The foggy world(s) of p63 isoform regulation in normal cells and cancer

. 2021 Jul ; 254 (4) : 454-473. [epub] 20210407

Jazyk angličtina Země Anglie, Velká Británie Médium print-electronic

Typ dokumentu časopisecké články, práce podpořená grantem, přehledy

Perzistentní odkaz   https://www.medvik.cz/link/pmid33638205

The p53 family member p63 exists as two major protein variants (TAp63 and ΔNp63) with distinct expression patterns and functional properties. Whilst downstream target genes of p63 have been studied intensively, how p63 variants are themselves controlled has been relatively neglected. Here, we review advances in understanding ΔNp63 and TAp63 regulation, highlighting their distinct pathways. TAp63 has roles in senescence and metabolism, and in germ cell genome maintenance, where it is activated post-transcriptionally by phosphorylation cascades after DNA damage. The function and regulation of TAp63 in mesenchymal and haematopoietic cells is less clear but may involve epigenetic control through DNA methylation. ΔNp63 functions to maintain stem/progenitor cells in various epithelia and is overexpressed in squamous and certain other cancers. ΔNp63 is transcriptionally regulated through multiple enhancers in concert with chromatin modifying proteins. Many signalling pathways including growth factors, morphogens, inflammation, and the extracellular matrix influence ΔNp63 levels, with inconsistent results reported. There is also evidence for reciprocal regulation, including ΔNp63 activating its own transcription. ΔNp63 is downregulated during cell differentiation through transcriptional regulation, while post-transcriptional events cause proteasomal degradation. Throughout the review, we identify knowledge gaps and highlight discordances, providing potential explanations including cell-context and cell-matrix interactions. Identifying individual p63 variants has roles in differential diagnosis and prognosis, and understanding their regulation suggests clinically approved agents for targeting p63 that may be useful combination therapies for selected cancer patients. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Zobrazit více v PubMed

Levine AJ. p53: 800 million years of evolution and 40 years of discovery. Nat Rev Cancer 2020; 20: 471-480.

Kastenhuber ER, Lowe SW. Putting p53 in context. Cell 2017; 170: 1062-1078.

Schmale H, Bamberger C. A novel protein with strong homology to the tumor suppressor p53. Oncogene 1997; 15: 1363-1367.

Yang A, Kaghad M, Wang Y, et al. p63, a p53 homolog at 3q27-29, encodes multiple products with transactivating, death-inducing, and dominant-negative activities. Mol Cell 1998; 2: 305-316.

Melino G, Memmi EM, Pelicci PG, et al. Maintaining epithelial stemness with p63. Sci Signal 2015; 8: re9.

Fisher ML, Balinth S, Mills AA. p63-related signaling at a glance. J Cell Sci 2020; 133: jcs228015.

Yi M, Tan Y, Wang L, et al. TP63 links chromatin remodeling and enhancer reprogramming to epidermal differentiation and squamous cell carcinoma development. Cell Mol Life Sci 2020; 77: 4325-4346.

Beyer U, Moll-Rocek J, Moll UM, et al. Endogenous retrovirus drives hitherto unknown proapoptotic p63 isoforms in the male germ line of humans and great apes. Proc Natl Acad Sci U S A 2011; 108: 3624-3629.

Sethi I, Romano R-A, Gluck C, et al. A global analysis of the complex landscape of isoforms and regulatory networks of p63 in human cells and tissues. BMC Genomics 2015; 16: 584.

eGTEx Project. Enhancing GTEx by bridging the gaps between genotype, gene expression, and disease. Nat Genet 2017; 49: 1664-1670.

Ghandi M, Huang FW, Jané-Valbuena J, et al. Next-generation characterization of the Cancer Cell Line Encyclopedia. Nature 2019; 569: 503-508.

Di Como CJ, Urist MJ, Babayan I, et al. p63 expression profiles in human normal and tumor tissues. Clin Cancer Res 2002; 8: 494-501.

Karni-Schmidt O, Castillo-Martin M, Shen TH, et al. Distinct expression profiles of p63 variants during urothelial development and bladder cancer progression. Am J Pathol 2011; 178: 1350-1360.

Tacha D, Bremer R, Haas T, et al. An immunohistochemical analysis of a newly developed, mouse monoclonal p40 (BC28) antibody in lung, bladder, skin, breast, prostate, and head and neck cancers. Arch Pathol Lab Med 2014; 138: 1358-1364.

Nekulova M, Holcakova J, Nenutil R, et al. Characterization of specific p63 and p63-N-terminal isoform antibodies and their application for immunohistochemistry. Virchows Arch 2013; 463: 415-425.

Galoczova M, Nenutil R, Pokorna Z, et al. TAp63 and ΔNp63 (p40) in prostate adenocarcinomas: ΔNp63 associates with a basal-like cancer stem cell population but not with metastasis. Virchows Arch 2020; 478: 627-636.

Uchida K, Ross H, Lotan T, et al. ΔNp63 (p40) expression in prostatic adenocarcinoma with diffuse p63 positivity. Hum Pathol 2015; 46: 384-389.

Coates PJ, Nenutil R, Holcakova J, et al. p63 isoforms in triple-negative breast cancer: ΔNp63 associates with the basal phenotype whereas TAp63 associates with androgen receptor, lack of BRCA mutation, PTEN and improved survival. Virchows Arch 2018; 472: 351-359.

Liu Y, Nekulova M, Nenutil R, et al. ΔNp63/p40 correlates with the location and phenotype of basal/mesenchymal cancer stem-like cells in human ER+ and HER2+ breast cancers. J Pathol Clin Res 2020; 6: 83-93.

Kim J, Villadsen R, Sørlie T, et al. Tumor initiating but differentiated luminal-like breast cancer cells are highly invasive in the absence of basal-like activity. Proc Natl Acad Sci U S A 2012; 109: 6124-6129.

Jo VY, Fletcher CD. p63 immunohistochemical staining is limited in soft tissue tumors. Am J Clin Pathol 2011; 136: 762-766.

Orzol P, Holcakova J, Nekulova M, et al. The diverse oncogenic and tumour suppressor roles of p63 and p73 in cancer: a review by cancer site. Histol Histopathol 2015; 30: 503-521.

Loljung L, Coates PJ, Nekulova M, et al. High expression of p63 is correlated to poor prognosis in squamous cell carcinoma of the tongue. J Oral Pathol Med 2014; 43: 14-19.

Bishop JA, Teruya-Feldstein J, Westra WH, et al. p40 (ΔNp63) is superior to p63 for the diagnosis of pulmonary squamous cell carcinoma. Mod Pathol 2012; 25: 405-415.

Aubry M-C, Roden A, Murphy SJ, et al. Chromosomal rearrangements and copy number abnormalities of TP63 correlate with p63 protein expression in lung adenocarcinoma. Mod Pathol 2015; 28: 359-366.

Crum CP, McKeon FD. p63 in epithelial survival, germ cell surveillance, and neoplasia. Annu Rev Pathol 2010; 5: 349-371.

Nekulova M, Holcakova J, Coates P, et al. The role of p63 in cancer, stem cells and cancer stem cells. Cell Mol Biol Lett 2011; 16: 296-327.

Amelio I, Grespi F, Annicchiarico-Petruzzelli M, et al. p63 the guardian of human reproduction. Cell Cycle 2012; 11: 4545-4551.

Xian W, McKeon F. TAp63 as a guardian of female germ line integrity. Nat Struct Mol Biol 2018; 25: 201-202.

Cefalù S, Lena AM, Vojtesek B, et al. TAp63gamma is required for the late stages of myogenesis. Cell Cycle 2015; 14: 894-901.

Rouleau M, Medawar A, Hamon L, et al. TAp63 is important for cardiac differentiation of embryonic stem cells and heart development. Stem Cells 2011; 29: 1672-1683.

Ciuffoli V, Lena AM, Gambacurta A, et al. Myoblasts rely on TAp63 to control basal mitochondria respiration. Aging (Albany NY) 2018; 10: 3558-3573.

Terrinoni A, Serra V, Bruno E, et al. Role of p63 and the Notch pathway in cochlea development and sensorineural deafness. Proc Natl Acad Sci U S A 2013; 110: 7300-7305.

Su X, Gi YJ, Chakravarti D, et al. TAp63 is a master transcriptional regulator of lipid and glucose metabolism. Cell Metab 2012; 16: 511-525.

Guo X, Keyes WM, Papazoglu C, et al. TAp63 induces senescence and suppresses tumorigenesis in vivo. Nat Cell Biol 2009; 11: 1451-1457.

Su X, Paris M, Gi YJ, et al. TAp63 prevents premature aging by promoting adult stem cell maintenance. Cell Stem Cell 2009; 5: 64-75.

Xue Y, Barker N, Hoon S, et al. Bortezomib stabilizes and activates p53 in proliferative compartments of both normal and tumor tissues in vivo. Cancer Res 2019; 79: 3595-3607.

Meek DW. Regulation of the p53 response and its relationship to cancer. Biochem J 2015; 469: 325-346.

Kouwenhoven EN, Oti M, Niehues H, et al. Transcription factor p63 bookmarks and regulates dynamic enhancers during epidermal differentiation. EMBO Rep 2015; 16: 863-878.

ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 2012; 489: 57-74.

Dreos R, Ambrosini G, Cavin Périer R, et al. EPD and EPDnew, high-quality promoter resources in the next-generation sequencing era. Nucleic Acids Res 2013; 41: D157-D164.

Wen J, Yan H, He M, et al. GSK-3β protects fetal oocytes from premature death via modulating TAp63 expression in mice. BMC Biol 2019; 17: 23.

Lantner F, Starlets D, Gore Y, et al. CD74 induces TAp63 expression leading to B-cell survival. Blood 2007; 110: 4303-4311.

Lewis TS, McCormick RS, Stone IJ, et al. Proapoptotic signaling activity of the anti-CD40 monoclonal antibody dacetuzumab circumvents multiple oncogenic transformation events and chemosensitizes NHL cells. Leukemia 2011; 25: 1007-1016.

Wu J, Bergholz J, Lu J, et al. TAp63 is a transcriptional target of NF-kappaB. J Cell Biochem 2010; 109: 702-710.

Yao J-Y, Pao C-C, Chen J-K. Transcriptional activity of TAp63 promoter is regulated by c-jun. J Cell Physiol 2010; 225: 898-904.

Park E, Kim H, Kim JM, et al. FANCD2 activates transcription of TAp63 and suppresses tumorigenesis. Mol Cell 2013; 50: 908-918.

Gaudio E, Paduano F, Pinton S, et al. TCL1A interacts with TP63 and enhances the survival of Raji Burkitt lymphoma cell line. Br J Haematol 2018; 183: 509-512.

Stavniichuk R, DeLaForest A, Thompson CA, et al. GATA4 blocks squamous epithelial cell gene expression in human esophageal squamous cells. Sci Rep 2021; 11: 3206.

Papakonstantinou N, Ntoufa S, Tsagiopoulou M, et al. Integrated epigenomic and transcriptomic analysis reveals TP63 as a novel player in clinically aggressive chronic lymphocytic leukemia. Int J Cancer 2019; 144: 2695-2706.

Humphries LA, Godbersen JC, Danilova OV, et al. Pro-apoptotic TP53 homolog TAp63 is repressed via epigenetic silencing and B-cell receptor signalling in chronic lymphocytic leukaemia. Br J Haematol 2013; 163: 590-602.

Bethge N, Honne H, Hilden V, et al. Identification of highly methylated genes across various types of B-cell non-Hodgkin lymphoma. PLoS One 2013; 8: e79602.

Harmes DC, Bresnick E, Lubin EA, et al. Positive and negative regulation of ΔN-p63 promoter activity by p53 and ΔN-p63-α contributes to differential regulation of p53 target genes. Oncogene 2003; 22: 7607-7616.

Li N, Li H, Cherukuri P, et al. TA-p63-γ regulates expression of ΔN-p63 in a manner that is sensitive to p53. Oncogene 2006; 25: 2349-2359.

Waltermann A, Kartasheva NN, Dobbelstein M. Differential regulation of p63 and p73 expression. Oncogene 2003; 22: 5686-5693.

Ruptier C, De Gaspéris A, Ansieau S, et al. TP63 P2 promoter functional analysis identifies β-catenin as a key regulator of ΔNp63 expression. Oncogene 2011; 30: 4656-4665.

Lanza M, Marinari B, Papoutsaki M, et al. Cross-talks in the p53 family: ΔNp63 is an anti-apoptotic target for ΔN63α and p53 gain-of-function mutants. Cell Cycle 2006; 5: 1996-2004.

Romano R-A, Birkaya B, Sinha S. Defining the regulatory elements in the proximal promoter of ΔNp63 in keratinocytes: potential roles for Sp1/Sp3, NF-Y, and p63. J Invest Dermatol 2006; 126: 1469-1479.

Yoh KE, Regunath K, Guzman A, et al. Repression of p63 and induction of EMT by mutant Ras in mammary epithelial cells. Proc Natl Acad Sci U S A 2016; 113: E6107-E6116.

Chu W-K, Lee K-C, Chow S-E, et al. Dual regulation of the ΔNp63 transcriptional activity by ΔNp63 in human nasopharyngeal carcinoma cell. Biochem Biophys Res Commun 2006; 342: 1356-1360.

Chu W-K, Dai P-M, Li H-L, et al. Transcriptional activity of the ΔNp63 promoter is regulated by STAT3. J Biol Chem 2008; 283: 7328-7337.

Ripamonti F, Albano L, Rossini A, et al. EGFR through STAT3 modulates ΔN63α expression to sustain tumor-initiating cell proliferation in squamous cell carcinomas. J Cell Physiol 2013; 228: 871-878.

Hsueh Y-J, Chen H-C, Chu W-K, et al. STAT3 regulates the proliferation and differentiation of rabbit limbal epithelial cells via a ΔNp63-dependent mechanism. Invest Ophthalmol Vis Sci 2011; 52: 4685-4693.

Borrelli S, Testoni B, Callari M, et al. Reciprocal regulation of p63 by C/EBP delta in human keratinocytes. BMC Mol Biol 2007; 8: 85.

Barbaro V, Testa A, Di Iorio E, et al. C/EBPdelta regulates cell cycle and self-renewal of human limbal stem cells. J Cell Biol 2007; 177: 1037-1049.

Hsueh Y-J, Kuo P-C, Chen J-K. Transcriptional regulators of the ΔNp63: their role in limbal epithelial cell proliferation. J Cell Physiol 2013; 228: 536-546.

Li X, Ottosson S, Wang S, et al. Wilms' tumor gene 1 regulates p63 and promotes cell proliferation in squamous cell carcinoma of the head and neck. BMC Cancer 2015; 15: 342.

Chin SS, Romano R-A, Nagarajan P, et al. Aberrant epidermal differentiation and disrupted ΔNp63/Notch regulatory axis in Ets1 transgenic mice. Biol Open 2013; 2: 1336-1345.

Hu L, Liang S, Chen H, et al. ΔNp63α is a common inhibitory target in oncogenic PI3K/Ras/Her2-induced cell motility and tumor metastasis. Proc Natl Acad Sci U S A 2017; 114: E3964-E3973.

Mehrazarin S, Chen W, Oh J-E, et al. The p63 gene is regulated by Grainyhead-Like 2 (GRHL2) through reciprocal feedback and determines the epithelial phenotype in human keratinocytes. J Biol Chem 2015; 290: 19999-20008.

Laronda MM, Unno K, Ishi K, et al. Diethylstilbestrol induces vaginal adenosis by disrupting SMAD/RUNX1-mediated cell fate decision in the Müllerian duct epithelium. Dev Biol 2013; 381: 5-16.

Fukunishi N, Katoh I, Tomimori Y, et al. Induction of ΔNp63 by the newly identified keratinocyte-specific transforming growth factor β signaling pathway with Smad2 and IκB kinase α in squamous cell carcinoma. Neoplasia 2010; 12: 969-979.

Qian Y, Jung Y-S, Chen X. ΔNp63, a target of DEC1 and histone deacetylase 2, modulates the efficacy of histone deacetylase inhibitors in growth suppression and keratinocyte differentiation. J Biol Chem 2011; 286: 12033-12041.

Nguyen B-C, Lefort K, Mandinova A, et al. Cross-regulation between Notch and p63 in keratinocyte commitment to differentiation. Genes Dev 2006; 20: 1028-1042.

Fontemaggi G, Gurtner A, Damalas A, et al. δEF1 repressor controls selectively p53 family members during differentiation. Oncogene 2005; 24: 7273-7280.

Kataoka K, Matsushima T, Ito Y, et al. Bhlha9 regulates apical ectodermal ridge formation during limb development. J Bone Miner Metab 2018; 36: 64-72.

Wang Y, Lin X, Gong X, et al. Atypical GATA transcription factor TRPS1 represses gene expression by recruiting CHD4/NuRD(MTA2) and suppresses cell migration and invasion by repressing TP63 expression. Oncogenesis 2018; 7: 96.

Bao X, Rubin AJ, Qu K, et al. A novel ATAC-seq approach reveals lineage-specific reinforcement of the open chromatin landscape via cooperation between BAF and p63. Genome Biol 2015; 16: 284.

Rinaldi L, Datta D, Serrat J, et al. Dnmt3a and Dnmt3b associate with enhancers to regulate human epidermal stem cell homeostasis. Cell Stem Cell 2016; 19: 491-501.

Pattison JM, Melo SP, Piekos SN, et al. Retinoic acid and BMP4 cooperate with TP63 to alter chromatin dynamics during surface epithelial commitment. Nat Genet 2018; 50: 1658-1665.

Hamdan FH, Johnsen SA. ΔNp63-dependent super enhancers define molecular identity in pancreatic cancer by an interconnected transcription factor network. Proc Natl Acad Sci U S A 2018; 115: E12343-E12352.

Qu J, Tanis SEJ, Smits JPH, et al. Mutant p63 affects epidermal cell identity through rewiring the enhancer landscape. Cell Rep 2018; 25: 3490-3503.e4.

Li L, Wang Y, Torkelson JL, et al. TFAP2C- and p63-dependent networks sequentially rearrange chromatin landscapes to drive human epidermal lineage commitment. Cell Stem Cell 2019; 24: 271-284.e8.

Santos-Pereira JM, Gallardo-Fuentes L, Neto A, et al. Pioneer and repressive functions of p63 during zebrafish embryonic ectoderm specification. Nat Commun 2019; 10: 3049.

Qu J, Yi G, Zhou H. p63 cooperates with CTCF to modulate chromatin architecture in skin keratinocytes. Epigenetics Chromatin 2019; 12: 31.

Lin-Shiao E, Lan Y, Welzenbach J, et al. p63 establishes epithelial enhancers at critical craniofacial development genes. Sci Adv 2019; 5: eaaw0946.

Jiang Y-Y, Jiang Y, Li C-Q, et al. TP63, SOX2, and KLF5 establish a core regulatory circuitry that controls epigenetic and transcription patterns in esophageal squamous cell carcinoma cell lines. Gastroenterology 2020; 159: 1311-1327.e19.

Antonini D, Rossi B, Han R, et al. An autoregulatory loop directs the tissue-specific expression of p63 through a long-range evolutionarily conserved enhancer. Mol Cell Biol 2006; 26: 3308-3318.

Antonini D, Sirico A, Aberdam E, et al. A composite enhancer regulates p63 gene expression in epidermal morphogenesis and in keratinocyte differentiation by multiple mechanisms. Nucleic Acids Res 2015; 43: 862-874.

Leslie EJ, Carlson JC, Shaffer JR, et al. Genome-wide meta-analyses of nonsyndromic orofacial clefts identify novel associations between FOXE1 and all orofacial clefts, and TP63 and cleft lip with or without cleft palate. Hum Genet 2017; 136: 275-286.

Sastre-Perona A, Hoang-Phou S, Leitner M-C, et al. De novo PITX1 expression controls bi-stable transcriptional circuits to govern self-renewal and differentiation in squamous cell carcinoma. Cell Stem Cell 2019; 24: 390-404.e8.

Ochieng JK, Schilders K, Kool H, et al. Sox2 regulates the emergence of lung basal cells by directly activating the transcription of Trp63. Am J Respir Cell Mol Biol 2014; 51: 311-322.

Bhattacharya S, Serror L, Nir E, et al. SOX2 regulates P63 and stem/progenitor cell state in the corneal epithelium. Stem Cells 2019; 37: 417-429.

Wu M, Ingram L, Tolosa EJ, et al. Gli transcription factors mediate the oncogenic transformation of prostate basal cells induced by a Kras-androgen receptor axis. J Biol Chem 2016; 291: 25749-25760.

Dudek AM, Vermeulen SH, Kolev D, et al. Identification of an enhancer region within the TP63/LEPREL1 locus containing genetic variants associated with bladder cancer risk. Cell Oncol (Dordr) 2018; 41: 555-568.

Li F, Yuan Q, Di W, et al. ERG orchestrates chromatin interactions to drive prostate cell fate reprogramming. J Clin Invest 2020; 130: 5924-5941.

Duteil D, Tourrette Y, Eberlin A, et al. The histone acetyltransferase inhibitor Nir regulates epidermis development. Development 2018; 145: dev158453.

Stefanowicz D, Lee JY, Lee K, et al. Elevated H3K18 acetylation in airway epithelial cells of asthmatic subjects. Respir Res 2015; 16: 95.

Andricovich J, Perkail S, Kai Y, et al. Loss of KDM6A activates super-enhancers to induce gender-specific squamous-like pancreatic cancer and confers sensitivity to BET inhibitors. Cancer Cell 2018; 33: 512-526.e8.

Shi Y-X, Wang Y, Li X, et al. Genome-wide DNA methylation profiling reveals novel epigenetic signatures in squamous cell lung cancer. BMC Genomics 2017; 18: 901.

Campbell JD, Yau C, Bowlby R, et al. Genomic, pathway network, and immunologic features distinguishing squamous carcinomas. Cell Rep 2018; 23: 194-212.e6.

Zhang H, Jin Z, Cheng L, et al. Integrative analysis of methylation and gene expression in lung adenocarcinoma and squamous cell lung carcinoma. Front Bioeng Biotechnol 2020; 8: 3.

Park BJ, Lee SJ, Kim JI, et al. Frequent alteration of p63 expression in human primary bladder carcinomas. Cancer Res 2000; 60: 3370-3374.

Gallant-Behm CL, Ramsey MR, Bensard CL, et al. ΔNp63α represses anti-proliferative genes via H2A.Z deposition. Genes Dev 2012; 26: 2325-2336.

Hazawa M, Lin D-C, Kobayashi A, et al. ROCK-dependent phosphorylation of NUP62 regulates p63 nuclear transport and squamous cell carcinoma proliferation. EMBO Rep 2018; 19: 73-88.

Li J, Xu X, Tiwari M, et al. SPT6 promotes epidermal differentiation and blockade of an intestinal-like phenotype through control of transcriptional elongation. Nat Commun 2021; 12: 784.

Candi E, Amelio I, Agostini M, et al. MicroRNAs and p63 in epithelial stemness. Cell Death Differ 2015; 22: 12-21.

Lin C, Li X, Zhang Y, et al. The microRNA feedback regulation of p63 in cancer progression. Oncotarget 2015; 6: 8434-8453.

Sakaram S, Craig MP, Hill NT, et al. Identification of novel ΔNp63α-regulated miRNAs using an optimized small RNA-Seq analysis pipeline. Sci Rep 2018; 8: 10069.

Su X, Chakravarti D, Cho MS, et al. TAp63 suppresses metastasis through coordinate regulation of Dicer and miRNAs. Nature 2010; 467: 986-990.

Chakravarti D, Su X, Cho MS, et al. Induced multipotency in adult keratinocytes through down-regulation of ΔNp63 or DGCR8. Proc Natl Acad Sci U S A 2014; 111: E572-E581.

Zhou R, Gao Y, Lv D, et al. METTL3 mediated m6A modification plays an oncogenic role in cutaneous squamous cell carcinoma by regulating ΔNp63. Biochem Biophys Res Commun 2019; 515: 310-317.

Ke S, Pandya-Jones A, Saito Y, et al. m6A mRNA modifications are deposited in nascent pre-mRNA and are not required for splicing but do specify cytoplasmic turnover. Genes Dev 2017; 31: 990-1006.

Lin S, Choe J, Du P, et al. The m6A methyltransferase METTL3 promotes translation in human cancer cells. Mol Cell 2016; 62: 335-345.

González S, Halabi M, Ju D, et al. Role of Jagged1-mediated Notch signaling activation in the differentiation and stratification of the human limbal epithelium. Cell 2020; 9: 1945.

Tadeu AMB, Horsley V. Notch signaling represses p63 expression in the developing surface ectoderm. Development 2013; 140: 3777-3786.

Yugawa T, Narisawa-Saito M, Yoshimatsu Y, et al. ΔNp63alpha repression of the Notch1 gene supports the proliferative capacity of normal human keratinocytes and cervical cancer cells. Cancer Res 2010; 70: 4034-4044.

Herrick DB, Lin B, Peterson J, et al. Notch1 maintains dormancy of olfactory horizontal basal cells, a reserve neural stem cell. Proc Natl Acad Sci U S A 2017; 114: E5589-E5598.

Thorpe LM, Yuzugullu H, Zhao JJ. PI3K in cancer: divergent roles of isoforms, modes of activation and therapeutic targeting. Nat Rev Cancer 2015; 15: 7-24.

Madsen RR. PI3K in stemness regulation: from development to cancer. Biochem Soc Trans 2020; 48: 301-315.

Matheny KE, Barbieri CE, Sniezek JC, et al. Inhibition of epidermal growth factor receptor signaling decreases p63 expression in head and neck squamous carcinoma cells. Laryngoscope 2003; 113: 936-939.

Barbieri CE, Barton CE, Pietenpol JA. ΔNp63α expression is regulated by the phosphoinositide 3-kinase pathway. J Biol Chem 2003; 278: 51408-51414.

Ma J, Meng Y, Kwiatkowski DJ, et al. Mammalian target of rapamycin regulates murine and human cell differentiation through STAT3/p63/Jagged/Notch cascade. J Clin Invest 2010; 120: 103-114.

D'Amico S, Shi J, Martin BL, et al. STAT3 is a master regulator of epithelial identity and KRAS-driven tumorigenesis. Genes Dev 2018; 32: 1175-1187.

Galoczova M, Coates P, Vojtesek B. STAT3, stem cells, cancer stem cells and p63. Cell Mol Biol Lett 2018; 23: 12.

Galvis LA, Holik AZ, Short KM, et al. Repression of Igf1 expression by Ezh2 prevents basal cell differentiation in the developing lung. Development 2015; 142: 1458-1469.

Günschmann C, Stachelscheid H, Akyüz MD, et al. Insulin/IGF-1 controls epidermal morphogenesis via regulation of FoxO-mediated p63 inhibition. Dev Cell 2013; 26: 176-187.

Nagarajan S, Bedi U, Budida A, et al. BRD4 promotes p63 and GRHL3 expression downstream of FOXO in mammary epithelial cells. Nucleic Acids Res 2017; 45: 3130-3145.

Khan IA, Yoo BH, McPhee M, et al. ErbB2-driven downregulation of the transcription factor Irf6 in breast epithelial cells is required for their 3D growth. Breast Cancer Res 2018; 20: 151.

Fichter CD, Przypadlo CM, Buck A, et al. A new model system identifies epidermal growth factor receptor-human epidermal growth factor receptor 2 (HER2) and HER2-human epidermal growth factor receptor 3 heterodimers as potent inducers of oesophageal epithelial cell invasion. J Pathol 2017; 243: 481-495.

Vasilaki E, Morikawa M, Koinuma D, et al. Ras and TGF-β signaling enhance cancer progression by promoting the ΔNp63 transcriptional program. Sci Signal 2016; 9: ra84.

Sundqvist A, Vasilaki E, Voytyuk O, et al. TGFβ and EGF signaling orchestrates the AP-1- and p63 transcriptional regulation of breast cancer invasiveness. Oncogene 2020; 39: 4436-4449.

Holcakova J, Nekulova M, Orzol P, et al. ΔNp63 activates EGFR signaling to induce loss of adhesion in triple-negative basal-like breast cancer cells. Breast Cancer Res Treat 2017; 163: 475-484.

Citro S, Bellini A, Miccolo C, et al. Synergistic antitumour activity of HDAC inhibitor SAHA and EGFR inhibitor gefitinib in head and neck cancer: a key role for ΔNp63α. Br J Cancer 2019; 120: 658-667.

Tomoshige K, Guo M, Tsuchiya T, et al. An EGFR ligand promotes EGFR-mutant but not KRAS-mutant lung cancer in vivo. Oncogene 2018; 37: 3894-3908.

The Cancer Genome Atlas Research Network. Comprehensive genomic characterization of squamous cell lung cancers. Nature 2012; 489: 519-525.

Dotto GP, Rustgi AK. Squamous cell cancers: a unified perspective on biology and genetics. Cancer Cell 2016; 29: 622-637.

Jiang Y, Jiang Y-Y, Xie J-J, et al. Co-activation of super-enhancer-driven CCAT1 by TP63 and SOX2 promotes squamous cancer progression. Nat Commun 2018; 9: 3619.

Sato T, Yoo S, Kong R, et al. Epigenomic profiling discovers trans-lineage SOX2 partnerships driving tumor heterogeneity in lung squamous cell carcinoma. Cancer Res 2019; 79: 6084-6100.

Segrelles C, Moral M, Lara MF, et al. Molecular determinants of Akt-induced keratinocyte transformation. Oncogene 2006; 25: 1174-1185.

Danilov AV, Neupane D, Nagaraja AS, et al. ΔNp63α-mediated induction of epidermal growth factor receptor promotes pancreatic cancer cell growth and chemoresistance. PLoS One 2011; 6: e26815.

Carroll DK, Carroll JS, Leong C-O, et al. p63 regulates an adhesion programme and cell survival in epithelial cells. Nat Cell Biol 2006; 8: 551-561.

Forster N, Saladi SV, van Bragt M, et al. Basal cell signaling by p63 controls luminal progenitor function and lactation via NRG1. Dev Cell 2014; 28: 147-160.

Ogawa E, Okuyama R, Ikawa S, et al. p51/p63 inhibits ultraviolet B-induced apoptosis via Akt activation. Oncogene 2008; 27: 848-856.

Leonard MK, Kommagani R, Payal V, et al. ΔNp63α regulates keratinocyte proliferation by controlling PTEN expression and localization. Cell Death Differ 2011; 18: 1924-1933.

Chikama T, Liu C-Y, Meij JTA, et al. Excess FGF-7 in corneal epithelium causes corneal intraepithelial neoplasia in young mice and epithelium hyperplasia in adult mice. Am J Pathol 2008; 172: 638-649.

Yamamoto-Fukuda T, Akiyama N, Takahashi M, et al. Keratinocyte growth factor (KGF) modulates epidermal progenitor cell kinetics through activation of p63 in middle ear cholesteatoma. J Assoc Res Otolaryngol 2018; 19: 223-241.

Cheng C-C, Wang D-Y, Kao M-H, et al. The growth-promoting effect of KGF on limbal epithelial cells is mediated by upregulation of ΔNp63α through the p38 pathway. J Cell Sci 2009; 122: 4473-4480.

Rosato B, Ranieri D, Nanni M, et al. Role of FGFR2b expression and signaling in keratinocyte differentiation: sequential involvement of PKCδ and PKCα. Cell Death Dis 2018; 9: 565.

Ranieri D, Rosato B, Nanni M, et al. Expression of the FGFR2c mesenchymal splicing variant in human keratinocytes inhibits differentiation and promotes invasion. Mol Carcinog 2018; 57: 272-283.

Restelli M, Lopardo T, Lo Iacono N, et al. DLX5, FGF8 and the Pin1 isomerase control ΔNp63α protein stability during limb development: a regulatory loop at the basis of the SHFM and EEC congenital malformations. Hum Mol Genet 2014; 23: 3830-3842.

Restelli M, Molinari E, Marinari B, et al. FGF8, c-Abl and p300 participate in a pathway that controls stability and function of the ΔNp63α protein. Hum Mol Genet 2015; 24: 4185-4197.

Robertson DM, Ho S-I, Cavanagh HD. C-terminal cleavage of ΔNp63α is associated with TSA-induced apoptosis in immortalized corneal epithelial cells. Invest Ophthalmol Vis Sci 2010; 51: 3977-3985.

Volckaert T, Yuan T, Chao C-M, et al. Fgf10-Hippo epithelial-mesenchymal crosstalk maintains and recruits lung basal stem cells. Dev Cell 2017; 43: 48-59.e5.

Huang Y, Hamana T, Liu J, et al. Prostate sphere-forming stem cells are derived from the P63-expressing basal compartment. J Biol Chem 2015; 290: 17745-17752.

Metzger DE, Xu Y, Shannon JM. Elf5 is an epithelium-specific, fibroblast growth factor-sensitive transcription factor in the embryonic lung. Dev Dyn 2007; 236: 1175-1192.

Seoane J, Gomis RR. TGF-β family signaling in tumor suppression and cancer progression. Cold Spring Harb Perspect Biol 2017; 9: a022277.

Derynck R, Budi EH. Specificity, versatility, and control of TGF-β family signaling. Sci Signal 2019; 12: eaav5183.

Murata K, Ota S, Niki T, et al. p63 - key molecule in the early phase of epithelial abnormality in idiopathic pulmonary fibrosis. Exp Mol Pathol 2007; 83: 367-376.

Hu M, Yao J, Carroll DK, et al. Regulation of in situ to invasive breast carcinoma transition. Cancer Cell 2008; 13: 394-406.

Yip YL, Tsao SW. Regulation of p63 expression in primary and immortalized nasopharyngeal epithelial cells. Int J Oncol 2008; 33: 713-724.

Mou H, Vinarsky V, Tata PR, et al. Dual SMAD signaling inhibition enables long-term expansion of diverse epithelial basal cells. Cell Stem Cell 2016; 19: 217-231.

Adorno M, Cordenonsi M, Montagner M, et al. A mutant-p53/Smad complex opposes p63 to empower TGFbeta-induced metastasis. Cell 2009; 137: 87-98.

Bui NHB, Napoli M, Davis AJ, et al. Spatiotemporal regulation of ΔNp63 by TGFβ-regulated miRNAs is essential for cancer metastasis. Cancer Res 2020; 80: 2833-2847.

Zhang Y, Yeh L-K, Zhang S, et al. Wnt/β-catenin signaling modulates corneal epithelium stratification via inhibition of Bmp4 during mouse development. Development 2015; 142: 3383-3393.

Zhang L, Wang Y-C, Okada Y, et al. Aberrant expression of a stabilized β-catenin mutant in keratocytes inhibits mouse corneal epithelial stratification. Sci Rep 2019; 9: 1919.

Min S, Oyelakin A, Gluck C, et al. p63 and its target follistatin maintain salivary gland stem/progenitor cell function through TGF-β/Activin signaling. iScience 2020; 23: 101524.

Abraham CG, Ludwig MP, Andrysik Z, et al. ΔNp63α suppresses TGFB2 expression and RHOA activity to drive cell proliferation in squamous cell carcinomas. Cell Rep 2018; 24: 3224-3236.

Hill NT, Gracia-Maldonado GH, Leonard MK, et al. Role of vitamin D3 in modulation of ΔNp63α expression during UVB induced tumor formation in SKH-1 mice. PLoS One 2014; 9: e107052.

Hill NT, Zhang J, Leonard MK, et al. 1α,25-Dihydroxyvitamin D₃ and the vitamin D receptor regulates ΔNp63α levels and keratinocyte proliferation. Cell Death Dis 2015; 6: e1781.

Shan NL, Minden A, Furmanski P, et al. Analysis of the transcriptome: regulation of cancer stemness in breast ductal carcinoma in situ by vitamin D compounds. Cancer Prev Res (Phila) 2020; 13: 673-686.

Curtis KM, Aenlle KK, Frisch RN, et al. TAp63γ and ΔNp63β promote osteoblastic differentiation of human mesenchymal stem cells: regulation by vitamin D3 metabolites. PLoS One 2015; 10: e0123642.

Kommagani R, Leonard MK, Lewis S, et al. Regulation of VDR by ΔNp63α is associated with inhibition of cell invasion. J Cell Sci 2009; 122: 2828-2835.

Bamberger C, Pollet D, Schmale H. Retinoic acid inhibits downregulation of ΔNp63α expression during terminal differentiation of human primary keratinocytes. J Invest Dermatol 2002; 118: 133-138.

Metallo CM, Ji L, de Pablo JJ, et al. Retinoic acid and bone morphogenetic protein signaling synergize to efficiently direct epithelial differentiation of human embryonic stem cells. Stem Cells 2008; 26: 372-380.

Kirschner RD, Rother K, Müller GA, et al. The retinal dehydrogenase/reductase retSDR1/DHRS3 gene is activated by p53 and p63 but not by mutants derived from tumors or EEC/ADULT malformation syndromes. Cell Cycle 2010; 9: 2177-2188.

Ma S, Meng Z, Chen R, et al. The Hippo pathway: biology and pathophysiology. Annu Rev Biochem 2019; 88: 577-604.

Zhao R, Fallon TR, Saladi SV, et al. Yap tunes airway epithelial size and architecture by regulating the identity, maintenance, and self-renewal of stem cells. Dev Cell 2014; 30: 151-165.

Saladi SV, Ross K, Karaayvaz M, et al. ACTL6A is co-amplified with p63 in squamous cell carcinoma to drive YAP activation, regenerative proliferation, and poor prognosis. Cancer Cell 2017; 31: 35-49.

Wang Y, Li J, Gao Y, et al. Hippo kinases regulate cell junctions to inhibit tumor metastasis in response to oxidative stress. Redox Biol 2019; 26: 101233.

Fisher ML, Ciavattone N, Grun D, et al. Sulforaphane reduces YAP/ΔNp63α signaling to reduce cancer stem cell survival and tumor formation. Oncotarget 2017; 8: 73407-73418.

Yuan M, Luong P, Hudson C, et al. c-Abl phosphorylation of ΔNp63α is critical for cell viability. Cell Death Dis 2010; 1: e16.

Chatterjee A, Sen T, Chang X, et al. Yes-associated protein 1 regulates the stability of ΔNp63α. Cell Cycle 2010; 9: 162-167.

Mendoza-Rodríguez M, Arévalo Romero H, Fuentes-Pananá EM, et al. IL-1β induces up-regulation of BIRC3, a gene involved in chemoresistance to doxorubicin in breast cancer cells. Cancer Lett 2017; 390: 39-44.

Mendoza-Rodríguez MG, Ayala-Sumuano JT, García-Morales L, et al. IL-1β inflammatory cytokine-induced TP63 isoform ΔNP63α signaling cascade contributes to cisplatin resistance in human breast cancer cells. Int J Mol Sci 2019; 20: 270.

Nelson AM, Katseff AS, Ratliff TS, et al. Interleukin 6 and STAT3 regulate p63 isoform expression in keratinocytes during regeneration. Exp Dermatol 2016; 25: 155-157.

Stadnikova A, Trosan P, Skalicka P, et al. Interleukin-13 maintains the stemness of conjunctival epithelial cell cultures prepared from human limbal explants. PLoS One 2019; 14: e0211861.

Ekman A-K, Bivik Eding C, Rundquist I, et al. IL-17 and IL-22 promote keratinocyte stemness in the germinative compartment in psoriasis. J Invest Dermatol 2019; 139: 1564-1573.e8.

Koshiba S, Ichimiya S, Nagashima T, et al. Tonsillar crypt epithelium of palmoplantar pustulosis secretes interleukin-6 to support B-cell development via p63/p73 transcription factors. J Pathol 2008; 214: 75-84.

Wu L, Chen X, Zhao J, et al. A novel IL-17 signaling pathway controlling keratinocyte proliferation and tumorigenesis via the TRAF4-ERK5 axis. J Exp Med 2015; 212: 1571-1587.

Gu X, Coates PJ, MacCallum SF, et al. TRAF4 is potently induced by TAp63 isoforms and localised according to differentiation in SCCHN. Cancer Biol Ther 2007; 6: 1986-1990.

Brauweiler AM, Leung DYM, Goleva E. The transcription factor p63 is a direct effector of IL-4/IL-13 mediated repression of keratinocyte differentiation. J Invest Dermatol 2021; 141: 770-778.

Yang X, Lu H, Yan B, et al. ΔNp63 versatilely regulates a broad NF-κB gene program and promotes squamous epithelial proliferation, migration, and inflammation. Cancer Res 2011; 71: 3688-3700.

Kumar S, Wilkes DW, Samuel N, et al. ΔNp63-driven recruitment of myeloid-derived suppressor cells promotes metastasis in triple-negative breast cancer. J Clin Invest 2018; 128: 5095-5109.

Chae Y-S, Kim H, Kim D, et al. Cell density-dependent acetylation of ΔNp63α is associated with p53-dependent cell cycle arrest. FEBS Lett 2012; 586: 1128-1134.

Salehi-Had H, Alvarenga LS, Isseroff R, et al. Factors modulating p63 expression in cultured limbal epithelial cells. Cornea 2005; 24: 845-852.

Ding L, Su Y, Fassl A, et al. Perturbed myoepithelial cell differentiation in BRCA mutation carriers and in ductal carcinoma in situ. Nat Commun 2019; 10: 4182.

Fisher ML, Kerr C, Adhikary G, et al. Transglutaminase interaction with α6/β4-integrin stimulates YAP1-dependent ΔNp63α stabilization and leads to enhanced cancer stem cell survival and tumor formation. Cancer Res 2016; 76: 7265-7276.

Grun D, Adhikary G, Eckert RL. NRP-1 interacts with GIPC1 and α6/β4-integrins to increase YAP1/ΔNp63α-dependent epidermal cancer stem cell survival. Oncogene 2018; 37: 4711-4722.

Chen J, Lan J, Liu D, et al. Ascorbic acid promotes the stemness of corneal epithelial stem/progenitor cells and accelerates epithelial wound healing in the cornea. Stem Cells Transl Med 2017; 6: 1356-1365.

Li H, Yun H-Y, Baek KJ, et al. Avian collagen is useful for the construction of skin equivalents. Cells Tissues Organs 2017; 204: 261-269.

Gouveia RM, Vajda F, Wibowo JA, et al. YAP, ΔNp63, and β-catenin signaling pathways are involved in the modulation of corneal epithelial stem cell phenotype induced by substrate stiffness. Cell 2019; 8: 347.

Kim Y-Y, Li H, Song YS, et al. Laminin peptide YIGSR enhances epidermal development of skin equivalents. J Tissue Viability 2018; 27: 117-121.

Hsueh Y-J, Huang S-F, Lai J-Y, et al. Preservation of epithelial progenitor cells from collagenase-digested oral mucosa during ex vivo cultivation. Sci Rep 2016; 6: 36266.

Nam SM, Maeng Y-S, Kim EK, et al. Ex vivo expansion of human limbal epithelial cells using human placenta-derived and umbilical cord-derived mesenchymal stem cells. Stem Cells Int 2017; 2017: 4206187.

Westfall MD, Joyner AS, Barbieri CE, et al. Ultraviolet radiation induces phosphorylation and ubiquitin-mediated degradation of ΔNp63α. Cell Cycle 2005; 4: 710-716.

Cherukuri P, DeCastro AJ, Balboni AL, et al. Phosphorylation of ΔNp63α via a novel TGFβ/ALK5 signaling mechanism mediates the anti-clonogenic effects of TGFβ. PLoS One 2012; 7: e50066.

Yen C-S, Chen J-C, Chang Y-F, et al. Lovastatin causes FaDu hypopharyngeal carcinoma cell death via AMPK-p63-survivin signaling cascade. Sci Rep 2016; 6: 25082.

Suzuki D, Senoo M. Increased p63 phosphorylation marks early transition of epidermal stem cells to progenitors. J Invest Dermatol 2012; 132: 2461-2464.

Choo M-K, Kraft S, Missero C, et al. The protein kinase p38α destabilizes p63 to limit epidermal stem cell frequency and tumorigenic potential. Sci Signal 2018; 11: eaau0727.

Huang Y-P, Kim Y, Li Z, et al. AEC-associated p63 mutations lead to alternative splicing/protein stabilization of p63 and modulation of Notch signaling. Cell Cycle 2005; 4: 1440-1447.

Huang Y, Sen T, Nagpal J, et al. ATM kinase is a master switch for the ΔNp63α phosphorylation/degradation in human head and neck squamous cell carcinoma cells upon DNA damage. Cell Cycle 2008; 7: 2846-2855.

Lazzari C, Prodosmo A, Siepi F, et al. HIPK2 phosphorylates ΔNp63α and promotes its degradation in response to DNA damage. Oncogene 2011; 30: 4802-4813.

Di Costanzo A, Festa L, Duverger O, et al. Homeodomain protein Dlx3 induces phosphorylation-dependent p63 degradation. Cell Cycle 2009; 8: 1185-1195.

Suh E-K, Yang A, Kettenbach A, et al. p63 protects the female germ line during meiotic arrest. Nature 2006; 444: 624-628.

Kim D-A, Lee B-L, Suh E-K. Ionizing radiation-induced TAp63α phosphorylation at C-terminal S/TQ motifs requires the N-terminal transactivation (TA) domain. Cell Cycle 2011; 10: 840-849.

Kim D-A, Suh E-K. Defying DNA double-strand break-induced death during prophase I meiosis by temporal TAp63α phosphorylation regulation in developing mouse oocytes. Mol Cell Biol 2014; 34: 1460-1473.

Tuppi M, Kehrloesser S, Coutandin DW, et al. Oocyte DNA damage quality control requires consecutive interplay of CHK2 and CK1 to activate p63. Nat Struct Mol Biol 2018; 25: 261-269.

Deutsch GB, Zielonka EM, Coutandin D, et al. DNA damage in oocytes induces a switch of the quality control factor TAp63α from dimer to tetramer. Cell 2011; 144: 566-576.

MacPartlin M, Zeng SX, Lu H. Phosphorylation and stabilization of TAp63gamma by IkappaB kinase-beta. J Biol Chem 2008; 283: 15754-15761.

Liao J-M, Zhang Y, Liao W, et al. IκB kinase β (IKKβ) inhibits p63 isoform γ (TAp63γ) transcriptional activity. J Biol Chem 2013; 288: 18184-18193.

Yabuta N, Ota C, Sasakura T, et al. Late cornified envelope 1C (LCE1C), a transcriptional target of TAp63 phosphorylated at T46/T281, interacts with PRMT5. Sci Rep 2018; 8: 4892.

Komatsu S, Takenobu H, Ozaki T, et al. Plk1 regulates liver tumor cell death by phosphorylation of TAp63. Oncogene 2009; 28: 3631-3641.

Li D, Li C, Wu M, et al. PKCδ stabilizes TAp63 to promote cell apoptosis. FEBS Lett 2015; 589: 2094-2099.

Armstrong SR, Wu H, Wang B, et al. The regulation of tumor suppressor p63 by the ubiquitin-proteasome system. Int J Mol Sci 2016; 17: 2041.

Rokudai S, Li Y, Otaka Y, et al. STXBP4 regulates APC/C-mediated p63 turnover and drives squamous cell carcinogenesis. Proc Natl Acad Sci U S A 2018; 115: E4806-E4814.

Caratozzolo MF, Marzano F, Abbrescia DI, et al. TRIM8 blunts the pro-proliferative action of ΔNp63α in a p53 wild-type background. Front Oncol 2019; 9: 1154.

He Y, Tai S, Deng M, et al. Metformin and 4SC-202 synergistically promote intrinsic cell apoptosis by accelerating ΔNp63 ubiquitination and degradation in oral squamous cell carcinoma. Cancer Med 2019; 8: 3479-3490.

He H, Peng Y, Fan S, et al. Cullin3/KCTD5 induces monoubiquitination of ΔNp63α and impairs its activity. FEBS Lett 2018; 592: 2334-2340.

Bilguun E-O, Kaira K, Kawabata-Iwakawa R, et al. Distinctive roles of syntaxin binding protein 4 and its action target, TP63, in lung squamous cell carcinoma: a theranostic study for the precision medicine. BMC Cancer 2020; 20: 935.

Prieto-Garcia C, Hartmann O, Reissland M, et al. Maintaining protein stability of ∆Np63 via USP28 is required by squamous cancer cells. EMBO Mol Med 2020; 12: e11101.

Ranieri M, Vivo M, De Simone M, et al. Sumoylation and ubiquitylation crosstalk in the control of ΔNp63α protein stability. Gene 2018; 645: 34-40.

Asatsuma-Okumura T, Ando H, De Simone M, et al. p63 is a cereblon substrate involved in thalidomide teratogenicity. Nat Chem Biol 2019; 15: 1077-1084.

Jeon YJ, Jo MG, Yoo HM, et al. Chemosensitivity is controlled by p63 modification with ubiquitin-like protein ISG15. J Clin Invest 2012; 122: 2622-2636.

MacPartlin M, Zeng S, Lee H, et al. p300 regulates p63 transcriptional activity. J Biol Chem 2005; 280: 30604-30610.

Ianakiev P, Kilpatrick MW, Toudjarska I, et al. Split-hand/split-foot malformation is caused by mutations in the p63 gene on 3q27. Am J Hum Genet 2000; 67: 59-66.

Stacy AJ, Zhang J, Craig MP, et al. TIP60 up-regulates ΔNp63α to promote cellular proliferation. J Biol Chem 2019; 294: 17007-17016.

Heyne K, Willnecker V, Schneider J, et al. NIR, an inhibitor of histone acetyltransferases, regulates transcription factor TAp63 and is controlled by the cell cycle. Nucleic Acids Res 2010; 38: 3159-3171.

LeBoeuf M, Terrell A, Trivedi S, et al. Hdac1 and Hdac2 act redundantly to control p63 and p53 functions in epidermal progenitor cells. Dev Cell 2010; 19: 807-818.

Ramsey MR, He L, Forster N, et al. Physical association of HDAC1 and HDAC2 with p63 mediates transcriptional repression and tumor maintenance in squamous cell carcinoma. Cancer Res 2011; 71: 4373-4379.

Fomenkov A, Zangen R, Huang Y-P, et al. RACK1 and stratifin target ΔNp63α for a proteasome degradation in head and neck squamous cell carcinoma cells upon DNA damage. Cell Cycle 2004; 3: 1285-1295.

Napoli M, Venkatanarayan A, Raulji P, et al. ΔNp63/DGCR8-dependent microRNAs mediate therapeutic efficacy of HDAC inhibitors in cancer. Cancer Cell 2016; 29: 874-888.

Yi Y, Chen D, Ao J, et al. Metformin promotes AMP-activated protein kinase-independent suppression of ΔNp63α protein expression and inhibits cancer cell viability. J Biol Chem 2017; 292: 5253-5261.

Zangen R, Ratovitski E, Sidransky D. ΔNp63α levels correlate with clinical tumor response to cisplatin. Cell Cycle 2005; 4: 1313-1315.

Rocca A, Viale G, Gelber RD, et al. Pathologic complete remission rate after cisplatin-based primary chemotherapy in breast cancer: correlation with p63 expression. Cancer Chemother Pharmacol 2008; 61: 965-971.

Rocco JW, Leong C-O, Kuperwasser N, et al. p63 mediates survival in squamous cell carcinoma by suppression of p73-dependent apoptosis. Cancer Cell 2006; 9: 45-56.

Isakoff SJ, Mayer EL, He L, et al. TBCRC009: a multicenter phase II clinical trial of platinum monotherapy with biomarker assessment in metastatic triple-negative breast cancer. J Clin Oncol 2015; 33: 1902-1909.

Gunaratne PH, Pan Y, Rao AK, et al. Activating p53 family member TAp63: a novel therapeutic strategy for targeting p53-altered tumors. Cancer 2019; 125: 2409-2422.

Galoczová M, Nenutil R, Coates P, et al. Possible usage of p63 in bioptic diagnostics. Klin Onkol 2018; 31: 27-31.

Abbas HA, Bui NHB, Rajapakshe K, et al. Distinct TP63 isoform-driven transcriptional signatures predict tumor progression and clinical outcomes. Cancer Res 2018; 78: 451-462.

Bankhead A 3rd, McMaster T, Wang Y, et al. TP63 isoform expression is linked with distinct clinical outcomes in cancer. EBioMedicine 2020; 51: 102561.

Rosenbluth JM, Johnson K, Tang L, et al. Evaluation of p63 and p73 antibodies for cross-reactivity. Cell Cycle 2009; 8: 3702-3706.

Baker M. When antibodies mislead: the quest for validation. Nature 2020; 585: 313-314.

Andersson S, Sundberg M, Pristovsek N, et al. Insufficient antibody validation challenges oestrogen receptor beta research. Nat Commun 2017; 8: 15840.

Belyi VA, Levine AJ. One billion years of p53/p63/p73 evolution. Proc Natl Acad Sci U S A 2009; 106: 17609-17610.

Chillemi G, Kehrloesser S, Bernassola F, et al. Structural evolution and dynamics of the p53 proteins. Cold Spring Harb Perspect Med 2017; 7: a028308.

Wang C, Teo CR, Sabapathy K. p53-related transcription targets of TAp73 in cancer cells - bona fide or distorted reality? Int J Mol Sci 2020; 21: 1346.

Mellacheruvu D, Wright Z, Couzens AL, et al. The CRAPome: a contaminant repository for affinity purification-mass spectrometry data. Nat Methods 2013; 10: 730-736.

Monaco G, Lee B, Xu W, et al. RNA-Seq signatures normalized by mRNA abundance allow absolute deconvolution of human immune cell types. Cell Rep 2019; 26: 1627-1640.e7.

Orzol P, Nekulova M, Holcakova J, et al. ΔNp63 regulates cell proliferation, differentiation, adhesion, and migration in the BL2 subtype of basal-like breast cancer. Tumour Biol 2016; 37: 10133-10140.

Zhang J, Jun Cho S, Chen X. RNPC1, an RNA-binding protein and a target of the p53 family, regulates p63 expression through mRNA stability. Proc Natl Acad Sci U S A 2010; 107: 9614-9619.

Xu E, Zhang J, Zhang M, et al. RNA-binding protein RBM24 regulates p63 expression via mRNA stability. Mol Cancer Res 2014; 12: 359-369.

Yan W, Zhang Y, Chen X. TAp63γ and ΔNp63γ are regulated by RBM38 via mRNA stability and have an opposing function in growth suppression. Oncotarget 2017; 8: 78327-78339.

Zhang Y, Feng X, Sun W, et al. Serine 195 phosphorylation in the RNA-binding protein Rbm38 increases p63 expression by modulating Rbm38's interaction with the Ago2-miR203 complex. J Biol Chem 2019; 294: 2449-2459.

Yan W, Zhang Y, Zhang J, et al. HuR is necessary for mammary epithelial cell proliferation and polarity at least in part via ΔNp63. PLoS One 2012; 7: e45336.

Cho S-J, Jung Y-S, Chen X. Poly (C)-binding protein 1 regulates p63 expression through mRNA stability. PLoS One 2013; 8: e71724.

Mohibi S, Zhang J, Chen X. PABPN1, a target of p63, modulates keratinocyte differentiation through regulation of p63α mRNA translation. J Invest Dermatol 2020; 140: 2166-2177.e6.

Zhang Y, Feng X, Zhang J, et al. Iron regulatory protein 2 exerts its oncogenic activities by suppressing TAp63 expression. Mol Cancer Res 2020; 18: 1039-1049.

Huang Y, Chuang AY, Romano R-A, et al. Phospho-ΔNp63α/NF-Y protein complex transcriptionally regulates DDIT3 expression in squamous cell carcinoma cells upon cisplatin exposure. Cell Cycle 2010; 9: 328-338.

Melino S, Bellomaria A, Nepravishta R, et al. p63 threonine phosphorylation signals the interaction with the WW domain of the E3 ligase Itch. Cell Cycle 2014; 13: 3207-3217.

Li Y, Peart MJ, Prives C. Stxbp4 regulates ΔNp63 stability by suppression of RACK1-dependent degradation. Mol Cell Biol 2009; 29: 3953-3963.

Huang Y-P, Wu G, Guo Z, et al. Altered sumoylation of p63α contributes to the split-hand/foot malformation phenotype. Cell Cycle 2004; 3: 1587-1596.

Ghioni P, D'Alessandra Y, Mansueto G, et al. The protein stability and transcriptional activity of p63α are regulated by SUMO-1 conjugation. Cell Cycle 2005; 4: 183-190.

Najít záznam

Citační ukazatele

Nahrávání dat ...

Možnosti archivace

Nahrávání dat ...