HHS Public Access Author manuscript J Invest Dermatol. Author manuscript; available in PMC 2012 June 01.

Similar documents
mirna Dr. S Hosseini-Asl

Wikipedia Article on Epigenetics and Melanoma

p53 and Apoptosis: Master Guardian and Executioner Part 2

Normal RAS-RAF (MAPK) pathway signaling

MAPK Pathway. CGH Next Generation Sequencing. Molecular Tools in Care of Patients with Pigmented Lesions 7/20/2017

Chapter 2 Melanoma Pathogenesis

HALLA KABAT * Outreach Program, mircore, 2929 Plymouth Rd. Ann Arbor, MI 48105, USA LEO TUNKLE *

BIO360 Fall 2013 Quiz 1

HHS Public Access Author manuscript J Invest Dermatol. Author manuscript; available in PMC 2016 February 01.

BRaf V600E cooperates with Pten silencing to elicit metastatic melanoma (Nature Genetics Supplementary Information)

10/31/2017. micrornas and cancer. From the one gene-one enzyme hypothesis to. microrna DNA RNA. Transcription factors.

CELL CYCLE MOLECULAR BASIS OF ONCOGENESIS

CDH1 truncating alterations were detected in all six plasmacytoid-variant bladder tumors analyzed by whole-exome sequencing.

Hands-On Ten The BRCA1 Gene and Protein

Ch. 18 Regulation of Gene Expression

The PI3K/AKT axis. Dr. Lucio Crinò Medical Oncology Division Azienda Ospedaliera-Perugia. Introduction

TUMOR-SUPPRESSOR GENES. Molecular Oncology Michael Lea

A particular set of insults induces apoptosis (part 1), which, if inhibited, can switch to autophagy. At least in some cellular settings, autophagy se

Regulation of Gene Expression in Eukaryotes

The mutations that drive cancer. Paul Edwards. Department of Pathology and Cancer Research UK Cambridge Institute, University of Cambridge

Supplementary Information Titles Journal: Nature Medicine

Osamu Tetsu, MD, PhD Associate Professor Department of Otolaryngology-Head and Neck Surgery School of Medicine, University of California, San

Characterisation of structural variation in breast. cancer genomes using paired-end sequencing on. the Illumina Genome Analyser

New Developments in the Treatment of Colorectal Cancer. Jonathan Loree, MD, MS, FRCPC Department of Medical Oncology BC Cancer Vancouver Centre

Introduction to Cancer Biology

Insights from Sequencing the Melanoma Exome

RAS Genes. The ras superfamily of genes encodes small GTP binding proteins that are responsible for the regulation of many cellular processes.

Quantification of early stage lesions for loss of p53 should be shown in the main figures.

Bersagli molecolari nel melanoma

Supplementary Online Content

MRC-Holland MLPA. Description version 23; 26 January 2017

Post-transcriptional regulation of an intronic microrna

Problem Set 8 Key 1 of 8

Corporate Medical Policy Genetic Testing for Cutaneous Malignant Melanoma

OverView Circulating Nucleic Acids (CFNA) in Cancer Patients. Dave S.B. Hoon John Wayne Cancer Institute Santa Monica, CA, USA

BIO360 Fall 2013 Quiz 1

a) List of KMTs targeted in the shrna screen. The official symbol, KMT designation,

MOLECULAR BASIS OF ONCOGENESIS

Oncogenes and Tumor Suppressors MCB 5068 November 12, 2013 Jason Weber

The Biology and Genetics of Cells and Organisms The Biology of Cancer

SUPPLEMENTARY INFORMATION

Next Generation Sequencing in Clinical Practice: Impact on Therapeutic Decision Making

FILE // WHAT ARE TUMOR SUPPRESSOR GENES EBOOK

DETERMINATION OF K-RAS MUTATION IN COLORECTAL AND LUNG CANCER

KIT Inhibition in Advanced Melanoma: Rationale and Clinical i l Results

Molecular Cell Biology. Prof. D. Karunagaran. Department of Biotechnology. Indian Institute of Technology Madras

AP VP DLP H&E. p-akt DLP

p53 cooperates with DNA methylation and a suicidal interferon response to maintain epigenetic silencing of repeats and noncoding RNAs

Tumor suppressor genes D R. S H O S S E I N I - A S L

Introduction to Genetics

High AU content: a signature of upregulated mirna in cardiac diseases

Multistep nature of cancer development. Cancer genes

Analysis of Massively Parallel Sequencing Data Application of Illumina Sequencing to the Genetics of Human Cancers

Efficacy of IGFBP7 for Treatment of Metastatic Melanoma and other Cancers in Mouse Models and Human Cell Lines

Genetic Interaction Between NRAS and BRAF Mutations and PTEN/MMAC1 Inactivation in Melanoma

Activation of cellular proto-oncogenes to oncogenes. How was active Ras identified?

