Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
2.
Mol Biol Cell ; 23(8): 1467-74, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22379106

ABSTRACT

In vivo imaging involving the use of genetically engineered animals is an innovative powerful tool for the noninvasive assessment of the molecular and cellular events that are often targets of therapy. On the basis of the knowledge that the activity of the nuclear factor-Y (NF-Y) transcription factor is restricted in vitro to proliferating cells, we have generated a transgenic reporter mouse, called MITO-Luc (for mitosis-luciferase), in which an NF-Y-dependent promoter controls luciferase expression. In these mice, bioluminescence imaging of NF-Y activity visualizes areas of physiological cell proliferation and regeneration during response to injury. Using this tool, we highlight for the first time a role of NF-Y activity on hepatocyte proliferation during liver regeneration. MITO-Luc reporter mice should facilitate investigations into the involvement of genes in cell proliferation and provide a useful model for studying aberrant proliferation in disease pathogenesis. They should be also useful in the development of new anti/proproliferative drugs and assessment of their efficacy and side effects on nontarget tissues.


Subject(s)
CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/metabolism , Cell Proliferation , Liver Regeneration , Molecular Imaging , Transcription, Genetic , Animals , Cell Cycle/genetics , Cell Line , Cyclin B2/genetics , DNA-Binding Proteins/genetics , Genes, Reporter , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Transgenic , Promoter Regions, Genetic
3.
J Thorac Oncol ; 6(3): 583-91, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21289517

ABSTRACT

PURPOSE: The aim of this study was to investigate the expression and biological activity of aromatase (CYP19A1) in malignant mesothelioma (MM). EXPERIMENTAL DESIGN: We found CYP19A1 in five human MM cell lines using reverse transcription polymerase chain reaction and Western immunoblots and in a group of samples from patients with MM by immunohistochemistry. Aromatization activity was determined in MM cells by enzyme-linked immunosorbent assay as a measure of estradiol (E2) product, in basal condition and after addition of cytokine, prostaglandin-E2, and epidermal growth factor to MM cells. Treatment of MM cells with exemestane, a CYP19A1 inhibitor, was assessed by cell proliferation kit, cell cycle analysis, and Western blot for caspase, poly(ADP-ribose)polymerase, Bcl-xL, and v-akt murin thymoma viral oncogene homolog (Akt). RESULTS: Biological activity of CYP19A1, already present in basal condition, was increased in MPP89 and Ist-Mes1 cells after treatment with cytokine, in all MM cells on prostaglandin-E2 treatment, and in MPP89, Ist-Mes2, and Ist-Mes1 after addition of epidermal growth factor. Treatment of MM cells with exemestane led to significant reduction of tumor cell growth, perturbation of cell cycle, caspase activation, poly(ADP-ribose)polymerase cleavage, and down-regulation of phosphorylation of Akt and Bcl-xL. In tumor tissues, we found a cytoplasmic localization of CYP19A1. By univariate analysis, overall survival resulted to be strongly influenced by high CYP19A1 expression (p = 0.001). CONCLUSION: These findings show that CYP19A1 is present in MM and that cell growth can be down-regulated by exemestane. As Akt pathway and Bcl-xL are implicated in conferring resistance to conventional chemotherapy, exemestane could open new treatment strategies to be associated with standard therapy for patients afflicted with MM.


Subject(s)
Androstadienes/pharmacology , Aromatase Inhibitors/pharmacology , Aromatase/chemistry , Aromatase/metabolism , Cell Proliferation/drug effects , Mesothelioma/drug therapy , Aromatase/genetics , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Cycle , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Follow-Up Studies , Humans , Immunoenzyme Techniques , Mesothelioma/enzymology , Mesothelioma/pathology , Prognosis , RNA, Messenger/genetics , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured
4.
Cancer Res ; 70(23): 9711-20, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20952509

ABSTRACT

The CCAAT-binding transcription factor NF-Y plays a central role in regulating cellular proliferation by controlling the expression of genes required for cell-cycle progression such as cyclin A, cyclin B1, cyclin B2, cdc25A, cdc25C, and cdk1. Here we show that unrestricted NF-Y activity leads to apoptosis in an E2F1- and wild-type p53 (wtp53)-dependent manner. Unrestricted NF-Y activity induced an increase in E2F1 mRNA and protein levels. Furthermore, NF-Y directly bound the E2F1 promoter and this correlated with the appearance of open chromatin marks. The ability of NF-Y to induce apoptosis was impaired in cells lacking E2F1 and wtp53. Moreover, NF-Y overexpression elicited phosphorylation of wt p53Ser18 in an E2F1-dependent manner. Our findings establish that NF-Y acts upstream of E2F1 in p53-mediated apoptosis.


