1
|
Li T, Liu W, Hui W, Shi T, Liu H, Feng Y, Gao F. Integrated Analysis of Ulcerative Colitis Revealed an Association between PHLPP2 and Immune Infiltration. DISEASE MARKERS 2022; 2022:4983471. [PMID: 35308140 PMCID: PMC8931176 DOI: 10.1155/2022/4983471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/08/2022] [Accepted: 02/17/2022] [Indexed: 02/07/2023]
Abstract
Ulcerative colitis (UC) is a progressive intestine inflammatory disease that is prone to recur. Herein, we utilize microarray technology and bioinformatics to reveal the underlying pathogenesis of UC and provide novel markers. Colonic biopsies were taken from eight UC patients and eight healthy controls. Three differentially expressed miRNAs (DEMIs) and 264 differentially expressed genes (DEGs) were screened using mRNA and miRNA microarray. Most DEGs were significantly associated with immune response and were markedly enriched in the IL-17 signaling pathway. Among the target genes of DEMIs, PHLPP2 overlapped with DEGs and the downregulation of PHLPP2 group was mainly involved in the epithelial-mesenchymal transition. PHLPP2 was downregulated in UC patients, which was validated in 5 GEO datasets and qRT-PCR. The ROC curve demonstrated that PHLPP2 has a perfect ability to distinguish UC patients from healthy controls. Moreover, PHLPP2 was low expression in patients with active UC. CIBERSORT algorithm indicated that the abundance of gamma delta T cells (P = 0.04), M0 macrophages (P = 0.01), and activated mast cells (P < 0.01) was significantly greater than that of the control group. The Spearman correlation analysis showed that PHLPP2 was positively correlated with the proportion of activated NK cells (rho = 0.62, P = 0.013) and Tregs (rho = 0.55, P = 0.03), but negatively correlated with those of activated mast cells (rho = -0.8, P < 0.01) and macrophages (rho = -0.73, P < 0.01). These results indicate that PHLPP2 is associated with immune cells in the pathogenesis of UC, as well as provide new prospects and future directions of investigation.
Collapse
Affiliation(s)
- Ting Li
- Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Weidong Liu
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
- Xinjiang Clinical Research Center for Digestive Diseases, China
| | - Wenjia Hui
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
- Xinjiang Clinical Research Center for Digestive Diseases, China
| | - Tian Shi
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
- Xinjiang Clinical Research Center for Digestive Diseases, China
| | - Huan Liu
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
- Xinjiang Clinical Research Center for Digestive Diseases, China
| | - Yan Feng
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
- Xinjiang Clinical Research Center for Digestive Diseases, China
| | - Feng Gao
- Xinjiang Medical University, Urumqi, Xinjiang, China
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
- Xinjiang Clinical Research Center for Digestive Diseases, China
| |
Collapse
|
2
|
The PHLPP1 N-Terminal Extension Is a Mitotic Cdk1 Substrate and Controls an Interactome Switch. Mol Cell Biol 2021; 41:e0033320. [PMID: 33397691 PMCID: PMC8088274 DOI: 10.1128/mcb.00333-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
PH domain leucine-rich repeat protein phosphatase 1 (PHLPP1) is a tumor suppressor that directly dephosphorylates a wide array of substrates, most notably the prosurvival kinase Akt. However, little is known about the molecular mechanisms governing PHLPP1 itself. Here, we report that PHLPP1 is dynamically regulated in a cell cycle-dependent manner and deletion of PHLPP1 results in mitotic delays and increased rates of chromosomal segregation errors. We show that PHLPP1 is hyperphosphorylated during mitosis by Cdk1 in a functionally uncharacterized region known as the PHLPP1 N-terminal extension (NTE). A proximity-dependent biotin identification (BioID) interaction screen revealed that during mitosis, PHLPP1 dissociates from plasma membrane scaffolds, such as Scribble, by a mechanism that depends on its NTE and gains proximity to kinetochore and mitotic spindle proteins such as KNL1 and TPX2. Our data are consistent with a model in which phosphorylation of PHLPP1 during mitosis regulates binding to its mitotic partners and allows accurate progression through mitosis. The finding that PHLPP1 binds mitotic proteins in a cell cycle- and phosphorylation-dependent manner may have relevance to its tumor-suppressive function.
Collapse
|
3
|
Baffi TR, Cohen-Katsenelson K, Newton AC. PHLPPing the Script: Emerging Roles of PHLPP Phosphatases in Cell Signaling. Annu Rev Pharmacol Toxicol 2021; 61:723-743. [PMID: 32997603 PMCID: PMC11003498 DOI: 10.1146/annurev-pharmtox-031820-122108] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Whereas protein kinases have been successfully targeted for a variety of diseases, protein phosphatases remain an underutilized therapeutic target, in part because of incomplete characterization of their effects on signaling networks. The pleckstrin homology domain leucine-rich repeat protein phosphatase (PHLPP) is a relatively new player in the cell signaling field, and new roles in controlling the balance among cell survival, proliferation, and apoptosis are being increasingly identified. Originally characterized for its tumor-suppressive function in deactivating the prosurvival kinase Akt, PHLPP may have an opposing role in promoting survival, as recent evidence suggests. Additionally, identification of the transcription factor STAT1 as a substrate unveils a role for PHLPP as a critical mediator of transcriptional programs in cancer and the inflammatory response. This review summarizes the current knowledge of PHLPP as both a tumor suppressor and an oncogene and highlights emerging functions in regulating gene expression and the immune system. Understanding the context-dependent functions of PHLPP is essential for appropriate therapeutic intervention.
Collapse
Affiliation(s)
- Timothy R Baffi
- Department of Pharmacology, University of California, San Diego, La Jolla, California 92093-0721, USA;
| | - Ksenya Cohen-Katsenelson
- Department of Pharmacology, University of California, San Diego, La Jolla, California 92093-0721, USA;
| | - Alexandra C Newton
- Department of Pharmacology, University of California, San Diego, La Jolla, California 92093-0721, USA;
| |
Collapse
|
4
|
Berberine induced modulation of PHLPP2-Akt-MST1 kinase signaling is coupled with mitochondrial impairment and hepatoma cell death. Toxicol Appl Pharmacol 2018; 347:92-103. [DOI: 10.1016/j.taap.2018.03.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/28/2018] [Accepted: 03/30/2018] [Indexed: 12/11/2022]
|
5
|
PHLPPing through history: a decade in the life of PHLPP phosphatases. Biochem Soc Trans 2017; 44:1675-1682. [PMID: 27913677 DOI: 10.1042/bst20160170] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 08/12/2016] [Accepted: 08/16/2016] [Indexed: 01/30/2023]
Abstract
In the decade since their discovery, the PH domain leucine-rich repeat protein phosphatases (PHLPP) have emerged as critical regulators of cellular homeostasis, and their dysregulation is associated with various pathophysiologies, ranging from cancer to degenerative diseases, such as diabetes and heart disease. The two PHLPP isozymes, PHLPP1 and PHLPP2, were identified in a search for phosphatases that dephosphorylate Akt, and thus suppress growth factor signaling. However, given that there are over 200 000 phosphorylated residues in a single cell, and fewer than 50 Ser/Thr protein phosphatases, it is not surprising that PHLPP has many other cellular functions yet to be discovered, including a recently identified role in regulating the epigenome. Both PHLPP1 and PHLPP2 are commonly deleted in human cancers, supporting a tumor suppressive role. Conversely, the levels of one isozyme, PHLPP1, are elevated in diabetes. Thus, mechanisms to correctly control PHLPP activity in cells are critical for normal cellular homeostasis. This review summarizes the known functions of PHLPP and its role in disease.
Collapse
|
6
|
Stankiewicz E, Mao X, Mangham DC, Xu L, Yeste-Velasco M, Fisher G, North B, Chaplin T, Young B, Wang Y, Kaur Bansal J, Kudahetti S, Spencer L, Foster CS, Møller H, Scardino P, Oliver RT, Shamash J, Cuzick J, Cooper CS, Berney DM, Lu YJ. Identification of FBXL4 as a Metastasis Associated Gene in Prostate Cancer. Sci Rep 2017; 7:5124. [PMID: 28698647 PMCID: PMC5505985 DOI: 10.1038/s41598-017-05209-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 05/24/2017] [Indexed: 01/26/2023] Open
Abstract
Prostate cancer is the most common cancer among western men, with a significant mortality and morbidity reported for advanced metastatic disease. Current understanding of metastatic disease is limited due to difficulty of sampling as prostate cancer mainly metastasizes to bone. By analysing prostate cancer bone metastases using high density microarrays, we found a common genomic copy number loss at 6q16.1-16.2, containing the FBXL4 gene, which was confirmed in larger series of bone metastases by fluorescence in situ hybridisation (FISH). Loss of FBXL4 was also detected in primary tumours and it was highly associated with prognostic factors including high Gleason score, clinical stage, prostate-specific antigen (PSA) and extent of disease, as well as poor patient survival, suggesting that FBXL4 loss contributes to prostate cancer progression. We also demonstrated that FBXL4 deletion is detectable in circulating tumour cells (CTCs), making it a potential prognostic biomarker by 'liquid biopsy'. In vitro analysis showed that FBXL4 plays a role in regulating the migration and invasion of prostate cancer cells. FBXL4 potentially controls cancer metastasis through regulation of ERLEC1 levels. Therefore, FBXL4 could be a potential novel prostate cancer suppressor gene, which may prevent cancer progression and metastasis through controlling cell invasion.