The clinical relevance of circulating, cell-free and exosomal micrornas as biomarkers for gynecological tumors

The pseudogene TUSC2P promotes TUSC2 function by binding multiple micrornas

Reviewers' comments: Reviewer #1 (Remarks to the Author):

Cell Cycle Checkpoint Genes and Cancer

Supplementary Material

Profiles of gene expression & diagnosis/prognosis of cancer. MCs in Advanced Genetics Ainoa Planas Riverola

THE FUTURE OF IMMUNOTHERAPY IN COLORECTAL CANCER. Prof. Dr. Hans Prenen, MD, PhD Oncology Department University Hospital Antwerp, Belgium

7/6/2015. Cancer Related Deaths: United States. Management of NSCLC TODAY. Emerging mutations as predictive biomarkers in lung cancer: Overview

Lecture 8 Neoplasia II. Dr. Nabila Hamdi MD, PhD

mirna-guided regulation at the molecular level

Supplementary Figure 1. a. b. Relative cell viability. Nature Genetics: doi: /ng SCR shyap1-1 shyap

Cell cycle, signaling to cell cycle, and molecular basis of oncogenesis

SUPPLEMENTARY INFORMATION

Clonal evolution of human cancers

Predisposition of Melanoma

Myelodysplastic Syndromes: Hematopathology. Analysis of SHIP1 as a potential biomarker of Disease Progression

C) The graph should look exactly like the graph on the left (Mut1 cells + Mating Pheromone for 3 hours at 25 degrees). The cells arrest in G1.

Selective filtering defect at the axon initial segment in Alzheimer s disease mouse models. Yu Wu

Hyperactivation of Ha-ras oncogene, but not Ink4a/Arf deficiency, triggers bladder tumorigenesis

microrna Presented for: Presented by: Date:

CCCTC-Binding Factor. Synonyms. Definition. Characteristics

MEDICAL POLICY. SUBJECT: GENOTYPING - RAS MUTATION ANALYSIS IN METASTATIC COLORECTAL CANCER (KRAS/NRAS) POLICY NUMBER: CATEGORY: Laboratory

Contents. Preface XV Acknowledgments XXI List of Abbreviations XXIII About the Companion Website XXIX

Response and resistance to BRAF inhibitors in melanoma

Lombardi Cancer Center Georgetown University Washington, DC

MicroRNA dysregulation in cancer. Systems Plant Microbiology Hyun-Hee Lee

CONTENTS. Preface... xii

EXAMPLE. - Potentially responsive to PI3K/mTOR and MEK combination therapy or mtor/mek and PKC combination therapy. ratio (%)

Deciphering the Role of micrornas in BRD4-NUT Fusion Gene Induced NUT Midline Carcinoma

Patricia Chevez-Barrrios AAOOP-USCAP /12/2016

Differential AKT dependency displayed by mouse models of BRAF V600E -initiated melanoma

Karyotype analysis reveals transloction of chromosome 22 to 9 in CML chronic myelogenous leukemia has fusion protein Bcr-Abl

Genetics and Genomics in Medicine Chapter 6 Questions

RESISTANCE TO TARGETED THERAPIES IN MELANOMA: NEW INSIGHTS

Post-transcriptional and translational regulation of mrna-like long non-coding RNAs by micrornas in early developmental stages of zebrafish embryos

HHS Public Access Author manuscript J Invest Dermatol. Author manuscript; available in PMC 2015 January 01.

Session 2: Biomarkers of epigenetic changes and their applicability to genetic toxicology

Src-INACTIVE / Src-INACTIVE

Supplementary Information

Defining how BRAF and NRAS mutations cooperate with UVA and UVB light to initiate melanoma.

Molecular Markers. Marcie Riches, MD, MS Associate Professor University of North Carolina Scientific Director, Infection and Immune Reconstitution WC

PIK3CA mutations are found in approximately 7% of

Regulation of p16 INK4A

Signaling Networks that Induce Melanomagenesis and Metastasis that can be Exploited for Therapeutic Benefit

Transcription:

Deletion of PTENP1 pseudogene in human melanoma Laura Poliseno 1, Adele Haimovic 1, Paul J. Christos 2, Eleazar C. Vega y Saenz de Miera 1, Richard Shapiro 1, Anna Pavlick 1, Russell S. Berman 1, Farbod Darvishian 1, and Iman Osman 1 1 The Interdisciplinary Melanoma Cooperative Group of the New York University School of Medicine, New York, NY, USA 2 Division of Biostatistics and Epidemiology, Weill Medical College of Cornell University, New York, NY, USA HHS Public Access Author manuscript Published in final edited form as: J Invest Dermatol. 2011 December ; 131(12): 2497 2500. doi:10.1038/jid.2011.232. Dear Editor, The aberrant activation of the PI3K/AKT pathway in melanoma is known to be caused by genomic deletion, promoter methylation or loss-of-function mutations of phosphatase and tensin homolog on chromosome 10 (PTEN) (see ref. (Madhunapantula and Robertson, 2009) for a review). More recently, PTEN protein abundance was shown to be decreased at the posttranscriptional level by a complex microrna network (He, 2010). The processed pseudogene of PTEN, PTENP1, is a modulator of the interaction between PTEN mrna and PTEN-targeting micrornas (Poliseno et al., 2010). PTENP1 shows extensive sequence similarity with PTEN. The high level of conservation in the most upstream region of the 3 UTR allows PTENP1 mrna to be bound by many of the microrna families that bind PTEN mrna (Figure 1 and Supplemental Text S1). Therefore, PTENP1 can protect PTEN from microrna-mediated downregulation. Consistent with its activity as a decoy for PTEN-targeting micrornas, PTENP1 is a bona fide tumor suppressor gene that causes growth inhibition in prostate cancer cells and undergoes deletion in various human malignancies (Poliseno et al, 2010). As recently pointed out (Chen, 2010), the post-transcriptional regulation of PTEN levels in melanoma has not been studied yet and the status of PTENP1 on 9p13 is unknown. Here, we report the analysis of PTENP1 locus in human melanoma cell lines and tissues (Table S1). We also analyze the relationship between: 1) PTENP1 deletion and CDKN2A deletion, as they are located approximately 20cM apart on chromosome 9p (Bennett, 2008); 2) PTENP1 deletion and PTEN deletion, as 9p and 10q losses often coexist in melanoma (Indsto et al., Users may view, print, copy, and download text and data-mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use:http://www.nature.com/authors/editorial_policies/license.html#terms Corresponding Author: Iman Osman, MD, New York University School of Medicine, 522 First Avenue, Smilow 405, New York, NY 10016, Phone: (212) 263-9076, Fax: (212) 263-9090, iman.osman@nyumc.org. CONFLICT OF INTEREST The authors declare no conflict of interest

Poliseno et al. Page 2 1998); 3) PTENP1 deletion and BRAF/NRAS mutation, as the combination of mutated BRAF (but not NRAS) and loss of PTEN expression is a common event in human melanoma (Tsao et al., 2004). Our genomic analysis revealed partial deletion of PTENP1 locus in 14.3% of the cell lines tested (Figure 2a). In addition, PTENP1 was found to be deleted in 20.9% of melanoma tissues (8 partial and 1 complete deletion) (Figure 2b). These results illustrate that PTENP1 is under selective pressure to undergo copy number losses (Poliseno et al., 2010). The average PTEN expression level in the 9 samples with either partial or complete deletion of PTENP1 (P5, P9, P11, P12, P20, M1, M9, M14 and M16) was lower than in the remaining 34 samples without any PTENP1 deletion (5.22 ± 2.30 N=9 vs. 20.59 ± 3.98 N=34, p = 0.06), confirming the protective effect of PTENP1 on PTEN levels (Poliseno et al., 2010). The details of the analyses of CDKN2A deletion, PTEN deletion, and BRAF and NRAS mutation in the human melanoma cell lines and tissues are provided in Supplemental Text S2. We observed that the partial or complete deletion of PTENP1 is always concomitant with the partial or complete deletion of CDKN2A in human melanoma cell lines, but not in human melanoma tissues. The focal deletion of PTENP1 (5/9 cases) has already been reported in other cancer types, such as breast and colon cancer (Poliseno et al., 2010), but is particularly interesting in melanoma, because it happens in spite of the fact that CDKN2A deletions are the most common alterations (Bennett, 2008). As far as the relationship between the partial or complete deletion of PTENP1 and that of PTEN is concerned, we observed that PTEN is deleted in 7/9 melanoma tissues that show PTENP1 deletion (Figure 2b). This result is supported by previous loss-of-heterozygosity analyses (Herbst et al., 1999) and is in agreement with the observation that 9p and 10q losses often coexist in melanoma (Indsto et al., 1998). The co-deletion of PTEN and PTENP1 can be reconciled with the function of PTENP1 as decoy for PTEN-targeting micrornas. PTEN deletion, either partial or complete, was found in 23 melanoma tissues. Excluding the cases in which PTEN deletion is complete and therefore PTENP1 deletion cannot affect PTEN levels, on average we observed lower PTEN protein expression in the samples showing both PTEN partial deletion and PTENP1 deletion compared to those showing PTEN partial deletion only, as exemplified by P5 and M5 tissues in Figure 2b. In the context of partial PTEN deletion, PTENP1 deletion may cause a further decrease in PTEN levels, possibly due to less efficient sheltering of PTEN-targeting micrornas. Of note, PTENP1 deletion may also affect PTEN levels when PTEN is downregulated by mechanisms other than genomic deletion, such as inactivating mutations. The biological effects of PTENP1 deletion are likely to be particularly strong on PTEN because of the intrinsic nature of this haploinsufficient tumor suppressor whose variations of even 20% can have profound consequences on cancer onset and progression (Berger and Pandolfi, 2011). Nonetheless, the concomitant deletion of PTENP1 and PTEN points towards PTEN-independent functions of PTENP1 as well. Two possible explanations can be

Poliseno et al. Page 3 invoked. First, PTENP1 is likely to function as decoy for additional microrna families which are not PTEN-targeting. These micrornas should preferentially bind to the R2 region of the 3 UTR that shows low homology with the corresponding one on PTEN (Figure 1a) and might affect targets that belong to other signaling cascades, so that the concomitant deletion of PTENP1 and PTEN causes the activation of independent pathways that possibly cooperate in tumorigenesis. Second, the PTEN-targeting micrornas for which PTENP1 acts as decoy have additional targets. Therefore, the deletion of PTENP1 might be advantageous for the tumor because of the decrease of other oncosuppressor genes besides PTEN (Poliseno et al., 2010). Examples of these genes are BIM, p21 and Sprouty2. The pro-apoptotic factor BIM is a mir-17 family-target (Fontana et al., 2008). Mutant BRAF and NRAS induce the MAPKdependent phosphorylation and consequent proteosomal degradation of BIM in order to provide melanoma cells with resistance to anoikis (Akiyama et al., 2009). The CDK inhibitor p21, another target of mir-17 family (Fontana et al., 2008), is repressed transcriptionally by TBX2 and TBX3 transcription factors which are frequently amplified in melanoma and are downstream effectors of the MAPK pathway (Bennett, 2008). Sprouty2 is a wt BRAF inhibitor that is targeted by mir-21. The downregulation of Sprouty2 observed in melanoma has been hypothesized as an alternative mechanism responsible for the aberrant activation of MAPK pathway besides BRAF mutation (Tsavachidou et al., 2004). Of note, the three genes described above share a common feature as antagonists of the MAPK pathway. Since the PI3K/AKT and the MAPK pathways have been shown to cooperate in melanomagenesis both in vitro and in mouse models (Dankort et al., 2009; Meier et al., 2007), it is tempting to hypothesize that, in the context of PTEN deletion, the deletion of PTENP1 might be an alternative mechanism evolved by the tumor to activate the MAPK pathway. In this respect, it is worth noticing that most of the cases showing both PTENP1 and PTEN deletion (71.4%) harbor wt BRAF. In conclusion, our data indicate that the recently identified tumor suppressor gene PTENP1 undergoes genomic deletion in human melanoma. Our data also suggest that PTENP1 deletion might be advantageous for the tumor not only because of its PTEN-related function, but also for PTEN-unrelated ones. Supplementary Material Refer to Web version on PubMed Central for supplementary material. ACKNOWLEDGEMENTS REFERENCES We thank Ting Tu, Daniel Lackaye and Holly Greenwald for the management of the clinical data of the IMCG patients. Dr. Paul J. Christos was partially supported by the Clinical Translational Science Center (CTSC) grant UL1-RR024996. This work was supported by the National Cancer Institute Cancer Center Support Grant (Grant No. 5 P30 CA 016087-27) and the Marc Jacobs Campaign to support the Interdisciplinary Melanoma Cooperative Group. Akiyama T, Dass CR, Choong PF. Bim-targeted cancer therapy: a link between drug action and underlying molecular changes. Mol Cancer Ther. 2009; 8:3173 3180. [PubMed: 19934277]

