The Roles of FOXM1 in Pancreatic Stem Cells and Carcinogenesis Ming Quan1,3, Peipei Wang2, Jiujie Cui1,3, Yong Gao2 and Keping Xie3*

Total Page:16

File Type:pdf, Size:1020Kb

The Roles of FOXM1 in Pancreatic Stem Cells and Carcinogenesis Ming Quan1,3, Peipei Wang2, Jiujie Cui1,3, Yong Gao2 and Keping Xie3* Quan et al. Molecular Cancer 2013, 12:159 http://www.molecular-cancer.com/content/12/1/159 REVIEW Open Access The roles of FOXM1 in pancreatic stem cells and carcinogenesis Ming Quan1,3, Peipei Wang2, Jiujie Cui1,3, Yong Gao2 and Keping Xie3* Abstract Pancreatic ductal adenocarcinoma (PDAC) has one of the poorest prognoses among all cancers. Over the past several decades, investigators have made great advances in the research of PDAC pathogenesis. Importantly, identification of pancreatic cancer stem cells (PCSCs) in pancreatic cancer cases has increased our understanding of PDAC biology and therapy. PCSCs are responsible for pancreatic tumorigenesis and tumor progression via a number of mechanisms, including extensive proliferation, self-renewal, high tumorigenic ability, high propensity for invasiveness and metastasis, and resistance to conventional treatment. Furthermore, emerging evidence suggests that PCSCs are involved in the malignant transformation of pancreatic intraepithelial neoplasia. The molecular mechanisms that control PCSCs are related to alterations of various signaling pathways, for instance, Hedgehog, Notch, Wnt, B-cell-specific Moloney murine leukemia virus insertion site 1, phosphoinositide 3-kinase/AKT, and Nodal/Activin. Also, authors have reported that the proliferation-specific transcriptional factor Forkhead box protein M1 is involved in PCSC self-renewal and proliferation. In this review, we describe the current knowledge about the signaling pathways related to PCSCs and the early stages of PDAC development, highlighting the pivotal roles of Forkhead box protein M1 in PCSCs and their impacts on the development and progression of pancreatic intraepithelial neoplasia. Keywords: Transcription factors, Oncogenic switch, Progression, Stem cells, Therapeutic targets, Molecular biomarkers Introduction roles of pancreatic CSCs (PCSCs) in pancreatic cancer de- The incidence of pancreatic cancer is increasing annu- velopment and progression is imperative. ally, especially in industrialized countries [1]. Despite CSC research has resulted in many advances in the ever-increasing research efforts over the past few de- fundamental understanding and clinical management of cades, prognoses for pancreatic cancer remain among several solid tumors, including brain, breast, head and the poorest for all cancers. It is also one of the leading neck, lung, prostate, colon, ovarian, and pancreatic can- causes of cancer-related mortality in developed coun- cer [5-11]. CSCs are now widely accepted to be a sub- tries, with a median survival duration of 6 months and population of tumor cells with the capacity for extensive 5-year overall survival rate of less than 5% [2,3]. Conven- proliferation, self-renewal, multipotency, high tumori- tional therapies, such as surgery, radiation therapy, chemo- genicity, and treatment resistance. Moreover, CSCs have therapy, and combinations of them, have had a limited a high propensity for invasiveness and metastasis [12]. impact on the course of this aggressive neoplasm, which is CSCs in pancreatic cancer cases are characterized by ex- characterized by rapid metastasis and resistance to these pression of the cell surface markers CD44, CD24, and therapies [4]. Researchers recently demonstrated that the epithelial-specific antigen (ESA; epithelial cell adhesion presence of cancer stem cells (CSCs) in pancreatic tumors molecule [EpCAM]) [13]. Authors