Collapse
Affiliation(s)
- Elzbieta Stankiewicz
- Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Xueying Mao
- Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - D Chas Mangham
- The Robert Jones and Agnes Hunt Orthopaedic Hospital, Department of Pathology, Oswestry, Shropshire, SY10 7AG, UK
| | - Lei Xu
- Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Marc Yeste-Velasco
- Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Gabrielle Fisher
- Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, EC1 6BQ, UK
| | - Bernard North
- Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, EC1 6BQ, UK
| | - Tracy Chaplin
- Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Bryan Young
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Yuqin Wang
- Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Jasmin Kaur Bansal
- Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Sakunthala Kudahetti
- Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Lucy Spencer
- Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Christopher S Foster
- Division of Cellular and Molecular Pathology, University of Liverpool, Liverpool, L69 3BX, UK
- HCA Pathology Laboratories, Shropshire House, Capper Street, London, WC1E6JA, UK
| | - Henrik Møller
- King's College London, Cancer Epidemiology and Population Health, London, SE1 9RT, UK
| | - Peter Scardino
- Department of Urology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - R Tim Oliver
- Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Jonathan Shamash
- Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Jack Cuzick
- Cancer Research UK Centre for Epidemiology, Mathematics and Statistics, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, EC1 6BQ, UK
| | - Colin S Cooper
- School of Medicine, University of East Anglia, Norwich, NR4 7TJ, UK
| | - Daniel M Berney
- Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Yong-Jie Lu
- Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
7
|
Kamal M, Holliday DL, Morrison EE, Speirs V, Toomes C, Bell SM. Loss of CSMD1 expression disrupts mammary duct formation while enhancing proliferation, migration and invasion. Oncol Rep 2017; 38:283-292. [PMID: 28534981 DOI: 10.3892/or.2017.5656] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 05/11/2017] [Indexed: 11/06/2022] Open
Abstract
The CUB and sushi multiple domains 1 (CSMD1) gene maps to chromosome 8p23, a region deleted in many cancers. Loss of CSMD1 expression is associated with poor prognosis in breast cancer suggesting that it acts as a tumour suppressor in this cancer. However, the function of CSMD1 is largely unknown. Herein, we investigated CSMD1 functions in cell line models. CSMD1 expression was suppressed in MCF10A and LNCaP cells using short hairpin RNA. Functional assays were performed focusing on the 'normal' MCF10A cell line. Suppression of CSMD1 significantly increased the proliferation, cell migration and invasiveness of MCF10A cells compared to shcontrols. shCSMD1 cells also showed significantly reduced adhesion to Matrigel and fibronectin. In a three-dimensional Matrigel model of MCF10A cells, reduced CSMD1 expression resulted in the development of larger and more poorly differentiated breast acini-like structures that displayed impaired lumen formation. Loss of CSMD1 expression disrupts a model of mammary duct formation while enhancing proliferation, migration and invasion. Our data suggest that CSMD1 is involved in the suppression of a transformed phenotype.
Collapse
Affiliation(s)
- Mohamed Kamal
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Deborah L Holliday
- Leeds Institute of Cancer and Pathology, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Ewan E Morrison
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Valerie Speirs
- Leeds Institute of Cancer and Pathology, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Carmel Toomes
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Sandra M Bell
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds LS9 7TF, UK
| |
Collapse
|
8
|
Regulation of PI3K effector signalling in cancer by the phosphoinositide phosphatases. Biosci Rep 2017; 37:BSR20160432. [PMID: 28082369 PMCID: PMC5301276 DOI: 10.1042/bsr20160432] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 01/10/2017] [Accepted: 01/12/2017] [Indexed: 12/24/2022] Open
Abstract
Class I phosphoinositide 3-kinase (PI3K) generates phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P3) at the plasma membrane in response to growth factors, activating a signalling cascade that regulates many cellular functions including cell growth, proliferation, survival, migration and metabolism. The PI3K pathway is commonly dysregulated in human cancer, and drives tumorigenesis by promoting aberrant cell growth and transformation. PtdIns(3,4,5)P3 facilitates the activation of many pleckstrin homology (PH) domain-containing proteins including the serine/threonine kinase AKT. There are three AKT isoforms that are frequently hyperactivated in cancer through mutation, amplification or dysregulation of upstream regulatory proteins. AKT isoforms have converging and opposing functions in tumorigenesis. PtdIns(3,4,5)P3 signalling is degraded and terminated by phosphoinositide phosphatases such as phosphatase and tensin homologue (PTEN), proline-rich inositol polyphosphate 5-phosphatase (PIPP) (INPP5J) and inositol polyphosphate 4-phosphatase type II (INPP4B). PtdIns(3,4,5)P3 is rapidly hydrolysed by PIPP to generate phosphatidylinositol 3,4-bisphosphate (PtdIns(3,4)P2), which is further hydrolysed by INPP4B to form phosphatidylinositol 3-phosphate (PtdIns3P). PtdIns(3,4)P2 and PtdIns3P are also important signalling molecules; PtdIns(3,4)P2 together with PtdIns(3,4,5)P3 are required for maximal AKT activation and PtdIns3P activates PI3K-dependent serum and glucocorticoid-regulated kinase (SGK3) signalling. Loss of Pten, Pipp or Inpp4b expression or function promotes tumour growth in murine cancer models through enhanced AKT isoform-specific signalling. INPP4B inhibits PtdIns(3,4)P2-mediated AKT activation in breast and prostate cancer; however, INPP4B expression is increased in acute myeloid leukaemia (AML), melanoma and colon cancer where it paradoxically promotes cell proliferation, transformation and/or drug resistance. This review will discuss how PTEN, PIPP and INPP4B distinctly regulate PtdIns(3,4,5)P3 signalling downstream of PI3K and how dysregulation of these phosphatases affects cancer outcomes.
Collapse
|
9
|
Thériault BL, Cybulska P, Shaw PA, Gallie BL, Bernardini MQ. The role of KIF14 in patient-derived primary cultures of high-grade serous ovarian cancer cells. J Ovarian Res 2014; 7:123. [PMID: 25528264 PMCID: PMC4302703 DOI: 10.1186/s13048-014-0123-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 12/12/2014] [Indexed: 02/07/2023] Open
Abstract
Objective Previously, it has been shown that KIF14 mRNA is overexpressed in ovarian cancer (OvCa), regardless of histological subtype. KIF14 levels are independently predictive of poor outcome and increased rates of recurrence in serous OvCa patients. Furthermore, it has been shown that KIF14 also controls the in vivo tumorigenicity of OvCa cell lines. In this study, we evaluate the potential of KIF14 as a therapeutic target through selective inhibition of KIF14 in primary high-grade serous patient-derived OvCa cells. Methods To assess the dependence of primary serous OvCa cultures on KIF14, protein levels in 11 prospective high grade serous ovarian cancer samples were increased (KIF14 overexpression by transfection) or decreased (anti-KIF14 shRNA) in vitro, and proliferative capacity, anchorage independence and xenograft growth were assessed. Results Seven of eleven samples demonstrated increased/decreased in vitro proliferation in response to KIF14 overexpression/knockdown, respectively. When examining in vitro tumorigenicity (colony formation) and in vivo growth (subcutaneous xenografts) in response to KIF14 manipulation, none of the samples demonstrated growth in soft agar (11 samples), or xenograft growth (4 samples). Conclusions Although primary high-grade serous OvCa cells may depend on KIF14 for in vitro proliferation we were unable to demonstrate a role for KIF14 on tumorigenicity or develop an in vivo model for assessment. We have, however developed an effective in vitro method to evaluate the effect of target gene manipulation on the proliferative capacity of primary OvCa cultures. Electronic supplementary material The online version of this article (doi:10.1186/s13048-014-0123-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Brigitte L Thériault
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada.
| | - Paulina Cybulska
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada. .,Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada. .,Division of Gynecological Oncology, University Health Network, Toronto, ON, Canada.
| | - Patricia A Shaw
- Department of Pathology, University Health Network, Toronto, ON, Canada. .,Princess Margaret Hospital, University Health Network Tissue Bank, Toronto, ON, Canada.
| | - Brenda L Gallie
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada. .,Division of Visual Science, Toronto Western Hospital Research Institute, Toronto, ON, Canada. .,Departments of Medical Biophysics, Molecular Genetics, and Ophthalmology, University of Toronto, Toronto, ON, Canada.
| | - Marcus Q Bernardini
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada. .,Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada. .,Division of Gynecological Oncology, University Health Network, Toronto, ON, Canada. .,Princess Margaret Cancer Centre, Rm M700, 610 University Ave, Toronto, Ontario, M5G 2M9, Canada.
| |
Collapse
|
10
|
Abstract
Precise control of the balance between protein phosphorylation, catalyzed by protein kinases, and protein dephosphorylation, catalyzed by protein phosphatases, is essential for cellular homeostasis. Dysregulation of this balance leads to pathophysiological states, driving diseases such as cancer, heart disease, and diabetes. Aberrant phosphorylation of components of the pathways that control cell growth and cell survival are particularly prevalent in cancer. One of the most studied tumor suppressors in these pathways is the lipid phosphatase PTEN (phosphatase and tensin homolog deleted on chromosome ten), which dephosphorylates the lipid second messenger phosphatidylinositol 3,4,5-trisphosphate (PIP3), thus preventing activation of the oncogenic kinase AKT (v-akt murine thymoma viral oncogene homolog). In 2005, the discovery of a family of protein phosphatases whose members directly dephosphorylate and inactivate AKT introduced a new negative regulator of the phosphoinositide 3-kinase (PI3K) oncogenic pathway. Pleckstrin homology domain leucine-rich repeat protein phosphatase (PHLPP) isozymes comprise a novel tumor suppressor family whose two members, PHLPP1 and PHLPP2, are deleted as frequently as PTEN in cancers such as those of the prostate. PHLPP is thus a novel therapeutic target to suppress oncogenic pathways and is a potential candidate biomarker to stratify patients for the appropriate targeted therapeutics. This review discusses the role of PHLPP in terminating AKT signaling and how pharmacological intervention would impact this pathway.
