Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Cell Biol ; 147(5): 1063-72, 1999 Nov 29.
Article in English | MEDLINE | ID: mdl-10579725

ABSTRACT

Although the interaction of matrix proteins with integrins is known to initiate signaling pathways that are essential for cell survival, a role for tumor suppressors in the regulation of these pathways has not been established. We demonstrate here that p53 can inhibit the survival function of integrins by inducing the caspase-dependent cleavage and inactivation of the serine/threonine kinase AKT/PKB. Specifically, we show that the alpha6beta4 integrin promotes the survival of p53-deficient carcinoma cells by activating AKT/PKB. In contrast, this integrin does not activate AKT/PKB in carcinoma cells that express wild-type p53 and it actually stimulates their apoptosis, in agreement with our previous findings (Bachelder, R.E., A. Marchetti, R. Falcioni, S. Soddu, and A.M. Mercurio. 1999. J. Biol. Chem. 274:20733-20737). Interestingly, we observed reduced levels of AKT/PKB protein after antibody clustering of alpha6beta4 in carcinoma cells that express wild-type p53. In contrast, alpha6beta4 clustering did not reduce the level of AKT/PKB in carcinoma cells that lack functional p53. The involvement of caspase 3 in AKT/PKB regulation was indicated by the ability of Z-DEVD-FMK, a caspase 3 inhibitor, to block the alpha6beta4-associated reduction in AKT/PKB levels in vivo, and by the ability of recombinant caspase 3 to promote the cleavage of AKT/PKB in vitro. In addition, the ability of alpha6beta4 to activate AKT/PKB could be restored in p53 wild-type carcinoma cells by inhibiting caspase 3 activity. These studies demonstrate that the p53 tumor suppressor can inhibit integrin-associated survival signaling pathways.


Subject(s)
Antigens, Surface/physiology , Integrins/physiology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins , Signal Transduction/physiology , Tumor Suppressor Protein p53/physiology , Antigens, Neoplasm/metabolism , Biomarkers, Tumor , Caspase 3 , Caspases/biosynthesis , Cell Survival/physiology , Colorectal Neoplasms , Enzyme Induction , Enzyme Inhibitors/pharmacology , Epitopes/metabolism , Humans , Integrin alpha6beta4 , Proto-Oncogene Proteins c-akt , Tumor Cells, Cultured
2.
J Cell Biol ; 151(6): 1295-304, 2000 Dec 11.
Article in English | MEDLINE | ID: mdl-11121443

ABSTRACT

The p53 oncosuppressor protein regulates cell cycle checkpoints and apoptosis, but increasing evidence also indicates its involvement in differentiation and development. We had previously demonstrated that in the presence of differentiation-promoting stimuli, p53-defective myoblasts exit from the cell cycle but do not differentiate into myocytes and myotubes. To identify the pathways through which p53 contributes to skeletal muscle differentiation, we have analyzed the expression of a series of genes regulated during myogenesis in parental and dominant-negative p53 (dnp53)-expressing C2C12 myoblasts. We found that in dnp53-expressing C2C12 cells, as well as in p53(-/-) primary myoblasts, pRb is hypophosphorylated and proliferation stops. However, these cells do not upregulate pRb and have reduced MyoD activity. The transduction of exogenous TP53 or Rb genes in p53-defective myoblasts rescues MyoD activity and differentiation potential. Additionally, in vivo studies on the Rb promoter demonstrate that p53 regulates the Rb gene expression at transcriptional level through a p53-binding site. Therefore, here we show that p53 regulates myoblast differentiation by means of pRb without affecting its cell cycle-related functions.


Subject(s)
Muscle, Skeletal/cytology , Myogenic Regulatory Factors/metabolism , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Cycle , Cell Differentiation , Mice , Mice, Mutant Strains , Models, Biological , Muscle, Skeletal/metabolism , Promoter Regions, Genetic , RNA Processing, Post-Transcriptional , Retinoblastoma Protein/genetics , Signal Transduction , Stem Cells , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Up-Regulation
3.
J Cell Biol ; 134(1): 193-204, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8698814