Subject(s)
Apoptosis/physiology , CCAAT-Binding Factor/physiology , E2F1 Transcription Factor/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Blotting, Western , CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , E2F1 Transcription Factor/genetics , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , HCT116 Cells , HeLa Cells , Humans , Mice , Mice, Knockout , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Tumor Suppressor Protein p53/genetics
5.
Lung Cancer ; 70(3): 271-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20371132

ABSTRACT

The Aurora proteins are a small family of serine/threonine kinase that function in various stages of mitosis. Current interest in Aurora kinase relates to its role in tumours, and its potential as a therapeutic target. In this work we studied the expression of Aurora kinases A and B and related genes in human mesothelioma tissues and in five mesothelioma cell lines. Moreover, we analyzed the effects of ZM447439 (ZM), an Aurora kinase inhibitor, on cellular growth. Results evidenced an over-expression of Aurora kinase A and related genes in human mesothelioma tissues and an over-expression of Aurora kinases A and B in all cell lines. Moreover, we demonstrated that ZM447439 was able to inhibit cell growth in all cell lines and that this inhibition was due to a specific effect as demonstrated by the reduction in the level of Histone H3 phosphorylation. Our findings support a role of Aurora kinase in mesothelioma and the possibility of using Aurora kinase inhibitors in therapeutic modalities.


Subject(s)
Histones/metabolism , Mesothelioma/metabolism , Pleural Neoplasms/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Aurora Kinase A , Aurora Kinases , Benzamides/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Computational Biology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mesothelioma/drug therapy , Mesothelioma/genetics , Mesothelioma/pathology , Mesothelioma/physiopathology , Microarray Analysis , Phosphorylation/drug effects , Pleural Neoplasms/drug therapy , Pleural Neoplasms/genetics , Pleural Neoplasms/pathology , Pleural Neoplasms/physiopathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Quinazolines/pharmacology
6.
J Biol Chem ; 285(19): 14160-9, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20223820

ABSTRACT

Mitogen-activated protein kinase kinase 3 (MAP2K3) is a member of the dual specificity kinase group. Growing evidence links MAP2K3 to invasion and tumor progression. Here, we identify MAP2K3 as a transcriptional target of endogenous gain-of-function p53 mutants R273H, R175H, and R280K. We show that MAP2K3 modulation occurred at the mRNA and protein levels and that endogenous mutant p53 proteins are capable of binding to and activate the MAP2K3 promoter. In addition, we found that the studied p53 mutants regulate MAP2K3 gene expression through the involvement of the transcriptional cofactors NF-Y and NF-kappaB. Finally, functional studies showed that endogenous MAP2K3 knockdown inhibits proliferation and survival of human tumor cells, whereas the ectopic expression of MAP2K3 can rescue the proliferative defect induced by mutant p53 knockdown. Taken together, our findings define a novel player through which mutant p53 exerts its gain-of-function activity in cancer cells.


Subject(s)
Gene Expression Regulation, Neoplastic , MAP Kinase Kinase 3/genetics , Mutation/genetics , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Chromatin Immunoprecipitation , Flow Cytometry , Humans , MAP Kinase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase 3/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasms/pathology , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Transcriptional Activation , Up-Regulation
7.
Mol Cancer ; 9: 27, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20122271

ABSTRACT

BACKGROUND: Malignant mesothelioma (MM) is an aggressive tumor that is resistant to conventional modes of treatment with chemotherapy, surgery or radiation. Research into the molecular pathways involved in the development of MM should yield information that will guide therapeutic decisions. Epidermal growth factor receptor (EGFR) and cyclooxygenase-2 (COX-2) are involved in the carcinogenesis of MM. Combination of COX-2 and EGFR inhibitors, therefore, could be an effective strategy for reducing cell growth in those lines expressing the two molecular markers. RESULTS: In order to verify the effect of COX-2 and EGFR inhibitors, five MM cell lines NCI-2452, MPP89, Ist-Mes-1, Ist-Mes-2 and MSTO-211 were characterized for COX-2 and EGFR and then treated with respective inhibitors (rofecoxib and gefitinib) alone and in combination. Only MPP89, Ist-Mes-1 and Ist-Mes-2 were sensitive to rofecoxib and showed growth-inhibition upon gefitinib treatment. The combination of two drugs demonstrated synergistic effects on cell killing only in Ist-Mes-2, the cell line that was more sensitive to gefitinib and rofecoxib alone. Down-regulation of COX-2, EGFR, p-EGFR and up-regulation of p21 and p27 were found in Ist-Mes-2, after treatment with single agents and in combination. In contrast, association of two drugs resulted in antagonistic effect in Ist-Mes-1 and MPP89. In these cell lines after rofecoxib exposition, only an evident reduction of p-AKT was observed. No change in p-AKT in Ist-Mes-1 and MPP89 was observed after treatment with gefitinib alone and in combination with rofecoxib. CONCLUSIONS: Gefitinib and rofecoxib exert cell type-specific effects that vary between different MM cells. Total EGFR expression and downstream signalling does not correlate with gefitinib sensitivity. These data suggest that the effect of gefitinib can be potentiated by rofecoxib in MM cell lines where AKT is not activated.


