Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.626
Filter
1.
Proc Natl Acad Sci U S A ; 121(21): e2318591121, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38739802

ABSTRACT

The transcription factor p73, a member of the p53 tumor-suppressor family, regulates cell death and also supports tumorigenesis, although the mechanistic basis for the dichotomous functions is poorly understood. We report here the identification of an alternate transactivation domain (TAD) located at the extreme carboxyl (C) terminus of TAp73ß, a commonly expressed p73 isoform. Mutational disruption of this TAD significantly reduced TAp73ß's transactivation activity, to a level observed when the amino (N)-TAD that is similar to p53's TAD, is mutated. Mutation of both TADs almost completely abolished TAp73ß's transactivation activity. Expression profiling highlighted a unique set of targets involved in extracellular matrix-receptor interaction and focal adhesion regulated by the C-TAD, resulting in FAK phosphorylation, distinct from the N-TAD targets that are common to p53 and are involved in growth inhibition. Interestingly, the C-TAD targets are also regulated by the oncogenic, amino-terminal-deficient DNp73ß isoform. Consistently, mutation of C-TAD reduces cellular migration and proliferation. Mechanistically, selective binding of TAp73ß to DNAJA1 is required for the transactivation of C-TAD target genes, and silencing DNAJA1 expression abrogated all C-TAD-mediated effects. Taken together, our results provide a mechanistic basis for the dichotomous functions of TAp73 in the regulation of cellular growth through its distinct TADs.


Subject(s)
Cell Proliferation , Protein Domains , Transcriptional Activation , Tumor Protein p73 , Tumor Protein p73/metabolism , Tumor Protein p73/genetics , Humans , Cell Movement/genetics , Mutation , Cell Line, Tumor , Protein Isoforms/metabolism , Protein Isoforms/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Phosphorylation , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics
2.
J Pathol ; 263(3): 328-337, 2024 07.
Article in English | MEDLINE | ID: mdl-38629257

ABSTRACT

Peritoneal metastasis of colorectal origin appears in ~10-15% of patients at the time of diagnosis and in 30-40% of cases with disease progression. Locoregional spread through the peritoneum is considered stage IVc and is associated with a poor prognosis. The development of a regional therapeutic strategy based on cytoreductive surgery, and hyperthermic intra-abdominal chemotherapy has significantly altered the course of the disease. Although recent evidence supports the benefits of cytoreductive surgery, the benefits of hyperthermic intra-abdominal chemotherapy are, however, still a matter of debate. Understanding the molecular alterations underlying the disease is crucial for developing new therapeutic strategies. Here, we evaluated the involvement in peritoneal dissemination of the oncogenic isoform of TP73, ΔNp73, and its effector targets in in vitro and mouse models, and in 30 patients diagnosed with colorectal peritoneal metastasis. In an orthotopic mouse model, we observed that tumor cells overexpressing ΔNp73 present a higher avidity for the peritoneum and that extracellular vesicles secreted by ΔNp73-upregulating tumor cells enhance their dissemination. In addition, we identified that tumor cells overexpressing ΔNp73 present with dysregulation of genes associated with an epithelial/mesothelial-to-mesenchymal transition (MMT) and that mesothelial cells exposed to the conditioned medium of tumor cells with upregulated ΔNp73 present a mesenchymal phenotype. Lastly, ΔNp73 and its effector target RNAs were dysregulated in our patient series, there were positive correlations between ΔNp73 and its effector targets, and MSN and ITGB4 (ΔNp73 effectors) predicted patient survival. In conclusion, ΔNp73 and its effector targets are involved in the peritoneal dissemination of colorectal cancer and predict patient survival. The promotion of the EMT/MMT and modulation of the adhesion capacity in colorectal cancer cells might be the mechanisms triggered by ΔNp73. Remarkably, ΔNp73 protein is a druggable protein and should be the focus of future studies. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Colorectal Neoplasms , Epithelial-Mesenchymal Transition , Peritoneal Neoplasms , Tumor Protein p73 , Humans , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/pathology , Animals , Male , Female , Tumor Protein p73/metabolism , Tumor Protein p73/genetics , Middle Aged , Aged , Mice , Gene Expression Regulation, Neoplastic , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor
3.
Proc Natl Acad Sci U S A ; 119(22): e2123202119, 2022 May 31.
Article in English | MEDLINE | ID: mdl-35617425