Collapse
Affiliation(s)
- Alexandra C Newton
- Department of Pharmacology, University of California, San Diego, La Jolla, California 92093;
| | | |
Collapse
|
11
|
Gangula NR, Maddika S. WD repeat protein WDR48 in complex with deubiquitinase USP12 suppresses Akt-dependent cell survival signaling by stabilizing PH domain leucine-rich repeat protein phosphatase 1 (PHLPP1). J Biol Chem 2013; 288:34545-54. [PMID: 24145035 PMCID: PMC3843068 DOI: 10.1074/jbc.m113.503383] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
PHLPP1 (PH domain leucine-rich repeat protein phosphatase 1) is a protein-serine/threonine phosphatase and a negative regulator of the PI3-kinase/Akt pathway. Although its function as a suppressor of tumor cell growth has been established, the mechanism of its regulation is not completely understood. In this study, by utilizing the tandem affinity purification approach we have identified WDR48 and USP12 as novel PHLPP1-associated proteins. The WDR48·USP12 complex deubiquitinates PHLPP1 and thereby enhances its protein stability. Similar to PHLPP1 function, WDR48 and USP12 negatively regulate Akt activation and thus promote cellular apoptosis. Functionally, we show that WDR48 and USP12 suppress proliferation of tumor cells. Importantly, we found a WDR48 somatic mutation (L580F) that is defective in stabilizing PHLPP1 in colorectal cancers, supporting a WDR48 role in tumor suppression. Together, our results reveal WDR48 and USP12 as novel PHLPP1 regulators and potential suppressors of tumor cell survival.
Collapse
Affiliation(s)
- Narmadha Reddy Gangula
- From the Laboratory of Cell Death and Cell Survival, Centre for DNA Fingerprinting and Diagnostics (CDFD), Nampally, Hyderabad 500001, India
| | | |
Collapse
|
12
|
Yuan Y, Failmezger H, Rueda OM, Ali HR, Gräf S, Chin SF, Schwarz RF, Curtis C, Dunning MJ, Bardwell H, Johnson N, Doyle S, Turashvili G, Provenzano E, Aparicio S, Caldas C, Markowetz F. Quantitative image analysis of cellular heterogeneity in breast tumors complements genomic profiling. Sci Transl Med 2013; 4:157ra143. [PMID: 23100629 DOI: 10.1126/scitranslmed.3004330] [Citation(s) in RCA: 278] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Solid tumors are heterogeneous tissues composed of a mixture of cancer and normal cells, which complicates the interpretation of their molecular profiles. Furthermore, tissue architecture is generally not reflected in molecular assays, rendering this rich information underused. To address these challenges, we developed a computational approach based on standard hematoxylin and eosin-stained tissue sections and demonstrated its power in a discovery and validation cohort of 323 and 241 breast tumors, respectively. To deconvolute cellular heterogeneity and detect subtle genomic aberrations, we introduced an algorithm based on tumor cellularity to increase the comparability of copy number profiles between samples. We next devised a predictor for survival in estrogen receptor-negative breast cancer that integrated both image-based and gene expression analyses and significantly outperformed classifiers that use single data types, such as microarray expression signatures. Image processing also allowed us to describe and validate an independent prognostic factor based on quantitative analysis of spatial patterns between stromal cells, which are not detectable by molecular assays. Our quantitative, image-based method could benefit any large-scale cancer study by refining and complementing molecular assays of tumor samples.
Collapse
Affiliation(s)
- Yinyin Yuan
- Cancer Research UK Cambridge Research Institute, Cambridge CB2 0RE, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Ibeawuchi C, Schmidt H, Voss R, Titze U, Abbas M, Neumann J, Eltze E, Hoogland AM, Jenster G, Brandt B, Semjonow A. Genome-wide investigation of multifocal and unifocal prostate cancer-are they genetically different? Int J Mol Sci 2013; 14:11816-29. [PMID: 23736690 PMCID: PMC3709757 DOI: 10.3390/ijms140611816] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 05/20/2013] [Accepted: 05/27/2013] [Indexed: 11/18/2022] Open
Abstract
Prostate cancer is widely observed to be biologically heterogeneous. Its heterogeneity is manifested histologically as multifocal prostate cancer, which is observed more frequently than unifocal prostate cancer. The clinical and prognostic significance of either focal cancer type is not fully established. To investigate prostate cancer heterogeneity, the genetic profiles of multifocal and unifocal prostate cancers were compared. Here, we report observations deduced from tumor-tumor comparison of copy number alteration data of both focal categories. Forty-one fresh frozen prostate cancer foci from 14 multifocal prostate cancers and eight unifocal prostate cancers were subjected to copy number variation analysis with the Affymetrix SNP 6.0 microarray tool. With the investigated cases, tumors obtained from a single prostate exhibited different genetic profiles of variable degrees. Further comparison identified no distinct genetic pattern or signatures specific to multifocal or unifocal prostate cancer. Our findings suggest that samples obtained from multiple sites of a single unifocal prostate cancer show as much genetic heterogeneity and variability as separate tumors obtained from a single multifocal prostate cancer.
Collapse
Affiliation(s)
- Chinyere Ibeawuchi
- Prostate Center, Department of Urology, University Hospital Muenster, Albert-Schweitzer-Campus 1, Gebaeude 1A, Muenster D-48149, Germany; E-Mail:
| | - Hartmut Schmidt
- Center for Laboratory Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, Gebaeude 1A, Muenster D-48149, Germany; E-Mail:
| | - Reinhard Voss
- Interdisciplinary Center for Clinical Research, University of Muenster, Albert-Schweitzer-Campus 1, Gebaeude D3, Domagkstrasse 3, Muenster D-48149, Germany; E-Mail:
| | - Ulf Titze
- Gerhard-Domagk Institute of Pathology, University Hospital Muenster, Domagkstrasse 17, Muenster D-48149, Germany; E-Mail:
| | - Mahmoud Abbas
- Institute of Pathology, University Hospital Hannover, Carl-Neuberg-Strasse 1, Hannover D-30625, Germany; E-Mail:
| | - Joerg Neumann
- Institute of Pathology, Klinikum Osnabrueck, Am Finkenhuegel 1, Osnabrueck D-49076, Germany; E-Mail:
| | - Elke Eltze
- Institute of Pathology, Saarbrücken-Rastpfuhl, Rheinstrasse 2, Saarbrücken D-66113, Germany; E-Mail:
| | - Agnes Marije Hoogland
- Department of Pathology, Erasmus Medical Center, ‘s-Gravendijkwal 230, 3015-CE Rotterdam, The Netherlands; E-Mail:
| | - Guido Jenster
- Department of Urology, Erasmus Medical Center, ‘s-Gravendijkwal 230, 3015-CE Rotterdam, The Netherlands; E-Mail:
| | - Burkhard Brandt
- Institute for Clinical Chemistry, University Clinic Schleswig-Holsteins, Arnold-Heller-Strasse 3, Haus 17, Kiel D-24105, Germany; E-Mail:
| | - Axel Semjonow
- Prostate Center, Department of Urology, University Hospital Muenster, Albert-Schweitzer-Campus 1, Gebaeude 1A, Muenster D-48149, Germany; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +49-251-83-47443; Fax: +49-251-83-45540
| |
Collapse
|
14
|
Analysis of genetic aberrations on chromosomal region 8q21–24 identifies E2F5 as an oncogene with copy number gain in prostate cancer. Med Oncol 2013; 30:465. [DOI: 10.1007/s12032-013-0465-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 01/09/2013] [Indexed: 10/27/2022]
|
15
|
O'Neill AK, Niederst MJ, Newton AC. Suppression of survival signalling pathways by the phosphatase PHLPP. FEBS J 2012; 280:572-83. [PMID: 22340730 DOI: 10.1111/j.1742-4658.2012.08537.x] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The recently discovered pleckstrin homology (PH) domain leucine-rich repeat protein phosphatase (PHLPP) family is emerging as a central component in suppressing cell survival pathways. Originally discovered in a rational search for a phosphatase that directly dephosphorylates and inactivates Akt, PHLPP is now known to potently suppress cell survival both by inhibiting proliferative pathways and by promoting apoptotic pathways. In the first instance, PHLPP directly dephosphorylates a conserved regulatory site (termed the hydrophobic motif) on Akt, protein kinase C and S6 kinase, thereby terminating signalling by these pro-survival kinases. In the second instance, PHLPP dephosphorylates and thus activates the pro-apoptotic kinase Mst1, thereby promoting apoptosis. PHLPP is deleted in a large number of cancers and the genetic deletion of one isozyme in a PTEN (phosphatase and tensin homologue located on chromosome 1) +/- (or heterozygous) prostate cancer model results in increased tumourigenesis, underscoring the role of PHLPP as a tumour suppressor. This review summarizes the targets and cellular actions of PHLPP, with emphasis on its role as a tumour suppressor in the oncogenic phosphoinositide 3-kinase (PI3K)/Akt signalling cascade.
Collapse
Affiliation(s)
- Audrey K O'Neill
- Biomedical Sciences Graduate Program, University of California, San Diego, CA 92093-0721, USA
| | | | | |
Collapse
|
16
|
Cheng I, Levin AM, Tai YC, Plummer S, Chen GK, Neslund-Dudas C, Casey G, Rybicki BA, Witte JS. Copy number alterations in prostate tumors and disease aggressiveness. Genes Chromosomes Cancer 2012; 51:66-76. [PMID: 21965145 PMCID: PMC3209417 DOI: 10.1002/gcc.20932] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 08/24/2011] [Indexed: 12/22/2022] Open
Abstract
Detecting genomic alterations that result in more aggressive prostate cancer may improve clinical treatment and our understanding of the biology underlying this common but complex disease. To this end, we undertook a genome-wide copy number alterations (CNAs) study of clinicopathological characteristics of 62 prostate tumors using the Illumina 1M single nucleotide polymorphism array. The highest overall frequencies of CNAs were on chromosomes 8q (gains), 8p (loss and copy-neutral), and 6q (copy-loss). Combined loss and copy-neutral events were associated with increasing disease grade (P = 0.03), stage (P = 0.01), and diagnostic prostate specific antigen (PSA) (P = 0.01). Further evaluation of CNAs using gene ontology identified pathways involved with disease aggressiveness. The "regulation of apoptosis" pathway was associated with stage of disease (P = 0.004), while the "reproductive cellular process" pathway was associated with diagnostic PSA (P = 0.00038). Specific genes within these pathways exhibited strong associations with clinical characteristics; for example, in the apoptosis pathway BNIP3L was associated with increasing prostate tumor stage (P = 0.007). These findings confirm known regions of CNAs in prostate cancer and localize additional regions and possible genes (e.g., BNIP3L, WWOX, and GATM) that may help to clarify the genetic basis of prostate cancer aggressiveness.
