1
|
Low-Calle AM, Ghoneima H, Ortega N, Cuibus AM, Katz C, Prives C, Prywes R. A Non-Canonical Hippo Pathway Represses the Expression of ΔNp63. Mol Cell Biol 2024; 44:27-42. [PMID: 38270135 PMCID: PMC10829837 DOI: 10.1080/10985549.2023.2292037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 11/28/2023] [Indexed: 01/26/2024] Open
Abstract
The p63 transcription factor, a member of the p53 family, plays an oncogenic role in squamous cell carcinomas, while in breast cancers its expression is often repressed. In the canonical conserved Hippo pathway, known to play a complex role in regulating growth of cancer cells, protein kinases MST1/2 and LATS1/2 act sequentially to phosphorylate and inhibit the YAP/TAZ transcription factors. We found that in MCF10A mammary epithelial cells as well as in squamous and breast cancer cell lines, expression of ΔNp63 RNA and protein is strongly repressed by inhibition of the Hippo pathway protein kinases. While MST1/2 and LATS1 are required for p63 expression, the next step of the pathway, namely phosphorylation and degradation of the YAP/TAZ transcriptional activators is not required for p63 repression. This suggests that regulation of p63 expression occurs by a noncanonical version of the Hippo pathway. We identified similarly regulated genes, suggesting the broader importance of this pathway. Interestingly, lowering p63 expression lead to increased YAP protein levels, indicating crosstalk of the YAP/TAZ-independent and -dependent branches of the Hippo pathway. These results, which reveal the intersection of the Hippo and p63 pathways, may prove useful for the control of their activities in cancer cells.
Collapse
Affiliation(s)
- Ana Maria Low-Calle
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Hana Ghoneima
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Nicholas Ortega
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Adriana M. Cuibus
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Chen Katz
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Carol Prives
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Ron Prywes
- Department of Biological Sciences, Columbia University, New York, New York, USA
| |
Collapse
|
2
|
Li Y, Giovannini S, Wang T, Fang J, Li P, Shao C, Wang Y, Shi Y, Candi E, Melino G, Bernassola F. p63: a crucial player in epithelial stemness regulation. Oncogene 2023; 42:3371-3384. [PMID: 37848625 PMCID: PMC10638092 DOI: 10.1038/s41388-023-02859-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/26/2023] [Accepted: 10/02/2023] [Indexed: 10/19/2023]
Abstract
Epithelial tissue homeostasis is closely associated with the self-renewal and differentiation behaviors of epithelial stem cells (ESCs). p63, a well-known marker of ESCs, is an indispensable factor for their biological activities during epithelial development. The diversity of p63 isoforms expressed in distinct tissues allows this transcription factor to have a wide array of effects. p63 coordinates the transcription of genes involved in cell survival, stem cell self-renewal, migration, differentiation, and epithelial-to-mesenchymal transition. Through the regulation of these biological processes, p63 contributes to, not only normal epithelial development, but also epithelium-derived cancer pathogenesis. In this review, we provide an overview of the role of p63 in epithelial stemness regulation, including self-renewal, differentiation, proliferation, and senescence. We describe the differential expression of TAp63 and ΔNp63 isoforms and their distinct functional activities in normal epithelial tissues and in epithelium-derived tumors. Furthermore, we summarize the signaling cascades modulating the TAp63 and ΔNp63 isoforms as well as their downstream pathways in stemness regulation.
Collapse
Affiliation(s)
- Yanan Li
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, 215000, China
| | - Sara Giovannini
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Tingting Wang
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, 215000, China
| | - Jiankai Fang
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, 215000, China
| | - Peishan Li
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, 215000, China
| | - Changshun Shao
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, 215000, China
| | - Ying Wang
- Shanghai Institute of Nutrition and Health, Shanghai, 200031, China
| | - Yufang Shi
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, 215000, China.
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy.
- Biochemistry Laboratory, Istituto Dermopatico Immacolata (IDI-IRCCS), 00100, Rome, Italy.
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy.
| | - Francesca Bernassola
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy.
| |
Collapse
|
3
|
Fisher ML, Balinth S, Mills AA. ΔNp63α in cancer: importance and therapeutic opportunities. Trends Cell Biol 2023; 33:280-292. [PMID: 36115734 PMCID: PMC10011024 DOI: 10.1016/j.tcb.2022.08.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/09/2022] [Accepted: 08/22/2022] [Indexed: 10/14/2022]
Abstract
Our understanding of cancer and the key pathways that drive cancer survival has expanded rapidly over the past several decades. However, there are still important challenges that continue to impair patient survival, including our inability to target cancer stem cells (CSCs), metastasis, and drug resistance. The transcription factor p63 is a p53 family member with multiple isoforms that carry out a wide array of functions. Here, we discuss the critical importance of the ΔNp63α isoform in cancer and potential therapeutic strategies to target ΔNp63α expression to impair the CSC population, as well as to prevent metastasis and drug resistance to improve patient survival.
Collapse
Affiliation(s)
- Matthew L Fisher
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Seamus Balinth
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Molecular and Cellular Biology Program, Stony Brook University, Stony Brook, NY 11794, USA
| | - Alea A Mills
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
4
|
Low-Calle AM, Ghoneima H, Ortega N, Cuibus AM, Katz C, Tong D, Prives C, Prywes R. A non-canonical Hippo pathway represses the expression of ΔNp63. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.13.528336. [PMID: 36824867 PMCID: PMC9949004 DOI: 10.1101/2023.02.13.528336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
The p63 transcription factor, a member of the p53 family, plays an oncogenic role in squamous cancers, while in breast cancers its expression is often repressed. In the canonical conserved Hippo pathway, known to play a complex role in regulating growth of cancer cells, the protein kinases MST1/2 and LATS1/2 act sequentially to phosphorylate and inhibit the YAP/TAZ transcription factors. We found that in the MCF10A mammary epithelial cell line as well as in squamous and breast cancer cell lines, expression of ΔNp63 RNA and protein is strongly repressed by inhibition of the Hippo pathway protein kinases in a manner that is independent of p53. While MST1/2 and LATS1 are required for p63 expression, the next step of the pathway, namely phosphorylation and degradation of the YAP/TAZ transcriptional activators is not required for repression of p63. This suggests that regulation of p63 expression occurs by a non-canonical version of the Hippo pathway. We additionally identified additional genes that were similarly regulated suggesting the broader importance of this pathway. Interestingly, we observed that experimentally lowering p63 expression leads to increased YAP protein levels, thereby constituting a feedback loop. These results, which reveal the intersection of the Hippo and p63 pathways, may prove useful for the control of their activities in cancer cells. One Sentence Summary Regulation of p63 expression occurs by a non-canonical version of the Hippo pathway in mammary epithelial, breast carcinoma and head and neck squamous carcinoma cells.
Collapse
|
5
|
Xu Y, Yang X, Xiong Q, Han J, Zhu Q. The dual role of p63 in cancer. Front Oncol 2023; 13:1116061. [PMID: 37182132 PMCID: PMC10174455 DOI: 10.3389/fonc.2023.1116061] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 04/13/2023] [Indexed: 05/16/2023] Open
Abstract
The p53 family is made up of three transcription factors: p53, p63, and p73. These proteins are well-known regulators of cell function and play a crucial role in controlling various processes related to cancer progression, including cell division, proliferation, genomic stability, cell cycle arrest, senescence, and apoptosis. In response to extra- or intracellular stress or oncogenic stimulation, all members of the p53 family are mutated in structure or altered in expression levels to affect the signaling network, coordinating many other pivotal cellular processes. P63 exists as two main isoforms (TAp63 and ΔNp63) that have been contrastingly discovered; the TA and ΔN isoforms exhibit distinguished properties by promoting or inhibiting cancer progression. As such, p63 isoforms comprise a fully mysterious and challenging regulatory pathway. Recent studies have revealed the intricate role of p63 in regulating the DNA damage response (DDR) and its impact on diverse cellular processes. In this review, we will highlight the significance of how p63 isoforms respond to DNA damage and cancer stem cells, as well as the dual role of TAp63 and ΔNp63 in cancer.
Collapse
Affiliation(s)
- Yongfeng Xu
- Abdominal Oncology Ward, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xiaojuan Yang
- Abdominal Oncology Ward, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Qunli Xiong
- Abdominal Oncology Ward, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Junhong Han
- State Key Laboratory of Biotherapy and Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Qing Zhu, ; Junhong Han,
| | - Qing Zhu
- Abdominal Oncology Ward, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Qing Zhu, ; Junhong Han,
| |
Collapse
|
6
|
Barrier-Forming Potential of Epithelial Cells from the Exstrophic Bladder. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:943-955. [PMID: 35358476 PMCID: PMC9194657 DOI: 10.1016/j.ajpath.2022.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/04/2022] [Accepted: 03/03/2022] [Indexed: 11/20/2022]
Abstract
Bladder exstrophy (BEX) is a rare developmental abnormality resulting in an open, exposed bladder plate. Although normal bladder urothelium is a mitotically quiescent barrier epithelium, histologic studies of BEX epithelia report squamous and proliferative changes that can persist beyond surgical closure. The current study examined whether patient-derived BEX epithelial cells in vitro were capable of generating a barrier-forming epithelium under permissive conditions. Epithelial cells isolated from 11 BEX samples, classified histologically as transitional (n = 6) or squamous (n = 5), were propagated in vitro. In conditions conducive to differentiated tight barrier formation by normal human urothelial cell cultures, 8 of 11 BEX lines developed transepithelial electrical resistances of more than 1000 Ω.cm2, with 3 squamous lines failing to generate tight barriers. An inverse relationship was found between expression of squamous KRT14 transcript and barrier development. Transcriptional drivers of urothelial differentiation PPARG, GATA3, and FOXA1 showed reduced expression in squamous BEX cultures. These findings implicate developmental interruption of urothelial transcriptional programming in the spectrum of transitional to squamous epithelial phenotypes found in BEX. Assessment of BEX epithelial phenotype may inform management and treatment strategies, for which distinction between reversible versus intractably squamous epithelium could identify patients at risk of medical complications or those who are most appropriate for reconstructive tissue engineering strategies.