Subject(s)
Lactones/pharmacology , Mesothelioma/pathology , Quinazolines/pharmacology , Sulfones/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclooxygenase 2/metabolism , Drug Synergism , ErbB Receptors/metabolism , Gefitinib , Humans , Mesothelioma/enzymology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
8.
Free Radic Biol Med ; 48(10): 1338-46, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20171273

ABSTRACT

HIPK2 is a stress-induced kinase and a transcriptional corepressor that functionally cooperates with p53 to suppress cancer. Activation of the p53 proapoptotic function requires a cascade of phosphorylations and acetylations, and HIPK2 takes part in both modifications in that it phosphorylates p53 Ser46 and induces p53 Lys382 acetylation. Here, to further investigate the role of HIPK2 in p53 activation, we started with the finding that HIPK2 inhibition upregulated Nox1, a homolog of the catalytic subunit of the superoxide-generating NADPH oxidase, involved in tumor progression and ROS production. We found that Nox1 inhibited p53 Lys382 acetylation, which is a target of SIRT1 deacetylase, and impaired p53 proapoptotic transcriptional activity. By the use of either small interfering RNAs to target SIRT1 or the SIRT1 inhibitor nicotinamide we found that Nox1-dependent inhibition of p53 transcriptional activity was SIRT1-dependent. Thus, Nox1 was unable to inhibit p53 when coexpressed with a SIRT1 deacetylase-defective mutant (SIRT1HY), suggesting a link between Nox1 and SIRT1 activity. Finally, recovery of HIPK2 function downregulated Nox1 expression with rescue of p53 Lys382 acetylation and p53 activity. Together, our findings indicate that Nox1 upregulation may activate SIRT1 and inhibit p53 and that Lys382 is important for p53 proapoptotic function.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma/metabolism , Colonic Neoplasms/metabolism , Lung Neoplasms/metabolism , NADPH Oxidases/biosynthesis , Tumor Suppressor Protein p53/metabolism , Acetylation/drug effects , Apoptosis/drug effects , Apoptosis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma/genetics , Carcinoma/pathology , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Line, Tumor , Cloning, Molecular , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutant Proteins/genetics , Mutant Proteins/metabolism , NADPH Oxidase 1 , NADPH Oxidases/genetics , Niacinamide/pharmacology , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Sirtuin 1/metabolism , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Tumor Suppressor Protein p53/genetics
9.
Cell Transplant ; 19(10): 1225-35, 2010.
Article in English | MEDLINE | ID: mdl-21208530

ABSTRACT

Techniques for medical tissue regeneration require an abundant source of human adult stem cells. There is increasing evidence that adipose stem cells contribute to restoration of tissue vascularization and organ function. The object of our study was to isolate and characterize adult adipose-derived stem cells from patients undergoing on lipoaspirate transplant with the aim to improve tissue regeneration. Adipose-derived stem cells were isolated and purified from the lipoaspirate of 15 patients and characterized for CD markers and the ability to differentiate toward the adipogenic lineage. We found that purified adipose stem cells express high level of CD49d, CD44, CD90, CD105, CD13, and CD71 and these markers of staminality were maintained at high level for at least 3 months and seven passages of in vitro culture. As expected, these cells resulted negative for the endothelial and hematopoietic-specific markers CD31, CD106, CD34, and CD45. Differentiation towards adipogenic lineage demonstrated that purified adipose-derived stem cells are still able to become adipocytes at least 3 months after in vitro culture. The analysis of Akt and MAPK phosphorylation confirmed a modulation of their activity during differentiation. Interestingly, we established for the first time that, among the p53 family members, a strong upregulation of p63 expression occurs in adipocytic differentiation, indicating a role for this transcription factor in adipocytic differentiation. Taken together, these data indicate that purified lipoaspirate-derived stem cells maintain their characteristic of staminality for a long period of in vitro culture, suggesting that they could be applied for cell-based therapy to improve autologous lipoaspirate transplant.