Collapse
Affiliation(s)
- Iona Cheng
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Albert M. Levin
- Department of Biostatistics and Research Epidemiology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Yu Chuan Tai
- Departments of Epidemiology & Biostatistics and Urology, Institute for Human Genetics, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158-9001, USA
| | - Sarah Plummer
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Gary K. Chen
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Christine Neslund-Dudas
- Department of Biostatistics and Research Epidemiology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Graham Casey
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Benjamin A. Rybicki
- Department of Biostatistics and Research Epidemiology, Henry Ford Health System, Detroit, MI 48202, USA
- Population Studies and Prevention Program, Karmanos Cancer Institute, Wayne State University, School of Medicine, Detroit, MI 48201, USA
| | - John S. Witte
- Departments of Epidemiology & Biostatistics and Urology, Institute for Human Genetics, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158-9001, USA
| |
Collapse
|
17
|
Barnabas N, Xu L, Savera A, Hou Z, Barrack ER. Chromosome 8 markers of metastatic prostate cancer in African American men: gain of the MIR151 gene and loss of the NKX3-1 gene. Prostate 2011; 71:857-71. [PMID: 21456068 DOI: 10.1002/pros.21302] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Accepted: 10/05/2010] [Indexed: 12/19/2022]
Abstract
BACKGROUND Radical prostatectomy (RP) is not curative if patients have undetected metastatic prostate cancer. Markers that indicate the presence of metastatic disease would identify men who may benefit from systemic adjuvant therapy. Our approach was to analyze the primary tumors of men with metastatic disease versus organ-confined disease to identify molecular changes that distinguish between these groups. METHODS Patients were identified based on long-term follow-up of serum prostate specific antigen (PSA) levels following RP. We compared the tumors of African American (AA) men with undetectable serum PSA for >9 year after RP (good outcome) versus those of AA men with a rising PSA and recurrence after radiation or androgen ablation or both (poor outcome). We used real-time quantitative PCR to assay gene copy number alterations in tumor DNA relative to patient-matched non-tumor DNA isolated from paraffin-embedded tissue. We assayed several genes located in the specific regions of chromosome 8p and 8q that frequently undergo loss and/or gain, respectively, in prostate cancer, and the androgen receptor gene at Xq12. RESULTS Gain of the MIR151 gene at 8q24.3 (in 33% of poor outcome vs. 6% of good outcome tumors) and/or loss of the NKX3-1 gene at 8p21.2 (in 39% of poor outcome vs. 11% of good outcome tumors) affected 67% of poor outcome tumors, compared to only 17% of good outcome tumors. CONCLUSIONS Copy number gain of the MIR151 gene and/or loss of the NKX3-1 gene in the primary tumor may indicate the presence of metastatic disease.
Collapse
Affiliation(s)
- Nandita Barnabas
- Vattikuti Urology Institute, Henry Ford Hospital, Detroit, Michigan 48202-3450, USA
| | | | | | | | | |
Collapse
|
18
|
Hansen RL, Heeboll S, Ottosen PD, Dyrskjøt L, Borre M. Smarcc1 expression: a significant predictor of disease-specific survival in patients with clinically localized prostate cancer treated with no intention to cure. ACTA ACUST UNITED AC 2010; 45:91-6. [PMID: 21087120 DOI: 10.3109/00365599.2010.530295] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE The clinical outcome of prostate cancer (PC) is extremely variable and therefore difficult to predict at the early stage of the disease. Since curative-intended therapies are bound up with the risk of severe adverse events, identification of new prognostic markers in PC is essential in individualized clinical treatment. The Smarcc1 protein, a part of the intranuclear SWI/SNF complex, is up-regulated in PC, and has been suggested to be implicated in tumour dedifferentiation, progression and biochemical recurrence. This makes Smarcc1 a possible candidate marker for PC survival. MATERIAL AND METHODS Immunohistochemistry was used to measure protein expression levels of Smarcc1in on a tissue microarray containing specimens from 100 patients suffering from clinically localized PC treated with no intention to cure and followed to death. RESULTS The median age at diagnosis was 75.5 years (55-95 years) and the median survival time was 5 years (0.01-15 years). In total, 41 patients (41%) died of PC. Statistically, there was no significant association between Smarcc1 immunostaining (negative/positive) and Gleason score (p = 0.7/0.8) or the clinical T stage (p = 0.9). Positive staining for Smarcc1 in patients with clinically localized PC correlated with a prolonged disease-free survival as opposed to negative staining (p = 0.025). CONCLUSION In patients with clinically localized PC treated without intention of cure, Smarcc1 expression was a statistically significant and independent predictor of disease-specific survival.
Collapse
|
19
|
Su Y, Meador JA, Calaf GM, Proietti De-Santis L, Zhao Y, Bohr VA, Balajee AS. Human RecQL4 helicase plays critical roles in prostate carcinogenesis. Cancer Res 2010; 70:9207-17. [PMID: 21045146 DOI: 10.1158/0008-5472.can-10-1743] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Prostate cancer is the second leading cause of cancer-associated deaths among men in the western countries. Here, we report that human RecQL4 helicase, which is implicated in the pathogenesis of a subset of cancer-prone Rothmund-Thomson syndrome, is highly elevated in metastatic prostate cancer cell lines. Increased RecQL4 expression was also detected in human prostate tumor tissues as a function of tumor grade with the highest expression level in metastatic tumor samples, suggesting that RecQL4 may be a potential prognostic factor for advanced stage of prostate cancer. Transient and stable suppression of RecQL4 by small interfering RNA and short hairpin RNA vectors drastically reduced the growth and survival of metastatic prostate cancer cells, indicating that RecQL4 is a prosurvival factor for prostate cancer cells. RecQL4 suppression led to increased poly(ADP-ribose) polymerase (PARP) synthesis and RecQL4-suppressed prostate cancer cells underwent an extensive apoptotic death in a PARP-1-dependent manner. Most notably, RecQL4 knockdown in metastatic prostate cancer cells drastically reduced their cell invasiveness in vitro and tumorigenicity in vivo, showing that RecQL4 is essential for prostate cancer promotion. Observation of a direct interaction of retinoblastoma (Rb) and E2F1 proteins with RecQL4 promoter suggests that Rb-E2F1 pathway may regulate RecQL4 expression. Collectively, our study shows that RecQL4 is an essential factor for prostate carcinogenesis.
Collapse
Affiliation(s)
- Yanrong Su
- Center for Radiological Research, Department of Radiation Oncology, Columbia University Medical Center, New York, New York 10032, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Li C, Xin W, Sy MS. Binding of pro-prion to filamin A: by design or an unfortunate blunder. Oncogene 2010; 29:5329-45. [PMID: 20697352 DOI: 10.1038/onc.2010.307] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Over the last decades, cancer research has focused on tumor suppressor genes and oncogenes. Genes in other cellular pathways has received less attention. Between 0.5% to 1% of the mammalian genome encodes for proteins that are tethered on the cell membrane via a glycosylphosphatidylinositol (GPI)-anchor. The GPI modification pathway is complex and not completely understood. Prion (PrP), a GPI-anchored protein, is infamous for being the only normal protein that when misfolded can cause and transmit a deadly disease. Though widely expressed and highly conserved, little is known about the functions of PrP. Pancreatic cancer and melanoma cell lines express PrP. However, in these cell lines the PrP exists as a pro-PrP as defined by retaining its GPI anchor peptide signal sequence (GPI-PSS). Unexpectedly, the GPI-PSS of PrP has a filamin A (FLNA) binding motif and binds FLNA. FLNA is a cytolinker protein, and an integrator of cell mechanics and signaling. Binding of pro-PrP to FLNA disrupts the normal FLNA functions. Although normal pancreatic ductal cells lack PrP, about 40% of patients with pancreatic ductal cell adenocarcinoma express PrP in their cancers. These patients have significantly shorter survival time compared with patients whose cancers lack PrP. Pro-PrP is also detected in melanoma in situ but is undetectable in normal melanocyte, and invasive melanoma expresses more pro-PrP. In this review, we will discuss the underlying mechanisms by which binding of pro-PrP to FLNA disrupts normal cellular physiology and contributes to tumorigenesis, and the potential mechanisms that cause the accumulation of pro-PrP in cancer cells.
Collapse
Affiliation(s)
- C Li
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-7288, USA
| | | | | |
Collapse
|
21
|
Qiao M, Wang Y, Xu X, Lu J, Dong Y, Tao W, Stein J, Stein GS, Iglehart JD, Shi Q, Pardee AB. Mst1 Is an Interacting Protein that Mediates PHLPPs' Induced Apoptosis. Mol Cell 2010; 38:512-23. [DOI: 10.1016/j.molcel.2010.03.017] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2009] [Revised: 12/09/2009] [Accepted: 03/17/2010] [Indexed: 11/24/2022]
|
22
|
Snail regulates cell survival and inhibits cellular senescence in human metastatic prostate cancer cell lines. Cell Biol Toxicol 2010; 26:553-67. [PMID: 20397042 DOI: 10.1007/s10565-010-9163-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2009] [Accepted: 03/24/2010] [Indexed: 01/14/2023]
Abstract
The epithelial-mesenchymal transition (EMT) is regarded as an important step in cancer metastasis. Snail, a master regulator of EMT, has been recently proposed to act additionally as a cell survival factor and inducer of motility. We have investigated the function of Snail (SNAI1) in prostate cancer cells by downregulating its expression via short (21-mer) interfering RNA (siRNA) and measuring the consequences on EMT markers, cell viability, death, cell cycle, senescence, attachment, and invasivity. Of eight carcinoma cell lines, the prostate carcinoma cell lines LNCaP and PC-3 showed the highest and moderate expression of SNAI1 mRNA, respectively, as measured by quantitative RT-PCR. Long-term knockdown of Snail induced a severe decline in cell numbers in LNCaP and PC-3 and caspase activity was accordingly enhanced in both cell lines. In addition, suppression of Snail expression induced senescence in LNCaP cells. SNAI1-siRNA-treated cells did not tolerate detachment from the extracellular matrix, probably due to downregulation of integrin α6. Expression of E-cadherin, vimentin, and fibronectin was also affected. Invasiveness of PC-3 cells was not significantly diminished by Snail knockdown. Our data suggest that Snail acts primarily as a survival factor and inhibitor of cellular senescence in prostate cancer cell lines. We therefore propose that Snail can act as early driver of prostate cancer progression.