ABSTRACT

The involvement of p53 protein in cell differentiation has been recently suggested by some observations made with tumor cells and the correlation found between differentiation and increased levels of p53. However, the effect of p53 on differentiation is in apparent contrast with the normal development of p53-null mice. To test directly whether p53 has a function in cell differentiation, we interfered with the endogenous wt-p53 protein of nontransformed cells of two different murine histotypes: 32D myeloid progenitors, and C2C12 myoblasts. A drastic inhibition of terminal differentiation into granulocytes or myotubes, respectively, was observed upon expression of dominant-negative p53 proteins. This inhibition did not alter the cell cycle withdrawal typical of terminal differentiation, nor p21(WAF1/CIP1) upregulation, indicating that interference with endogenous p53 directly affects cell differentiation, independently of the p53 activity on the cell cycle. We also found that the endogenous wt-p53 protein of C2C12 cells becomes transcriptionally active during myogenesis, and this activity is inhibited by p53 dominant-negative expression. Moreover, we found that p53 DNA-binding and transcriptional activities are both required to induce differentiation in p53-negative K562 cells. Taken together, these data strongly indicate that p53 is a regulator of cell differentiation and it exerts this role, at least in part, through its transcriptional activity.


Subject(s)
Hematopoiesis , Muscle, Skeletal/cytology , Tumor Suppressor Protein p53/physiology , Animals , Base Sequence , Cell Differentiation , Cell Division , Cell Survival , Cells, Cultured , DNA Primers/chemistry , Gene Expression Regulation, Developmental , Genes, p53 , Granulocytes/cytology , Humans , Mice , Molecular Sequence Data , RNA, Messenger/genetics , Transcription, Genetic
4.
Cell Death Differ ; 13(9): 1554-63, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16341121

ABSTRACT

HMGA gene overexpression and rearrangements are frequent in several tumours, but their oncogenic function is still unclear. Here we report of a physical and functional interaction between High Mobility Group A1 (HMGA1) protein and p53 oncosuppressor. We found that HMGA1 binds p53 in vitro and in vivo, and both proteins are present in the same complexes bound to the Bax gene promoter. HMGA1 interferes with the p53-mediated transcription of p53 effectors Bax and p21(waf1) while cooperates with p53 in the transcriptional activation of the p53 inhibitor mdm2. This transcriptional modulation is associated with a reduced p53-dependent apoptosis in cells expressing exogenous HMGA1 and p53, or in cells expressing endogenously the proteins and in which p53 was activated by UV-irradiation. Furthermore, antisense inhibition of HMGA1b expression dramatically increases the UV-induced p53-mediated apoptosis. These data define a new physical and functional interaction between HMGA1 and p53 that modulates transcription of p53 target genes and inhibits apoptosis.


Subject(s)
Apoptosis , HMGA1b Protein/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Line , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Humans , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-mdm2/metabolism , Transcription, Genetic , Transcriptional Activation , Ultraviolet Rays , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
5.
J Clin Invest ; 98(5): 1165-73, 1996 Sep 01.
Article in English | MEDLINE | ID: mdl-8787680

ABSTRACT

Lonidamine, a dichlorinated derivative of indazole-3-carboxylic acid, was shown to play a significant role in reversing or overcoming multidrug resistance. Here, we show that exposure to 50 microg/ml of lonidamine induces apoptosis in adriamycin and nitrosourea-resistant cells (MCF-7 ADR(r) human breast cancer cell line, and LB9 glioblastoma multiform cell line), as demonstrated by sub-G1 peaks in DNA content histograms, condensation of nuclear chromatin, and internucleosomal DNA fragmentation. Moreover, we find that apoptosis is preceded by accumulation of the cells in the G0/G1 phase of the cell cycle. Interestingly, lonidamine fails to activate the apoptotic program in the corresponding sensitive parental cell lines (ADR-sensitive MCF-7 WT, and nitrosourea-sensitive LI cells) even after long exposure times. The evaluation of bcl-2 protein expression suggests that this different effect of lonidamine treatment in drug-resistant and -sensitive cell lines might not simply be due to dissimilar expression levels of bcl-2 protein. To determine whether the lonidamine-induced apoptosis is mediated by p53 protein, we used cells lacking endogenous p53 and overexpressing either wild-type p53 or dominant-negative p53 mutant. We find that apoptosis by lonidamine is independent of the p53 gene.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Carmustine/pharmacology , Doxorubicin/pharmacology , Indazoles/pharmacology , Tumor Suppressor Protein p53/metabolism , Breast Neoplasms/pathology , Cell Line , Drug Resistance , Female , Glioblastoma/pathology , Humans , Treatment Failure , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
6.
Mol Cell Biol ; 16(2): 487-95, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8552075

