Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 259
Filter
1.
Sci Rep ; 9(1): 17933, 2019 11 29.
Article in English | MEDLINE | ID: mdl-31784573

ABSTRACT

In this report we describe the development of a Fluorescent Protein-Protein Interaction-visualization (FLUOPPI) to enable the simultaneous measurement of both Mdm2:p53 and Mdm4:p53 interactions in order to assess the relative efficiencies of mimetic molecules of the p53 peptide helix against both PPIs. Mdm2 and Mdm4 overexpression frequently leads to the inactivation of non-mutated p53 in human cancers, via inhibition of its transcriptional activity, enhancing its degradation by the proteasome or by preventing its nuclear import. Development of inhibitors to disrupt the binding of one or both of these protein interactions have been the subject of intensive pharmaceutical development for anti-cancer therapies. Using the bimodal FLUOPPI system we have characterised compounds that were either monospecific for Mdm2 or bispecific for both Mdm2 and Mdm4. We have also demonstrated that the FLUOPPI assay can reliably differentiate between specific and non-specific disruption of these protein complexes via accurate assessment and normalization to the cell population under measurement. We envision that this methodology will increase the efficiency of identifying compounds that are either specific against a single PPI from a closely related family of interactions or compounds that interact across multiple related PPI pairs, depending on which is more desirable.


Subject(s)
Cell Cycle Proteins/metabolism , Protein Interaction Mapping/methods , Protein Interaction Maps , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , CHO Cells , Cell Line , Cricetulus , Fluorescence , Humans , Microscopy, Fluorescence/methods , Neoplasms/metabolism , Protein Interaction Maps/drug effects
2.
Protein Eng Des Sel ; 31(7-8): 301-312, 2018 07 01.
Article in English | MEDLINE | ID: mdl-30169723

ABSTRACT

Engineered non-antibody scaffold proteins constitute a rapidly growing technology for diagnostics and modulation/perturbation of protein function. Here, we describe the rapid and systematic development of high-affinity 10FN3 domain inhibitors of the MDM2 and MDMX proteins. These are often overexpressed in cancer and represent attractive drug targets. Using facile in vitro expression and pull-down assay methodology, numerous design iterations addressing insertion site(s) and spacer length were screened for optimal presentation of an MDM2/X dual peptide inhibitor in the 10FN3 scaffold. Lead inhibitors demonstrated robust, on-target cellular inhibition of MDM2/X leading to activation of the p53 tumor suppressor. Significant improvement to target engagement was observed by increasing valency within a single 10FN3 domain, which has not been demonstrated previously. We further established stable reporter cell lines with tunable expression of EGFP-fused 10FN3 domain inhibitors, and showed their intracellular location to be contingent on target engagement. Importantly, competitive inhibition of MDM2/X by small molecules and cell-penetrating peptides led to a readily observable phenotype, indicating significant potential of the developed platform as a robust tool for cell-based drug screening.


Subject(s)
Proto-Oncogene Proteins c-mdm2/immunology , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Active Transport, Cell Nucleus , Amino Acid Sequence , Cell Line , Cell Nucleus/metabolism , Models, Molecular , Protein Domains
3.
Sci Rep ; 7(1): 1763, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28496125

ABSTRACT

Linear peptides can mimic and disrupt protein-protein interactions involved in critical cell signaling pathways. Such peptides however are usually protease sensitive and unable to engage with intracellular targets due to lack of membrane permeability. Peptide stapling has been proposed to circumvent these limitations but recent data has suggested that this method does not universally solve the problem of cell entry and can lead to molecules with off target cell lytic properties. To address these issues a library of stapled peptides was synthesized and screened to identify compounds that bound Mdm2 and activated cellular p53. A lead peptide was identified that activated intracellular p53 with negligible nonspecific cytotoxicity, however it still bound serum avidly and only showed a marginal improvement in cellular potency. These hurdles were overcome by successfully identifying a pyridinium-based cationic lipid formulation, which significantly improved the activity of the stapled peptide in a p53 reporter cell line, principally through increased vesicular escape. These studies underscore that stapled peptides, which are cell permeable and target specific, can be identified with rigorous experimental design and that these properties can be improved through use with lipid based formulations. This work should facilitate the clinical translation of stapled peptides.


Subject(s)
Drug Delivery Systems , Hydrocarbons/chemistry , Intracellular Space/metabolism , Lipids/chemistry , Multiprotein Complexes/metabolism , Peptides/chemistry , Cations , Cell Survival , Endosomes/metabolism , Genes, Reporter , HEK293 Cells , Humans , Inhibitory Concentration 50 , Peptide Library , Proto-Oncogene Proteins c-mdm2/metabolism , Pyridines/chemistry , Transcriptional Activation/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
4.
Oncogene ; 36(14): 2002-2013, 2017 04 06.
Article in English | MEDLINE | ID: mdl-27869164

ABSTRACT

Overexpression of mutant p53 is a common finding in most cancers but testicular tumours accumulate wild-type p53 (wtp53). In contrast to the accepted concept that p53 homozygous mutant mice do not accumulate mutant p53 in normal cells, our study on a mutant p53 mouse model of Li-Fraumeni syndrome harbouring the hot-spot p53R172H mutation described an elevated level of mutant p53 in non-cancerous mouse tissues. Here we use detailed immunohistochemical analysis to document the expression of p53R172H in mouse testis. In developing and adult testes, p53R172H was expressed in gonocytes, type A, Int, B spermatogonia as well as in pre-Sertoli cells and Leydig cells but was undetectable in spermatocytes and spermatids. A similar staining pattern was demonstrated for wtp53. However, the intensity of wtp53 staining was generally weaker than that of p53R172H, which indicates that the expression of p53R172H can be a surrogate marker of p53 gene transcription. Comparing the responses of wtp53 and p53R172H to irradiation, we found persistent DNA double-strand breaks in p53R172H testes and the formation of giant spermatogonia (GSG) following persistent DNA damage in p53R172H and p53-null mice. Strikingly, we found that p53R172H promotes spontaneous formation of GSG in non-stressed p53R172H ageing mice. Two types of GSG: Viable and Degenerative GSG were defined. We elucidate the factors involved in the formation of GSG: the loss of p53 function is a requirement for the formation of GSG whereas DNA damage acts as a promoting trigger. The formation of GSG does not translate to higher efficacy of testicular tumorigenesis arising from mutant p53 cells, which might be due to the presence of delayed-onset of p53-independent apoptosis.


Subject(s)
DNA Damage/physiology , Genes, p53/physiology , Mutant Proteins/physiology , Spermatogonia/pathology , Amino Acid Substitution , Animals , Animals, Newborn , Apoptosis/genetics , Arginine/genetics , Embryo, Mammalian , Histidine/genetics , Male , Mice , Mice, Transgenic , Mutant Proteins/genetics , Mutation Rate , Spermatogonia/metabolism , Testicular Neoplasms/genetics , Testicular Neoplasms/pathology , Testis/metabolism , Testis/pathology
5.
Cell Death Differ ; 23(9): 1515-28, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27104929

ABSTRACT

Bidirectional interactions between astrocytes and neurons have physiological roles in the central nervous system and an altered state or dysfunction of such interactions may be associated with neurodegenerative diseases, such as Alzheimer's disease (AD) and amyotrophic lateral sclerosis (ALS). Astrocytes exert structural, metabolic and functional effects on neurons, which can be either neurotoxic or neuroprotective. Their neurotoxic effect is mediated via the senescence-associated secretory phenotype (SASP) involving pro-inflammatory cytokines (e.g., IL-6), while their neuroprotective effect is attributed to neurotrophic growth factors (e.g., NGF). We here demonstrate that the p53 isoforms Δ133p53 and p53ß are expressed in astrocytes and regulate their toxic and protective effects on neurons. Primary human astrocytes undergoing cellular senescence upon serial passaging in vitro showed diminished expression of Δ133p53 and increased p53ß, which were attributed to the autophagic degradation and the SRSF3-mediated alternative RNA splicing, respectively. Early-passage astrocytes with Δ133p53 knockdown or p53ß overexpression were induced to show SASP and to exert neurotoxicity in co-culture with neurons. Restored expression of Δ133p53 in near-senescent, otherwise neurotoxic astrocytes conferred them with neuroprotective activity through repression of SASP and induction of neurotrophic growth factors. Brain tissues from AD and ALS patients possessed increased numbers of senescent astrocytes and, like senescent astrocytes in vitro, showed decreased Δ133p53 and increased p53ß expression, supporting that our in vitro findings recapitulate in vivo pathology of these neurodegenerative diseases. Our finding that Δ133p53 enhances the neuroprotective function of aged and senescent astrocytes suggests that the p53 isoforms and their regulatory mechanisms are potential targets for therapeutic intervention in neurodegenerative diseases.


Subject(s)
Tumor Suppressor Protein p53/metabolism , Alternative Splicing , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Autophagy/drug effects , Brain/metabolism , Brain/pathology , Cells, Cultured , Cellular Senescence , Coculture Techniques , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Leupeptins/pharmacology , Neurons/cytology , Neurons/metabolism , Neuroprotection/physiology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Sequestosome-1 Protein/antagonists & inhibitors , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Serine-Arginine Splicing Factors/antagonists & inhibitors , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
6.
Oncogene ; 34(48): 5933-41, 2015 Nov 26.
Article in English | MEDLINE | ID: mdl-25746004

ABSTRACT

In mice, the deletion of either Mdm2 or Mdm4 results in a p53-dependent embryonic lethality. We used zinc-finger nucleases to construct mutations in the mdm2 and mdm4 genes of zebrafish. Although the loss of mdm2 results in a p53-dependent early embryonic lethality, mdm4 mutant fish are viable and grow to adulthood. We also found that an in-frame five-amino acid deletion in mdm2 creates a novel hypomorphic allele. The lethal phenotype observed in the mdm2 mutant fish could be partially rescued by injecting mRNA encoding functional Mdm2, and this required the E3 ligase activity of the protein. Complete rescue was obtained by crossing the mdm2 mutant fish onto a p53M214K mutant background. Although p53 mutant fish on a wild-type mdm2 background were shown to accumulate high levels of p53 protein specifically in tumor tissues, we detected extensive staining of p53 in many normal tissues of the mdm2-p53M214K double-mutant fish. Our results are suggestive of the hypothesis that p53 protein accumulates during tumor formation as a result of tumor-specific inactivation of the Mdm2 pathway.


Subject(s)
Animals, Genetically Modified/metabolism , Eye Neoplasms/pathology , Mutation/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Animals, Genetically Modified/embryology , Animals, Genetically Modified/genetics , Blotting, Western , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/pathology , Eye Neoplasms/genetics , Eye Neoplasms/metabolism , Gene Expression Regulation, Developmental , Mice , Phenotype , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Suppressor Protein p53/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
7.
Cell Death Differ ; 22(7): 1081-93, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25526089

ABSTRACT

P53 is critically important in preventing oncogenesis but its role in inflammation in general and in the function of inflammatory macrophages in particular is not clear. Here, we show that bone marrow-derived macrophages exhibit endogenous p53 activity, which is increased when macrophages are polarized to the M2 (alternatively activated macrophage) subtype. This leads to reduced expression of M2 genes. Nutlin-3a, which destabilizes the p53/MDM2 (mouse double minute 2 homolog) complex, promotes p53 activation and further downregulates M2 gene expression. In contrast, increased expression of M2 genes was apparent in M2-polarized macrophages from p53-deficient and p53 mutant mice. Furthermore, we show, in mice, that p53 also regulates M2 polarization in peritoneal macrophages from interleukin-4-challenged animals and that nutlin-3a retards the development of tolerance to Escherichia coli lipopolysaccharide. P53 acts via transcriptional repression of expression of c-Myc (v-myc avian myelocytomatosis viral oncogene homolog) gene by directly associating with its promoter. These data establish a role for the p53/MDM2/c-MYC axis as a physiological 'brake' to the M2 polarization process. This work reveals a hitherto unknown role for p53 in macrophages, provides further insight into the complexities of macrophage plasticity and raises the possibility that p53-activating drugs, many of which are currently being trialled clinically, may have unforeseen effects on macrophage function.


Subject(s)
Macrophage Activation , Macrophages/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Cell Polarity , Gene Expression Regulation , Imidazoles/pharmacology , Interleukin-4/metabolism , Lipopolysaccharides , Macrophages/immunology , Mice , Mice, Transgenic , Piperazines/pharmacology , Proto-Oncogene Proteins c-mdm2/immunology , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/immunology , Signal Transduction/immunology , Tumor Suppressor Protein p53/physiology
8.
Cell Death Differ ; 21(9): 1377-87, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24926616

ABSTRACT

In addition to the tumor suppressor p53 protein, also termed p53α, the TP53 gene produces p53ß and p53γ through alternative splicing of exons 9ß and 9γ located within TP53 intron 9. Here we report that both TG003, a specific inhibitor of Cdc2-like kinases (Clk) that regulates the alternative splicing pre-mRNA pathway, and knockdown of SFRS1 increase expression of endogenous p53ß and p53γ at mRNA and protein levels. Development of a TP53 intron 9 minigene shows that TG003 treatment and knockdown of SFRS1 promote inclusion of TP53 exons 9ß/9γ. In a series of 85 primary breast tumors, a significant association was observed between expression of SFRS1 and α variant, supporting our experimental data. Using siRNA specifically targeting exons 9ß/9γ, we demonstrate that cell growth can be driven by modulating p53ß and p53γ expression in an opposite manner, depending on the cellular context. In MCF7 cells, p53ß and p53γ promote apoptosis, thus inhibiting cell growth. By transient transfection, we show that p53ß enhanced p53α transcriptional activity on the p21 and Bax promoters, while p53γ increased p53α transcriptional activity on the Bax promoter only. Moreover, p53ß and p53γ co-immunoprecipitate with p53α only in the presence of p53-responsive promoter. Interestingly, although p53ß and p53γ promote apoptosis in MCF7 cells, p53ß and p53γ maintain cell growth in response to TG003 in a p53α-dependent manner. The dual activities of p53ß and p53γ isoforms observed in non-treated and TG003-treated cells may result from the impact of TG003 on both expression and activities of p53 isoforms. Overall, our data suggest that p53ß and p53γ regulate cellular response to modulation of alternative splicing pre-mRNA pathway by a small drug inhibitor. The development of novel drugs targeting alternative splicing process could be used as a novel therapeutic approach in human cancers.


Subject(s)
Alternative Splicing/genetics , Tumor Suppressor Protein p53/genetics , Cell Line, Tumor , Humans , MCF-7 Cells , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tumor Suppressor Protein p53/metabolism
9.
Oncogene ; 32(29): 3461-9, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-22986526

ABSTRACT

The RNA helicase p68 (DDX5) is an established co-activator of the p53 tumour suppressor that itself has a pivotal role in orchestrating the cellular response to DNA damage. Although several factors influence the biological outcome of p53 activation, the mechanisms governing the choice between cell-cycle arrest and apoptosis remain to be elucidated. In the present study, we show that, while p68 is critical for p53-mediated transactivation of the cell-cycle arrest gene p21(WAF1/CIP1), it is dispensable for induction of several pro-apoptotic genes in response to DNA damage. Moreover, p68 depletion results in a striking inhibition of recruitment of p53 and RNA Pol II to the p21 promoter but not to the Bax or PUMA promoters, providing an explanation for the selective effect on p21 induction. Importantly, these findings are mirrored in a novel inducible p68 knockout mouse model in which p68 depletion results in a selective inhibition of p21 induction in several tissues. Moreover, in the bone marrow, p68 depletion results in an increased sensitivity to γ-irradiation, consistent with an increased level of apoptosis. These data highlight a novel function of p68 as a modulator of the decision between p53-mediated growth arrest and apoptosis in vitro and in vivo.


Subject(s)
Apoptosis/physiology , Cell Cycle Checkpoints/physiology , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , DEAD-box RNA Helicases/metabolism , DNA Damage/physiology , Animals , Blotting, Western , Chromatin Immunoprecipitation , Flow Cytometry , Immunohistochemistry , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Transcriptional Activation/physiology , Transfection , Tumor Suppressor Protein p53/metabolism
10.
Oncogene ; 32(10): 1252-65, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-22580601

ABSTRACT

Tumour-derived mutant p53 proteins promote invasion, in part, by enhancing Rab coupling protein (RCP)-dependent receptor recycling. Here we identified MET as an RCP-binding protein and showed that mutant p53 promoted MET recycling. Mutant p53-expressing cells were more sensitive to hepatocyte growth factor, the ligand for MET, leading to enhanced MET signalling, invasion and cell scattering that was dependent on both MET and RCP. In cells expressing the p53 family member TAp63, inhibition of TAp63 also lead to cell scattering and MET-dependent invasion. However, in cells that express very low levels of TAp63, the ability of mutant p53 to promote MET-dependent cell scattering was independent of TAp63. Taken together, our data show that mutant p53 can enhance MET signalling to promote cell scattering and invasion through both TAp63-dependent and -independent mechanisms. MET has a predominant role in metastatic progression and the identification of mechanisms through which mutations in p53 can drive MET signalling may help to identify and direct therapy.


Subject(s)
Mutation , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Movement , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , HCT116 Cells , HT29 Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Neoplasm Invasiveness , Phosphorylation , Signal Transduction , Transcription Factors/metabolism , Transfection , Tumor Suppressor Proteins/metabolism
11.
Oncogene ; 32(25): 3101-10, 2013 Jun 20.
Article in English | MEDLINE | ID: mdl-22824801

ABSTRACT

Heat shock proteins Hsp90 and Hsp70 facilitate protein folding but can also direct proteins for ubiquitin-mediated degradation. The mechanisms regulating these opposite activities involve Hsp binding to co-chaperones including CHIP and HOP at their C-termini. We demonstrated that the extreme C-termini of Hsp70 and Hsp90 contain phosphorylation sites targeted by kinases including CK1, CK2 and GSK3-ß in vitro. The phosphorylation of Hsp90 and Hsp70 prevents binding to CHIP and thus enhances binding to HOP. Highly proliferative cells contain phosphorylated chaperones in complex with HOP and phospho-mimetic and non-phosphorylable Hsp mutant proteins show that phosphorylation is directly associated with increased proliferation rate. We also demonstrate that primary human cancers contain high levels of phosphorylated chaperones and show increased levels of HOP protein and mRNA. These data identify C-terminal phosphorylation of Hsp70 and Hsp90 as a switch for regulating co-chaperone binding and indicate that cancer cells possess an elevated protein folding environment by the concerted action of co-chaperone expression and chaperone modifications. In addition to identifying the pathway responsible for regulating chaperone-mediated protein folding/degradation balances in normal cells, the data provide novel mechanisms to account for the aberrant chaperone activities observed in human cancer cells and have implications for the application of anti-chaperone therapies in cancer treatment.


Subject(s)
Breast Neoplasms/metabolism , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Line, Tumor , Female , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Heat-Shock Proteins/genetics , Humans , Phosphorylation , Protein Binding , Protein Folding , RNA, Messenger/biosynthesis
12.
Oncogene ; 32(17): 2150-60, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-22733133

ABSTRACT

The tumour suppressor p53, involved in DNA repair, cell cycle arrest and apoptosis, also inhibits blood vessel formation, that is, angiogenesis, a process strongly contributing to tumour development. The p53 gene expresses 12 different proteins (isoforms), including TAp53 (p53 (or p53α), p53ß and p53γ) and Δ133p53 isoforms (Δ133p53α, Δ133p53ß and Δ133p53γ). The Δ133p53α isoform was shown to modulate p53 transcriptional activity and is overexpressed in various human tumours. However, its role in tumour progression is still unexplored. In the present study, we examined the involvement of Δ133p53 isoforms in tumoural angiogenesis and tumour growth in the highly angiogenic human glioblastoma U87. Our data show that conditioned media from U87 cells depleted for Δ133p53 isoforms block endothelial cell migration and tubulogenesis without affecting endothelial cell proliferation in vitro. The Δ133p53 depletion in U2OS osteosarcoma cells resulted in a similar angiogenesis blockade. Furthermore, using conditioned media from U87 cells ectopically expressing each Δ133p53 isoform, we determined that Δ133p53α and Δ133p53γ but not Δ133p53ß, stimulate angiogenesis. Our in vivo data using the chicken chorio-allantoic membrane and mice xenografts establish that angiogenesis and growth of glioblastoma U87 tumours are inhibited upon depletion of Δ133p53 isoforms. By TaqMan low-density array, we show that alteration of expression ratio of Δ133p53 and TAp53 isoforms differentially regulates angiogenic gene expression with Δ133p53 isoforms inducing pro-angiogenic gene expression and repressing anti-angiogenic gene expression.


Subject(s)
Brain Neoplasms/blood supply , Glioblastoma/blood supply , Neovascularization, Pathologic/metabolism , Tumor Suppressor Protein p53/genetics , Angiogenic Proteins/genetics , Angiogenic Proteins/metabolism , Animals , Brain Neoplasms/pathology , Cattle , Cell Line, Tumor , Cell Movement , Cell Proliferation , Chick Embryo , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/metabolism , Gene Expression , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tumor Burden , Tumor Suppressor Protein p53/metabolism
13.
Oncogene ; 32(34): 4009-16, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23069659

ABSTRACT

Mutant p53 proteins accumulate to high levels in human tumors and in preneoplastic lesions in the skin and fallopian tube. However examination of tissues from mice and fish that are homozygous for mutant p53 surprisingly showed that the protein was present only at low levels except in the tumors that arose in these animals. The mutant protein did accumulate, however, following treatment with ionizing radiation in the same tissues in which the wild-type protein is induced. Here we study in detail the accumulation of mutant and wild-type p53 proteins following ionizing radiation in zebrafish embryos. We found that the mutant protein was induced by lower levels of radiation and reached higher levels than the wild-type protein. Morpholino knockdown of the zebrafish homologs of Mdm2 and Mdm4 caused dramatic accumulation of mutant p53 protein. The most remarkable results were observed by examining p53 protein levels over an extended time course. Mutant p53 protein increased and persisted for days after irradiation and this was accompanied by persistent elevation of phosphorylated H2AX (γH2AX), implying that the resolution of DNA damage signaling in these embryos is severely compromised by mutations in p53. Thus mutation in p53 results in an exaggerated and persistent damage response, which could in turn drive the process of cancer development as high levels of mutant p53 can act as an oncoprotein to drive invasion and metastasis.


Subject(s)
DNA Damage , Radiation, Ionizing , Signal Transduction/radiation effects , Tumor Suppressor Protein p53/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Blotting, Western , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/radiation effects , Eye Neoplasms/genetics , Eye Neoplasms/metabolism , Gene Knockdown Techniques , Histones/metabolism , Imidazoles/pharmacology , Immunohistochemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation/radiation effects , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/metabolism , Phosphoproteins/metabolism , Piperazines/pharmacology , Protein Binding/drug effects , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/genetics , Zebrafish/embryology , Zebrafish Proteins/genetics
14.
Cell Death Differ ; 19(5): 816-26, 2012 May.
Article in English | MEDLINE | ID: mdl-22075982

ABSTRACT

In response to stress, p53 binds and transactivates the internal TP53 promoter, thus regulating the expression of its own isoform, Δ133p53α. Here, we report that, in addition to p53, at least four p63/p73 isoforms regulate Δ133p53 expression at transcriptional level: p63ß, ΔNp63α, ΔNp63ß and ΔNp73γ. This regulation occurs through direct DNA-binding to the internal TP53 promoter as demonstrated by chromatin immunoprecipitation and the use of DNA-binding mutant p63. The promoter regions involved in the p63/p73-mediated transactivation were identified using deleted, mutant and polymorphic luciferase reporter constructs. In addition, we observed that transient expression of p53 family members modulates endogenous Δ133p53α expression at both mRNA and protein levels. We also report concomitant variation of p63 and Δ133p53 expression during keratinocyte differentiation of HaCat cells and induced pluripotent stem cells derived from mutated p63 ectodermal dysplasia patients. Finally, proliferation assays indicated that Δ133p53α isoform regulates the anti-proliferative activities of p63ß, ΔNp63α, ΔNp63ß and ΔNp73γ. Overall, this study shows a strong interplay between p53, p63 and p73 isoforms to orchestrate cell fate outcome.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Isoforms/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Proliferation , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , Humans , Nuclear Proteins/genetics , Protein Isoforms/genetics , Real-Time Polymerase Chain Reaction , Transcription Factors/genetics , Tumor Protein p73 , Tumor Suppressor Proteins/genetics
15.
J Mol Biol ; 405(3): 736-53, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21094167

ABSTRACT

Eukaryotic initiation factor (eIF)4E is overexpressed in many types of cancer such as breast, head and neck, and lung. A consequence of increased levels of eIF4E is the preferential translation of pro-tumorigenic proteins such as c-Myc, cyclin D1, and vascular endothelial growth factor. Inhibition of eIF4E is therefore a potential therapeutic target for human cancers. A novel peptide based on the eIF4E-binding peptide eIF4G1, where the α-helix was stabilized by the inclusion of α-helix inducers as shown by CD measurements, was synthesized. The helically stabilized peptide binds with an apparent K(d) of 9.43±2.57 nM, which is ∼15.7-fold more potent than the template peptide from which it is designed. The helically stabilized peptide showed significant biological activity at a concentration of 400 µM, unlike the naturally occurring eIFG1 peptide when measured in cell-based cap-dependent translational reporter and WST-1 (4-[3-(4-iodophenyl)-2-(4-nitrophenyl)-2H-5-tetrazolio]-1,3-benzene disulfonate) assays. Fusion of the template peptide and the stabilized peptide to the cell-penetrating peptide TAT produced more active but equally potent inhibitors of cap-dependent translation in cell lines. They also equally disrupted cell metabolism as measured in a WST-1 assay. Propidium iodide staining revealed that the TAT-fused, helically stabilized peptide caused more cell death than the TAT-fused eIF4G1 template peptide with substantial decreases in the G1 and G2 cell populations. Annexin-staining experiments also indicated that the TAT-fused eIF4G1 derivative peptides caused cell death by apoptosis. The results presented should offer further insight into peptidomimetics development for eIF4E.


Subject(s)
Antineoplastic Agents/chemistry , Drug Design , Eukaryotic Initiation Factor-4E/chemistry , Eukaryotic Initiation Factor-4G/chemistry , Peptidomimetics/chemistry , Recombinant Fusion Proteins/chemistry , Amino Acid Sequence , Antineoplastic Agents/metabolism , Apoptosis , Cell Line, Tumor , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4G/genetics , Gene Products, tat/chemistry , Gene Products, tat/genetics , Gene Products, tat/metabolism , Humans , Molecular Sequence Data , Peptidomimetics/metabolism , Protein Binding , Protein Biosynthesis/drug effects , Protein Structure, Secondary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
16.
Cell Death Differ ; 18(2): 248-58, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20689555

ABSTRACT

We have previously reported that the human p53 gene encodes at least nine different p53 isoforms, including Δ133p53α, which can modulate p53 transcriptional activity and apoptosis. In this study, we aimed to investigate the regulation of Δ133p53α isoform expression and its physiological role in modulating cell cycle arrest and apoptosis. We report here that in response to a low dose of doxorubicin (which induces cell cycle arrest without promoting apoptosis), p53 directly transactivates the human p53 internal promoter, inducing Δ133p53α protein expression. The induced Δ133p53α then inhibits p53-dependent apoptosis and G1 arrest without inhibiting p53-dependent G2 arrest. Therefore, endogenous Δ133p53α does not exclusively function in a dominant-negative manner toward p53, but differentially regulates cell cycle arrest and apoptosis.


Subject(s)
DNA Damage , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , Apoptosis , Base Sequence , Cell Line, Tumor , Doxorubicin/pharmacology , G1 Phase , Genes, p53 , Humans , Introns , Molecular Sequence Data , Promoter Regions, Genetic , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics
17.
Cell Death Differ ; 17(9): 1486-500, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20203688

ABSTRACT

Chemotherapeutics (e.g., aurora kinase inhibitors) designed to target proliferative cells are often nonspecific for tumor cells as normal cycling cells are also susceptible. Indeed, one of the major dose-limiting toxicities of aurora kinase inhibitors is a dangerous depletion of neutrophils in patients. In this study we proposed a strategy to selectively target p53 mutant cells while sparing normal ones. The strategy is based on the understanding that normal cells have an intact p53 pathway but not tumor cells carrying p53 mutations. Nongenotoxic activation of p53 using nutlin led to a reversible activation of G1 and G2 arrest in normal cells, which prevents them from entering mitosis, thus protecting them from the side effects of aurora kinase inhibition (VX-680), namely endoreduplication and apoptosis. Cells carrying mutant p53 are selectively killed by the nutlin/VX-680 combination, whereas p53 wild-type cells retain their proliferative capacity. The major implications drawn from these results are: (1) reversible nongenotoxic activation of p53 may be used as a strategy for the chemoprotection of normal tissues, and (2) aurora kinase inhibitors may have alleviated side effects when used in combination with nutlin-like inhibitors. We highlight the distinct roles of p53 and p73 in mediating the cellular responses to VX-680 and suggest that dual protection by p53 and p73 are needed to guard against endoreduplication and polyploidy.


Subject(s)
Apoptosis/drug effects , Imidazoles/pharmacology , Mutation/physiology , Piperazines/pharmacology , Tumor Suppressor Protein p53/genetics , Apoptosis/genetics , Aurora Kinases , Caffeine/pharmacology , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Cyclin A2/genetics , Cyclin A2/metabolism , Cyclin B1/genetics , Cyclin B1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA-Binding Proteins/genetics , G2 Phase/drug effects , Gene Expression/drug effects , Gene Expression/genetics , Humans , Imidazoles/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Proteins/genetics , Piperazines/therapeutic use , Polyploidy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , RNA, Small Interfering/genetics , Tetraploidy , Tumor Protein p73 , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics
18.
Oncogene ; 29(3): 432-41, 2010 Jan 21.
Article in English | MEDLINE | ID: mdl-19838211

ABSTRACT

It has previously been shown that ubiquitin-specific protease 2a (USP2a) is a regulator of the Mdm2/p53 pathway. USP2a binds to Mdm2 and can deubiquitinate Mdm2 without reversing Mdm2-mediated p53 ubiquitination. Overexpression of USP2a causes accumulation of Mdm2 and promotes p53 degradation. We now show that MdmX is also a substrate for USP2a. MdmX associates with USP2a independently of Mdm2. Ectopic expression of wild-type USP2a but not a catalytic mutant prevents Mdm2-mediated degradation of MdmX. This correlates with the ability of wild-type USP2a to deubiquitinate MdmX. siRNA-mediated knockdown of USP2a in NTERA-2 testicular embryonal carcinoma cells and MCF7 breast cancer cells causes destabilization of MdmX and results in a decrease in MdmX protein levels, showing that endogenous USP2a participates in the regulation of MdmX stability. The therapeutic drug, cisplatin decreases MdmX protein expression. USP2a mRNA and protein levels were also reduced after cisplatin exposure. The magnitude and time course of USP2a downregulation suggests that the reduction in USP2a levels could contribute to the decrease in MdmX expression following treatment with cisplatin. Knockdown of USP2a increases the sensitivity of NTERA-2 cells to cisplatin, raising the possibility that suppression of USP2a in combination with cisplatin may be an approach for cancer therapy.


Subject(s)
Endopeptidases/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins/metabolism , Antineoplastic Agents/pharmacology , Blotting, Western , Catalytic Domain/genetics , Cell Cycle Proteins , Cell Line, Tumor , Cisplatin/pharmacology , Down-Regulation/drug effects , Endopeptidases/genetics , Humans , Immunoprecipitation , Mutation , Nuclear Proteins/genetics , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Substrate Specificity , Transfection , Ubiquitin Thiolesterase , Ubiquitination
19.
Br J Cancer ; 98(1): 4-8, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18182973

ABSTRACT

The p53 pathway is aberrant in most human tumours with over 50% expressing mutant p53 proteins. The pathway is critically controlled by protein degradation. Here, we discuss the latest developments in the search for small molecules that can modulate p53 pathway protein stability and restore p53 activity for cancer therapy.


Subject(s)
Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Humans , Neoplasms/therapy , Proto-Oncogene Proteins c-mdm2/metabolism
20.
Oncogene ; 27(24): 3371-83, 2008 May 29.
Article in English | MEDLINE | ID: mdl-18223694

ABSTRACT

p53 missense mutant proteins commonly show increased stability compared to wild-type p53, which is thought to depend largely on the inability of mutant p53 to induce the ubiquitin ligase MDM2. However, recent work using mouse models has shown that the accumulation of mutant p53 occurs only in tumour cells, indicating that stabilization requires additional factors. To clarify the stabilization of p53 mutants in tumours, we analysed factors that affect their folding and degradation. Although all missense mutants that we studied are more stable than wild-type p53, the levels correlate with individual structural characteristics, which may be reflected in different gain-of-function properties. In the absence of Hsp90 activity, the less stable unfolded p53 mutants preferentially associate in a complex with Hsp70 and CHIP (carboxy terminus of Hsp70-interacting protein), and we show that CHIP is responsible for ubiquitination and degradation of these mutants. The demonstration of a complex interplay between Hsp90, Hsp70 and CHIP that regulate the stability of different p53 mutant proteins improves our understanding of the pro-tumorigenic effects of increased Hsp90 activity during multi-stage carcinogenesis. Understanding the roles of Hsp90, Hsp70 and CHIP in cancers may also provide an important avenue through which to target p53 to enhance treatment of human cancers.


Subject(s)
HSC70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Mutation/genetics , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Benzoquinones/pharmacology , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Fibroblasts/cytology , Fibroblasts/metabolism , HSC70 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , Humans , Immunoblotting , Immunoprecipitation , Lactams, Macrocyclic/pharmacology , Mice , Mice, Knockout , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Protein Conformation , Proto-Oncogene Proteins c-mdm2/physiology , Ubiquitin-Protein Ligases/genetics , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...