Collapse
|
23
|
Fiorentino M, Capizzi E, Loda M. Blood and tissue biomarkers in prostate cancer: state of the art. Urol Clin North Am 2010; 37:131-41, Table of Contents. [PMID: 20152526 DOI: 10.1016/j.ucl.2009.11.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The prevalence of prostate cancer (PCa) is high and increases with age. PCa is the most common cutaneous cancer in American men. Prostate-specific antigen (PSA) screening has impacted the detection of PCa and is directly responsible for a dramatic decrease in stage at diagnosis. Gleason score and stage at the time of diagnosis remain the mainstay to predict prognosis, in the absence of more accurate and reliable tissue or blood biomarkers. Despite extensive research efforts, very few biomarkers of PCa have been introduced to date in clinical practice. Even screening with PSA has recently been questioned. A thorough analysis of all tissue and serum biomarkers in prostate cancer research cannot be easily synthesized, and goes beyond the scope of the present article. Therefore the authors focus here on the most recently reported tissue and circulating biomarkers for PCa whose application in clinical practice is either current or expected in the near future.
Collapse
Affiliation(s)
- Michelangelo Fiorentino
- Department of Pathology and Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, D1536, 44 Binney Street, Boston, MA 02115, USA
| | | | | |
Collapse
|
24
|
Chattopadhyay I, Singh A, Phukan R, Purkayastha J, Kataki A, Mahanta J, Saxena S, Kapur S. Genome-wide analysis of chromosomal alterations in patients with esophageal squamous cell carcinoma exposed to tobacco and betel quid from high-risk area in India. Mutat Res 2010; 696:130-8. [PMID: 20083228 DOI: 10.1016/j.mrgentox.2010.01.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2009] [Revised: 01/04/2010] [Accepted: 01/10/2010] [Indexed: 01/19/2023]
Abstract
Genomic alterations such as chromosomal amplifications, deletions and loss of heterozygosity play an important role in the pathogenesis and progression of cancer. Environmental risk factors contribute to the development and progression of tumors by facilitating the loss of tumor suppressor genes and amplification of oncogenes. In this current study, Affymetrix 10K single nucleotide polymorphism (SNP) arrays were used to evaluate genomic alterations in 20 pairs of matched germ-line and tumor DNA obtained from patients with esophageal squamous cell carcinoma (ESCC) from high-risk area of India where tobacco, betel quid and alcohol use are widespread. Twenty-two amplified regions and 16 deleted regions identified across chromosomal arms were biologically relevant. The candidate genes located at amplified regions of chromosomes or low-level gain regions such as PLA2G5 (1p36-p34), COL11A1 (1p21), KCNK2 (1q41), S100A3 (1q21), ENAH (1q42.12), RGS1 (1q31), KCNH1 (1q32-q41), INSIG2 (2q14.1), FGF12 (3q28), TRIO (5p15.2), RNASEN (5p15.2), FGF10 (5p13-p12), EDN1(6p24.1-p22.3), SULF1 (8q13.2-13.3), TLR4 (9q32-q33), TNC (9q33), NTRK2 (9q22.1), CD44 (11p13), NCAM1 (11q23.1), TRIM29 (11q22-q23), PAK1 (11q13-q14) and RAB27A (15q15-q21.1), are found to be associated with cellular migration and proliferation, tumor cell metastasis and invasion, anchorage independent growth and inhibition of apoptosis. The candidate genes located at deleted regions of chromosomes, such as FBLN2 (3p25.1), WNT7A (3p25), DLC1 (8p22), LZTS1 (8p22), CDKN2A (9p21), COL4A1 (13q34), CDK8 (13q12) and DCC (18q21.3), are found to be associated with the suppression of tumor. The suggested candidate genes were mostly involved in potential signaling pathways such as focal adhesion (COL4A1), tight junction (CLDN10), MAPK signaling pathway (FGF12) and neuroactive ligand receptor interaction pathway (CCKAR). Expression of FGF12 and COL4A1 was validated by tissue microarray. These unique copy number alteration profiles should be taken into consideration when developing biomarkers for the early detection of ESCC in high-risk areas of India in association with tobacco and betel quid use.
Collapse
|
25
|
Watson SK, Woolcock BW, Fee JN, Bainbridge TC, Webber D, Kinahan TJ, Lam WL, Vielkind JR. Minimum altered regions in early prostate cancer progression identified by high resolution whole genome tiling path BAC array comparative hybridization. Prostate 2009; 69:961-75. [PMID: 19267368 DOI: 10.1002/pros.20949] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Carcinoma of the prostate (CaP) is a serious health problem. The altered molecular mechanisms that lead to this disease are poorly understood. METHODS Specimens from radical prostatectomies and blood were collected from 18 CaP surgery patients. For CGH studies, 20 CaP-related samples (16 Gleason grade 3, 3 higher grades, 1 BPH sample) and 18 samples of patient-matched normal epithelial cells were obtained by laser-assisted microdissection from frozen sections of the 18 prostatectomy specimens. High resolution SMRT aCGH was used to compare genomic profiles of prostatic samples to patient-matched blood and pooled female DNA. TMPRSS2-ERG fusion transcript analysis was performed by RT-PCR in relation to alterations detected at the TMPRSS2 locus. RESULTS Our comprehensive aCGH approach allowed us to define 35 regions of recurrent alterations while excluding germline copy number polymorphisms. Novel regions identified include 2q14.2, containing INHBB, and 17q21.31. The TMPRSS2 locus at 21q22.3 may be a hotspot for rearrangements with 75% of the alterations resulting in the expression of a TMPRSS2-ERG fusion transcript. Differences in fusion expression in different areas in an individual tumor focus and expression in adjacent normal epithelium supported intrafocal heterogeneity and field cancerization, respectively. Both features challenge our efforts to develop more objective markers for diagnosis and prediction of the severity of CaP. CONCLUSION The high-density array enabled precise mapping of genomic alterations and consequently definition of minimum altered regions smaller than previously reported thus facilitating identification of those genes that contribute to the cancer transformation process.
Collapse
Affiliation(s)
- Spencer K Watson
- Department of Cancer Genetics and Developmental Biology, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Huang YT, Heist RS, Chirieac LR, Lin X, Skaug V, Zienolddiny S, Haugen A, Wu MC, Wang Z, Su L, Asomaning K, Christiani DC. Genome-wide analysis of survival in early-stage non-small-cell lung cancer. J Clin Oncol 2009; 27:2660-7. [PMID: 19414679 PMCID: PMC2690391 DOI: 10.1200/jco.2008.18.7906] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Accepted: 12/02/2008] [Indexed: 01/02/2023] Open
Abstract
PURPOSE Lung cancer, of which 85% is non-small-cell (NSCLC), is the leading cause of cancer-related death in the United States. We used genome-wide analysis of tumor tissue to investigate whether single nucleotide polymorphisms (SNPs) in tumors are prognostic factors in early-stage NSCLC. PATIENTS AND METHODS One hundred early-stage NSCLC patients from Massachusetts General Hospital (MGH) were used as a discovery set and 89 NSCLC patients collected by the National Institute of Occupational Health, Norway, were used as a validation set. DNA was extracted from flash-frozen lung tissue with at least 70% tumor cellularity. Genome-wide genotyping was done using the high-density SNP chip. Copy numbers were inferred using median smoothing after intensity normalization. Cox models were used to screen and validate significant SNPs associated with the overall survival. RESULTS Copy number gains in chromosomes 3q, 5p, and 8q were observed in both MGH and Norwegian cohorts. The top 50 SNPs associated with overall survival in the MGH cohort (P < or = 2.5 x 10(-4)) were selected and examined using the Norwegian cohort. Five of the top 50 SNPs were validated in the Norwegian cohort with false discovery rate lower than 0.05 (P < .016) and all five were located in known genes: STK39, PCDH7, A2BP1, and EYA2. The numbers of risk alleles of the five SNPs showed a cumulative effect on overall survival (P(trend) = 3.80 x 10(-12) and 2.48 x 10(-7) for MGH and Norwegian cohorts, respectively). CONCLUSION Five SNPs were identified that may be prognostic of overall survival in early-stage NSCLC.