Subject(s)
Adipocytes/cytology , Adult Stem Cells/cytology , Adipocytes/metabolism , Adipose Tissue/cytology , Adult , Adult Stem Cells/metabolism , Aged , Biomarkers/metabolism , Cell Differentiation , Cells, Cultured , Flow Cytometry , Humans , Membrane Proteins/metabolism , Membrane Proteins/physiology , Middle Aged , Tissue and Organ Harvesting
10.
J Cell Mol Med ; 14(10): 2417-35, 2010 Oct.
Article in English | MEDLINE | ID: mdl-19863697

ABSTRACT

Even though several studies highlighted the role of maternal thyroid hormones (THs) during embryo-foetal development, direct evidence of their interaction with embryonic thyroid receptors (TRs) is still lacking. We generated a transgenic mouse model ubiquitously expressing a reporter gene tracing TH action during development. We engineered a construct (TRE2×) containing two TH-responsive elements controlling the expression of the LacZ reporter gene, which encodes ß-galactosidase (ß-gal). The specificity of the TRE2× activation by TH was evaluated in NIH3T3 cells by cotransfecting TRE2× along with TRs, retinoic or oestrogen receptors in the presence of their specific ligands. TRE2× transgene was microinjected into the zygotes, implanted in pseudopregnant BDF1 (a first-generation (F1) hybrid from a cross of C57BL/6 female and a DBA/2 male) mice and transgenic mouse models were developed. ß-gal expression was assayed in tissue sections of transgenic mouse embryos at different stages of development. In vitro, TRE2× transactivation was observed only following physiological T3 stimulation, mediated exclusively by TRs. In vivo, ß-gal staining, absent until embryonic day 9.5-10.5 (E9.5-E10.5), was observed as early as E11.5-E12.5 in different primordia (i.e. central nervous system, sense organs, intestine, etc.) of the TRE2× transgenic embryos, while the foetal thyroid function (FTF) was still inactive. Immunohistochemistry for TRs essentially colocalized with ß-gal staining. No ß-gal staining was detected in embryos of hypothyroid transgenic mice. Importantly, treatment with T3 in hypothyroid TRE2× transgenic mice rescued ß-gal expression. Our results provide in vivo direct evidence that during embryonic life and before the onset of FTF, maternal THs are transcriptionally active through the action of embryonic TRs. This model may have clinical relevance and may be employed to design end-point assays for new molecules affecting THs action.


Subject(s)
Embryonic Development , Gene Expression Regulation, Enzymologic , Thyroid Hormones/genetics , Transcriptional Activation , Animals , Central Nervous System/embryology , Central Nervous System/metabolism , Female , Gene Expression Regulation, Developmental , Genes, Reporter , Genetic Engineering , Immunohistochemistry , Lac Operon , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Pregnancy , Promoter Regions, Genetic , Thyroid Hormones/metabolism , Transgenes , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
11.
Mol Cancer ; 8: 85, 2009 Oct 14.
Article in English | MEDLINE | ID: mdl-19828042

ABSTRACT

BACKGROUND: Activation of p53-mediated gene transcription is a critical cellular response to DNA damage and involves a phosphorylation-acetylation cascade of p53. The discovery of differences in the response to different agents raises the question whether some of the p53 oncosuppressor functions might be exerted by different posttranslational modifications. Stress-induced homeodomain-interacting protein kinase-2 (HIPK2) phosphorylates p53 at serine-46 (Ser46) for p53 apoptotic activity; p53 acetylation at different C-terminus lysines including p300-mediated lysine-382 (Lys382) is also required for full activation of p53 transcriptional activity. The purpose of the current study was to evaluate the interplay among HIPK2, p300, and p53 in p53 acetylation and apoptotic transcriptional activity in response to drug by using siRNA interference, p300 overexpression or deacetylase inhibitors, in cancer cells. RESULTS: Knockdown of HIPK2 inhibited both adriamycin-induced Ser46 phosphorylation and Lys382 acetylation in p53 protein; however, while combination of ADR and zinc restored Ser46 phosphorylation it did not recover Lys382 acetylation. Chromatin immunoprecipitation studies showed that HIPK2 was required in vivo for efficient p300/p53 co-recruitment onto apoptotic promoters and that both p53 modifications at Ser46 and Lys382 were necessary for p53 apoptotic transcription. Thus, p53Lys382 acetylation in HIPK2 knockdown as well as p53 apoptotic activity in response to drug could be rescued by p300 overexpression. Similar effect was obtained with the Sirt1-inhibitor nicotinamide. Interestingly trichostatin A (TSA), the inhibitor of histone deacetylase complexes (HDAC) did not have effect, suggesting that Sirt1 was the deacetylase involved in p53 deacetylation in HIPK2 knockdown. CONCLUSION: These results reveal a novel role for HIPK2 in activating p53 apoptotic transcription. Our results indicate that HIPK2 may regulate the balance between p53 acetylation and deacetylation, by stimulating on one hand co-recruitment of p300 and p53Lys382 on apoptotic promoters and on the other hand by inhibiting Sirt1 deacetylase activity. We attempted to reactivate p53 apoptotic transcriptional activity by rescuing both Ser46 and Lys382 modification in response to drug. Our data propose combination strategies for the treatment of tumors with dysfunctional p53 and/or HIPK2 that include classical chemotherapy with pharmacological or natural agents such as Sirt1-deacetylase inhibitors or zinc, respectively.