reported that CD133+ contributes to the early metastasis and chemotherapeutic cells in primary pancreatic tumors and pancreatic cancer drug resistance of pancreatic cancer [5]. Therefore, eluci- cell lines represent those with enhanced, potent prolifer- dating the molecular mechanisms underlying the critical ative capacity [14]. Increasingly, studies have demon- strated that the presence of PCSCs combined with drug * Correspondence: [email protected] resistance and high levels of metastasis contribute to 3Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX therapy failure, resulting in the high mortality rates for 77030, USA pancreatic cancer [5]. Furthermore, researchers have Full list of author information is available at the end of the article © 2013 Quan et al.; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated. Quan et al. Molecular Cancer 2013, 12:159 Page 2 of 14 http://www.molecular-cancer.com/content/12/1/159 proposed that Forkhead box protein M1 (FOXM1) is in- Further categorization of these alterations using whole volved in the self-renewal of PCSCs, and tumorigenesis exome sequencing revealed that they corresponded to 12 and metastasis of pancreatic cancer cells [15]. core signaling pathways, which contained several import- FOXM1 is a member of the Forkhead box transcrip- ant genes already confirmed to be drivers in the early tion factor superfamily, which consists of more than stages of PDAC development, such as cellular processes 50 members sharing a conserved winged-helix DNA- (KRAS), DNA damage control (tumor suppressor 53 binding domain. FOXM1 is a proliferation-specific tran- [TP53]), cell-cycle regulation (cyclin-dependent kinase scription factor whose expression is correlated with the inhibitor 2a [CDKN2A]), and transforming growth factor proliferative ability of cells [16]. FOXM1 is well known (TGF)-β signaling (SMAD4) [29]. The progression of to be a key cell-cycle regulator for both transition from PanIN from trivial dysplastic epithelium (PanIN-1) to cy- G1 to S phase and progression from G2 phase to mitosis tologic and nuclear atypia (PanIN-2 and −3, respectively) [17]. Increasing evidence suggests that FOXM1 expres- and, ultimately, invasive carcinoma corresponds to the sion is substantially elevated in most human malignan- gradual accumulation of genetic alterations. These al- cies, such as glioblastoma, lung cancer, hepatocellular terations include activating mutation of the oncogene carcinoma (HCC), breast cancer, and pancreatic cancer, KRAS and inactivation of the tumor suppressor genes and plays a crucial role in tumorigenesis, angiogenesis, CDKN2A, TP53, and SMAD4 [28,30,31]. invasion, and metastasis [17-23]. Also, several recent studies suggested that FOXM1 is involved in self-renewal KRAS and proliferation of CSCs [15,24,25]. However, the mo- The earliest and most important genetic alterations ob- lecular mechanisms by which FOXM1 signaling regulates served in pancreatic carcinogenesis are activating muta- PCSCs in pancreatic cancer development and progres- tions of KRAS [32-34]. At least 90% of PanIN-1 lesions sion remain poorly understood. harbor mutations of KRAS, and the average concentra- A deeper comprehension of PSCSs would likely pro- tion of mutant KRAS alleles in PanIN lesions increases vide a new perspective on and increased understanding markedly along with the PanIN grade; ultimately, almost of the mechanisms that govern the development of pan- all PanIN lesions have KRAS mutations [30,35]. Re- creatic cancer. In this review, we briefly describe the searchers have identified three major point mutations in crucial role of FOXM1 in PCSCs in pancreatic cancer codons 12, 13, and 61 that lead to the association of ab- development and