Collapse
MESH Headings
- Aged
- Biomarkers, Tumor/genetics
- Cadherins/genetics
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Non-Small-Cell Lung/pathology
- Chromosomes, Human, Pair 3
- Chromosomes, Human, Pair 5
- Chromosomes, Human, Pair 8
- Cohort Studies
- Female
- Gene Expression Profiling/methods
- Gene Expression Regulation, Neoplastic
- Gene Frequency
- Genetic Predisposition to Disease
- Genome-Wide Association Study
- Humans
- Intracellular Signaling Peptides and Proteins/genetics
- Kaplan-Meier Estimate
- Lung Neoplasms/genetics
- Lung Neoplasms/mortality
- Lung Neoplasms/pathology
- Male
- Massachusetts/epidemiology
- Middle Aged
- Neoplasm Staging
- Norway/epidemiology
- Nuclear Proteins/genetics
- Oligonucleotide Array Sequence Analysis
- Polymorphism, Single Nucleotide
- Predictive Value of Tests
- Prognosis
- Proportional Hazards Models
- Protein Serine-Threonine Kinases/genetics
- Protein Tyrosine Phosphatases/genetics
- Protocadherins
- RNA Splicing Factors
- RNA-Binding Proteins/genetics
- Reproducibility of Results
- Risk Assessment
- Risk Factors
- Time Factors
Collapse
Affiliation(s)
- Yen-Tsung Huang
- From the Department of Epidemiology, Department of Biostatistics and Department of Environmental Health, Harvard School of Public Health; the Department of Pathology, Brigham and Women's Hospital; and the Cancer Center and the Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA; and the Section of Toxicology, Department of Biological and Chemical Working Environment, National Institute of Occupational Health, Oslo, Norway
| | - Rebecca S. Heist
- From the Department of Epidemiology, Department of Biostatistics and Department of Environmental Health, Harvard School of Public Health; the Department of Pathology, Brigham and Women's Hospital; and the Cancer Center and the Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA; and the Section of Toxicology, Department of Biological and Chemical Working Environment, National Institute of Occupational Health, Oslo, Norway
| | - Lucian R. Chirieac
- From the Department of Epidemiology, Department of Biostatistics and Department of Environmental Health, Harvard School of Public Health; the Department of Pathology, Brigham and Women's Hospital; and the Cancer Center and the Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA; and the Section of Toxicology, Department of Biological and Chemical Working Environment, National Institute of Occupational Health, Oslo, Norway
| | - Xihong Lin
- From the Department of Epidemiology, Department of Biostatistics and Department of Environmental Health, Harvard School of Public Health; the Department of Pathology, Brigham and Women's Hospital; and the Cancer Center and the Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA; and the Section of Toxicology, Department of Biological and Chemical Working Environment, National Institute of Occupational Health, Oslo, Norway
| | - Vidar Skaug
- From the Department of Epidemiology, Department of Biostatistics and Department of Environmental Health, Harvard School of Public Health; the Department of Pathology, Brigham and Women's Hospital; and the Cancer Center and the Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA; and the Section of Toxicology, Department of Biological and Chemical Working Environment, National Institute of Occupational Health, Oslo, Norway
| | - Shanbeh Zienolddiny
- From the Department of Epidemiology, Department of Biostatistics and Department of Environmental Health, Harvard School of Public Health; the Department of Pathology, Brigham and Women's Hospital; and the Cancer Center and the Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA; and the Section of Toxicology, Department of Biological and Chemical Working Environment, National Institute of Occupational Health, Oslo, Norway
| | - Aage Haugen
- From the Department of Epidemiology, Department of Biostatistics and Department of Environmental Health, Harvard School of Public Health; the Department of Pathology, Brigham and Women's Hospital; and the Cancer Center and the Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA; and the Section of Toxicology, Department of Biological and Chemical Working Environment, National Institute of Occupational Health, Oslo, Norway
| | - Michael C. Wu
- From the Department of Epidemiology, Department of Biostatistics and Department of Environmental Health, Harvard School of Public Health; the Department of Pathology, Brigham and Women's Hospital; and the Cancer Center and the Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA; and the Section of Toxicology, Department of Biological and Chemical Working Environment, National Institute of Occupational Health, Oslo, Norway
| | - Zhaoxi Wang
- From the Department of Epidemiology, Department of Biostatistics and Department of Environmental Health, Harvard School of Public Health; the Department of Pathology, Brigham and Women's Hospital; and the Cancer Center and the Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA; and the Section of Toxicology, Department of Biological and Chemical Working Environment, National Institute of Occupational Health, Oslo, Norway
| | - Li Su
- From the Department of Epidemiology, Department of Biostatistics and Department of Environmental Health, Harvard School of Public Health; the Department of Pathology, Brigham and Women's Hospital; and the Cancer Center and the Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA; and the Section of Toxicology, Department of Biological and Chemical Working Environment, National Institute of Occupational Health, Oslo, Norway
| | - Kofi Asomaning
- From the Department of Epidemiology, Department of Biostatistics and Department of Environmental Health, Harvard School of Public Health; the Department of Pathology, Brigham and Women's Hospital; and the Cancer Center and the Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA; and the Section of Toxicology, Department of Biological and Chemical Working Environment, National Institute of Occupational Health, Oslo, Norway
| | - David C. Christiani
- From the Department of Epidemiology, Department of Biostatistics and Department of Environmental Health, Harvard School of Public Health; the Department of Pathology, Brigham and Women's Hospital; and the Cancer Center and the Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA; and the Section of Toxicology, Department of Biological and Chemical Working Environment, National Institute of Occupational Health, Oslo, Norway
| |
Collapse
|
27
|
Genomic profiling of prostate cancers from African American men. Neoplasia 2009; 11:305-12. [PMID: 19242612 DOI: 10.1593/neo.81530] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2008] [Revised: 01/11/2009] [Accepted: 01/12/2009] [Indexed: 11/18/2022] Open
Abstract
African American (AA) men have a higher incidence and significantly higher mortality rates from prostate cancer than white men, but the biological basis for these differences are poorly understood. Few studies have been carried out to determine whether there are areas of allelic loss or gain in prostate cancers from AA men that are overrepresented in or specific to this group. To better understand the molecular mechanisms of prostate cancer in AA men, we have analyzed 20 prostate cancers from AA men with high-density single-nucleotide polymorphism arrays to detect genomic copy number alterations. We identified 17 regions showing significant loss and 4 regions with significant gains. Most of these regions had been linked to prostate cancer by previous studies of copy number alterations of predominantly white patients.We identified a novel region of loss at 4p16.3, which has been shown to be lost in breast, colon, and bladder cancers. Comparison of our primary tumors with tumors from white patients from a previously published cohort with similar pathological characteristics showed higher frequency of loss of at numerous loci including 6q13-22, 8p21, 13q13-14, and 16q11-24 and gains of 7p21 and 8q24, all of which had higher frequencies in metastatic lesions in this previously published cohort. Thus, the clinically localized cancers from AA men more closely resembled metastatic cancers from white men. This difference may in part explain the more aggressive clinical behavior of prostate cancer in AA men.
Collapse
|
28
|
Heebøll S, Borre M, Ottosen PD, Dyrskjøt L, Orntoft TF, Tørring N. Snail1 is over-expressed in prostate cancer. APMIS 2009; 117:196-204. [PMID: 19245592 DOI: 10.1111/j.1600-0463.2008.00007.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Transcription factor Snail1 is a mediator of cell migration and survival, and expression is elevated in several cancer types. The Snail1 gene is reportedly amplified in prostate cancer (PC), and we investigated Snail1 expression in PC. Immunohistochemical Snail1 staining was determined on a tissue microarray which includes 327 specimens of PC, 30 specimens from patients with benign prostatic hyperplasia (BPH), benign tissue from 30 PC patients and 15 high-grade prostate intraepithelial neoplasia (high-grade PIN) specimens. Clinicopathological and follow-up data were available for all patients. No BPH specimen and only 21% of benign tissue from PC patients showed high expression of Snail1. Only 7% of high-grade PIN patients expressed a high level of Snail1. In contrast, approximately 50% of PC tissue from patients with PC showed marked nuclear immunostaining. Snail1 immunostaining was significantly associated with Gleason score (p<0.05). Snail1 expression was not correlated to T stage, metastasis at time of diagnosis, risk of or time to recurrence. Snail1 expression was significantly increased in PC with a positive correlation to dedifferentiation, but not to cancer progression or prognosis. The presented data indicate that Snail1 expression is upregulated from the early stages of PC.
Collapse
Affiliation(s)
- Sara Heebøll
- Molecular Diagnostic Laboratory, Department of Clinical Biochemistry, Aarhus University Hospital, Skejby, Brenstrupgaardsvej 100, Aarhus N, Denmark.
| | | | | | | | | | | |
Collapse
|
29
|
Hirano I, Nakamura S, Yokota D, Ono T, Shigeno K, Fujisawa S, Shinjo K, Ohnishi K. Depletion of Pleckstrin homology domain leucine-rich repeat protein phosphatases 1 and 2 by Bcr-Abl promotes chronic myelogenous leukemia cell proliferation through continuous phosphorylation of Akt isoforms. J Biol Chem 2009; 284:22155-22165. [PMID: 19261608 DOI: 10.1074/jbc.m808182200] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The constitutive activation of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway commonly occurs in cancers and is a crucial event in tumorigenesis. Chronic myelogenous leukemia (CML) is characterized by a reciprocal chromosomal translocation (9;22) that generates the Bcr-Abl fusion gene. The PI3K/Akt pathway is activated by Bcr-Abl chimera protein and mediates the leukemogenesis in CML. However, the mechanism by which Bcr-Abl activates the PI3K/Akt pathway is not completely understood. In the present study, we found that pleckstrin homology domain leucine-rich repeat protein phosphatases 1 and 2 (PHLPP1 and PHLPP2) were depleted in CML cells. We investigated the interaction between PHLPPs and Bcr-Abl in CML cell lines and Bcr-Abl+ progenitor cells from CML patients. The Abl kinase inhibitors and depletion of Bcr-Abl induced the expression of PHLPP1 and PHLPP2, which dephosphorylated Ser-473 on Akt1, -2, and -3, resulting in inhibited proliferation of CML cells. The reduction of PHLPP1 and PHLPP2 expression by short interfering RNA in CML cells weakened the Abl kinase inhibitor-mediated inhibition of proliferation. In colony-forming unit-granulocyte, erythroid, macrophage, megakaryocyte; colony-forming unit-granulocyte, macrophage; and burst-forming unit-erythroid, treatment with the Abl kinase inhibitors and depletion of Bcr-Abl induced PHLPP1 and PHLPP2 expression and inhibited colony formation of Bcr-Abl+ progenitor cells, whereas depletion of PHLPP1 and PHLPP2 weakened the inhibition of colony formation activity by the Abl kinase inhibitors in Bcr-Abl+ progenitor cells. Thus, Bcr-Abl represses the expression of PHLPP1 and PHLPP2 and continuously activates Akt1, -2, and -3 via phosphorylation on Ser-473, resulting in the proliferation of CML cells.