Subject(s)
Apoptosis/genetics , Carrier Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Acetylation/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Doxorubicin/pharmacology , E1A-Associated p300 Protein/metabolism , Gene Deletion , Gene Knockdown Techniques , Humans , Hydroxamic Acids/pharmacology , Lysine/metabolism , Niacinamide/pharmacology , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Sirtuin 1/metabolism , Transcription, Genetic/drug effects , Zinc/pharmacology , bcl-2-Associated X Protein/metabolism
12.
PLoS One ; 4(8): e6819, 2009 Aug 28.
Article in English | MEDLINE | ID: mdl-19714248

ABSTRACT

BACKGROUND: The tumor suppressor homeodomain-interacting protein kinase-2 (HIPK2) by phosphorylating serine 46 (Ser46) is a crucial regulator of p53 apoptotic function. HIPK2 is also a transcriptional co-repressor of hypoxia-inducible factor-1alpha (HIF-1alpha) restraining tumor angiogenesis and chemoresistance. HIPK2 can be deregulated in tumors by several mechanisms including hypoxia. Here, we sought to target hypoxia by restoring HIPK2 function and suppressing HIF-1alpha, in order to provide evidence for the involvement of both HIPK2 and p53 in counteracting hypoxia-induced chemoresistance. METHODOLOGY/PRINCIPAL FINDINGS: Upon exposure of colon and lung cancer cells to hypoxia, by either low oxygen or cobalt, HIPK2 function was impaired allowing for increased HIF-1alpha expression and inhibiting the p53-apoptotic response to drug. Cobalt suppressed HIPK2 recruitment onto HIF-1alpha promoter. Hypoxia induced expression of the p53 target MDM2 that downregulates HIPK2, thus MDM2 inhibition by siRNA restored the HIPK2/p53Ser46 response to drug. Zinc supplementation to hypoxia-treated cells increased HIPK2 protein stability and nuclear accumulation, leading to restoration of HIPK2 binding to HIF-1alpha promoter, repression of MDR1, Bcl2, and VEGF genes, and activation of the p53 apoptotic response to drug. Combination of zinc and ADR strongly suppressed tumor growth in vivo by inhibiting HIF-1 pathway and upregulating p53 apoptotic target genes. CONCLUSIONS/SIGNIFICANCE: We show here for the first time that hypoxia-induced HIPK2 deregulation was counteracted by zinc that restored HIPK2 suppression of HIF-1 pathway and reactivated p53 apoptotic response to drug, underscoring the potential use of zinc supplementation in combination with chemotherapy to address hypoxia and improve tumor treatment.


Subject(s)
Carrier Proteins/metabolism , Cell Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/pharmacology , Cobalt/pharmacology , Doxorubicin/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-mdm2/genetics , Transcription, Genetic/drug effects , Zinc/pharmacology
13.
FASEB J ; 23(11): 3957-66, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19608627

ABSTRACT

MicroRNAs (miRs) are 21- to 23-nucleotide RNA molecules that regulate the stability or translational efficiency of target messenger RNAs of proteins involved in cell growth and apoptosis. miR-92 is part of the mir-17-92 cluster, which comprises members with an effect on cell proliferation. However, the role of miR-92 is unknown, and its targets have not been identified. Here, we describe a mechanism through which miR-92 contributes to regulate cell proliferation. Using a miR-92 synthetic double-strand oligonucleotide, we demonstrate that miR-92 increases 32D myeloid cell proliferation and 5-bromo-2-deoxyuridine (BrdU) incorporation and inhibits cell death. The effect is miR-92 specific since the miR-92 antagomir inhibits cell proliferation. Moreover, we show that miR-92 acts by modulating p63-isoform abundance through down-regulatation of endogenous DeltaNp63beta. Using luciferase reporters containing p63 3'UTR fragments with wild-type or mutant miR-92 complementary sites, we demonstrate that the wild-type 3'UTR is a direct target of miR-92. Finally, we observed that a miR-92-resistant DeltaNp63beta isoform (without 3'UTR) inhibits cell proliferation and parallels the effect of the antagomir. We conclude that one of the molecular mechanisms through which miR-92 increases cell proliferation is by negative regulation of an isoform of the cell-cycle regulator p63.


Subject(s)
Antigens, CD/metabolism , MicroRNAs/physiology , Myeloid Cells/drug effects , Platelet Membrane Glycoproteins/metabolism , 3' Untranslated Regions , Antigens, CD/drug effects , Cell Proliferation/drug effects , Down-Regulation , Gene Knockdown Techniques , HCT116 Cells , Humans , Platelet Membrane Glycoproteins/drug effects , Protein Isoforms/metabolism , Tetraspanin 30
14.
Cancer Res ; 69(14): 5978-86, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19567674

ABSTRACT

Increased expression of alpha(6)beta(4) integrin in several epithelial cancers promotes tumor progression; however, the mechanism underlying its transcriptional regulation remains unclear. Here, we show that depletion of homeodomain-interacting protein kinase 2 (HIPK2) activates beta(4) transcription that results in a strong increase of beta(4)-dependent mitogen-activated protein kinase and Akt phosphorylation, anchorage-independent growth, and invasion. In contrast, stabilization of HIPK2 represses beta(4) expression in wild-type p53 (wtp53)-expressing cells but not in p53-null cells or cells expressing mutant p53, indicating that HIPK2 requires a wtp53 to inhibit beta(4) transcription. Consistent with our in vitro findings, a strong correlation between beta(4) overexpression and HIPK2 inactivation by cytoplasmic relocalization was observed in wtp53-expressing human breast carcinomas. Under loss of function of HIPK2 or p53, the p53 family members TAp63 and TAp73 strongly activate beta(4) transcription. These data, by revealing that beta(4) expression is transcriptionally repressed in tumors by HIPK2 and p53 to impair beta(4)-dependent tumor progression, suggest that loss of p53 function favors the formation of coactivator complex with the TA members of the p53 family to allow beta(4) transcription.


Subject(s)
Carrier Proteins/metabolism , Integrin beta4/metabolism , Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carrier Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Cytoplasm/metabolism , Disease Progression , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Histone Acetyltransferases/metabolism , Humans , Immunohistochemistry , Integrin beta4/genetics , Mitogen-Activated Protein Kinases/metabolism , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , p300-CBP Transcription Factors/metabolism
15.
Cancer Res ; 69(15): 6241-8, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19638586

ABSTRACT

In the past few years, much effort has been devoted to show the single-target specificity of nongenotoxic, p53 reactivating compounds. However, the divergent biological responses induced by the different compounds, even in the same tumor cells, demand additional mechanistic insights, whose knowledge may lead to improved drug design or selection of the most potent drug combinations. To address the molecular mechanism underlying induction of mitotic arrest versus clinically more desirable apoptosis, we took advantage of two MDM2 antagonists, Nutlin-3 and RITA, which respectively produce these two outcomes. We show that, along with p53 reactivation, the proapoptotic p53-activator HIPK2 is degraded by MDM2 in Nutlin-3-treated cells, but activated by transiently reduced MDM2 levels in RITA-treated ones. Gain- and loss-of-function experiments revealed the functional significance of MDM2-mediated HIPK2 regulation in cell decision between mitotic arrest and apoptosis in both types of p53 reactivation. These data indicate that strategies of p53 reactivation by MDM2 inhibition should also take into consideration MDM2 targets other than p53, such as the apoptosis activator HIPK2.


Subject(s)
Carrier Proteins/biosynthesis , Furans/pharmacology , Imidazoles/pharmacology , Piperazines/pharmacology , Protein Serine-Threonine Kinases/biosynthesis , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , HCT116 Cells , Humans , Mitosis/drug effects , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Up-Regulation/drug effects
16.
EMBO J ; 28(13): 1926-39, 2009 Jul 08.
Article in English | MEDLINE | ID: mdl-19521340

ABSTRACT

MDM4 is a key regulator of p53, whose biological activities depend on both transcriptional activity and transcription-independent mitochondrial functions. MDM4 binds to p53 and blocks its transcriptional activity; however, the main cytoplasmic localization of MDM4 might also imply a regulation of p53-mitochondrial function. Here, we show that MDM4 stably localizes at the mitochondria, in which it (i) binds BCL2, (ii) facilitates mitochondrial localization of p53 phosphorylated at Ser46 (p53Ser46(P)) and (iii) promotes binding between p53Ser46(P) and BCL2, release of cytochrome C and apoptosis. In agreement with these observations, MDM4 reduction by RNA interference increases resistance to DNA-damage-induced apoptosis in a p53-dependent manner and independently of transcription. Consistent with these findings, a significant downregulation of MDM4 expression associates with cisplatin resistance in human ovarian cancers, and MDM4 modulation affects cisplatin sensitivity of ovarian cancer cells. These data define a new localization and function of MDM4 that, by acting as a docking site for p53Ser46(P) to BCL2, facilitates the p53-mediated intrinsic-apoptotic pathway. Overall, our results point to MDM4 as a double-faced regulator of p53.


Subject(s)
Mitochondria/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Animals , Antineoplastic Agents/metabolism , Apoptosis , Carcinoma/genetics , Carcinoma/metabolism , Cell Line, Tumor , Cells, Cultured , Cisplatin/metabolism , Cytochromes c/metabolism , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mitochondria/ultrastructure , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins c-bcl-2 , Ubiquitin-Protein Ligases/analysis
17.
J Clin Invest ; 119(5): 1093-108, 2009 May.
Article in English | MEDLINE | ID: mdl-19363294

ABSTRACT

The identification of biomarkers that distinguish between aggressive and indolent forms of prostate cancer (PCa) is crucial for diagnosis and treatment. In this study, we used cultured cells derived from prostate tissue from patients with PCa to define a molecular mechanism underlying the most aggressive form of PCa that involves the functional activation of eNOS and HIFs in association with estrogen receptor beta (ERbeta). Cells from patients with poor prognosis exhibited a constitutively hypoxic phenotype and increased NO production. Upon estrogen treatment, formation of ERbeta/eNOS, ERbeta/HIF-1alpha, or ERbeta/HIF-2alpha combinatorial complexes led to chromatin remodeling and transcriptional induction of prognostic genes. Tissue microarray analysis, using an independent cohort of patients, established a hierarchical predictive power for these proteins, with expression of eNOS plus ERbeta and nuclear eNOS plus HIF-2alpha being the most relevant indicators of adverse clinical outcome. Genetic or pharmacologic modulation of eNOS expression and activity resulted in reciprocal conversion of the transcriptional signature in cells from patients with bad or good outcome, respectively, highlighting the relevance of eNOS in PCa progression. Our work has considerable clinical relevance, since it may enable the earlier diagnosis of aggressive PCa through routine biopsy assessment of eNOS, ERbeta, and HIF-2alpha expression. Furthermore, proposing eNOS as a therapeutic target fosters innovative therapies for PCa with NO inhibitors, which are employed in preclinical trials in non-oncological diseases.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Estrogen Receptor beta/metabolism , Gene Expression Regulation, Neoplastic/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Nitric Oxide Synthase Type III/metabolism , Prostatic Neoplasms/diagnosis , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers , Cell Hypoxia/physiology , Cell Line, Tumor , Cell Nucleus/metabolism , Chromatin Assembly and Disassembly/physiology , Cytoplasm/metabolism , Enzyme Inhibitors/pharmacology , Estradiol/pharmacology , Gene Expression/drug effects , Gene Expression/genetics , Glucose Transporter Type 1/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/genetics , Prognosis , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/metabolism , Protein Binding/drug effects , Protein Binding/genetics , Response Elements/genetics , Telomerase/genetics , Telomerase/metabolism , Tissue Array Analysis , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
18.
Mol Cancer Ther ; 8(3): 543-51, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19258428

ABSTRACT

We reported previously that the disruption of c-Myc through mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) inhibition blocks the expression of the transformed phenotype in the embryonal rhabdomyosarcoma (ERMS) cell line (RD), thereby inducing myogenic differentiation in vitro. In this article, we investigate whether MEK/ERK inhibition, by the MEK/ERK inhibitor U0126, affects c-Myc protein level and growth of RMS tumor in an in vivo xenograft model. U0126 significantly reduced RMS tumor growth in RD cell line-xenotransplanted mice. Immunobiochemical and immunohistochemical analysis showed (a) phospho-active ERK levels were reduced by U0126 therapy and unaltered in normal tissues, (b) phospho-Myc and c-Myc was reduced commensurate with phospho-ERK inhibition, and (c) reduction in Ki-67 and endothelial (CD31) marker expression. These results indicate that MEK/ERK inhibition affects growth and angiogenic signals in tumor. The RD-M1 cultured xenograft tumor-derived cell line and the ERMS cell line TE671 responded to U0126 by arresting growth, down-regulating c-Myc, and initiating myogenesis. All these results suggest a tight correlation of MEK/ERK inhibition with c-Myc down-regulation and arrest of tumor growth. Thus, MEK inhibitors may be investigated for a signal transduction-based targeting of the c-Myc as a therapeutic strategy in ERMS.


Subject(s)
Butadienes/pharmacology , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/antagonists & inhibitors , Muscle Neoplasms/pathology , Nitriles/pharmacology , Rhabdomyosarcoma, Embryonal/pathology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Butadienes/therapeutic use , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Nude , Muscle Neoplasms/drug therapy , Muscle Neoplasms/genetics , Nitriles/therapeutic use , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Rhabdomyosarcoma, Embryonal/drug therapy , Rhabdomyosarcoma, Embryonal/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
Mol Cancer ; 8: 1, 2009 Jan 07.
Article in English | MEDLINE | ID: mdl-19128456

ABSTRACT

BACKGROUND: Homeodomain-interacting protein kinase-2 (HIPK2), a transcriptional co-repressor with apoptotic function, can affect hypoxia-inducible factor 1 (HIF-1) transcriptional activity, through downmodulation of its HIF-1alpha subunit, in normoxic condition. Under hypoxia, a condition often found in solid tumors, HIF-1alpha is activated to induce target genes involved in chemoresistance, inhibition of apoptosis and tumor progression. Here, we investigated whether the HIPK2 overexpression could downregulate HIF-1alpha expression and activity in tumor cells treated with hypoxia-mimicking condition, and evaluated whether HIPK2-dependent downregulation of HIF-1alpha could sensitize chemoresistant tumor cells to adriamycin (ADR)-induced apoptosis. METHODS: Tumor cell lines carrying wild-type p53, siRNA p53, or mutant p53 were overexpressed with HIPK2 (full length or catalytic inactive mutant) and treated with cobalt chloride (CoCl2) to mimic hypoxia, in the presence or absence of ADR treatment. HIF-1alpha expression was measured by semiquantitative reverse-transcriptase (RT)-PCR and Western immunoblotting and HIF-1 activity was evaluated by luciferase assay using reporter plasmid containing hypoxia response elements (HREs) upstream of luciferase gene. HIF-1 target genes, including multidrug resistance 1 (MDR1) and the antiapoptotic Bcl2 were determined by RT-PCR. Cell survival and apoptosis were measured by colony assay and cleavage of the caspase-3 substrate PARP, respectively. RESULTS: Overexpression of HIPK2 resulted in downmodulation of cobalt-stabilized HIF-1alpha protein and HIF-1alpha mRNA levels, with subsequent inhibition of HIF-1 transcriptional activity. MDR1 and Bcl-2 gene expression was downmodulated by HIPK2 overexpression in cobalt-treated cells. Inhibition of HIF-1 transcriptional activity was dependent on HIPK2 catalytic activity. HIPK2 overexpression did not induce per se apoptosis of cobalt-treated cells, on the contrary it sensitized cobalt-treated cells to ADR-induced apoptosis, regardless of their p53 status. CONCLUSION: The ability of HIPK2 to restore the apoptosis-inducing potential of chemotherapeutic drug in hypoxia-mimicking condition and therefore to sensitize chemoresistant tumor cells suggests that HIPK2 may induce fundamental alterations in cell signaling pathways, involving or not p53 function. Thus potential use of HIPK2 is promising for cancer treatment by potentiating cytotoxic therapies, regardless of p53 cell status.


Subject(s)
Apoptosis/physiology , Carrier Proteins/metabolism , Drug Resistance, Neoplasm/physiology , Gene Expression Regulation, Neoplastic/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Protein Serine-Threonine Kinases/metabolism , Antibiotics, Antineoplastic/pharmacology , Blotting, Western , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Line, Tumor , Doxorubicin/pharmacology , Humans , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Suppressor Protein p53/metabolism
20.
Exp Cell Res ; 315(1): 67-75, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18996371

ABSTRACT

The maintenance of p53 transactivation activity is important for p53 apoptotic function. We have shown that stable knockdown of HIPK2 induces p53 misfolding with inhibition of p53 target gene transcription. In this study we established a lentiviral-based system for doxycyclin (Dox)-induced conditional interference of HIPK2 expression to evaluate the molecular mechanisms involved in p53 deregulation. We found that HIPK2 knockdown induced metallothionein 2A (MT2A) upregulation as assessed by RT-PCR analysis, increased promoter acetylation, and increased promoter luciferase activity. The MT2A upregulation correlated with resistance to Adriamycin (ADR)-driven apoptosis and with p53 inhibition. Thus, acute knockdown of HIPK2 (HIPK2i) induced misfolded p53 protein in MCF7 breast cancer cells and inhibited p53 DNA-binding and transcription activities in response to ADR treatment. Previous works show that MT may modulate p53 activity through zinc exchange. Here, we found that inhibition of MT2A expression by siRNA in the HIPK2i cells restored p53 transcription activity. Similarly zinc supplementation to HIPK2i cells restored p53 transcription activity and drug-induced apoptosis. These data support the notion that MT2A is involved in p53 deregulation and strengthen the possibility that combination of chemotherapy and zinc might be useful to treat tumors with inactive wtp53.


Subject(s)
Metallothionein/metabolism , Protein Serine-Threonine Kinases/deficiency , Tumor Suppressor Protein p53/metabolism , Zinc/metabolism , Apoptosis/drug effects , Carrier Proteins/metabolism , Cell Line, Tumor , DNA, Neoplasm/metabolism , Doxorubicin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lentivirus , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Binding/drug effects , Protein Conformation/drug effects , Protein Folding/drug effects , Protein Serine-Threonine Kinases/metabolism , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Up-Regulation/drug effects , Zinc/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...