia Nigra/metabolism , Transcription Factors/genetics , Two-Hybrid System Techniques , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Cell Death Differ ; 13(6): 984-93, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16575404

ABSTRACT

Four sets of p53-binding proteins are discussed in this review. These are the E2F family, the ASPP family, Y-box-binding protein YB1, and the prolyl isomerase Pin1. Each appears to play a role in the decision by p53 to induce an arrest of cell proliferation or apoptosis and they may also be independent markers of cancer. Their activities appear to be linked with the cell cycle and they may also interact with each other. In this review, the properties of each protein class are discussed as well as how they affect p53 functions. A model is proposed as to how their activities might be coordinated.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , E2F Transcription Factors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Peptidylprolyl Isomerase/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Humans , NIMA-Interacting Peptidylprolyl Isomerase , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Repressor Proteins , Transcription, Genetic , Y-Box-Binding Protein 1
10.
EMBO J ; 20(11): 2702-14, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11387205

ABSTRACT

Gas2 is a caspase-3 substrate that plays a role in regulating microfilament and cell shape changes during apoptosis. Here we provide evidence that overexpression of Gas2 efficiently increases cell susceptibility to apoptosis following UV irradiation, etoposide and methyl methanesulfonate treatments, and that these effects are dependent on increased p53 stability and transcription activity. To investigate possible pathways linking Gas2 to p53, a yeast two-hybrid screen swas performed, indicating m-calpain as a strong Gas2- interacting protein. Moreover, we demonstrate that Gas2 physically interacts with m-calpain in vivo and that recombinant Gas2 inhibits calpain-dependent processing of p53. Importantly, the Gas2 dominant-negative form (Gas2171-314) that binds calpain but is unable to inhibit its activity abrogates Gas2's ability to stabilize p53, to enhance p53 transcriptional activity and to induce p53-dependent apoptosis. Finally, we show that Gas2 is able to regulate the levels of p53 independently of Mdm2 status, suggesting that, like calpastatin, it may enhance p53 stability by inhibiting calpain activity.


Subject(s)
Apoptosis/physiology , Calpain/metabolism , Microfilament Proteins/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Calpain/genetics , Cell Line , Cell Survival , Etoposide/pharmacology , Genes, Reporter , Humans , Luciferases/genetics , Methyl Methanesulfonate/pharmacology , Mice , Mice, Inbred BALB C , Osteosarcoma , Recombinant Proteins/metabolism , Saccharomyces cerevisiae , Sequence Deletion , Transfection , Tumor Cells, Cultured , Ultraviolet Rays
11.
EMBO J ; 19(22): 6185-95, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11080164

ABSTRACT

Covalent modification of the promyelocytic leukaemia protein (PML) by SUMO-1 is a prerequisite for the assembly of nuclear bodies (NBs), subnuclear structures disrupted in various human diseases and linked to transcriptional and growth control. Here we demonstrate that p53 is recruited into NBs by a specific PML isoform (PML3) or by coexpression of SUMO-1 and hUbc9. NB targeting depends on the direct association of p53, through its core domain, with a C-terminal region of PML3. The relocalization of p53 into NBs enhances p53 transactivation in a promoter-specific manner and affects cell survival. Our results indicate the existence of a cross-talk between PML- and p53-dependent growth suppression pathways, implying an important role for NBs and their resident proteins as modulators of p53 functions.


Subject(s)
Neoplasm Proteins/metabolism , Nuclear Proteins , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Conjugating Enzymes , Biological Transport, Active , Cell Nucleus/metabolism , Cell Survival , Humans , Ligases/genetics , Ligases/metabolism , Neoplasm Proteins/genetics , Promyelocytic Leukemia Protein , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , SUMO-1 Protein , Transcription Factors/genetics , Transcription, Genetic , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins , Ubiquitins/genetics , Ubiquitins/metabolism
12.
FEBS Lett ; 481(2): 152-8, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10996315

ABSTRACT

Growth arrest specific (gas) 1 gene product is expressed in non-transformed fibroblasts in response to stimuli driving cells into Go phase. Gas1 has been demonstrated to inhibit cell proliferation when over-expressed in proliferating fibroblasts. This activity depends on a function of the p53 protein independent of its transactivating ability. To better define the pathway leading from Gas1, which is located on the plasma membrane, to p53, we have undertaken a detailed characterization of its topology. We demonstrate that the protein undergoes cotranslational modifications in the endoplasmic reticulum, consisting of signal peptide cleavage, N-linked glycosylation and glycosyl-phosphatidylinositol anchor addition. Immunoelectron microscopy shows that, in its mature form, Gas1 is randomly distributed over the outer leaflet of the plasma membrane and that upon antibody-induced clustering it relocalizes to caveolae.