progression with a focus on recent normal KRAS protein products with malignant trans- insight into the cross-talk between FOXM1 and signal- formation, with mutations in codon 12 being the most ing pathways in PCSCs here and below. important to pancreatic carcinogenesis [32,33]. KRAS encodes for a guanosine triphosphate (GTP)-binding The roles of FOXM1 and signaling pathways in protein that functions as a crucial modulator of a series the early stages of pancreatic ductal of cellular processes, such as proliferation, survival, and adenocarcinoma development motility. Ras activity in cells is tightly regulated, and Ras Over the past few decades, increasing evidence has dem- is normally bound to guanosine diphosphate (GDP) in an onstrated that almost all pancreatic cancers progress inactive state. Exogenous signals such as growth factor from diverse premalignant lesions to invasive carcin- stimulation trigger the removal of GDP from Ras, allow- omas. Precursors of pancreatic cancer include pancreatic ing for GTP to bind and activate downstream effectors. intraepithelial neoplasia (PanIN), intraductal papillary Activated ras feeds multiple signaling pathways, including mucinous neoplasms (IPMNs), mucinous cystic neo- the Ras/Raf/mitogen-activated protein kinase (MAPK) plasms (MCNs), and intraductal tubular papillary neo- pathway and the phosphoinositide 3-kinase (PI3K)/AKT plasms (ITPNs) [26-28]. Pancreatic carcinoma in general signaling pathway, which have diverse roles in cytoskeletal may arise from any of these precursor lesions, yet pan- alterations, cell-cycle progression, and apoptosis inhib- creatic ductal adenocarcinoma (PDAC) in particular is ition. Activating mutations of KRAS contribute to loss of much more closely associated with PanIN than with the intrinsic GTPase activity of Ras protein; consequently, other precursor lesions. PanIN lesions
Recommended publications
  • Predicting FOXM1-Mediated Gene Regulation Through the Analysis of Genome-Wide FOXM1 Binding Sites in MCF-7, K562, SK-N-SH, GM12878 and ECC-1 Cell Lines
    International Journal of Molecular Sciences Article Predicting FOXM1-Mediated Gene Regulation through the Analysis of Genome-Wide FOXM1 Binding Sites in MCF-7, K562, SK-N-SH, GM12878 and ECC-1 Cell Lines 1, , 2, 1 3,4 Keunsoo Kang * y, Yoonjung Choi y, Hoo Hyun Kim , Kyung Hyun Yoo and Sungryul Yu 5,* 1 Department of Microbiology, College of Science & Technology, Dankook University, Cheonan 31116, Korea; [email protected] 2 Deargen Inc., Daejeon 34051, Korea; [email protected] 3 Laboratory of Biomedical Genomics, Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; [email protected] 4 Research Institute of Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea 5 Department of Clinical Laboratory Science, Semyung University, Jecheon 27136, Korea * Correspondence: [email protected] (K.K.); [email protected] (S.Y.); Tel.: +82-41-550-3456 (K.K.); +82-43-649-1418 (S.Y.) These authors contributed equally to the work. y Received: 22 June 2020; Accepted: 24 August 2020; Published: 26 August 2020 Abstract: Forkhead box protein M1 (FOXM1) is a key transcription factor (TF) that regulates a common set of genes related to the cell cycle in various cell types. However, the mechanism by which FOXM1 controls the common gene set in different cellular contexts is unclear. In this study, a comprehensive meta-analysis of genome-wide FOXM1 binding sites in ECC-1, GM12878, K562, MCF-7, and SK-N-SH cell lines was conducted to predict FOXM1-driven gene regulation. Consistent with previous studies, different TF binding motifs were identified at FOXM1 binding sites, while the NFY binding motif was found at 81% of common FOXM1 binding sites in promoters of cell cycle-related genes.