Collapse
|
7
|
Huang YJ, Lee TC, Pai YC, Lin BR, Turner JR, Yu LCH. A novel tumor suppressor role of myosin light chain kinase splice variants through downregulation of the TEAD4/CD44 axis. Carcinogenesis 2021; 42:961-974. [PMID: 34000008 PMCID: PMC8283729 DOI: 10.1093/carcin/bgab038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 05/03/2021] [Accepted: 05/12/2021] [Indexed: 12/24/2022] Open
Abstract
Myosin light chain kinase (MLCK) regulates actinomyosin contraction. Two splice variants of long MLCK are expressed in epithelial cells and divergently regulate gut barrier functions; reduced MLCK levels in human colorectal cancers (CRC) with unclarified significance have been reported. CRC are solid tumors clonally sustained by stem cells highly expressing CD44 and CD133. The aim was to investigate the role of MLCK splice variants in CRC tumorigenesis. We found lower MLCK1/2 and higher CD44 expression in human CRC, but no change in CD133 or LGR5. Large-scale bioinformatics showed an inverse relationship between MYLK and CD44 in human sample gene datasets. A 3-fold increased tumor burden was observed in MLCK(-/-) mice compared with wild-type (WT) mice in a chemical-induced CRC model. Primary tumorspheres derived from the MLCK(-/-) mice displayed larger sizes and higher CD44 transcript levels than those from the WT mice. Bioinformatics revealed binding of TEAD4 (a transcriptional enhancer factor family member in the Hippo pathway) to CD44 promoter, which was confirmed by luciferase reporter assay. Individually expressing MLCK1 and MLCK2 variants in the MLCK-knockout (KO) Caco-2 cells inhibited the nuclear localization of TEAD4 cofactors, VGLL3 and YAP1, respectively, and both variants reduced the CD44 transcription. Accelerated cell cycle transit was observed in the MLCK-KO cells, whereby expression of MLCK1/2 variants counterbalanced the cell hyperproliferation. In conclusion, MLCK1/2 variants are novel tumor suppressors by downregulating the TEAD4/CD44 axis via reducing nuclear translocation of distinct transcriptional coactivators. The reduction of epithelial MLCKs, especially isoform 2, may drive cancer stemness and tumorigenesis.
Collapse
Affiliation(s)
- Yen-Ju Huang
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan ROC
| | - Tsung-Chun Lee
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan ROC.,Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan ROC
| | - Yu-Chen Pai
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan ROC
| | - Been-Ren Lin
- Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan ROC
| | - Jerrold R Turner
- Brigham's Women Hospital, Harvard Medical School, Boston, MA, USA
| | - Linda Chia-Hui Yu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan ROC
| |
Collapse
|
8
|
Cam M, Charan M, Welker AM, Dravid P, Studebaker AW, Leonard JR, Pierson CR, Nakano I, Beattie CE, Hwang EI, Kambhampati M, Nazarian J, Finlay JL, Cam H. ΔNp73/ETS2 complex drives glioblastoma pathogenesis- targeting downstream mediators by rebastinib prolongs survival in preclinical models of glioblastoma. Neuro Oncol 2021; 22:345-356. [PMID: 31763674 DOI: 10.1093/neuonc/noz190] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) remains one of the least successfully treated cancers. It is essential to understand the basic biology of this lethal disease and investigate novel pharmacological targets to treat GBM. The aims of this study were to determine the biological consequences of elevated expression of ΔNp73, an N-terminal truncated isoform of TP73, and to evaluate targeting of its downstream mediators, the angiopoietin 1 (ANGPT1)/tunica interna endothelial cell kinase 2 (Tie2) axis, by using a highly potent, orally available small-molecule inhibitor (rebastinib) in GBM. METHODS ΔNp73 expression was assessed in glioma sphere cultures, xenograft glioblastoma tumors, and glioblastoma patients by western blot, quantitative reverse transcription PCR, and immunohistochemistry. Immunoprecipitation, chromatin immunoprecipitation (ChiP) and sequential ChIP were performed to determine the interaction between ΔNp73 and E26 transformation-specific (ETS) proto-oncogene 2 (ETS2) proteins. The oncogenic consequences of ΔNp73 expression in glioblastomas were examined by in vitro and in vivo experiments, including orthotopic zebrafish and mouse intracranial-injection models. Effects of rebastinib on growth of established tumors and survival were examined in an intracranial-injection mouse model. RESULTS ΔNp73 upregulates both ANGPT1 and Tie2 transcriptionally through ETS conserved binding sites on the promoters by interacting with ETS2. Elevated expression of ΔNp73 promotes tumor progression by mediating angiogenesis and survival. Therapeutic targeting of downstream ΔNp73 signaling pathways by rebastinib inhibits growth of established tumors and extends survival in preclinical models of glioblastoma. CONCLUSION Aberrant expression of ΔNp73 in GBM promotes tumor progression through autocrine and paracrine signaling dependent on Tie2 activation by ANGPT1. Disruption of this signaling by rebastinib improves tumor response to treatment in glioblastoma.
Collapse
Affiliation(s)
- Maren Cam
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio
| | - Manish Charan
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio
| | - Alessandra M Welker
- Cancer Center and Regenerative Medicine, Massachusetts General Hospital, Boston
| | - Piyush Dravid
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio
| | - Adam W Studebaker
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio
| | - Jeffrey R Leonard
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio
| | - Christopher R Pierson
- Department of Pathology & Laboratory Medicine, Nationwide Children's Hospital, Columbus, Ohio
| | - Ichiro Nakano
- Comprehensive Cancer Center, University of Alabama, Birmingham, Alabama
| | - Christine E Beattie
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Eugene I Hwang
- Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC
| | - Madhuri Kambhampati
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC
| | - Javad Nazarian
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC
| | - Jonathan L Finlay
- Neuro-oncology Program, Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Hakan Cam
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| |
Collapse
|
9
|
Morand GB, Ikenberg K, Vital DG, Cardona I, Moch H, Stoeckli SJ, Huber GF. Preoperative assessment of CD44‐mediated depth of invasion as predictor of occult metastases in early oral squamous cell carcinoma. Head Neck 2018; 41:950-958. [DOI: 10.1002/hed.25532] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 07/31/2018] [Accepted: 09/28/2018] [Indexed: 12/14/2022] Open
Affiliation(s)
- Grégoire B. Morand
- Department of Otorhinolaryngology, Head and Neck SurgeryUniversity Hospital Zurich Zurich Switzerland
| | - Kristian Ikenberg
- Institute of Pathology and Molecular PathologyUniversity Hospital Zurich Zurich Switzerland
| | - Domenic G. Vital
- Department of Otorhinolaryngology, Head and Neck SurgeryUniversity Hospital Zurich Zurich Switzerland
| | - Isabel Cardona
- Department of Otolaryngology, Head and Neck SurgeryMcGill University Montreal Québec Canada
| | - Holger Moch
- Institute of Pathology and Molecular PathologyUniversity Hospital Zurich Zurich Switzerland
| | - Sandro J. Stoeckli
- Department of Otorhinolaryngology, Head and Neck SurgeryKantonsspital St. Gallen St. Gallen Switzerland
| | - Gerhard F. Huber
- Department of Otorhinolaryngology, Head and Neck SurgeryUniversity Hospital Zurich Zurich Switzerland
| |
Collapse
|
10
|
Curtarelli RB, Gonçalves JM, dos Santos LGP, Savi MG, Nör JE, Mezzomo LAM, Rodríguez Cordeiro MM. Expression of Cancer Stem Cell Biomarkers in Human Head and Neck Carcinomas: a Systematic Review. Stem Cell Rev Rep 2018; 14:769-784. [DOI: 10.1007/s12015-018-9839-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
11
|
Dudek AM, Vermeulen SH, Kolev D, Grotenhuis AJ, Kiemeney LALM, Verhaegh GW. Identification of an enhancer region within the TP63/LEPREL1 locus containing genetic variants associated with bladder cancer risk. Cell Oncol (Dordr) 2018; 41:555-568. [PMID: 29956121 PMCID: PMC6153957 DOI: 10.1007/s13402-018-0393-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2018] [Indexed: 12/24/2022] Open
Abstract
Purpose Genome-wide association studies (GWAS) have led to the identification of a bladder cancer susceptibility variant (rs710521) in a non-coding intergenic region between the TP63 and LEPREL1 genes on chromosome 3q28, suggesting a role in the transcriptional regulation of these genes. In this study, we aimed to functionally characterize the 3q28 bladder cancer risk locus. Methods Fine-mapping was performed by focusing on the region surrounding rs710521, and variants were prioritized for further experiments using ENCODE regulatory data. The enhancer activity of the identified region was evaluated using dual-luciferase assays. CRISPR/Cas9-mediated deletion of the enhancer region was performed and the effect of this deletion on cell proliferation and gene expression levels was evaluated using CellTiter-Glo and RT-qPCR, respectively. Results Fine-mapping of the GWAS signal region led to the identification of twenty SNPs that showed a stronger association with bladder cancer risk than rs710521. Using publicly available data on regulatory elements and sequences, an enhancer region containing the bladder cancer risk variants was identified. Through reporter assays, we found that the presence of the enhancer region significantly increased ΔNTP63 promoter activity in bladder cancer-derived cell lines. CRISPR/Cas9-mediated deletion of the enhancer region reduced the viability of bladder cancer cells by decreasing the expression of ΔNTP63 and p63 target genes. Conclusions Taken together, our data show that bladder cancer risk-associated variants on chromosome 3q28 are located in an active enhancer region. Further characterization of the allele-specific activity of the identified enhancer and its target genes may lead to the identification of novel signaling pathways involved in bladder carcinogenesis. Electronic supplementary material The online version of this article (10.1007/s13402-018-0393-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Aleksandra M Dudek
- Department of Urology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Sita H Vermeulen
- Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dimitar Kolev
- Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anne J Grotenhuis
- Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lambertus A L M Kiemeney
- Department of Urology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
- Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gerald W Verhaegh
- Department of Urology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands.
| |
Collapse
|
12
|
Mohajertehran F, Sahebkar A, Zare R, Mohtasham N. The promise of stem cell markers in the diagnosis and therapy of epithelial dysplasia and oral squamous cell carcinoma. J Cell Physiol 2018; 233:8499-8507. [PMID: 29797575 DOI: 10.1002/jcp.26789] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/30/2018] [Indexed: 12/16/2022]
Abstract
Oral squamous cell carcinoma (OSCC) is the most common type of head and neck cancer. Epithelial dysplasia is often initiated in the cells and cell nuclei adjacent to the epithelial cell membrane. Reduced cell-cell adhesions enable cancer cells to detach from the tumor and disseminate to other organs. The mutations in epithelial dysplasia markers such as E-cadherin and epithelial cell adhesion molecules (CD326) can lead to proliferation, growth and survival of the tumor cells and persistence of numerous malignancies that play a key role in epithelial dysplasia of OSCC. Accordingly, these genes can be considered prognostic markers or potential therapeutic targets for the tailored management of patients with OSCC. The gene expression profile of OSCC stem cells indicates a differential pattern that facilitates establishing a cell signature. Owing to the highly tumorigenic behavior of cancer stem cells and the role of these cells in tumor differentiation, treatment resistance, relapse, and metastasis, we reviewed the role of stem cell markers in epithelial dysplasia and OSCC.