ABSTRACT

Expression of exogenous wild-type (wt) p53 in different leukemia cell lines can induce growth arrest, apoptotic cell death, or cell differentiation. The hematopoietic cell lines that have been used so far to study wt p53 functions have in common the characteristic of not expressing endogenous p53. However, the mechanisms involved in the transformation of these cells are different, and the cells are at different stages of tumor progression. It can be postulated that each type of neoplastic cell offers a particular environment in which p53 might generate different effects. To test this hypothesis, we introduced individual oncogenes into untransformed, interleukin-3 (IL-3)-dependent myeloid precursor 32D cells to have a single transforming agent at a time. The effects induced by wt p53 overexpression were subsequently evaluated in each oncogene-expressing 32D derivative. We found that in not fully transformed, v-ras-expressing 32D cells, as already shown for the parental 32D cells, overexpression of the wt p53 gene caused no phenotypic changes and no reduction of the proliferative rate as long as the cells were maintained in their normal culture conditions (presence of IL-3 and serum). An accelerated rate of apoptosis was observed after IL-3 withdrawal. In contrast, in transformed, IL-3-independent 32D cells, wt p53 overexpression induced different effects. The v-abl-transformed cells manifested a reduction in growth rate, while the v-src-transformed cells underwent monocytic differentiation. These results show that the phenotype effects of wt p53 action(s) can vary as a function of the cellular environment.


Subject(s)
Bone Marrow/metabolism , Oncogene Proteins/biosynthesis , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Blotting, Northern , Blotting, Western , Bone Marrow/drug effects , Bone Marrow Cells , Cell Differentiation , Cell Division , Cell Survival , Cells, Cultured , Interleukin-3/pharmacology , Mice , Monocytes/physiology , Oncogene Protein p21(ras)/biosynthesis , Oncogene Protein p21(ras)/genetics , Oncogene Protein pp60(v-src)/biosynthesis , Oncogene Protein pp60(v-src)/genetics , Oncogene Proteins/genetics , Oncogene Proteins v-abl/biosynthesis , Oncogene Proteins v-abl/genetics , Phosphorylation , Recombinant Proteins/biosynthesis , Transfection , Tumor Suppressor Protein p53/genetics
7.
Int J Biol Markers ; 22(4): 302-6, 2007.
Article in English | MEDLINE | ID: mdl-18161662

ABSTRACT

Mutations in the TP53 gene are the most common genetic alterations in cancer. Accumulation of mutated protein may induce circulating anti-p53 antibodies (anti-p53Ab) in sera of cancer patients. The aim of our work was to evaluate the presence and prognostic value of anti-p53Ab in gastric cancer patients and to investigate whether their presence is related to p53 overexpression in tumor tissue. Anti-p53Ab were analyzed in sera from 111 patients with gastric carcinoma and from 64 healthy donors by ELISA. p53 expression was also quantified by ELISA in biopsies of 54 gastric cancers and 22 healthy gastric mucosas. Significant anti-p53Ab levels were found in 15.3% of patients, whereas none of the 64 donor sera were positive. High levels of p53 expression were detected only in tumor tissue, in 72.2% of cases. A significant correlation was observed between anti-p53Ab and high levels of mutated p53 in tissue (p<0.05). The survival time of serum-positive patients was significantly longer than that of patients with low/negative serum levels, with a survival rate of 41.2% and 14.9%, respectively, over 48 months (p<0.05). Thus, detection of serum anti-p53Ab in gastric cancer patients can be useful to identify a subset of patients with better prognosis.


Subject(s)
Adenocarcinoma/blood , Adenocarcinoma/diagnosis , Gene Expression Regulation, Neoplastic , Genes, p53 , Stomach Neoplasms/blood , Stomach Neoplasms/diagnosis , Tumor Suppressor Protein p53/chemistry , Adenocarcinoma/genetics , Adult , Aged , Biopsy , Case-Control Studies , Disease Progression , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Prognosis , Stomach Neoplasms/genetics , Tumor Suppressor Protein p53/immunology
8.
Mol Biol Cell ; 15(8): 3751-7, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15181149