    [Show full text]
  • Activation of AKT/AP1/Foxm1 Signaling Confers Sorafenib Resistance to Liver Cancer Cells
    ONCOLOGY REPORTS 42: 785-796, 2019 Activation of AKT/AP1/FoxM1 signaling confers sorafenib resistance to liver cancer cells DONGJING YAN1*, XIAOJING YAN2*, XUFANG DAI3,4*, LINGXI CHEN2, LIANGBO SUN2, TAO LI2, FENGTIAN HE2, JIQIN LIAN2 and WANGWEI CAI1 1Department of Biochemistry and Molecular Biology, Hainan Medical College, Haikou, Hainan 571199; 2Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038; 3Department of Educational Science College, Chongqing Normal University; 4Chongqing Key Laboratory of Psychological Diagnosis and Educational Technology for Children with Special Needs, Chongqing 400047, P.R. China Received December 5, 2018; Accepted June 6, 2019 DOI: 10.3892/or.2019.7192 Abstract. Sorafenib is the first‑line drug used in the treatment the AKT/AP1/FoxM1 signaling axis is an important determi- of liver cancer; however, drug resistance seriously limits the nant of sorafenib tolerance. clinical response to sorafenib. The present study investigated the molecular mechanisms of sorafenib resistance in liver Introduction cancer cells. The data indicated that forkhead box M1 (FoxM1) was significantly overexpressed in sorafenib‑resistant cells, at Liver cancer, the most common primary liver tumor, is the third the mRNA and protein levels. Knockdown of FoxM1 rendered leading cause of cancer mortality globally (1). Surgical resec- drug‑tolerant cells sensitive to sorafenib. Furthermore, FoxM1 tion, thermal ablation and liver transplantation are the current was upregulated at the transcriptional
    [Show full text]
  • SUPPLEMENTAL FIGURE LEGENDS Supplemental Figure S1. RBPJ
    Xie et al. SUPPLEMENTAL FIGURE LEGENDS Supplemental Figure S1. RBPJ correlates with BTIC marker expression. A-D. The TCGA GBM dataset was downloaded and correlations analyzed by R. RBPJ mRNA levels were highly correlated with (A) Olig2, (B) Sox2, (C) CD133, and (D) Sox4 levels. E. RBPJ is preferentially expressed in proneural glioblastomas. The glioblastoma TCGA dataset was interrogated for RBPJ mRNA expression segregated by transcriptional profile. The proneural tumors were further divided into G-CIMP (glioma CpG-island methylator phenotype) or non-G-CIMP. **, p < 0.01. ****, p < 0.0001. *****, p < 0.00001. Supplemental Figure S2. Targeting RBPJ induces BTIC apoptosis. A. 3691 BTICs were transduced with shCONT, shRBPJ-1, or shRBPJ-2. Lysates were prepared and immunoblotted with the indicated antibodies. shRNA-mediated knockdown of RBPJ was associated with increased cleaved (activated) PARP. B. 3691 BTICs were transduced with shCONT, shRBPJ-1, or shRBPJ-2. Apoptosis measured by Annexin V staining. Data are presented as mean ± SEM (two- way ANOVA; **, p < 0.01; n = 3). Supplemental Figure S3. Targeting RBPJ does not affect non-BTIC proliferation. Non-BTICs (Top, 3691; Bottom, 4121) were transduced with shCONT, shRBPJ-1, or shRBPJ-2. Cell proliferation was measured by CellTiter-Glo. 42 Xie et al. Supplemental Figure S4. RBPJ induces transcriptional profiles in BTICs distinct from Notch activation. A. In parallel experiments, 3691 BTICs were either treated with DAPT (at either 5 μM or 10 μM) vs. vehicle control (DMSO) or transduced with shRBPJ vs. shCONT. RNA-Seq was performed and the results displayed as a heat map with normalization to the relevant control.
    [Show full text]
  • FOXM1 Is a Transcriptional Target of Erα and Has a Critical Role in Breast
    Oncogene (2010) 29, 2983–2995 & 2010 Macmillan Publishers Limited All rights reserved 0950-9232/10 $32.00 www.nature.com/onc ORIGINAL ARTICLE FOXM1 is a transcriptional target of ERa and has a critical role in breast cancer endocrine sensitivity and resistance J Millour1, D Constantinidou1, AV Stavropoulou1, MSC Wilson1, SS Myatt1, JM-M Kwok1, K Sivanandan1, RC Coombes1, RH Medema2, J Hartman3, AE Lykkesfeldt4 and EW-F Lam1 1Department of Surgery and Cancer, Imperial College London, London, UK; 2Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands; 3Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden and 4Department of Breast Cancer Research, Institute of Cancer Biology, Danish Cancer Society, Copenhagen, Denmark In this study, we investigated the regulation of FOXM1 Introduction expression by estrogen receptor a (ERa) and its role in hormonal therapy and endocrine resistance. FOXM1 Breast cancer is the second most prevalent cause of protein and mRNA expression was regulated by ER- cancer death in the western hemisphere and displays a ligands, including estrogen, tamoxifen (OHT) and fulves- complex aeitology. The forkhead box (FOX) family trant (ICI182780; ICI) in breast carcinoma cell lines. member FOXM1 was previously reported to be elevated Depletion of ERa by RNA interference (RNAi) in MCF-7 in breast cancer as well as in carcinomas of other origins cells downregulated FOXM1 expression. Reporter gene (Pilarsky et al., 2004). FOXM1 is expressed in prolifer- assays showed that ERa activates FOXM1 transcription ating adult tissues and in response to injury or repair through an estrogen-response element (ERE) located (Korver et al., 1997a, b; Ye et al., 1999; Leung et al., within the proximal promoter region.