ABSTRACT

p73, a p53 family member, undergoes alternative splicing at the 3' end to produce multiple isoforms, but their expression and activity are largely unknown. Thus, CRISPR was used to knock out exon 12 (E12) in human cancer cell lines and mice, leading to isoform switch from p73α to isoform p73α1. We found that p73α1 is naturally expressed and induced by DNA damage. We also found that knockout of E12 suppresses cell growth and migration in H1299 and MIA PaCa-2 cells and promotes cellular senescence in mouse embryonic fibroblasts. Similarly, ectopic expression of p73α1 suppresses cell proliferation, whereas knockdown of p73α1 restores the cell proliferative and migratory capacities of E12−/− cells. Consistently, we found that E12+/− mice are not prone to spontaneous tumors. Instead, E12+/− mice are prone to systemic inflammation and exhibit elevated TNFα expression in inflamed tissues. Moreover, we found that Notch1, a master regulator of the inflammatory response, is regulated by p73α1 and highly expressed in E12−/− cells and inflamed E12+/− mouse tissues. Furthermore, through knockdown of p73α1 and/or Notch1 in E12−/− cells, we found that Notch1 is necessary for p73α1-mediated growth suppression. Together, these data suggest that p73α1 plays a critical role in tumor suppression and the inflammatory response via Notch1.


Subject(s)
Genes, Tumor Suppressor , Inflammation , Neoplasms , Receptor, Notch1 , Tumor Protein p73 , Animals , Cell Line, Tumor , DNA Damage , Exons/genetics , Gene Knockout Techniques , Humans , Inflammation/genetics , Mice , Mice, Knockout , Neoplasms/genetics , Neoplasms/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Tumor Protein p73/genetics , Tumor Protein p73/metabolism
4.
Genes Dev ; 31(17): 1715-1716, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28982757

ABSTRACT

Medulloblastomas are among the most common malignant brain cancers in the pediatric population and consist of at least four distinct subgroups with unique molecular and genetic features and clinical outcomes. In this issue of Genes & Development, Niklison-Chirou and colleagues (pp. 1738-1753) identify the p53 family member and p73 isoform TAp73 as a crucial factor causing glutamine addiction in aggressive medulloblastomas. Their findings pave the way for the use of glutamine restriction as an adjuvant treatment for TAp73-expressing medulloblastomas.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Child , DNA-Binding Proteins , Diet , Glutamine , Humans , Nuclear Proteins , Tumor Protein p73 , Tumor Suppressor Proteins
5.
Genes Dev ; 31(17): 1738-1753, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28971956

ABSTRACT

Medulloblastoma is the most common solid primary brain tumor in children. Remarkable advancements in the understanding of the genetic and epigenetic basis of these tumors have informed their recent molecular classification. However, the genotype/phenotype correlation of the subgroups remains largely uncharacterized. In particular, the metabolic phenotype is of great interest because of its druggability, which could lead to the development of novel and more tailored therapies for a subset of medulloblastoma. p73 plays a critical role in a range of cellular metabolic processes. We show overexpression of p73 in a proportion of non-WNT medulloblastoma. In these tumors, p73 sustains cell growth and proliferation via regulation of glutamine metabolism. We validated our results in a xenograft model in which we observed an increase in survival time in mice on a glutamine restriction diet. Notably, glutamine starvation has a synergistic effect with cisplatin, a component of the current medulloblastoma chemotherapy. These findings raise the possibility that glutamine depletion can be used as an adjuvant treatment for p73-expressing medulloblastoma.