Collapse
Affiliation(s)
- Isao Hirano
- Department of Internal Medicine III, Hamamatsu, Shizuoka 431-3192, Japan; Cancer Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Satoki Nakamura
- Department of Internal Medicine III, Hamamatsu, Shizuoka 431-3192, Japan
| | - Daisuke Yokota
- Department of Internal Medicine III, Hamamatsu, Shizuoka 431-3192, Japan; Cancer Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Takaaki Ono
- Department of Internal Medicine III, Hamamatsu, Shizuoka 431-3192, Japan; Cancer Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Kazuyuki Shigeno
- Department of Internal Medicine III, Hamamatsu, Shizuoka 431-3192, Japan
| | - Shinya Fujisawa
- Division of Hematology, Hamamatsu Medical Center, 328 Tomitsuka, Naka-ku, Hamamatsu, Shizuoka 432-8580, Japan
| | - Kaori Shinjo
- Department of Internal Medicine III, Hamamatsu, Shizuoka 431-3192, Japan
| | - Kazunori Ohnishi
- Department of Internal Medicine III, Hamamatsu, Shizuoka 431-3192, Japan; Cancer Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| |
Collapse
|
30
|
Sørensen KD, Wild PJ, Mortezavi A, Adolf K, Tørring N, Heebøll S, Ulhøi BP, Ottosen P, Sulser T, Hermanns T, Moch H, Borre M, Ørntoft TF, Dyrskjøt L. Genetic and Epigenetic SLC18A2 Silencing in Prostate Cancer Is an Independent Adverse Predictor of Biochemical Recurrence after Radical Prostatectomy. Clin Cancer Res 2009; 15:1400-10. [DOI: 10.1158/1078-0432.ccr-08-2268] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
31
|
Sirintrapun SJ, Parwani AV. Molecular Pathology of the Genitourinary Tract: Prostate and Bladder. Surg Pathol Clin 2008; 1:211-36. [PMID: 26837907 DOI: 10.1016/j.path.2008.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The knowledge of cellular mechanisms in tumors of the prostate and bladder has grown exponentially. Molecular technologies have led to the discovery of TMPRSS2 in prostate cancer and the molecular pathways distinguishing low- and high-grade urothelial neoplasms. UroVysion with fluorescence in situ hybridization is already commonplace as an adjunct to cytologic diagnosis of urothelial neoplasms. This trend portends the future in which classification and diagnosis of tumors of the prostate and bladder through morphologic analysis will be supplemented by molecular information correlating with prognosis and targeted therapy. This article outlines tumor molecular pathology of the prostate and bladder encompassing current genomic, epigenomic, and proteonomic findings.
Collapse
Affiliation(s)
- S Joseph Sirintrapun
- Pathology Informatics, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Anil V Parwani
- Department of Pathology, University of Pittsburgh Medical Center Shadyside Hospital, Room WG 07, 5230 Centre Avenue, Pittsburgh, PA 15232, USA.
| |
Collapse
|
32
|
Holcomb IN, Grove DI, Kinnunen M, Friedman CL, Gallaher IS, Morgan TM, Sather CL, Delrow JJ, Nelson PS, Lange PH, Ellis WJ, True LD, Young JM, Hsu L, Trask BJ, Vessella RL. Genomic alterations indicate tumor origin and varied metastatic potential of disseminated cells from prostate cancer patients. Cancer Res 2008; 68:5599-608. [PMID: 18632612 DOI: 10.1158/0008-5472.can-08-0812] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Disseminated epithelial cells can be isolated from the bone marrow of a far greater fraction of prostate-cancer patients than the fraction of patients who progress to metastatic disease. To provide a better understanding of these cells, we have characterized their genomic alterations. We first present an array comparative genomic hybridization method capable of detecting genomic changes in the small number of disseminated cells (10-20) that can typically be obtained from bone marrow aspirates of prostate-cancer patients. We show multiple regions of copy-number change, including alterations common in prostate cancer, such as 8p loss, 8q gain, and gain encompassing the androgen-receptor gene on Xq, in the disseminated cell pools from 11 metastatic patients. We found fewer and less striking genomic alterations in the 48 pools of disseminated cells from patients with organ-confined disease. However, we identify changes shared by these samples with their corresponding primary tumors and prostate-cancer alterations reported in the literature, evidence that these cells, like those in advanced disease, are disseminated tumor cells (DTC). We also show that DTCs from patients with advanced and localized disease share several abnormalities, including losses containing cell-adhesion genes and alterations reported to associate with progressive disease. These shared alterations might confer the capability to disseminate or establish secondary disease. Overall, the spectrum of genomic deviations is evidence for metastatic capacity in advanced-disease DTCs and for variation in that capacity in DTCs from localized disease. Our analysis lays the foundation for elucidation of the relationship between DTC genomic alterations and progressive prostate cancer.
Collapse
Affiliation(s)
- Ilona N Holcomb
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Brognard J, Newton AC. PHLiPPing the switch on Akt and protein kinase C signaling. Trends Endocrinol Metab 2008; 19:223-30. [PMID: 18511290 PMCID: PMC2963565 DOI: 10.1016/j.tem.2008.04.001] [Citation(s) in RCA: 148] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2007] [Revised: 03/26/2008] [Accepted: 04/03/2008] [Indexed: 12/16/2022]
Abstract
The Ser/Thr-specific phosphatase PHLPP [pleckstrin homology (PH) domain leucine-rich repeat protein phosphatase] provides 'the brakes' for Akt and protein kinase C (PKC) signaling. The two isoforms of this recently discovered family, PHLPP1 and PHLPP2, control the amplitude and duration of signaling of Akt and PKC by catalyzing the dephosphorylation of the hydrophobic phosphorylation motif, a C-terminal phosphorylation switch that controls these kinases. Aberrant regulation of either kinase accompanies many diseases, notably diabetes and cancer. By specifically dephosphorylating the hydrophobic motif, PHLPP controls the degree of agonist-evoked signaling by Akt and the cellular levels of PKC. This review focuses on the function of PHLPP1 and PHLPP2 in modulating signaling by Akt and PKC.
Collapse
Affiliation(s)
| | - Alexandra C. Newton
- To whom correspondence should be addressed: (858) 534-4527, FAX: (858) 822-5888,
| |
Collapse
|
34
|
Lin LJ, Wang YF, Zheng CQ, Jin Y, Hu GZ, Liu X, Lin Y. Genome-wide gene loss in human pancreatic cancer cells. Shijie Huaren Xiaohua Zazhi 2008; 16:1849-1854. [DOI: 10.11569/wcjd.v16.i17.1849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate genome-wide loss of heterozygosity (LOH) and homozygous deletion in human pancreatic cancer cell lines.
METHODS: Genome-wide LOH and homozygous deletion in 17 pancreatic cancer cell lines were studied using high-density single nucleotide polymorphism array and the data were analyzed using a special analytical software. PCR was performed to verify homozygous deletion following screening for potential genetic domains associated with development of pancreatic cancer.
RESULTS: A total of 26 homozygous deletions were verified by PCR and the accuracy of the chip was 83.9% (26/31). On average, 1.29 genes were involved in each region. Each pancreatic cancer cell line had different LOH. Different chromosome arms presented with various LOH frequency with the most common abnormalities in 9p and 18q, occurring in 16 cell lines (94.1%).
CONCLUSION: Genome-wide LOH and homozygous deletions are common in pancreatic cancer cell lines, indicating existence of novel tumor suppressor genes.
Collapse
|
35
|
Bibliography. Current world literature. Adrenal cortex. Curr Opin Endocrinol Diabetes Obes 2008; 15:284-299. [PMID: 18438178 DOI: 10.1097/med.0b013e3283040e80] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
36
|
Assié G, LaFramboise T, Platzer P, Bertherat J, Stratakis CA, Eng C. SNP arrays in heterogeneous tissue: highly accurate collection of both germline and somatic genetic information from unpaired single tumor samples. Am J Hum Genet 2008; 82:903-15. [PMID: 18355774 DOI: 10.1016/j.ajhg.2008.01.012] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2007] [Revised: 01/28/2008] [Accepted: 01/29/2008] [Indexed: 11/17/2022] Open
Abstract
SNP arrays provide reliable genotypes and can detect chromosomal aberrations at a high resolution. However, tissue heterogeneity is currently a major limitation for somatic tissue analysis. We have developed SOMATICs, an original program for accurate analysis of heterogeneous tissue samples. Fifty-four samples (42 tumors and 12 normal tissues) were processed through Illumina Beadarrays and then analyzed with SOMATICs. We demonstrate that tissue heterogeneity-related limitations not only can be overcome but can also be turned into an advantage. First, admixture of normal cells with tumor can be used as an internal reference, thereby enabling highly sensitive detection of somatic deletions without having corresponding normal tissue. Second, the presence of normal cells allows for discrimination of somatic from germline aberrations, and the proportion of cells in the tissue sample that are harboring the somatic events can be assessed. Third, relatively early versus late somatic events can also be distinguished, assuming that late events occur only in subsets of cancer cells. Finally, admixture by normal cells allows inference of germline genotypes from a cancer sample. All this information can be obtained from any cancer sample containing a proportion of 40-75% of cancer cells. SOMATICs is a ready-to-use open-source program that integrates all of these features into a simple format, comprehensively describing each chromosomal event.
