<<

Central Journal of Dermatology and Clinical Research

Review Article *Corresponding author Edward A Ratovitski, Head and Neck Research Division, Johns Hopkins University School of Medicine, Tumor P63 is a Key Cancer Research Building-II, Room 5M05, 1550 Orleans Street, Baltimore, MD 21231, USA. Tel: 410-491-9802; Fax: 410-614-1411; E-mail: [email protected] Regulator of Skin Functions in Submitted: 29 October 2013 Accepted: 31 October 2013 Ectodermal Dysplasia Published: 04 November 2013 Copyright Edward A Ratovitski* © 2013 Ratovitski Head and Neck Cancer Research Division, Johns Hopkins University School of Medicine, USA OPEN ACCESS

Abstract Keywords Tumor protein (TP)-p63 has been discovered as TP53 homolog more than fifteen • p63 years ago and has become a master regulator of skin development, proliferation and stem • MicroRNA maintenance. While TP53 is known to be the most mutated in human cancer, TP63 • are mostly associated with the various types of ectodermal dysplasia. All TP53 • arrest family members, TP53, TP63 and TP73, function as factors that regulate the cell • cycle arrest, apoptosis, autophagy or metabolism through activation/repression of downstream • Epigenetics target or protein-protein interactions with other protein regulators of transcription and • Keratinocytes splicing. Several downstream target genes or protein interactors of TP63 are involved in the • Ectodermal dysplasia molecular mechanisms underlying the ectodermal dysplasia phenotypes. This mini-review underlines a few venues of investigations about the key role for TP63 in skin biology and pathology.

INTRODUCTION TP63 exert their functions through the transcriptional regulation (activation of repression) by binding Multiple molecular mechanisms are underlying the skin and differentiation, while the master gene in the direct control of the skin epithelial sheet architecture, stem cellto the renewal, specific cell downstream cycle arrest, gene apoptosis, targets, senescence,many of them autophagy, involved genes, tumor protein (TP)-p63 is being investigated for the last skin aging, cellular response to ultraviolet (UV)- irradiation, decadeinvolved [1-4]. in genetic TP63 andgene epigenetic encoding regulationtranscription of factors skin-specific with thereby contributing to the molecular pathogenesis of several mutated in cancer, however is often mutated in heritable skin geneticand epigenetic diseases controls of skin of and thereof epithelial via tissues and [15-26]. microRNAs, Among disorders,the transactivation such as ectodermaldomain (TA-) dysplasia and without [3,4]. it TP63 (ΔN-), mutations is rarely TP63 responsive gene targets are the following genes implicated can cause several different disorders: ectrodactyly, ectodermal in skin development and functions: ALOX12B, AQP3, BPAG1, dysplasia and cleft lip/palate syndrome (EEC), ankyloblepharon- CASP14, CDH1, CLDN1, DLX3, 5 and 6, DSG, DSP, EVL, HBP1, IKK, ectodermal defects-cleft lip/palate syndrome (AEC), limb IVL, IRF6, KRT5, and 14, LSH, PPL, PERP, SATB1, TPRG1, and mammary syndrome (LMS), Acro-Dermato-Ungual-Lacrimal- ZNF750 [17,20,21,27-37]. Tooth syndrome (ADULT), Rapp- Hodgkin syndrome (RHS), and split hand/foot malformation (SHFM4) [1,3,4]. Each group TP63 also engages in numerous protein-protein interactions

ability to bind TP63 responsive elements in gene promoters splicing, protein degradation, and signaling [6-8,10,38-40]. TP63 of mutations affects specific functions of TP63 protein, such its that contribute to chromatin remodeling, RNA transcription, RNA and to form protein/protein complexes with other regulatory of gene expression. For example, the AEC-derived TP63 components(DNA-binding (SAM domain), domain), to be as properly reviewed ubiquitinated in [5-10]. (TI-domain), mutationsassociates with altered many the proteins ability involved for TP63 in toa fine-tuned physically regulation associate The key studies show that null mice display single- leading to a deregulated splicing of FGFR2 responsible forwith epithelial/mesenchymal mRNA processing/splicing transition proteins (EMT) (e.g. [6,8]. ABBP1, AEC-derived SRA4) leading to a breakage of the epidermal barrier formation that layered and translucent skin, with an altered stratified epithelia mutations in TP63 were also found to alter the TP63 binding to

protects against dehydration [11,12]. Homozygous ΔNp63 null palate, and down regulation of PERP [38,39]. disorganized epithelial cells exhibiting a premature expression SATB2, which has been implicated in the development of cleft mice show under-developed stratified epidermis with clusters of TP63 is over expressed in multiple human epithelial , expressing transgenic mice show an abnormal hair follicle including head and neck, esophagus and lung cancers, cervix and developmentof terminal differentiationand altered cell markersfate of follicular [13]. However,keratinocytes ΔNp63α [14]. breast tumors [40]. Accumulating evidence shows that TP63

Cite this article: Ratovitski EA (2013) Tumor Protein P63 is a Key Regulator of Skin Functions in Ectodermal Dysplasia. J Dermatolog Clin Res 1(1): 1006. Ratovitski (2013) Email: [email protected] Central and especially its ∆ ∆ N isoforms play a critical role in head and remodeling complexes, noncoding microRNAs implicated in neck squamous cell carcinomas (HNSCC). Np63a, the most regulationAccumulating of RNA transcription evidence shows and thatRNA epigeneticprocessing mechanisms[50]. predominantly expressed isoform in HNSCC, is over expressed are involved in the control of epidermal development and andRole sometimes of autophagy amplified in cutaneousin HNSCC tumors functions [41]. and TP63 terminal keratinocyte differentiation [51]. Multiple elements of First indication of autophagy in skin cells was produced almost thirty years ago showing the melanin macro globule (MMG), epigenetic machinery (e.g. DNMT1, JMJD3, SETD8, HDAC1 and 2, formerly called “macromelanosome,” a cytoplasmic spherical underlyingEHMT1 and epidermal2, BRG1, CHD1, differentiation 3, and 4, BMI1, programs, CBX2, as4, and reviewed 6, EZH2, in granule formed in the melanocyte [42]. MMG are retained within JARID2, and SATB1) were implicated in molecular mechanisms melanocytes or transferred to keratinocytes and Langerhans cells number of genes involved in the control of nuclear/chromatin in the epidermis, and macrophages in the dermis, where they [51]. In the epidermis of p63-null mice the expression of a large can fuse with other heterolysosomes [41]. Cutaneous malignant wild-type controls [39]. On one hand, TP63 directly controls the melanomas often exhibit pigmented regions that are darker assembly and remodeling is significantly changed compared to than the surrounding skin [43]. While melanoma cells are the order chromatin structure in central domain of the epidermal original source of the melanin, keratinocytes and melanophages SATB1 transcription and promotes establishing of specific higher- also contribute to the tumor color because they contain melanin balance of gene expression during terminal keratinocyte obtained from melanoma cells [42]. Moreover, autophagy differentiation complex [39]. On required the other for hand,maintenance TP63 expressionof the proper in appears to be a common trait of invasive melanoma cells in the keratinocytes is regulated by histone methyltransferase SETD8, dermis [42]. Strikingly, the Caucasian skin keratinocytes exhibit which, in turn, is a target of c- and higher autophagic activity than those from African American skin mediates its effects on epidermal differentiation [52]. Another [42]. Melanosome accumulation in keratinocytes was accelerated by treatment with lysosomal inhibitors or with small interfering remodeling ATPase encoded by the HELLS gene, is essential for TP63 transcriptional target LSH, an SNF2-like chromatin essential for autophagy supporting its pivotal role in skin color determination,RNAs specific as for well autophagy-related as in chemo- or proteins, immuno- therapeuticwhich are normal levels of DNA and to drive skin stem cell treatment of melanoma patients [43-45]. proliferation and tumorigenesis [40]. LSH acts as an efficient TP63transcriptional was shown repressor to play a bycritical cooperating role in regulation with the of DNMT1,various epigeneticDNMT3B, HDAC1 enzymes and andHDAC2 accessory to silence proteins gene transcription by activating [40]. or UVB, a major cause of skin damage, which accompanies repressing their transcription, as well as modulating their levels showncomplex to alterationsinduce macroautophagy in irradiated and skin mitochondrial cells (e.g. DNA autophagy lesions, [46,47].oxidative Furthermore, stress, inflammation the epidermal and caspase expression activation), of SQSTM1, was also the TP63by TP63-dependent and microRNAs microRNAs in [53], skin Ratovitski, proliferation in preparation]. and differentiation psoriatic skin than in skin affected by atopic dermatitis or from healthykey regulator controls, of autophagy, suggesting isthe found role tofor be autophagy significantly in cutaneous higher in and migration of skin stem cells supported by the function induced autophagy in epidermal keratinocytes leading to of ArgonauteEssential role (AGO) of proteins microRNAs as essentialin governing components a self-renewal of the ainflammation massive accumulation [48]. Both UVA- of and high-molecular-weight UVA-oxidized phospholipids protein AGO1 and 2 aggregates containing the autophagy-adaptor protein SQSTM1 in ATG7-/- keratinocytes [49]. RNA-induced silencing complex [54]. When both are ablated in the skin, the global expression of microRNAs is onsignificantly epidermal compromised and hair and development causes severe was defects demonstrated in skin displayed a dramatic ATM-dependent of morphogenesis [54]. The overall profound impact of microRNAs Squamous cell carcinoma (SCC) cells exposed to cisplatin

by generating mice with skin-specific targeting of theDICER two ΔNp63α leading to a transcriptional regulation of downstream major components of the microRNA processing machinery, mRNA and microRNA gene targets in SCC cells [26,49]. and DGCR8 resulted in dramatic and similar skin phenotypes expression (e.g. ATG1/ULK1, ATG3, ATG4A, ATG5, ATG6/BECN1, affectingDICER and the DGCR8.follicular Keratinocyte-specificepithelium and inter-follicular deletion epidermis of ATG7,Phosphorylated ATG9A and ΔNp63α ATG10 )was in cisplatin-treatedshown to regulate SCC autophagic cells through gene both transcriptional and post-transcriptional mechanisms transcription of DICER, while directly repressing the expression [26,50]. of[55,56]. miR-34 Intriguingly, encoding genes both [57-60]. TAp63 and ΔNp63 directly regulate Epigenetics, skin cell malfunction, and TP63 Epigenetic regulation mediates genomic adaption to the keratinocyte cell cycle progression and the expression of several environment and epigenetic alterations (heritable changes in Inhibition of miR-34a and miR-34c activity restored is modulated by miR-720 and miR-574-3p in the suprabasal through changes in chromatin structure) can contribute to the layerscell cycle of the regulators, epidermis CCND1 [56]. and MiR-205 CDK4 is [60]. broadly TP63 expressed expression in developmentgene function of that disease occur phenotypes, without any as change can mutations in DNA sequence[50]. The layers and skin stem cells [60]. Genetic deletion of miR-205 causesthe epidermis, neonatal includinglethality with the basal,severely suprabasal compromised and superficialepidermal epigenetic molecular mechanisms include DNA methylation, modifications of histones, histone code, chromatin and polycomb J Dermatolog Clin Res 1(1): 1006 (2013) 2/6 Ratovitski (2013) Email: [email protected] Central

negative effect on the wild-type TP63 protein [69]. The AEC TP63 mutations exert a selective dominant-negative function on and hair follicle growth [60]. In the miR-205 knockout skin cells, phospho-AKT is significantly down regulated, while cells factor signaling resulting from reduced expression of FGFR2 and prematurely exit the cell cycle and become quiescent [60]. MiR- 3wild-type TP63 associated with impairment of fibroblastin vitro growth and in of205 skin targets cells negative [59]. Moreover, regulators the of lossPI(3)K of miR-205signaling isthat associated mediate vivo studies of the potential biological treatments modulating the withthe repression melanoma of progression phospho-AKT [61]. and MiR-205 restrict was the also proliferation reported effect, direct of AEC-derivedTP63 target genesTP63 mutations [68]. In summary, on FGFR2b the splicing needed to promote keratinocyte migration through downregulation of to shed a light on the AEC pathogenesis. AEC mouse models also display a deregulation of the TP63 MiR-31 is highly expressed in ‘activated’ skin conditions (e.g. woundlipid phosphatase healing, , INPPL1 and and modulating psoriasis), AKT and signaling elevated miR- [62]. 31 levels in a transgenic mouse model results in aberrant wound istranscriptional essential for thetarget barrier ZNF750, function which of is skin associated [70]. TP63 with with familial AEC healing and hair loss [56,63]. MiR-31 is also present in anagen psoriasis [36,70]. ZNF750 is reported to induce KLF4, which hair follicles and exerts inhibitory effects on hair follicle growth an organotypic human tissue model, while the forced expression by targeting multiple genes [56,63]. mutations prevented the transcriptional induction of ZNF750 in MiR-200a, miR-200b, miR-200c, miR-141, miR-429, miR-19b, miR-20, miR-17-5p, and miR-93 are preferentially expressed in of ZNF750 rescued impaired epidermal differentiation resulting the epidermis, while miR-199a, and miR-199b are exclusively [36,70].from AEC Among mutants the suggesting genes that the deregulated existence of in a AEC p63–ZNF750– are those expressed in the hair follicle, as reviewed in [56]. The skin- implicatedKLF4 network in in epidermal human epidermal adhesion, development skin barrier and formation homeostasis and hair follicle biology (e.g. FRAS1, COL7A, CDH3, SPRR1A and 4, of the epidermis, hair follicle and sebaceous gland in mice, as LCE5A, HRNR, ALOX12B, GATA3, KRT 25, 27, 33B, 34, 35, 81 and specific miR-203 is abundantly expressed in differentiating cells 85, and PERP), some of them are controlled directly or indirectly by TP63 [70-72]. molecularwell as in theswitch suprabasal between epidermal proliferative layer basal in human cells and skin terminally [64]. By differentiatedtargeting ΔNp63, suprabasal miR-203 keratinocytes may control [65]. ‘stemness’ and act as a FUTURE STUDIES Among the ninety genes associated with the genetic skin transcription regulated by TP53 family factors (TP53, TP63, and Accumulating evidence supports that microRNAs, whose TP73), could contribute in multiple signaling pathways involved in disorders, several functional pathways were defined to play a cell cycle arrest, apoptosis, autophagy, metabolism and epigenetic 70 TP63role in exerts epidermal its function differentiation, through including a transcriptional NOTCH, TGFβ, regulation IKK, transcriptional regulation, thereby potentially underlying the MAPK, PI3K, TP63, and WNT), as reviewed in [ ]. In general, mechanisms leading to epithelial cell maintenance, and tumor of downstream gene targets. For example, the deregulation of development and chemoresistance [50]. As a member of the KRT1, 5, 10, 14, and PLEC1 is implicated in epidermolysis bullosa TP53 family, TP63 transcription factor is likely to play its decisive simplex or epidermolytic ichthyosis altering the basement role in transcriptional and post-transcriptional regulation of membrane zone adhesion and hemidesmosomes and causing epidermal blistering [70]. Deregulation of ALOX12B was shown to cause lamellar ichthyosis affecting cholesterol metabolism andmicroRNAs -130b expression in epithelial by cancers, binding epithelial directly to differentiation TP63-responsive and [70], while CLDN1 is involved in neonatal ichthyosis sclerosing elementsEMT [50]. located ΔNp63α in wasclose shown proximity to inhibit to the miR-138, genomic -181a, loci of -181b, these cholangitis syndrome showing ichthyosis, hypotrichosis, and dental abnormalities [70]. KRT1, 9, 16 and 17 are implicated in palmoplantar keratoderma and pachyonychia congenital microRNAsThe AEC syndrome in primary keratinocytesand TP63 [66]. showing alteration if desmosomes affecting palms, soles, and hair and causing nail dystrophy [70]. Mutations in the TP63-regulated The most severe cutaneous manifestation of the AEC is the CDH3 long-lasting skin fragility with severe skin erosions after birth ectodermal dysplasia, ectrodactyly, macular dystrophy altering [67-69]. Additionally, the skin of the AEC patients shows an cause hypotrichosis with juvenile macular dystrophy and impaired expression of differentiation markers involved in TP63, as well as miR-203, miR-21 and miR-125b implicated cell adhesion and barrier formation, as well as a dysregulated indevelopment the regulation of the of hair TP63 and or retinaTP53, has[71-73]. been Interestingly, suggested in that the cell cycle regulation [67,68]. The new knock-in mouse AEC pathogenesis of psoriasis [74-77]. models were recently offered for investigation of the molecular mechanisms underlying the AEC, as well as providing potential Disrupted skin barrier due to altered keratinocyte therapeutic approaches [67-69]. These models show the skin differentiation is common in pathologic conditions such as fragility associated with microscopic blistering between the basal atopic dermatitis, ichthyosis and psoriasis [78]. The dominant and suprabasal compartments of the epidermis and reduced in the TP63 target ZNF750 leads to a clinical phenotype desmosomal contacts [69]. Expression of desmosomal CDH reminiscent of psoriasis and seborrheic dermatitis [78]. and DSP was strongly reduced in knock-in mouse AEC mutant Furthermore, the deregulation of the TP63/hedgehog cross keratinocytes and in newborn epidermis similarly to human talk was reported to underlie the pathogenesis of basal cell keratinocytes isolated from AEC patients, in TP63-depleted carcinoma of the skin through regulation of SUFU expression keratinocytes and in p63 null embryonic skin, indicating that [78 TP63 mutations causative of AEC syndrome have a dominant- molecular pathways underlying the role for TP63 transcriptional ]. Evidently, the additional studies needed to define the J Dermatolog Clin Res 1(1): 1006 (2013) 3/6 Ratovitski (2013) Email: [email protected] Central targets in the pathogenesis of the above-mentioned high 15. Truon morbidity skin abnormalities that could be targeted as potential proliferation and differentiation of developmentally mature keratinocytes.g AB, Kretz Genes M, RidkyDev. 2006; TW, Kimmel20: 3185-3197. R, Khavari PA. p63 regulates reagents [50,56,63,66,79]. Finally, the TP63 antibodies have been 16. provenfields ofto be intervention useful tool in using the diagnosis TP63-related of a primary biomolecules cutaneous or carcinosarcoma, spindle cell SCC, and in distinguishing sclerosing Viganò MA, Lamartine J, Testoni B, Merico D, Alotto D, Castagnoli C, et al. New p63 targets in keratinocytes identified by a genome-wide cutaneous tumors [79-82]. 17. approach. EMBO J. 2006; 25: 5105-5116. ACKNOWLEDGEMENTS Birkaya B, Ortt K, Sinha S. Novel in vivo targets of DeltaNp63 in keratinocytes identified by a modified chromatin immunoprecipitation This study was supported in part by the Flight Attendant 18. approach. BMC Mol Biol. 2007; 8: 43. (#082469). regulation of epithelial integrity and cancer. Cell Cycle. 2007; 6: 240- 245.Trink B, Osada M, Ratovitski E, Sidransky D. p63 transcriptional ResearchREFERENCES Institutions grant 19. 1. Heterozygous germline mutations in the homolog p63 are the 129:Senoo 523-536. M, Pinto F, Crum CP, McKeon F. p63 Is essential for the causeCelli J, of Duijf EEC syndrome. P, Hamel BC,Cell. Bamshad 1999; 99: M, 143-153. Kramer B, Smits AP, et al. proliferative potential of stem cells in stratified epithelia. Cell. 2007; 20. 2. Mikkola ML. p63 in skin appendage development. Cell Cycle. 2007; 6: 285-290. Koster MI, Dai D, Marinari B, Sano Y, Costanzo A, Karin M, et al. p63 3. induces key target genes required for epidermal morphogenesis. Proc 21. Cycle. 2007; 6: 262-268. Natl Acad Sci U S A. 2007; 104: 3255-3260. Rinne T, Brunner HG, van Bokhoven H. p63-associated disorders. Cell Transcriptional network of p63 in human keratinocytes. PLoS One. 4. 2009;Pozzi S,4: 5008. Zambelli F, Merico D, Pavesi G, Robert A, Maltère P, et al.

Vanbokhoven H, Melino G, Candi E, Declercq W. p63, a story of mice 22. 5. and men. J Invest Dermatol. 2011; 131: 1196-1207. al. Differential altered stability and transcriptional activity of ∆ Guo X, Keyes WM, Papazoglu C, Zuber J, Li W, Lowe SW, et al. TAp63 Browne G, Cipollone R, Lena AM, Serra V, Zhou H, van Bokhoven H, et induces senescence and suppresses tumorigenesis in vivo. Nat Cell 23. 2207. Np63 Biol. 2009; 11: 1451-1457. mutants in distinct ectodermal dysplasias. J Cell Sci. 2011; 124: 2200- ∆ α 6. promotesHa L, Ponnamperuma malignant conversion RM, Jay S, Ricci of keratinocytes. MS, Weinberg PLoS WC. One.Dysregulated 2011; 6: Fomenkova T, et al. P63 alpha mutations lead to aberrant splicing of e21877.Np63 ± inhibits expression of Ink4a/arf, blocks senescence, and Fomenkov A, Huang YP, Topaloglu O, Brechman A, Osada M, Chem. 2003; 278: 23906-23914. 24. Paris M, Rouleau M, Pucéat M, Aberdam D. Regulation of skin aging keratinocyte receptor in the Hay-Wells syndrome. J Biol and heart development by TAp63. Cell Death Differ. 2012; 19: 186- 7. 193. Altered sumoylation of p63alpha contributes to the split-hand/foot malformationHuang YP, Wu phenotype. G, Guo Z, Cell Osada Cycle. M, 2004; Fomenkov 3: 1587-1596. T, Park HL, et al. 25.

8. embryonicMedawar A, stem Virolle cells. T, RostagnoPLoS One. P, 2008; de la 3:Forest-Divonne e3441. S, Gambaro K, AEC-associated p63 mutations lead to alternative splicing/protein Rouleau M, et al. DeltaNp63 is essential for epidermal commitment of Huang YP, Kim Y, Li Z, Fomenkov T, Fomenkov A, Ratovitski EA. 26. Huang Y, Guerrero-Preston R, Ratovitski EA. Phospho-∆ α- 2005; 4: 1440-1447. dependent regulation of autophagic signaling through transcription stabilization of p63 and modulation of Notch signaling. Cell Cycle. Np63 9. Vivo M, Di Costanzo A, Fortugno P, Pollice A, Calabrò V, La Mantia 27. and micro-RNA modulation. Cell Cycle. 2012; 11: 1247-1259. RT, et al. Perp is a p63-regulated gene essential for epithelial integrity. Ihrie RA, Marques MR, Nguyen BT, Horner JS, Papazoglu C, Bronson 3537-3543.G. Downregulation of DeltaNp63alpha in keratinocytes by p14ARF- Cell. 2005; 120: 843-856. mediated SUMO-conjugation and degradation. Cell Cycle. 2009; 8: 10. Rossi M, De Simone M, Pollice A, Santoro R, La Mantia G, Guerrini L, et 28. WC, et al. Homeobox gene Dlx3 is regulated by p63 during ectoderm Cell Cycle. 2006; 5: 1816-1822. development:Radoja N, Guerrini relevance L, Lo in Iacono the pathogenesis N, Merlo GR, of ectodermalCostanzo A, dysplasias. Weinberg al. Itch/AIP4 associates with and promotes p63 protein degradation. Development. 2007; 134: 13-18. 11. 29. 1999;Mills AA, 398: Zheng 708-713. B, Wang XJ, Vogel H, Roop DR, Bradley A. p63 is a p53 et al. Regulation of Dlx5 and Dlx6 gene expression by p63 is involved homologue required for limb and epidermal morphogenesis. Nature. inLo EECIacono and N, SHFM Mantero congenital S, Chiarelli limb A,defects. Garcia Development. E, Mills AA, Morasso 2008; 135: MI, 12. 1377-1388. p63 is essential for regenerative proliferation in limb, craniofacial and Yang A, Schweitzer R, Sun D, Kaghad M, Walker N, Bronson RT, et al. 30. et al. Claudin-1 is a p63 target gene with a crucial role in epithelial 13. epithelial development. Nature. 1999; 398: 714-718. development.Lopardo T, Lo PLoS Iacono One. N, 2008;Marinari 3: e2715. B, Giustizieri ML, Cyr DG, Merlo G, et al. ∆ Romanoregulator RA,of epithelial Smalley K,development Magraw C, and Serna differentiation. VA, Kurita T, Development. Raghavan S, 31. 2012; 139:Np63 772-782. knockout mice reveal its indispensable role as a master andRomano directing RA, Ortt epidermal K, Birkaya cell B,fate. Smalley PLoS K,One. Sinha 2009; S. An 4: e5623.active role of the 14. DeltaN isoform of p63 in regulating basal keratin genes K5 and K14 development and altered cell fate of follicular keratinocytes in 32. Romano RA, Smalley K, Liu S, Sinha S. Abnormal hair follicle 137: 1431-1439. ofMarinari ectodermal B, Ballaro dysplas C, Koster MI, Giustizieri ML, Moretti F, Crosti F, et al. transgenic mice expressing DeltaNp63alpha. Development. 2010; IKKalpha is a p63 transcriptional target involved in the pathogenesis ias. J Invest Dermatol. 2009; 129: 60-69. J Dermatolog Clin Res 1(1): 1006 (2013) 4/6 Ratovitski (2013) Email: [email protected] Central

33. Antonini D, Dentice M, Mahtani P, De Rosa L, Della Gatta G, Mandinova Autophagy is induced by UVA and promotes removal of oxidized A, et al. Tprg, a gene predominantly expressed in skin, is a direct target

1685. phospholipids and protein aggregates in epidermal keratinocytes. J of the transcription factor p63. J Invest Dermatol. 2008; 128: 1676- 50. Invest Dermatol. 2013; 133: 1629-1637. 34. cancer cells. Curr Genomics. 2013; 14: 441-452. Ratovitski EA. Tumor Protein p63/microRNA network in epithelial 51. DeathBorrelli Differ. S, Candi 2010; E, Hu17: B, 1896-1907. Dolfini D, Ravo M, Grober OM, et al. The p63 target HBP1 is required for skin differentiation and stratification. Cell 35. Dermatol.Botchkarev 2012; VA, 132: Gdula 2505-2521. MR, Mardaryev AN, Sharov AA, Fessing MY. Epigenetic regulation of gene expression in keratinocytes. J Invest Thomason HA, Zhou H, Kouwenhoven EN, Dotto GP, Restivo G, Nguyen 52. BC, et al. Cooperation between the transcription factors p63 and IRF6 histone methyltransferase Setd8 acts in concert with c-Myc and is 1561-1569. Driskell I, Oda H, Blanco S, Nascimento E, Humphreys P, Frye M. The is essential to prevent cleft palate in mice. J Clin Invest. 2010; 120: 36. 53. required to maintain skin. EMBO J. 2012; 31: 616-629. Zarnegar BJ, Webster DE, Lopez-Pajares V, Vander Stoep Hunt B, Qu Phospho-∆ α Huang Y, Kesselman D, Kizub D, Guerrero-Preston R, Ratovitski EA. K, Yan KJ, et al. Genomic profiling of a human organotypic model of carcinoma cells upon cisplatin exposure. Cell Cycle. 2013; 12: 684- Np63 ±/microRNA feedback regulation in squamous AEC syndrome reveals ZNF750 as an essential downstream target of 697. 37. mutant TP63. Am J Hum Genet. 2012; 91: 435-443. 54. Ratovitski EA. Phospho-ΔNp63α regulates AQP3, ALOX12B, CASP14 al. Quantitative functions of Argonaute proteins in mammalian Wang D, Zhang Z, O’Loughlin E, Lee T, Houel S, O’Carroll D, et and CLDN1 expression through transcription and microRNA development. Genes Dev. 2012; 26: 693-704. 38. modulation. FEBS Lett. 2013; 587: 3581-6. 55. Chung J, Grant RI, Kaplan DR, Irwin MS. Special AT-rich binding Yi R, Pasolli HA, Landthaler M, Hafner M, Ojo T, Sheridan R, et protein-2 (SATB2) differentially affects disease-causing p63 mutant al. DGCR8-dependent microRNA biogenesis is essential for skin 39. proteins. J Biol Chem. 2011; 286: 40671-40680. 56. development. Proc Natl Acad Sci U S A. 2009; 106: 498-502. Fessing MY, Mardaryev AN, Gdula MR, Sharov AA, Sharova TY, of skin development and regeneration. Cell Cycle. 2012; 11; 468-74. Botchkareva NV. MicroRNA/mRNA regulatory networks in the control Rapisarda V, et al. p63 regulates Satb1 to control tissue-specific 57. chromatin remodeling during development of the epidermis. J Cell 40. Biol. 2011; 194: 825-839. Su X, Chakravarti D, Cho MS, Liu L, Gi YJ, Lin YL, et al. TAp63 suppresses metastasis through coordinate regulation of Dicer and miRNAs. Keyes WM, Pecoraro M, Aranda V, Vernersson-Lindahl E, Li W, Vogel to drive skin stem cell proliferation and tumorigenesis. Cell Stem Cell. 58. Nature. 2010; 467: 986-990. 2011;H, et al. 8: ΔNp63α 164-176. is an that targets chromatin remodeler Lsh ∆ α cancerHuang cells.Y, Chuang Cell Death A, Hao Differ. H, Talbot 2011; C, 18: Sen 1220-1230. T, Trink B, et al. Phospho- 41. Np63 ± is a key regulator of the cisplatin-induced microRNAome in 59. Antonini D, Russo MT, De Rosa L, Gorrese M, Del Vecchio L, Missero AcadHibi K, Sci Trink U S A. B, 2000; Patturajan 97: 5462-5467. M, Westra WH, Caballero OL, Hill DE, et al. C. Transcriptional repression of miR-34 family contributes to p63- AIS is an oncogene amplified in squamous cell carcinoma. Proc Natl 42. 2010; 130: 1249-1257. mediated cell cycle progression in epidermal cells. J Invest Dermatol. 134-139.Nakagawa H, Hori Y, Sato S, Fitzpatrick TB, Martuza RL. The nature 60. and origin of the melanin macroglobule. J Invest Dermatol. 1984; 83: 43. Wang D, Zhang Z, O’Loughlin E, Wang L, Fan X, Lai EC, et al. MicroRNA-205 controls neonatal expansion of skin stem cells by Murase D, Hachiya A, Takano K, Hicks R, Visscher MO, Kitahara T, 61. modulating the PI(3)K pathway. Nat Cell Biol. 2013; 15: 1153-1163. Dermatol.et al. Autophagy 2013; 133: has 2416-2424. a significant role in determining skin color by regulating melanosome degradation in keratinocytes. J Invest Liu S, Tetzlaff MT, Liu A, Liegl-Atzwanger B, Guo J, Xu X. Loss of 44. microRNA-205 expression is associated with melanoma progression. regulation of autophagy-related protein 5 (ATG5) contributes to the 62. Lab Invest. 2012; 92: 1084-1096. pathogenesisLiu H, He Z, von of early-stage Rütte T, Yousefi cutaneous S, Hunger melanoma. RE, Simon Sci Transl HU. Down- Med. 2013; 5: 202ra123. Yu J, Peng H, Ruan Q, Fatima A, Getsios S, Lavker RM. MicroRNA-205 promotes keratinocyte migration via the lipid phosphatase SHIP2. 45. 63. FASEB J. 2010; 24: 3950-3959.

360.Liu H, He Z, Simon HU. Targeting autophagy as a potential therapeutic Mardaryev AN, Ahmed MI, Vlahov NV, Fessing MY, Gill JH, Sharov AA, approach for melanoma therapy. Semin Cancer Biol. 2013; 23: 352- 3869-3881.et al. Micro-RNA-31 controls hair cycle-associated changes in gene 46. expression programs of the skin and hair follicle. FASEB J. 2010; 24: mitochondrial morphology in human melanoma-derived cells post 64. Zanchetta LM, Garcia A, Lyng F, Walsh J, Murphy JE. Mitophagy and Yi R, Poy MN, Stoffel M, Fuchs E. A skin microRNA promotes 47. exposure to simulated sunlight. Int J Radiat Biol. 2011; 87: 506-517. 65. differentiationLena AM, Shalom-Feuerstein by repressing ‘stemness’. R, Rivetti Nature. di Val Cervo 2008; P,452: Aberdam 225-229. D,

irradiationMisovic M, induces Milenkovic alteration D, Martinovic in cytoskeleton T, Ciric D, and Bumbasirevic autophagy inV, Knight RA, Melino G, et al. miR-203 represses ‘stemness’ by repressing humanKravic-Stevovic keratinocytes. T. Short-term Ultrastruct exposure Pathol. 2013; to UV-A, 37: 241-248. UV-B, and UV-C 66. DeltaNp63. Cell Death Differ. 2008; 15: 1187-1195. 48. AcadRivetti Sci di U Val S A. Cervo 2012; P, 109: Lena 1133-1138. AM, Nicoloso M, Rossi S, Mancini M, Zhou Lee HM, Shin DM, Yuk JM, Shi G, Choi DK, Lee SH, et al. Autophagy H, et al. p63-microRNA feedback in keratinocyte senescence. Proc Natl negatively regulates keratinocyte inflammatory responses via 67. 49. scaffolding protein p62/SQSTM1. J Immunol. 2011; 186: 1248-1258. Pourreyron C, et al. Mutations in AEC syndrome skin reveal a role for Clements SE, Techanukul T, Lai-Cheong JE, Mee JB, South AP, Zhao Y, Zhang CF, Rossiter H, Eckhart L, König U, Karner S, et al. J Dermatolog Clin Res 1(1): 1006 (2013) 5/6 Ratovitski (2013) Email: [email protected] Central

p63 in basement membrane adhesion, skin barrier integrity and hair 76. expression of p63 isoforms in normal skin and hyperproliferative Kim SY, Cho HJ, Kim DS, Choi HR, Kwon SB, Na JI, et al. Differential 68. follicle biology. Br J Dermatol. 2012; 167: 134-144. Mutant p63 causes defective expansion of ectodermal progenitor cells 77. conditions. J Cutan Pathol. 2009; 36: 825-830. Ferone G, Thomason HA, Antonini D, De Rosa L, Hu B, Gemei M, et al. 4: 192-205. 125b)Gu X, expression Nylander E,in Coatespsoriatic PJ, epidermis. Nylander Acta K. Effect Derm ofVenereol. narrow-band 2011; and impaired FGF signalling in AEC syndrome. EMBO Mol Med. 2012; 91:ultraviolet 392-397. B phototherapy on p63 and microRNA (miR-21 and miR- 69. R, et al. p63 control of desmosome gene expression and adhesion is 78. compromisedFerone G, Mollo in AEC MR, syndrome. Thomason Hum HA, Mol Antonini Genet. D, 2013; Zhou 22: H, 531-543. Ambrosio determinantChari NS, Romano of epidermal RA, Koster homeostasis. MI, Jaks V,Cell Roop Death D, Differ. Flores 2013; ER, et 20: al. 70. 1080-1088.Interaction between the TP63 and SHH pathways is an important pathways in disorders of epidermal differentiation. Trends Genet. 2013;Lopez-Pajares 29: 31-40. V, Yan K, Zarnegar BJ, Jameson KL, Khavari PA. Genetic 79. 71. differentiationShen J, van den in Bogaard skin keratinocytes EH, Kouwenhoven derived EN,from Bykov EEC syndrome VJ, Rinne is expressed in differentiated keratinocytes and regulates epidermal T, Zhang Q, et al. APR-246/PRIMA-1(MET) rescues epidermal lateCohen differentiation I, Birnbaum genes.RY, Leibson PLoS One.K, Taube 2012; R, 7: Sivan e42628. S, Birk OS. ZNF750 2157-2162. 72. patients with p63 mutations. Proc Natl Acad Sci U S A. 2013; 110: 80. Romanelli P, Miteva M, Schwartzfarb E, Ricotti C, Sullivan T, Abenoza Dermatol Sci. 2012; 66: 89-97. Zhang X. Genome-wide association study of skin complex diseases. J P, et al. P63 is a helpful tool in the diagnosis of a primary cutaneous 73. target gene with a crucial role in the developing human limb bud and 81. hairShimomura follicle. Y,Development. Wajid M, Shapiro 2008; L, 135: Christiano 743-753. AM. P-cadherin is a p63 carcinosarcoma. J Cutan Pathol. 2009; 36: 280-282. 74. Gleason BC, Calder KB, Cibull TL, Thomas AB, Billings SD, Morgan Pathol.MB, et 2009; al. Utility 36: 543-547. of p63 in the differential diagnosis of atypical 32:Shen 236-242. CS, Tsuda T, Fushiki S, Mizutani H, Yamanishi K. The expression fibroxanthoma and spindle cell squamous cell carcinoma. J Cutan of p63 during epidermal remodeling in psoriasis. J Dermatol. 2005; 82. 75. Vidal CI, Goldberg M, Burstein DE, Emanuel HJ, Emanuel PO. p63 Dermatol.Gu X, Lundqvist 2006; EN,126: Coates 137-141. PJ, Thurfjell N, Wettersand E, Nylander K. Immunohistochemistry is a useful adjunct in distinguishing sclerosing Dysregulation of TAp63 mRNA and protein levels in psoriasis. J Invest cutaneous tumors. Am J Dermatopathol. 2010; 32: 257-261.

Cite this article Ratovitski EA (2013) Tumor Protein P63 is a Key Regulator of Skin Functions in Ectodermal Dysplasia. J Dermatolog Clin Res 1(1): 1006.

J Dermatolog Clin Res 1(1): 1006 (2013) 6/6