    [Show full text]
  • The Forkhead Transcription Factor FOXM1 Promotes Endocrine
    Bergamaschi et al. Breast Cancer Research 2014, 16:436 http://breast-cancer-research.com/content/16/5/436 RESEARCH ARTICLE Open Access The forkhead transcription factor FOXM1 promotes endocrine resistance and invasiveness in estrogen receptor–positive breast cancer by expansion of stem-like cancer cells Anna Bergamaschi1, Zeynep Madak-Erdogan1, Yu Jin Kim2, Yoon-La Choi2,3, Hailing Lu1,4 and Benita S Katzenellenbogen1* Abstract Introduction: The forkhead transcription factor FOXM1 coordinates expression of cell cycle–related genes and plays a pivotal role in tumorigenesis and cancer progression. We previously showed that FOXM1 acts downstream of 14-3-3ζ signaling, the elevation of which correlates with a more aggressive tumor phenotype. However, the role that FOXM1 might play in engendering resistance to endocrine treatments in estrogen receptor–positive (ER+) patients when tumor FOXM1 is high has not been clearly defined yet. Methods: We analyzed FOXM1 protein expression by immunohistochemistry in 501 ER-positive breast cancers. We also mapped genome-wide FOXM1, extracellular signal-regulated kinase 2 and ERα binding events by chromatin immunoprecipitation followed by high-throughput sequencing (ChIP-seq) in hormone-sensitive and resistant breast cancer cells after tamoxifen treatment. These binding profiles were integrated with gene expression data derived from cells before and after FOXM1 knockdown to highlight specific FOXM1 transcriptional networks. We also modulated the levels of FOXM1 and newly discovered FOXM1-regulated genes and examined their impact on the cancer stem-like cell population and on cell invasiveness and resistance to endocrine treatments. Results: FOXM1 protein expression was high in 20% of the tumors, which correlated with significantly reduced survival in these patients (P = 0.003 by logrank Mantel-Cox test).
    [Show full text]
  • FOXM1: a Multifunctional Oncoprotein and Emerging Therapeutic Target in Ovarian Cancer
    cancers Review FOXM1: A Multifunctional Oncoprotein and Emerging Therapeutic Target in Ovarian Cancer Cassie Liu, Carter J. Barger and Adam R. Karpf * Eppley Institute and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68918-6805, USA; [email protected] (C.L.); [email protected] (C.J.B.) * Correspondence: [email protected]; Tel.: +1-402-559-6115 Simple Summary: Ovarian cancer is a lethal disease in women with a 10-year survival rate of <40% worldwide. A key molecular alteration in ovarian cancer is the aberrant overexpression and activation of the transcription factor forkhead box M1 (FOXM1). FOXM1 regulates the expression of a multitude of genes that promote cancer, including those that increase the growth, survival, and metastatic spread of cancer cells. Importantly, FOXM1 overexpression is a robust biomarker for poor prognosis in pan-cancer and ovarian cancer. In this review, we first discuss the molecular mechanisms controlling FOXM1 expression and activity, with a specific emphasis on ovarian cancer. We then discuss the evidence for and the manner by which FOXM1 expression promotes aggressive cancer biology. Finally, we discuss the clinical utility of FOXM1, including its potential as a cancer biomarker and as a therapeutic target in ovarian cancer. Abstract: Forkhead box M1 (FOXM1) is a member of the conserved forkhead box (FOX) transcription factor family. Over the last two decades, FOXM1 has emerged as a multifunctional oncoprotein and a robust biomarker of poor prognosis in many human malignancies. In this review article, we address the current knowledge regarding the mechanisms of regulation and oncogenic functions of FOXM1, Citation: Liu, C.; Barger, C.J.; Karpf, particularly in the context of ovarian cancer.
    [Show full text]
  • Loss of the Forkhead Transcription Factor Foxm1 Causes Centrosome Amplification and Mitotic Catastrophe
    Research Article Loss of the Forkhead Transcription Factor FoxM1 Causes Centrosome Amplification and Mitotic Catastrophe Diane R. Wonsey and Maximillian T. Follettie Department of Discovery Medicine, Wyeth Research, Cambridge, Massachusetts Abstract with a targeted deletion of FoxM1 in the liver show decreased Expression of the forkhead transcription factor FoxM1 bromodeoxyuridine (BrdUrd) incorporation and fewer mitotic cells correlates with proliferative status in a variety of normal compared with wild-type controls following partial hepatectomy (6). and transformed cell types. Elevated expression of FoxM1 has Conversely, premature expression of FoxM1 in transgenic mice been noted in both hepatocellular carcinoma and basal cell accelerates hepatocyte DNA replication and the expression of cell carcinoma. However, whether FoxM1 expression is essential cycle regulatory proteins following partial hepatectomy (7). for the viability of transformed cells is unknown. We report Consistent with a role in proliferation, elevated expression of here that the expression of FoxM1 is significantly elevated in FoxM1 has been reported in both basal cell carcinoma (8) and in primary breast cancer. Microarray analysis shows that FoxM1 hepatocellular carcinoma (9). In addition, FoxM1 expression is re- regulates genes that are essential for faithful chromosome quired for the proliferative expansion of hepatocellular carcinoma segregation and mitosis, including Nek2, KIF20A, and CENP-A. in a mouse model of tumor induction (10). The observation that a p19ARF peptide fragment physically interacts with FoxM1, sup- Loss of FoxM1 expression generates mitotic spindle defects, delays cells in mitosis, and induces mitotic catastrophe. Time- presses FoxM1 transcriptional activity, and inhibits FoxM1- lapse microscopy indicates that depletion of FoxM1 generates enhanced anchorage-independent growth (10) suggests that cells that enter mitosis but are unable to complete cell FoxM1 may be an attractive target for cancer therapy.
    [Show full text]
  • FAM188B Expression Is Critical for Cell Growth Via FOXM1 Regulation in Lung Cancer
    biomedicines Article FAM188B Expression Is Critical for Cell Growth via FOXM1 Regulation in Lung Cancer Young Eun Choi 1, Hamadi Madhi 2 , HaEun Kim 1,3 , Jeon-Soo Lee 1, Myung-Hee Kim 2, Yong-Nyun Kim 1,* and Sung-Ho Goh 1,* 1 Research Institute, National Cancer Center, 323 Ilsan-ro, Goyang 10408, Korea; [email protected] (Y.E.C.); [email protected] (H.K.); [email protected] (J.-S.L.) 2 Department of Anatomy, Yonsei University College of Medicine, Seoul 03722, Korea; [email protected] (H.M.); [email protected] (M.-H.K.) 3 Department of Experimental Medicine, McGill University, Canada 845 Rue Sherbrooke O, Montréal, QC H3A 0G4, Canada * Correspondence: [email protected] (Y.-N.K.); [email protected] (S.-H.G.); Tel.: +82-31-920-2477 (S.-H.G.) Received: 7 October 2020; Accepted: 30 October 2020; Published: 31 October 2020 Abstract: Although family with sequence similarity 188 member B (FAM188B) is known to be a member of a novel putative deubiquitinase family, its biological role has not been fully elucidated. Here, we demonstrate the oncogenic function of FAM188B via regulation of forkhead box M1 (FOXM1), another oncogenic transcription factor, in lung cancer cells. FAM188B knockdown induced the inhibition of cell growth along with the downregulation of mRNA and protein levels of FOXM1. FAM188B knockdown also resulted in downregulation of Survivin and cell cycle-related proteins, which are direct targets of FOXM1. Interestingly, FOXM1 co-immunoprecipitated with FAM188B, and the levels of FOXM1 ubiquitination increased with FAM188B knockdown but decreased with FAM188B overexpression.