Collapse
Affiliation(s)
- Guillaume Assié
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | | | | | | | | | | |
Collapse
|
37
|
Lu T, Hano H. Deletion at chromosome arms 6q16-22 and 10q22.3-23.1 associated with initiation of prostate cancer. Prostate Cancer Prostatic Dis 2008; 11:357-61. [PMID: 18227855 DOI: 10.1038/pcan.2008.4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Loss of heterozygosity (LOH) at 6q16-22 and 10q22.3-23.1 is common chromosomal alteration in advanced prostate cancer and suggests that one or more tumor suppressor genes may lie within these chromosome arms. However, the genetic changes in early stage prostate cancer and premalignant lesions remain to be investigated. We used 11 informative microsatellite markers at 6q16-22 and 10q22.3-23.1 in Japanese patients to compare the frequency of LOH in 53 lesions of high-grade prostatic intraepithelial neoplasia (HGPIN), 38 cases (38 lesions) of incidental prostate cancer (IPC) and 107 cases (168 lesions) of clinical prostate cancer (CPC). The frequency of LOH at 6q16-22 with at least one marker was 38 and 49% in IPC and CPC cases, respectively. Similarly, allelic loss at 10q22.3-23.1 was present in 35 and 39% of IPC and CPC, respectively. High-frequency LOH was detected in both the clinically insignificant and significant prostate cancers at 6q16-22 and 10q22.3-23.1 (P>0.05). However, no allelic loss was detected in any markers at the same regions in HGPIN (0%), which is usually considered a premalignant lesion to prostate cancer. Deletions of both the chromosome regions, 6q16-22 and 10q22.3-23.1, are more likely important events in the initiation and/or promotion of prostate cancer.
Collapse
Affiliation(s)
- T Lu
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan.
| | | |
Collapse
|
38
|
Lamy P, Andersen CL, Dyrskjot L, Torring N, Wiuf C. A Hidden Markov Model to estimate population mixture and allelic copy-numbers in cancers using Affymetrix SNP arrays. BMC Bioinformatics 2007; 8:434. [PMID: 17996079 PMCID: PMC2206057 DOI: 10.1186/1471-2105-8-434] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Accepted: 11/09/2007] [Indexed: 01/27/2023] Open
Abstract
Background Affymetrix SNP arrays can interrogate thousands of SNPs at the same time. This allows us to look at the genomic content of cancer cells and to investigate the underlying events leading to cancer. Genomic copy-numbers are today routinely derived from SNP array data, but the proposed algorithms for this task most often disregard the genotype information available from germline cells in paired germline-tumour samples. Including this information may deepen our understanding of the "true" biological situation e.g. by enabling analysis of allele specific copy-numbers. Here we rely on matched germline-tumour samples and have developed a Hidden Markov Model (HMM) to estimate allelic copy-number changes in tumour cells. Further with this approach we are able to estimate the proportion of normal cells in the tumour (mixture proportion). Results We show that our method is able to recover the underlying copy-number changes in simulated data sets with high accuracy (above 97.71%). Moreover, although the known copy-numbers could be well recovered in simulated cancer samples with more than 70% cancer cells (and less than 30% normal cells), we demonstrate that including the mixture proportion in the HMM increases the accuracy of the method. Finally, the method is tested on HapMap samples and on bladder and prostate cancer samples. Conclusion The HMM method developed here uses the genotype calls of germline DNA and the allelic SNP intensities from the tumour DNA to estimate allelic copy-numbers (including changes) in the tumour. It differentiates between different events like uniparental disomy and allelic imbalances. Moreover, the HMM can estimate the mixture proportion, and thus inform about the purity of the tumour sample.
Collapse
Affiliation(s)
- Philippe Lamy
- Bioinformatics Research Center, University of Aarhus, Hoegh-Guldbergsgade 10, Bldg 1090, 8000 Aarhus C, Denmark.
| | | | | | | | | |
Collapse
|
39
|
Heaphy CM, Bisoffi M, Griffith JK. Diagnostic significance of allelic imbalance in cancer. EXPERT OPINION ON MEDICAL DIAGNOSTICS 2007; 1:159-68. [PMID: 23489303 DOI: 10.1517/17530059.1.2.159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Allelic imbalance (AI), a deviation from the normal 1:1 ratio of maternal and paternal alleles, occurs in virtually all solid and blood-borne malignancies. The frequency and spectrum of AI in a tumor cell reflects the karyotypic complexity of the cancer genome. Hence, many investigations have assessed the extent of AI to analyze differences between normal and tumor tissues in a variety of different organs. In this review, the authors describe established and emerging technologies used to assess the extent of AI in human tissues, and their application in the diagnosis of cancer. The four major methods to be reviewed represent powerful and widely used tools for the identification of allelic imbalances accompanying cancer initiation and progression. These are fluorescent in situ hybridization, comparative genomic hybridization, single nucleotide polymorphism arrays and the use of microsatellite markers. For each method, the authors provide a brief description of the approach and elaborate on specific studies that highlight its utility in the diagnosis of human cancers.
Collapse
Affiliation(s)
- Christopher M Heaphy
- Graduate Research Assistant, University of New Mexico School of Medicine, Department of Biochemistry and Molecular Biology, MSC08 4670, 1 University of New Mexico, Albuquerque, New Mexico 87131-0001, USA +1 505 272 5090 ; +1 505 272 6587 ;
| | | | | |
Collapse
|
40
|
D'Amico F, Biancolella M, Margiotti K, Reichardt JK, Novelli G. Genomic biomarkers, androgen pathway and prostate cancer. Pharmacogenomics 2007; 8:645-61. [PMID: 17559353 DOI: 10.2217/14622416.8.6.645] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Prostate cancer is the most frequent male malignancy diagnosed in western countries and the second leading cause of cancer-related deaths. The growth and function of the prostate gland depends on androgens. Owing to the importance of androgens in prostate development, genes involved in androgen biosynthesis and metabolism have been extensively studied. In this review, we address recent progress toward the use of inherited and acquired genetic variants to predict susceptibility and clinical outcomes of prostate cancer patients. Many of these genetic variants involve several genes related to the biosynthesis and metabolism of androgens, such as steroid-5-α-reductase, α polypeptide 2 (SRD5A2), cytochrome P450 (CYP)19A1, CYP17A1, hydroxy-δ-5-steroid dehydrogenase, 3 β- and steroid δ-isomerase 2 (HSD3B2) and androgen receptor (AR). With increasing knowledge, it may be possible to distinguish indolent from aggressive prostate tumors by molecular fingerprinting. Furthermore, with the emergence of new investigative tools, such as microarray platforms and comparative genomic hybridization (CGH) array, a variety of new genomic biomarkers will be available in the future to provide accurate prognostic and monitoring solutions for individualized patient care.
Collapse
Affiliation(s)
- Franca D'Amico
- Dipartimento di Biopatologia e Diagnostica per Immagini, Università di Roma Tor Vergata, 00133 Roma, Italy
| | | | | | | | | |
Collapse
|
41
|
Sørensen FJ, Andersen CL, Wiuf C. SNPTools: a software tool for visualization and analysis of microarray data. Bioinformatics 2007; 23:1550-2. [PMID: 17384422 DOI: 10.1093/bioinformatics/btm122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
SUMMARY We have created a software tool, SNPTools, for analysis and visualization of microarray data, mainly SNP array data. The software can analyse and find differences in intensity levels between groups of arrays and identify segments of SNPs (genes, clones), where the intensity levels differ significantly between the groups. In addition, SNPTools can show jointly loss-of-heterozygosity (LOH) data (derived from genotypes) and intensity data for paired samples of tumour and normal arrays. The output graphs can be manipulated in various ways to modify and adjust the layout. A wizard allows options and parameters to be changed easily and graphs replotted. All output can be saved in various formats, and also re-opened in SNPTools for further analysis. For explorative use, SNPTools allows various genome information to be loaded onto the graphs. AVAILABILITY The software, example data sets and tutorials are freely available from http://www.birc.au.dk/snptools
Collapse
Affiliation(s)
- Frank J Sørensen
- Bioinformatics Research Center, University of Aarhus, Høegh Guldbergs Gade 10, Aarhus C, Denmark
| | | | | |
Collapse
|
42
|
Sørensen KD, Borre M, Ørntoft TF, Dyrskjøt L, Tørring N. Chromosomal deletion, promoter hypermethylation and downregulation ofFYN in prostate cancer. Int J Cancer 2007; 122:509-19. [DOI: 10.1002/ijc.23136] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
43
|
Lamy P, Andersen CL, Dyrskjot L, Torring N, Orntoft T, Wiuf C. Are microRNAs located in genomic regions associated with cancer? Br J Cancer 2006; 95:1415-8. [PMID: 17003783 PMCID: PMC2360608 DOI: 10.1038/sj.bjc.6603381] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
We report on the location of 283 miRNAs in the human genome in relation to copy number changes in three distinct types of tumours: prostate, bladder and colon. In prostate and colon tumours, we find miRNAs over-represented in regions with copy number gain and under-represented in regions with copy number loss. Surprisingly this pattern appears to be reversed in bladder cancer. We compared our miRNA copy number data to published miRNA expression data; unexpectedly, we did not find a statistically significant relationship between miRNA copy number and expression level. This suggests that miRNA expression is regulated through different mechanisms than mRNA expression.
Collapse
Affiliation(s)
- P Lamy
- Bioinformatics Research Center (BiRC), University of Aarhus, Hoegh-Guldbergs Gade 10, Bldg 1090, 8000 Aarhus C, Denmark
| | - C L Andersen
- Molecular Diagnostic Laboratory, Aarhus University Hospital, Skejby, Brendstrupgaardsvej 100, 8200 Aarhus N, Denmark
| | - L Dyrskjot
- Molecular Diagnostic Laboratory, Aarhus University Hospital, Skejby, Brendstrupgaardsvej 100, 8200 Aarhus N, Denmark
| | - N Torring
- Molecular Diagnostic Laboratory, Aarhus University Hospital, Skejby, Brendstrupgaardsvej 100, 8200 Aarhus N, Denmark
| | - T Orntoft
- Molecular Diagnostic Laboratory, Aarhus University Hospital, Skejby, Brendstrupgaardsvej 100, 8200 Aarhus N, Denmark
| | - C Wiuf
- Bioinformatics Research Center (BiRC), University of Aarhus, Hoegh-Guldbergs Gade 10, Bldg 1090, 8000 Aarhus C, Denmark
- Molecular Diagnostic Laboratory, Aarhus University Hospital, Skejby, Brendstrupgaardsvej 100, 8200 Aarhus N, Denmark
- E-mail:
| |
Collapse
|