Subject(s)
Cell Membrane/metabolism , Glycosylphosphatidylinositols/metabolism , Membrane Glycoproteins/metabolism , Saccharomyces cerevisiae Proteins , 3T3 Cells , Animals , COS Cells , Cell Cycle Proteins , Cell Division , Consensus Sequence/physiology , Endoplasmic Reticulum/metabolism , GPI-Linked Proteins , Glutaral , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/ultrastructure , Membrane Proteins , Mice , Microscopy, Immunoelectron , Palmitic Acid/metabolism , Phosphatidylinositol Diacylglycerol-Lyase , Precipitin Tests , Protein Binding , Protein Sorting Signals/physiology , Tissue Fixation , Transfection , Type C Phospholipases/metabolism
13.
Oncogene ; 19(2): 188-99, 2000 Jan 13.
Article in English | MEDLINE | ID: mdl-10644996

ABSTRACT

Control of cell growth and division by the p53 tumor suppressor protein requires its abilities to transactivate and repress specific target genes and to associate in complex with other proteins. Here we demonstrate that p53 binds to the E1A-regulated transcription factor p120E4F, a transcriptional repressor of the adenovirus E4 promoter. The interaction involves carboxy-terminal half of p120E4F and sequences located at the end of the sequence-specific DNA-binding domain of p53. Ectopic expression of p120E4F leads to a block of cell proliferation in several human and murine cell lines and this effect requires the association with wild-type (wt) p53. Although p120E4F can also bind to mutant p53, the growth suppression induced by overexpression of the protein is severely reduced in a cell line that contains mutant p53. These data suggest that p120E4F may represent an important element within the complex network of p53 checkpoint functions.


Subject(s)
Adenovirus E4 Proteins/physiology , Growth Inhibitors/physiology , Tumor Suppressor Protein p53/physiology , 3T3 Cells , Adenovirus E4 Proteins/biosynthesis , Adenovirus E4 Proteins/genetics , Adenovirus E4 Proteins/isolation & purification , Amino Acids/physiology , Animals , Growth Inhibitors/genetics , Humans , Mice , Mice, Inbred BALB C , Peptide Fragments/physiology , Protein Binding/genetics , Transcriptional Activation , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Zinc Fingers/genetics , Zinc Fingers/physiology
14.
EMBO J ; 18(22): 6462-71, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10562558

ABSTRACT

The growth-suppressive properties of p53 are controlled by posttranslational modifications and by regulation of its turnover rate. Here we show that p53 can be modified in vitro and in vivo by conjugation to the small ubiquitin-like protein SUMO-1. A lysine residue at amino acid position 386 of p53 is required for this previously undescribed modification, strongly suggesting that this lysine residue serves as the major attachment site for SUMO-1. Unlike ubiquitin, attachment of SUMO-1 does not appear to target proteins for rapid degradation but rather, has been proposed to change the ability of the modified protein to interact with other cellular proteins. Accordingly, we provide evidence that conjugation of SUMO-1 to wild-type p53 results in an increased transactivation ability of p53. We suggest that posttranslational modification of p53 by SUMO-1 conjugation provides a novel mechanism to regulate p53 activity.


Subject(s)
Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism , Ubiquitins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Cell Line , Humans , Kinetics , Lysine , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , SUMO-1 Protein , Transfection , Tumor Cells, Cultured , Ubiquitins/chemistry
16.
Proc Natl Acad Sci U S A ; 94(9): 4675-80, 1997 Apr 29.
Article in English | MEDLINE | ID: mdl-9114050

ABSTRACT

The involvement of p53 in regulating diverse cellular processes dictates that it must respond to multiple signaling mechanisms, thus coordinating the response to various "stress conditions." Genotoxic stress has served as a paradigm to dissect the transactivation-dependent branch of the pathway by which p53 can induce growth arrest. Alternate mechanisms have been invoked to explain transactivation-independent effects of p53, especially in the context of apoptosis. We have identified a p53-dependent pathway initiated by the gas1 product, a plasma membrane protein highly expressed during G0, which activates a transactivation-independent p53 growth arrest function. Through a detailed deletional analysis and site-specific mutagenesis of p53 we show that the Gas1-dependent signal transduction relies on a proline-rich region (amino acids 63-85) of murine p53. In vivo competition experiments using combinations of such mutants implicate this functional domain of p53 as a docking site in the transmission of antiproliferative signals.


Subject(s)
Cell Cycle/physiology , Membrane Proteins/metabolism , Proline , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Cycle Proteins , DNA Mutational Analysis , GPI-Linked Proteins , Mice , Mice, Inbred BALB C , Models, Biological , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Signal Transduction , Transcriptional Activation , Tumor Suppressor Protein p53/genetics
17.
Adv Clin Path ; 1(2): 123-136, 1997 Apr.
Article in English | MEDLINE | ID: mdl-10352476

ABSTRACT

In eukariotic cells each phase of the cell cycle is controlled by the sequential activation of various cyclin-dependent kinases (Cdks). These kinases, which are dimers of a catalytic (cdc) and a regulatory (cyclin) subunit, are known to phosphorylate various substrates whose activity is crucial for cell cycle progression. As key regulators of the cell cycle cdks must be under control by both extracellular and intracellular signals. There are several distinct mechanisms for controlling the activity of the different cdks: regulated synthesis and destruction of the regulatory subunit (cyclin), regulated activation and destruction of inhibitory subunits (cki) and postranslational modifications of the catalytic subunit by specific kinases and phosphatases. In this review I will discuss the elements controlling the mammalian cell cycle and their relationship with the genesis of cancer.