ABSTRACT

We recently demonstrated that the p53 oncosuppressor associates to centrosomes in mitosis and this association is disrupted by treatments with microtubule-depolymerizing agents. Here, we show that ATM, an upstream activator of p53 after DNA damage, is essential for p53 centrosomal localization and is required for the activation of the postmitotic checkpoint after spindle disruption. In mitosis, p53 failed to associate with centrosomes in two ATM-deficient, ataxiatelangiectasia-derived cell lines. Wild-type ATM gene transfer reestablished the centrosomal localization of p53 in these cells. Furthermore, wild-type p53 protein, but not the p53-S15A mutant, not phosphorylatable by ATM, localized at centrosomes when expressed in p53-null K562 cells. Finally, Ser15 phosphorylation of endogenous p53 was detected at centrosomes upon treatment with phosphatase inhibitors, suggesting that a p53 dephosphorylation step at centrosome contributes to sustain the cell cycle program in cells with normal mitotic spindles. When dissociated from centrosomes by treatments with spindle inhibitors, p53 remained phosphorylated at Ser15. AT cells, which are unable to phosphorylate p53, did not undergo postmitotic proliferation arrest after nocodazole block and release. These data demonstrate that ATM is required for p53 localization at centrosome and support the existence of a surveillance mechanism for inhibiting DNA reduplication downstream of the spindle assembly checkpoint


Subject(s)
Centrosome/chemistry , Mitosis , Protein Serine-Threonine Kinases/metabolism , Serine/metabolism , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , Cell Line , Centrosome/metabolism , DNA-Binding Proteins , Humans , Mutation/genetics , Nocodazole/pharmacology , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Serine/genetics , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Tubulin/analysis , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Proteins
10.
Oncogene ; 35(2): 228-40, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-25961923

ABSTRACT

The p53 inhibitor, MDM4 (MDMX) is a cytoplasmic protein with p53-activating function under DNA damage conditions. Particularly, MDM4 promotes phosphorylation of p53 at Ser46, a modification that precedes different p53 activities. We investigated the mechanism by which MDM4 promotes this p53 modification and its consequences in untransformed mammary epithelial cells and tissues. In response to severe DNA damage, MDM4 stimulates p53Ser46(P) by binding and stabilizing serine-threonine kinase HIPK2. Under these conditions, the p53-inhibitory complex, MDM4/MDM2, dissociates and this allows MDM4 to promote p53/HIPK2 functional interaction. Comparative proteomic analysis of DNA damage-treated cells versus -untreated cells evidenced a diffuse downregulation of proteins with anti-apoptotic activity, some of which were targets of p53Ser46(P)/HIPK2 repressive activity. Importantly, MDM4 depletion abolishes the downregulation of these proteins indicating the requirement of MDM4 to promote p53-mediated transcriptional repression. Consistently, MDM4-mediated HIPK2/p53 activation precedes HIPK2/p53 nuclear translocation and activity. Noteworthy, repression of these proteins was evident also in mammary glands of mice subjected to γ-irradiation and was significantly enhanced in transgenic mice overexpressing MDM4. This study evidences the flexibility of MDM2/MDM4 heterodimer, which allows the development of a positive activity of cytoplasmic MDM4 towards p53-mediated transcriptional function. Noteworthy, this activity uncovers coordinated repression of molecules with shared anti-apoptotic function which precedes active cell apoptosis and that are frequently overexpressed and/or markers of tumour phenotype in human cancer.


Subject(s)
Apoptosis/physiology , Carrier Proteins/metabolism , DNA Damage/physiology , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Base Sequence , Carrier Proteins/genetics , Cell Cycle Proteins , Cytoplasm/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , HCT116 Cells , Humans , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Serine/metabolism , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
11.
Oncogene ; 35(7): 887-96, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-25961924

ABSTRACT

Human Mena (hMENA), an actin regulatory protein of the ENA/VASP family, cooperates with ErbB receptor family signaling in breast cancer. It is overexpressed in high-risk preneoplastic lesions and in primary breast tumors where it correlates with HER2 overexpression and an activated status of AKT and MAPK. The concomitant overexpression of hMENA and HER2 in breast cancer patients is indicative of a worse prognosis. hMENA is expressed along with alternatively expressed isoforms, hMENA(11a) and hMENAΔv6 with opposite functions. A novel role for the epithelial-associated hMENA(11a) isoform in sustaining HER3 activation and pro-survival pathways in HER2-overexpressing breast cancer cells has been identified by reverse phase protein array and validated in vivo in a series of breast cancer tissues. As HER3 activation is crucial in mechanisms of cell resistance to PI3K inhibitors, we explored whether hMENA(11a) is involved in these resistance mechanisms. The specific hMENA(11a) depletion switched off the HER3-related pathway activated by PI3K inhibitors and impaired the nuclear accumulation of HER3 transcription factor FOXO3a induced by PI3K inhibitors, whereas PI3K inhibitors activated hMENA(11a) phosphorylation and affected its localization. At the functional level, we found that hMENA(11a) sustains cell proliferation and survival in response to PI3K inhibitor treatment, whereas hMENA(11a) silencing increases molecules involved in cancer cell apoptosis. As shown in three-dimensional cultures, hMENA(11a) contributes to resistance to PI3K inhibition because its depletion drastically reduced cell viability upon treatment with PI3K inhibitor BEZ235. Altogether, these results indicate that hMENA(11a) in HER2-overexpressing breast cancer cells sustains HER3/AKT axis activation and contributes to HER3-mediated resistance mechanisms to PI3K inhibitors. Thus, hMENA(11a) expression can be proposed as a marker of HER3 activation and resistance to PI3K inhibition therapies, to select patients who may benefit from these combined targeted treatments. hMENA(11a) activity could represent a new target for antiproliferative therapies in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Microfilament Proteins/metabolism , Receptor, ErbB-3/genetics , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Cell Line, Tumor , Electrophoresis, Gel, Two-Dimensional , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Microfilament Proteins/genetics , Phosphoinositide-3 Kinase Inhibitors , Protein Isoforms , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Receptor, ErbB-2/genetics , Transfection
12.
Oncogene ; 18(42): 5831-5, 1999 Oct 14.
Article in English | MEDLINE | ID: mdl-10523864

ABSTRACT

Experiments previously performed on 32D and C2C12 cell lines indicated that wild type p53 (wtp53) protein has a role in granulocyte and myotube differentiation. Since these are immortal cells, we asked whether the inhibition of differentiation induced by the expression of dominant-negative p53 (dnp53) proteins was dependent on the immortalization-determined microenvironment. Thus, we evaluated the effects produced by interfering with the endogenous p53 gene in murine primary hemopoietic and muscle cells. Expression of dnp53 protein reduced the differentiation of bone marrow cells into granulocytes and macrophages. Moreover, p53 activation was measurable during the differentiation process of primary myoblasts, while interference with this activation led to a consistent slow down of terminal differentiation. Since the impairment of the differentiation was not accompanied by alterations in the cell cycle withdrawal and in the rate of apoptosis which are coupled with these types of differentiation, the data here reported support a specific role for p53 in the differentiation process. However, the difference in the intensity of inhibition between immortal and primary cells, i. e., complete versus slow down, respectively, suggests that the immortalization process might render the cells more sensitive to the loss of wtp53 activity for the differentiation process.


Subject(s)
Hematopoietic Stem Cells/cytology , Muscles/cytology , Tumor Suppressor Protein p53/physiology , Animals , Bone Marrow Cells/cytology , Cell Differentiation/physiology , Cell Division/physiology , Cell Line , Cell Line, Transformed , Cell Survival/physiology , Coculture Techniques , Mice , Stem Cells/cytology , Transcription, Genetic
13.
Oncogene ; 10(4): 731-7, 1995 Feb 16.
Article in English | MEDLINE | ID: mdl-7862450

ABSTRACT

Apoptotic cell death is an active process which regulates the maintenance of the hematopoietic homeostasis. It has been reported that wild-type p53 (wt-p53) protein induces apoptosis in leukemia cells. To assess whether p53 is involved in the apoptotic process of normal hematopoietic cells, we introduced the temperature-sensitive p53Val135 mutant into the murine myeloid precursor cell line 32Dcl3. These are diploid, non-tumorigenic cells whose survival and proliferation are dependent upon growth factor supply (IL-3 and serum). Overexpression of wt-p53 protein does not affect morphology and proliferation of 32D cells as long as they are maintained in the presence of IL-3. However, after IL-3 withdrawal, wt-p53 overexpression significantly accelerates apoptosis. This phenomenon is IL-3 specific since no differences in death rates induced by serum starvation are found between parental cells and p53-transfectants. When the latter experiments are carried out at 37 degrees C with p53 protein in mutant conformation, an extended survival of 32D cells is observed after IL-3 deprivation, but not after serum withdrawal. Taken together, these results show that wt-p53 actively mediates the apoptosis due to the absence of specific growth factors, such as IL-3, suggesting that p53 might be involved in the response of myeloid precursors to environmental cytokines for the maintenance of the hematopoietic homeostasis.