Collapse
Affiliation(s)
- Farnaz Mohajertehran
- Oral and Maxillofacial Disease Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Dental Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Zare
- Dental Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nooshin Mohtasham
- Dental Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
13
|
ΔNp63 drives metastasis in breast cancer cells via PI3K/CD44v6 axis. Oncotarget 2018; 7:54157-54173. [PMID: 27494839 PMCID: PMC5342332 DOI: 10.18632/oncotarget.11022] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 07/22/2016] [Indexed: 01/03/2023] Open
Abstract
P63 is a transcription factor belonging to the family of p53, essential for the development and differentiation of epithelia. In recent years, it has become clear that altered expression of the different isoforms of this gene can play an important role in carcinogenesis. The p63 gene encodes for two main isoforms known as TA and ΔN p63 with different functions. The role of these different isoforms in sustaining tumor progression and metastatic spreading however has not entirely been clarified. Here we show that breast cancer initiating cells express ΔNp63 isoform that supports a more mesenchymal phenotype associated with a higher tumorigenic and metastatic potential. On the contrary, the majority of cells within the tumor appears to express predominantly TAp63 isoform. While ΔNp63 exerts its effects by regulating a PI3K/CD44v6 pathway, TAp63 modulates this pathway in an opposite fashion. As a result, tumorigenicity and invasive capacity of breast cancer cells is a balance of the two isoforms. Finally, we found that tumor microenvironmental cytokines significantly contribute to the establishment of breast cancer cell phenotype by positively regulating ΔNp63 and CD44v6 expression.
Collapse
|
14
|
Sasaki Y, Tamura M, Takeda K, Ogi K, Nakagaki T, Koyama R, Idogawa M, Hiratsuka H, Tokino T. Identification and characterization of the intercellular adhesion molecule-2 gene as a novel p53 target. Oncotarget 2018; 7:61426-61437. [PMID: 27556181 PMCID: PMC5308662 DOI: 10.18632/oncotarget.11366] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 08/08/2016] [Indexed: 12/19/2022] Open
Abstract
The p53 tumor suppressor inhibits cell growth through the activation of both cell cycle arrest and apoptosis, which maintain genome stability and prevent cancer development. Here, we report that intercellular adhesion molecule-2 (ICAM2) is transcriptionally activated by p53. Specifically, ICAM2 is induced by the p53 family and DNA damage in a p53-dependent manner. We identified a p53 binding sequence located within the ICAM2 gene that is responsive to wild-type p53, TAp73, and TAp63. In terms of function, we found that the ectopic expression of ICAM2 inhibited cancer cell migration and invasion. In addition, we demonstrated that silencing endogenous ICAM2 in cancer cells caused a marked increase in extracellular signal-regulated kinase (ERK) phosphorylation levels, suggesting that ICAM2 inhibits migration and invasion of cancer cells by suppressing ERK signaling. Moreover, ICAM2 is underexpressed in human cancer tissues containing mutant p53 as compared to those with wild-type p53. Notably, the decreased expression of ICAM2 is associated with poor survival in patients with various cancers. Our findings demonstrate that ICAM2 induction by p53 has a key role in inhibiting migration and invasion.
Collapse
Affiliation(s)
- Yasushi Sasaki
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| | - Miyuki Tamura
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| | - Kousuke Takeda
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan.,Department of Oral Surgery, Sapporo Medical University, Sapporo, Japan
| | - Kazuhiro Ogi
- Department of Oral Surgery, Sapporo Medical University, Sapporo, Japan
| | - Takafumi Nakagaki
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan.,Department of Oral Surgery, Sapporo Medical University, Sapporo, Japan
| | - Ryota Koyama
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| | - Masashi Idogawa
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| | | | - Takashi Tokino
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| |
Collapse
|
15
|
Jimenez-Hernandez LE, Vazquez-Santillan K, Castro-Oropeza R, Martinez-Ruiz G, Muñoz-Galindo L, Gonzalez-Torres C, Cortes-Gonzalez CC, Victoria-Acosta G, Melendez-Zajgla J, Maldonado V. NRP1-positive lung cancer cells possess tumor-initiating properties. Oncol Rep 2017; 39:349-357. [PMID: 29138851 PMCID: PMC5783600 DOI: 10.3892/or.2017.6089] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 10/13/2017] [Indexed: 12/21/2022] Open
Abstract
Tumor-initiating cells possess the capacity for self-renewal and to create heterogeneous cell lineages within a tumor. Therefore, the identification and isolation of cancer stem cells is an essential step in the analysis of their biology. The aim of the present study was to determine whether the cell surface protein neuropilin 1 (NRP1) can be used as a biomarker of stem-like cells in lung cancer tumors. For this purpose, NRP1-negative (NRP1-) and NRP1-positive (NRP1+) cell subpopulations from two lung cancer cell lines were sorted by flow cytometry. The NRP1+ cell subpopulation showed an increased expression of pluripotency markers OCT-4, Bmi-1 and NANOG, as well as higher cell migration, clonogenic and self-renewal capacities. NRP1 gene knockdown resulted not only in a decreased expression of stemness markers but also in a decrease in the clonogenic, cell migration and self-renewal potential. In addition, the NRP1+ cell subpopulation exhibited dysregulated expression of epithelial-to-mesenchymal transition-associated genes, including the ΔNp63 isoform protein, a previously reported characteristic of cancer stem cells. Notably, a genome-wide expression analysis of NRP1-knockdown cells revealed a potential new NRP1 pathway involving OLFML3 and genes associated with mitochondrial function. In conclusion, we demonstrated that NRP1+ lung cancer cells have tumor-initiating properties. NRP1 could be a useful biomarker for tumor-initiating cells in lung cancer tumors.
Collapse
|
16
|
Yan W, Zhang Y, Chen X. TAp63γ and ΔNp63γ are regulated by RBM38 via mRNA stability and have an opposing function in growth suppression. Oncotarget 2017; 8:78327-78339. [PMID: 29108232 PMCID: PMC5667965 DOI: 10.18632/oncotarget.18463] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 04/23/2017] [Indexed: 11/25/2022] Open
Abstract
The p63 gene is expressed as TAp63 from the P1 promoter and as ΔNp63 from the P2 promoter. Through alternative splicing, five TA and five ΔN isoforms (α-ε) are expressed. Isoforms α-β and δ share an identical 3’ untranslated region (3’UTR) whereas isoform γ has a unique 3’UTR. Recently, we found that RBM38 RNA-binding protein is a target of p63 and RBM38 in turn regulates p63α/β expression via mRNA stability. However, it is uncertain whether p63γ has a unique biological activity and whether p63γ is regulated by RBM38. Here, we found that the levels of ΔNp63γ transcript and protein are induced upon overexpression of RBM38 but decreased by RBM38 knockdown. Conversely, we found that the levels of ΔNp63β transcript and protein are decreased by ectopic expression of RBM38 but increased by RBM38 knockdown, consistent with our previous report. Interestingly, RBM38 increases the half-life of p63γ mRNA by binding to a GU-rich element in p63γ 3’UTR. In contrast, our previous studies showed that RBM38 decreases the half-life of p63α/β mRNAs by binding to AU-/U-rich elements in their 3’UTR. We also found that knockout of p63γ in ME180 and HaCaT cells, in which ΔNp63 isoforms are predominant, inhibits cell proliferation and migration, suggesting that ΔNp63γ has a pro-growth activity. In contrast, we found that knockout of TAp63γ in MIA PaCa-2 cells, in which TAp63 isoforms are predominant, promotes cell proliferation, migration, and inhibits cellular senescence. Taken together, we conclude that ΔNp63γ has an oncogenic potential whereas TAp63γ is a tumor suppressor.
Collapse
Affiliation(s)
- Wensheng Yan
- The Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, California, USA
| | - Yanhong Zhang
- The Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, California, USA
| | - Xinbin Chen
- The Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, California, USA
| |
Collapse
|
17
|
Nekulova M, Holcakova J, Gu X, Hrabal V, Galtsidis S, Orzol P, Liu Y, Logotheti S, Zoumpourlis V, Nylander K, Coates PJ, Vojtesek B. ΔNp63α expression induces loss of cell adhesion in triple-negative breast cancer cells. BMC Cancer 2016; 16:782. [PMID: 27724925 PMCID: PMC5057421 DOI: 10.1186/s12885-016-2808-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 09/23/2016] [Indexed: 12/31/2022] Open
Abstract
Background p63, a member of the p53 protein family, plays key roles in epithelial development and carcinogenesis. In breast cancer, p63 expression has been found predominantly in basal-A (epithelial-type) triple-negative breast carcinomas (TNBC). To investigate the functional role of p63 in basal-A TNBC, we created MDA-MB-468 cell lines with inducible expression of the two major N-terminal p63 isoforms, TAp63α and ∆Np63α. Results TAp63α did not have significant effect on gene expression profile and cell phenotype, whilst the main effect of ΔNp63α was reduction of cell adhesion. Gene expression profiling revealed genes involved in cell adhesion and migration whose expression relies on overexpression of ΔNp63α. Reduced cell adhesion also led to decreased cell proliferation in vitro and in vivo. Similar data were obtained in another basal-A cell line, BT-20, but not in BT-549 basal-B (mesenchymal-like) TNBC cells. Conclusions In basal-A TNBC cells, ∆Np63α has much stronger effects on gene expression than TAp63α. Although p63 is mentioned mostly in connection with breast cell differentiation and stem cell regulation, we showed that a major effect of p63 is regulation of cell adhesion, a process important in metastasis and invasion of tumour cells. That this effect is not seen in mesenchymal-type TNBC cells suggests lineage-dependent functions, mirroring the expression of ∆Np63α in primary human breast cancers. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2808-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marta Nekulova
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno, 65653, Czech Republic
| | - Jitka Holcakova
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno, 65653, Czech Republic
| | - Xiaolian Gu
- Department of Medical Biosciences, Umeå University, Umeå, 90185, Sweden
| | - Vaclav Hrabal
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno, 65653, Czech Republic
| | - Sotiris Galtsidis
- Institute of Biology, Medicinal Chemistry & Biotechnology, NHRF, Athens, Greece
| | - Paulina Orzol
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno, 65653, Czech Republic
| | - Yajing Liu
- NCRC, 026-329S, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Stella Logotheti
- Institute of Biology, Medicinal Chemistry & Biotechnology, NHRF, Athens, Greece
| | | | - Karin Nylander
- Department of Medical Biosciences, Umeå University, Umeå, 90185, Sweden
| | - Philip J Coates
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno, 65653, Czech Republic
| | - Borivoj Vojtesek
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno, 65653, Czech Republic.
| |
Collapse
|
18
|
Mishra DK, Veena U, Kaliki S, Kethiri AR, Sangwan VS, Ali MH, Naik MN, Singh V. Differential Expression of Stem Cell Markers in Ocular Surface Squamous Neoplasia. PLoS One 2016; 11:e0161800. [PMID: 27584160 PMCID: PMC5008752 DOI: 10.1371/journal.pone.0161800] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 08/12/2016] [Indexed: 12/12/2022] Open
Abstract
Ocular Surface Squamous Neoplasm (OSSN) is the neoplasia arising from the conjunctiva, cornea and limbus. OSSN ranges from mild, moderate, severe dysplasia, carcinoma in situ (CIS) to squamous cell carcinoma (SCC). Recent findings on cancer stem cells theory indicate that population of stem-like cell as in neoplasia determines its heterogeneity and complexity leading to varying tumor development of metastatic behavior and recurrence. Cancer stem cell markers are not much explored in the cases of OSSN. In the present study, we aim to evaluate the expression of stem cells using stem cell markers mainly p63, ABCG2, c-KIT (CD117) and CD44 in OSSN tissue, which could have prognostic significance. The present study tries for the first time to explore expression of these stem markers in the cases of OSSN. These cases are subdivided into two groups. One group comprises of carcinoma in situ (n = 6) and the second group comprises of invasive carcinoma (n = 6). The mean age at presentation was 52 years; with 53 years for CIS group and 52 years for SCC group. From each group section from the paraffin block were taken for the IHC staining of p63, c-Kit, ABCG2 and CD44. Our experiments show high expression of P63 and CD44 in the cases of CIN and SCC. Both CIS and SCC displayed positive staining with p63, with more than 80% cells staining positive. However minimal expression of c-kit in both CIN and SCC. But surprisingly we got high expression of ABCG2 in cases of carcinoma in situ as compared to that of invasive squamous cell carcinoma. More than 50% of cells showed CD44 positivity in both CIS and SCC groups. Our results show for the first time that these four stem cells especially the limbal epithelium stem cells play a vital role in the genesis of OSSN but we need to explore more cases before establishing its clinical and biological significance.
Collapse
Affiliation(s)
- Dilip Kumar Mishra
- Ophthalmic Pathology Laboratory, L V Prasad Eye Institute, Hyderabad, Telangana, India
| | - Uppala Veena
- Ophthalmic Pathology Laboratory, L V Prasad Eye Institute, Hyderabad, Telangana, India
| | - Swathi Kaliki
- The Operation Eyesight Universal Institute for Eye cancer, L V Prasad Eye Institute, Hyderabad, Telangana, India
| | - Abhinav Reddy Kethiri
- Champalimaud Translational Centre for Eye Research and Tej Kohli Cornea Institute, L V Prasad Eye Institute, Hyderabad, Telangana, India
- Prof. Brien Holden Eye Research Center, and Center for Ocular Regeneration, L V Prasad Eye Institute, Hyderabad, Telangana, India
- Manipal University, Manipal, Karnataka, India
| | - Virender S. Sangwan
- Champalimaud Translational Centre for Eye Research and Tej Kohli Cornea Institute, L V Prasad Eye Institute, Hyderabad, Telangana, India
- Prof. Brien Holden Eye Research Center, and Center for Ocular Regeneration, L V Prasad Eye Institute, Hyderabad, Telangana, India
| | - Mohammed Hasnat Ali
- Clinical Epidemiology and Bio-Statistics, L V Prasad Eye Institute, Hyderabad, Telangana, India
| | - Milind N. Naik
- The Operation Eyesight Universal Institute for Eye cancer, L V Prasad Eye Institute, Hyderabad, Telangana, India
| | - Vivek Singh
- Champalimaud Translational Centre for Eye Research and Tej Kohli Cornea Institute, L V Prasad Eye Institute, Hyderabad, Telangana, India
- Prof. Brien Holden Eye Research Center, and Center for Ocular Regeneration, L V Prasad Eye Institute, Hyderabad, Telangana, India
- Manipal University, Manipal, Karnataka, India
| |
Collapse
|
19
|
Orzol P, Nekulova M, Holcakova J, Muller P, Votesek B, Coates PJ. ΔNp63 regulates cell proliferation, differentiation, adhesion, and migration in the BL2 subtype of basal-like breast cancer. Tumour Biol 2016; 37:10133-40. [PMID: 26825981 DOI: 10.1007/s13277-016-4880-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/15/2016] [Indexed: 12/21/2022] Open
Abstract
Triple-negative breast cancers (TNBC) comprise a heterogeneous subgroup of tumors with a generally poor prognosis. Subclassification of TNBC based on genomic analyses shows that basal-like TNBCs, specifically the basal A or BL2 subtype, are characterized by the expression of ΔNp63, a transcription factor that has been attributed a variety of roles in the regulation of proliferation, differentiation, and cell survival. To investigate the role(s) of p63 in basal-like breast cancers, we used HCC1806 cells that are classified as basal A/BL2. We show that these cells endogenously express p63, mainly as the ΔNp63α isoform. TP63 gene knockout by CRISPR resulted in viable cells that proliferate more slowly and adhere less tightly, with an increased rate of migration. Analysis of adhesion-related gene expression revealed a complex set of alterations in p63-depleted cells, with both increased and decreased adhesion molecules and adhesion substrates compared to parental cells expressing p63. Examination of the phenotype of these cells indicated that endogenous p63 is required to suppress the expression of luminal markers and maintain the basal epithelial phenotype, with increased levels of both CK8 and CK18 and a reduction in N-cadherin levels in cells lacking p63. On the other hand, the level of CK5 was not decreased and ER was not increased, indicating that p63 loss is insufficient to induce full luminal-type differentiation. Taken together, these data demonstrate that p63 exerts multiple pro-oncogenic effects on cell differentiation, proliferation and adhesion in basal-like breast cancers.
Collapse
Affiliation(s)
- Paulina Orzol
- Regional Centre of Applied and Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53, Brno, Czech Republic
| | - Marta Nekulova
- Regional Centre of Applied and Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53, Brno, Czech Republic
| | - Jitka Holcakova
- Regional Centre of Applied and Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53, Brno, Czech Republic
| | - Petr Muller
- Regional Centre of Applied and Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53, Brno, Czech Republic
| | - Borivoj Votesek
- Regional Centre of Applied and Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53, Brno, Czech Republic.
| | - Philip J Coates
- Regional Centre of Applied and Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53, Brno, Czech Republic.
| |
Collapse
|
20
|
Abstract
In stratified epithelial and glandular tissues, homeostasis relies on the self-renewing capacity of stem cells, which are within the basal layer. The p53 family member p63 is an indispensable transcription factor for epithelial morphogenesis and stemness. A splice variant of the transcription factor p63 that lacks an amino-terminal domain, ΔNp63, is selectively found in the basal compartments of several ectoderm-derived tissues such as stratified and glandular epithelia, in which it is required for the replenishment of stem cells. Thus far, the transcriptional programs downstream of p63 in stemness regulation remain incompletely defined. Unveiling the molecular basis of stem cell self-renewal may be relevant in understanding how this process may contribute to cancer development. In this review, we specifically highlight experimental investigations, which suggest that p63 is a marker of normal epithelial stem cells and describe p63 transcriptional targets that may be involved in stemness regulation. Finally, we discuss relevant findings implicating p63 in epithelial cancer stem cell biology.
Collapse
Affiliation(s)
- Gerry Melino
- Biochemistry Laboratory, Istituto Dermopatico dell'Immacolata-Istituto di Ricovero e Cura a Carattere Scientifico (IDI-IRCCS), c/o Department of Experimental Medicine and Biochemical Sciences, University of Rome "Tor Vergata," 00133 Rome, Italy. Toxicology Unit, Medical Research Council, Leicester University, Hodgkin Building, P.O. Box 138, Leicester LE1 9HN, UK
| | - Elisa Maria Memmi
- Biochemistry Laboratory, Istituto Dermopatico dell'Immacolata-Istituto di Ricovero e Cura a Carattere Scientifico (IDI-IRCCS), c/o Department of Experimental Medicine and Biochemical Sciences, University of Rome "Tor Vergata," 00133 Rome, Italy
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, European Institute of Oncology, 20141 Milan, Italy. Department of Health Sciences, Milan University, 20142 Milan, Italy
| | - Francesca Bernassola
- Biochemistry Laboratory, Istituto Dermopatico dell'Immacolata-Istituto di Ricovero e Cura a Carattere Scientifico (IDI-IRCCS), c/o Department of Experimental Medicine and Biochemical Sciences, University of Rome "Tor Vergata," 00133 Rome, Italy. Department of Experimental Oncology, European Institute of Oncology, 20141 Milan, Italy.
| |
Collapse
|
21
|
|
22
|
Allegra A, Alonci A, Penna G, Innao V, Gerace D, Rotondo F, Musolino C. The cancer stem cell hypothesis: a guide to potential molecular targets. Cancer Invest 2014; 32:470-95. [PMID: 25254602 DOI: 10.3109/07357907.2014.958231] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Common cancer theories hold that tumor is an uncontrolled somatic cell proliferation caused by the progressive addition of random mutations in critical genes that control cell growth. Nevertheless, various contradictions related to the mutation theory have been reported previously. These events may be elucidated by the persistence of residual tumor cells, called Cancer Stem Cells (CSCs) responsible for tumorigenesis, tumor maintenance, tumor spread, and tumor relapse. Herein, we summarize the current understanding of CSCs, with a focus on the possibility to identify specific markers of CSCs, and discuss the clinical application of targeting CSCs for cancer treatment.
Collapse
|
23
|
D'Aguanno S, Barcaroli D, Rossi C, Zucchelli M, Ciavardelli D, Cortese C, De Cola A, Volpe S, D'Agostino D, Todaro M, Stassi G, Di Ilio C, Urbani A, De Laurenzi V. p63 isoforms regulate metabolism of cancer stem cells. J Proteome Res 2014; 13:2120-36. [PMID: 24597989 DOI: 10.1021/pr4012574] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
p63 is an important regulator of epithelial development expressed in different variants containing (TA) or lacking (ΔN) the N-terminal transactivation domain. The different isoforms regulate stem-cell renewal and differentiation as well as cell senescence. Several studies indicate that p63 isoforms also play a role in cancer development; however, very little is known about the role played by p63 in regulating the cancer stem phenotype. Here we investigate the cellular signals regulated by TAp63 and ΔNp63 in a model of epithelial cancer stem cells. To this end, we used colon cancer stem cells, overexpressing either TAp63 or ΔNp63 isoforms, to carry out a proteomic study by chemical-labeling approach coupled to network analysis. Our results indicate that p63 is implicated in a wide range of biological processes, including metabolism. This was further investigated by a targeted strategy at both protein and metabolite levels. The overall data show that TAp63 overexpressing cells are more glycolytic-active than ΔNp63 cells, indicating that the two isoforms may regulate the key steps of glycolysis in an opposite manner. The mass-spectrometry proteomics data of the study have been deposited to the ProteomeXchange Consortium ( http://proteomecentral.proteomexchange.org ) via the PRIDE partner repository with data set identifiers PXD000769 and PXD000768.
Collapse
Affiliation(s)
- Simona D'Aguanno
- Department of Experimental and Clinical Sciences, "G. d'Annunzio University" , Via dei Vestini 31, Chieti-Pescara 66100, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Choi W, Porten S, Kim S, Willis D, Plimack ER, Hoffman-Censits J, Roth B, Cheng T, Tran M, Lee IL, Melquist J, Bondaruk J, Majewski T, Zhang S, Pretzsch S, Baggerly K, Siefker-Radtke A, Czerniak B, Dinney CPN, McConkey DJ. Identification of distinct basal and luminal subtypes of muscle-invasive bladder cancer with different sensitivities to frontline chemotherapy. Cancer Cell 2014; 25:152-65. [PMID: 24525232 PMCID: PMC4011497 DOI: 10.1016/j.ccr.2014.01.009] [Citation(s) in RCA: 1233] [Impact Index Per Article: 123.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 10/17/2013] [Accepted: 01/13/2014] [Indexed: 12/11/2022]
Abstract
Muscle-invasive bladder cancers (MIBCs) are biologically heterogeneous and have widely variable clinical outcomes and responses to conventional chemotherapy. We discovered three molecular subtypes of MIBC that resembled established molecular subtypes of breast cancer. Basal MIBCs shared biomarkers with basal breast cancers and were characterized by p63 activation, squamous differentiation, and more aggressive disease at presentation. Luminal MIBCs contained features of active PPARγ and estrogen receptor transcription and were enriched with activating FGFR3 mutations and potential FGFR inhibitor sensitivity. p53-like MIBCs were consistently resistant to neoadjuvant methotrexate, vinblastine, doxorubicin and cisplatin chemotherapy, and all chemoresistant tumors adopted a p53-like phenotype after therapy. Our observations have important implications for prognostication, the future clinical development of targeted agents, and disease management with conventional chemotherapy.
Collapse
MESH Headings
- Aged
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor/genetics
- Blotting, Western
- Carcinoma, Basal Cell/drug therapy
- Carcinoma, Basal Cell/pathology
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/pathology
- Cell Differentiation
- Cell Proliferation
- Cisplatin/administration & dosage
- Clinical Trials, Phase II as Topic
- Cohort Studies
- Doxorubicin/administration & dosage
- Drug Resistance, Neoplasm/genetics
- Female
- Gene Expression Profiling
- Humans
- Male
- Methotrexate/administration & dosage
- MicroRNAs/genetics
- Muscle Neoplasms/classification
- Muscle Neoplasms/drug therapy
- Muscle Neoplasms/pathology
- Mutation/genetics
- Neoadjuvant Therapy
- Neoplasm Invasiveness
- Neoplasm Staging
- PPAR gamma/genetics
- PPAR gamma/metabolism
- Prognosis
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, Fibroblast Growth Factor, Type 3/metabolism
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Tumor Suppressor Protein p53/genetics
- Urinary Bladder Neoplasms/classification
- Urinary Bladder Neoplasms/drug therapy
- Urinary Bladder Neoplasms/pathology
- Vinblastine/administration & dosage
Collapse
Affiliation(s)
- Woonyoung Choi
- Department of Urology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sima Porten
- Department of Urology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Seungchan Kim
- Computational Biology Division, Translational Genomics Research Institute, 445N, Fifth Street, Phoenix, AZ 85004, USA
| | - Daniel Willis
- Department of Urology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elizabeth R Plimack
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111-2497, USA
| | - Jean Hoffman-Censits
- Department of Medical Oncology, Thomas Jefferson University Hospital, 1025 Walnut Street, Suite 700, Philadelphia, PA 19107, USA
| | - Beat Roth
- Department of Urology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Tiewei Cheng
- Department of Urology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas-Graduate School of Biomedical Sciences (GSBS) at Houston, Houston, TX 77030, USA
| | - Mai Tran
- Department of Urology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas-Graduate School of Biomedical Sciences (GSBS) at Houston, Houston, TX 77030, USA
| | - I-Ling Lee
- Department of Urology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jonathan Melquist
- Department of Urology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jolanta Bondaruk
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Tadeusz Majewski
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shizhen Zhang
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shanna Pretzsch
- Department of Urology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Keith Baggerly
- Department of Bioinformatics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Arlene Siefker-Radtke
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bogdan Czerniak
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Colin P N Dinney
- Department of Urology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - David J McConkey
- Department of Urology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas-Graduate School of Biomedical Sciences (GSBS) at Houston, Houston, TX 77030, USA.
| |
Collapse
|
25
|
|
26
|
Downregulation of keratin 76 expression during oral carcinogenesis of human, hamster and mouse. PLoS One 2013; 8:e70688. [PMID: 23936238 PMCID: PMC3728316 DOI: 10.1371/journal.pone.0070688] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 06/21/2013] [Indexed: 11/19/2022] Open
Abstract
Background Keratins are structural marker proteins with tissue specific expression; however, recent reports indicate their involvement in cancer progression. Previous study from our lab revealed deregulation of many genes related to structural molecular integrity including KRT76. Here we evaluate the role of KRT76 downregulation in oral precancer and cancer development. Methods We evaluated KRT76 expression by qRT-PCR in normal and tumor tissues of the oral cavity. We also analyzed K76 expression by immunohistochemistry in normal, oral precancerous lesion (OPL), oral squamous cell carcinoma (OSCC) and in hamster model of oral carcinogenesis. Further, functional implication of KRT76 loss was confirmed using KRT76-knockout (KO) mice. Results We observed a strong association of reduced K76 expression with increased risk of OPL and OSCC development. The buccal epithelium of DMBA treated hamsters showed a similar trend. Oral cavity of KRT76-KO mice showed preneoplastic changes in the gingivobuccal epithelium while no pathological changes were observed in KRT76 negative tissues such as tongue. Conclusion The present study demonstrates loss of KRT76 in oral carcinogenesis. The KRT76-KO mice data underlines the potential of KRT76 being an early event although this loss is not sufficient to drive the development of oral cancers. Thus, future studies to investigate the contributing role of KRT76 in light of other tumor driving events are warranted.
Collapse
|
27
|
Loljung L, Coates PJ, Nekulova M, Laurell G, Wahlgren M, Wilms T, Widlöf M, Hansel A, Nylander K. High expression of p63 is correlated to poor prognosis in squamous cell carcinoma of the tongue. J Oral Pathol Med 2013; 43:14-9. [PMID: 23607508 DOI: 10.1111/jop.12074] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2013] [Indexed: 01/09/2023]
Abstract
BACKGROUND p63 proteins are important in formation of the oral mucosa. Normal oral mucosa shows a balance between the six protein isoforms, whereas an imbalance between them is seen in squamous cell carcinomas (SCC). There is controversy over the clinical impact of p63 in SCC, which may relate to different expression in different areas. In addition, p63 isoforms can act as p53-like molecules (TAp63) or can inhibit p53 functions (ΔNp63) and expression of these isoforms varies in different tumours. Here, we chose to concentrate on the most common intra-oral sub-site, SCC of the mobile tongue. METHODS Total p63, ΔNp63 and TAp63 were analysed separately using immunohistochemistry. The percentage of cells and intensity of expression of different isoforms of p63 was evaluated using a quick score method and correlated with clinical data in a group of 87 patients with tongue SCC. RESULTS All tumours expressed p63 in at least 60% of the cells when using two different antibodies detecting all 6 isoforms. p63 expression correlated significantly with 2-year survival (P = 0.018), with fewer patients surviving 2 years if their tumours expressed p63 with strong intensity in at least 80% of the cells (quick score 18). Looking at 5-year survival, this was even more emphasized. ΔNp63 was expressed in all tumours, whereas expression of TAp63 was seen only in 59/87 patients, usually at very low levels. CONCLUSIONS Based on the present data, we recommend using expression of p63 as an additional factor contributing prognostic information in analysis of SCC in the tongue.
Collapse
Affiliation(s)
- Lotta Loljung
- Department of Medical Biosciences/Pathology, Umeå University, Umeå, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Privette Vinnedge LM, Kappes F, Nassar N, Wells SI. Stacking the DEK: from chromatin topology to cancer stem cells. Cell Cycle 2013; 12:51-66. [PMID: 23255114 PMCID: PMC3570517 DOI: 10.4161/cc.23121] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Stem cells are essential for development and tissue maintenance and display molecular markers and functions distinct from those of differentiated cell types in a given tissue. Malignant cells that exhibit stem cell-like activities have been detected in many types of cancers and have been implicated in cancer recurrence and drug resistance. Normal stem cells and cancer stem cells have striking commonalities, including shared cell surface markers and signal transduction pathways responsible for regulating quiescence vs. proliferation, self-renewal, pluripotency and differentiation. As the search continues for markers that distinguish between stem cells, progenitor cells and cancer stem cells, growing evidence suggests that a unique chromatin-associated protein called DEK may confer stem cell-like qualities. Here, we briefly describe current knowledge regarding stem and progenitor cells. We then focus on new findings that implicate DEK as a regulator of stem and progenitor cell qualities, potentially through its unusual functions in the regulation of local or global chromatin organization.
Collapse
Affiliation(s)
- Lisa M Privette Vinnedge
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | | | | | | |
Collapse
|
29
|
Yan W, Chen X, Zhang Y, Zhang J, Jung YS, Chen X. Arsenic suppresses cell survival via Pirh2-mediated proteasomal degradation of ΔNp63 protein. J Biol Chem 2012; 288:2907-13. [PMID: 23271742 DOI: 10.1074/jbc.m112.428607] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Transcription factor p63, a member of the p53 family, shares a high degree of sequence similarity with p53. Because of transcription from two distinct promoters, the p63 gene encodes two isoforms, TAp63 and ΔNp63. Although TAp63 acts as a tumor suppressor, ΔNp63 functions as an oncogene and is often overexpressed in squamous cell carcinomas. Thus, therapeutic agents targeting ΔNp63 might be used to manage tumors that overexpress ΔNp63. Here we found that arsenic trioxide, a frontline agent for acute promyelocytic leukemia, inhibits ΔNp63 but not TAp63 expression in time- and dose-dependent manners. In addition, we found that arsenic trioxide decreases the stability of ΔNp63 protein via a proteasome-dependent pathway but has little effect on the level of ΔNp63 transcript. Furthermore, we found that arsenic trioxide activates the Pirh2 promoter and consequently induces Pirh2 expression. Consistent with this, we found that knockdown of Pirh2 inhibits, whereas ectopic expression of Pirh2 enhances, arsenic-induced degradation of ΔNp63 protein. Importantly, we found that knockdown of ΔNp63 sensitizes, whereas ectopic expression of ΔNp63 inhibits, growth suppression induced by arsenic. Together, these data suggest that arsenic degrades ΔNp63 protein at least in part via Pirh2-dependent proteolysis and that inhibition of ΔNp63 expression facilitates tumor cells to arsenic-induced death.
Collapse
Affiliation(s)
- Wensheng Yan
- Comparative Oncology Laboratory, University of California at Davis, Davis, California 95616, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
The p53 homolog p73 is frequently overexpressed in cancers. Especially the transactivation domain truncated isoform ΔNp73 has oncogenic properties and its upregulation is associated with poor patient survival. It has been shown that ΔNp73 has an inhibitory effect on the transactivation capacity of p53 and other p73 isoforms. Here, we confirm this finding but surprisingly find that ΔNp73 may also stimulate the expression of TGF-β signaling targets. Promoter-reporter analysis indicated that the presence of Smad Binding Elements (SBE) in the promoter is sufficient for stimulation of gene expression by ΔNp73. TGF-β signaling was less efficient in ΔNp73 downregulated cells, whereas tetracycline induced ΔNp73 increased expression of endogenous TGF-β regulated genes PAI-1 and Col1a1. Pull-down assays with SBE DNA suggest that ΔNp73 enhances smad3/4 binding to SBEs, thereby stimulating TGF-β signaling. Chromatin immunoprecipitation assays confirmed a direct interaction between ΔNp73 and SBE. Given the role of TGF-β signaling in carcinogenesis, tumor invasion and metastasis via targets like PAI-1 and Col1a1, our data suggest a model on how this effect of ΔNp73 could be a contributing factor in cancer progression.
Collapse
|
31
|
Wray H, Mackenzie IC, Storey A, Navsaria H. α6 Integrin and CD44 enrich for a primary keratinocyte population that displays resistance to UV-induced apoptosis. PLoS One 2012; 7:e46968. [PMID: 23071680 PMCID: PMC3468583 DOI: 10.1371/journal.pone.0046968] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 09/10/2012] [Indexed: 12/01/2022] Open
Abstract
Epidermal human keratinocytes are exposed to a wide range of environmental genotoxic insults, including the UV component of solar radiation. Epidermal homeostasis in response to cellular or tissue damage is maintained by a population of keratinocyte stem cells (KSC) that reside in the basal layer of the epithelium. Using cell sorting based on cell-surface markers, we have identified a novel α6 integrinhigh+/CD44+ sub-population of basal keratinocytes. These α6 integrinhigh+/CD44+ keratinocytes have both high proliferative potential, form colonies in culture that have characteristics of holoclones and have a unique pattern of resistance to apoptosis induced by UVB radiation or by agents that induce single- or double strand DNA breaks. Resistance to UVB induced apoptosis in the α6 integrinhigh+/CD44+ cells involved increased expression of TAp63 and was overcome by PI-3 kinase inhibition. In marked contrast, the α6 integrinhigh+/CD44+ cells were sensitive to apoptosis induced by the cross-linking agent cisplatin, and imatinib inhibition of c-Abl blocked the ability of cisplatin to kill α6 integrinhigh+/CD44+ cells. Our findings reveal a population of basal keratinocytes with long-term proliferative properties that display specific patterns of apoptotic resistance that is dependent upon the genotoxic stimulus, and provide insights into how these cells can be targeted with chemotherapeutic agents.
Collapse
Affiliation(s)
- Helen Wray
- Blizard Institute of Cell and Molecular Science, Queen Mary’s School of Medicine and Dentistry, Whitechapel, London, United Kingdom
| | - Ian C. Mackenzie
- Blizard Institute of Cell and Molecular Science, Queen Mary’s School of Medicine and Dentistry, Whitechapel, London, United Kingdom
| | - Alan Storey
- Department of Molecular Oncology, Weatherall Institute of Molecular Medicine (WIMM), University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Harshad Navsaria
- Blizard Institute of Cell and Molecular Science, Queen Mary’s School of Medicine and Dentistry, Whitechapel, London, United Kingdom
- * E-mail:
| |
Collapse
|
32
|
Ferronika P, Triningsih FE, Ghozali A, Moeljono A, Rahmayanti S, Shadrina AN, Naim AE, Wudexi I, Arnurisa AM, Nanwani ST, Harijadi A. p63 Cytoplasmic Aberrance is Associated with High Prostate Cancer Stem Cell Expression. Asian Pac J Cancer Prev 2012; 13:1943-8. [DOI: 10.7314/apjcp.2012.13.5.1943] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
33
|
Reduction of NOTCH1 expression pertains to maturation abnormalities of keratinocytes in squamous neoplasms. J Transl Med 2012; 92:688-702. [PMID: 22330335 DOI: 10.1038/labinvest.2012.9] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Notch is a transmembrane receptor functioning in the determination of cell fate. Abnormal Notch signaling promotes tumor development, showing either oncogenic or tumor suppressive activity. The uncertainty about the exact role of Notch signaling, partially, stems from inconsistencies in descriptions of Notch expression in human cancers. Here, we clarified basal-cell dominant expression of NOTCH1 in squamous epithelium. NOTCH1 was downregulated in squamous neoplasms of oral mucosa, esophagus and uterine cervix, compared with the normal basal cells, although the expression tended to be retained in cervical lesions. NOTCH1 downregulation was observed even in precancers, and there was little difference between cancers and high-grade precancerous lesions, suggesting its minor contribution to cancer-specific events such as invasion. In culture experiments, reduction of NOTCH1 expression resulted in downregulation of keratin 13 and keratin 15, and upregulation of keratin 17, and NOTCH1 knockdown cells formed a dysplastic stratified epithelium mimicking a precancerous lesion. The NOTCH1 downregulation and the concomitant alterations of those keratin expressions were confirmed in the squamous neoplasms both by immunohistochemical and cDNA microarray analyses. Our data indicate that reduction of NOTCH1 expression directs the basal cells to cease terminal differentiation and to form an immature epithelium, thereby playing a major role in the histopathogenesis of epithelial dysplasia. Furthermore, downregulation of NOTCH1 expression seems to be an inherent mechanism for switching the epithelium from a normal and mature state to an activated and immature state, suggesting its essential role in maintaining the epithelial integrity.
Collapse
|
34
|
Zhang Y, Gu C, Shi H, Zhang A, Kong X, Bao W, Deng D, Ren L, Gu D. Association between C3orf21, TP63 polymorphisms and environment and NSCLC in never-smoking Chinese population. Gene 2012; 497:93-7. [DOI: 10.1016/j.gene.2012.01.053] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Revised: 12/21/2011] [Accepted: 01/21/2012] [Indexed: 01/01/2023]
|
35
|
Wiklund ED, Gao S, Hulf T, Sibbritt T, Nair S, Costea DE, Villadsen SB, Bakholdt V, Bramsen JB, Sørensen JA, Krogdahl A, Clark SJ, Kjems J. MicroRNA alterations and associated aberrant DNA methylation patterns across multiple sample types in oral squamous cell carcinoma. PLoS One 2011; 6:e27840. [PMID: 22132151 PMCID: PMC3222641 DOI: 10.1371/journal.pone.0027840] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 10/26/2011] [Indexed: 01/08/2023] Open
Abstract
Background MicroRNA (miRNA) expression is broadly altered in cancer, but few studies have investigated miRNA deregulation in oral squamous cell carcinoma (OSCC). Epigenetic mechanisms are involved in the regulation of >30 miRNA genes in a range of tissues, and we aimed to investigate this further in OSCC. Methods TaqMan® qRT-PCR arrays and individual assays were used to profile miRNA expression in a panel of 25 tumors with matched adjacent tissues from patients with OSCC, and 8 control paired oral stroma and epithelium from healthy volunteers. Associated DNA methylation changes of candidate epigenetically deregulated miRNA genes were measured in the same samples using the MassArray® mass spectrometry platform. MiRNA expression and DNA methylation changes were also investigated in FACS sorted CD44high oral cancer stem cells from primary tumor samples (CSCs), and in oral rinse and saliva from 15 OSCC patients and 7 healthy volunteers. Results MiRNA expression patterns were consistent in healthy oral epithelium and stroma, but broadly altered in both tumor and adjacent tissue from OSCC patients. MiR-375 is repressed and miR-127 activated in OSCC, and we confirm previous reports of miR-137 hypermethylation in oral cancer. The miR-200 s/miR-205 were epigenetically activated in tumors vs normal tissues, but repressed in the absence of DNA hypermethylation specifically in CD44high oral CSCs. Aberrant miR-375 and miR-200a expression and miR-200c-141 methylation could be detected in and distinguish OSCC patient oral rinse and saliva from healthy volunteers, suggesting a potential clinical application for OSCC specific miRNA signatures in oral fluids. Conclusions MiRNA expression and DNA methylation changes are a common event in OSCC, and we suggest miR-375, miR-127, miR-137, the miR-200 family and miR-205 as promising candidates for future investigations. Although overall activated in OSCC, miR-200/miR-205 suppression in oral CSCs indicate that cell specific silencing of these miRNAs may drive tumor expansion and progression.
Collapse
Affiliation(s)
- Erik D Wiklund
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Role of p63 in cancer development. Biochim Biophys Acta Rev Cancer 2011; 1816:57-66. [PMID: 21515338 DOI: 10.1016/j.bbcan.2011.04.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Revised: 04/05/2011] [Accepted: 04/08/2011] [Indexed: 12/22/2022]
Abstract
Since their initial identification p53 homologues p63 and p73 have been expected to play a role in cancer development due to their close homology to p53, notoriously one of the most mutated genes in cancer. However soon after their discovery the awareness that these genes were rarely mutated in cancer seemed to indicate that they did not play a role in its development. However a large number of data collected in the following years indicated that altered expression rather than mutation could be found in different neoplasia and play a role in its biology. In particular p63 due to its fundamental role in epithelial development seems to play a role in a number of tumors of epithelial origin. In this review we summarize some of the evidence linking p63 to carcinogenesis.
Collapse
|
37
|
The role of p63 in cancer, stem cells and cancer stem cells. Cell Mol Biol Lett 2011; 16:296-327. [PMID: 21442444 PMCID: PMC6275999 DOI: 10.2478/s11658-011-0009-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Accepted: 03/07/2011] [Indexed: 01/01/2023] Open
Abstract
The transcription factor p63 has important functions in tumorigenesis, epidermal differentiation and stem cell self-renewal. The TP63 gene encodes multiple protein isoforms that have different or even antagonistic roles in these processes. The balance of p63 isoforms, together with the presence or absence of the other p53 family members, p73 and p53, has a striking biological impact. There is increasing evidence that interactions between p53-family members, whether cooperative or antagonistic, are involved in various cell processes. This review summarizes the current understanding of the role of p63 in tumorigenesis, metastasis, cell migration and senescence. In particular, recent data indicate important roles in adult stem cell and cancer stem cell regulation and in the response of cancer cells to therapy.
Collapse
|
38
|
Liu X, Kolokythas A, Wang J, Huang H, Zhou X. Gene Expression Signatures of Lymph Node Metastasis in Oral Cancer: Molecular Characteristics and Clinical Significances. CURRENT CANCER THERAPY REVIEWS 2010; 6:294-307. [PMID: 21709736 PMCID: PMC3122885 DOI: 10.2174/157339410793358066] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Even though lymph node metastasis accounts for the vast majority of cancer death in patients with oral cancer (OC), the molecular mechanisms of lymph node metastasis remain elusive. Genome-wide microarray analyses and functional studies in vitro and in vivo, along with detailed clinical observations, have identified a number of molecules that may contribute to lymph node metastasis. These include lymphangionenic cytokines, cell adhesion molecules, basement membrane-interacting molecules, matrix enzymes and relevant downstream signaling pathways. However, defined gene signatures from different studies are highly variable, which hinders their translation to clinically relevant applications. To date, none of the identified signatures or molecular biomarkers has been successfully implemented as a diagnostic or prognostic tool applicable to routine clinical practice. In this review, we will first introduce the significance of lymph node metastasis in OC, and clinical/experimental evidences that support the underlying molecular mechanisms. We will then provide a comprehensive review and integrative analysis of the existing gene expression studies that aim to identify the metastasis-related signatures in OC. Finally, the remaining challenges will be discussed and our insights on future directions will be provided.
Collapse
Affiliation(s)
- Xiqiang Liu
- Center for Molecular Biology of Oral Diseases, College of Dentistry, University of Illinois at Chicago, Chicago, IL
- Research Institute & the Affiliated Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Antonia Kolokythas
- Department of Oral and Maxillofacial Surgery, College of Dentistry, University of Illinois at Chicago, Chicago, IL
| | - Jianguang Wang
- Department of Oral and Maxillofacial Surgery, the Second Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hongzhang Huang
- Research Institute & the Affiliated Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Xiaofeng Zhou
- Center for Molecular Biology of Oral Diseases, College of Dentistry, University of Illinois at Chicago, Chicago, IL
- Research Institute & the Affiliated Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Graduate College, and UIC Cancer Center, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
39
|
Abstract
Small populations within an increasing array of solid tumors, labeled cancer stem cells (CSC) or tumor-initiating cells (TIC), have the ability to differentiate, self-renew, and replicate the original tumor in vivo. To date, these cells have been distinguished from the bulk-tumor population by the expression pattern of cell-surface proteins (e.g., CD24, CD44, CD133) and cellular activities, such as the efflux of Hoechst dye or aldehyde dehydrogenase activity. Recent data have shown that these markers are inducible by exposure to anticancer agents; this finding highlights not only the potential fluidity of the CSC compartment, but also the functionality of these markers. The involvement of CD44 in invasion, adhesion, and metastasis, or the role of CD24 in modulation of src, FAK, and GLI1 are examples of these relevant roles. Instead of looking solely at the marker expression in these populations, we hope to clarify the biologically significant roles these markers and activities play in tumor progression, metastases, and as possible targets for therapy.
Collapse
Affiliation(s)
- Stephen B Keysar
- University of Colorado Cancer Center, Aurora, Colorado 80045, USA
| | | |
Collapse
|
40
|
Herfs M, Hubert P, Suarez-Carmona M, Reschner A, Saussez S, Berx G, Savagner P, Boniver J, Delvenne P. Regulation of p63 isoforms by snail and slug transcription factors in human squamous cell carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:1941-9. [PMID: 20150431 DOI: 10.2353/ajpath.2010.090804] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
TP63 is a p53-related gene that contains two alternative promoters, which give rise to transcripts that encode proteins with (TAp63) or without (DeltaNp63) an amino-transactivating domain. Whereas the expression of p63 is required for proper development of epithelial structures, the role of p63 in tumorigenesis remains unclear. Here, we investigated the role of Snail and Slug transcription factors, known to promote epithelial-to-mesenchymal transitions during development and cancer, in the regulation of p63 isoforms in human squamous cell carcinoma (SCC). In the present study, we observed that the expressions of DeltaN and TAp63 isoforms were, respectively, down- and up-regulated by both Snail and Slug. However, the induction of TAp63 was not directly caused by these two transcription factors but resulted from the loss of DeltaNp63, which acts as dominant-negative inhibitor of TAp63. In SCC cell lines and cancer tissues, high expression of Snail and Slug was also significantly associated with altered p63 expression. Finally, we showed that DeltaNp63 silencing reduced cell-cell adhesion and increased the migratory properties of cancer cells. These data suggest that the disruption of p63 expression induced by Snail and Slug plays a crucial role in tumor progression. Therefore, p63 and its regulating factors could constitute novel prognosis markers in patients with SCC and attractive targets for the therapeutic modulation of neoplastic cell invasiveness.
Collapse
Affiliation(s)
- Michael Herfs
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, 4000 Liege, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Amoresano A, Di Costanzo A, Leo G, Di Cunto F, La Mantia G, Guerrini L, Calabrò V. Identification of ΔNp63α Protein Interactions by Mass Spectrometry. J Proteome Res 2010; 9:2042-8. [DOI: 10.1021/pr9011156] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Angela Amoresano
- Dipartimento di Chimica Organica e Biologica, Università Federico II, Napoli, Italy, Dipartimento di Biologia Strutturale e Funzionale, Università di Napoli, Federico II, Italy, Centro di Biotecnologie Molecolari, Università di Torino, Torino, Italy, and Dipartimento di Scienze Biomolecolari e Biotecnologie, Università di Milano, Italy
| | - Antonella Di Costanzo
- Dipartimento di Chimica Organica e Biologica, Università Federico II, Napoli, Italy, Dipartimento di Biologia Strutturale e Funzionale, Università di Napoli, Federico II, Italy, Centro di Biotecnologie Molecolari, Università di Torino, Torino, Italy, and Dipartimento di Scienze Biomolecolari e Biotecnologie, Università di Milano, Italy
| | - Gabriella Leo
- Dipartimento di Chimica Organica e Biologica, Università Federico II, Napoli, Italy, Dipartimento di Biologia Strutturale e Funzionale, Università di Napoli, Federico II, Italy, Centro di Biotecnologie Molecolari, Università di Torino, Torino, Italy, and Dipartimento di Scienze Biomolecolari e Biotecnologie, Università di Milano, Italy
| | - Ferdinando Di Cunto
- Dipartimento di Chimica Organica e Biologica, Università Federico II, Napoli, Italy, Dipartimento di Biologia Strutturale e Funzionale, Università di Napoli, Federico II, Italy, Centro di Biotecnologie Molecolari, Università di Torino, Torino, Italy, and Dipartimento di Scienze Biomolecolari e Biotecnologie, Università di Milano, Italy
| | - Girolama La Mantia
- Dipartimento di Chimica Organica e Biologica, Università Federico II, Napoli, Italy, Dipartimento di Biologia Strutturale e Funzionale, Università di Napoli, Federico II, Italy, Centro di Biotecnologie Molecolari, Università di Torino, Torino, Italy, and Dipartimento di Scienze Biomolecolari e Biotecnologie, Università di Milano, Italy
| | - Luisa Guerrini
- Dipartimento di Chimica Organica e Biologica, Università Federico II, Napoli, Italy, Dipartimento di Biologia Strutturale e Funzionale, Università di Napoli, Federico II, Italy, Centro di Biotecnologie Molecolari, Università di Torino, Torino, Italy, and Dipartimento di Scienze Biomolecolari e Biotecnologie, Università di Milano, Italy
| | - Viola Calabrò
- Dipartimento di Chimica Organica e Biologica, Università Federico II, Napoli, Italy, Dipartimento di Biologia Strutturale e Funzionale, Università di Napoli, Federico II, Italy, Centro di Biotecnologie Molecolari, Università di Torino, Torino, Italy, and Dipartimento di Scienze Biomolecolari e Biotecnologie, Università di Milano, Italy
| |
Collapse
|
42
|
Shimada Y, Ishii G, Nagai K, Atsumi N, Fujii S, Yamada A, Yamane Y, Hishida T, Nishimura M, Yoshida J, Ikeda N, Ochiai A. Expression of podoplanin, CD44, and p63 in squamous cell carcinoma of the lung. Cancer Sci 2009; 100:2054-9. [PMID: 19681903 PMCID: PMC11158176 DOI: 10.1111/j.1349-7006.2009.01295.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2009] [Revised: 07/02/2009] [Accepted: 07/07/2009] [Indexed: 11/28/2022] Open
Abstract
Recent molecular biological studies have identified podoplanin as a candidate cancer stem cell (CSC) marker in squamous cell carcinoma (SqCC). The purpose of this study was to examine the expression pattern of podoplanin, and the other stem cell markers CD44 and p63, and their relationship to clinico-pathological features including survival in pulmonary SqCC. We examined histologically the expression of podoplanin, CD44, and p63 in 162 consecutive SqCC by immunostaining. Podoplanin expression was observed in 107 (66%) tumors, CD44 in 145 (89.5%), and p63 in 151 (93.2%), respectively. In 95.3% of the podoplanin-positive tumors, tumor cells showing strong expression were localized in the periphery of the tumor nests. However, this peripheral localization was observed in only 55.9% of the CD44-positive and 43% of p63-positive tumors. In 88.8% of the podoplanin-positive tumors, positive cells were localized more peripherally in the tumor nests than CD44- or p63-positive cells and when CD44 and p63 expressions were compared in these podoplanin-positive tumors, p63-positive layers in the periphery of the tumor nests were broader compared to CD44-positive layers. These findings suggest tumor cells are aligned in the "hierarchical distribution pattern" according to the expression of these three markers. Patients who had podoplanin-positive tumors with the "hierarchical pattern" resulted in significantly better overall survival than those who had podoplanin-negative tumors (P = 0.043). These results suggest that podoplanin expression would reflect the most immature status in the differentiation process of SqCC, and SqCC with hierarchical expression would be a well-organized tumor group with lower biological aggressiveness based on the CSC concept.
Collapse
Affiliation(s)
- Yoshihisa Shimada
- Pathology Division, Research Center for Innovative Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Saintigny P, El-Naggar AK, Papadimitrakopoulou V, Ren H, Fan YH, Feng L, Lee JJ, Kim ES, Hong WK, Lippman SM, Mao L. DeltaNp63 overexpression, alone and in combination with other biomarkers, predicts the development of oral cancer in patients with leukoplakia. Clin Cancer Res 2009; 15:6284-91. [PMID: 19773378 DOI: 10.1158/1078-0432.ccr-09-0498] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The risk of malignant transformation of oral preneoplastic lesion (OPL) is difficult to assess. DeltaNp63 is an early oncoprotein associated with mucosal tumorigenesis. The purpose of this study was to assess DeltaNp63 expression in OPL and its role as a marker of oral cancer risk. EXPERIMENTAL DESIGN DeltaNp63 expression was determined using immunohistochemistry in 152 OPL patients included in a clinical trial comparing retinyl palmitate alone or plus beta-carotene with low-dose 13-cis-retinoic acid. The associations between DeltaNp63 expression as well as DeltaNp63 expression with other potential risk factors for oral cancer development were analyzed. RESULTS DeltaNp63 expression was positive in 41 (27%) patients, clusters of intraepithelial inflammatory cells (EIC) were noted in 37 (26%) patients, and podoplanin (previously reported) was positive in 56 (37%) patients. Significantly more patients whose lesions were DeltaNp63 positive or exhibited EIC developed oral cancers. In the multicovariate analysis including age, treatment, and histologic status as cofactors, positive DeltaNp63 expression was associated with an increased hazard ratio of 3.308 (95% confidence interval, 1.663-6.580; P = 0.0007). Patients whose lesions showed positive DeltaNp63, podoplanin, and EIC had the highest oral cancer risk with a hazard ratio of 4.372 (95% confidence interval, 1.912-9.992; P = 0.0005) and 61% oral cancer development rate at 5 years compared with 15% of other OPL patients (P < 0.0001). CONCLUSION DeltaNp63 overepression in OPL is associated with increased oral cancer risk. Together, DeltaNp63, podoplanin, and EIC may be used as biomarkers to identify OPL patients with substantially high oral cancer risk.
Collapse
Affiliation(s)
- Pierre Saintigny
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas M D Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Mangiulli M, Valletti A, Caratozzolo MF, Tullo A, Sbisà E, Pesole G, D'Erchia AM. Identification and functional characterization of two new transcriptional variants of the human p63 gene. Nucleic Acids Res 2009; 37:6092-104. [PMID: 19700772 PMCID: PMC2764424 DOI: 10.1093/nar/gkp674] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
p63 belongs to a family of transcription factors, which, while demonstrating striking conservation of functional domains, regulate distinct biological functions. Its principal role is in the regulation of epithelial commitment, differentiation and maintenance programs, during embryogenesis and in adult tissues. The p63 gene has a complex transcriptional pattern, producing two subclasses of N-terminal isoforms (TA and DeltaN) which are alternatively spliced at the C-terminus. Here, we report the identification of two new C-terminus p63 variants, we named p63 delta and epsilon, that increase from 6 to 10 the number of the p63 isoforms. Expression analysis of all p63 variants demonstrates a tissue/cell-type-specific nature of p63 alternative transcript expression, probably related to their different cellular functions. We demonstrate that the new p63 variants as DeltaN isoforms are active as transcription factors as they have nuclear localization and can modulate the expression of p63 target genes. Moreover, we report that, like DeltaNp63alpha, DeltaNp63delta and epsilon sustain cellular proliferation and that their expression decreases during keratinocyte differentiation, suggesting their involvement in this process. Taken together, our results demonstrate the existence of novel p63 proteins whose expression should be considered in future studies on the roles of p63 in the regulation of cellular functions.
Collapse
Affiliation(s)
- Marina Mangiulli
- Dipartimento di Biochimica e Biologia Molecolare E. Quagliariello, Università degli Studi di Bari, via Orabona 4, 70126 Bari, Italy
| | | | | | | | | | | | | |
Collapse
|
45
|
Ratushny V, Astsaturov I, Burtness BA, Golemis EA, Silverman JS. Targeting EGFR resistance networks in head and neck cancer. Cell Signal 2009; 21:1255-68. [PMID: 19258037 PMCID: PMC2770888 DOI: 10.1016/j.cellsig.2009.02.021] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2009] [Accepted: 02/17/2009] [Indexed: 01/01/2023]
Abstract
A core set of oncoproteins is overexpressed or functionally activated in many types of cancer, and members of this group have attracted significant interest as subjects for development of targeted therapeutics. For some oncoproteins such as EGFR/ErbB1, both small molecule and antibody agents have been developed and applied in the clinic for over a decade. Analysis of clinical outcomes has revealed an initially unexpected complexity in the response of patients to these agents. Diverse factors, including developmental lineage of the tumor progenitor cell, co-mutation or epigenetic modulation of genes encoding proteins in an extended EGFR signaling network or regulating core survival responses in individual tumors, and environmental factors including inflammatory agents and viral infection, all have been identified as modulating response to treatment with EGFR-targeted drugs. Second and third generation therapeutic strategies increasingly incorporate knowledge of cancer type-specific signaling environments, in a more personalized treatment approach. This review takes squamous cell carcinoma of the head and neck (SCCHN) as a specific example of an EGFR-involved cancer with idiosyncratic biological features that influence design of treatment modalities, with particular emphasis on commonalities and differences with other cancer types.
Collapse
Affiliation(s)
- Vladimir Ratushny
- Programs in Head and Neck Cancer and Molecular Medicine, Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA, 19111, USA
- Program in Molecular and Cell Biology and Genetics, Drexel University College of Medicine, 2900 W. Queen Lane, Philadelphia, PA 19129
| | - Igor Astsaturov
- Programs in Head and Neck Cancer and Molecular Medicine, Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA, 19111, USA
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA, 19111, USA
| | - Barbara A. Burtness
- Programs in Head and Neck Cancer and Molecular Medicine, Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA, 19111, USA
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA, 19111, USA
| | - Erica A. Golemis
- Programs in Head and Neck Cancer and Molecular Medicine, Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA, 19111, USA
| | - Joshua S. Silverman
- Programs in Head and Neck Cancer and Molecular Medicine, Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA, 19111, USA
- Department of Radiation Oncology, Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA, 19111, USA
| |
Collapse
|
46
|
Boldrup L, Coates PJ, Gu X, Nylander K. ΔNp63 isoforms differentially regulate gene expression in squamous cell carcinoma: identification ofCox-2as a novel p63 target. J Pathol 2009; 218:428-36. [DOI: 10.1002/path.2560] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
47
|
An active role of the DeltaN isoform of p63 in regulating basal keratin genes K5 and K14 and directing epidermal cell fate. PLoS One 2009; 4:e5623. [PMID: 19461998 PMCID: PMC2680039 DOI: 10.1371/journal.pone.0005623] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Accepted: 04/22/2009] [Indexed: 11/19/2022] Open
Abstract
Background One major defining characteristic of the basal keratinocytes of the stratified epithelium is the expression of the keratin genes K5 and K14. The temporal and spatial expression of these two genes is usually tightly and coordinately regulated at the transcriptional level. This ensures the obligate pairing of K5 and K14 proteins to generate an intermediate filament (IF) network that is essential for the structure and function of the proliferative keratinocytes. Our previous studies have shown that the basal-keratinocyte restricted transcription factor p63 is a direct regulator of K14 gene. Methodology/Principal Findings Here we provide evidence that p63, specifically the ΔN isoform also regulates the expression of the K5 gene by binding to a conserved enhancer within the 5′ upstream region. By using specific antibodies against ΔNp63, we show a concordance in the expression between basal keratins and ΔNp63 proteins but not the TAp63 isoforms during early embryonic skin development. We demonstrate, that contrary to a previous report, transgenic mice expressing ΔNp63 in lung epithelium exhibit squamous metaplasia with de novo induction of K5 and K14 as well as transdifferentiation to the epidermal cell lineage. Interestingly, the in vivo epidermal inductive properties of ΔNp63 do not require the C-terminal SAM domain. Finally, we show that ΔNp63 alone can restore the expression of the basal keratins and reinitiate the failed epidermal differentiation program in the skin of p63 null animals. Significance ΔNp63 is a critical mediator of keratinocyte stratification program and directly regulates the basal keratin genes.
Collapse
|
48
|
The alpha/beta carboxy-terminal domains of p63 are required for skin and limb development. New insights from the Brdm2 mouse which is not a complete p63 knockout but expresses p63 gamma-like proteins. Cell Death Differ 2009; 16:1108-17. [PMID: 19300453 PMCID: PMC2778344 DOI: 10.1038/cdd.2009.25] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
p63, an ancestral transcription factor of the p53 family, has three C-terminal isoforms whose relative in vivo functions are elusive. The p63 gene is essential for skin and limb development, as vividly shown by two independent global knockout mouse models. Both strains, although constructed differently, have identical and severe phenotypes, characterized by absent epidermis and hindlimbs and only rudimentary forelimbs at birth. Here we show that mice from one model, Brdm2, express normal levels of truncated p63 proteins that contain the DNA binding and oligomerization domain but lack the long carboxy-terminal SAM (sterile alpha-motif) and post-SAM domains that are specific for the alpha and beta isoforms. As such, transcriptionally active p63 proteins from Brdm2 mice resemble the naturally occurring p63gamma isoforms, which of all the p63 isoforms most closely resemble p53. Thus, Brdm2 mice are p63alpha/beta isoform-specific knockout mice, gaining unexpected new importance. Our studies identify that p63alpha/beta but not p63gamma are absolutely required for proper skin and limb development.
Collapse
|
49
|
Godar S, Ince TA, Bell GW, Feldser D, Donaher JL, Bergh J, Liu A, Miu K, Watnick RS, Reinhardt F, McAllister SS, Jacks T, Weinberg RA. Growth-inhibitory and tumor- suppressive functions of p53 depend on its repression of CD44 expression. Cell 2008; 134:62-73. [PMID: 18614011 DOI: 10.1016/j.cell.2008.06.006] [Citation(s) in RCA: 337] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2007] [Revised: 03/17/2008] [Accepted: 06/04/2008] [Indexed: 01/15/2023]
Abstract
The p53 tumor suppressor is a key mediator of cellular responses to various stresses. Here, we show that under conditions of basal physiologic and cell-culture stress, p53 inhibits expression of the CD44 cell-surface molecule via binding to a noncanonical p53-binding sequence in the CD44 promoter. This interaction enables an untransformed cell to respond to stress-induced, p53-dependent cytostatic and apoptotic signals that would otherwise be blocked by the actions of CD44. In the absence of p53 function, the resulting derepressed CD44 expression is essential for the growth and tumor-initiating ability of highly tumorigenic mammary epithelial cells. In both tumorigenic and nontumorigenic cells, CD44's expression is positively regulated by p63, a paralogue of p53. Our data indicate that CD44 is a key tumor-promoting agent in transformed tumor cells lacking p53 function. They also suggest that the derepression of CD44 resulting from inactivation of p53 can potentially aid the survival of immortalized, premalignant cells.
Collapse
Affiliation(s)
- Samuel Godar
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
|