Subject(s)
Cerebellar Neoplasms/diet therapy , Cerebellar Neoplasms/physiopathology , Glutamine/metabolism , Medulloblastoma/diet therapy , Medulloblastoma/physiopathology , Tumor Protein p73/genetics , Tumor Protein p73/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic/genetics , Glutaminase/genetics , Glutaminase/metabolism , Heterografts , Humans , Mice , Mitochondria/genetics , Mitochondria/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Survival Analysis , TOR Serine-Threonine Kinases/metabolism , Treatment Outcome , Tumor Cells, Cultured
6.
Am J Hum Genet ; 108(7): 1318-1329, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34077761

ABSTRACT

TP73 belongs to the TP53 family of transcription factors and has therefore been well studied in cancer research. Studies in mice, however, have revealed non-oncogenic activities related to multiciliogenesis. Utilizing whole-exome sequencing analysis in a cohort of individuals with a mucociliary clearance disorder and cortical malformation, we identified homozygous loss-of-function variants in TP73 in seven individuals from five unrelated families. All affected individuals exhibit a chronic airway disease as well as a brain malformation consistent with lissencephaly. We performed high-speed video microscopy, immunofluorescence analyses, and transmission electron microscopy in respiratory epithelial cells after spheroid or air liquid interface culture to analyze ciliary function, ciliary length, and number of multiciliated cells (MCCs). The respiratory epithelial cells studied display reduced ciliary length and basal bodies mislocalized within the cytoplasm. The number of MCCs is severely reduced, consistent with a reduced number of cells expressing the transcription factors crucial for multiciliogenesis (FOXJ1, RFX2). Our data demonstrate that autosomal-recessive deleterious variants in the TP53 family member TP73 cause a mucociliary clearance disorder due to a defect in MCC differentiation.


Subject(s)
Lissencephaly/genetics , Mucociliary Clearance/genetics , Respiratory Mucosa/metabolism , Tumor Protein p73/genetics , Cell Differentiation/genetics , Cells, Cultured , Ciliopathies/genetics , Genes, Recessive , Homozygote , Humans , Loss of Function Mutation , Microscopy, Video , Respiratory Mucosa/cytology , Respiratory Mucosa/ultrastructure , Exome Sequencing
7.
BMC Cancer ; 24(1): 1164, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300412

ABSTRACT

The intricate interplay of cancer stem cell plasticity, along with the bidirectional transformation between epithelial-mesenchymal states, introduces further intricacy to offer insights into newer therapeutic approaches. Differentiation therapy, while successful in targeting leukemic stem cells, has shown limited overall success, with only a few promising instances. Using colon carcinoma cell strains with sequential p53/p73 knockdowns, our study underscores the association between p53/p73 and the maintenance of cellular plasticity. Morphological alterations corresponding with cell surface marker expressions, transcriptome analysis and functional assays were performed to access stemness and EMT (Epithelial-Mesenchymal Transition) characteristics in the spectrum of cells exhibiting sequential p53 and p73 knockdowns. Notably, our investigation explores the effectiveness of esculetin in reversing the shift from an epithelial to a mesenchymal phenotype, characterized by stem cell-like traits. Esculetin significantly induces enterocyte differentiation and promotes epithelial cell polarity by altering Wnt axes in Cancer Stem Cell-like cells characterized by high mesenchymal features. These results align with our previous findings in leukemic blast cells, establishing esculetin as an effective differentiating agent in both Acute Myeloid Leukemia (AML) and solid tumor cells.


Subject(s)
Cell Differentiation , Cell Plasticity , Epithelial-Mesenchymal Transition , Gene Knockdown Techniques , Neoplastic Stem Cells , Tumor Protein p73 , Tumor Suppressor Protein p53 , Umbelliferones , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Humans , Umbelliferones/pharmacology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Cell Differentiation/drug effects , Tumor Protein p73/metabolism , Tumor Protein p73/genetics , Cell Plasticity/drug effects , Cell Line, Tumor , Phenotype , Cell Transformation, Neoplastic/genetics , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism
8.
BMC Cancer ; 24(1): 587, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38741073