Subject(s)
Apoptosis , Hematopoiesis , Hematopoietic Stem Cells/cytology , Interleukin-3/physiology , Tumor Suppressor Protein p53/physiology , Animals , Apoptosis/drug effects , Cell Cycle , Cell Division , DNA Damage , In Vitro Techniques , Mice , Transfection
14.
Oncogene ; 15(5): 607-11, 1997 Jul 31.
Article in English | MEDLINE | ID: mdl-9247315

ABSTRACT

Expression of exogenous wt-p53 in different tumor cell lines can induce growth arrest, apoptosis, or differentiation. Several experimental works have highlighted the relevance of cellular context in the determination of p53-mediated final outcomes. We recently observed that these diverse wt-p53 effects can also be induced by overexpressing wt-p53 in a single cell type-the 32D myeloid progenitors-transformed with different activated oncogenes. Here we show that 32D cells transformed with two different oncogenes, v-src or c-fms [S301,F969], both belonging to the CSF-1 transduction pathway, respond to exogenous wt-p53 expression with the same final outcome-monocytic differentiation. This result is particularly significant since 32D cells do not spontaneously express the CSF-1 receptor, whereas they undergo granulocytic differentiation upon G-CSF stimulation. These data strongly support the idea that wt-p53 suppressing effects result from interactions between p53 activity and the signaling pathways activated in different transformed cells.


Subject(s)
Genes, Tumor Suppressor , Macrophage Colony-Stimulating Factor/metabolism , Monocytes/cytology , Stem Cells/cytology , Tumor Suppressor Protein p53/metabolism , Cell Differentiation/genetics , Cell Division/drug effects , Cell Division/genetics , Cell Line, Transformed , Cell Survival/drug effects , Cell Survival/genetics , Gene Expression Regulation , Genes, fms , Genes, src , Granulocyte Colony-Stimulating Factor/pharmacology , Interleukin-3/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Stem Cells/drug effects , Stem Cells/physiology , Tumor Suppressor Protein p53/genetics
15.
Oncogene ; 14(6): 729-40, 1997 Feb 13.
Article in English | MEDLINE | ID: mdl-9038381

ABSTRACT

Alterations of the tumor suppressor gene p53 are uncommon in differentiated thyroid neoplasia but are detected at high frequency in anaplastic thyroid carcinoma suggesting that impaired p53 function may contribute to the undifferentiated and highly aggressive phenotype of these tumors. Effects of wild type p53 (wt-p53) re-expression were investigated in a human anaplastic thyroid carcinoma cell line (ARO) expressing a mutated p53. ARO cells were stably transfected with the temperature-sensitive p53 Val135 gene (ts-p53) which exhibits wild type-like activity at 32 degrees C. Exogenous wt-p53 function in ARO-tsp53 clones was assessed by evaluating its transcriptional activity on a CAT reporter vector containing p53 binding sites. At 32 degrees C, a significant reduction in the proliferation rate (approximately or equal to 50%) was observed, with accumulation of cells in the G0/G1 phase of the cell cycle. This effect was accompanied by induction of the expression of the growth inhibitor p21/Waf1 gene. At 32 degrees C, ARO-tsp53 clones also showed a marked impairment of their tumorigenic potential. Furthermore, transfected clones re-acquired the ability to respond to thyrotropin (TSH) stimulation showing an increased expression of thyroid-specific genes (thyroglobulin, thyroperoxidase and TSH receptor). In conclusion, re-expression of wt-p53 activity in ARO cells, inhibits cell proliferation and restores responsiveness to physiological stimuli.


Subject(s)
Carcinoma/metabolism , Carcinoma/pathology , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Tumor Suppressor Protein p53/physiology , Cell Differentiation/physiology , Cell Division/physiology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Gene Expression , Genes, p53 , Humans , Mutation , Phenotype , Temperature , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
16.
Oncogene ; 19(29): 3245-55, 2000 Jul 06.
Article in English | MEDLINE | ID: mdl-10918581

ABSTRACT

32D cells expressing v-Ha-Ras fail to show a transformed phenotype. Since Ras requires an active IGF-1R for transformation of fibroblasts, we asked whether expression of IRS-1 or Shc (two of the major substrates of the IGF-1R) could co-operate with oncogenic Ras in transforming 32D cells. We find that IRS-1, but not Shc, in combination with v-Ha-Ras generates a fully transformed phenotype in 32D cells. 32D cells expressing both IRS-1 and v-Ha-Ras (32D/IRS1/Ras) survive and proliferate in the absence of IL-3, do not undergo granulocytic differentiation in the presence of G-CSF and form tumors in nu/nu and syngeneic mice. In contrast, 32D cells expressing singly IRS-1 or v-Ha-Ras exhibit only a block in differentiation capacity. Over-expression of Shc proteins, by itself, promotes differentiation of 32D cells. Concomitant expression of IRS-1 and v-Ha-Ras synergistically phosphorylates ERK-1 and ERK-2 whereas a MEK inhibitor rapidly induces death of 32D/IRS1/Ras transformed cells. Furthermore, transformed 32D/IRS1/Ras cells display high levels of PI3-K activation and undergo rapid apoptosis when exposed to PI3-K inhibitors. The data indicate that: (1) a fully transformed phenotype in 32D cells is generated when a block in differentiation (v-Ha-Ras) is coupled with another differentiation block (IRS-1); (2) PI3-K and MAPK activity are required for the survival of transformed cells; (3) the signals generated by IRS-1 and oncogenic Ras converge on ERK and PI3-K resulting in high levels of activation.


Subject(s)
Cell Transformation, Neoplastic , MAP Kinase Kinase Kinase 1 , Oncogene Protein p21(ras)/genetics , Phosphoproteins/genetics , Androstadienes/pharmacology , Animals , Cell Differentiation , Cell Survival , Chromones/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Gene Expression , Insulin Receptor Substrate Proteins , Mice , Mice, Nude , Morpholines/pharmacology , Oncogene Protein p21(ras)/biosynthesis , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphoproteins/biosynthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Rabbits , Transfection , Tumor Cells, Cultured , Wortmannin
17.
Cell Death Differ ; 11(6): 596-607, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15150542

ABSTRACT

Stimulation of the Ras/MAPK cascade can either activate p53 and promote replicative senescence and apoptosis, or degrade p53 and promote cell survival. Here we show that p53 can directly counteract the Ras/MAPK signaling by inactivating ERK2/MAPK. This inactivation is due to a caspase cleavage of the ERK2 protein and contributes to p53-mediated growth arrest. We found that in Ras-transformed cells, growth arrest induced by p53, but not p21(Waf1), is associated with a strong reduction in ERK2 activity, phosphorylation, and protein half-life, and with the appearance of caspase activity. Likewise, DNA damage-induced cell cycle arrest correlates with p53-dependent ERK2 downregulation and caspase activation. Furthermore, caspase inhibitors or expression of a caspase-resistant ERK2 mutant interfere with ERK2 cleavage and restore proliferation in the presence of p53 activation, indicating that caspase-mediated ERK2 degradation contributes to p53-induced growth arrest. These findings strongly point to ERK2 as a novel p53 target in growth suppression.


Subject(s)
Caspases/metabolism , Cell Division/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Caspase 3 , Cell Division/drug effects , Cell Line, Transformed , DNA Damage/physiology , Down-Regulation , Doxorubicin/pharmacology , Mice
18.
Clin Cancer Res ; 6(2): 671-80, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10690553

ABSTRACT

The contributions of defective mismatch repair (MMR) and the p53-response to cell killing by N-(2-chloroethyl)-N'-cyclohexyl-N-nitrosourea (CCNU) were evaluated. MMR defects were previously shown to be associated with CCNU sensitivity (G. Aquilina et al., Cancer Res., 58: 135-141, 1998). Unexpectedly, eight MMR-deficient variants of the A2780 human ovarian carcinoma cell line were 3-fold more resistant to CCNU than the MMR-proficient parental cells. The variants were members of a preexisting subpopulation of drug-resistant A2780 cells. In addition to deficient expression of the MMR protein hMLH1, an essential component of the hMutL alpha repair complex, the variants exhibited alterations in the expression of other genes that influence drug sensitivity. Although A2780 cells possess a wild-type p53 gene, all of the clones contained a heterozygous G to T tranversion at codon 172. This change resulted in a Val to Phe substitution and was associated with a constitutive production of high levels of p53, which was inactive as a transcriptional activator of bax and p21. The hMLH1/p53 defective variants displayed a less prominent cell cycle arrest and reduced apoptosis after CCNU treatment. In contrast, MMR-defective A2780 variants, which had a similar hMutL alpha defect but retained a wild-type p53, did exhibit the expected CCNU sensitivity. Expression of a dominant-negative p53val135 increased CCNU resistance of both MMR-proficient and MMR-deficient A2780 cells. Thus, defective MMR and p53 influence CCNU sensitivity in opposite directions. Their effects are independent, and sensitization by defective MMR does not require a functional p53 response.