    [Show full text]
  • Plk1 Regulates the Repressor Function of Foxm1b by Inhibiting Its
    www.nature.com/scientificreports OPEN Plk1 Regulates the Repressor Function of FoxM1b by inhibiting its Interaction with the Received: 27 June 2016 Accepted: 09 March 2017 Retinoblastoma Protein Published: 07 April 2017 Nishit K. Mukhopadhyay1,*, Vaibhav Chand1,*, Akshay Pandey1, Dragana Kopanja1, Janai R. Carr2, Yi-Ju Chen3, Xiubei Liao1 & Pradip Raychaudhuri1,4 FoxM1b is a cell cycle-regulated transcription factor, whose over-expression is a marker for poor outcome in cancers. Its transcriptional activation function requires phosphorylation by Cdk1 or Cdk2 that primes FoxM1b for phosphorylation by Plk1, which triggers association with the co-activator CBP. FoxM1b also possesses transcriptional repression function. It represses the mammary differentiation gene GATA3 involving DNMT3b and Rb. We investigated what determines the two distinct functions of FoxM1b: activation and repression. We show that Rb binds to the C-terminal activation domain of FoxM1b. Analyses with phospho-defective and phospho-mimetic mutants of FoxM1b identified a critical role of the Plk1 phosphorylation sites in regulating the binding of FoxM1b to Rb and DNMT3b. That is opposite of what was seen for the interaction of FoxM1b with CBP. We show that, in addition to GATA3, FoxM1b also represses the mammary luminal differentiation marker FoxA1 by promoter- methylation, and that is regulated by the Plk1 phosphorylation sites in FoxM1b. Our results show that the Plk1 phosphorylation sites in FoxM1b serve as a regulator for its repressor function, and they provide insights into how FoxM1b inhibits differentiation genes and activates proliferation genes during cancer progression. The transcription factor FoxM1 is a driver of cancer progression. Its over-expression coincides with high-grade progression in almost all types of cancers, including those in breast1,2, liver3,4, lung5,6, prostate7,8, pancreas9,10, ovary11, brain12, nasopharynx13, esophagus14, and also in certain leukemia15.
    [Show full text]
  • Inhibitor of Differentiation 4 (ID4) Represses Myoepithelial Differentiation Of
    bioRxiv preprint doi: https://doi.org/10.1101/2020.04.06.026963; this version posted April 10, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 1 Inhibitor of Differentiation 4 (ID4) represses myoepithelial differentiation of 2 mammary stem cells through its interaction with HEB 1,2 1,2 1,2 1,2 3 Holly Holliday , Daniel Roden , Simon Junankar , Sunny Z. Wu , Laura A. 1,2 3,4 1 1 1 4 Baker , Christoph Krisp , Chia-Ling Chan , Andrea McFarland , Joanna N. Skhinas , 1,2 5 6 1,2 5 Thomas R. Cox , Bhupinder Pal , Nicholas Huntington , Christopher J. Ormandy , 7 5 3 1,2 6 Jason S. Carroll , Jane Visvader , Mark P. Molloy , Alexander Swarbrick 7 1. The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, 8 NSW 2010, Australia 9 2. St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 10 2010, Australia 11 3. Australian Proteome Analysis Facility, Macquarie University, Sydney, NSW 12 2109, Australia 13 4. Institute of Clinical Chemistry and Laboratory Medicine, Mass Spectrometric 14 Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg 20251, 15 Germany 16 5. ACRF Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of 17 Medical Research, Parkville, Victoria 3052, Australia 18 6. Biomedicine Discovery Institute, Department of Biochemistry and Molecular 19 Biology, Monash University, Clayton, VIC 3168, Australia 20 7. Cancer Research UK Cambridge Institute, University of Cambridge, Robinson 21 Way, Cambridge CB2 0RE, UK 22 bioRxiv preprint doi: https://doi.org/10.1101/2020.04.06.026963; this version posted April 10, 2020.
    [Show full text]
  • The Role of Inhibitor of DNA Binding 4 (Id4) in Mammary Gland Development and Breast Cancer
    The role of Inhibitor of DNA binding 4 (Id4) in mammary gland development and breast cancer Simon Junankar A thesis in fulfilment of the requirements for the degree of Doctor of Philosophy UNSW Garvan Institute of Medical Research St. Vincent’s Hospital Clinical School Faculty of Medicine November 2012 Copyright and Authenticity statement I Originality Statement ‘I hereby declare that this submission is my own work and to the best of my knowledge it contains no materials previously published or written by another person, or substantial proportions of material which have been accepted for the award of any other degree or diploma at UNSW or any other educational institution, except where due acknowledgement is made in the thesis. Any contribution made to the research by others, with whom I have worked at UNSW or elsewhere, is explicitly acknowledged in the thesis. I also declare that the intellectual content of this thesis is the product of my own work, except to the extent that the assistance from others in project’s design and conception or in style, presentation and linguistic expression is acknowledged.’ Signed: Date: 14/11/12 II Acknowledgements Firstly I would like to thank Alex Swarbrick for being a great supervisor, friend and mentor. You gave me the freedom to follow my own ideas and also gave direction when I needed it. I would also like to thank the other members of the Swarbrick group for their help, advice and friendship. In particular I would like to thank Radhika for conceptualising the Id4 project and for so much advice and help along the way.
    [Show full text]
  • Single-Cell RNA Landscape of Intratumoral Heterogeneity and Immunosuppressive Microenvironment in Advanced Osteosarcoma
    ARTICLE https://doi.org/10.1038/s41467-020-20059-6 OPEN Single-cell RNA landscape of intratumoral heterogeneity and immunosuppressive microenvironment in advanced osteosarcoma Yan Zhou1,11, Dong Yang2,11, Qingcheng Yang2,11, Xiaobin Lv 3,11, Wentao Huang4,11, Zhenhua Zhou5, Yaling Wang 1, Zhichang Zhang2, Ting Yuan2, Xiaomin Ding1, Lina Tang 1, Jianjun Zhang1, Junyi Yin1, Yujing Huang1, Wenxi Yu1, Yonggang Wang1, Chenliang Zhou1, Yang Su1, Aina He1, Yuanjue Sun1, Zan Shen1, ✉ ✉ ✉ ✉ Binzhi Qian 6, Wei Meng7,8, Jia Fei9, Yang Yao1 , Xinghua Pan 7,8 , Peizhan Chen 10 & Haiyan Hu1 1234567890():,; Osteosarcoma is the most frequent primary bone tumor with poor prognosis. Through RNA- sequencing of 100,987 individual cells from 7 primary, 2 recurrent, and 2 lung metastatic osteosarcoma lesions, 11 major cell clusters are identified based on unbiased clustering of gene expression profiles and canonical markers. The transcriptomic properties, regulators and dynamics of osteosarcoma malignant cells together with their tumor microenvironment particularly stromal and immune cells are characterized. The transdifferentiation of malignant osteoblastic cells from malignant chondroblastic cells is revealed by analyses of inferred copy-number variation and trajectory. A proinflammatory FABP4+ macrophages infiltration is noticed in lung metastatic osteosarcoma lesions. Lower osteoclasts infiltration is observed in chondroblastic, recurrent and lung metastatic osteosarcoma lesions compared to primary osteoblastic osteosarcoma lesions. Importantly, TIGIT blockade enhances the cytotoxicity effects of the primary CD3+ T cells with high proportion of TIGIT+ cells against osteo- sarcoma. These results present a single-cell atlas, explore intratumor heterogeneity, and provide potential therapeutic targets for osteosarcoma. 1 Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China.
    [Show full text]