ABSTRACT

YAP and TAZ, the Hippo pathway terminal transcriptional activators, are frequently upregulated in cancers. In tumor cells, they have been mainly associated with increased tumorigenesis controlling different aspects from cell cycle regulation, stemness, or resistance to chemotherapies. In fewer cases, they have also been shown to oppose cancer progression, including by promoting cell death through the action of the p73/YAP transcriptional complex, in particular after chemotherapeutic drug exposure. Using HCT116 cells, we show here that oxaliplatin treatment led to core Hippo pathway down-regulation and nuclear accumulation of TAZ. We further show that TAZ was required for the increased sensitivity of HCT116 cells to oxaliplatin, an effect that appeared independent of p73, but which required the nuclear relocalization of TAZ. Accordingly, Verteporfin and CA3, two drugs affecting the activity of YAP and TAZ, showed antagonistic effects with oxaliplatin in co-treatments. Importantly, using several colorectal cell lines, we show that the sensitizing action of TAZ to oxaliplatin is dependent on the p53 status of the cells. Our results support thus an early action of TAZ to sensitize cells to oxaliplatin, consistent with a model in which nuclear TAZ in the context of DNA damage and p53 activity pushes cells towards apoptosis.


Subject(s)
Antineoplastic Agents , Colonic Neoplasms , Hippo Signaling Pathway , Oxaliplatin , Protein Serine-Threonine Kinases , Signal Transduction , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Tumor Suppressor Protein p53 , Humans , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Colonic Neoplasms/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Hippo Signaling Pathway/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/therapeutic use , Oxaliplatin/pharmacology , Porphyrins/pharmacology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Trans-Activators/metabolism , Trans-Activators/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Tumor Protein p73/metabolism , Tumor Protein p73/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Verteporfin/pharmacology , Verteporfin/therapeutic use , YAP-Signaling Proteins/metabolism
9.
Mol Biol Rep ; 51(1): 886, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39105958

ABSTRACT

Cancer is considered the uncontrolled growth and spread of cells into neighboring tissues, a process governed at the molecular level by many different factors, including abnormalities in the protein family's death-associated kinase (DAPK). DAPK2 is a member of the DAPK protein family, which plays essential roles in several cellular processes. DAPK2 acts as a tumor suppressor, interacting with several proteins, such as TNF, IFN, etc. during apoptosis and autophagy. Expression of DAPK2 causes changes in the structure of the cell, ultimately leading to cell death by apoptosis. In this essay, studies are obtained from Scopus, PubMed, and the Web of Science. According to these investigations, DAPK2 activates autophagy by interacting with AMPK, mTORC1, and p73. Furthermore, DAPK2 induces apoptosis pathway via interacting with the p73 family and JNK. In general, due to the vital role of DAPK2 in cell physiology and its effect on various factors and signaling pathways, it can be a potent target in the treatment of various cancers, including gastric, ovarian, breast, and other prominent cancers.


Subject(s)
Apoptosis , Autophagy , Death-Associated Protein Kinases , Neoplasms , Signal Transduction , Humans , Death-Associated Protein Kinases/metabolism , Death-Associated Protein Kinases/genetics , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Apoptosis/genetics , Autophagy/genetics , Tumor Protein p73/metabolism , Tumor Protein p73/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Gene Expression Regulation, Neoplastic
10.
PLoS Genet ; 17(5): e1009553, 2021 05.
Article in English | MEDLINE | ID: mdl-33945523

ABSTRACT

The CBFB gene is frequently mutated in several types of solid tumors. Emerging evidence suggests that CBFB is a tumor suppressor in breast cancer. However, our understanding of the tumor suppressive function of CBFB remains incomplete. Here, we analyze genetic interactions between mutations of CBFB and other highly mutated genes in human breast cancer datasets and find that CBFB and TP53 mutations are mutually exclusive, suggesting a functional association between CBFB and p53. Integrated genomic studies reveal that TAp73 is a common transcriptional target of CBFB and p53. CBFB cooperates with p53 to maintain TAp73 expression, as either CBFB or p53 loss leads to TAp73 depletion. TAp73 re-expression abrogates the tumorigenic effect of CBFB deletion. Although TAp73 loss alone is insufficient for tumorigenesis, it enhances the tumorigenic effect of NOTCH3 overexpression, a downstream event of CBFB loss. Immunohistochemistry shows that p73 loss is coupled with higher proliferation in xenografts. Moreover, TAp73 loss-of-expression is a frequent event in human breast cancer tumors and cell lines. Together, our results significantly advance our understanding of the tumor suppressive functions of CBFB and reveal a mechanism underlying the communication between the two tumor suppressors CBFB and p53.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Core Binding Factor beta Subunit/genetics , Gene Expression Regulation, Neoplastic , Tumor Protein p73/genetics , Tumor Suppressor Protein p53/genetics , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor beta Subunit/deficiency , Core Binding Factor beta Subunit/metabolism , Female , Genes, Tumor Suppressor , Humans , Immunohistochemistry , Mice , Mutation , Receptor, Notch3/genetics , Receptor, Notch3/metabolism , Transcription, Genetic , Tumor Protein p73/deficiency , Tumor Protein p73/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
11.
Proc Natl Acad Sci U S A ; 118(44)2021 11 02.
Article in English | MEDLINE | ID: mdl-34716260

ABSTRACT

The p53 tumor suppressor protein, known to be critically important in several processes including cell-cycle arrest and apoptosis, is highly regulated by multiple mechanisms, most certifiably the Murine Double Minute 2-Murine Double Minute X (MDM2-MDMX) heterodimer. The role of MDM2-MDMX in cell-cycle regulation through inhibition of p53 has been well established. Here we report that in cells either lacking p53 or expressing certain tumor-derived mutant forms of p53, loss of endogenous MDM2 or MDMX, or inhibition of E3 ligase activity of the heterocomplex, causes cell-cycle arrest. This arrest is correlated with a reduction in E2F1, E2F3, and p73 levels. Remarkably, direct ablation of endogenous p73 produces a similar effect on the cell cycle and the expression of certain E2F family members at both protein and messenger RNA levels. These data suggest that MDM2 and MDMX, working at least in part as a heterocomplex, may play a p53-independent role in maintaining cell-cycle progression by promoting the activity of E2F family members as well as p73, making them a potential target of interest in cancers lacking wild-type p53.


Subject(s)
Cell Cycle Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Protein p73/metabolism , Animals , Apoptosis , Cell Cycle/physiology , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , DNA-Binding Proteins/metabolism , E2F1 Transcription Factor/metabolism , Humans , Nuclear Proteins/metabolism , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Tumor Protein p73/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
12.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33649219

ABSTRACT

Infiltration of tumor-promoting immune cells is a strong driver of tumor progression. Especially the accumulation of macrophages in the tumor microenvironment is known to facilitate tumor growth and to correlate with poor prognosis in many tumor types. TAp73, a member of the p53/p63/p73 family, acts as a tumor suppressor and has been shown to suppress tumor angiogenesis. However, what role TAp73 has in regulating immune cell infiltration is unknown. Here, we report that low levels of TAp73 correlate with an increased NF-κB-regulated inflammatory signature in breast cancer. Furthermore, we show that loss of TAp73 results in NF-κB hyperactivation and secretion of Ccl2, a known NF-κB target and chemoattractant for monocytes and macrophages. Importantly, TAp73-deficient tumors display an increased accumulation of protumoral macrophages that express the mannose receptor (CD206) and scavenger receptor A (CD204) compared to controls. The relevance of TAp73 expression in human breast carcinoma was further accentuated by revealing that TAp73 expression correlates negatively with the accumulation of protumoral CD163+ macrophages in breast cancer patient samples. Taken together, our findings suggest that TAp73 regulates macrophage accumulation and phenotype in breast cancer through inhibition of the NF-κB pathway.


Subject(s)
Breast Neoplasms/immunology , NF-kappa B/immunology , Signal Transduction/immunology , Tumor Microenvironment/immunology , Tumor Protein p73/immunology , Tumor-Associated Macrophages/immunology , Animals , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Breast Neoplasms/pathology , Chemokine CCL2/immunology , Female , Humans , Membrane Glycoproteins/immunology , Mice , Receptors, Cell Surface/immunology , Receptors, Immunologic/immunology , Scavenger Receptors, Class A/immunology , Tumor-Associated Macrophages/pathology
13.
Genes Dev ; 30(11): 1253-4, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27298333

ABSTRACT

Multiciliogenesis is essential for the function of different epithelia, and its failure results in brain defects, respiratory diseases, and infertility. In this issue of Genes & Development, Nemajerova and colleagues (pp. 1300-1312) reveal the p53 family member and p73 isoform TAp73 as a transcription factor dictating the differentiation of multiciliated cells. Their findings provide the long-awaited unifying explanation for the diverse phenotypes of the p73 knockout mice.


Subject(s)
Nuclear Proteins/genetics , Tumor Protein p73 , Animals , Cell Differentiation , DNA-Binding Proteins/genetics , Phenotype , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics
14.
Semin Cell Dev Biol ; 110: 51-60, 2021 02.
Article in English | MEDLINE | ID: mdl-32362381

ABSTRACT

Multiciliated cells (MCC) project dozens to hundreds of motile cilia from the cell surface to generate fluid flow across epithelial surfaces or turbulence to promote the transport of gametes. The MCC differentiation program is initiated by GEMC1 and MCIDAS, members of the geminin family, that activate key transcription factors, including p73 and FOXJ1, to control the multiciliogenesis program. To support the generation of multiple motile cilia, MCCs must undergo massive centriole amplification to generate a sufficient number of basal bodies (modified centrioles). This transcriptional program involves the generation of deuterosomes, unique structures that act as platforms to regulate centriole amplification, the reactivation of cell cycle programs to control centriole amplification and release, and extensive remodeling of the cytoskeleton. This review will focus on providing an overview of the transcriptional regulation of MCCs and its connection to key processes, in addition to highlighting exciting recent developments and open questions in the field.


Subject(s)
Cell Cycle Proteins/genetics , Centrioles/metabolism , Cilia/metabolism , Ciliopathies/genetics , Transcription Factors/genetics , Transcription, Genetic , Animals , Cell Cycle/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Centrioles/ultrastructure , Cilia/ultrastructure , Ciliopathies/metabolism , Ciliopathies/pathology , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Humans , Signal Transduction , Transcription Factors/metabolism , Tumor Protein p73/genetics , Tumor Protein p73/metabolism
15.
Cancer Metastasis Rev ; 41(4): 853-869, 2022 12.
Article in English | MEDLINE | ID: mdl-35948758

ABSTRACT

Cancer largely adheres to Darwinian selection. Evolutionary forces are prominent during metastasis, the final and incurable disease stage, where cells acquire combinations of advantageous phenotypic features and interact with a dynamically changing microenvironment, in order to overcome the metastatic bottlenecks, while therapy exerts additional selective pressures. As a strategy to increase their fitness, tumors often co-opt developmental and tissue-homeostasis programs. Herein, 25 years after its discovery, we review TP73, a sibling of the cardinal tumor-suppressor TP53, through the lens of cancer evolution. The TP73 gene regulates a wide range of processes in embryonic development, tissue homeostasis and cancer via an overwhelming number of functionally divergent isoforms. We suggest that TP73 neither merely mimics TP53 via its p53-like tumor-suppressive functions, nor has black-or-white-type effects, as inferred by the antagonism between several of its isoforms in processes like apoptosis and DNA damage response. Rather, under dynamic conditions of selective pressure, the various p73 isoforms which are often co-expressed within the same cancer cells may work towards a common goal by simultaneously activating isoform-specific transcriptional and non-transcriptional programs. Combinatorial co-option of these programs offers selective advantages that overall increase the likelihood for successfully surpassing the barriers of the metastatic cascade. The p73 functional pleiotropy-based capabilities might be present in subclonal populations and expressed dynamically under changing microenvironmental conditions, thereby supporting clonal expansion and propelling evolution of metastasis. Deciphering the critical p73 isoform patterns along the spatiotemporal axes of tumor evolution could identify strategies to target TP73 for prevention and therapy of cancer metastasis.


Subject(s)
Neoplasms , Tumor Suppressor Proteins , Humans , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Tumor Protein p73/genetics , Tumor Suppressor Protein p53/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Genes, Tumor Suppressor , Neoplasms/genetics , Neoplasms/pathology , Tumor Microenvironment
16.
Nat Rev Mol Cell Biol ; 12(4): 259-65, 2011 04.
Article in English | MEDLINE | ID: mdl-21427767

ABSTRACT

The p53 family of proteins consists of p53, p63 and p73, which are transcription factors that affect both cancer and development. It is now emerging that these proteins also regulate maternal reproduction. Whereas p63 is important for maturation of the egg, p73 ensures normal mitosis in the developing blastocyst. p53 subsequently regulates implantation of the embryo through transcriptional control of leukaemia inhibitory factor. Elucidating the cell biological basis of how these factors regulate female fertility may lead to new approaches to the control of human maternal reproduction.


Subject(s)
DNA-Binding Proteins/physiology , Nuclear Proteins/physiology , Trans-Activators/physiology , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/physiology , Animals , Blastocyst/cytology , Blastocyst/physiology , DNA-Binding Proteins/genetics , Female , Fertility/genetics , Fertility/physiology , Humans , Male , Mice , Mice, Knockout , Models, Biological , Nuclear Proteins/genetics , Oocytes/cytology , Oocytes/physiology , Phosphoproteins/genetics , Phosphoproteins/physiology , Reproduction/genetics , Reproduction/physiology , Trans-Activators/genetics , Transcription Factors , Tumor Protein p73 , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics
17.
Proc Natl Acad Sci U S A ; 117(27): 15694-15701, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32571922

ABSTRACT

The p53 family member p73 has a complex gene structure, including alternative promoters and alternative splicing of the 3' UTR. This results in a complex range of isoforms whose biological relevance largely remains to be determined. By deleting exon 13 (which encodes a sterile α motif) from the Trp73 gene, we selectively engineered mice to replace the most abundantly expressed C-terminal isoform, p73α, with a shorter product of alternative splicing, p73ß. These mice (Trp73Δ13/Δ13 ) display severe neurodevelopmental defects with significant functional and morphological abnormalities. Replacement of p73α with p73ß results in the depletion of Cajal-Retzius (CR) cells in embryonic stages, thus depriving the developing hippocampus of the pool of neurons necessary for correct hippocampal architecture. Consequently, Trp73Δ13/Δ13 mice display severe hippocampal dysgenesis, reduced synaptic functionality and impaired learning and memory capabilities. Our data shed light on the relevance of p73 alternative splicing and show that the full-length C terminus of p73 is essential for hippocampal development.


Subject(s)
Alternative Splicing/genetics , Embryonic Development/genetics , Hippocampus/growth & development , Tumor Protein p73/genetics , Animals , Apoptosis/genetics , Hippocampus/metabolism , Humans , Interstitial Cells of Cajal/metabolism , Learning/physiology , Memory/physiology , Mice , Neurons/metabolism , Promoter Regions, Genetic
18.
Mol Carcinog ; 61(7): 629-642, 2022 07.
Article in English | MEDLINE | ID: mdl-35560453

ABSTRACT

Members of the p53 family of transcription factors-p53, p63, and p73-share a high degree of homology; however, members can be activated in response to different stimuli, perform distinct (sometimes opposing) roles and are expressed in different tissues. The level of complexity is increased further by the transcription of multiple isoforms of each homolog, which may interact or interfere with each other and can impact cellular outcome. Proteins perform their functions through interacting with other proteins (and/or with nucleic acids). Therefore, identification of the interactors of a protein and how they interact in 3D is essential to fully comprehend their roles. By utilizing an in silico protein-protein interaction prediction method-HMI-PRED-we predicted interaction partners of p53 family members and modeled 3D structures of these protein interaction complexes. This method recovered experimentally known interactions while identifying many novel candidate partners. We analyzed the similarities and differences observed among the interaction partners to elucidate distinct functions of p53 family members and provide examples of how this information may yield mechanistic insight to explain their overlapping versus distinct/opposing outcomes in certain contexts. While some interaction partners are common to p53, p63, and p73, the majority are unique to each member. Nevertheless, most of the enriched pathways associated with these partners are common to all members, indicating that the members target the same biological pathways but through unique mediators. p63 and p73 have more common enriched pathways compared to p53, supporting their similar developmental roles in different tissues.


Subject(s)
Transcription Factors , Tumor Suppressor Protein p53 , DNA-Binding Proteins/metabolism , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Protein p73/genetics , Tumor Protein p73/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
19.
BMC Cancer ; 22(1): 581, 2022 May 25.
Article in English | MEDLINE | ID: mdl-35614413

ABSTRACT

BACKGROUND: Long non-coding RNA P73 antisense RNA 1 T (non-protein coding), also known as Lnc RNA TP73-AS1, is dysregulated in various tumors but the correlation between its expression and clinicopathological parameters and/or prognoses in cancer patients is inconclusive. Here, we performed a meta-analysis to evaluate the prognostic value of Lnc RNA TP73-AS1 for malignancies. METHODS: We systematically searched four online databases including PubMed, the Web of Science, Embase, and the Cochrane Library for eligible articles published up to June 29/2020. Odds ratios (ORs) and Pooled hazard ratios (HRs) with 95% confidence intervals (95% CIs) were used to assess the association of TP73-AS1 expression with prognostic and clinicopathological parameters. We further validated TP73-AS1 expression in various malignancies and its potential prognostic value using the GEPIA online database. We predicted potential biological processes and relevant signal mechanisms through the public databases. RESULTS: A total of 26 studies examining 14 cancers were analyzed to evaluate the relationship between TP73-AS1 expression, clinicopathological features and prognostic indicators. The results indicated that TP73-AS1 expression markedly correlates with TNM stage (OR = 3.27,95% CI:2.43-4.39, P < 0.00001), tumor size (OR = 3.00, 95%CI:2.08-4.35, P < 0.00001), lymph node metastasis (OR = 2.77, 95%CI:1.42-5.38,P < 0.00001) and distant metastasis (OR = 4.50,95%CI:2. 62-7.73,P < 0.00001). No correlation with age (OR = 1.12,95%CI:0.77-1.64, P > 0.05), gender (OR = 1.08, 95%CI:0.84-1.38, P > 0.05) or differentiation (OR = 1.39, 95%CI:0.71-2.70, P = 0.340) was observed. TP73-AS1 overexpression was a biomarker of poor Overall survival(OS)(HR = 1.85,95%CI:1.53-2.22, P < 0.00001) and Disease-Free-Survival (DFS) (HR = 1.57,95%CI:1.03-2.42, P < 0.05). Dysregulated TP73-AS1 expression and its prognostic value in various cancers was validated based on The Cancer Genome Atlas (TCGA). Further biological function predictions indicated that TP73-AS1 was involved in pro-oncogenic signaling. CONCLUSIONS: The upregulation of Lnc RNA TP73-AS1 was related to detrimental clinicopathological parameters and can be considered an indicator of poor prognosis for cancer malignancies.


Subject(s)
Neoplasms , RNA, Long Noncoding , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Computational Biology , Humans , Lymphatic Metastasis , Neoplasms/pathology , Prognosis , RNA, Long Noncoding/metabolism , Tumor Protein p73/genetics
20.
Mol Biol Rep ; 49(7): 6859-6869, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35138524

ABSTRACT

BACKGROUND: Prostate cancer is a malignant disease that severely affects the health and comfort of the male population. The long non-coding RNA TP73-AS1 has been shown to be involved in the malignant transformation of various human cancers. However, whether TP73-AS1 contributes to prostate cancer progression has not been reported yet. Accordingly, here we aimed to report the role of TP73-AS1 in the development and progression of prostate cancer and determine its relationship with TP73. METHODS AND RESULTS: TP73-AS1-specific siRNA oligo duplexes were used to silence TP73-AS1 in DU-145 and PC-3 cells. Results indicated that TP73-AS1 was upregulated whereas TP73 was downregulated in prostate cancer cells compared to normal prostate cells and there was a negative correlation between them. Besides, loss of function experiments of TP73-AS1 in prostate cancer cells strongly induced cellular apoptosis, interfered with the cell cycle progression, and modulated related pro- and anti-apoptotic gene expression. Colony formation and migration capacities of TP73-AS1-silenced prostate cancer cells were also found to be dramatically reduced. CONCLUSIONS: Our findings provide novel evidence that suggests a chief regulatory role for the TP73-TP73-AS1 axis in prostate cancer development and progression, suggesting that the TP73/TP73-AS1 axis can be a promising diagnostic and therapeutic target for prostate cancer.


Subject(s)
MicroRNAs , Prostatic Neoplasms , RNA, Long Noncoding , Tumor Protein p73/metabolism , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , MicroRNAs/genetics , Prostatic Neoplasms/genetics , RNA, Long Noncoding/genetics
SELECTION OF CITATIONS
SEARCH DETAIL