Poliseno et al. Page 4 Bennett DC. How to make a melanoma: what do we know of the primary clonal events? Pigment Cell Melanoma Res. 2008; 21:27 38. [PubMed: 18353141] Berger AH, Pandolfi PP. Haplo-insufficiency: a driving force in cancer. J Pathol. 2011; 223:137 146. [PubMed: 21125671] Chen M. Smoke and mirrors: pseudogenes tricking mirnas. Pigment Cell Melanoma Res. 2010; 23:583 584. [PubMed: 20701690] Dankort D, Curley DP, Cartlidge RA, Nelson B, Karnezis AN, Damsky WE Jr. et al. Braf(V600E) cooperates with Pten loss to induce metastatic melanoma. Nat Genet. 2009; 41:544 552. [PubMed: 19282848] Fontana L, Fiori ME, Albini S, Cifaldi L, Giovinazzi S, Forloni M, et al. Antagomir-17-5p abolishes the growth of therapy-resistant neuroblastoma through p21 and BIM. PLoS ONE. 2008; 3:e2236. [PubMed: 18493594] He L. Posttranscriptional regulation of PTEN dosage by noncoding RNAs. Sci Signal. 2010; 3:pe39. [PubMed: 21045203] Herbst RA, Podewski EK, Mommert S, Kapp A, Weiss J. PTEN and MXI1 allelic loss on chromosome 10q is rare in melanoma in vivo. Arch Dermatol Res. 1999; 291:567 569. [PubMed: 10552217] Indsto JO, Holland EA, Kefford RF, Mann GJ. 10q deletions in metastatic cutaneous melanoma. Cancer Genet Cytogenet. 1998; 100:68 71. [PubMed: 9406584] Madhunapantula SV, Robertson GP. The PTEN-AKT3 signaling cascade as a therapeutic target in melanoma. Pigment Cell Melanoma Res. 2009; 22:400 419. [PubMed: 19493313] Meier F, Busch S, Lasithiotakis K, Kulms D, Garbe C, Maczey E, et al. Combined targeting of MAPK and AKT signalling pathways is a promising strategy for melanoma treatment. Br J Dermatol. 2007; 156:1204 1213. [PubMed: 17388918] Poliseno L, Salmena L, Zhang J, Carver B, Haveman WJ, Pandolfi PP. A coding-independent function of gene and pseudogene mrnas regulates tumour biology. Nature. 2010; 465:1033 1038. [PubMed: 20577206] Tsao H, Goel V, Wu H, Yang G, Haluska FG. Genetic interaction between NRAS and BRAF mutations and PTEN/MMAC1 inactivation in melanoma. J Invest Dermatol. 2004; 122:337 341. [PubMed: 15009714] Tsavachidou D, Coleman ML, Athanasiadis G, Li S, Licht JD, Olson MF, et al. SPRY2 is an inhibitor of the ras/extracellular signal-regulated kinase pathway in melanocytes and melanoma cells with wild-type BRAF but not with the V599E mutant. Cancer Res. 2004; 64:5556 5559. [PubMed: 15313890]

Poliseno et al. Page 5 Figure 1. PTEN- and PTENP1-targeting micrornas a. Schematic representation of PTEN and PTENP1 3 UTR. The most upstream region of the 3 UTR (R1, dark grey) is highly conserved between PTEN and PTENP1. In the middle R2 region (light grey), the conservation drops dramatically. The downstream R3 region (white) is present only in PTEN (modified from Poliseno et al., 2010). b. List of the PTEN-targeting microrna families validated so far. The region of the 3 UTR where the microrna binding sites are located and the conservation of these sites in PTENP1 3 UTR are indicated. Out of a total of 18 PTEN-targeting microrna families, 12 are PTENP1-targeting as well (grey). All these families have at least 1 binding site in the high homology R1 region.

Poliseno et al. Page 6 Figure. 2. Analysis of CDKN2A, CDKN2B, MTAP, PTENP1, PTEN, BRAF and NRAS status in human melanoma cell lines (a) and tissues (b) a. 14 primary (WM series) and 14 metastatic (Sk-Mel series, 501Mel, A375 and 451Lu) melanoma cell lines and b. 23 primary (P1-P23) and 20 metastatic (M1-M20) melanoma tissues were analyzed by qpcr on genomic DNA to identify deletions at 9p21 (CDKN2A, CDKN2B and MTAP loci), 9p13 (PTENP1 locus) and 10q (PTEN locus). Mutations identified in exon 11 and 15 of BRAF and exon 2 and 3 of NRAS are reported. PTEN protein levels were analyzed in melanoma tissues by immunohistochemistry (IHC) and the percentage of positive cells is indicated. N/A: genomic qpcr not performed due to constraints on DNA availability. On the left, IHC shows lower PTEN levels in P5 tissue harboring partial PTEN and PTENP1 deletion compared to M5 tissue harboring partial PTEN deletion only.