18.
Oncogene ; 12(1): 177-85, 1996 Jan 04.
Article in English | MEDLINE | ID: mdl-8552389

ABSTRACT

Overexpression of the wild type p53 gene in normal and transformed cells induces G1 arrest of cellular proliferation. In cell lines carrying the valine 135 temperature-sensitive p53 mutant gene, restoration of wild type p53 protein conformation at the permissive temperature causes an increase in the levels of cyclin D1, as well as the cyclin/cdk inhibitor p21/waf1. Accumulation of cyclin D1 is the result both of (post)transcriptional and post-translational regulatory mechanisms. Ablation of cyclin D1 induction by antisense cDNA microinjection significantly delays the onset of growth arrest, indicating that increased cyclin D1 levels likely contribute to wild type p53 G1 arrest. Whereas antisense ablation of either cyclin D1 or p21/waf1 can delay the onset of p53-induced growth arrest, ablation of neither is able to overcome a pre-existing p53-induced G1 block. In summary, the accumulated evidence indicate that induction of both cyclin D1 and p21/waf1 are involved in establishing the p53-mediated growth arrest in murine cell lines expressing temperature sensitive p53 protein.


Subject(s)
Cyclins/physiology , Oncogene Proteins/physiology , Tumor Suppressor Protein p53/physiology , Amino Acid Sequence , Cell Division , Cyclin D1 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , G1 Phase , Humans , Molecular Sequence Data , Oncogene Proteins/genetics , RNA, Messenger/analysis
19.
Mol Cell Biol ; 15(12): 7152-60, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8524283

ABSTRACT

In normal cells, induction of quiescence is accompanied by the increased expression of growth arrest-specific genes (gas). One of them, gas1, is regulated at the transcriptional level and codes for a membrane-associated protein (Gas1) which is down regulated during the G0-to-S phase transition in serum-stimulated cells. Gas1 is not expressed in growing or transformed cells, and when overexpressed in normal fibroblasts, it blocks the G0-to-S phase transition. Moreover, Gas1 blocks cell proliferation in several transformed cells with the exception of simian virus 40- or adenovirus-transformed cell lines. In this paper, we demonstrate that overexpression of Gas1 blocks cell proliferation in a p53-dependent manner and that the N-terminal domain-dependent transactivating function of p53 is dispensable for Gas1-induced growth arrest. These data therefore indicate that the other intrinsic transactivation-independent functions of p53, possibly related to regulation of apoptosis, should be involved in mediating Gas1-induced growth arrest.


Subject(s)
Cell Cycle , Cell Division/physiology , Membrane Proteins/physiology , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Antibodies , Base Sequence , Blotting, Western , Cell Cycle Proteins , Cell Line, Transformed , Chloramphenicol O-Acetyltransferase/analysis , Chloramphenicol O-Acetyltransferase/biosynthesis , DNA Primers , GPI-Linked Proteins , Gene Expression Regulation , Membrane Proteins/biosynthesis , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/immunology , Polymerase Chain Reaction , Recombinant Proteins/analysis , Recombinant Proteins/biosynthesis , Resting Phase, Cell Cycle , S Phase , Transcription, Genetic , Transfection
20.
Science ; 269(5224): 682-5, 1995 Aug 04.
Article in English | MEDLINE | ID: mdl-7624798

ABSTRACT

The p27 mammalian cell cycle protein is an inhibitor of cyclin-dependent kinases. Both in vivo and in vitro, p27 was found to be degraded by the ubiquitin-proteasome pathway. The human ubiquitin-conjugating enzymes Ubc2 and Ubc3 were specifically involved in the ubiquitination of p27. Compared with proliferating cells, quiescent cells exhibited a smaller amount of p27 ubiquitinating activity, which accounted for the marked increase of p27 half-life measured in these cells. Thus, the abundance of p27 in cells is regulated by degradation. The specific proteolysis of p27 may represent a mechanism for regulating the activity of cyclin-dependent kinases.


Subject(s)
Cell Cycle Proteins , Cyclin-Dependent Kinases/antagonists & inhibitors , Cysteine Endopeptidases/metabolism , Microtubule-Associated Proteins/metabolism , Multienzyme Complexes/metabolism , Tumor Suppressor Proteins , Ubiquitin-Protein Ligase Complexes , Ubiquitins/metabolism , Adenosine Triphosphate/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , Cell Line , Cyclin-Dependent Kinase Inhibitor p27 , Electroporation , Enzyme Inhibitors/metabolism , Humans , Kinetics , Leupeptins/pharmacology , Ligases/metabolism , Mice , Proteasome Endopeptidase Complex , Rabbits , Recombinant Proteins/metabolism , Succinates/pharmacology , Tumor Cells, Cultured , Ubiquitin-Conjugating Enzymes , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL
...