Subject(s)
Apoptosis/drug effects , Base Pair Mismatch , Cell Cycle/drug effects , Genes, p53 , Lomustine/toxicity , Proto-Oncogene Proteins c-bcl-2 , Tumor Suppressor Protein p53/genetics , Adaptor Proteins, Signal Transducing , Amino Acid Substitution , Carrier Proteins , Cell Survival/drug effects , Codon , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Female , Humans , In Situ Nick-End Labeling , Methylnitrosourea/toxicity , MutL Protein Homolog 1 , Neoplasm Proteins/genetics , Nuclear Proteins , Ovarian Neoplasms , Proto-Oncogene Proteins/genetics , Recombinant Proteins/metabolism , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein
19.
Oncogene ; 34(19): 2493-504, 2015 May 07.
Article in English | MEDLINE | ID: mdl-24998848

ABSTRACT

The TP53 tumor-suppressor gene is frequently mutated in human cancer. Missense mutations can add novel functions (gain-of-function, GOF) that promote tumor malignancy. Here we report that mutant (mut) p53 promotes tumor malignancy by suppressing the expression of a natural occurring anti-inflammatory cytokine, the secreted interleukin-1 receptor antagonist (sIL-1Ra, IL1RN). We show that mutp53 but not wild-type (wt) p53 suppresses the sIL-1Ra production in conditioned media of cancer cells. Moreover, mutp53, but not wtp53, binds physically the sIL-1Ra promoter and the protein-protein interaction with the transcriptional co-repressor MAFF (v-MAF musculoaponeurotic fibrosarcoma oncogene family, protein F) is required for mutp53-induced sIL-1Ra suppression. Remarkably, when exposed to IL-1 beta (IL-1ß) inflammatory stimuli, mutp53 sustains a ready-to-be-activated in vitro and in vivo cancer cells' response through the sIL-1Ra repression. Taken together, these results identify sIL-1Ra as a novel mutp53 target gene, whose suppression might be required to generate a chronic pro-inflammatory tumor microenvironment through which mutp53 promotes tumor malignancy.


Subject(s)
DNA-Binding Proteins/genetics , Inflammation/genetics , Interleukin 1 Receptor Antagonist Protein/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Cell Line, Tumor , HT29 Cells , Hep G2 Cells , Humans , Inflammation/immunology , Interleukin 1 Receptor Antagonist Protein/biosynthesis , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin-1beta/pharmacology , MCF-7 Cells , MafF Transcription Factor/metabolism , Mutation , Neoplasms/genetics , Neoplasms/mortality , Nuclear Proteins/metabolism , Prognosis , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , RNA, Small Interfering , Tumor Microenvironment/immunology
20.
FEBS Lett ; 490(3): 163-70, 2001 Feb 16.
Article in English | MEDLINE | ID: mdl-11223031

ABSTRACT

Most genes are members of a family. It is generally believed that a gene family derives from an ancestral gene by duplication and divergence. The tumor suppressor p53 was a striking exception to this established rule. However, two new p53 homologs, p63 and p73, have recently been described [1-6]. At the sequence level, p63 and p73 are more similar to each other than each is to p53, suggesting the possibility that the ancestral gene is a gene resembling p63/p73, while p53 is phylogenetically younger [1,2].The complexity of the family has also been enriched by the alternatively spliced forms of p63 and p73, which give rise to a complex network of proteins involved in the control of cell proliferation, apoptosis and development [1,2,4,7-9]. In this review we will mainly focus on similarities and differences as well as relationships among p63, p73 and p53.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins/metabolism , Evolution, Molecular , Membrane Proteins , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Trans-Activators , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , DNA Damage , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , E2F Transcription Factors , Gene Deletion , Genes, Tumor Suppressor , Humans , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Oncogene Proteins/metabolism , Phosphoproteins/chemistry , Phosphoproteins/genetics , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/metabolism , Tumor Protein p73 , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL