Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Chembiochem ; 21(13): 1843-1851, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32185845

ABSTRACT

We describe a molecular characterization of the interaction between the cancer-related proteins WWOX and p73. This interaction is mediated by the first of two WW domains (WW1) of WWOX and a PPXY-motif-containing region in p73. While phosphorylation of Tyr33 of WWOX and association with p73 are known to affect apoptotic activity, the quantitative effect of phosphorylation on this specific interaction is determined here for the first time. Using ITC and fluorescence anisotropy, we measured the binding affinity between WWOX domains and a p73 derived peptide, and showed that this interaction is regulated by Tyr phosphorylation of WW1. Chemical synthesis of the phosphorylated domains of WWOX revealed that the binding affinity of WWOX to p73 is decreased when WWOX is phosphorylated. This result suggests a fine-tuning of binding affinity in a differential, ligand-specific manner: the decrease in binding affinity of WWOX to p73 can free both partners to form new interactions.


Subject(s)
Tumor Protein p73/metabolism , WW Domain-Containing Oxidoreductase/metabolism , Amino Acid Motifs , Calorimetry , Fluorescence Polarization , Humans , Models, Molecular , Phosphorylation , Protein Binding , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Tumor Protein p73/chemistry , WW Domain-Containing Oxidoreductase/chemistry , WW Domain-Containing Oxidoreductase/genetics
2.
Pharmacol Res ; 162: 105245, 2020 12.
Article in English | MEDLINE | ID: mdl-33069756

ABSTRACT

p73, along with p53 and p63, belongs to the p53 family of transcription factors. Besides the p53-like tumor suppressive activities, p73 has unique roles, namely in neuronal development and differentiation. In addition, the TP73 gene is rarely mutated in tumors. This makes p73 a highly appealing therapeutic target, particularly towards cancers with a null or disrupted p53 pathway. Distinct isoforms are transcribed from the TP73 locus either with (TAp73) and without (ΔNp73) the N-terminal transactivation domain. Conversely to TA tumor suppressors, ΔN proteins exhibit oncogenic properties by inhibiting p53 and TA protein functions. As such, p73 isoforms compose a puzzled and challenging regulatory pathway. This state-of-the-art review affords an update overview on p73 structure, biological functions and pharmacological regulation. Importantly, it addresses the relevance of p73 isoforms in carcinogenesis, highlighting their potential as drug targets in anticancer therapy. A critical discussion of major pharmacological approaches to promote p73 tumor suppressive activities, with relevant survival outcomes for cancer patients, is also provided.


Subject(s)
Neoplasms/metabolism , Tumor Protein p73/metabolism , Animals , Humans , Neoplasms/drug therapy , Signal Transduction , Tumor Protein p73/chemistry , Tumor Suppressor Protein p53/metabolism
3.
J Biol Chem ; 293(51): 19546-19558, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30429221

ABSTRACT

In response to genotoxic stress, the tumor suppressor protein p73 induces apoptosis and cell cycle arrest. Despite extensive studies on p73-mediated apoptosis, little is known about the cytoplasmic apoptotic function of p73. Here, using H1299 lung cancer cells and diverse biochemical approaches, including colony formation, DNA fragmentation, GST pulldown, and apoptosis assays along with NMR spectroscopy, we show that p73 induces transcription-independent apoptosis via its transactivation domain (TAD) through a mitochondrial pathway and that this apoptosis is mediated by the interaction between p73-TAD and the anti-apoptotic protein B-cell lymphoma-extra large (Bcl-XL or BCL2L1). This binding disrupted an interaction between Bcl-XL and the pro-apoptotic protein BH3-interacting domain death agonist (Bid). In particular, we found that a 16-mer p73-TAD peptide motif (p73-TAD16) mediates transcription-independent apoptosis, accompanied by cytochrome c release from the mitochondria, by interacting with Bcl-XL Interestingly, the structure of the Bcl-XL-p73-TAD16 peptide complex revealed a novel mechanism of Bcl-XL recognition by p73-TAD. We observed that the α-helical p73-TAD16 peptide binds to a noncanonical site in Bcl-XL, comprising the BH1, BH2, and BH3 domains in an orientation opposite to those of pro-apoptotic BH3 peptides. Taken together, our results indicate that the cytoplasmic apoptotic function of p73 is mediated through a noncanonical mode of Bcl-XL recognition. This finding sheds light on a critical transcription-independent, p73-mediated mechanism for apoptosis induction, which has potential implications for anticancer therapy.


Subject(s)
Apoptosis , Cytoplasm/metabolism , Tumor Protein p73/metabolism , bcl-X Protein/metabolism , Cell Line, Tumor , Cytoplasm/pathology , Humans , Mitochondria/metabolism , Mitochondria/pathology , Models, Molecular , Protein Binding , Protein Domains , Transcription, Genetic , Tumor Protein p73/chemistry , bcl-X Protein/genetics
4.
BMC Evol Biol ; 17(1): 177, 2017 08 03.
Article in English | MEDLINE | ID: mdl-28774266

ABSTRACT

BACKGROUND: The p53 signalling pathway, which controls cell fate, has been extensively studied due to its prominent role in tumor development. The pathway includes the tumor supressor protein p53, its vertebrate paralogs p63 and p73, and their negative regulators MDM2 and MDM4. The p53/p63/p73-MDM system is ancient and can be traced in all extant animal phyla. Despite this, correct phylogenetic trees including both vertebrate and invertebrate species of the p53/p63/p73 and MDM families have not been published. RESULTS: Here, we have examined the evolution of the p53/p63/p73 protein family with particular focus on the p53/p63/p73 transactivation domain (TAD) and its co-evolution with the p53/p63/p73-binding domain (p53/p63/p73BD) of MDM2. We found that the TAD and p53/p63/p73BD share a strong evolutionary connection. If one of the domains of the protein is lost in a phylum, then it seems very likely to be followed by loss of function by the other domain as well, and due to the loss of function it is likely to eventually disappear. By focusing our phylogenetic analysis to p53/p63/p73 and MDM proteins from phyla that retain the interaction domains TAD and p53/p63/p73BD, we built phylogenetic trees of p53/p63/p73 and MDM based on both vertebrate and invertebrate species. The trees follow species evolution and contain a total number of 183 and 98 species for p53/p63/p73 and MDM, respectively. We also demonstrate that the p53/p63/p73 and MDM families result from whole genome duplications. CONCLUSIONS: The signaling pathway of the TAD and p53/p63/p73BD in p53/p63/p73 and MDM, respectively, dates back to early metazoan time and has since then tightly co-evolved, or disappeared in distinct lineages.


Subject(s)
Evolution, Molecular , Proto-Oncogene Proteins c-mdm2/genetics , Signal Transduction , Tumor Suppressor Protein p53/genetics , Amino Acid Sequence , Animals , Gene Duplication , Humans , Phosphorylation , Phylogeny , Protein Binding , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-mdm2/chemistry , Species Specificity , Tumor Protein p73/chemistry , Tumor Protein p73/genetics , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
5.
Arch Biochem Biophys ; 619: 1-9, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28235466

ABSTRACT

The alpha splice variant of p73 (p73α), a homologue of the tumour suppressor p53, has at its C terminus a sterile alpha motif (SAM); this domain, SAMp73, is involved in lipid binding and it is thought to mediate in protein-protein interactions. As SAMp73 is a 68-residue-long helical bundle, it could be a good model to study the (2,2,2-trifluoroethanol) TFE-induced conformational transitions of α-helical proteins. Furthermore, as SAMp73 binds to lipids through a well-known polypeptide patch, we can test whether TFE is a good mimic of lipids and membranes. To address those questions, we used several biophysical probes, namely, fluorescence, circular dichroism, 1D, 2D and 3D-NMR spectroscopies, and dynamic light scattering. The TFE-induced conformational transition of SAMp73 was complex, involving several species as detected by the biophysical probes. The last TFE-induced transition occurred at a concentration of TFE of ∼20% (v/v), where the protein lost its compactness. None of those TFE-induced species accumulated during the two-state folding of SAMp73 in aqueous solution. The final state at 40% TFE was highly helical, but its structure was not rigid. For SAMp73, TFE did not properly mimic a membrane-like environment, since at very low TFE concentrations, other residues, together with those known to interact with lipids, were also affected by the co-solvent. Comparison with studies on isolated peptides, comprising the helical regions of SAMp73, suggests that peptides were good models of the intact protein in TFE.


Subject(s)
Sterile Alpha Motif/genetics , Trifluoroethanol/chemistry , Tumor Protein p73/chemistry , Cell Membrane/metabolism , Circular Dichroism , Humans , Light , Magnetic Resonance Spectroscopy , Peptides/chemistry , Protein Binding , Protein Domains , Protein Folding , Protein Interaction Mapping , Protein Structure, Secondary , Scattering, Radiation , Solvents/chemistry , Spectrometry, Fluorescence
6.
Biochim Biophys Acta Gen Subj ; 1865(7): 129914, 2021 07.
Article in English | MEDLINE | ID: mdl-33872756

ABSTRACT

BACKGROUND: Plakophilin 1 (PKP1) is a component of desmosomes, which are key structural components for cell-cell adhesion, and can also be found in other cell locations. The p53, p63 and p73 proteins belong to the p53 family of transcription factors, playing crucial roles in tumour suppression. The α-splice variant of p73 (p73α) has at its C terminus a sterile alpha motif (SAM); such domain, SAMp73, is involved in the interaction with other macromolecules. METHODS: We studied the binding of SAMp73 with the armadillo domain of PKP1 (ARM-PKP1) in the absence and the presence of 100 mM NaCl, by using several biophysical techniques, namely fluorescence, far-ultraviolet circular dichroism (CD), nuclear magnetic resonance (NMR), isothermal titration calorimetry (ITC), and molecular docking and simulations. RESULTS: Association was observed between the two proteins, with a dissociation constant of ~5 µM measured by ITC and fluorescence in the absence of NaCl. The binding region of SAMp73 involved residues of the so-called "middle-loop-end-helix" binding region (i.e., comprising the third helix, together with the C terminus of the second one, and the N-cap of the fourth), as shown by 15N, 1H- HSQC-NMR spectra. Molecular modelling provided additional information on the possible structure of the binding complex. CONCLUSIONS: This newly-observed interaction could have potential therapeutic relevance in the tumour pathways where PKP1 is involved, and under conditions when there is a possible inactivation of p53. GENERAL SIGNIFICANCE: The discovery of the binding between SAMp73 and ARM-PKP1 suggests a functional role for their interaction, including the possibility that SAMp73 could assist PKP1 in signalling pathways.


Subject(s)
Armadillo Domain Proteins/metabolism , Plakophilins/metabolism , Protein Interaction Domains and Motifs , Sterile Alpha Motif , Tumor Protein p73/metabolism , Armadillo Domain Proteins/chemistry , Humans , Models, Molecular , Molecular Docking Simulation , Plakophilins/chemistry , Protein Binding , Protein Conformation , Protein Domains , Tumor Protein p73/chemistry
7.
Cell Cycle ; 19(14): 1833-1845, 2020 07.
Article in English | MEDLINE | ID: mdl-32584647

ABSTRACT

The p53 family transcriptional factor p73 plays a pivotal role in development. Ablation of p73 results in severe neurodevelopmental defects, chronic infections, inflammation and infertility. In addition to this, Trp73-\- mice display severe alteration in the ciliated epithelial lining and the full-length N-terminal isoform TAp73 has been implicated in the control of multiciliogenesis transcriptional program. With our recently generated Trp73Δ13/Δ13 mouse model, we interrogate the physiological role of p73 C-terminal isoforms in vivo. Trp73Δ13/Δ13 mice lack exon 13 in Trp73 gene, producing an ectopic switch from the C-terminal isoforms p73α to p73ß. Trp73Δ13/Δ13 mice show a pattern of expression of TAp73 comparable to the wild-type littermates, indicating that the α to ß switch does not significantly alter the expression of the gene in this cell type. Moreover, Trp73Δ13/Δ13 do not display any significant alteration in the airway ciliated epithelium, suggesting that in this context p73ß can fully substitute the function of the longer isoform p73α. Similarly, Trp73Δ13/Δ13 ciliated epithelium of the brain ependyma also does appear defective. In this district however expression of TAp73 is not detectable, indicating that expression of the gene might be compensated by alternative mechanisms. Overall our work indicates that C-terminus p73 is dispensable for the multiciliogenesis program and suggests a possible tissue-specific effect of p73 alternative splicing.


Subject(s)
Cilia/metabolism , Organogenesis , Tumor Protein p73/chemistry , Tumor Protein p73/metabolism , Animals , Cell Line , Ependyma/metabolism , Epithelium/metabolism , Epithelium/ultrastructure , Mice , Mitochondria/metabolism , Mitochondria/ultrastructure , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Structure-Activity Relationship , Trachea/metabolism
8.
Nat Commun ; 11(1): 1475, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32193462

ABSTRACT

Inter-individual differences in T helper (Th) cell responses affect susceptibility to infectious, allergic and autoimmune diseases. To identify factors contributing to these response differences, here we analyze in vitro differentiated Th1 cells from 16 inbred mouse strains. Haplotype-based computational genetic analysis indicates that the p53 family protein, p73, affects Th1 differentiation. In cells differentiated under Th1 conditions in vitro, p73 negatively regulates IFNγ production. p73 binds within, or upstream of, and modulates the expression of Th1 differentiation-related genes such as Ifng and Il12rb2. Furthermore, in mouse experimental autoimmune encephalitis, p73-deficient mice have increased IFNγ production and less disease severity, whereas in an adoptive transfer model of inflammatory bowel disease, transfer of p73-deficient naïve CD4+ T cells increases Th1 responses and augments disease severity. Our results thus identify p73 as a negative regulator of the Th1 immune response, suggesting that p73 dysregulation may contribute to susceptibility to autoimmune disease.


Subject(s)
Cell Differentiation , Th1 Cells/cytology , Th1 Cells/metabolism , Tumor Protein p73/metabolism , Alleles , Animals , Base Sequence , Binding Sites , Colitis/pathology , DNA/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Deletion , Gene Expression Regulation , Interferon-gamma/metabolism , Mice , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Binding , Protein Domains , Severity of Illness Index , Tumor Protein p73/chemistry , Tumor Protein p73/deficiency , Tumor Protein p73/genetics , Tumor Suppressor Protein p53/metabolism
9.
Sci Rep ; 9(1): 950, 2019 01 30.
Article in English | MEDLINE | ID: mdl-30700826

ABSTRACT

More than 50% of colon cancers bear mutations in p53, one of the most important tumor suppressors, and its family members p63 or p73 are expected to contribute to inhibiting the progression of colon cancers. The AP2 family also acts as a tumor suppressor. Here we found that p73 and AP2 are able to activate NEU4, a neuraminidase gene, which removes the terminal sialic acid residues from cancer-associated glycans. Under serum starvation, NEU4 was up-regulated and one of the NEU4 target glycans, sialyl Lewis X, was decreased, whereas p73 and AP2 were up-regulated. Sialyl Lewis X levels were not, however, decreased under starvation conditions in p73- or AP2-knockdown cells. p53 and AP2 underwent protein-protein interactions, exerting synergistic effects to activate p21, and interaction of p53 with AP2 was lost in cells expressing the L350P mutation of p53. The homologous residues in p63 and p73 are L423 and L377, respectively. The synergistic effect of p53/p63 with AP2 to activate genes was lost with the L350P/L423P mutation in p53/p63, but p73 bearing the L377P mutation was able to interact with AP2 and exerted its normal synergistic effects. We propose that p73 and AP2 synergistically activate the NEU4 promoter in colon cancer cells.


Subject(s)
Colonic Neoplasms/genetics , Neuraminidase/genetics , Promoter Regions, Genetic , Transcription Factor AP-2/metabolism , Tumor Protein p73/metabolism , Amino Acid Sequence , Base Sequence , Cell Line, Tumor , Down-Regulation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Models, Biological , Neuraminidase/metabolism , Protein Binding , Protein Interaction Mapping , Response Elements/genetics , Sialyl Lewis X Antigen/metabolism , Transcription Factor AP-2/genetics , Tumor Protein p73/chemistry , Tumor Protein p73/genetics , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism
10.
Phytomedicine ; 55: 179-190, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30668428

ABSTRACT

BACKGROUND: Epithelial-to-mesenchymal transition (EMT), a key step in oral cancer progression, is associated with invasion, metastasis, and therapy resistance, thus targeting the EMT represents a critical therapeutic strategy for the treatment of oral cancer metastasis. Our previous study showed that Abrus agglutinin (AGG), a plant lectin, induces both intrinsic and extrinsic apoptosis to activate the tumor inhibitory mechanism. OBJECTIVE: This study aimed to investigate the role of AGG in modulating invasiveness and stemness through EMT inhibition for the development of antineoplastic agents against oral cancer. METHODS: The EMT- and stemness-related proteins were studied in oral cancer cells using Western blot analysis and fluorescence microscopy. The potential mechanisms of Snail downregulation through p73 activation in FaDu cells were evaluated using Western blot analysis, immunoprecipitation, confocal microscopy, and molecular docking analysis. Immunohistochemical staining of the tumor samples of AGG-treated FaDu-xenografted nude mice was performed. RESULTS: At the molecular level, AGG-induced p73 suppressed Snail expression, leading to EMT inhibition in FaDu cells. Notably, AGG promoted the translocation of Snail from the nucleus to the cytoplasm in FaDu cells and triggered its degradation through ubiquitination. In this setting, AGG inhibited the interaction between Snail and p73 in FaDu cells, resulting in p73 activation and EMT inhibition. Moreover, in epidermal growth factor (EGF)-stimulated FaDu cells, AGG abolished the upregulation of extracellular signal-regulated kinase (ERK)1/2 that plays a pivotal role in the upregulation of Snail to regulate the EMT phenotypes. In immunohistochemistry analysis, FaDu xenografts from AGG-treated mice showed decreased expression of Snail, SOX2, and vimentin and increased expression of p73 and E-cadherin compared with the control group, confirming EMT inhibition as part of its anticancer efficacy against oral cancer. CONCLUSION: In summary, AGG stimulates p73 in restricting EGF-induced EMT, invasiveness, and stemness by inhibiting the ERK/Snail pathway to facilitate the development of alternative therapeutics for oral cancer.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Mouth Neoplasms/drug therapy , Plant Lectins/pharmacology , Snail Family Transcription Factors/metabolism , Tumor Protein p73/metabolism , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Epidermal Growth Factor/metabolism , Humans , Mice, Nude , Molecular Docking Simulation , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Plant Lectins/chemistry , Snail Family Transcription Factors/chemistry , Snail Family Transcription Factors/genetics , Tumor Protein p73/chemistry , Tumor Protein p73/genetics , Ubiquitination , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
11.
J Mol Biol ; 430(13): 1829-1838, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29733853

ABSTRACT

p73 (encoded by TP73 gene) is a p53 related protein that functions as a transcriptional factor. Similarly to p53, following DNA damage, p73 is stabilized and activated and controls expression of target genes that are involved in the regulation of cycle arrest and apoptosis. However, great complexity to the function of this gene is given by the wide range of its non-tumor-related roles, which include neurological development, ciliogenesis and fertility. From the structural point of view, p73 displays an intricate range of regulations because it can be expressed both as an N-terminally deleted dominant-negative isoforms and as multiple alternatively spliced C-terminal isoforms, which can include or not a sterile alpha motif domain. More is known about the functions of the N-terminal isoforms of p73 (TAp73 and ΔNp73) and their opposing pro- and anti-apoptotic roles, whereas the functional differences of the distinct C-terminal splice forms of p73 are very far away from been defined. Here we summarize the current available literature regarding p73 C-terminal isoforms and the contribution of the sterile alpha motif domain to p73 function, trying to provide an unified view in this complex and sometime controversial field. Current data indicate that the full-length, TAp73α, is the major, if not the exclusive, isoform detected in physiological systems, indicating that detailed spatio-temporal expression analysis and functional studies are highly demanded to support a physiological role for the p73 alternative splicing. With this article, we also aim to emphasize the need to further investigation on the topic, refocusing the attention on what we believe are the most relevant unanswered questions.


Subject(s)
Alternative Splicing , Tumor Protein p73/chemistry , Tumor Protein p73/metabolism , Cell Death , Cell Differentiation , Cell Proliferation , Humans , Protein Domains , Tumor Protein p73/genetics
12.
Cell Death Differ ; 25(1): 144-153, 2018 01.
Article in English | MEDLINE | ID: mdl-29077094

ABSTRACT

The p53 family of transcription factors (p53, p63 and p73) covers a wide range of functions critical for development, homeostasis and health of mammals across their lifespan. Beside the well-established tumor suppressor role, recent evidence has highlighted novel non-oncogenic functions exerted by p73. In particular, p73 is required for multiciliated cell (MCC) differentiation; MCCs have critical roles in brain and airways to move fluids across epithelial surfaces and to transport germ cells in the reproductive tract. This novel function of p73 provides a unifying cellular mechanism for the disparate inflammatory and immunological phenotypes of p73-deficient mice. Indeed, mice with Trp73 deficiency suffer from hydrocephalus, sterility and chronic respiratory tract infections due to profound defects in ciliogenesis and complete loss of mucociliary clearance since MCCs are essential for cleaning airways from inhaled pollutants, pathogens and allergens. Cross-species genomic analyses and functional rescue experiments identify TAp73 as the master transcriptional integrator of ciliogenesis, upstream of previously known central nodes. In addition, TAp73 shows a significant ability to regulate cellular metabolism and energy production through direct transcriptional regulation of several metabolic enzymes, such as glutaminase-2 and glucose-6 phosphate dehydrogenase. This recently uncovered role of TAp73 in the regulation of cellular metabolism strongly affects oxidative balance, thus potentially influencing all the biological aspects associated with p73 function, including development, homeostasis and cancer. Although through different mechanisms, p63 isoforms also contribute to regulation of cellular metabolism, thus indicating a common route used by all family members to control cell fate. At the structural level, the complexity of p73's function is further enhanced by its ability to form heterotetramers with some p63 isoforms, thus indicating the existence of an intrafamily crosstalk that determines the global outcome of p53 family function. In this review, we have tried to summarize all the recent evidence that have emerged on the novel non-oncogenic roles of p73, in an attempt to provide a unified view of the complex function of this gene within its family.


Subject(s)
Cilia/physiology , Tumor Protein p73/physiology , Amino Acids/physiology , Animals , Axoneme/physiology , Cilia/ultrastructure , Epidermis/growth & development , Humans , Metabolism , Mice , Oxidative Stress , Respiratory System/ultrastructure , Transcription Factors/physiology , Transcription, Genetic , Tumor Protein p73/chemistry , Tumor Protein p73/genetics
13.
PLoS One ; 13(4): e0195835, 2018.
Article in English | MEDLINE | ID: mdl-29668749

ABSTRACT

p73 is a member of the p53 protein family and has essential functions in several signaling pathways involved in development, differentiation, DNA damage responses and cancer. As a transcription factor, p73 achieves these functions by binding to consensus DNA sequences and p73 shares at least partial target DNA binding sequence specificity with p53. Transcriptional activation by p73 has been demonstrated for more than fifty p53 targets in yeast and/or human cancer cell lines. It has also been shown previously that p53 binding to DNA is strongly dependent on DNA topology and the presence of inverted repeats that can form DNA cruciforms, but whether p73 transcriptional activity has similar dependence has not been investigated. Therefore, we evaluated p73 binding to a set of p53-response elements with identical theoretical binding affinity in their linear state, but different probabilities to form extra helical structures. We show by a yeast-based assay that transactivation in vivo correlated more with the relative propensity of a response element to form cruciforms than to its expected in vitro DNA binding affinity. Structural features of p73 target sites are therefore likely to be an important determinant of its transactivation function.


Subject(s)
Binding Sites , Inverted Repeat Sequences , Tumor Protein p73/metabolism , Base Sequence , Humans , Nucleic Acid Conformation , Protein Binding , Transcriptional Activation , Tumor Protein p73/chemistry , Tumor Protein p73/genetics , Tumor Suppressor Protein p53/metabolism , Yeasts/genetics , Yeasts/metabolism
14.
Sci Signal ; 11(524)2018 04 03.
Article in English | MEDLINE | ID: mdl-29615516

ABSTRACT

The transcription factors p53 and p73 are critical to the induction of apoptotic cell death, particularly in response to cell stress that activates c-Jun N-terminal kinase (JNK). Mutations in the DNA-binding domain of p53, which are commonly seen in cancers, result in conformational changes that enable p53 to interact with and inhibit p73, thereby suppressing apoptosis. In contrast, wild-type p53 reportedly does not interact with p73. We found that JNK-mediated phosphorylation of Thr81 in the proline-rich domain (PRD) of p53 enabled wild-type p53, as well as mutant p53, to form a complex with p73. Structural algorithms predicted that phosphorylation of Thr81 exposes the DNA-binding domain in p53 to enable its binding to p73. The dimerization of wild-type p53 with p73 facilitated the expression of apoptotic target genes [such as those encoding p53-up-regulated modulator of apoptosis (PUMA) and Bcl-2-associated X protein (BAX)] and, subsequently, the induction of apoptosis in response to JNK activation by cell stress in various cells. Thus, JNK phosphorylation of mutant and wild-type p53 promotes the formation of a p53/p73 complex that determines cell fate: apoptosis in the context of wild-type p53 or cell survival in the context of the mutant. These findings refine our current understanding of both the mechanistic links between p53 and p73 and the functional role for Thr81 phosphorylation.


Subject(s)
Apoptosis , JNK Mitogen-Activated Protein Kinases/metabolism , Tumor Protein p73/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Binding Sites/genetics , Cell Line, Tumor , Cell Survival , Humans , JNK Mitogen-Activated Protein Kinases/chemistry , JNK Mitogen-Activated Protein Kinases/genetics , Models, Molecular , Mutation , Phosphorylation , Protein Binding , Protein Conformation , Signal Transduction , Tumor Protein p73/chemistry , Tumor Protein p73/genetics , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics
15.
Structure ; 26(8): 1091-1100.e4, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30099987

ABSTRACT

Despite high sequence homology among the p53 family members, the regulation of their transactivation potential is based on strikingly different mechanisms. Previous studies revealed that the activity of TAp63α is regulated via an autoinhibitory mechanism that keeps inactive TAp63α in a dimeric conformation. While all p73 isoforms are constitutive tetramers, their basal activity is much lower compared with tetrameric TAp63. We show that the dimeric state of TAp63α not only reduces DNA binding affinity, but also suppresses interaction with the acetyltransferase p300. Exchange of the transactivation domains is sufficient to transfer the regulatory characteristics between p63 and p73. Structure determination of the transactivation domains of p63 and p73 in complex with the p300 Taz2 domain further revealed that, in contrast to p53 and p73, p63 has a single transactivation domain. Sequences essential for stabilizing the closed dimer of TAp63α have evolved into a second transactivation domain in p73 and p53.


Subject(s)
DNA/chemistry , E1A-Associated p300 Protein/chemistry , Transcription Factors/chemistry , Transcriptional Activation , Tumor Protein p73/chemistry , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Proteins/chemistry , Amino Acid Sequence , Binding Sites , Cell Line, Tumor , Cloning, Molecular , DNA/genetics , DNA/metabolism , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Models, Molecular , Neurons , Osteoblasts , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Thermodynamics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Protein p73/genetics , Tumor Protein p73/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
16.
Article in English | MEDLINE | ID: mdl-27091942

ABSTRACT

The family of the p53 tumor suppressive transcription factors includes p73 and p63 in addition to p53 itself. Given the high degree of amino-acid-sequence homology and structural organization shared by the p53 family members, they display some common features (i.e., induction of cell death, cell-cycle arrest, senescence, and metabolic regulation in response to cellular stress) as well as several distinct properties. Here, we describe the structural evolution of the family members with recent advances on the molecular dynamic studies of p53 itself. A crucial role of the carboxy-terminal domain in regulating the properties of the DNA-binding domain (DBD) supports an induced-fit mechanism, in which the binding of p53 on individual promoters is preferentially regulated by the KOFF over KON.


Subject(s)
Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Binding Sites , Humans , Molecular Dynamics Simulation , Promoter Regions, Genetic , Trans-Activators/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Tumor Protein p73/chemistry , Tumor Protein p73/genetics , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
17.
Sci Rep ; 7: 44532, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28291250

ABSTRACT

Disease associated genetic variations often cause intracellular enzyme inactivation, dysregulation and instability. However, allosteric communication of mutational effects to distant functional sites leading to loss-of-function remains poorly understood. We characterize here interdomain site-to-site communication by which a common cancer-associated single nucleotide polymorphism (c.C609T/p.P187S) reduces the activity and stability in vivo of NAD(P)H:quinone oxidoreductase 1 (NQO1). NQO1 is a FAD-dependent, two-domain multifunctional stress protein acting as a Phase II enzyme, activating cancer pro-drugs and stabilizing p53 and p73α oncosuppressors. We show that p.P187S causes structural and dynamic changes communicated to functional sites far from the mutated site, affecting the FAD binding site located at the N-terminal domain (NTD) and accelerating proteasomal degradation through dynamic effects on the C-terminal domain (CTD). Structural protein:protein interaction studies reveal that the cancer-associated polymorphism does not abolish the interaction with p73α, indicating that oncosuppressor destabilization largely mirrors the low intracellular stability of p.P187S. In conclusion, we show how a single disease associated amino acid change may allosterically perturb several functional sites in an oligomeric and multidomain protein. These results have important implications for the understanding of loss-of-function genetic diseases and the identification of novel structural hot spots as targets for pharmacological intervention.


Subject(s)
NAD(P)H Dehydrogenase (Quinone)/genetics , Neoplasms/genetics , Protein Conformation , Tumor Protein p73/genetics , Allosteric Regulation/genetics , Flavin-Adenine Dinucleotide/chemistry , Flavin-Adenine Dinucleotide/genetics , Genetic Predisposition to Disease , Humans , Mutation , NAD(P)H Dehydrogenase (Quinone)/chemistry , Neoplasms/pathology , Polymorphism, Single Nucleotide , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/genetics , Protein Binding , Protein Domains/genetics , Protein Interaction Domains and Motifs/genetics , Tumor Protein p73/chemistry , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics
18.
Biomol NMR Assign ; 10(1): 49-51, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26294377

ABSTRACT

p53, p63, p73 family of proteins are transcription factors with crucial roles in regulating cellular processes such apoptosis, proliferation, differentiation, and DNA damage response. The three family members have both overlapping and unique biological functions. Sequence and structural homology are greatest in the DNA binding domains (DBD), which is the site of the majority of p53 mutations. Structurally unstable p53 DBD mutants can associate with themselves or p63 and p73 DBDs, impeding tumor suppressor functions. Evidence suggests that these proteins associate to form amyloid-like oligomers and fibrils through an aggregation-prone sequence within the DBDs. Despite having high sequence and structure similarities, p63 and p73 DBDs appear to have considerably lower tendencies to be incorporated into p53 aggregates, relative to p53. The backbone resonance assignments of p73 DBD reported here complement those previously reported for p53 and p63, allowing comparisons and providing molecular insights into their biological functions and roles in aggregation and tumor development.


Subject(s)
DNA/metabolism , Nuclear Magnetic Resonance, Biomolecular , Tumor Protein p73/chemistry , Tumor Protein p73/metabolism , Humans , Protein Binding , Protein Domains
19.
Cell Death Differ ; 23(12): 1952-1960, 2016 12.
Article in English | MEDLINE | ID: mdl-27447112

ABSTRACT

The high percentage of p53 missense mutations found in cancer has been attributed to mutant acquired oncogenic gain of functions. Different aspects of these tumour-promoting functions are caused by repression of the transcriptional activity of p53 family members p63 and p73. A subset of frequently occurring p53 mutations results in thermodynamic destabilisation of the DNA-binding domain (DBD) rendering this domain highly unstable. These conformational mutants (such as p53R175H) have been suggested to directly bind to p63 and p73 via a co-aggregation mechanism mediated by their DBDs. Although the DBDs of p63 and p73 are in fact not sufficient for the interaction as shown previously, we demonstrate here that the transactivation inhibitory (TI) domains within the α-isoform-specific C termini of p63 and p73 are essential for binding to p53R175H. Hence, the closed dimeric conformation of inactive TAp63α that renders the TI domain inaccessible prevents efficient interaction. We further show that binding to p53R175H correlates with an intrinsic aggregation propensity of the tetrameric α-isoforms conferred by an openly accessible TI domain again supporting interaction via a co-aggregation mechanism.


Subject(s)
Mutant Proteins/metabolism , Protein Aggregates , Transcription Factors/chemistry , Transcription Factors/metabolism , Tumor Protein p73/chemistry , Tumor Protein p73/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Cell Line, Tumor , Humans , Models, Biological , Mutant Proteins/chemistry , Peptides/metabolism , Protein Binding , Protein Domains , Protein Multimerization
20.
Cell Death Differ ; 23(12): 1930-1940, 2016 12.
Article in English | MEDLINE | ID: mdl-27716744

ABSTRACT

Members of the p53 tumor-suppressor family are expressed as multiple isoforms. Isoforms with an N-terminal transactivation domain are transcriptionally active, while those ones lacking this domain often inhibit the transcriptional activity of other family members. In squamous cell carcinomas, the high expression level of ΔNp63α inhibits the tumor-suppressor function of TAp73ß. This can in principle be due to blocking of the promoter or by direct interaction between both proteins. p63 and p73 can hetero-oligomerize through their tetramerization domains and a hetero-tetramer consisting of two p63 and two p73 molecules is thermodynamically more stable than both homo-tetramers. Here we show that cells expressing both p63 and p73 exist in mouse epidermis and hair follicle and that hetero-tetramer complexes can be detected by immunoprecipitation in differentiating keratinocytes. Through structure determination of the hetero-tetramer, we reveal why this hetero-tetramer is the thermodynamically preferred species. We have created mutants that exclusively form either hetero-tetramers or homo-tetramers, allowing to investigate the function of these p63/p73 hetero-tetramers. Using these tools, we show that inhibition of TAp73ß in squamous cell carcinomas is due to promoter squelching and not direct interaction.


Subject(s)
Phosphoproteins/chemistry , Phosphoproteins/metabolism , Protein Multimerization , Trans-Activators/chemistry , Trans-Activators/metabolism , Tumor Protein p73/chemistry , Tumor Protein p73/metabolism , Amino Acid Sequence , Animals , Humans , Mice , Mutation/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Salts/chemistry , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL