201
|
Induced multipotency in adult keratinocytes through down-regulation of ΔNp63 or DGCR8. Proc Natl Acad Sci U S A 2014; 111:E572-81. [PMID: 24449888 DOI: 10.1073/pnas.1319743111] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The roles of microRNAs (miRNAs) and the miRNA processing machinery in the regulation of stem cell biology are not well understood. Here, we show that the p53 family member and p63 isoform, ΔNp63, is a transcriptional activator of a cofactor critical for miRNA processing (DGCR8). This regulation gives rise to a unique miRNA signature resulting in reprogramming cells to multipotency. Strikingly, ΔNp63(-/-) epidermal cells display profound defects in terminal differentiation and express a subset of markers and miRNAs present in embryonic stem cells and fibroblasts induced to pluripotency using Yamanaka factors. Moreover, ΔNp63(-/-) epidermal cells transduced with an inducible DGCR8 plasmid can differentiate into multiple cell fates in vitro and in vivo. We found that human primary keratinocytes depleted of ΔNp63 or DGCR8 can be reprogrammed in 6 d and express a unique miRNA and gene expression signature that is similar but not identical to human induced pluripotent stem cells. Our data reveal a role for ΔNp63 in the transcriptional regulation of DGCR8 to reprogram adult somatic cells into multipotent stem cells.
Collapse
|
202
|
Yallowitz AR, Alexandrova EM, Talos F, Xu S, Marchenko ND, Moll UM. p63 is a prosurvival factor in the adult mammary gland during post-lactational involution, affecting PI-MECs and ErbB2 tumorigenesis. Cell Death Differ 2014; 21:645-54. [PMID: 24440910 DOI: 10.1038/cdd.2013.199] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 12/13/2013] [Accepted: 12/13/2013] [Indexed: 01/31/2023] Open
Abstract
In embryogenesis, p63 is essential to develop mammary glands. In the adult mammary gland, p63 is highly expressed in the basal cell layer that comprises myoepithelial and interspersed stem/progenitor cells, and has limited expression in luminal epithelial cells. In adult skin, p63 has a crucial role in the maintenance of epithelial stem cells. However, it is unclear whether p63 also has an equivalent role as a stem/progenitor cell factor in adult mammary epithelium. We show that p63 is essential in vivo for the survival and maintenance of parity-identified mammary epithelial cells (PI-MECs), a pregnancy-induced heterogeneous population that survives post-lactational involution and contain multipotent progenitors that give rise to alveoli and ducts in subsequent pregnancies. p63+/- glands are normal in virgin, pregnant and lactating states. Importantly, however, during the apoptotic phase of post-lactational involution p63+/- glands show a threefold increase in epithelial cell death, concomitant with increased activation of the oncostatin M/Stat3 and p53 pro-apoptotic pathways, which are responsible for this phase. Thus, p63 is a physiologic antagonist of these pathways specifically in this regressive stage. After the restructuring phase when involution is complete, mammary glands of p63+/- mice again exhibit normal epithelial architecture by conventional histology. However, using Rosa(LSL-LacZ);WAP-Cre transgenics (LSL-LacZ, lox-stop-lox β-galactosidase), a genetic in vivo labeling system for PI-MECs, we find that p63+/- glands have a 30% reduction in the number of PI-MEC progenitors and their derivatives. Importantly, PI-MECs are also cellular targets of pregnancy-promoted ErbB2 tumorigenesis. Consistent with their PI-MEC pool reduction, one-time pregnant p63+/- ErbB2 mice are partially protected from breast tumorigenesis, exhibiting extended tumor-free and overall survival, and reduced tumor multiplicity compared with their p63+/+ ErbB2 littermates. Conversely, in virgin ErbB2 mice p63 heterozygosity provides no survival advantage. In sum, our data establish that p63 is an important survival factor for pregnancy-identified PI-MEC progenitors in breast tissue in vivo.
Collapse
Affiliation(s)
- A R Yallowitz
- Department of Pathology, Stony Brook University, School of Medicine, Stony Brook, NY 11794, USA
| | - E M Alexandrova
- Department of Pathology, Stony Brook University, School of Medicine, Stony Brook, NY 11794, USA
| | - F Talos
- Department of Pathology, Stony Brook University, School of Medicine, Stony Brook, NY 11794, USA
| | - S Xu
- Department of Pathology, Stony Brook University, School of Medicine, Stony Brook, NY 11794, USA
| | - N D Marchenko
- Department of Pathology, Stony Brook University, School of Medicine, Stony Brook, NY 11794, USA
| | - U M Moll
- Department of Pathology, Stony Brook University, School of Medicine, Stony Brook, NY 11794, USA
| |
Collapse
|
203
|
Du J, Romano RA, Si H, Mattox A, Bian Y, Yang X, Sinha S, Van Waes C, Chen Z. Epidermal overexpression of transgenic ΔNp63 promotes type 2 immune and myeloid inflammatory responses and hyperplasia via NF-κB activation. J Pathol 2014; 232:356-68. [DOI: 10.1002/path.4302] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 10/15/2013] [Accepted: 11/11/2013] [Indexed: 12/20/2022]
Affiliation(s)
- Jihui Du
- Clinical Genomics Unit and Tumor Biology Section, Head and Neck Surgery Branch; National Institute on Deafness and Other Communication Disorders; NIH, Bethesda MD 20892 USA
- Central Laboratory, Nanshan Hospital; Guangdong Medical College; Shenzhen Guangdong 518052 China
| | - Rose-Anne Romano
- Department of Biochemistry, State University of New York at Buffalo; Center for Excellence in Bioinformatics and Life Sciences; Buffalo New York USA
| | - Han Si
- Clinical Genomics Unit and Tumor Biology Section, Head and Neck Surgery Branch; National Institute on Deafness and Other Communication Disorders; NIH, Bethesda MD 20892 USA
| | - Austin Mattox
- Clinical Genomics Unit and Tumor Biology Section, Head and Neck Surgery Branch; National Institute on Deafness and Other Communication Disorders; NIH, Bethesda MD 20892 USA
| | - Yansong Bian
- Clinical Genomics Unit and Tumor Biology Section, Head and Neck Surgery Branch; National Institute on Deafness and Other Communication Disorders; NIH, Bethesda MD 20892 USA
| | - Xinping Yang
- Clinical Genomics Unit and Tumor Biology Section, Head and Neck Surgery Branch; National Institute on Deafness and Other Communication Disorders; NIH, Bethesda MD 20892 USA
| | - Satrajit Sinha
- Department of Biochemistry, State University of New York at Buffalo; Center for Excellence in Bioinformatics and Life Sciences; Buffalo New York USA
| | - Carter Van Waes
- Clinical Genomics Unit and Tumor Biology Section, Head and Neck Surgery Branch; National Institute on Deafness and Other Communication Disorders; NIH, Bethesda MD 20892 USA
| | - Zhong Chen
- Clinical Genomics Unit and Tumor Biology Section, Head and Neck Surgery Branch; National Institute on Deafness and Other Communication Disorders; NIH, Bethesda MD 20892 USA
| |
Collapse
|
204
|
Fischer B, Metzger M, Richardson R, Knyphausen P, Ramezani T, Franzen R, Schmelzer E, Bloch W, Carney TJ, Hammerschmidt M. p53 and TAp63 promote keratinocyte proliferation and differentiation in breeding tubercles of the zebrafish. PLoS Genet 2014; 10:e1004048. [PMID: 24415949 PMCID: PMC3886889 DOI: 10.1371/journal.pgen.1004048] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 11/04/2013] [Indexed: 11/18/2022] Open
Abstract
p63 is a multi-isoform member of the p53 family of transcription factors. There is compelling genetic evidence that ΔNp63 isoforms are needed for keratinocyte proliferation and stemness in the developing vertebrate epidermis. However, the role of TAp63 isoforms is not fully understood, and TAp63 knockout mice display normal epidermal development. Here, we show that zebrafish mutants specifically lacking TAp63 isoforms, or p53, display compromised development of breeding tubercles, epidermal appendages which according to our analyses display more advanced stratification and keratinization than regular epidermis, including continuous desquamation and renewal of superficial cells by derivatives of basal keratinocytes. Defects are further enhanced in TAp63/p53 double mutants, pointing to partially redundant roles of the two related factors. Molecular analyses, treatments with chemical inhibitors and epistasis studies further reveal the existence of a linear TAp63/p53->Notch->caspase 3 pathway required both for enhanced proliferation of keratinocytes at the base of the tubercles and their subsequent differentiation in upper layers. Together, these studies identify the zebrafish breeding tubercles as specific epidermal structures sharing crucial features with the cornified mammalian epidermis. In addition, they unravel essential roles of TAp63 and p53 to promote both keratinocyte proliferation and their terminal differentiation by promoting Notch signalling and caspase 3 activity, ensuring formation and proper homeostasis of this self-renewing stratified epithelium. The mammalian epidermis is a stratified self-renewing epithelium, in which cell loss at the surface is properly balanced by cell proliferation in basal layers to ensure tissue homeostasis. But how is this balance genetically controlled? Here, we address this question in zebrafish breeding tubercles, epidermal appendages in which keratinocytes undergo more advanced differentiation processes than in regular fish epidermis, sharing crucial features with the cornified mammalian skin. We identify a linear pathway consisting of the transcription factor p53 and its close relative TAp63, which activate Notch signalling and thereby caspase 3 to promote terminal differentiation and eventual shedding of keratinocytes in upper tubercle layers, while at the same time employing non-cell autonomous mechanisms to promote keratinocyte proliferation at the tubercle base, thereby ensuring proper development and homeostasis of this self-renewing tissue. Such a two-fold function of the pathway is consistent with the formerly reported dual role of a caspase during wing regeneration in the fruitfly. Our findings will help to better understand the seemingly contrary effects described for TAp63 in different mammalian systems, while demonstrating partial functional redundancy between p53 and TAp63 during epidermal development in fish.
Collapse
Affiliation(s)
- Boris Fischer
- Institute of Developmental Biology, University of Cologne, Cologne, Germany
| | - Manuel Metzger
- Institute of Developmental Biology, University of Cologne, Cologne, Germany
| | - Rebecca Richardson
- Institute of Developmental Biology, University of Cologne, Cologne, Germany
| | - Philipp Knyphausen
- Institute of Developmental Biology, University of Cologne, Cologne, Germany
| | - Thomas Ramezani
- Institute of Developmental Biology, University of Cologne, Cologne, Germany
| | - Rainer Franzen
- Cell Biology, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Elmon Schmelzer
- Cell Biology, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Wilhelm Bloch
- Institute of Cardiology and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | | | - Matthias Hammerschmidt
- Institute of Developmental Biology, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
- * E-mail:
| |
Collapse
|
205
|
Muller PAJ, Trinidad AG, Caswell PT, Norman JC, Vousden KH. Mutant p53 regulates Dicer through p63-dependent and -independent mechanisms to promote an invasive phenotype. J Biol Chem 2014; 289:122-32. [PMID: 24220032 PMCID: PMC3879536 DOI: 10.1074/jbc.m113.502138] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 10/27/2013] [Indexed: 12/13/2022] Open
Abstract
The control and processing of microRNAs (miRs) is critical in the regulation of all cellular responses. Previous studies have suggested that a reduction in the expression of certain miRs, or an overall decrease in miR processing through the partial depletion of Dicer, can promote enhanced metastatic potential. We show here that Dicer depletion can promote the invasive behavior of cells that is reflected in enhanced recycling and activation of the growth factor receptors Met and EGF receptor. These responses are also seen in response to the expression of tumor-derived mutant p53s, and we show that mutant p53 can down-regulate Dicer expression through both direct inhibition of the TAp63-mediated transcriptional activation of Dicer and a TAp63-independent control of Dicer protein expression. Our results delineate a clear relationship between mutant p53, TAp63, and Dicer that might contribute to the metastatic function of mutant p53 but, interestingly, also reveal TAp63-independent functions of mutant p53 in controlling Dicer activity.
Collapse
Affiliation(s)
- Patricia A. J. Muller
- From the Cancer Research UK Beatson Institute, Glasgow G61 1BD, Scotland, United Kingdom and
| | - Antonio G. Trinidad
- From the Cancer Research UK Beatson Institute, Glasgow G61 1BD, Scotland, United Kingdom and
| | - Patrick T. Caswell
- the Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Jim C. Norman
- From the Cancer Research UK Beatson Institute, Glasgow G61 1BD, Scotland, United Kingdom and
| | - Karen H. Vousden
- From the Cancer Research UK Beatson Institute, Glasgow G61 1BD, Scotland, United Kingdom and
| |
Collapse
|
206
|
Girardini JE, Walerych D, Del Sal G. Cooperation of p53 mutations with other oncogenic alterations in cancer. Subcell Biochem 2014; 85:41-70. [PMID: 25201188 DOI: 10.1007/978-94-017-9211-0_3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Following the initial findings suggesting a pro-oncogenic role for p53 point mutants, more than 30 years of research have unveiled the critical role exerted by these mutants in human cancer. A growing body of evidence, including mouse models and clinical data, has clearly demonstrated a connection between mutant p53 and the development of aggressive and metastatic tumors. Even if the molecular mechanisms underlying mutant p53 activities are still the object of intense scrutiny, it seems evident that full activation of its oncogenic role requires the functional interaction with other oncogenic alterations. p53 point mutants, with their pleiotropic effects, simultaneously activating several mechanisms of aggressiveness, are engaged in multiple cross-talk with a variety of other cancer-related processes, thus depicting a complex molecular landscape for the mutant p53 network. In this chapter revealing evidence illustrating different ways through which this cooperation may be achieved will be discussed. Considering the proposed role for mutant p53 as a driver of cancer aggressiveness, disarming mutant p53 function by uncoupling the cooperation with other oncogenic alterations, stands out as an exciting possibility for the development of novel anti-cancer therapies.
Collapse
Affiliation(s)
- Javier E Girardini
- Molecular Oncology Group, Institute of Molecular and Cell Biology of Rosario, IBR-CONICET, Rosario, Argentina
| | | | | |
Collapse
|
207
|
Xu E, Zhang J, Zhang M, Jiang Y, Cho SJ, Chen X. RNA-binding protein RBM24 regulates p63 expression via mRNA stability. Mol Cancer Res 2013; 12:359-69. [PMID: 24375645 DOI: 10.1158/1541-7786.mcr-13-0526] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
UNLABELLED p63, a p53 family member, plays pivotal roles in epidermal development, aging, and tumorigenesis. Thus, understanding how p63 expression is controlled has biological and clinical importance. RBM24 is an RNA-binding protein and shares a high sequence similarity with RBM38, a critical regulator of p63. In this study, we investigated whether RBM24 is capable of regulating p63 expression. Indeed, we found that ectopic expression of RBM24 decreased, whereas knockdown of RBM24 increased, the levels of p63 transcript and protein. To explore the underlying mechanism, we found that RBM24 was able to bind to multiple regions in the p63 3' untranslated region and, subsequently, destabilize p63 transcript. Furthermore, we showed that the 3' untranslated region in p63 transcript and the RNA-binding domain in RBM24 were required for RBM24 to bind p63 transcript and consequently, inhibit p63 expression. Taken together, our data provide evidence that RBM24 is a novel regulator of p63 via mRNA stability. IMPLICATIONS Our study suggests that p63 is regulated by RBM24 via mRNA stability, which gives an insight into understanding how posttranscriptional regulatory mechanisms contribute to p63 expression.
Collapse
Affiliation(s)
- Enshun Xu
- Comparative Oncology Laboratory, University of California at Davis, Davis, CA 95616.
| | | | | | | | | | | |
Collapse
|
208
|
Chin SS, Romano RA, Nagarajan P, Sinha S, Garrett-Sinha LA. Aberrant epidermal differentiation and disrupted ΔNp63/Notch regulatory axis in Ets1 transgenic mice. Biol Open 2013; 2:1336-45. [PMID: 24337118 PMCID: PMC3863418 DOI: 10.1242/bio.20135397] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The transcription factor Ets1 is expressed at low levels in epidermal keratinocytes under physiological conditions, but is over-expressed in cutaneous squamous cell carcinoma (SCC). We previously showed that over-expression of Ets1 in differentiated keratinocytes of the skin leads to significant pro-tumorigenic alterations. Here, we further extend these studies by testing the effects of over-expressing Ets1 in the proliferative basal keratinocytes of the skin, which includes the putative epidermal stem cells. We show that induction of the Ets1 transgene in the basal layer of skin during embryogenesis results in epidermal hyperplasia and impaired differentiation accompanied by attenuated expression of spinous and granular layer markers. A similar hyper-proliferative skin phenotype was observed when the transgene was induced in the basal layer of the skin of adult mice leading to hair loss and open sores. The Ets1-mediated phenotype is accompanied by a variety of changes in gene expression including alterations in Notch signaling, a crucial mediator of normal skin differentiation. Finally, we show that Ets1 disrupts Notch signaling in part via its ability to upregulate ΔNp63, an established transcriptional repressor of several of the Notch receptors. Given the established tumor suppressive role for Notch signaling in skin tumorigenesis, the demonstrated ability of Ets1 to interfere with this signaling pathway may be important in mediating its pro-tumorigenic activities.
Collapse
Affiliation(s)
- Shu Shien Chin
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | | | | | | | | |
Collapse
|
209
|
Ng GY, Yeh LK, Zhang Y, Liu H, Feng GS, Kao WWY, Liu CY. Role of SH2-containing tyrosine phosphatase Shp2 in mouse corneal epithelial stratification. Invest Ophthalmol Vis Sci 2013; 54:7933-42. [PMID: 24204042 DOI: 10.1167/iovs.13-12646] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
PURPOSE Shp2 protein tyrosine phosphatase mediates a wide variety of receptor tyrosine kinases (RTK) cell signaling. Herein, we investigate the role of Shp2 in corneal morphogenesis and homeostasis. METHODS Shp2 was conditionally knocked out (Shp2(cko)) in Krt14-rtTA;tet-O-Cre;Shp2(f/f) triple transgenic mice administrated with doxycycline (Dox) from postnatal day 1 (P1) to P10, P15, and P25, respectively. In addition, corneal epithelial debridement was performed in adult (P42) mice treated with or without Dox for 8 days (from P42-P50). Mouse eyes were then subjected to histology and immunohistochemistry. RESULTS Shp2(cko) revealed impaired stratification of conjunctival and corneal epithelia during morphogenesis. Likewise, Shp2(cko) failed to restore epithelial stratification after a corneal epithelial wound in adult Shp2(cko). At the cellular level, the ratio of proliferating cell nuclear antigen (PCNA-positive)/total basal cells remained unchanged, but cells in G2/M (survivin-positive) phase was significantly increased in Shp2(cko) as compared with those in the control littermate. Interestingly, deltaN-p63 (ΔNp63) expression and the asymmetric division of the basal cells were coincidentally dampened in Shp2(cko). Transmission electron microscopic study showed that desmosome and hemidesmosome densities were reduced in the corneal epithelium of Shp2(cko). Immunohistochemistry also demonstrated that expression of E-cadherin/β-catenin junction and laminin-β1 was extensively downregulated in Shp2(cko). On the other hand, corneal epithelium lacking Shp2 remained positive for K14, Pax-6, and keratin 12 (K12), suggesting that Shp2 was dispensable for the corneal epithelial-type differentiation. CONCLUSIONS These data argued that Shp2 deficiency predominantly impacted p63-dependent cell division and cell adhesive ability, which resulted in the impairment of stratification during corneal epithelial development and wound healing.
Collapse
Affiliation(s)
- Gracia Y Ng
- Edith J. Crawley Vision Research Center/Department of Ophthalmology, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | | | | | | | | | | | | |
Collapse
|
210
|
Gu J, Lu Y, Qiao L, Ran D, Li N, Cao H, Gao Y, Zheng Q. Mouse p63 variants and chondrogenesis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2013; 6:2872-2879. [PMID: 24294373 PMCID: PMC3843267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 11/08/2013] [Indexed: 06/02/2023]
Abstract
As a critical member of the p53 family of transcription factors, p63 has been implicated a role in development than in tumor formation, because p63 is seldom mutated in human cancers, while p63 null mice exhibit severe developmental abnormalities without increasing cancer susceptibility. Notably, besides the major epithelial and cardiac defect, p63 deficient mice show severe limb and craniofacial abnormalities. In addition, humans with p63 mutations also show severe limb and digit defects, suggesting a putative role of p63 in skeletal development. There are eight p63 variants which encode for the TAp63 and ΔNp63 isoforms by alternative promoters. How these isoforms function during skeletal development is currently largely unknown. Our recent transgenic studies suggest a role of TAP63α, but not ΔNP63α, during embryonic long bone development. However, the moderate skeletal phenotypes in the TAP63α transgenic mice suggest requirement of additional p63 isoform(s) for the limb defects in p63 null mice. Here, we report analysis of mouse p63 variants in MCT and ATDC5 cells, two cell models undergo hypertrophic differentiation and mimic the process of endochondral bone formation upon growth arrest or induction. We detected increased level of p63 variants in hypertrophic MCT cells by regular RT-PCR analysis. Further analysis by qRT-PCR, we detected significantly upregulated level of γ variant (p<0.05), but not α or β variant (p>0.05), in hypertrophic MCT cells than in proliferative MCT cells. Moreover, we detected upregulated TAP63γ in ATDC5 cells undergoing hypertrophic differentiation. Our results suggest that TAp63γ plays a positive role during endochondral bone formation.
Collapse
Affiliation(s)
- Junxia Gu
- Department of Hematology and Hematological Laboratory Science, School of Medical Science and Laboratory Medicine, Jiangsu UniversityZhenjiang 212013, China
| | - Yaojuan Lu
- Department of Hematology and Hematological Laboratory Science, School of Medical Science and Laboratory Medicine, Jiangsu UniversityZhenjiang 212013, China
- Department of Anatomy and Cell Biology, Rush University Medical CenterChicago, IL 60612, USA
| | - Longwei Qiao
- Department of Hematology and Hematological Laboratory Science, School of Medical Science and Laboratory Medicine, Jiangsu UniversityZhenjiang 212013, China
| | - Deyuan Ran
- Department of Hematology and Hematological Laboratory Science, School of Medical Science and Laboratory Medicine, Jiangsu UniversityZhenjiang 212013, China
| | - Na Li
- Department of Hematology and Hematological Laboratory Science, School of Medical Science and Laboratory Medicine, Jiangsu UniversityZhenjiang 212013, China
| | - Hong Cao
- Department of Hematology and Hematological Laboratory Science, School of Medical Science and Laboratory Medicine, Jiangsu UniversityZhenjiang 212013, China
| | - Yan Gao
- Department of Hematology and Hematological Laboratory Science, School of Medical Science and Laboratory Medicine, Jiangsu UniversityZhenjiang 212013, China
| | - Qiping Zheng
- Department of Hematology and Hematological Laboratory Science, School of Medical Science and Laboratory Medicine, Jiangsu UniversityZhenjiang 212013, China
- Department of Anatomy and Cell Biology, Rush University Medical CenterChicago, IL 60612, USA
| |
Collapse
|
211
|
Frank SB, Miranti CK. Disruption of prostate epithelial differentiation pathways and prostate cancer development. Front Oncol 2013; 3:273. [PMID: 24199173 PMCID: PMC3813973 DOI: 10.3389/fonc.2013.00273] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 10/18/2013] [Indexed: 12/14/2022] Open
Abstract
One of the foremost problems in the prostate cancer (PCa) field is the inability to distinguish aggressive from indolent disease, which leads to difficult prognoses and thousands of unnecessary surgeries. This limitation stems from the fact that the mechanisms of tumorigenesis in the prostate are poorly understood. Some genetic alterations are commonly reported in prostate tumors, including upregulation of Myc, fusion of Ets genes to androgen-regulated promoters, and loss of Pten. However, the specific roles of these aberrations in tumor initiation and progression are poorly understood. Likewise, the cell of origin for PCa remains controversial and may be linked to the aggressive potential of the tumor. One important clue is that prostate tumors co-express basal and luminal protein markers that are restricted to their distinct cell types in normal tissue. Prostate epithelium contains layer-specific stem cells as well as rare bipotent cells, which can differentiate into basal or luminal cells. We hypothesize that the primary oncogenic cell of origin is a transient-differentiating bipotent cell. Such a cell must maintain tight temporal and spatial control of differentiation pathways, thus increasing its susceptibility for oncogenic disruption. In support of this hypothesis, many of the pathways known to be involved in prostate differentiation can be linked to genes commonly altered in PCa. In this article, we review what is known about important differentiation pathways (Myc, p38MAPK, Notch, PI3K/Pten) in the prostate and how their misregulation could lead to oncogenesis. Better understanding of normal differentiation will offer new insights into tumor initiation and may help explain the functional significance of common genetic alterations seen in PCa. Additionally, this understanding could lead to new methods for classifying prostate tumors based on their differentiation status and may aid in identifying more aggressive tumors.
Collapse
Affiliation(s)
- Sander B Frank
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute , Grand Rapids, MI , USA ; Genetics Graduate Program, Michigan State University , East Lansing, MI , USA
| | | |
Collapse
|
212
|
Bid HK, Roberts RD, Cam M, Audino A, Kurmasheva RT, Lin J, Houghton PJ, Cam H. ΔNp63 promotes pediatric neuroblastoma and osteosarcoma by regulating tumor angiogenesis. Cancer Res 2013; 74:320-9. [PMID: 24154873 DOI: 10.1158/0008-5472.can-13-0894] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The tumor suppressor gene p53 and its family members p63/p73 are critical determinants of tumorigenesis. ΔNp63 is a splice variant of p63, which lacks the N-terminal transactivation domain. It is thought to antagonize p53-, p63-, and p73-dependent translation, thus blocking their tumor suppressor activity. In our studies of the pediatric solid tumors neuroblastoma and osteosarcoma, we find overexpression of ΔNp63; however, there is no correlation of ΔNp63 expression with p53 mutation status. Our data suggest that ΔNp63 itself endows cells with a gain-of-function that leads to malignant transformation, a function independent of any p53 antagonism. Here, we demonstrate that ΔNp63 overexpression, independent of p53, increases secretion of interleukin (IL)-6 and IL-8, leading to elevated phosphorylation of STAT3 (Tyr-705). We show that elevated phosphorylation of STAT3 leads to stabilization of hypoxia-inducible factor 1α (HIF-1α) protein, resulting in VEGF secretion. We also show human clinical data, which suggest a mechanistic role for ΔNp63 in osteosarcoma metastasis. In summary, our studies reveal the mechanism by which ΔNp63, as a master transcription factor, modulates tumor angiogenesis.
Collapse
Affiliation(s)
- Hemant K Bid
- Authors' Affiliations: Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital; and Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| | | | | | | | | | | | | | | |
Collapse
|
213
|
Ratovitski EA. Phospho-ΔNp63α regulates AQP3, ALOX12B, CASP14 and CLDN1 expression through transcription and microRNA modulation. FEBS Lett 2013; 587:3581-6. [PMID: 24070899 DOI: 10.1016/j.febslet.2013.09.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 09/14/2013] [Accepted: 09/16/2013] [Indexed: 11/30/2022]
Abstract
Cisplatin-induced and ATM-phosphorylated (p)-ΔNp63α regulates the expression of epidermal differentiation and skin barrier regulators (AQP3, CASP14, ALOX12B, and CLDN1) in squamous cell carcinoma (SCC) cells by dual transcriptional and post-transcriptional mechanisms. We found that p-ΔNp63α bound to target gene promoters, and regulated the activity of the tested promoters in vitro. P-ΔNp63α was shown to upregulate miR-185-5p and downregulate let7-5p, which subsequently modulated AQP3, CASP14, ALOX12B and CLDN1 through their respective 3'-untranslated regions. The introduction of miR-185-5p into resistant SCC-11M cells, which are unable to phosphorylate ΔNp63α, render these cells more sensitive to cisplatin treatment. Further studies of the AQP3, CASP14, ALOX12B, and CLDN1 contributions to chemoresistance may assist in developing novel microRNA-based therapies for human SCC.
Collapse
Affiliation(s)
- Edward A Ratovitski
- Head and Neck Cancer Research Division, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
214
|
ΔNp63 regulates select routes of reprogramming via multiple mechanisms. Cell Death Differ 2013; 20:1698-708. [PMID: 24013722 DOI: 10.1038/cdd.2013.122] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 07/30/2013] [Accepted: 07/31/2013] [Indexed: 12/12/2022] Open
Abstract
Somatic cells can be converted into induced pluripotent stem cells (iPSCs) by forced expression of various combinations of transcription factors, but the molecular mechanisms of reprogramming are poorly understood. Specifically, evidence that the reprogramming process can take many distinct routes only begins to emerge. It is definitively established that p53 deficiency greatly enhances reprogramming, revealing p53's barrier function for induced pluripotency, but the role of its homologs p63 and p73 are unknown. Here we report that in stark contrast to p53, p73 has no role in reprogramming. However, p63 is an enabling (rather than a barrier) factor for Oct4, Sox2 and Klf4 (OSK) and Oct4 and Sox2 (OS), but not for Oct4 and Klf4 (OK) reprogramming of mouse embryonic fibroblasts. Specifically, p63 is essential during reprogramming for maximum efficiency, albeit not for the ability to reprogram per se, and is dispensable for maintaining stability and pluripotency of established iPSC colonies. ΔNp63, but not TAp63, is the principal isoform involved. Loss of p63 can affect reprogramming via several mechanisms such as reduced expression of mesenchymal-epithelial transition and pluripotency genes, hypoproliferation and loss of the most reprogrammable cell populations. During OSK and OS reprogramming, different mechanisms seem to be critical, such as regulation of epithelial and pluripotency genes in OSK reprogramming versus regulation of proliferation in OS reprogramming. Finally, our data reveal three different routes of reprogramming by OSK, OS or OK, based on their differential p63 requirements for iPSC efficiency and pluripotency marker expression. This supports the concept that many distinct routes of reprogramming exist.
Collapse
|
215
|
Li Y, Moretto-Zita M, Soncin F, Wakeland A, Wolfe L, Leon-Garcia S, Pandian R, Pizzo D, Cui L, Nazor K, Loring JF, Crum CP, Laurent LC, Parast MM. BMP4-directed trophoblast differentiation of human embryonic stem cells is mediated through a ΔNp63+ cytotrophoblast stem cell state. Development 2013; 140:3965-76. [PMID: 24004950 DOI: 10.1242/dev.092155] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The placenta is a transient organ that is necessary for proper fetal development. Its main functional component is the trophoblast, which is derived from extra-embryonic ectoderm. Little is known about early trophoblast differentiation in the human embryo, owing to lack of a proper in vitro model system. Human embryonic stem cells (hESCs) differentiate into functional trophoblast following BMP4 treatment in the presence of feeder-conditioned media; however, this model has not been widely accepted, in part owing to a lack of proof for a trophoblast progenitor population. We have previously shown that p63, a member of the p53 family of nuclear proteins, is expressed in proliferative cytotrophoblast (CTB), precursors to terminally differentiated syncytiotrophoblast (STB) in chorionic villi and extravillous trophoblast (EVT) at the implantation site. Here, we show that BMP4-treated hESCs differentiate into bona fide CTB by direct comparison with primary human placental tissues and isolated CTB through gene expression profiling. We show that, in primary CTB, p63 levels are reduced as cells differentiate into STB, and that forced expression of p63 maintains cyclin B1 and inhibits STB differentiation. We also establish that, similar to in vivo events, hESC differentiation into trophoblast is characterized by a p63(+)/KRT7(+) CTB stem cell state, followed by formation of functional KLF4(+) STB and HLA-G(+) EVT. Finally, we illustrate that downregulation of p63 by shRNA inhibits differentiation of hESCs into functional trophoblast. Taken together, our results establish that BMP4-treated hESCs are an excellent model of human trophoblast differentiation, closely mimicking the in vivo progression from p63(+) CTB stem cells to terminally differentiated trophoblast subtypes.
Collapse
Affiliation(s)
- Yingchun Li
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
216
|
Poligone B, Hayden MS, Chen L, Pentland AP, Jimi E, Ghosh S. A role for NF-κB activity in skin hyperplasia and the development of keratoacanthomata in mice. PLoS One 2013; 8:e71887. [PMID: 23977171 PMCID: PMC3747062 DOI: 10.1371/journal.pone.0071887] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 07/10/2013] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Previous studies have implicated NF-κB signaling in both cutaneous development and oncogenesis. However, these studies have been limited in part by the lethality that results from extreme over- or under-expression of NF-κB in available mouse models. Even cre-driven tissue specific expression of transgenes, or targeted deletion of NF-κB can cause cell death. Therefore, the present study was undertaken to evaluate a novel mouse model of enhanced NF-κB activity in the skin. METHODS A knock-in homologous recombination technique was utilized to develop a mouse model (referred to as PD mice) with increased NF-κB activity. RESULTS The data show that increased NF-κB activity leads to hyperproliferation and dysplasia of the mouse epidermis. Chemical carcinogenesis in the context of enhanced NF-κB activity promotes the development of keratoacanthomata. CONCLUSION Our findings support an important role for NF-κB in keratinocyte dysplasia. We have found that enhanced NF-κB activity renders keratinocytes susceptible to hyperproliferation and keratoacanthoma (KA) development but is not sufficient for transformation and SCC development. We therefore propose that NF-κB activation in the absence of additional oncogenic events can promote TNF-dependent, actinic keratosis-like dysplasia and TNF-independent, KAs upon chemical carcinogensis. These studies suggest that resolution of KA cannot occur when NF-κB activation is constitutively enforced.
Collapse
Affiliation(s)
- Brian Poligone
- Department of Dermatology and the James P. Wilmot Cancer Center, University of Rochester School of Medicine, Rochester, New York, United States of America
- * E-mail:
| | - Matthew S. Hayden
- Department of Dermatology, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| | - Luojing Chen
- Department of Dermatology and the James P. Wilmot Cancer Center, University of Rochester School of Medicine, Rochester, New York, United States of America
| | - Alice P. Pentland
- Department of Dermatology and the James P. Wilmot Cancer Center, University of Rochester School of Medicine, Rochester, New York, United States of America
| | - Eijiro Jimi
- Division of Molecular Signaling and Biochemistry, Kyushu Dental College, Kitakyushu, Fukuoka, Japan
| | - Sankar Ghosh
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| |
Collapse
|
217
|
Poly (C)-binding protein 1 regulates p63 expression through mRNA stability. PLoS One 2013; 8:e71724. [PMID: 23940783 PMCID: PMC3737132 DOI: 10.1371/journal.pone.0071724] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 07/09/2013] [Indexed: 11/19/2022] Open
Abstract
p63, a transcription factor and p53 family protein, plays a crucial role in tumor suppression and development of various epithelial tissues. While p63 expression is controlled mostly by post-translational modifications, recent studies indicate that transcriptional and posttranscriptional regulations are essential for proper p63 expression. Here, we investigated the regulation of p63 expression by poly (C)-binding protein 1 (PCBP1, also known as hnRNP-E1 and αCP1). We found that knockdown of PCBP1 decreases the level of p63 transcript and protein. We also found that PCBP1 regulates the stability of p63 mRNA via binding to p63 3’UTR. Additionally, we found that a CU-rich element (CUE) in p63 3′UTR is bound by and responsive to PCBP1. Together, we conclude that PCBP1 regulates p63 expression via mRNA stability.
Collapse
|
218
|
Tadeu AMB, Horsley V. Notch signaling represses p63 expression in the developing surface ectoderm. Development 2013; 140:3777-86. [PMID: 23924630 DOI: 10.1242/dev.093948] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The development of the mature epidermis requires a coordinated sequence of signaling events and transcriptional changes to specify surface ectodermal progenitor cells to the keratinocyte lineage. The initial events that specify epidermal keratinocytes from ectodermal progenitor cells are not well understood. Here, we use both developing mouse embryos and human embryonic stem cells (hESCs) to explore the mechanisms that direct keratinocyte fate from ectodermal progenitor cells. We show that both hESCs and murine embryos express p63 before keratin 14. Furthermore, we find that Notch signaling is activated before p63 expression in ectodermal progenitor cells. Inhibition of Notch signaling pharmacologically or genetically reveals a negative regulatory role for Notch signaling in p63 expression during ectodermal specification in hESCs or mouse embryos, respectively. Taken together, these data reveal a role for Notch signaling in the molecular control of ectodermal progenitor cell specification to the epidermal keratinocyte lineage.
Collapse
Affiliation(s)
- Ana Mafalda Baptista Tadeu
- Department of Molecular, Cell and Developmental Biology, Yale University, 219 Prospect Street, Box 208103, New Haven, CT 06520, USA
| | | |
Collapse
|
219
|
Ramsey MR, Wilson C, Ory B, Rothenberg SM, Faquin W, Mills AA, Ellisen LW. FGFR2 signaling underlies p63 oncogenic function in squamous cell carcinoma. J Clin Invest 2013; 123:3525-38. [PMID: 23867503 DOI: 10.1172/jci68899] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 05/08/2013] [Indexed: 02/06/2023] Open
Abstract
Oncogenic transcription factors drive many human cancers, yet identifying and therapeutically targeting the resulting deregulated pathways has proven difficult. Squamous cell carcinoma (SCC) is a common and lethal human cancer, and relatively little progress has been made in improving outcomes for SCC due to a poor understanding of its underlying molecular pathogenesis. While SCCs typically lack somatic oncogene-activating mutations, they exhibit frequent overexpression of the p53-related transcription factor p63. We developed an in vivo murine tumor model to investigate the function and key transcriptional programs of p63 in SCC. Here, we show that established SCCs are exquisitely dependent on p63, as acute genetic ablation of p63 in advanced, invasive SCC induced rapid and dramatic apoptosis and tumor regression. In vivo genome-wide gene expression analysis identified a tumor-survival program involving p63-regulated FGFR2 signaling that was activated by ligand emanating from abundant tumor-associated stroma. Correspondingly, we demonstrate the therapeutic efficacy of extinguishing this signaling axis in endogenous SCCs using the clinical FGFR2 inhibitor AZD4547. Collectively, these results reveal an unanticipated role for p63-driven paracrine FGFR2 signaling as an addicting pathway in human cancer and suggest a new approach for the treatment of SCC.
Collapse
Affiliation(s)
- Matthew R Ramsey
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | |
Collapse
|
220
|
Diethylstilbestrol induces vaginal adenosis by disrupting SMAD/RUNX1-mediated cell fate decision in the Müllerian duct epithelium. Dev Biol 2013; 381:5-16. [PMID: 23830984 DOI: 10.1016/j.ydbio.2013.06.024] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 06/24/2013] [Accepted: 06/25/2013] [Indexed: 11/21/2022]
Abstract
Women exposed to diethylstilbestrol (DES) in utero frequently develop vaginal adenosis, from which clear cell adenocarcinoma can arise. Despite decades of extensive investigation, the molecular pathogenesis of DES-associated vaginal adenosis remains elusive. Here we report that DES induces vaginal adenosis by inhibiting the BMP4/Activin A-regulated vaginal cell fate decision through a downregulation of RUNX1. BMP4 and Activin A produced by vaginal mesenchyme synergistically activated the expression of ΔNp63, thus deciding vaginal epithelial cell fate in the Müllerian duct epithelial cells (MDECs) via direct binding of SMADs on the highly conserved 5' sequence of ΔNp63. Therefore, mice in which Smad4 was deleted in MDECs failed to express ΔNp63 in vaginal epithelium and developed adenosis. This SMAD-dependent ΔNp63 activation required RUNX1, a binding partner of SMADs. Conditional deletion of Runx1 in the MDECs induced adenosis in the cranial portion of vagina, which mimicked the effect of developmental DES-exposure. Furthermore, neonatal DES exposure downregulated RUNX1 in the fornix of the vagina, where DES-associated adenosis is frequently found. This observation strongly suggests that the downregulation of RUNX1 is the cause of vaginal adenosis. However, once cell fate was determined, the BMP/Activin-SMAD/RUNX1 signaling pathway became dispensable for the maintenance of ΔNp63 expression in vaginal epithelium. Instead, the activity of the ΔNp63 locus in vaginal epithelium was maintained by a ΔNp63-dependent mechanism. This is the first demonstration of a molecular mechanism through which developmental chemical exposure causes precancerous lesions by altering cell fate.
Collapse
|
221
|
Senoo M. Epidermal Stem Cells in Homeostasis and Wound Repair of the Skin. Adv Wound Care (New Rochelle) 2013; 2:273-282. [PMID: 24527349 DOI: 10.1089/wound.2012.0372] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Indexed: 12/17/2022] Open
Abstract
SIGNIFICANCE The skin interfollicular epidermis (IFE) is an organism's first line of defense against a harmful environment and physical damage. During homeostasis and wound repair, the IFE is rejuvenated constantly by IFE stem cells (SCs) that are capable of both proliferation and differentiation. However, the identity and behavior of IFE SCs remain controversial. RECENT ADVANCES Two opposing theories exist regarding homeostasis of the IFE. On the basis of morphological and proliferative characteristics, one posits that the IFE is composed of a discrete epidermal proliferative unit comprised of ∼10 transit-amplifying (TA) cells and a centrally located SC in the basal layer. The other suggests that homeostasis of the IFE is maintained by a single progenitor population in the basal layer. A recent study has challenged these two apparently distinct models and demonstrated that the basal layer of the IFE contains both SCs and TA cells, which make distinct contributions to tissue homeostasis and repair. Moreover, phosphorylation levels of the transcription factor p63, the master regulator of the proliferative potential of epidermal SCs, can be used to distinguish self-renewing SCs from TA cells with more limited proliferative potential. CRITICAL ISSUES As technologies advance, IFE SCs can be identified at a single-cell level. Refinements of their identification and characterization are critical, not only for SC biology but also for the development of novel clinical applications. FUTURE DIRECTIONS Understanding the signaling pathways that control self-renewal and differentiation of IFE SCs will aid in developing novel cell-based therapeutics targeting degenerative epidermal diseases and wound repair.
Collapse
Affiliation(s)
- Makoto Senoo
- Institute for Regenerative Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania
- Department of Animal Biology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
222
|
Vernersson Lindahl E, Garcia EL, Mills AA. An allelic series of Trp63 mutations defines TAp63 as a modifier of EEC syndrome. Am J Med Genet A 2013; 161A:1961-71. [PMID: 23775923 DOI: 10.1002/ajmg.a.36074] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 05/03/2013] [Indexed: 01/01/2023]
Abstract
Human Ectrodactyly, Ectodermal dysplasia, Clefting (EEC) syndrome is an autosomal dominant developmental disorder defined by limb deformities, skin defects, and craniofacial clefting. Although associated with heterozygous missense mutations in TP63, the genetic basis underlying the variable expressivity and incomplete penetrance of EEC is unknown. Here, we show that mice heterozygous for an allele encoding the Trp63 p.Arg318His mutation, which corresponds to the human TP63 p.Arg279His mutation found in patients with EEC, have features of human EEC. Using an allelic series, we discovered that whereas clefting and skin defects are caused by loss of Trp63 function, limb anomalies are due to gain- and/or dominant-negative effects of Trp63. Furthermore, we identify TAp63 as a strong modifier of EEC-associated phenotypes with regard to both penetrance and expressivity.
Collapse
|
223
|
Celardo I, Grespi F, Antonov A, Bernassola F, Garabadgiu AV, Melino G, Amelio I. Caspase-1 is a novel target of p63 in tumor suppression. Cell Death Dis 2013; 4:e645. [PMID: 23703390 PMCID: PMC3674380 DOI: 10.1038/cddis.2013.175] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
p63 is a p53 family transcription factor, which besides unique roles in epithelial development, shares tumor suppressive activity with its homolog p53. The p63 gene has different transcriptional start sites, which generate two N-terminal isoforms (transactivation domain (TA)p63 and amino terminal truncated protein(ΔN)p63); in addition alternative splicing at the 5′-end give rise to at least five C-terminal isoforms. This complexity of gene structure has probably fostered the debate and controversy on p63 function in cancer, with TP63-harboring two distinctive promoters, codifying for the TAp63 and ΔNp63 isoforms, and having discrete functions. However, ΔNp63 also drives expression of target genes that have a relevant role in cancer and metastasis. In this study, we identified a novel p63 transcriptional target, caspase-1. Caspase-1 is proinflammatory caspase, which functions in tumor suppression. We show that both p63 isoforms promote caspase-1 expression by physical binding to its promoter. Consistent with our in vitro findings, we also identified a direct correlation between p63 and caspase-1 expression in human cancer data sets. In addition, survival estimation analysis demonstrated that functional interaction between p63 and caspase-1 represents a predictor of positive survival outcome in human cancers. Overall, our data report a novel p63 target gene involved in tumor suppression, and the clinical analysis underlines the biological relevance of this finding and suggests a further clinically predictive biomarker.
Collapse
Affiliation(s)
- I Celardo
- Medical Research Council, Toxicology Unit, Leicester University, Leicester, UK
| | | | | | | | | | | | | |
Collapse
|
224
|
Interaction between the TP63 and SHH pathways is an important determinant of epidermal homeostasis. Cell Death Differ 2013; 20:1080-8. [PMID: 23686138 DOI: 10.1038/cdd.2013.41] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 03/06/2013] [Accepted: 04/08/2013] [Indexed: 11/09/2022] Open
Abstract
Deregulation of the hedgehog (HH) pathway results in overexpression of the GLI target BCL2 and is an initiating event in specific tumor types including basal cell carcinoma of the skin. Regulation of the HH pathway during keratinocyte differentiation is not well understood. We measured HH pathway activity in response to differentiation stimuli in keratinocytes. An upregulation of suppressor of fused (SUFU), a negative regulator of the HH pathway, lowered HH pathway activity and was accompanied by loss of BCL2 expression associated with keratinocyte differentiation. We used in vitro and in vivo models to demonstrate that ΔNp63α, a crucial regulator of epidermal development, activates SUFU transcription in keratinocytes. Increasing SUFU protein levels inhibited GLI-mediated gene activation in suprabasal keratinocytes and promoted differentiation. Loss of SUFU expression caused deregulation of keratinocyte differentiation and BCL2 overexpression. Using in vivo murine models, we also provide evidence of GLI-mediated regulation of the TP63 pathway. p63 expression appears essential to establish an optimally functioning HH pathway. These observations present a regulatory mechanism by which SUFU acts as an interacting node between the HH and TP63 pathways to mediate differentiation and maintain epidermal homeostasis. Disruption of this regulatory node can be an important contributor to multistep carcinogenesis.
Collapse
|
225
|
p63-expressing cells are the stem cells of developing prostate, bladder, and colorectal epithelia. Proc Natl Acad Sci U S A 2013; 110:8105-10. [PMID: 23620512 DOI: 10.1073/pnas.1221216110] [Citation(s) in RCA: 171] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The tumor protein p63 (p63), and more specifically the NH2-terminal truncated (ΔN) p63 isoform, is a marker of basal epithelial cells and is required for normal development of several epithelial tissues, including the bladder and prostate glands. Although p63-expressing cells are proposed to be the stem cells of the developing prostate epithelium and bladder urothelium, cell lineages in these endoderm-derived epithelia remain highly controversial, and rigorous lineage tracing studies are warranted. Here, we generated knock-in mice expressing Cre recombinase (Cre) under the control of the endogenous ΔNp63 promoter. Heterozygote ΔNp63(+/Cre) mice were phenotypically normal and fertile. Cre-mediated recombination in ΔNp63(+/Cre);ROSA26(EYFP) reporter mice faithfully recapitulated the pattern of ΔNp63 expression and were useful for genetic lineage tracing of ΔNp63-expressing cells of the caudal endoderm in vivo. We found that ΔNp63-positive cells of the urogenital sinus generated all epithelial lineages of the prostate and bladder, indicating that these cells represent the stem/progenitor cells of those epithelia during development. We also observed ΔNp63 expression in caudal gut endoderm and the contribution of ΔNp63-positive cells to the stem/progenitor compartment of adult colorectal epithelium. Because p63 is a master regulator of stratified epithelial development, this finding provides a unique developmental insight into the cell of origin of squamous cell metaplasia and squamous cell carcinoma of the colon.
Collapse
|
226
|
Delineating Molecular Mechanisms of Squamous Tissue Homeostasis and Neoplasia: Focus on p63. J Skin Cancer 2013; 2013:632028. [PMID: 23710361 PMCID: PMC3655637 DOI: 10.1155/2013/632028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 03/14/2013] [Indexed: 11/18/2022] Open
Abstract
Mouse models have informed us that p63 is critical for normal epidermal development and homeostasis. The p53/p63/p73 family is expressed as multiple protein isoforms due to a combination of alternative promoter usage and C-terminal alternative splicing. These isoforms can mimic or interfere with one another, and their balance ultimately determines biological outcome in a context-dependent manner. While not frequently mutated, p63, and in particular the ΔNp63 subclass, is commonly overexpressed in human squamous cell cancers. In vitro keratinocytes and murine transgenic and transplantation models have been invaluable in elucidating the contribution of altered p63 levels to cancer development, and studies have identified the roles for ΔNp63 isoforms in keratinocyte survival and malignant progression, likely due in part to their transcriptional regulatory function. These findings can be extended to human cancers; for example, the novel recognition of NFκB/c-Rel as a downstream effector of p63 has identified a role for NFκB/c-Rel in human squamous cell cancers. These models will be critical in enhancing the understanding of the specific molecular mechanisms of cancer development and progression.
Collapse
|
227
|
Kim SY, Cordeiro MH, Serna VA, Ebbert K, Butler LM, Sinha S, Mills AA, Woodruff TK, Kurita T. Rescue of platinum-damaged oocytes from programmed cell death through inactivation of the p53 family signaling network. Cell Death Differ 2013; 20:987-97. [PMID: 23598363 DOI: 10.1038/cdd.2013.31] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 02/12/2013] [Accepted: 03/20/2013] [Indexed: 12/24/2022] Open
Abstract
Non-proliferating oocytes within avascular regions of the ovary are exquisitely susceptible to chemotherapy. Early menopause and sterility are unintended consequences of chemotherapy, and efforts to understand the oocyte apoptotic pathway may provide new targets for mitigating this outcome. Recently, the c-Abl kinase inhibitor imatinib mesylate (imatinib) has become the focus of research as a fertoprotective drug against cisplatin. However, the mechanism by which imatinib protects oocytes is not fully understood, and reports of the drug's efficacy have been contradictory. Using in vitro culture and subrenal grafting of mouse ovaries, we demonstrated that imatinib inhibits the cisplatin-induced apoptosis of oocytes within primordial follicles. We found that, before apoptosis, cisplatin induces c-Abl and TAp73 expression in the oocyte. Oocytes undergoing apoptosis showed downregulation of TAp63 and upregulation of Bax. While imatinib was unable to block cisplatin-induced DNA damage and damage response, such as the upregulation of p53, imatinib inhibited the cisplatin-induced nuclear accumulation of c-Abl/TAp73 and the subsequent downregulation of TAp63 and upregulation of Bax, thereby abrogating oocyte cell death. Surprisingly, the conditional deletion of Trp63, but not ΔNp63, in oocytes inhibited apoptosis, as well as the accumulation of c-Abl and TAp73 caused by cisplatin. These data suggest that TAp63 is the master regulator of cisplatin-induced oocyte death. The expression kinetics of TAp63, c-Abl and TAp73 suggest that cisplatin activates TAp63-dependent expression of c-Abl and TAp73 and, in turn, the activation of TAp73 by c-Abl-induced BAX expression. Our findings indicate that imatinib protects oocytes from cisplatin-induced cell death by inhibiting c-Abl kinase, which would otherwise activate TAp73-BAX-mediated apoptosis. Thus, imatinib and other c-Abl kinase inhibitors provide an intriguing new way to halt cisplatin-induced oocyte death in early follicles and perhaps conserve the endocrine function of the ovary against chemotherapy.
Collapse
Affiliation(s)
- S-Y Kim
- Division of Reproductive Biology and Clinical Research, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
228
|
Pelosi G, Rossi G, Cavazza A, Righi L, Maisonneuve P, Barbareschi M, Graziano P, Pastorino U, Garassino M, de Braud F, Papotti M. ΔNp63 (p40) distribution inside lung cancer: a driver biomarker approach to tumor characterization. Int J Surg Pathol 2013; 21:229-39. [PMID: 23486764 DOI: 10.1177/1066896913476750] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
ΔNp63 (henceforth simply p40) is a squamous/basal-type biomarker corresponding to nontransactivating (non-TA) isoforms of p63 gene. Its prospective relevance as driver biomarker in lung cancer has not yet been thoroughly investigated. In all, 72 adenocarcinomas (ADs), 27 squamous cell carcinomas (SQCs), 13 pleomorphic carcinomas (PLCs), 10 small-cell lung carcinomas (SCLCs), 5 large-cell neuroendocrine carcinomas (LCNECs), 5 adenosquamous carcinomas (ADSQCs), 3 large-cell carcinomas with basaloid features (B-LCC), 2 carcinoids, 2 carcinosarcomas (CS), 2 salivary-gland type tumors (SGTTs) of the lung, and 5 pleural malignant epithelioid mesotheliomas (MEMs) were prospectively diagnosed by morphology and verified by immunohistochemistry for p40, p63, and thyroid transcription factor 1 (TTF1). Histological scores (HS) were devised by multiplying the percentage of immunoreactive cells (0 to 100%) by immunostaining intensity (low = 1 vs strong = 2, according to internal controls). There was a nonrandom distribution of p40 across the diverse tumor groups and cell differentiation lineages, with p40-HS > 100 closely paralleling squamous or myoepithelial carcinomas (SQC, B-LCC, SQC-containing PLC, ADSQC with predominant SQC, SGTT), and p40-HS ≤ 10 pinpointing AD, AD-containing PLC, or CS and neuroendocrine (NE) tumors. At variance, p63-HS was significantly higher than p40 in AD (P < .0001) and NE tumors (P = .0156), with positive predictive value being 83% and 95% and overall accuracy being 95% and 99%, respectively. Also, TTF1 was shared by gland-differentiated and NE tumors. MEM cases were always negative for all biomarkers. The HS-guided assessment of p40 allowed an effective orientation among thoracic malignancies at the level of individual tumor patients thereby contributing to prospectively realize a driver, holistic approach to cancer characterization.
Collapse
Affiliation(s)
- Giuseppe Pelosi
- Dipartimento di Patologia Diagnostica e Laboratorio, Fondazione IRCCS Istituto Nazionale dei Tumori e Università degli Studi, Via G. Venezian, 1, I-20133 Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
229
|
Weiss RM, Guo S, Shan A, Shi H, Romano RA, Sinha S, Cantley LG, Guo JK. Brg1 determines urothelial cell fate during ureter development. J Am Soc Nephrol 2013; 24:618-26. [PMID: 23449535 DOI: 10.1681/asn.2012090902] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Developing and adult ureters express the epigenetic regulator Brg1, but the role of Brg1 in ureter development is not well understood. We conditionally ablated Brg1 in the developing ureter using Hoxb7-Cre and found that Brg1 expression is upstream of p63, Pparγ, and sonic hedgehog (Shh) expression in the ureteral epithelium. In addition, epithelial stratification in the basal cells required Brg1-dependent p63 expression, whereas terminal differentiation of the umbrella cells required Brg1-dependent Pparγ expression. Furthermore, the loss of ureteric Brg1 resulted in failure of Shh expression, which correlated with reduced smooth muscle cell development and hydroureter. Taken together, we conclude that Brg1 expression unifies three aspects of ureter development: maintenance of the basal cell population, guidance for terminal differentiation of urothelial cells, and proper investment of ureteral smooth muscle cells.
Collapse
Affiliation(s)
- Robert M Weiss
- Department of Urology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | | | | | |
Collapse
|
230
|
Lu Y, Abbassi S, Li F, Ding M, Wu G, Gu J, Zheng Q. Distinct function of P63 isoforms during embryonic skeletal development. Gene 2013; 519:251-9. [PMID: 23481305 DOI: 10.1016/j.gene.2013.02.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 02/15/2013] [Indexed: 11/26/2022]
Abstract
P63 belongs to the P53 family of transcription factors. There are multiple P63 isoforms that play important functions both in cancer and development. The obvious limb defect in p63 null mice and in human skeletal syndromes with P63 mutations suggest its essential role in long bone development. However, how the different P63 isoforms function during long bone development is largely unknown. We have previously shown that TAP63α, the longest P63 isoform, plays a positive role in embryonic skeletal development, since targeting TAP63α expression in hypertrophic chondrocytes accelerates endochondral ossification at both E17.5 and P1 stages. Here, we report transgenic studies of ΔNP63α, another P63 isoform which lacks the N-terminal transactivation domain compared to TAP63α, using the same hypertrophic chondrocyte-specific Col10a1 control element. No skeletal abnormalities were detected in these Col10a1-ΔNP63α transgenic mice at both E17.5 and P1 stages, suggesting less importance of ΔNP63α during late embryonic skeletal development. To further investigate the function of P63 isoforms during early skeletal development, we have generated ΔNP63α and TAP63α transgenic mice using a chondrocyte-specific Col2a1 control element. Surprisingly, while no skeletal defect was shown in the Col2a1-ΔNP63α transgenic mice, reduced ossification was observed in the digit and tail bones of Col2a1-TAP63α transgenic mice at both E17.5 and P1 stages compared to their wild-type littermates. Expression profiling and immunohistochemical analysis detected upregulated expression of Sox9, a major negative regulator of endochondral ossification, in Col2a1-TAP63α transgenic mice. Taken together, our results suggest a distinct function of P63 isoforms, herein, ΔNP63α and TAP63α, during endochondral ossification.
Collapse
Affiliation(s)
- Yaojuan Lu
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL 60612, USA
| | | | | | | | | | | | | |
Collapse
|
231
|
Su X, Chakravarti D, Flores ER. p63 steps into the limelight: crucial roles in the suppression of tumorigenesis and metastasis. Nat Rev Cancer 2013; 13:136-43. [PMID: 23344544 PMCID: PMC4181578 DOI: 10.1038/nrc3446] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The role of p63 in cancer has been an area of intense debate and controversy. Is TP63 (which encodes p63) a tumour suppressor gene or an oncogene? This debate is partly due to the complexity of the gene. There are several p63 isoforms - some with tumour suppressive functions and others with oncogenic functions. In this Opinion article, we focus on the recent advances in understanding p63 biology and its roles in cancer. In this regard, we discuss the role of p63 in multiple stem cell compartments, ageing, in the response to DNA damage and in DNA repair. Finally, we highlight the importance of understanding the interactions between all three p53 family members and the potential impact of this knowledge on cancer therapy and regenerative medicine.
Collapse
Affiliation(s)
- Xiaohua Su
- Department of Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | | | | |
Collapse
|
232
|
Lee B, Dai X. Transcriptional control of epidermal stem cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 786:157-73. [PMID: 23696356 DOI: 10.1007/978-94-007-6621-1_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Transcriptional regulation is fundamentally important for the progression of tissue stem cells through different stages of development and differentiation. Mammalian skin epidermis is an excellent model system to study such regulatory mechanisms due to its easy accessibility, stereotypic spatial arrangement, and availability of well-established cell type/lineage differentiation markers. Moreover, epidermis is one of the few mammalian tissues the stem cells of which can be maintained and propagated in culture to generate mature cell types and a functional tissue (reviewed in [1]), offering in vitro and ex vivo platforms to probe deep into the underlying cell and molecular mechanisms of biological functions.
Collapse
Affiliation(s)
- Briana Lee
- Department of Biological Chemistry, School of Medicine, University of California, D250 Med Sci I, Irvine 92697-1700, CA, USA
| | | |
Collapse
|
233
|
Abstract
In the past fifteen years, it has become apparent that tumour-associated p53 mutations can provoke activities that are different to those resulting from simply loss of wild-type tumour-suppressing p53 function. Many of these mutant p53 proteins acquire oncogenic properties that enable them to promote invasion, metastasis, proliferation and cell survival. Here we highlight some of the emerging molecular mechanisms through which mutant p53 proteins can exert these oncogenic functions.
Collapse
Affiliation(s)
- Patricia A J Muller
- The Beatson Institute for Cancer Research, Switchback Road, Bearsden, Glasgow, G61 1BD, UK
| | | |
Collapse
|
234
|
Antonini D, Sibilio A, Dentice M, Missero C. An Intimate Relationship between Thyroid Hormone and Skin: Regulation of Gene Expression. Front Endocrinol (Lausanne) 2013; 4:104. [PMID: 23986743 PMCID: PMC3749490 DOI: 10.3389/fendo.2013.00104] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 08/05/2013] [Indexed: 12/23/2022] Open
Abstract
Skin is the largest organ of the human body and plays a key role in protecting the individual from external insults. The barrier function of the skin is performed primarily by the epidermis, a self-renewing stratified squamous epithelium composed of cells that undergo a well-characterized and finely tuned process of terminal differentiation. By binding to their receptors thyroid hormones (TH) regulate epidermal cell proliferation, differentiation, and homeostasis. Thyroid dysfunction has multiple classical manifestations at skin level. Several TH-responsive genes, as well as genes critical for TH metabolism and action, are expressed at epidermal level. The role of TH in skin is still controversial, although it is generally recognized that TH signaling is central for skin physiology and homeostasis. Here we review the data on the epidermis and its function in relation to TH metabolism and regulation of gene expression. An understanding of the cellular and molecular basis of TH action in epidermal cells may lead to the identification of putative therapeutical targets for treatment of skin disorders.
Collapse
Affiliation(s)
| | - Annarita Sibilio
- Department of Clinical Medicine Surgery, University of Naples Federico II, Napoli, Italy
| | - Monica Dentice
- Department of Clinical Medicine Surgery, University of Naples Federico II, Napoli, Italy
| | - Caterina Missero
- CEINGE Biotecnologie Avanzate, Napoli, Italy
- Fondazione IRCCS SDN, Napoli, Italy
- *Correspondence: Caterina Missero, CEINGE Biotecnologie Avanzate, via Gaetano Salvatore 486, Napoli 80145, Italy e-mail:
| |
Collapse
|
235
|
Abstract
p63 is a transcriptional factor implicated in cancer and development. The presence in TP63 gene of alternative promoters allows expression of one isoform containing the N-terminal transactivation domain (TA isoform) and one N-terminal truncated isoform (ΔN isoform). Complete ablation of all p63 isoforms produced mice with fatal developmental abnormalities, including lack of epidermal barrier, limbs and other epidermal appendages. Specific TAp63-null mice, although they developed normally, failed to undergo in DNA damage-induced apoptosis during primordial follicle meiotic arrest, suggesting a p63 involvement in maternal reproduction. Recent findings have elucidated the role in DNA damage response of a novel Hominidae p63 isoform, GTAp63, specifically expressed in human spermatic precursors. Thus, these findings suggest a unique strategy of p63 gene, to evolve in order to preserve the species as a guardian of reproduction. Elucidation of the biological basis of p63 function in reproduction may provide novel approaches to the control of human fertility.
Collapse
Affiliation(s)
- Ivano Amelio
- Medical Research Council; Toxicology Unit; Leicester University; Leicester, UK
- Department for Molecular Biomedical Research; VIB; Ghent University; Ghent, Belgium
- Department of Biomedical Molecular Biology; Ghent University; Ghent, Belgium
| | - Francesca Grespi
- Medical Research Council; Toxicology Unit; Leicester University; Leicester, UK
- Department for Molecular Biomedical Research; VIB; Ghent University; Ghent, Belgium
- Department of Biomedical Molecular Biology; Ghent University; Ghent, Belgium
| | | | - Gerry Melino
- Medical Research Council; Toxicology Unit; Leicester University; Leicester, UK
- Department for Molecular Biomedical Research; VIB; Ghent University; Ghent, Belgium
- Department of Biomedical Molecular Biology; Ghent University; Ghent, Belgium
- Biochemistry IDI-IRCCS Laboratory and Department of Experimental Medicine and Surgery; University of Rome “Tor Vergata;” Rome, Italy
| |
Collapse
|
236
|
Balboni AL, Hutchinson JA, DeCastro AJ, Cherukuri P, Liby K, Sporn MB, Schwartz GN, Wells WA, Sempere LF, Yu PB, DiRenzo J. ΔNp63α-mediated activation of bone morphogenetic protein signaling governs stem cell activity and plasticity in normal and malignant mammary epithelial cells. Cancer Res 2012; 73:1020-30. [PMID: 23243027 DOI: 10.1158/0008-5472.can-12-2862] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Genetic analysis of TP63 indicates that ΔNp63 isoforms are required for preservation of regenerative stasis within diverse epithelial tissues. In squamous carcinomas, TP63 is commonly amplified, and ΔNp63α confers a potent survival advantage. Genome-wide occupancy studies show that ΔNp63 promotes bidirectional target gene regulation by binding more than 5,000 sites throughout the genome; however, the subset of targets mediating discreet activities of TP63 remains unclear. We report that ΔNp63α activates bone morphogenic proteins (BMP) signaling by inducing the expression of BMP7. Immunohistochemical analysis indicates that hyperactivation of BMP signaling is common in human breast cancers, most notably in the basal molecular subtype, as well as in several mouse models of breast cancer. Suppression of BMP signaling in vitro with LDN193189, a small-molecule inhibitor of BMP type I receptor kinases, represses clonogenicity and diminishes the cancer stem cell-enriched ALDH1(+) population. Importantly, LDN193189 blocks reconstitution of mixed ALDH1(+)/ALDH1(-) cultures indicating that BMP signaling may govern aspects of cellular plasticity within tumor hierarchies. These results show that BMP signaling enables reversion of committed populations to a stem-like state, potentially supporting progression and maintenance of tumorigenesis. Treatment of a mouse model of breast cancer with LDN193189 caused reduced expression of markers associated with epithelial-to-mesenchymal transition (EMT). Furthermore, in vivo limiting dilution analysis assays revealed that LDN193189 treatment suppressed tumor-initiating capacity and increased tumor latency. These studies support a model in which ΔNp63α-mediated activation of BMP signaling governs epithelial cell plasticity, EMT, and tumorigenicity during breast cancer initiation and progression.
Collapse
Affiliation(s)
- Amanda L Balboni
- Program in Experimental and Molecular Medicine, Department of Pharmacology and Toxicology, The Audrey and Theodor Geisel School of Medicine at Dartmouth, NH 03755, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
237
|
Ferone G, Mollo MR, Thomason HA, Antonini D, Zhou H, Ambrosio R, De Rosa L, Salvatore D, Getsios S, van Bokhoven H, Dixon J, Missero C. p63 control of desmosome gene expression and adhesion is compromised in AEC syndrome. Hum Mol Genet 2012; 22:531-43. [PMID: 23108156 PMCID: PMC3542863 DOI: 10.1093/hmg/dds464] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Ankyloblepharon, ectodermal defects, cleft lip/palate (AEC) syndrome is a rare autosomal dominant disorder caused by mutations in the p63 gene, essential for embryonic development of stratified epithelia. The most severe cutaneous manifestation of this disorder is the long-lasting skin fragility associated with severe skin erosions after birth. Using a knock-in mouse model for AEC syndrome, we found that skin fragility was associated with microscopic blistering between the basal and suprabasal compartments of the epidermis and reduced desmosomal contacts. Expression of desmosomal cadherins and desmoplakin was strongly reduced in AEC mutant keratinocytes and in newborn epidermis. A similar impairment in desmosome gene expression was observed in human keratinocytes isolated from AEC patients, in p63-depleted keratinocytes and in p63 null embryonic skin, indicating that p63 mutations causative of AEC syndrome have a dominant-negative effect on the wild-type p63 protein. Among the desmosomal components, desmocollin 3, desmoplakin and desmoglein 1 were the most significantly reduced by mutant p63 both at the RNA and protein levels. Chromatin immunoprecipitation experiments and transactivation assays revealed that p63 controls these genes at the transcriptional level. Consistent with reduced desmosome function, AEC mutant and p63-deficient keratinocytes had an impaired ability to withstand mechanical stress, which was alleviated by epidermal growth factor receptor inhibitors known to stabilize desmosomes. Our study reveals that p63 is a crucial regulator of a subset of desmosomal genes and that this function is impaired in AEC syndrome. Reduced mechanical strength resulting from p63 mutations can be alleviated pharmacologically by increasing desmosome adhesion with possible therapeutic implications.
Collapse
|
238
|
Gallant-Behm CL, Ramsey MR, Bensard CL, Nojek I, Tran J, Liu M, Ellisen LW, Espinosa JM. ΔNp63α represses anti-proliferative genes via H2A.Z deposition. Genes Dev 2012; 26:2325-36. [PMID: 23019126 DOI: 10.1101/gad.198069.112] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
ΔNp63α is a member of the p53 family of transcription factors that functions as an oncogene in squamous cell carcinomas (SCCs). Because ΔNp63α and p53 bind virtually identical DNA sequence motifs, it has been proposed that ΔNp63α functions as a dominant-negative inhibitor of p53 to promote proliferation and block apoptosis. However, most SCCs concurrently overexpress ΔNp63α and inactivate p53, suggesting the autonomous action of these oncogenic events. Here we report the discovery of a novel mechanism of transcriptional repression by ΔNp63α that reconciles these observations. We found that although both proteins bind the same genomic sites, they regulate largely nonoverlapping gene sets. Upon activation, p53 binds all enhancers regardless of ΔNp63α status but fails to transactivate genes repressed by ΔNp63α. We found that ΔNp63α associates with the SRCAP chromatin regulatory complex involved in H2A/H2A.Z exchange and mediates H2A.Z deposition at its target loci. Interestingly, knockdown of SRCAP subunits or H2A.Z leads to specific induction of ΔNp63α-repressed genes. We identified SAMD9L as a key anti-proliferative gene repressed by ΔNp63α and H2A.Z whose depletion suffices to reverse the arrest phenotype caused by ΔNp63α knockdown. Collectively, these results illuminate a molecular pathway contributing to the autonomous oncogenic effects of ΔNp63α.
Collapse
|
239
|
Romano RA, Smalley K, Magraw C, Serna VA, Kurita T, Raghavan S, Sinha S. Δ Np63 knockout mice reveal its indispensable role as a master regulator of epithelial development and differentiation. J Cell Sci 2012. [DOI: 10.1242/jcs.108647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|